+ All Categories
Home > Documents > Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/...

Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/...

Date post: 27-Jun-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
287
Preparation and In Vitro Evaluation of a Polymer Based Controlled Release Dry Powder Inhaler Formulation for Pulmonary Delivery A thesis submitted in fulfilment of the requirements for the degree of Doctor of Philosophy By Mohammad Didare Alam Muhsin B. Pharm. (Honours), M. Pharm. Institute of Health and Biomedical Innovation School of Clinical Sciences Faculty of Health Queensland University of Technology Brisbane, Australia May, 2014
Transcript
Page 1: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Preparation and In Vitro Evaluation of a Polymer Based Controlled Release Dry Powder Inhaler

Formulation for Pulmonary Delivery

A thesis submitted in fulfilment of the requirements for the degree of Doctor

of Philosophy

By

Mohammad Didare Alam Muhsin B. Pharm. (Honours), M. Pharm.

Institute of Health and Biomedical Innovation

School of Clinical Sciences

Faculty of Health

Queensland University of Technology

Brisbane, Australia

May, 2014

Page 2: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 3: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Abstract

I

Abstract Dry powder inhalers (DPIs) are easier to use, more stable and efficient systems with better

lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention

of researchers in the field of pulmonary drug delivery. Present-day research in this area is

predominantly directed towards designing controlled release DPI formulations using

polymeric micro- or nanoparticles. Nanoparticulate systems can escape mucociliary

clearance and phagocytic removal that favour a prolonged residence of the delivered

formulation in the lungs for drug release. The natural polymer chitosan has been found

highly promising as a matrix for fabricating micro-/ nanoparticle based controlled release

DPI formulations. Analogous to conventional DPIs, the major challenge with formulation of a

controlled release DPI is cohesive agglomeration and consequent poor aerosolization of

micronized particles. Recent research has shown that addition of L-leucine to a chitosan-

based DPI formulation greatly enhances its dispersibility. This observation encouraged to

undertake this work aimed at investigation of the impact of chemical conjugation of

L-leucine to chitosan on dispersibility of particles from a DPI system. The major focus of this

work was to synthesize an L-leucine conjugate of chitosan and prepare controlled release

nanoparticulate DPI formulations thereof in order to compare their aerosolization and drug

release. Diltiazem hydrochloride (DH), an antihypertensive agent, was chosen as the model

drug for this study. The oral administration of this drug suffers from high degree of hepatic

first-pass metabolism and poor bioavailability that make it a good candidate for designing

controlled release DPI formulation. An attempt was also made to investigate in vitro the

safety of chitosan, its L-leucine conjugate and their nanoparticles for respiratory delivery

using a non-malignant bronchial epithelial cell line, BEAS-2B.

The first part of this work involved selective conjugation of L-leucine to the C-2 of chitosan

by reacting with Boc-L-leucine succinimide (Boc-leu-OSu). A protection-deprotection

strategy was adopted with phthalic anhydride and trityl chloride as the protecting groups at

C-2 and C-6 for ensuring regional selectivity. Hydrazine hydrate was used for removal of

phthaloyl protection while trityl and Boc (in Boc-leu-OSu) groups were deprotected

simultaneously by 4M HCl in 1, 4-dioxane. The product was adequately characterized by

Fourier Transform Infrared (FT-IR), 1H, 13C and 2D 1H-13C HSQC Nuclear Magnetic Resonance

Page 4: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Abstract

II

(NMR) spectroscopy, Elemental Analysis and X-ray Photoelectron Spectroscopy (XPS). The

results of the analyses confirmed conjugation of L-leucine to chitosan.

The second part of this work involved preparation of nanoparticle formulation of chitosan

and its L-leucine conjugate and comparison of their aerosolization and drug release

characteristics. Nanoparticles were prepared by two different approaches based on

water-in-oil (W/O) emulsification of aqueous polymeric solution in heavy mineral oil, viz.

W/O emulsion-solvent evaporation and W/O emulsion-glutaradehyde cross-linking. Loading

of the model drug, DH, into the nanoparticles was successfully done by glutaraldehyde

cross-linking technique, but the drug could not be loaded into particles by solvent

evaporation technique. The nanoparticles were characterized for size, shape and surface

morphology by scanning electron microscopy (SEM) and Malvern Zetasizer Nano-S. The drug

loading and entrapment efficiency were estimated by an indirect method based on

quantification of drug lost in the external oil phase by UV spectrophotometry. The

aerosolization of nanoparticles was studied by a Twin Stage Impinger (TSI) with a Rotahaler

at an airflow rate of 65±5 L/min. Drug release study was conducted in phosphate buffered

saline (PBS, pH 7.3±0.2) at 37 °C with gentle stirring and the samples withdrawn at intervals

were quantified by UV spectrophotometry. The drug loading and entrapment efficiency of

conjugate nanoparticles were significantly (p<0.05) higher than those of chitosan

nanoparticles (drug loading: 20±1% vs. 16±1%; entrapment efficiency: 46±1% vs. 38±1%).

The aerosolization study with glutaraldehyde cross-linked particles showed significantly

(p<0.05) higher Fine Particle Fraction (FPF) for both the blank and drug-loaded conjugate

nanoparticles compared to corresponding chitosan nanoparticles (blank nanoparticles:

24±0.8% vs. 19±1.01%; drug-loaded nanoparticles: 21±0.7% vs. 15±1.5%). The conjugate

nanoparticles also showed twice as much higher and more prolonged drug release as

chitosan nanoparticles (52%, 16 days vs. 23%, 8 days).

The third part of this work studied in vitro the effect of chitosan, its L-leucine conjugate and

their nanoparticles on the cell viability, trans-epithelial permeability and chemokine (IL-8)

release by MTT assay, sodium fluorescein transport assay and enzyme-linked

immunosorbent assay (ELISA), respectively using the in vitro cell model, BEAS-2B. For MTT

Page 5: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Abstract

III

assay, a series of concentrations (between 0.125 and 16 mg/mL) of the test samples were

applied to 24-hour old cultures of the cell for a period of 12, 24 and 48 hours. Based on the

MTT assay results, 4 concentrations (0.5, 1, 2 and 4 mg/mL) were chosen to perform sodium

fluorescein transport assay and ELISA. These were the highest concentrations at which, for

most of the test samples, the % survival of treated cells was above 50% and beyond which

the % survival was found to fall abruptly. For sodium fluorescein transport assay, confluent

cell monolayers grown on transwell inserts were apically treated by test samples mixed with

sodium fluorescein and aliquots of medium withdrawn from the basolateral chambers at

various time intervals (0.5-48 h) were analysed by spectrofluorometry. ELISA for IL-8 was

performed on culture supernatants harvested after exposure of the cell cultures to the

samples for 24 h period. The results of MTT assay showed that all the 4 samples had a

concentration- and time-dependent effect on cell viability. Chitosan was found to have low

toxicity with IC50 values of 48, 24 and 17 mg/mL for 12, 24 and 48 h treatment durations,

respectively. Other 3 samples were more toxic, but still showed a low toxicity with IC50

values being in the range of 2-7 mg/mL. In general, the rank order of their cytotoxic effect

was: chitosan nanoparticles < conjugate < conjugate nanoparticles. According to the results

of sodium fluorescein transport assay, chitosan and its nanoparticles did not cause any

significant change in transepithelial permeability (p>0.05). The conjugate produced a mild,

but significant increase in the permeability (p<0.05) up to a concentration of 2 mg/mL.

Paradoxically, cell monolayers treated with conjugate nanoparticles showed lower

permeability than that of control. However, there was a concentration-depended gradual

increase in the permeability, with the highest concentration of 4 mg/mL exhibiting a

permeability close to that of control. According to ELISA performed on cell culture

supernatants, all the 4 samples significantly induced IL-8 release (p<0.05). The rank order of

IL-8 induction of the 4 samples were conjugate < chitosan = chitosan nanoparticles

<conjugate nanoparticles.

To conclude, the body of work presented in this dissertation showed successful conjugation

of L-leucine to chitosan. The nanoparticle formulation prepared from the L-leucine

conjugate was found to be more dispersible from a DPI than that prepared from the parent

chitosan corroborating the hypothesis previously made in this connection. The conjugate

Page 6: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Abstract

IV

was also found to hold a strong promise in controlling drug release. However, nanoparticles

made of the conjugate appeared to be relatively more toxic, but still within an acceptable

limit (IC50 of 2 mg/mL after 48 h exposure). So, the product could be considered to have a

good prospect for controlled release pulmonary drug delivery.

Page 7: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Keywords

V

Keywords Chitosan L-leucine Conjugation Nanoparticle Dry powder inhaler (DPI) Pulmonary delivery Aerosolization Dispersion Controlled release Diltiazem hydrochloride BEAS-2B

Page 8: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

VI

Table of Contents Abstract ............................................................................................................................ I

Keywords ......................................................................................................................... V

Table of Contents ............................................................................................................ VI

List of Figures ................................................................................................................. XII

List of Tables ................................................................................................................. XVI

List of Abbreviations ..................................................................................................... XVII

Statement of Originality ................................................................................................ XXI

Acknowledgements ...................................................................................................... XXII

Dedications ................................................................................................................. XXIV

Publications and Communications ................................................................................ XXV

Chapter 1: Introduction

1.1 Background ....................................................................................................................... 1

1.2 Hypothesis ........................................................................................................................ 4

1.3 Aims of the Project ........................................................................................................... 5

1.3.1 Generic Aim ............................................................................................................................. 5

1.3.2 Specific Aims ............................................................................................................................ 6

Chapter 2: Literature Review

2.1 The Architecture of the Respiratory Airways ................................................................... 7

2.2 The Respiratory Tract as a Portal for Drug Delivery ......................................................... 8

2.3 Different Types of Devices for Respiratory Drug Delivery ............................................. 10

2.4 Formulation Challenges of Successful Drug Delivery from a DPI ................................... 14

2.5 Use of Aerosolization Enhancers for Maximizing Drug Delivery from a DPI .................. 16

2.6 Controlled Release Dry Powder Inhalers — Current status, Challenges and Future Prospect .......................................................................................................................... 18

2.7 The Role of Biodegradable Polymers in Designing a Controlled Release DPI ................ 20

2.8 Chitosan ― a Promising Natural Biodegradable Polymer for Sustained Release DPI Formulation .................................................................................................................... 20

2.9 Conjugation of L-Leucine with Chitosan ― a Novel Approach for Maximizing Drug Delivery from a Controlled Release DPI ......................................................................... 26

Page 9: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

VII

2.10 The Human Bronchial Epithelial Cell Line, BEAS-2B for the Assessment of the Toxicity and Inflammatory Activity of the DPI formulation ......................................................... 29

2.10.1 MTT Cell Viability Assay ..................................................................................................... 30

2.10.2 Sodium Fluorescein Transport Assay ............................................................................... 33

2.10.3 Release of Proinflammatory Mediators ........................................................................... 35

2.10.4 Interleukin-8 (IL-8) .............................................................................................................. 37

2.10.5 BEAS-2B – the In Vitro Cell Model for the Assessment of Toxicity and Inflammatory Activity .................................................................................................................................. 37

2.11 Model Drug: Diltiazem Hydrochloride (a systemically acting antihypertensive Agent) 38

Chapter 3: General Methods

3.1 REAGENTS AND MATERIALS ........................................................................................... 40

3.1.1 For Synthesis .......................................................................................................................... 40

3.1.2 For Pharmaceutical Studies ................................................................................................... 40

3.1.3 For Cell Line Studies ............................................................................................................... 41

3.2 METHODS ....................................................................................................................... 41

3.2.1 Conjugation of Chitosan with L-Leucine ................................................................................ 42

3.2.1.1 Characterization ............................................................................................................. 42

3.2.1.1.1 Fourier Transform Infrared (FTIR) Spectroscopy ................................................... 43

3.2.1.1.2 Nuclear Magnetic Resonance (NMR) Spectroscopy .............................................. 43

3.2.1.1.3 Elemental Analysis ................................................................................................. 44

3.2.1.1.4 X-ray Photoelectron Spectroscopy (XPS) ............................................................... 44

3.2.1.2 Synthesis ........................................................................................................................ 44

3.2.1.2.1 N-Phthaloyl-chitosan (2) ........................................................................................ 46

3.2.1.2.2 N-Phthaloyl-3,6-Di-O-acetyl-chitosan (2a) ............................................................. 46

3.2.1.2.3 N-Phthaloyl-6-O-trityl-chitosan (3) ........................................................................ 47

3.2.1.2.4 N-Phthaloyl-3-O-acetyl-6-O-trityl-chitosan (3a)..................................................... 47

3.2.1.2.5 6-O-Trityl-chitosan (4) ............................................................................................ 48

3.2.1.2.6 6-O-Trityl-chitosan-N-Boc-L-leucine (5) .................................................................. 48

3.2.1.2.7 Chitosan-N-L-leucine.HCl (6) .................................................................................. 49

Page 10: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

VIII

3.2.2 Preparation of Chitosan and Chitosan-L-Leucine Conjugate Nanoparticles and In Vitro Studies on their Aerosolization Performance and Drug Release .......................................... 50

3.2.2.1 Preparation of Chitosan and Chitosan-L-leucine Conjugate Nanoparticles ........... 50

3.2.2.1.1 W/O Emulsion-Solvent Evaporation Method ........................................................ 50

3.2.2.1.2 W/O Emulsion-Glutaraldehyde Crosslinking Method ............................................ 51

3.2.2.2 Scanning Electron Microscopy ....................................................................................... 52

3.2.2.3 Zetasizer Analysis ........................................................................................................... 53

3.2.2.4 Estimation of Production Yield, Drug Loading and Entrapment Efficiency ................... 53

3.2.2.4.1 Production Yield ..................................................................................................... 53

3.2.2.4.2 Drug Loading and Entrapment Efficiency ............................................................... 54

3.2.2.5 In Vitro Drug Release Study ........................................................................................... 54

3.2.2.6 In Vitro Evaluation of Aerosolization and Lung Deposition ........................................... 56

3.2.3 In Vitro Evaluation of Toxicity and Inflammatory Activity on the Pulmonary Epithelial Cell Line BEAS-2B .......................................................................................................................... 59

3.2.3.1 Cell line ........................................................................................................................... 59

3.2.3.2 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay ...................................................................................................................... 59

3.2.3.3 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B Cell Monolayers ..................................... 61

3.2.3.3.1 Determination of the Polarisation Time of the BEAS-2B Cell Culture by Trans-Epithelial Electrical Resistance (TEER) Measurement .................................. 61

3.2.3.3.2 Comparison of the Permeability of a Transwell Containing BEAS-2B Cell Monolayer and a Blank Transwell by Sodium Fluorescein Transport Assay .......... 62

3.2.3.3.3 Effect of Chitosan, Conjugate and their Nanoparticles on the Permeability of BEAS-2B Cell Monolayer......................................................................................... 62

3.2.3.4 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study ........... 64

3.2.4 Statistical Analysis ................................................................................................................. 65

Chapter 4: Method Validation

4.1 Summary ......................................................................................................................... 66

4.2 Analytical Validation ....................................................................................................... 66

4.2.1 UV Spectrophotometric Assay ............................................................................................... 66

4.2.2 Spectrofluorometric Assay .................................................................................................... 68

4.2.3 Enzyme-linked Immunosorbent Assay (ELISA) ...................................................................... 69

Page 11: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

IX

4.2.4 In Vitro Aerosolization Study ................................................................................................. 70

4.2.5 Preparation of Nanoparticles — Batch-to-Batch Variability ................................................ 70

Chapter 5: Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

5.1 INTRODUCTION .............................................................................................................. 72

5.2 RESULTS AND DISCUSSION ............................................................................................. 73

5.2.1 Synthesis and Characterization of N-Phthaloyl-Chitosan ...................................................... 74

5.2.2 Synthesis and Characterization of N-Phthaloyl-6-O-Trityl-Chitosan ..................................... 80

5.2.3 Synthesis and Characterization of 6-O-Trityl-Chitosan ......................................................... 83

5.2.4 Synthesis and Characterization of 6-O-Trityl-Chitosan-N-Boc-L-Leucine .............................. 85

5.2.5 Synthesis and Characterization of Chitosan-N-L-Leucine.HCl ............................................... 89

5.3 CONCLUSION .................................................................................................................. 94

Chapter 6: Preparation of Chitosan and Chitosan-L-leucine Conjugate Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

6.1 INTRODUCTION .............................................................................................................. 95

6.2 RESULTS .......................................................................................................................... 96

6.2.1 Preparation of Chitosan and Chitosan-L-leucine Conjugate Nanoparticles .......................... 96

6.2.2 Morphology and Particle Size Analysis .................................................................................. 96

6.2.2.1 Scanning Electron Microscopy ....................................................................................... 96

6.2.2.2 Zetasizer Analysis ........................................................................................................... 97

6.2.3 Production Yield, Drug Loading and Entrapment Efficiency ............................................... 100

6.2.4 In Vitro Drug Release Study ................................................................................................. 100

6.2.5 In Vitro Aerosolization Study ............................................................................................... 105

6.3 DISCUSSION .................................................................................................................. 111

6.3.1 Preparation of Chitosan and Chitosan-L-Leucine Conjugate Nanoparticles........................ 111

6.3.2 Particle Size and Morphology .............................................................................................. 112

6.3.3 Production Yield, Drug Loading and Entrapment Efficiency ............................................... 115

6.3.4 In Vitro Drug Release Study ................................................................................................. 116

6.3.5 In Vitro Aerosolization Study ............................................................................................... 121

6.4 CONCLUSION ................................................................................................................ 125

Page 12: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

X

Chapter 7: In Vitro Evaluation of Toxicity and Inflammatory Activity on the Pulmonary Epithelial Cell Line Beas-2B

7.1 INTRODUCTION ............................................................................................................ 127

7.2 RESULTS ........................................................................................................................ 128

7.2.1 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay .................................................................................................................................... 128

7.2.1.1 MTT Assay of Chitosan, Chitosan-L-Leucine Conjugate and their Nanoparticles ........ 128

7.2.1.2 MTT Assay of Diltiazem Hydrochloride ........................................................................ 133

7.2.2 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B cell Monolayer ............................................. 134

7.2.2.1 Determination of the Polarisation Point of the BEAS-2B Cell Culture by TEER Measurement .............................................................................................................. 134

7.2.2.2 Comparison of the Permeability of a Transwell Containing Confluent BEAS-2B Cell Monolayer and a Blank Transwell................................................................................ 134

7.2.2.3 Effect of Chitosan, Conjugate and their Nanoparticles on Transport of Na Flu across BEAS-2B Cell Monolayer .............................................................................................. 135

7.2.3 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study ................. 141

7.3 DISCUSSION .................................................................................................................. 142

7.3.1 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay .................................................................................................................................... 142

7.3.2 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B Cell Monolayer ............................................. 146

7.3.3 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study ................. 148

7.4 CONCLUSION ....................................................................................................................... 149

Chapter 8: Overall Conclusions and Further Directions

8.1 Summary .......................................................................................................................... 151

8.1.1 Conjugation of L-leucine with Chitosan ................................................................................... 152

8.1.2 Preparation and Characterization of Chitosan and Conjugate Nanoparticles and their Comparison in Terms of Aerosolization and Drug Release .................................................... 155

8.1.3 Toxicity and Inflammatory Effect ............................................................................................ 157

8.2 Overall Conclusion ........................................................................................................... 158

8.3 Limitation of this Study .................................................................................................... 159

Page 13: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Table of Contents

XI

8.4 Future Directions ............................................................................................................. 159

8.4.1 Synthesis of an L-Leucine Conjugate at C-6 of Chitosan or a Bis-Conjugate at C-2 and C-6……. ............................................................................................................................................. 159

8.4.2 Conjugation of Chitosan with other Amino Acids ............................................................... 160

8.4.3 Loading the Drug into Blank Nanoparticles ......................................................................... 163

8.4.4 Emulsion- Solvent Evaporation Technique for Direct Loading of other Drugs .................... 164

8.4.5 Aerodynamic Particle Sizing of the Aerosol Plume ............................................................. 164

8.4.6 Mixing of Nanoparticles with Interactive Carrier Particles ................................................. 165

8.4.7 Incorporation of Lactose, Mannitol or Similar Substances into the Nanoparticles as a Component of the Matrix .................................................................................................... 165

8.4.8 Incorporation of Antiadherents into the Nanoparticles...................................................... 166

8.4.9 Stability Study of the Nanoparticles .................................................................................... 166

Bibliography……..……………………………………………………………………………………………………168-211

Appendices

Appendix 5-1 .......................................................................................................................... 212

Appendix 5-2 .......................................................................................................................... 216

Appendix 5-3 .......................................................................................................................... 219

Appendix 5-4 .......................................................................................................................... 221

Appendix 5-5 .......................................................................................................................... 223

Appendix 6 ............................................................................................................................. 224

Appendix 7-1 .......................................................................................................................... 227

Appendix 7-2 .......................................................................................................................... 228

Appendix 7-3 .......................................................................................................................... 230

Page 14: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Figures

XII

List of Figures Figure 1- 1: Hypothesized Inter-particle Interaction Following Conjugation of L-leucine to Chitosan ... 4 Figure 2- 1: Human Respiratory System ................................................................................................. 7 Figure 2- 2 : Structure of the Respiratory Airways according to the Model of Weibel ........................... 8 Figure 2- 3: Nebulizer ............................................................................................................................ 11 Figure 2- 4: Metered Dose Inhaler (MDI) .............................................................................................. 12 Figure 2- 5: Dry Powder Inhaler (DPI) formulation ............................................................................... 13 Figure 2- 6: Dry Powder Inhaler (DPI) ................................................................................................... 13 Figure 2- 7: Problems Ensuing in a DPI due to Micronization of Particles ............................................ 15 Figure 2- 8: L-Leucine Coating and Inter-particle Interaction ............................................................... 17 Figure 2- 9: Ideal Release Profile of a Controlled Release Dosage Form .............................................. 19 Figure 2- 10: The Structure of Chitosan ................................................................................................ 21 Figure 2- 11: Mechanism of Drug Release from Chitosan Micro- and Nanoparticles .......................... 25 Figure 2- 12: The Structure of L-Leucine ............................................................................................... 26 Figure 2- 13: (a) & (b) Orientation of a surfactant molecule at oil-water and air-water interfaces

respectively, (c) & (d) Micellar orientation of a surfactant molecule in water and non-polar solvents respectively ....................................................................................... 28

Figure 2- 14: Probable orientation of hydrophilic and hydrophobic domains of L-leucine conjugated to chitosan in particle-air (a) and particle-water (b) interfaces. ...................................... 28

Figure 2- 15: Conversion of MTT to Formazan ..................................................................................... 31 Figure 2- 16: The Structure of Diltiazem Hydrochloride ....................................................................... 38 Figure 3- 1: A Bird's Eye View of the Whole Project ............................................................................. 42 Figure 3- 2: Synthetic Pathway for Conjugation of L-leucine to Chitosan (1) ....................................... 45 Figure 3- 3: O-Acetylation of N-phthaloyl-chitosan (2) and N-phthaloyl-6-O-trityl-chitosan (3) ......... 45 Figure 3- 4: A Simplified Scheme of Methods Used for the Preparation of Nanoparticles .................. 52 Figure 3- 5: Twin Stage Impinger (TSI) .................................................................................................. 58 Figure 3- 6: A Simplified Scheme of MTT Cell Viability Assay ............................................................... 60 Figure 3- 7: Transwell Insert ................................................................................................................. 61 Figure 3- 8: A Simplified Scheme of Na Flu Transport Assay ................................................................ 63 Figure 3- 9: A Simplified Scheme for Evaluation of IL-8 Induction in BEAS-2B Cells by ELISA .............. 64 Figure 4- 1: UV Scan of Diltiazem Hydrochloride in PBS over the Range of 200-400 nm ..................... 67 Figure 4- 2: Beer-Lambert’s Calibration Curve of Diltiazem Hydrochloride Solution in PBS ................ 67 Figure 4- 3: Calibration Curve of Na Flu in 1:1 RPMI-NaOH (1mM) ...................................................... 68 Figure 4- 4: Calibration Curve for Estimation of Human IL-8 by ELISA ................................................. 69 Figure 4- 5: Comparison of TSI Deposition from 3 Replicates of Chitosan Nanoparticle Formulation

(determined by gravimetric analysis) ................................................................................. 70 Figure 4- 6: (a) Production Yield (%), (b) Drug Loading (%) and (c) Entrapment Efficiency (%) of 3

Batches of Drug-loaded Chitosan Nanoparticles ................................................................ 71 Figure 4- 7: SEM Micrographs of 3 Batches of Drug-loaded Chitosan Nanoparticles (at x100,000

magnification) ..................................................................................................................... 71 Figure 5- 1: Structure of Chitosan and L-Leucine .................................................................................. 72 Figure 5- 2: Hydrogen Bonding in Chitosan and its Disruption by N-Phthaloylation ............................ 74

Page 15: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Figures

XIII

Figure 5- 3: A Combined Presentation of the FT-IR Spectra of: (A) N-Phthaloyl-Chitosan, (B) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan, (C) N-Phthaloyl-6-O-Trityl-Chitosan, (D) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan, (E) 6-O-Trityl-Chitosan, (F) 6-O-Tritylchtiosan-N-Boc-L-Leucine and (G) Chitosan-N-L-Leucine.HCl ......................... 76

Figure 5- 4: Determination of Degree of Substitution (DS) of N-Phthaloyl-Chitosan from C/N Ratio Obtained by Elemental Analysis ......................................................................................... 77

Figure 5- 5: A Combined Presentation of the 1H NMR Spectra of (A) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan (in CDCl3), (B) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan (in CDCl3), (C) 6-O-Trityl-Chitosan (in Pyridine-d5), (D) 6-O-Trityl-Chitosan-N-Boc-L-Leucine (in Pyridine-d5) and (E) Chitosan-N-L-Leucine.HCl (in D2O) ................................................. 78

Figure 5- 6: A Combined Presentation of 13C NMR Spectra of (A) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan (in CDCl3), (B) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan (in CDCl3), (C) 6-O-Trityl-Chitosan-N-Boc-L-Leucine (in Pyridine-d5) and (D) Chitosan-N-L-Leucine.HCl (in D2O) ................................................................................................................................ 82

Figure 5- 7: 1H-13C HSQC NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5 ........ 88 Figure 5- 8: 1H-13C HSQC NMR Spectrum of Chitosan-N-L-Leucine.HCl in D2O ..................................... 90 Figure 5- 9: Structure of Chitosan, L-Leucine and and Chitosan-N-L-Leucine.HCl ................................ 91 Figure 5- 10: XPS Multiplex spectra of Chitosan, L-Leucine and Chitosan-N-L-leucine.HCl for Nitrogen

.......................................................................................................................................... 92 Figure 5- 11: XPS Multiplex Spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl for Carbon

.......................................................................................................................................... 93 Figure 5- 12: (a) C/N and (b) C/O Ratios of Chitosan-N-L-Leucine.HCl at Different DS Ratios .............. 94 Figure 6- 1: Scanning Electron Micrographs of A) Blank Chitosan Nanoparticles (Method-1),

B) Blank Conjugate Nanoparticles (Method-1), C) Blank Chitosan Nanoparticles (Method-2), D) Blank Conjugate Nanoparticles (Method-2), E) Drug-loaded Chitosan Nanoparticles (Method-2) and F) Drug-loaded Conjugate Nanoparticles (Method-2) at X100,000 magnification ...................................................................................................... 98

Figure 6- 2: Particle Size Distribution of A) Blank Chitosan Nanoparticles (Method-1), B) Blank Conjugate Nanoparticles (Method-1), C) Blank Chitosan Nanoparticles (Method-2), D) Blank Conjugate Nanoparticles (Method-2), E) Drug-loaded Chitosan Nanoparticles (Method-2) and F) Drug-loaded Conjugate Nanoparticles (Method-2) ...... 99

Figure 6-3. 1: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Zero Order Model) ........ 102

Figure 6-3. 2: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (1st Order Model) ........... 103

Figure 6-3. 3: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Higuchi Model) .............. 103

Figure 6-3. 4: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Hixson-Crowell Model) .. 104

Figure 6-3. 5: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Peppas-Korsmeyer Model) ........................................................................................................................................ 104

Figure 6-4. 1: Particle Deposition in Different Stages of TSI (estimated by gravimetric analysis) ...... 106 Figure 6-4. 2: Recovered Doses (RD) from Different Formulations (estimated by gravimetric analysis

of TSI depositions) .......................................................................................................... 106

Page 16: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Figures

XIV

Figure 6-4. 3: Emitted Doses (ED) from Different Formulations (estimated by gravimetric analysis of TSI depositions) .............................................................................................................. 107

Figure 6-4. 4: Fine Particle Fraction (FPF) of Different Formulations (estimated by gravimetric analysis of TSI depositions) .......................................................................................................... 107

Figure 6-5. 1: Drug Deposition in Different Stages of TSI (estimated by UV spectrophotometric analysis) .......................................................................................................................... 108

Figure 6-5. 2: Recovered Doses (RD) from Different Formulations (determined by UV spectrophotometric analysis of TSI depositions) ........................................................... 108

Figure 6-5. 3: Emitted Doses (RD) from Different Formulations (estimated by UV spectrophotometric analysis of TSI depositions) ............................................................................................ 109

Figure 6-5. 4: Fine Particle Fraction (FPF) of Different Formulations (estimated by UV spectrophotometric analysis of TSI depositions) ........................................................... 109

Figure 6-6. 1: Particle Deposition in Different Stages of TSI from Inteactive Mixtures of Drug-loaded Chitosan and Conjugate Nanoparticles with Lactose Monohydrate (estimated by gravimetric analysis) ....................................................................................................... 110

Figure 6-6. 2: (A) Recovered Dose (RD), (B) Emitted Dose (ED) and (C) Fine Particle Fraction (FPF) of Inteactive Mixtures of Drug-loaded Chitosan and Conjugate Nanoparticles with Lactose Monohydrate (estimated by gravimetric analysis) ........................................................ 110

Figure 7-1.1 a: Effect of Chitosan on the Viability of BEAS-2B Cell Line ............................................. 129 Figure 7-1.1 b: Effect of Chitosan Nanoparticle on the Viability of BEAS-2B Cell Line ....................... 130 Figure 7-1.1 c: Effect of Chitosan-L-Leucine Conjugate on the Viability of BEAS-2B Cell Line ............ 131 Figure 7-1.1 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticle on the Viability of BEAS-2B Cell

Line ............................................................................................................................... 132 Figure 7-1. 2: Effect of Diltiazem HCl on the Viability of BEAS-2B Cell Line ........................................ 133 Figure 7-2. 1: Time Course of TEER Development in BEAS-2B Cell Monolayers Grown on Transwell

Inserts ............................................................................................................................. 134 Figure 7-2. 2: Permeability of Na Flu across a Blank Transwell and a Transwell Containing Confluent

BEAS-2B Cell Monolayer ................................................................................................. 135 Figure 7-2.3 a: Effect of Chitosan on Na Flu Transport across the BEAS-2B Cell Monolayer ............. 136 Figure 7-2.3 b: Effect of Chitosan Nanoparticle on Na Flu Transport across the BEAS-2B Cell

Monolayer .................................................................................................................... 137 Figure 7-2.3 c: Effect of Chitosan-L-Leucine Conjugate on Na Flu Transport across the BEAS-2B Cell

Monolayer .................................................................................................................... 138 Figure 7-2.3 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticle on Na Flu Transport across the

BEAS-2B Cell Monolayer .............................................................................................. 139 Figure 7-2. 4: Comparison of the Effect of Chitosan, Chitosan-L-Leucine Conjugate and their Nanoparticles on Na Flu Transport across the BEAS-2B Cell Monolayer…..…………………140 Figure 7- 3: Effect of Chitosan, its L-Leucine Conjugate and their Nanoparticles on IL-8 Release by

BEAS-2B Cells .................................................................................................................... 141 Figure 7- 4: Kinetic Scheme for Drug Disposition from Immediate Release and Controlled Release

Dosage Forms .................................................................................................................... 146 Figure 8- 1: Selective Conjugation of L-Leucine to 6-OH of Chitosan ................................................. 161 Figure 8- 2: Simultaneous conjugation of leucine on C-2 and C-6 of chitosan ................................... 162 Figure 8- 3: A few amino acids with different types of side chains .................................................... 163

Page 17: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Figures

XV

Figure 8- 4: Loading drug into blank chitosan nanoparticles by passive absorption from an aqueous solution ............................................................................................................................................... 164 Figure A5-1. 1: FT-IR (ATR) Spectrum of N-Phthaloyl-Chitosan .......................................................... 212 Figure A5-1. 2: FT-IR (ATR) Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan ................................ 212 Figure A5-1. 3: FT-IR (ATR) Spectrum of N-Phthaloyl-6-O-Trityl-Chitosan.......................................... 213 Figure A5-1. 4: FT-IR (ATR) Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan ....................... 213 Figure A5-1. 5: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan .............................................................. 214 Figure A5-1. 6: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan [run with hydrazine hydrate solution

(40-60%), diluted further with water (1:1)] ................................................................. 214 Figure A5-1. 7: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine................................... 215 Figure A5-1. 8: FT-IR (ATR) Spectrum of Chitosan-N-L-Leucine.HCl .................................................... 215 Figure A5-2. 1: 1H NMR Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan in CDCl3........................ 216 Figure A5-2. 2: 1H NMR Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan in CDCl3............... 216 Figure A5-2. 3: 1H NMR Spectrum of 6-O-Trityl-Chitosan in Pyridine-d5 ............................................ 217 Figure A5-2. 4: 1H NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5 ................. 217 Figure A5-2. 5: 1H NMR spectrum of Chitosan-N-L-leucine.HCl in D2O ............................................... 218 Figure A5-3. 1: 13C NMR Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan in CDCl3 ....................... 219 Figure A5-3. 2: 13C NMR Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan in CDCl3 .............. 219 Figure A5-3. 3: 13C NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5 ................ 220 Figure A5-3. 4: 13C NMR Spectrum of Chitosan-N-L-Leucine.HCl in D2O ............................................. 220 Figure A5-4. 1: Determination of Degree of Substitution (DS) of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan

from C/N Ratio Obtained by Elemental Analysis ......................................................... 221 Figure A5-4. 2: Determination of Degree of Trityl Substitution (DS) of N-Phthaloyl-6-O-Trityl-Chitosan

from C/N Ratio Obtained by Elemental Analysis ......................................................... 221 Figure A5-4. 3: Determination of Degree of Dephthaloylation of 6-O-Trityl-Chitosan from C/N Ratio

Obtained by Elemental Analysis .................................................................................. 222 Figure A5-4. 4: Determination of Degree of Substitution of 6-O-Trityl-Chitosan-N-Boc-L-Leucine from

C/N Ratio Obtained by Elemental Analysis .................................................................. 222 Figure A5-4. 5: Determination of Degree of Trityl and Boc Deprotection in Chitosan-N-L-Leucine.HCl

from C/N Ratio Obtained by Elemental Analysis ......................................................... 222 Figure A5-5. 1: XPS Survey spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl ................. 223 Figure A5-5. 2: XPS Multiplex spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl for Oxygen

..................................................................................................................................... 223

Page 18: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Tables

XVI

List of Tables Table 4- 1: Accuracy and Precision for UV assay of Diltiazem Hydrochloride ...................................... 67 Table 4- 2: Accuracy and Precision for Spectrofluorometric Assay of Na Flu in 1:1 RPMI-NaOH (1mM)

............................................................................................................................................... 68 Table 4- 3: Accuracy and Precision for Estimation of Human IL-8 by ELISA (n=2) .............................. 69 Table 5- 1: Stoichiometric and Experimental C/N And C/O Ratios of Chitosan and the Synthesized

Chitosan-N-L-Leucine.HCl ...................................................................................................... 93 Table 6- 1: Production Yield, Drug Loading and Entrapment Efficiency of Chitosan and Conjugate

Nanoparticles....................................................................................................................... 100 Table 6- 2: Mathematical Models Applied to the Release Data of Diltiazem Hydrochloride Loaded

into Chitosan and Conjugate Nanoparticles ........................................................................ 101 Table 6- 3: Release Rate Constants and Determination Coefficients for Drug Release Profile according

to Various Kinetic Models .................................................................................................... 102 Table A6- 1: Zetasizer Analysis of Blank and Drug-loaded Chitosan and Conjugate Nanoparticles ... 224 Table A6- 2: In Vitro Release of Diltiazem HCl from Chitosan and Conjugate Nanoparticles in PBS at

37 °C .................................................................................................................................. 225 Table A6- 3: Aerosolization Study of Blank and Drug-Loaded Chitosan and Conjugate Nanoparticles

Using Twin-Stage Impinger (TSI) ....................................................................................... 226 Table A7-1.1 a: MTT Assay of Chitosan on BEAS-2B Cell Line ............................................................. 227 Table A7-1.1 b: MTT Assay of Chitosan Nanoparticles on BEAS-2B Cell Line ..................................... 227 Table A7-1.1 c: MTT Assay of Chitosan-L-Leucine Conjugate on BEAS-2B Cell Line ........................... 227 Table A7-1.1 d: MTT Assay of Chitosan-L-Leucine Conjugate Nanoparticles on BEAS-2B Cell Line ... 227 Table A7-1. 2: MTT Assay of Diltiazem HCl on BEAS-2B Cell Line ....................................................... 227 Table A7-2. 1: TEER of BEAS-2B Cell Lines Grown on Transwell Inserts at Different Time Intervals .. 228 Table A7-2. 2: Permeability of Na Flu across a Blank Transwell and a Transwell Containig Confluent

BEAS-2B Monolayer ........................................................................................................ 228 Table A7-2.3 a: Effect of Chitosan on Na Flu Transport across the BEAS-2B Monolayer ................... 228 Table A7-2.3 b: Effect of Chitosan Nanoparticles on Na Flu Transport across the BEAS-2B Monolayer

..................................................................................................................................... 228 Table A7-2.3 c: Effect of Chitosan-L-Leucine Conjugate on Na Flu Transport across the BEAS-2B

Monolayer .................................................................................................................... 229 Table A7-2.3 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticles on Na Flu Transport across the

BEAS-2B Monolayer ..................................................................................................... 229 Table A7- 3: ELISA for IL-8 Released by BEAS-2B Cells upon Treatment with Chitosan, its L-Leucine

Conjugate and their Nanoparticles ................................................................................... 230

Page 19: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Abbreviations

XVII

List of Abbreviations 2D Two Dimensional

Ad12SV40 Adenovirus 12- Simian Virus 40 hybrid

ANOVA Analysis of Variance

arom Aromatic

ATR Attenuated Total Reflection

BALF Bronchioalveolar Lavage Fluid

BALF-P Bronchoalveolar Lavage Fluid Protein

BDP Beclomethasone Dipropionate

BE Binding Energy

BEAS-2B S.6 BEAS-2B Sublcone 6

Boc Butyloxycarbonyl

Boc-leu-OSu Boc-L-Leucine Succinimide

BSA Bovine Serum Albumin

ca. Circa (means about)

Calcein AM Calcein Acetoxymethyl Ester

CCM Cell Culture Medium

CFC Chlorofluorocarbon

COPD Chronic Obstructive Pulmonary Disease

cps Centipoise

cST Centistoke

CV Coefficient of Variation

DDA Degree of Deacetylation

DLS Dynamic Light Scattering

DMF N, N-Dimethylformamide

DMSO Dimethyl Sulfoxide

DMSO-d6 Hexadeuterodimethyl sulfoxide

DPI Dry Powder Inhaler

DS Degree of Substitution

DH Diltiazem Hydrochloride

Page 20: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Abbreviations

XVIII

ECH Epichlorohydrin

ED Emitted Dose

EDTA Ethylenediaminetetraacetic acid

EGDE Ethylene Glycol Diglycidyl Ether

ELISA Enzyme-Linked Immunosorbent Assay

ERS Electrical Resistance System

Et2O Di-ethyl Ether

EtOH Ethanol

eV Electron Volt

FBS Foetal Bovine Serum

FD Fluorescein Isothiocyanate Dextran

FD10 Fluorescein Isothiocyanate Dextran 10

FD4 Fluorescein Isothiocyanate Dextran 4

FDA Food and Drug Administration

FITC Fluorescein Isothiocyanate

FPF Fine Particle Fraction

FTIR Fourier Transform Infrared

GL Glutaraldehyde

GM-CSF Granulocyte Macrophage Colony Stimulating Factor

He–Ne Helium-Neon

HFA Hydrofluoroalkane

HMW High Molecular Weight

HPC Hydroxypropyl Cellulose

HSQC Heteronuclear Single Quantum Correlation

IC50 Median Inhibitory Concentration

IL-1 Interleukin -1

IL-6 Interleukin-6

Inc. Incorporated

IRE Internal Reflection Element

KBr Potassium Bromide

LB Line Broadening

Page 21: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Abbreviations

XIX

LD50 Median Lethal Dose

LDH Lactate Dehydrogenease

leu Leucine

LMW Low Molecular Weight

LOD Limit of Detection

LOQ Limit of Quantitation

mA Milliampere

MIP-2 Macrophage Inflammatory Protein-2

mg/kg Milligram/Kilogram

mg/mL Milligram/Milliliter

MHz Megahertz

MIP-1α Macrophage Inflammatory Protein-1α

MMAD Mass Median Aerodynamic Diameter

MMW Medium Molecular Weight

MPO Myeloperoxidase

MTT 3-(4,5 dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide

MW Molecular Weight

mW Milliwatt

MXF Moxifloxacin

Na Flu Sodium fluorescein

NADH Nicotinamide Adenine Dinucleotide

NADPH Nicotinamide Adenine Dinucleotide Phosphate

NHAc N-acetyl

nm Nanometer

NMR Nuclear Magnetic Resonance

Nothridge, CA Northridge, California

OAc O-Acetyl

Papp Apparent Permeability Coefficient

PCCA Professional Compounding Chemists of Australia

PDI Polydispersity Index

PEG Polyethylene Glycol

Page 22: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

List of Abbreviations

XX

phth Phthaloyl

PKC Protein Kinase C

PLA Poly Lactic Acid

PLGA Poly(D,L-lactide-co-glycolide)

ppm Parts Per Million

PVA Polyvinyl Alcohol

PVD Physical Vapour Deposition

Pyridine-d5 Deuterated Pyridine

r.nm Radius in Nanometer

r.t. Room Temperature

RD Recovered Dose

rpm Rotation Per Minute

RPMI Rosewell Park Memorial Institute

S1 Stage-1

S2 Stage-2

SE Standard Error

SEM Scanning Electron Microscopy

SV40 Simian Virus 40

TEER Transepithelial Electrical Resistance

TMC Trimethyl Chitosan Chloride

TNF-α Tumour Necrosis Factor-α

Tr Trityl

TS Terbutaline Sulfate

UV Ultraviolet

v/v Volume/Volume

VSMC Vascular Smooth Muscle Cell

w/w Weight/Weight

WD Working Distance

W/O Water-in-Oil

XPS X-ray Photoelectron Spectroscopy

ZO-1 Zonula Occludens-1

Page 23: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Statement of Originality

XXI

Statement of Originality The studies described in this thesis have not previously been submitted for a degree or

diploma at this or any other university or tertiary institution. To the best of my knowledge, it

contains no material previously published or written except where due reference has been

made.

Date: 05.05.2014

QUT Verified Signature

Page 24: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Acknowledgements

XXII

Acknowledgements In the Name of Allah, the Most Beneficent, the Most Merciful

I would like to express the deepest appreciation to my principal supervisor, Dr Nazrul Islam

for giving me an opportunity to pursue a PhD under him. I am grateful to him for

recommending me for International Postgraduate Research Scholarship (IPRS) and

Queensland University of Technology Postgraduate Research Award (QUT PRA) to pursue

this program. Without his guidance and persistent support this dissertation would not have

been possible.

My sincerest gratitude to Prof Graeme George whose constant encouragement and

enthusiastic support enabled me to face my challenges in polymer work with courage. He

continuously conveyed a spirit of adventure in regard to research for making this work a

success.

Heart-felt thanks to Prof Kenneth Beagley who introduced me to the arena of cell line work.

His intelligent guidance and organizational support played a significant part in advancing the

work forward to this stage.

The involvement of Associate Prof Vito Ferro was an important event in the progress of this

research. His analytical ability and strong command of synthetic chemistry was the

cornerstone in solving the challenges that I faced time and again in carrying out the

polymeric synthesis performed in this work.

I express my gratitude to Dr Mark Wellard, Dr Chris Carvalho, Dr Llew Rintoul, Dr Tim

Dargaville, Dr John Colwell, Dr Ferry Melchels, Dr Peter Hines, Dr Leonore de Boer, Ms

Rachel Hancock, Mr Eric Martinez, Mr Nathaniel Raup, Mr Shane Russel, Mr David Boatfield,

Mr David Smith, Mr Scott Tucker and all others from Queensland University of Technology

(QUT) who helped me in various ways by providing laboratory and technical assistance

during performing this work.

Page 25: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Acknowledgements

XXIII

I am grateful to Dr Barry Wood and Mr George Blazak from University of Queensland who

helped me in performing X-ray photoelectron spectroscopic (XPS) and elemental analysis of

my synthetic products.

I would like to acknowledge Queensland University of Technology (QUT) for granting me

International Postgraduate Research Scholarship (IPRS) and Queensland University of

Technology Postgraduate Research Award (QUT PRA) and the Discipline of Pharmacy and

the Faculty of Health for providing extended financial support during this work. I would like

to specially note here Assoc Prof Faraser Ross (Ex-Head, Pharmacy Descipline) and Prof

Michele Clark (Ex-Head, School of Clinical Sciences) for their support to extend my

scholarship during the extended candidature period.

This acknowledgement will remain incomplete if I do not thank all my colleagues, peers and

friends for their support and help throughout this research. I would like to specially note

Dr Rinku Tuli, Dr Babak Radi and Dr Charles Armitage for their valuable help at different

stages of this work.

Finally, I would like to pay tribute to my deceased father whose memory has always been a

source of inspiration for me. Special thanks to my mother who has always been praying for

my success, my beloved wife, Dr Shamah Marzuqah, who has been deprived of due

attention during this long journey and all other relations whose support and blessings

encouraged me to stay firm during ups and downs of this work.

Page 26: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Dedications

XXIV

Dedications

To

Almighty Allah

and

My beloved parents

Page 27: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Publications and Communications

XXV

Publications and Communications

Conference Abstracts Muhsin, M., George, G., Beagley, K., Ferro, V. and Islam, N. (2013): Effect of chemical

conjugation of L-leucine to chitosan on the dispersibility and drug release of a dry powder inhaler nanoparticle formulation. A poster abstract accepted for presentation at American Association of Pharmaceutical Scientists (AAPS) Annual Meeting and Exposition 2013 being held November 10-14, 2013 in San Antonio, Texas, USA.

Muhsin, M., George, G., Beagley, K., Ferro, V. and Islam, N. (2012): In vitro biocompatibility

study of chitosan, its L-leucine conjugate and their nanoparticles for pulmonary delivery using the human bronchial epithelial cell line, BEAS-2B. A poster presentation in IHBI Inspires Postgrdauate Conference 2012 held November 22-23, 2012 at Gold Coast, Australia.

Muhsin, M., George, G., Beagley, K., Ferro, V. and Islam, N. (2010): Preparation of a

leucine-conjugated chitosan for use in dry powder inhaler (DPI) formulations. An oral presentation in Australian Pharmaceutical Science Association (APSA) Annual Conference 2010 held December 6-9, 2010 at Brisbane, Australia.

Award 2013 International Pharmaceutical Excipients Council (IPEC) Foundation Graduate Student

Scholarship Award

Page 28: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 29: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 30: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 31: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

1

1.1 Background Pulmonary route is receiving increasing acceptance as a convenient, non-invasive way of

drug delivery to the lung tissue and/or the systemic circulation. The route has successfully

been exploited for targeting drugs to the lung in the treatment of asthma and chronic

obstructive pulmonary disease (COPD) and quite a few drugs, such as salbutamol sulfate,

terbutaline sulfate, budesonide etc., are already available for administration through this

route. Because of the unique features of the lung, such as its large surface area, high

permeability and wide blood supply, the route appears to be of great potential for systemic

delivery of poorly absorbable drugs too. Considerable efforts have been made during the

last few decades for developing appropriate formulations for pulmonary delivery of many

therapeutic agents such as insulin, calcitonin, interferons, parathyroid hormone, leuprolide

etc. for systemic action (Agu et al., 2001; Damms & Bains, 1995; Okamoto et al., 2011;

Patton, 1997; Patton, 2000; Shahiwala & Misra, 2005).

The three most common types of devices available for pulmonary drug delivery include dry

powder inhaler (DPI), pressurized metered dose inhaler (pMDI) and nebulizer. pMDIs can

deliver more consistently a predetermined dose of drug (Leach, 2007), but these devices

need synchronization of actuation and inhalation, failing which a significant proportion of

the delivered dose is wasted as oropharyngeal deposition (Newman, 2005). Besides, these

devices require a chemical propellant (e.g. CFC or HFA) to dispense the inhaled

formulations. Nebulizers aerosolize drug from an aqueous solution or suspension and

require a jet of gas or ultrasonic wave as the dispersing force. In general, nebulizers are not

very portable. They are more often used in hospital settings for emergency treatment of

acute pulmonary conditions. DPI is superior to the two other common pulmonary drug

delivery systems in terms of not requiring any propellant or actuation-inhalation

coordination (Li et al., 2005). Moreover, it is easy to use and portable. With the FDA

approval and advent of first inhaled insulin dry powder aerosol, Exubera (Pfizer) in the

market in 2006 (albeit withdrawn later for some unknown reason), dry powder inhaler drug

delivery has become a subject of tremendous interest and there has been an increased drive

in research for developing new systems for delivering therapeutic agents by inhalation for

Page 32: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

2

systemic action (Chougule et al., 2007; Hickey, 2013; Kandasamy & Chandrasekaran, 2013;

Taneja et al., 2012). In addition to insulin, drug molecules that have already been

investigated for delivery by a DPI for systemic action include: anti-malarial vaccine (Edwards

et al., 2005), human growth hormone (Blizzard et al., 2003), morphine (Dershwitz et al.,

2000), meglumine complexes of amphotericin B and mycoheptine (Ekzemplyarov et al.,

1977), heparin (Bai et al., 2010) etc.

An important drawback of many currently available pulmonary therapies is their short

duration of action and need for administration 3-4 times a day. For instance, the effect of

inhaled salbutamol sulfate, one of the most popular bronchodilators used in the treatment

of asthma and COPD, lasts only several hours, requiring repeated administration every 4 to

6 h to sustain a bronchodilator response (Jantikar et al., 2007). To overcome the problem,

an increasing trend is seen for designing these therapies in controlled release forms suitable

for pulmonary delivery (Salama et al., 2009a). Controlled pulmonary therapy could be

particularly beneficial for enhancing systemic bioavailability of drugs, like diltiazem or

morphine, which undergo extensive hepatic first-pass metabolism following oral delivery.

Although there is a continued effort in this direction, no controlled release inhalation

system has yet found its way to the market.

Micro- and nanoparticles of biodegradable polymers have drawn much attention for

pulmonary delivery of drugs in controlled release form (Fiegel et al., 2004; Fu et al., 2002;

Salama et al., 2009a; Sivadas et al., 2008). These systems could be widely varied in size,

shape and other characteristics to control drug release and target the deeper regions of

lungs and obviate the need for removal because they ultimately degrade and are

metabolised by the body. Nanoparticulate systems have two important advantages over

microparticles: first, they can escape mucociliary clearance and second, they can bypass

phagocytic recognition by alveolar macrophages (Ahsan et al., 2002; Grenha et al., 2005).

These attributes can significantly contribute to enhancing residence of these systems in the

lungs for extended drug release. Chitosan, a widely available natural biopolymer, has gained

special attention as matrix for designing controlled release micro- and nanoparticles. This

polymer has been reported to have low toxicity, biocompatibility and biodegradability (Tong

Page 33: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

3

et al., 2009). It has also been reported to have some additional attractive features like

mucoadhesiveness and trans-epithelial permeability enhancement (Amidi et al., 2008b;

Learoyd et al., 2009; Naikwade & Bajaj, 2009). Moreover, chitosan has also been shown to

enhance aerosolization and lung deposition of a DPI formulation (Li & Birchall, 2006).

One important challenge associated with a DPI formulation is cohesive aggregation of

micronized particles that affects the aerosolization and subsequent lung deposition of the

delivered drug. It has been reported that addition of the amino acid, L-leucine to chitosan-

based DPI formulations significantly enhance aerosolization of chitosan particles in vitro

presumably by migration of L-leucine to the particle surface owing to its surfactant-like

structure; this gives the particles a pitted appearance and in turn reduces inter-particle

interaction (Learoyd et al., 2008a, 2009; Rabbani & Seville, 2005; Seville & Learoyd, 2007). It

can be intriguing to investigate the effect of chemical conjugation of L-leucine to chitosan on

aerosolization and lung deposition of such a system.

For any system to be administered by inhalation, it is important to assess their safety in the

respiratory system. The pulmonary epithelium serves as the barrier to the permeation of

any inhaled substance into the lungs (Patton et al., 2004). In addition, it also responds to

toxic substances by releasing various inflammatory mediators (Ghio et al., 2002). Thus, it is

customary to explore in vitro the effect of products intended for inhalation on the

pulmonary epithelium as a primary approach to investigate their safety for the respiratory

administration. Many tests have been reported in the literature to this end, of which three

commonly used tests are MTT assay to assess the effect on the cell viability (Zhang et al.,

2009), sodium fluorescein (Na Flu) transport assay to assess any untoward effect on

trans-epithelial permeability (Grenha et al., 2007) and induction of the inflammatory

mediators such as the chemokine interleukin-8 (IL-8) which is often observed in lung

inflammation (Sivadas et al., 2008).

Based on these considerations, this study attempted to synthesize an L-leucine conjugate of

chitosan and assess the impact of such conjugation on dispersibility and drug release profile

of a nanoparticulate DPI formulation. Diltiazem hydrochloride (DH), an antihypertensive

Page 34: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

4

agent, was chosen as a model drug for the formulations considering its high degree of

hepatic first-pass metabolism giving poor bioavailability (~40%) following oral

administration. The biosafety of the particles was also assessed in vitro in terms of cell

viability, transepithelial permeability and inflammatory chemokine (IL-8) release using a

bronchial epithelial cell line, BEAS-2B. This is a non-malignant cell line and has not been

used before for studying the safety of chitosan or any of its derivatives. So, this study is

expected to complement previous reports in this line on various malignant epithelial cell

lines like calu-3 and A-549.

1.2 Hypothesis Because of the surfactant-like property of L-leucine (Matubayasi et al., 2002; Watry &

Richmond, 2002), upon conjugation to chitosan, it will enhance the dispersibility of the

nanoparticles (made of conjugated chitosan) from a DPI formulation. In addition, the

leucinization of chitosan could also be expected to increase the hydrophilicity of the

polymer and in turn increase the loading of a water-soluble drug, which in consequence

might provide a more prolonged release.

OHO

O

OH

n

NH

NH2

O

Chitosan-N-L-leucine

Figure 1- 1: Hypothesized Inter-particle Interaction Following Conjugation of L-leucine to Chitosan

Hydrophobic Side-chain

Page 35: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

5

The essence of the first part of the hypothesis is enhanced dispersion and lung deposition of

the L-leucine conjugated chitosan particles from a DPI (in comparison to unconjugated

chitosan particles). This is based on the concept that, being surfactant in nature (Matubayasi

et al., 2002; Watry & Richmond, 2002), L-leucine conjugated to chitosan will give an

amphiphilic environment around the chitosan backbone where its hydrophobic domain

would be projected outward giving a pitted appearance to the particle surface and so

reduced inter-particle interaction (Fig. 1-1). This concept stems from the very basic

principles of surfactant chemistry (Sinko, 2006) and has also been supported by

observations of other investigators regarding orientation of L-leucine (Raula et al., 2010) and

its analogue tri-leucine (Lechuga-Ballesteros et al., 2008) at a particle-air interface. The

deposition of L-leucine (added to a particle formulation) onto the surface of the drying

particle giving it a pitted appearance, in turn reducing inter-particle interaction and

enhancing particle dispersion has also been well-documented in the literature (Columbano

et al., 2003; Learoyd et al., 2008a, 2009; Rabbani & Seville, 2005; Seville et al., 2007). The

second part of the hypothesis was concerned with the expected release of the model drug,

DH from L-leucine conjugated chitosan particles compared to those made from chitosan

itself. This is also based on the expected amphiphilic environment created by L-leucine

around the chitosan backbone where its hydrophilic domain is supposed to be associated

with the surrounding media in an aqueous environment, increasing its hydrophilicity and in

turn facilitating increased loading of a water-soluble drug and consequently a more

prolonged release. An elaborate discussion of these aspects making the background of the

hypothesis has been presented in the Paragraph 2 of the Section 2.5 [p. 17] and the

Paragraph 3 of the Section 2.9 [p. 27-29] and associated diagrams [Fig. 2-8, Fig. 2-13 (a-d)

and Fig. 2-14 (a-b)] in the Chapter 2.

1.3 Aims of the Project

1.3.1 Generic Aim Considering the high potential of the natural, biodegradable polymer chitosan as a release

modifier, postulated benefits of conjugating L-leucine to chitosan and probable advantages

of delivering the antihypertensive drug, diltiazem hydrochloride (DH) through the

pulmonary route, this work was aimed at performing research in order to achieve

Page 36: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 1 Introduction

6

controlled release DPI formulations of DH using drug-loaded nanoparticles of chitosan and

its L-leucine conjugate for the management of systemic hypertension. The generic aim of

this study was to gain a better understanding of the mechanism of dispersion and drug

release of a polymer based DPI formulation.

1.3.2 Specific Aims The following specific aims were set forth in order to address the hypothesis proposed for

this research:

(a) To synthesize L-leucine conjugated chitosan

(b) To prepare drug-loaded polymeric nanoparticulate DPI formulation using chitosan and

its L-leucine conjugate

(c) To study the physicochemical properties of polymeric nanoparticulate DPI formulation

prepared from chitosan and its L-leucine conjugate

(d) To investigate in vitro the influence of chemical conjugation of L-leucine with chitosan on

aerosolization, lung deposition, and release-retarding features of a nanoparticle based

DPI formulation

(e) To evaluate in vitro the potential toxicity of the prepared DPI formulation on a

non-malignant bronchial epithelial cell line, BEAS-2B.

The aims (a), (b) and (c) were directed towards chemical conjugation of L-leucine with

chitosan and preparation of the nanoparticulate DPI formulations of chitosan and its

L-leucine conjugate that were prerequisites for performing the experiments relating the aim

(d). The aim (d) was set to test the hypotheses, viz. (a) enhanced dispersibility of

nanoparticles made of conjugated chitosan and (b) increased hydrophilicity of the

conjugated chitosan leading to increased loading and consequent more prolonged release

of a water-soluble drug. The experimental results, as discussed in the Chapter 6, Sections

6.2.3-6.2.5 and 6.3.3-6.3.5, supported the hypotheses. The 5th aim (e) addressed the safety

issues considering that the formulations are meant for administering as a DPI through the

pulmonary route.

Page 37: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 38: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 39: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 40: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

7

2.1 The Architecture of the Respiratory Airways Anatomically, the respiratory system may be divided into two regions: 1. conducting zone

and 2. respiratory zone. The conducting airways consist of the air-transmitting passages of

the nose, nasopharynx, larynx, trachea, bronchi and bronchioles (Gray, 2000; Itoh et al.,

2004). The last three form the tracheo-bronchial region of the lung (generations 1–16 in the

bifurcating airway model) (Fig. 2-2). The primary function of the conducting airways is to act

as conduit for air movement. Additionally, this region provides filtration, warming and

humidification of inhaled air.

Mouth/Nose Trachea Bronchioles Alveoli

Figure 2- 1: Human Respiratory System

[Reproduced from (Fishbein)]

The actual exchange of gases (oxygen and CO2) occurs across the distal lung respiratory

epithelium comprised of the alveolar ducts and saccules (the alveoli, generations 17–23 in

the bifurcating airway model) (Fig.2-2).

Page 41: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

8

Figure 2- 2 : Structure of the Respiratory Airways according to the Model of Weibel

[adapted from: Weibel (1963)]

2.2 The Respiratory Tract as a Portal for Drug Delivery The concept of administering drugs via the lung is not new. It has been used for centuries to

deliver many drugs conveniently and effectively for medical and recreational purposes.

Smoking of tobacco products is one of the most common examples, and before the advent

of modern inhalers, some asthma medications were administered in the form of cigarettes

(Sanjar & Matthews, 2001). But, inhaled medications did not reach the status of being safe,

effective, and convenient enough for widespread public use until 1956 when 3M Riker

(Northridge, CA) scientists invented the metered dose inhaler (MDI) (Leach, 2007). Since

then there has been an enormous effort for using this route for delivery of drugs and, by

this time, inhalation of aerosolized drugs (e.g. β2-agonists, corticosteroids, and mucolytics)

has become a well-established and preferred means of treating localized disease states

within the lung such as asthma and COPD (Learoyd et al., 2008a). The main reason lying

behind this is the fact that local targeting of drugs into the lungs quickens the effect,

reduces the dose required and systemic exposure and, in turn, results in reduced systemic

side effects (Sahib et al., 2012; Shaikh et al., 2010). One can easily envisage the difference

by comparing the 100-200 µg inhalation dose of the anti-asthmatic drug, salbutamol sulfate

to the usual oral dose of 2-4 mg which is larger by about 20 times.

