dendrimer functionalized magnetic nanoparticles as …...(saumya nigam) (iitb: 09411414) (mu:...

175
Dendrimer functionalized magnetic nanoparticles as promising platforms for cancer theranostics Submitted in the partial fulfilment of the requirements of the degree of Doctor of Philosophy of the Indian Institute of Technology Bombay, India and Monash University, Australia by Saumya Nigam Supervisors: Prof. D. Bahadur (IIT Bombay) Dr. X. Chen (Monash University) The course of study for this award was developed jointly by the Indian Institute of Technology, Bombay and Monash University, Australia and given academic recognition by each of them. The programme was administered by The IITB-Monash Research Academy (Year 2016)

Upload: others

Post on 02-Mar-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

Dendrimer functionalized magnetic nanoparticles

as promising platforms for cancer theranostics

Submitted in the partial fulfilment of the requirements of the degree of

Doctor of Philosophy

of the

Indian Institute of Technology Bombay, India

and

Monash University, Australia

by

Saumya Nigam

Supervisors:

Prof. D. Bahadur (IIT Bombay)

Dr. X. Chen (Monash University)

The course of study for this award was developed jointly by

the Indian Institute of Technology, Bombay and Monash University, Australia

and given academic recognition by each of them.

The programme was administered by The IITB-Monash Research Academy

(Year 2016)

Page 2: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

Dedicated to...

My Grandfather

Page 3: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical
Page 4: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

Declaration

I declare that this written submission represents my ideas in my own words and where others’

ideas or words have been included, I have adequately cited and referenced the original

sources. I also declare that I have adhered to all principles of academic honesty and integrity

and have not misrepresented or fabricated or falsified any idea/data/fact/source in my

submission. I understand that any violation of the above will be cause for disciplinary action

by the Institute and can also evoke penal action from the sources which have thus not been

properly cited or from whom proper permission has not been taken when needed.

Notice 1

Under the Copyright Act 1968, this thesis must be used only under the normal conditions of

scholarly fair dealing. In particular no results or conclusions should be extracted from it, nor

should it be copied or closely paraphrased in whole or in part without the written consent of

the author. Proper written acknowledgement should be made for any assistance obtained

from this thesis.

Notice 2

I certify that I have made all reasonable efforts to secure copyright permissions for third-

party content included in this thesis and have not knowingly added copyright content to my

work without the owner’s permission.

(Saumya Nigam)

(IITB: 09411414)

(MU: 22667563)

Date: 24.10.2016

Page 5: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

i

Abstract

Cancer therapeutics deals with development of preclinical therapeutic drugs for

cancer which are developed through innovative research and technologies. This itself is a

very challenging task and it involves various approaches to prevent, diagnose and treat

cancer. Various functional nanomaterials have been developed for improving the efficiency

of therapeutic drugs against cancer. For use in biomedical applications, the nanomaterials

must exhibit unique properties like reduced size, aqueous stability, biocompatibility, and

interactive functional groups. The ‘active’ surfaces of nanoparticles could be modified by

organic or inorganic materials, such as macromolecules, bio-molecules, drugs, etc. Amongst

various functional materials, magnetic nanomaterials have emerged as versatile nanosystems

promising for the detection, diagnosis and treatment of cancers. Superparamagnetic iron

oxide (Fe3O4) nanoparticles have been thoroughly investigated as drug delivery vectors,

magnetic drug targeting agents, contrast agents in magnetic resonance imaging (MRI) and

hyperthermia treatment of cancer. Dendrimers are another emerging class of functional

nanomaterials which are hyper branched, mostly symmetrical polymers with repetitive

branching units. The presence of multiple functional groups makes them ideal candidates for

anchoring guest molecules and therefore, they are assessed as delivery vectors, MR imaging

agents, stabilizers of molecules, catalysis, sensing etc. Combining these two nanomaterials

would contribute towards the development of ‘smart’ and versatile nanosystems with desired

properties.

Citric acid functionalized Fe3O4 (CA–Fe3O4) and glutamic acid functionalized Fe3O4

(Glu–Fe3O4) aqueous colloidal magnetic nanoparticles were synthesized. The surface

engineering made the Fe3O4 hydrophilic and facilitated its aqueous suspensions. Their

successful synthesis was confirmed by X-Ray diffraction (XRD) studies while infrared

spectroscopy (FTIR) was used to confirm the surface modification. The electron

micrographs showed that the nanoparticles were spherically-shaped, evenly dispersed and

magnetometry confirmed their superparamagnetic nature (MS = 57 emu/g at 20 kOe). Time-

dependent calorimetric measurements determined the specific absorption rate (SAR) of CA–

Fe3O4 and Glu–Fe3O4 which is an important parameter in evaluating their heating efficacy

in the presence of alternating magnetic field (ACMF). The SAR of CA–Fe3O4 was found to

Page 6: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

ii

be 49.24 W/g at an applied field of 10 kA/m. On the other hand, SAR of Glu–Fe3O4

nanoparticles was ~134 W/g. These values strongly suggested that these nanoparticles could

also act as effective heating source for magnetic hyperthermia. Doxorubicin hydrochloride

(DOX) was used as a model drug to evaluate their performance for drug delivery. The DOX

molecules were released in substantial amounts in the mild acidic environments. These

nanoparticles showed no loss of cell proliferation activity with the mouse fibroblast (L929),

human cervical carcinoma (HeLa), oral carcinoma (KB), prostate cancer (PC-3) and human

breast cancer (MCF-7) cells.

A peptide dendrimer of second generation was synthesized by divergent method

following the condensation reaction of L-lysine and L-arginine as monomeric units. Its

structural characterization was carried out by various sophisticated techniques such as

nuclear magnetic resonance (NMR), FTIR and X-ray photoelectron spectroscopy (XPS).

The Glu–Fe3O4 were functionalized by various generations of polyamidoamine

(PAMAM) and as-prepared 2nd generation peptide dendrimer and their performances as

delivery vectors for cationic (doxorubicin) and anionic (epigallocatechin gallate) drugs were

assessed. It was seen that the drug loading and release efficiency increased with an increase

in the dendrimer generation. Further, the peptide dendrimers exhibited similar drug loading

efficiency as the PAMAM dendrimers, however, their drug release capacity was

significantly improved. These nanosystems were also found to be potentially effective in

magnetic hyperthermia and were thus used towards in vitro combinatorial chemo-thermo

therapy of cancer using human cervical cancer (HeLa) cells as exemplary model. For the

magnetic hyperthermia treatment, the exposure of these cells to ACMF for 10 min was

successful in reducing the viable cell population by 50% (LD50). While exploring the

combinatorial therapy, it was seen that DOX in synergism with magnetic hyperthermia,

enhanced therapeutic effects and successfully reduced the viable cell population to ~2%.

The in vivo studies investigated the performance of dendrimer functionalized Fe3O4

nanoparticles in a subcutaneous syngeneic murine melanoma model. The systemic exposure

of these nanoparticles (15 mg/kg body weight) caused changes in various blood and serum

parameters. This assisted with the information about their toxicity and non-specific uptake

by various organs. All the vital organs (heart, kidneys, lungs, liver, spleen, brain, stomach

and thigh muscles) showed no loss of activity. The biochemical parameters also remained

Page 7: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

iii

unaltered in the treated mice in comparison to the control mice population confirming a

healthy liver and renal activity. The atomic emission spectroscopy (ICP-AES) of the organs

showed that the magnetic nanoparticles were mainly accumulated in the liver, lung and

spleen of the mice with a meagre amount also seen in heart and kidneys. The evaluation of

efficient magnetic drug targeting (MDT) revealed ~6-fold accumulation of iron in the

tumour region when compared to the tumour of mice which were not exposed to the

magnetic field. This high localisation pattern led to high concentrations of DOX in the

tumour and thus was effective in arresting the tumour growth significantly. It was seen that

lower number of doses are sufficient to suppress the tumour growth in combination with

magnetic field than that required without the magnetic field. By the end of 14th day, the

average tumour volume was 55 ± 8.3 mm3 as compared to the control animals in which the

tumour volume was seen to be 4794 ± 844 mm3 (~88-fold decrease).

Furthermore, the dendrimer functionalized Fe3O4 nanoparticles were also seen to be

MR active and showed higher relaxivities when compared to the commercial magnetic

contrast agent. Various physico-chemical parameters potentiate and affect the MR contrast

properties. The relaxivities of the nanoparticles were evaluated under the varying parameters

of iron concentration, buffer environments (ultrapure water, buffered saline and simulated

body fluid) and temperatures (25, 37 and 45 °C). It was seen that under the conditions of

simulated body fluid environment at 37 °C, the peptide dendrimer functionalized Fe3O4

nanoparticles show higher r2 (spin-spin) relaxivity of 220 mM-1s-1. In vitro T2 weighted MR

imaging of dendrimer functionalized Fe3O4 treated (for various treatment times) HeLa cells

showed increased contrast when compared to the untreated cells. Owing to the high

magnetisation, high specific absorption rate and shorter transverse relaxation time, the

dendrimer functionalized Fe3O4 nanoparticles could be a suitable platform for MR imaging

and multimodal cancer theranostics.

Keywords: Cancer, Chemotherapy, Contrast agent, Dendrimers, Drug delivery, Functional

nanomaterials, Hyperthermia, Iron oxide, Magnetic drug targeting, Magnetic

nanoparticles, Magnetic resonance imaging, Superparamagnetism, Theranostics.

Page 8: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

iv

Table of Contents

Abstract ................................................................................................................................. i

List of Figures ................................................................................................................... viii

List of Tables .................................................................................................................... xiv

Abbreviations and Nomenclatures ................................................................................... xv

Chapter 1 .............................................................................................................................. 1

Functional Nanomaterials and their Applications in Cancer Theranostics ................... 1

1.1 Superparamagnetic Iron Oxide Nanoparticles ............................................................. 1

1.2 Dendrimers ................................................................................................................... 2

1.3 Cancer ........................................................................................................................... 3

1.4 Cancer Theranostics ..................................................................................................... 5

1.4.1 Chemotherapy ........................................................................................................ 7

1.4.2 Magnetic Hyperthermia and Chemo-thermotherapy ........................................... 13

1.4.3 Diagnostic Bio-imaging ....................................................................................... 16

1.5 Objectives of the Thesis ............................................................................................. 20

1.6 Outline of the Thesis .................................................................................................. 21

1.7 References .................................................................................................................. 21

Chapter 2 ............................................................................................................................ 28

Citrate-stabilised Fe3O4 Nanoparticles: Conjugation and Release of Doxorubicin for

Cancer Therapeutics .......................................................................................................... 28

2.1 Introduction ................................................................................................................ 28

2.2 Experimental and Characterisation Techniques ......................................................... 30

2.2.1 Synthesis of Citrate-stabilised Fe3O4 Nanoparticles ............................................ 30

2.2.2 Drug Loading and Release ................................................................................... 30

2.2.3 Calorimetric Measurements ................................................................................. 31

2.2.4 In vitro Evaluation ............................................................................................... 32

2.2.5 Characterisation Techniques ................................................................................ 32

2.3 Results and Discussions ............................................................................................. 33

2.3.1 Nanoparticle Characterisation.............................................................................. 33

2.3.2 Drug Loading and Release ................................................................................... 36

2.3.3 Calorimetric Measurements ................................................................................. 38

2.3.4 In vitro Evaluation ............................................................................................... 39

Page 9: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

v

2.4 Summary .................................................................................................................... 40

2.5 References .................................................................................................................. 40

Chapter 3 ............................................................................................................................ 44

PEG-Modified PAMAM-Fe3O4-drug Triads with the Potential for Improved

Therapeutic Efficacy .......................................................................................................... 44

3.1 Introduction ................................................................................................................ 44

3.2 Experimental Techniques ........................................................................................... 47

3.2.1 Synthesis and surface modification of Fe3O4 nanoparticles ................................ 47

3.2.2 Analysis of Drug Loading and Release ............................................................... 48

3.2.3 Evaluation of Carriers’ Biocompatibility and Therapeutic Efficacy ................... 49

3.3 Results and Discussions ............................................................................................. 49

3.3.1 Characterisation of Fe3O4 Nanoparticles ............................................................. 49

3.3.2 DOX Loading and Release .................................................................................. 53

3.3.3 In vitro Evaluation of Cellular Toxicity and the Therapeutic Effect ................... 57

3.4 Summary .................................................................................................................... 59

3.5 References .................................................................................................................. 60

Chapter 4 ............................................................................................................................ 65

Section A ............................................................................................................................. 65

4.1 Synthesis of Peptide Dendrimer and its in vitro Characteristics ................................ 65

4.1.1 Introduction .......................................................................................................... 65

4.1.2 Experimental and Characterisation Techniques .................................................. 67

4.1.3 Results and Discussions ....................................................................................... 69

4.1.4 Summary .............................................................................................................. 72

4.1.5 References ............................................................................................................ 73

Chapter 4 ............................................................................................................................ 75

Section B .............................................................................................................................. 75

4.2 Dendritic Fe3O4 Nanoparticles for Combinatorial Therapy: Peptide Dendrimers with

Enhanced Efficiency as Alternative Platforms for PAMAM Dendrimers ....................... 75

4.2.1 Introduction .......................................................................................................... 75

4.2.2 Experimental Techniques .................................................................................... 78

4.2.3 Results and Discussions ....................................................................................... 81

4.2.4 Summary .............................................................................................................. 95

Page 10: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

vi

4.2.5 References ............................................................................................................ 95

Chapter 5 ............................................................................................................................ 98

Assessment of Doxorubicin-loaded Dendritic Fe3O4 Nanoparticles for Magnetic Drug

Targeting in Murine Melanoma Model............................................................................ 98

5.1 Introduction ................................................................................................................ 98

5.2 Experimental and Characterisation Techniques ....................................................... 100

5.2.1 In vitro Evaluation ............................................................................................. 100

5.2.2 In vivo Therapeutic Efficacy Studies ................................................................. 101

5.3 Results and Discussion ............................................................................................. 106

5.3.1 In vitro Evaluation ............................................................................................. 106

5.3.2 In vivo Therapeutic Efficacy Studies ................................................................. 109

5.4 Summary .................................................................................................................. 115

5.5 References ................................................................................................................ 116

Chapter 6 .......................................................................................................................... 118

MR Contrast Properties of Dendritic Fe3O4 Nanoparticles ......................................... 118

6.1 Introduction .............................................................................................................. 118

6.2 Experimental Techniques ......................................................................................... 120

6.3 Results and Discussions ........................................................................................... 120

6.3.1 Relaxivity studies ............................................................................................... 120

6.3.2 In vitro T2 weighted imaging ............................................................................. 123

6.4 Summary .................................................................................................................. 124

6.5 References ................................................................................................................ 124

Chapter 7 .......................................................................................................................... 126

Conclusions and future scope .......................................................................................... 126

7.1 Conclusions .............................................................................................................. 126

7.2 Future scope ............................................................................................................. 128

Appendix I ........................................................................................................................ 130

Thermally-activated delivery of curcumin using magnetic liposomes ........................ 130

AI.1 Introduction ............................................................................................................ 130

AI.2 Experimental Techniques ....................................................................................... 133

AI.2.1 Synthesis of Fe3O4 Nanoparticles and Magnetic Liposomes .......................... 133

AI.2.2 Analysis of Drug Loading and Release ........................................................... 134

Page 11: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

vii

AI.2.4 Evaluation of Biocompatibility and Therapeutics ........................................... 135

AI.3 Results and Discussion ........................................................................................... 136

AI.3.1 Characterisation of the Synthesised Nanoparticles ......................................... 136

AI.3.2 Encapsulation (Loading) of Curcumin ............................................................ 142

AI.3.3 Release of Curcumin at Elevated Temperatures .............................................. 143

AI.3.4 Biocompatibility of Magnetic Liposomes and Anticancer Therapeutic Effect of

Curcumin-Loaded Magnetic Liposomes .................................................................... 144

AI.4 Summary ................................................................................................................ 146

AI.5 References .............................................................................................................. 146

List of Publications ........................................................................................................... 151

Acknowledgments ............................................................................................................ 154

Page 12: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

viii

List of Figures

Figure 1.1 Crystal Structure of Magnetite (© University of Minnesota) .............................. 2

Figure 1.2 Example of Generation 2 PAMAM dendrimer (© Dendritech Inc.) ................... 3

Figure 1.3 A schematic representation of the growth of a tumour (© National Cancer

Institute) ................................................................................................................................. 4

Figure 1.4 Schematic representation of the surface modification process of SPIONs53 ....... 9

Figure 1.5 Drug release (%) in the absence of an AC magnetic field (at room temperature)

and presence of an AC magnetic field (at 42° C) in an acetate buffer (pH 5). Inset:

Fluorescence spectra of the released drug from the DMNCs in the absence and presence of

an AC magnetic field54 ......................................................................................................... 10

Figure 1.6 Intracellular uptake of G3- (a) and G3-FA- (c) stabilised Fe3O4 NPs. (b) and (d)

panels show the binding of G3- and G3-FA-stabilised Fe3O4 NPs with the pre-incubation of

free FA, respectively. Panel (e) shows the morphology of control cells without treatment56

.............................................................................................................................................. 11

Figure 1.7 Schematic representation of the MNP-g-MPS-MTX58 ...................................... 12

Figure 1.8 (a) Confocal laser scanning microscopy image of GL261 mouse glioma cells

following a 2 h incubation with nanoparticle and (b) MRI signal intensity of unlabelled cells

and cells labelled with nanoparticle, measured at 3T using a spin echo sequence91 ........... 19

Figure 1.9 T2-weighted fast spin echo images after injection of 2.5 mg/m1 conjugates per

mouse in 1 h (a) FA-PEG-G3.5 (b) PEG-G3.5@IONPs (c) FA-PEG-G3.5@IONPs. The

arrows denote allograft tumours, which are marked by circles52 ......................................... 20

Figure 2.1 (a) XRD pattern and (b) TEM micrograph of CA–Fe3O4 (Inset of (b) shows the

selected area electron diffraction pattern of CA–Fe3O4)...................................................... 33

Figure 2.2 FTIR spectra of pure CA and CA–Fe3O4 .......................................................... 34

Figure 2.3 (A) TGA-DTA plots of CA–Fe3O4 (B) Zeta-potential of CA–Fe3O4 at different

pH values. Inset ‘a’ shows the hydrodynamic diameter of CA–Fe3O4 obtained from DLS

measurements; inset ‘b’ shows the possible schematic representation of CA–Fe3O4 (CA

coating on the surface of the Fe3O4) .................................................................................... 35

Figure 2.4 Field dependence of magnetisation (M vs. H) plot of CA–Fe3O4 at 300 K. Inset

shows the temperature dependence of magnetisation (M vs. T) measurement .................... 36

Page 13: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

ix

Figure 2.5 (a) Normalised fluorescence spectra of (a) 10 mg/ml of DOX (1 ml) reacted with

different amounts (0, 20, 40, 60, 80 and 100 mg) of CA–Fe3O4 for 15 min. Inset shows the

loading efficiency (binding isotherm of DOX with CA–Fe3O4) obtained from quenching of

fluorescence intensities; (b) 10 mg/ml of pure DOX (1 ml) at different time intervals; and

(c) Drug release profile of DOX-loaded CA–Fe3O4 in a cell mimicking environment

(reservoir: pH 5 and sink: pH 7.3 at 37 °C) ......................................................................... 38

Figure 2.6 (a) Time-dependent calorimetric measurements of CA–Fe3O4; and (b) Viabilities

of HeLa cells incubated with medium that contains CA–Fe3O4 .......................................... 39

Figure 3.1 (a) XRD pattern of Glu–Fe3O4 nanoparticles; TEM micrographs showing (b)

Glu–Fe3O4 nanoparticles (c) particle size distribution (d) selected area electron diffraction

(SAED) pattern, and (e) high-resolution TEM image of Glu–Fe3O4 nanoparticles ............ 50

Figure 3.2 FTIR spectra of (a) glutamic acid (Glu) and glutamic acid-coated Fe3O4 (Glu–

Fe3O4) nanoparticles, (b) PEG-PAMAM of generations 3, 5, 6, and (c) Fe3O4–DGX

nanoparticles ........................................................................................................................ 51

Figure 3.3 TGA-DTA of glutamic acid-coated Fe3O4 (Glu–Fe3O4) nanoparticles ............. 52

Figure 3.4 (a) Room-temperature field-dependent magnetisation of Glu–Fe3O4 and Fe3O4

nanoparticles coated with dendrimers of generations 3, 5, and 6 (Fe3O4–DGX) at 20 kOe (b)

Time-dependent calorimetric measurements of Glu–Fe3O4 in different ACMFs. The SAR

was calculated to be ∼134 W/g from the initial slope of the time-temperature curve. ........ 52

Figure 3.5 (a,b,c) Fluorescence intensity of DOX-loaded Fe3O4–DGX nanoparticles. (d)

Drug loading efficiency versus dendrimer generation. The difference between DG3 and DG6

was significant (p < 0.05). There were no significant differences between DG3 and DG5, and

DG5 and DG6 (p > 0.05) ....................................................................................................... 54

Figure 3.6 (a) Loading efficiencies of EGCG on to Fe3O4–DGX nanoparticles (b) Loading

efficiencies of three groups of Fe3O4–DGX (x = 3, 5 and 6) nanoparticles against dendrimer

generation. The difference between DG3 and DG6 was significant (p < 0.05). ................... 55

Figure 3.7 Drug-release profile of (a) DOX-loaded Fe3O4–DGX nanoparticles at pH 5.0 and

7.4 and (b) EGCG-loaded Fe3O4–DGX nanoparticles at pH 7.4 ......................................... 57

Figure 3.8 Percentage relative cell viability versus the concentration of Fe3O4–DGX

nanoparticles. A decreasing trend of biocompatibility of these nanoparticles is seen with

increase in dendrimer generation and concentration. ........................................................... 58

Page 14: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

x

Figure 3.9 Confocal laser scanning images of (a) control HeLa cells and (b) treated HeLa

cells after 5 h of incubation (scale−20 μm) (c) Viability of HeLa cells in a medium that

contained either Fe3O4–DG5 or DOX–Fe3O4–DG5 nanoparticles in various amounts. The

difference between any pair of data at each concentration was insignificant (p > 0.05). .... 59

Figure 4.1 Schematic representation of the synthesis of the EDA-KR2 dendrimer ............ 68

Figure 4.2 High-resolution X-ray Photoelectron spectra showing C1s, N1s and O1s core

levels of the EDA-KR2 dendrimer ....................................................................................... 70

Figure 4.3 (a) FTIR spectra (b) thermal degradation profile of the EDA-KR2 dendrimer . 71

Figure 4.4 Cell viabilities incubated with peptide dendrimer. The dendrimer is seen to be

biocompatible for a variety of cell lines, even at concentrations of as high as 20 mg/ml. .. 72

Figure 4.5 (a) XRD pattern; (b) FTIR spectra; (c) thermal degradation profiles and (d, e, f)

electron micrographs of Glu–Fe3O4, PAMAM–IO, and KR2–IO nanoparticles, respectively

(σ ≤ 15%) ............................................................................................................................. 82

Figure 4.6 High resolution XPS spectra for C1s, Fe2p, O1s and N1s of the Glu–Fe3O4,

PAMAM–IO and KR2–IO nanoparticles ............................................................................. 84

Figure 4.7 Drug loading profiles of (a) PAMAM–IO and (b) KR2–IO nanoparticles;

Gaussian profiles of (c) DOX-PAMAM–IO and (d) DOX-KR2–IO; and Stern–Volmer plots

of DOX with (e) PAMAM–IO (R2=0.973) and (f) KR2–IO (R2=0.995) .............................. 85

Figure 4.8 Drug release profile of (a) PAMAM–IO and (b) KR2–IO nanoparticles under the

stimulus of pH 5.0 and 7.3 ................................................................................................... 87

Figure 4.9 Time-dependent calorimetric measurements of (A) PAMAM–IO and (B) KR2–

IO nanoparticles at different ACMF and concentrations ..................................................... 89

Figure 4.10 Dependence of SAR on the strength of applied ACMF for (a) PAMAM–IO and

(b) KR2–IO ........................................................................................................................... 90

Figure 4.11 (a) Synergistic effects of DOX-loaded nanoparticles and hyperthermia on HeLa

cells (b) Viable HeLa cell population in the presence of ACMF for varying treatment times

for both PAMAM–IO and KR2–IO nanoparticles ............................................................... 92

Figure 4.12 Synergistic effects of DOX and magnetic hyperthermia (MHT) on viable HeLa

cell population after exposure to both, DOX-loaded dendritic nanoparticles and ACMF... 92

Page 15: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xi

Figure 4.13 Laser Scanning Confocal microscopy images of treated HeLa cells with FITC-

nanoparticles (control), DOX-PAMAM–IO and MHT, and DOX-KR2–IO and MHT. The

scale bar of the images is 50 μm. ......................................................................................... 94

Figure 5.1 Melanoma tumour bearing C57BL/6 mouse being administered intravenously

with DOX-loaded KR2–IO nanoparticles. The skin around the tumour region was shaved

and neodymium magnet was stuck using a medical tape for 6 h. ...................................... 105

Figure 5.2 (a, b) Biocompatibility profile of PAMAM–IO and KR2–IO with B16F10

melanoma cells after 24 and 48 h respectively (c) Dose-dependent cell viability profile of

DOX-loaded dendritic Fe3O4 nanoparticles after 24 and 48 h. Their IC50 values showed

significant difference for 24 h and 48 h (p < 0.05) while there was no significance between

the DOX-PAMAM–IO and DOX-KR2–IO at same time point (p > 0.05).. ...................... 106

Figure 5.3 (a) Schematic representation of magnetically-guided cellular internalization of

dendritic Fe3O4 nanoparticles, (b) (A, C) Confocal micrographs and (B, D) fluorescence

profiles of PAMAM–IO and KR2–IO nanoparticles internalized by B16F10 melanoma cells

after 24 h (c) Amounts of iron internalized by the melanoma cells estimated using atomic

emission spectroscopy……………………………………………………………………108

Figure 5.4 Histograms depicting variations in blood parameters for (a) red blood cells, (b)

haemoglobin, (c) Platelets, (d) white blood cells, (e) lymphocytes and (f) monocytes. The

study showed variations across 14 days of study after intravenous administration of dendritic

Fe3O4 nanoparticles. The results are expressed as mean ± s.d. (n=3) with statistical

significance *p < 0.05 and **p < 0.01 with respect to control animals. ........................ 11010

Figure 5.5 Histograms showing (a) mean corpuscular volume, (b) mean corpuscular

haemoglobin and (c) mean corpuscular haemoglobin. The results are expressed as mean ±

s.d. (n=3) with statistical significance *p < 0.05 and **p < 0.01 with respect to control

animals. .......................................................................................................................... 11010

Figure 5.6 Histograms depicting variations in serum biochemical parameters for (a) SGPT,

(b) SGOT, (c) ALP, (d) Creatinine and (e) blood urea nitrogen (over different time points

during the 14-day study period). The results are expressed as mean ± s.d. (n=3) with

statistical significance *p < 0.05 and **p < 0.01 with respect to control animals. .......... 1111

Figure 5.7 Biodistribution of (a) PAMAM–IO and (b) KR2–IO nanoparticles through

quantification of iron accumulated in various vital organs represented at different time points

Page 16: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xii

post intravenous administration. The quantified iron is represented as mean ± s.d. after

control values were deducted from the plot. (c) Body weight profiles of mice across 14 days

of study. The changes in weight of animals are non-significant (p > 0.05) and does not show

any critical changes. ........................................................................................................... 113

Figure 5.8 (a) Body weight profiles of mice across 35 days of study. The changes in weight

of control animals are due to the uncontrolled tumour size. The weight of animals of group

II, III and IV are significantly decreased (*p < 0.05) in comparison to the control animals,

(b) tumour volume profiles of untreated control animals against the treated groups II, III and

IV. Tumour growth was significantly inhibited with DOX-loaded KR2–IO nanoparticles as

against animals treated with free DOX. (c) Quantification of iron accumulated in various

vital organs. In absence of magnetic field, the thigh muscles (tumour) tend to accumulate

small amount of iron which is significantly elevated with application of magnetic field. 114

Figure 5.9 Histological analyses of the excised organs after 24 h. Optical micrographs of

the treated tumor exhibited zones with compromised cellular cohesion and structural

integrity with significantly different iron content in absence and presence of magnetic field

(MF). Other vital organs showed minimal accumulation of iron causing little or no toxicity

to normal tissues and bodily functions……………………………………………………115

Figure 6.1 Plots of transverse relaxivity (r2) values of (a) PAMAM–IO and (b) KR2–IO in

ultrapure water ................................................................................................................... 121

Figure 6.2 Plots of transverse relaxivity (r2) values of (a) PAMAM-IO and (b) KR2-IO in

phosphate buffered saline (pH 7.3) (c) phantom images showing substantial reduction in

transverse relaxation times of both the nanoparticles ........................................................ 122

Figure 6.3 Plots of transverse relaxivity (r2) values of (a) PAMAM-IO and (b) KR2-IO in

simulated body fluid (pH 7.4) (c) phantom images showing substantial reduction in

transverse relaxation times of both the nanoparticles ........................................................ 123

Figure 6.4 T2-weighted MR images of HeLa cells with PAMAM-IO (a,c) and KR2-IO (b,d)

in the absence (a,b) and presence (c,d) of magnetic field .................................................. 124

Figure AI.1 (a) XRD pattern of Dx–Fe3O4, TEM micrographs of (b) Dx–Fe3O4 (inset shows

the selected area diffraction pattern of Dx–Fe3O4 and (c) magnetic liposomes (inset shows

magnified image of MLs)................................................................................................... 136

Page 17: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xiii

Figure AI.2 FTIR spectra of (a) dextrin and Dx–Fe3O4 nanoparticles (b) MLs and Cur-MLs

............................................................................................................................................ 138

Figure AI.3 Thermal degradation profiles of (a) Dx–Fe3O4 and (b) magnetic liposomes 140

Figure AI.4 Zeta potential of Dx–Fe3O4 and MLs as a function of pH. Inset shows the

hydrodynamic diameter of the MLs obtained from DLS measurements (1.2 µm) ............ 141

Figure AI.5 (a) Field-dependent magnetisation (M vs. H) plot of Dx–Fe3O4 and MLs at room

temperature (b) Time-dependent specific absorption measurements of Dx–Fe3O4

nanoparticles (Inset depicts the dependence of heat generation on the applied ACMF) ... 142

Figure AI.6 (a) Absorbance spectra of curcumin-loaded MLs against pure curcumin (b)

Drug release profiles of curcumin-MLs at physiological and hyperthermic temperatures 143

Figure AI.7 (a) Percentage of the cell viability of MLs incubated with mouse fibroblasts and

cervical cancer cells for 24 h (b) Dose-dependent evaluation of Curcumin-MLs for

determination of IC50 with HeLa cells ............................................................................... 145

Page 18: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xiv

List of Tables

Table 4.1 The area under the peaks of DOX-loaded PAMAM–IO and KR2–IO nanoparticles

(σ≤10%) ............................................................................................................................... 86

Table 4.2 IC50 values of (a) DOX-PAMAM–IO and (b) DOX-KR2–IO nanoparticles with

various cancer cells. The values are represented in µg/ml of formulations with ≤ 15% (IC50

of pure DOX was 1.1-1.5μM with different cell lines) ........................................................ 88

Table 5.1 Animal denomination, dosage of dendritic nanoparticles and time points of sample

collection for biocompatibility and biodistribution studies (h=hour; d=day) .................... 102

Table 5.2 Animal denomination, dosage of dendritic nanoparticles and time points of sample

collection for biocompatibility and biodistribution studies (h=hour; MF=magnetic field)

............................................................................................................................................ 104

Table 5.3 IC50 values of DOX-PAMAM–IO and DOX-KR2–IO nanoparticles with B16F10

melanoma cells. The values are represented in mg/ml of formulations with ≤ 15% (IC50 of

pure DOX was 0.11 – 0.17 μM25) ...................................................................................... 107

Page 19: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xv

Abbreviations and Nomenclatures

ACMF Alternating Current Magnetic Field

ALP Alkaline Phosphatase

BBB Blood Brain Barrier

BUN Blood Urea Nitrogen

CA Citric Acid

CBC Complete Blood Count

DAPI 4′, 6-Diamidino-2-phenylindole

DCM Dicholoromethane

DOX Doxorubicin

DLS Dynamic Light Scattering

DMSO Dimethyl sulfoxide

DTA Differential Thermal Analysis

EC Epicatechin

ECG Epicatechin-3-gallate

EGC Epigallocatechin

EGCG Epigallocatechin-3-gallate

EPR Enhanced Permeation and Retention

FBS Foetal Bovine Serum

FITC Fluorescein isothiocyanate

FTIR Fourier Transform Infrared Spectrometer

Glu Glutamic Acid

GRAN Granulocytes

Hb Haemoglobin

HCT Haematocrit

HRTEM High-Resolution Transmission Electron Microscope

ICP-AES Inductive Coupled Plasma - Atomic Emission Spectroscopy

LSCM Laser Scanning Confocal Microscope

LYM Lymphocytes

MCH Mean Corpuscular Haemoglobin

Page 20: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xvi

MCHC Mean Corpuscular Haemoglobin Concentration

MCV Mean Corpuscular Volume

MDT Magnetic Drug Targeting

MF Magnetic Field

MHT Magnetic Hyperthermia

MNP Magnetic Nanoparticles

MON Monocytes

MRI Magnetic Resonance Imaging

NMR Nuclear Magnetic Resonance

PAMAM Polyamidoamine

PBS Phosphate Buffered Saline

PEG Polyethylene Glycol

PEI Polyethylene Imine

PI Propidium Iodide

PLT Platelets

PPI Polypropylene Imine

RBC Red Blood Cells

RES Reticulo-endothelial System

SAED Selected Area Electron Diffraction

SAR Specific Absorption Rate

SBF Simulated Body Fluid

SGOT Serum Glutamic Oxaloacetic Transaminase

SGPT Serum Glutamic Pyruvic Transaminase

SPION Superparamagnetic Iron Oxide Nanoparticles

SRB Sulforhodamine-B

TC Curie Temperature

TE Echo Time

TEM Transmission Electron Microscope

TGA Thermo-gravimetric Analysis

TR Repetition Time

VSM Vibrating Sample Magnetometer

Page 21: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

xvii

WBC White Blood Cells

XRD X-Ray Diffraction

XPS X-ray Photoelectron Spectroscopy

Page 22: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

Parts of this chapter have been published in J. Biomed. Nanotech, 2014, 10, 32-49.

Chapter 1

Functional Nanomaterials and their Applications in

Cancer Theranostics

The fabrication of materials along with a scaling down of their size to nanometres imparts

certain distinctive physical and chemical properties to them due to size and surface effects.

These enhanced spectrum of properties of functional materials show promise of improvements

in diverse applications. At nanoscale dimensions, the physico-chemical, optical, mechanical,

catalytic, electrical, electronic and magnetic behaviour of materials is no longer similar or

dependent on their bulk behaviour. This has led to an increase in the number of materials, both

inorganic and organic. These materials are tailored and applied to various applications. This

chapter introduces two promising functional nanomaterials briefly and elaborately discusses

the various aspects of their use in cancer therapeutics and imaging diagnostics.

1.1 Superparamagnetic Iron Oxide Nanoparticles

Iron oxide nanoparticles primarily exist in two forms: magnetite (Fe3O4) and

maghemite (γ-Fe2O3). Fe3O4 nanoparticles have an inverse spinel crystal structure, with oxygen

forming a face-centred cubic system in which the tetrahedral sites are occupied by Fe3+ and the

octahedral sites are filled with both Fe2+ and Fe3+ ions (Figure 1.1). At nanoscale sizes,

magnetic nanoparticles have a single magnetic domain. The strongly coupled magnetic spins

on each atom combine to produce a particle with a single ‘giant’ spin. This effect is known as

Page 23: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

2

superparamagnetism. Magnetic nanoparticles (MNPs) show superparamagnetic behaviour at

room temperature, which is attributed to the size reduction in the material; that is, these

particles show no hysteresis loop in nanoscale dimensions. In recent years, researchers have

optimised the properties of these nanoparticles for the improvement of their multimodal

performance. MNPs have undergone thorough investigation and evaluation in a variety of

biomedical applications such as hyperthermia treatment of cancer, contrast agents for magnetic

resonance imaging (MRI), magnetic separation and sorting of cells and proteins (bio-

recognition), and controlled and targeted drug delivery1-3. This has led to the rise of an entire

field of nanobiomedicine.