Page 42: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

9

More recently, it has been demonstrated that the lung may also be an ideal site for the

non-invasive delivery of therapeutic molecules, including peptides and proteins, to the

systemic circulation (Agu et al., 2001; Damms & Bains, 1995; Okamoto et al., 2011; Patton,

1997; Patton, 2000; Shahiwala & Misra, 2005). In fact, as a non-invasive route of medication,

pulmonary delivery is drawing greater attention of scientists day by day, because it has

many attractive features including an enormous surface area (almost 140 m2), thin (0.1-0.5

μm) alveolar epithelial membrane, extensive vasculature, low extracellular and intracellular

enzymatic activity and absence of first-pass metabolism, and thus permits rapid absorption

and onset of action (Agu et al., 2001). These features, in addition, may necessitate a

relatively lower dose resulting in reduced systemic side effects compared to other

non-injection routes of administration (Bi et al., 2009). Small molecules deposited in the

lungs are very rapidly absorbed into the systemic circulation, with the fastest uptake of any

route of delivery other than the intravenous route (Patton et al., 2004). Examples of rapidly

absorbed inhaled drugs include nicotine (Burch et al., 1989), rizatriptan (Rabinowitz et al.,

2004), morphine (Dershwitz et al., 2000) and fentanyl (Mather et al., 1998). The use of the

lungs for the delivery of macromolecules like peptides and proteins, which otherwise must

be injected, is one of the most exciting new areas in pulmonary delivery. For reasons that

are not completely understood, the lungs provide higher bioavailability for peptides and

proteins than any other non-invasive route of drug delivery (Patton, 1996; Patton et al.,

1998). However, unlike the situation with small molecules, for which lung metabolism is

minimal, enzymatic hydrolysis of small natural peptides (less than 30 amino acids) can be

very high unless they are chemically engineered (blocked) to inhibit peptidases (Patton et

al., 1998). Exubera, a fast-acting inhaled insulin was the first ever macromolecular inhalable

product to hit the market in January 2006 (Freemantle & Strack, 2006). The product had

glycemic control comparable to subcutaneous insulin (Patton & Byron, 2007), but in less

than 2 years after its introduction the product was withdrawn from the market because of

disappointing sales. Other peptides and proteins investigated for pulmonary delivery include

growth hormone (Bosquillon et al., 2004), parathyroid hormone (Codrons et al., 2004),

erythropoietin (Dumont et al., 2005) and quite a few others (Patton & Byron, 2007).

Page 43: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

10

Although promising, delivery of therapeutics to the lungs faces several anatomical and

physiological challenges (Patton, 1996). To deposit in the lungs, drugs must traverse a

complex lung structure that is heterogeneous in geometry and environment from patient to

patient. Once deposited, natural clearance mechanisms, including the ‘‘mucociliary

escalator’’, work to expel particles from the upper airways (Amidi et al., 2008b; Koushik &

Kompella, 2004), while alveolar macrophages rapidly (often within minutes) engulf particles

between 1 and 5 μm that reach the deep lungs (Koushik & Kompella, 2004). Additional drug

loss may occur in the inhaler device due to inefficient aerosolization, or in the mouth,

throat, and upper airways due to sub-optimal aerosol characteristics or improper

coordination of aerosol activation and breathing (Edwards et al., 1998). Consequently,

optimized formulation can play vital role to maximize delivery efficiency and eliminate

irreproducibility that can limit the practicality of new pulmonary therapies.

2.3 Different Types of Devices for Respiratory Drug Delivery There are three main categories of devices in common use for pulmonary drug delivery, viz.

nebulizers, pressurized metered dose inhalers (pMDIs) and dry powder inhalers (DPIs).

Nebulizers (Fig. 2-3) are probably one of the oldest forms of pulmonary drug delivery

devices (Dolovich et al., 2005). These devices aerosolize the drug from an aqueous

solution/suspension. The device requires a dispersing force (either a jet of gas or ultrasonic

waves) for aerosolization (O'Riordan, 2002). In terms of drug delivery, nebulizers are less

efficient and more time-consuming compared to DPIs (Le Brun et al., 2000). They are not

portable and so their use is limited to the treatment of hospitalized patients (Islam &

Rahman, 2008). However, nebulizers can be designed to make the best use of a patient’s

breathing pattern, by using the so-called ‘breath-assisted nebulizers’ (O'Callaghan & Barry,

1997). Further, with jet nebulizers, adjustments in drug dosing are easier to achieve (Rau,

2002). Thus, Nebulizers may be best suited for inhaling high-dose drugs in hospital settings

with little patient co-ordination/skill.

Page 44: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

11

Figure 2- 3: Nebulizer

pMDIs (also called MDIs) spray a pre-determined dose of drug, dissolved/suspended in

liquefied propellant(s), from a pressurized canister (Fig. 2-4). Since their introduction in

1956 by 3M Riker (Northridge, CA), pMDIs have been the mainstay of the treatment of

asthma for >40 years (Smyth & Hickey, 2005). These devices can more consistently deliver

doses per inhalation (Leach, 2007). But, they require synchronization of actuation and

inhalation to achieve successful lung deposition and, therefore, a large number of drug

particles may deposit onto the oropharyngeal areas due to a lack of co-ordination between

actuation and inhalation. Besides, liquefied chlorofluorocarbons (CFC), the propellants

originally used in pMDI formulations, are believed to have adverse effects on the

stratospheric ozone layer (Sivasakthivel & Reddy, 2011) and have been banned by an

international agreement — The Montreal Protocol (1987) (Smyth & Hickey, 2005).

However, CFCs have since gradually been replaced in MDIs by environment friendly

propellants, hydrofluoroalkanes (HFAs).

Page 45: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

12

Figure 2- 4: Metered Dose Inhaler (MDI)

The latest addition to the respiratory drug delivery technology is Dry Powder Inhalers (DPIs)

(Fig. 2-6). Since its introduction in 1967 (Altounyan, 1967), there has been a continuously

growing interest in this new technology, especially after the signing of the Montreal Protocol

banning CFCs ― the major propellants used in MDIs (Leach, 2007). Basically, DPI

formulations can be categorized into two types; one consists of loose agglomerates of

micronized drug particles (<5 μm) having controlled flow properties, and the other is carrier-

based interactive mixtures that consist of micronized (<5 μm) drug particles mixed with

larger particles of a carrier (e.g. lactose) (Hersey, 1975) (Fig. 2-5). The formulation is

contained within a device, that upon inhalation provides sufficient de-agglomeration forces

to de-aggregate micronized drug particles or separate them from the carrier and, in turn,

results in dispersion and respiratory deposition of a therapeutic dose of micronized drug

particles (Young et al., 2008) (Fig. 2-6). In carrier based DPIs, drug particles (<5 μm) are

blended with a suitable larger carrier in order to improve flow properties and dose

uniformity (French et al., 1996).

Page 46: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

13

A. Loose Agglomerate of Micronized Drug Particles B. Carrier-based Interactive Mixtures

Figure 2- 5: Dry Powder Inhaler (DPI) formulation

In recent years, DPIs represent the most rapidly expanding field in pulmonary drug delivery,

largely as a result of the perceived limitations in pMDIs and nebulisers. Unlike pMDIs, DPIs

avoid problems inherent in the use of propellant gases and the need for coordination of

inhalation and actuation (Hickey et al., 1994). DPIs are also very portable, patient friendly

and easy to use (Geller, 2005). Moreover, since drugs are kept in DPIs in solid state, they

exhibit high physicochemical stability, particularly for proteins and peptides (Manning et al.,

1989).

Figure 2- 6: Dry Powder Inhaler (DPI)

Page 47: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

14

2.4 Formulation Challenges of Successful Drug Delivery from a DPI For efficient delivery of drugs to the deep lungs, aerosol particles should be small enough to

pass through the mouth, throat, and conducting airways and deposit in the deep lung. The

site of particle deposition within the lungs is clearly demarcated based on particle size. The

upper airways (nose, mouth, larynx, and pharynx) and the branched anatomy of the

tracheobronchial tree act as a series of filters for inhaled particles (Byron et al., 1986). Thus,

aerosol particles with mass median aerodynamic diameter (MMAD) > 100 μm are trapped in

the naso/oropharynx and generally do not enter the respiratory tract. Particles with MMAD

> 10 μm come into contact with the upper respiratory tract (above trachea) and are

eliminated quickly by mucociliary clearance. Particles with MMAD in the size range of 5 to

10 μm are deposited in the large ciliated airways, and only particles having an MMAD 1-5

μm reach the alveolar space (Schlesinger, 1985). Although it is generally accepted that the

particles must be in the range of 1-5 µm for effective delivery to the deep lung, an increase

in deep lung deposition is observed as the particle size is reduced still further into the

nanometer range and an efficient particle deposition in the alveoli can also be achieved with

nanoparticles of the size <100 nm (Jain, 2008; Pirooznia et al., 2012; Rogueda & Traini, 2007;

Todoroff & Vanbever, 2011).

From the above discussion it is evident that, for application of drugs to the deeper parts of

the lungs, a micronization process has to be employed to produce particles of 1-5 μm or of

further smaller diameters in the nano range. Unfortunately, such micronization produces

particles of high surface area, resulting in a highly cohesive system where the particles show

a strong tendency of adhering to each other. The flowability and dispersion properties of

these systems are poor, causing manufacturing problems, dose variability and poor

aerosolization/ lung deposition (Malcolmson & Embleton, 1998) (Fig. 2-7).

The most common formulation approach employed to solve the ensuing problem is to blend

the drug with a larger inert carrier, such as α-lactose monohydrate (lactose), giving a drug-

carrier binary mixture (Fig. 2-5). This ordered mixing process aims to reduce the high

cohesion forces present between micronized drug particles, improve powder flow and allow

accurate metering (Hersey, 1975). During the inhalation manoeuvre, energy supplied to

the dry powder inhaler (DPI) by the patient’s inspiration must overcome the adhesive

Page 48: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

15

Figure 2- 7: Problems Ensuing in a DPI due to Micronization of Particles

forces between drug and carrier, allowing liberation of drug from the device and dispersion

of the drug particles into the deep lung. Thus, a balance between the cohesive (drug-drug)

and adhesive (drug-excipient) forces is critically important for optimum fluidization of a

carrier-based DPI formulation. In one hand, excessive adhesive forces may prevent

detachment of the respirable particles from the carrier surfaces, resulting in their deposition

together with carrier particles in the upper airways. On the other hand, strong cohesive

forces may enhance segregation and agglomeration, which could also directly affect the

fluidization and dispersion properties of the formulation (Byron, 1986; Hickey et al., 1994).

To further complicate the issue, the cohesion-adhesion balance in a carrier-based

interactive mixture can additionally be influenced by the drug concentration in the

formulation. Due to differences in the dose size of various drugs, a large variation is

observed in the drug content in commercially available DPI formulations; however, for a

carrier-based interactive mixture the drug content typically ranges from 0.1 to 4%

(Grasmeijer et al., 2013). Interactive mixing of drug and carrier involves a dynamic process

of ordered mixing (drug-carrier association) and randomization (drug-carrier dissociation)

that ultimately determines the overall outcome of the blending process (Staniforth, 1981).

With an increase in drug content, the equilibrium is shifted towards randomization (Hersey,

1975; Staniforth, 1981; Staniforth, 1987) resulting in less homogeneous mixtures that are

more prone to segregation of drug and carrier particles during handling (e.g. filling, storage

and transportation) (Bryan et al., 1979). The upside is that this apparent adverse effect may

come along with better detachment of drug from carrier particles when the mixture is

Page 49: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

16

subjected to detachment forces (Kulvanich & Stewart, 1987). However, there are mixed

reports in the literature regarding the effect of the drug content on the aerosolization

performance of DPI formulations containing lactose as the carrier. While Steckel et al.

(2004) reported an improvement in fine particle fraction (FPF) of salbutamol sulfate upon

increasing the drug content from 0.25% to 2.8% in a lactose-based interactive mixture,

other studies showed unfavorable effects with increased drug contents (Le et al., 2012;

Steckel & Mueller, 1997).

In an attempt to address the problem of poor dipersion of drug from a DPI formulation, a

wide variety of approaches, to engineer the drug or carrier, have been investigated by

formulation scientists. In general, the researchers have primarily focussed on the influence

of the physicochemical properties of the carrier (Steckel et al., 2004; Young et al., 2005) or

the addition of ternary components to the drug/carrier mixture (Begat et al., 2005; Louey et

al., 2003). Many researchers have also focussed on producing particles with increased

stability and reduced interparticulate adhesion with diameters conducive to respiratory

therapy by employing such techniques as supercritical fluid extraction (Jung & Perrut, 2001),

high gravity precipitation (Chiou et al., 2007) and sonication (Kaerger & Price, 2004). In

addition, controlled spray drying, with or without excipients, has also shown promise for

producing drug particles with specific morphology (such as Nektars PulmoSphere) (Duddu et

al., 2002) and density parameters (such as large porous particles) (Edwards et al., 1997). In

spite of these multifarious approaches, the respirable fraction from currently available DPI

formulations still remains not more than 40% (Lavorini, 2013; Shah & Misra, 2004; Tronde et

al., 2008). So, it is evident that a lot of work is yet to be done to optimize delivery of drug

from a DPI.

2.5 Use of Aerosolization Enhancers for Maximizing Drug Delivery from a DPI One of the well-characterized approaches attempted by formulation scientists to increase

aerosolization from DPIs involves the addition of a co-excipient as a ternary component to

binary drug-lactose mixes in order to impart modified surface properties to the primary

excipient particles (Labiris & Dolovich, 2003). Many compounds have been shown to

increase dispersibility of dry powders when employed in this fashion. Among these

compounds, the amino acid, L-leucine and its analogues appeared very promising in

Page 50: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

17

enhancing aerosolization & lung deposition and have drawn a lot of attention of researchers

in this area (Alhusban & Seville, 2009; Learoyd et al., 2009). Other compounds used for this

purposes include: phenylalanine (Li et al., 2005), arginine (Li et al., 2005), cyclodextrins (Li et

al., 2005; Srichana et al., 2001), polyethylene glycols (Ely et al., 2007; Gilani et al., 2004),

Mg stearate (Begat et al., 2009; Begat et al., 2005) and phospholipids (Begat et al., 2009;

Staniforth et al., 2004). These compounds act as lubricants/antiadherents between surfaces,

thus reducing cohesion forces and improving deagglomeration and flowability that in turn

results in increased dispersibility of powders (Najafabadi et al., 2004; Staniforth & Morton,

2002).

Figure 2- 8: L-Leucine Coating and Inter-particle Interaction

In the spray-drying process, because of its predominantly hydrophobic nature (Black &

Mould, 1991) and surfactant-like properties (Glinski et al., 2000), L-leucine migrates to the

surface of the drying droplet and hence influences the surface characteristics of the

resultant particle (Columbano et al., 2003; Rabbani & Seville, 2005), and in turn generates

highly dispersible particles with optimal aerosolization properties (Learoyd et al., 2008a). It

has been observed that accumulation of L-leucine on the spray-dried particle surface

imparts a pitted/wrinkled appearance to the surface that reduces inter-particulate contact

Hydrophobic Hydrophilic

Page 51: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

18

points for aggregation resulting in a reduction of surface forces, which in turn improves

flowability and dispersibility (Learoyd et al., 2009; Seville et al., 2007) (Fig. 2-8). Apart from

changing the surface morphology, L-leucine-induced reduction in the surface free energy

may also contribute to reduced inter-particulate interaction leading to an enhanced

aerosolization of the particles. A number of studies have demonstrated L-leucine’s ability to

reduce the surface free energy, however no direct correlation was observed between

aerosolization performances of particles and dispersive surface energies; the key factor

contributing to enhanced aerosolization appeared to be the change in the surface

morphology, leading to reduced inter-particle contact (Chew et al., 2005; Paajanen et al.,

2009; Raula et al., 2010). Some researchers have also shown concentration-dependent

gradation in the effect of L-leucine on the enhancement of aerosolization of particles from a

dry powder inhaler. For instance, Seville et al. (2007) reported in their study a progressive

increase in the FPF of spray-dried powders for inhalation with an increase in L-leucine

content from 5 to 20% using salbutamol sulphate as a model drug and lactose as a bulking

agent. On the contrary, however, Chew et al. (2005) did not find any correlation between

L-leucine concentration and dispersive surface free energy of disodium chromoglycate

powders co-spray dried with L-leucine.

Recently, Raula et al. (2008) investigated the effect of direct coating of drug particles with

L-leucine vapours by a novel one-step, aerosol flow reactor-based method. They found that,

upon deposition, L-leucine formed a discrete crystalline surface layer around individual

particles. This coated powder was found to flow and disperse well and appeared promising

for use in dry powder inhaler formulations even without the addition of a coarse lactose

carrier. When dispersed from a DPI at 60 L/min air flow without any added carrier, the fine

particle fraction (FPF) of the coated salbutamol sulfate powder was found to be 35-48% and,

upon blending with lactose carriers, the FPF reached as high as 90%.

2.6 Controlled Release Dry Powder Inhalers — Current status, Challenges and Future Prospect

Controlled release delivery (Fig. 2-9) is one of the most advanced and widely acclaimed

therapeutic strategies in the present day medicinal and pharmaceutical arena. These

systems have a number of overwhelming advantages over conventional treatment

Page 52: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

19

modalities including extended duration of action, reduction in dosing frequency, improved

management of therapy, improved compliance, reduction in side effects (Hardy & Chadwick,

2000), together with potential cost savings in the long run (Saks & Gardner, 1997).

Figure 2- 9: Ideal Release Profile of a Controlled Release Dosage Form

Controlled drug release to the lung is an emerging research field that may offer new

opportunities for enhanced clinical responses for both local and systemic treatments.

However, due to the efficiency of lung clearance mechanisms, our ability to control release

of drug within the lung is considered one of the major challenges in pulmonary drug delivery

(Martonen Ted et al., 2005; Smyth & Hickey, 2005). As a consequence of these powerful

clearance mechanisms (as already noted in the section 2.2), researchers are very limited in

their capacity to control drug release in the lung environment. However, by utilizing the

current understanding of lung–particle interactions, some strategies evolved to overcome

these limitations. For instance, pulmonary controlled release can be achieved by using

formulation approaches like liposomes (Wong et al., 2003), polymeric micro-/nanoparticles

(Florea et al., 2006; Jaspart et al., 2007; Yamamoto et al., 2005), swellable microgels (El-

Sherbiny & Smyth, 2012), large/low-density porous particle or aerodynamically light particle

(Edwards et al., 1997), complexation (Aguiar et al., 2004), and drug conjugates (Williams &

Taylor, 1992). However, although a significant amount of research has been directed

towards delivery of drug in controlled release form to the respiratory tract, no product has

yet found its way into the market.

Page 53: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

20

2.7 The Role of Biodegradable Polymers in Designing a Controlled Release DPI The use of biodegradable polymers is an exciting approach to achieve controlled pulmonary

delivery. Polymers can be tailored into micro- or nanoparticulate systems to deliver precise

amount of drugs over a prolonged period of time (Kim & Pack, 2006). In the perspective of

pulmonary delivery, apart from providing controlled drug release, they also offer other

advantages such as increased drug penetration to the distal parts of the lung, prolonged

residence time of the drug in situ, and improved in vivo drug stability (Sivadas, 2010).

Attempts have been made for controlled pulmonary therapy for both local respiratory

disorders and some systemic diseases using polymeric micro-/ nanoparticles. These included

the topical treatment of asthma (Beck-Broichsitter et al., 2010), local infectious diseases

(Suarez et al., 2001), pulmonary hypertension (Kimura et al., 2009), and the systemic use of

insulin (Aguiar et al., 2004), low molecular weight heparin (Patel et al., 2012) and calcitonin

(Yamamoto et al., 2005). A wide range of natural and synthetic polymers have so far been

investigated as carrier materials for sustained pulmonary delivery with encouraging results.

Examples include: chitosan(Learoyd et al., 2009; Naikwade & Bajaj, 2009), poly(dl-lactide-co-

glycolide) (PLGA) (Sung et al., 2009; Ungaro et al., 2006), sodium hyaluronate (Sivadas et al.,

2008), polyvinyl alcohol (PVA)(Salama et al., 2008), poly(ether-anhydride)(Fiegel et al.,

2004), poly lactic acid (PLA) (Coowanitwong et al., 2007), albumin (Li et al., 2001), etc.

2.8 Chitosan ― a Promising Natural Biodegradable Polymer for Sustained Release DPI Formulation

Chitosan (Fig. 2-10) is an amino-polysaccharide produced by alkaline N-deacetylation of the

naturally occurring polymer chitin which is a major component of the shells of

crustaceans like crabs and shrimps and the second most abundant biopolymer after

cellulose in the world. It is a copolymer of (β1→4)-linked D-glucosamine with a smaller

amount of N-acetyl-D-glucosamine from incomplete deacetylation (Armiji & Patel, 1994;

Choi et al., 2004). The glucosamine residue of chitosan has a pKa value of 6-7, resulting in

protonation of the amino groups in acidic condition. As a consequence, chitosan dissolves in

acidic solutions; but, due to suppression of ionization of the amino groups, it does not

dissolve in neutral to alkaline solutions (Agnihotri & Aminabhavi, 2004; Amidi et al., 2008b;

Asada et al., 2004; Germershaus et al., 2008; Learoyd et al., 2009); the rigid crystalline

structure of chitosan (Saito et al., 1981; Saito et al., 1987) and extensive intra- and inter-

Page 54: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

21

chain hydrogen bonding (Liu et al., 2004; Nishimura et al., 1991) have also been reported to

make important contribution to this intractability of the polymer. These chemical properties

allow chitosan to play an effective role in controlling drug delivery. It can be processed in

acidic conditions where it is soluble; by contrast, as products made of chitosan are insoluble

in neutral or alkaline pH, it behaves as a sustained release delivery system under such

conditions (Onishi & Machida, 2006). It would be relevant to note here that, though

chitosan is typically insoluble in water, it could be prepared in some water-soluble forms

(the so-called ‘water-soluble chitosan — WSC) by controlling its degree of deacetylation

(DDA) and/or molecular weight (MW). Thus, chitosan with a DDA of about 50% is water

soluble and can be obtained from chitin by hydrolysis with alkali (Kurita et al., 1977) or from

chitosan by N-acetylation with acetic anhydride (Kurita et al., 1989). Similarly, low molecular

weight water-soluble forms can be prepared by chemical or enzymatic depolymerisation of

insoluble chitosan (Qin et al., 2002; Vikhoreva et al., 1999).

OOHO

NHRO

OH

nR= H, -COCH31

234

5

6

Figure 2- 10: The Structure of Chitosan

Because of multifarious industrial sources and preparation methods and conditions,

chitosan widely varies in its physicochemical properties (Kumirska et al., 2009). Commercial

chitosan is characterized by a DDA between 70 and 95% and a MW between 50 and 2,000

kDa (Sun et al., 2009). A knowledge of DDA and MW is important for its applications and, by

far, the MW is the most important factor that control its properties (Yaghobi, 2012). Based

on the MW, chitosan is divided into three categories, namely low molecular weight (LMW),

medium molecular weight (MMW) and high molecular weight (HMW), but these categories

are rather arbitrary — there are no well-recognized boundaries between them. Thus,

various researchers reported different and sometimes overlapping values for the three

categories. For example, when some investigators termed 50-190 kDa as low-, 190-310 kDa

as medium-, and 310-375 kDa as high-molecular-weight chitosan (Shuai et al., 2013), others

used the values <10 kDa, 20-50 kDa, and 1,000 kDa, respectively, for indicating the same

categories (Lee et al., 2005). In spite of this ambiguity, the classification has been widely

Page 55: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

22

used in the literature and served as a useful tool for describing differences observed in

physicochemical properties with a change in the molecular weight of the polymer. In

general, it has been shown that solubilization of chitosan gets increasingly difficult with an

increase in MW (El-Hefian et al., 2009; Pillai et al., 2009), making chitosan LMW easier to

process for drug delivery applications compared with MMW or HMW ones (Kumirska et al.,

2011), but lower molecular weights of the polymer offer relatively less drug encapsulation

efficiency and poorer control over drug release (Kumirska et al., 2011; Learoyd et al.,

2008a). These shortcomings, however, might be compensated by adequately crosslinking

the polymer with an appropriate agent (such as glutaraldehyde) (Bansal et al., 2011).

Chitosan has been shown to be both biocompatible and biodegradable (Dodane & Vilivalam,

1998; Grenha et al., 2007). The 16 g/kg oral LD50 of the polymer for mice indicates a very

low toxicity potential for this product (De Jesus Valle et al., 2008; Huang et al., 2005).

Furthermore, its degradation products are also considered to be non-toxic and

non-immunogenic (Muzzarelli, 1993) — the degradation products being oligosaccharides of

variable length (Dumitriu, 2002). Because of its cationic nature, It can also bind to mucosal

surfaces, which leads to bioadhesion, and in turn reduces mucociliary clearance, thereby

providing a longer contact time for drugs (Davis, 1999). In addition, it has also been found to

improve the drug absorption by opening the intercellular tight junctions of the lung

epithelium (Yamamoto et al., 2005). Moreover, in a recent study, it has been shown that

chitosan can also be employed as a ‘dispersibilty enhancer’ in dry powder inhaler

formulations (Li & Birchall, 2006).

Chitosan has widely been used as sustained release carrier systems in several

pharmaceutical formulations such as beads (Aiedeh & Taha, 2001; Srinatha et al., 2008),

gels (Patel & Amiji, 1996; Ruel-Gariepy et al., 2000), films (Ji et al., 2009; Puttipipatkhachorn

et al., 2001), sponges (Oungbho & Muller, 1997; Phaechamud & Charoenteeraboon, 2008)

and micro- and nanoparticles (Barakat & Almurshedi, 2011; Saha et al., 2010) for its unique

properties like low toxicity, biocompatibility, biodegradability and mucoadhesion. Given that

chitosan not only acts as a drug release modifier (Learoyd et al., 2008a), but also has

mucoadhesive properties (Sigurdsson et al., 2006) and potential for enhancing

Page 56: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

23

aerosolization (Li & Birchall, 2006), it can be regarded as a highly promising carrier for

preparing sustained release formulations for pulmonary drug delivery.

Of various formulation strategies, micro- and nano-particulate systems have attracted much

attention as systems for drug inclusion for controlled delivery of drugs (Agnihotri et al.,

2004; Mitra & Dey, 2011). Micro- and nanoparticles can be prepared in a variety of size,

shape, and porosity by varying the process parameters (DeLuca et al., 1993). By

appropriately controlling the properties, particles can be designed to provide a steady

release of encapsulated drugs over prolonged periods of time to local tissues or the

systemic circulation (Chow et al., 2007). In addition to controlling drug release, they can also

offer protection to the encapsulated drug from enzymatic degradation (Abd El-Hameed &

Kellaway, 1997; Sadeghi et al., 2008). Most importantly, such systems can potentially be

designed for deposition of drugs to the targeted regions of lungs (Fu et al., 2002; Madlova et

al., 2009). Finally, being biodegradable, the particles ultimately degrade and are

metabolized by the body obviating need for removal (Budhian et al., 2005; Fu et al., 2004b).

Nanoparticles have widely been studied in drug delivery research for site-specific targeting

and controlled release (Swami et al., 2012). They have been proposed as valuable vehicles

for efficient drug transport to the lungs (Sham et al., 2004). By controlling particle

dispersion, breathing and particle size, 90% of an inhaled nanoparticle dose can specifically

be deposited to lower regions of lungs (Madlova et al., 2009). Moreover, nanoparticles

exhibit some characteristics that make them ideal candidates for pulmonary drug delivery.

Studies have shown that nanoparticles can escape mucociliary clearance and recognition by

alveolar macrophages (Ahsan et al., 2002; Grenha et al., 2005; Mao et al., 2009). However,

exploitation of this system in therapeutic setting could not yet be done to its potential

because of technological limitation of producing stable, non-aggregating nanoparticle

formulations (Madlova et al., 2009).

Chitosan micro- and nano-particles have been formulated by a variety of techniques

including spray drying (Khan & Vishakante, 2013; Learoyd et al., 2009), ionotropic gelation

(Boonyo et al., 2008; Liu & Gao, 2009), emulsion-solvent evaporation (Panos et al., 2008),

emulsion-crosslinking (Dhawan & Singla, 2003; Xu et al., 2012), coacervation (Gupta &

Page 57: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

24

Jabrail, 2007a; Tavares et al., 2012), etc. The release of drug from the particles is controlled

by three different mechanisms: (a) release from the surface of microspheres, (b) diffusion

through a swollen rubbery matrix and (c) erosion (Fig. 2-11). In most cases, the release may

involve more than one mechanism. Release from the surface layer of the matrix gives an

initial burst effect; this effect can be reduced by cross-linking. The diffusion of the drug

through the gel diffusion layer caused by polymer swelling takes place slowly over a

prolonged period of time and results in a sustained drug release effect. Three steps are

involved in the diffusion of drug out of the matrix: first, penetration of water into the matrix

causing swelling; secondly, transition from glassy to rubbery polymer, and finally, the

diffusion of drug out of the swollen rubbery matrix. Thus, the release follows a typical

hydrogel release profile (Agnihotri et al., 2004; Learoyd et al., 2008a).

The molecular weight of chitosan, the ratio of polymer to drug, solubility of the

encapsulated molecule in the release media and extent of cross-linking — all these factors

play their role in controlling the release of the loaded drug from the particle. Due to the

cationic nature of the polymer, the pH of the release media also influences the drug release

profile.

An increase in chitosan molecular weight has been found to increase duration of drug

release. For example, Learoyd et al. (2009) have shown for chitosan-lactose based

formulations that chitosan LMW (low molecular weight: <190 kDa) took 2.5 h of time to

release 100% BDP (beclometasone dipropionate) from spray-dried microspheres, whereas

time required by chitosan MMW (medium molecular weight: 190–310 kDa) and chitosan

HMW (high molecular weight: >310 kDa) were 5 h and 12 h, respectively. Increasing the

amount of chitosan in the particle increases the thickness of the diffusion layer, resulting in

slower release of drug. Martinac et al. (2005) studied the drug release profile of loratadine-

loaded spray-dried chitosan microspheres in two different drug:polymer ratios (1:6 and 1:8).

The result showed that 50% drug release from the two samples took 45 and 80 minutes,

respectively. The solubility of the encapsulated drug is another factor that has a marked

effect on drug release. Drugs with low solubility in water are most slowly released.

Page 58: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

25

Figure 2- 11: Mechanism of Drug Release from Chitosan Micro- and Nanoparticles

In vitro release study of terbutaline sulfate (TS) and beclometasone dipropionate (BDP) in

phosphate buffer solution (PBS, pH 6.8) from spray-dried chitosan microspheres showed

that the release of BDP was much slower than TS (Learoyd et al., 2009). The investigators

attributed it to partial solubility of BDP in PBS unlike TS which is freely soluble in this

medium. Cross-linking with an agent like glutaraldehyde (GL) can greatly increase the

duration of drug release. Ventura et al. (2008) studied the in vitro release of moxifloxacin

from spray-dried glutaraldehyde-crosslinked chitosan microspheres in phosphate buffer

solution (PBS, pH 7.4). The results showed that cross-linked microspheres gave a burst effect

followed by a prolonged moxifloxacin (MXF) release. About 60%, 40% and 25% (w/w) of

MXF was released within the first hour for the systems prepared in the presence of 4, 2 and

0.5 mL of GL, respectively, while a further release of only 10% (w/w), 22% (w/w) and 32%

(w/w) of loaded MXF, respectively, took place in 4 days. On the other hand, the entire drug

loaded into the un-crosslinked microspheres was released within 24 h. Since chitosan is

more soluble in acidic pH than alkaline one, drug release in the acidic environment occurs

more quickly. A study on theophylline-loaded chitosan/β-cyclodextrin (1:3:1) microspheres

showed that the release of the drug was more than 61% in the first hour and attained 74%

within 8 h at pH 1.2, while the drug release was only 39% and 60%, respectively at pH 6.8

(Zhang et al., 2008b).

Page 59: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

26

2.9 Conjugation of L-Leucine with Chitosan ― a Novel Approach for Maximizing Drug Delivery from a Controlled Release DPI

As mentioned in the section 2.5, L-leucine (Fig. 2-12) ― a predominantly hydrophobic amino

acid with surfactant like property ― has been found to be a very efficient aerosolization

enhancer when added to a DPI formulation by blending, spray drying or even by direct

coating on drug particles by physical vapour deposition (PVD). In recent years, some

researchers attempted to combine the potential of chitosan as a release modifier with the

aerosolization enhancing ability of L-leucine and found it to be very effective (Learoyd et al.,

2008a, 2009; Seville & Learoyd, 2007). Learoyd et al. (2008a) reported that spray-drying

30% v/v aqueous ethanol formulations, containing terbutaline sulfate as a model drug,

chitosan as a drug release modifier, L-leucine as an aerosolization enhancer and lactose as a

bulking agent, produced highly dispersible sustained release powder for pulmonary delivery

with emitted doses of over 90% and FPFs of up to 82% of the total loaded dose. In another

study, these investigators demonstrated that combining L-leucine and chitosan together in a

spray-dried lactose-based formulation of beclomethasone dipropionate could give 64% FPF

with 2 hour dissolution time for low MW (<190 kDa) chitosan and 54% FPF with 12 hour

dissolution time for high MW (>310 kDa) chitosan (Learoyd et al., 2008b). In a subsequent

study, Learoyd et al. (2009) reported over 90% of emitted doses and up to 76% FPF of the

loaded dose from a spray-dried powder containing both the hydrophilic drug, terbutaline

sulfate and the hydrophobic drug, beclomethsone dipropionate along with chitosan,

L-leucine and lactose. The formulation showed sustained release profiles for both drugs in

dissolution tests.

H3COH

CH3 NH2

O

Figure 2- 12: The Structure of L-Leucine

Hydrophilic Domain

Hydrophobic Domain

Page 60: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

27

It is evident that these workers concentrated their attention mainly on physically combining

chitosan and L-leucine in a DPI formulation. No attempt has so far been made to chemically

conjugate chitosan with L-leucine in order to investigate its impact on aerosolization and

lung deposition from a DPI formulation. It can be an interesting question to explore if

chemical conjugation of chitosan with L-leucine can still maintain this effect on dispersibility

of chitosan from a DPI which is a very likely possibility in view of the structural features of

L-leucine that could be expected to orient the molecule around the chitosan backbone with

its hydrophobic part being projected outward. There is good possibility that this will in turn

reduce the inter-particle cohesion and may even add further to dispersiblity enhancing

property of L-leucine found with simple physical addition. In addition to this possible effect

on the surface morphology, the ability of L-leucine to reduce the surface tension, as detailed

in the section 2.5 of this chapter, could also be expected to play a positive role in enhancing

the dispersion of particles.

It would be relevant here to shed some more light on the probable interfacial orientation of

the hydrophilic (─NH2─COOH) and hydrophobic (─CH2─CH(CH3)2) domains of L-leucine

conjugated to chitosan. As shown in the Fig. 2-13 (a) and (b), as a general rule, the

hydrophobic domain of an amphiphilic agent is projected towards air at a water-air

interface or towards the oil phase at a water-oil interface, while the hydrophilic domain is

associated with the aqueous phase (Sinko, 2006). Because of this tendency of the two

different domains to get associated with two different phases, when this type of molecules

are added to a liquid they are oriented at the interface rather than the bulk. However, as

the concentration is increased, the interface becomes saturated at a certain point (called

critical micelle concentration, CMC) and the molecules begin to form in the bulk of liquid

special type of aggregates called micelles where the domain liked by the surrounding

medium is organized at the periphery in association with it but the one disliked by the

medium is oriented towards the core of the micelle in order to keep itself away from the

surrounding medium ((Fig. 2-13 (c) and (d)) (Sinko, 2006). So, it is obvious that in an aerial

environment (for example, when aerosolized from a DPI), the polymer chitosan — being

hydrophilic in nature (Wen & Park, 2011) — would behave as the hydrophilic phase and so

the hydrophobic domain of L-leucine conjugated to it would be projected towards the air

(Fig. 2-14(a)). This is in conformity with the observations of other investigators for the

Page 61: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

28

Figure 2- 13: (a) & (b) Orientation of a surfactant molecule at oil-water and air-water interfaces respectively, (c) & (d) Micellar orientation of a surfactant molecule in water and non-polar solvents respectively [adapted from Sinko (2006)]

Figure 2- 14: Probable orientation of hydrophilic and hydrophobic domains of L-leucine conjugated to chitosan in particle-air (a) and particle-water (b) interfaces.

Page 62: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

29

orientation of L-leucine (Raula et al., 2010) and its tri-leucine analogue (Lechuga-Ballesteros

et al., 2008) at a particle-air interface. On the other hand, although hydrophilic in nature

(Wen & Park, 2011), the polymer chitosan is poorly soluble in water due to its rigid

crystalline structure (Saito et al., 1981; Saito et al., 1987) and extensive intra- and inter-

chain hydrogen bonding (Liu et al., 2004; Nishimura et al., 1991). So, in an aqueous

environment (for example, in the lung fluid or in vitro release medium), the polymer would

behave as a hydrophobic pahse in relation to the surrounding water and the orientation of

the hydrophobic and hydrophilic domains of L-leucine would be just opposite to what is

seen in a particle-air interface (that is, the hydrophilic domain would be associated with the

surrounding aqueous medium and the hydrophobic domain would tend to burry itself into

the polymer backbone within the particle) (Fig. 2-14(b)). It would be worth noting here that

lung fluid — the in vivo release medium (as well as PBS used as in vitro release medium in

this study)— has a pH of 7.3 that is slightly higher than pure water (pH 7.0) and may also

contain substances like enzymes, but a pH variation or presence of enzymes (which are

protein in nature and so water-soluble macromolecules) does not bring any change to the

hydrophilic nature of the aqueous phase and so its interfacial interaction with the

suspended polymer particles in terms of hydrophilicity/ hydrophobicity remains all the

same. However, at a pH of 7.3 the ionization of the amino group of L-leucine may be

suppressed causing it to be less associated with the surrounding medium, but the carboxylic

acid group would be ionized and so well associated with the the medium. The pulmonary

route is considered to be free from the drawback of first-pass metabolism (Agu et al., 2001)

and so there should not be any significant enzymatic degradation of the administered

particles in the lung fluid until they are engulfed by macrophages. However, for the sake of

argument, even if the particles are assumed to be ezymatically metablized to give chitosan

monomers or oligomers, it would rather increase their hydrophilicity.

2.10 The Human Bronchial Epithelial Cell Line, BEAS-2B for the Assessment of the Toxicity and Inflammatory Activity of the DPI formulation

As already noted, pulmonary route of drug delivery has drawn immense attention of

formulation scientists for its interesting and attractive features, and polymer-based drug

delivery to the respiratory system for local or systemic action is an exciting area of present

Page 63: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

30

day research. However, it is important to ensure in the first place that the formulation/

system intended for inhalation does not have any adverse reaction to the respiratory

airways. The safety of inhaled formulations is best evaluated in vitro by using a variety of

toxicological tests on respiratory epithelial cell lines to provide complementary information.

Literature reports suggest many different toxicological tests for the assessment of the safety

of polymeric nanoparticles in respiratory epithelial cell lines. Three most commonly used

and well-characterised tests in this series are MTT cell viability assay, sodium fluorescein

transport assay and release of proinflammatory mediators.

2.10.1 MTT Cell Viability Assay The determination of cell viability is a common method for in vitro evaluation of the

cytotoxicity of biomaterials (Wu et al., 2008). In vitro assays based on cultured cells are

often used to screen potential drugs and excipients for any adverse biological effects

towards body cells and tissues before in vivo testing to reduce untoward use of research

animals. Researchers have widely used cell viability assays to evaluate the safety of inhaled

materials (Amidi et al., 2006; Bivas-Benita et al., 2004; Forbes et al., 2000; Grenha et al.,

2007; Stone et al., 1998; Westmoreland et al., 1999). Traditionally, a number of in vitro

cytotoxicity assays have been used to this end. These assays are based on assessing

membrane integrity (e.g., neutral red, calcein AM) or cell metabolism (e.g. MTT, alamar

Blue) by using colorimetric or fluorescent dyes as marker (Monteiro-Riviere et al., 2009).

The predictive value of these tests is based on the concept that toxic chemicals affect the

basic functions of cells and so the toxicity can be measured by assessing cellular damage

(Mao et al., 2005). The changes in metabolic activity has been shown to be superior

indicators of early cell injury, because disruption of membrane integrity reflects more

serious damage leading to cell death (Davila et al., 1990).

MTT cell proliferation assay, originally developed by Tim Mosmann in 1983 (Mosmann,

1983), is a quick effective method for testing mitochondrial impairment and correlates well

with cell proliferation (Muzzarelli et al., 2005), and therefore is often performed for

evaluation of the cytotoxicity of polymers (Mosmann, 1983). The assay integrates both the

number of cells and mitochondrial activity per cell (Wan et al., 1994). Central to this assay is

Page 64: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

31

a water-soluble, yellow tetrazole dye, 3-(4,5 dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium

bromide (MTT), that is metabolically reduced by living cells to water insoluble, purple MTT-

formazan (Fig. 2-15) (Mosmann, 1983). Damaged or dead cells show reduced or no

dehydrogenase activity and thus fail to reduce MTT to formazan (Florea et al., 2006). The

MTT formazan is extracted from cells with an organic solvent (e.g. DMSO, 1-propanol etc.)

and its concentration is determined by measuring the absorbance with a

spectrophotometer at ~550 nm. In general, the absorbance is a linear function of the

number of metabolically active cells (Monteiro-Riviere et al., 2009; Zhang et al., 2011).

Decreased absorbance reflects reduced mitochondrial activity, and, by inference, decreased

cell health.

N NNN

S

N

Br

MitochondrialReductase

MTT

NNH

NN

S

N

Formazan Product Figure 2- 15: Conversion of MTT to Formazan

Referring to an article of Slater et al. (1963), some authors incorrectly attributed the

reduction process to the mitochondrial enzyme, succinate dehydrogenase (Denizot & Lang,

1986). But, based on cell homogenates and isolated mitochondria, two coupling points have

been found between MTT reduction and the mitochondrial respiratory chain — coenzyme

Q: cytochrome c (Liu et al., 1997; Marshall et al., 1995; Slater et al., 1963). In addition, there

has been increasing evidence that denies exclusive role of mitochondria and suggests for an

even more important role of NADH- and NADPH-dependent mechanisms (Berridge & Tan,

1993; Liu et al., 1997; Vistica et al., 1991). Considering these facts, the MTT assay can be

regarded as a quantitative measure of a combination of cellular proliferation and an

integrated set of enzyme activities that are related to cell metabolism in various ways

(Berridge et al., 2005). In addition, its rapidity, precision and lack of requirement of any

radioisotope are important advantages of this test (Mosmann, 1983).

Page 65: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

32

There have been quite a few studies on the safety and biocompatibility of chitosan and its

derivatives in pulmonary drug delivery systems both in neat and particulate forms. In light of

these studies, chitosan has generally been recognised as a biocompatible and biodegradable

polysaccharide with low toxicity (Dornish et al., 1997; Grenha et al., 2007; Hirano et al.,

1988; Huang et al., 2004; Mansouri et al., 2004; Mi et al., 2001; Nordtveit et al., 1996; Wang

et al., 2007). Chitosan in solution has been reported to exhibit low or no toxicity in

respiratory cell lines like A-549 and 16HBE14o- (Florea et al., 2006; Grenha et al., 2007;

Huang et al., 2004; Lim et al., 2001; Smith et al., 2004; Smith et al., 2005). A high

concentration of chitosan has, however, been reported to exert some degree of cytotoxic

effect. For instance, chitosan solution at a concentration of 15 mg/mL has been reported to

reduce Calu-3 cell viability to approximately 70% (Florea et al., 2006). Contradictory reports

are seen in the literature about the compatibility of particulate chitosan with the respiratory

system. Chitosan nanoparticles, prepared by an ionic gelation method, were shown to

reduce the viability of A-549 cells by approximately 70% at a concentration of about

1 mg/mL (Huang et al., 2004). Another report from this group noted that chitosan

microparticles induced pro-inflammatory responses in rat lungs (Huang et al., 2005). In

contrast, other studies reported a low toxicity of chitosan nanoparticles in respiratory cell

lines (Grenha et al., 2007; Smith et al., 2005). Grenha et al. (2007) found <50% reduction in

Calu-3 or A-549 cell viability with chitosan nanoparticles. Further, Vllasaliu et al. (2010)

observed the effect of chitosan nanoparticles on Calu-3 cell viability to be slightly lower

compared to chitosan solution. While in some cases this difference did not reach a statistical

significance, in other cases the discrepancy was clearly apparent (Vllasaliu et al., 2010). In

another report, the authors noted that chitosan/cyclodextrin (‘hybrid’) nanoparticles

exhibited a significantly lower cytotoxicity than those based on chitosan only (Teijeiro-

Osorio et al., 2009a, 2009b), with the IC50 values for chitosan/cyclodextrin-containing

nanoparticles being 3-fold higher compared to the chitosan-only nanoparticles. However, it

should be emphasized here that direct comparisons between these studies are difficult

because of differences in experimental parameters including different MWs and DDAs of

chitosan, different chitosan formulations and finally different cytotoxicity assays and cell

lines.

Page 66: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

33

One of the best characterized derivatives of chitosan is TMC (trimethyl chitosan chloride) —

a compound obtained by quaternization of the –NH2 group at C-2. The outstanding

biocompatibility of unmodified chitosan is accompanied by its low solubility at physiological

pH. Quaternization improves solubility (Thanou et al., 2002), but on the other hand

significantly increases its cytotoxicity (Godbey et al., 2001; Mao et al., 2005). In general,

polycations are considered to be cytotoxic (Morgan et al., 1989). It is suggested that TMC,

like most cationic macromolecules such as protamine and polylysine, produces cytotoxic

effects by interacting with anionic components (sialic acid) of the glycoproteins on the

surface of epithelial cells (Choksakulnimitr et al., 1995; Fischer et al., 2003; Mao et al.,

2005). There have been extensive studies with TMC and it was found that the toxicity varies

with various factors like molecular weight, degree of quaternization, duration and dose etc.

(Mao et al., 2005; Mourya & Inamdar, 2009). Some investigators, however, found TMC to be

low or non-toxic in the forms and assay conditions they applied (Mao et al., 2005; Mourya &

Inamdar, 2009). PEGylation was found to considerably decrease the cytotoxicity of TMC.

PEG is considered to shield a proportion of the positive charges present on TMC (Mao et al.,

2005). A similar effect was found upon complexation of TMC with insulin. This is again

attributable to the electrostatic interaction between TMC and insulin, which decreased the

interaction of the cationic amino groups of TMC with the anionic components of the cell

membrane, leading to higher cell viability (Mao et al., 2005).

2.10.2 Sodium Fluorescein Transport Assay Apart from the effect on the cell viability, one important indicator of adverse effects of an

inhaled material on pulmonary epithelium is any detrimental change in the epithelial barrier

function, because the epithelium serves as the protective barrier against inhaled particles

and acts as a major determinant of the interaction of the particles with other body

compartments (Hermans & Bernard, 1999; Strengert & Knaus, 2011). Permeation of a

substance across an epithelial barrier can occur through two different routes, viz.

1) transcellular transport across the cell, passing through both the apical and basolateral

membranes, and 2) paracellular transport through intercellular junctions (Krug et al., 2009).

In functional epithelial tissues, the paracellular route is regulated mainly by tight-junctional

complexes, which confer a virtually impermeable barrier (Florea et al., 2006) to

Page 67: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

34

macromolecules larger than 1000 Da (Illum, 2000; Stolnik & Shakesheff, 2009). In vitro, the

tightness of this intercellular barrier can be determined by measuring the transepithelial

electrical resistance (TEER) and the paracellular apparent permeability (Papp) of a fluid phase

marker (Florea et al., 2006) like fluorescein isothiocyanate (FITC)-dextran (FD) (Vllasaliu et

al., 2010), [14C]-mannitol (Thanou et al., 2000a; van der Merwe et al., 2004), [14C]-PEG4000

(van der Merwe et al., 2004) or sodium fluorescein (Na Flu) (Sivadas et al., 2008) which are

normally unable to permeate through tight junctions.

It is worth noting here that a mild, transient enhancement of trans-epithelial permeability

can rather be used to the benefit of increasing drug permeability. From this perspective,

chitosan and its derivatives have long been studied for their ability to enhance epithelial

permeability to investigate their potential as a possible carrier or excipient capable of

enhancing drug absorption (Boonyo et al., 2007; Florea et al., 2006; Hamman et al., 2002;

Illum, 1998; Illum et al., 1994; Illum et al., 2001; Kotze et al., 1997a; Kotze et al., 1998b;

Thanou et al., 2001a; Thanou et al., 2000b; Thanou et al., 2001b; van der Merwe et al.,

2004; Vllasaliu et al., 2010). Many of these studies demonstrated that chitosan can improve

paracellular permeability of drugs or fluid phase markers through polarized pulmonary

epithelial cell line, Calu-3 (Bonferoni et al., 2008; Florea et al., 2006; Vllasaliu et al., 2010).

Vllasaliu et al. (2010) also reported a significant bronchoepithelial permeation enhancement

of FD4 and FD10 with chitosan nanoparticles at a concentration of 0.003% (w/v) (the

permeability of FD4 was enhanced 7.6-fold while there was a 6.5-fold increase in the

permeability of FD10). However, they found chitosan solution to be more efficient in

improving the permeability of both FD4 and FD10, with a 10.1-fold improvement in

permeability across the cells observed for both FITC-dextrans, relative to their respective

controls. Some investigators, however, observed that although particulate chitosan caused

an increase in permeability, it was not significant (Grenha et al., 2007; Sivadas et al., 2008).

When chitosan dissolves in an aqueous medium, its amino groups are protonated giving

positive charges (Illum et al., 2001). The opening of tight junctions by chitosan results from

its interaction with negative charges on cell membrane, which causes a redistribution in

cytoskeletal F-actin, thereby resulting in a structural reorganization of tight junction

associated proteins like ZO-1 (Illum, 1998; Schipper et al., 1997). In addition, Smith and co-

workers (2005) have also shown that chitosan causes a loss of tight junction integrity

Page 68: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

35

through inactivation of a protein kinase C (PKC). But, chitosan is soluble only at a pH below

5.6, limiting its use in body compartments (Yadav et al., 2011). It has been found that, at pH

7.4 (the more commonly found pH in body compartments) chitosan is ineffective as

permeability enhancer (Borchard et al., 1996; Kotze et al., 1998a). Evidently, at lower pH

values a higher charge density, in combination with a better solubility, provides a better

environment for intimate contact with the epithelial membrane (Artursson et al., 1994).

Experiments with TMC — which is more soluble than chitosan, especially at neutral and

basic pH values (Kotze et al., 1997b)― demonstrated strong permeation enhancement

effect for small hydrophilic compounds such as [14C]-mannitol, large molecules such as [14C]-

PEG4000, and peptide drugs, buserelin and octreotide, across the cell monolayers (Kotze et

al., 1997a; Thanou et al., 2000a; van der Merwe et al., 2004).

2.10.3 Release of Proinflammatory Mediators Epithelial cells of the respiratory tract provide an excellent structural barrier to inhaled

agents, but they also play a crucial role in the lung function, partly through production of

various pro-inflammatory mediators such as tumour necrosis factor-α (TNF-α), interleukin

(IL)-1, and IL-8 (Ghio et al., 2002; Rahman & MacNee, 2000). These mediators are basically

released as a protective response of the host to destroy and remove injurious agents and in

turn promote tissue repair. But, when these crucial and normally beneficial agents are

generated in excessive amounts or at inappropriate times or places, they can cause

untoward damage to the host tissue and exacerbate or perpetuate the injury and may even

lead to chronic inflammation (Laskin et al., 2007; Rahman & MacNee, 2000).

Many studies have emphasised the ability of inhaled particles to induce inflammation and

other toxicity in the lung (Borm, 2002; Churg & Brauer, 2000; Oberdorster, 2001).

Interactions with particles can induce various types of lung cells, including lung

monocytes/macrophages and pulmonary epithelial cells, to increase their release of

inflammatory mediators (Becher et al., 2001; Driscoll et al., 1997; Hetland et al., 2004;

Mazzarella et al., 2012; Monn & Becker, 1999; Takizawa et al., 2000; Tal et al., 2010; Tsukue

et al., 2010; Vanhee et al., 1995). Increased levels of proinflammatory mediators, such as

interleukin-8 (IL-8)/macrophage inflammatory protein-2 (MIP-2) and interleukin-6 (IL-6),

have also been demonstrated in both animal and human bronchoalveolar lavage fluid (BALF)

Page 69: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

36

after particle exposure (Becher et al., 2001; Ulrich et al., 2002; Vanhee et al., 1995). Thus, it

is imperative to ascertain for any particulate matter intended for delivery through

pulmonary route that it would not induce undue inflammatory response. This can be

accomplished by checking the release of inflammatory mediators like IL-8 or IL-6 by an

immunological technique like enzyme-linked immunosorbent assay (ELISA) (Saedisomeolia

et al., 2009; Sivadas et al., 2008).

There have been a limited number of works on inflammatory activity of chitosan, both neat

and particulate, on the lung and pulmonary epithelial cell lines (Choi et al., 2010; Florea et

al., 2006; Huang et al., 2005; Sivadas et al., 2008; Witschi & Mrsny, 1999). Florea et al.

(2006) performed in vivo studies by administering chitosan and its derivative, TMC, into rat

lungs. Histopathological analysis of lung tissue showed that chitosan elicited neutrophil

infiltration and structural damage to the lung parenchyma; the effects were relatively milder

with TMC. In another study, Huang et al. (2005) have shown that inhaled chitosan

microparticles at a dose of 2-10 mg/kg induced dose-dependent proinflammatory effects in

rat lungs which were manifested by an increase in biochemical parameters like

bronchoalveolar lavage fluid protein (BALF-P) and lactate dehydrogenease activity (BALF-

LDH) and increases in lung tissue myeloperoxidase (MPO) activity and leukocyte migration.

Choi et al. (2010) reported increased expression of pro-inflammatory cytokines (IL-1β, IL-6,

and TNF-α) and chemokine (MIP-1α) in lung 1 to 24 h after intratracheal instillation of

hydrophobically modified glycol-chitosan nanoparticles into mice. According to a report by

Witschi & Mrsny (1999), chitosan microparticles, applied apically, induced basolateral

release of IL-6 and IL-8 from polarized Calu-3 cells. However, release of other cytokines, such

as IL-1β, TNF-α, GM-CSF and TGF-β, were not affected. Some authors, however, reported

opposite findings. For example, Sivadas et al. (2008) studied IL-8 release from Calu-3 cell

monolayers treated with protein-loaded inhalable microparticles of a range of polymers

including chitosan, hydroxypropyl cellulose (HPC), hyaluronic acid, alginate, gelatin,

ovalbumin and poly(lactide-co-glycolide) (PLGA) on the pulmonary epithelium. After 4 h of

apical exposure to the polymeric microparticles, only PLGA and gelatin microparticles were

found to cause a significant increase in IL-8 release. In a study by Mura et al. (2011) PLGA-

chitosan nanoparticles were demonstrated not to induce an increase in IL-6 or IL-8 levels

upon incubation with Calu-3 cells in concentrations up to 0.2 mg/mL.

Page 70: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

37

2.10.4 Interleukin-8 (IL-8) IL-8 is the best characterized member of the family of chemokines — the proinflammatory

cytokines that chemoattract and activate blood cells (Ben-Baruch et al., 1995; Oppenheim et

al., 1991). This chemokine has a wide range of proinflammatory properties ranging from its

main function as chemoattractant for neutrophils, basophils and a subpopulation of

lymphocytes to activation of neutrophils (Fitzgerald, 2001). Besides its central role in

inflammation, other biological functions of IL-8 include T cell chemotaxis (Taub et al., 1996),

angiogenesis (Koch et al., 1992), and hematopoiesis (Cacalano et al., 1994). Expression of

this chemokine is a typical characteristic of pulmonary inflammatory and immune conditions

(Erger & Casale, 1998; Pease & Sabroe, 2002). It is produced in abundance by mononuclear

phagocytic cells as well as a number of non-inflammatory cells including fibroblasts and

pulmonary epithelial cells (Kunkel et al., 1991).

2.10.5 BEAS-2B – the In Vitro Cell Model for the Assessment of Toxicity and Inflammatory Activity

BEAS-2B (Reddel et al., 1988) is an adenovirus 12-SV40 virus hybrid (Ad12SV40) transformed

human epithelial cell line, which was isolated from normal human bronchial epithelium

obtained from autopsy of noncancerous individuals. It has been reported that cultures of

BEAS-2B S.6 cells can develop significant trans-epithelial resistance and exhibit tight junction

formation (Wan et al., 2000). The cell line has also been used as an established in vitro

model of induced IL-8 secretion after treatment with various model inflammatory stimuli

such as tumor necrosis factor-α (TNF-α), ozone, or air pollution particles (Devlin et al., 1994;

Lakshminarayanan et al., 1997; Quay et al., 1998). Unlike some malignant pulmonary

epithelial cell lines like Calu-3 and A-549 as reported above, no study has so far been

performed on this cell line to investigate the impact of chitosan or its derivatives on the cell

viability, trans-epithelial permeability or inflammatory response. It can be considered

intriguing to explore the effect of chitosan and its L-leucine conjugate on this cell line that

will not merely assess their safety, but will also provide information about the behaviour of

a non-malignant respiratory epithelial cell line in response to them.

Page 71: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

38

2.11 Model Drug: Diltiazem Hydrochloride (a systemically acting antihypertensive Agent)

Diltiazem hydrochloride (DH) (Fig. 2-16) is a benzothiazepine calcium channel blocker. It has

widely been used in the treatment of hypertension, angina and cardiac arrhythmias (Buckley

et al., 1990). In recent past, a large prospective study demonstrated that diltiazem was as

effective as β-blockers and thiazide diuretics in preventing cardiovascular events in

hypertensive patients (Hansson et al., 2000). Thus, from an evidence-based point of view,

diltiazem appears to be a first-line alternative among antihypertensive drugs. The drug has a

relatively short half-life (3–5 h) and is usually administered 3–4 times daily in the form of an

immediate release formulation (Buckley et al., 1990). After oral administration, the drug

undergoes extensive first-pass metabolism giving an absolute bioavailability of only about

40% with a large inter-individual variation (Buckley et al., 1990; Yeung et al., 1993). Lee et al.

(1991) reported that the extraction ratios of diltiazem in the small intestine and the liver

were about 85% and 63%, respectively after an oral administration to rats, suggesting that

both the small intestine and the liver had a significant contribution to the first-pass

elimination of the drug.

S

N

N

O

O

O

O

. HCl

Figure 2- 16: The Structure of Diltiazem Hydrochloride

In order to improve its bioavailability and ensure a more uniform blood profile, researchers

are trying to employ other routes, including transdermal, buccal and nasal routes, as non-

invasive alternatives to the oral route for the delivery of the drug. But, to our knowledge, no

effort has so far been made to explore the potential of the pulmonary route as a mode of

delivery of the drug for the treatment of systemic hypertension. Given the enormous

vasculature and extensive surface area for absorption, absence of variability of pH found in

different regions of GIT and avoidance of extensive intestinal and hepatic metabolism, the

Page 72: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 2 Literature Review

39

lungs can be a promising alternative to the oral route that has the potential to assure better

bioavailability of the drug with a reduced dose. So, it will be interesting to explore the

potential of designing a DPI formulation using diltiazem as the model drug.

Page 73: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 74: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 75: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 76: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

40

3.1 REAGENTS AND MATERIALS

3.1.1 For Synthesis Low molecular weight chitosan (DDA 92%, MW 50-190 kDa), used as the starting material

for the synthetic studies, was obtained from Sigma-Aldrich, Australia. Phthalic anhydride,

acetic anhydride, trityl chloride, aqueous hydrazine hydrate (50-60%), Boc-L-leucine

succinimide (Boc-leu-OSu) and 4M HCl in 1, 4-dioxane were used as the reagents at various

steps of the synthesis. Phthalic anhydride was a Merck Schuchardt (Germany) product. All

other reagents were supplied by Sigma-Aldrich (Australia) and were used as received

without any further purification. N, N-Dimethylformamide (DMF) and pyridine were used as

solvents. Both were Sigma-Aldrich (Australia) products. Pyridine was stored over 4 Å

molecular sieves to prevent moisture absorption. Methanol (Chem-Supply, Australia),

ethanol (Recochem, Australia) and diethyl ether (Merck, Australia) were used for

precipitation and purification of reaction products. CDCl3, Pyridine-d5 and D2O were used for

NMR characterization of synthetic products. They all were obtained from Sigma-Aldrich

(Australia).

3.1.2 For Pharmaceutical Studies Chitosan (as above) was used as the polymeric matrix for the preparation of nanoparticles.

2% aq. solution of glacial acetic acid (Merck, Australia) was used for dissolution of chitosan.

Paraffin oil heavy 68, viscosity: 66.0-70.0 cST @ 40 °C (Chem-Supply, Australia) was used as

the external phase for the emulsification process and as the dispersant for Zetasizer analysis

of nanoparticles. Span 80 (Professional Compounding Chemists of Australia — PCCA) was

used as the emulsifier. 50% aqueous glutaradehyde (Merck, Australia) was used as cross-

linker for nanoparticle fabrication. Hexane (Merck, Australia) was used for washing off oil

from nanoparticles and for preparing nanoparticle suspensions for SEM analysis. Di-ethyl

ether (as above) was used for removing residual glutaraldehyde from nanoparticles.

Diltiazem HCl (DH — a gift from Alpha-Pharm, Australia) was used as the model drug for

nanoparticle formulations. Phosphate-buffered saline (PBS), pH 7.3±0.2 (Oxoid, England)

was used as the release medium for the drug release study. The lactose monohydrate

(Inhalac® 120) used in carrier-based dispersibility tests was obtained from Meggle Excipients

and Technology, Germany.

Page 77: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

41

3.1.3 For Cell Line Studies The bronchial epithelial cell line, BEAS-2B was a gift from Prof Philip Hansbro (School of

Biomedical Sciences and Pharmacy, The University of Newcastle, Australia). Rosewell Park

Memorial Institute, RPMI 1640 (Gibco®), foetal bovine serum, FBS (Lonza), L-glutamine

(Gibco®) and Penicillin-Streptomycin (Gibco®) were used as the ingredients of the cell

culture medium. These were supplied by Life Technologies Australia Pty. Ltd. 3(4, 5-

dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide, MTT and dimethyl sulfoxide, DMSO

(spectrophotometric grade), used for cytotoxicity assay, were obtained from Sigma-Aldrich

(Australia). Sodium fluorescein, Na Flu (Sigma-Aldrich, Australia) was used as a paracellular

marker for trans-epithelial permeability experiments. Analysis of the cytokine, IL-8 was done

by Human IL-8 ELISA MAXTM Deluxe kit (Biolegend, USA). 0.5% Trypsin-EDTA (Gibco®, Life

Technologies, Australia), diluted to 0.01% in PBS, was used for splitting cells.

3.2 METHODS This section describes the experimental methods followed in course of this research project

to achieve the stated aims and objectives. Broadly speaking, the experiments performed to

this end may be considered in three general headings:

a) Conjuation of chitosan with L-leucine

b) Preparation of chitosan and chitosan-L-Leucine conjugate nanoparticles and in vitro

studies on their aerosolization performance and drug release

c) In vitro evaluation of toxicity and inflammatory activity on the pulmonary epithelial

cell line, BEAS-2B

A bird’s eye view of the methods followed in this work is presented in the Fig.3-1.

Page 78: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

42

Figure 3- 1: A Bird's Eye View of the Whole Project

3.2.1 Conjugation of Chitosan with L-Leucine

3.2.1.1 Characterization The synthetic products were characterized by Fourier transform infrared (FTIR) and 1H and 13C nuclear magnetic resonance (NMR) spectroscopy and elemental analysis. The last two

products in the synthetic pathway, 6-O-trityl-chitosan-N-Boc-L-leucine and chitosan-L-

leucine.HCl, being new to the existing literature, were also characterized by 2D 1H-13C

gradient-enhanced heteronuclear single quantum correlation (ge-HSQC) NMR spectroscopy.

The final product, chitosan-N-L-leucine. HCl was further characterized by X-ray

photoelectron spectroscopy (XPS). XPS analysis was also performed on chitosan and

L-Leucine for comparison.

Page 79: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

43

3.2.1.1.1 Fourier Transform Infrared (FTIR) Spectroscopy Infrared spectra were recorded on a Nicolet 5700 FT-IR ATR spectrophotometer (Thermo

Electron Corporation, Madison, Wisconsin, USA) equipped with a Smart Endurance diamond

internal reflection element (IRE) under dry air at ambient temperature (25 ͦC). The spectra

were collected by using standard spectral collection techniques and the software Omnic 32

over the wave number range 4000–650 cm−1 using 64 scans at a resolution of 4 cm−1.

3.2.1.1.2 Nuclear Magnetic Resonance (NMR) Spectroscopy 1H, 13C and 2D HSQC NMR spectra were recorded on a Bruker AVANCE 400 high resolution

spectrometer (Bruker BioSpin GmbH, Germany) using QNP quad nucleus probe (tunable for 1H, 19F, 31P, 13C) under a static magnetic field of 400 and 100 MHz, respectively, at 298K.

Spectra of 16384 data points were recorded over a spectral width of 12.0144 ppm using the

accumulation of 32 transients and an exponential line broadening (LB) of 0.30 Hz was

applied prior to Fourier transformation for 1H NMR. 13C NMR spectra were recorded over a

spectral window of 219.8552 ppm with 16384 data points and 6144 transients using proton

decoupling. Prior to Fourier transformation, 2.00 Hz LB was applied. The 1H-13C ge-HSQC

experiments were performed in phase-sensitive mode using Echo/Antiecho-TPPI gradient

selection. The data matrix was 256 x 2048 with spectral widths of 4807.69 Hz for proton and

18,115.94 Hz for carbon. The evolution time was set to 1/(8JCH) = 0.89 ms. Four gradients

pulses were applied along the z axis with ratios of 80, 20.1, 11 and -5, respectively and a

duration of 1 ms. 18 transients were accumulated for each increment. A square sine window

function was applied in both dimensions prior to Fourier transformation. CDCl3, pyridine-d5

and D2O were the solvents used for preparing NMR solutions of the intermediates and the

final product according to their solubility. A concentration of approximately 1 mmol was

used for preparing the NMR samples. After adding the sample to the solvent in a vial, it was

stirred for several hours on a magnetic stirrer to thoroughly mix and dissolve it, and finally

centrifuged to settle down any un-dissolved solid. Approximately 0.6 mL of the sample

solution was transferred to a 5 mm NMR tube, which was inserted in the magnet to perform

the experiment. Chemical shifts were recorded in parts per million (ppm) and were

referenced to solvent peaks (at 7.24 ppm and 77.23 ppm, for example, for 1H and 13C NMR in

CDCl3).

Page 80: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

44

3.2.1.1.3 Elemental Analysis Elemental analyses were performed by the Microanalytical Service, School of Chemistry and

Molecular Biosciences, the University of Queensland. The samples were extensively (ca.

24 h) dried in vacuo at 60 °C. The C/N ratio determined with elemental analysis was used to

evaluate the degree of substitution (DS) of the intermediates and the final product.

3.2.1.1.4 X-ray Photoelectron Spectroscopy (XPS) The XPS analysis of chitosan, L-leucine and chitosan-N-L-leucine.HCl was performed with a

Kratos Axis Ultra Spectrophotometer (Kratos Analytical, Manchester, UK) equipped with

monochromatized Aluminium X-ray source (powered at 10 mA and 15 KV), 165 mm radius

hemispherical analyser (HSA) and 8-channel electron multiplier (channeltron) detection

system. Samples were mounted onto stainless steel sample holders using double-sided

adhesive tape and the spectra were collected using an analysis area of 700 x 300 µm. Low-

resolution wide scans (survey spectrum) in the binding energy scale (0–1200 eV with 1.0 eV

steps) were collected at a constant analyser pass energy of 160 eV to identify the elements

present. High-resolution narrow scans (multiplex spectra) were collected at 20 eV with

0.05 eV steps for O1s, N1s, C1s, and Cl2p to identify their chemical status. The data were

processed using Casa XPS software Version 2.3.14. Binding energies (BE) of the various

elements were referenced to the C1s line at 285.0 eV.

3.2.1.2 Synthesis The sequence of steps followed in the synthesis of chitosan-N-L-Leucine.HCl (6) from

chitosan (1) are shown in the Fig. 3-2. Fig. 3-3 shows the schemes followed for the synthesis

of N-phthaloyl-3,6-di-O-acetyl-chitosan (2a) and N-phthaloyl-3-O-acetyl-6-O-trityl-chitosan

(3a) — two additional products with enhanced solubility synthesized as a tool for further

structural elucidation and confirmation of the intermediates, N-phthaloyl-chitosan (2) and

N-phthaloyl-6-O-trityl-chitosan (3).

Page 81: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

45

O NHN

OO

O

Pyridine/Ar0°C, 3

h

r.t., 21 h

Boc-leu-OSuO

HOO

OTr

n

NH

NH

O

OO

4M HCl

in

Dioxane

r.t., 24 h/

Ar

OHO

O

OH

n

NH

NH2.HClO

6-O-Trityl-chitosan-N-Boc-L-leucineChitosan-N-L-leucine.HCl

O

HONH2

O

OH

123

45

6 OO

O

Chitosan

DMF/130°C/Ar8

h

OHO

N

O

OH

n

OO

N-Phthaloyl-chitosan

Pyridine/ Reflux

(115°C) /Ar

24 h

H2O/ Reflux

(115°C) /Ar

18 h

NH2NH2.H2O

OHO

NH2

O

OTr

n

6-O-Trityl-chitosan

n

OHO

N

O

OTr

n

OO

N-phthaloyl-6-O-Trityl-chitosan

Cl

1

2

4

56

O

O

3

Figure 3- 2: Synthetic Pathway for Conjugation of L-leucine to Chitosan (1)

OOH

NHAcHOO

HON

O

OH

O n

OO

N-Phthaloyl-chitosan

Ac2O, Pyridine

OOAc

NHAcOAcO

OAcN

O

OAc

O n

OO

N-Phthaloyl-3,6-Di-O-acetyl-chitosan

2

130°C, 24 h

2a

OOTr

NHAcHOO

HON

O

OTr

O n

OO

N-Phthaloyl-6-O-trityl-chitosan

Ac2O, Pyridine

OOTr

NHAcAcOO

AcON

O

OTr

O n

OO

N-Phthaloyl-3-O-acetyl-6-O-trityl-chitosan

1300C, 24 h

3 3a

Figure 3- 3: O-Acetylation of N-phthaloyl-chitosan (2) and N-phthaloyl-6-O-trityl-chitosan (3)

[N.B.: Ac = Acetyl, Tr = Trityl]

Page 82: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

46

3.2.1.2.1 N-Phthaloyl-chitosan (2) N-Phthaloyl-chitosoan (2) was synthesized according to the method of Holappa et al. (2004)

with some modifications. Briefly, phthalic anhydride (2.43 g, 16.4 mmol) was added to a

dispersion of chitosan obtained by overnight stirring of chitosan (1 g, 5.48 mmol free –NH2

group) in DMF (20 mL) containing 5% (v/v) water. The mixture was heated with stirring at

130 °C for 8 h, cooled to room temperature and finally poured into ice water. The

precipitate was collected by filtration, washed with copious amounts of methanol and dried

to give 1.56 g (94.70%) of a pale tan powder. DS: 0.91. IR (ATR): ν 3700-3100 (O─H stretch

overlapping N─H stretch), 2980-2830 (C─H stretch, pyranose), 1774 (C=O stretch, imide),

1703 (C=O stretch, imide), 1612 (amide I, N-acetyl), 1547 (N─H bend overlapping amide II),

1468 (asymmetric C─H bend in CH2), 1385 (C=C, phth), 1200-800 (C-O stretch, pyranose),

718 cm-1 (arom, phthaloyl). Elemental Analysis: Calcd.: C, 53.73; H, 5.00; N, 4.63. Found: C,

51.25; H, 4.87; N, 4.44.

3.2.1.2.2 N-Phthaloyl-3,6-Di-O-acetyl-chitosan (2a) N-Phthaloyl-3,6-Di-O-acetyl-chitosan (2a) was synthesized following the method described

by Nishimura et al. (1991) with few modifications. In brief, acetic anhydride (10 mL, 105.8

mmol) was added to a suspension of N-phthaloyl-chitosan 2 (100 mg, 0.33 mmol) in pyridine

(20 mL) and heated with stirring at 130 °C overnight. The resulting homogeneous mixture

was cooled to room temperature and precipitated in ice-water (60 mL). The precipitate was

washed successively with ethanol and ether and dried to give 124 mg of 2a (97%). DS: 1.97.

IR (ATR): ν 3700-3100 (O─H stretch overlapping N─H stretch), 2980-2830 (C─H stretch,

pyranose), 1777 (C=O stretch, imide), 1743(C=O stretch, O-acetyl ester), 1711(C=O stretch,

imide), 1613 (amide I, N-acetyl), 1543 (N─H bend overlapping amide II), 1469 (asymmetric

C─H bend in CH2), 1430 (asymmetric C─H bend in CH3), 1385 (C=C, phth), 1371 (symmetric

C─H bend in CH3), 1219 (C─O stretch, O-acetyl), 1200-800 (C-O stretch, pyranose), 722 cm-1

(arom, phthaloyl). 1H NMR (CDCl3): δ 1.63-2.19 (0- and N-acetyl), 2.80-5.90 (pyranose), 7.68,

7.74 ppm (arom, phth). 13C NMR (CDCl3): δ 20.4 -20.8 (0- and N-acetyl), 55.3 (C-2), 62.1 (C-6),

70.2 (C-3), 72.4 (C-5), and 75.3 (C-4), 97.2 (C-1), 124.0, 131.3 and 134.6 (arom, phth), 167.7-

170.2 ppm (C=O). Elemental Analysis: Calcd.: C, 54.47; H, 4.96; N, 3.63. Found: C, 54.48; H,

4.88; N, 3.66.

Page 83: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

47

3.2.1.2.3 N-Phthaloyl-6-O-trityl-chitosan (3) N-Phthaloyl-6-O-trityl-chitosan (3) was synthesized from N-phthaloyl-chitosan (2) according

to the method of Zhang et al. (2008a) with few modifications. Briefly, N-phthaloyl-chitosan 2

(1 g, 3.32 mmol free ─OH), suspended in pyridine (47 mL), was treated with trityl chloride

(9.23 g, 33.1 mmol), stirred with heating at reflux (~115 °C) for 24 hours under Argon. After

cooling to room temperature, the reaction mixture was poured into EtOH. The precipitate

was collected by filtration and washed successively with EtOH and Et2O. The yield of the

product was 1.45 g (80%). DS: 1.06. IR (KBR): ν 3700-3100 (O─H stretch overlapping N─H

stretch), 3100-3000 (C─H, trityl), 2980-2830 (C─H, pyranose and CH3), 1776 (C=O, imide),

1712 (C=O, imide), 1611 (amide I, N-acetyl), 1591 (N─H bend overlapping amide II), 1490

(C=C, trityl), 1468 (asymmetric C─H bend in CH2), 1448 (C=C, trityl), 1384 (C=C, phth) 1200-

800 (C-O, pyranose), 764 (arom, trityl), 746 (arom, trityl), 719 (arom, phth), 699 cm-1 (arom,

trityl). Elemental Analysis: Calcd.: C, 71.72; H, 5.38; N, 2.57. Found: C, 71.91; H, 5.27; N,

2.50.

3.2.1.2.4 N-Phthaloyl-3-O-acetyl-6-O-trityl-chitosan (3a) N-Phthaloyl-3-O-acetyl-6-O-trityl-chitosan (3a) was synthesized following the same

procedure as described above for N-phthaloyl-3,6-di-O-acetyl-chitosan (2a). In brief, acetic

anhydride (10 mL, 105.8 mmol) was added to a suspension of N-phthaloyl-6-O-trityl-chitosan

3 (100 mg, 0.18 mmol) in pyridine (20 mL) and heated overnight at 130 °C with reflux. The

resulting solution was cooled to room temperature and precipitated by pouring into ice-

water (60 mL). The precipitate was collected by filtration, washed successively with ethanol

and ether and dried to give a yield of 90 mg. This was 83% of the theoretical yield assuming

that 100% substitution has taken place. The microanalysis results were not satisfactory and

so could not be used for calculating the actual DS. IR (ATR): ν 3700-3100 (O─H stretch

overlapping N─H stretch), 3100-3000 (C─H, trityl), 2980-2830 (C-H stretch, pyranose and

CH3), 1777 (C=O stretch, imide), 1745 (C=O stretch, O-acetyl ester), 1714 (C=O stretch,

imide), 1612 (amide I, N-acetyl), 1595 (N─H bend), 1547 (amide II, N-acetyl), 1529 (C=C,

trityl), 1491 (C=C, trityl), 1468 (asymmetric C─H bend in CH2), 1449 (C=C, trityl), 1385 (C=C,

phth), 1220 (C─O stretch, O-acetyl), 1200-800 (C-O stretch, pyranose), 765 (arom, trityl),

747 (arom, trityl), 721 (arom, phthaloyl), 704 cm-1 (arom, trityl). 1H NMR (CDCl3): δ 1.63-2.05

Page 84: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

48

(0- and N-acetyl), 3.00-5.80 (pyranose), 6.89-7.28 (arom, trityl associated with residual

solvent peak at 7.24), 7.68, 7.74 ppm (arom, phth). 13C NMR (CDCl3): δ 20.4 -20.8 (0- and N-

acetyl), 55.3 (C-2), 62.1 (C-6), 70.2 (C-3), 72.4 (C-5), and 75.3 (C-4), 97.2 (C-1), 123.9-147.0

(arom, trityl and phth), 167.8-170.3 (C=O, N-acetyl and phth). Elemental Analysis: Calcd.:

C, 70.68; H, 5.33; N, 2.39. Found: C, 61.65; H, 4.89; N, 3.05.

3.2.1.2.5 6-O-Trityl-chitosan (4) Deprotection of the phthaloyl group from N-phthaloyl-6-O-trityl-chitosan (3) to produce

6-O-trityl-chitosan (4) was performed according to the methods reported by other

investigators (Nishimura et al., 1991; Zhang et al., 2008a) with some modifications. Briefly, a

suspension of N-phthaloyl-6-O-trityl-chitosan 3 (1 g, 1.84 mmol ─NH2 group equivalent) in

hydrazine hydrate (50-60%, 50 mL) was stirred with heating at reflux (~110 °C) for 18 hours

under an argon atmosphere. The reaction mixture was cooled to room temperature and

poured into water (500 mL). The precipitate was filtered off and washed with water (2 x 500

mL) and finally with ethanol and ether. The yield of the product was 0.75 g (96%).

Microanalytical data was unsatisfactory to calculate the actual degree of dephthaloylation.

However, FT-IR and 1H NMR spectra indicate complete removal of the phthaloyl group. IR

(ATR): ν 3700-3100 (O─H stretch overlapping N─H stretch), 3100-3000 (C─H, trityl), 2980-

2830 (C─H, pyranose and CH3), 1661 (amide I, N-acetyl), 1596 (amide II overlapping N─H

bend), 1490 (C=C, trityl), 1448 (C=C, trityl), 1374 (symmetric C─H bend in CH3), 1318

(amide III), 1200-800 (C-O, pyranose), 763 (arom, trityl), 746 (arom, trityl), 697 cm-1 (arom,

trityl). 1H NMR: δ 1.67-2.13 (CH3, N-acetyl), 3.00-5.79 (pyranose), 7.28, 7.38, 7.79 ppm

(arom, trityl). Elemental Analysis: Calcd.: C, 71.13; H, 6.44; N, 3.30. Found: C, 71.14; H, 6.13;

N, 3.54.

3.2.1.2.6 6-O-Trityl-chitosan-N-Boc-L-leucine (5) A solution of Boc-leu-OSu (1.16 g, 3.53 mmol) in pyridine (15 mL) was added drop-wise to

6-O-trityl-chitosan 4 (500 mg, 1.18 mmol -NH2 group equivalent) at an ice-cooled

temperature over a period of about 1 h under an argon atmosphere and the reaction

mixture was stirred for 3 h at 0 °C. An additional 772.88 mg (2.35 mmol) of Boc-leu-OSu in

pyridine (10 mL) was then added drop-wise to the reaction mixture and the reaction was

Page 85: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

49

continued at room temperature under argon for a further 21 h until a clear solution was

obtained. Finally, the product was precipitated by pouring the reaction mixture into diethyl

ether (250 mL). The precipitate was washed with di-ethyl ether (x 3) and dried to give

485 mg (70%) of 6-O-trityl-chitosan-N-Boc-L-leucine (5) as a solid. DS: 0.74. IR (ATR): ν 3700-

3100 (O─H stretch overlapping N─H stretch), 3100-3000 (C─H, trityl), 2980-2830 (C─H,

pyranose & Boc-Leucine), 1683 (amide I, N-acetyl, N-L-leucine and N-carbamide), 1597

(amide II overlapping N─H bend), 1491 (C=C, trityl), 1448 (C=C, trityl), 1390 and 1367 (Boc

t-butyl and leucine isopropyl), 1319 (amide III), 1200-800 (C-O, pyranose), 764 (arom, trityl),

747 (arom, trityl), 702cm-1 (arom, trityl). 1H NMR: δ 0.90 (H-δ1, leu), 1.27 (H-γ, leu

overlapped with t-butyl, Boc), 1.44 (t-butyl, Boc overlapped with CH3, N-acetyl at 1.66 ppm),

1.92 (H-β, leu overlapped with H-δ1), 2.15 (CH3, N-acetyl), 2.65 (H-δ2, leu), 3.00-5.5

(pyranose protons overlapped with H-α, leu at 4.51), 7.28, 7.40 and 7.78 ppm (arom, trityl). 13C NMR (CDCl3): δ 20.7-23.8 (CH3, N-acetyl), 25.3 & 26.4 (C-δ1&2, leu), 28.9 (t-butyl, Boc),

30.3 (C-γ, leu), 43.0 (C-β, leu), 54.5 (C-α, leu), 56.3 (C-2), 58.4 (C-6), 72.6 (C-3), 75.8 (C-5),

77.4 (C-4), 79.2 (C(CH3)3), 87.1 (C-Ph3), 97.4, 101.0, 102.0, 104.5 and 104.8 (C-1), 127.8,

128.7, 129.9, & 145.0 (arom, trityl), 156.9 (C=O, carbamide), 173.6 (C=O, N-leu), 176.3 ppm

(C=O, N-acetyl). Elemental Analysis: Calcd.: C, 68.23; H, 7.25; N, 4.33. Found: C, 67.98; H,

6.78; N, 4.14.

3.2.1.2.7 Chitosan-N-L-leucine.HCl (6) 4M HCl in 1, 4-dioxane (4 mL) was added drop-wise to 6-O-trityl-chitosan-N-Boc-L-Leucine 5

(200 mg, 343 µmol) at ice-cold temperature under an argon atmosphere and stirred for

15 min at this temperature. Stirring was continued at room temperature under argon for

24 h. The reaction mixture was poured into di-ethyl ether (40 mL), filtered off, washed with

ether (x 3) and dried to give the product, chitosan-N-L-Leucine.HCl 6 (80 mg, 66%). The

microanlaytical data suggest that 91% of the protecting Boc and triytl groups have been

removed. IR (ATR): ν 3700-3100 (O─H stretch overlapping N─H stretch), 2980-2830 (C─H,

pyranose & L-leucine), 1672 & 1628 (amide I, N-acetyl overlapping N-L-leucine), 1558 & 1512

(amide II overlapping symmetric N─H bend in NH3+), 1390 and 1372 (isopropyl, leu), 1322

(amide III), 1200-800 cm-1 (C-O, pyranose). 1H NMR: δ 0.98 (H-δ1&2, leu), 1.78 (H-δ2 and H-β

of leu with some overlapping with H-δ1 and H-γ), 2.07 (N-acetyl and H-δ1, leu), 2.80 (H-γ,

Page 86: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

50

leu), 3.19 (H-2, pyranose), 3.30-4.34 (H-2 to H-6, pyranose overlapped with H-α and H-β of

leu), 4.50-5.54 ppm (H-1, pyranose), 13C NMR (CDCl3): δ 20.7 (CH3, N-acetyl), 21.7 & 23.5

(C-δ1&2, leu), 25.0 (C-γ, leu), 40.0 (C-β, leu), 52.0 (C-α, leu), 55.6 (C-2), 59.8 (C-6), 69.-76.1

(C-3, C-5, C-4), 97.3-101.1 (C-1), 170.8 (C=O, N-leu), 174.4 ppm (C=O, N-acetyl). Elemental

Analysis: Calcd.: C, 43.81; H, 7.62; N, 8.40. Found: C, 36.84; H, 7.14; N, 6.93. XPS Analysis

(Atom%): Chitosan: O, 28.59; C, 63.02; N, 7.34, Cl 1.05. L-leucine: O, 19.40; C, 69.44;

N, 11.55. Chitosan-N-L-leucine.HCl: O, 25.68; C, 60.99; N, 9.31; F, 0.64; Cl, 3.38.

3.2.2 Preparation of Chitosan and Chitosan-L-Leucine Conjugate Nanoparticles and In Vitro Studies on their Aerosolization Performance and Drug Release

3.2.2.1 Preparation of Chitosan and Chitosan-L-leucine Conjugate Nanoparticles

Chitosan and chitosan-L-leucine conjugate nanoparticles were prepared by water-in-oil

(W/O) emulsification process in two different ways, viz. 1) emulsion-solvent evaporation and

2) emulsion-glutaraldehyde cross-linking. Both the approaches shared a common initial step

of emulsifying an aqueous solution of the polymer (with or without drug) into nano-droplets

in heavy paraffin oil with a high-shear homogenizer, Heidolph Diax 900 (Sigma-Aldrich Pty

Ltd., Australia). The droplets in the resulting water-in-oil (W/O) emulsion were then

hardened into nanoparticles either by applying heat to evaporate the aqueous phase

(Method-1) or by cross-linking with glutaraldehyde (Method-2).

3.2.2.1.1 W/O Emulsion-Solvent Evaporation Method In this approach, chitosan and conjugate nanoparticles were prepared by a technique

modified from that reported previously for fabrication of chitosan microparticles (Abd El-

Hameed & Kellaway, 1997; Lim et al., 2000; Onishi et al., 2005). In brief, for fabricating blank

chitosan nanoparticles, 2.5 mL of a 2% solution of chitosan in 2% acetic acid was added

drop-wise at a temperature of 60 °C to 100 g heavy paraffin oil containing 1 mL span 80 and

homogenized into a W/O emulsion at 11,000 rpm for 10 min. The emulsion was stirred

overnight at 5000 rpm and 60 °C using an IKa® Eurostar Digital overhead stirrer (IKA® Works

(Asia), Inc.) to remove the internal water phase and harden the droplets into nanoparticles.

Then, the nanoparticles were separated from the oil phase by centrifugation overnight at

5,000 x g using an Allegra X15R Bench top Centrifuge (Beckman-Coulter, Inc. USA), washed 3

times with hexane to remove excess oil and dried under vacuum at 60 °C.

Page 87: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

51

For making blank chitosan-L-leucine conjugate nanoparticles, the same protocol was

followed except that, being freely soluble in water, no acetic acid was required for preparing

the solution of the conjugate in water.

For preparing drug-loaded chitosan and conjugate nanoparticles, 50% of the polymer was

replaced by the drug, DH. All other conditions were kept unchanged. To clarify, a solution

containing 1% each of chitosan and DH in 2% acetic acid was added drop-wise at a

temperature of 60 °C to 100 g heavy paraffin oil containing 1 mL span 80 and homogenized

into a W/O emulsion at 11,000 rpm for 10 min. The emulsion was stirred overnight at 5000

rpm and 60 °C using an IKa® Eurostar Digital overhead stirrer (IKA® Works (Asia), Inc.) to

remove the internal water phase and harden the droplets into nanoparticles. Then, the

nanoparticles were separated from the oil phase by centrifugation overnight at 5,000 x g

using an Allegra X15R Bench top Centrifuge (Beckman-Coulter, Inc. USA), washed 3 times

with hexane to remove excess oil and dried under vacuum at 60 °C.

3.2.2.1.2 W/O Emulsion-Glutaraldehyde Crosslinking Method In this approach, the same protocol was followed as described above except that no heat

was applied either during emulsification of the aqueous polymeric solution into the oil or

later. Instead, for hardening of the droplets into particles, a 50% aqueous solution of

glutaraldehyde (1 mL and 0.75 mL per 25 mg of chitosan and conjugate, respectively) was

added and the mixture was stirred as above at 5000 rpm for 12 h. The addition of the

glutaraldehyde solution was done in 3 portions (1 × 0.5 mL and 2 × 0.25 mL for chitosan and

3 × 0.25 mL for the conjugate) at intervals of 10 minutes, 1 hour and 2 hour as reported

previously by Deveswaran et al. (2007). Then, the nanoparticles were separated from the oil

phase by centrifugation overnight at 5,000 x g using an Allegra X15R Bench top Centrifuge

(Beckman-Coulter, Inc. USA), washed 3 times with hexane to remove excess oil. Finally, the

isolated particles were additionally washed 3-5 times with diethyl ether to remove

residual glutaraldehyde. Water and ethanol were not considered appropriate for this

purpose to avoid leaching of the drug, DH, which is highly soluble in these solvents.

Page 88: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

52

Figure 3- 4: A Simplified Scheme of Methods Used for the Preparation of Nanoparticles

[N.B. : Method 1 — Emusion- solvent evaporation, Method-2 — Emulsion- glutaraldehyde crosslinking]

3.2.2.2 Scanning Electron Microscopy The size, shape and surface morphology of chitosan and chitosan-L-leucine conjugate

nanoparticles were investigated by scanning electron microscopy (SEM). The SEM imaging

was done on an ultra-high resolution scanning electron microscope, JEOL 7001F (JEOL

Australasia Pty Ltd, Frenchs Forest, Australia). A tiny drop (5 μL) of nanoparticle suspension

in hexane (100 µg/mL) was placed on carbon adhesive tape adhered onto a properly labelled

aluminium stub. The specimen stubs were then sputter coated with a thin layer of gold with

a Leica EM SCD005 sputter coater (Leica Microsystems, North Ryde, Australia) at 30 mA for

1.5 min using an argon gas purge. The specimens were examined with the SEM under high

vacuum using a spot size of 10 with an accelerating voltage of 20.0 kV and a working

distance (WD) of 10 mm. Several photomicrographs (secondary electron images) of the

samples were recorded at different magnifications.

Page 89: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

53

3.2.2.3 Zetasizer Analysis The size distribution and polydispersity index (PDI) of chitosan and chitosan-L-leucine

conjugate nanoparticles were measured by dynamic light scattering (DLS) using a ZetaSizer

Nano S instrument (Malvern Instruments Ltd., UK) equipped with a 4 mW He–Ne laser

operating at a wavelength of 633 nm. The analysis was performed at 25 °C with a detection

angle of 173°. For analysis, the particles were diluted to an appropriate concentration

(ca. 0.1%) with heavy mineral oil containing 1% span 80 as the dispersant and placed in a

glass cuvette with square aperture (Sarstedt AG & Co., Nümbrecht, Germany). The

measurement was done in triplicate and averaged. The reported size is the Z-average (or

“cumulants mean”) for the particle hydrodynamic diameters calculated from the intensity of

scattered light, and the PDI is a width parameter, which was also calculated from the

cumulants analysis of the signal intensity. Data acquisition and analysis was done by Malvern

Zetasizer Software version 6.32. The viscosity and refractive index values of the dispersant,

heavy paraffin oil with 1% span 80 (used for data analysis) were determined to be 73.00 cps

(at 25 °C) and 1.482, respectively by a Cannon-Fenske Routine Viscometer (Poulten, Selfe

and Lee, Ltd. England) and an Abbe Refractometer (Atago Co., Ltd., Tokyo, Japan),

respectively.

3.2.2.4 Estimation of Production Yield, Drug Loading and Entrapment Efficiency

3.2.2.4.1 Production Yield The yields of nanoparticles were quantified as the percentage of anticipated yields. The total

amount of nanoparticles obtained was weighed and the percentage yield was calculated

using the following formula (Dandge & Dehghan, 2009):

Production yield (%) = 𝑊1𝑊2

x 100 (1)

Where W1 = weight of dried nanoparticle

W2 = sum of dry weight of the starting materials.

The measurement was done in triplicate (n=3).

Page 90: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

54

3.2.2.4.2 Drug Loading and Entrapment Efficiency To determine drug loading and entrapment efficiency, an aliquot (5 mL) of the external oil

phase was centrifuged to remove the particles. Then, the free drug in the supernatant was

extracted into an excess of PBS (1:4) and analyzed by UV spectrophotometry (Beckman

Coulter DU 800 UV/Vis Spectrophotometer, USA) at 235 nm. Each sample was assayed in

triplicate (n = 3). Amount of drug in the supernatant was subtracted from the total amount

of drug initially added for making nanoparticles. The drug loading and entrapment efficiency

were calculated as follows (Chimote & Banerjee, 2009; Grenha et al., 2007):

Drug loading (%) = Total drug−Free drugNanoparticle weight

x 100 (2)

Entrapment efficiency (%) = Total drug−Free drugTotal drug

x 100 (3)

3.2.2.5 In Vitro Drug Release Study Drug release from the nanoparticles was studied according to the method reported by

Masotti et al. (2007) with few modifications. Briefly, an aliquot of nanoparticles equivalent

to 2.5 mg DH was incubated in 50 mL PBS (pH = 7.3±0.2, 37 °C) under gentle magnetic

stirring (100 rpm). At fixed time intervals (0.5 h, 1 h, 2 h, 4 h, 6 h, 12 h, 24 h and later every

24 h up to 30 days), the samples were centrifuged and 5 mL aliquots of supernatant was

removed. An equal volume of fresh buffer was replaced immediately. The withdrawn

samples were analysed by UV spectrophotometry. All measurements were performed in

triplicate (n=3).

To ascertain the release kinetics and the mechanism of release, the release data were

analysed by fitting to commonly used mathematical models, viz. zero order model, 1st order

model, Higuchi’s square root model (Higuchi, 1963), Hixson-Crowell model (Hixson &

Crowell, 1931) and Korsmeyer-Peppas model (Korsmeyer et al., 1983) according to the

following equations:

Page 91: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

55

Zero order model: F=kot (4)

1st order model: ln (1-F)=-k1t (5)

Higuchi model: F=KHt1/2 (6)

Hixson-Crowell model: 1-(1-F)1/3=k1/3t (7)

Korsmeyer-Peppas model: F=kK-Ptn (8)

where F denotes cumulative fraction of drug released at time t. k0, k1, kH, k1/3 and kK-P are

the apparent release rate constants of the respective mathematical models.

n is the release exponent of the Korsmeyer-Peppas model, indicative of the mechanism of

drug release. n ≤ 0.45 corresponds to a Fickian diffusion (case I transport), 0.45 < n ≤ 0.89 to

an anomalous (non-Fickian) transport, n = 0.89 to a zero-order (case II) release kinetics, and

n > 0.89 to a super case II transport. Case II and supercase II transports indicate involvement

of polymer chain relaxation and erosion in drug release.

The initial loading dose (DL) providing for prompt achievement of the desired blood level and

the desired maintenance dose release rate (MDR) to maintain the desired blood level over

extended period of time can be given by the following equations (Lingam et al., 2008; Prasad

& Kishore, 2012):

DL = Cd.Vd/F (9)

MDR = ke.Cd.Vd (10)

where Cd is the desired blood level, Vd is the apparent volume of distribution, ke is the first

order rate constant for overall drug elimination and F is the fraction of the dose absorbed.

For DH, Cd = 0.05 μg/mL, Vd = 3.1 L/Kg and ke = 0.19/h (Prasad & Kishore, 2012). Assuming

F=1 for the pulmonary route, for a 70-Kg healthy adult the DL and MDR for DH stand out to

be 10.85 mg and 2.06 mg/h, respectively.

Page 92: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

56

3.2.2.6 In Vitro Evaluation of Aerosolization and Lung Deposition The aerosolization efficiency of the dry powder inhaler (DPI) formulations (chitosan and

chitosan-L-leucine conjugate nanoparticles) was investigated in vitro by a Twin Stage

Impinger (TSI, Copley Scientific, Nottingham, UK) (Fig. 3-5) using the method described in

the British Pharmacopoeia (TSI apparatus, British Pharmacopoeia, 2013). Rotahaler® (Glaxo

Wellcome) was used as the DPI device and PBS as the collection liquid in the upper and

lower stages of the apparatus. 7 mL of PBS was placed in stage-1 (S1) and 30 mL in stage-2

(S2) of the TSI. PBS was used to maintain a consistency with the spectrometric analyses

made during drug release study and determination of entrapment efficiency. The airflow

through the TSI was controlled at 65±5 L/min by a vacuum pump (D-63150, Erweka,

Germany) using a calibrated digital flow meter (Fisher and Porter, Model 10A3567SAX, UK).

For each actuation, a size 3 hard gelatin capsule (Fawns and McAllan Pty Ltd., Australia) filled

with 20 mg of nanoparticle formulation was inserted into the Rotahaler placed into a

moulded mouthpiece attached to the TSI apparatus. The capsule was twisted to release the

powder into the body of the DPI device. The liberated powder was drawn through the TSI at

a flow rate of 65±5 L/min for 5 seconds. This procedure was repeated at least three times

for each formulation.

Each stage (Rotahaler, S1 and S2) was washed separately and collected in appropriate

vessels. Washings from each stage was filtered repeatedly for at least 5 times through

pre-weighed nylon filters of 0.20 µm pore size and 47 mm diameter (Phenomenex, USA)

(that were dried at 60 °C temperature until reaching a constant weight). The filtration was

repeated until the filtrate became completely free from any sign of turbidity. The filters,

with retained particles, were dried at 60 °C until a constant weight was reached. The weight

of the particles deposited was determined by subtracting the weight of blank filter from the

total weight.

In case of drug-loaded nanoparticles, before collecting by filtration, the volume of washings

from each section was adjusted to 50 mL and incubated with gentle stirring for an

appropriate length of time (2 weeks for chitosan nanoparticles and 3 weeks for conjugate

nanoparticles) to allow for the drug release. Finally, the filtrate was analysed for drug

content by UV spectrophotometry at 235 nm.

Page 93: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

57

For carrier-based dispersibility tests, the drug-loaded chitosan and conjugate nanoparticles

were mixed with inhalation grade lactose monohydrate (Inhalac® 120) microparticles used

as carriers to make binary interactive mixtures for inhalation by a hand mixing technique

(Alway et al., 1996; Liu & Stewart, 1998). The nanoparticles (5%) were placed between two

layers of lactose particles in a glass test tube together with 3 ceramic beads of

approximately 10 mm in diameter and shaken vigorously for 10 minutes to ensure adequate

mixing. The ceramic beads provide a ball milling effect for breaking up the aggregates

formed during the mixing process.

The total amount of particles or drug collected from the inhaler, stage-1 (S1) and stage-2

(S2) are termed as the recovered dose (RD).

The emitted dose (ED) refers to the fraction of the RD delivered from the inhaler into stages

1 and 2 expressed as a percentage:

ED = S1+S2 RD

x 100 (11)

The fine particle fraction (FPF) is defined as the fraction of the RD deposited in the stage-2 of

TSI expressed as percentage:

FPF = S2RD

x 100 (12)

Although the rotahaler®, a unit-dose capsule-based DPI device, has been reported to be less

efficient in producing respirable fraction than many of the recently developed devices (e.g.

Aeroliser®, Dinkihaler®, Inhalator®) (Louey et al., 2004; Tang et al., 2009), it has been chosen

for this study because of its simple internal geometry and gentle mechanism for capsule

emptying and powder deagglomeration (Chan et al., 1997; Xu & Hickey, 2013). Moreover, it

represents a low-resistance device and so could be readily used by patients with impaired

lung function at the inspiratory flow rate of 60 L/min used in this study (Clark & Bailey,

1996).

Page 94: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

58

Figure 3- 5: Twin Stage Impinger (TSI)

TSI is an officially approved instrument for the assessment of deposition of the emitted dose

from inhalation aerosols (British Pharmacopoeia, 2013). Since its first description in the

literature it has been extensively used and shown to be a convenient and reliable tool for

distinguishing ‘good’ and ‘poor’ aerosols (Hallworth & Westmoreland, 1987). The apparatus

was chosen for this study because of its simplicity and ease of operation. It is true that it

divides the whole sample only into two size categories with a cut-off diameter of 6.4 µm

(Hiller et al., 1980; Timsina et al., 1994) and the separation between the two categories is

also not perfectly sharp; but, though multistage devices can give actual size distribution,

they are slow and tedious because chemical and physical quantification of collected particles

is very time consuming (Miller et al., 1992). Besides, some investigators (Holzner & Mueller,

1995) have shown that the fractions below and above 6.4 μm in the TSI and the MSLI

(multi-stage liquid impinger) were identical.

Page 95: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

59

3.2.3 In Vitro Evaluation of Toxicity and Inflammatory Activity on the Pulmonary Epithelial Cell Line, BEAS-2B

3.2.3.1 Cell line For in vitro toxicological and immunological evaluation of chitosan, its L-leucine conjugate

and their nanoparticles, the bronchial epithelial cell line, BEAS-2B was used. This is a non-

malignant cell line derived from normal human bronchial epithelial cells immortalized by an

SV40/adenovirus-12 hybrid virus (Graness et al., 2002; Murphy, 2007). Cell cultures were

grown using 75 cm2 flasks in a humidified incubator in an atmosphere of 5% CO2/95% air at

37 °C. The same conditions were maintained for incubation of the cells in the experiments to

follow.

The cell culture medium (CCM) used consisted of 500 mL RPMI 1640, 50mL FBS (heat-

inactivated), 5 mL L-glutamine (200 mM) and 5 mL penicillin-streptomycin (10K µg/mL). Cells

were subcultured every 2-3 days when they became nearly confluent.

3.2.3.2 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay

The MTT assay was performed according to previous reports (Grenha et al., 2007; Manca et

al., 2008) with some modifications. 100 µl of the culture medium containing 5 X 104 cells

was placed in each of 11 wells in a 96-well plate (Costar®, Corning Incorporated, USA) and

incubated for 24 hours. Then the medium in 10 wells (each) was replaced with 100 µl fresh

medium containing a range of concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0,

12.0 and 16.0 mg/mL) of the test material leaving one well untreated for control. After 12,

24 or 48 h of cell incubation with the test materials, 50 μl of MTT solution (0.5 mg/mL in

PBS) was added to each well. The cells were incubated for a further 4 h and the medium was

removed. The formazan crystals generated by the cells were solubilised with 100 μl DMSO

and the absorbance of each well was measured by an xMarkTM Microplate

Spectrophotometer (Bio-Rad Laboratories, USA) at 550 nm and corrected for background

absorbance using DMSO as the blank.

Page 96: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

60

The relative cell viability (%) was calculated as follows:

Viability (%) = 𝐴−𝑆CM−𝑆

x 100 (13)

Where,

A is the absorbance obtained for the test substance

S is the absorbance obtained for DMSO (blank) and

CM is the absorbance obtained for untreated cells (incubated with CCM).

The latter reading was assumed to correspond to 100% cell viability.

Figure 3- 6: A Simplified Scheme of MTT Cell Viability Assay

Page 97: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

61

The concentration causing 50% inhibition of cell growth (IC50) was determined by regression

analysis using the points on the steep portion of the concentration effect curve according to

Adamson et al. (1991).

The assay was performed on two occasions with 3 replicates of each concentration of the

test substance in each instance.

3.2.3.3 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B Cell Monolayers

3.2.3.3.1 Determination of the Polarisation Time of the BEAS-2B Cell Culture by Trans-Epithelial Electrical Resistance (TEER) Measurement

To determine the time taken for the cells to get polarised, 200 μl of BEAS-2B cell suspension

in the cell culture medium was seeded in triplicate at a density of 5 × 105 cells/cm2 in the

apical chamber of Corning® collagen-coated PTFE transwell inserts (0.4 µm, 0.33 cm2) placed

on a 24-well plate (Costar®, Corning Incorporated, USA) and 700 μl of medium was added to

the basolateral chamber. The cells were incubated and the media were replaced every

alternate day both in the apical and basolateral chambers. A blank (no cell) insert was

maintained under the same conditions. The TEER across the inserts was measured by

Millicell ERS Volt-Ohm Meter (Millipore Australia Pty. Ltd.) at 24 h intervals for 10 days. Net

TEER was determined by subtracting that measured for a blank insert. The experiment was

performed twice.

Figure 3- 7: Transwell Insert

(adapted from Transwell® Permeable Supports Selection and Use Guide – Corning)

Page 98: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

62

3.2.3.3.2 Comparison of the Permeability of a Transwell Containing BEAS-2B Cell Monolayer and a Blank Transwell by Sodium Fluorescein Transport Assay

To compare the permeability of a transwell containing a confluent BEAS-2B cell monolayer

with that of a blank transwell, the permeability to Na Flu was tested according to the

method described by Grenha et al. (2007) with some modifications. Following the same

protocol as described in the section 3.2.3.3.1, cells were apically seeded in triplicate in

transwells and incubated. Blank transwell inserts were maintained under the same

conditions. After 4-5 days when the TEER reached a stable maximum value, 200 μl of

medium, containing 0.2 mg/mL of Na Flu, was replaced in the apical chamber and incubated

for equilibration for a further 30 min. Then, 100 μl samples were transferred from

basolateral chambers to black 96-well plates (Greiner Bio One, Germany) and diluted with

100 μl of 1 mM NaOH solution. The fluorescence was measured using a fluorometer,

POLARstar OPTIMA microplate reader (BMG LABTECH, Inc., USA) setting the excitation and

emission wavelengths at 485 and 530 nm, respectively. The experiment was repeated twice.

The concentration of Na Flu was determined from a standard curve drawn concurrently

from a series of Na Flu concentrations.

The apparent permeability coefficient (Papp) was measured using the following equation:

Papp (cm/s) = d𝑄d𝑡

x 1𝐴𝐶0

(14)

where d𝑄d𝑡

is the transport rate (cumulative amount of Na Flu, Q permeated across the cell

monolayers over time, t)

A is the surface area of the transwell insert (0.33 cm2) and

C0 is the initial concentration of Na Flu in the apical chamber.

3.2.3.3.3 Effect of Chitosan, Conjugate and their Nanoparticles on the Permeability of BEAS-2B Cell Monolayer

The effect of different concentrations of chitosan, conjugate and their nanoparticles on the

permeability of polarised BEAS-2B cell monolayer was investigated using Na Flu as a

permeability marker following the protocol described in the section 3.2.3.3.2. The cells were

seeded in 5 transwell inserts and incubated. After the cells became confluent, 200 µl of CCM

Page 99: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

63

containing the test substance with 0.2 mg/mL of Na Flu was replaced in the apical chamber.

Four concentrations of the test substance (viz. 0.5, 1, 2 and 4 mg/mL) were applied leaving

one insert untreated for control. No blank inserts were maintained. The cells were incubated

for 48 h and 100 μl media were withdrawn from the basolateral chambers at 0.5, 1, 2 4, 6,

12, 24 and 48 h time-points. The samples were assayed by fluorimety as described in the

previous section. The experiment was performed on two occasions and each sample was

repeated in each instance in triplicate.

Figure 3- 8: A Simplified Scheme of Na Flu Transport Assay

Page 100: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

64

3.2.3.4 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study The potential inflammatory effect of the test materials was evaluated on BEAS-2B cell line

according to the method reported (Fujisawa et al., 2000; Schulz et al., 2002) with some

modifications. Briefly, 100 µl of cell culture medium containing 5 X 104 cells was placed in

each of 5 wells in a 96-well plate (Costar®, Corning Incorporated, USA) and incubated. When

the cells reached 80-90% confluency, the medium was replaced by 100 μl of medium

containing a range of concentrations (0.5, 1, 2 and 4 mg/mL) of the test material leaving one

well untreated for control. After 24 h of incubation, supernatants were harvested and stored

at -20 °C until analysis. The supernatants were analyzed for IL-8 levels using ELISA MAX™ Kit

(Biolegend, Inc., USA) following manufacturer’s protocol (Fig. 3-9) (2011). The experiments

were done on two occasions with three replicates for each concentration.

Figure 3- 9: A Simplified Scheme for Evaluation of IL-8 Induction in BEAS-2B Cells by ELISA

Page 101: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 3 General Methods

65

3.2.4 Statistical Analysis All the pharmaceutical and cell line experiments were performed in triplicates and the

results were expressed as mean ± standard error (SE). The statistical significance of the

differences was assessed by one-way and two-way Analysis of Variance (ANOVA) with

post-hoc (Tukey-HSD) analysis; differences with a p-value of 0.05 or less were considered as

significant.

Page 102: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 103: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 104: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 105: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

66

4.1 Summary The objective of validation of an analytical method is to demonstrate that the procedure,

when correctly applied, produces results that are fit for the intended purpose. To be fit for

the intended purpose, the method must meet certain validation criteria. Typical validation

criteria, which should be considered, are: linearity, accuracy, precision, limit of detection

(LOD) and limit of quantitation (LOQ).

This chapter details the analytical validation for the following:

1) UV assay of diltiazem hydrochloride (DH) for determination of drug loading and

entrapment efficiency and in in vitro drug release and aerosolization studies.

2) Spectrofluorometric assay of sodium fluorescein (Na Flu) in trans-epithelial permeability

studies.

3) Enzyme-linked immunosorbent assay (ELISA) of Interleukin-8 (IL-8)

4) In vitro aerosolization study

5) Nanoparticle preparation — batch-to-batch variability

4.2 Analytical Validation

4.2.1 UV Spectrophotometric Assay The UV spectrum of DH in PBS was determined and the wavelength of maximum

absorbance (λmax) was found to be 235 nm (Fig. 4-1). Beer-Lambert’s calibration curve of DH

solution in PBS representing the UV absorbance at 235nm in a concentration range of 0-

25µg/mL is presented in the Fig. 4-2. A linear curve was obtained with a coefficient of

determination (r2) of 0.9989. The accuracy and precision of the assay were examined using

low, medium and high concentrations (Table 4-1). The accuracy was in the range of

97.8-102.5% and the coefficient of variation ranged from 0.002-0.006. The limit of

detection (LOD) and limit of quantitation (LOQ) were estimated to be 0.85 µg/mL and 2.82

µg/mL, respectively from the standard calibration curve.

Page 106: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

67

Table 4- 1: Accuracy and Precision for UV assay of Diltiazem Hydrochloride (n=3)

Concentration (µg/mL) Accuracy (%) Coefficient of Variation

(CV)

5 102.5 0.002

15 98.0 0.006

25 97.8 0.004

Figure 4- 1: UV Scan of Diltiazem Hydrochloride in PBS over the Range of 200-400 nm

Figure 4- 2: Beer-Lambert’s Calibration Curve of Diltiazem Hydrochloride Solution in PBS (n=3)

y = 0.0542x + 0.0105 r² = 0.9989

0.0

0.2

0.4

0.6

0.8

1.0

1.2

0 10 20 30

Abso

rban

ce

Concentration (µg/ml)

Page 107: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

68

4.2.2 Spectrofluorometric Assay The fluorescence intensity was measured for a series of concentrations of Na Flu in 1:1

mixture of RPMI and 1mM NaOH at excitation and emission wavelengths of 485 and

530 nm, respectively. A linear relationship was found between the fluorescence intensity

and Na Flu concentration in the range of 0.02-2 µg/mL with a coefficient of determination

(r²) of 0.9999 (Fig. 4-3). The accuracy for concentrations 0.02, 0.2 and 2 µg/mL was 99.1%,

100.4% and 100.0%, respectively. The coefficient of variation for these concentrations was

0.40, 0.05 and 0.01, respectively (Table 4-2). The LOD and LOQ were estimated to be 0.004

µg/mL and 0.015 µg/mL, respectively.

Figure 4- 3: Calibration Curve of Na Flu in 1:1 RPMI-NaOH (1mM) (n=3)

Table 4- 2: Accuracy and Precision for Spectrofluorometric Assay of Na Flu in 1:1 RPMI-NaOH (1mM) (n=3)

Concentration (µg/mL) Accuracy (%) Coefficient of Variation

(CV)

0.02 99.1 0.40

0.2 100.4 0.05

2 100.0 0.01

y = 4450.4x + 392.41 r² = 0.9999

0

2500

5000

7500

10000

0.0 0.5 1.0 1.5 2.0

Flur

esce

nce

inte

nsity

(a.u

.)

Concentration (µg/ml)

Page 108: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

69

4.2.3 Enzyme-linked Immunosorbent Assay (ELISA) Fig. 4-4 presents the log-log plot of the UV absorbance at 450 nm for a series of

concentrations of Human Interleukin-8 (IL-8) analysed by ELISA using Human IL-8 ELISA

MaxTM Deluxe Kit (Biolegend, USA) according to manufacturer’s protocol (2011). A linear

relationship was obtained in the concentration range of 15.625-1000 pg/mL with a

coefficient of determination (r2) of 0.9998. The accuracy and precision (coefficient of

variation) were in the range of 98.7-104.3% and 0.0007-0.0304, respectively (Table 4-3). The

LOD and LOQ were estimated to be 13.92 and 46.40 pg/mL, respectively.

Figure 4- 4: Calibration Curve for Estimation of Human IL-8 by ELISA (n=2)

Table 4- 3: Accuracy and Precision for Estimation of Human IL-8 by ELISA (n=2)

Concentration (pg/mL) Accuracy (%) Coefficient of variation (CV)

15.625 101.3 0.0021

31.25 98.7 0.0007

62.5 100.0 0.0021

125 102.7 0.0099

250 104.3 0.0007

500 101.8 0.0304

1000 100.3 0.0099

y = 0.0006x r² = 0.9998

0.001

0.01

0.1

1

10 100 1000

Abso

rban

ce

Human IL-8 Concentration (pg/ml)

Page 109: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

70

4.2.4 In Vitro Aerosolization Study The in vitro aerosol depositions from 3 replicates of chitosan nanoparticle formulations are

presented in the Fig. 4-5. The coefficient of variation (CV) for the emitted dose, stage-1 and

stage-2 deposition were 0.01, 0.04 and 0.08, respectively. This implies that there was no

significant variation among the 3 replicates in terms of aerosolization and deposition of

nanoparticles. This, in turn, indicates that the overall aerosolization, washing and analytical

procedures were reliable and reproducible.

Figure 4- 5: Comparison of TSI Deposition from 3 Replicates of Chitosan Nanoparticle Formulation (determined by gravimetric analysis)

4.2.5 Preparation of Nanoparticles — Batch-to-Batch Variability The Figs. 4-6 a, b and c presents the production yield, drug loading and entrapment

efficiency of 3 batches of drug-loaded chitosan nanoparticle formulations. The coefficients

of variation were 0.08, 0.11 and 0.05.

Fig. 4-7 presents the SEM micrographs of 3 different batches of drug-loaded chitosan

nanoparticles. The particles are similar in size (10-20 nm), shape and surface morphology

suggesting batch-to-batch reproducibility of the particles.

39 42

19

38

46

17

39 41

20

0

5

10

15

20

25

30

35

40

45

50

Rotahaler Stage-1 Stage-2

% D

epos

ition

Page 110: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 4 Method Validation

71

a b c

Figure 4- 6: (a) Production Yield (%), (b) Drug Loading (%) and (c) Entrapment Efficiency (%) of 3 Batches of Drug-loaded Chitosan Nanoparticles

Batch-1 Batch-2 Batch-3

Figure 4- 7: SEM Micrographs of 3 Batches of Drug-loaded Chitosan Nanoparticles (at x100,000 magnification)

0

20

40

60

80

100

120

140Pr

oduc

tion

Yiel

d (%

)

0

5

10

15

20

Drug

load

ing

(%)

05

1015202530354045

Entr

apm

ent e

ffici

ency

(%)

Page 111: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 112: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 113: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 114: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

72

5.1 INTRODUCTION As already deatailed in Chpater 2, the natural biopolymer chitosan has got many attractive

features and thus earned a special attention of scientists in diverse fields including

medicine, pharmacy and biotechnology (Denkbas & Ottenbrite, 2006; Krajewska, 2004; Lim

& Hudson, 2003; Roy et al., 1999; van der Lubben et al., 2001). Over the years, the polymer

has been the subject of an active interest of synthetic chemists for chemical derivatizatrion

in order to improve its functional applicability (Bobu et al., 2011; Jayakumar, 2007; Kim &

Choi, 1998 ; Kurita et al., 2002b; Mourya et al., 2010; Nud'ga et al., 1973).

For many years, there has been a keen interest in chemical conjugation of amino acids with

chitosan for potential utilization in various fields including adsorption of heavy metals (Ishii

et al., 1995; Oshita et al., 2007b; Oshita et al., 2003), removal of low density lipoproteins (Fu

et al., 2004a) and immobilization of lipases (Yi et al., 2009). Recent reports on enhanced

dispersibility from a chitosan-based dry powder inhaler (DPI) upon addition of the amino

acid, L-leucine (Learoyd et al., 2008a, 2008b, 2009) prompted this investigator to explore

the impact of chemical conjugation of chitosan with L-leucine on the dispersibility of

particles from a DPI. As noted in the Chapter 1, based on the structural features of

L-leucine and previously reported orientation of the hydrophobic part of L-leucine (added to

a particle formulation) at the particle-air interface, it was assumed that, upon conjugation,

the L-leucine residue will be oriented around the chitosan backbone with its hydrophobic

part being projected outward, resulting in a reduced inter-particle interaction (between

particles made of conjugated chitosan) (Section 1.2, Fig. 1-1). Based on this educated guess,

it was hypothesized that the L-leucine conjugation of chitosan would enhance the

dispersiblity of the particles (made of the conjugate) from a DPI (Section 1.2).

OOHO

NHRO

OH

nR= H, -COCH3

Chitosan

123

45

6

H3COH

CH3 NH2

O

L-Leucine

Figure 5- 1: Structure of Chitosan and L-Leucine

Page 115: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

73

For testing the hypothesis, this work was designed to synthesize a conjugate of L-leucine to

chitosan that was later used for fabrication of a nanoparticulate DPI formulation. As a first

approach to this end, a conjugate of L-leucine with chitosan was synthesized by amidation

with the primary amino group at C-2. The product was found to have improved solubility in

water and was adequately characterised by FT-IR, 1H, 13C and 2D 1H-13C HSQC NMR

spectroscopy, elemental analysis and XPS.

Following the style of synthetic chemistry, the analytical data have been presented in the

experimental sections (Sections 3.2.1.2.1 to 3.2.1.2.7) in the Chapter 3 and the results are

discussed here without making separate sections for results and discussion as have been

done for formulation and toxicological works to be presented in the forthcoming Chapters 6

and 7.

5.2 RESULTS AND DISCUSSION There are three available options for conjugating L-leucine to chitosan: first, selectively to

the free ─amino group at the C-2 position; second, selectively to the primary ─OH group at

C-6; and third, simultaneously both at the C-2 and C-6 positions. Because of the insolubility

of chitosan in ordinary organic solvents and the presence of three different kinds of reactive

groups (─NH2 and primary and secondary ─OH) in its repeating units, it is not very easy to

effect controlled chemical modification reactions. For regioselective modification of the

polymer at a specified position, it is imperative to apply appropriate protection-

deprotection strategies and to perform reactions in solution using organosoluble

intermediates. Thus, for controlled functionalization at C-2 of chitosan with L-leucine, it was

important to protect the C-6 position with an appropriate group. The trityl moiety was

selected for this purpose, because this bulky, lipophilic group gives sufficient

organosolubility to chitosan for carrying out controlled functionalization of the free amino

group at C-2 (Holappa et al., 2004; Holappa et al., 2005). But, since chitosan per se is not

sufficiently organosoluble to allow its reaction with trityl chloride, it was first reacted with

phthalic anhydride to give N-phthaloyl-chitosan, which has been found to be the most

attractive option from the viewpoints of solubilization and subsequent deprotection (Kurita

et al., 1982; Nishimura et al., 1991). The deprotection of the phthaloyl group was effected

Page 116: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

74

by hydrazinolysis in aqueous solution and the resulting amine was coupled with Boc-L-

leucine succinimide (Boc-leu-OSu). Finally, both the Boc and trityl protection was removed

simultaneously by treating with 4M HCl in 1,4-dioxane to give the target conjugate as the

hydrochloride salt.

5.2.1 Synthesis and Characterization of N-Phthaloyl-Chitosan The intractability of chitosan has been attributed to its rigid crystalline structure (Saito et al.,

1981; Saito et al., 1987) and the amino functionality of the glucosamine residue is

considered to have an important contribution to it by forming intra- and inter-chain

hydrogen bonding as shown in Fig. 5-2 (Liu et al., 2004; Nishimura et al., 1991). Therefore,

chemical modification of the amino group, e.g. by N-phthaloylation, can be expected to

disrupt its inherent crystalline structure and improve solubility in general organic solvents

(Fig. 5-2) (Kurita et al., 1982; Nishimura et al., 1991). The resulting N-phthaloyl-chitosan

shows much improved solubility in organic solvents like DMF, DMAc, DMSO and pyridine

(Nishimura et al., 1991; Nishimura et al., 1990). The phthaloyl group can later be

deprotected easily to regenerate the free amino group paving way for further

functionalization at this site. Because of these advantages, N-phthaloyl-chitosan has been

utilized as a key intermediate for some regioselective and quantitative chemical

modification of chitosan (Kurita et al., 2002b; Kurita et al., 2000a; Kurita et al., 1998;

Nishimura et al., 1991; Yoksan et al., 2001; Yoksan et al., 2003).