Figure 1.1 Crystal Structure of Magnetite (© University of Minnesota)

1.2 Dendrimers

Dendrimers are monodisperse, branched macromolecules with three-dimensional

spatial conformations that have a defined radial symmetry4, 5. This structural uniqueness

imparts a wide range of physical and chemical properties to dendritic molecules. They also

characteristically set them apart from the classical polymeric molecules. Dendrimers consist of

three architectural denominations: the central core, the branching units and the terminal

functional groups. The number of repeated branching molecules assigns generation to the

dendrimers and is also responsible for their globular shape. It is the hyperbranching of the

molecule from the centre of the dendrimer towards the periphery that results in homostructural

layers between the focal points (branching points). The number of focal points from the core

towards the outer surface is the generation number. Thus, generation refers to the number of

repeated branching cycles performed during the synthesis. The core of the dendrimer is denoted

as generation ‘zero’ (G0). The branches of lower generation dendrimers tend to radiate out

Page 24: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

3

towards the periphery and exist in open conformation. On the other hand, since the number of

the generation increases, the branches tend to retract and adopt globular conformations in three-

dimensional space, with intramolecular hydrogen bonding governing the structures. The

generation-dependent conformational changes that are confirmed by X-ray analysis

demonstrated that the higher generations are more spherical when compared to the lower linear

generations6. Due to their structural and functional uniqueness, dendrimers have found

successful biomedical applications7 in the fields of MRI8, drug delivery9, nucleotide delivery10,

11 and the like.

Figure 1.2 Example of Generation 2 PAMAM dendrimer (© Dendritech Inc.)

1.3 Cancer

The body is made up of many types of cells that grow and divide in a controlled fashion,

according to the body’s requirement and to keep it healthy. When cells become old or damaged,

they are replaced with new cells, which maintains the cycle of life. When this functional order

process goes out of control, the cells bypass their death and new cells are continuously formed

even though the body does not need them. The cells that have disrupted cellular machinery

divide uncontrollably and become autonomous, which gives rise to a diseased state and is

termed as cancer. Cancerous cells and tissues have abnormal growth rates, shapes, sizes, and

functioning. Cancer cells do not grow faster than normal cells; rather their growth is just

uncontrolled. These extra, unwanted and anomalous cells form a solid mass that is known as a

tumour. These cells also have the tendency to spread to and accumulate in other parts of the

Page 25: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

4

body and are responsible for tumours at sites that are at a distance from the origin (metastasis).

This uncontrolled cell proliferation disrupts the balance between cell growth rates and cell

mortality rates, giving rise to dysfunctional cell masses that have irregular shapes and sizes

(Figure 1.3). This disorder can be a result of (a) uncontrolled cell growth and (b) loss of a cell’s

ability to undergo apoptosis, which is programmed cell death. The immune system of the body

is unable to recognise and remove these cells because they are erroneously identified as ‘self’

and not foreign, which poses a major challenge in the treatment of cancer.

Figure 1.3 A schematic representation of the growth of a tumour (© National Cancer

Institute)

Tumours are further classified into following types:

(a) Benign tumours: Are not cancerous. They can often be removed, and, in most cases,

they do not come back. Cells in benign tumours do not spread to other parts of the body.

(b) Malignant tumours: Are cancerous. Cells in these tumours can invade nearby tissues

and spread to other parts of the body.

Cancer types can be grouped into broader categories. The main categories of cancer include:

(a) Carcinoma: Cancer that begins in the skin or in tissues that line or cover internal organs.

(b) Sarcoma: Cancer that begins in bone, cartilage, fat, muscle, blood vessels, or other

connective or supportive tissue.

(c) Leukemia: Cancer that starts in blood-forming tissue such as the bone marrow and

causes large numbers of abnormal blood cells to be produced and enter the blood.

(d) Lymphoma and myeloma: Cancers that begin in the cells of the immune system.

Page 26: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

5

(e) Central nervous system cancers: Cancers that begin in the tissues of the brain and spinal

cord.

The cancer therapies that are currently under investigation consist primarily of the drugs

or other molecules that block the growth and spread of cancer. These molecules act by

interfering with biochemical signal cascades that are involved in tumour proliferation and

growth. Thus, these target molecules are called as “molecular targets,” targeted therapies are

referred to as “molecularly targeted therapies”. The molecular and cellular changes that the cell

undergoes are specific to the type of cancer. Therefore, these targeted therapies may be more

effective than classical therapies (including chemotherapy and radiotherapy) due to their

specificity and selectivity. Another approach focuses on the concept of starvation as the rapidly

dividing tumor cells require nutrients proportionately. This requirement leads to rapid

proliferation of blood vessels in the region, angiogenesis, due to increased levels of angiogenic

growth factors locally. Blocking of these local signal molecules, in theory, should lead to death

of cancer cells due to starvation. The general approach in overcoming the current hindrances

is to attach the therapeutic molecules to a nanoscale carrier that will release them at the target

site over an extended period of time or when specifically triggered to do so12. In addition, the

surfaces of these nanoscale carriers may be engineered to seek out and become localised at a

disease site, for example, by identifying and attaching to cancer sites.

1.4 Cancer Theranostics

In the recent past, significant advances have been made in the design and development

of drug delivery systems through the use of multidisciplinary teams that utilise biological,

chemical, physical, and engineering sciences. The underpinning science aims to achieve a

greater understanding of the physico-chemical properties of drugs, their interaction with the

delivery systems, their biological fate and the targeting of drugs at the molecular, membrane

and cellular levels. The rise of modern pharmaceutical and nanobiotechnology promotes the

development of novel nanomaterials-based delivery systems that can overcome biological

barriers, particularly in cancer therapy13-16. The ultimate requirement is to formulate a system

that can circulate through the blood stream, detect molecular changes due to cancer, image the

targeted cancer, deliver the therapeutic agent to the desired location and then monitor the

effectiveness of the therapy. Keeping this in mind, researchers are now inclined towards the

tailoring of nanomaterials with systematic variation and properties, which leads to unique and

varied applications that are well beyond the traditional ones. Among the long list of nanoscale

Page 27: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

6

materials that are currently being explored for their prospective use in nanomedicine17-20,

superparamagnetic Fe3O4 nanoparticles are widely favoured because of their biocompatibility

profile21, 22 and reactive surface that can be modified with a variety of molecules23-26. This

flexibility of nanostructure design has led to the use of these nanoparticles in diverse

applications that range from magnetic separation27, biosensors28, 29, medical imaging30-34, drug

delivery33, 35-38 and tissue repair39 to hyperthermia40-42.

MNPs are chosen as drug carriers due to their specific nanostructure fabrication,

tailored drug release properties, and lower toxicity and immunogenicity, which results in

improved treatment efficacy and reduced side effects. These drug loaded MNPs can be guided

to the desired target area using an external magnetic field while simultaneous tracking of the

biodistribution of the particles takes place. Besides this, these nanoparticles respond

significantly and generate heat when exposed to an alternating current magnetic field (ACMF),

which can provide promising therapeutic solutions in the issue of hyperthermia42, 43. However,

for use in therapeutic applications, the colloidal stability of these nanoparticles in aqueous and

physiological mediums, which is usually obtained by either charging the surface or by creating

steric hindrance using surfactant molecules, is crucial. In order to control the surface properties

of these nanoparticles, the surface may be modified with different types of molecules, which

prevents large agglomerates, changes in the structural configuration, biodegradation, and

anchors various cargo molecules44. The main hindrance faced by these magnetic nanoparticles

in delivering the intended cargo to the desired location is their non-specific internalisation by

the cells45. Dendrimers have received significant attention for their ability to functionalise and

stabilise nanoparticles. Their peculiar architecture and flexibility of modification in different

ways have provided them with unique properties46 and could possibly be used in biomedical

applications. They have an edge over other macromolecules and polymers due to their

capability to provide varied functional groups. A second advantage is that they have an internal

cavity for entrapment of guest molecules such as drugs, imaging molecules and targeting

ligands. The presence of multiple functional groups with symmetric perfection and nanometre-

scale internal cavities enables the use of dendritic stabilised nanoparticles in remarkable

biomedical applications such as drug delivery, imaging and sensing44. Lately, researchers have

started to attempt the integration of the unique features of dendrimer chemistry with the

versatile magnetic nanoparticles in a single nanovehicle, in order to enhance their properties;

this broadens the scope of their biomedical applications.

Page 28: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

7

Dendrimer-functionalised magnetic nanoparticles can be coupled with target

molecules, drugs and imaging moiety to provide a multifunctional platform for real-time

monitoring and targeted drug delivery. Such delivery, controls the release of the therapeutic

agent and enables its release only at the target site without healthy tissue being exposed to the

drug, thereby minimising side effects. This chapter reviews the recent progress in the design

and fabrication of dendrimer-based magnetic nanoparticles for biomedical applications. An

extensive literature survey shows that there is only a small number of works available on this

particular subject, indicating that these nanomaterials are still in the nascent stage. It also

indicates that and plentiful work, from fundamental biological scientific studies to

commercially viable nanobiotechnology, can be carried out using these exceptionally attractive

materials.

1.4.1 Chemotherapy

As the name suggests, chemotherapy involves the use of various chemicals for the

treatment of diseases. These chemicals interfere with the internal machinery of the cells to

rectify the diseased state to a normal state. Chemotherapy is one of the classical therapies in

the treatment of cancer. A wide spectra of molecules have been explored and used as

therapeutic agents in a variety of cancers. These chemicals damage the internal biochemical

cascades (that is, DNA, RNA and proteins) to initiate apoptosis. However, the main challenge

that is faced by chemotherapy is its unspecific distribution and action in the body as these

molecules are as detrimental to normal cells as to cancer cells. This demands the development

of carriers and delivery vectors for these therapeutic agents, which can identify and selectively

deliver these agents only to the cancer sites47. The development of drug delivery vectors is

currently dominated by liposomes48 and polymeric systems22, which is marked by some critical

disadvantages. Therefore, researchers are in constant search of efficient drug carriers.

Normally, a drug delivery system comprises three components: a therapeutic agent that

is, the drug, a targeting moiety and a carrier. The drug is either incorporated by passive

absorption or chemical conjugation to the carrier. The choice of the carrier molecule is crucial

because it significantly affects the pharmacokinetics and pharmacodynamics of the drugs. A

wide range of materials such as natural or synthetic polymers, lipids, surfactants and

dendrimers have been employed as drug carriers49. One of the major limitations of a drug

delivery system is the challenge of releasing the desired amount of drug to its actual site of

action at a specific rate within a stipulated time. In the current scenario of drug delivery, the

Page 29: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

8

bottleneck is caused due to poor solubility and high toxicity of drugs, requirement of high

dosages, non-specific delivery, in vivo degradation and short circulating of the half-lives of the

delivery system50. Therefore, advances in biotechnology and nanomedicine aim to design a

targeted drug delivery system in which the drugs can be effectively and conveniently conveyed

to the desired site, and to improve the pharmacokinetics of easily degradable materials, increase

patient compliance and reduce healthcare costs.

1.4.1.1 Non-targeted drug delivery

Dendrimer-modified magnetic nanoparticles have been explored recently for their

potential as drug delivery vehicles. Wu et al.51 investigated the preparation of dendritic-linear

block copolymer-modified superparamagnetic iron oxide nanoparticles (SPIONs) that have a

core of Fe3O4 nanoparticles and a dendritic-linear block copolymer, polyamidoamine-type

dendron-b-poly(2-dimethylaminoethylmethacrylate)-b-poly(N-isopropylacrylamide)

(PAMAM-b-PDMAEMA-b-PNIPAM) as a shell in order to stabilise the nanoparticles in an

aqueous medium. In the buffer solution, the release amounts of doxorubicin (DOX) from

uncross-linked block copolymer-modified SPIONs were 28.8 and 15.5% at 25 °C and 37 °C,

respectively, while the release amounts from the cross-linked block copolymer-modified

SPIONs were 26.8 and 13.7% at 25 °C and 37 °C, respectively. These results indicate that both,

the uncross-linked and cross-linked polymer-modified SPIONs show thermo-sensitive drug

release behaviours (due to the presence of the thermo-sensitive PNIPAM component). Chang

et al.52 developed a pH-responsive drug release system by conjugating PAMAM dendrimers

with DOX (PAMAM–DOX) and superparamagnetic Fe3O4 nanoparticles. PAMAM

(generation 2.5) was modified by PEG and further modified by hydrazine in order to anchor

DOX molecules. The formulations that were synthesised were found to be sensitive towards

the change in proton concentration in the surrounding medium (pH sensitivity). An interesting

feature of this result was that, after 15 h, around 80% of the DOX was released in pH 5, while

less than 8% of the DOX was released in pH 7.4. The amount of the drug released was governed

by the acidic hydrolysis of these hydrazone bonds.

He et al.53 described dendritic−linear−brush-like triblock copolymer polyamidoamine-

b-poly(2-(dimethylamino)-ethyl methacrylate)-b-poly(poly(ethylene glycol) methyl ether

methacrylate) (PAMAM-b-PDMAEMA-b-PPEGMA)-grafted SPIONs and studied their drug

release properties. PDMAEMA and PPEGMA were added step-by-step onto the surface of

SPIONs using a Cu-mediated radical polymerisation method (Figure 2.1). The release amounts

Page 30: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

9

of DOX from the copolymer-modified SPIONs for a period of 48 h at pH 4.7, 7.4 and 11.0

were 83.1, 64.7 and 8.3%, respectively. The results showed that free DOX was able to inhibit

the cell proliferation of NIH 3T3 and HeLa cells more efficiently when compared to the DOX

released from the formulations; this indicated a delay in the release of the drug due to a lower

diffusion rate from the polymeric shell, which showed promise of lower toxicity to the normal

cells. Therefore, on the basis of the biocompatibility and drug release effects, the modified

SPIONs could provide a unique opportunity to design excellent drug delivery systems for

therapeutic applications.

Figure 1.4 Schematic representation of the surface modification process of SPIONs53

Chandra et al.54 synthesised dendrimer-functionalised Fe3O4 nanoparticles for delivery

of DOX. The nanoparticles were synthesised and modified by 3-aminopropyltrimethoxysilane

(APTS). The dendritic structures were grown on these silane-coated Fe3O4 nanoparticles by

using methylacrylate and arginine as the monomeric units. The drug release revealed a

sustained release profile and achieved a plateau after 8 h for a maximum release of 54% of the

drug. Chandra et al. also evaluated the efficacy of the drug release under ACMF, marking the

efficiency of these platforms in combination therapy of cancer for hyperthermia. In the absence

of any external magnetic field, the percentage of drug release was approximately 35% for the

initial 45 mins, which shot up to ~80% on application of ACMF for the next 15 min. Under the

applied field conditions, acid hydrolysis and the increase in temperature contribute

synergistically towards the drug release profile (Figure 1.5).

Page 31: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

10

Figure 1.5 Drug release (%) in the absence of an AC magnetic field (at room temperature)

and presence of an AC magnetic field (at 42° C) in an acetate buffer (pH 5). Inset:

Fluorescence spectra of the released drug from the DMNCs in the absence and presence of an

AC magnetic field54

Another important parameter related to drug delivery is the biodistribution and toxicity

of the nanocarriers. It has been found in dendrimer-based MNPs that smaller generation

dendrimers show rapid renal elimination while higher generation dendrimers show

accumulation in the liver55. The binding of the ethylene glycol units renders positive charges

in the dendrimers, which enhances permeation and retention (EPR) and increases circulation

time in the blood. The encapsulation of the drugs within the ethylene glycol arms of the

dendrimers leads to reduced toxicity and lesser accumulation in body organs. Therefore, it is

expected that if the dendrimers are covered with ethylene glycol entity, it can make the

dendrimers attractive drug carriers in vivo.

1.4.1.2 Targeted drug delivery

1.4.1.2.1 Ligand-targeted Drug Delivery

The specificity of the target location still remains a challenge in cancer therapy. Non-

targeted systems tend to deliver the therapeutic molecules to the cancer as well as the

neighbouring normal cells, which leads to side effects. To overcome this barrier, delivery

systems are modified by biomolecules that identify the target cell and, thus, enhance the

specificity of the chemotherapy. Along these lines, Shi et al.56 reported dendrimer-modified

Page 32: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

11

Fe3O4 nanoparticles that utilise folic acid as the targeting ligand. Folic acid (FA) was

conjugated with the PAMAM (generation 3) using amide bonding and then incubated with

Fe3O4 nanoparticles. The cell uptake studies reveal that both G3- and G3-FA-stabilised Fe3O4

nanoparticles bind to the KB cells that overexpress FA receptors (Figure 1.6). The presence of

free FA molecules in the growth media also affected (lowered) by the binding of nanoparticles

to the receptors because free FA is the preferred ligand for the receptors. Thus, the results of

Shi et al. suggest the potential use of dendrimer-functionalised Fe3O4 nanoparticles in targeted

delivery of various therapeutic agents.

Figure 1.6 Intracellular uptake of G3- (a) and G3-FA- (c) stabilised Fe3O4 NPs. (b) and (d)

panels show the binding of G3- and G3-FA-stabilised Fe3O4 NPs with the pre-incubation of

free FA, respectively. Panel (e) shows the morphology of control cells without treatment56

Chang et al.57 described a PAMAM-modified Fe3O4 nanocarrier system for targeted

delivery of doxorubicin to MCF-7 cells. The drug release profiles were investigated in a buffer

of pH 5.0 and pH 7.4 at 37°C. After 15 h, about 75% of the DOX was released in low pH while

less than 8% of the DOX release was observed in a physiological pH (7.4), thus establishing

the pH-sensitive drug release behaviour. On similar lines, Li et al.58 fabricated polyglycerol-

conjugated Fe3O4 nanoparticles for targeted delivery of methotrexate (MTX) to cancer cells.

Page 33: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

12

An esterification reaction was carried out to form conjugates of 3-(trimethoxysilyl)

propylmethacrylate-coated MNPs (MNP-g-MPS) and MTX, which involved the reaction

between the hydroxyl groups of hyperbranched polyglycerol (HPG) and the carboxylic acid

groups of MTX (Figure 1.7). The drug release profiles were evaluated in a phosphate buffer of

pH 7.4.

Figure 1.7 Schematic representation of the MNP-g-MPS-MTX58

The formulations were evaluated for their cytotoxicity in RAW mouse macrophages, 3T3

fibroblasts and KB cells. The uptake of iron from MNP-g-HPG nanoparticles was very low and

showed the evasion of endocytosis by the cells (0.4-0.5 pg/cell). The conjugation of MTX, on

the other hand, significantly increased the endocytosed nanoparticles for KB cells, (~1.2

pg/cell) while for 3T3 fibroblasts and RAW macrophages, it was less than 0.7 pg/cell. The

formulation of MNP-g-HPG-MTX2 (12.5 µg/mg of nanoparticle) showed a 20-fold increase

in the uptake by the KB cells (12.1 pg/cell) when compared to MNP-g-HPG nanoparticles,

while for 3T3 fibroblasts and RAW macrophages, the uptake was 11 and 8 times lower,

respectively. Depending on the amounts of MTX internalised, the nanoparticles were capable

of killing ~50% of the KB cells while exhibiting low cytotoxicity towards 3T3 fibroblasts and

RAW macrophages.

1.4.1.2.2 Magnetically-guided (Magnetic Drug Targeting) Drug Delivery

Magnetic drug targeting (MDT) in cancer therapy refers to the attachment of

therapeutic molecules to magnetic delivery vectors, after which the molecules are concentrated

Page 34: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

13

and released at the cancer site under the guidance of an external homogenous magnetic field59,

60. Ideally, the drugs should be chemically bound to the magnetic nanocarriers, before

administering them into the systemic circulation, magnetically guiding them along with the

blood flow and trapping them around the tumour region. The magnetic field gradient interacts

with these magnetic nanocarriers in the circulation and leads them to accumulate in the tumour

region. For all in vivo applications, it is desirable that MNPs retain sufficient hydrophilicity

and, do not exceed 100 nm so that rapid clearance by the reticulo-endothelial system (RES)

can be avoided. This can be achieved by stabilising the magnetic nanoparticles with dendrimers

through which a smaller, more neutral and hydrophilic surface, which has a longer plasma half-

life61, can be obtained. It has also been shown that a strong magnetic field at the site of the

tumour induces accumulation of drug-loaded magnetic nanoparticles. However, as a proof of

concept, Alexiou and co-workers found quite interesting and encouraging results when they

employed the MDT in the local accumulation of mitoxantrone in the squamous cell carcinoma

implanted in New Zealand White rabbits62. They administered their nanoparticles in the

femoral artery while an external magnetic field was focussed on the tumour. They found a

significant (p < 0.05), complete, and permanent remission of the squamous cell carcinoma as

compared to the control group of animals. They successfully combined intratumoural

nanoparticle accumulation and locoregional cancer treatment without any kind of systemic

toxicity. The principles of magnetic guidance of MNP-conjugated drugs have reached clinical

trials for cancer therapy. MDT has been known to result in desired therapeutics in superficial

tumours, but its ability to effectively reach the deep-seated tissues/cancer sites remains

inadequate63. Despite the advantages of the dendrimer-stabilised magnetic nanoparticles, we

could hardly find any work utilising these nanoparticles for magnetic drug targeting, which

suggested that this area is still unexplored and has promising prospects. Most of the recent

research has centred on drug delivery and, therefore, it is time to devote studies to be carried

out beyond ‘proof of concept’.

1.4.2 Magnetic Hyperthermia and Chemo-thermotherapy

The classical routes of cancer treatment include chemotherapy, radiotherapy and

surgical removal of the tumour mass. These methods of combating cancer are non-selective

and cannot differentiate between cancer and normal cells. A solid tumour has very poor

vasculature, which makes the cells compete for nutrients and oxygen for their high energy

demands. The intercellular interactions are disrupted and the cells become autonomous. The

Page 35: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

14

continued, uncontrolled cell division results in a solid tumour mass that turns hypoxic (low on

oxygen) and is responsible for a large number of chemical and metabolic differences between

cancerous and normal cells64. Although this hypoxic nature makes these cells insensitive to

radiations and chemotherapeutic agents, which also hampers its clinical treatment, it also

makes these cancer cells sensitive towards a rise in temperatures65. Due to their compact three-

dimensional structure, it is difficult for these cells to dissipate the applied heat, which in turn

proves lethal to the cells, resulting in apoptosis as a direct response. The heating causes the

coagulation of proteins, denaturing them and obstructing the cellular machinery, leading to cell

death. Even if the cells do not die, this rise in temperatures in combination with the hypoxic

conditions renders these cells vulnerable to chemotherapeutic agents or to being radio-

sensitised. To overcome the shortcomings of classical cancer therapies and to improve

therapeutic effectiveness, hyperthermia has been explored as an alternative therapy in cancer

treatment.

Hyperthermia literally translates to ‘elevated temperatures’. This heat generation, when

obtained through power dissipation of magnetisation reversal processes of nanoparticles, is

termed as magnetic hyperthermia. When a colloidal suspension of magnetic nanoparticles is

exposed to an alternating current magnetic field (ACMF), the reversal of magnetisation occurs

through hysteresis loss and Neel and Brownian relaxation losses66. During the undertaking of

these processes, substantial amounts of energy is dissipated in the microenvironment as heat

and has found applications in hyperthermia treatment of cancer67-70. In ferromagnetic materials,

the heat is generated due to susceptibility and hysteresis losses. Therefore, the power generated

in the case of ferromagnetic materials is proportional to the product of the frequency of ACMF

and the area of the hysteresis loop. There are no Brownian losses (whole particles rotation),

and the vector sum of the rotational losses of the magnetic moments within the particle (Néel

relaxation) tend to cancel each other because the magnetic domains are randomly aligned in a

multi-domain particle system. On the other hand, in superparamagnetic materials this heat is

a direct result of power dissipation due to Brownian and rotational relaxations of single domain

particles. This heating power is governed by various factors and is primarily dependent on the

particle size, magnetic properties of the material, strength (H) and frequency (ω) of the applied

ACMF and solvent properties71-74. Despite being more proficient in their heat-generating

properties, ferromagnetic materials are not chosen over superparamagnetic nanoparticles for

magnetic hyperthermia applications. Recently, various researchers have explored the

possibility of combining magnetic hyperthermia with the chemotherapy of cancer48, 75. It is

Page 36: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

15

seen that they act synergistically, which yields more promising results than when they function

in separate therapy options. Since hyperthermia causes severe changes in cellular

macromolecules, affecting all cellular functions, there are two primary concerns in its clinical

use. First, similar to classical therapies, hyperthermia also misses selectivity and affects both,

cancer and normal cells. Second, the foremost challenge in applying hyperthermia to cancer

treatment is the maintaining of local temperatures below 45 °C, so that the neighbouring normal

cells suffer minimally. Also, the combining of chemotherapy with thermotherapy is expected

to result in improved therapeutic performances and in the regression of tumours.

Guardia et al.76 studied the heating behaviour of iron oxide nanocrystals as a function

of size. Between the sizes of 13 and 40 nm, the nanoparticles with an average diameter of 19±3

nm showed high clinically significant SAR values and were able to check cell proliferation in

oral carcinoma cells in vitro efficiently. To address the challenge of localised heating,

multifunctional magnetic nanoparticles have been explored for their heat-generating property

that can be also be modulated according to the required clinical settings. Baba et al.77 undertook

a comparative study to evaluate the uptake and hyperthermic efficiency of superparamagnetic

and ferromagnetic nanoparticles with human breast cancer cells (MCF-7). Their results

demonstrated that the cell death due to ferromagnetic particles was significantly higher than

the cell death due to superparamagnetic nanoparticles that reached temperatures above 45 °C.

Sadhukha and co-workers synthesised 12±3 nm-sized aqueous-stable Fe3O4 nanoparticles with

high magnetisation and SAR properties78. They evaluated the hyperthermic efficiency of these

nanoparticles with human lung adenocarcinoma (A549) and human mammary adenocarcinoma

(MDA-MB-231) cell lines and found magnetic hyperthermia to be much more effective than

conventional hyperthermia (water bath). They concluded that magnetic hyperthermia treatment

resulted in pleiotropic effects with induction of apoptosis and generation of reactive oxygen

species as the mechanistic causes. In one of their other works, Sadhukha and co-workers

developed epidermal growth factor receptor (EGFR)-targeted inhalable Fe3O4 nanoparticles to

target non-small cell lung cancer in order to selectively target the epithelial linings of tumours

in lungs79. Female Fox Chase SCID® Beige mice were chosen to generate an orthotopic lung

tumour model and were subjected to magnetic hyperthermia after successful inhalation and

deposition of these nanoparticles. Exposure of ACMF in these animals was seen to cause an

insignificant decrease in tumour size (p>0.05). This decrease in tumour size was relative to the

control animals that received the same dose but were not exposed to ACMF.

Page 37: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

16

It has been seen that the synergistic action of chemotherapy and magnetic hyperthermia

yield much more promising results than as separate therapy options. Qu et al.80 evaluated

camptothecin in combination with hyperthermia against ovarian and liver cancer cells, while

Li et al.81 used 5-fluorouracil, which specifically targeted cancer cells by the anti-human

epidermal growth factor receptor 2 (anti-HER2). These nanoparticles were therapeutically

evaluated with murine bladder cancer cells (MBT-2) and were able to reduce the viable cell

population by 70% after an ACMF exposure of 15 min. In male C3H/HeN mice, these

nanoparticles also showed some promising local hyperthermia effects when administered both

intratumourally as well as intravenously, with significant reduction in the tumour volume in

small tumours (<50 mm3). On similar lines, Kossatz and co-workers found that hyperthermia

along with doxorubicin successfully reduced the tumour volume by 40% in subcutaneous

MDA-MB-231 tumour-bearing female athymic nude mice82. Various formulations have been

fabricated and studied for applications in hyperthermia and combinatorial therapy, but

dendrimer functionalised magnetic nanoparticles have not yet been explored much.

1.4.3 Diagnostic Bio-imaging

An accurate diagnosis and detection of cancer at an early stage is one of the major

limitations in existent cancer therapy. Typically, the tumours that grow in the deep-seated

tissues/organs go undetected, leading to inadequate treatment measures that have high

morbidity, which leads to lower chances of the survival of the patient. Tumour detection, thus,

would require a non-invasive diagnostic technique with high spatial resolution, which would

investigate the anatomy and physiology of the tumour even in the early stages. New and

customised nanoparticulate formulations exploit the properties of multiple imaging modalities

in order to improve cancer diagnosis and to monitor the response to chemotherapy. Two such

imaging techniques are optical imaging and magnetic resonance imaging (MRI). Optical

imaging primarily utilises the fluorophores or the tagging molecules that provide sensitivity

and spatial and temporal resolution to the nanoparticles; however, their imaging through

penetration is limited to a few millimetres below the tissue.

MRI is a high-resolution medical imaging technique that is widely used in clinical

settings for the physiology of both healthy and diseased bodies. It uses the magnetic relaxation

of the proton nuclei of the water in the body and generates three-dimensional tomographic

images. Proton relaxation is a very slow process that reduces the resolution of the images,

resulting in compromised quality and unclear information. In order to improve the contrast of

Page 38: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

17

the images obtained, additional mediator materials are required. These are known as contrast

agents. Gadolinium-based paramagnetic compounds have been used to aid the quickening of

proton relaxation, which results in adequate MR signals that generate high-quality, brighter

images83, 84. Another class of contrast agents are superparamagnetic nanoparticles. Magnetic

nanoparticles have not only shown potential in selective delivery of anticancer drugs but have

also exhibited highly desirable sensitivity and specific imaging capabilities. The higher

magnetic moment of these nanoparticles gives rise to alterations in the surrounding magnetic

fields of the MRI machine. This alteration in the field gradient results in the shortening of the

relaxation time of the proton nuclei. As a result, an image that has high contrast characteristics

is generated along with sufficient information about the region in question, especially in the

case of tumours85, 86. MRI results provide improved image resolution and tissue contrast

through the use of nanoparticles and contribute towards the revelation of detailed tissue

morphology and anatomy for whole body imaging of animals and humans.

1.4.3.1 Optical Imaging

Tumour detection by using fluorescent probes may be quite advantageous in terms of

improved biocompatibility when compared to other types of contrast agents, but they suffer

from poor penetration of light through tissues. To provide a minimally invasive solution to this

barrier, nanotechnology has enabled the use of water-soluble, functionalised MNPs that are

highly stable and, when tagged with fluorescent molecules, can manifest extensive fluorescent

properties for in vivo imaging of live cells. Although dendrimers have not been explored much

in the context of MNPs, they have been used as matrices for the synthesis of lanthanide-

magnetic nanohybrids for imaging. Luwang et al.87 reported the use of a poly (amido amine)

(PAMAM) dendrimer for the first time as a cross-linking agent for the synthesis of a

luminescent lanthanide-based multifunctional nanohybrids (YPO4:Tb3+@Fe3O4) with strong

luminescence properties that show their ability to label cervical cancer cells for bio-imaging.

In this respect, Wate et al.88 envisaged the immense biological scope of dendrimers and Fe3O4

nanoparticles. They realised that, if used concurrently with graphene oxide (GO), the

dendrimer-modified Fe3O4 nanoparticles would provide physico-chemical advantages such as

a solution/dispersion state and a superparamagnetic property, especially with regard to their

biological implications and in reducing the cellular toxicity of GO. Hence, a multicomponent

GO nanostructured system that consists of Fe3O4 nanoparticles, the PAMAM-G4-NH2

dendrimer and Cy5 has been fabricated, which exhibited high dispersion in an aqueous

Page 39: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

18

medium, strong NIR optical absorbance and magnetically responsive properties. In vitro

experiments provide a clear indication of successful uptake of the nanoparticles by MCF-7

breast cancer cells, and this multicomponent GO nanostructured system is seen to behave as a

bright and stable fluorescent marker.

1.4.3.2 Magnetic Resonance imaging (MRI)

MRI is a non-invasive imaging technique with high spatial resolution and tomographic

capabilities. The technique utilises the magnetic relaxation of water protons in the body to

generate images. However, the signals generated from the water protons are not enough for

accurate image construction, which might lead to improper diagnostics. This problem is

addressed by the use of contrast agents. In simplistic terms, the contrast agents create a local

magnetic field, which in turn affects the relaxation of water protons. These enhanced signals

are, thus, used to provide better images with improved contrast. The sensitivity in biological

targets depends on the specific and selective accumulation of the contrast agents at the target

site. Since quite some time, targeted MNPs have been used to enhance the MR (magnetic

resonance) signal sensitivity for in vivo tumour detection. In this direction, dendrimers have

been seen to have a significant effect on the corresponding MR relaxivities and physiological

properties of MNPs89. Thus, dendrimer-MNPs agents are expected to provide sharper images

with physiologically relevant contrast, enhanced blood-pool retention time and specific organ

uptake. The fine tuning of the size and end-group functionalities of the dendrimers provide

added advantage in this respect90. Superparamagnetic Fe3O4 nanoparticles have been shown to

be effective contrast agents in labelling cells in order to provide high sensitivity in MRI, but

this sensitivity depends on their ability to label cells with sufficient quantities of the

nanoparticles, which is otherwise challenging.

To address this issue, a cell-penetrating polyester dendron with peripheral guanidines

was developed and conjugated to the nanoparticles surface by Martin and his co-workers91. In

GL261 mouse glioma cells, the dendritic guanidine exhibited remarkable cell-penetrating

capabilities to the HIV-Tat 47-57 peptide for the transport of fluorescein, and when conjugated

to the nanoparticles, it exhibited significantly enhanced uptake, in comparison to nanoparticles

that have no dendron or dendrons with hydroxyl or amine peripheries. T2 values of the cell

Page 40: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

19

pellets, using a spin echo sequence on a 3T MRI scanner, were found to be 65 and 6.1 ms for

unlabelled and labelled cell pellets, respectively (Figure 1.8). The substantial decrease

observed in the transverse relaxation time (T2) of labelled cells that are relative to control cells

illustrates the potential utility of these nanoparticles in labelling cells for detection by MRI.

Figure 1.8 (a) Confocal laser scanning microscopy image of GL261 mouse glioma cells

following a 2 h incubation with nanoparticle and (b) MRI signal intensity of unlabelled cells

and cells labelled with nanoparticle, measured at 3T using a spin echo sequence91

Duanmu et al.92 reported water-soluble Fe3O4 nanoparticles that were coated with three

different generations (G1, G2 and G3) of melamine dendrons, and investigated the MRI

contrast-enhancement potential of dendron-functionalised nanoparticles. The R2 relaxivities for

the G2 and G3-modified nanoparticles were found to be significantly larger than those with G1

(with the G2, R2 value being the largest at ~260 mM-1s-1), which may be likely due to the

physico-chemical nature of the dendron. Though a detailed understanding of these generation-

specific effects requires further study, the strong transverse relaxivities with generation-

specific values and tunable physico-chemical properties that were obtained made them an ideal

choice as contrast agents in MRI applications. Acknowledging the status mentioned above,

Chang et al.52, 57 presented a breakthrough in the development of new synthetic dendrimer-

modified magnetic Fe3O4 conjugates in the enhancement of anatomical MR contrast. The

system is based on the conjugates of FA), poly (ethylene glycol) (PEG)-modified dendrimers

(PAMAM) with DOX and superparamagnetic Fe3O4 (FA-PEG-PAMAM-DOX@IONPs). For

MR imaging, C57BL/6 female mice (6–7 weeks) were implanted with B16F10 melanoma

cancer cells. Tumours are seen as hyper-intense areas in T2-weighted MR images, and for all

mice injected with IONPs, darkening of T2-weighted MR images from the tumour areas at 1 h

Page 41: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

20

post-injection relative to pre-injection indicated that the IONPs accumulated and made the

tumours clearly detectable (Figure 1.9).