OO OO

H H N

O

O

H

H

HO

HO

NH H

O

O

O

ODMF

OO OO

H N

O

O

H

HO

HO

NO

OO

O O

Figure 5- 2: Hydrogen Bonding in Chitosan and its Disruption by N-Phthaloylation [adapted from Nishimura et al. (1991)]

Protection of the C-2 amino group of chitosan (DDA 92%, MW 50-190 kDa) was successfully

performed by employing a 3-fold excess of phthalic anhydride in DMF containing 5% (v/v)

Page 117: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

75

water at 130 °C without any added base according to the method of Holappa et al. (2004)

with some modifications. Under these conditions, the reaction mixture turned to a viscous

gel after 7-8 hours. Before adding phthalic anhydride, chitosan was soaked in the solvent

overnight to let it swell after Rout et al. (1993) who reported that pre-swelling of chitosan

enhanced the reaction process by increasing the accessibility of the amino groups to the

reagent.

The structure of the compound was confirmed by FT-IR spectroscopy and elemental

analysis. Figs. 5-3 A and A 5-1.1 (Appendix 5-1) show the FTIR spectrum of N-phthaloyl-

chitosan. The peaks at 1703 and 1774 cm-1 for phthalimide carbonyl groups and at 718 cm-1

for the aromatic ring confirm successful substitution of the phthaloyl group for the amino

group of chitosan. Replacement of the twin broad band observed in chitosan at ~3700-3100

cm-1 region (due to overlapping O-H and N-H stretches) by a single broad band also indicates

removal of the free amino group. In addition, the peak for the primary amino group at

1584 cm−1 also almost disappeared. The peaks at 1612 and 1547 cm-1 are assigned to amide

I and amide II functionalities, respectively. The sharp band at 1385 cm-1 is assigned to

phthaloyl C=C. No absorptions around 1850 cm-1 and 1790 cm-1 (ascribable to aromatic

anhydride carbonyl bond) were observed in the spectrum indicating that purification of the

reaction product by washing with methanol completely removed any un-reacted phthalic

anhydride. These interpretations are in agreement with previously published data (Holappa

et al., 2004; Stefanescu et al., 2008; Zhang et al., 2003).

The microanalytical data of the product (C, 51.25%; H, 4.87%; N, 4.44%) closely resemble

the theoretical values (C, 53.73%; H, 5.00%; N, 4.63%) calculated for complete

phthaloylation of starting chitosan (DDA 0.92). To determine the degree of substitution

(DS), the expected C/N values were calculated for different levels of DS (0 to 0.92) and a

standard calibration curve was drawn by plotting the DS values against C/N ratios. The DS

value found by plotting the C/N ratio (51.25%/4.44%=11.54) on the standard calibration

curve was 0.91 (Fig. 5-4). With 92% DDA of the starting material, this means that 99% of the

free amino groups were substituted. The yield of the product based on this DS value was

calculated to be 95%.

Page 118: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

76

Figure 5- 3: A Combined Presentation of the FT-IR Spectra of: (A) N-Phthaloyl-Chitosan, (B) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan, (C) N-Phthaloyl-6-O-Trityl-Chitosan, (D) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan, (E) 6-O-Trityl-Chitosan, (F) 6-O-Tritylchtiosan-N-Boc-L-Leucine and (G) Chitosan-N-L-Leucine.HCl

C=O,Phth

arom,Phth

C=O,OAc

C─O,OAc

C=C,Tr arom,Tr

C=O,OAc

C─O,OAc

C=O,Phth

arom,Phth

C=O,Phth

C=O,Phth

arom,Phth

arom,Tr arom,Phth

C=C,Tr

arom,Tr

C─H,Tr

C─H,Tr

C=O,Amide

Isopropyl (Leu)

C=O, Amide

Isopropyl (Leu)/ t-butyl (Boc)

C=C,Tr arom,Tr

C─H,Tr

Page 119: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

77

Figure 5- 4: Determination of Degree of Substitution (DS) of N-Phthaloyl-Chitosan from C/N Ratio Obtained by Elemental Analysis

It has been reported that treatment of chitosan with phthalic anhydride often results in

partial O-phthaloylation in addition to N-substitution and shows some characteristic signs in

the IR spectrum of the product [viz. weak bands at 2600−2700 cm-1 (free carboxyl), medium

bands at 1250 -1290 cm-1 (ester), broadened imide carbonyl band at 1710 cm-1 owing to

overlapping with the resulting ester linkage (Kurita et al., 2002b; Kurita et al., 2000b)].

Although the mixed N,O-phthaloylated product is still a convenient organosoluble precursor

for some modification reactions (Rout et al., 1994; Rout et al., 1993), the O-phthaloyl group

is an obstacle in most cases to quantitative and regioselective substitution. However, it has

been demonstrated that this side reaction could be avoided by using a mixed solvent

containing DMF and water at 95:5 ratio (Kurita et al., 2002b). This protocol was followed

here and minimal reaction at C-3 and C-6 were detected by IR spectroscopy. Weak bands

were observed at 1250 -1290 cm-1; these may be due to trace amounts of O-phthaloylation.

There is no other sign indicative of O-phthaloylation as evident from the absence of bands

characteristic of carboxyl groups at 2600-2700 cm-1, presence of strong C-H aromatic bands

due to aromatic rings at around 2900 cm-1 and the sharp imide carbonyl band at 1703 cm-1

Page 120: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

78

Figure 5- 5: A Combined Presentation of the 1H NMR Spectra of (A) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan (in CDCl3), (B) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan (in CDCl3), (C) 6-O-Trityl-Chitosan (in Pyridine-d5), (D) 6-O-Trityl-Chitosan-N-Boc-L-Leucine (in Pyridine-d5) and (E) Chitosan-N-L-Leucine.HCl (in D2O)

(Ikeda et al., 2002; Kurita et al., 2007; Kurita et al., 2002b; Kurita et al., 2000b). This is

further supported by microanalytical data giving a DS of 0.91, which otherwise might have

exceeded the DDA value of 0.92.

Although the phthaloylated product was soluble enough for further modification reactions,

its solubility did not prove sufficient for solution-state NMR characterization. NMR

characterization of the product in a solvent like DMSO-d6 has been reported (Huang & Fang,

Arom, phth

Arom, phth Arom, Tr

Arom, Tr

Arom, Tr t-butyl, Boc

H2 to H5 H1

Pyranose

H2 to H5 H1

Pyranose

CH3 (N, O-Ac)

CH3 (N-Ac) H1 to H6 (pyranose)

CH3 (N-Ac) + H-δ1 (leu)

H-γ (leu)

CDCl3 (residual)

CDCl3 (residual)

Pyridine-d5

(residual)

Pyridine-d5

(residual)

H-β (leu)

H- δ2 (leu)

H- α (leu)

H1 to H6 (pyranose)

CH3 (N-Ac) +

H-δ1 (leu) +

H-δ2 (leu)

H- δ2 (leu) + H-β (leu)

H-γ (leu) H2 H2 to H6

+ H- α (leu)

H1

CH3 (N, O-Ac)

H2O

H2O

H2O

Pyranose

Page 121: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

79

2006; Liu et al., 2004; Zhang et al., 2003). It is possible that this difference in solubility arose

from differences in the MW and DDA of the chitosan used as the starting material.

In order to obtain a more soluble compound to enable further structural characterization of

the product by 1H and 13C NMR spectroscopy, the 3,6-di-O-acetylated derivative of the

N-phthaloylated chitosan was prepared. Complete O-acetylation was achieved by treating

the compound with excess acetic anhydride and pyridine at 130 °C for 12 h as reported by

Nishimura et al. (1991; 1990). However, the reaction could not be effected at room

temperature as was reported. This is again probably because of the differences in MW and

DDA of chitosan used for the reaction. The IR spectrum of the derivative showed

characteristic absorptions of O-acetyl groups at 1743 and 1219 cm-1 (Figs. 5-3 B and A 5-1.2

in Appendix 5-1). As expected and reported earlier by Nishimura et al. (1991; 1990), the

peracetate derivative showed appreciable solubility in chloroform, allowing for a more

definitive analysis of the structure by 1H and 13C NMR spectroscopy. The 1H NMR spectrum

of the derivative in CDCl3 (Figs. 5-5 A and A 5-2.1 in Appendix 5-2) showed conspicuous

peaks at 7.68 and 7.74 ppm for phthaloyl groups in addition to peaks for pyranose ring

protons at 2.80-5.90 ppm. The peaks for N-acetyl and O-acetyl protons appeared at

1.63-2.19 ppm. The 13C NMR data of the compound in CDCl3 was very conspicuous

(Figs. 5-6 A and A 5-3.1 in Appendix 5-3). The spectrum shows typical signals for phthaloyl

group at 124.0, 131.3 and 134.6 ppm. The signals for CH3 carbons and C=O carbons for

N,O-acetyl groups appeared at 20.4-20.8 and 167.7-170.2 ppm, respectively. The signals due

to ring carbons of hexosamine residues (55.3-97.2 ppm) were also readily assigned by

comparison with the literature. The IR and NMR data are in good agreement with that

reported earlier by Nishimura et al. (1991; 1990).

The microanalytical data (C, 54.48%; H, 4.88%; N, 3.66%) are in good agreement with

theoretical values (C, 54.47%; H, 4.96%; N, 3.63%). The DS value as calculated from the C/N

ratio (54.48%/3.66%=14.89) of analysis was 1.97 per monosaccharide residue (Fig. A 5-4.1 in

Appendix 5-4). Based on this DS value, the yield was calculated to be 97%. This means,

under the conditions applied, the reaction proceeded to completion (the theoretical

maximum DS for O-acetylation being 2.0). This further suggests that phthaloylation has

taken place exclusively at C-2.

Page 122: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

80

5.2.2 Synthesis and Characterization of N-Phthaloyl-6-O-Trityl-Chitosan Once the N-phthaloylation of chitosan had been achieved, the next step in the sequence

was introduction of trityl group at C-6. The resulting product, upon subsequent

dephthaloylation, gives trityl-chitosan, which is soluble in organic solvents and allows

modification of the free amino group in solution. The trityl group can be easily removed at a

later stage by heating in aqueous acetic acid or by treatment with stronger acids without

heating (Nishimura et al., 1991). Due to the higher reactivity of the primary hydroxyl group

and steric bulk of the trityl moiety, the reaction takes place selectively at C-6 (Nishimura et

al., 1990). However, the substitution gradually extends to the secondary hydroxyl functions

with increasing DS levels. In addition, the selectivity can also be affected upon using a larger

excess of trityl chloride (Finch, 1999).

The reaction was successfully performed by heating N-phthaloyl-chitosan in pyridine at

reflux (~115 °C) with a 10-fold excess of trityl chloride under an argon atmosphere for 24 h.

The procedure, except using the reflux condition, was based on that reported by Zhang et

al. (2008a). Other researchers (Holappa et al., 2004; Kurita et al., 2007; Nishimura et al.,

1991; Nishimura et al., 1990; Zhang et al., 2008a) conducted the reaction at 80 or 90 °C with

a 3-fold excess of trityl chloride, but under these conditions conversion to the product was

low. The reaction was very sensitive to the presence of moisture necessitating strict

measures for its exclusion, including dying the solvent, pyridine, over molecular sieves and

running the reaction under a constant flow of argon.

The product was characterized by IR and gave satisfactory elemental analysis. As shown in

the Figs. 5-3 C and A 5-1.3 (Appendix 5-1), in addition to peaks attributed to the phthaloyl

absorption, the IR spectrum also shows characteristic new peaks at 3100-2900, 1490, 1448,

764, 746 and 699 cm-1 that unambiguously confirm the presence of substituted

triphenylmethyl (trityl) group on the N-phthaloylated glucosamine residues. The peaks at

3100-2900 cm-1 are attributed to the stretching vibrations of trityl CH. The bands at 1490

and 1448 cm-1 are characteristic of the aromatic C=C stretch. The peaks at 764, 746 and 699

cm-1 correspond to monosubstitued aromatic rings. These assignments agree well with

previous reports in the literature (Holappa et al., 2004; Stefanescu et al., 2008; Yu et al.,

2006; Zhang et al., 2008a).

Page 123: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

81

The microanalytical data (C, 71.91%; H, 5.27%; N, 2.50%) are close to the theoretical values

(C, 71.72%; H, 5.38%; N, 2.57%) calculated taking 92% degree of deacetylation and 91%

degree of N-phthaloylation into consideration. The degree of substitution, as calculated

from C/N ratio of elemental analysis (71.91%/2.50%=28.76), was 1.06 (Fig. A 5-4.2 in

Appendix 5-4). Assuming that tritylation has taken place selectively at 6-OH, this is slightly

higher than the theoretical maximum of 1.0. This means that, in addition to the primary

─OH groups at C-6, some tritylation has also taken place at the secondary ─OH at C-3,

though to a very small extent (6%). Based on the DS value, the yield of the product was

calculated to be 80%.

As expected, this product was more organosoluble than the starting N-phthaloyl-chitosan,

but still not enough for obtaining satisfactory solution NMR spectra. So, again, following the

report of Nishimura (1991), the strategy of acetylating the 3-OH group with excess acetic

anhydride and pyridine was undertaken. As evident from the 1H NMR spectrum (CDCl3)

presented in the Figs. 5-5 B and A 5-2.2 (Appendix 5-2), there appeared additional peaks in

the aromatic region at 6.89-7.28 ppm for monosubstituted phenyl rings of the trityl moiety

(associated with residual solvent peak at 7.24 ppm) in addition to the phthaloyl peaks at

7.68 and 7.74 ppm. The signals for pyranose protons appeared at 3.00-5.80 ppm. The peaks

at 1.63-2.05 ppm are for N- and O-acetyl groups. The 13C NMR spectrum (Figs. 5-6 B and

A 5-3.2 in Appendix 5-3) shows a series of signals (123.9-147.0 ppm) in the aromatic region.

There are more signals in this region compared to the 13C spectrum of 3,6-di-O-acetyl-N-

pthaloyl-chitosan presented in the Figs. 5-6 A and 5-3.1 (Appendix 5-3). The additional

signals (127.5-129.9, 145.4 and 147.0 ppm) are assigned to the trityl group. The spectrum

shows other expected signals as usual (viz. signals for pyranose ring carbons at

55.3-97.2 ppm, signal for CH3 at 20.3 -20.8 ppm and signals for C=O at 167.8-170.3 ppm).

The IR spectrum (Figs. 5-3 D and A 5-1.4 in Appendix 5-1) shows characteristic absorptions

at 1745 and 1220 cm-1 due to C=O and C-O vibrations of O-acetyl groups and a decrease is

observed in the ─OH stretch vibration at 3700-3100 cm-l.

Page 124: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

82

Figure 5- 6: A Combined Presentation of 13C NMR Spectra of (A) N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan (in CDCl3), (B) N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan (in CDCl3), (C) 6-O-Trityl-Chitosan-N-Boc-L-Leucine (in Pyridine-d5) and (D) Chitosan-N-L-Leucine.HCl (in D2O)

C-2 C-1

N,O

-Ac

C-6

C-4

C-5

C-3

Arom

(p

hth)

C=O

N,O

-Ac

C-2

C-6

C-3

C-5

C-4

C-1

Arom

(p

hth)

Arom

(T

r)

C=O

N-A

c

t-bu

tyl

(Boc

)

C-δ 1

&2

(leu)

C-γ

(leu)

C-β

(leu)

C-α

(leu)

C-

2 C-

6 C-3

C-5

C-4 C(

CH3)

3

C-Ph

3

C-1

Arom

(T

r)

C=O

(car

bam

ide)

C=O

(N-le

u)

C=O

(N-A

c)

N-A

c

C-δ 1

&2

(leu)

C-γ

(leu)

C-β

(leu)

C-α

(leu)

C-

2 C-

6

C-3,

C-5

& C

-4

C-1

C=O

(N-le

u)

C=O

(N-A

c)

Page 125: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

83

It is noteworthy that some earlier workers (Kurita et al., 2007; Yu et al., 2006) reported NMR

characterization of N-phthaloyl-6-O-trityl-chitosan in solution in DMSO-d6 and pyridine-d5

without any further modification. This again may be due to the differences in the MW and

DDA of the chitosan used.

The microanalytical data for the compound (C, 61.65%; H, 4.89%; N, 3.05%) was not close

enough to the theoretical values (C, 70.68%; H, 5.33%; N, 2.39%) to be considered

satisfactory and an attempt to calculate the degree of substitution based on C/N ratio found

by analysis failed to produce any meaningful outcome. The results indicate that there is less

C and more N than expected. One possible explanation for this may be that some

detritylation has taken place during the acetylation process because of the high

temperature (130 °C) applied. This explanation is supported by the changes in the relative

intensities of aromatic phthaloyl and trityl peaks in the IR spectrum of N-phthaloyl-3-O-

acetyl-6-O-trityl-chitosan compared to those in the spectrum of N-phthaloyl-6-O-trityl-

chitosan (Fig. 5-3 (C) and (D)).

5.2.3 Synthesis and Characterization of 6-O-Trityl-Chitosan The next step in the sequence of reactions was selective deprotection of the phthaloyl

group from the C-2 position in order to make the ─NH2 group free for subsequent

conjugation to L-leucine. Removal of the phthaloyl group from N-phthaloyl-6-O-trityl-

chitosan was efficiently done by heating with an aqueous solution of hydrazine hydrate at

reflux (110 °C) for 18 h under an argon atmosphere.

As evident from the IR spectrum (Figs. 5-3 E and A 5-1.5 in Appendix 5-1) of the resulting

product, 6-O-trityl-chitosan, absorptions at 1776 and 1712 cm-1 due to phthalimido groups

and at 719 cm-1 due to phthaloyl aromatic ring disappeared completely. On the other hand,

it shows a characteristic absorption at 1596 cm-1 for the deprotected primary amino group,

in addition to those due to trityl groups (at 3100-3000, 1490, 1448, 763, 746 and 697 cm-1).

These spectral data agree well with the literature (Hu et al., 2005; Nishimura et al., 1991; Yu

et al., 2006).

Page 126: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

84

The degree of organosolubility of the product permitted the running of the 1H NMR

spectrum in pyridine-d5, but it was not enough for obtaining satisfactory 13C NMR data.

Attempted peracetylation proved unsuccessful for this product. Some workers (Yu et al.,

2006), however, reported 13C NMR analysis for this product in pyridine-d5. The Figs. 5-5 C

and A 5-2.3 (Appendix 5-2) present the 1H NMR spectrum of the compound in pyrindine-d5.

The spectrum shows trityl peaks at 7.28, 7.38 and 7.79 ppm. A broad set of peaks appear at

3.00-5.78 ppm for pyranose protons. The peaks at 1.67-2.13 ppm are for acetyl protons.

There are peaks at 5.06, 7.22, 7.59 and 8.73 for residual solvent (pyridine-d5). The signals

shifted a little downfield compared to their position in the NMR spectrum of N-phthaloyl-3-

O-acetyl-6-O-trityl-chitosan in CDCl3 (Figs. 5-5 B and A 5-2.2 in Appendix 5-2). This sort of

downfield shift of NMR signals in pyridine-d5 has previously been reported by Bernet et al.

(2000); they attributed it to the anisotropy effect of the pyridine ring.

The microanalytical data (C, 71.14%; H, 6.13%; N, 3.54%) shows slightly lower percentage of

H and higher percentage of N compared with the expected theoretical values (C, 71.13%; H,

6.44%; N, 3.30%). This suggests a greater degree of depthaloylation than the theoretical

maximum (0.91) when the observed C/N value (71.14%/3.54% = 20.10) was plotted in the

standard calibration curve (Fig. A 5-4.3 in Appendix 5-4). But, this is practically unlikely. This

aberration may again be due to removal of some trityl groups during dephthaloylation by

hydrazinolysis at high temperature condition (110 °C).

Use of appropriate concentration of hydrazine hydrate solution appeared to be crucial for

completion of the dephthaloylation process. Some earlier workers (Holappa et al., 2004;

Nishimura et al., 1991) reported dilution of hydrazine hydrate with 2 times water when

adding to the reaction mixture. The reagent used in this work contained hydrazine hydrate

in 50-60% concentration. Any further dilution of the reagent resulted in incomplete

deprotection of phthaloyl groups leaving a weak band at 1716 cm-1 in the IR spectrum (Fig. A

5-1.6 in Appendix 5-1).

Page 127: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

85

5.2.4 Synthesis and Characterization of 6-O-Trityl-Chitosan-N-Boc-L-Leucine The next step in the synthetic pathway was conjugation of L-leucine to the C-2 position of

6-O-trityI-chitosan. There are numerous reports in the literature on conjugation of various

amino acids with glucosamine or other amino sugars (Bergmann & Zervas, 1932; Kochetkov

et al., 1962; Ohnishi et al., 2000; Sosnovsky & Gnewuch, 1994). Jeon & Kim (2001) reported

conjugation of a number of amino acids to chitooligosaccharides (<10kDa). Some workers

also reported conjugation of peptides/ oligopeptides to 6-O-trityl-chitosan (Nishiyama et al.,

1999; Nishiyama et al., 2000). Some other studies described conjugation of L-leucine and

other amino acids onto the chitosan beads using epichlorohydrin (ECH) or ethylene glycol

diglycidyl ether (EGDE) as a spacer arm (Fu et al., 2004a; Oshita et al., 2007a; Yi et al., 2009).

But, no attempt has so far been reported for direct conjugation of L-leucine to chitosan or

6-O-trityl-chitosan.

The conjugation was performed by reacting 6-O-trityl-chitosan with 3 equivalents of Boc-L-

leucine-succinimide in pyridine under an atmosphere of argon. A solution of Boc-L-leucine-

succinimide in pyridine was added drop-wise to 6-O-trityl-chitosan at an extremely slow rate

(10-12 drops/min) at 0 °C. The reaction was conducted at this temperature for the first

three hours and then continued for a further 21 h at room temperature. The method was

primarily based on an earlier report of Kurita et al. (2002a) on an analogous amidation

reaction of 6-O-trityl-chitosan with N-nicotinoyl-phenylalanine. Various conditions were

investigated to optimize the reaction. Addition of further 2 equivalents of Boc-L-leucine-

succinimide 2-3 h after starting the reaction resulted in an improved yield. A constant flow

of Argon to eliminate moisture from the system was found to be critical for the success of

the reaction. Both DMF and pyridine have been found to be effective as solvents, but

pyridine gave a better dispersion of 6-O-tritylchtosan. When the reaction was run at room

temperature right from the very beginning, it gave a relatively poor yield and also resulted

in reduced intensity of the amide peak in the IR spectrum. Use of low temperature

throughout the whole length of the reaction also was not useful for an efficient outcome.

Running the experiment at an increased temperature of 60 °C was found to cause failure of

the reaction presumably by decomposition of the reagent, Boc-L-leucine-succinimide.

Addition of the reagent, Boc-L-leucine-succinimide, to 6-O-trityl-chitosan in one portion

(rather than drop-wise) was devastating; the IR spectrum suggests that even the trityl

Page 128: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

86

groups were removed from the chitosan backbone. It indicates that such quick addition of

the reagent caused vigorous reaction causing generation of heat that decomposed Boc-L-

leucine-succinimide. The generated free amino acid probably in turn cleaved the trityl

groups off the chitosan backbone. This also justifies the need for adding the reagent drop-

wise at 0 °C. Diethyl ether was found to be useful for precipitation of the product and

washing off the impurities. 10 volumes of di-ethyl ether were required for complete

precipitation of the product from the reaction mixture. It was followed by further treatment

with a modest amount (2-3 volumes) of ether for removing any remaining impurities. Use of

acetone or alcohols (methanol/ethanol) for purification reduced the yield, apparently by

dissolving part of the product. Methyl and ethyl alcohol seemed to soften the product

indicating a better solubility of the product in these solvents.

The structure of the product was confirmed by IR spectroscopy in combination with 1H and 13C NMR spectroscopy and elemental analysis. Two dimensional 1H-13C HSQC NMR

spectroscopy was also run to further confirm 1H and 13C NMR assignments and resolve

overlaps in the 1H NMR spectrum. Appearance of a strong amide band at 1683 cm-1 and a

doublet for isopropyl (L-leucine)/ t-butyl (Boc) at 1390 cm-1 and 1367 cm-1 in the IR spectrum

(Figs. 5-3 F and A 5-1.7 in Appendix 5-1) confirms successful conjugation of Boc-L-leucine to

6-O-trityl-chitosan. However, a weak band at 1597 cm-1 indicates that some unconjugated

free amino groups are left. The bands at 2980-2830 cm-1 represent aliphatic C─H stretches

of the CH, CH2 and CH3 groups in the pyranose ring and Boc-L-leucine side chain. The

expected bands for trityl groups are also evident.

The organosolubility of the product was poor for running NMR spectra, especially for

resolving the pyranose signals. In the 1H NMR spectrum (Figs. 5-5 D and A 5-2.4 in Appendix

5-2), the pyranose protons gave rise to a few extremely broad peaks at 3.0-5.8 ppm, while

peaks for pyranose carbons in the 13C NMR spectrum (Figs. 5-6 C and A 5-3.3 in Appendix

5-3) were barely discernible even after 60 h. However, both proton and carbon NMR

spectra showed well defined peaks for Boc t-butyl group and L-leucine residue, in addition

to the trityl group, at appropriate positions. The peaks for Boc t-butyl appeared at 1.44 ppm

and 28.9 ppm in 1H and 13C NMR spectra, respectively. The peaks at 25.3, 26.4, 30.3, 43.0

Page 129: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

87

and 54.5 ppm in the 13C NMR spectrum were assigned to C-δ1, C-δ2, C-γ, C-β, and C-α of L-

leucine residue. The chemical shifts at 22.7 and 23.8 in the 13C NMR spectrum were

assigned to –CH3 (N-acetyl) carbon. Pyranose and trityl carbons resonated at their typical

positions. NMR signals were also observed for C=O of carbamide, L-leucine and N-acetyl at

156.9, 173.6 and 176.3 ppm respectively as well as for the quaternary carbons of Boc and

trityl moieties at 79.2 and 87.1 ppm, respectively. These assignments for L-leucine, Boc and

trityl residues are based on some products reported in the literature, which contained the

same residues as part of their structures (Braga et al., 2004 ; Krohn et al., 2012; Perich et al.,

1987). However, as compared to these reports, the signals seem to have shifted slightly

downfield because of the anisotropic effect of the pyridine ring as shown before for 6-O-

trityl-chitosan too. The 1H-13C 2D correlation data (Fig. 5-7) was helpful in confirming the

chemical shifts of some carbons that did not give very well-defined peaks in the 1-D 13C

NMR spectrum (viz. pyranose carbons). The 2D data were also useful in assigning the signals

of different protons in the 1H NMR spectrum that showed a lot of overlap. The N-acetyl peak

at 1.66 ppm is overlapped by t-butyl, Boc signal, while the peaks for H-γ, leu and t-butyl, Boc

overlap at 1.27 ppm. On the other hand, the signals for H-β and H-δ1 of L-leucine overlap at

1.92 ppm. In addition, the chemical shift of H-α of L-leucine (4.32-4.87 ppm) overlaps on H-2

(4.37-4.58 ppm) and H-4 (4.45-4.65 ppm) of the pyranose. One limitation of the 2D

spectrum was the absence of any data points corresponding to C-1 signals. However, this

shortcoming was circumvented by the 2D data of the next and final compound in the

pathway (chitosan-N-L-leucine.HCl) that show clear data points correlating C-1 signals to

corresponding H-1 peaks (Fig. 5-8).

Page 130: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

88

O

O

NHCOCH3HOO

HOO

O

Oy

123

45

6

123

4 5

6NH

NH

O

OO

6-O-Trityl-chitosan-N-Boc-L-leucine

Tr

α

βγ

δ 1

δ 2

x

Figure 5- 7: 1H-13C HSQC NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5

H-δ

1

H-γ

t-bu

tyl (

Boc)

CH3

(N-A

c)

H-β

+ H

-δ1

CH3

(N-A

c)

H-δ

2

H-α

H-2

+ H

-6

H-3

+ H

-5 +

H-4

Arom

(Tr)

CH3 (N-Ac) C-δ1&2

C-t-butyl (Boc) C-γ

C-β

C-α C-2 C-6

C-3 + C-5 + C-4 C(CH3)3

C-Ph3

Arom (Tr)

C=O (carbamide)

C=O (N-leu) C=O (N-Ac)

Page 131: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

89

The microanalytical data of the product (C, 67.98%; H, 6.78%; N, 4.14%) were in close

agreement with the expected values (C, 68.23%; H, 7.25%; N, 4.33%). The plot of obtained

C/N ratio (67.98%/4.14%=16.42) in the standard calibration curve drawn gave a DS value of

0.74 (Fig. A 5-4.4 in Appendix 5-4). With a DDA of 0.92 and assuming that complete

dephthaloylation has been achieved in the previous step, this means 80% of the available

primary ─NH2 groups have been substituted by Boc-L-leucine residues. On the basis of this

degree of substitution, the yield was calculated to be 70%.

5.2.5 Synthesis and Characterization of Chitosan-N-L-Leucine.HCl The protecting groups, trityl and Boc, were removed from 6-O-trityl-chitosan-N-Boc-L-

Leucine by global deprotection using an excess of 4M HCl in Dioxane. Though other

researchers reported deprotection of these groups by half an hour to 2 h treatment with 4M

HCl in Dioxane (Dellaria et al., 1990; Lee & Boger, 2009; Scozzafava & Supuran, 2000 ; Tan et

al., 2007), in the present work incomplete removal of the trityl group was detected by NMR

spectroscopy even after running the experiment for 24 hours. The resulting final product

chitosan-N-L-leucine.HCl showed an appreciable water solubility that enabled

characterisation by 1H and 13C NMR spectroscopy in addition to IR spectroscopic

characterisation.

The structural details of the compound were elucidated by IR, 1H and 13C NMR spectroscopy,

elemental analysis and XPS. The IR spectrum of the product (Figs. 5-3 G and A 5-1.8 in

Appendix 5-1) shows that HCl treatment of 6-O-trityl-chitosan-N-Boc-L-leucine removed all

the characteristic peaks attributable to trityl groups without breaking the amide linkage.

Since both the isopropyl group of L-leucine and t-butyl group of Boc show characteristic

doublet peaks at the same position (around 1366 and 1390 cm-1), it was difficult to infer

from the IR spectrum if Boc protection has been removed or not and the role of NMR

spectra became more significant to reach a decision here. It is evident both from the 1H and 13C NMR spectra (Fig. 5-5 E, Fig. 5-6 D and Figs. A 5-2.5 and A 5-3.4 in Appendices 5-2 and

5-3, respectively) that peaks attributable to Boc t-butyl (1.44 ppm in 1H NMR and 28.9 ppm

in 13C NMR) have completely been removed. The NMR spectra also confirm nearly complete

Page 132: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

90

OOH

NHCOCH3HOO

HONH

O

OH

O y123

45

6

123

45

6

NH2.HClO

Chitosan-N-L-leucine.HCl

x

αβ

γδ 1

δ 2

Figure 5- 8: 1H-13C HSQC NMR Spectrum of Chitosan-N-L-Leucine.HCl in D2O

removal of all the trityl peaks from the aromatic region. In the 13C NMR spectrum, many

carbon signals, especially those from pyranose carbons, were doubled up. Apparently, this

is due to the presence of three different kinds of monomeric units in the product, viz.

unsubstituted, N-acetyl substituted and L-leucine substituted gulosamine residues. This kind

of multiplicity of carbon peaks in chitosan derivatives has also been reported previously

H-δ

1&2

H-δ

2 + H

CH3

(N-A

c) +

H-δ

1

H-γ

H-2

H-2

to

H-6

+ H

-β +

H-α

H-1

CH3 (N-Ac)

C-δ1&2 C-γ

C-β

C-α C-2 C-6

C-3 + C-5 + C-4

C-1

C=O (leu) C=O (N-Ac)

Page 133: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

91

(Crini et al., 1997; Heras et al., 2000). The HSQC spectrum (Fig. 5-8) shows that, in contrast

to the multiplicity of the carbon signals, the proton signals were overlapped. The resonance

of H-δ1&2 of L-leucine overlaps to give the intense signal at 0.98 ppm. H-δ2 and H-β of

L-leucine combined to give the signal at 1.78 ppm. This signal also has slight overlapping

with H-δ1 and H-γ. Finally, the composite signal from H2-H6 of pyranose (3.38-4.33 ppm)

overlaps with H-α and H-β of L-leucine at 3.87-4.30 and 4.04 ppm, respectively. The 2D

spectrum also reveals shifting in the resonances of some protons from their original position

in the starting material, 6-O-trityl-chitosan-N-Boc-L-leucine. Most prominent was the H-γ of

L-leucine that shifts from 1.27 ppm (most upfield of all L-leucine protons) downfield to 2.80

ppm just before the broad composite signal for H2-6 of pyranose, H-α and H-β of L-leucine.

For further confirmation of conjugation of L-leucine to chitosan, X-ray photoelectron

spectroscopic analysis (XPS) was performed on chitosan, L-leucine and the synthetic

conjugate, chitosan-N-L-Leucine.HCl. A low-resolution wide (survey) scan was performed to

detect the elements present and their relative content (Fig. A 5-5.1 in Appendix 5-5) and

then high-resolution narrow scan was performed on the major elements, viz. C, N and O to

have an in-depth look at their chemical status and to compare the spectra obtained for the

conjugate with those of chitosan and L-leucine. Figs. 5-10 and 5-11 present the multiplex

spectra for N and C. The multiplex spectrum for O is presented in Fig. A 5-5.2 in the

Appendix 5-5.

O

OH

NHCOCH3HOO

HONH2

O

OH

O123

45

6

123

4 56

Chitosan

n(1-n)

(n=0.92)Chitosan-N-L-leucine.HCl (n=0.92)

OOH

NHCOCH3HOOHO

NH

O

OH

O (1-n)123

45

6

123

4 5

6

NH2.HClO

nH3C

OHCH3 NH2

O

L-Leucine

Figure 5- 9: Structure of Chitosan, L-Leucine and and Chitosan-N-L-Leucine.HCl

The N atom in chitosan exists in three different chemical states: NH (i.e. N in free ─NH2),

Namide (i.e. N in amide linkage, ─NHCO), and N+ (i.e. quaternized N in ─NH3Cl, the amino

group attached to HCl present as impurity). As it is evident from structures of chitosan,

Page 134: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

92

L-leucine and chitosan-L-leucine.HCl (Fig. 5-9), the conjugation of L-leucine to chitosan

transforms free ─NH2 into an amide (─NHCO), thus there is a reduction in NH and a

simultaneous increase in Namide. Besides, one N+ is added per molecule of L-leucine and

hence there is an elevation in the quantity of N+. So, changes in signal intensity of different

chemical states of N could be taken as an indication of conjugation of L-leucine to chitosan.

As shown in the Fig. 5-10, XPS analysis shows that the signal intensity of NH is reduced from

5.94% (in chitosan) to 0.68% (in conjugate). On the other hand, there is a great elevation in

the intensity of the signals for Namide (from 0.72% to 5.14%) and N+ (from 0.78% to 2.99%).

These changes in the signal intensity of different chemical states of N clearly support

successful conjugation of L-leucine to chitosan.

A comparative analysis of the structures of chitosan, L-leucine and chitosan-N-L-leucine.HCl

(Fig. 5-9) further reveals that conjugation of L-leucine to chitosan adds 3 C-C carbon atoms

per molecule that resonates at 285 eV and so an elevation is expected in the signal intensity

of carbon atom in this state. Fig. 5-11 shows that the signal intensity of C-C has been

enhanced from 12.39% (in chitosan) to 19.14% (in conjugate) as expected from their

chemical structure. This provides a further evidence of conjugation of L-leucine to chitosan.

Figure 5- 10: XPS Multiplex spectra of Chitosan, L-Leucine and Chitosan-N-L-leucine.HCl for Nitrogen

N+ (0.78%) Namide (0.72%)

N-H (5.94%)

N-H (10.48%

N+ (2.99%)

Namide (5.14%)

N-H (0.68%)

Page 135: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

93

Figure 5- 11: XPS Multiplex Spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl for Carbon

An analysis of stoichiometric C/N and C/O ratios of chitosan-N-L-leucine.HCl at different

degrees of substitution (from 0 to 100%) indicates that, with increased degree of

conjugation, the C/N ratio decreases while the C/O ratio increases (Fig. 5-12 (a) and (b)).

The stoichiometric and experimental C/N and C/O ratios of the chitosan and its conjugate

under consideration are listed in the Table 5-1. Although the C/N and C/O ratios

determined by XPS are larger than those predicted from their structures, there is reduction

in C/N and elevation in C/O as expected. The deviation of experimental values from the

theoretical ones can be attributed to the presence of possible carbonaceous impurities with

chitosan and traces of trityl protection remaining with the conjugate. Furthermore, since

XPS is a surface analysis covering only a depth of 5-10 nm from the sample surface, it cannot

be expected to represent the whole sample quantitatively.

Table 5- 1: Stoichiometric and Experimental C/N And C/O Ratios of Chitosan and the Synthesized Chitosan-N-L-Leucine.HCl

Samples Stoichiometrical Experimental

C/N C/O C/N C/O

Chitosan 6.16 1.21 8.59 2.20

Chitosan-N-L-leucine.HCl 6.10 1.75 6.55 2.38

Camide (10.44%)

C-O (39.82%)

C-C (12.39%)

COO (11.65%)

C-N (15.26%)

C-C (43.68%

COO (1.21%)

Camide (9.45%)

C-C (19.14%)

C-O (33.29%)

Page 136: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 5 Synthesis and Characterization of a Chitosan-L-Leucine Conjugate

94

a

b

Figure 5- 12: (a) C/N and (b) C/O Ratios of Chitosan-N-L-Leucine.HCl at Different DS Ratios

5.3 CONCLUSION A conjugate of L-leucine and chitosan was successfully synthesized and characterised by IR

and NMR spectroscopy, XPS and elemental analysis. Among a few drawbacks in the work

was the failure to obtain satisfactory elemental analysis data for N-phthaloyl-3-O-acetyl-6-

O-trityl-chitosan. However, the purpose of synthesizing this product was to further

characterize the precursor N-phthaloyl-6-O-trityl-chitosan by 1H and 13C NMR spectroscopy

and the compound served the purpose adequately. Moreover, the parent compound

N-phthaloyl-6-O-trityl-chitosan provided satisfactory elemental analysis data. Another

limitation was the higher than expected value in elemental analysis for the dephthaloylated

product. This was attributed to probable removal of some of the trityl groups due to high

temperature conditions applied during the reaction. However, the IR and NMR data for the

compound confirms that the phthaloyl group was efficiently removed.

6.06

6.08

6.10

6.12

6.14

6.16

6.18

0.00 0.50 1.00

C/N

DS

0.00

0.50

1.00

1.50

2.00

2.50

0.00 0.50 1.00

C/O

DS

Page 137: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 138: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 139: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 140: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

95

6.1 INTRODUCTION Chitosan micro- and nano-particles have drawn special attention for controlled drug delivery

applications through the respiratory system (Al-Qadi et al., 2012; Andrade et al., 2011). In

addition to well reported biocompatibility and biodegradability, one important feature of

the polymer is its mucoadhesiveness that helps to retain the administered formulation at

the absorbing epithelial membrane, thereby prolonging drug release (Huang et al., 2003;

Learoyd et al., 2008a). Because of this interesting feature, in addition to being used as a tool

for controlling drug release, chitosan has also been widely investigated as a formulation

excipient intended to prolong drug residence at the absorbing mucosal surfaces (Boonyo et

al., 2007; Dyer et al., 2002; Illum, 1998; Soane et al., 1999).

The use of nanoparticulate systems is particularly promising both from the point of drug

transport to the lung and sustained drug action. Upon normal respiration, inhaled

nanoparticles of different sizes can target all the three regions (viz. nasopharyngeal,

tracheobronchial and alveolar) of the respiratory airways; however, nanoparticles of less

than 100 nm diameter are deposited mainly in the alveolar region (Hoet et al., 2004;

Oberdorster et al., 2005). Moreover, nanoparticles can escape mucociliary clearance and

phagocytic removal by alveolar macrophages ensuring prolonged residence of the

administered drug in the lungs for release and absorption (Ahsan et al., 2002; Grenha et al.,

2005).

One major challenge associated with a DPI system is strong cohesion shown by micronized

particles that affects their dispersibility and greatly reduces deposition of inhaled particles

to lungs. Various measures have been tried to overcome the problem which include, among

others, addition of an interactive carrier (e.g. lactose) and/or a ternary component like

magnesium stearate or L-leucine (Begat et al., 2005). Considering the promising effect of

L-leucine as an aerosolization enhancer upon physical addition to a chitosan-based

controlled release DPI formulation, as previously reported in the literature (Learoyd et al.,

2008a, 2008b, 2009), this work was designed to explore the effect of chemical conjugation

of L-leucine to chitosan from this perspective.

Page 141: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

96

Detailed literature review addressing these points has already been presented in the

Chapter 2. This chapter details the preparation of nanoparticles from chitosan and its

L-leucine conjugate as controlled release DPI formulations using the antihypertensive drug

diltiazem HCl (DH) as the model drug. In view of the extensive first-pass metabolism of the

drug following oral administration that could potentially be bypassed by respiratory

administration, it was considered as a good candidate for this work. The suitability of the

particles for pulmonary delivery is ascertained in terms of their size and morphology. A

comprehensive comparison is made between the dispersibility and drug release pattern of

the nanoparticle formulations of the two species.

6.2 RESULTS

6.2.1 Preparation of Chitosan and Chitosan-L-leucine Conjugate Nanoparticles Nanoparticles of chitosan and its L-leucine conjugate were successfully prepared using both

approaches, viz. emulsion- solvent evaporation (method-1) and emulsion- glutaraldehyde

cross-linking (method-2), as described in the Section 3.2.2.1 of the Chapter 3. However, the

solvent evaporation technique could not load the model drug, DH into chitosan particles

and showed extensive agglomeration of conjugate particles, upon incorporation of the drug,

so that the particles turned into a lump during isolation.

6.2.2 Morphology and Particle Size Analysis

6.2.2.1 Scanning Electron Microscopy Figs. 6-1 A and B present the SEM micrographs of blank chitosan and chitosan-L-leucine

conjugate nanoparticles, respectively, prepared by the method-1. The particles are mostly

spherical with a diameter predominantly in the range of 10-30 nm. On average, the

conjugate nanoparticles tend to be slightly larger than those made of unmodified chitosan.

Many particles, particularly ones with larger diameters, seem to be oblate, rather than

perfect spheres. The surface of the particles is smooth and no pores or cracks are observed.

Occasionally, some of the big particles seem to have indentations on their surface. They

appear to have arisen from fusion of multiple small particles into bigger ones. The particles

showed an aggregation tendency, though there appears to be some isolated single ones,

too.

Page 142: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

97

SEM images of blank chitosan and conjugate nanoparticles prepared by method-2 are

shown in the Figs. 6-1 C and D, respectively. They are similar to ones made by method-1 in

size, shape and surface morphology.

Drug loaded nanoparticles of chitosan and conjugate made by method-2 are presented in

the Figs. 6-1 E and F, respectively. The particles appear to be slightly smaller in diameter

(approximately 10-20 nm) than the blank nanoparticles made by either method. They

appear to consist of more regular spheres. The particles are dense with no pores,

depressions or indentations on their surface. There is no evidence of drug crystals deposited

on the particle surface.

6.2.2.2 Zetasizer Analysis The zetasizer analysis of chitosan and conjugate nanoparticles is presented in the

Figs. 6-2 A-F and the corresponding numerical data are summarized in the Table A6-1

(Apppendix-6).

The zetasizer analysis of both the blank chitosan and conjugate nanoparticles made by the

method-1 showed a uni-modal size distribution with Z-averages of 67.25±2.82 r.nm and

76.61±1.23 r.nm, respectively; corresponding polydispersity indices were 0.58±0.26 and

0.35±0.08, respectively (Table A6-1 in Appendix-6, Figs. 6-2 A and B). On the other hand,

both the blank and drug-loaded nanoparticles made of chitosan and the conjugate by the

method-2 showed multi-modal size distribution (Figs. 6-2 C-F). Three of these formulations,

viz. blank and drug-loaded chitosan nanoparticles and blank conjugate nanoparticles

showed bi-modal distributions, comprising a population of particles at 10-16 r.nm size range

and another between 194-263 r.nm (Table A6-1 in Appendix-6, Figs. 6-2 C-E). The overall

Z-averages for the three formulations were 70.34±3.99 r.nm, 32.23±1.29 r.nm and

21.17±0.52 r.nm, respectively (Table A6-1 in Appendix-6). The drug-loaded nanoparticles of

conjugate showed a tri-modal distribution with mean sizes of 0.49±0.01 r.nm, 15.02±0.93

r.nm and 248.57±10.67 r.nm, the overall Z-average being 9.26±0.99 r.nm (Fig. 6-2 F). All the

four samples showed the highest polydispersity index of 1.00 (Table A6-1 in Appendix-6).

Page 143: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

98

A B

C D

E F

Figure 6- 1: Scanning Electron Micrographs of A) Blank Chitosan Nanoparticles (Method-1), B) Blank Conjugate Nanoparticles (Method-1), C) Blank Chitosan Nanoparticles (Method-2), D) Blank Conjugate Nanoparticles (Method-2), E) Drug-loaded Chitosan Nanoparticles (Method-2) and F) Drug-loaded Conjugate Nanoparticles (Method-2) at X100,000 magnification

[Note: Dimension bar is shown under each micrograph. The cracks in the images are artefacts of processing for microscopy.]

Size:10-30 nm Size:10-30 nm

Size:10-30 nm Size:10-30 nm

Size:10-20 nm Size:10-20 nm

Page 144: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

99

A B

C D

E F

Figure 6- 2: Particle Size Distribution of A) Blank Chitosan Nanoparticles (Method-1), B) Blank Conjugate Nanoparticles (Method-1), C) Blank Chitosan Nanoparticles (Method-2), D) Blank Conjugate Nanoparticles (Method-2), E) Drug-loaded Chitosan Nanoparticles (Method-2) and F) Drug-loaded Conjugate Nanoparticles (Method-2)

[Note: Particle size distribution was determined in heavy paraffin oil suspension by dynamic light scattering with Malvern Zetasizer Nano-S. The sizes are presented as r.nm (hydrodynamic radius in nm).]

0

5

10

15

20

25

30

35

40

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

0

5

10

15

20

25

30

35

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

0

5

10

15

20

25

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

0

2

4

6

8

10

12

14

16

18

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

0

2

4

6

8

10

12

14

16

18

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

0

2

4

6

8

10

12

0.1 1 10 100 1000 10000

Inte

nsity

(%)

Size (r.nm)

49 r.nm (n=3) 70 r.nm (n=3)

16 r.nm (n=3)

194 r.nm (n=3)

10 r.nm (n=3)

255 r.nm (n=3)

16 r.nm (n=3)

263 r.nm (n=3)

0.5 r.nm (n=3)

15 r.nm (n=3)

249 r.nm (n=3)

Page 145: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

100

6.2.3 Production Yield, Drug Loading and Entrapment Efficiency The yield of both the blank and drug-loaded nanoparticles of chitosan and the conjugate

prepared by either method and the drug loading and entrapment efficiency of drug-loaded

nanoparticles prepared by the method-2 were determined using the equations (1), (2) and

(3), respectively as described in the Section 3.2.2.4 of the Chapter 3. The results are

summarized in the Table 6-1 and discussed in the Section 6.3.3.

Table 6- 1: Production Yield, Drug Loading and Entrapment Efficiency of Chitosan and Conjugate Nanoparticles (mean±SE, n=3)

Method of Preparation

Formulation Production Yield (%)

Drug loading (%)

Entrapment efficiency (%)

Method-1 Chitosan nanoparticles (blank) 69±2 - -

Conjugate nanoparticles (blank) 57±2 - -

Method-2

Chitosan nanoparticles (blank) 191±5 - -

Conjugate nanoparticles (blank) 169±4 - -

Chitosan nanoparticles (drug-loaded) 125±6 16±1 38±1

Conjugate nanoparticles (drug-loaded) 119±5 20±1 46±1

6.2.4 In Vitro Drug Release Study The release of the drug, DH from glutaraldehyde cross-linked chitosan and

chitosan-L-leucine conjugate nanoparticles was studied in phosphate buffered saline (pH

7.3±0.2) at 37 °C as described in the Section 3.2.2.5 of the Chapter 3 and the data are

presented in the Table A6-2 (Appendix 6) as cumulative % of drug released and retained at

different time points. The release data are plotted according to various kinetic models [zero

order, first order, Higuchi (Higuchi, 1963) and Hixson-Crowell (Hixson & Crowell, 1931)] in

Figs. 6-3.1 to 6-3.4. For gaining an insight into the mechanism of drug release, the data were

also analysed by Korsmeyer-Peppas model (Korsmeyer et al., 1983) (Fig. 6-3.5). A

summarized account of the applied kinetic models is given in the Table 6-2.

As shown in the Table A6-2 (Appendix 6) and the Fig. 6-3.1, both chitosan and conjugate

nanoparticles exhibited a large initial burst followed by a controlled release at a slow rate

over a period of approximately 1-2 weeks . Chitosan nanoparticles showed an initial burst

Page 146: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

101

release of 16±0.32% and a maximum cumulative % release of 23±0.31% at the end of 192

hours (8 days). The corresponding values for conjugate nanoparticles were 31±1.98% and

52±1.55%, respectively. The release from conjugate nanoparticles continued for a longer

period and reached the maximum at the end of 384 hours (16 days). Although the

experiment was continued for a period of 30 days, no further increase in the amount of drug

released was observed after 8 and 16 days periods from chitosan and conjugate

nanoparticles, respectively.

Table 6- 2: Mathematical Models Applied to the Release Data of Diltiazem Hydrochloride Loaded into Chitosan and Conjugate Nanoparticles

Mathematical models Equation Zero order model F=kot First order model ln (1-F)=-k1t Higuchi model F=KHt1/2 Hixson-Crowell model 1-(1-F)1/3=k1/3t Korsmeyer-Peppas model F=kK-Ptn

Note: F is cumulative fraction of drug released at time t. k0, k1, kH, k1/3 and kK-P are the apparent release rate

constants of the respective mathematical models. n is the release exponent of the Korsmeyer-Peppas model,

indicative of the mechanism of drug release.

To determine the goodness of fit of various models, the release data were analysed by

simple linear regression (using the data points until the time at which cumulative drug

release reached its maximum — i.e. 8 and 16 days for chitosan and conjugate nanoparticles

respectively). The determination coefficients (r2) and release rate constants determined by

fitting the release data to various kinetic models and regression analysis are presented in

the Table 6-3. For Korsmeyer-Peppas model, the table records the values of release

exponent, n.

Page 147: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

102

Table 6- 3: Release Rate Constants and Determination Coefficients for Drug Release Profile according to Various Kinetic Models

Models Zero order First order Higuchi Hixson-Crowell Korsmeyer-Peppas

Formulations k0

(×10-2

h-1)

r2 k1

(×10-2

h-1)

r2 kH

(×10-2

h-1)

r2 K1/3

(×10-2

h-1)

r2 n r2

Chitosan

nanoparticle

0.0313 0.8868 0.0005 0.8921 0.4709 0.9808 0.0006 0.8903 0.0568 0.9611

Conjugate

nanoparticle

0.0513 0.8334 0.0009 0.8593 1.1159 0.9661 0.0012 0.8509 0.0856 0.9709

Note: k0, k1, kH, k1/3 and kK-P are the apparent release rate constants of the respective mathematical models. n

is the release exponent of the Korsmeyer-Peppas model, indicative of the mechanism of drug release.

Figure 6-3. 1: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Zero Order Model)

[Note: Samples were analysed by UV spectrophotometry at intervals of 0.5, 1, 2, 4, 6, 12 and 24h time-points and later every 24 h upto 30 days. Drug loading: chitosan nanoparticles, 16% and conjugate nanoparticles, 20%. Each point in the graphs represents mean±SE (n=3). The linear regression was performed for the data points until the time at which the cumulative drug release reached its maximum, i.e. 8 and 16 days for chitosan and conjugate nanoparticles, respectively.]

Page 148: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

103

Figure 6-3. 2: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (1st Order Model)

[Note: Samples were analysed by UV spectrophotometry at intervals of 0.5, 1, 2, 4, 6, 12 and 24 h time-points and later every 24 h upto 30 days. Drug loading: chitosan nanoparticles, 16% and conjugate nanoparticles, 20%. Each point in the graphs represents mean±SE (n=3). The linear regression was performed for the data points until the time at which the cumulative drug release reached its maximum, i.e. 8 and 16 days for chitosan and conjugate nanoparticles, respectively.]

Figure 6-3. 3: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Higuchi Model)

[Note: Samples were analysed by UV spectrophotometry at intervals of 0.5, 1, 2, 4, 6, 12 and 24 h time-points and later every 24 h upto 30 days. Drug loading: chitosan nanoparticles, 16% and conjugate nanoparticles, 20%. Each point in the graphs represents mean±SE (n=3). The linear regression was performed for the data points until the time at which the cumulative drug release reached its maximum, i.e. 8 and 16 days for chitosan and conjugate nanoparticles respectively.]

Page 149: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

104

Figure 6-3. 4: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Hixson-Crowell Model)

[Note: Samples were analysed by UV spectrophotometry at intervals of 0.5, 1, 2, 4, 6, 12 and 24h time-points and later every 24h upto 30 days. Drug loading: chitosan nanoparticles, 16% and conjugate nanoparticles, 20%. Each point in the graphs represents mean±SE (n=3). The linear regression was performed for the data points until the time at which the cumulative drug release reached its maximum, i.e. 8 and 16 days for chitosan and conjugate nanoparticles respectively.]

Figure 6-3. 5: Release Profile of Diltiazem HCl from Chitosan ( ) and Chitosan-L-Leucine Conjugate ( ) Nanoparticles in PBS (pH 7.3±0.2, 37 °C) (Peppas-Korsmeyer Model)

[Note: Samples were analysed by UV spectrophotometry at intervals of 0.5, 1, 2, 4, 6, 12 and 24 h time-points and later every 24 h upto 30 days. Drug loading: chitosan nanoparticles, 16% and conjugate nanoparticles, 20%. Each point in the graphs represents mean±SE (n=3). The linear regression was performed for the data points until the time at which the cumulative drug release reached its maximum, i.e. 8 and 16 days for chitosan and conjugate nanoparticles respectively.]

Page 150: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

105

6.2.5 In Vitro Aerosolization Study The dispersibility of both the blank and drug-loaded chitosan and conjugate nanoparticles

prepared by emulsification glutaraldehyde cross-linking was studied using a twin-stage

impinger (TSI). The particles deposited in different stages were collected using PBS as the

solvent and analysed gravimetrically using 0.2 µm membrane filters for collecting particles.

For further confirmation, drug loaded particles were also analysed spectrophotometrically

by determining the amount of drug released from the particles. PBS was used as the

collection solvent in the stages I and II of the TSI to maintain a consistency with the

spectrophotometric analyses made during drug release study and determination of

entrapment efficiency. Although 0.2 µm is a much higher pore size compared to the size of

the prepared nanoparticles, repeated circulation of the liquid containing the particles

through the filter enabled complete removal of particles, because of the tendency of

particles to form agglomerates and gradually block the pores. The filtrate was finally

checked both visually and under microscope to ensure freedom from any remaining

particles.

Table A6-3 (Appendix 6) summarises the numerical data representing relative particle

deposition in different stages of the TSI apparatus, recovered dose (RD) and emitted dose

(ED) and fine particle fraction (FPF) expressed in percentage. Fine particle fraction (FPF)

actually represents the stage II deposition as a fraction of the total recovered dose (RD).

Fig. 6-4.1 illustrates the relative deposition of particles in different stages of the TSI as

estimated by gravimetric analysis. The recovered dose (RD), emitted dose (ED) and fine

particle fraction (FPF) are presented in Figs. 6-4.2, 6-4.3 and 6-4.4, respectively.

As per gravimetric analysis, the total doses of the formulations recovered from all the stages

of TSI ranged between of 74 and 87% of the loaded doses (Fig. 6-4.2, Table A6-3 in

Appendix 6). The fraction of the total dose emitted in the upper and lower stages was in the

range of 73-85%, with the exception of the blank chitosan nanoparticle that had a relatively

low emitted dose of 62% only (Fig. 6-4.3, Table A6-3 in Appendix 6). Statistical analyses

showed that there was no difference between emitted doses of drug-loaded chitosan and

conjugate nanoparticles (73±1.96% vs. 79±2.52%, p>0.05), but the emitted dose of blank

Page 151: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

106

conjugate nanoparticles was significantly higher than that of blank chitosan nanoparticles

(85±0.94% vs. 62±0.39%, p<0.05). The fine particle fractions (FPF) of the formulations were

between 15 and 24% (Fig. 6-4.4, Table A6-3 in Appendix 6). The FPFs of both blank and

drug-loaded conjugate nanoparticles were significantly higher than those of corresponding

chitosan nanoparticles (24±0.8% and 21±0.7% vs. 19±1.01% and 15±1.5%, respectively;

p<0.05). The drug-loaded nanoparticles of both chitosan and the conjugate had lower FPFs

than corresponding blank ones (15±1.5% and 21±0.7% vs. 19±1.01% and 24±0.8%,

respectively), but analysis of variance showed the difference to be insignificant (p>0.05).

Figure 6-4. 1: Particle Deposition in Different Stages of TSI (estimated by gravimetric analysis)

[Note: Particle deposition in different stages was collected in PBS, flitered using 0.2 μm filters and weighed after drying. The net weight of particles was determined by subtracting the weight of blank filters. Data represent mean±SE, n=3.]

Figure 6-4. 2: Recovered Doses (RD) from Different Formulations (estimated by gravimetric analysis of TSI depositions)

[Note: Data represent mean ± SE, n=3.]

38

15

27 22

43

61 58 57

19 24

15 21

0

10

20

30

40

50

60

70

BlankChitosan

Nanoparticles

BlankConjugate

Nanoparticles

Drug-loadedChitosan

Nanoparticles

Drug-loadedConjugate

Nanoparticles

%Pa

rtic

le D

epos

ition

Rotahaler (R)Stage-1 (S1)Stage-2 (S2)

84 86 81 74

0

20

40

60

80

100

Blank ChitosanNanoparticle

Blank ConjugateNanoparticle

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticles

Reco

vere

d Do

se (%

)

Page 152: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

107

Figure 6-4. 3: Emitted Doses (ED) from Different Formulations (estimated by gravimetric analysis of TSI depositions)

[Note: Data represent mean ± SE, n=3. Significance level, p < 0.05.]

Figure 6-4. 4: Fine Particle Fraction (FPF) of Different Formulations (estimated by gravimetric analysis of TSI depositions)

[Note: Data represent mean ± SE, n=3. Significance level, p < 0.05.]

62

85

73 79

0

10

20

30

40

50

60

70

80

90

100

Blank ChitosanNanoparticle

Blank ConjugateNanoparticle

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticles

Emitt

ed D

ose

(%)

19

24

15

21

0

5

10

15

20

25

30

Blank ChitosanNanoparticle

Blank ConjugateNanoparticle

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticles

Fine

Par

ticle

Fra

ctio

n (%

) p > 0.05 p < 0.05

p < 0.05 p < 0.05

Page 153: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

108

Figs. 6-5.1 to 6-5.4 present the results of drug deposition from drug-loaded chitosan and

conjugate nanoparticles in different stages of the TSI according to spectrophotometric

analysis.

Figure 6-5. 1: Drug Deposition in Different Stages of TSI (estimated by UV spectrophotometric analysis)

[Note: Particles deposited in different stages of TSI were collected in PBS, incubated at 37 °C for 2 weeks (for chitosan) / 3 weeks (for conjugate), after which the release medium was analysed spectrophotometrically to determine the amount of DH. Data represent mean ± SE, n=3.]

Figure 6-5. 2: Recovered Doses (RD) from Different Formulations (determined by UV spectrophotometric analysis of TSI depositions)

[Note: Data represent mean ± SE, n=3.]

28 21

56 58

16 21

0

10

20

30

40

50

60

70

Drug-loaded ChitosanNanoparticles

Drug-loaded ConjugateNanoparticles

% D

rug

Depo

sitio

n

Rotahaler (R)

Stage-1 (S1)

Stage-2 (S2)

81 87

0

10

20

30

40

50

60

70

80

90

100

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticles

Reco

vere

d Do

se (%

)

Page 154: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

109

Figure 6-5. 3: Emitted Doses (RD) from Different Formulations (estimated by UV spectrophotometric analysis of TSI depositions)

[Note: Data represent mean±SE, n=3.]

Figure 6-5. 4: Fine Particle Fraction (FPF) of Different Formulations (estimated by UV spectrophotometric analysis of TSI depositions)

[Note: Data represent mean±SE, n=3.]

72 79

0

10

20

30

40

50

60

70

80

90

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticle

Emitt

ed D

ose

(%)

16

21

0

5

10

15

20

25

Drug-loaded ChitosanNanoparticle

Drug-loaded ConjugateNanoparticles

Fine

Par

ticle

Fra

ctio

n (%

)

Page 155: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

110

Figs. 6-6.1 and 6-6.2 (A-C) present the results of particle deposition from interactive

mixtures of drug-loaded chitosan and conjugate nanoparticles with lactose monohydrate in

different stages of the TSI according to gravimetric analysis.

Figure 6-6. 1: Particle Deposition in Different Stages of TSI from Inteactive Mixtures of Drug-loaded Chitosan and Conjugate Nanoparticles with Lactose Monohydrate (estimated by gravimetric analysis)

[Note: Particle deposition in different stages was collected in PBS, flitered using 0.2 μm filters and weighed after drying. The net weight of particles was determined by subtracting the weight of blank filters. Data represent mean±SE, n=3.]