Figure 1.9 T2-weighted fast spin echo images after injection of 2.5 mg/m1 conjugates per

mouse in 1 h (a) FA-PEG-G3.5 (b) PEG-G3.5@IONPs (c) FA-PEG-G3.5@IONPs. The

arrows denote allograft tumours, which are marked by circles52

Another encouraging attempt was made to achieve dual objectives of demonstrating

magnetic resonance and fluorescence imaging simultaneously by grafting small-sized dendrons

on the surface of Fe3O4 nanoparticles93. From the T1 and T2 values measured by relaxometry at

1.5T, mean relaxivity values of 272 mM-1s-1for r2 and with a 26.4 r2/r1 ratio were obtained for

the nanoparticles bearing carboxylate functions at their periphery. The authors attributed the

improved relaxivity values to the optimal design of the small-sized dendritic organic shell,

attractive magnetic properties of the inorganic core and the dendrons anchored through

phosphonate functionalities. This study confirmed the superiority of the dendritic approach to

develop new, smart and multimodal contrast agents.

1.5 Objectives of the Thesis

(a) Synthesis and characterisation of magnetite nanoparticles by the soft chemical route;

(b) Synthesis and characterisation of the peptide dendrimer;

(c) Development of multifunctional dendritic magnetic nanoparticles and optimisation of their

properties relevant for various cancer theranostic applications;

(d) Thorough in vitro assessment of these formulations for cancer therapeutics, magnetic

resonance imaging and in vivo assessment of their compatibility, biodistribution and magnetic

drug targeting; and

Page 42: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

21

(e) Establishing the comparative potential of as-prepared peptide dendrimers against the widely

used commercial polyamidoamine (PAMAM) dendrimers.

1.6 Outline of the Thesis

This thesis is organised into seven chapters and an appendix. The first chapter

introduces the functional nanomaterials, Fe3O4 nanoparticles and dendrimers, and thoroughly

reviews their current and probable use as theranostic agents in combating cancer. In the chapter

two, the synthesis and characterisation of aqueous colloidal suspension of citric acid-coated

Fe3O4 is reported. The chapter evaluates the biocompatibility, heating ability and therapeutic

efficacy of the drug-loaded nanoparticles against cervical cancer cells. Chapter three describes

the functionalisation of Fe3O4 with polyamidoamine dendrimer nanosystems to develop a

conjugate drug delivery platform. A comparative therapeutic evaluation of this system is dealt

with on the basis of the generation of the dendrimer. Section one of the fourth chapter, describes

the elaborate synthesis and characterisation of the peptide dendrimer. The properties of this

dendrimer were optimised so as to enhance the biocompatibility while minimally

compromising on their physico-chemical properties. Section two of the fourth chapter

describes the synthesis and characterisation of dendrimer-functionalised Fe3O4 nanoparticles

(both PAMAM and as-prepared peptide). An elaborate evaluation of their biocompatibility,

drug delivery efficiency, therapeutic efficacy and their potential use in in vitro combinatorial

chemo-thermotherapy is evaluated. Taking a step forward with these formulations, chapter five

reports the comparative in vivo assessment of biocompatibility, bio-distribution and therapeutic

efficacy of these nanoparticles in C57BL/6 black mice. The efficacy of DOX-loaded dendritic

Fe3O4 nanoparticles were evaluated for tumor regression via intravenous administration, based

on the concept of MDT. Chapter six evaluates the MR relaxivity properties of these

nanoparticles and its dependence on various parameters, mainly elevated temperatures. In

chapter seven, the conclusions of the present work and their intended prospective investigation

are discussed. The appendix one explores the combination of magnetic nanoparticles with lipid

vesicles that form magnetic liposomes for therapeutic efficacy of a hydrophobic drug.

1.7 References

1. K. C. Barick, M. Aslam, Y.-P. Lin, D. Bahadur, P. V. Prasad and V. P. Dravid, Journal

of Materials Chemistry, 2009, 19, 7023-7029.

Page 43: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

22

2. H. Gu, K. Xu, C. Xu and B. Xu, Chemical Communications, 2006, DOI:

10.1039/B514130C, 941-949.

3. F. Q. Hu, L. Wei, Z. Zhou, Y. L. Ran, Z. Li and M. Y. Gao, Advanced Materials, 2006,

18, 2553-2556.

4. D. A. Tomalia, H. Baker, J. Dewald, M. Hall, G. Kallos, S. Martin, J. Roeck, J. Ryder

and P. Smith, Polym J, 1985, 17, 117-132.

5. R. Esfand and D. A. Tomalia, Drug Discovery Today, 2001, 6, 427-436.

6. V. Percec, W. D. Cho, P. E. Mosier, G. Ungar and D. J. P. Yeardley, Journal of the

American Chemical Society, 1998, 120, 11061-11070.

7. P. Kesharwani and A. K. Iyer, Drug Discovery Today, 2015, 20, 536-547.

8. C.-H. Huang, K. Nwe, A. Al Zaki, M. W. Brechbiel and A. Tsourkas, ACS Nano, 2012,

6, 9416-9424.

9. J. Zhu and X. Shi, Journal of Materials Chemistry B, 2013, 1, 4199-4211.

10. A. Lakshminarayanan, V. K. Ravi, R. Tatineni, Y. B. R. D. Rajesh, V. Maingi, K. S.

Vasu, N. Madhusudhan, P. K. Maiti, A. K. Sood, S. Das and N. Jayaraman,

Bioconjugate Chemistry, 2013, 24, 1612-1623.

11. Y. Tang, Y.-B. Li, B. Wang, R.-Y. Lin, M. van Dongen, D. M. Zurcher, X.-Y. Gu, M.

M. Banaszak Holl, G. Liu and R. Qi, Molecular Pharmaceutics, 2012, 9, 1812-1821.

12. H. Nehoff, N. N. Parayath, L. Domanovitch, S. Taurin and K. Greish, International

journal of nanomedicine, 2014, 9, 2539-2555.

13. M. Ferrari, Nat Rev Cancer, 2005, 5, 161-171.

14. O. C. Farokhzad and R. Langer, Advanced Drug Delivery Reviews, 2006, 58, 1456-

1459.

15. V. P. Torchilin, Advanced Drug Delivery Reviews, 2006, 58, 1532-1555.

16. L. Dykman and N. Khlebtsov, Chemical Society Reviews, 2012, 41, 2256-2282.

17. D. K. Chatterjee, M. K. Gnanasammandhan and Y. Zhang, Small, 2010, 6, 2781-2795.

18. N. Chekina, D. Horak, P. Jendelova, M. Trchova, M. J. Benes, M. Hruby, V. Herynek,

K. Turnovcova and E. Sykova, Journal of Materials Chemistry, 2011, 21, 7630-7639.

19. A. Anitha, N. Deepa, K. P. Chennazhi, S. V. Nair, H. Tamura and R. Jayakumar,

Carbohydrate Polymers, 2011, 83, 66-73.

20. M. Shen and X. Shi, Nanoscale, 2010, 2, 1596-1610.

21. N. Lewinski, V. Colvin and R. Drezek, Small, 2008, 4, 26-49.

22. M. K. Jaiswal, R. Banerjee, P. Pradhan and D. Bahadur, Colloids and Surfaces B:

Biointerfaces, 2010, 81, 185-194.

Page 44: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

23

23. A. K. Gupta and M. Gupta, Biomaterials, 2005, 26, 3995-4021.

24. S. Laurent, D. Forge, M. Port, A. Roch, C. Robic, L. Vander Elst and R. N. Muller,

Chemical Reviews, 2008, 108, 2064-2110.

25. J. R. McCarthy, K. A. Kelly, E. Y. Sun and R. Weissleder, Nanomedicine, 2007, 2, 153-

167.

26. M. Mahmoudi, S. Sant, B. Wang, S. Laurent and T. Sen, Advanced Drug Delivery

Reviews, 2011, 63, 24-46.

27. H. Xu, Z. P. Aguilar, L. Yang, M. Kuang, H. Duan, Y. Xiong, H. Wei and A. Wang,

Biomaterials, 2011, 32, 9758-9765.

28. J. M. Perez, L. Josephson, T. O'Loughlin, D. Hogemann and R. Weissleder, Nat

Biotech, 2002, 20, 816-820.

29. H. Li, Q. Wei, J. He, T. Li, Y. Zhao, Y. Cai, B. Du, Z. Qian and M. Yang, Biosensors

and Bioelectronics, 2011, 26, 3590-3595.

30. C. Sun, J. S. H. Lee and M. Zhang, Advanced Drug Delivery Reviews, 2008, 60, 1252-

1265.

31. C. G. Hadjipanayis, R. Machaidze, M. Kaluzova, L. Wang, A. J. Schuette, H. Chen, X.

Wu and H. Mao, Cancer Research, 2010, 70, 6303-6312.

32. B. H. Kim, N. Lee, H. Kim, K. An, Y. I. Park, Y. Choi, K. Shin, Y. Lee, S. G. Kwon,

H. B. Na, J.-G. Park, T.-Y. Ahn, Y.-W. Kim, W. K. Moon, S. H. Choi and T. Hyeon,

Journal of the American Chemical Society, 2011, 133, 12624-12631.

33. X. Yang, H. Hong, J. J. Grailer, I. J. Rowland, A. Javadi, S. A. Hurley, Y. Xiao, Y.

Yang, Y. Zhang, R. J. Nickles, W. Cai, D. A. Steeber and S. Gong, Biomaterials, 2011,

32, 4151-4160.

34. H. Wei, O. T. Bruns, O. Chen and M. G. Bawendi, Integrative Biology, 2013, 5, 108-

114.

35. F. Dilnawaz, A. Singh, C. Mohanty and S. K. Sahoo, Biomaterials, 2010, 31, 3694-

3706.

36. B. Chertok, A. E. David and V. C. Yang, Biomaterials, 2010, 31, 6317-6324.

37. D. Losic, Y. Yu, M. S. Aw, S. Simovic, B. Thierry and J. Addai-Mensah, Chemical

Communications, 2010, 46, 6323-6325.

38. Q. Quan, J. Xie, H. Gao, M. Yang, F. Zhang, G. Liu, X. Lin, A. Wang, H. S. Eden, S.

Lee, G. Zhang and X. Chen, Molecular Pharmaceutics, 2011, 8, 1669-1676.

39. A. Solanki, J. D. Kim and K.-B. Lee, Nanomedicine, 2008, 3, 567-578.

Page 45: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

24

40. P. Pradhan, J. Giri, R. Banerjee, J. Bellare and D. Bahadur, Journal of Magnetism and

Magnetic Materials, 2007, 311, 208-215.

41. L. Lartigue, C. Innocenti, T. Kalaivani, A. Awwad, M. d. M. Sanchez Duque, Y. Guari,

J. Larionova, C. Guérin, J.-L. G. Montero, V. Barragan-Montero, P. Arosio, A.

Lascialfari, D. Gatteschi and C. Sangregorio, Journal of the American Chemical

Society, 2011, 133, 10459-10472.

42. M. Levy, A. Quarta, A. Espinosa, A. Figuerola, C. Wilhelm, M. García-Hernández, A.

Genovese, A. Falqui, D. Alloyeau, R. Buonsanti, P. D. Cozzoli, M. A. García, F.

Gazeau and T. Pellegrino, Chemistry of Materials, 2011, 23, 4170-4180.

43. L. A. Thomas, L. Dekker, M. Kallumadil, P. Southern, M. Wilson, S. P. Nair, Q. A.

Pankhurst and I. P. Parkin, Journal of Materials Chemistry, 2009, 19, 6529-6535.

44. S. Chandra, K. C. Barick and D. Bahadur, Advanced Drug Delivery Reviews, 2011, 63,

1267-1281.

45. Y. Zhang, N. Kohler and M. Zhang, Biomaterials, 2002, 23, 1553-1561.

46. D. A. Tomalia, A. M. Naylor and W. A. Goddard, Angewandte Chemie International

Edition in English, 1990, 29, 138-175.

47. D. Ling and T. Hyeon, Small, 2013, 9, 1449-1449.

48. L. Pradhan, R. Srivastava and D. Bahadur, Acta Biomaterialia, 2014, 10, 2976-2987.

49. J. H. Park, G. Saravanakumar, K. Kim and I. C. Kwon, Advanced Drug Delivery

Reviews, 2010, 62, 28-41.

50. G. Orive, R. M. Hernández, A. R. g. Gascón, A. Domı́nguez-Gil and J. L. Pedraz,

Current Opinion in Biotechnology, 2003, 14, 659-664.

51. X. Wu, X. He, L. Zhong, S. Lin, D. Wang, X. Zhu and D. Yan, Journal of Materials

Chemistry, 2011, 21, 13611-13620.

52. Y. Chang, X. Meng, Y. Zhao, K. Li, B. Zhao, M. Zhu, Y. Li, X. Chen and J. Wang,

Journal of Colloid and Interface Science, 2011, 363, 403-409.

53. X. He, X. Wu, X. Cai, S. Lin, M. Xie, X. Zhu and D. Yan, Langmuir, 2012, 28, 11929-

11938.

54. S. Chandra, S. Mehta, S. Nigam and D. Bahadur, New Journal of Chemistry, 2010, 34,

648-655.

55. R. Duncan and L. Izzo, Advanced Drug Delivery Reviews, 2005, 57, 2215-2237.

56. X. Shi, T. P. Thomas, L. A. Myc, A. Kotlyar and J. J. R. Baker, Physical Chemistry

Chemical Physics, 2007, 9, 5712-5720.

Page 46: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

25

57. Y. Chang, N. Liu, L. Chen, X. Meng, Y. Liu, Y. Li and J. Wang, Journal of Materials

Chemistry, 2012, 22, 9594-9601.

58. M. Li, K.-G. Neoh, R. Wang, B.-Y. Zong, J. Y. Tan and E.-T. Kang, European Journal

of Pharmaceutical Sciences, 2013, 48, 111-120.

59. C. Alexiou, R. Tietze, E. Schreiber, R. Jurgons, H. Richter, L. Trahms, H. Rahn, S.

Odenbach and S. Lyer, Journal of Magnetism and Magnetic Materials, 2011, 323,

1404-1407.

60. C. Alexiou and R. Jurgons, in Magnetism in Medicine, Wiley-VCH Verlag GmbH &

Co. KGaA, 2007, DOI: 10.1002/9783527610174.ch4h, pp. 596-605.

61. D. S. Mathew and R.-S. Juang, Chemical Engineering Journal, 2007, 129, 51-65.

62. C. Alexiou, W. Arnold, R. J. Klein, F. G. Parak, P. Hulin, C. Bergemann, W. Erhardt,

S. Wagenpfeil and A. S. Lübbe, Cancer Research, 2000, 60, 6641-6648.

63. B. Shapiro, S. Kulkarni, A. Nacev, S. Muro, P. Y. Stepanov and I. N. Weinberg, Wiley

Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, 2015, 7, 446-457.

64. K. A. Kennedy, B. A. Teicher, S. Rockwell and A. C. Sartorelli, Biochemical

Pharmacology, 1980, 29, 1-8.

65. G. Hegyi, G. P. Szigeti, Sz, #xe1, A. sz and #xe1, Evidence-Based Complementary and

Alternative Medicine, 2013, 2013, 12.

66. A. Jordan, R. Scholz, P. Wust, H. Fähling and F. Roland, Journal of Magnetism and

Magnetic Materials, 1999, 201, 413-419.

67. K. H. Bae, M. Park, M. J. Do, N. Lee, J. H. Ryu, G. W. Kim, C. Kim, T. G. Park and

T. Hyeon, ACS Nano, 2012, 6, 5266-5273.

68. G. Béalle, R. Di Corato, J. Kolosnjaj-Tabi, V. Dupuis, O. Clément, F. Gazeau, C.

Wilhelm and C. Ménager, Langmuir, 2012, 28, 11834-11842.

69. R. Love, R. Z. Soriano and R. J. Walsh, Cancer Research, 1970, 30, 1525-1533.

70. F. K. Storm, W. H. Harrison, R. S. Elliott and D. L. Morton, Cancer Research, 1979,

39, 2245-2251.

71. G. Glöckl, R. Hergt, M. Zeisberger, S. Dutz, S. Nagel and W. Weitschies, Journal of

Physics: Condensed Matter, 2006, 18, S2935.

72. M. Jeun, S. Bae, A. Tomitaka, Y. Takemura, K. H. Park, S. H. Paek and K.-W. Chung,

Applied Physics Letters, 2009, 95, 082501-082503.

73. H. Mamiya and B. Jeyadevan, Scientific Reports, 2011, 1, 157.

74. P. de la Presa, Y. Luengo, M. Multigner, R. Costo, M. P. Morales, G. Rivero and A.

Hernando, The Journal of Physical Chemistry C, 2012, 116, 25602-25610.

Page 47: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

26

75. N. K. Prasad, K. Rathinasamy, D. Panda and D. Bahadur, Journal of Materials

Chemistry, 2007, 17, 5042-5051.

76. P. Guardia, R. Di Corato, L. Lartigue, C. Wilhelm, A. Espinosa, M. Garcia-Hernandez,

F. Gazeau, L. Manna and T. Pellegrino, ACS Nano, 2012, 6, 3080-3091.

77. D. Baba, Y. Seiko, T. Nakanishi, H. Zhang, A. Arakaki, T. Matsunaga and T. Osaka,

Colloids and Surfaces B: Biointerfaces, 2012, 95, 254-257.

78. T. Sadhukha, L. Niu, T. S. Wiedmann and J. Panyam, Molecular Pharmaceutics, 2013,

10, 1432-1441.

79. T. Sadhukha, T. S. Wiedmann and J. Panyam, Biomaterials, 2013, 34, 5163-5171.

80. Y. Qu, J. Li, J. Ren, J. Leng, C. Lin and D. Shi, Nanoscale, 2014, 6, 12408-12413.

81. T.-J. Li, C.-C. Huang, P.-W. Ruan, K.-Y. Chuang, K.-J. Huang, D.-B. Shieh and C.-S.

Yeh, Biomaterials, 2013, 34, 7873-7883.

82. S. Kossatz, J. Grandke, P. Couleaud, A. Latorre, A. Aires, K. Crosbie-Staunton, R.

Ludwig, H. Dahring, V. Ettelt, A. Lazaro-Carrillo, M. Calero, M. Sader, J. Courty, Y.

Volkov, A. Prina-Mello, A. Villanueva, A. Somoza, A. Cortajarena, R. Miranda and I.

Hilger, Breast Cancer Research, 2015, 17, 66.

83. K. S. Kim, W. Park, J. Hu, Y. H. Bae and K. Na, Biomaterials, 2014, 35, 337-343.

84. N. Raghunand, J. Scicinski, B. Oronsky, G. Guntle, E. Bruckheimer and R. Korn,

Cancer Research, 2014, 74, 2068.

85. X. Wang, D. Niu, Q. Wu, S. Bao, T. Su, X. Liu, S. Zhang and Q. Wang, Biomaterials,

2015, 53, 349-357.

86. Z. Li, K. Dong, S. Huang, E. Ju, Z. Liu, M. Yin, J. Ren and X. Qu, Advanced Functional

Materials, 2014, 24, 3612-3620.

87. M. N. Luwang, S. Chandra, D. Bahadur and S. K. Srivastava, Journal of Materials

Chemistry, 2012, 22, 3395-3403.

88. S. W. Prateek, S. B. Shashwat, J.-B. Archana, R. M. Russel, R. Z. Khushbu, K. Jayant

and R. D. K. Misra, Nanotechnology, 2012, 23, 415101.

89. E. Strable, J. W. M. Bulte, B. Moskowitz, K. Vivekanandan, M. Allen and T. Douglas,

Chemistry of Materials, 2001, 13, 2201-2209.

90. J. W. Bulte, T. Douglas, B. Witwer, S. C. Zhang, E. Strable, B. K. Lewis, H. Zywicke,

B. Miller, P. van Gelderen, B. M. Moskowitz, I. D. Duncan and J. A. Frank, Nature

biotechnology, 2001, 19, 1141-1147.

91. A. L. Martin, L. M. Bernas, B. K. Rutt, P. J. Foster and E. R. Gillies, Bioconjugate

Chemistry, 2008, 19, 2375-2384.

Page 48: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

27

92. C. Duanmu, I. Saha, Y. Zheng, B. M. Goodson and Y. Gao, Chemistry of Materials,

2006, 18, 5973-5981.

93. G. Lamanna, M. Kueny-Stotz, H. Mamlouk-Chaouachi, C. Ghobril, B. Basly, A.

Bertin, I. Miladi, C. Billotey, G. Pourroy, S. Begin-Colin and D. Felder-Flesch,

Biomaterials, 2011, 32, 8562-8573.

Page 49: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

This chapter has been published in J. Magn. Magn. Mater., 2011, 323, 237–243.

Chapter 2

Citrate-stabilised Fe3O4 Nanoparticles: Conjugation and

Release of Doxorubicin for Cancer Therapeutics

2.1 Introduction

Magnetic nanoparticles have received a great deal of attention due to their potential in

biomedical applications such as hyperthermia treatment of cancer, as a contrast agent for

magnetic resonance imaging, magnetic separation and sorting of cells and proteins (bio

recognition), and controlled and targeted drug delivery1-6. In the recent past, among the vast

varieties of magnetic nanoparticles, superparamagnetic Fe3O4 nanoparticles have emerged as

an excellent candidate for biomedical applications due to their better chemical stability and

biocompatibility in comparison to other metallic magnetic nanoparticles7, 8. Many methods

have been developed to prepare superparamagnetic Fe3O4 nanoparticles9. The thermal

decomposition of organometallic precursors in an organic solvent (that has a high boiling point)

at elevated temperatures in the presence of surfactants has been successfully used for the

synthesis of monodisperse Fe3O4 nanoparticles3, 10. However, the Fe3O4 nanoparticles prepared

by these methods are highly hydrophobic, which hampers their biomedical applications,

especially, in drug delivery, hyperthermia treatment of cancer and magnetic resonance

imaging. Although many ligand-exchange processes have been established to offer them a

hydrophilic surface characteristic for aqueous stability, their magnetic field responsiveness has

Page 50: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

29

not been effectively improved3, 9. Therefore, much effort has been focused on the fabrication

of biocompatible aqueous stable superparamagnetic Fe3O4 nanoparticles and engineering them

with the desired functionality, which offer good magnetic responses, through soft-chemical

routes2, 11-16.

The strategies used for surface functionalisation comprise grafting of, or coating with

organic species, such as surfactants or polymers, or coating with an inorganic layer such as

silica or gold13-22. Further, the presence of these biocompatible layers on the surface not only

stabilises the Fe3O4 nanoparticles, but also provides an accessible surface for routine

conjugation of biomolecules through the well-developed bioconjugation chemistry. Thus, the

stability of the bonding between functional molecules and Fe3O4 nanoparticles is crucial from

the point of view of the application. The small-molecule targeting groups are attractive due to

their ease of preparation and simple conjugation chemistry18, 23, 24. Further, multiple grafting or

coating of small molecules can provide multivalent systems that exhibit significantly enhanced

efficacy in drugs and biomolecules25. On the other hand, some binding affinity may be lost

through steric hindrances by large surfactant molecules or long polymer chains, which could

be easily overcome by the use of small molecules that have multiple functional groups such as

carboxyl (COOH), amine (NH2), thiol (SH) and the like. Furthermore, the presence of a large

number of uncoordinated functional groups on the surface of the magnetic nanoparticle can be

used for the linking of various biomolecules and drugs.

Among various small molecules, citrate moiety has been extensively used in the

preparation of aqueous stable Fe3O4 nanoparticles and in their conjugation to biomolecules and

drugs by exploiting the uncoordinated carboxyl groups present on the surface of nanoparticles.

Liu et al.26 fabricated highly water-dispersible Fe3O4 particles that have a uniform size by

solvothermal reaction at 200 °C through the reduction of FeCl3 in the presence of trisodium

citrate as a stabiliser. They found that these Fe3O4 nanoparticles with surface citrate groups can

effectively enrich peptides at a trace level. Munnier et al.12 developed a new method for

reversible association of drug (DOX) to citrate-stabilised Fe3O4 nanoparticles that were

prepared by agitating bare nanoparticles in a citric acid solution. Khosroshahi and Ghazanfari17

fabricated citrate-modified Fe3O4 nanoparticles by stirring bare Fe3O4 nanoparticles in

trisodium citrate solution as an intermediate to obtain silica-coated Fe3O4 core-shell

nanoparticles. However, most of these works on the fabrication of aqueous stabilised Fe3O4

nanoparticles are achieved at either elevated temperatures2, 3, 26 or involve multiple synthesis

steps12, 13, 15. The goal of this chapter was to develop a facile single-step process for the

preparation of highly biocompatible citric acid functionalised (citrate-stabilised) Fe3O4

Page 51: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

30

aqueous colloidal magnetic nanoparticles (CA–Fe3O4), with optimal magnetisation at low

temperatures, as potential drug carriers, which can also be used as effective heating source for

the hyperthermic treatment of cancer.

2.2 Experimental and Characterisation Techniques

2.2.1 Synthesis of Citrate-stabilised Fe3O4 Nanoparticles

In a typical synthesis, 4.44 g of FeCl3 and 1.732 g of FeCl2 were dissolved in 80 ml of

water in a round-bottomed flask and the temperature was slowly increased to 70 °C in a

refluxing condition under a nitrogen atmosphere with constant mechanical stirring at 1000 rpm.

The temperature was maintained at 70 °C for 30 min. Further, 20 ml of ammonia solution was

added promptly to the reaction mixture and kept at the same temperature for another 30 min.

Then 4 ml of an aqueous solution of citric acid (0.5 g/ml) was added to the reaction mixture

mentioned above and the reaction temperature was slowly raised to 90 °C under reflux and

reacted for 60 min with continuous stirring. Black precipitates were obtained by cooling the

reaction mixture to room temperature, followed by thorough rinsing with water. During each

rinsing step, samples were separated from the supernatant by using a permanent magnet.

2.2.2 Drug Loading and Release

The anticancer agent, doxorubicin hydrochloride (DOX) was used as a model drug to

estimate the drug release behaviour of the CA–Fe3O4. In order to investigate the interaction of

the drug molecules with CA–Fe3O4, we have studied the fluorescence spectra of pure DOX and

DOX loaded CA–Fe3O4 in addition to zeta-potential measurements. The aqueous dispersion of

different amounts of CA–Fe3O4 (0, 20, 40, 60, 80 and 100 mg from a stock suspension of 2

mg/ml) was added to 1 ml of the DOX solution (10 µg/ml) and mixed thoroughly by shaking

at room temperature for 15 min. The fluorescence spectra of the supernatant (obtained after

magnetic sedimentation of the drug loaded CA–Fe3O4) were then recorded. The fluorescence

spectra of 1 ml of pure DOX (10 mg/ml) were also taken at different time intervals for

comparative studies. The fluorescence intensities of supernatants (washed drug molecules were

also taken into consideration for calculations) against those of the pure DOX solution were

used to determine the loading efficiency (binding isotherm of DOX with CA–Fe3O4). The

loading efficiency (w/w%) was calculated using the following relation:

Page 52: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

31

% 𝐿𝑜𝑎𝑑𝑖𝑛𝑔 𝑒𝑓𝑓𝑖𝑐𝑖𝑒𝑛𝑐𝑦 = 𝐼𝐷𝑂𝑋− 𝐼𝑆− 𝐼𝑊

𝐼𝐷𝑂𝑋 × 100 (Eqn. 2.1)

where, IDOX is the fluorescence intensity of the pure DOX solution; IS, the fluorescence

intensity of supernatant; and IW, the fluorescence intensity of washed DOX (physically

adsorbed DOX molecules).

The drug release study was carried out under a reservoir-sink conditions. For the release

study, we have quantified the amount of DOX-loaded CA–Fe3O4 according to the binding

isotherm. The loading was carried out at an increased scale by incubating 2 ml of the aqueous

solution of DOX (2 mg/ml) with 1 ml of the aqueous suspension of CA–Fe3O4 (10 mg/ml) for

1 h in the dark (however, no decrease in fluorescence intensities was observed after 15 min of

incubation). The drug loaded CA–Fe3O4 (10 mg) was immersed in 5 ml of an acetate buffer

(pH 5), and then put into a dialysis bag. The dialysis was performed against 200 ml of

phosphate buffered saline (PBS) with a pH of 7.3, under continuous stirring at 37 °C to mimic

the environment of the cell. 1 ml of the external medium was withdrawn and replaced with

fresh PBS at fixed times in order to maintain the sink conditions. The amount of doxorubicin

released was determined by measuring the fluorescence intensity at λex = 490 nm and λem =

535±35 nm against the standard plot that was prepared under similar conditions. Each

experiment was performed in triplicates; the standard deviation is given in the plot.

2.2.3 Calorimetric Measurements

Time-dependent calorimetric measurements were done to evaluate the heating

capabilities of these CA–Fe3O4. Towards this end, 1 ml (10 mg/ml of Fe) of Fe3O4 colloidal

suspension was taken in a glass sample holder with suitable arrangements to minimise the heat

loss. An alternating current magnetic field (ACMF) of 7.64, 8.2 and 10.0 kA/m, and a fixed

frequency of 425 kHz were used to evaluate the specific absorption rate (SAR). The SAR was

calculated using the following equation3, 16, 27:

𝑆𝐴𝑅 = 𝐶 × ∆𝑇

∆𝑡 ×

1

𝑚𝐹𝑒 (Eqn. 2.2)

where, C is the specific heat of the solvent (C=Cwater=4.18 J/g °C); ΔT/Δt is the initial slope of

the time-dependent temperature curve; and mFe is the mass fraction of Fe in the sample.

Page 53: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

32

2.2.4 In vitro Evaluation

The Sulforhodamine-B (SRB) assay was performed to evaluate the cytocompatibility

of the CA–Fe3O4 with HeLa cells. The cells were seeded into 96-well plates at densities of

1×104 cells per well for 24 h. Further, different concentrations of the CA–Fe3O4 colloidal

suspension (0, 31.250, 15.625, 7.813, 3.906, 1.953, 0.977, and 0.488 mg/ml of Fe) were added

to the cells and incubated for 24 h at 37 °C and 5% CO2. Thereafter, the cells were washed

thrice with PBS and processed for the SRB assay to determine the cell viability. For this, cells

were fixed with a solution of 10% trichloroacetic acid and stained with 0.4% SRB that was

dissolved in 1% acetic acid. The cell-bound dye was extracted with 10 mM unbuffered Tris

buffer solution (pH 10.5), after which the absorbance was measured at 560 nm by using a

multiwell plate reader. The cell viability was calculated using the following formula:

% 𝐶𝑒𝑙𝑙 𝑉𝑖𝑎𝑏𝑖𝑙𝑖𝑡𝑦 = 𝐴𝑏𝑠𝑜𝑟𝑏𝑎𝑛𝑐𝑒 𝑜𝑓 𝑡𝑟𝑒𝑎𝑡𝑒𝑑 𝑐𝑒𝑙𝑙𝑠

𝐴𝑏𝑠𝑜𝑟𝑏𝑎𝑛𝑐𝑒 𝑜𝑓 𝑐𝑜𝑛𝑡𝑟𝑜𝑙 𝑐𝑒𝑙𝑙𝑠 × 100 (Eqn. 2.3)

2.2.5 Characterisation Techniques

The X-ray diffraction (XRD) pattern was recorded on a Philips powder diffractometer

PW3040/60 with Cu Kα radiation. The surface modification of the nanoparticles was analysed

using a Fourier transform infrared spectrometer (FTIR) (Magna 550, Nicolet Instruments

Corporation, USA) in the range of 4000–400 cm-1. The particle size was determined by using

a high-resolution transmission electron microscope (HRTEM), JEOL JAM 2100F, which

operated at 200 kV. The thermal analyses were performed by a TA Instruments SDT Q600

analyser under N2 atmosphere from room temperature to 800 °C, with a heating rate of 10

°C/min. The elemental analysis was carried out by the FLASH EA 1112 series CHNS (O)

analyser (Thermo Fennigan, Italy). The hydrodynamic diameter and zeta-potential were

determined by dynamic light scattering (DLS) and Zeta PALS, respectively, (BI-200

Brookhaven Instruments Corp). The magnetic measurements of dried and powdered samples

were carried out using a vibrating sample magnetometer (VSM, LakeShore, Model-7410). The

Curie temperature was measured in an applied field of 100 Oe. In order to evaluate the SAR,

the amount of iron in the nanoparticle suspension was determined by UV (Cecil, Model No.

CE3021) by using the established phenanthroline spectrophotometric method28. The standard

curve was prepared from a stock iron solution under similar conditions (R2=0.998). The heat-

generating capability of the nanoparticles was evaluated by their SAR under the ACMF by a

radio frequency generator (EASY HEAT, EZLI5060) that operated at a fixed frequency of 247

Page 54: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

33

kHz. All the fluorescence spectra were recorded on the Hitachi F 2500 fluorescence

spectrophotometer. The fluorescence intensities of aliquots for determination of the amounts

of the released drug were recorded using a Perkin Elmer 1420 multilabel counter.

2.3 Results and Discussions

2.3.1 Nanoparticle Characterisation

Figure 2.1 shows (a) XRD pattern and (b) TEM micrograph of CA–Fe3O4. The XRD

pattern reveals the formation of a single-phase Fe3O4 inverse spinel structure with a lattice

constant, a = ~8.378 Å, which is very close to the reported value of magnetite (JCPDS Card

No. 88-0315, a = 8.375 Å). The presence of sharp and intense peaks confirmed the formation

of highly crystalline nanoparticles. The crystallite size of CA–Fe3O4 is estimated to be

approximately 8 nm from X-ray line broadening using the Scherrer formula. From the TEM

micrograph, it is clearly observed that Fe3O4 nanoparticles are almost spherical and are around

8–10 nm (σ≤10%) in diameter. The selected area electron diffraction pattern (SAED) of this

sample (inset of Figure 2.1b) can be indexed to the reflections of the inverse spinel Fe3O4

structure and shows only the diffraction intensity that is associated with highly crystalline

Fe3O4, which is consistent with the XRD result.

Figure 2.1 (a) XRD pattern and (b) TEM micrograph of CA–Fe3O4 (Inset of (b) shows the

selected area electron diffraction pattern of CA–Fe3O4)

30 40 50 60 70

Inte

ns

ity

(a.u

.)

2 (Degree)

(22

0)

(31

1)

(40

0)

(42

2)

(51

1)

(44

0)

(a)

(b)

Page 55: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

34

Figure 2.2 shows the FTIR spectra of pure CA and CA–Fe3O4. The absorption bands

for the pure CA are well resolved, but those of the CA–Fe3O4 are rather broad and few. The

1710 cm-1 peak assignable to the C=O vibration (asymmetric stretching) from the COOH group

of CA shifts to an intense band at about 1600 cm-1 for CA–Fe3O4, revealing the binding of a

CA radical to surface of Fe3O4 nanoparticles by chemisorption of carboxylate (citrate) ions 18,

29. Carboxylate groups of CA form complexes with Fe atoms on the surface of Fe3O4, imparting

a partial single bond character to the C=O bond. This results in the weakening of the C=O

bond, which shifts the stretching frequency to a lower value. Furthermore, the vibrational

modes that appear at 1400, 1250 and 1065 cm-1 in CA–Fe3O4 corresponds to the symmetric

stretching of COO–, symmetric stretching of C–O, and the OH group of CA30. The strong IR

band observed at around 575 cm-1 in CA–Fe3O4 can be ascribed to the Fe–O stretching

vibrational mode of Fe3O4.

Figure 2.2 FTIR spectra of pure CA and CA–Fe3O4

Figure 2.3A shows the TGA-DTA plots of CA–Fe3O4. A weight loss of ~7.5% with a

sharp endothermic peak at ~60 °C can be ascribed to the removal of physically absorbed water

and CA molecules on the Fe3O4 nanoparticles. The weight loss of about 13.5% with a broad

exothermic peak at ~260 °C can be associated with the removal of chemically attached CA

molecules from the surface of Fe3O4 nanoparticles. The weight loss of ~3.0% beyond 400 °C,

with a sharp exothermic peak at ~500 °C is associated with the phase transformation of Fe3O4–

Fe2O3. Furthermore, elemental analysis shows the presence of organic components (carbon and

Page 56: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

35

hydrogen) on CA–Fe3O4. Thus, the FTIR, TGA and CHNS(O) results confirmed that Fe3O4

nanoparticles have been functionalised with citric acid during the course of the synthesis.

Figure 2.3 (A) TGA-DTA plots of CA–Fe3O4 (B) Zeta-potential of CA–Fe3O4 at different

pH values. Inset ‘a’ shows the hydrodynamic diameter of CA–Fe3O4 obtained from DLS

measurements; inset ‘b’ shows the possible schematic representation of CA–Fe3O4 (CA

coating on the surface of the Fe3O4)

Figure 2.3B shows the zeta-potential of CA–Fe3O4 at different pH values. From zeta-

potential measurements, it has been observed that adsorption of CA onto the surface of Fe3O4

nanoparticles results in a highly negative surface charge; also, the isoelectric point is not

observed in the measured pH range of 3–6 (isoelectric point of bare Fe3O4 nanoparticles is 6.7).

These highly negative values of the zeta-potential for the CA–Fe3O4 further confirmed the

presence of negatively charged carboxylate groups on the surface of the Fe3O4 nanoparticles.

Furthermore, DLS measurements (Inset ‘a’ of Figure 2.3B) indicate that these samples result

in an aqueous colloidal suspension with mean hydrodynamic diameters (almost constant with

invariable change in the polydispersity index) of about 25 nm (σ≤10%) due to the presence of

associated and hydrated organic layers3, 31. Specifically, some of the carboxylate groups of

citric acid strongly coordinate to iron cations on the Fe3O4 surface to form a robust coating (a

possible schematic representation of CA–Fe3O4 is shown in the inset ‘b’ of Figure 2.3B), while

uncoordinated carboxylate groups extend into the water medium, conferring a high degree of

water stability to the Fe3O4 nanoparticles. Further, the electrostatic repulsive forces originate

among the highly negatively charged CA–Fe3O4 (–25.5 mV at pH 6) in aqueous suspensions,

which also play an important role in their water stabilisation.

Page 57: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

36

Figure 2.4 shows the field-dependent magnetisation (M vs. H) plot of CA–Fe3O4 at 300

K. The CA–Fe3O4 exhibits superparamagnetic behaviour without magnetic hysteresis and

remanence. The maximum magnetisation of CA–Fe3O4 was found to be 57 emu/g at 20 kOe.

The observed magnetisation is comparable to that of neat Fe3O4 nanoparticles (60.5 emu/g),

which is obtained by the co-precipitation method32 and is higher than the aqueous stable Fe3O4

nanoparticles (43.2 emu/g) that are obtained by the high-temperature thermal decomposition

method followed by subsequent surface functionalisation through ligand-exchange strategy3.

Thus, these aqueous stable Fe3O4 nanoparticles that have a high magnetic response can be

exploited for magnetic drug targeting, hyperthermia treatment and magnetic resonance

imaging. Further, the temperature dependence of magnetisation (M vs. T) measurement (inset

of Figure 2.4) shows that the Curie temperature (TC) of CA–Fe3O4 is around 580 °C, which is

in agreement with that reported for Fe3O43, 33, whereas, the TC of γ-Fe2O3 is around 645 °C34.

These results confirm that the phase formed is Fe3O4 rather than γ-Fe2O3. The slight hump

observed in the M vs. T curve at around 300 °C may be assigned to an increase in inter-particle

interaction near the surface due to the removal of organic moiety, (citric acid) as is suggested

by the TGA-DTA analysis.

Figure 2.4 Field dependence of magnetisation (M vs. H) plot of CA–Fe3O4 at 300 K. Inset

shows the temperature dependence of magnetisation (M vs. T) measurement

2.3.2 Drug Loading and Release

We have used the fluorescence spectra and zeta-potential analyses to investigate the

interactions of drug molecules with CA–Fe3O4, and their loading efficiency. At a low pH (4–

Page 58: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

37

6), the protonated primary amine present on the drug induces a positive charge in the

doxorubicin molecule (cationic DOX)35. While at low pH, the carboxylic moiety of citric acid

(pK1=3.13, pK2=4.76 and pK3=6.40) is deprotonated and carries a negative charge36. The

surface charge of the CA–Fe3O4 at pH 6 (the pH at which the drug loading experiment was

carried out) was found to be negative (–25.5 mV) from the zeta-potential measurement. An

increase in the surface charge, from –25.5 to –10.5 mV, is observed in the CA–Fe3O4 after drug

loading (100 µg DOX reacted with 200 µg CA–Fe3O4 from their stock solutions of 2 mg/ml)

through the zeta-potential measurement. This result strongly suggested that positively charged

drug molecules are bound to negatively charged CA–Fe3O4 through electrostatic interactions.

The impressive affinity of doxorubicin for negatively charged molecules such as oleate ions

and phospholipids has been the subject of numerous investigations37-39. The interaction of DOX

molecules with CA–Fe3O4 was also evident from the predominant quenching of DOX

fluorescence in the presence of CA–Fe3O4 (Figure 2.5a), whereas, self-quenching (DOX–DOX

interaction due to π–π stacking) of pure DOX is not observed (Figure 2.5b). Furthermore, the

fluorescence intensity of DOX decreases (till saturation loading is achieved) on increasing the

concentration of CA–Fe3O4, which is obvious due to the increase in the loading efficiency of

DOX into CA–Fe3O4. The fluorescence spectroscopy (the fluorescence intensity is highly

dependent on the state of the molecule, that is, free or in the attached form) has been

successfully used to study interactions between DOX and its surrounding in previous literature,

for instance, when the drug intercalates DNA or penetrates within membrane models or

liposomal drug carriers40-43. The loading efficiency (binding isotherm of DOX with CA–Fe3O4)

obtained from quenching of fluorescence intensities is shown in the inset of Figure 2.5a. From

the inset of Figure 2.5a, it has been observed that loading efficiency is strongly dependent on

the ratio of the particles to DOX in the reaction solution and a maximum of around 90% (σ≤5%)

drug loading efficiency (w/w) could be achieved by electrostatic interactions. The obtained

loading efficiency is much higher than that reported (14%) by Munnier et al.12. They have

stated that the drug molecules (DOX–Fe2+ complex) were attached to the surface –OH groups

of the citric acid treated Fe3O4 nanoparticles through Fe2+ ions. The presence of citric acid

moieties on the surface of Fe3O4 nanoparticles may provide steric hindrance to the attachment

of the DOX–Fe2+ complex with the surface –OH groups, thereby reducing the loading of the

drug onto the nanoparticles. However, the drug loading is attributed primarily to the

electrostatic interactions between positively charged DOX molecules and negatively charged

carboxyl moieties that are present on the surface of Fe3O4 nanoparticles (as is suggested from

zeta-potential measurements), showing comparatively higher drug loading.

Page 59: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

38

Figure 2.5 (a) Normalised fluorescence spectra of (a) 10 mg/ml of DOX (1 ml) reacted with

different amounts (0, 20, 40, 60, 80 and 100 mg) of CA–Fe3O4 for 15 min. Inset shows the

loading efficiency (binding isotherm of DOX with CA–Fe3O4) obtained from quenching of

fluorescence intensities; (b) 10 mg/ml of pure DOX (1 ml) at different time intervals; and (c)

Drug release profile of DOX-loaded CA–Fe3O4 in a cell mimicking environment (reservoir:

pH 5 and sink: pH 7.3 at 37 °C)

Figure 2.5c shows the drug release profile of DOX-loaded CA–Fe3O4 in a cell mimicking

environment (reservoir: pH 5 and sink: pH 7.3 at 37 °C). It has been observed that drug

molecules release slowly over a period of 50 h; the shape of the release profile suggests that

the complete release of drug was not attained. The drug loaded CA–Fe3O4 released around 60%

of the loaded drug in acetate buffer (pH 5) against the PBS (pH 7.3) after 50 h. The release of

DOX could be attributed to the weakening of the electrostatic interactions between the drug

and the partially neutralised carboxyl groups on the nanoparticle surface, which is due to an

increase in the protons in the colloidal solution. Munnier et al.12 discussed that release of DOX

molecules (released gradually over a period of 1 h and, thereby, attaining a plateau) from loaded

particles (achieved by chelation of the DOX–Fe2+ complex with –OH groups on the surface of

citric acid treated Fe3O4 nanoparticles) is primarily due to stimulated hydrolysis of drug

molecules. However, in this study, it was seen that weakening of the electrostatic interactions

is a slower process, which leads to the sustained release of drug molecules over a period of 50

h. These results indicate that the bound drug molecules will be released in appreciable amounts

in the mild acidic environments of the tumours.

2.3.3 Calorimetric Measurements

Time-dependent calorimetric measurements were performed on a suspension of CA–

Fe3O4 in order to investigate their heating efficacy (Figure 2.6a) as an additional functionality.

The SAR of CA–Fe3O4 was found to be 32.26, 38.63 and 49.24 W/g of Fe with an applied field

c

Page 60: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

39

(H) of 7.64, 8.82 and 10.0 kA/m, respectively (at a fixed frequency of 425 kHz). It has been

observed that the time required to reach 43 °C (hyperthermia temperature) decreases with an

increase in the field (Figure 2.6a), which is obvious as the heat generation is proportional to

the square of the applied ACMF. These SAR values should not be viewed in terms of

performances, but only as the demonstration of the fact that these nanoparticles are effective

heating sources for hyperthermia treatment of cancer.

Figure 2.6 (a) Time-dependent calorimetric measurements of CA–Fe3O4; and (b) Viabilities

of HeLa cells incubated with medium that contains CA–Fe3O4

2.3.4 In vitro Evaluation

Figure 2.6b shows the viabilities of HeLa cells incubated with medium that contains

CA–Fe3O4. The SRB assay results indicate that the viability of the HeLa cells is not affected

by the mere presence of CA–Fe3O4, which results in the registering of normal growth in the

cells, suggesting that nanoparticles are reasonably biocompatible, and do not have a toxic

effect, and may be used under further in vivo settings. The percentage of cell viability is slightly

above 100% at lower concentration, which may be due to the presence of iron (Fe3O4

nanoparticles) that sometimes facilitates cell growth3, 31.

This study discussed the formation of aqueous-stable, highly crystalline, biocompatible

citric acid functionalised Fe3O4 nanoparticles that have optimal magnetisation, higher drug

loading efficiencies and a good SAR, which could find promising applications in drug delivery

and hyperthermia treatment of cancer.

Page 61: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

40

2.4 Summary

A simple facile approach for the preparation of citrate stabilized Fe3O4 aqueous

colloidal nanoparticles of 8–10 nm by using a soft chemical approach is described in this

chapter. The detailed structural analyses by FTIR, TGA-DTA, CHNS(O) and zeta-potential

confirmed the functionalisation of Fe3O4 nanoparticles with citric acid. These nanoparticles

exhibited good colloidal stability, optimal magnetisation, in vitro biocompatibility and good

specific absorption rate (under an external ACMF). It was evident that the positively charged

drug molecules such as DOX could easily bound to the negatively charged CA–Fe3O4 through

electrostatic interactions. More specifically, a drug loading efficiency of about 90% (w/w) was

achieved by electrostatic interactions of drug molecules (DOX) with CA–Fe3O4. The drug

release profile in a cell mimicking environment indicated that the bound drug molecules will

be released in appreciable amounts in the mildly acidic environments of the tumours. Thus,

CA–Fe3O4 can be used as a potential carrier for effective magnetic drug targeting and

hyperthermia treatment of cancer.

However, the drug loaded on to CA–Fe3O4 is attached to the surface functional groups

and, thus, is very vulnerable to pH change in its immediate microenvironment. In order to avoid

this undesired release of DOX, various macromolecules could be further used to bind on the

nanoparticles surface. These macromolecules could effectively provide an accessible surface

for routine conjugation of biomolecules through the well-developed bioconjugation chemistry.

Therefore, dendrimers were used to conjugate to the surface groups of Fe3O4 nanoparticles in

the subsequent work. This also enhanced the aqueous stability and physico-chemical properties

of Fe3O4, and developed them into an improved platform as drug delivery vectors.

2.5 References

1. J. Cheon and J.-H. Lee, Accounts of Chemical Research, 2008, 41, 1630-1640.

2. K. C. Barick, M. Aslam, P. V. Prasad, V. P. Dravid and D. Bahadur, Journal of

Magnetism and Magnetic Materials, 2009, 321, 1529-1532.

3. K. C. Barick, M. Aslam, Y.-P. Lin, D. Bahadur, P. V. Prasad and V. P. Dravid, Journal

of Materials Chemistry, 2009, 19, 7023-7029.

4. H. Gu, K. Xu, C. Xu and B. Xu, Chemical Communications, 2006, DOI:

10.1039/B514130C, 941-949.

Page 62: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

41

5. F. Q. Hu, L. Wei, Z. Zhou, Y. L. Ran, Z. Li and M. Y. Gao, Advanced Materials, 2006,

18, 2553-2556.

6. S. Mornet, S. Vasseur, F. Grasset and E. Duguet, Journal of Materials Chemistry, 2004,

14, 2161-2175.

7. Z. Liu, J. Ding and J. Xue, New Journal of Chemistry, 2009, 33, 88-92.

8. A. Petri-Fink, M. Chastellain, L. Juillerat-Jeanneret, A. Ferrari and H. Hofmann,

Biomaterials, 2005, 26, 2685-2694.

9. D. Ling, N. Lee and T. Hyeon, Accounts of Chemical Research, 2015, 48, 1276-1285.

10. O. Bixner, A. Lassenberger, D. Baurecht and E. Reimhult, Langmuir, 2015, 31, 9198-

9204.

11. D. Ling and T. Hyeon, Small, 2013, 9, 1449-1449.

12. E. Munnier, S. Cohen-Jonathan, C. Linassier, L. Douziech-Eyrolles, H. Marchais, M.

Soucé, K. Hervé, P. Dubois and I. Chourpa, International Journal of Pharmaceutics,

2008, 363, 170-176.

13. D. Yang, J. Hu and S. Fu, The Journal of Physical Chemistry C, 2009, 113, 7646-7651.

14. A. Bee, R. Massart and S. Neveu, Journal of Magnetism and Magnetic Materials, 1995,

149, 6-9.

15. S. Chandra, S. Mehta, S. Nigam and D. Bahadur, New Journal of Chemistry, 2010, 34,

648-655.

16. P. Pradhan, J. Giri, G. Samanta, H. D. Sarma, K. P. Mishra, J. Bellare, R. Banerjee and

D. Bahadur, Journal of Biomedical Materials Research Part B: Applied Biomaterials,

2007, 81B, 12-22.

17. M. E. Khosroshahi and L. Ghazanfari, Physica E: Low-dimensional Systems and

Nanostructures, 2010, 42, 1824-1829.

18. Y. Sahoo, A. Goodarzi, M. T. Swihart, T. Y. Ohulchanskyy, N. Kaur, E. P. Furlani and

P. N. Prasad, The Journal of Physical Chemistry B, 2005, 109, 3879-3885.

19. M. Carmen Bautista, O. Bomati-Miguel, M. del Puerto Morales, C. J. Serna and S.

Veintemillas-Verdaguer, Journal of Magnetism and Magnetic Materials, 2005, 293,

20-27.

20. S. Li, Y. Ma, X. Yue, Z. Cao and Z. Dai, New Journal of Chemistry, 2009, 33, 2414-

2418.

21. A. I. Lesnikovich, T. M. Shunkevich, V. N. Naumenko, S. A. Vorobyova and M. V.

Baykov, Journal of Magnetism and Magnetic Materials, 1990, 85, 14-16.

Page 63: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

42

22. J. L. Lyon, D. A. Fleming, M. B. Stone, P. Schiffer and M. E. Williams, Nano Letters,

2004, 4, 719-723.

23. Y. Sahoo, H. Pizem, T. Fried, D. Golodnitsky, L. Burstein, C. N. Sukenik and G.

Markovich, Langmuir, 2001, 17, 7907-7911.

24. S. Nayak, H. Lee, J. Chmielewski and L. A. Lyon, Journal of the American Chemical

Society, 2004, 126, 10258-10259.

25. M. Mammen, S.-K. Choi and G. M. Whitesides, Angewandte Chemie International

Edition, 1998, 37, 2754-2794.

26. J. Liu, Z. Sun, Y. Deng, Y. Zou, C. Li, X. Guo, L. Xiong, Y. Gao, F. Li and D. Zhao,

Angewandte Chemie International Edition, 2009, 48, 5875-5879.

27. M. Ma, Y. Wu, J. Zhou, Y. Sun, Y. Zhang and N. Gu, Journal of Magnetism and

Magnetic Materials, 2004, 268, 33-39.

28. B. Ding, S. Xia, K. Hayat and X. Zhang, Journal of Agricultural and Food Chemistry,

2009, 57, 2938-2944.

29. M. Răcuciu, D. E. Creangă and A. Airinei, Eur. Phys. J. E, 2006, 21, 117-121.

30. J.-J. Max and C. Chapados, The Journal of Physical Chemistry A, 2004, 108, 3324-

3337.

31. J. Wan, W. Cai, X. Meng and E. Liu, Chemical Communications, 2007, DOI:

10.1039/B712795B, 5004-5006.

32. Y.-Q. Zhang, X.-W. Wei and R. Yu, Catal Lett, 2010, 135, 256-262.

33. K. C. Barick and D. Bahadur, Bull Mater Sci, 2006, 29, 595-598.

34. Ö. Özdemir, Physics of the Earth and Planetary Interiors, 1990, 65, 125-136.

35. F. Tewes, E. Munnier, B. Antoon, L. Ngaboni Okassa, S. Cohen-Jonathan, H. Marchais,

L. Douziech-Eyrolles, M. Soucé, P. Dubois and I. Chourpa, European Journal of

Pharmaceutics and Biopharmaceutics, 2007, 66, 488-492.

36. F. Cássio and C. Leáo, Applied and Environmental Microbiology, 1991, 57, 3623-3628.

37. E. Munnier, F. Tewes, eacute, eacute, ric, S. Cohen-Jonathan, C. Linassier, L.

Douziech-Eyrolles, H. Marchais, eacute, Souc, eacute, Martin, Herv, eacute, Katel, P.

Dubois and I. Chourpa, Chemical and Pharmaceutical Bulletin, 2007, 55, 1006-1010.

38. F. A. De Wolf, K. Nicolay and B. De Kruijff, Biochemistry, 1992, 31, 9252-9262.

39. G. Speelmans, R. W. H. M. Staffhorst, B. de Kruijff and F. A. de Wolf, Biochemistry,

1994, 33, 13761-13768.

40. K. K. Karukstis, E. H. Z. Thompson, J. A. Whiles and R. J. Rosenfeld, Biophysical

Chemistry, 1998, 73, 249-263.

Page 64: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

43

41. M. Pietrzak, Z. Wieczorek, A. Stachelska and Z. Darzynkiewicz, Biophysical

Chemistry, 2003, 104, 305-313.

42. A. Szulawska, M. Gniazdowski and M. Czyz, Biochemical Pharmacology, 2005, 69,

7-17.

43. K. Das, B. Jain, A. Dube and P. K. Gupta, Chemical Physics Letters, 2005, 401, 185-

188.

Page 65: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

Parts of this chapter has been published in Langmuir, 2014, 30, 1004-1011.

Parts of this chapter has been “Just Accepted” in IEEE Trans. Magn. DOI: 10.1109/TMAG.2016.2517602.

Chapter 3

PEG-Modified PAMAM-Fe3O4-drug Triads with the

Potential for Improved Therapeutic Efficacy

3.1 Introduction

Chemotherapy uses chemicals to damage the internal machinery (that is, DNA, RNA

and proteins) of cells in order to trigger cell cycle arrest or apoptosis, however, such agents

generally induce apoptosis in both cancer and normal cells1. Doxorubicin (DOX), for example,

is an anthracycline molecule, which is a widely used cationic anticancer drug that interferes

with the replication process by intercalating DNA, resulting in cell apoptosis. DOX is

selectively destructive to rapidly proliferating cells, but is also toxic to cardiac tissue, which

might result in life-threatening heart damage2, 3. These side effects of DOX could be reduced

by the use of a delivery system that can safely and specifically deliver it to the targeted tissue.

Catechins, which belong to one of the sub-classes of polyphenols, are the major alkaloids found

in extracts of green tea (Camellia sinensis). Catechins comprise of epigallocatechin-3-gallate

(EGCG), epigallocatechin (EGC), epicatechin-3-gallate (ECG), epicatechin (EC) and

theaflavins4, 5. EGCG is an anionic polyphenol and has been explored as a chemopreventive

and chemotherapeutic drug for cancer6-8. It was found to inhibit the production of the vascular

endothelial growth factor (VEGF) in some types of cancer cells9-11 and to activate VEGF

receptors in leukemia12 and colon13 cancer cells. Catechins are highly unstable in an aqueous

Page 66: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

45

solution and degrade through oxidative processes14. Their rapid degradation in blood and other

body fluids hampers their efficient delivery by traditional routes of administration, such as

orally, through enteric absorption, or by intravenous injection to the systemic circulation. The

absorption and bioavailability of catechins, across intestinal membranes, have been reported to

vary largely based on their molecular weight and lipophilicity15. Indeed, the aqueous instability

and uncontrollable absorption of EGCG limit their practical use as cancer therapeutics. The

challenges in the delivery of both DOX and EGCG, their chemical stability, their

bioavailability and bioactivity have necessitated a delivery system. This delivery system should

be able to deliver these drugs to the targeted tissue and selectively release them by responding

to local microenvironment conditions such as temperature and pH values.

In the last chapter, we evaluated CA–Fe3O4 nanoparticles for their potential as a

delivery agent for cationic DOX. The findings concluded that these nanoparticles have

appropriate properties and performances for use as drug delivery vehicles and can therefore,

be customised for better performances. The desired tailoring in the physico-chemical properties

and performances can be achieved by the surface modification of the Fe3O4 nanoparticles using

a variety of molecules. Without surface modification, bare Fe3O4 nanoparticles agglomerate

due to van der Waals forces and magnetic dipole-dipole interactions because of their high

surface energy and large surface area-to-volume ratio. When modified, the surface energy of

Fe3O4 is reduced and the nanoparticles form a stable, uniform dispersion in aqueous

suspensions16, which is a desirable performance for the administration of drugs. The two

primary strategies to engineer the surface of nanoparticles are (i) embedding the nanoparticles

in matrices17 or (ii) encapsulating them within macromolecules18, 19. Based on these

approaches, the development of surface-modified drug delivery systems has been dominated

by liposomal and polymeric systems20, 21. Both, however, have critical disadvantages.

Liposome capsules, although possessing a uniform size, generally have a poor physiological

stability in blood, which hampers efficient delivery22. The nanocarrier systems prepared from

polymers, on the other hand, have a tuneable stability but high water/pH degradability and

uncontrolled absorption after administration; these systems are also highly polydispersed and

have a large size distribution23. The drawbacks mentioned above, however, could be obviated

by the use of branched macromolecules (for example, dendrimer)24, 25. The dendrimers not only

reduce particle agglomeration, but also provide high number of functional groups. In fact, over

the past two decades, dendrimers have been intensively investigated as drug delivery systems

because of their unique structures and properties, monodispersity, water solubility,

multivalency and encapsulation ability to entrap hydrophobic drugs26. Among a number of

Page 67: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

46

dendrimers, polyamidoamine (PAMAM) dendrimers are the most studied as drug delivery

vehicles27-29. The PAMAM dendrimer-drug system is also often loaded with magnetic

nanoparticles (for example, Fe3O4) as an agent that performs additional functions, such as

magnetic resonance imaging or hyperthermia treatment30, 31. In addition, dendrimers can be

modified with poly (ethylene glycol) (PEG) to decrease the toxicity of the dendrimers and to

improve their systemic circulation time, resulting in enhanced retention effects32, 33.

There are two strategies in the loading of a drug onto dendrimers: encapsulation and

conjugation. In the process of encapsulation, drug molecules are internalised by a carrier.

Encapsulation involves either hydrophobic interactions between the relatively non-polar

cavities of dendrimers and a hydrophobic drug, or hydrogen bonding between dendrimeric

cavities and drug molecules. Conjugation is a process of loading drug molecules onto the

surface of a carrier. It can be achieved either by covalent or by electrostatic interactions

between the dendrimer and drug. While EGCG is still in its preliminary stages of evaluation as

an anticancer drug, DOX has been successfully delivered using dendritic nanosystems.

PAMAM-based dendritic-linear block copolymers (that is, dendrons) have been investigated

to encapsulate both DOX and Fe3O4 particles34, 35. These works showed that while the PAMAM

dendrimers showed improved encapsulation efficiency, their drug release performance was

unsatisfactory. Their findings demonstrated that the releasing efficiency of dendritic systems

was acceptably high at a temperature that was much lower (25 °C) than 37 °C, which would

make clinical administration difficult34. Also, while the system could also be pH-responsive,

the high releasing efficiency (> 50 %) at normal physiological conditions, that is, pH 7.434, 35,

predicted that much of the loaded DOX, if administrated systemically in vivo, would be

released immediately and uncontrollably into the bloodstream, resulting in a severe side effect.

The release of conjugated drugs from a dendrimer system has been investigated under

various external stimuli such as temperature and, especially, pH36-41 based on the fact that the

microenvironments around a tumour and inside cells are typically acidic42. In general, the

release efficiency of covalently conjugated drugs is lower than that of electrostatically bound

drugs. For example, when DOX molecules were covalently bound to PEG-modified PAMAM

by thiolated transferrin molecules by Michael addition, the release of DOX was only 30% at

pH 4.543. Recent work on electrostatic conjugation of DOX with various generations (for

example, G3, G5 and G6) of dendrimers demonstrated that the release efficiency of DOX was

40% in a PBS buffer at pH 5.218, 44, but was increased to 80 % when pH was reduced to 4.218.

Note that the acidic pH around 5 is found neither extracellularly (normally) nor within most of

the cell cytoplasm; this is, however, the pH of the lysosome, which is where many internalised

Page 68: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

47

molecules end up and are broken down by the acid hydrolases and other acid-active enzymes.

Further, the PEGylation of PAMAM dendrimers increases the drug loading by enhancing

hydration around the dendrimer periphery. It also induces the release of the drug in a controlled

fashion over a prolonged time. Cationic PAMAM interacts with negatively charged plasma

membranes, thereby promoting cell death by interfering with membrane fluidity. In contrast,

anionic PEG-PAMAM maintains the integrity of the plasma membrane, thus, reducing the

cytotoxicity 45, 46.

The primary objective of this chapter, therefore, was to establish optimal fabrication

procedures to develop multifunctional nanocarriers that are capable of carrying and delivering

both cationic and anionic drugs. To this end, different generations (G3, G5 and G6) of PEG-

PAMAM were used to modify the surface of Fe3O4, and DOX and EGCG were electrostatically

conjugated on the surface of the PAMAM-Fe3O4 nanocarriers. The PAMAM-Fe3O4-drug triads

were thoroughly characterised and explored in vitro as drug delivery systems for cancer

chemotherapeutics.

3.2 Experimental Techniques

3.2.1 Synthesis and surface modification of Fe3O4 nanoparticles

Iron oxide (Fe3O4) nanoparticles were synthesised using a soft chemical route. In this

process, 4.44 g of FeCl3 and 1.732 g of FeCl2 were dissolved in 80 ml of water in a round-

bottomed flask, and the temperature was slowly increased to 60 °C with refluxing under a

nitrogen atmosphere and mechanical stirring at 1000 rpm. After 30 min, 25 ml of ammonia

solution was added to the reaction mixture for precipitation of magnetite. An aqueous solution

of 5 ml of glutamic acid (0.5 g/ml) was then added to the reaction mixture mentioned above,

and the temperature was raised to 95 °C under reflux and maintained for a further 90 min with

continuous stirring. A black precipitate of glutamic acid-coated iron oxide nanoparticles (Glu–

Fe3O4) was obtained and then was thoroughly washed with ultrapure water. During each

washing step, the supernatant was decanted using a permanent magnet.

The Glu–Fe3O4 were then modified with PEG-PAMAM dendrimers to anchor the drug

molecules. In this process, 500 µl of Glu-Fe3O4 (at 1 mg/ml) was mixed with 200 µl of PEG-

PAMAM (0.1 mg/ml; generation 3, 5, or 6, marked as GX, where x=3, 5, or 6), and the volume

was made up to 1 ml by ultrapure water. The mixture was incubated at room temperature

overnight under continuous shaking. The dendrimer modified-Fe3O4 nanoparticles

Page 69: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

48

(abbreviated as Fe3O4–DGX, where x=3, 5, or 6) were then collected by a permanent magnet

and rinsed with ultrapure water 3–4 times.

3.2.2 Analysis of Drug Loading and Release

In order to evaluate the interactions of the drug molecules with the Fe3O4–DGX

nanoparticles, absorption and fluorescence spectroscopy was followed for EGCG and DOX,

respectively. The host-guest molecular complexation affects the characteristic electronic

absorption spectra of the reaction system (Benesi–Hildebrand method) and, therefore, was

employed to study the binding of EGCG to the Fe3O4–DGX nanoparticles. The fluorescence

intensity is highly dependent on the free or attached state of the molecule and, thus, has been

extensively used to study interactions between fluorophores and quencher47-50. The aqueous

suspension of varying amounts of Fe3O4–DGX nanoparticles (200, 400, 600, 800, 1000 and

1200 µg from a stock solution of 1 mg/ml) were added to 1 ml of each of the drug solutions

(10 µg/ml). The mixture was incubated at room temperature for 15 min. The absorbance and

fluorescence spectra of the supernatant after magnetic sedimentation were recorded for EGCG

and DOX, respectively. The intensities of the supernatant against the pure drug solution were

used to determine the loading efficiency of the Fe3O4–DGX nanoparticles. The loading

efficiency (w/w %) was calculated using the following relation;

% 𝐿𝑜𝑎𝑑𝑖𝑛𝑔 𝐸𝑓𝑓𝑖𝑐𝑖𝑒𝑛𝑐𝑦 = (𝐼𝐷𝑟𝑢𝑔− 𝐼𝑆)

𝐼𝐷𝑟𝑢𝑔 × 100 (Eqn. 3.1)

where IDrug is the intensity of the pure drug, and Is is the intensity of the supernatant.

For drug release experiments, the amounts of the drug and the Fe3O4–DGX

nanoparticles were quantified on the basis of loading efficiency. Along the lines of typical drug

loading experiment, 500 µg of Fe3O4–DGX (x = 3, 5 & 6) nanoparticles were incubated with

the 500 µg of the drug (1 mg/ml) in the dark for 3 h. The drug-loaded nanoparticles were

collected with a permanent magnet and were utilised for drug release experiments. For a typical

drug release study, drug-loaded Fe3O4–DGX nanoparticles were immersed in 5 ml of a sodium

acetate buffer (pH 5) and put into a dialysis bag, which was then suspended in 200 ml PBS (pH

7.3) to form the reservoir-sink settings. The dialysis was performed under continuous stirring

at 37 °C. At different time intervals, 1 ml aliquots from the sink (PBS) were withdrawn and

replaced with equal amounts of fresh PBS in order to maintain the interfacial concentration

gradient across the semi-permeable membrane. The amount of the drug released was

determined by the measurement of absorbance (λmax = 274 nm) and fluorescence (λex = 490 nm

Page 70: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

49

and λem = 560 nm) of the aliquots respectively, against standard plots that were prepared under

similar conditions (EGCG: R2= 0.996; DOX: R2= 0.999).

3.2.3 Evaluation of Carriers’ Biocompatibility and Therapeutic Efficacy

The biocompatibility of the Fe3O4–DGX (x = 3, 5 & 6) was assessed with the human

cervical cancer cell line (HeLa). In order to establish the potential of drug-loaded Fe3O4–DGX

in releasing the respective drugs, a dose-dependent study was undertaken to evaluate its 50%

inhibitory concentration (IC50) values over 24 h. The HeLa cells were grown in a supplemented

essential medium at 37 °C at 5% CO2 in a humidified atmosphere. Once they reached 90%

confluency, they were treated with a trypsin-EDTA solution in order to initiate detachment

from the substrate surface. The obtained cell suspension was centrifuged and the supernatant

was discarded. The cells were then re-suspended in a fresh supplemented medium and were

counted by the trypan blue exclusion method51. The cells were then re-seeded in 96-well plates

at a cell density of 2×104 cells per well and grown for a further 24 h. After 24 h, the spent

growth medium was discarded and replaced by a fresh supplemented medium that contained

different concentrations of the Fe3O4–DGX and drug-loaded Fe3O4–DGX (individual, serially

diluted test samples); this was followed by an additional 24 h of incubation. The cells were

then carefully washed with sterile PBS and the viable cell population was determined by the

sulforhodamine B (SRB) colorimetric assay52 as described in section 2.2.4. The IC50 value was

determined from dose-responsive sigmoidal curves that were generated from the spectroscopic

data obtained from Origin 8.0 software.

3.3 Results and Discussions

3.3.1 Characterisation of Fe3O4 Nanoparticles

The characterisation of the prepared nanomaterials was undertaken as described in

section 2.2.5. The XRD pattern revealed the formation of single-phase Fe3O4 nanoparticles

with an inverse spinel structure that had a lattice constant, a = ~8.38 Å, which was consistent

with the reported value of magnetite (JCPDS Card No. 88-0315, a = 8.375 Å). A high degree

of crystallinity in nanoparticles was indicated by the presence of sharp and intense peaks

(Figure 3.1a). The Scherrer formula was used to calculate the crystallite size of Glu–Fe3O4

(diffraction line broadening), which was ~8 nm53.

Page 71: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

50

Figure 3.1 (a) XRD pattern of Glu–Fe3O4 nanoparticles; TEM micrographs showing (b) Glu–

Fe3O4 nanoparticles (c) particle size distribution (d) selected area electron diffraction (SAED)

pattern, and (e) high-resolution TEM image of Glu–Fe3O4 nanoparticles

The microstructure examination in TEM showed that the particles were spherical with

regular morphology (Figure 3.1b), and their size ranged between 10 and 15 nm (Figure 3.1c).

The SAED rings in Figure 3.1d were indexed to be (220), (311), (400), (511) and (440)

diffraction planes of the inverse spinel Fe3O4, which was consistent with the XRD pattern.

From the high-resolution micrograph, the lattice spacing of the crystallite (Figure 3.1e) was

measured to be 2.5 Å, which corresponds to the (311) plane of Fe3O4.

The FTIR spectra of the synthesised nanoparticles showed successful grafting of

glutamic acid onto the surface of Fe3O4 nanoparticles. Figure 3.2a shows that the absorption

bands for the pure glutamic acid are well resolved when compared to the Glu–Fe3O4, which

are broadened and few. The strong IR band observed around 580 cm-1 in Glu–Fe3O4 could be

ascribed to the Fe–O stretching vibration mode of the magnetite. The spectral band at ~1650

cm-1 (C=O vibration of –COOH groups for glutamic acid) shifted to 1620 cm-1, indicating the

binding of glutamic acid to the surface of Fe3O4 nanoparticles by chemical absorption of

carboxylate (glutamate) ions. The carboxylate groups of the glutamic acid forms complexes

with Fe atoms that are present on the surface of Fe3O4, rendering a partial single bond character

to the C=O bond54. This is expected to weaken the C=O bond, which shifts the stretching

frequency to a lower value. The frequencies that appear at the 1540, 1420 and 1260 cm-1 in

Glu–Fe3O4 correspond to the symmetric stretching of COO–, C-O and OH groups, respectively,

confirming the successful grafting of glutamic acid on the surface of Fe3O4 nanoparticles. The

FTIR spectra of the pure PEG-PAMAM (Figure 3.2b) and PEG-PAMAM-modified Fe3O4

(Figure 3.2c) nanoparticles show well-resolved characteristic vibrations of PAMAM

Page 72: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

51

dendrimers, which broadened after conjugation with the Glu–Fe3O4 nanoparticles. Fe3O4–DGX

samples revealed a broad N–H stretching vibrations at 3400 cm-1, C–H stretching vibrations at

2880 cm-1 (2900 cm-1), COO- asymmetric stretching at 1625 cm-1, C=O stretching at 1550 cm-

1, N-O stretching at 1325 cm-1 (1350 cm-1), C–O–C stretching vibrations at 1250 cm-1, and

interfering C–O stretching with C–N stretches at 1100 cm-1. The values in parenthesis represent

the vibrational peaks of pure PEG-PAMAM. Due to the intense fingerprint region of PEG-

PAMAM dendrimers, the metal oxygen vibrations of iron oxide have been masked.