Figure 6-6. 2: (A) Recovered Dose (RD), (B) Emitted Dose (ED) and (C) Fine Particle Fraction (FPF) of Inteactive Mixtures of Drug-loaded Chitosan and Conjugate Nanoparticles with Lactose Monohydrate (estimated by gravimetric analysis)

[Note: Data represent mean ± SE, n=3.]

21

16

50 47

28

37

0

10

20

30

40

50

60

Chitosan NP-LactoseInteractive Mixture

Conjugate NP-LactoseInteractive Mixture

% P

artic

le D

epos

ition

Rotahaler (R)Stage-1 (S1)Stage-2 (S2)

A B C

p<0.05 p>0.05 p>0.05

Page 156: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

111

6.3 DISCUSSION

6.3.1 Preparation of Chitosan and Chitosan-L-Leucine Conjugate Nanoparticles This work was aimed at preparing nanoparticulate DPI formulation loaded with the model

drug, DH. Of various techniques reported in the literature, emulsification was chosen for this

work. The underlying reason was that the technique is simple and inexpensive and has been

used both at the bench-top and industrial scale for fabrication of polymeric particles

(Berkland et al., 2001). The method allows getting particles in desired size range by

adjusting the time and/or speed of stirring during emulsification (Dubey & Parikh, 2004).

Besides, it also allows fabrication of particles with a more optimized release of the

encapsulated drug by ensuring a uniform dispersion of the drug throughout the polymeric

matrix (Obeidat, 2009).

Considering the concerns raised by some investigators on possible harmful effect (e.g.

irritation of mucous membranes) of any residual glutaraldehyde in particles (Lim et al.,

1997; Richards & Knowles, 1968), this work was primarily focussed on fabrication of

nanoparticles by the solvent evaporation approach (method-1). To generate discrete

particles in the desired size range, three major changes were made in the previously

reported methods (Abd El-Hameed & Kellaway, 1997; Lim et al., 2000; Onishi et al., 2005) by

progressive trial and error. These included: 1) use of heavy mineral oil instead of light

mineral oil or light-heavy mixture, 2) increasing the speed of the overhead stirrer to 5000

rpm and 3) reducing the total solid content to a maximum of 50 mg/100g of oil. These

changes prevented coalescence and agglomeration of particles during the hardening phase.

Although the method was successful to generate discrete particles in the desired size range,

the attempts to load the model drug, DH failed. Microscopic examination of the emulsion

samples at intervals during the hardening phase showed precipitation of the drug as needle-

shaped crystals at a certain point of time (often within 2-6 h after starting the stirring

process). The problem was not encountered when glutaraldehyde cross-linking was applied

instead for hardening of the particles. It seems that the drug crystallized under heat and

precipitated out before solidification of the polymer. Another possibility is failure of the

particles to retain the drug crystals formed because of their small size. It would be relevant

to note here that previous reports on this technique described preparation of particles in

Page 157: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

112

micrometer range and this attempt was, in fact, the first of its kind to prepare nanoparticles

in this way. Although the method failed to incorporate DH — the model drug chosen for this

work, it might prove useful for incorporating other drugs with different physicochemical

properties. Besides, attempts could also be made to load the drug into pre-formed particles

by passive absorption from aqueous solution as has been reported previously by some

investigators for loading various drugs in chitosan particles (Gupta & Jabrail, 2006, 2007a,

2007b; Jameela et al., 1994b; Ubaidulla et al., 2007). When attempt was made to fabricate

drug-loaded conjugate particles by this method, the particles formed aggregates and turned

into a lump during isolation process. This difference could be due to higher hydrophilicity of

the conjugate that was further augmented upon incorporation of DH — a highly water

soluble drug. An increased agglomeration of chitosan particles has been reported previously

upon incorporation of the hydrophilic drug, propranolol hydrochloride (Patel et al., 2007).

The glutaraldehyde cross-linking method was found to be superior, especially in the sense

that it did not encounter problem with drug loading as found with solvent evaporation

approach. Although there are concerns about possible toxic effect of residual

glutaraldehyde upon administration of the particles in vivo, some investigators have shown

absence of any untoward effect on living tissues with particles made by this technique

(Jameela & Jayakrishnan, 1995; Jameela et al., 1994a). In addition, this technique has an

added advantage of avoiding the use of heat which could be harmful, if sufficiently high, for

a drug like DH.

6.3.2 Particle Size and Morphology Scanning electron microscopy (SEM) was used in this study to visualize the size, shape, and

surface morphology of the nanoparticles. The size and size distribution were further

analysed by dynamic light scattering (DLS) using a Zetasizer. It may be noted here that SEM

has widely been used for size analysis in conjunction with laser diffraction/dynamic light

scattering. Some investigators depended solely on SEM for this purpose (Gupta & Jabrail,

2006; Janoria & Mitra, 2007; Nie et al., 2008; Onishi et al., 2005).

Page 158: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

113

The blank particles, prepared by either method, were found to be spherical-to-oblate in

shape (Figs. 6-1 A-D), suggesting that these particles were weak in mechanical strength and

had a tendency of deformation under pressure. In contrast, drug-loaded particles were

found to be nearly perfect spheres (Figs. 6-1 E-F). This implies that the drug-polymer

mixture increased the compactness and rigidity of the particles compared to the polymers

alone.

With few exceptions, the surface of the particles was smooth with no cracks or pores (Figs.

6-1 A-F). This apparently suggests that the internal structure of the particles was compact

and nonporous. There was no sign of the presence of any drug crystal on the particle

surfaces. This suggests that the drug and polymer have uniformly been mixed together in

the process of particle formation.

As shown in the Figs. 6-1 A-F, the size of the nanoparticles was below 100 nm implying that

they were well-suited for pulmonary delivery, because it has been reported that particles of

this size range are primarily deposited in the alveolar region of the lungs upon inhalation

(Hoet et al., 2004; Oberdorster et al., 2005). The size was only slightly affected by the

nature of the polymeric matrix (i.e. chitosan/ conjugate) or the method of preparation (i.e.

solvent evaporation / glutaraldehyde cross-linking). However, drug loading brought a

reduction in the size of both chitosan and conjugate nanoparticles (Figs. 6-1 E & F). This

effect could be attributed to reduced viscosity of the solution due to replacement of half of

the polymeric material with drug that made dispersion of the aqueous phase into oil easier

giving rise to smaller sizes of droplets, ultimately giving smaller particles. This is supported

by the findings of Denkbas & Odabasi (2000) who demonstrated a decrease in particle size

with decrease in chitosan concentration in the aqueous phase. One negative aspect of all

the formulations was that the particles showed a tendency to form agglomerates (Figs. 6-1

A-F). However, this is a common phenomenon with nanoparticles, which results from high

surface area and associated surface energy of particles in this size range.

Page 159: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

114

Zetasizer analysis of blank chitosan and conjugate nanoparticles made by the method-1

gave a unimodal particle size distribution with a Z-average of ~70 r.nm, which was much

higher compared to the diameter of the particles observed by SEM (10-30 nm) (Table A6-1

in Appendix 6, Figs. 6-1 A and B, 6-2 A and B). However, consistent with SEM micrographs,

the Z-average of conjugate nanoparticles appeared to be larger than that of chitosan

nanoparticles (76.61±1.23 r.nm vs. 67.25±2.82 r.nm) (Table A6-1). On the other hand, all the

formulations made by method-2 showed multimodal distribution with a population of

particles having Z-average in the range of 9-16 r.nm and another population with

Z-averages between 194 and 263 r.nm. The drug-loaded nanoparticles of the conjugate

showed an additional population with Z-average of ~0.5 r.nm (Table A6-1 in Appendix 6,

Figs. 6-2 C-F). SEM micrographs of these formulations clearly indicate that the diameter of

the blank chitosan and conjugate nanoparticles made by this method predominantly fall in

the range of 10-30 nm (Figs. 6-1 C-D), while the drug-loaded particles had a close, but

relatively narrower, diameter range of 10-20 nm (Figs. 6-1 E-F). This suggests that the larger

size populations observed during particle sizing reflects aggregates arising from cohesion of

small individual particles. Similar observations have been reported by a number of

investigators in relation to various polymeric and non-polymeric microparticles (Li & Birchall,

2006; Li et al., 2005; Rabbani & Seville, 2005; Steckel et al., 2003). The high polydispersity

index (1.00) of the samples also gives an indication of a high agglomeration tendency of the

particles. This phenomenon implies potential poor performance of the particles during

aerosolization, which is explained later in the Section 6.3.5. However, as long as the

agglomerates had their sizes in the order of 200-250 r.nm, the particles are expected be

good enough to reach deeper regions of the respiratory system. It remains a question here

why particles prepared by method-1 do not show a peak at 10-30 nm diameter despite the

fact that they have lower polydispersity indices in the range of 0.3-0.5; however, the

agglomerate sizes obtained for them (49 and 70 r.nm for chitosan and conjugate

nanoparticles, respectively) are much smaller than those of the nanoparticles prepared by

the method-2 (200-250 r.nm) which is consistent with their smaller polydispersity indices

(Table A6-1 in Appendix 6).

Page 160: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

115

6.3.3 Production Yield, Drug Loading and Entrapment Efficiency The solvent evaporation procedure produced a yield of 69±2% and 57±2% for chitosan and

conjugate nanoparticles, respectively (Table 6-1). This apparently low yield can be

attributed to the small scale of production that could have resulted in relatively large losses

of materials through adhesion to the vessel walls and propeller shaft and blades and

subsequent centrifugation performed for isolation and purification. On the contrary, the

glutaraldehyde cross-linking technique apparently produced more than 100% yield

(chitosan, 191±5% and conjugate, 169±4%) (Table 6-1); this could be attributed to an

increase in the bulk of the starting polymers due to incorporation of glutaraldehyde

moieties between polymer chains. Again, although still more than 100%, drug-loaded

nanoparticles had relatively lower yield (chitosan, 125±6% and conjugate, 119±5%) (Table 6-

1). This could reasonably be attributed to replacement of 50% polymer with the drug, DH

that did not interact with the cross-linker, glutaraldehyde in the way that the polymers did.

It would be pertinent to note here that, in addition to incorporation of crooslinked

glutaraldehyde moieties between the polymer chains, residual moisture left in the particles

might also have contributed to the increase in the bulk of polymeric materials resulting in

more than 100% percent yield compared to the starting material.

The entrapment efficiency is the fraction of the total drug input that has been encapsulated

into the particles. During the process of forming drug-loaded nanoparticles, the drug is often

lost to some extent, and thus a 100% entrapment efficiency of any drug is unlikely. The

entrapment efficiency of 38±1% and 46±1% as achieved here for chitosan and conjugate

nanoparticles (Table 6-1) means that about 60% of the drug has practically been lost. Many

factors could be responsible for this relatively low incorporation efficiency. Firstly, some

drug is obviously partitioned out to the external oil phase. Though the solubility of the drug,

DH in oil was negligible, the oil phase was still able to accommodate a good deal of the drug

because of its large volume (79 times higher than the drug-containing water phase).

Secondly, during the cross-linking and hardening process, water is exuded from the particles

taking along the dissolved drug; this could also contribute to decrease in the incorporation

efficiency (Jameela & Jayakrishnan, 1995). Finally, small size of the particles might also have

contributed to the low entrapment. It has been reported that efficiency of particles to

entrap drugs falls with decrease in particle size (Hadinoto et al., 2007).

Page 161: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

116

The drug loading (%) refers to the amount of drug per 100 g of particle. Drug loading

obtained for chitosan and conjugate nanoparticles was 16±1% and 20±1%, respectively

(Table 6-1). As explained above, this apparently low % loading was due to the loss of some

drug to the external oil phase and, possibly, also for anticipated poor entrapment of drug

because of small particle size. However, these figures could be higher if the particle yield did

not exceed 100% (chitosan, 125±6% and conjugate, 119±5%) (Table 6-1) owing possibly to

glutaraldehyde cross-linking.

The higher entrapment efficiency and drug loading of conjugate nanoparticles compared to

chitosan nanoparticles (46±1% and 20±1% vs. 38±1% and 16±1%, respectively) (Table 6-1)

could be attributed to the higher solubility of the conjugate in water that enabled its better

association with the water soluble drug, DH. This is because the entrapment of drug is

dependent on the successful molecular-level association of the drug with the polymer

(Kockisch et al., 2005).

6.3.4 In Vitro Drug Release Study Ideally, a controlled release formulation should initially provide quick release of an amount

of drug that would allow to promptly achieve an effective plasma concentration followed by

a slow, continued release of an amount that would be adequate to replace the amount of

drug eliminated from the body and maintain a constant blood level of the drug over time

within the therapeutic window. As shown in the results section (Section 6.2.4), although the

rate and extent of drug release varied, both chitosan and conjugate nanoparticles showed a

bi-phasic drug release with a large initial burst followed by a slow, controlled release over a

period of 8 and 16 days, respectively (Fig. 6-3.1, Table A6-2 in Appendix 6). The initial burst

accounted for 16±0.32%, and 31±1.98% of drug release from chitosan and conjugate

nanoparticles, respectively within the first 30 min, leaving 84±0.55% and 69±3.42% back in

the particles for slow, steady release. This kind of bi-phasic release might prove useful to

satisfy the pattern of release required of a controlled release formulation to quickly achieve

a therapeutic blood level (burst release) and maintain it over time (controlled release).

Previous investigators also observed similar type of release for various drugs (e.g.

5-fluorouracil, methotrexate, centchroman, mitoxantrone, and bovine serum albumin (BSA))

Page 162: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

117

from chitosan particles made by glutaraldehyde cross-linking (Dubey & Parikh, 2004; Gupta

& Jabrail, 2007b; Jameela & Jayakrishnan, 1995; Jameela et al., 1994a) as well as for some

drugs (e.g. for clozapine, BSA) from particles made by other techniques (Agnihotri &

Aminabhavi, 2004; Gan & Wang, 2007). The phenomenon has also been reported for drug

release from other polymeric particles (such as PLGA and PLA) (Erden & Celebi, 1996;

Ibrahim et al., 2005). The large initial burst release is attributed to the drug located at or

near the surface (Agnihotri & Aminabhavi, 2004). It could be reasoned that the hydrophilic

nature of chitosan and high aqueous solubility of DH allowed the hydration of the polymer

surface and subsequent dissolution and release of the drug to occur in an exceedingly quick

succession. The subsequent sustained release is attributed to the swelling of the polymer to

form a gel layer that offers a hindrance to the outward diffusion of the drug molecule from

the core and thereby slows down the drug release (Denkbas et al., 1999; Dhawan & Singla,

2003; Learoyd et al., 2008a; Lim et al., 2000).

The release of drug both from chitosan and conjugate nanoparticles was not complete even

after running the experiment for one month. The maximum cumulative release obtained

was only 23% for chitosan nanoparticles and 52% for conjugate ones leaving back 77% and

48% drug, respectively, still unreleased (Fig. 6-3.1, Table A6-2 in Appendix 6). This sort of

incomplete release has also been reported previously for chitosan particles (Agnihotri &

Aminabhavi, 2004; Lim et al., 2000; Torres et al., 1998 ). This is attributed to binding of a

fraction of incorporated drug to some components of the matrix though electrostatic or

covalent interaction (Jameela et al., 1994a; Lim et al., 2000). In their studies, Jameela et al.

(1994a) accounted interaction with the aldehyde moieties of the residual glutaraldehyde for

incomplete release (~50%) of BSA.

Both the initial burst and total release from conjugate nanoparticles were approximately

twice as much higher as from chitosan nanoparticles (31% and 52% vs. 16% and 23%)

(Fig. 6-3.1, Table A6-2 in Appendix 6)). This can be attributed to improved aqueous

solubility shown by the conjugate compared to chitosan. While chitosan needed an

overnight stirring with dilute acetic acid for dissolution, the conjugate dissolved immediately

upon adding to water. This higher solubility of conjugate, in one hand, allowed for

Page 163: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

118

increased drug loading (Table 6-1) into the particles as has already been explained in the

Section 6.3.3 and, on the other hand, conferred to them more wettability in water. Being a

water soluble drug, DH could be expected to easily dissolve in hydrated polymeric

environment. Therefore, the higher the drug loading, the more the drug dissolved in the

hydrated polymeric matrix. This would have resulted in a higher diffusional driving force and

in turn caused a faster drug release (31% for conjugate vs. 16% for chitosan within the first

30 min) (Fig. 6-3.1, Table A6-2 in Appendix 6). Similar reasoning has been made earlier by

Patel et al. (2007) for explaining the release of another hydrophilic drug, propranolol

hydrochloride from chitosan-based matrix. Additionally, a better wettability of conjugate

nanoparticles meant their quicker hydration and so faster dissolution and release of the

incorporated drug. The higher solubility shown by the conjugate could be related to the

surface changes induced by L-leucine conjugation. It has previously been hypothesized that,

because of its surfactant like property, L-leucine conjugated to chitosan would impart an

amphiphilic environment around the chitosan backbone (Section 1.2 and Fig. 1-1 in Chapter

1). This amphipathic environment could duly be expected to increase the solubility of the

polymer.

Besides exhibiting a higher rate and extent of drug release, the conjugate nanoparticles also

continued releasing drug for a period twice as long as was shown by chitosan nanoparticles

(16 days, 52% vs. 8 days, 23%) (Fig. 6-3.1, Table A6-2 in Appendix 6). Two factors could have

contributed to this. First, conjugate nanoparticles had a higher percentage of drug loading

(20±1% vs. 16±1%) (Table 6-1). Second, conjugate nanoparticles released twice as much

drug as chitosan nanoparticles (52% vs. 23%) (Fig. 6-3.1, Table A6-2 in Appendix 6). This

again apparently was due to the higher hydrophilicity of the conjugate that permitted easier

access of the release media and in turn created a more favourable environment for drug

dissolution and diffusion.

For elucidating the kinetic pattern of drug release from both the chitosan and conjugate

nanoparticles, the release data were fitted to 4 different kinetic models reported in the

literature, including zero-order, 1st order, square root of time or Higuchi model (Higuchi,

1963) and cube root or Hixson-Crowell model (Hixson & Crowell, 1931) (Figs. 6-3.1 to 6-3.4).

Page 164: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

119

Of these, the first three models are used for studying kinetics of diffusion-controlled release

from a matrix-type system, while Hixson-Crowell model is used to describe dissolution–type

release kinetics where the drug is released by dissolution with change in the surface area or

diameter of the particles (Asada et al., 2004; Filipovic-Grcic et al., 1996). Although literature

reports suggest that controlled release of drug from chitosan-based systems in PBS/alkaline

pH is governed by diffusion of the drug through hydrated swollen matrix (Agnihotri &

Aminabhavi, 2004; Learoyd et al., 2008a; Lim et al., 2000), Hixson-Crowell model was also

included in this study for kinetic analysis considering higher water solubility shown by the

conjugate. However, it would be relevant to note here that neither chitosan nor conjugate

nanoparticles have practically shown any visible sign of matrix dissolution. The values of r2

for zero order, first order, Higuchi and Hixson-Crowell models were 0.8868, 0.8921, 0.9808

and 0.8903, respectively for chitosan nanoparticles and 0.8334, 0.8593, 0.9661and 0.8509,

respectively for conjugate nanoparticles (Table 6-3). From the r2 values it can be inferred

that, after the initial burst, the release of the drug from both chitosan and conjugate

nanoparticles followed Higuchi’s square root of time kinetics. This is in agreement with

previous reports on drug release kinetics of chitosan micro- and nanoparticles (Dubey &

Parikh, 2004; Filipovic-Grcic et al., 1996; Jameela & Jayakrishnan, 1995; Learoyd et al.,

2008a; Lim et al., 2000; Peniche et al., 2003). The release rate constants (KH) determined by

fitting the data to the model were 0.4709 × 10-2 h-1 for chitosan nanoparticles and 1.1159 ×

10-2 h-1 for conjugate nanoparticles (Table 6-3). The small values of the rate constants

further confirm that, after the large initial burst, the drug release from the nanoparticles

proceeded at an extremely slow rate. Besides, their relative magnitude also corroborates

that the release rate of drug from conjugate nanoparticles was nearly twice as high as that

from chitosan nanoparticles.

To better characterise the drug release behaviour for the polymeric systems under study,

and particularly to gain some insight into the corresponding mechanism, the release data

were also applied to the semi-empirical Korsmeyer-Peppas model (Korsmeyer et al., 1983)

(Fig. 6-3.5). This model uses the value of the release exponent (n) obtained from the slope

of a plot of log % cumulative release versus log time in order to characterize different

release mechanisms. If n is 0.45 or less, the release mechanism is considered to follow

Page 165: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

120

Fickian diffusion or case I transport, where drug release occurs through usual molecular

diffusion because of a concentration (or chemical potential) gradient. Higher values of n

between 0.45 and 0.89 indicate non-Fickian or anomalous transport, where release is

controlled by a combination of diffusion and polymer relaxation/erosion. When n reaches a

value of 0.89 or above, the mechanism of drug release is regarded as case-II or super case-II

transport which mean that the drug release involves polymer relaxation and chain

disentanglement/erosion and the rate does not change over time (Cox et al., 1999; Harland

et al., 1988). As shown in the Table 6-3, application of drug release data to the model and

regressional analysis resulted in a reasonably good fit giving coefficient of determination (r2)

values of 0.9611 for chitosan nanoparticles and 0.9709 for conjugate nanoparticles. The

values for release exponent (n) were found to be 0.0568 and 0.0856, respectively. This

suggests that after the initial burst, the release of drug from both the formulations occurred

by Fickian diffusion through hydrated polymeric matrix. Similar findings on the mechanism

of drug release from chitosan micro-/ nanoparticles have been reported by other

investigators. For instance, in tracine-loaded chitosan nanoparticles prepared by a similar

method, Wilson et al. (2010) have shown that after an initial burst the particles offered a

continuous, slow release and the mechanism of drug release as per the Korsmeyer model

was Fickian. In another study, Sivadas et al. (2008) reported Fickian diffusion-based release

of BSA from chitosan microparticles prepared by spray drying. However, there are also

reports in the literature describing non-Fickian release of drugs from chitosan micro- or

nanoparticles (Dhawan & Singla, 2003; Patel et al., 2007; Sezer & Akbuga, 1995). On the

other hand, Gupta & Jabrail (2006) reported a case of centchroman-loaded chitosan

microspheres where the release of the drug initially was Fickian and became non-Fickian at

later stage, which the authors attributed to ensuing structural variation in microspheres

with progressive swelling and dissolution. It is, however, important to note here that these

apparent disagreements of these reports are not surprising if we consider different types of

preparation methods employed for making chitosan micro- and nanoparticles and variation

in the physicochemical properties of incorporated drugs, all of which are intimately related

to the mechanism of drug release from the particles.

Page 166: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

121

With 16 and 20% drug loading (Table 6-1) and 16 and 31% burst release (Fig. 6-3.1) for

chitosan and conjugate nanoparticles, respectively, the amounts of the drug released as the

burst effect per 100 mg of chitosan and conjugate nanoparticles stand out to be 2.56 and

6.20 mg, respectively. So, to provide an initial loading dose of 10.85 mg DH (as calculated in

section 3.2.2.5 of chapter 3), a lung deposition of 423.83 and 175.00 mg of drug-loaded

chitosan and conjugate nanoparticles would be required. However, with a release rate

constant (kH) of 0.4709 ×10-2 and 1.1159×10-2 per hour (as determined by Higuchi model) for

the controlled release period of chitosan and conjugate nanoparticles, the amount of drug

released from 423 and 175 mg doses of chitosan and conjugate nanoparticles would be 1.99

and 1.95 mg per hour, respectively. These values are close to the required maintenance

dose release rate (MDR) of 2.06 mg/h for DH (as calculated in Section 3.2.2.5 of Chapter 3).

So, it could be expected that, once the patient has received the required amount of lung

deposition of drug-loaded chitosan and conjugate nanoparticles (423.83 and 175.00 mg,

respectively), the concentration of the drug in blood would be maintained close to the

desired level (0.05 μg/mL) during the controlled release period (8 and 16 days for chitosan

and conjugate nanoparticles, respectively as shown in the Fig. 6-3.1). However, in

consideration of patient convenience, further studies are required to increase drug

entrapment/ loading and the rate and extent of drug release in order to reduce the dose

size. One approach to this end may be to replace part of the polymer by a bulking agent like

lactose or mannitol. According to previous literature, these agents can help increase the

drug release by increasing the hydrophilicity of the matrix (Abd El-Hameed & Kellaway,

1997; Jain et al., 2000; Li et al., 2009). In addition, they can probably also help improving

drug loading and entrapment in the same way. It would be relevant to note that the present

level of drug loading and release might prove satisfactory for potent drugs with smaller dose

sizes.

6.3.5 In Vitro Aerosolization Study The aerosolization performance of different formulations was characterised in terms of

three parameters, viz. recovered dose (RD), emitted dose (ED) and fine particle fraction

(FPF).

Page 167: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

122

The recovered dose (RD) was presented as the percentage of the actual dose actuated from

the capsule by Rotahaler® device. As per gravimetric analysis, the recovered doses from

different formulations were between 74% and 86% (Fig. 6-4.2, Table A6- 3 in Appendix 6),

indicating that about 15 to 25% of the actuated particles remained in the inhaler device and

TSI chambers. Sticking of some particles to the glass surfaces of the TSI chambers might

have primarily accounted for this loss. For drug-loaded particles, drug released from the

particles in the collecting solvent (PBS) might also have some contribution to the loss. Thus,

the relatively lower value of 74% for drug-loaded conjugate nanoparticles could be related

to their higher drug content and quicker drug release rate as discussed in the Section 6.3.4.

It would be interesting to note here that the addition of 16 and 31% of drug released in

burst in the first 30 min from chitosan and conjugate nanoparticles respectively (Fig. 6-3.1)

to the corresponding doses of 81 and 74% recovered from them in TSI study gives a total

dose of 97 and 105%, respectively which are within ± 5% of their total doses.

The emitted dose (ED) was the amount of drug released from the inhaler device, i.e. the

sum of drug collected at the upper and lower stages of the TSI modelling for the upper and

lower respiratory airways. The emission of particles from the inhaler device was reasonably

good for most formulations with the values ranging from 73 to 85% (Fig. 6-4.3, Table A6-3 in

Appendix 6). However, blank chitosan particles showed a poor performance and the

emission was found to be significantly lower than corresponding conjugate particles

(62±0.39% vs. 85±0.94%; p<0.05) (Fig. 6-4.3, Table A6-3 in Appendix 6). Analogous to the

difference observed with water solubility, this difference in emission could also be due to

leucinization induced surface changes (an amphiphilic environment around the chitosan

backbone) as previously hypothesized, which is expected to reduce the interparticle

interaction and increase the aerosolization of the particles (Section 1.2 and Fig. 1-1 in

Chapter 1). On the contrary, no difference was seen between chitosan and the conjugate in

the emission of drug-loaded nanoparticles (73±1.96% vs. 79±2.52%; p>0.05) (Fig. 6-4.3,

Table A6-3 in Appendix 6). SEM studies showed that drug loading reduced the particle size

of both chitosan and conjugate and they had very similar particle sizes (Fig 6-1 E-F). This

could be related to the similar emission shown by the two formulations.

Page 168: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

123

Although drug-loaded chitosan and conjugate nanoparticles showed equivalent emission

from the DPI device, both blank and drug-loaded conjugate nanoparticles showed a

significantly (p<0.05) higher FPF than corresponding chitosan nanoparticles (24±0.8% and

21±0.7% vs. 19±1.01% and 15±1.5%, respectively) (Fig. 6-4.4, Table A6- 3 in Appendix 6).

This is an important finding that further corroborates the hypothesis made as the original

background of this project (Section 1.2 and Fig. 1-1 in Chapter 1). The higher FPF values of

conjugate nanoparticles establish higher dispersibility of the compound. However, the

increase in FPF shown by the conjugate nanoparticles was much less than that reported for

microparticles prepared by physical addition of L-leucine to chitosan in a DPI formulation.

Learoyd et al. (2008a, 2008b, 2009) demonstrated 82% FPF for a hydrophilic drug,

terbutaline sulphate, 64% for a hydrophobic drug, beclomethasone diproprionate, and 76%

for a combination of the two drugs from spray-dried chitosan (LMW: <190 kDa)

microparticles containing L-leucine as an aerosolization enhancer. These formulations,

however, also contained lactose as a bulking agent that might also have a significant

contribution to the high degree of FPF observed.

The results also show that, albeit statistically not significant, blank nanoparticles of both

chitosan and conjugate, showed higher FPF than corresponding drug-loaded nanoparticles

(19±1.01% and 24±0.8% vs. 15±1.5% and 21±0.7%, respectively; p>0.05) (Fig. 6-4.4, Table

A6-3 in Appendix 6). The lower FPF of drug-loaded particles may be due to a higher

agglomeration tendency caused by incorporation of DH — a highly water-soluble drug. Lim

& Wan (1998) reported increased agglomeration of chitosan-based microspheres upon

addition of a hydrophilic drug, propranolol hydrochloride. Besides, as per SEM analysis,

drug-loaded nanoparticles had a smaller particle size than blank nanoparticles

(Figs. 6-1 A-F). The smaller particle size might also have contributed to their higher

agglomeration limiting their transit from stage I to stage II of the TSI.

Finally, in consideration of the 6.4 µm cut-off value for the lower impingement chamber, it

may look a bit disappointing for particles of the order of 10-30 nm size to have an FPF of

only 15-24%. But, as already explained in the literature review (Section 2.4 in Chapter 2), it

is related to the higher cohesiveness exerted by micronized particles that makes the most

Page 169: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

124

important challenge of a DPI system and made the background of this research as well. In

this view, it is rather encouraging that the nanoparticle formulations of neither chitosan nor

conjugate, prepared under this project, needed a carrier like lactose or mannitol to aid their

dispersion from the inhaler depositing a substantial fraction into the stage II of the TSI. For

free drugs of this size, it is unexpected to get such deposition in the lower stage of the TSI

without being blended with a carrier.

For further confirmation of the results of TSI depositions estimated by gravimetric method,

the depositions from different stages were additionally examined by UV spectrophotometric

estimation of drug released. The results obtained for drug deposition in different stages of

TSI were similar to those obtained by gravimetric analysis (Figs. 6-5.1 to 6-5.4). This

confirms the reliability of results obtained by gravimetric analysis.

To investigate the possible effect of an interactive carrier on the dispersion of the

nanoparticles, drug-loaded chitosan and conjugate nanoparticles were mixed (5%) with

inhalation grade lactose monohydrate microparticles. As shown in Fig. 6-6.2(C), mixing with

lactose carrier particles produced a significant (p<0.05) elevation in dispersion of both

chitosan and conjugate nanoparticles (FPF: 28±0.71% & 37±0.58%) in comparison with

carrier-free particles (FPF: 15±1.20% & 21±0.58%). This enhancement in nanoparticle

dispersion could be attributed to the probable reduction of the strong cohesive force among

the polymer nanoparticles and their de-agglomeration by the lactose monohydrate carrier

microparticles as has been suggested previously for DPI formulations containing micronized

drug particles with lactose as a carrier (Bisgaard et al., 2002; Hersey, 1975). Ideally, mixing

of small quantities of micronized drug particles with larger carrier particles form an ordered

blend, where a perfect mix consists in a regular coating of fine particles on the coarser

constituent (Berard et al., 2002). The various forces contributing to this interaction include

van der walls forces, capillary forces, electrostatic forces and mechanical interlocking (Desai

et al., 2013). During inhalation, the particles are detached from the carrier surface by the

inspiratory force (Pilcer & Amighi, 2010). However, the extent of detachment is affected by

the strength of the drug-carrier adhesive forces. A strong adhesion resists drug detachment

and dispersion in the inhalation airstream and in turn reduces the overall lung deposition of

Page 170: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

125

drug particles (Desai et al., 2013). Thus, the enhancement of nanoparticle dispersion in this

study suggests formation of stable, but sufficiently weak bonds with the carrier

microparticles to allow their release upon application of dispersive force (vacuum air-flow).

Further studies using different nanoparticle- carrier ratios (e.g. 2.5 or 10%) and by changing

carrier type, such as using different grades of lactose (e.g. anhydrous lactose, medium

lactose, regular lactose, lactose crystals, foremost lactose) or other carriers (e.g., mannitol,

sorbitol) could be tried to find best possible nanoparticle-microcarrier combination. Finally,

once again, the conjugate nanoparticles showed significantly higher dispersibility compared

with chitosan nanoparticles (28±0.71% vs. 37±0.58%, p<0.05). This further supports

enhancement in dispersibility of chitosan upon leucinization.

6.4 CONCLUSION Nanoparticles of chitosan and its conjugate with L-leucine were prepared and the model

drug, DH was successfully loaded into them by emulsification- glutaraldehyde cross-linking

method. The particles were less than 100 nm in size suggesting their suitability for

deposition to deeper regions of lungs. Aerosolization studies with TSI showed that

formulations were able to produce 15-25% FPF without addition of any carrier like lactose or

mannitol. Nanoparticles made of conjugate were found to be more dispersible than those of

chitosan, generating a higher FPF. However, if we consider the current challenges of DPI in

terms of drug deposition in the lower airways, an FPF of 15-24% is not sufficiently high and

so more studies are needed in that direction. One attempt in this case was mixing the

polymer nanoparticles with lactose monohydrate microparticles that demonstrated an

encouraging enhancement in dispersibility of particles. Drug release studies showed a

higher drug loading as well as higher percentage of drug release for an extended period of

time for drug-loaded conjugate nanoparticles compared to chitosan nanoparticles. But, for a

drug like DH, with bulky therapeutic dose, a drug loading of 15-19% cannot be deemed well

enough and will require multiple puffs to make the required dose. Therefore, more

extensive studies are required to optimize drug loading if the formulation is intended for

pulmonary administration of this drug. However, the formulation could be a good ploy for

respiratory administration of potent drugs with small dose sizes. Although the drug-loading

Page 171: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 6 Preparation of Nanoparticles and In Vitro Studies on their Aerosolization and Drug Release

126

was unsuccessful with emulsion- solvent evaporation technique, the method could be useful

for other drugs with different physicochemical properties. The prepared blank particles

could also be tried for drug loading by passive absorption.

Page 172: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 173: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 174: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 175: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

127

7.1 INTRODUCTION The respiratory system is already an established route of drug delivery for lung diseases like

asthma and COPD and is gaining more attention for the delivery of drugs to systemic

circulation. However, it is a relatively new portal of drug administration compared to oral or

parenteral route and, so, to avoid any untoward effect on the respiratory system, it is

important to ensure in the first place the biosafety of any formulation intended for

inhalation.

As discussed in details in the literature review in the Chapter 2, the biosafety of inhaled

formulations can best be evaluated in vitro by a combination of a variety of toxicological

tests on respiratory epithelial cell lines to provide complementary information. Cell viability

assays, e.g. the MTT assay, are widely used to evaluate the biosafety of inhaled materials

(Salama et al., 2009b; Sivadas et al., 2008; Zhang et al., 2009). The MTT assay relies on

mitochondrial reduction of a yellow tertrazole dye, MTT [3(4, 5-dimethylthiazol-2-yl)-2, 5-

diphenyltetrazolium bromide] into insoluble formazan crystal as an indicator of cell viability.

Another indicator of adverse effects of inhaled agents on epithelial cell layers is any

pernicious change in the permeability of the epithelial barrier. This results from disruption

of intercellular tight junctions and can be assessed by measuring the permeability of a fluid

phase marker like sodium fluorescein across a confluent cell monolayer (Gartlon & Clothier,

2008 ; Grenha et al., 2007; Sivadas et al., 2008). Yet another important concern is to rule out

any untoward inflammatory response of the respiratory epithelium to the inhaled product.

This can be accomplished by checking the release of chemokines like IL-8 by an

immunological technique such as ELISA (Saedisomeolia et al., 2009; Sivadas et al., 2008).

IL-8 is a chemokine whose release is often associated with initiation of inflammatory

processes in the lung tissue (Drumm et al., 2000).

Previous studies on chitosan and its micro-/ nanoparticles on pulmonary epithelial cell lines

like Calu-3 and A-549 indicate their low toxicity on respiratory airways (Grenha et al., 2007;

Sivadas et al., 2008). TMC, a water soluble derivative of chitosan, has been reported to be

more toxic than the precursor chitosan (Choksakulnimitr et al., 1995; Fischer et al., 2003;

Page 176: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

128

Mao et al., 2005). The L-leucine conjugate of chitosan synthesized under this project is a

new compound and has shown improved water solubility compared to chitosan. So, it

warrants a thorough characterisation of its safety profile for using in a pulmonary delivery

formulation.

The respiratory epithelial cell models, used in previous toxicological investigations of

chitosan and TMC (e.g. calu-3 and A-549), were derived from human lung tumor. BEAS-2B

(Reddel et al., 1988) is a non-malignant human bronchial epithelial cell line immortalized

using an SV40/adenovirus-12 hybrid virus (Graness et al., 2002). It has been widely used as

an in vitro bronchial epithelial cell model (Wong et al., 2006). But, no study has so far been

reported to investigate the toxicity of chitosan or any of its derivatives using this cell line. It

can, therefore, be expected that a study of the toxicity of chitosan and the synthesized

L-leucine conjugate using this cell line would enrich and complement the existing wealth of

information.

This chapter evaluates the biosafety of chitosan, its L-leucine conjugate and their

nanoparticles made by emulsion-solvent evaporation technique in terms of the three

well-recognised toxicity indicators (cell viability, trans-epithelial permeability and

chemokine release) using BEAS-2B as an in vitro model.

7.2 RESULTS

7.2.1 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay

7.2.1.1 MTT Assay of Chitosan, Chitosan-L-Leucine Conjugate and their Nanoparticles The Figs. 7-1.1 a, b, c and d present the results of MTT assay of neat chitosan, chitosan

nanoparticles, neat chitosan-L-leucine conjugate and chitosan-L-leucine conjugate

nanoparticles, respectively in terms of % survival of BEAS-2B cells at progressively increased

concentrations (between 0.125 and 16 mg/mL) applied over a period of 12, 24 and 48

hours. The corresponding numerical data are presented in the Tables A7-1.1 (a), (b), (c) and

(d), respectively in the Appendix 7-1.

Page 177: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

129

As it is evident from the Fig. 7-1.1 a, the percent survival of the cells progressively decreased

with an increase in concentration of chitosan, but it did not fall below 50% in any of the

three treatment durations even at the highest concentration applied. Apparently, there was

an increased effect on cell survival with an increase in duration of treatment, but analysis of

variance (ANOVA) suggests that the effects were not significantly different (p >0.05). The

IC50 of the sample for the 12, 24 and 48 h treatment durations was determined to be 48, 24

and 17 mg/mL respectively.

Figure 7-1.1 a: Effect of Chitosan on the Viability of BEAS-2B Cell Line

The % survival of BEAS-2B cells was determined by MTT assay after 12, 24 and 48h exposure to increasing concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0, 12.0 and 16.0 mg/mL) of chitosan suspension in cell culture medium. Results presented as % survival relative to controls and expressed as the mean ± SE (n = 3).

0%

10%

20%

30%

40%

50%

60%

70%

80%

90%

100%

% S

urvi

val

Conc. (mg/ml)

12 Hours 24 Hours 48 Hours

Statistics: 12h=24h=48h @ significance level, α=0.05

IC50: • 12h — 48 mg/ mL • 24h — 24 mg/ mL • 48h — 17 mg/ mL

Page 178: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

130

As shown in the Fig. 7-1.1 b, up to a concentration of 4 mg/mL, chitosan nanoparticles also

showed a rather gradual decline in the cell survival with increasing concentration and the

value was 50% or above . However, beyond a concentration of 4 mg/mL, percentage of cell

survival abruptly fell reaching nearly zero at a concentration of 8 mg/mL. As regards time

effect, there was no significant difference between 12 and 24 h time periods (p > 0.5), but

the effect on cell survival produced following 48 h incubation was significantly higher

compared to other two durations (p < 0.05). The IC50 of the sample for the 12, 24 and 48 h

treatment was determined to be 6, 7 and 3 mg/mL, respectively.

Figure 7-1.1 b: Effect of Chitosan Nanoparticle on the Viability of BEAS-2B Cell Line

The % survival of BEAS-2B cells was determined by MTT assay after 12, 24 and 48h exposure to increasing

concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0, 12.0 and 16.0 mg/mL) of chitosan nanoparticle

suspension in cell culture medium. Results presented as % survival relative to controls and expressed as the

mean ± SE (n = 3).

-20%

0%

20%

40%

60%

80%

100%

120%

% S

urvi

val

Conc. (mg/ml)

12 Hours 24 Hours 48 Hours

IC50: • 12h — 6 mg/mL • 24h — 7 mg/mL • 48h — 3 mg/mL

Statistics: 12h=24h>48h @ significance level, α=0.05

Page 179: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

131

As evident from the Fig. 7-1.1 c, the chitosan-L-leucine conjugate also showed a

concentration dependent effect on the cell survival and the effect appeared to be more

pronounced than its precursor, chitosan. The effect on cell survival increased with increase

in the duration of treatment. In case of the 12 h treatment, the percentage of viable cells

was still higher than 50% up to a concentration of 4 mg/mL. The percentage of cell survival

showed a linear decline beyond the concentration of 2 mg/mL, reaching to nearly zero at a

concentration of 12 mg/mL. Prolonged treatments over 24h and 48h periods showed a

more rapid decline in cell survival reaching a percentage of 50% at about just 2 mg/mL and it

culminated to nearly zero at a concentration of 8 mg/mL. Apparently, the effects over 24 h

and 48 h periods were close to each other, but statistical analysis shows that the effects

were significantly different for all the three durations (p < 0.05). The IC50 of the samples for

the 12, 24 and 48 h incubation was determined to be 6, 4 and 4 mg/mL, respectively.

Figure 7-1.1 c: Effect of Chitosan-L-Leucine Conjugate on the Viability of BEAS-2B Cell Line

The % survival of BEAS-2B cells was determined by MTT assay after 12, 24 and 48h exposure to increasing

concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0, 12.0 and 16.0 mg/mL) of chitosan-L-leucine

conjugate solution in cell culture medium. Results presented as % survival relative to controls and expressed as

the mean ± SE (n = 3).

0%

10%

20%

30%

40%

50%

60%

70%

80%

90%

100%

% S

urvi

val

Conc. (mg/ml)

12 Hours 24 Hours 48 Hours

IC50: • 12h — 6 mg/mL • 24h — 4 mg/mL • 48h — 4 mg/mL

Statistics: 12h>24h>48h @ significance level, α=0.05

Page 180: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

132

Conjugate nanoparticles also showed a concentration- and time- dependent effect on the

cell viability (Fig. 7-1.1 d). However, the effect appeared to be more pronounced than neat

conjugate, the cell viability falling to about 50% or below at just 2 mg/mL concentration at

all the three treatment durations. For 12h and 24h durations, concentration required to

cause nearly 100% cell death was 12 mg/mL, while it was 8 mg/mL for 48h treatment

duration. According to analysis of variance, there was no significant difference between the

effects on cell viability observed over 12h and 24h durations (p>0.05); 48h effects were

significantly higher than 12h or 24h effects (p<0.05). The IC50 of the sample for 12, 24 and

48 h durations was determined to be 5, 4 and 2 mg/mL, respectively.

Figure 7-1.1 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticle on the Viability of BEAS-2B Cell Line

The % survival of BEAS-2B cells was determined by MTT assay after 12, 24 and 48h exposure to increasing

concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0, 12.0 and 16.0 mg/mL) of chitosan-L-leucine

conjugate nanoparticle suspension in cell culture medium. Results presented as % survival relative to controls

and expressed as the mean ± SE (n = 3).

0%

10%

20%

30%

40%

50%

60%

70%

80%

90%

% S

urvi

val

Conc. (mg/ml)

12 Hours 24 Hours 48 Hours

IC50: • 12h — 5 mg/mL • 24h — 4 mg/mL • 48h — 2 mg/mL

Statistics: 12h=24h>48h @ significance level, α=0.05

Page 181: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

133

7.2.1.2 MTT Assay of Diltiazem Hydrochloride Fig. 7-1.2 presents the effect of different concentrations of diltiazem hydrochloride (DH) on

the viability of BEAS-2B cell line after incubation for the 3 different time intervals (12, 24

and 48 h). Corresponding tabular data are presented in the Table A7-1.2 in the Appendix

7-1. As shown in the Fig. 7-1.2 and further confirmed by ANOVA post-hoc analysis, the

percentage of cell death progressively increased with increase in incubation period. In all

the three cases, cell viability appeared to remain more or less unaffected (rather slightly

enhanced) at 0.125 and 0.25 mg/mL concentration. But, beyond the concentration of 0.375

mg/mL % survival abruptly fell to nearly zero. The IC50 of the sample for 12, 24 and 48 h

treatment periods was determined to be 0.47, 0.43 and 0.37 mg/mL, respectively.

Figure 7-1. 2: Effect of Diltiazem HCl on the Viability of BEAS-2B Cell Line

The % survival of BEAS-2B cells was determined by MTT assay after 12, 24 and 48h exposure to increasing

concentrations (viz. 0.125, 0.25, 0.375, 0.50, 1.0, 2.0, 4.0, 8.0, 12.0 and 16.0 mg/mL) of diltiazem hydrochloride

solution in cell culture medium. Results presented as % survival relative to controls and expressed as the

mean ± SE (n = 3).

-20%

0%

20%

40%

60%

80%

100%

120%

140%

160%

% S

urvi

val

Conc. (mg/ml

12 hour 24 hour 48 hour

IC50: 12h — 0.47 mg/mL 24h — 0.43 mg/mL 48h — 0.37 mg/mL

Statistics: 12h>24h>48h @ significance level, α = 0.05

Page 182: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

134

7.2.2 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B cell Monolayer

7.2.2.1 Determination of the Polarisation Point of the BEAS-2B Cell Culture by TEER Measurement

Fig. 7-2.1 presents 10 days’ transepithelial electrical resistance (TEER) measurement across

BEAS-2B cell monolayer grown over 0.4 µm transwell inserts using RPMI as the culture

medium. Corresponding numerical data are presented in the Table A7-2.1 in Appendix 7-2.

The results suggest that the cell monolayers reach their maximum TEER at 4 days.

Figure 7-2. 1: Time Course of TEER Development in BEAS-2B Cell Monolayers Grown on Transwell Inserts

The TEER of cell monolayers was determined at 24 h intervals after seeding the inserts with BEAS-2B cells at a density of 5 × 105 cells/cm2. The monolayer TEER was corrected for the resistance of the filter support alone. Results expressed as the mean ± SE (n = 3).

7.2.2.2 Comparison of the Permeability of a Transwell Containing Confluent BEAS-2B Cell Monolayer and a Blank Transwell

The Fig. 7-2.2 presents the apparent permeability of the blank inserts (no cell) and seeded

inserts to Na Flu at the polarization point. Corresponding tabular data are presented in the

Table A7-2.2 in the Appendix 7-2. It clearly indicates that there was a comprehensive

reduction to the permeability of seeded inserts compared to the blank inserts, confirming

development of an effective barrier by BEAS-2B cells upon polarization.

0.002.004.006.008.00

10.0012.0014.0016.0018.0020.00

1 2 3 4 5 6 7 8 9 10

TEER

(Ohm

.cm

2)

Days

Page 183: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

135

Figure 7-2. 2: Permeability of Na Flu across a Blank Transwell and a Transwell Containing Confluent BEAS-2B Cell Monolayer

The amount of Na Flu transported to the basolateral chambers of blank transwells and transwells containing confluent cell monolayers was determined spectrofluorometrically 0.5 h after apical treatment with cell culture medium containing Na Flu (0.2 mg/mL). The apparent permeability coefficient (Papp) values were calculated using the equation (14) as described in detail in the Chapter 3, Section 3.2.3.3.2. Results expressed as the mean ± SE (n = 3).

7.2.2.3 Effect of Chitosan, Conjugate and their Nanoparticles on Transport of Na Flu across BEAS-2B Cell Monolayer

Figs. 7-2.3 a, b, c and d present the effect of chitosan, its L-leucine conjugate and their

nanoparticles, respectively on the permeability of sodium fluorescein across polarized

BEAS-2B cell monolayers at various concentrations (0.5-4 mg/mL) and time-points

(0.5-48 h). Corresponding tabular data are presented in the Tables A7-2.3 a, b, c and d,

respectively in the Appendix 7-2.

703

110

0

100

200

300

400

500

600

700

800

Blank Transwell Transwell containing polarizedBEAS-2B monolayer

Appa

rent

Per

mea

bilit

y C

oeff

icie

nt, P

app

( nm

/s)

Page 184: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

136

As shown in the Fig. 7-2.3 a and further confirmed by ANOVA with post-hoc analysis, there

was no significant change in the permeability of BEAS-2B cell monolayers treated with

chitosan suspension compared to untreated cell monolayer irrespective of concentration

and time (p value for concentration = 0.124 and p for concentration-time interaction =

0.123).

Figure 7-2.3 a: Effect of Chitosan on Na Flu Transport across the BEAS-2B Cell Monolayer

Cumulative amounts of Na Flu transported to the basolateral chambers of transwells containing polarized cell monolayers were determined by spectrofluorometry at 0.5, 1, 2 4, 6, 12, 24 and 48 h time-points after apical treatment with increasing concentrations (viz. 0, 0.5, 1, 2 and 4 mg/mL) of chitosan suspension in cell culture medium containing Na Flu (0.2 mg/mL). Results expressed as the mean ± SE (n = 3).

0.000

0.001

0.002

0.003

0.004

0.005

0.006

0.007

0.008

Cum

ulat

ive

amou

nt o

f Na

Flu

tran

spor

ted

(mg)

Time (Hour)

0 mg/ml

0.5 mg/ml

1 mg/ml

2 mg/ml

4 mg/ml

Statistics: Chitosan=Control (p >0.05) (@ all conc. and time-points)

Page 185: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

137

Fig. 7-2.3 b suggests that chitosan nanoparticles also did not cause any appreciable change

in the permeability of the cell monolayer compared to control. However, statistical analyses

indicate that the permeability of the cell monolayer treated with a concentration of 4

mg/mL was significantly lower than control (0 mg/mL) at initial time-points from 0.5 h up to

6 hours (p < 0.05), but it levels off with passage of more time at 12h time-point (p = 0.317,

0.780 and 0.079 at 12, 24 and 48 h, respectively).

Figure 7-2.3 b: Effect of Chitosan Nanoparticle on Na Flu Transport across the BEAS-2B Cell Monolayer

Cumulative amounts of Na Flu transported to the basolateral chambers of transwells containing polarized cell monolayers were determined by spectrofluorometry at 0.5, 1, 2 4, 6, 12, 24 and 48 h time-points after apical treatment with increasing concentrations (viz. 0, 0.5, 1, 2 and 4 mg/mL) of chitosan nanoparticle suspension in cell culture medium containing Na Flu (0.2 mg/mL). Results expressed as the mean ± SE (n = 3).

0.000

0.005

0.010

0.015

0.020

0.025

0.030

Cum

ulat

ive

amou

nt o

f Na

Flu

tran

spor

ted

(mg)

Time (Hour)

0 mg/ml

0 .5 mg/ml

1 mg/ml

2 mg/ml

4 mg/ml

Statistics: • 0.5, 1 and 2 mg/mL: Chitosan NP=Control • 4 mg/mL: Chitosan NP < Control (0.5h-6h)

@ significance level, α = 0.05

Page 186: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

138

Fig. 7-2.3 c presenting the effect of the chitosan-L-leucine conjugate apparently shows no

difference between the treatments and control. But, statistical analysis indicates that

permeability of the cell monolayers receiving 0.5, 1 and 2 mg/mL concentrations of the

conjugate were significantly higher than the control at most time-points (p = 0.003, 0.000,

0.000 respectively). But, there was found no significant difference among these 3

concentrations (p > 0.05). On the other hand, the permeability of the cell monolayer at 4

mg/mL concentration appeared not to be statistically different from that of the control (p =

0.999).

Figure 7-2.3 c: Effect of Chitosan-L-Leucine Conjugate on Na Flu Transport across the BEAS-2B Cell Monolayer

Cumulative amounts of Na Flu transported to the basolateral chambers of transwells containing polarized cell monolayers were determined by spectrofluorometry at 0.5, 1, 2 4, 6, 12, 24 and 48 h time-points after apical treatment with increasing concentrations (viz. 0, 0.5, 1, 2 and 4 mg/mL) of chitosan-L-leucine conjugate solution in cell culture medium containing Na Flu (0.2 mg/mL). Results expressed as the mean ± SE (n = 3).

0.000

0.002

0.004

0.006

0.008

0.010

0.012

0.014

0.5 1.0 2.0 4.0 8.0 16.0 32.0 64.0

Cum

ulat

ive

amou

nt o

f Na

Flu

tran

spor

ted

(mg)

Time (Hour)

0 mg/ml

0.5 mg/ml

1 mg/ml

2 mg/ml

4 mg/ml

Statistics: • 0.5 = 1 = 2 mg/mL: Conjugate>Control • 4 mg/mL: Conjugate = Control

@ significance level, α = 0.05

Page 187: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

139

In case of conjugate nanoparticles (Fig. 7-2.3 d), the permeability of the cell monolayers was

found to be significantly lower than control (p = 0.000) at all the 4 concentrations tested.

However, there was a very clear-cut concentration-dependent gradation in the permeability

— the highest concentration used in this experiment (4 mg/mL) showing a maximum that

approached close to the control at the initial time-points and became equal to that at 24

and 48 hour time-points.

Figure 7-2.3 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticle on Na Flu Transport across the BEAS-2B Cell Monolayer

Cumulative amounts of Na Flu transported to the basolateral chambers of transwells containing polarized cell monolayers were determined by spectrofluorometry at 0.5, 1, 2 4, 6, 12, 24 and 48 h time-points after apical treatment with increasing concentrations (viz. 0, 0.5, 1, 2 and 4 mg/mL) of chitosan-L-leucine conjugate nanoparticle suspension in cell culture medium containing Na Flu (0.2 mg/mL). Results expressed as the mean ± SE (n = 3).

0.000

0.001

0.002

0.003

0.004

0.005

0.006

0.007

0.008

0.009

0.010

Cum

ulat

ive

amou

nt o

f Na

Flu

tran

spor

ted

(mg)

Time (Hour)

0 mg/ml

0.5 mg/ml

1 mg/ml

2 mg/ml

4 mg/ml

Statistics: 0.5 < 1 < 2 < 4 mg/mL < Control @ significance level, α=0.05

Page 188: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

140

Fig. 7-2.4 compares the effects of the 4 different samples on the permeability of BEAS-2B

cell monolayer in terms of the relative permeability (Papp, polymer/Papp, control) of the

samples (at 2 mg/mL concentration) plotted against time. The concentration has been

chosen based on MTT assay results that suggest that the percentage of cell death caused by

all the four samples at this concentration is either below or around 50%. As evident from

the figure, the chitosan-L-leucine conjugate caused the highest permeation and its

nanoparticles the lowest. The chitosan and its nanoparticles demonstrated a comparable

permeability at most of the time-points and statistically there is no significant difference

between them (p = 0.589).

Figure 7-2. 4: Comparison of the Effect of Chitosan, Chitosan-L-Leucine Conjugate and their Nanoparticles on Na Flu Transport across the BEAS-2B Cell Monolayer

Cumulative amounts of Na Flu in basolateral chambers of transwells containing confluent cell monolayers were determined by spectrofluorometry at 0.5, 1, 2 4, 6, 12, 24 and 48 h time-points after apical treatment with the test samples (2 mg/mL) suspended/dissolved in cell culture medium containing Na Flu (0.2 mg/mL). The apparent permeability coefficient (Papp) values were calculated using the equation (14) as described in detail in the Section 3.2.3.3.2 of Chapter 3. Results presented as apparent permeability of cell monolayer to Na Flu relative to control and expressed as the mean ± SE (n = 3).

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

Papp

(pol

ymer

)/Pa

pp (c

ontr

ol)

Time (Hour)

ChitosanChitosan nanoparticlesConjugateConjugate nanoparticle

Statistics: Conjugate>Chitosan=Chitosan NP>Conjugate NP

(Conc. 2 mg/ mL; α = 0.05)

Page 189: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

141

7.2.3 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study Fig. 7-3 demonstrates the effect of increasing concentrations (0.5-4 mg/mL) of chitosan,

chitosan-L-leucine conjugate and their nanoparticles on IL-8 release from BEAS-2B cells upon

incubation for 24 hours compared to the no-treatment control (0 mg/mL). The

corresponding numerical data are presented in the Table A7-3 in the Appendix 7-3.

Figure 7- 3: Effect of Chitosan, its L-Leucine Conjugate and their Nanoparticles on IL-8 Release by BEAS-2B Cells

The IL-8 levels in culture supernatants were determined by ELISA 24 h after exposure of the cells with increasing concentrations (0, 0.5, 1, 2 and 4 mg/mL) of the test samples suspended/dissolved in the cell culture medium. Results expressed as the mean ± SE (n = 3).

As shown in the Fig. 7-3, chitosan caused mild, but significant, induction of IL-8 release

compared to control (0 mg/mL) at all the 4 concentrations applied. Although apparently

there was a progressive increase in lL-8 release with increasing concentration, there was no

0

500

1,000

1,500

2,000

2,500

3,000

3,500

0 1 2 3 4 5

Conc

entr

atio

n of

IL-8

(pg/

ml)

Concentration of the treatments (mg/ml)

Chitosan

Chitosan nanoparticles

Conjugate

Conjugate nanoparticles

Statistics: Conjugate NP > Conjugate > Chitosan = Chitosan NP> Control @ significance level, α = 0.05

Page 190: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

142

significant difference in responses to the 4 different concentrations as per statistical analysis

(p>0.05). Chitosan nanoparticles apparently elicited a higher response than neat chitosan at

the lowest concentration applied (0.5 mg/mL), but statistical analysis suggests that there

was no difference between the effects of chitosan and its nanoparticles at all the

concentrations tested (p>0.05). The chitosan-L-leucine conjugate caused a concentration-

depended and statistically significant induction of IL-8 release (p>0.05) up to the

concentration of 2 mg/mL, after which the response fell down at the concentration of 4

mg/mL. The response to conjugate was slightly lower than chitosan and its nanoparticles;

the difference was found to be statistically significant (p<0.05). In a pattern similar to neat

conjugate, its nanoparticles also caused a concentration-dependent induction of IL-8

expression up to a concentration of 2 mg/mL and a reduction in the effect at further higher

concentration of 4 mg/mL. The response was particularly strong and much higher than all

other 3 samples at 1 and 2 mg/mL concentration.

7.3 DISCUSSION

7.3.1 In Vitro Evaluation of Cytotoxicity on the Respiratory Epithelial Cell Line BEAS-2B by MTT Assay

This experiment was meant to investigate in vitro the potential of the chemically

synthesized chitosan-L-leucine conjugate and its nanoparticles to cause any toxicity to

pulmonary epithelium in comparison to the parent commercially available polymer,

chitosan and its nanoparticles. This was a prologue to other experiments designed in this

series to evaluate the effect of these materials on trans-epithelial permeability and to assess

their inflammatory potential. The study assessed effects of increasing concentrations of

chitosan, its L-leucine conjugate and their nanoparticles on cell viability in order to elucidate

the concentration-effect relationship and identify the highest concentration of the samples

that would exhibit a tolerable level of toxicity towards the cell line. The concentrations

exhibiting an accepted level of toxicity were then used to investigate the potential of the

samples to modulate epithelial permeability or elicit inflammatory response.

Page 191: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

143

As shown in the Figs. 7-1.1 a, b, c and d, in general, all the 4 samples viz. chitosan, chitosan

nanoparticles, conjugate and conjugate nanoparticles, showed a concentration- and time-

dependent effect on the viability of cells — the percentage of cell survival showing a

progressive decrease with increased concentration and longer exposure, although the

magnitude of cell death varied with different samples. The intensity of the effect, as per

statistical analysis, was progressively higher in the order of chitosan, chitosan nanoparticles,

conjugate and conjugate nanoparticles (p<0.05). As shown in the Fig. 7-1.1 a, in agreement

with previous literature that reported low cytotoxicity of chitosan in various respiratory cell

lines like calu-3 or A-549 (Florea et al., 2006; Grenha et al., 2007; Huang et al., 2004; Lim et

al., 2001; Smith et al., 2004; Smith et al., 2005), the BEAS-2B cell line used in this study also

showed a high percentage of cell survival following treatment with chitosan over the entire

concentration range applied and all the three time periods tested. The cell survival did not

fall in any case below 50% (Fig. 7-1.1 a). Chitosan nanoparticles also showed a high

percentage of cell survival up to a concentration of 4 mg/mL. However, beyond the

concentration of 4 mg/mL, percentage of cell survival abruptly declined reaching nearly zero

at a concentration of 8 mg/mL (Fig. 7-1.1 b). Although the nanoparticles caused higher

percentage of cell death compared to the neat chitosan, it is consistent with the findings of

some previous investigators in consideration of dose applied. For instance, Huang et al.

(2004) reported a 70% reduction in the viability of A-549 cells by chitosan nanoparticles at a

concentration of about 1 mg/mL. But, it is in contradiction to the report of Vllasaliu et al.

(2010) who found the effect of the nanoparticles on the calu-3 cells to be lower than

chitosan solution. Fig. 7-1.1 c suggests that the L-leucine conjugate of chitosan was more

toxic than its precursor, chitosan. This finding appears to be reasonable if it is compared

with another water-soluble derivative, trimethylchtiosan chloride (TMC) that has been

reported to be more toxic than chitosan (Choksakulnimitr et al., 1995; Fischer et al., 2003;

Mao et al., 2005). The increased toxicity has been attributed to its increased solubility that

allows a better interaction with anionic cell surface components. The conjugate

nanoparticles appeared to be more toxic than neat conjugate. Nonetheless, they still had an

IC50 value of ≥ 2 mg/mL even at the longest exposure of 48 h (Fig. 7-1.1 d).

Page 192: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

144

Considering that the particles are intended for the delivery of the drug, DH to the lungs, an

attempt was made to assess the effect of the drug as well on the viability of BEAS-2B cell

line. As shown in the Fig. 7-1.2, the drug appeared to be rather more toxic to the cell line

causing complete cell death above a concentration of 0.375 mg/mL. No study has been

reported in the literature on the effect of DH on any of the pulmonary cell lines. There are,

however, a few studies reporting significant inhibition of vascular smooth muscle cells

(VSMCs) by the drug (Salabei et al., 2012; Yan et al., 2007). It would be appropriate to note

here that the toxicity profile might improve upon loading the drug into polymeric

nanoparticles. It has been shown earlier by Manca et al. (2008) that incorporation of the

drug rifampicin into polymeric microparticles (PLGA, chitosan and chitosan-coated PLGA)

substantially reduced drug toxicity to alveolar epithelial cell line, A-549.

Using chitosan/ conjugate nanoparticles loaded with the model drug (DH), no experiments

on the toxicity have been performed in this study. Therefore, it would be relevant to make a

preliminary theoretical assessment of the toxicity status of such particles on the basis of the

experimental data obtained on drug release from drug-loaded particles and cytotoxicity of

blank particles. As shown in the Section 3.2.2.5 of the Chapter 3, DH would require a

loading dose (DL) of 10.85 mg to initially achieve the desired blood level of 0.05 µg/ mL and

maintenance dose release rate (MDR) of 2.06 mg/h for maintaining this blood level over an

extended period of time. With an MDR of 2.06 mg/h, the amount of DH to be added as the

maintenance dose (Dm) would be 24.72, 49.44 and 98.88 mg for 12, 24 and 48h,

respectively. Added these values to the DL of 10.85 mg, the total doses of DH for a

controlled release formulation stand out to be 35.57, 60.29 and 109.73 mg for 12, 24 and

48h, respectively. On the other hand, the IC50 values of blank chitosan and conjugate

nanoparticles obtained in this study were 6, 7 and 3 mg/mL for chitosan NP and 5, 4 and 2

mg/mL for conjugate NP for 12, 24 and 48h treatments, respectively (Figs. 7-1.1 b & d). It

has been reported that, in a healthy adult, lungs have an air surface liquid (ASL) volume of

3.6 mL (Widdicombe, 2002). Therefore, taking primarily the IC50 values as the maximum

permissible limits, it can be said that the amounts of nanoparticles to be deposited in the

lungs should not exceed 21.6, 25.2 and 10.8 mg for chitosan NP and 18, 14.4 and 7.2 mg for

conjugate NP for 12, 24 and 48h exposures, respectively. So, to achieve a DH blood level of

0.05 µg/mL and maintain it over a period of 12, 24 and 48h duration, it will be required to

Page 193: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

145

prepare nanoparticle formulations containing 35.57, 60.29 and 109.73 mg of DH with an

amount of chitosan not exceeding 21.6, 25.2 and 10.8 mg or an amount of conjugate not

exceeding 18, 14.4 and 7.2 mg, respectively. In addition, the release profiles of the prepared

nanoparticle formulations should be such that 10.85 mg of drug would be released in burst

and the rest in a controlled manner at a rate of 2.06 mg/h. As discussed in the Section 6.3.4

of the Chapter 6, with the release profiles demonstrated by the prepared nanoparticles

formulaitons, a lung deposition of 423.83 and 175.00 mg of DH-loaded chitosan and

conjugate nanoparticles, respectively, would be required to achieve the desired blood level

of 0.05 µg/mL, although, once deposited, they could be expected to release the drug at a

rate close to the required MDR of 2.06 mg/h for a prolonged period of time (8 days for

chitosan and 16 days for conjugate nanoparticles). These doses of chitosan and conjugate

nanoparticle formulations are much higher than that could be considered acceptable from

toxicity point of view as per above discussion. Therefore, further studies are required to

reduce the dose size and increase the drug loading into the particles. One option in this

direction could be to add in the nanoparticle formulations an agent like lactose or mmanitol

either as a bulking agent or partial replacement of some polymer. As discussed in the

Section 6.3.4 of Chapter 6, this can help in increasing drug loading and release. Additionally,

they can also play a role in reducing the toxicity by reducing the polymer content in the

formulation. It would be pertinent to note here that the toxicity level shown by the chitosan

and conjugate nanoparticles could still be good enough for drugs with small dose sizes (such

as salbutamol sulphate, morphine etc.). Another concern here is the cytotoxicity profile of

the model drug, DH showing an IC50 value of 0.47, 0.43 and 0.37 mg/mL for 12, 24 and 48 h,

respectively. It has, however, already been noted that incorporation of the drug into the

particles could potentially reduce its toxicity to a substantial extent (Manca et al., 2008).

Besides, for a controlled release dosage form, kr (release rate constant) <<< ka (absorption

rate constant). Thus, in the kinetic scheme of drug delivery from such a dosage form, the

absorptive phase essentially becomes insignificant compared with the drug release phase

(Fig. 7-4) (Troy & Beringer, 2006). So, the drug could be expected to be absorbed from the

lungs into the blood stream as soon as it is released. This transient stay of the drug released

from the nanoparticles in to the lungs could also be expected to reduce its toxicity on the

pulmonary epithelium.

Page 194: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

146

Figure 7- 4: Kinetic Scheme for Drug Disposition from Immediate Release and Controlled Release Dosage Forms

7.3.2 In Vitro Evaluation of the Effect on the Integrity of Respiratory Epithelium by Sodium Fluorescein Transport Assay across BEAS-2B Cell Monolayer

Chitosan, its L-leucine conjugate and their nanoparticles were investigated for their effect

on the transepithelial permeability across polarized BEAS-2B cell monolayers by Na Flu

transport assay. Before initiating the actual experiments, a preliminary study was performed

to determine the time-point by which the cells reach the confluence level under the

experimental conditions to be employed (Fig. 7-2.1). In addition, the sodium fluorescein

transport assay was also performed to confirm the development of an effective

permeability barrier upon polarization of cells (Fig. 7-2.2). Based on the MTT assay results,

4 concentrations, viz. 0.5, 1, 2 and 4 mg/mL were selected for the experiment at which most

samples showed a cell viability of 50% or above and beyond which there was an abrupt

decline in % cell survival.

As shown in the Fig. 7-2.3 a and confirmed by statistical analysis, in contradiction to

previous literature reports on other cell lines (Bonferoni et al., 2008; Florea et al., 2006;

Vllasaliu et al., 2010), chitosan did not cause any significant change to the permeability of

BEAS-2B cell monolayer irrespective of concentration and time (p>0.05). This apparent

contradiction might be related to the cell line difference, but the finding is not unexpected if

Page 195: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

147

we consider the experimental pH of 7.4 — at which chitosan has been reported by some

investigators (Borchard et al., 1996; Kotze et al., 1998a) to be ineffective as a permeability

enhancer owing to its poor solubility that hinders an intimate contact (molecular

interaction) between the substance and the cell. A similar trend was shown by chitosan

nanoparticles (Fig. 7-2.3 b) except at the highest concentration of 4 mg/mL applied that

showed a statistically significant reduction in permeability at initial time-points up to a

period of 6 h (p<0.05). There are contradictory reports in the literature about the effect of

particulate chitosan on enhancing permeability across monolayers of the pulmonary

epithelial cell line, calu-3 (Grenha et al., 2007; Sivadas et al., 2008; Vllasaliu et al., 2010).

While Vllasaliu et al. (2010) reported a significant bronchoepithelial permeation

enhancement across calu-3 cell monolayer, others (Grenha et al., 2007; Sivadas et al., 2008)

observed that enhancement in permeability was statistically insignificant. Unlike neat

chitosan, the L-leucine conjugate caused significant enhancement in permeability (p>0.05)

up to a concentration of 2 mg/mL, but no difference was seen with further increase in

concentration (p<0.05) (Fig. 7-2.3 c). A higher concentration of 4 mg/mL again showed a

deviation from the usual trend demonstrating no significant difference from the control

(p>0.05). Considering higher solubility of the conjugate in water, an enhancement in cell

layer permeability was a reasonable outcome if we compare with the other

well-investigated water-soluble chitosan derivative, TMC that has been reported to have a

strong permeation enhancing effect (Kotze et al., 1997a; Thanou et al., 2000a; van der

Merwe et al., 2004). In contrast to neat chitosan-L-leucine conjugate, its nanoparticles

showed a significant reduction in permeability at all concentrations (p<0.05). However,

there was a concentration-dependent increase in the permeability and, at the highest

concentration of 4 mg/mL the permeability was very close to that of the control

(Fig. 7-2.3 d). The reduction in the permeability does not fit with the reports on the micro-/

nanoparticles of the other water-soluble derivative, TMC that have been found to induce

permeation enhancement similar to neat TMC (Amidi et al., 2008a; Amidi et al., 2008b;

Chen et al., 2008; Coco et al., 2013; Mi et al., 2008; Sandri et al., 2010; Sandri et al., 2007).

Further studies are required to understand this apparently deviant behaviour of conjugate

nanoparticles. To summarise, chitosan and chitosan nanoparticles did not cause much

difference in the permeability. The conjugate caused a slight, but significant enhancement

Page 196: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

148

of the permeability, while the conjugate nanoparticles had the opposite effect. Thus, the

rank order of the 4 samples in terms of associated transepithelial permeability was:

conjugate>chitosan=chitosan nanoparticle>conjugate nanoparticles.

It is evident that, with particulate chitosan and both the neat and particulate conjugate, the

permeability effect at the concentration of 4 mg/mL deviated from the pattern shown by

the lower concentrations of 0.5, 1 and 2 mg/mL (Fig. 7-2.3 b, c, d). This apparent aberration

could be related to the higher level of cell toxicity observed at such a high concentration (as

already shown under the MTT assay results in Fig. 7-1.1 a, b, c and d), but further studies

are required to elucidate the exact cause of this deviation.

7.3.3 In Vitro Evaluation of Inflammatory Effect by Chemokine (IL-8) Release Study This experiment attempted to investigate in vitro the inflammatory potential of the novel

chitosan-L-leucine conjugate synthesized under this project and its nanoparticles on

pulmonary epithelium in comparison with parent chitosan and its nanoparticles. Release of

the chemokine interleukin-8 (IL-8) was taken as an indicator of inflammatory response and

was estimated by enzyme-linked immunosorbent assay (ELISA). As for the Na Flu transport

assay described in the previous section, a series of concentrations, viz. 0.5, 1, 2 and 4

mg/mL, of the samples were chosen for the experiments on the basis of MTT assay results.

As shown in the Fig. 7-3, chitosan and its nanoparticles showed a mild, but significant

induction of IL-8 release; however there was no significant increase in the effect with

concentration (p>0.05). Statistical analysis also shows that there was no difference between

the effects of neat and particulate chitosan (p>0.05). Previous studies with chitosan

microparticles on calu-3 cell line reports contradictory outcomes in relation to IL-8 release.

While Witschi & Mrsny (1999) reported induction of IL-8 by chitosan microparticles, other

investigators did not find any significant difference compared to control (Sivadas et al.,

2008). Although the neat conjugate appeared to be less inflammatory than both the neat

and particulate chitosan, conjugate nanoparticles showed a completely opposite trend.

While the IL-8 release caused by conjugate nanoparticles at 0.05 mg/mL (917±50 pg/mL)

was statistically indifferent from that of control (768±76 pg/mL), the IL-8 levels observed for

1, 2 and 4 mg/mL concentration of the conjugate nanoparticles were 2230±251, 2957±150

Page 197: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

149

and 1959±64 pg/mL respectively, which were in the order of 3-4 times the IL-8 level induced

by the control (Table A7-3 in Appendix 7-3). This is a concern in consideration of the large

dose size of the model drug, DH that could zeopradize the pharmaceutical benefits offered

by the conjugate nanoparticles. Further studies are required to minimize the dose size of

conjugate nanoparticles required. Replacement of part of the conjugate by a bulking agent

like lactose or mannitol, as suggested for increasing the drug loading and release in

chapter 6, might prove beneficial in this context too. However, since there was no

significant induction of IL-8 at 0.5 mg/mL, the material might still be sufficiently safe for

potent drugs with small dose sizes (e.g. salbutamol sulphate, morphine).

The tapering down of the effect of both the neat and particulate forms of the conjugate

beyond 2 mg/mL concentration may be related to the high cytotoxicity and considerable cell

death taking place at such a high concentration as evidenced by MTT assay. It is possible

that due to high toxicity exerted by such a high concentration the cells fail to respond

normally to the treatment applied.

7.4 CONCLUSION The biosafety of chitosan, its L-leucine conjugate and their nanoparticles, made by

emulsification solvent evaporation, have been studied in vitro in terms of cell viability,

transepithelial permeability and the chemokine, IL-8 release using the bronchial epithelial

cell line, BEAS-2B as an in vitro model. In accordance with previous literatures on other

respiratory epithelial cell lines like calu-3 and A-549, chitosan and its nanoparticles showed

a low toxicity on BEAS-2B cell line. Similarly, the synthesised L-leucine conjugate of chitosan

also demonstrated a low toxicity. The conjugate nanoparticle appeared to be relatively

more toxic and inflammatory. However, its IC50 did not fall below 2 mg/mL even at 48 h

treatment duration. Besides, it did not cause any significant induction of IL-8 on 24 h

treatment of BEAS-2B cell line at 0.5 mg/mL concentration. So, it could still be considered

to have an acceptable level of toxicity, particularly for respiratory delivery of potent, low-

dose drugs (such as salbutamol sulphate or morphine). Further, epithelial cells in vitro can

be more sensitive to toxicological insult compared to the epithelium in situ. In this context, a

low level of toxicity demonstrated in vitro by these substances is an encouraging indicator of

Page 198: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 7 In Vitro Evaluation of Toxicity and Inflammatory Activity

150

their safety for respiratory delivery. However, the large dose size of the model drug, DH

would potentially require a large volume of lung deposition of the nanoparticle formulation

and so further studies are required to minimize the required dose of the formulation if this

is to be considered for pulmonary delivery of DH. Replacement of part of the polymer in the

nanoparticle formulaion with a bulking agent like lactose or mannitol could be a useful ploy

to be considered in this line.

Page 199: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 200: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 201: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 202: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

151

8.1 Summary This study revealed new strategies for addressing the issues of controlling drug release and

enhancing aerosolization — two major concerns of present-day research on DPI

formulation. Because of many unique advantages of controlled release therapy leading to

enhanced patient compliance and convenience and better therapeutic outcomes,

formulation scientists are in constant endeavour of developing DPIs in a controlled release

form, although no such formulation has yet been launched to the market. Since the natural

polymer chitosan has many attractive features including low toxicity, biocompatibility,

biodegradability and mucoadhesiveness, its micro-/ nanoparticles have drawn special

attention for formulating controlled release DPIs. The poor dispersibility of DPIs stems from

agglomeration of micronized particles (<5 µm) required for their deposition into the deeper

regions of respiratory airways. The commonly used approach to improve dispersion of

micronized particles from a DPI is mixing with an interactive carrier (e.g. lactose, mannitol)

that acts to diffuse the cohesive force between like particles. Another popular approach is

addition of an agent like L-leucine, magnesium stearate or PEG 6000 in the formulation as

aerosolization enhancer. Besides, many other techniques including addition of fines and

multifarious engineering of drug or carrier particles have been employed. Of late, addition

of L-leucine in the formulation as an aerosolization enhancer has been shown to greatly

reduce particle agglomeration and improve dispersibility of chitosan-based DPI

formulations. This study demonstrated that chemical conjugation of L-leucine to chitosan

can also enhance dispersibility of particles from a DPI formulation. Using diltiazem

hydrochloride — an antihypertensive agent undergoing extensive first-pass metabolism

following oral administration — as the model drug, this work has further revealed that

L-leucine conjugated chitosan nanoparticles can offer better controlled release profile

compared to those prepared from the parent chitosan.

The scientific basis of this work was the hypothesis that L-leucine conjugation to chitosan

would create an amphiphilic environment around the chitosan backbone resulting in

reduced inter-particle interaction and so enhanced dispersion of particles. Another

consideration here was freedom of the pulmonary route from the drawback of hepatic first-

Page 203: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

152

pass metabolism that potentially could improve bioavailability of a drug like diltiazem

hydrochloride. Based on these considerations, the first step in course of this work was

synthesis of a chitosan-L-leucine conjugate. The next step to follow was preparation of

nanoparticulate formulations of both chitosan and its conjugate and assessment of their

suitability for lung deposition in terms of particle size, shape and surface characteristics.

Then, the aerosolization property and drug release profiles of the two formulations were

studied and compared. Finally, the nanoparticle formulations were also assessed for their

safety for respiratory delivery.

Forthcoming sections summarize the key findings of this research and their implications.

This is followed by an account of the potential future research in this connection.

8.1.1 Conjugation of L-leucine with Chitosan This study has demonstrated successful synthesis of a chitosan-L-leucine conjugate. It was a

new compound with improved aqueous solubility that allowed its prompt dissolution in

water or PBS without the requirement of any acid as for chitosan. This higher solubility of

the conjugate is attributable, at least in part, to the amphiphilic environment created by the

conjugated L-leucine around the chitosan backbone. It is a great finding with potentially far-

reaching impact in view of the poor solubility of chitosan in both aqueous and organic

solvents that restricted so far its utilization to the potential in pharmaceutical, biomedical

and other fields. This discovery could be expected to open a new window in the effort of

scientists for producing soluble derivatives of the polymer for its better exploitation.

The structure of chitosan offers three possibilities for conjugation of L-leucine — at C-2, C-6

or both. In consideration of the higher reactivity of the ─NH2 group, this study performed, as

the first approach, selective conjugation at C-2. A protection-deprotection technique was

employed to ensure regioselectivity. Initially, the C-2 was protected by N-phthaloylation and

after tritylation at C-6 it was removed by hydrazinalysis. Then, L-leucine was conjugated at

C-2 by reacting with Boc-L-leucine succinimide where the Boc was another protecting group

for preventing polymerization of L-leucine. Finally, both trityl and Boc groups were

deprotected simultaneously by 4M HCl in Dioxane. The intermediates and the final product,

Page 204: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

153

Chitosan-N-L-leucine HCl were characterized by a combination of FT-IR, 1H and 13C NMR

spectroscopy and elemental analysis. As the last two products in the pathway were new to

the existing literature, they were further characterized by 2D 1H-13C HSQC spectroscopy.

The FTIR spectra showed characteristic absorption for N-phthaloyl-chitosan (phthaloyl C=C

1703 and 1774 cm-1, phthaloyl aromatic 718 cm-1), N-phthaloyl-6-O-trityl-chtiosan (trityl C=C

1448 and 1490 cm-1, trityl aromatics 699, 746 and 764 cm-1 in addition to phthaloyl signals),

6-O-trityl-chitosan-N-Boc-L-leucine (intensified amide-I signal at 1683 cm-1, Boc t-butyl/

L-leucine isopropyl at 1367 and 1390 cm-1 in addition to trityl signals) and chitosan-N-L-

leucine.HCl (amide-1 1628 and 1672, L-leucine isopropyl 1372 and 1390 cm-1). The IR spectra

of 6-O-trityl-chitosan confirmed disappearance of the phthaloyl signals (at 1712 and

1776 cm-1) while still keeping the trityl signals in place. The spectrum of chitosan-

N-L-leucine.HCl also showed disappearance of trityl signals (at 702, 747 and 764 cm-1) and

reduction in the intensity of the doublet at 1372 and 1390 cm-1 giving an indication that the

Boc group has been removed and the signal arose from L-leucine isopropyl only.

The solubility of N-phthaloyl-chitosan and N-phthaloyl-6-O-trityl-chitosan in common NMR

solvents was not good enough for getting 1H and 13C NMR spectra in solution and they were

acetylated to improve their solubility and get NMR spectrum in solution. The 1H and 13CNMR

spectra of the peracetate derivatives in CDCl3 showed characteristic chemical shifts for the

phthaloyl and trityl aromatic protons and carbons, respectively, in appropriate regions in

addition to those of pyranose. The proton NMR spectrum of 6-O-trityl-chitosan in

pyridine-d5 confirmed the removal of phthaloyl peaks while still retaining the trityl peaks.

The compound failed to show enough solubility either as such or as peracetate derivative to

get 13C NMR spectrum in solution. The 1H and 13C NMR spectra of 6-O-trityl-chitosan-Boc-

N-L-leucine in pyridine-d5 showed characteristic signals at 1.44 and 28.9 ppm for Boc t-butyl

in addition to the signals for trityl aromatics, pyranose and L-leucine protons and carbons at

appropriate places, thereby confirming conjugation of Boc-L-leucine to 6-O-trityl-chitosan.

On the other hand, the 1H and 13C NMR spectra of chitosan-N-L-leucine.HCl in D2O

confirmed removal of the Boc and trityl peaks while still retaining the pyranose and

L-leucine proton and carbon signals. As Boc t-butyl and L-leucine isopropyl had IR absorption

at the same positions, the NMR spectra was important to definitively ascertain the

Page 205: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

154

conjugation of L-leucine moiety and ultimate deprotection of the Boc group. The 2D spectra

of 6-O-trityl-chitosan-N-Boc-L-leucine and chitosan-N-L-leucine.HCl showed clear-cut data

points correlating the carbon signals to proton signals. The 2D spectrum of 6-O-trityl-

chitosan-N-Boc-L-leucine, however, missed the data points for the anomeric carbon, but the

presence of corresponding data points in the 2D spectrum of the final product, chitosan-

N-L-leucine.HCl (the next compound in the pathway) confirmed that their absence from the

2D spectrum of the former was simply due to lack of enough solubility. Taken together, the

IR and NMR data conclusively confirmed the identity of the intermediates and the final

product.

For further confirmation of conjugation of L-leucine to chitosan and having an insight into

the phenomenon at the surface level, the final product chitosan-L-leucine.HCl was also

investigated by XPS analysis. The high resolution XPS scan of N showed a reduction in the

intensity of NNH signal and elevation in the intensity of NN+ and Namide, as was expected from

conjugation of L-leucine to chitosan in consideration of the chemical structure of chitosan,

L-leucine and the conjugate. Moreover, high resolution scan of C also showed expected

elevation in C-C carbon signal, further confirming the conjugation.

The elemental analysis of most intermediates and the final product provided satisfactory

results allowing calculation of the degrees of substitution and elimination of the substituent

groups. The DS values for the substitution of phthaloyl, trityl and Boc-L-leucine moieties in

N-phthaloyl-chitosan, N-phthaloyl-6-O-trityl-chitosan and 6-O-trityl-chitosan-N-Boc-L-leucine

were determined from the C/N ratios of elemental analysis to be 0.91, 1.06 and 0.74,

respectively. The degree of elimination of Boc and trityl moieties from 6-O-trityl-chitosan-

N-Boc-L-leucine to give chitosan-N-L-leucine.HCl was calculated to be 91%. However, the

C/N ratio from the microanalytical data of 6-O-trityl-chitosan gave a value of 1.13 as the

degree of elimination of phthaloyl groups from N-phthaloyl-6-O-trityl-chitosan, which was

higher than the theoretical maximum of 0.91. This deviation was probably due to some

detritylation taking place at the high temperature condition of ~110 °C at which the reaction

was conducted. So, based on IR and NMR data, suggesting complete removal of phthaloyl

groups, the degree of dephthaloylation was taken to be 0.91.

Page 206: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

155

The yield of the reaction products were determined on the basis of their calculated degrees

of substitution to be 94.70% for N-phthaloyl-chitosan, 80% for N-phthaloyl-6-O-trityl-

chitosan, 96% for 6-O-trityl-chitosan, 70% for 6-O-tityl-chitosan-N-Boc-L-Leucine and 66% for

chitosan-N-L-leucine.HCl. This means that the reactions proceeded with reasonably good

yields confirming their soundness from quantitative point of view too.

8.1.2 Preparation and Characterization of Chitosan and Conjugate Nanoparticles and their Comparison in Terms of Aerosolization and Drug Release The study showed successful preparation of chitosan and conjugate nanoparticles with a

particle size of less than 100 nm that indicated suitability of the particles for deposition into

deeper regions of lungs. Of the two methods employed for nanoparticle preparation,

however, the W/O emulsion- solvent evaporation method failed to load the model drug

diltiazem hydrochloride into the particles, while the other method W/O emulsion-

glutaraldehyde crosslinking was successful both in generating the particles of desired size

and loading the drug into them. Although the emulsion- solvent evaporation method failed

to load the drug, it was still a significant, novel outcome in the sense that it was the first

report of its kind for generating chitosan nanoparticles by this method using paraffin oil as

the external phase. The method might be useful for loading other drugs with different

physicochemical properties. It could also be tried for loading diltiazem hydrochloride or

other drugs by passive absorption from an aqueous solution.

The study demonstrated superiority of conjugate nanoparticles over those of chitosan in

terms of drug entrapment/loading, drug dispersion and controlled release. The conjugate

nanoparticles showed an entrapment efficiency of 46±1%, which was significantly higher

(p<0.05) than that of chitosan nanoparticles (38±1%). Similarly, the % loading of conjugate

nanoparticles (20±1%) was significantly higher (p<0.05) than that of chitosan nanoparticles

(16±1%). This higher entrapment efficiency and drug loading (%) can be taken as a sequel of

the higher aqueous solubility of the conjugate that allowed better association of the drug

molecules with the polymer.

Page 207: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

156

The drug release studies in PBS (pH 7.3±0.2, 37 °C) showed twice as much higher (52%) and

more prolonged (16 days) drug release from conjugate nanoparticles compared to that from

chitosan nanoparticles (23%, 8 days). In both the cases, there was an initial burst release

(chitosan 16%, conjugate 31%) within the first 30 min followed by a slow, controlled release

over a prolonged period (chitosan 7%, conjugate 21%). Such a biphasic release can

conveniently meet the requirements of a controlled release formulation, with the initial

burst release offering the loading dose (DL) required for a prompt achievement of the

desired blood level and the subsequent controlled release maintaining this level over time.

The higher and more prolonged drug release from conjugate nanoparticles is again

attributable to higher solubility of the conjugate that allowed for quicker hydration of the

polymeric matrix promoting faster drug dissolution and release. On the other hand, the

higher drug loading of conjugate nanoparticles, which is also attributed to higher solubility

of the conjugate, produced a stronger driving force for drug release upon dissolution of the

drug in the matrix. Kinetic analysis with different models demonstrated that the drug

release occurred by Fickian diffusion (release exponent, n <0.45) and the release rate can

best be described by square root of time kinetics (Higuchi model) with a release rate

constant (kH) of 0.4709 × 10-2 and 1.1159 × 10-2 per hour for chitosan and conjugate

nanoparticles, respectively. In consideration of the minimum effective concentration, Cd of

0.05 μg/mL, apparent volume of distribution, Vd of 3.1 L/Kg and first order overall

elimination rate constant, ke of 0.19/h for DH and assuming 100% absorption from the

lungs, the required initial loading dose (DL) and the maintenance dose release rate (MDR) of

DH stand out to be 10.85 mg and 2.06 mg/h, respectively. It is estimated that, with 16 and

20% drug loading and 16 and 31% burst release for chitosan and conjugate nanoparticles,

respectively, a lung deposition of 423 and 175 mg of drug-loaded chitosan and conjugate

nanoparticles, respectively, would be required to provide an initial DH loading dose of 10.85

mg. With a release rate constant (kH) of 0.4709 × 10-2 and 1.1159 × 10-2 per hour for

chitosan and conjugate nanoparticles, respectively, these doses can provide a release rate

of 1.99 and 1.95 mg per hour, respectively. These values are close to the required

maintenance dose release rate (MDR) of 2.06 mg/h for DH. This means that, although

inhalation of such a bulky dose might be inconvenient for the patient, once inhaled the

plasma concentration would be maintained close to the desired level over time for the

Page 208: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

157

length of the estimated controlled release period (8 and 16 days for chitosan and conjugate

nanoparticles, respectively). Further studies are required, however, to increase the drug

loading and release so that the dose size could be reduced to a more convenient level. Use

of bulking agents like lactose or mannitol to replace part of the polymer component might

help in increasing drug loading and release and in turn minimizing the dose size. The present

level of drug loading and pattern of drug release can still be useful for more potent drugs

with small dose sizes (e.g. salbutamol sulphate, morphine).

The aerosolization study of nanoparticle formulations with the in vitro lung model, Twin

Stage Impinger (TSI) revealed that both the blank and drug-loaded nanoparticles of

conjugate could produce significantly higher (p<0.05) fine particle fraction (FPF) than

corresponding chitosan nanoparticles (Blank nanoparticles: chitosan 21±0.7%, conjugate

24±0.8%; Drug-loaded nanoparticles: chitosan 15±1.5%, conjugate 19±1.01%). This was an

important finding that corroborated the hypothesis made in this regard as the background

of this project. Although these FPF values do not appear to be very great in comparison with

those available from currently available DPIs, they bear special significance from two

perspectives: firstly, this finding opened a new window in current efforts for increasing

dispersion from a DPI and secondly, this level of FPF cannot be expected with conventional

DPIs containing drug particles without mixing with an interactive carrier like mannitol or

lactose.

8.1.3 Toxicity and Inflammatory Effect In the final part of this research, the polymers were assessed, both in neat and particulate

forms, for their safety for pulmonary delivery using cytotoxicity, trans-epithelial

permeability and chemokine release as the indicators of their toxicity and inflammatory

effect. The bronchial epithelial cell line, BEAS-2B was chosen as the in vitro model in

consideration of the fact that no study on the toxicity and inflammatory effect of chitosan or

any of its derivatives has previously been reported for this cell line. Moreover, being a

non-malignant respiratory epithelial cell line, investigation performed on this cell line was

expected to complement previous reports on cell lines like calu-3 or A-549 which are

malignant in nature.

Page 209: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

158

The results of MTT cell viability assay and the assessment of the chemokine, IL-8 induction

indicated that particulate forms of the polymers were more cytotoxic and inflammatory

than the neat polymers, where conjugate nanoparticles appeared to be the most cytotoxic

and inflammatory. However, IC50 value of the conjugate nanoparticles for 48h exposure was

determined to be 2 mg/mL, which indicted that, although more toxic than other samples,

they still had a low level of toxicity. On the other hand, IL-8 release caused by conjugate

nanoparticles were 2230±251 and 2957±150 pg/mL at 1 and 2 mg/mL concentrations which

were 3 to 4 times higher than the basal level of IL-8 release (768±76 pg/mL) and so raises

some concern about their safety for respiratory delivery. However, there was no significant

induction of IL-8 at 0.5 mg/mL concentration. So, the nanoparticles could still be safe

enough and useful for potent drugs with small dose sizes. For drugs like DH with bulky dose

sizes, the idea of replacing part of the polymer with a bulking agent like lactose or mannitol

is worth consideration.

The trans-epithelial permeability study was performed using sodium fluorescein as a fluid

phase marker. The results indicated that chitosan and its nanoparticles do not cause any

significant change in the permeability, the conjugate showed mild but significant increase in

the permeability while the conjugate nanoparticles rather caused a reduction in the

permeability.

8.2 Overall Conclusion This research showed successful conjugation of L-leucine to chitosan, giving a new

compound with improved solubility and thus paving a way for better utilization of chitosan.

The work established a methodology for preparing nanoparticles of both chitosan and its

conjugate of sizes less than 100 nm indicating their suitability for deposition to the deeper

regions of the respiratory airways. The conjugate nanoparticles showed higher drug loading,

better controlled release profile and higher dispersibility suggesting their strong promise as

a controlled release DPI formulation. However, they appeared to be more toxic and

inflammatory than chitosan nanoparticles. So, in consideration of the large dose size of the

model drug diltiazem hydrochloride, further studies are required to increase drug loading,

release and dispersion from a DPI in order to reduce the required lung deposition of the

nanoparticle formulation. However, the present level of drug loading, release and toxicity

could still be fine for the delivery of potent drugs with small dose sizes.

Page 210: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

159

8.3 Limitation of this Study One limitation of this work was that the conclusions drawn about the safety of the

nanoparticle formulations for respiratory delivery were based on in vitro studies performed

only on blank nanoparticles prepared by emulsion- solvent evaporation method. The

responses could be different upon treating the cells with drug-loaded nanoparticles.

Besides, the nanoparticles prepared by emulsion- glutaraldehyde technique may also

behave differently.

Secondly, the inferences drawn on the efficacy and safety of the prepared nanoparticle

formulations of chitosan and its synthetic conjugate were all based on in vitro experiments.

In vivo studies are required to prove their potential in a biological system.

8.4 Future Directions The success of this effort to synthesize a chitosan-L-leucine conjugate and the promise

shown by the derivative in terms of providing controlled release of drug and enhancing

dispersion from a DPI warrants further studies to be carried out based on this research that

could be expected to advance and enrich our current understanding and could result in an

improved performance of the developed formulation. In particular, there is need of

well-designed efforts for further increasing drug loading, release and aerosolization in order

to reduce the amount of polymer needed in a given dose. Following are some suggestions in

this line that might be worthy of endeavour and consideration.

8.4.1 Synthesis of an L-Leucine Conjugate at C-6 of Chitosan or a Bis-Conjugate at C-2 and C-6

This work reported selective conjugation of L-leucine at C-2 of chitosan. Conjugation of

L-leucine selectively at C-6 or simultaneously both at C-2 and C-6 might come up with

derivatives having relatively different physicochemical and biological attributes. This may

open new opportunities to improve the solubility, dispersibility and drug release and toxicity

profiles and minimize the limitations of the conjugate already synthesized.

Page 211: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

160

Proposed below are two schemes (Figs. 8-1 & 8-2), based on this work and related literature

reports (Li et al., 2003; Satoh et al., 2006), which could be taken as guidelines for performing

L-leucine conjugation selectively at C-2 or simultaneously both at C-2 and C-6:

8.4.2 Conjugation of Chitosan with other Amino Acids In addition to L-leucine, other amino acids have also been investigated by researchers for

improving dispersion of micronized particles from a DPI with varying degrees of success

(Chew et al., 2005; Li et al., 2005). In nature, there are more than 100 amino acids, all of

which have the common structural features comprising amine (-NH2) and carboxylic acid

(-COOH) functional groups, but they widely vary in the nature of the side chains giving each

amino acid its own peculiarity in terms of physicochemical properties. Phenylalanine

(non-polar aromatic side-chain), L-serine (polar uncharged side-chain), L-lysine (positively

charged side-chain) and L-glutamate (negatively charged side-chain) are a few examples, for

illustration (Fig. 8-3). It would be an interesting idea to conjugate a range of different types

of amino acids to chitosan and assess their potential in increasing solubility, dispersibility

and sustaining drug release from a controlled release DPI formulation.

Page 212: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

161

NMPr.t., 12h

OOH

NHAcHOO

HONH2

O

OH

O n123

45

6

123

4 56

OO

O

Chitosan

DMF/130 °C/Ar8h

OOH

NHAcHOO

HON

O

OH

O n

OO

N-Phthaloyl-chitosan

OOH

NHAcHOO

HON

O

X

O n

OO

6-halo-6-deoxy-N-phthaloyl-chitosan

NMP80 °C, 4h

NaN3

N-halosuccinimide,Triphenylphosphine

NMP80 °C, 2h

OOH

NHAcHOO

HON

O

N3

O n

OO

6-azido-6-deoxy-N-phthaloyl-chitosan

Triphenylphosphine

OOH

NHAcHOO

HON

O

NH2

O n

OO

6-amino-6-deoxy-N-phthaloyl-chitosan

O NHN

OO

O

Pyridine/Ar0 °C, 3hr.t., 21h

Boc-leu-OSu

OOH

NHAcHOO

HON

O

NH

O n

OO

6-N-Boc-L-leucine-6-deoxy-N-phthaloyl-chitosan

OHNO

O

NH2NH2.H2O NMP/H2O (1:1), 100 °C, 4h

OOH

NHAcHOO

HONH2

O

NH

O n123

45

6

6-N-Boc-L-leucine-6-deoxy-chitosan

OHNO

O

5-8M HCl r.t., 24h

OOH

NHAcHOO

HONH2

O

NH

O n

OClH.H2N

6-N-L-leucine-6-deoxy-chitosan

X= Cl/ Br/ I

O

O

Figure 8- 1: Selective Conjugation of L-Leucine to 6-OH of Chitosan

[Ac = ─COCH3, DMF = N,N-Dimethylformamide, NMP = N-methyl-2-pyrrolidone]

Page 213: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

162

OOH

NHAcHOO

HON

O

N3

O n123

45

6

123

4 56OO

6-azido-6-deoxy-N-phthaloyl-chitosan

(i) Triphenylphosphine, NMP, r.t., 12h(ii) NH2NH2.H2O, NMP/Water (1:1), 100 °C, 4h

OOH

NHAcHOO

HONH2

O

NH2

O n1

6-amino-6-deoxy-chitosan

O NHN

O O

O

Pyridine/Ar0 °C, 3hr.t., 21h

Boc-leu-OSu

OOH

NHAcHOO

HOO

NHO n

2,6-Di-N-Boc-L-leucine-6-deoxy-chitosan

NH

NH

OOO

OHNO

O

5-8M HCl, r.t., 24h

OOH

NHAcHOO

HOO

NHO n

2,6-Di-N-L-leucine-6-deoxy-chitosan.HCl

NH

NH2.HClO

OClH.H2N

O

O

Figure 8- 2: Simultaneous conjugation of leucine on C-2 and C-6 of chitosan

[Ac = ─COCH3, NMP = N-methyl-2-pyrrolidone]

Page 214: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

163

O-

NH3+

O

L-Phenylalanine

(Non-polar aromatic side chain)

HO O-

NH3+

O

L-Serine

(Polar un-charged side chain)

+H3NO-

NH3+

O

L-Lysine

(Positively charged side chain)

-O O-

O O

NH3+

L-Glutamate

(Negatively charged side chain)

Figure 8- 3: A few amino acids with different types of side chains

8.4.3 Loading the Drug into Blank Nanoparticles This study attempted to load the drug into the nanoparticles by mixing the drug with the

polymer in the aqueous phase prior to emulsification. As noted before, the emulsion-

solvent evaporation technique failed to load the model drug, DH due to precipitation of the

drug out of the particles into the external phase as needle like crystals at the high

temperature condition (60 °C) applied for hardening the particles. A number of investigators

have previously reported loading of other drugs (e.g. hydroxyurea, rifampicin, centchroman,

Bovine serum albumin (BSA), diphtheria toxoid (DT) and insulin) into chitosan particles by

passive absorption from aqueous solution (Fig. 8-4) (Gupta & Jabrail, 2006, 2007a, 2007b;

Jameela et al., 1994b; Ubaidulla et al., 2007). Considering the advantage of avoiding the use

of a possibly irritant agent like glutaraldehyde offered by the solvent evaporation technique

and the novelty of making the particles of nanometre size by this technique, the passive

absorption approach is worth giving a try. Additionally, by applying this approach, the drug

could be saved from exposure to elevated temperature that could be a source of instability

for many drugs. Moreover, it also gives freedom of raising the temperature to a higher level

that may help in more rapid solidification of the particles and may also add to their

compactness. The possible destabilization of the model drug diltiazem hydrochloride was

Page 215: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

164

the consideration behind not raising the temperature above 60 °C in this study. A

comparative study could also be performed by loading diltiazem hydrochloride by passive

absorption into blank particles made by glutaraldehyde crosslinking. This approach could

also be attempted for loading other drugs, especially those susceptible to high temperature

(such as peptides and proteins), into blank nanoparticles made by either method.

Figure 8- 4: Loading drug into blank chitosan nanoparticles by passive absorption from an aqueous solution

8.4.4 Emulsion- Solvent Evaporation Technique for Direct Loading of other Drugs Although the model drug (DH) precipitated out of the nanoparticles during the hardening

phase in the emulsion- solvent evaporation method, the principles of this method might still

be useful for generating chitosan nanoparticles loaded with other drugs (e.g. salbutamol

sulphate, morphine etc.). Experiments with different drugs could also be expected to give a

better understanding of the factors affecting drug loading into the particles by this method.

8.4.5 Aerodynamic Particle Sizing of the Aerosol Plume Because of their extremely high surface area and associated surface free energy, it is not

unusual that nanoparticles would fail to break their agglomerates when they are subjected

to dispersive forces during aerosolization. Many of these agglomerates are ultimately

deposited in the upper airways and in turn reduce the fraction of the delivered dose

Page 216: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

165

reaching to the lungs. Zetasizer analysis of both the blank and drug-loaded chitosan and

conjugate nanoparticles showed that the particles underwent agglomeration giving one or

more additional peaks beyond the original size range (Fig. 6-2 in Chapter 6). The FPF

obtained for the particles (15-24%) also indicated formation of particle agglomerates.

Although zetasizer analysis gave a rough estimate of the agglomeration tendency of the

nanoaparticles, a more conclusive idea of the extent of agglomeration of the particles could

be obtained by aerodynamic particle sizing of the aerosol plume using instruments like

Andersen cascade impactor, multi-stage liquid impinger (MSLI), time-of-flight (TOF) aerosol

analysers or laser diffraction based aerosol spray particle sizer (e.g. Aerotrac or SprayTech).

8.4.6 Mixing of Nanoparticles with Interactive Carrier Particles It is a common practice in DPI technology to mix the micronized drug particles with larger

particles of a carrier substance such as lactose or mannitol to reduce the cohesive attraction

between the drug particles that in turn reduce the agglomeration tendency of the particles

and improve their dispersion. In this study, both chitosan and conjugate nanoparticles

showed a higher dispersion when mixed with the lactose microcarrier (Inhalac® 120) at a 5%

concentration of nanoparticles (Fig. 6-6.2(C). Mixing of the nanoparticles with large carriers

at different ratios might come up with an appropriate combination with better lung

deposition. In addition, other types of carriers including mannitol or sorbitol could also be

investigated for searching out a better combination.

8.4.7 Incorporation of Lactose, Mannitol or Similar Substances into the Nanoparticles as a Component of the Matrix

In addition to adding lactose or mannitol in form of large interactive carrier particles to

reduce inter-particle attraction of micronized particles, the literature also reports their

incorporation into DPI micro-/ nanoparticles as a component of their matrix. Such

incorporation can help to improve the particle characteristics in a number of ways. First,

they can serve as a bulking agent and in turn can minimize the amount of total polymer to

be used. Secondly, their incorporation into the particle matrix has also been shown to help

increase the rate and extent of drug release from the particles by increasing their

hydrophilicity (Abd El-Hameed & Kellaway, 1997; Jain et al., 2000; Li et al., 2009). Thirdly,

Page 217: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

166

this increased hydrophilicity could be expected to increase in turn the loading of the model

drug, DH which is a highly water soluble substance. Finally, Incorporation of mannitol into

particles has also been reported to improve aerosolization of the particles from a DPI (Zhao

et al., 2008). All these factors can combine together to ultimately reduce the dose size of

the nanoparticle formulation. This is important in consideration of the toxicity profile of the

conjugate nanoparticles observed in this study and the large dose size required for

delivering the model drug, DH. So, it could be a good idea to investigate the impact of

incorporation of these agents (in different percentages) as a component of the chitosan and

conjugate nanoparticles on their drug loading, release and aerosolization.

8.4.8 Incorporation of Antiadherents into the Nanoparticles Besides interactive carrier particles like lactose or mannitol, another popular approach for

improving aerosolization of the prepared nanoparticles is incorporation of a small

percentage of an antiadherent substance, such as L-leucine, PEG 6000 or magnesium

stearate as a component of the particle formulations (Ely et al., 2007; Learoyd et al., 2009;

Lim & Wan, 1998). This sort of agents has widely been reported to reduce inter-particle

interaction and increase aerosolization of micronized particles from a DPI, often by

depositing on the particle surface and in turn reducing the cohesive force between the

particles. Therefore, it would make an interesting study to investigate the effect of mixing or

incorporating these agents on the aerosolization of prepared chitosan and conjugate

nanoparticles.

8.4.9 Stability Study of the Nanoparticles It is important to ensure that the nanoparticle formulations would be able to maintain their

physicochemical properties such as size and morphology, flowability and dispersibility, drug

release behaviour during the storage period. It is also important to ensure the

physicochemical stability of the incorporated drug. Besides, considerations also should be

given to the fact that the formulations might be exposed to various extreme conditions of

temperature and humidity during this period. A well designed stability study is therefore

intended to predict stability of the formulations. Roughly, the formulations could be stored

for a period of 3 to 6 months at different storage conditions such as room temperature

Page 218: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Chapter 8 Overall Conclusions and Further Directions

167

(25 °C), refrigerator (4-8 °C) and accelerated temperature and humidity (40 °C/ 75% RH) and

samples could be assessed for various characteristics as noted above at intervals (say, 0, 1, 3

and 6 months).

Page 219: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 220: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 221: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 222: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

168

Abd El-Hameed, M. D., & Kellaway, I. W. (1997). Preparation and in vitro characterization of mucoadhesive polymeric microspheres as intra-nasal delivery systems. Eur. J. Pharm. Biopharm., 44(1), 53-60.

Adamson, P. C., Balis, F. M., Arndt, C. A., Holcenberg, J. S., Narang, P. K., Murphy, R. F.,

Gillespie, A. J., Poplack, D. G. (1991). Intrathecal 6-mercaptopurine: preclinical pharmacology, phase I/II trial, and pharmacokinetic study. Cancer Res., 51(22), 6079-6083.

Agnihotri, S. A., & Aminabhavi, T. M. (2004). Controlled release of clozapine through

chitosan microparticles prepared by a novel method. J. Controlled Release, 96(2), 245-259.

Agnihotri, S. A., Mallikarjuna, N. N., & Aminabhavi, T. M. (2004). Recent advances on

chitosan-based micro- and nanoparticles in drug delivery. J. Controlled Release 100(1), 5-28.

Agu, R. U., Ugwoke, M. I., Armand, M., Kinget, R., & Verbeke, N. (2001). The lung as a route

for systemic delivery of therapeutic proteins and peptides. Respir. Res., 2(4), 198-209.

Aguiar, M. M. G., Rodrigues, J. M., & Silva Cunha, A. (2004). Encapsulation of insulin-

cyclodextrin complex in PLGA microspheres: a new approach for prolonged pulmonary insulin delivery. J. Microencapsulation, 21(5), 553-564.

Ahsan, F., Rivas, I. P., Khan, M. A., & Torres Suarez, A. I. (2002). Targeting to macrophages:

role of physicochemical properties of particulate carriers-liposomes and microspheres-on the phagocytosis by macrophages. J. Controlled Release, 79(1-3), 29-40.

Aiedeh, K., & Taha, M. O. (2001). Synthesis of iron-crosslinked chitosan succinate and iron-

crosslinked hydroxamated chitosan succinate and their in vitro evaluation as potential matrix materials for oral theophylline sustained-release beads. Eur. J. Pharm. Sci., 13(2), 159-168.

Al-Qadi, S., Grenha, A., Carrion-Recio, D., Seijo, B., & Remunan-Lopez, C. (2012).

Microencapsulated chitosan nanoparticles for pulmonary protein delivery: In vivo evaluation of insulin-loaded formulations. J. Controlled Release, 157(3), 383-390.

Page 223: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

169

Alhusban, F. A., & Seville, P. C. (2009). Carbomer-modified spray-dried respirable powders for pulmonary delivery of salbutamol sulphate. J. Microencapsulation, 26(5), 444-455.

Altounyan, R. E. C. (1967). Inhibition of experimental asthma by a new compound sodium

cromoglycate. Intal. Acta Allergol., 22, 487–489.

Alway, B., Sangchantra, R., & Stewart, P. J. (1996). Modeling the dissolution of diazepam in

lactose interactive mixtures. Int. J. Pharm., 130(2), 213-224.

Amidi, M., Krudys, K. M., Snel, C. J., Crommelin, D. J. A., Della Pasqua, O. E., Hennink, W. E.,

Jiskoot, W. (2008a). Efficacy of pulmonary insulin delivery in diabetic rats: Use of a model-based approach in the evaluation of insulin powder formulations. J. Controlled Release, 127(3), 257-266.

Amidi, M., Pellikaan, H. C., de Boer, A. H., Crommelin, D. J. A., Hennink, W. E., Jiskoot, W.

(2008b). Preparation and physicochemical characterization of supercritically dried insulin-loaded microparticles for pulmonary delivery. Eur. J. Pharm. Biopharm., 68(2), 191-200.

Amidi, M., Romeijn, S. G., Borchard, G., Junginger, H. E., Hennink, W. E., Jiskoot, W. (2006).

Preparation and characterization of protein-loaded N-trimethyl chitosan nanoparticles as nasal delivery system. J. Controlled Release, 111(1-2), 107-116.

Andrade, F., Goycoolea, F., Chiappetta, D. A., das Neves, J., Sosnik, A., Sarmento, B. (2011).

Chitosan-grafted copolymers and chitosan-ligand conjugates as matrices for pulmonary drug delivery. Int. J. Carbohydr. Chem., Article ID 865704, 14 pp.

Armiji, M. M., & Patel, V. R. (1994). Chitosan-poly(ethylene oxide) semi-IPNs as a pH-

sensitive drug delivery system. Polym. Prepr. (Am. Chem. Soc., Div. Polym. Chem.), 35(2), 403-404.

Artursson, P., Lindmark, T., Davis, S. S., & Illum, L. (1994). Effect of chitosan on the

permeability of monolayers of intestinal epithelial cells (Caco-2). Pharm. Res., 11(9), 1358-1361.

Asada, M., Takahashi, H., Okamoto, H., Tanino, H., & Danjo, K. (2004). Theophylline particle

design using chitosan by the spray drying. Int. J. Pharm., 270(1-2), 167-174.

Page 224: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

170

Bai, S., Gupta, V., & Ahsan, F. (2010). Inhalable Lactose-Based Dry Powder Formulations of Low Molecular Weight Heparin. J. Aerosol Med. Pulm. Drug Delivery, 23(2), 97-104.

Bansal, V., Sharma, P. K., Sharma, N., Pal, O. P., & Malviya, R. (2011). Applications of

Chitosan and Chitosan derivatives in drug delivery. Adv. Biol. Res., 5(1), 28-37.

Barakat, N. S., & Almurshedi, A. S. (2011). Preparation and characterization of chitosan

microparticles for oral sustained delivery of gliclazide: in vitro/in vivo evaluation. Drug Dev. Res., 72(3), 235-246.

Becher, R., Hetland, R. B., Refsnes, M., Dahl, J. E., Dahlman, H. J., Schwarze, P. E. (2001). Rat

lung inflammatory responses after in vivo and in vitro exposure to various stone particles. Inhalation Toxicol., 13(9), 789-805.

Beck-Broichsitter, M., Gauss, J., Gessler, T., Seeger, W., Kissel, T., Schmehl, T. (2010).

Pulmonary Targeting with Biodegradable Salbutamol-Loaded Nanoparticles. J. Aerosol Med. Pulm. Drug Delivery, 23(1), 47-57.

Begat, P., Morton, D. A. V., Shur, J., Kippax, P., Staniforth, J. N., Price, R. (2009). The role of

force control agents in high-dose dry powder inhaler formulations. J. Pharm. Sci., 98(8), 2770-2783.

Begat, P., Price, R., Harris, H., Morton, D. A. V., & Staniforth, J. N. (2005). The influence of

force control agents on the cohesive-adhesive balance in dry powder inhaler formulations. Kona, 23, 109-121.

Ben-Baruch, A., Michiel, D. F., & Oppenheim, J. J. (1995). Signals and receptors involved in

recruitment of inflammatory cells. J. Biol. Chem., 270(20), 11703-11706.

Berard, V., Lesniewska, E., Andres, C., Pertuy, D., Laroche, C., Pourcelot, Y. (2002). Affinity

scale between a carrier and a drug in DPI studied by atomic force microscopy. Int. J. Pharm., 247(1-2), 127-137.

Bergmann, M., & Zervas, L. (1932). Synthesis of glucopeptides of d-glucosamine (N-glycyl-d-

glucosamine and N-d-alanyl-d-glucosamine). Ber. Dtsch. Chem. Ges. B, 65B, 1201-1205.

Berkland, C., Kim, K., & Pack, D. W. (2001). Fabrication of PLG microspheres with precisely

controlled and monodisperse size distributions. J. Controlled Release, 73(1), 59-74.

Page 225: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

171

Bernet, B., Xu, J., & Vasella, A. (2000). Oligosaccharide analogs of polysaccharides part 20-NMR analysis of templated cellodextrins possessing two parallel chains: a mimic for cellulose I? Helv. Chim. Acta, 83(9), 2072-2114.

Berridge, M. V., Herst, P. M., & Tan, A. S. (2005). Tetrazolium dyes as tools in cell biology:

new insights into their cellular reduction. Biotechnol. Annu. Rev., 11, 127-152.

Berridge, M. V., & Tan, A. S. (1993). Characterization of the cellular reduction of 3-(4,5-

dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT): Subcellular localization, substrate dependence, and involvement of mitochondrial electron transport in MTT reduction. Arch. Biochem. Biophys., 303(2), 474-482.

Bi, R., Shao, W., Wang, Q., & Zhang, N. (2009). Solid lipid nanoparticles as insulin inhalation

carriers for enhanced pulmonary delivery. J. Biomed. Nanotechnol., 5(1), 84-92.

Bisgaard, H., O'Callaghan, C., Smaldone, G. C. (Eds.). (2002). Drug Delivery to the Lung. [In:

Lung Biol. Health Dis., 2002; 162].

Bivas-Benita, M., Romeijn, S., Junginger, H. E., & Borchard, G. (2004). PLGA-PEI nanoparticles

for gene delivery to pulmonary epithelium. Eur. J. Pharm. Biopharm., 58(1), 1-6.

Black, S. D., & Mould, D. R. (1991). Development of hydrophobicity parameters to analyze

proteins which bear post- or cotranslational modifications. Anal. Biochem., 193(1), 72-82.

Blizzard, C. D., Chipman, J. J., Cutler, G. B., Jr., Jackson, B., Johnston, L., Lucas, R. A.,

Mintzes, J. (2003). Method for administration of growth hormone via pulmonary delivery for the treatment of growth disorder (pp. 20 pp). PCT Int. Appl.: WO 2003079991 A2 20031002.

Bobu, E., Nicu, R., Lupei, M., Ciolacu, F., & Desbrieres, J. (2011). Synthesis and

characterization of n-alkyl chitosan for papermaking applications Cell. Chem. Technol., 45(9-10), 619-625.

Bonferoni, M. C., Sandri, G., Rossi, S., Ferrari, F., Gibin, S., Caramella, C. (2008). Chitosan

citrate as multifunctional polymer for vaginal delivery. Evaluation of penetration enhancement and peptidase inhibition properties. Eur. J. Pharm. Sci., 33(2), 166-176.

Page 226: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

172

Boonyo, W., Junginger, H. E., Waranuch, N., Polnok, A., & Pitaksuteepong, T. (2007). Chitosan and trimethyl chitosan chloride (TMC) as adjuvants for inducing immune responses to ovalbumin in mice following nasal administration. J. Controlled Release 121(3), 168-175.

Boonyo, W., Junginger, H. E., Waranuch, N., Polnok, A., & Pitaksuteepong, T. (2008).

Preparation and characterization of particles from chitosan with different molecular weights and their trimethyl chitosan derivatives for nasal immunization. J. Met., Mater. Miner. , 18(2), 59-65.

Borchard, G., Luessen, H. L., de Boer, A. G., Verhoef, J. C., Lehr, C.-M., Junginger, H. E.

(1996). The potential of mucoadhesive polymers in enhancing intestinal peptide drug absorption. III: Effects of chitosan-glutamate and carbomer on epithelial tight junctions in vitro. J. Controlled Release, 39(2,3), 131-138.

Borm, P. J. A. (2002). Particle toxicology: from coal mining to nanotechnology. Inhalation

Toxicol., 14(3), 311-324.

Bosquillon, C., Preat, V., & Vanbever, R. (2004). Pulmonary delivery of growth hormone

using dry powders and visualization of its local fate in rats. J. Controlled Release, 96(2), 233-244.

Braga, V. M. L., de Melo, S. J., Srivastava, R. M., & Falcao, E. P. d. S. (2004 ). Synthesis of new

1,2,4-oxadiazoles carrying (1'S,2'S)-t-butyloxycarbonyl-1-amino-2-methyl-1-butyl and (1'S)-t-butyloxycarbonyl-1'-amino-1'-ethyl groups at C-5 J. Braz. Chem. Soc., 15(4), 603-607.

British Pharmacopoeia (2013). Appendix XII C7 Aerodynamic Assessment of Fine Particles -

Fine Particle Dose and Particle Size Distribution. London: The Stationary Office

Bryan, L., Rungvejhavuttivittaya, Y., & Stewart, P. J. (1979). Mixing and demixing of

microdose quantities of sodium salicylate in a direct compression vehicle. Powder Technol. , 22(2), 147-151.

Buckley, M. M. T., Grant, S. M., Goa, K. L., McTavish, D., & Sorkin, E. M. (1990). Diltiazem. A

reappraisal of its pharmacological properties and therapeutic use. Drugs, 39(5), 757-806.

Budhian, A., Siegel, S. J., & Winey, K. I. (2005). Production of haloperidol-loaded PLGA

nanoparticles for extended controlled drug release of haloperidol. J. Microencapsulation, 22(7), 773-785.

Page 227: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

173

Burch, S. G., Erbland, M. L., Gann, L. P., & Hiller, F. C. (1989). Plasma nicotine levels after inhalation of aerosolized nicotine. Am. Rev. Respir. Dis., 140(4), 955-957.

Byron, P. R. (1986). Some future perspectives for unit dose inhalation aerosols. Drug Dev.

Ind. Pharm. , 12(7), 993-1015.

Byron, P. R., Roberts, N. S. R., & Clark, A. R. (1986). An isolated perfused rat lung preparation

for the study of aerosolized drug deposition and absorption. J. Pharm. Sci. , 75(2), 168-171.

Cacalano, G., Lee, J., Kikly, K., Ryan, A. M., Pitts-Meek, S., Hultgren, B., Wood, W. I.,

Moore, M. W. (1994). Neutrophil and B cell expansion in mice that lack the murine IL-8 receptor homolog. Science (Washington, D. C.), 265(5172), 682-684.

Chan, H.-K., Clark, A., Gonda, I., Mumenthaler, M., & Hsu, C. (1997). Spray dried powders

and powder blends of recombinant human deoxyribonuclease (rhDNase) for aerosol delivery. Pharm. Res., 14(4), 431-497.

Chen, F., Zhang, Z.-R., Yuan, F., Qin, X., Wang, M., Huang, Y. (2008). In vitro and in vivo study

of N-trimethyl chitosan nanoparticles for oral protein delivery. Int. J. Pharm., 349(1-2), 226-233.

Chew, N. Y. K., Shekunov, B. Y., Tong, H. H. Y., Chow, A. H. L., Savage, C., Wu, J., Chan, H.-K.

(2005). Effect of amino acids on the dispersion of disodium cromoglycate powders. J. Pharm. Sci., 94(10), 2289-2300.

Chimote, G., & Banerjee, R. (2009). Evaluation of antitubercular drug-loaded surfactants as

inhalable drug-delivery systems for pulmonary tuberculosis. J. Biomed. Mater. Res., Part A, 89A(2), 281-292.

Chiou, H., Li, L., Hu, T., Chan, H.-K., Chen, J.-F., Yun, J. (2007). Production of salbutamol

sulfate for inhalation by high-gravity controlled antisolvent precipitation. Int. J. Pharm. , 331(1), 93-98.

Choi, M., Cho, M., Han, B. S., Hong, J., Jeong, J., Park, S., Cho, M.-H., Kim, K., Cho, W.-S.

(2010). Chitosan nanoparticles show rapid extrapulmonary tissue distribution and excretion with mild pulmonary inflammation to mice. Toxicol. Lett., 199(2), 144-152.

Page 228: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

174

Choi, Y. J., Kim, E. J., Piao, Z., Yun, Y. C., & Shin, Y. C. (2004). Purification and characterization of chitosanase from Bacillus sp. strain KCTC 0377BP and its application for the production of chitosan oligosaccharides. Appl. Environ. Microbiol., 70(8), 4522-4531.

Choksakulnimitr, S., Masuda, S., Tokuda, H., Takakura, Y., & Hashida, M. (1995). In vitro

cytotoxicity of macromolecules in different cell culture systems. J. Controlled Release, 34(3, br273), 233-241.

Chougule, M. B., Padhi, B. K., Jinturkar, K. A., & Misra, A. (2007). Development of dry

powder inhalers. Recent Pat. Drug Deliv. Formul., 1(1), 11-21.