Figure 3.2 FTIR spectra of (a) glutamic acid (Glu) and glutamic acid-coated Fe3O4 (Glu–

Fe3O4) nanoparticles, (b) PEG-PAMAM of generations 3, 5, 6, and (c) Fe3O4–DGX

nanoparticles

Thermo-gravimetric analysis (TGA) and differential thermal analysis (DTA)

degradation profiles (Figure 3.3) show a weight loss of 9.6% with a sharp DTA endothermic

peak at ~80 °C, which could be attributed to the removal of physically absorbed water

molecules on the Fe3O4 nanoparticles. The weight loss of 5.0% with a broad DTA exothermic

peak at ~290 °C could be associated with the removal of chemically attached glutamic acid

molecules from the surface of Fe3O4 nanoparticles during the degradation of Glu–Fe3O4

nanoparticles. The weight loss of ~2.8% beyond the temperature of ~400 °C, indicated by a

sharp DTA exothermic peak at ~565 °C, is likely associated with the phase transformation of

magnetite to maghemite (Fe2O3)55.

Page 73: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

52

Figure 3.3 TGA-DTA of glutamic acid-coated Fe3O4 (Glu–Fe3O4) nanoparticles

In order to investigate whether the magnetic performance of Fe3O4 nanoparticles is

compromised by coatings, the field-dependent magnetisation (M vs. H) of Glu–Fe3O4 and

Fe3O4–DGX nanoparticles was measured at 300 K at a field of 20 kOe (Figure 3.4a). The Glu–

Fe3O4 nanoparticles exhibited superparamagnetic behaviour that was characterised by zero

coercivity and remenance, and had a maximum magnetisation of 57 emu/g at 20 kOe. The

magnetisations of the Fe3O4–DGX nanoparticles were calculated to be approximately 48, 37

and 33 emu/g for generations 3, 5 and 6, respectively. The presence of PEG-PAMAM on the

surface of these nanoparticles resulted in a decrease in the saturation magnetisation value due

to the increase in the non-magnetic components. This decrease is seen to be in proportion to

the generation of the PEG-PAMAM.

Figure 3.4 (a) Room-temperature field-dependent magnetisation of Glu–Fe3O4 and Fe3O4

nanoparticles coated with dendrimers of generations 3, 5, and 6 (Fe3O4–DGX) at 20 kOe (b)

Time-dependent calorimetric measurements of Glu–Fe3O4 in different ACMFs. The SAR was

calculated to be ∼134 W/g from the initial slope of the time-temperature curve.

Page 74: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

53

The ability of the superparamagnetic nanoparticles to generate localised heat in the

presence of ACMF makes them potentially useful in killing tumour cells. To investigate their

potential in the hyperthermic treatment of cancer, time-dependent calorimetric measurements

were carried out with Glu–Fe3O4 nanoparticle aqueous suspensions (typically ~45°C)56. SAR

was calculated from the initial slope of the temperature-time curve (Figure 3.4b), and was

found to be 134.3 W/g. The SAR value could be optimised by varying parameters such as

frequency, applied field and physical properties of the nanoparticles57, 58. The field strength had

a considerable effect on the heating capability. Figure 3.4b shows that at an applied field of

309Oe (400 A), the temperature of the aqueous suspension with high concentrations of iron

oxide nanoparticles (5 mg/ml) reaches 45 °C within 2.3 min. When the applied field was

reduced to 270 Oe (350A) and 232 Oe (300 A), the time taken to reach 45 °C increased to 3.3

and 4.5 min, respectively.

The colloidal stability and dispersion of the nanoparticles are associated with the

electric charge of the particle surface. The zeta potential of the different Glu–Fe3O4 and Fe3O4–

DGX (x = 3, 5 and 6) nanoparticle systems were measured and found to be −26.4 5.6, −19.0

2.0, −20.4 4.9 and −23.9 6.4, respectively. Apparently, the attachment of glutamic acid

on the surface of Fe3O4 nanoparticles resulted in a negatively charged surface (−26.4 mV).

Glutamic acid is a bi-carboxylic amino acid, that is, it has two –COOH groups per molecule.

Some of these carboxylate groups bind with the Fe cations in situ, while those that remain

unbound extend freely into the surrounding aqueous medium. These free carboxylate groups

not only impart an anionic surface to the nanoparticles but also play an important role in the

enhanced aqueous stability of the suspension of the nanoparticles. Zeta potential analysis also

showed that all the Fe3O4–DGX nanoparticles exhibited positive potential until pH 4; however,

their surface charge became negative with increasing pH. Around the physiologically relevant

pH of 7.4, the surface charges of the nanoparticles were measured and found to be −19.0, −20.4

or −23.9 mV for G3, G5 and G6, respectively. This change in the surface charge of the three

generations was due to the abundance of PEG-moiety on the dendrimers. The large negative

surface charge on the surface is believed to have enhanced the colloidal stability of the

nanoparticles.

3.3.2 DOX Loading and Release

Fluorescence spectroscopy was used to investigate the interactions of DOX with

Fe3O4–DGX nanoparticles (Figure 3.5). The interactions of DOX with the nanoparticles were

Page 75: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

54

evident by the dropping of fluorescence in the presence of the nanoparticles. The loading

interactions were evaluated at λex = 490 nm and λem = 590 nm of the DOX. The drug loading

efficiencies of the Fe3O4–DGX nanoparticle systems were calculated to be 86.9, 93.2 and 96.1

%, respectively, for generation 3, 5 and 6, using eqn. 3.1 (Figure 3.5d). It was found that the

average loading efficiency of DOX onto Fe3O4–DGX increased with the increase in the

generation of PAMAM, and was up to ~96 % for Fe3O4-DG6 carriers, which differs from

previous work18, 59. However, we also noticed that the increments were only statistically

significant between DG3 and DG6.

Figure 3.5 (a,b,c) Fluorescence intensity of DOX-loaded Fe3O4–DGX nanoparticles. (d) Drug

loading efficiency versus dendrimer generation. The difference between DG3 and DG6 was

significant (p < 0.05). There were no significant differences between DG3 and DG5, and DG5

and DG6 (p > 0.05)

It has been reported that dendrimers of higher generations are more capable of

encapsulating drugs than lower generation ones, while lower generations facilitate the

conjugation of drugs18, 59. In the previous works, the conjugation capacities of higher

generations of dendrimers were impaired by their more compact packing and steric hindrances

that were posed by the surface amine groups18. The amine groups present on the surface of the

PAMAM dendrimers play an important role in interacting with the drug molecules. It was

Page 76: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

55

observed that the electrostatic interactions between the −NH2 (δ+) groups of DOX with the –

OH (δ−) groups of the PEG-PAMAM are responsible for the high drug loading capacities. The

number of available PEG chains increase with an increase in the generation of the dendrimer.

Thus, the enhanced conjugation capacity of higher generations of dendrimers could be

attributed to the PEGylation on the surface of the PAMAM dendrimer The benefit from the

loosely packed long chains of PEG is two-fold. First they offer more space to host drug

molecules; second, they reduce steric hindrance when compared to the bare PAMAM

dendrimers.

Absorbance spectroscopy was utilised to evaluate the interactions of EGCG molecules

with Fe3O4–DGX nanoparticles and to calculate their loading efficiencies. Figure 3.6 shows the

drug loading efficiency obtained from the spectral changes in the EGCG with increasing

amounts of Fe3O4–DGX nanoparticles, calculated against absorbance of the free EGCG

solution. The drug molecules are expected to interact with the dendrimers electrostatically,

resulting in a drug-nanoparticle conjugate60. The loading interactions were evaluated at the

absorption maxima of EGCG, λmax = 274 nm. As the amount of Fe3O4–DGX was increased in

the solution, a slow decrease in the absorbance peak of the pure EGCG was observed, thereby

suggesting drug-nanoparticle interaction. A saturation point was reached when no further

decrease in absorbance was observed even after the addition of more Fe3O4–DGX

nanoparticles. The absorbance value of the supernatant of the particles was used to calculate

the drug loading capacities.

Figure 3.6 (a) Loading efficiencies of EGCG on to Fe3O4–DGX nanoparticles (b) Loading

efficiencies of three groups of Fe3O4–DGX (x = 3, 5 and 6) nanoparticles against dendrimer

generation. The difference between DG3 and DG6 was significant (p < 0.05).

Page 77: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

56

The loading of EGCG on the Fe3O4–DGX nanoparticles could be explained by the

formation of hydrogen bonds between the –OH groups of EGCG and PEG-PAMAM. At the

same time, a repulsive force exists between the –OH groups of both the molecules, which limits

the drug loading capacity to a certain extent. Statistical analysis revealed that the difference

between the loading capacities of Fe3O4–DG3 and Fe3O4–DG6 was significant (p<0.05), while

no significant difference was seen between the loading capacities of Fe3O4-DG3 and Fe3O4-

DG5, and Fe3O4-DG5 and Fe3O4-DG6 (p>0.05).

Three main types of interactions are expected between the dendrimer-nanoparticle

drug, namely, electrostatic interactions between the dendrimer and the charged functional

groups present on the drug; hydrophobic interactions between relative non-polar cavities of

dendrimers and hydrophobic end groups of the drug; and hydrogen-bond interactions between

dendritic cavities and drug molecules. Among these interaction mechanisms, the electrostatic

interactions contribute more to the solubility enhancement of the drugs than do the hydrophobic

and hydrogen-bond interactions. The loading of DOX on Fe3O4–DGX is higher than EGCG,

which could be explained on the basis of attractive interactions between the functional groups

of the drug molecules and PEG-PAMAM. Electrostatic interactions develop between the −NH2

(δ+) groups of DOX and the –OH (δ−) groups of the PEG-PAMAM, while the –OH groups of

EGCG interact through the formation of hydrogen bonds with PEG-PAMAM.

The drug release experiments were carried out in reservoir-sink conditions at 37 °C,

which mimics the cellular environment (reservoir: pH 5 and sink: pH 7.3). Figure 3.7a shows

that the release of DOX from Fe3O4–DGX was collectively faster at pH 5 than at pH 7.3. The

electrostatic conjugation is through the interaction of the cationic ends of DOX with the anionic

ends on the surface of PEG-PAMAM. The lowering in the pH value leads to protonation of the

PEG-modified dendrimers, which results in the cleavage of the bond of PEG with the DOX

molecules. It also shows that at pH 5, DOX was released rapidly for the initial 10 h before a

plateau was reached. The plateau percentages of DOX release observed over a period of 24h

were 608, 689 and 809 % by the Fe3O4−DG3, Fe3O4−DG5 and Fe3O4−DG6, respectively.

The release efficacy of DOX from Fe3O4–DGX increased with the generation of PAMAM,

which again contradicted previous work18, 59.

Page 78: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

57

Figure 3.7 Drug-release profile of (a) DOX-loaded Fe3O4–DGX nanoparticles at pH 5.0 and

7.4 and (b) EGCG-loaded Fe3O4–DGX nanoparticles at pH 7.4

In general, EGCG molecules were released slowly in the acidic environment, although,

in appreciable amounts (Figure 3.7b). It can be seen that up to 40–60% of EGCG molecules

were released over the first 24 h. The release of EGCG was slow and the release profile was

seen to follow a linear trend, which did not attain saturation/plateau even after 24 h. The release

mechanism of EGCG in an acidic environment involves the protonation of the dendrimers,

which results in weakening of the interactions between the EGCG molecules and the PEG-

PAMAM dendrimers. Hence, the slow releasing profile indicates that the cleavage of the

binding interactions is a slow process. The release of both of these drugs is stimulated by the

high [H+] concentration in the surrounding medium. The ease of protonation of DOX molecules

enhances their release in comparison to the EGCG molecules, which are not protonated but

released only when the dendrimeric chains are protonated. The PEGylation of PAMAM chains

also plays a role in the slow release of EGCG molecules as against the higher DOX release.

3.3.3 In vitro Evaluation of Cellular Toxicity and the Therapeutic Effect

The Fe3O4–DGX nanoparticles with or without the drugs were incubated with HeLa

cells in order to evaluate their compatibility or toxicity to the cell proliferation activity.

Quantitative evaluation showed that at physiological pH (7.2-7.4), the drug-free Fe3O4–DGX

nanoparticles had no significant effect on the proliferation of HeLa cells (Figure 3.8). Also, no

change in cellular morphology was observed, suggesting that the nanoparticles alone were

biocompatible. Although the lower generations were biocompatible, the higher generations of

PEG-PAMAM revealed moderate levels of cytotoxicity. The degradation of PAMAM in a

cellular environment could lead to the generation of toxic products, namely, methyl

Page 79: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

58

methacrylate and ethylene diamine, leading to elevated toxicity levels, depending on the

generation of the dendrimer.

Figure 3.8 Percentage relative cell viability versus the concentration of Fe3O4–DGX

nanoparticles. A decreasing trend of biocompatibility of these nanoparticles is seen with

increase in dendrimer generation and concentration.

The internalisation of the nanoparticles was examined using DOX-loaded Fe3O4–DG5

nanoparticles under confocal laser scanning microscopy. HeLa cells (2×103) were incubated

with DOX-loaded Fe3O4–DG5 for 5 h. Due to the inherent fluorescence of DOX, no tagging

moiety was required. The confocal images (Figure 3.9a) demonstrated that the control cells

showed no discernible changes in cytoplasm, nucleus, nuclear membrane and nucleoli. Figure

3.9b demonstrates that the particles were internalised by the cells. Though the incubation time

was not long enough for the drug to kill the cell (Figure 3.9b), the DOX-loaded nanoparticles

had caused significant changes in cellular morphology and related features, namely,

granulation of cytoplasm and degradation of the nuclear membrane.

Page 80: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

59

Figure 3.9 Confocal laser scanning images of (a) control HeLa cells and (b) treated HeLa

cells after 5 h of incubation (scale−20 μm) (c) Viability of HeLa cells in a medium that

contained either Fe3O4–DG5 or DOX–Fe3O4–DG5 nanoparticles in various amounts. The

difference between any pair of data at each concentration was insignificant (p > 0.05).

Under physiological conditions, PAMAM–Fe3O4–drug nanoparticles would cause a very mild

side effect to normal tissues due to low levels of drug release. Hence, the drug molecules can

be carried in a reasonably benign manner and not released until the triads are taken up into the

cell. The inhibition of cell proliferation activity is observed with both the drug molecules.

EGCG is seen to have less toxicity when compared to DOX molecules. At high doses of

PAMAM–Fe3O4–drug nanoparticles, EGCG reduced the cell population by 40%, while DOX

reduces the population by 80%, reiterating the effectiveness of DOX over EGCG. The IC50 was

calculated to be 11.8±5.3 µg/ml (R2 = 0.966) and 56.3±2.7 µg/ml (R2 = 0.982) of DOX-loaded

and EGCG-loaded Fe3O4-DGX nanoparticles, respectively.

3.4 Summary

The last chapter concluded that Fe3O4 nanoparticles were good drug delivery vectors,

but their properties and performances can be improved further by using macromolecules. In

this chapter, we have presented the dendrimer–Fe3O4 system as an improved platform for the

delivery of both cationic and anionic molecules. The chapter established the fabrication

procedures of the PAMAM-Fe3O4-drug nanosystem and the therapeutic efficiency of pH-

responsive triads by using different generations (G3, G5 and G6) of PEG-PAMAM. The

nanocarriers were synthesised by using the PEG-modified PAMAM dendrimers to encapsulate

Page 81: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

60

glutamic acid-modified Fe3O4 nanoparticles, and the drug molecules were electrostatically

conjugated to the surface of the nanocarriers. It was observed that the PAMAM of higher

generations possesses more favourable qualities in terms of both higher conjugation capacity

and greater efficiency in the release of the drugs. These nanocarriers are evidently capable of

carrying both cationic and anionic therapeutic molecules. At pH 7.4, the drug release was as

low as 15 % with sustained (DOX) and linear (EGCG) drug release profiles that were obtained

in acidic environment. The nanocarriers alone (that is, drug-free) are biocompatible and have

little or no adverse effect on cellular proliferation and morphology when incubated with HeLa

cells. The PAMAM-Fe3O4-drug triad showed controlled anticancer activity with a sigmoidal

(DOX) and approximately linear (EGCG) dose-response profile. Moreover, the

superparamagnetic behaviour and calorimetric measurements of nanocarriers suggest their

probable use in the hyperthermic treatment of cancer. These dendrimer-Fe3O4 nanosystems

showed excellent physico-chemical properties, and drug carrying and delivery performances,

but raised demands of improved biocompatibility, when higher generations of dendrimers were

in question. Towards this end, we aimed to synthesise a peptide dendrimer with an internal

chemical environment and physico-chemical properties similar to PAMAM. This peptide

dendrimer is expected to match PAMAM in its drug delivery performances with enhanced

biocompatibility.

3.5 References

1. T. Vial and J. Descotes, Toxicology, 2003, 185, 229-240.

2. E. Raschi, V. Vasina, M. G. Ursino, G. Boriani, A. Martoni and F. De Ponti,

Pharmacology & Therapeutics, 2010, 125, 196-218.

3. R. Hayward, D. Hydock, N. Gibson, S. Greufe, E. Bredahl and T. Parry, J Physiol

Biochem, 2013, 69, 177-187.

4. S. Bansal, N. Syan, P. Mathur and S. Choudhary, Med Chem Res, 2012, 21, 3347-3360.

5. K. Fraser, G. A. Lane, D. E. Otter, Y. Hemar, S.-Y. Quek, S. J. Harrison and S.

Rasmussen, Food Research International, 2013, 53, 827-835.

6. Y.-S. Chiou, S. Sang, K.-H. Cheng, C.-T. Ho, Y.-J. Wang and M.-H. Pan,

Carcinogenesis, 2013, 34, 1315-1322.

7. D.-S. Hsieh, H. Wang, S.-W. Tan, Y.-H. Huang, C.-Y. Tsai, M.-K. Yeh and C.-J. Wu,

Biomaterials, 2011, 32, 7633-7640.

Page 82: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

61

8. R. Shukla, N. Chanda, A. Zambre, A. Upendran, K. Katti, R. R. Kulkarni, S. K. Nune,

S. W. Casteel, C. J. Smith, J. Vimal, E. Boote, J. D. Robertson, P. Kan, H. Engelbrecht,

L. D. Watkinson, T. L. Carmack, J. R. Lever, C. S. Cutler, C. Caldwell, R. Kannan and

K. V. Katti, Proceedings of the National Academy of Sciences, 2012, 109, 12426-

12431.

9. Y. Shirakami, M. Shimizu, S. Adachi, H. Sakai, T. Nakagawa, Y. Yasuda, H. Tsurumi,

Y. Hara and H. Moriwaki, Cancer Science, 2009, 100, 1957-1962.

10. B. H. Zhu, W. H. Zhan, Z. R. Li, Z. Wang, Y. L. He, J. S. Peng, S. R. Cai, J. P. Ma and

C. H. Zhang, World journal of gastroenterology : WJG, 2007, 13, 1162-1169.

11. F. Spinella, L. Rosanò, V. Di Castro, S. Decandia, A. Albini, M. R. Nicotra, P. G. Natali

and A. Bagnato, Molecular Cancer Therapeutics, 2006, 5, 1483-1492.

12. Y. K. Lee, N. D. Bone, A. K. Strege, T. D. Shanafelt, D. F. Jelinek and N. E. Kay,

Blood, 2004, 104, 788-794.

13. M. Shimizu, Y. Shirakami, H. Sakai, Y. Yasuda, M. Kubota, S. Adachi, H. Tsurumi,

Y. Hara and H. Moriwaki, Chemico-Biological Interactions, 2010, 185, 247-252.

14. Z.-Y. Chen, Q. Y. Zhu, D. Tsang and Y. Huang, Journal of Agricultural and Food

Chemistry, 2001, 49, 477-482.

15. M. N. Clifford, J. J. van der Hooft and A. Crozier, The American Journal of Clinical

Nutrition, 2013, 98, 1619S-1630S.

16. B.-F. Pan, F. Gao and L.-M. Ao, Journal of Magnetism and Magnetic Materials, 2005,

293, 252-258.

17. X. L. Liu, E. S. G. Choo, A. S. Ahmed, L. Y. Zhao, Y. Yang, R. V. Ramanujan, J. M.

Xue, D. D. Fan, H. M. Fan and J. Ding, Journal of Materials Chemistry B, 2014, 2,

120-128.

18. K. Rouhollah, M. Pelin, Y. Serap, U. Gozde and G. Ufuk, Journal of Pharmaceutical

Sciences, 2013, 102, 1825-1835.

19. X. Lu, R. Jiang, M. Yang, Q. Fan, W. Hu, L. Zhang, Z. Yang, W. Deng, Q. Shen, Y.

Huang, X. Liu and W. Huang, Journal of Materials Chemistry B, 2014, 2, 376-386.

20. M. Shen and X. Shi, Nanoscale, 2010, 2, 1596-1610.

21. S. Chandra, K. C. Barick and D. Bahadur, Advanced Drug Delivery Reviews, 2011, 63,

1267-1281.

22. K.-E. Løkling, R. Skurtveit, S. L. Fossheim, G. Smistad, I. Henriksen and J. Klaveness,

Magnetic Resonance Imaging, 2003, 21, 531-540.

Page 83: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

62

23. T. Pakula, P. Minkin and K. Matyjaszewski, in Advances in Controlled/Living Radical

Polymerization, American Chemical Society, 2003, vol. 854, ch. 26, pp. 366-382.

24. S. Mignani, S. E. Kazzouli, M. Bousmina and J.-P. Majoral, Progress in Polymer

Science, 2013, 38, 993-1008.

25. J. Zhu and X. Shi, Journal of Materials Chemistry B, 2013, 1, 4199-4211.

26. J. Li, J. Li, S. Xu, D. Zhang and D. Liu, Colloids and Surfaces B: Biointerfaces, 2013,

110, 183-190.

27. M. Labieniec and C. Watala, cent.eur.j.biol., 2009, 4, 434-451.

28. R. Esfand and D. A. Tomalia, Drug Discovery Today, 2001, 6, 427-436.

29. E. MIKICIUK-OLASIK, P. SZYMAŃSKI and M. MARKOWICZ, Nano, 2011, 06,

509-539.

30. B. Pan, D. Cui, Y. Sheng, C. Ozkan, F. Gao, R. He, Q. Li, P. Xu and T. Huang, Cancer

Research, 2007, 67, 8156-8163.

31. M. Mahmoudi, A. Simchi, A. S. Milani and P. Stroeve, Journal of Colloid and Interface

Science, 2009, 336, 510-518.

32. P. Pradhan, J. Giri, F. Rieken, C. Koch, O. Mykhaylyk, M. Döblinger, R. Banerjee, D.

Bahadur and C. Plank, Journal of Controlled Release, 2010, 142, 108-121.

33. G. M. Sinéad M Ryan, Xuexuan Wang, David M Haddleton & David J Brayden, Expert

Opinion on Drug Delivery, 2008, 5, 371-383.

34. X. Wu, X. He, L. Zhong, S. Lin, D. Wang, X. Zhu and D. Yan, Journal of Materials

Chemistry, 2011, 21, 13611-13620.

35. M. A. Lopez-Heredia, M. Bongio, V. M. J. I. Cuijpers, N. W. M. van Dijk, J. J. J. P.

van den Beucken, J. G. C. Wolke and J. A. Jansen, Tissue engineering. Part C, Methods,

2012, 18, 369-373.

36. D. Castelli, C. Boffa, P. Giustetto, E. Terreno and S. Aime, J Biol Inorg Chem, 2013,

DOI: 10.1007/s00775-013-1042-0, 1-8.

37. F. P. Wang, H. P. Mu, J. Y. Zhang, W. X. Li, Q. Z. Wang and X. Z. Du, Journal of

Applied Polymer Science, 2013, 130, 1981-1989.

38. F. Ding, X. Shi, Z. Jiang, L. Liu, J. Cai, Z. Li, S. Chen and Y. Du, Journal of Materials

Chemistry B, 2013, 1, 1729-1737.

39. J. Peng, T. Qi, J. Liao, B. Chu, Q. Yang, W. Li, Y. Qu, F. Luo and Z. Qian,

Biomaterials, 2013, 34, 8726-8740.

40. M. K. Jaiswal, R. Banerjee, P. Pradhan and D. Bahadur, Colloids and Surfaces B:

Biointerfaces, 2010, 81, 185-194.

Page 84: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

63

41. M. Gary-Bobo, Y. Mir, C. Rouxel, D. Brevet, O. Hocine, M. Maynadier, A. Gallud, A.

Da Silva, O. Mongin, M. Blanchard-Desce, S. Richeter, B. Loock, P. Maillard, A.

Morère, M. Garcia, L. Raehm and J.-O. Durand, International Journal of

Pharmaceutics, 2012, 432, 99-104.

42. I. F. Tannock and D. Rotin, Cancer Res, 1989, 49, 4373-4384.

43. Y. Li, H. He, X. Jia, W.-L. Lu, J. Lou and Y. Wei, Biomaterials, 2012, 33, 3899-3908.

44. H. He, Y. Li, X.-R. Jia, J. Du, X. Ying, W.-L. Lu, J.-N. Lou and Y. Wei, Biomaterials,

2011, 32, 478-487.

45. M. A. Dobrovolskaia, A. K. Patri, J. Simak, J. B. Hall, J. Semberova, S. H. De Paoli

Lacerda and S. E. McNeil, Molecular Pharmaceutics, 2011, 9, 382-393.

46. S. Bhattacharjee, I. M. C. M. Rietjens, M. P. Singh, T. M. Atkins, T. K. Purkait, Z. Xu,

S. Regli, A. Shukaliak, R. J. Clark, B. S. Mitchell, G. M. Alink, A. T. M. Marcelis, M.

J. Fink, J. G. C. Veinot, S. M. Kauzlarich and H. Zuilhof, Nanoscale, 2013, 5, 4870-

4883.

47. S. Chandra, S. Dietrich, H. Lang and D. Bahadur, Journal of Materials Chemistry,

2011, 21, 5729-5737.

48. M. Pietrzak, Z. Wieczorek, A. Stachelska and Z. Darzynkiewicz, Biophysical

Chemistry, 2003, 104, 305-313.

49. A. Szulawska, M. Gniazdowski and M. Czyz, Biochemical Pharmacology, 2005, 69,

7-17.

50. K. Das, B. Jain, A. Dube and P. K. Gupta, Chemical Physics Letters, 2005, 401, 185-

188.

51. W. Strober, in Current Protocols in Immunology, John Wiley & Sons, Inc., 2001, DOI:

10.1002/0471142735.ima03bs21.

52. V. Vichai and K. Kirtikara, Nat. Protocols, 2006, 1, 1112-1116.

53. A. L. Patterson, Physical Review, 1939, 56, 978-982.

54. J.-J. Max and C. Chapados, The Journal of Physical Chemistry A, 2004, 108, 3324-

3337.

55. F. Fajaroh, H. Setyawan, A. Nur and I. W. Lenggoro, Advanced Powder Technology,

2013, 24, 507-511.

56. Z. S. Deng and J. Liu, J Healthc Eng, 2013, 4, 409-426.

57. R. E. Rosensweig, Journal of Magnetism and Magnetic Materials, 2002, 252, 370-374.

58. G. Glöckl, R. Hergt, M. Zeisberger, S. Dutz, S. Nagel and W. Weitschies, Journal of

Physics: Condensed Matter, 2006, 18, S2935.

Page 85: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

64

59. M. Markowicz, P. Szymański, M. Ciszewski, A. Kłys and E. Mikiciuk-Olasik, J Biol

Phys, 2012, 38, 637-656.

60. A. D'Emanuele and D. Attwood, Advanced Drug Delivery Reviews, 2005, 57, 2147-

2162.

Page 86: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

65

Chapter 4

Section A

4.1 Synthesis of Peptide Dendrimer and its in vitro Characteristics

4.1.1 Introduction

The physico-chemical properties of dendrimers are primarily governed by their

repetitive units of monomers (internal chemical environment) and the functional groups that

are present on their surfaces. To achieve a high degree of precision and structural order,

dendrimers are synthesised in a stepwise fashion. Generally, two different methods, namely,

divergent and convergent, are adopted for the synthesis of dendrimers. These two synthetic

methods have inherent advantages and disadvantages. Using the divergent synthesis method,

the dendritic molecule is formed from a central core, which then extends radially outwards

through addition of branching molecules. The main advantage of the divergent method is that

highly molecular nanoscaffold architecture is attained with desired repetitive branching

monomers. Thus, the dendrimer can be tailored to achieve maximum functionalities and

properties. However, two major challenges are encountered in divergent synthesis. First, the

number of reaction points increases in geometric progression with every generation, followed

by an increase in molecular weight. This compromises the reaction kinetics, making it slower;

the synthesis of high generation dendrimers then becomes difficult, further lowering the yield

of the desired product. The addition of each branching unit requires care and precision in order

to prevent structural defects and asymmetry in the structure of the dendrimer. Secondly, the

separation of the desired dendrimer from the by-products is hindered due to the molecular

Page 87: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

66

similarity exhibited by the by-product as well as the desired dendrimer. On the other hand, the

convergent method employs synthesis of small dendrites from the exterior, and the reaction

then proceeds inwards to the central core. The convergent procedure results in lesser structural

defects and easy purification of dendrimers, resulting in a high degree of monodispersity.

Despite the possibility of purer and flawless dendrimers, the convergent method falls short in

the synthesis of higher generation dendrimers. This choice is limited due to the steric forces

that crowd the dendrites around the central core molecule.

Despite the difficulties, these macromolecules have gained interest over classical

polymers due to the varied options presented by dendritic macromolecules. The vast pool of

molecules offers freedom of choice for the central core, the branching monomeric units and the

surface functional groups and gives rise to a multivalent system. This, in turn, has a wide

variety of possible chemical compositions, internal chemical environments, tailorable surface

groups, structural properties and architecture. Multi-functionalised dendrimers have also been

successfully synthesised, mainly, by modifying the terminal functional groups, which offers

adjustments in physical and chemical properties as required1, 2. Methyl acrylate alternating with

ethylene diamine forms the most widely synthesised, studied and used class of polyamidoamine

(PAMAM) dendrimers3; the internal amide groups provide an abundance of lone pairs of

electrons. Another popular class of amine-terminated dendrimers is the poly-(propylene imine)

(PPI) synthesised by Michael's addition of primary amines to acrylonitrile, followed by

subsequent hydrogenation by Raney cobalt or Raney nickel catalyst4. The interiors of PPI

dendrimers are the tertiary nitrogen atoms with lone pairs of electrons that contribute to their

reactive cavities. Both the classes of dendrimers have primary amine groups on the surfaces,

which govern the surface properties, reactivity and surface charge. These polyamidoamine

(PAMAM), polyethylene imine (PEI), polypropylene imine (PPI) dendrimers have found their

way into biomedical applications such as drug delivery 5-7, gene delivery8-10, antimicrobials11,

bioseparation12, biosensing13-15 and magnetic resonance imaging16-19. The world of biomedical

applications of dendrimers is currently dominated by the PAMAM dendrimers.

PAMAM surpasses other dendritic systems due to the ease of its preparation, desirable

chemical and physical properties, surface functional groups, and comparatively lower toxicity

to other dendrimers. Despite the lower cytotoxicity in comparison with other dendrimers and

dendritic systems, the degradation products of PAMAM show significant toxicity, which limits

their use in biological systems 20, 21. However, when various factors (such as biocompatibility,

haemocompatibility, immunogenicity, and biodistribution) are taken into consideration,

Page 88: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

67

cytotoxicity remains the most critical factor in the successful use of any dendritic system in

any living system. The cytotoxicity of these dendrimers is not only attributed to the charges of

the surface functional groups but also to the number of generations, and has been well reported

20-23. Thus, the primary aim of this chapter was to fabricate dendrimers with enhanced

biocompatibility without compromising their chemical composition, internal and external

environment, physical and chemical properties, and efficacy in biomedical applications.

4.1.2 Experimental and Characterisation Techniques

4.1.2.1 Synthesis of Peptide Dendrimer

The structure of the dendrimer was tailored by the use of ethylene diamine as the core

molecule along with L-lysine and L-arginine as its branching monomers. The synthesis of the

peptide dendrimer was undertaken as described elsewhere, with minor modifications24. For a

typical synthesis, 1 mmol of ethylene diamine and 4 mmol of ester-activated di-Boc-L-lysine

were dissolved in dimethyl formamide. The reaction was maintained at 0–5 °C for 24 h under

constant stirring and a nitrogen atmosphere. The synthesis scheme of peptide dendrimer is

elaborately depicted in Figure 4.1. Compound 1 precipitates at the end of the coupling reaction

and is washed with 15% sodium chloride, 5% aqueous citric acid, 5% sodium bicarbonate

solution and ultrapure water successively, and subsequently dried under high vacuum. It is then

purified by column chromatography by using 3% ethyl acetate in PET ether as the eluent, and

subjected to deprotection to remove the boc- groups in order to yield compound 2 (generation

1: 72.1% yield). For the synthesis of the second generation dendrimer, 8 mmol of di-Fmoc-L-

Arginine was dissolved in anhydrous dichloromethane and activated in the presence of 1-ethyl-

3-(3-dimethylaminopropyl) carbodiimide and N-hydroxysuccinimide for 45 minutes.

Simultaneously, 1 mmol of compound 2 was dissolved in anhydrous dichloromethane and

stirred under a nitrogen atmosphere in an ice bath. The activated arginine solution was added

to the solution under continuous stirring under a nitrogen atmosphere, and maintained between

0–5 °C for 24 hours. After completion of the reaction, a white compound 3 was precipitated,

dried under reduced pressure and washed thoroughly in a manner similar to compound 1.

Compound 3 was then purified by column chromatography with 7% ethyl acetate in PET ether

as the eluent, and subjected to deprotection to remove the Fmoc- groups, in order to yield

compound 4 (EDA-KR2 dendrimer) (generation 2: 61.7% yield).

Page 89: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

68

Figure 4.1 Schematic representation of the synthesis of the EDA-KR2 dendrimer

4.1.2.2 Evaluation of Biocompatibility of the Dendrimer

Cell Culture: The toxicity of functional nanomaterials in a living system is an important factor

that limits their use in biomedical applications. To meet this end, an as-prepared peptide

dendrimer was assessed for its biocompatibility with murine fibroblast (L929), human cervical

cancer (HeLa), human oral carcinoma (KB), human breast adenocarcinoma (MCF-7), and

human prostate cancer (PC-3) cell lines. The cell lines were cultured in appropriate growth

medium supplemented with 10% FBS and antibiotics at 37 °C in a 5% CO2 environment in a

humidified incubator. When confluent, the cells were treated with trypsin-EDTA solution for

detachment and counted by the trypan blue exclusion method25.

For a typical experiment, 2×104 cells/well were seeded in a 96-well plate and were

incubated in a suitable tissue culture medium for 24 h at 37 °C in a 5% CO2 environment. The

formulations were suspended in the appropriate growth medium and serially diluted (20.0,

10.0, 5.0, 2.0, 1.0, 0.5, 0.25, 0.125, 0.625 and 0.3125 mg/ml). The exhausted media in the wells

was replaced with 200 µl of this mixture and the cells were allowed to grow for an additional

24 h in the presence of the nanoparticles in the growth media under similar growth conditions.