Chow, A. H. L., Tong, H. H. Y., Chattopadhyay, P., & Shekunov, B. Y. (2007). Particle

Engineering for Pulmonary Drug Delivery. Pharm. Res., 24(3), 411-437.

Churg, A., & Brauer, M. (2000). Ambient atmospheric particles in the airways of human

lungs. Ultrastruct. Pathol., 24(6), 353-361.

Clark, A. R., & Bailey, R. (1996). In: P. R. Byron, R. N. Dalby & S. J. Farr (Eds.), Respiratory

Drug Delivery V (pp. 221-230). Buffalo Grove, IL: Interpharm Press, Inc. .

Coco, R., Plapied, L., Pourcelle, V., Jerome, C., Brayden, D. J., Schneider, Y.-J., Preat, V.

(2013). Drug delivery to inflamed colon by nanoparticles: Comparison of different strategies. Int. J. Pharm. (Amsterdam, Neth.), 440(1), 3-12.

Codrons, V., Vanderbist, F., Ucakar, B., Preat, V., & Vanbever, R. (2004). Impact of

formulation and methods of pulmonary delivery on absorption of parathyroid hormone (1-34) from rat lungs. J. Pharm. Sci., 93(5), 1241-1252.

Columbano, A., Buckton, G., & Wikeley, P. (2003). Characterization of surface modified

salbutamol sulphate-alkylpolyglycoside microparticles prepared by spray drying. Int. J. Pharm., 253(1-2), 61-70.

Coowanitwong, I., Arya, V., Patel, G., Kim, W.-S., Craciun, V., Rocca, J. R., Singh, R.,

Hochhaus, G. (2007). Laser-ablated nanofunctional polymers for the formulation of slow-release powders for dry powder inhalers: physicochemical characterization and slow-release characteristics. J. Pharm. Pharmacol., 59(11), 1473-1484.

Cox, P. J., Khan, K. A., Munday, D. L., & Sujja-areevath, J. (1999). Development and

evaluation of a multiple-unit oral sustained release dosage form for S(+)-ibuprofen: preparation and release kinetics. Int. J. Pharm., 193(1), 73-84.

Page 229: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

175

Crini, G., Torri, G., Guerrini, M., Morcellet, M., Weltrowski, M., Martel, B. (1997). NMR characterization of N-benzyl sulfonated derivatives of chitosan. Carbohydr. Polym., 33(2/3), 145-151.

Damms, B., & Bains, W. (1995). The cost of delivering drugs without needles: noninvasive

methods may be more convenient, but are they economically practical? Bio/Technology, 13(13), 1438-1440.

Dandge, B. H., & Dehghan, M. H. G. (2009). Formulation and evaluation of nasal

mucoadhesive microparticles of diltiazem hydrochloride. Int. J. ChemTech Res. , 1(4), 1036-1042.

Davila, J. C., Reddy, C. G., Davis, P. J., & Acosta, D. (1990). Toxicity assessment of papaverine

hydrochloride and papaverine-derived metabolites in primary cultures of rat hepatocytes. In Vitro Cell. Dev. Biol., 26(5), 515-524.

Davis, S. S. (1999). Delivery of peptide and non-peptide drugs through the respiratory tract.

Pharm. Sci. Technol. Today, 2(11), 450-456.

De Jesus Valle, M. J., Dinis-Oliveira, R. J., Carvalho, F., Bastos, M. L., & Navarro, A. S. (2008).

Toxicological evaluation of lactose and chitosan delivered by inhalation. J. Biomater. Sci., Polym. Ed., 19(3), 387-397.

Dellaria, J. F., Jr., Maki, R. G., Stein, H. H., Cohen, J., Whittern, D., Marsh, K., Hoffman, D. J.,

Plattner, J. J., Perun, T. J. (1990). New inhibitors of renin that contain novel phosphostatine Leu-Val replacements. J. Med. Chem., 33(2), 534-542.

DeLuca, P. P., Mehta, R. C., Hausberger, A. G., & Thanoo, B. C. (1993). Biodegradable

Polyesters for Drug and Polypeptide Delivery. In M. A. El-Aly, D. M. Piatt & B. A. Charpentier (Eds.), Polymeric Delivery Systems, Properties and Applications (pp. 53–79). Washington, DC: American Chemical Society.

Denizot, F., & Lang, R. (1986). Rapid colorimetric assay for cell growth and survival.

Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J. Immunol. Methods, 89(2), 271-277.

Denkbas, E. B., & Odabasi, M. (2000). Chitosan microspheres and sponges: preparation and

characterization. J. Appl. Polym. Sci., 76(11), 1637-1643.

Page 230: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

176

Denkbas, E. B., & Ottenbrite, R. M. (2006). Perspectives on: chitosan drug delivery systems based on their geometries. J. Bioact. Compat. Pol., 21(4), 351-368.

Denkbas, E. B., Seyyal, M., & Piskin, E. (1999). 5-Fluorouracil loaded chitosan microspheres

for chemoembolization. J. Microencapsulation, 16(6), 741-749.

Dershwitz, M., Walsh, J. L., Morishige, R. J., Connors, P. M., Rubsamen, R. M., Shafer, S. L.,

Rosow, C. E. (2000). Pharmacokinetics and pharmacodynamics of inhaled versus intravenous morphine in healthy volunteers. Anesthesiology, 93(3), 619-628.

Desai, S. S., Aher, A. A., & Kadaskar, P. T. (2013). Methods for reduction of cohesive forces

between carrier and drug in DPI formulation. Drug Dev. Ind. Pharm., 39(11), 1589-1598.

Deveswaran, R., Basavaraj, B. V., Bharath, S., Abraham, S., & Madhavan, V. (2007).

Diltiazem-loaded chitosan microspheres - a long acting controlled delivery system. Int. J. Chem. Sci., 5(2), 874-880.

Devlin, R. B., McKinnon, K. P., Noah, T., Becker, S., & Koren, H. S. (1994). Ozone-induced

release of cytokines and fibronectin by alveolar macrophages and airway epithelial cells. Am. J. Physiol., 266(6, Pt. 1), L612-L619.

Dhawan, S., & Singla, A. K. (2003). Nifedipine loaded chitosan microspheres prepared by

emulsification phase-separation. Biotech. Histochem. , 78(5), 243-254.

Dodane, V., & Vilivalam, V. D. (1998). Pharmaceutical applications of chitosan. Pharm. Sci.

Technol. Today, 1(6), 246-253.

Dolovich, M. B., Ahrens, R. C., Hess, D. R., Anderson, P., Dhand, R., Rau, J. L., Smaldone, G.

C., Guyatt, G. (2005). Device selection and outcomes of aerosol therapy: Evidence-based guidelines: American College of Chest Physicians/American College of Asthma, Allergy, and Immunology. Chest, 127(1), 335-371.

Dornish, M., Hagen, A., Hansson, E., Pecheur, C., Verdier, F., Skaugrud, O. (1997). Safety of

Protasan: ultrapure chitosan salts for biomedical and pharmaceutical use. Adv. Chitin Sci., 2, 664-670.

Driscoll, K. E., Carter, J. M., Hassenbein, D. G., & Howard, B. (1997). Cytokines and particle-

induced inflammatory cell recruitment. Environ. Health Perspect. Suppl., 105(5), 1159-1164.

Page 231: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

177

Drumm, K., Attia, D. I., Kannt, S., Micke, P., Buhl, R., Kienast, K. (2000). Soot-exposed mononuclear cells increase inflammatory cytokine mRNA expression and protein secretion in cocultured bronchial epithelial cells. Respiration, 67(3), 291-297.

Dubey, R. R., & Parikh, R. H. (2004). Two-stage optimization process for formulation of

chitosan microspheres. AAPS PharmSciTech, 5(1), 20-28.

Duddu, S. P., Sisk, S. A., Walter, Y. H., Tarara, T. E., Trimble, K. R., Clark, A. R., Eldon, M. A.,

Elton, R. C., Pickford, M., Hirst, P. H., Newman, S. P., Weers, J. G. (2002). Improved Lung Delivery from a Passive Dry Powder Inhaler Using an Engineered PulmoSphere® Powder. Pharm. Res., 19(5), 689-695.

Dumitriu, S. (Ed.). (2002). Polymeric Biomaterials (Second Edition, Revised and Expanded

ed.): Marcel Dekker, Inc., New York.

Dumont, J. A., Bitonti, A. J., Clark, D., Evans, S., Pickford, M., Newman, S. P. (2005). Delivery

of an erythropoietin-Fc fusion protein by inhalation in humans through an immunoglobulin transport pathway. J. Aerosol Med. , 18(3), 294-303.

Dyer, A. M., Hinchcliffe, M., Watts, P., Castile, J., Jabbal-Gill, I., Nankervis, R., Smith, A., Illum,

L. (2002). Nasal Delivery of Insulin Using Novel Chitosan Based Formulations: A Comparative Study in Two Animal Models Between Simple Chitosan Formulations and Chitosan Nanoparticles. Pharm. Res., 19(7), 998-1008.

Edwards, D. A., Ben-Jebria, A., & Langer, R. (1998). Recent advances in pulmonary drug

delivery using large, porous inhaled particles. J. Appl. Physiol., 85(2), 379-385.

Edwards, D. A., Hanes, J., Caponetti, G., Hrkach, J., Ben-Jebria, A., Eskew, M. L., Mintzes, J.,

Deaver, D., Lotan, N., Langer, R. (1997). Large porous particles for pulmonary drug delivery. Science (Washington, D. C.), 276(5320), 1868-1871.

Edwards, D. A., Sung, J., Pulliam, B., Wehrenberg-Klee, E., Schwartz, E., Dreyfuss, P.,

Kulkarni, S., Lieberman, E. (2005). Pulmonary delivery of malarial vaccine in the form of particulates (pp. 25 pp). PCT Int. Appl.: WO 2005110379 A2 20051124.

Ekzemplyarov, O. N., Etingov, E. D., Kholodova, G. V., Kokushina, T. M., Kulbakh, V. O.,

Lagert, I. K., Lushitskaya, I. M., Mikhailets, G. A., Sokolov, L. B., Zelmanov, R. B. (1977). Meglumine complex fungicidal polyene macrolide antibiotic compositions and treatment method. Publication No. US4002741 A.

Page 232: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

178

El-Hefian, E. A., Yahaya, A. H., & Misran, M. (2009). Characterisation of chitosan solubilised in aqueous formic and acetic acids. Maejo Int. J. Sci. Technol., 3(3), 415-425.

El-Sherbiny, I. M., & Smyth, H. D. C. (2012). Controlled Release Pulmonary Administration of

Curcumin Using Swellable Biocompatible Microparticles. Mol. Pharmaceutics, 9(2), 269-280.

Ely, L., Roa, W., Finlay, W. H., & Loebenberg, R. (2007). Effervescent dry powder for

respiratory drug delivery. Eur. J. Pharm. Biopharm., 65(3), 346-353.

Erden, N., & Celebi, N. (1996). Factors influencing release of salbutamol sulfate from

poly(lactide-co-glycolide) microspheres prepared by water-in-oil-in-water emulsion technique. Int. J. Pharm., 137(1), 57-66.

Erger, R. A., & Casale, T. B. (1998). Interleukin-8 plays a significant role in IgE-mediated lung

inflammation. Eur. Respir. J., 11(2), 299-305.

Fiegel, J., Fu, J., & Hanes, J. (2004). Poly(ether-anhydride) dry powder aerosols for sustained

drug delivery in the lungs. J. Controlled Release, 96(3), 411-423.

Filipovic-Grcic, J., Becirevic-Lacan, M., Skalko, N., & Jalsenjak, I. (1996). Chitosan

microspheres of nifedipine and nifedipine-cyclodextrin inclusion complexes. Int. J. Pharm., 135(1,2), 183-190.

Finch, P., (Editor), (1999). Carbohydrates, Structures, Syntheses and Dynamics. Dordrecht,

The Netherlands: Kluwer Academic Publishers.

Fischer, D., Li, Y., Ahlemeyer, B., Krieglstein, J., & Kissel, T. (2003). In vitro cytotoxicity testing

of polycations: influence of polymer structure on cell viability and hemolysis. Biomaterials, 24(7), 1121-1131.

Fishbein, M. C. Smoker's Lung: Pathology Photo Essay. Retrieved September 20, 2013, from

http://www.medicinenet.com/smokers_lung_pathology_photo_essay/article.htm

Fitzgerald, K. A. (Ed.). (2001). The Cytokine: Facts Book (2, illustrated ed.): Academic Press,

London.

Page 233: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

179

Florea, B. I., Thanou, M., Junginger, H. E., & Borchard, G. (2006). Enhancement of bronchial octreotide absorption by chitosan and N-trimethyl chitosan shows linear in vitro/in vivo correlation. J. Controlled Release, 110(2), 353-361.

Forbes, B., Hashmi, N., Martin, G. P., & Lansley, A. B. (2000). Formulation of inhaled

medicines: effect of delivery vehicle on immortalized epithelial cells. J. Aerosol Med. , 13(3), 281-288.

Freemantle, N., & Strack, T. R. (2006). Will availability of inhaled human insulin (Exubera)

improve management of type 2 diabetes? The design of the Real World trial. Trials, 7, 25.

French, D. L., Edwards, D. A., & Niven, R. W. (1996). The influence of formulation on

emission, deaggregation and deposition of dry powders for inhalation. J. Aerosol Sci., 27(5), 769-783.

Fu, G., Yu, H., Yuan, Z., Liu, B., Shen, B., He, B. (2004a). Chitosan Adsorbents Carrying Amino

Acids for Selective Removal of Low Density Lipoprotein. Artif. Cells, Blood Substitutes, Biotechnol., 32(2), 303-313.

Fu, J., Fiegel, J., & Hanes, J. (2004b). Synthesis and characterization of PEG-based ether-

anhydride terpolymers: novel polymers for controlled drug delivery. Macromolecules, 37(19), 7174-7180.

Fu, J., Fiegel, J., Krauland, E., & Hanes, J. (2002). New polymeric carriers for controlled drug

delivery following inhalation or injection. Biomaterials, 23(22), 4425-4433.

Fujisawa, T., Kato, Y., Atsuta, J., Terada, A., Iguchi, K., Kamiya, H., Yamada, H., Nakajima, T.,

Miyamasu, M., Hirai, K. (2000). Chemokine production by the BEAS-2B human bronchial epithelial cells: differential regulation of eotaxin, IL-8, and RANTES by TH2- and TH1-derived cytokines. J. Allergy Clin. Immunol. , 105(1, Pt. 1), 126-133.

Gan, Q., & Wang, T. (2007). Chitosan nanoparticle as protein delivery carrier-Systematic

examination of fabrication conditions for efficient loading and release. Colloids Surf., B, 59(1), 24-34.

Gartlon, J., & Clothier, R. (2008 ). Fluorescein Leakage assay Background Review Document

as an alternative method for eye irritation testing. Ispra, Italy: European Centre for the Validation of Alternative Methods (ECVAM), In-vitro Methods Unit, Institute for Health and Consumer Protection, European Commission’s Joint Research Centre (EC-JRC), Ispra, Italy.

Page 234: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

180

Geller, D. E. (2005). Comparing clinical features of the nebulizer, metered-dose inhaler, and dry powder inhaler. Respir. Care, 50(10), 1313-1321; discussion 1321-1312.

Germershaus, O., Mao, S., Sitterberg, J., Bakowsky, U., & Kissel, T. (2008). Gene delivery

using chitosan, trimethyl chitosan or polyethylenglycol-graft-trimethyl chitosan block copolymers: Establishment of structure–activity relationships in vitro. J. Controlled Release, 125(2), 145-154.

Ghio, A. J., Silbajoris, R., Carson, J. L., & Samet, J. M. (2002). Biologic effects of oil fly ash

[Review]. Environ. Health Perspect., 110 Suppl 1, 89-94.

Gilani, K., Rouholamini Najafabadi, A., Barghi, M., & Rafiee-Tehrani, M. (2004).

Aerosolisation of beclomethasone dipropionate using spray dried lactose/polyethylene glycol carriers. Eur. J. Pharm. Biopharm., 58(3), 595-606.

Glinski, J., Chavepeyer, G., & Platten, J. K. (2000). Surface properties of aqueous solutions of

l-leucine. Biophys. Chem., 84(2), 99-103.

Godbey, W. T., Wu, K. K., & Mikos, A. G. (2001). Poly(ethylenimine)-mediated gene delivery

affects endothelial cell function and viability. Biomaterials, 22(5), 471-480.

Graness, A., Chwieralski, C. E., Reinhold, D., Thim, L., & Hoffmann, W. (2002). Protein kinase

C and ERK activation are required for TFF-peptide-stimulated bronchial epithelial cell migration and tumor necrosis factor-α-induced interleukin-6 (IL-6) and IL-8 secretion. J. Biol. Chem. , 277(21), 18440-18446.

Grasmeijer, F., Hagedoorn, P., Frijlink, H. W., & de Boer, A. H. (2013). Drug content effects

on the dispersion performance of adhesive mixtures for inhalation. PloS one, 8(8), e71339.

Gray, H. (2000). Anatomy of the human body: Lea & Febiger, Philadelphia, Bartleby.

Grenha, A., Grainger, C. I., Dailey, L. A., Seijo, B., Martin, G. P., Remunan-Lopez, C., Forbes, B.

(2007). Chitosan nanoparticles are compatible with respiratory epithelial cells in vitro. Eur. J. Pharm. Sci., 31(2), 73-84.

Grenha, A., Seijo, B., & Remunan-Lopez, C. (2005). Microencapsulated chitosan

nanoparticles for lung protein delivery. Eur. J. Pharm. Sci. , 25(4-5), 427-437.

Page 235: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

181

Gupta, K. C., & Jabrail, F. H. (2006). Glutaraldehyde and glyoxal cross-linked chitosan microspheres for controlled delivery of centchroman. Carbohydr. Res., 341(6), 744-756.

Gupta, K. C., & Jabrail, F. H. (2007a). Controlled-release formulations for hydroxy urea and

rifampicin using polyphosphate-anion-crosslinked chitosan microspheres. J. Appl. Polym. Sci., 104(3), 1942-1956.

Gupta, K. C., & Jabrail, F. H. (2007b). Glutaraldehyde cross-linked chitosan microspheres for

controlled release of centchroman. Carbohydr. Res., 342(15), 2244-2252.

Hadinoto, K., Zhu, K., & Tan, R. B. H. (2007). Drug release study of large hollow

nanoparticulate aggregates carrier particles for pulmonary delivery. Int. J. Pharm., 341(1-2), 195-206.

Hallworth, G. W., & Westmoreland, D. G. (1987). The twin impinger: a simple device for

assessing the delivery of drugs from metered dose pressurized aerosol inhalers. J. Pharm. Pharmacol., 39(12), 966-972.

Hamman, J. H., Stander, M., & Kotze, A. F. (2002). Effect of the degree of quaternization of

N-trimethyl chitosan chloride on absorption enhancement: in vivo evaluation in rat nasal epithelia. Int. J. Pharm., 232(1-2), 235-242.

Hansson, L., Hedner, T., Lund-Johansen, P., Kjeldsen, S. E., Lindholm, L. H., Syvertsen, J. O.,

Lanke, J., de Faire, U., Dahlof, B., Karlberg, B. E. (2000). Randomized trial of effects of calcium antagonists compared with diuretics and β-blockers on cardiovascular morbidity and mortality in hypertension: the Nordic Diltiazem (NORDIL) study. Lancet, 356(9227), 359-365.

Hardy, J. G., & Chadwick, T. S. (2000). Sustained release drug delivery to the lungs: an option

for the future. Clin. Pharmacokinet., 39(1), 1-4.

Harland, R. S., Gazzaniga, A., Sangalli, M. E., Colombo, P., & Peppas, N. A. (1988).

Drug/polymer matrix swelling and dissolution. Pharm. Res., 5(8), 488-494.

Heras, A., Rodriguez, N. M., Ramos, V. M., & Agullo, E. (2000). N-Methylene phosphonic

chitosan: a novel soluble derivative. Carbohydr. Polym., 44(1), 1-8.

Hermans, C., & Bernard, A. (1999). Lung epithelium-specific proteins: characteristics and

potential applications as markers. Am. J. Respir. Crit. Care Med., 159(2), 646-678.

Page 236: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

182

Hersey, J. A. (1975). Ordered mixing: a new concept in powder mixing practice. Powder Technol. , 11, 41-44.

Hetland, R. B., Cassee, F. R., Refsnes, M., Schwarze, P. E., Lag, M., Boere, A. J., Dybing, E.

(2004). Release of inflammatory cytokines, cell toxicity and apoptosis in epithelial lung cells after exposure to ambient air particles of different size fractions. Toxicol. In Vitro, 18(2), 203-212.

Hickey, A. J. (2013). Back to the future: Inhaled drug products. J. Pharm. Sci., 102(4),

1165-1172.

Hickey, A. J., Concessio, N. M., VanOort, M. M., & Platz, R. M. (1994). Factors influencing the

dispersion of dry powders as aerosols. J. Pharm. Technol., 18, 58–64.

Higuchi, T. (1963). Mechanism of sustained-action medication. Theoretical analysis of rate of

release of solid drugs dispersed in solid matrices. J. Pharm. Sci., 52(12), 1145-1149.

Hiller, F. C., Mazumder, M. K., Wilson, J. D., & Bone, R. C. (1980). Aerodynamic size

distribution, hygroscopicity, and deposition estimation of beclomethasone dipropionate aerosol. J. Pharm. Pharmacol., 32(9), 605-609.

Hirano, S., Seino, H., Akiyama, Y., & Nonaka, I. (1988). Biocompatibility of chitosan by oral

and intravenous administrations. Polym. Mater. Sci. Eng., 59, 897-901.

Hixson, A. W., & Crowell, J. H. (1931). Dependence of reaction velocity upon surface and

agitation. I. Theoretical considerations. Ind. Eng. Chem., 23, 923-931.

Hoet, P. H. M., Brueske-Hohlfeld, I., & Salata, O. V. (2004). Nanoparticles-known and

unknown health risks. J. Nanobiotechnol., 2(12), 1-15.

Holappa, J., Nevalainen, T., Savolainen, J., Soininen, P., Elomaa, M., Safin, R., Suvanto, S.,

Pakkanen, T., Masson, M., Loftsson, T., Jaervinen, T. (2004). Synthesis and Characterization of Chitosan N-Betainates Having Various Degrees of Substitution. Macromolecules, 37(8), 2784-2789.

Holappa, J., Nevalainen, T., Soininen, P., Elomaa, M., Safin, R., Masson, M., Jaervinen, T.

(2005). N-Chloroacyl-6-O-triphenylmethylchitosans: Useful Intermediates for Synthetic Modifications of Chitosan. Biomacromolecules, 6(2), 858-863.

Page 237: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

183

Holzner, P. M., & Mueller, B. W. (1995). Particle size determination of metered dose inhalers with inertial separation methods: Apparatus A and B (BP), Four Stage Impinger and Andersen Mark II Cascade Impactor. Int. J. Pharm., 116(1), 11-18.

Hu, Y., Jiang, H., Xu, C., Wang, Y., & Zhu, K. (2005). Preparation and characterization of

poly(ethylene glycol)-g-chitosan with water- and organo solubility. Carbohydr. Polym., 61(4), 472-479.

Huang, M., & Fang, Y. e. (2006). Preparation, characterization, and properties of chitosan-g-

polyvinyl alcohol copolymer. Biopolymers, 81(3), 160-166.

Huang, M., Khor, E., & Lim, L.-Y. (2004). Uptake and Cytotoxicity of Chitosan Molecules and

Nanoparticles: Effects of Molecular Weight and Degree of Deacetylation. Pharm. Res., 21(2), 344-353.

Huang, Y. C., Vieira, A., Huang, K. L., Yeh, M. K., & Chiang, C. H. (2005). Pulmonary

inflammation caused by chitosan microparticles. J. Biomed. Mater. Res., Part A, 75A(2), 283-287.

Huang, Y. C., Yeh, M. K., Cheng, S. N., & Chiang, C. H. (2003). The characteristics of

betamethasone-loaded chitosan microparticles by spray-drying method. J. Microencapsulation 20(4), 459-472.

Human IL-8 ELISA MAX™ Deluxe Kit. Manufacturer's Protocol (2011). BioLegend, Inc., San

Diego, USA (http://www.biolegend.com/media_assets/pro_detail/datasheets/ 431504.pdf).

Ibrahim, M. A., Ismail, A., Fetouh, M. I., & Goepferich, A. (2005). Stability of insulin during

the erosion of poly(lactic acid) and poly(lactic-co-glycolic acid) microspheres. J. Controlled Release 106(3), 241-252.

Ikeda, H., Harata, M., & Kurita, K. (2002). Regioselective one-step N-phthaloylation of

chitosan and some modification reactions of the product. Adv. Chitin Sci., 6, 243-244.

Illum, L. (1998). Chitosan and its use as a pharmaceutical excipient. Pharm. Res., 15(9),

1326-1331.

Illum, L. (2000). Transport of drugs from the nasal cavity to the central nervous system.

Eur. J. Pharm. Sci., 11(1), 1-18.

Page 238: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

184

Illum, L., Farraj, N., & Davis, S. S. (1994). Chitosan as a novel nasal delivery system for peptide drugs. Pharm. Res., 11(8), 1186-1184.

Illum, L., Jabbal-Gill, I., Hinchcliffe, M., Fisher, A. N., & Davis, S. S. (2001). Chitosan as a novel

nasal delivery system for vaccines. Adv. Drug Delivery Rev., 51(1-3), 81-96.

Ishii, H., Minegishi, M., Lavitpichayawong, B., & Mitani, T. (1995). Synthesis of chitosan-

amino acid conjugates and their use in heavy metal uptake. Int. J. Biol. Macromol., 17(1), 21-23.

Islam, N., & Rahman, S. (2008). Pulmonary drug delivery: Implication for new strategy for

pharmacotherapy for neurodegenerative disorders. Drug Discov. Ther., 2(5), 264-276.

Itoh, H., Nishino, M., & Hatabu, H. (2004). Architecture of the lung: morphology and

function. J. Thorac. Imaging, 19(4), 221-227.

Jain, K. K. (2008). The Handbook of Nanomedicine (Illustrated ed.). New Jersey, USA:

Humana Press.

Jain, R. A., Rhodes, C. T., Railkar, A. M., Malick, A. W., & Shah, N. H. (2000). Controlled

release of drugs from injectable in situ formed biodegradable PLGA microspheres: effect of various formulation variables. Eur. J. Pharm. Biopharm., 50(2), 257-262.

Jameela, S. R., & Jayakrishnan, A. (1995). Glutaraldehyde cross-linked chitosan microspheres

as a long acting biodegradable drug delivery vehicle: studies on the in vitro release of mitoxantrone and in vivo degradation of microspheres in rat muscle. Biomaterials, 16(10), 769-775.

Jameela, S. R., Misra, A., & Jayakrishnan, A. (1994a). Cross-linked chitosan microspheres as

carriers for prolonged delivery of macromolecular drugs. J. Biomater. Sci. Polym. Ed., 6(7), 621-632.

Jameela, S. R., Misra, A., & Jayakrishnan, A. (1994b). Crosslinked chitosan microspheres as

carriers for prolonged delivery of macromolecular drugs. J. Biomater. Sci., Polym. Ed., 6(7), 621-632.

Janoria, K. G., & Mitra, A. K. (2007). Effect of lactide/glycolide ratio on the in vitro release of

ganciclovir and its lipophilic prodrug (GCV-monobutyrate) from PLGA microspheres. Int. J. Pharm., 338(1-2), 133-141.

Page 239: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

185

Jantikar, A., Brashier, B., Maganji, M., Raghupathy, A., Mahadik, P., Gokhale, P., Gogtay, J., Salvi, S. (2007). Comparison of bronchodilator responses of levosalbutamol and salbutamol given via a pressurized metered dose inhaler: a randomized, double blind, single-dose, crossover study. Resp. Med., 101(4), 845-849.

Jaspart, S., Bertholet, P., Piel, G., Dogne, J.-M., Delattre, L., Evrard, B. (2007). Solid lipid

microparticles as a sustained release system for pulmonary drug delivery. Eur. J. Pharm. Biopharm., 65(1), 47-56.

Jayakumar, R. N., N.; Tokura, S.; Tamura, H. (2007). Sulfated chitin and chitosan as novel

biomaterials Int. J. Biol. Macromol., 40 (3), 175-181.

Jeon, Y.-J., & Kim, S.-K. (2001). Effect of antimicrobial activity by chitosan oligosaccharide

N-conjugated with asparagine. J. Microbiol. Biotechnol., 11(2), 281-286.

Ji, J., Ding, Z., & Yang, X. (2009). Preparation and properties of chitosan film as a drug

sustained- release system. Huaxi Kouqiang Yixue Zazhi, 27(3), 248-251.

Jung, J., & Perrut, M. (2001). Particle design using supercritical fluids: Literature and patent

survey. J. Supercrit. Fluids 20(3), 179-219.

Kaerger, J. S., & Price, R. (2004). Processing of Spherical Crystalline Particles via a Novel

Solution Atomization and Crystallization by Sonication (SAXS) Technique. Pharm. Res., 21(2), 372-381.

Kandasamy, R., & Chandrasekaran, K. (2013). Sustained release aerosol for pulmonary drug

delivery system: a review. Int. J. Pharm. Pharm. Sci., 5(3), 126-130.

Khan, M. S., & Vishakante, G. D. (2013). Development and evaluation of porous chitosan

nanoparticles for treatment of enterotoxigenic Escherichia coli infection. J. Biomed. Nanotechnol., 9(1), 107-114.

Kim, C. H., & Choi, K. S. (1998 ). Synthesis and properties of carboxyalkyl chitosan derivatives

J. Ind. Eng. Chem. (Seoul) 4(1), 19-25.

Kim, K. K., & Pack, D. W. (2006). Microspheres for drug delivery. BioMEMS Biomed.

Nanotechnol., 1, 19-50.

Page 240: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

186

Kimura, S., Egashira, K., Chen, L., Nakano, K., Iwata, E., Miyagawa, M., Tsujimoto, H., Hara, K., Morishita, R., Sueishi, K., Tominaga, R., Sunagawa, K. (2009). Nanoparticle-Mediated Delivery of Nuclear Factor κB Decoy Into Lungs Ameliorates Monocrotaline-Induced Pulmonary Arterial Hypertension. Hypertension, 53(5), 877-883.

Koch, A. E., Polverini, P. J., Kunkel, S. L., Harlow, L. A., DiPietro, L. A., Elner, V. M., Elner, S. G.,

Strieter, R. M. (1992). Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science (Washington, D. C., 1883-), 258(5089), 1798-1789.

Kochetkov, N. K., Derevitskaya, V. A., & Molodtsov, N. V. (1962). Gycopeptides. III. Synthesis

of N-aminoacyl derivatives of amino sugars. Zh. Obshch. Khim., 32, 2500-2505.

Kockisch, S., Rees, G. D., Tsibouklis, J., & Smart, J. D. (2005). Mucoadhesive, triclosan-loaded

polymer microspheres for application to the oral cavity: preparation and controlled release characteristics. Eur. J. Pharm. Biopharm., 59(1), 207-216.

Korsmeyer, R. W., Gurny, R., Doelker, E., Buri, P., & Peppas, N. A. (1983). Mechanisms of

solute release from porous hydrophilic polymers. Int. J. Pharm., 15(1), 25-35.

Kotze, A. F., de Leeuw, B. J., Luessen, H. L., de Boer, A. G., Verhoef, J. C., Junginger, H. E.

(1997a). Chitosans for enhanced delivery of therapeutic peptides across intestinal epithelia: in vitro evaluation in Caco-2 cell monolayers. Int. J. Pharm., 159(2), 243-253.

Kotze, A. F., Luessen, H. L., De Leeuw, B. J., De Boer, B. G., Verhoef, C. J., Junginger, H. E.

(1998a). Comparison of the effect of different chitosan salts and N-trimethyl chitosan chloride on the permeability of intestinal epithelial cells (Caco-2). J. Controlled Release 51(1), 35-46.

Kotze, A. F., Luessen, H. L., De Leeuw, B. J., De Boer, B. G., Verhoef, J. C., Junginger, H. E.

(1997b). N-Trimethyl chitosan chloride as a potential absorption enhancer across mucosal surfaces: in vitro evaluation in intestinal epithelial cells (Caco-2). Pharm. Res., 14(9), 1197-1202.

Kotze, A. F., Thanou, M., Verhoef, J. C., & Junginger, H. E. (1998b). Chitosan and N-trimethyl

chitosan chloride as absorption enhancers for nasal and rectal delivery of insulin. Proceedings of the International Symposium on Controlled Release of Bioactive Materials, 25th, 479-480.

Page 241: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

187

Koushik, K., & Kompella, U. B. (2004). Particle & device engineering for inhalation drug delivery. Drug Delivery Technol., 4(2), 40,42,44-50.

Krajewska, B. (2004). Application of chitin- and chitosan-based materials for enzyme

immobilizations: a review Enzyme Microb. Tech. , 35(2-3), 126-139.

Krohn, K., Shuklov, I., Ahmed, I., & Voss, A. (2012). The use of hypophosphorous acid in

radical chain deoxygenation carbohydrates. In Carbohydrate Chemistry: Proven Synthetic Methods (Vol. 1 pp. 53-65).

Krug, S. M., Fromm, M., & Guenzel, D. (2009). Two-path impedance spectroscopy for

measuring paracellular and transcellular epithelial resistance. Biophys. J., 97(8), 2202-2211.

Kulvanich, P., & Stewart, P. J. (1987). The effect of particle size and concentration on the

adhesive characteristics of a model drug-carrier interactive system. J. Pharm. Pharmacol., 39(9), 673-678.

Kumirska, J., Weinhold, M. X., Czerwicka, M., Kaczynski, Z., Bychowska, A., Brzozowski, K.,

Thoeming, J., Stepnowski, P. (2011). Influence of the chemical structure and physicochemical properties of chitin- and chitosan-based materials on their biomedical activity. Biomed. Eng., Trends Mater. Sci., 25-64.

Kumirska, J., Weinhold, M. X., Steudte, S., Thoeming, J., Brzozowski, K., Stepnowski, P.

(2009). Determination of the pattern of acetylation of chitosan samples: Comparison of evaluation methods and some validation parameters. Int. J. Biol. Macromol., 45(1), 56-60.

Kunkel, S. L., Standiford, T., Kasahara, K., & Strieter, R. M. (1991). Interleukin-8 (IL-8): the

major neutrophil chemotactic factor in the lung. Exp. Lung Res., 17(1), 17-23.

Kurita, K., Hayakawa, M., Nishiyama, Y., & Harata, M. (2002a). Polymeric asymmetric

reducing agents: preparation and reducing performance of chitosan/dihydronicotinamide conjugates having L- and D-phenylalanine spacer arms. Carbohydr. Polym., 47(1), 7-14.

Kurita, K., Ichikawa, H., Ishizeki, S., Fujisaki, H., & Iwakura, Y. (1982). Studies on chitin.

8. Modification reaction of chitin in highly swollen state with aromatic cyclic carboxylic acid anhydrides Makromol. Chem., 183(5), 1161-1169.

Page 242: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

188

Kurita, K., Ikeda, H., Shimojoh, M., & Yang, J. (2007). N-phthaloylated chitosan as an essential precursor for controlled chemical modifications of chitosan: synthesis and evaluation. Polym. J. (Tokyo, Jpn.), 39(9), 945-952.

Kurita, K., Ikeda, H., Yoshida, Y., Shimojoh, M., & Harata, M. (2002b). Chemoselective

protection of the amino groups of chitosan by controlled phthaloylation: facile preparation of a precursor useful for chemical modifications. Biomacromolecules, 3(1), 1-4.

Kurita, K., Kojima, T., Nishiyama, Y., & Shimojoh, M. (2000a). Synthesis and Some Properties

of Non-natural Amino Polysaccharides: Branched Chitin and Chitosan. Macromolecules, 33(13), 4711-4716.

Kurita, K., Koyama, Y., Nishimura, S., & Kamiya, M. (1989). Facile preparation of water-

soluble chitin from chitosan. Chem. Lett.(9), 1597-1598.

Kurita, K., Sannan, T., & Iwakura, Y. (1977). Studies on chitin, 4. Evidence for formation of

block and random copolymers of N-acetyl-D-glucosamine and D-glucosamine by hetero- and homogeneous hydrolyses. Makromol. Chem., 178(12), 3197-3202.

Kurita, K., Shimada, K., Nishiyama, Y., Shimojoh, M., & Nishimura, S.-I. (1998). Nonnatural

Branched Polysaccharides: Synthesis and Properties of Chitin and Chitosan Having α-Mannoside Branches. Macromolecules, 31(15), 4764-4769.

Kurita, K., Uno, M., Saito, Y., & Nishiyama, Y. (2000b). Regioselectivity in protection of

chitosan with the phthaloyl group. Kichin, Kitosan Kenkyu, 6(2), 43-50.

Labiris, N. R., & Dolovich, M. B. (2003). Pulmonary drug delivery. Part II: the role of inhalant

delivery devices and drug formulations in therapeutic effectiveness of aerosolized medications. Br. J. Clin. Pharmacol., 56(6), 600-612.

Lakshminarayanan, V., Beno, D. W. A., Costa, R. H., & Roebuck, K. A. (1997). Differential

regulation of interleukin-8 and intercellular adhesion molecule-1 by H2O2 and tumor necrosis factor-α in endothelial and epithelial cells. J. Biol. Chem., 272(52), 32910-32918.

Laskin, D. L., Sunil, V. R., Laumbach, R. J., & Kipen, H. M. (2007). Inflammatory Cytokines and

Lung Toxicity, In: R. V. House & J. Descotes (Eds.), Cytokines in Human Health: Immunotoxicology, Pathology, and Therapeutic Applications (pp. 83-112): Humana Press.

Page 243: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

189

Lavorini, F. (2013). The challenge of delivering therapeutic aerosols to asthma patients. ISRN Allergy, 102418, 102418 pp.

Le Brun, P. P. H., de Boer, A. H., Heijerman, H. G. M., & Frijlink, H. W. (2000). A review of the

technical aspects of drug nebulization. Pharm. World Sci. , 22(3), 75-81.

Le, V. N. P., Thi, T. H. H., Robins, E., & Flament, M. P. (2012). In vitro evaluation of powders

for inhalation: The effect of drug concentration on particle detachment. Int. J. Pharm. (Amsterdam, Neth.), 424(1-2), 44-49.

Leach, C. L. (2007). Inhalation aspects of therapeutic aerosols. Toxicol. Pathol., 35(1), 23-26.

Learoyd, T. P., Burrows, J. L., French, E., & Seville, P. C. (2008a). Chitosan-based spray-dried

respirable powders for sustained delivery of terbutaline sulfate. Eur. J. Pharm. Biopharm. , 68(2), 224-234.

Learoyd, T. P., Burrows, J. L., French, E., & Seville, P. C. (2008b). Modified release of

beclometasone dipropionate from chitosan-based spray-dried respirable powders. Powder Technol., 187(3), 231-238.

Learoyd, T. P., Burrows, J. L., French, E., & Seville, P. C. (2009). Sustained delivery by leucine-

modified chitosan spray-dried respirable powders. Int. J. Pharm. , 372(1-2), 97-104.

Lechuga-Ballesteros, D., Charan, C., Stults Cheryl, L. M., Stevenson Cynthia, L., Miller

Danforth, P., Vehring, R., Tep, V., Kuo, M.-C. (2008). Trileucine improves aerosol performance and stability of spray-dried powders for inhalation. J. Pharm. Sci., 97(1), 287-302.

Lee, S. Y., & Boger, D. L. (2009). Synthesis of the chlorofusin cyclic peptide. Tetrahedron,

65(16), 3281-3284.

Lee, Y.-S., Kim, Y.-H., & Kim, S.-B. (2005). Changes in the respiration, growth, and Vitamin C

content of soybean sprouts in response to chitosan of different molecular weights. HortScience, 40(5), 1333-1335.

Lee, Y. H., Lee, M. H., & Shim, C. K. (1991). Pharmacokinetics of diltiazem and

deacetyldiltiazem in rats. Int. J. Pharm. , 76(1-2), 71-76.

Page 244: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

190

Li, C. B., Zheng, P. W., Zhao, Z. X., Zhang, W. Q., Li, M. B., Yang, Q. C., Cui, Y., Xu, Y. L. (2003). Reduction of azides to amines with new metal/Lewis acid systems in H2O or aqueous EtOH. Chin. Chem. Lett., 14(8), 773-775.

Li, F. Q., Hu, J. H., Lu, B., Yao, H., & Zhang, W. G. (2001). Ciprofloxacin-loaded bovine serum

albumin microspheres: preparation and drug-release in vitro. J. Microencapsul., 18(6), 825-829.

Li, H.-Y., & Birchall, J. (2006). Chitosan-Modified Dry Powder Formulations for Pulmonary

Gene Delivery. Pharm. Res., 23(5), 941-950.

Li, H. Y., Seville, P. C., Williamson, I. J., & Birchall, J. C. (2005). The use of amino acids to

enhance the aerosolisation of spray-dried powders for pulmonary gene therapy. J. Gene Med., 7(3), 343-353.

Li, X. Y., Guo, Q. F., Zheng, X. L., Kong, X. Y., Shi, S., Chen, L., Zhao, X., Wei, Y. Q., Qian, Z. Y.

(2009). Preparation of honokiol-loaded chitosan microparticles via spray-drying method intended for pulmonary delivery. Drug Delivery, 16(3), 160-166.

Lim, L. Y., & Wan, L. S. C. (1998). Effect of magnesium stearate on chitosan microspheres

prepared by an emulsification-coacervation technique. J. Microencapsulation, 15(3), 319-333.

Lim, L. Y., Wan, L. S. C., & Thai, P. Y. (1997). Chitosan microspheres prepared by

emulsification and ionotropic gelation. Drug Dev. Ind. Pharm., 23(10), 981-985.

Lim, S.-H., & Hudson, S. M. (2003). Review of chitosan and its derivatives as antimicrobial

agents and their uses as textile chemicals. J. Macromol. Sci. Polymer Rev., C43 (2), 223-269.

Lim, S. T., Forbes, B., Martin, G. P., & Brown, M. B. (2001). In vivo and in vitro

characterization of novel microparticulates based on hyaluronan and chitosan hydroglutamate. AAPS PharmSciTech, 2(4), 20.

Lim, S. T., Martin, G. P., Berry, D. J., & Brown, M. B. (2000). Preparation and evaluation of

the in vitro drug release properties and mucoadhesion of novel microspheres of hyaluronic acid and chitosan. J. Controlled Release, 66(2-3), 281-292.

Page 245: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

191

Lingam, M., Ashok, T., Venkateswarlu, V., & Rao, Y. M. (2008). Design and evaluation of a novel matrix type multiple units as biphasic gastroretentive drug delivery systems. AAPS PharmSciTech, 9(4), 1253-1261.

Liu, H., & Gao, C. (2009). Preparation and properties of ionically cross-linked chitosan

nanoparticles. Polym. Adv. Technol., 20(7), 613-619.

Liu, J., & Stewart, P. J. (1998). Deaggregation during the Dissolution of Benzodiazepines in

Interactive Mixtures. J. Pharm. Sci., 87(12), 1632-1638.

Liu, L., Li, Y., Li, Y., & Fang, Y.-E. (2004). Rapid N-phthaloylation of chitosan by microwave

irradiation. Carbohydr. Polym., 57(1), 97-100.

Liu, Y., Peterson, D. A., Kimura, H., & Schubert, D. (1997). Mechanism of cellular 3-(4,5-

dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J. Neurochem., 69(2), 581-593.

Louey, M. D., Razia, S., & Stewart, P. J. (2003). Influence of physico-chemical carrier

properties on the in vitro aerosol deposition from interactive mixtures. Int. J. Pharm., 252(1-2), 87-98.

Louey, M. D., Van Oort, M., & Hickey, A. J. (2004). Aerosol Dispersion of Respirable Particles

in Narrow Size Distributions Produced by Jet-Milling and Spray-Drying Techniques. Pharm. Res., 21(7), 1200-1206.

Madlova, M., Jones, S. A., Zwerschke, I., Ma, Y., Hider, R. C., Forbes, B. (2009). Poly(vinyl

alcohol) nanoparticle stability in biological media and uptake in respiratory epithelial cell layers in vitro. Eur. J. Pharm. Biopharm. , 72(2), 438-443.

Malcolmson, R. J., & Embleton, J. K. (1998). Dry powder formulations for pulmonary

delivery. Pharm. Sci. Technol. Today, 1(9), 394-398.

Manca, M.-L., Mourtas, S., Dracopoulos, V., Fadda, A. M., & Antimisiaris, S. G. (2008). PLGA,

chitosan or chitosan-coated PLGA microparticles for alveolar delivery? A comparative study of particle stability during nebulization. Colloids Surf., B, 62(2), 220-231.

Manning, M. C., Patel, K., & Borchardt, R. T. (1989). Stability of protein pharmaceuticals.

Pharm. Res. , 6(11), 903-918.

Page 246: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

192

Mansouri, S., Lavigne, P., Corsi, K., Benderdour, M., Beaumont, E., Fernandes, J. C. (2004). Chitosan-DNA nanoparticles as non-viral vectors in gene therapy: strategies to improve transfection efficacy. Eur. J. Pharm. Biopharm., 57(1), 1-8.

Mao, S., Cun, D., & Kawashima, Y. (2009). Novel non-injectable formulation approaches of

peptides and proteins. In: L. Jorgensen & H. M. Nielsen (Eds.), Delivery Technol. Biopharm. (pp. 29-67). West Sussex, UK: John Wiley & Sons Ltd.

Mao, S., Shuai, X., Unger, F., Wittmar, M., Xie, X., Kissel, T. (2005). Synthesis,

characterization and cytotoxicity of poly(ethylene glycol)-graft-trimethyl chitosan block copolymers. Biomaterials, 26(32), 6343-6356.

Marshall, N. J., Goodwin, C. J., & Holt, S. J. (1995). A critical assessment of the use of

microculture tetrazolium assays to measure cell growth and function. Growth Regul., 5(2), 69-84.

Martinac, A., Filipovic-Grcic, J., Perissutti, B., Voinovich, D., & Pavelic, Z. (2005). Spray-dried

chitosan/ethyl cellulose microspheres for nasal drug delivery: Swelling study and evaluation of in vitro drug release properties. J. Microencapsul. , 22(5), 549-561.

Martonen Ted, B., Smyth Hugh, D., Isaacs Kristin, K., & Burton Ray, T. (2005). Issues in drug

delivery: concepts and practice. Respir. Care, 50(9), 1228-1252.

Masotti, A., Marino, F., Ortaggi, G., & Palocci, C. (2007). Fluorescence and Scanning Electron

Microscopy of Chitosan/DNA Nanoparticles for Biological Applications. In: A. Mendes-Vilas & J. Diaz (Eds.), Modern Research and Educational Topics in Microscopy (Vol. 2, pp. 690-696): Formatex, Badajoz, Spain.

Mather, L. E., Woodhouse, A., Ward, M. E., Farr, S. J., Rubsamen, R. A., Eltherington, L. G.

(1998). Pulmonary administration of aerosolized fentanyl: pharmacokinetic analysis of systemic delivery. Br. J. Clin. Pharmacol., 46(1), 37-43.

Matubayasi, N., Miyamoto, H., Namihira, J., Yano, K., & Tanaka, T. (2002). Thermodynamic

Quantities of Surface Formation of Aqueous Electrolyte Solutions. J. Colloid Interface Sci., 250(2), 431-437.

Mazzarella, G., Esposito, V., Bianco, A., Ferraraccio, F., Prati, M. V., Lucariello, A., Manente,

L., Mezzogiorno, A., De Luca, A. (2012). Inflammatory effects on human lung epithelial cells after exposure to diesel exhaust micron sub particles (PM1.0) and pollen allergens. Environ. Pollut. (Oxford, U. K.), 161, 64-69.

Page 247: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

193

Mi, F.-L., Tan, Y.-C., Liang, H.-F., & Sung, H.-W. (2001). In vivo biocompatibility and degradability of a novel injectable-chitosan-based implant. Biomaterials, 23(1), 181-191.

Mi, F.-L., Wu, Y.-Y., Lin, Y.-H., Sonaje, K., Ho, Y.-C., Chen, C.-T., Juang, J.-H., Sung, H.-W.

(2008). Oral Delivery of Peptide Drugs Using Nanoparticles Self-Assembled by Poly(γ-glutamic acid) and a Chitosan Derivative Functionalized by Trimethylation. Bioconjugate Chem., 19(6), 1248-1255.

Miller, N. C., Marple, V. A., Schultz, R. K., & Poon, W. S. (1992). Assessment of the twin

impinger for size measurement of metered-dose inhaler sprays. Pharm. Res., 9(9), 1123-1127.

Mitra, A., & Dey, B. (2011). Chitosan microspheres in novel drug delivery systems. Indian J.

Pharm. Sci., 73(4), 355-366.

Monn, C., & Becker, S. (1999). Cytotoxicity and Induction of Proinflammatory Cytokines from

Human Monocytes Exposed to Fine (PM2.5) and Coarse Particles (PM10-2.5) in Outdoor and Indoor Air. Toxicol. Appl. Pharmacol., 155(3), 245-252.

Monteiro-Riviere, N. A., Inman, A. O., & Zhang, L. W. (2009). Limitations and relative utility

of screening assays to assess engineered nanoparticle toxicity in a human cell line. Toxicol. Appl. Pharmacol., 234(2), 222-235.

Morgan, D. M. L., Larvin, V. L., & Pearson, J. D. (1989). Biochemical characterization of

polycation-induced cytotoxicity to human vascular endothelial cells. J. Cell Sci., 94(3), 553-559.

Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: application to

proliferation and cytotoxicity assays. J. Immunol. Methods, 65(1-2), 55-63.

Mourya, V. K., & Inamdar, N. N. (2009). Trimethyl chitosan and its applications in drug

delivery. J. Mater. Sci.: Mater. Med. , 20(5), 1057-1079.

Mourya, V. K., Inamdar, N. N., & Tiwari, A. (2010). Carboxymethyl chitosan and its

applications. Adv. Mat. Lett., 1(1), 11-33.

Mura, S., Hillaireau, H., Nicolas, J., Le Droumaguet, B., Gueutin, C., Zanna, S., Tsapis, N.,

Fattal, E. (2011). Influence of surface charge on the potential toxicity of PLGA nanoparticles towards Calu-3 cells. Int. J. Nanomedicine, 6, 2591-2605.

Page 248: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

194

Murphy, J. G. (2007). Responses of Respiratory System Cells In Vitro and In Vivo to Petrochemical Combustion-Derived Ultrafine Particles. Unpublished Ph D thesis, Louisiana State University and Agricultural and Mechanical College.

Muzzarelli, R. A. A. (1993). Biochemical significance of exogenous chitins and chitosans in

animals and patients. Carbohydr. Polym., 20(1), 7-16.

Muzzarelli, R. A. A., Guerrieri, M., Goteri, G., Muzzarelli, C., Armeni, T., Ghiselli, R.,

Cornelissen, M. (2005). The biocompatibility of dibutyryl chitin in the context of wound dressings. Biomaterials, 26(29), 5844-5854.

Naikwade, S., & Bajaj, A. (2009). Preparation and in vitro evaluation of budesonide spray

dried microparticles for pulmonary delivery. Sci. Pharm. , 77(2), 419-441.

Najafabadi, A. R., Gilani, K., Barghi, M., & Rafiee-Tehrani, M. (2004). The effect of vehicle on

physical properties and aerosolization behaviour of disodium cromoglycate microparticles spray dried alone or with L-leucine. Int. J. Pharm., 285(1-2), 97-108.

Newman, S. P. (2005). Principles of metered-dose inhaler design. Respir. Care, 50(9),

1177-1190.

Nie, H., Lee, L. Y., Tong, H., & Wang, C.-H. (2008). PLGA/chitosan composites from a

combination of spray drying and supercritical fluid foaming techniques: New carriers for DNA delivery. J. Controlled Release, 129(3), 207-214.

Nishimura, S., Kohgo, O., Kurita, K., & Kuzuhara, H. (1991). Chemospecific manipulations of a

rigid polysaccharide: syntheses of novel chitosan derivatives with excellent solubility in common organic solvents by regioselective chemical modifications. Macromolecules, 24(17), 4745-4748.

Nishimura, S., Kohgo, O., Kurita, K., Vittavatvong, C., & Kuzuhara, H. (1990). Syntheses of

novel chitosan derivatives soluble in organic solvents by regioselective chemical modifications. Chem. Lett. (2), 243-246.

Nishiyama, Y., Yoshikawa, T., Kurita, K., Hojo, K., Kamada, H., Tsutsumi, Y., Mayumi, T.,

Kawasaki, K. (1999). Regioselective conjugation of chitosan with a laminin-related peptide, Tyr-Ile-Gly-Ser-Arg, and evaluation of its inhibitory effect on experimental cancer metastasis. Chem. Pharm. Bull., 47(3), 451-453.

Page 249: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

195

Nishiyama, Y., Yoshikawa, T., Ohara, N., Kurita, K., Hojo, K., Kamada, H., Tsutsumi, Y., Mayumi, T., Kawasaki, K. (2000). A conjugate from a laminin-related peptide, Tyr-Ile-Gly-Ser-Arg, and chitosan: efficient and regioselective conjugation and significant inhibitory activity against experimental cancer metastasis. J. Chem. Soc., Perkin 1(7), 1161-1165.

Nordtveit, R. J., Vaarum, K. M., & Smidsroed, O. (1996). Degradation of partially

N-acetylated chitosans with hen egg white and human lysozyme. Carbohydr. Polym., 29(2), 163-167.

Nud'ga, L. A., Plisko, E. A., & Danilov, S. N. (1973). N-Alkylation of chitosan. Zhur. Obshch.

Khim 43(12), 2756-2760.

O'Callaghan, C., & Barry, P. W. (1997). The science of nebulised drug delivery. Thorax, 52

Suppl 2, S31-44.

O'Riordan, T. G. (2002). Formulations and nebulizer performance. Respir. Care 47(11),

1305-1312; discussion 1312-1303.

Obeidat, W. M. (2009). Recent patents review in microencapsulation of pharmaceuticals

using the emulsion solvent removal methods. Recent Pat. Drug Deliv. Formul., 3(3), 178-192.

Oberdorster, G. (2001). Pulmonary effects of inhaled ultrafine particles. Int. Arch. Occup.

Environ. Health, 74(1), 1-8.

Oberdorster, G., Oberdorster, E., & Oberdorster, J. (2005). Nanotoxicology: an emerging

discipline evolving from studies of ultrafine particles. Environ. Health Perspect., 113(7), 823-839.

Ohnishi, T., Maruyama, T., Higashi, S., & Awazu, S. (2000). Blood-brain barrier transport of

L-tyrosine conjugates: a model study for the brain targeting using large neutral amino acid transport system. J. Drug Target., 8(6), 395-401.

Okamoto, H., Shiraki, K., Yasuda, R., Danjo, K., & Watanabe, Y. (2011). Chitosan-interferon-β

gene complex powder for inhalation treatment of lung metastasis in mice. J. Controlled Release, 150(2), 187-195.

Page 250: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

196

Onishi, H., & Machida, Y. (2006). Improvement of Oral Drug Absorption by Chitosan and Its Derivatives. In: E. Touitou & Brian W. Barry (Editors) (Eds.), Enhancement in Drug Delivery (pp. 57): CRC Press, Taylor & Francis Group.

Onishi, H., Oosegi, T., Machida, Y., & McGinity, J. W. (2005). Preparation and in vitro

evaluation of chitosan microspheres containing prednisolone: Comparison of simple and conjugate microspheres. Drug Dev. Ind. Pharm., 31(7), 597-605.

Oppenheim, J. J., Zachariae, C. O. C., Mukaida, N., & Matsushima, K. (1991). Properties of

the novel proinflammatory supergene "intercrine" cytokine family. Annu. Rev. Immunol., 9, 617-648.

Oshita, K., Noguchi, O., Oshima, M., & Motomizu, S. (2007a). Synthesis of cross-linked

chitosan modified with the glycine moiety for the collection/concentration of bismuth in aquatic samples for ICP-MS determination. Anal. Sci., 23(10), 1203-1208.

Oshita, K., Takayanagi, T., Oshima, M., & Motomizu, S. (2007b). Adsorption behavior of

cationic and anionic species on chitosan resins possessing amino acid moieties. Anal. Sci , 23(12), 1431-1434.

Oshita, K., Xu, J., Gao, Y.-H., Lee, K.-H., Oshima, M., Motomizu, S. (2003). Synthesis of cross-

linked chitosan resin possessing leucine moiety for the column collection/concentration of molybdenum and determination of molybdenum by ICP-MS, ICP-AES and GFAAS. Bull. Chem. Soc. Jpn. , 76(8), 1555-1560.

Oungbho, K., & Muller, B. W. (1997). Chitosan sponges as sustained-release drug carriers.

Int. J. Pharm., 156(2), 229-237.

Paajanen, M., Katainen, J., Raula, J., Kauppinen, E. I., & Lahtinen, J. (2009). Direct evidence

on reduced adhesion of Salbutamol sulphate particles due to L-leucine coating. Powder Technol., 192(1), 6-11.

Panos, I., Acosta, N., & Heras, A. (2008). New drug delivery systems based on chitosan. Curr.

Drug Discovery Technol., 5(4), 333-341.

Patel, B., Gupta, V., & Ahsan, F. (2012). PEG-PLGA Based Large Porous Particles for

Pulmonary Delivery of a Highly Soluble Drug, Low Molecular Weight Heparin. J. Controlled Release, 162(2), 310-320.

Page 251: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

197

Patel, V. M., Prajapati, B. G., & Patel, M. M. (2007). Design and characterization of chitosan-containing mucoadhesive buccal patches of propranolol hydrochloride. Acta Pharm. (Zagreb, Croatia), 57(1), 61-72.

Patel, V. R., & Amiji, M. M. (1996). Preparation and characterization of freeze-dried

chitosan-poly(ethylene oxide) hydrogels for site-specific antibiotic delivery in the stomach. Pharm. Res., 13(4), 588-593.

Patton, J. S. (1996). Mechanisms of macromolecule absorption by the lungs. Adv. Drug

Delivery Rev., 19(1), 3-36.

Patton, J. S. (1997). Deep-lung delivery of therapeutic proteins. Chemtech, 27(12), 34-38.

Patton, J. S. (2000). Pulmonary delivery of drugs for bone disorders. Adv. Drug Delivery Rev.,

42(3), 239-248.

Patton, J. S., & Byron, P. R. (2007). Inhaling medicines: delivering drugs to the body through

the lungs. Nat. Rev. Drug Discovery, 6(1), 67-74.

Patton, J. S., Fishburn, C. S., & Weers, J. G. (2004). The lungs as a portal of entry for systemic

drug delivery. Proc. Am. Thorac. Soc., 1(4), 338-344.

Patton, J. S., Nagarajan, S., & Clark, A. (1998). Pulmonary absorption and metabolism of

peptides and proteins. Respir. Drug Delivery VI, Int. Symp., 6th, 17-24.

Pease, J. E., & Sabroe, I. (2002). The role of interleukin-8 and its receptors in inflammatory

lung disease: Implications for therapy. Am. J. Respir. Med., 1(1), 19-25.

Peniche, C., Arguelles-Monal, W., Peniche, H., & Acosta, N. (2003). Chitosan: An attractive

biocompatible polymer for microencapsulation. Macromol. Biosci. , 3(10), 511-520.

Perich, J. W., Langford, N. J., & Johns, R. B. (1987). Synthesis of casein-related peptides and

phosphopeptides. II. The solid-phase synthesis of the non-phosphorylated octapeptide corresponding to region 14-21 of bovine β-casein A2. Aust. J. Chem., 40(8), 1389-1397.

Phaechamud, T., & Charoenteeraboon, J. (2008). Antibacterial activity and drug release of

chitosan sponge containing doxycycline hyclate. AAPS PharmSciTech, 9(3), 829-835.

Page 252: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

198

Pilcer, G., & Amighi, K. (2010). Formulation strategy and use of excipients in pulmonary drug delivery. Int. J. Pharm., 392(1–2), 1-19.

Pillai, C. K. S., Paul, W., & Sharma, C. P. (2009). Chitin and chitosan polymers: Chemistry,

solubility and fiber formation. Prog. Polym. Sci., 34(7), 641-678.

Pirooznia, N., Hasannia, S., Sahebghadam Lotfi, A., & Ghanei, M. (2012). Encapsulation of

Alpha 1-antitrypsin in PLGA nanoparticles: in vitro characterization as an effective aerosol formulation in pulmonary diseases. J. Nanobiotechnol., 10, 20.

Prasad, S. R., & Kishore, V. S. (2012). Enhancement of skin permeation of Diltiazem

Hydrochloride gels through mouse skin by using olive oil as permeation enhancers. Int. J. Pharm., Chem. Biol. Sci., 2(3), 348-353.

Puttipipatkhachorn, S., Nunthanid, J., Yamamoto, K., & Peck, G. E. (2001). Drug physical

state and drug-polymer interaction on drug release from chitosan matrix films. J. Controlled Release, 75(1-2), 143-153.