Page 90: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

69

The cells were then washed with 1X PBS (pH 7.3) carefully and the viable cell population was

determined by the sulforhodamine-B colorimetric assay26 described in section 2.2.4.

4.1.2.3 Characterisation Techniques

The 1H and 13C NMR spectra were recorded by the Brüker Avance 500WB, 500 MHz

spectrometer. The chemical shifts, δ, are denoted in ppm (parts per million). The X-ray

Photoelectron Spectroscopy (XPS) data was recorded by Multilab 2000, Thermo VG

Scientific, USA, using 250 W micro-focused monochromatic Al Kα as an X-ray source

(hν=1486.6 eV). The binding energies obtained from the XPS analyses were standardised using

C1s core levels at 284.6 eV. Fourier-transform infrared (FTIR) spectroscopy measurements,

thermal degradation profiles, measurements of hydrodynamic diameter and the zeta potential

and the absorbance measurements of the SRB assay were recorded as described in section 2.2.5.

4.1.3 Results and Discussions

4.1.3.1 Characterisation of the peptide dendrimer

Compound 2 was a white solid (4.6 g, yield 72.1%) after purification and deprotection

of compound 1 (Figure 4.1). 1H NMR (δ, CDCl3): 1.277 – 1.809 (m, -CH2; Lys), 2.531 (m,

NCH2NHCO; amide), 2.853 – 3.279 (s, CH2N; EDA and m, CH2NH; Lys &NCH2CH2NHCO),

3.813 – 3.967 (m, COCH(R)NH), 5.11 (s, -NH2; Lys). 13C NMR (δ, CDCl3): 21.77 (4C5, -CH2),

29.204 (4C6, -CH2), 31.115 (4C4, -CH2), 40.840 (4C7, -CH2-CH2-NH2), 47.006 (2C1, -CH2

EDA), 52.230 (4C3, -CO-CH(NH2)CH2), 176.199 (4C1, CH2(NH2)CON).

Compound 4 was also obtained as a white solid (2.8 g, yield 61.7%) after deprotection

of compound 3 (Figure 4.1). 1H NMR (δ, d6-DMSO): 1.034 – 1.702 (m, -CH2; Lys & Arg),

1.938 (d, CH2NHC(R); Arg), 2.563 (t, CH2CH2NH; Arg), 3.363 (t, CH2CH(NH2)CO; Lys &

Arg), 3.689 (s, CH2N; EDA), 4.120 (t, CH2CH2N(R), Lys), 5.264 (d, COCH(NH)CH2, Lys

&Arg), 8.653 (s, NHC(NH)NH2, Arg). 13C NMR (δ, CDCl3): 22.868 (4C4, CH2CH2CH2, Lys),

24.342 (8C4, CH2CH2CH2, Arg), 25.310 (8C3, CH(NH2)CH2, Arg), 29.183 (4C5, CH2CH2CH2,

Lys), 34.264 (4C3, CH(NH2)CH2, Lys), 40.840 (8C5, CH2CH2NH, Arg), 47.006 (4C6,

CH2CH2N(R), Lys), 48.034 (2C, CH2CH2, EDA), 53.838 (8C2, COCH2(NH2)CH2, Arg),

60.446 (4C2, COCH2(NH2)CH2, Lys), 156.909 (8C6, NHC(NH)NH2, Arg), 175.864 (12C1,

NOCH2(NH2), Lys & Arg).

Page 91: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

70

XPS was used to study and confirm the formation of the amide bonds between the

monomers to form the EDA-KR2 (Figure 4.2 (a, b and c)). The C1s core level of EDA-KR2

shows three deconvoluted peaks at 284.6, 286.5, and 289.6 eV, respectively. The peak at 284.6

eV corresponds to the C-H and C-C skeleton of the peptide dendrimer, while the intermediate

one at 286.5 eV is associated with the C-N bond. The peak towards higher binding energy

could be assigned to N-C=O and –O-C=O bonds of EDA-KR2. The N1s core levels of EDA-

KR2 show a sharp peak that reveals two deconvoluted peaks at 400.1 and 401.7 eV, which

correspond to the C-N-H and N-C=O bonds, respectively. The O1s core level of EDA-KR2

shows a single broad peak that is further deconvoluted into three components that appear at

532.4, 533.7, and 535.2 eV and are attributed to the N-C=O, –O-C=O and –O-C=O bonds,

respectively. The XPS spectrum of the EDA-KR2 dendrimer shows asymmetric features, which

indicates the presence of multiple oxygen bonds. All of these chemical bonds show a slight

shift in their binding energies, which confirms the successful synthesis of the peptide

dendrimer.

Figure 4.2 High-resolution X-ray Photoelectron spectra showing C1s, N1s and O1s core

levels of the EDA-KR2 dendrimer

The FTIR spectrum of the EDA-KR2dendrimer is shown in Figure 4.3a. The spectral

bands of the monomeric units of lysine and arginine separately are appropriately resolved,

whereas, the bands seen in the peptide dendrimer are less resolved. The FTIR of generation

one (G1) dendrimer primarily shows vibrational bands due to the presence of lysine, which

masks the bands of EDA. The spectrum of EDA-KR2 depicts the vibrational bands of both

lysine and arginine, which occur at 3333 cm-1 (3151 cm-1) due to NH- stretching, 2969 cm-1

(2928 cm-1) due to asymmetric stretching of CH3 and 1710 cm-1 (1680 cm-1) due to out-of-

plane bending of -NH2, while the band that appears at 1581 cm-1 (1574 cm-1) represents the

stretching of the C=O present in the dendrimer. The bands observed at 1450 cm-1 (1464 cm-1)

arise due to asymmetric bending of CH3, and at 1410 and 1376 cm-1, due to symmetric, in-

Page 92: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

71

plane bending of CH3 groups, respectively. The characteristic peaks of the secondary amide

(1544 cm-1) and the primary amide (1645 cm-1) confirmed the successful binding of arginine

to the lysine end groups through amide/peptide bonds. The values in parentheses represent the

values reported in earlier published works27, 28.

Figure 4.3 (a) FTIR spectra (b) thermal degradation profile of the EDA-KR2 dendrimer

The thermal analysis profile of the EDA-KR2 dendrimer (Figure 4.3b) showed a weight

loss of ~0.6% with a corresponding endothermic peak at ~50 °C, which could be ascribed to

the removal of adsorbed moisture. The weight loss of ~7.6% with a sharp endothermic peak

seen at ~180 °C is associated with the disintegration and removal of arginine molecules from

the surface of the peptide dendrimer. A further weight loss of 50.2% by 250 °C, with a broader

endothermic peak is attributed to the complete degradation of the constituent molecules of the

dendrimer. Therefore, the thermal degradation profile of the EDA-KR2 molecule suggested

that the synthesised dendrimer is stable up to the temperatures of ~150 °C. In addition, the zeta

potential measurements showed that the EDA-KR2 dendrimer carries a positive surface charge

of +45 mV at a near-physiological pH (pH 7.3), which arises due to the resonating secondary

amine groups of arginine, free on the surface of the EDA-KR2 dendrimer and extending into

the surrounding aqueous medium.

4.1.3.2 In vitro assessment of the peptide dendrimer

The cells were exposed to very high concentrations of the EDA-KR2 dendrimer in their

growth medium. Concentrations of as high as 20 mg/ml of the dendrimer showed minimal

effect on L929, PC3 and MCF-7 cells, with an approximate of 10% of decrease in the viable

cell population of HeLa and KB cells. As the concentration of the dendrimer was reduced, it

was observed that there was no hindrance to the cell proliferation activity, irrespective of the

Page 93: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

72

cell line. The viable cell population and the cellular morphology also remained unchanged and,

therefore, it was safely concluded that these dendrimers can be safely used with a wide variety

of cancer cells in in vivo settings.

Figure 4.4 Cell viabilities incubated with peptide dendrimer. The dendrimer is seen to be

biocompatible for a variety of cell lines, even at concentrations of as high as 20 mg/ml.

4.1.4 Summary

In the current chapter, an attempt was made to synthesise a biocompatible peptide

dendrimer that had amide interiors and amine exteriors. NMR, FTIR, XPS, Zeta potential

measurements, and thermogravimetry were utilised to confirm the synthesis and to characterise

these peptide dendrimers. Since amino acids were used as branching units, the dendrimer was

found to be biocompatible even at higher concentrations. Amino acids were chosen as the

branching units because, they could be utilised by the cells for their metabolism after

degradation of the dendrimer, thereby reducing the toxicity. The amide bond established

between amino acids is a peptide bonds and thus, these dendrimers can also be categorised as

peptide dendrimers. The synthesised peptide dendrimers have an edge over the widely used

PAMAM dendrimers due to better biocompatibility.

Based on these results, we expect this peptide dendrimer to behave in a fashion similar

to that of PAMAM dendrimers. This necessitates a thorough comparison of their performances

as drug delivery vectors and other biomedical applications. To this end, the forthcoming section

demonstrates a comparative study of PAMAM (commercial) and EDA-KR2 (as-prepared) in

Page 94: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

73

their ability to conjugate with Fe3O4 nanoparticles and successfully deliver DOX. These

platforms were also compared for combinatorial thermo-chemo therapy in in vitro settings.

4.1.5 References

1. P. Goyal, K. Yoon and M. Weck, Chemistry – A European Journal, 2007, 13, 8801-

8810.

2. N. Fischer-Durand, M. Salmain, B. Rudolf, L. Jugé, V. Guérineau, O. Laprévote, A.

Vessières and G. Jaouen, Macromolecules, 2007, 40, 8568-8575.

3. R. Esfand and D. A. Tomalia, in Dendrimers and Other Dendritic Polymers, John

Wiley & Sons, Ltd, 2002, DOI: 10.1002/0470845821.ch25, pp. 587-604.

4. E. M. M. de Brabander-van den Berg and E. W. Meijer, Angewandte Chemie

International Edition in English, 1993, 32, 1308-1311.

5. K. Rouhollah, M. Pelin, Y. Serap, U. Gozde and G. Ufuk, Journal of Pharmaceutical

Sciences, 2013, 102, 1825-1835.

6. C. M. Paleos, D. Tsiourvas, Z. Sideratou and L. Tziveleka, Biomacromolecules, 2004,

5, 524-529.

7. J. Zhu and X. Shi, Journal of Materials Chemistry B, 2013, 1, 4199-4211.

8. Y. Zhang, C. Zhou, K. Kwak, X. Wang, B. Yung, L. J. Lee, Y. Wang, P. Wang and R.

Lee, Pharm Res, 2012, 29, 1627-1636.

9. L.-A. Tziveleka, A.-M. G. Psarra, D. Tsiourvas and C. M. Paleos, Journal of Controlled

Release, 2007, 117, 137-146.

10. A. Lakshminarayanan, V. K. Ravi, R. Tatineni, Y. B. R. D. Rajesh, V. Maingi, K. S.

Vasu, N. Madhusudhan, P. K. Maiti, A. K. Sood, S. Das and N. Jayaraman,

Bioconjugate Chemistry, 2013, 24, 1612-1623.

11. A. Felczak, N. Wronska, A. Janaszewska, B. Klajnert, M. Bryszewska, D. Appelhans,

B. Voit, S. Rozalska and K. Lisowska, New Journal of Chemistry, 2012, 36, 2215-2222.

12. F. Qie, G. Zhang, J. Hou, X. Sun, S.-z. Luo and T. Tan, Talanta, 2012, 93, 166-171.

13. S. Chandra, N. Barola and D. Bahadur, Chemical Communications, 2011, 47, 11258-

11260.

14. P. Dey, M. Adamovski, S. Friebe, A. Badalyan, R.-C. Mutihac, F. Paulus, S.

Leimkühler, U. Wollenberger and R. Haag, ACS Applied Materials & Interfaces, 2014,

6, 8937-8941.

Page 95: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

74

15. G. Castillo, K. Spinella, A. Poturnayová, M. Šnejdárková, L. Mosiello and T. Hianik,

Food Control, 2015, 52, 9-18.

16. V. J. Venditto, C. A. S. Regino and M. W. Brechbiel, Molecular Pharmaceutics, 2005,

2, 302-311.

17. S. Langereis, Q. G. de Lussanet, M. H. P. van Genderen, E. W. Meijer, R. G. H. Beets-

Tan, A. W. Griffioen, J. M. A. van Engelshoven and W. H. Backes, NMR in

Biomedicine, 2006, 19, 133-141.

18. C. D. Malone, E. S. Olson, R. F. Mattrey, T. Jiang, R. Y. Tsien and Q. T. Nguyen, PLoS

ONE, 2015, 10, e0137104.

19. Q. Chen, H. Wang, H. Liu, S. Wen, C. Peng, M. Shen, G. Zhang and X. Shi, Analytical

Chemistry, 2015, 87, 3949-3956.

20. N. Malik, R. Wiwattanapatapee, R. Klopsch, K. Lorenz, H. Frey, J. W. Weener, E. W.

Meijer, W. Paulus and R. Duncan, Journal of Controlled Release, 2000, 65, 133-148.

21. R. Duncan and L. Izzo, Advanced Drug Delivery Reviews, 2005, 57, 2215-2237.

22. R. B. Kolhatkar, K. M. Kitchens, P. W. Swaan and H. Ghandehari, Bioconjugate

Chemistry, 2007, 18, 2054-2060.

23. S. Parimi, T. J. Barnes, D. F. Callen and C. A. Prestidge, Biomacromolecules, 2009,

11, 382-389.

24. K. Luo, C. Li, G. Wang, Y. Nie, B. He, Y. Wu and Z. Gu, Journal of Controlled

Release, 2011, 155, 77-87.

25. W. Strober, in Current Protocols in Immunology, John Wiley & Sons, Inc., 2001, DOI:

10.1002/0471142735.ima03bs21.

26. V. Vichai and K. Kirtikara, Nat. Protocols, 2006, 1, 1112-1116.

27. M. Jackson, P. I. Haris and D. Chapman, Biochimica et Biophysica Acta (BBA) -

Protein Structure and Molecular Enzymology, 1989, 998, 75-79.

28. M. W. Forbes, M. F. Bush, N. C. Polfer, J. Oomens, R. C. Dunbar, E. R. Williams and

R. A. Jockusch, The Journal of Physical Chemistry A, 2007, 111, 11759-11770.

Page 96: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

75

Chapter 4

Section B

4.2 Dendritic Fe3O4 Nanoparticles for Combinatorial Therapy:

Peptide Dendrimers with Enhanced Efficiency as Alternative

Platforms for PAMAM Dendrimers

4.2.1 Introduction

In the earlier chapters, we demonstrated the need for surface engineering of Fe3O4

nanoparticles and their successful use in drug delivery applications. Our work in chapter three

of this thesis demonstrated that the encapsulation of these nanoparticles by PAMAM dendrimer

presents itself as a promising alternative platform in drug delivery. We evaluated the drug

delivery performance of these nanoparticles thoroughly, and it was observed that they were

capable of carrying both cationic and anionic therapeutic payloads without any loss in the

anticancer activity of the drug molecules. In the last section, we synthesised a peptide

dendrimer in order to contest its biomedical performance against PAMAM dendrimer. Also,

we intend to look into alternative therapeutic strategies with this peptide dendrimer. The

classical therapies that combat cancer in clinics are radiotherapy and chemotherapy.

Radiotherapy utilises high-energy radiations that induce cell death by sensitising the cancer

cells1. Chemotherapy, on the other hand, involves the use of a variety of chemical molecules

(singularly or in combination) to kill cancer cells2, 3. Also, surgical removal of the tumour mass

is undertaken but is limited to solid tumours. These therapies do not destroy cancer cells alone,

Page 97: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

76

but are detrimental to normal cells. This lack of differentiation between cancerous and normal

cells in classical therapies has forced researchers to look for alternative therapies that

selectively act on tumour cells only. This search has led to the development of multifunctional

nanomaterials that could be exploited for targeted cancer therapeutics that have improved

clinical significance.

The magnetic nanoparticles in colloidal suspensions generate heat when exposed to an

alternating magnetic field4. Both ferromagnetic as well as superparamagnetic nanoparticles are

capable of generating heat and could be used for magnetic hyperthermia. But, so far, only

superparamagnetic nanoparticles have been clinically used. Two major factors dictate this

choice of particles. First, on application of ACMF, due to the high hysteresis losses the

ferromagnetic particles would heat uncontrollably, which in clinical settings will prove lethal

to normal cells in addition to the cancer cells. Second, on removal of the external magnetic

field, the particles will be still magnetised (remnant magnetisation) and will agglomerate due

to interparticle magnetic interactions. This agglomeration is highly undesirable inside a

biological system. On the other hand, superparamagnetic particles heat only due to rotational

losses, that tend to saturate and, therefore, cannot generate heat beyond a certain temperature.

Also, due to zero hysteresis, these particles show no residual magnetisation after removal of

ACMF and do not agglomerate.

When the difficulties mentioned above are taken into account, multifunctional

superparamagnetic nanomaterials enter the picture of cancer therapeutics. These ‘smart’ or

‘multifunctional’ nanomaterials are widely used in various biomedical applications5-10. These

nanomaterials could be targeted to the cancer site by using an external magnet (MDT), which

makes them selective to some extent. Due to the external heating of these nanoparticles, the

rise in temperature can be successfully limited to the area in which these particles are

accumulated. Studies about the optimising of the working parameters of magnetic

hyperthermia hypothesise that high magnetisation and higher SAR values are the prerequisites

for these nanoparticles, towards their clinical use. However, after accumulation in target cells,

these superparamagnetic nanoparticles behave very differently, and the observed SAR and the

resultant heating then are lower than required. Thus, the working parameters need to be

redefined for successful magnetic hyperthermia11-13. Since there are no existing strict protocols

for the clinical use of hyperthermia, this limitation is dealt with by using a high concentration

of nanoparticles and amplified strengths of applied magnetic fields. In addition to being

successful drug carriers, superparamagnetic magnetic nanoparticles have also been widely used

as hyperthermic agents in cancer therapeutics14. Shah et al.15 developed core-shell

Page 98: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

77

hyperthermic nanoplatforms, which were successfully targeted to the mitochondria inside the

cell. The presence of pro-apoptotic amphipathic tail-anchoring peptide (ATAP)-targeting

ligand improved tumour targeting and cellular uptake in malignant brain cancer cells

(glioblastomamultiforme, U87vIII) and metastatic breast cancer cells (MDA-MB-231).

Hyperthermia was seen to sensitise the cells towards ATAP and to act synergistically with the

ATAP moieties, inducing apoptosis by mitochondrial membrane permeation.

The enhanced sensitivity towards chemotherapeutic drugs by magnetic hyperthermia

opened up avenues of combination therapy, which involved a variety of therapeutic molecules

in combination with the magnetic hyperthermia for superior anticancer treatment. The use of

magnetic hyperthermia by Barick et al.16 to sensitise human cervical cancer (HeLa) cells

towards doxorubicin effected results that were quite interesting. The peptide-mimicking cross-

linked macromolecules on the surface of nanocarriers were seen to behave like amphiphilic

cell-penetrating peptides; these macromolecules facilitated cellular internalisation, enhanced

accumulation of doxorubicin molecules, and in combination with ACMF, showed significantly

high cell toxicity. Kim et al.17 fabricated copolymer of N-isopropylacrylamide (NIPAAm) and

N-hydroxy-methylacrylamide (HMAAm) (poly(NIPAAm-co-HMAAm)) with magnetic

nanoparticles and used them for ‘switchable’ delivery of DOX. The population of viable human

melanoma cells (COLO 679) decreased by 30% just after the exposure to ACMF for 5 min.

This decrease was attributed to the induction of apoptosis by synergistic action of DOX and

hyperthermia. Apart from doxorubicin, hyperthermia was seen to sensitise and enhance the

anticancer capability of other therapeutic drugs as well. Rao et al.18 used thermally-responsive

polymeric nanoparticle-encapsulated curcumin against human prostate adenocarcinoma. The

application of ACMF and the subsequent hyperthermia was seen to significantly enhance the

anticancer capability of curcumin, decreasing its inhibitory concentration 7-fold. Recently,

researchers have also combined magnetic hyperthermia with the delivery of other therapeutic

molecules such as oligonucleotides19, 20; this technique is in its nascent stage.

A variety of multifunctional nanoplatforms have been developed and studied for

magnetic hyperthermia and combinatorial therapy but dendrimer functionalised magnetic

nanoparticles have not yet been explored much. The current chapter thus demonstrates an

improved performance in anticancer combinatorial therapy through the use of dendritic

nanoparticles as carrier and hyperthermic platforms. The work undertaken in this chapter

demonstrates the fabrication of the dendrimer-modified Fe3O4 nanoparticles through the use of

as-prepared peptide dendrimers, which were then examined for their efficacy in chemotherapy

Page 99: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

78

and combinatorial thermo-chemotherapy in comparison with the widely used PAMAM-coated

Fe3O4 nanoparticles.

4.2.2 Experimental Techniques

4.2.2.1 Synthesis of Dendrimer-coated Fe3O4 Nanoparticles

Fe3O4 nanoparticles were synthesised by the conventional co-precipitation method and

stabilised by glutamic acid, as described in chapter three of this thesis. The surface of the

glutamic acid-modified Fe3O4 nanoparticles (Glu–Fe3O4) was further modified with

commercially available PAMAM dendrimers and as-prepared peptide dendrimers to yield

PAMAM-modified Fe3O4 (PAMAM–IO) and EDA-KR2-modified Fe3O4 (KR2–IO)

nanoparticles, respectively. For dendrimer modification, 500 µg of Glu–Fe3O4 nanoparticles (2

mg/ml) were incubated with varying amounts of both the dendrimers, PAMAM and EDA-KR2

(w/w ratios of 1, 2, 4, 6, 8 and 10); the volume of the mixture was made up to 1 ml. This

reaction mixture was incubated under shaking/rocking overnight to yield PAMAM–IO and

KR2–IO nanoparticles. These dendritic nanoparticles were then collected over a permanent

magnet and washed with ultrapure water 3–4 times. The absorption spectra of the supernatants

(and washings) were analysed at 280 (PAMAM) and 230 nm (EDA-KR2) in order to calculate

the bound dendrimers on the surface of the nanoparticles against the standard plot that was

prepared under similar conditions (R2=0.999 and 0.997, respectively). Each experiment was

performed in triplicates.

4.2.2.2 Drug Loading and Assessment of Binding Interactions

The carrier efficiency of the PAMAM–IO and KR2–IO nanoparticles was evaluated

using DOX as the therapeutic molecule. The drug loading efficiency of both the dendritic

nanoparticles was investigated by recording the fluorescence spectrum (ex=490 nm and

em=560 nm) of DOX. Each experiment was performed in triplicates and the standard deviation

was calculated. For a typical drug loading experiment, varying amounts of dendritic

nanoparticles (PAMAM–IO & KR2–IO; Stock: 2 mg/ml) were added to 1 ml of an aqueous

DOX solution (10 g/ml), mixed gently and then incubated for 10 min; the fluorescence

spectrum of the supernatant was then recorded from 520 to 800 nm after magnetic

sedimentation. The spectrum of the pure DOX solution (10 g/ml) was recorded for

Page 100: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

79

comparative evaluation and was used as a standard initial intensity. The addition of dendritic

nanoparticles showed a steady decrease in the fluorescence intensity of the DOX solution. The

process was continued until no further decrease in the intensity was observed. The DOX-loaded

dendritic nanoparticles were washed with ultrapure water 3–4 times over a permanent magnet,

and the washings were collected and analysed for loss of any superficially bound drug. The

loading efficiencies were calculated using eqn. 4.1.

The binding interactions of DOX molecules with both the dendritic nanoparticles were

further studied and understood by using a modified Stern–Volmer plot. For calculation of the

binding constant, 10 g of DOX was dissolved in 1 ml of ultrapure water and the fluorescence

spectrum was recorded. After 15 min, 20 g of PAMAM–IO and KR2–IO from the stock (2

mg/ml) were added and incubated for 15 min. The fluorescence spectrum of the supernatant

was recorded by magnetic sedimentation. The fluorescence intensities of the DOX molecules

were plotted against the corresponding total concentration of the nanoparticles. The data were

plotted and analysed according to the following relation21:

log [ΔF 𝐹⁄ ] = log 𝐾 + 𝑛 log [𝑄] (Eqn. 4.1)

where ΔF is the difference between the initial and final fluorescence intensity of DOX

(fluorophore) in the absence and presence of nanoparticles (quencher), K is the binding

constant, n is the binding affinity and [Q] is the total concentration of nanoparticles.

4.2.2.3 Drug Release Studies

The DOX-loaded dendritic nanoparticles were quantified according to their loading

efficiencies in order to perform the drug release experiments. The release behaviour of DOX

was assessed under reservoir-sink conditions using low pH as a stimulus. For evaluation of

drug release profiles, each of the DOX-loaded dendritic nanoparticles was suspended in 5 ml

of a 0.1X sodium acetate buffer (pH 5.0) in a dialysis bag (separately) that acted as reservoir.

The dialysis bag was then suspended in 200 ml 0.1X PBS (pH 7.3) in order to represent the

reservoir-sink conditions. Aliquots of 1 ml from the sink (PBS) were withdrawn and replaced

with an equal amount of fresh PBS simultaneously at fixed time intervals in order to maintain

the concentration gradient across the semi-permeable membrane. The aliquots were then

subjected to the analyses, and the amount of DOX released (cumulative release) was

determined by the measurement of fluorescence against the standard plot. Each experiment was

performed in triplicates and the standard deviation was calculated. For evaluation of drug

Page 101: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

80

release under neutral pH conditions, the DOX-loaded dendritic nanoparticles were suspended

in PBS (pH 7.3), and an experiment similar to the one described above was carried out.

4.2.2.4 Evaluation of Biocompatibility and Therapeutic Efficacy

The biocompatibility of PAMAM, EDA-KR2, PAMAM–IO and KR2–IO were assessed

with murine fibroblasts (L929), human cervical cancer (HeLa), human oral carcinoma (KB),

human breast adenocarcinoma (MCF-7), and human prostate cancer (PC-3) cell lines. The

formulations were suspended in an appropriate growth medium and serially diluted (2.0, 1.0,

0.5, 0.25, 0.125, 0.625 and 0.3125 mg/ml). 200 µl of this mixture was used to replace the spent

media in the wells, and the cells were allowed to grow for an additional 24 h in the presence of

the nanoparticles in the growth media under similar growth conditions. To evaluate the

potential of DOX-loaded dendritic nanoparticles to release DOX in the cancer cells, a dose-

dependent study was undertaken over 24 h. The therapeutic efficacy was evaluated with

different cancer cell lines (HeLa, KB, MCF-7 and PC-3). The concentration of the DOX-loaded

dendritic nanoparticles that reduced the cell population by 50% was referred to as the inhibitory

concentration (IC50) values of the said formulation, and was calculated by a dose-responsive

sigmoidal curve fitting by using Origin 8.0 software. The live cell population was determined

by the SRB assay22 and represented as relative percent viability against untreated cells as the

control (Eqn. 3.3).

4.2.2.5 Evaluation of Calorimetric Potential of Dendritic Nanoparticles

In order to evaluate time-dependent heat generation by the nanoparticles, 1 ml of an

aqueous suspension of different concentrations (1, 2 and 5 mg/ml) of the systems of both the

dendritic nanoparticles was placed along the radial centre of a solenoid coil of a radio frequency

generator; the suspension was then exposed to an ACMF of 232, 271 and 309 Oe (operating at

a current of 300, 350 and 400 A, respectively). The set-up was appropriately insulated in order

to minimise the heat loss. The time-dependent increase in the temperature of the suspension

was recorded, and the SAR was calculated by the initial slope using eqn. 3.2. The

concentrations of both, the dendritic nanoparticles and the applied ACMF, were optimised in

order to heat the suspension to 45±0.5 °C and to maintain it at that temperature for subsequent

magnetic hyperthermia and combinatorial therapeutic studies.

Page 102: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

81

4.2.2.6 In vitro Magnetic Hyperthermia (MHT) Studies

The effect of both the dendritic nanoparticles, with and without the exposure to ACMF,

was assessed on the live cell population using human cervical cancer (HeLa) cells as a model

system. To a cell suspension that contained 1×106 cells, 500µl (2 mg/ml) of each of the sterile

dendritic nanoparticles was added. The volume was made up to 1 ml by a supplemented growth

medium and mixed very gently to lessen the cell agglomeration. The cell suspensions were

then exposed to an ACMF of 271 Oe (350 A) initially for 8 min. (PAMAM–IO) and 12 min.

(KR2–IO) in order to reach 45 °C. The temperature was maintained at 45±0.5 °C for different

time intervals (5, 10, 15, 20, 25 and 30 min) by reducing the applied field strength to 193 Oe

(250 A). After this, the time for which the cell suspension was exposed to the ACMF (after

reaching 45 °C) is referred to as the treatment time. The treated cell suspension was then

incubated for 6 h at 37 °C in a 5% CO2 environment and then counted for viable cell population:

% 𝑉𝑖𝑎𝑏𝑙𝑒 𝐶𝑒𝑙𝑙𝑠 =𝑁𝑜. 𝑜𝑓 𝑙𝑖𝑣𝑒 𝐶𝑒𝑙𝑙𝑠

𝑇𝑜𝑡𝑎𝑙 𝑐𝑒𝑙𝑙𝑠 × 100 (Eqn. 4.2)

4.2.2.7 ACMF-triggered Combinatorial Therapy

The combined therapeutic effect of DOX-loaded dendritic nanoparticles and magnetic

hyperthermia was evaluated using HeLa cells. The experimentation performed was similar to

the one followed for magnetic hyperthermia studies. An aqueous suspension of DOX-loaded

dendritic nanoparticles (at IC50 concentration) was added to a cell suspension of 1×106 cells

and then exposed to ACMF for a treatment time of 10 min. (LD50). The cell suspension was

then incubated for 6 h and the viable cell population was calculated using eqn. 4.2. The results

were represented as the mean ± standard deviation of the mean (SD) from three independent

experiments that were performed at different times. The analyses were done using the statistics’

module of Origin 9.1 software in order to evaluate significant differences between a pair of

results. The levels of significance that were evaluated were p< 0.05 (*), p< 0.01 (**) and p<

0.001 (***).

4.2.3 Results and Discussions

4.2.3.1 Characterisation of Dendrimer-coated Fe3O4 Nanoparticles

The characterisation of the prepared nanomaterials was undertaken as described in

section 2.2.5. The XRD patterns of PAMAM–IO and KR2–IO nanoparticles were analysed to

Page 103: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

82

identify the crystal structure (Figure 4.5a). The diffraction peaks correspond to the inverse

spinel cubic crystal structure of Fe3O4. It was also observed that the characteristic XRD pattern

of Fe3O4 was not affected by the conjugation of dendrimers, maintaining the crystallinity of

Fe3O4 nanoparticles. Figure 4.5b shows the FTIR spectra of the glutamic acid, Glu–Fe3O4,

PAMAM, PAMAM–IO, EDA-KR2 and the KR2–IO nanoparticles. The FTIR of PAMAM

dendrimers show characteristic vibrations, as reported in earlier works23. The FTIR spectrum

of EDA-KR2 dendrimers is in accordance with the spectrum described earlier in chapter five.

The Glu–Fe3O4 nanoparticles showed few broad bands upon being coated with the dendrimers.

Thus, the results confirm the successful conjugation of the dendrimers onto the surface of Glu–

Fe3O4 nanoparticles. UV-Vis absorption spectroscopy was used to quantify the amount of

dendrimers that were utilised in the surface modification of Glu–Fe3O4 nanoparticles. The

percent conjugation of PAMAM and EDA-KR2 dendrimers with the surface of Glu–Fe3O4 was

found to be 34.0±5.3% and 32.5±5.6%, respectively, as calculated against the standard curves.

Figure 4.5 (a) XRD pattern; (b) FTIR spectra; (c) thermal degradation profiles and (d, e, f)

electron micrographs of Glu–Fe3O4, PAMAM–IO, and KR2–IO nanoparticles, respectively (σ

≤ 15%)

The thermal analyses profiles of both the dendritic magnetic nanoparticles show weight

loss, which is attributed to the loss of adsorbed moisture, the dendrimers (PAMAM and EDA-

KR2 each), and the phase transition of magnetite to maghemite (Figure 4.5c). PAMAM–IO

nanoparticles show an initial weight loss of 1.3%, which corresponds to the water molecules

(d) (e) (f)

Page 104: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

83

that are physically adsorbed onto the nanoparticles; a further loss of 7.7% in weight is observed

up to a temperature of 400 °C and is attributed to the degradation of PAMAM molecules that

are conjugated with the surface of Glu–Fe3O4 nanoparticles. The degradation profile of KR2–

IO nanoparticles reveals a higher weight loss of approx. 13.7%, when compared to the weight

loss of PAMAM–IO. This is due to the bulky architecture and molecular weight of the EDA-

KR2 dendrimer as opposed to the PAMAM dendrimer. The transmission electron micrographs

of Glu–Fe3O4, PAMAM–IO, and KR2–IO nanoparticles show spherical morphology with slight

agglomeration. The particle sizes of Glu–Fe3O4, PAMAM–IO and KR2–IO nanoparticles range

from 8–12 nm, 10–15 nm and 10–18 nm, respectively (Figure 4.5d, e and f).

XPS was used to analyse the binding energies of Glu–Fe3O4, PAMAM–IO and KR2–

IO nanoparticles (Figure 4.6). The spectrum of Glu–Fe3O4 nanoparticles reveals that the C1s

levels deconvoluted into three peaks that correspond to the C–H and C–C, C–N and N–C=O, -

O–C=O, respectively. The XPS spectra of PAMAM–IO and KR2–IO nanoparticles show that

the C1s core levels deconvoluted in two (284.6 and 287 eV) and three (283.8, 284.7 and 286.8

eV) peaks, respectively. The peaks that arise at lower energies correspond to the C–H and C–

C bonds of the dendrimers, while the intermediate peak in KR2–IO (284.7 eV) is associated

with the C–N bond (not visible in the PAMAM–IO). The higher binding energy peaks arise

due to the N–C=O and -O–C=O bonds present in the interiors of the dendrimers. The Fe2p

levels of Glu–Fe3O4 reveal the Fe3+ and Fe2+ oxidation states of magnetite, which is consistent

even after the conjugation of the dendrimers. This confirms that surface engineering by the

dendrimers does not alter the crystal structure of Glu–Fe3O4. The O1s core levels of both these

dendritic nanoparticles show asymmetric features, indicating the presence of multiple oxygen

bonds. The O1s levels show a single sharp peak with a shoulder, which is further deconvoluted

into two components that mark the presence of iron oxide-bound oxygen and the oxygen in the

dendrimer skeleton, respectively. The N1s levels of both the dendritic magnetic nanoparticles

show sharp peaks, which further reveal two deconvoluted peaks that represent the C–N–H and

N–C=O bonds, respectively. All of the binding energies show a slight shift, confirming the

successful conjugation of dendrimers to Glu–Fe3O4.

Page 105: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

84

Figure 4.6 High resolution XPS spectra for C1s, Fe2p, O1s and N1s of the Glu–Fe3O4,

PAMAM–IO and KR2–IO nanoparticles

4.2.3.2 Drug Loading and Assessment of Binding Interactions

Figure 4.7a and b shows the spectrum of the pure DOX solution against the spectra

obtained by incubating with different amounts of PAMAM–IO and KR2–IO, respectively. The

conjugation of DOX to these nanoparticles leads to the reduction in the number of free electrons

Glu-Fe3O4 PAMAM-IO KR2-IO

279 282 285 288 291 294

C1s

In

ten

sit

y (

a.u

.)

Binding Energy (eV)

(284.6 eV)

C-H, C-C

C-N

(285.9 eV)

N-C=O, -O-C=O

(288.3 eV)

278 280 282 284 286 288 290 292 294

C1s

Inte

ns

ity

(a

.u.)

Binding Energy (eV)

(284.6 eV)

C-C, C-H

(287 eV)

N-C=O, -O-C=O

282 283 284 285 286 287 288

C1s

Inte

nsit

y (

a.u

.)

Binding Energy (eV)

(283.8 eV)

C-C, C-H

(284.7 eV)

(286.8 eV)

N-C=O, -O-C=O

C-N

700 707 714 721 728 735 742

Fe2p

Inte

ns

ity

(a

.u.)

Binding Energy (eV)

(711.4 eV)

Fe in Fe3O4

700 705 710 715 720 725 730 735 740

Fe2p

Inte

ns

ity

(a

.u.)

Binding Energy (eV)

710.2 eV

Fe in Fe3O4

700 710 720 730 740

Fe2p

Inte

ns

ity

(a

.u.)

Binding Energy (eV)

527 528 529 530 531 532 533 534 535

O1s

Inte

ns

ity

(a.u

.)

Binding Energy (eV)

Fe3O

4 - Oxide

Hydroxides

(530 eV)

(531.6 eV)

524 526 528 530 532 534 536 538

O1s

Inte

nsit

y (

a.u

.)

Binding Energy (eV)

(529 eV)

Fe3O4 - Oxide

N-C=O

(530.5 eV)

524 526 528 530 532 534 536 538

O1s

Inte

nsit

y (

a.u

.)

Binding Energy (eV)

(530 eV)

N-C=O

(528.7 eV)

Fe3O4

396 397 398 399 400 401 402 403 404

N1s

Inte

nsit

y (

a.u

.)

Binding Energy (eV)

(399.9 eV)

C-N-H

(401.2 eV)

N-C=O

392 394 396 398 400 402 404

N1s

Inte

nsit

y (

a.u

.)

Binding Energy (eV)

C-N-H(398.8 eV)

N-C=O

(400.4 eV)

392 394 396 398 400 402 404

N1s

Inte

ns

ity

(a

.u.)

Binding Energy (eV)

(398.4 eV)

C-N-H

(399.3 eV)

N-C=O

Page 106: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

85

available for transition, which in turn is responsible for the lowering of the intensity of its

fluorescence. No further decrease even after the addition of nanoparticles indicated the

saturation of the drug loading capacities of the nanoparticles. The drug loading capacity of

PAMAM–IO was comparable to that of KR2–IO and found to be 90.1±2.5 % and 88.7±2.7%,

respectively. The slight difference in the loading efficiency of KR2–IO may be due to the steric

hindrance offered by resonating secondary amine groups of the arginine present on the surface

of EDA-KR2. The results show that the peptide dendrimer (EDA-KR2) is as effective as the

PAMAM dendrimers in conjugating with DOX.

Figure 4.7 Drug loading profiles of (a) PAMAM–IO and (b) KR2–IO nanoparticles;

Gaussian profiles of (c) DOX-PAMAM–IO and (d) DOX-KR2–IO; and Stern–Volmer plots

of DOX with (e) PAMAM–IO (R2=0.973) and (f) KR2–IO (R2=0.995)

Page 107: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

86

In order to confirm the conjugation of DOX with the dendrimers, the fluorescence

spectra were further deconvoluted using the Gaussian curve fitting (Figure 4.7c and d). The

emission spectrum of DOX exhibits three maxima peaks at 560 nm (P1), 590 nm (P2) and 630

nm (P3). The nature of the interactions of DOX with the dendritic nanoparticles is signified by

a change in the peak maxima position, the shape of the spectrum, and the ratios of the area

under each deconvoluted peak. From the spectrum of pure DOX, a gradual decrease is observed

in the values of A1/A2 and A3/A2 for the DOX–nanoparticles, which indicates their

conjugation (Table 4.1).

Table 4.1 The area under the peaks of DOX-loaded PAMAM–IO and KR2–IO nanoparticles

(σ≤10%)

PAMAM–IO KR2–IO

A1/A2 A3/A2 A1/A2 A3/A2

DOX 0.658 0.869 0.728 1.004

DOX-Dend. nanoparticles_25% 0.614 0.691 0.614 0.670

DOX-Dend. nanoparticles_50% 0.532 0.649 0.538 0.646

DOX-Dend. nanoparticles_75% 0.527 0.633 0.529 0.629

DOX-Dend. nanoparticles_90% 0.517 0.606 0.515 0.615

In order to understand the binding interactions between DOX and the dendritic

nanoparticles better, the Stern–Volmer plot was used to calculate the binding constant of the

drug loading process (Figure 4.7e and f). The y-intercept of the plot represents the logarithmic

value of the binding constant, which was 1.018 and 1.0009 (μg/ml)-1 for PAMAM–IO

(R2=0.973) and KR2–IO (R2=0.995), respectively. The slope of the plot is a representation of

the fraction of DOX that takes part in the binding or the binding affinity, which was 0.0442

and 0.0512 for PAMAM–IO and KR2–IO, respectively. The binding affinity of KR2–IO

towards DOX is seen to be more than that of the PAMAM–IO, confirming that KR2–IO

nanoparticles have better drug-carrying properties. The appropriate linear fitting of the

modified Stern–Volmer equation leads to the conclusion that the quenching observed is a

bimolecular process that involves DOX and dendritic nanoparticles.

Page 108: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

87

4.2.3.3 Drug Release Studies

Figure 4.8 shows the DOX release profiles from the DOX-loaded dendritic

nanoparticles under low pH conditions over a period of 48 h. This reservoir-sink system is

expected to behave and interact in a fashion similar to the lysosome-cytoplasm interface in a

living cell, because it mimics their [H+] environments that are separated by a semi-permeable

membrane. Thus, a low pH acts as an external trigger for on-demand delivery of the DOX

molecules. Large amounts of DOX are released (> 50%) within the initial 6–7 h, followed by

sustained release that attains a plateau after ~12 h. The increase of [H+] in the

microenvironments results in the protonation of DOX, which in turn initiates the weakening of

the non-covalent binding between the DOX and the dendritic nanoparticles. This weakening

results in the release of DOX molecules into the immediate environment and a subsequent slow

diffusion into the sink through the dialysis membrane.

Figure 4.8 Drug release profile of (a) PAMAM–IO and (b) KR2–IO nanoparticles under the

stimulus of pH 5.0 and 7.3

The release efficiency of DOX from KR2–IO is found to be 81.7±1.6%, which is substantially

more than the 64.4±3.2 % of PAMAM–IO, recorded over an experimental period of 48 h. This

indicates that, for similar amounts of DOX carried by these dendritic nanoparticles, the release

efficiency of the KR2–IO is significantly higher than PAMAM–IO nanoparticles. This elevated

drug release observed from KR2–IO could also be due to the acidic hydrolysis of peptide bonds

in the EDA-KR2 dendrimer and the disintegration of the whole dendrimeric architecture, which

may not occur in PAMAM–IO. On the other hand, at near physiological pH, the DOX released

from both the carriers is less than 10%, which confirms the pH responsive behaviour of these

Page 109: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

88

dendritic nanoparticles. When the DOX-loaded dendritic nanoparticles are delivered to the

cancer site, which has lower pH than normal body fluids, the release of DOX is expected to

initiate. This DOX release would be facilitated further upon the internalisation of the

nanoparticles in the lysosomes of the cancer cells.

4.2.3.4 Evaluation of Biocompatibility and Therapeutic Efficacy

The Glu–Fe3O4, EDA-KR2, PAMAM–IO, and KR2–IO nanoparticles are seen to be

non-toxic. No hindrance in cell proliferation activity and viable cell population was observed.

The cellular morphology was also unchanged and, therefore, it was concluded that these

nanoparticles can be safely used with a wide variety of cancer cells in in vivo applications.

Therapeutic efficacy studies reveal that the DOX-loaded nanoparticles are competent drug

carriers and can efficiently reduce the population of cancer cells. The IC50 values of DOX-

PAMAM–IO and DOX-KR2–IO were calculated by a dose-responsive sigmoidal curve fitting

from the Origin 8.0 software, and are listed in Table 4.2.

Table 4.2 IC50 values of (a) DOX-PAMAM–IO and (b) DOX-KR2–IO nanoparticles with

various cancer cells. The values are represented in µg/ml of formulations with ≤ 15% (IC50

of pure DOX was 1.1-1.5μM with different cell lines)

Cell Line/s DOX-PAMAM–IO DOX-KR2–IO

HeLa 90.0 125.7

MCF-7 59.9 67.0

PC-3 74.4 89.6

KB 63.0 77.3

4.2.3.5 Evaluation of Calorimetric Potential of Dendritic Nanoparticles

Superparamagnetic nanoparticles are known to generate heat when exposed to an

ACMF due to Brownian and Néel relaxations, which largely depends upon the anisotropy and

size of the nanoparticles4.The zero remanence in superparamagnetic nanoparticles ensures

minimal aggregation of the nanoparticles in the absence of the magnetic field, when present

within a biological system. This heat-generating property was investigated as a time-dependent

study for PAMAM–IO and KR2–IO. The SAR was calculated to be 110.36 W/g (PAMAM–

IO) and 106.12 W/g (KR2–IO), as against the ~134 W/g of Glu–Fe3O4. The observed decrease

Page 110: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

89

in the heating capability of the dendritic nanoparticles is attributed to the non-magnetic

dendrimer entities on their surfaces.

However, the SAR is dependent on various physical parameters such as the strength of

the ACMF (H), the frequency of the applied ACMF (ω), and the physical characteristics of the

magnetic nanoparticles, such as magnetisation, particle size and size distribution4, 24-26.

Nevertheless, practically, the heating proficiency of nanoparticles cannot be raised solely by

increasing the parameters mentioned above. Therefore, the SAR value needs to be optimised

by varying these parameters (frequency, field and physical properties of the nanoparticles)

(Figure 4.9A and B). The concentration of 5 mg/ml of dendritic nanoparticles in an aqueous

suspension was seen to be very efficient in raising the surrounding temperature to the desired

levels. However, this concentration is potentially toxic to normal cells. Hence, the subsequent

evaluation of the heating performance was focused on the aqueous suspensions of dendritic

nanoparticles at concentrations of 1 or 2 mg/ml. Under the conditions of applied fields, the

suspension of 2 mg/ml effectively reached 45°C in 7.5 min (309.33 Oe) and 10.5 min (271 Oe),

but failed to achieve this temperature at 232 Oe even after 25 min. The concentration of 1

mg/ml was found to be insufficient under any applied field strength, even after 30 min of

exposure. In summary, the 2 mg/ml concentration of the dendritic nanoparticle suspension and

the ACMF of 271 Oe were considered to be optimal working parameters that would achieve

the maximal hyperthermia treatment effect without being detrimental to the neighbouring

normal cells and were optimised for subsequent magnetic hyperthermia studies.

Figure 4.9 Time-dependent calorimetric measurements of (A) PAMAM–IO and (B) KR2–IO

nanoparticles at different ACMF and concentrations

Page 111: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

90

The time required to attain 45 °C (hyperthermia temperature) was observed to be

broadly dependent on the strength of the applied field (H) and the concentration of

nanoparticles in the suspension. Also, at a given concentration, the time required to reach 45

°C shows an inverse proportionality in relation to applied field and increases with a decrease

in the field. This behaviour is also in agreement with the fact that the quantity of heat produced

is directly proportional to the square of the applied ACMF (H) (Figure 4.10).

Figure 4.10 Dependence of SAR on the strength of applied ACMF for (a) PAMAM–IO and

(b) KR2–IO

4.2.3.6 In vitro Magnetic Hyperthermia (MHT) Studies

Hyperthermia induces three main cellular responses: cytotoxicity, radio-sensitisation,

and thermal tolerance27, 28. Cancer cells undertake the apoptotic route in direct response to

applied heat, while the normal cells adapt to the temperature changes much easily. The cells

that do not undergo immediate apoptosis become sensitised towards other forms of therapy,

such as certain chemotherapeutic drugs or radiations. The heat-generating property of magnetic

nanoparticles has been widely exploited in order to find an effective treatment for cancer11, 29-

31. The exposure of radio waves to a biological system is limited in terms of the SAR of the

nanomaterials, and is given below32:

𝑆𝐴𝑅 = 𝜎𝐸𝑖

2

𝜌 (Eqn. 4.3)

where Ei represents the rms value of the applied electric field strength in the tissue (V/m), σ is

the conductivity of body tissue (S/m), and ρ is the density of the body tissue (kg/m3). Its effect

on the cancer cells is assessed as the rate at which electromagnetic energy is absorbed by a unit

mass of the tissue.

Page 112: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

91

There are two possible means to induce heat in the cancer cells that are intended for

therapeutics; by heating its (a) extracellular and its (b) intracellular environment. Both the

methods have their own advantages and disadvantages, which are mainly based on their ability

to affect the viable cancer cell population. Extracellular heating was followed in the current

work and the results were encouraging. Two conditions served as the control for this study: (a)

cell suspension with the dendritic nanoparticles, but without the ACMF and (b) cell suspension

without dendritic nanoparticles, but with application of ACMF. The control cells do not show

any reduction in cell proliferation activity and cell population. This confirms that the

application of the ACMF and the administration of nanoparticles separately is not enough to

cause the desired cell death.

On the other hand, the nanoparticles in combination with ACMF led to much more

interesting and promising results (Figure 4.11a). The effect of MHT was established from the

change in the cellular morphology and integrity. Though the MHT (under the above mentioned

parameters) was not enough to kill the cells, the treatment caused discernible changes in the

cellular morphology, the plasma membrane and the cytoplasm, which led to cell death. Figure

4.11b depicts the effect of elevated temperatures on the HeLa cell population under the

influence of the ACMF for various treatment times. The reduction of ~20% of viable cells

instantaneously in the viable cell population is observed after a treatment time of 5 min. Even

a small treatment time of 10 min is sufficient to reduce the number of live cells by ~50%. The

treatment time at which the cell population is reduced to 50% is considered as its median lethal

dose (LD50) and is further used to evaluate the effects of combinatorial therapy in vitro. This

lethal effect of heat on the proliferation and the viability of cells attains saturation at 20 min

and shows minimal effects with further increase in the treatment time.

Page 113: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

92

Figure 4.11 (a) Synergistic effects of DOX-loaded nanoparticles and hyperthermia on HeLa

cells (b) Viable HeLa cell population in the presence of ACMF for varying treatment times

for both PAMAM–IO and KR2–IO nanoparticles

4.2.3.7 ACMF-triggered Combinatorial Therapy

The combinatorial effect of DOX-loaded dendritic nanoparticles and hyperthermia was

evaluated in HeLa cells. The treatment time at LD50 values and the formulation concentration

at IC50 values were used in the combining of chemotherapy with magnetic hyperthermia. Figure

4.12 summarises the effect of DOX-loaded nanoparticles in combination with the ACMF-

induced elevated temperatures. The enhanced cell death that is observed is a result of the fatal

synergistic contribution of DOX and high temperatures. The combinatorial treatment is seen to

reduce the viable cell population to 2.5 and 3.6 % for DOX-PAMAM–IO and DOX-KR2–IO,

respectively.

Figure 4.12 Synergistic effects of DOX and magnetic hyperthermia (MHT) on viable HeLa

cell population after exposure to both, DOX-loaded dendritic nanoparticles and ACMF.

Page 114: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

93

The laser scanning confocal microscopy (LSCM) images (Figure 4.13) of the control HeLa

cells against the treated cells confirm the detrimental effects of combinatorial therapy. LSCM

images were recorded using an inverted confocal microscope by Olympus, model IX 81. Due

to the fluorescent property of DOX, no tagging molecule was required; the nucleus was stained

using a 4′, 6-Diamidino-2-phenylindole (DAPI) solution. The images show the confocal images

as well as the graphical representation of the fluorescent intensity. These images confirm that

the DOX-loaded dendritic nanoparticles were successfully internalised by the HeLa cells and

caused detrimental changes. The images clearly depict that the plasma membrane is completely

disrupted after the combinatorial treatment, which is due to the toxic effect of DOX in addition

to the MHT. The nucleus is seen to be intact, which gradually degrades as the cells undergo

apoptosis. The graphical analyses of the fluorescent intensity of the control cells have a smooth

and collective intensity profile. In the treated cells, however, the intensity is scattered and

higher. This increase in the intensity of green fluorescent peaks suggested the degradation and

fragmentation of cells due to the released DOX molecules, which lead to the desired cell death.

FITC-labelled nanoparticles were used to tag the control cells. The DOX-loaded dendritic

nanoparticles in synergism with hyperthermia are seen to be successful in reducing the live cell

population to a minimum.

Page 115: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

94

Figure 4.13 Laser Scanning Confocal microscopy images of treated HeLa cells with FITC-

nanoparticles (control), DOX-PAMAM–IO and MHT, and DOX-KR2–IO and MHT. The

scale bar of the images is 50 μm.

Page 116: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

95

4.2.4 Summary

This section elaborates on our attempt to combine dendrimers with superparamagnetic

Fe3O4 nanoparticles, which resulted in the ‘best of both worlds’ and opened up new arenas in

the field of improved cancer therapeutics. This work demonstrated the fabrication of aqueous-

stable, magnetic and biocompatible formulation of dendritic nanoparticles through the use of

PAMAM and peptide dendrimers. These dendritic nanoparticles showed good SAR values

hence, better efficiency in magnetic hyperthermia. The peptide dendrimer is substantially

efficient than the PAMAM dendrimer in both loading/carrying and delivering DOX to the

desired cancer cells. These formulations were also successful in combining chemotherapy with

magnetic hyperthermia. The comparative evaluation of these dendritic nanoparticles for drug

delivery, magnetic hyperthermia and combination therapy established the superiority of the

peptide dendrimer over PAMAM dendrimers. Also the physico-chemical properties of peptide

dendrimer were minimally compromised as compared to its PAMAM counterpart. These

promising in vitro results established the dendritic-Fe3O4 nanoparticles attractive platforms for

further in vivo evaluations.

4.2.5 References

1. G. Delaney, S. Jacob, C. Featherstone and M. Barton, Cancer, 2005, 104, 1129-1137.

2. S. H. Kaufmann and W. C. Earnshaw, Experimental Cell Research, 2000, 256, 42-49.

3. T. Vial and J. Descotes, Toxicology, 2003, 185, 229-240.

4. R. E. Rosensweig, Journal of Magnetism and Magnetic Materials, 2002, 252, 370-374.

5. S. Chandra, N. Barola and D. Bahadur, Chemical Communications, 2011, 47, 11258-

11260.

6. A. P. Majewski, A. Schallon, V. Jérôme, R. Freitag, A. H. E. Müller and H. Schmalz,

Biomacromolecules, 2012, 13, 857-866.

7. C. Wang, S. Ravi, U. S. Garapati, M. Das, M. Howell, J. Mallela, S. Alwarappan, S. S.

Mohapatra and S. Mohapatra, Journal of Materials Chemistry B, 2013, 1, 4396-4405.

8. A. Mandal, S. Sekar, M. Kanagavel, N. Chandrasekaran, A. Mukherjee and T. P. Sastry,

Biochimica et Biophysica Acta (BBA) - General Subjects, 2013, 1830, 4628-4633.

9. L. Zhang, W.-F. Dong and H.-B. Sun, Nanoscale, 2013, 5, 7664-7684.

10. S. Yalçın, M. Erkan, G. Ünsoy, M. Parsian, J. Kleeff and U. Gündüz, Biomedicine &

Pharmacotherapy, 2014, 68, 737-743.

Page 117: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

96

11. A. Jordan, R. Scholz, P. Wust, H. Fähling and F. Roland, Journal of Magnetism and

Magnetic Materials, 1999, 201, 413-419.

12. L. Asín, M. R. Ibarra, A. Tres and G. F. Goya, Pharmaceutical Research, 2012, 29,

1319-1327.

13. R. Di Corato, A. Espinosa, L. Lartigue, M. Tharaud, S. Chat, T. Pellegrino, C. Ménager,

F. Gazeau and C. Wilhelm, Biomaterials, 2014, 35, 6400-6411.

14. I. Sharifi, H. Shokrollahi and S. Amiri, Journal of Magnetism and Magnetic Materials,

2012, 324, 903-915.

15. B. P. Shah, N. Pasquale, G. De, T. Tan, J. Ma and K.-B. Lee, ACS Nano, 2014, 8, 9379-

9387.

16. K. C. Barick, S. Singh, N. V. Jadhav, D. Bahadur, B. N. Pandey and P. A. Hassan,

Advanced Functional Materials, 2012, 22, 4975-4984.

17. Y.-J. Kim, M. Ebara and T. Aoyagi, Advanced Functional Materials, 2013, 23, 5753-

5761.

18. W. Rao, W. Zhang, I. Poventud-Fuentes, Y. Wang, Y. Lei, P. Agarwal, B. Weekes, C.

Li, X. Lu, J. Yu and X. He, Acta Biomaterialia, 2014, 10, 831-842.

19. P. T. Yin, S. Shah, N. J. Pasquale, O. B. Garbuzenko, T. Minko and K.-B. Lee,

Biomaterials, 2016, 81, 46-57.

20. P. T. Yin, B. P. Shah and K.-B. Lee, Small, 2014, 10, 4106-4112.

21. B. Ahmad, S. Parveen and R. H. Khan, Biomacromolecules, 2006, 7, 1350-1356.

22. V. Vichai and K. Kirtikara, Nat. Protocols, 2006, 1, 1112-1116.

23. M. C. Popescu, D. Filip, C. Vasile, C. Cruz, J. M. Rueff, M. Marcos, J. L. Serrano and

G. Singurel, The journal of physical chemistry. B, 2006, 110, 14198-14211.

24. G. Glöckl, R. Hergt, M. Zeisberger, S. Dutz, S. Nagel and W. Weitschies, Journal of

Physics: Condensed Matter, 2006, 18, S2935.

25. M. Jeun, S. Bae, A. Tomitaka, Y. Takemura, K. H. Park, S. H. Paek and K.-W. Chung,

Applied Physics Letters, 2009, 95, 082501-082503.

26. P. de la Presa, Y. Luengo, M. Multigner, R. Costo, M. P. Morales, G. Rivero and A.

Hernando, The Journal of Physical Chemistry C, 2012, 116, 25602-25610.

27. L. F. Fajardo L-G, Cancer Research, 1984, 44, 4826s-4835s.

28. J. Landry, S. Samson and P. Chrétien, Cancer Research, 1986, 46, 324-327.

29. N. K. Prasad, K. Rathinasamy, D. Panda and D. Bahadur, Journal of Materials

Chemistry, 2007, 17, 5042-5051.

Page 118: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

97

30. K. H. Bae, M. Park, M. J. Do, N. Lee, J. H. Ryu, G. W. Kim, C. Kim, T. G. Park and

T. Hyeon, ACS Nano, 2012, 6, 5266-5273.

31. E. Gkanas, cent.eur.j.chem., 2013, 11, 1042-1054.

32. J. M. Osepchuk and R. C. Petersen, Microwave Magazine, IEEE, 2001, 2, 57-69.

Page 119: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

98

Chapter 5

Assessment of Doxorubicin-loaded Dendritic Fe3O4

Nanoparticles for Magnetic Drug Targeting in Murine

Melanoma Model

5.1 Introduction

The previous chapters of this thesis have led to the conclusion that peptide dendrimer

based Fe3O4 nanoparticles (a) have superior drug delivery efficacy and (b) can synergistically

combine thermo- and chemotherapy for improved therapeutic performances. While the Fe3O4

nanoparticles actively contribute in magnetic hyperthermia, they are also capable of playing a

dual role as a targeting platform. A major challenge in stepping up the successful in vitro

therapies to in vivo scenarios is the uncontrolled biodistribution of the delivery vectors1, 2. This

random systemic dispersal of nanoparticles leads to the release of drug molecules in non-

specified locations causing adverse effects to otherwise healthy tissues. Towards this end,

specific targeting of these delivery vectors have been attempted using a variety of molecules

like antibodies3-6, ligands7, 8, peptides9, 10 and the like. These molecules are known to attach

specifically to the receptors present on the cell membranes and are thus, selectively internalised

by the cancer cells11. This reduces their uptake by the neighbouring normal cells thereby

reducing the side effects. But these targeting moieties also have critical disadvantages as they

are specific to only a certain kind of receptors expressed in cancer cells, that is, a receptor

Page 120: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

99

present on a prostate cancer cell may not be expressed by a breast cancer cell. Thus, one type

of targeting ligand would be specific to deliver drugs to the tumour depending on the type of

cancer dealt with.

To overcome this limitation, magnetic drug targeting presents itself as a promising

attempt. As the name suggests, this method involves passive targeting of the delivery vectors,

bound to the drug molecules, to the tumour region by the use of a strong external magnetic

field12. The magnetic nanoparticles are of immense importance because the movement of these

nanoparticles can be controlled and guided by an external magnetic field. In concept,

chemotherapeutic agents are bound to these nanoparticles and introduced into the arterial

circulation, supplying to the tumour, while guided externally by a strong magnetic field. It not

only leads to higher accumulation of drug in the tumour but also reduces the overall dose

administered in the body thus minimising the side effects. In order to be used for MDT, one of

the mandatory requirement for these nanoparticles is to exhibit magnetism only in the presence

of an external magnetic field. This requirement should be fulfilled in order to avoid any

undesirable magnetic agglomeration in the absence of magnetic fields. Thus,

superparamagnetic nanoparticles are chosen over the ferromagnetic nanoparticles. Many

attempts have been made recently to address the achievability of magnetic drug targeting13.

Nowicka and co-workers fabricated a doxorubicin-iron oxide conjugate and applied a

magnetic field to guarantee its efficient delivery to the desired human urinary bladder

carcinoma cell line14. Pourmehran et al.15, 16 used discrete phase modelling and one-way

coupling of particle–fluid phases to simulate the air flow and magnetic particle deposition and

tracking in the presence of an external non-uniform magnetic field in human lung model. Their

computational results showed that MDT technique is promising and has a direct relation with

increasing the particle diameter. Lunnoo et al.17 simulated and investigated the parameters,

which govern the capture efficiency of the drug carriers by MDT in mimicked arterial flow.

Their study concluded that the capture efficiency of small particles decreased with decreasing

size and was less than 5% for magnetic particles in the superparamagnetic regime. Though the

thickness of non-magnetic coating materials did not significantly influence the capture

efficiency, it was difficult to capture smaller nanoparticles (D<200 nm) in the arterial flow.

These results provided new insight into the mechanism and governing parameters of the MDT

for clinical settings. Arias et al.18 fabricated Fe3O4/chitosan nanocomposites, which not only

enhanced the intravenous delivery of gemcitabine to the cancer tissue but was also seen to be

hyperthermia responsive in an alternating magnetic field. The in vivo proof of concept, using

Page 121: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

100

Prussian blue staining, further confirmed the magnetic targeting capabilities of this

magnetite/chitosan core/shell nanocomposite. On the other hand, Hornung and co-workers

employed Fe3O4 nanoparticles to carry and deliver the cytotoxic drug mitoxantrone to human

colon carcinoma cells19. They developed three-dimensional multicellular tumour spheroids

from the colorectal cancer cells and studied the accumulation and delivery efficiency of these

nanoparticles. In an entirely different approach, Kempe et al.20 demonstrated the successful use

of tissue plasminogen activator (tPA) modified Fe3O4 nanoparticles in thrombolytic therapy by

employing magnetic drug targeting. Alexiou and co-workers found quite interesting and

encouraging results when they employed the MDT in the local accumulation of mitoxantrone

in the squamous cell carcinoma implanted in New Zealand White rabbits21. They administered

their nanoparticles in the femoral artery while an external magnetic field was applied on the

tumour. They found a significant (p < 0.05), complete, and permanent remission of the

squamous cell carcinoma as compared to the control group of animals. They successfully

combined intratumoural nanoparticle accumulation and locoregional cancer treatment without

systemic toxicity.

In the studies elaborating the application of MDT, the researchers have directly loaded

the therapeutic molecule on to the nanoparticle surface and administered in the artery supplying

the tumour for better accumulation. Though a variety of magnetic nanosystems are being

explored, dendrimer functionalised magnetic nanoparticles have not yet been evaluated as a

platform for MDT. The following sections thus, evaluates the performance of delivery of DOX-

loaded dendritic nanoparticles by magnetic drug targeting to the syngeneic melanoma mouse

model in C57BL/6 black mice. This chapter explores the bio-distribution and biocompatibility

of the dendritic nanoparticles, while assessing the therapeutic potential of DOX-loaded

nanoparticles in the presence and absence of external magnetic field.

5.2 Experimental and Characterisation Techniques

5.2.1 In vitro Evaluation

The biocompatibility of PAMAM–IO and KR2–IO was assessed with murine

melanoma (B16F10; skin cancer). The melanoma cells were grown in a 96-well microplate for

24 h prior to addition of the nanoparticles. The formulations were suspended in an appropriate

growth medium and serially diluted. 200 µl of this suspension was used to replace the spent

media in the wells, and the cells were allowed to grow for an additional 24 h and 48 h in the

Page 122: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

101

presence of the nanoparticles in humidified CO2 environment at 37 °C. To calculate the

therapeutic concentrations of DOX-PAMAM–IO and DOX-KR2–IO, a dose-dependent

evaluation was undertaken over 24 and 48 h. After the set time period (24 and 48 h), the MTS

colorimetric assay (CellTiter 96® Cell Proliferation Assay, Promega) was performed according

to the manufacturer’s protocol. The percentage viable cell population was estimated and

represented against control cells according to equation 2.3. The inhibitory concentration (IC50)

values of each of the nanoparticles was calculated by dose-responsive sigmoidal curve fitting

using Origin 9.1 software.

The effect of the magnetic field on the internalisation of these nanoparticles was

assessed. Towards this end, 1 × 106 cells were grown in 35 mm petridish for 24 h under

physiological conditions. After 24 h, the spent media was replaced by a suspension of

PAMAM–IO and KR2–IO nanoparticles (in the growth medium) of concentration 1 mg/ml.

These petridishes were then placed on a universal magnetic plate and the cells were incubated

in presence of magnetic field for 3, 6, 12 and 24 h. Another set of treated cells were grown for

similar time periods but without the magnetic plate. The untreated cells, unexposed to magnetic

field, were used as control. After the specific time periods, the cells were washed with PBS

(multiple times), trypsinised, centrifuged and the cell pellet was collected. The pellet was then

digested using 1 ml of conc. hydrochloric acid for 30 min. The volume of the cell lysate was

then made up to 10 ml with ultrapure water and the amount of internalised nanoparticles was

determined by the inductively coupled plasma-atomic emission spectroscopy (ICP-AES)

analysis. It should be noted that while rinsing the cells with PBS, some amount of nanoparticles

remains adhered to the cell membrane and cannot be washed out completely. These

nanoparticles are erroneous but still contribute to the final iron content calculated by ICP-AES.

5.2.2 In vivo Therapeutic Efficacy Studies

The animal studies were conducted in compliance with the Institutional Animal Ethical

Committee guidelines. 4-6 weeks old C57BL/6 mice were purchased, maintained and

experiments were carried out at National Toxicology Centre (NTC, Pune, India). Animals were

fed with food & water ad libitum and housed in a constant temperature and humidity conditions

with 12 h light and dark cycles. C57BL/6 black male mice (4-6 weeks’ age) were used for the

biocompatibility and bio-distribution study of the PAMAM–IO and KR2–IO (samples

described in earlier chapters). In vivo tumour inhibition studies were carried out in syngeneic

allograft of melanoma tumour model grown in the same species of black mice.

Page 123: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

102

5.2.2.1 Biocompatibility and Bio-distribution Studies

The animals were divided into three major experimental groups; Control, Sample I

(PAMAM–IO) and Sample II (KR2–IO). The detailed information about the grouping of

animals, dosing and sacrifice is listed in table 5.1.

Table 5.1 Animal denomination, dosage of dendritic nanoparticles and time points of sample

collection for biocompatibility and biodistribution studies (h=hour; d=day)

Grouping of animals Number of

animals

Time of

sample

collection

Group Sub-group

PAMAM–IO I 3 24 h

II 3 48 h

III 3 7 d

IV 3 14 d

KR2–IO V 3 24 h

VI 3 48 h

VII 3 7 d

VIII 3 14 d

Control IX 3 24 h

X 3 14 d

The dose for the biocompatibility and biodistribution studies were determined based on the

existing literature1 (15 mg/kg body weight). 200 µl of this concentration was administered

through the tail vein using a 27-gauge needle. The animals of the control group were injected

with 200 µl of sterile PBS. The animals were euthanised by cervical dislocation after the set

time points post dose-administration. Their tissues (heart, kidneys, lungs, liver, spleen, brain,

stomach and thigh muscles from right hind leg) were collected, weighed and stored at -20ºC

for further analysis and their blood was collected for immediate analysis for the biochemical

and haematological parameters. A complete analysis of the blood was performed (Mindray

BC2800 Vet Auto hematology Analyser) which included examination of white blood cells

(WBC), lymphocytes (LYM), monocytes (MON), granulocytes (GRAN), red blood cells

(RBC), haemoglobin (Hb), haematocrit (HCT), mean corpuscular volume (MCV), mean

Page 124: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

103

corpuscular haemoglobin (MCH), mean corpuscular haemoglobin concentration (MCHC) and

platelets (PLT). The activity of enzymes serum glutamic pyruvic transaminase (SGPT), serum

glutamic oxaloacetic transaminase (SGOT) and alkaline phosphatase (ALP) were analysed

from the plasma using aSmart-7 Semi auto Chemistry analyser, Pathozyme. The levels of

creatinine and blood urea nitrogen (BUN) were also evaluated. The activity of the above

mentioned enzymes, creatinine and BUN reflect on the activity and health of the liver, which

is a direct indicator of the biocompatibility of these nanoparticles. For sample preparation for

ICP-AES, all the tissues were dried at 55-60 °C, their dry organ weight was recorded, and

crushed to obtain powder. To a known amount of tissue powder, conc. hydrochloric acid was

added and kept overnight for complete dissolution and used for the estimation of their iron

content through ICP-AES. The iron accumulation in the vital organs were represented per mg

corresponding dry organ weight.

5.2.2.2 Tumour Regression and Therapeutic Efficacy Studies

Development of animal tumour model: The mice bearing subcutaneous melanoma were used

as the model. To male mice (4-6 weeks), 1×106 B16F10 melanoma cells (suspended in sterile

saline solution) were subcutaneously injected on the right flank and observed for tumour

growth. On the 12th – 15th day of inoculation, a palpable tumour appeared and the animals with

uniform tumour volume (~ 100 mm3) were selected for further studies.

Anti-tumour Efficacy and Magnetic Drug Targeting: Our earlier studies established that

KR2–IO nanoparticles showed superior performances over PAMAM–IO nanoparticles.

Henceforth, DOX-loaded KR2–IO nanoparticles were assessed for in vivo therapeutic efficacy

studies. The effect of magnetically guided passive targeting was evaluated in terms of

nanoparticle accumulation and corresponding tumour regression. The tumour bearing mice (on

15th day of tumour inoculation) were randomly divided into four groups with three animals

each. The details about the grouping of animals, dosage and magnetic exposure are listed in

table 5.2.

Page 125: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

104

Table 5.2 Animal denomination, dosage of dendritic nanoparticles and magnetic exposure for

therapeutic efficacy studies (h=hour; MF=magnetic field)

Group Sample Number of

animals Dose

Magnetic

exposure

I Control 3 Sterile PBS -

II Pure DOX 3

10 mg of

DOX per kg

body weight

-

III DOX-loaded

KR2–IO 3 -

IV DOX-loaded

KR2–IO + MF 3

6 h after every

dosage

Group I mice served as control and were injected with sterile PBS, group II with pure

DOX, group III with DOX-loaded KR2–IO and group IV with DOX-loaded KR2–IO in

presence of a neodymium-iron-boron based permanent magnet (250 mT for 6 h). The samples

were administered in the tail vein of the animals at a DOX concentration of 10 mg/kg body

weight22, 23 and a half dose was administered every 6th day. The animals of group IV were

exposed to the magnetic field along with every dosage. After every third day, from the start of

the study, the tumour dimensions were measured using a digital Vernier caliper and the tumour

volume was calculated according to equation 5.124.