Qin, C., Du, Y., Xiao, L., Li, Z., & Gao, X. (2002). Enzymic preparation of water-soluble

chitosan and their antitumor activity. Int. J. Biol. Macromol., 31(1-3), 111-117.

Quay, J. L., Reed, W., Samet, J., & Devlin, R. B. (1998). Air pollution particles induce IL-6 gene

expression in human airway epithelial cells via NF-κB activation. Am. J. Respir. Cell Mol. Biol., 19(1), 98-106.

Rabbani, N. R., & Seville, P. C. (2005). The influence of formulation components on the

aerosolization properties of spray-dried powders. J. Controlled Release, 110(1), 130-140.

Rabinowitz, J. D., Wensley, M., Lloyd, P., Myers, D., Shen, W., Lu, A., Hodges, C., Hale, R.,

Mufson, D., Zaffaroni, A. (2004). Fast onset medications through thermally generated aerosols. J. Pharmacol. Exp. Ther. , 309(2), 769-775.

Rahman, I., & MacNee, W. (2000). Oxidative stress and regulation of glutathione in lung

inflammation. Eur. Respir. J., 16(3), 534-554.

Rau, J. L. (2002). Design principles of liquid nebulization devices currently in use. Respir. Care

47(11), 1257-1275; discussion 1275-1258.

Page 253: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

199

Raula, J., Laehde, A., & Kauppinen, E. I. (2008). A Novel Gas Phase Method for the Combined Synthesis and Coating of Pharmaceutical Particles. Pharm. Res., 25(1), 242-245.

Raula, J., Thielmann, F., Naderi, M., Lehto, V.-P., & Kauppinen, E. I. (2010). Investigations on

particle surface characteristics vs. dispersion behaviour of L-leucine coated carrier-free inhalable powders. Int. J. Pharm., 385(1-2), 79-85.

Reddel, R. R., Ke, Y., Gerwin, B. I., McMenamin, M. G., Lechner, J. F., Su, R. T., Brash, D. E.,

Park, J. B., Rhim, J. S., Harris, C. C. (1988). Transformation of human bronchial epithelial cells by infection with SV40 or adenovirus-12 SV40 hybrid virus, or transfection via strontium phosphate coprecipitation with a plasmid containing SV40 early region genes. Cancer Res. , 48(7), 1904-1909.

Richards, F., & Knowles, J. R. (1968). Glutaraldehyde as a protein cross-linking reagent.

J. Mol. Biol., 37, 231-233.

Rogueda, P. G. A., & Traini, D. (2007). The nanoscale in pulmonary delivery. Part 1:

deposition, fate, toxicology and effects. Expert Opin. Drug Deliv., 4(6), 595-606.

Rout, D. K., Barman, S. P., Pulapura, S. K., & Gross, R. A. (1994). Cholesteric Mesophases

Formed by the Modified Biological Macromolecule 3,6-O-(Butyl Carbamate)-N-phthaloyl Chitosan. Macromolecules, 27(11), 2945-2950.

Rout, D. K., Pulapura, S. K., & Gross, R. A. (1993). Liquid-crystalline characteristics of site-

selectively-modified chitosan. Macromolecules, 26(22), 5999-6006.

Roy, K., Mao, H.-Q., Huang, S.-K., & Leong, K. W. (1999). Oral gene delivery with chitosan-

DNA nanoparticles generates immunological protection in a murine model of peanut allergy Nat. Med. (New York), 5(4), 387-391.

Ruel-Gariepy, E., Chenite, A., Chaput, C., Guirguis, S., & Leroux, J. C. (2000). Characterization

of thermosensitive chitosan gels for the sustained delivery of drugs. Int. J. Pharm., 203(1-2), 89-98.

Sadeghi, A. M. M., Dorkoosh, F. A., Avadi, M. R., Saadat, P., Rafiee-Tehrani, M., Junginger, H.

E. (2008). Preparation, characterization and antibacterial activities of chitosan, N-trimethyl chitosan (TMC) and N-diethylmethyl chitosan (DEMC) nanoparticles loaded with insulin using both the ionotropic gelation and polyelectrolyte complexation methods. Int. J. Pharm. , 355(1-2), 299-306.

Page 254: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

200

Saedisomeolia, A., Wood, L. G., Garg, M. L., Gibson, P. G., & Wark, P. A. B. (2009). Anti-inflammatory effects of long-chain n-3 PUFA in rhinovirus-infected cultured airway epithelial cells. Br. J. Nutr., 101(4), 533-540.

Saha, P., Goyal, A. K., & Rath, G. (2010). Formulation and evaluation of chitosan-based

ampicillin trihydrate nanoparticles. Trop. J. Pharm. Res., 9(5), 483-488.

Sahib, M. N., Abdalwahed, S., Abdulameer, Darwis, Y., Peh, K. K., Tan, Y. T. F. (2012).

Solubilization of beclomethasone dipropionate in sterically stabilized phospholipid nanomicelles (SSMs): physicochemical and in vitro evaluations. Drug Des., Dev. Ther., 6, 29-42.

Saito, H., Tabeta, R., & Hirano, S. (1981). Conformation of chitin and N-acyl chitosans in solid

state as revealed by carbon-13 cross polarization/magic angle spinning (CP/MAS) NMR spectroscopy. Chem. Lett.(10), 1479-1482.

Saito, H., Tabeta, R., & Ogawa, K. (1987). High-resolution solid-state carbon-13 NMR study

of chitosan and its salts with acids: conformational characterization of polymorphs and helical structures as viewed from the conformation-dependent carbon-13 chemical shifts. Macromolecules, 20(10), 2424-2430.

Saks, S. R., & Gardner, L. B. (1997). The pharmacoeconomic value of controlled-release

dosage forms. J. Controlled Release, 48(2,3), 237-242.

Salabei, J. K., Balakumaran, A., Frey, J. C., Boor, P. J., Treinen-Moslen, M., Conklin, D. J.

(2012). Verapamil stereoisomers induce antiproliferative effects in vascular smooth muscle cells via autophagy. Toxicol. Appl. Pharmacol., 262(3), 265-272.

Salama, R., Hoe, S., Chan, H.-K., Traini, D., & Young, P. M. (2008). Preparation and

characterisation of controlled release co-spray dried drug-polymer microparticles for inhalation 1: Influence of polymer concentration on physical and in vitro characteristics. Eur. J. Pharm. Biopharm. , 69(2), 486-495.

Salama, R., Traini, D., Chan, H.-K., & Young, P. M. (2009a). Recent advances in controlled

release pulmonary therapy. Curr. Drug Delivery, 6(4), 404-414.

Salama, R. O., Traini, D., Chan, H.-K., Sung, A., Ammit, A. J., Young, P. M. (2009b).

Preparation and evaluation of controlled release microparticles for respiratory protein therapy. J. Pharm. Sci. , 98(8), 2709-2717.

Page 255: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

201

Sandri, G., Bonferoni, M. C., Rossi, S., Ferrari, F., Boselli, C., Caramella, C. (2010). Insulin-loaded nanoparticles based on N-trimethyl chitosan: in vitro (Caco-2 model) and ex vivo (excised rat jejunum, duodenum, and ileum) evaluation of penetration enhancement properties. AAPS PharmSciTech, 11(1), 362-371.

Sandri, G., Bonferoni, M. C., Rossi, S., Ferrari, F., Gibin, S., Zambito, Y., Di Colo, G., Caramella,

C. (2007). Nanoparticles based on N-trimethylchitosan: evaluation of absorption properties using in vitro (Caco-2 cells) and ex vivo (excised rat jejunum) models. Eur. J. Pharm. Biopharm., 65(1), 68-77.

Sanjar, S., & Matthews, J. (2001). Treating systemic diseases via the lung. J. Aerosol Med.

Pulm. Drug Delivery, 14(Suppl. 1), S51-S58.

Satoh, T., Kano, H., Nakatani, M., Sakairi, N., Shinkai, S., Nagasaki, T. (2006). 6-Amino-6-

deoxy-chitosan. Sequential chemical modifications at the C-6 positions of N-phthaloyl-chitosan and evaluation as a gene carrier. Carbohydr. Res., 341(14), 2406-2413.

Schipper, N. G. M., Olsson, S., Hoogstraate, J. A., deBoer, A. G., Varum, K. M., Artursson, P.

(1997). Chitosans as absorption enhancers for poorly absorbable drugs. 2: Mechanism of absorption enhancement. Pharm. Res., 14(7), 923-929.

Schlesinger, R. B. (1985). Clearance from the respiratory tract. Fundam. Appl. Toxicol., 5(3),

435-450.

Schulz, C., Farkas, L., Wolf, K., Kraetzel, K., Eissner, G., Pfeifer, M. (2002). Differences in LPS-

induced activation of bronchial epithelial cells (BEAS-2B) and type II-like pneumocytes (A-549). Scand. J. Immunol., 56(3), 294-302.

Scozzafava, A., & Supuran, C. T. (2000 ). Carbonic anhydrase activators - Part 21. Novel

activators of isozymes I, II and IV incorporating carboxamido and ureido histamine moieties Eur. J. Med. Chem., 35(1), 31-39.

Seville, P. C., & Learoyd, T. P. (2007). Spray dried dispersible powder composition for

inhalation (pp. 33pp). PCT Int. Appl.: WO 2007060384 A2 20070531.

Seville, P. C., Learoyd, T. P., Li, H. Y., Williamson, I. J., & Birchall, J. C. (2007). Amino acid-

modified spray-dried powders with enhanced aerosolization properties for pulmonary drug delivery. Powder Technol., 178(1), 40-50.

Page 256: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

202

Sezer, A. D., & Akbuga, J. (1995). Controlled release of piroxicam from chitosan beads. Int. J. Pharm., 121(1), 113-116.

Shah, S. P., & Misra, A. (2004). Liposomal amikacin dry powder inhaler: effect of fines on in

vitro performance. AAPS PharmSciTech, 5(4), e65.

Shahiwala, A., & Misra, A. (2005). A preliminary pharmacokinetic study of liposomal

leuprolide dry powder inhaler: a technical note. AAPS PharmSciTech, 6(3), E482-486.

Shaikh, S., Nazim, S., Khan, T., Shaikh, A., Zameeruddin, M., Quazi, A. (2010). Recent

advances in pulmonary drug delivery system: a review. Int. J. Appl. Pharm., 2(4), 27-31.

Sham, J. O. H., Zhang, Y., Finlay, W. H., Roa, W. H., & Lobenberg, R. (2004). Formulation and

characterization of spray-dried powders containing nanoparticles for aerosol delivery to the lung. Int. J. Pharm., 269(2), 457-467.

Shuai, H.-H., Yang, C.-Y., Harn, H. I. C., York, R. L., Liao, T.-C., Chen, W.-S., Yeh, J. A., Cheng,

C.-M. (2013). Using surfaces to modulate the morphology and structure of attached cells - a case of cancer cells on chitosan membranes. Chem. Sci., 4(8), 3058-3067.

Sigurdsson, H. H., Loftsson, T., & Lehr, C.-M. (2006). Assessment of mucoadhesion by a

resonant mirror biosensor. Int. J. Pharm. , 325(1-2), 75-81.

Sinko, P. J. (2006). Martin's Physical Pharmacy and Pharmaceutical Sciences: Physical

Chemical and Biopharmaceutical Principles in the Pharmaceutical Sciences (Illustrated ed.). Philadelphia, PA: Lippincott Williams & Wilkins.

Sivadas, N. (2010). Bioengineered microparticles for controlled drug delivery to the lungs.

Ph.D. dissertation, National University of Ireland

Sivadas, N., O'Rourke, D., Tobin, A., Buckley, V., Ramtoola, Z., Kelly, J. G., Hickey, A. J., Cryan,

S.-A. (2008). A comparative study of a range of polymeric microspheres as potential carriers for the inhalation of proteins. Int. J. Pharm. , 358(1-2), 159-167.

Sivasakthivel, T., & Reddy, K. K. S. K. (2011). Ozone Layer Depletion and Its Effects: A Review.

Int. J. Environ. Sci. Technol., 2(1), 30-37.

Page 257: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

203

Slater, T. F., Sawyer, B., & Straeuli, U. (1963). Studies on Succinate-Tetrazolium Reductase Systems. Iii. Points of Coupling of Four Different Tetrazolium Salts. Biochim. Biophys. Acta, 77, 383-393.

Smith, J., Wood, E., & Dornish, M. (2004). Effect of Chitosan on Epithelial Cell Tight

Junctions. Pharm. Res., 21(1), 43-49.

Smith, J. M., Dornish, M., & Wood, E. J. (2005). Involvement of protein kinase C in chitosan

glutamate-mediated tight junction disruption. Biomaterials, 26(16), 3269-3276.

Smyth, H. D. C., & Hickey, A. J. (2005). Carriers in drug powder delivery: implications for

inhalation system design. Am. J. Drug Delivery, 3(2), 117-132.

Soane, R. J., Frier, M., Perkins, A. C., Jones, N. S., Davis, S. S., Illum, L. (1999). Evaluation of

the clearance characteristics of bioadhesive systems in humans. Int. J. Pharm., 178(1), 55-65.

Sosnovsky, G., & Gnewuch, C. T. (1994). In the Search for New Anticancer Drugs. 27.

Synthesis and Comparison of Anticancer Activity in vivo of Amino Acids, Carbohydrates, and Carbohydrate-Amino Acid Conjugates Containing the [N'-(2-Chloroethyl)-N'-nitrosoamino]Carbonyl Group. J. Pharm. Sci., 83(7), 989-998.

Srichana, T., Suedee, R., & Reanmongkol, W. (2001). Cyclodextrin as a potential drug carrier

in salbutamol dry powder aerosols: the in-vitro deposition and toxicity studies of the complexes. Respir. Med., 95(6), 513-519.

Srinatha, A., Pandit, J. K., & Singh, S. (2008). Ionic Cross-linked Chitosan Beads for Extended

Release of Ciprofloxacin: In vitro Characterization. Indian J. Pharm. Sci., 70(1), 16-21.

Staniforth, J., Morton, D., Tobyn, M., Eason, S., Harmer, Q., Ganderton, D. (2004). Devices

and pharmaceutical compositions containing amino acids, phospholipids or stearates for enhancing dosing efficiency (pp. 222 pp). PCT Int. Appl.: WO 2004093848 A2 20041104.

Staniforth, J. N. (1981). Total mixing. Int. J. Pharm. Tech. Prod. Mfr., 2(1), 7-12.

Staniforth, J. N. (1987). Order out of chaos. J. Pharm. Pharmacol., 39(5), 329-334.

Page 258: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

204

Staniforth, J. N., & Morton, D. A. V. (2002). Magnesium stearate, a phospholipid, or an amino acid in preparation of pharmaceutical particles for inhalation (pp. 39 pp). PCT Int. Appl.: WO 2002043700 A2 20020606.

Steckel, H., Markefka, P., teWierik, H., & Kammelar, R. (2004). Functionality testing of

inhalation grade lactose. Eur. J. Pharm. Biopharm., 57(3), 495-505.

Steckel, H., & Mueller, B. W. (1997). In vitro evaluation of dry powder inhalers II: influence

of carrier particle size and concentration on in vitro deposition. Int. J. Pharm., 154(1), 31-37.

Steckel, H., Rasenack, N., Villax, P., & Muller, B. W. (2003). In vitro characterization of jet-

milled and in-situ-micronized fluticasone-17-propionate. Int. J. Pharm., 258(1-2), 65-75.

Stefanescu, C., Daly, W., & Negulescu, I. (2008). Reaction of chitosan with trityl chloride and

phthalic anhydride in homogeneous ionic liquid solutions. Polym. Prepr. (Am. Chem. Soc., Div. Polym. Chem.), 49(2), 793-794.

Stolnik, S., & Shakesheff, K. (2009). Formulations for delivery of therapeutic proteins.

Biotechnol. Lett., 31(1), 1-11.

Stone, V., Shaw, J., Brown, D. M., Macnee, W., Faux, S. P., Donaldson, K. (1998). The role of

oxidative stress in the prolonged inhibitory effect of ultrafine carbon black on epithelial cell function. Toxicol. in Vitro, 12(6), 649-659.

Strengert, M., & Knaus, U. G. (2011). Analysis of epithelial barrier integrity in polarized lung

epithelial cells. Methods Mol. Biol. (N. Y., NY, U. S.), 763(Permeability Barrier), 195-206.

Suarez, S., O'Hara, P., Kazantseva, M., Newcomer, C. E., Hopfer, R., McMurray, D. N., Hickey,

A. J. (2001). Respirable PLGA microspheres containing rifampicin for the treatment of tuberculosis: screening in an infectious disease model. Pharm. Res., 18(9), 1315-1319.

Sun, Y., Cui, F., Shi, K., Wang, J., Niu, M., Ma, R. (2009). The effect of chitosan molecular

weight on the characteristics of spray-dried methotrexate-loaded chitosan microspheres for nasal administration. Drug Dev. Ind. Pharm. , 35(3), 379-386.

Page 259: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

205

Sung, J. C., Padilla, D. J., Garcia-Contreras, L., VerBerkmoes, J. L., Durbin, D., Peloquin, C. A., Elbert, K. J., Hickey, A. J., Edwards, D. A. (2009). Formulation and Pharmacokinetics of Self-Assembled Rifampicin Nanoparticle Systems for Pulmonary Delivery. Pharm. Res., 26(8), 1847-1855.

Swami, A., Shi, J., Gadde, S., Votruba, A. R., Kolishetti, N., Farokhzad, O. C. (2012).

Nanoparticles for Targeted and Temporally Controlled Drug Delivery. In: S. Svenson & R. K. Prud’homme (Eds.), Multifunctional Nanoparticles for Drug Delivery Applications Nanostructure Science and Technology (pp. 9-29 ). New York: Springer

Takizawa, H., Ohtoshi, T., Kawasaki, S., Abe, S., Sugawara, I., Nakahara, K., Matsushima, K.,

Kudoh, S. (2000). Diesel exhaust particles activate human bronchial epithelial cells to express inflammatory mediators in the airways: a review. Respirology (Carlton, Vic.), 5(2), 197-203.

Tal, T. L., Simmons, S. O., Silbajoris, R., Dailey, L., Cho, S.-H., Ramabhadran, R., Linak, W.,

Reed, W., Bromberg, P. A., Samet, J. M. (2010). Differential transcriptional regulation of IL-8 expression by human airway epithelial cells exposed to diesel exhaust particles. Toxicol. Appl. Pharmacol., 243(1), 46-54.

Tan, T. H. S., Worthington, R. J., Pritchard, R. G., Morral, J., & Micklefield, J. (2007).

Homopolymeric pyrrolidine-amide oligonucleotide mimics: Fmoc-synthesis and DNA/RNA binding properties Org. Biomol. Chem., 5(2), 239-248.

Taneja, R., Sachdeva, V., Khatri, N., & Singh, V. (2012). Recent trends in DPI Technology. Int.

Res. J. Pharm., 3(11), 27-34.

Tang, P., Chan, H. K., Chiou, H., Ogawa, K., Jones, M. D., Adi, H., Buckton, G., Prud'homme, R.

K., Raper, J. A. (2009). Characterization and aerosolization of mannitol particles produced via confined liquid impinging jets. Int. J. Pharm., 367(1-2), 51-57.

Taub, D. D., Anver, M., Oppenheim, J. J., Longo, D. L., & Murphy, W. J. (1996). T lymphocyte

recruitment by interleukin-8 (IL-8). IL-8-induced degranulation of neutrophils releases potent chemoattractants for human T lymphocytes both in vitro and in vivo. J. Clin. Invest., 97(8), 1931-1941.

Tavares, I. S., Caroni, A. L. P. F., Dantas Neto, A. A., Pereira, M. R., & Fonseca, J. L. C. (2012).

Surface charging and dimensions of chitosan coacervated nanoparticles. Colloids Surf., B., 90, 254-258.

Page 260: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

206

Teijeiro-Osorio, D., Remunan-Lopez, C., & Alonso, M. J. (2009a). Chitosan/cyclodextrin nanoparticles can efficiently transfect the airway epithelium in vitro. Eur. J. Pharm. Biopharm., 71(2), 257-263.

Teijeiro-Osorio, D., Remunan-Lopez, C., & Alonso, M. J. (2009b). New Generation of Hybrid

Poly/Oligosaccharide Nanoparticles as Carriers for the Nasal Delivery of Macromolecules. Biomacromolecules, 10(2), 243-249.

Thanou, M., Florea, B. I., Geldof, M., Junginger, H. E., & Borchard, G. (2002). Quaternized

chitosan oligomers as novel gene delivery vectors in epithelial cell lines. Biomaterials, 23(1), 153-159.

Thanou, M., Florea, B. I., Langemeyer, M. W. E., Verhoef, J. C., & Junginger, H. E. (2000a).

N-trimethylated chitosan chloride (TMC) improves the intestinal permeation of the peptide drug buserelin in vitro (Caco-2 cells) and in vivo (rats). Pharm. Res., 17(1), 27-31.

Thanou, M., Verhoef, J. C., & Junginger, H. E. (2001a). Oral drug absorption enhancement by

chitosan and its derivatives. Adv. Drug Delivery Rev., 52(2), 117-126.

Thanou, M., Verhoef, J. C., Marbach, P., & Junginger, H. E. (2000b). Intestinal absorption of

octreotide: N-trimethyl chitosan chloride (TMC) ameliorates the permeability and absorption properties of the somatostatin analogue in vitro and in vivo. J. Pharm. Sci., 89(7), 951-957.

Thanou, M., Verhoef, J. C., Verheijden, J. H. M., & Junginger, H. E. (2001b). Intestinal

absorption of octreotide using trimethyl chitosan chloride: studies in pigs. Pharm. Res., 18(6), 823-828.

Timsina, M. P., Martin, G. P., Marriott, C., Ganderton, D., & Yianneskis, M. (1994). Drug

delivery to the respiratory tract using dry powder inhalers. Int. J. Pharm., 101(1-2), 1-13.

Todoroff, J., & Vanbever, R. (2011). Fate of nanomedicines in the lungs. Curr. Opin. Colloid

Interface Sci., 16(3), 246-254.

Tong, H., Shi, Q., Fernandes, J. C., Liu, L., Dai, K., Zhang, X. (2009). Progress and prospects of

chitosan and its derivatives as non-viral gene vectors in gene therapy. Curr. Gene Ther., 9(6), 495-502.

Page 261: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

207

Torres, D., Baodo, L., Blanco, D., & Vila-Jato, J. (1998 ). Comparison between aqueous and non-aqueous solvent evaporation methods for microencapsulation of drug–resin complexes. Int. J. Pharm. , 173, 171–182.

Tronde, A., Gillen, M., Borgstroem, L., Loetvall, J., & Ankerst, J. (2008). Pharmacokinetics of

budesonide and formoterol administered via 1 pressurized metered-dose inhaler in patients with asthma and COPD. J. Clin. Pharmacol., 48(11), 1300-1308.

Troy, D. B., & Beringer, P. (Eds.). (2006). Remington: the Science and Practice of Pharmacy

(21 ed.). Philadelphia, PA: Lippincott Williams & Wilkins.

Tsukue, N., Okumura, H., Ito, T., Sugiyama, G., & Nakajima, T. (2010). Toxicological

evaluation of diesel emissions on A549 cells. Toxicol. in Vitro, 24(2), 363-369.

Ubaidulla, U., Khar, R. K., Ahmad, F. J., Sultana, Y., & Panda, A. K. (2007). Development and

characterization of chitosan succinate microspheres for the improved oral bioavailability of insulin. J. Pharm. Sci., 96(11), 3010-3023.

Ulrich, M. M. W., Alink, G. M., Kumarathasan, P., Vincent, R., Boere, A. J. F., Cassee, F. R.

(2002). Health effects and time course of particulate matter on the cardiopulmonary system in rats with lung inflammation. J. Toxicol. Environ. Health, Part A, 65(20), 1571-1595.

Ungaro, F., De Rosa, G., Miro, A., Quaglia, F., & La Rotonda, M. I. (2006). Cyclodextrins in the

production of large porous particles: Development of dry powders for the sustained release of insulin to the lungs. Eur. J. Pharm. Sci., 28(5), 423-432.

van der Lubben, I. M., Verhoef, J. C., Borchard, G., & Junginger, H. E. (2001). Chitosan for

mucosal vaccination Adv. Drug Delivery Rev. , 52(2), 139-144.

van der Merwe, S. M., Verhoef, J. C., Verheijden, J. H. M., Kotze, A. F., & Junginger, H. E.

(2004). Trimethylated chitosan as polymeric absorption enhancer for improved peroral delivery of peptide drugs. Eur. J. Pharm. Biopharm., 58(2), 225-235.

Vanhee, D., Gosset, P., Boitelle, A., Wallaert, B., & Tonnel, A. B. (1995). Cytokines and

cytokine network in silicosis and coal workers' pneumoconiosis. Eur. Respir. J., 8(5), 834-842.

Page 262: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

208

Ventura, C. A., Tommasini, S., Crupi, E., Giannone, I., Cardile, V., Musumeci, T., Puglisi, G. (2008). Chitosan microspheres for intrapulmonary administration of moxifloxacin: Interaction with biomembrane models and in vitro permeation studies. Eur. J. Pharm. Biopharm., 68(2), 235-244.

Vikhoreva, G. A., Gorbacheva, I. N., & Gal'braikh, L. S. (1999). Synthesis and properties of

water-soluble derivatives of chitin. A review. Fibre Chem., 31(4), 274-278.

Vistica, D. T., Skehan, P., Scudiero, D., Monks, A., Pittman, A., Boyd, M. R. (1991).

Tetrazolium-based assays for cellular viability: a critical examination of selected parameters affecting formazan production. Cancer Res., 51(10), 2515-2520.

Vllasaliu, D., Exposito-Harris, R., Heras, A., Casettari, L., Garnett, M., Illum, L., Stolnik, S.

(2010). Tight junction modulation by chitosan nanoparticles: Comparison with chitosan solution. Int. J. Pharm., 400(1-2), 183-193.

Wan, H., Williams, R., Doherty, P., & Williams, D. F. (1994). A study of the reproducibility of

the MTT test. J. Mater. Sci.: Mater. Med., 5 (3), 154-159.

Wan, H., Winton, H. L., Soeller, C., Stewart, G. A., Thompson, P. J., Gruenert, D. C., Cannell,

M. B., Garrod, D. R., Robinson, C. (2000). Tight junction properties of the immortalized human bronchial epithelial cell lines Calu-3 and 16HBE14o. Eur. Respir. J., 15(6), 1058-1068.

Wang, L.-C., Chen, X.-G., Zhong, D.-Y., & Xu, Q.-C. (2007). Study on poly(vinyl

alcohol)/carboxymethyl-chitosan blend film as local drug delivery system. J. Mater. Sci.: Mater. Med., 18(6), 1125-1133.

Watry, M. R., & Richmond, G. L. (2002). Orientation and Conformation of Amino Acids in

Monolayers Adsorbed at an Oil/Water Interface As Determined by Vibrational Sum-Frequency Spectroscopy. J. Phys. Chem. B, 106(48), 12517-12523.

Weibel, E. R. (1963). Principles and methods for the morphometric study of the lung and

other organs. Lab. Invest., 12, 131-155.

Wen, H., & Park, K. (2011). Oral Controlled Release Formulation Design and Drug Delivery:

Theory to Practice. Hoboken, New Jersey: John Wiley & Sons, Inc.

Page 263: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

209

Westmoreland, C., Walker, T., Matthews, J., & Murdock, J. (1999). Preliminary investigations into the use of a human bronchial cell line (16HBE14o-) to screen for respiratory toxins in vitro. Toxicol. in Vitro, 13(4/5), 761-764.

Widdicombe, J. H. (2002). Volume of airway surface liquid in health and disease. Am. J.

Respir. Crit. Care Med., 165(11), 1566.

Williams, A. S., & Taylor, G. (1992). Synthesis, characterization and release of cromoglycate

from dextran conjugates. Int. J. Pharm., 83(1-3), 233-239.

Wilson, B., Samanta, M. K., Santhi, K., Kumar, K. P. S., Ramasamy, M., Suresh, B. (2010).

Chitosan nanoparticles as a new delivery system for the anti-Alzheimer drug tacrine. Nanomedicine (Philadelphia, PA, U. S.), 6(1), 144-152.

Witschi, C., & Mrsny, R. J. (1999). In vitro evaluation of microparticles and polymer gels for

use as nasal platforms for protein delivery. Pharm. Res., 16(3), 382-390.

Wong, C. K., Li, M. L. Y., Wang, C. B., Ip, W. K., Tian, Y. P., Lam, C. W. K. (2006). House dust

mite allergen Der p 1 elevates the release of inflammatory cytokines and expression of adhesion molecules in co-culture of human eosinophils and bronchial epithelial cells. Int. Immunol., 18(8), 1327-1335.

Wong, J. P., Yang, H., Blasetti, K. L., Schnell, G., Conley, J., Schofield, L. N. (2003). Liposome

delivery of ciprofloxacin against intracellular Francisella tularensis infection. J. Controlled Release, 92(3), 265-273.

Wu, L., Bharatwaj, B., Panyam, J., & da Rocha, S. R. P. (2008). Core-shell Particles for the

Dispersion of Small Polar Drugs and Biomolecules in Hydrofluoroalkane Propellants. Pharm. Res., 25(2), 289-301.

Xu, J., Ma, L., Liu, Y., Xu, F., Nie, J., Ma, G. (2012). Design and characterization of antitumor

drug paclitaxel-loaded chitosan nanoparticles by W/O emulsions. Int. J. Biol. Macromol. , 50(2), 438-443.

Xu, Z., & Hickey, A. J. (2013). A comparison of aerosol performance using standardized

entrainment tubes vs. dry powder inhaler devices. KONA Powder Part. J., 30, 201-210.

Yadav, H. K. S., Joshi, G. B., Singh, M. N., & Shivakumar, H. G. (2011). Naturally occurring

chitosan and chitosan derivatives: a review. Curr. Drug Ther., 6(1), 2-11.

Page 264: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

210

Yaghobi, N. (2012). Controlling chitosan's molecular weight via multistage deacetylation.

Biotechnol. Bioprocess Eng., 17(4), 812-817.

Yamamoto, H., Kuno, Y., Sugimoto, S., Takeuchi, H., & Kawashima, Y. (2005). Surface-modified PLGA nanosphere with chitosan improved pulmonary delivery of calcitonin by mucoadhesion and opening of the intercellular tight junctions. J. Controlled Release, 102(2), 373-381.

Yan, Y., Zhang, Z., Li, Y., Zhao, X., Wang, X., Zhui, D. (2007). Effect of diltiazem on

proliferation of VSMC Chinese Journal of Gerontology, 27(23), 2280-2282.

Yeung, P. K. F., Prescott, C., Haddad, C., Montague, T. J., McGregor, C., Quilliam, M. A., Xei,

M., Li, R., Farmer, P., Klassen, G. A. (1993). Pharmacokinetics and metabolism of diltiazem in healthy males and females following a single oral dose. Eur. J. Drug Metab. Pharmacokinet. , 18(2), 199-206.

Yi, S.-S., Noh, J.-M., & Lee, Y.-S. (2009). Amino acid modified chitosan beads: Improved

polymer supports for immobilization of lipase from Candida rugosa. J. Mol. Catal. B: Enzym., 57(1-4), 123-129.

Yoksan, R., Akashi, M., Biramontri, S., & Chirachanchai, S. (2001). Hydrophobic Chain

Conjugation at Hydroxyl Group onto γ-Ray Irradiated Chitosan. Biomacromolecules, 2(3), 1038-1044.

Yoksan, R., Akashi, M., Hiwatari, K.-i., & Chirachanchai, S. (2003). Controlled

hydrophobic/hydrophilicity of chitosan for spheres without specific processing technique. Biopolymers, 69(3), 386-390.

Young, P. M., Edge, S., Traini, D., Jones, M. D., Price, R., El-Sabawi, D., Urry, C., Smith, C.

(2005). The influence of dose on the performance of dry powder inhalation systems. Int. J. Pharm. , 296(1-2), 26-33.

Young, P. M., Roberts, D., Chiou, H., Rae, W., Chan, H.-K., Traini, D. (2008). Composite

carriers improve the aerosolisation efficiency of drugs for respiratory delivery. J. Aerosol Sci., 39(1), 82-93.

Yu, H., Wang, W., Chen, X., Deng, C., & Jing, X. (2006). Synthesis and characterization of the

biodegradable polycaprolactone-graft-chitosan amphiphilic copolymers. Biopolymers, 83(3), 233-242.

Page 265: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Bibliography

211

Zhang, C., Ping, Q., Zhang, H., & Shen, J. (2003). Synthesis and characterization of water-soluble O-succinyl-chitosan. Eur. Polym. J., 39(8), 1629-1634.

Zhang, F., Bernet, B., Bonnet, V., Dangles, O., Sarabia, F., Vasella, A. (2008a). 2-Azido-2-

deoxycellulose: synthesis and 1,3-dipolar cycloaddition. Helv. Chim. Acta, 91(4), 608-618.

Zhang, W. F., Chen, X. G., Li, P. W., He, Q. Z., & Zhou, H. Y. (2008b). Preparation and

characterization of theophylline loaded chitosan/β-cyclodextrin microspheres. J. Mater. Sci.: Mater. Med., 19(1), 305-310.

Zhang, W. F., Zhou, H. Y., Chen, X. G., Tang, S. H., & Zhang, J. J. (2009). Biocompatibility

study of theophylline/chitosan/β-cyclodextrin microspheres as pulmonary delivery carriers. J. Mater. Sci.: Mater. Med., 20(6), 1321-1330.

Zhang, Y., Cai, X., Wang, Y., Zhang, C., Li, L., Choi, S.-W., Wang, L. V., Xia, Y. (2011).

Noninvasive Photoacoustic Microscopy of Living Cells in Two and Three Dimensions through Enhancement by a Metabolite Dye. Angew. Chem., Int. Ed., 50(32), 7359-7363, S7359/7351-S7359/7359.

Zhao, M., You, Y., Ren, Y., Zhang, Y., & Tang, X. (2008). Formulation, characteristics and

aerosolization performance of azithromycin DPI prepared by spray-drying. Powder Technol., 187(3), 214-221.

Page 266: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 267: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 268: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the
Page 269: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

212

Appendix 5-1

Figure A5-1. 1: FT-IR (ATR) Spectrum of N-Phthaloyl-Chitosan

Figure A5-1. 2: FT-IR (ATR) Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan

Page 270: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

213

Figure A5-1. 3: FT-IR (ATR) Spectrum of N-Phthaloyl-6-O-Trityl-Chitosan

Figure A5-1. 4: FT-IR (ATR) Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan

Page 271: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

214

Figure A5-1. 5: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan

Figure A5-1. 6: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan [run with hydrazine hydrate solution (40-60%), diluted further with water (1:1)]

Page 272: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

215

Figure A5-1. 7: FT-IR (ATR) Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine

Figure A5-1. 8: FT-IR (ATR) Spectrum of Chitosan-N-L-Leucine.HCl

Page 273: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

216

Appendix 5-2

Figure A5-2. 1: 1H NMR Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan in CDCl3

Figure A5-2. 2: 1H NMR Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan in CDCl3

Page 274: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

217

Figure A5-2. 3: 1H NMR Spectrum of 6-O-Trityl-Chitosan in Pyridine-d5

Figure A5-2. 4: 1H NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5

Page 275: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

218

Figure A5-2. 5: 1H NMR spectrum of Chitosan-N-L-leucine.HCl in D2O

Page 276: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

219

Appendix 5-3

Figure A5-3. 1: 13C NMR Spectrum of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan in CDCl3

Figure A5-3. 2: 13C NMR Spectrum of N-Phthaloyl-3-O-Acetyl-6-O-Trityl-Chitosan in CDCl3

Page 277: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

220

Figure A5-3. 3: 13C NMR Spectrum of 6-O-Trityl-Chitosan-N-Boc-L-Leucine in Pyridine-d5

Figure A5-3. 4: 13C NMR Spectrum of Chitosan-N-L-Leucine.HCl in D2O

Page 278: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

221

Appendix 5-4

Figure A5-4. 1: Determination of Degree of Substitution (DS) of N-Phthaloyl-3,6-Di-O-Acetyl-Chitosan from C/N Ratio Obtained by Elemental Analysis

Figure A5-4. 2: Determination of Degree of Trityl Substitution (DS) of N-Phthaloyl-6-O-Trityl-Chitosan from C/N Ratio Obtained by Elemental Analysis

0.00

0.50

1.00

1.50

2.00

2.50

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16

Degr

ee o

f Sub

stitu

tion

(DS)

C/N ratio

14.89

1.97

Page 279: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

222

Figure A5-4. 3: Determination of Degree of Dephthaloylation of 6-O-Trityl-Chitosan from C/N Ratio Obtained by Elemental Analysis

Figure A5-4. 4: Determination of Degree of Substitution of 6-O-Trityl-Chitosan-N-Boc- L-Leucine from C/N Ratio Obtained by Elemental Analysis

Figure A5-4. 5: Determination of Degree of Trityl and Boc Deprotection in Chitosan- N-L-Leucine.HCl from C/N Ratio Obtained by Elemental Analysis

Page 280: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

223

Appendix 5-5

Figure A5-5. 1: XPS Survey spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl

Figure A5-5. 2: XPS Multiplex spectra of Chitosan, L-Leucine and Chitosan-N-L-Leucine.HCl for Oxygen

Page 281: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

224

Appendix 6

Table A6- 1: Zetasizer Analysis of Blank and Drug-loaded Chitosan and Conjugate Nanoparticles

Method of Preparation Formulations Peaks Size (r.nm) % Intensity Width (r.nm) Z-Average (r.nm) PDI

Method-1

Blank Chitosan Nanoparticles Peak-1 48.96±1.56 100.00±0.00 7.48±0.41 67.25±2.82 0.58±0.26

Blank Conjugate

Nanoparticles Peak-1 70.28±2.06 100.00±0.00 12.62±0.57 76.61±1.23 0.35±0.08

Method-2

Blank Chitosan Nanoparticles

Peak-1 16.46±1.01 17.47±0.69 1.9±0.14 70.34±3.99 1.00±0.00

Peak-2 194.37±10.6 82.53±0.69 44.74±2.79

Blank Conjugate

Nanoparticles

Peak-1 9.93±0.19 22.27±0.82 1.40±0.07 21.17±0.52 1.00±0.00

Peak 2 254.77±9.55 77.73±0.82 65.75±4.6

Drug-loaded Chitosan

Nanoparticles

Peak-1 16.03±0.66 23.2±1.92 2.22±0.09 32.23±1.29 1.00±0.00

Peak 2 262.77±13.09 76.80±1.92 66.9±3.18

Drug-loaded Conjugate

Nanoparticles

Peak-1 0.49±0.01 16.1±0.55 0.11±0.01

9.26±0.99 1.00±0.00 Peak 2 15.02±0.93 9.83±0.43 3.28±0.38

Peak 3 248.57±10.67 74.07±0.88 93.68±6.08

Note: Data represent mean ± SE (n=3). The sizes are presented as r.nm (hydrodynamic radius in nm).

Page 282: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

225

Table A6- 2: In Vitro Release of Diltiazem HCl from Chitosan and Conjugate Nanoparticles in PBS at 37 °C

Formulation Chitosan nanoparticle Conjugate nanoparticle

Time Points (h) Cumulative % Drug Released

Cumulative % Drug Remaining

Cumulative % Drug Released

Cumulative % Drug Remaining

0.5 16±0.32 84±0.55 31±1.98 69±3.42

1 17±0.3 83±0.53 32±1.83 68±3.18

2 17±0.33 83±0.58 33±2.05 67±3.55

4 18±0.27 82±0.46 33±1.99 67±3.45

6 18±0.3 82±0.53 34±1.91 66±3.31

12 18±0.32 82±0.55 36±2.18 64±3.77

24 19±0.3 81±0.52 38±1.73 62±3

48 20±0.35 80±0.61 41±1.78 59±3.09

72 21±0.35 79±0.61 43±1.74 57±3.02

96 21±0.31 79±0.54 45±1.52 55±2.64

120 22±0.31 78±0.54 46±1.78 54±3.09

144 22±0.34 78±0.58 47±1.67 53±2.89

168 22±0.3 78±0.53 48±1.67 52±2.89

192 23±0.35 77±0.6 49±1.59 51±2.76

216 23±0.32 77±0.56 50±1.67 50±2.89

240 23±0.31 77±0.54 50±1.58 50±2.73

264 23±0.38 77±0.66 51±1.56 49±2.71

288 23±0.37 77±0.63 51±1.61 49±2.79

312 23±0.38 77±0.65 51±1.49 49±2.57

336 23±0.34 77±0.59 51±1.53 49±2.65

360 23±0.35 77±0.6 51±1.4 49±2.43

384 23±0.37 77±0.63 52±1.55 48±2.69

408 23±0.28 77±0.49 52±1.53 48±2.65

432 23±0.36 77±0.62 52±1.56 48±2.7

456 23±0.32 77±0.56 52±1.53 48±2.66

480 23±0.36 77±0.62 52±1.47 48±2.55

504 23±0.32 77±0.56 52±1.34 48±2.33

528 23±0.35 77±0.6 52±1.37 48±2.37

552 23±0.33 77±0.57 52±1.42 48±2.46

576 23±0.3 77±0.53 53±1.49 47±2.58

600 23±0.29 77±0.49 52±1.49 48±2.59

624 23±0.38 77±0.66 52±1.48 48±2.56

648 23±0.29 77±0.5 52±1.41 48±2.44

672 23±0.31 77±0.53 52±1.46 48±2.52

696 23±0.34 77±0.59 52±1.45 48±2.5

720 23±0.3 77±0.52 53±1.52 47±2.63

Note: Data represent mean ± SE (n=3).

Page 283: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

226

Table A6- 3: Aerosolization Study of Blank and Drug-Loaded Chitosan and Conjugate Nanoparticles Using Twin-Stage Impinger (TSI)

Method Formulation

% Deposition Recovered Dose (RD)

(%)

Emitted Dose (ED)

(%)

Fine Particle Fraction

(FPF) Rotahaler (R)

Stage-1 (S1)

Stage-2 (S2)

Gravimetric analysis

Blank chitosan

nanoparticle 38±0.4 43±1.4 19±1.01 84±1.32 62±0.39 19±1.01

Blank conjugate

nanoparticle 15±0.9 61±0.2 24±0.8 86±0.73 85±0.94 24±0.8

Drug-loaded chitosan

nanoparticle 27±2 58±2 15±1.5 81±1.92 73±1.96 15±1.5

Drug-loaded conjugate

nanoparticle 22±2.5 57±3.2 21±0.7 74±2.52 79±2.52 21±0.7

Spectrophotometric

analysis

Drug-loaded chitosan

nanoparticle 28±2.2 56±2.2 16±1.6 81±1.66 72±2.15 16±1.6

Drug-loaded conjugate

nanoparticle 21±2.4 58±3.1 21±0.7 87±2.97 79±2.39 21±0.7

Note: Data represent mean ± SE (n=3).

Page 284: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

227

Appendix 7-1

Table A7-1.1 a: MTT Assay of Chitosan on BEAS-2B Cell Line

Duration % Survival at different concentrations of the treatment (mean ± SE, n=3)

0.125 mg/mL 0.25 mg/mL 0.375 mg/mL 0.5 mg/mL 1 mg/mL 2 mg/mL 4 mg/mL 8 mg/mL 12 mg/mL 16 mg/mL

12 hour 99±0.36 94±0.58 96±0.03 91±2.05 93±1.77 88±3.74 85±1.81 85±2.50 82±1.97 83±3.58

24 hour 99±1.09 94±0.88 85±5.13 84±6.00 78±5.39 81±2.58 70±1.21 77±2.51 71±6.38 66±2.91

48 hour 97±0.62 85±0.29 84±1.79 82±4.22 84±1.80 84±2.38 70±3.52 68±3.49 64±2.30 53±1.54

Table A7-1.1 b: MTT Assay of Chitosan Nanoparticles on BEAS-2B Cell Line

Duration % Survival at different concentrations of the treatment (mean ± SE, n=3)

0.125 mg/mL 0.25 mg/mL 0.375 mg/mL 0.5 mg/mL 1 mg/mL 2 mg/mL 4 mg/mL 8 mg/mL 12 mg/mL 16 mg/mL

12 hour 98±8.66 93±1.39 101±6.57 90±9.37 88±10.36 77±6.61 60±2.41 10±6.06 1±3.03 -3±5.01

24 hour 102±10.55 101±3.38 96±2.87 104±3.86 95±6.63 85±5.08 84±8.80 11±4.50 -4±0.41 -1±0.46

48 hour 69±2.15 67±4.29 56±7.05 52±6.15 51±3.90 56±1.63 45±2.70 12±3.04 -1±1.29 -2±1.14

Table A7-1.1 c: MTT Assay of Chitosan-L-Leucine Conjugate on BEAS-2B Cell Line

Duration % Survival at different concentrations of the treatment (mean ± SE, n=3)

0.125 mg/mL 0.25 mg/mL 0.375 mg/mL 0.5 mg/mL 1 mg/mL 2 mg/mL 4 mg/mL 8 mg/mL 12 mg/mL 16 mg/mL

12 hour 82±1.62 79±0.38 78±2.48 83±2.81 78±1.18 77±2.59 59±2.64 32±2.25 5±0.78 6±0.56

24 hour 69±2.74 73±1.56 72±0.55 69±1.49 69±3.00 60±2.34 36±1.44 7±2.52 3±0.64 5±0.68

48 hour 80±2.82 72±0.48 73±1.21 69±0.57 64±4.98 54±1.50 23±2.99 5±2.81 2±1.11 2±0.90

Table A7-1.1 d: MTT Assay of Chitosan-L-Leucine Conjugate Nanoparticles on BEAS-2B Cell Line

Duration % Survival at different concentrations of the treatment (mean ± SE, n=3)

0.125 mg/mL 0.25 mg/mL 0.375 mg/mL 0.5 mg/mL 1 mg/mL 2 mg/mL 4 mg/mL 8 mg/mL 12 mg/mL 16 mg/mL

12 hour 72±3.51 71±2.91 66±4.97 63±4.44 53±7.01 56±4.03 50±4.64 39±1.62 9±0.48 8±0.91

24 hour 80±5.90 76±2.85 74±0.92 68±1.92 59±2.66 49±1.75 36±0.90 21±0.72 5±0.35 5±0.97

48 hour 66±1.06 64±3.36 57±3.19 53±0.11 43±3.59 38±2.80 25±2.63 5±1.50 2±0.75 2±0.60

Table A7-1. 2: MTT Assay of Diltiazem HCl on BEAS-2B Cell Line

Duration % Survival at different concentrations of the treatment (mean ± SE, n=3)

0.125 mg/mL 0.25 mg/mL 0.375 mg/mL 0.5 mg/mL 1 mg/mL 2 mg/mL 4 mg/mL 8 mg/mL 12 mg/mL 16 mg/mL

12 hour 124±17.30 139±9.90 112±3.23 29±2.54 5±0.19 5±0.40 5±0.40 5±0.19 5±0.26 6±0.71

24 hour 122±3.01 119±5.54 89±2.54 10±0.12 7±0.29 6±0.81 4±0.22 4±0.09 4±0.17 4±0.21

48 hour 93±3.89 81±0.58 63±2.94 0±1.45 0±1.19 0±1.19 0±1.30 0±1.19 1±1.20 2±1.03

Page 285: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

228

Appendix 7-2

Table A7-2. 1: TEER of BEAS-2B Cell Lines Grown on Transwell Inserts at Different Time Intervals

Day TEER (ohm-cm2)

1 2 3 Mean SE 1 3.74 1.54 3.74 3.01 0.73 2 4.07 3.52 6.27 4.62 0.84 3 4.18 6.49 10.56 7.08 1.86 4 12.65 16.83 10.23 13.24 1.93 5 8.25 12.54 5.50 8.76 2.05 6 9.24 8.91 1.87 6.67 2.40 7 4.40 3.41 3.96 3.92 0.29 8 3.08 3.63 4.73 3.81 0.49 9 2.31 1.10 1.87 1.76 0.35

10 0.66 1.10 2.20 1.32 0.46

Table A7-2. 2: Permeability of Na Flu across a Blank Transwell and a Transwell Containig Confluent BEAS-2B Monolayer

Type of Transwell Apparent Permeability Coefficient, Papp ( nm/s)

1 2 3 Mean SE

Blank Transwell 711 675 722 703 14

Transwell containing polarized BEAS-2B monolayer

108 107 115 110 2

Table A7-2.3 a: Effect of Chitosan on Na Flu Transport across the BEAS-2B Monolayer Concentration Time interval → 0.5 h 1 h 2 h 4 h 6 h 12 h 24 h 48 h

0 mg/mL Cumulative amount of Na Flu transported (mg) 0.0003±0 0.001±0 0.0016±0 0.0024±0 0.0037±0.0001 0.0053±0.0001 0.006±0 0.0059±0.0002 Apparent permeability coefficient, Papp (nm/s) 33±2.37 45±2.2 33±0.44 25±0.58 26±1.29 18±0.68 10±0.16 5±0.19

0.5 mg/mL Cumulative amount of Na Flu transported (mg) 0.0003±0 0.0009±0 0.0016±0.0001 0.0023±0.0001 0.0033±0.0001 0.0054±0.0002 0.0063±0.0001 0.0067±0.0002 Apparent permeability coefficient, Papp (nm/s) 33±1.9 40±1.68 33±2.22 24±1.32 23±1.16 19±0.73 11±0.33 5±0.19 Papp,polymer/Papp,control 1.01±0.13 0.9±0.04 1±0.05 0.94±0.06 0.9±0.04 1.02±0.01 1.05±0.02 1.13±0.01

1 mg/mL Cumulative amount of Na Flu transported (mg) 0.0004±0 0.0009±0 0.0015±0 0.0024±0.0001 0.0033±0.0001 0.0055±0.0001 0.0062±0.0002 0.0064±0.0002 Apparent permeability coefficient, Papp (nm/s) 36±0.62 40±2.64 32±1.3 25±1.47 23±0.96 19±0.58 10±0.46 5±0.17 Papp,polymer/Papp,control 1.1±0.08 0.89±0.02 0.95±0.05 1±0.03 0.9±0.04 1.04±0.04 1.04±0.03 1.1±0

2 mg/mL Cumulative amount of Na Flu transported (mg) 0.0003±0 0.0009±0 0.0014±0 0.0022±0.0001 0.0032±0 0.0056±0.0001 0.006±0.0002 0.0064±0.0003 Apparent permeability coefficient, Papp (nm/s) 28±6.54 40±1.33 30±0.6 24±1.36 22±0.49 19±0.64 10±0.36 5±0.26 Papp,polymer/Papp,control 0.83±0.13 0.89±0.01 0.91±0.01 0.93±0.04 0.87±0.03 1.06±0.02 1.01±0.04 1.09±0.01

4 mg/mL Cumulative amount of Na Flu transported (mg) 0.0003±0 0.0008±0 0.0013±0 0.0023±0 0.003±0.0001 0.0051±0 0.0061±0.0002 0.0066±0.0002 Apparent permeability coefficient, Papp (nm/s) 27±4.46 36±1.59 28±0.69 24±0.41 21±1.28 17±0.25 10±0.4 5±0.18 Papp,polymer/Papp,control 0.8±0.1 0.79±0 0.86±0.03 0.96±0.01 0.82±0.02 0.95±0.02 1.02±0.03 1.12±0.04

Note: The values represent mean of 3 replicates ± SE

Table A7-2.3 b: Effect of Chitosan Nanoparticles on Na Flu Transport across the BEAS-2B Monolayer

Concentration Time interval→ 0.5 h 1 h 2 h 4 h 6 h 12 h 24 h 48 h

0 mg/mL Cumulative amount of Na Flu transported (mg) 0.0057±0.0001 0.0073±0 0.0093±0.0001 0.0107±0 0.0155±0.0004 0.0212±0.0004 0.0255±0.0002 0.0241±0.0005 Apparent permeability coefficient, Papp (nm/s) 483±13.48 310±3.92 197±3.72 113±0.88 109±3.47 74±1.62 44±0.41 21±0.46

0 .5 mg/mL Cumulative amount of Na Flu transported (mg) 0.0052±0 0.0072±0.0002 0.0092±0.0002 0.0108±0.0004 0.0159±0.0005 0.0225±0.0004 0.0258±0 0.0253±0.0004 Apparent permeability coefficient, Papp (nm/s) 442±7.33 303±9.95 193±4.75 114±5.17 111±4.02 79±1.69 45±0 22±0.36 Papp,polymer/Papp,control 0.91±0.01 0.97±0.03 0.98±0.04 1.01±0.04 1.02±0.02 1.06±0.02 1.01±0 1.05±0.01

1 mg/mL Cumulative amount of Na Flu transported (mg) 0.0042±0.0001 0.0066±0.0003 0.0086±0.0004 0.0104±0.0004 0.0152±0.0005 0.0222±0.0003 0.0258±0 0.0257±0.0001 Apparent permeability coefficient, Papp (nm/s) 356±10.91 281±14.52 181±8.94 110±4.84 106±3.62 77±1.34 45±0.05 22±0.13 Papp,polymer/Papp,control 0.73±0.01 0.9±0.05 0.92±0.05 0.97±0.04 0.98±0.02 1.04±0.03 1±0.01 1.06±0.01

2 mg/mL Cumulative amount of Na Flu transported (mg) 0.0034±0 0.0062±0.0002 0.0086±0.0003 0.0101±0.0004 0.0143±0.0006 0.0215±0.0006 0.0258±0 0.0258±0 Apparent permeability coefficient, Papp (nm/s) 291±7.53 263±8.97 182±8.25 106±4.61 100±4.23 75±2.23 45±0.07 22±0 Papp,polymer/Papp,control 0.6±0.01 0.84±0.03 0.92±0.03 0.94±0.04 0.92±0.02 1.01±0.04 1.01±0.01 1.07±0.02

4 mg/mL Cumulative amount of Na Flu transported (mg) 0.003±0.0001 0.0055±0.0001 0.0074±0 0.009±0.0001 0.0117±0.0003 0.0205±0.0003 0.0256±0.0002 0.0255±0.0003 Apparent permeability coefficient, Papp (nm/s) 258±13.97 234±4.81 157±0.75 95±1.14 82±2.11 72±1.2 44±0.4 22±0.29 Papp,polymer/Papp,control 0.53±0.03 0.75±0.02 0.79±0.01 0.84±0.01 0.75±0.03 0.96±0.03 1±0 1.05±0.01

Note: The values represent mean of 3 replicates ± SE

Page 286: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

229

Table A7-2.3 c: Effect of Chitosan-L-Leucine Conjugate on Na Flu Transport across the BEAS-2B Monolayer

Concentration Time interval→ 0.5 h 1 h 2 h 4 h 6 h 12 h 24 h 48 h

0 mg/mL Cumulative amount of Na Flu transported (mg) 0.0008±0 0.0018±0 0.0024±0.0001 0.004±0 0.005±0.0001 0.0078±0.0001 0.0101±0.0001 0.011±0

Apparent permeability coefficient, Papp (nm/s) 68±5.04 78±3.62 51±2.81 42±0.91 35±1.16 27±0.45 17±0.22 9±0.08

0.5 mg/mL Cumulative amount of Na Flu transported (mg) 0.0008±0 0.0019±0 0.0025±0.0001 0.004±0.0002 0.0051±0.0002 0.008±0.0002 0.0103±0.0001 0.011±0.0002

Apparent permeability coefficient, Papp (nm/s) 74±6.98 81±0.9 54±2.78 42±2.32 35±2.01 28±0.86 18±0.24 9±0.24 Papp,polymer/Papp,control 1.1±0.1 1.04±0.05 1.07±0.1 1±0.03 1±0.02 1.03±0.01 1.01±0.02 1±0.02

1 mg/mL Cumulative amount of Na Flu transported (mg) 0.0008±0 0.0017±0 0.0023±0 0.004±0.0002 0.0047±0 0.0078±0.0001 0.0103±0.0003 0.011±0.0001

Apparent permeability coefficient, Papp (nm/s) 71±5.66 74±0.11 50±1.9 42±2.34 33±0.34 27±0.46 18±0.54 9±0.1

Papp,polymer/Papp,control 1.04±0.04 0.94±0.04 0.99±0.09 1.01±0.05 0.92±0.02 0.99±0.01 1.02±0.01 1±0

2 mg/mL Cumulative amount of Na Flu transported (mg) 0.0008±0 0.0018±0.0001 0.0024±0.0001 0.0039±0 0.0048±0.0002 0.0078±0 0.0101±0.0001 0.0111±0.0001

Apparent permeability coefficient, Papp (nm/s) 74±3.02 77±4.97 51±2.16 41±0.93 33±1.56 27±0.27 17±0.21 9±0.09

Papp,polymer/Papp,control 1.1±0.04 0.98±0.01 1.01±0.03 0.97±0.03 0.94±0.01 1±0.02 1±0 1.01±0.01

4 mg/mL Cumulative amount of Na Flu transported (mg) 0.0008±0 0.0019±0 0.0024±0.0001 0.0038±0.0002 0.0045±0.0001 0.0076±0.0002 0.0097±0.0003 0.0113±0.0002 Apparent permeability coefficient, Papp (nm/s) 69±4.47 81±3.45 52±2.69 41±2.95 32±1.01 26±0.8 17±0.7 9±0.21

Papp,polymer/Papp,control 1.01±0.03 1.04±0.08 1.03±0.1 0.96±0.04 0.89±0 0.97±0.01 0.96±0.04 1.02±0.02

Note: The values represent mean of 3 replicates ± SE

Table A7-2.3 d: Effect of Chitosan-L-Leucine Conjugate Nanoparticles on Na Flu Transport across the BEAS-2B Monolayer

Concentration Time interval→ 0.5 h 1 h 2 h 4 h 6 h 12 h 24 h 48 h

0 mg/mL Cumulative amount of Na Flu transported (mg) 0.0013±0 0.0028±0 0.0038±0.0001 0.0043±0 0.0059±0.0002 0.008±0.0001 0.0088±0 0.0088±0

Apparent permeability coefficient, Papp (nm/s) 110±2.46 118±3.73 80±3.41 45±0.68 42±1.51 28±0.41 15±0.17 7±0.07

0.5 mg/mL Cumulative amount of Na Flu transported (mg) 0.0001±0 0.0002±0 0.0004±0 0.0005±0 0.0009±0 0.0018±0 0.0023±0 0.0024±0

Apparent permeability coefficient, Papp (nm/s) 12±0.1 12±0.62 9±0.75 6±0.34 6±0.29 6±0.09 4±0.1 2±0.02

Papp,polymer/Papp,control 0.11±0 0.1±0 0.12±0.01 0.13±0 0.16±0 0.23±0 0.26±0 0.27±0

1 mg/mL Cumulative amount of Na Flu transported (mg) 0.0003±0 0.0006±0 0.0008±0 0.0011±0 0.0014±0 0.0031±0.0002 0.004±0 0.0041±0.0002 Apparent permeability coefficient, Papp (nm/s) 26±1.58 27±1.08 18±0.8 12±0.1 10±0.4 11±0.7 7±0.1 3±0.2

Papp,polymer/Papp,control 0.23±0.01 0.22±0 0.22±0 0.26±0 0.24±0.01 0.39±0.02 0.46±0 0.46±0.02

2 mg/mL Cumulative amount of Na Flu transported (mg) 0.0006±0 0.0013±0 0.0017±0 0.0024±0 0.0036±0.0001 0.0056±0 0.0066±0.0001 0.0069±0.0001

Apparent permeability coefficient, Papp (nm/s) 58±5.9 57±1.46 37±1.13 25±0.44 25±0.9 19±0.29 11±0.24 6±0.11 Papp,polymer/Papp,control 0.53±0.06 0.48±0.02 0.46±0.03 0.55±0.01 0.6±0 0.7±0.01 0.75±0.02 0.79±0.01

4 mg/mL Cumulative amount of Na Flu transported (mg) 0.0011±0 0.0025±0.0001 0.0032±0 0.0041±0 0.0048±0.0001 0.0067±0.0001 0.0086±0.0001 0.0089±0

Apparent permeability coefficient, Papp (nm/s) 99±3.07 107±6.32 68±1.29 43±0.96 33±1.4 23±0.59 15±0.2 7±0.01

Papp,polymer/Papp,control 0.89±0.02 0.9±0.06 0.84±0.02 0.95±0.03 0.8±0.04 0.84±0.03 0.98±0 1.01±0.01

Note: The values represent mean of 3 replicates ± SE

Page 287: Preparation and In Vitro Evaluation of a Polymer … Didare Alam...lung delivery than nebulizers/ metered dose inhalers (MDIs) and gained significant attention of researchers in the

Appendices

230

Appendix 7-3

Table A7- 3: ELISA for IL-8 Released by BEAS-2B Cells upon Treatment with Chitosan, its L-Leucine Conjugate and their Nanoparticles

Concentration of Treatment (mg/mL)

Concentration of IL-8 (pg/mL)

Chitosan Chitosan nanoparticles Conjugate Conjugate

nanoparticles 0 557±64 451±96 444±100

0.5 1105±86 1416±41 789±68 917±50 1 1262±79 1380±9 861±90 2230±251 2 1346±140 1136±61 1347±79 2957±150 4 1518±98 1230±64 1070±54 1959±64

N.B. The values represent mean of 3 replicates ± SE


Recommended