𝑇𝑢𝑚𝑜𝑢𝑟 𝑠𝑖𝑧𝑒 (𝑚𝑚3) = 𝜋

6 × 𝑚𝑎𝑗𝑜𝑟 𝑑𝑖𝑚𝑒𝑛𝑠𝑖𝑜𝑛 × 𝑚𝑖𝑛𝑜𝑟 𝑑𝑖𝑚𝑒𝑛𝑠𝑖𝑜𝑛2 (Eqn. 5.1)

It is noteworthy that caliper measurements of the subcutaneous allograft are superficial and are

affected by the contribution from the epidermis and underlying adipose tissue, as well as fur,

each of which adds some error and inconsistency in the volume calculations.

Page 126: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

105

Figure 5.1 Melanoma tumour bearing C57BL/6 mouse being administered intravenously

with DOX-loaded KR2–IO nanoparticles. The skin around the tumour region was shaved and

neodymium magnet was stuck using a medical tape for 6 h.

The treated animals were then kept under observation till all the animals of control

(group I) died. The tumour size of the animals of group II, III and IV was measured on regular

intervals and tumour regression/inhibition, if any, was duly recorded. The animals were

subjected to euthanasia by cervical dislocation and tissues (heart, kidneys, lungs, liver, spleen,

brain and tumour) were collected and stored for analyses.

Histopathological examination was carried out of the collected tissues in order to

confirm the iron accumulation visually. The tissue pieces from the vital organs, of suitable size,

from all the experimental animals were collected group-wise in 10% formalin in labelled plastic

container for fixation immediately after necropsy. The collected tissues were then processed

for histological procedure using alcohol-xylene protocol for 24 h in order to dehydrate and

clear the tissues. The tissues were initially placed in 50% absolute alcohol for 4 to 6 h for

removal of fixative, then in ascending grades of alcohol for 2 h each, and finally kept in

absolute alcohol for 2 h. After clearing with xylene, the tissues were embedded in paraffin

blocks, sectioned at 4–5 µm thickness using automated microtome (Leica, Germany). The

tissues’ sections were stained by standard Prussian Blue staining protocol and fixed for

observation. All the fixed slides were examined under a Binocular Light Microscope

Page 127: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

106

(Olympus, Japan) and the images were captured using a digital camera (Sony, Japan).

Histopathological evaluation was carried out by an expert qualified veterinary pathologist, who

was blinded to the groups. The iron estimation in the dried tissue powder was also undertaken

as described earlier.

Statistical Analyses: All the results were expressed as mean ± standard deviation. Statistical

significance of the results was computed by two sample t-test using Origin 9.1 software and

p<0.05 confidence intervals were considered significant.

5.3 Results and Discussion

5.3.1 In vitro Evaluation

The PAMAM–IO, and KR2–IO nanoparticles were seen to be non-toxic as the cell

proliferation activity and viable cell population remained unaffected (Figure 5.2 a, b). After 48

h of incubation, the higher concentrations of the PAMAM–IO nanoparticles showed meagre

loss in percent viable cells (~10%) while KR2–IO nanoparticles essentially maintained the

same viable cell population as control. The melanoma cells showed neither change in the

cellular morphology nor any other loss of structural integrity. Therefore, it was concluded that

these nanoparticles were safe to be used for in vivo studies. The DOX-loaded dendritic

nanoparticles are competent to release DOX which efficiently reduced the population of the

melanoma cells (Figure 5.2 c). The IC50 values of DOX-PAMAM–IO and DOX-KR2–IO are

summarised in table 5.3.

Figure 5.2 (a, b) Biocompatibility profile of PAMAM–IO and KR2–IO with B16F10

melanoma cells after 24 and 48 h respectively (c) Dose-dependent cell viability profile of

DOX-loaded dendritic Fe3O4 nanoparticles after 24 and 48 h. Their IC50 values showed

significant difference for 24 h and 48 h (p < 0.05) while there was no significance between

the DOX-PAMAM–IO and DOX-KR2–IO at same time point (p > 0.05).

Page 128: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

107

Table 5.3 IC50 values of DOX-PAMAM–IO and DOX-KR2–IO nanoparticles with B16F10

melanoma cells. The values are represented in mg/ml of formulations with ≤ 15% (IC50 of

pure DOX was 0.11 – 0.17 μM25)

24 h 48 h

DOX-PAMAM–IO 2.4±1.9 (R2 = 0.999) 0.7±0.1 (R2 = 0.992)

DOX-KR2–IO 3.2±1.5 (R2 = 0.999) 0.6±0.2 (R2 = 0.955)

These dendritic Fe3O4 are seen to be successfully internalised by the melanoma cells (Figure

5.3). The positively charged surface of the dendrimers present on these nanoparticles play a

very important role in facilitating their uptake. These nanoparticles interact electrostatically

with the negatively charged cell membrane and compromise its structural integrity which forms

the passage for their internalisation. Figure 5.3a schematically illustrates the concept of

magnetically-guided cellular internalization of nanoparticles. Figure 5.3b shows confocal

micrographs (A, C) of the melanoma cells and their corresponding fluorescence profile (B, D).

The green fluorescence of FITC-tagged dendritic Fe3O4 nanoparticles is seen to be evenly

distributed throughout the cytoplasm while blue fluorescence marked the nucleus. Though both

of the dendritic Fe3O4 nanoparticles are positively charged, KR2–IO show better uptake than

the PAMAM–IO. This behaviour could be attributed to the resonating amine groups of arginine

on the surface of KR2–IO which makes it more positively charged than PAMAM–IO (discussed

in chapter 4B).

Page 129: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

108

Figure 5.3 (a) Schematic representation of magnetically-guided cellular internalization of

dendritic Fe3O4 nanoparticles, (b) (A, C) Confocal micrographs and (B, D) fluorescence

profiles of PAMAM–IO and KR2–IO nanoparticles internalized by B16F10 melanoma cells

after 24 h (c) Amounts of iron internalized by the melanoma cells estimated using atomic

emission spectroscopy

This enhanced cellular uptake is further improved by the magnetic field, which aids in better

adherence of these nanoparticles on the cell membrane. The effect of presence of magnetic

field was evaluated on the amount of nanoparticles taken up by the melanoma cells as a function

of the magnetic field exposure time (Figure 5.3 c). The AES analyses show that control cells

have an iron content of 0.0625±2.24 µg and the internalisation of both the dendritic

nanoparticles increased the iron content of the cells substantially. The amount of iron was

represented as µg per mg of dendritic Fe3O4 nanoparticles. After 3h of incubation, there is no

significant effect of the magnetic field in the iron content of the cells. Only after 6 h of

incubation, a substantial increase in iron content is observed, i.e. ~33 µg/mg for PAMAM-IO

and ~38 µg/mg for KR2-IO, in the cells exposed to the magnetic field. When compared between

the formulations with and without MF, 6 h of incubation on the magnetic plate shows ~7-fold

increase in the amount of iron internalised by the cells which reduced to ~3-fold increase at 12

h and further decreased to ~1.6-fold after 24 h in comparison to the control cells. This is

indicative of the fact that magnetically-guided uptake of the nanoparticles could only be

Page 130: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

109

enhanced to a certain limit which is dependent on the time for which the magnetic field is

applied. This result proved to be the pivotal basis of our in vivo magnetic drug targeting study

in the mouse tumour model.

5.3.2 In vivo Therapeutic Efficacy Studies

5.3.2.1 Biocompatibility and Bio-distribution Studies

Haematological Parameters: A complete blood evaluation (CBC) were performed to study

the variations, if any, after the intravenous sample administration. Figure 5.4 shows changes

observed in counts of red blood cells (RBC), haemoglobin (Hb), platelets (PLT), white blood

cells (WBC), lymphocytes (LYM), monocytes (MON) while figure 5.5 depicts the summarised

values for mean corpuscular volume (MCV), mean corpuscular haemoglobin (MCH) and mean

corpuscular haemoglobin concentration (MCHC). The levels of RBC and haemoglobin were

minimally affected by the nanoparticles suggesting their haemocompatibility. The platelets’

count in the blood is a very critical parameter as these cells are responsible for haemostasis.

PAMAM–IO caused a sudden drop of platelets’ level in the first 24 h (p<0.01) which regained

its normal level within a week and is maintained thereafter. On the other hand, KR2–IO did not

show any significant effect on the count of platelets through the period of 14 days post-

administration. The introduction of these nanoparticles in the systemic circulation generates an

immune response, which is indicated by the elevated levels of white blood cells. This immune

response triggers specific cells (lymphocytes and monocytes) in blood which are responsible

for the clearing of the nanoparticles from the blood stream and take them to organs like liver,

lungs and spleen to be degraded and cleared out of the body. The levels of WBCs take

approximately 7 days to return to their normal levels, which is also in congruence with the iron

estimation analyses discussed later in this chapter. The administration of PAMAM–IO and

KR2–IO did not cause any significant variation in the MCV, MCH and MCHC.

Page 131: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

110

Figure 5.4 Histograms depicting variations in blood parameters for (a) red blood cells, (b)

haemoglobin, (c) Platelets, (d) white blood cells, (e) lymphocytes and (f) monocytes. The

study showed variations across 14 days of study after intravenous administration of dendritic

Fe3O4 nanoparticles. The results are expressed as mean ± s.d. (n=3) with statistical

significance *p < 0.05 and **p < 0.01 with respect to control animals.

Figure 5.5 Histograms showing (a) mean corpuscular volume, (b) mean corpuscular

haemoglobin and (c) mean corpuscular haemoglobin. The results are expressed as mean ± s.d.

(n=3) with statistical significance *p < 0.05 and **p < 0.01 with respect to control animals.

Serum Biochemistry: After intravenous administration of dendritic Fe3O4 nanoparticles to the

mice, the serum was analysed for the biochemical parameters. The activity of enzymes SGPT,

SGOT and ALP were evaluated over the course of the study (Figure 5.6 a,b,c). These enzymes

are suggestive of healthy liver functions. Any fluctuations in their levels primarily indicate the

inflammatory response of the body towards a foreign moiety, such as nanoparticles. Both

PAMAM–IO and KR2–IO cause an instant elevation in the levels of SGPT and SGOT (p <

Page 132: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

111

0.05) for the initial 24 h and resume their normal levels by the 7th day. This behaviour is also

suggestive of little or no toxicity of these nanoparticles to organs and regular bodily functions.

Another important parameter is the level of creatinine and urea in the blood, which is an

indicator of healthy renal function. No significant changes were observed in the creatinine

levels of the treated animals compared to the control (Figure 5.6 d). On the other hand, slightly

elevated levels of blood urea were observed for a period of 48 h (Figure 5.6 e) which declined

during the course of next 7 days and returned to their physiological levels (p<0.005). These

results clearly indicated healthy renal function in the mice after treatment with nanoparticles.

The results directly suggest the biocompatibility of both PAMAM-IO as well KR2-IO

nanoparticles.

Figure 5.6 Histograms depicting variations in serum biochemical parameters for (a) SGPT,

(b) SGOT, (c) ALP, (d) Creatinine and (e) blood urea nitrogen (over different time points

during the 14-day study period). The results are expressed as mean ± s.d. (n=3) with

statistical significance *p < 0.05 and **p < 0.01 with respect to control animals.

Bio-distribution assessment by iron estimation: The bio-distribution of dendritic Fe3O4

nanoparticles, administered at 10 mg per kg body weight, was evaluated after intravenous

injection in various vital organs (heart, kidneys, lungs, liver, spleen, brain, stomach and thigh

muscles). Post intravenous administration these dendritic Fe3O4 nanoparticles are freely

available to every organ in the body. Analysis and estimation of iron in the vital organs could

be a direct indication of the bio-distribution of these nanoparticles. A time-dependent

estimation of iron was carried out and is summarised in figure 5.7. Except lungs, liver and

Page 133: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

112

spleen, the level of iron in all the tested organs was very low (<10 µg/mg of dry organ weight),

suggesting non-specific uptake in these organs. Lungs, liver and spleen show substantially high

level of iron (> 20 µg/mg of dry organ weight) for the first 24 h which gradually declined over

the next 13 days attaining their least value (~5 µg/mg of dry organ weight). This trend of high

iron level in these vital organs provided a better understanding and suggested that these organs

are responsible for biodegradation of the nanoparticles with the passage of time. This suggests

that these organs play a very important role in degradation and clearance of the nanoparticles.

The macrophages and Kupffer cells are responsible for uptake of the nanoparticles by active

phagocytosis in the lungs and liver, which is responsible for their rapid clearance from the

systemic circulation and accumulation in the organs. The size of the nanoparticles plays a very

important role in their phagocytosis and clearance. Ultra-small nanoparticles (<20 nm) have

been known to evade these cells resulting in a longer circulation time as compared to their

bigger counterparts1. The brain exhibited an extremely low accumulation of iron. This could

be due to the blood brain barrier (BBB), which prevents the accumulation of unwanted foreign

matter in the brain. It should also be noted that a meagre amount of iron extravasated to the

thigh muscles extracted from the right flank of the mice. This trend suggests that the non-

specific uptake of dendritic Fe3O4 by the thigh muscles is negligible and displays a major

difference when compared to the magnetically-guided accumulation (discussed later). By the

end of 30th day, the residual iron content of all the analyzed organs was insignificant (<2 µg/mg

of organ) and similar to control animals which confirmed the biodegradability and clearance

of both our dendritic Fe3O4 nanoparticles. Figure 5.7c depicts the variations in the body weight

of the mice, and shows negligible changes in the body weight post-administration. From the

above results, it could be concluded that both of these dendritic Fe3O4 nanoparticles are

completely compatible and safe to be used with mice and do not have any detrimental effects

on the body.

Page 134: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

113

Figure 5.7 Biodistribution of (a) PAMAM–IO and (b) KR2–IO nanoparticles through

quantification of iron accumulated in various vital organs represented at different time points

post intravenous administration. The quantified iron is represented as mean ± s.d. after

control values were deducted from the plot. (c) Body weight profiles of mice across 14 days

of study. The changes in weight of animals are non-significant (p > 0.05) and does not show

any critical changes.

5.3.2.2 Tumour Regression and Therapeutic Efficacy Studies

Tumour regression and Magnetic Drug Targeting: The in vivo bio-distribution of the

PAMAM–IO and KR2–IO nanoparticles indicated that these nanoparticles are biocompatible

and could be successfully used as drug delivery vectors. Towards this end, the DOX-loaded

KR2–IO nanoparticles were assessed for in vivo anti-tumour therapeutic efficacy using a

syngeneic melanoma model in C57BL/6 mice. The administration of free DOX in group II

mice was seen to reduce the tumour size in comparison to the control animals (p<0.05) but also

affected the body weight after 14 days’ post-administration (Figure 5.8a). This result was in

accordance to the fact that when the free drug is administered, it is non-specifically taken up

not only by the tumour cells but also by all the other vital organs and thus, have unhindered

detrimental side effects on the bodily functions. DOX-loaded KR2–IO nanoparticles also

successfully delivered DOX to the tumour region which resulted in significant tumour growth

inhibition when compared to the untreated control animals (Figure 5.8b). In the animals of

group III, the tumor size remained static till 14th day of the treatment (tumor growth inhibition)

and only showed insignificant growth by the end of 35th day. Even after significant tumor

regression is observed and 35 days of treatment, the tumor is still tangible in the animals of

group III. The rate of survival of these animals was much higher than control and free DOX

Page 135: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

114

treated animals as 60% of animals (group III) were alive after 60 days with a small tangible

tumor.

Figure 5.8 (a) Body weight profiles of mice across 35 days of study. The changes in weight

of control animals are due to the uncontrolled tumour size. The weight of animals of group II,

III and IV are significantly decreased (*p < 0.05) in comparison to the control animals, (b)

tumour volume profiles of untreated control animals against the treated groups II, III and IV.

Tumour growth was significantly inhibited with DOX-loaded KR2–IO nanoparticles as

against animals treated with free DOX. (c) Quantification of iron accumulated in various vital

organs. In absence of magnetic field, the thigh muscles (tumour) tend to accumulate small

amount of iron which is significantly elevated with application of magnetic field.

The accumulation of iron in the tumour is observed in the animals of group III where

the amount of iron was found to be below 25 µg/mg dry organ weight (Figure 5.8 c). This

meagre increase could be attributed to the nanoparticle accumulation due to leaky tumour

vasculature. On the other hand, the application of magnetic field around the tumour region in

animals of group IV, caused a substantial increase in the amount of nanoparticles (161 µg of

Fe/mg dry organ weight) in the tumour. The application of magnetic field attracts the

nanoparticles from the blood stream to the tumour region and the accumulation is further

facilitated by the leaky vasculature at the tumour site. This elevated level of nanoparticles in

the tumour leads to high concentration of DOX released which in turn improves the tumour

inhibition efficacy of our DOX-loaded KR2-IO nanoparticles (Figure 5.8 c). Therefore, the rate

of tumour size reduction and growth regression was seen highest in the mice of group IV. By

the end of second dosing (14th day), the average tumour volume was 55 ± 8.3 mm3 as compared

to the control animals in which the tumour volume was seen to be 4794 ± 844 mm3 (~ 88-fold

decrease). The tumour was seen to disappear by the end of 20th day post-treatment but the mice

were kept under observation for evaluation of their survival and tumour re-lapse, if any. The

Page 136: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

115

mice of all the groups were kept under continuous observation until the control mice died which

marked the termination point of the study. We observed death of only one animal from the

treated group IV in entire study period on 37th day while,~100% survival rate was observed

with no tumor re-lapse even after 60 days.

Figure 5.9 shows the histopathological images of the tissue sections with Prussian blue

staining. The stained slides did not exhibit any signs of toxicity in terms of tissue damage and

structural integrity in the organs viz. kidney, lungs, heart, brain, spleen and liver. Further, the

tumor sections of the treated group without the magnetic field exhibited very less nanoparticle

accumulation against the group exposed to the magnetic field along with the nanoparticles.

These results further support the magnetically-guided nanoparticle accumulation data and

tumor regression, confirming the improved anti-tumor potential of peptide dendrimer-based

Fe3O4 nanoparticles.

Figure 5.9 Histological analyses of the excised organs after 24 h. Optical micrographs of the

treated tumor exhibited zones with compromised cellular cohesion and structural integrity

with significantly different iron content in absence and presence of magnetic field (MF).

Other vital organs showed minimal accumulation of iron causing little or no toxicity to

normal tissues and bodily functions.

5.4 Summary

After validating the in vitro efficacy of PAMAM–IO and KR2–IO nanoparticles, this

chapter focussed on in vivo investigation of these particles in a subcutaneous syngeneic murine

melanoma model. The systemic exposure of these nanoparticles caused changes in various

blood and serum parameters. This assisted with the information about their toxicity and non-

specific uptake by various organs. The variations in the complete blood counts and the enzymes

(SGPT and SGOT) showed only an initial change which resumed to their physiological levels

Page 137: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

116

within 7 days of treatment. Both the dendritic nanoparticles showed a tendency to localise in

lungs, liver and spleen which are responsible for their rapid clearance from the systemic

circulation. However, in tumour bearing mice, localisation of nanoparticles is manipulated by

the application of an external magnetic field. We observed ~6-fold higher nanoparticle

localisation in magnetically targeted tumour (right flank) as compared to passive localisation

in thigh muscles (right flank) of non-tumour bearing mice. This elevated levels of iron

accumulation in the tumour is due to the erratic angiogenesis causing leaky vasculature in the

tumour, which is further facilitated by the application of an external magnetic field. This high

localisation pattern of DOX-loaded KR2–IO led to high concentrations of DOX in the tumour

and thus was effective in arresting the tumour growth significantly. It was seen that lower

number of doses are sufficient to suppress the tumour growth in combination with magnetic

field than that required without the magnetic field. This elaborate study confirmed that KR2–

IO nanoparticles not only surpass PAMAM-IO as drug delivery vectors, but can also act

successfully as platforms for magnetic targeting of cancer.

5.5 References

1. H. Arami, A. Khandhar, D. Liggitt and K. M. Krishnan, Chemical Society Reviews,

2015, 44, 8576-8607.

2. L. Garza-Ocanas, D. A. Ferrer, J. Burt, L. A. Diaz-Torres, M. Ramirez Cabrera, V. T.

Rodriguez, R. Lujan Rangel, D. Romanovicz and M. Jose-Yacaman, Metallomics,

2010, 2, 204-210.

3. A. K. Patri, A. Myc, J. Beals, T. P. Thomas, N. H. Bander and J. R. Baker, Bioconjugate

Chemistry, 2004, 15, 1174-1181.

4. Y.-M. Huh, Y.-w. Jun, H.-T. Song, S. Kim, J.-s. Choi, J.-H. Lee, S. Yoon, K.-S. Kim,

J.-S. Shin, J.-S. Suh and J. Cheon, Journal of the American Chemical Society, 2005,

127, 12387-12391.

5. Y.-W. Chen, G.-G. Liou, H.-B. Pan, H.-H. Tseng, Y.-T. Hung and C.-P. Chou,

International Journal of Nanomedicine, 2015, 10, 6997-7018.

6. J. Guo, H. Hong, G. Chen, S. Shi, Q. Zheng, Y. Zhang, C. P. Theuer, T. E. Barnhart,

W. Cai and S. Gong, Biomaterials, 2013, 34, 8323-8332.

7. X. Shi, T. P. Thomas, L. A. Myc, A. Kotlyar and J. J. R. Baker, Physical Chemistry

Chemical Physics, 2007, 9, 5712-5720.

Page 138: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

117

8. Y. Chang, Y. Li, X. Meng, N. Liu, D. Sun, H. Liu and J. Wang, Polymer Chemistry,

2013, 4, 789-794.

9. A. M. Hamilton, S. Aidoudi-Ahmed, S. Sharma, V. R. Kotamraju, P. J. Foster, K. N.

Sugahara, E. Ruoslahti and B. K. Rutt, Journal of Molecular Medicine, 2015, 93, 991-

1001.

10. Y. Huang, Y. Jiang, H. Wang, J. Wang, M. C. Shin, Y. Byun, H. He, Y. Liang and V.

C. Yang, Advanced Drug Delivery Reviews, 2013, 65, 1299-1315.

11. J. F. Kukowska-Latallo, K. A. Candido, Z. Cao, S. S. Nigavekar, I. J. Majoros, T. P.

Thomas, L. P. Balogh, M. K. Khan and J. R. Baker, Cancer Research, 2005, 65, 5317-

5324.

12. C. Alexiou and R. Jurgons, in Magnetism in Medicine, Wiley-VCH Verlag GmbH &

Co. KGaA, 2007, DOI: 10.1002/9783527610174.ch4h, pp. 596-605.

13. V. V. Mody, A. Cox, S. Shah, A. Singh, W. Bevins and H. Parihar, Applied

Nanoscience, 2013, 4, 385-392.

14. A. M. Nowicka, A. Kowalczyk, A. Jarzebinska, M. Donten, P. Krysinski, Z. Stojek, E.

Augustin and Z. Mazerska, Biomacromolecules, 2013, 14, 828-833.

15. O. Pourmehran, M. Rahimi-Gorji, M. Gorji-Bandpy and T. B. Gorji, Journal of

Magnetism and Magnetic Materials, 2015, 393, 380-393.

16. O. Pourmehran, T. B. Gorji and M. Gorji-Bandpy, Biomechanics and Modeling in

Mechanobiology, 2016, DOI: 10.1007/s10237-016-0768-3, 1-20.

17. T. Lunnoo and T. Puangmali, Nanoscale Research Letters, 2015, 10, 1-11.

18. J. L. Arias, L. H. Reddy and P. Couvreur, Journal of Materials Chemistry, 2012, 22,

7622-7632.

19. A. Hornung, M. Poettler, R. Friedrich, J. Zaloga, H. Unterweger, S. Lyer, J. Nowak, S.

Odenbach, C. Alexiou and C. Janko, Molecules, 2015, 20, 18016.

20. H. Kempe and M. Kempe, Biomaterials, 2010, 31, 9499-9510.

21. C. Alexiou, W. Arnold, R. J. Klein, F. G. Parak, P. Hulin, C. Bergemann, W. Erhardt,

S. Wagenpfeil and A. S. Lübbe, Cancer Research, 2000, 60, 6641-6648.

22. P. B. Johansen, Cancer Chemotherapy and Pharmacology, 5, 267-270.

23. W. N. Keith, P. Joseph Mee and R. Brown, Cancer Research, 1990, 50, 6841-6847.

24. M. M. Tomayko and C. P. Reynolds, Cancer Chemother Pharmacol, 1989, 24, 148-

154.

25. J. D. Jun Shao, Donald Lamm, David N. Weissman, Carl J. Malanga, Yongyut

Rojanasakul, Joseph K. H. Ma, Drug Delivery, 2001, 8, 71-76.

Page 139: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

118

Chapter 6

MR Contrast Properties of Dendritic Fe3O4 Nanoparticles

6.1 Introduction

Magnetic Resonance Imaging (MRI) is a powerful technique used widely in clinical

settings to image interiors of human body. It is a non-invasive, tomographic technique, without

the dangers of ionising radiation that offers good spatial resolution1, 2. MRI is based on the

principles of nuclear magnetic resonance (NMR) which can also provide us with the chemical

and physical information about molecules. The human body comprises of mainly fat and water

molecules, contributing approximately 63% hydrogen atoms, magnetic relaxation of which

generates the NMR signal. When the proton is placed in an external magnetic field, the nuclear

spin vector of the particle aligns itself with the external field (either parallel or antiparallel to

the field), just like any other magnet would. This alignment initiates a precession at a constant

frequency. In a simplistic sense, MRI is just the application of proton NMR to biological

systems, which yields intensity maps of water proton spin relaxation in tissues. Contrast in the

images generated by MRI, is the consequence of proton density and relaxation time constants

that vary throughout the tissues.

However, MRI suffers from relatively low sensitivity that limits its utility as it relies

exclusively upon the above mentioned inherent contrast mechanisms. This flaw can be

addressed using exogenous agents that influence local proton spin relaxation dynamics thereby

Page 140: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

119

enhancing the contrast and ability to distinguish the structures within the images. There are

several different types of contrast agents currently in use, and the two main classes are based

on chelated paramagnetic ions, such as gadolinium (Gd)3, 4, and superparamagnetic

nanoparticles like Fe3O45, 6. Particulate superparamagnetic contrast agents offer advantages

over their paramagnetic counterparts in both efficiency and mechanism of action. Henceforth,

Fe3O4 nanoparticles are discussed as an exemplary contrast agent for the superparamagnetic

nanomaterials. Paramagnetic chelates produce local effects that are mediated by exchange of

water protons, but Fe3O4 nanoparticles produce magnetic field gradients which results in the

changes in overall tissue susceptibility thereby affecting the contrast. These magnetic field

gradients impact a larger region without the need for direct contact with water flux. Many

factors make Fe3O4 excellent materials for developing targeted MRI contrast agents. They have

been known to naturally occur in many organisms and are biodegradable, exhibiting neither

acute nor chronic toxicity. These properties make them the only inorganic particulate contrast

agent currently approved for clinical settings. In clinical scenario, the contrast arises due to the

disproportionate accumulation of Fe3O4 nanoparticles resulting from the reduced or eliminated

reticulo-endothelial system (RES) function of pathologic versus healthy tissue. Once these

nanoparticles reach the tumour site, they extravasate through the porous endothelium (leaky

vasculature) and are retained by the tumour due to its compromised clearance mechanisms

(poor drainage). Relaxivity is an extrinsic property of these nanomaterials and is dependent on

a variety of factors7-9. These parameters can be modulated accordingly to improve their MR

performance and quality of the images in the clinical scenarios.

To meet this end, a variety of agents have been evaluated and used to encapsulate the

Fe3O4 nanoparticles to the region of interest for MRI. Jaiswal et al.10 have developed thermo-

responsive polymeric nanohydrogels to encapsulate Fe3O4 nanoparticles to enhance the

contrast properties exhibiting relaxivity rate of 173 mM-1s-1. An et al.11 synthesised Fe-Co

nanoalloy system modified with dextran molecules as sensitive contrast agents for MRI. These

nanoalloy systems had a relaxivity of 17.4 mM-1s-1 and gave a negative contrast in in vitro and

in vivo systems. Mandal and co-workers attempted to develop multimodal nanocarriers capable

of carrying therapeutic load in addition to their MR contrast properties12. They combined FITC

labelled Fe3O4 nanoparticles with antibody and gemcitabine drug to Hep2 cells. Yang et al.13

attempted to develop a dual-contrast agent by conjugating superparamagnetic silica-coated

Fe3O4 core-shell nanoparticles with gadolinium complex. This hybrid material was seen to be

water-dispersible, stable, and biocompatible while displaying both T1- and T2- relaxivities.

Page 141: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

120

Relaxivity measurements show that they have a T1 relaxivity (r1) of 4.2 mM−1s−1 and T2

relaxivity (r2) of 17.4 mM−1s−1, suggesting a possibility to use them as both T1 positive and T2

negative contrast agents. Fe3O4 nanoparticles have been commercialised as various

formulations, such as Ferrixan® (Bayer Schering Pharma AG) which shows a negative

relaxivity of 150 mM-1s-1 and Feridex® (AMAG Pharmaceuticals, Inc.) having a negative

relaxivity of 160 mM-1s-1.

This chapter deals with the use of the formulations discussed earlier (in second section

of chapter four of this thesis) as potential MR contrast agents. These nanoformulations have

been shown to successfully deliver doxorubicin to cancer cells and useful in magnetic

hyperthermia. To establish the multimodality of these nanoparticles in diagnostic imaging,

their performance as contrast agents in MR imaging was assessed. Also, as the working

environment of these nanoparticles would be hyperthermic (therapeutics), the dependence of

their MR relaxivity was studied under the conditions of varying buffer ions and temperatures.

6.2 Experimental Techniques

The samples prepared in section two of chapter five of this report (PAMAM–IO and

KR2–IO) were used to study and evaluate their comparative MR relaxivity properties. The

samples were suspended in 0.05% agarose gel (in various buffer solutions) and fixed. The

liquid microenvironments used were ultrapure water, phosphate buffer and simulated body

fluid (SBF). The temperatures inside the magnetic bore were maintained at 25, 37 and 45 ±0.5

°C by air blower placed near the bore and monitored digitally. The samples were scanned using

a multi-echo T2-weighted fast spin echo imaging sequence (TR/TE=3500/30, 45, 60, 75, 90

and 105 ms, slice thickness = 2 mm) by a 9.4T Agilent small animal scanner. For in vitro T2-

weighted MR imaging of HeLa cells, the cells were incubated with PAMAM–IO and KR2–IO

in the presence and absence of magnetic field. The magnetic field was applied and maintained

using a universal magnet plate. After incubation, the cells were gently washed by phosphate

buffered saline and fixed in 0.05% agarose gel for MR imaging.

6.3 Results and Discussions

6.3.1 Relaxivity studies

The T2-weighted MR images of PAMAM–IO and KR2–IO at different iron

concentrations were recorded at 9.4T. A significant signal attenuation was seen with increase

Page 142: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

121

in the iron concentration of the recorded phantoms. The relaxation rates showed a linear

increase with an increase in iron concentrations. At 25 °C, in ultrapure water, the r2 values of

PAMAM–IO and KR2–IO were comparable and were calculated to be 302 and 321 mM-1s-1

respectively (Figure 7.1). On increasing the temperature to physiological (37 °C) and then to

hyperthermia (45 °C) temperature, a gradual decrease in relaxivity is seen. The values

decreased to 177.4 and 178.5 mM-1s-1 for PAMAM–IO and KR2–IO respectively.

Figure 6.1 Plots of transverse relaxivity (r2) values of (a) PAMAM–IO and (b) KR2–IO in

ultrapure water

A similar trend was observed when these samples were evaluated in phosphate buffered

saline of pH 7.3 (Figure 7.2). At 25 °C, the r2 values of PAMAM-IO and KR2-IO were

comparable and were calculated to be 306 and 326 mM-1s-1 respectively. On increasing the

temperature to 45 °C, the values decreased to 149 and 183 mM-1s-1 for PAMAM-IO and KR2-

IO respectively. The difference between these values suggest that KR2-IO nanoparticles would

show improved performance in buffered environment than in pure water environment. The

relaxivity of these nanoparticles is sufficiently high to provide good MR contrast than the

PAMAM-IO nanoparticles.

Page 143: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

122

Figure 6.2 Plots of transverse relaxivity (r2) values of (a) PAMAM-IO and (b) KR2-IO in

phosphate buffered saline (pH 7.3) (c) phantom images showing substantial reduction in

transverse relaxation times of both the nanoparticles

The obtained phantom images showed a significant signal reduction with increasing

concentration of Fe from 0.001 to 0.25 mM (Figure 7.3). At 25 °C, in simulated body fluid

(SBF), the r2 values of PAMAM-IO and KR2-IO were comparable and were calculated to be

284 and 287 mM-1s-1 respectively. On increasing the temperature to physiological 37 °C, the

values decreased to 221 and 213 mM-1s-1 respectively. At 45 °C, a further reduction in

relaxivity is seen. The values now decreased to 134 and 165 mM-1s-1 for PAMAM-IO and KR2-

IO respectively.

Page 144: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

123

Figure 6.3 Plots of transverse relaxivity (r2) values of (a) PAMAM-IO and (b) KR2-IO in

simulated body fluid (pH 7.4) (c) phantom images showing substantial reduction in

transverse relaxation times of both the nanoparticles

This decrease in relaxivity with increase in temperature is due to the thermal activation and

movement of the water molecules14. At elevated temperatures the magnetic spins are also

disordered leading to reduction in magnetic strength of the Fe3O4 nanoparticles. This in

accordance with the results observed with PAMAM-IO and KR2-IO. Under the working

conditions of hyperthermia, i.e. 45 °C and in actual physiological conditions, KR2-IO assures

better MR performance than PAMAM-IO nanoparticles.

6.3.2 In vitro T2 weighted imaging

The T2 weighted images reveal that both the dendritic Fe3O4 nanoparticles are efficient

in providing improved contrast for imaging the HeLa cells (Figure 6.4). It was seen that the

presence of magnetic field and the duration of exposure to the magnetic field, during the

incubation, influenced the cellular uptake of these nanoparticles. The exposure of MF for

longer duration caused substantial increase in the amounts of nanoparticles internalised by the

cells (discussed in detail in chapter 5). This led to the observation that larger number of

Page 145: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

124

nanoparticles inside the cell provided better MR contrast as compared to the cells with lower

nanoparticle content.

Figure 6.4 T2-weighted MR images of HeLa cells with PAMAM-IO (a,c) and KR2-IO (b,d)

in the absence (a,b) and presence (c,d) of magnetic field

6.4 Summary

This chapter focussed on the evaluation of transverse relaxation times of PAMAM–IO

and KR2–IO nanoparticles and their MR contrast properties. It was seen that the transverse

relaxation time of these nanoparticles was greatly dependent on the ambient temperature. As

these dendritic nanoparticles are intended to be used in hyperthermic environment, their MR

contrast at 45 °C plays a critical role in their MRI applications. A decreasing trend in the

relaxivity values of the nanoparticles was observed when the temperature was increased from

ambient (25 °C) to physiological (37 °C) and then to hyperthermia (45 °C) temperatures. This

observation was attributed to the disordering in the water molecules and the magnetic moments

caused by the elevated temperatures. Despite the reduction, these nanoparticles were expected

to be efficient under physiological environment and high temperatures due to their high

transverse relaxivity. This was also confirmed by the T2 imaging of the cervical cancer cells.

6.5 References

1. M. S. Shiroishi, G. Castellazzi, J. L. Boxerman, F. D'Amore, M. Essig, T. B. Nguyen,

J. M. Provenzale, D. S. Enterline, N. Anzalone, A. Dörfler, À. Rovira, M. Wintermark

and M. Law, Journal of Magnetic Resonance Imaging, 2015, 41, 296-313.

Page 146: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

125

2. D. T. Ginat, B. Swearingen, W. Curry, D. Cahill, J. Madsen and P. W. Schaefer, Journal

of Magnetic Resonance Imaging, 2014, 39, 1357-1365.

3. W.-L. Zhang, N. Li, J. Huang, S.-F. Luo, M.-X. Fan, S.-Y. Liu, B. Muir and J.-H. Yu,

Journal of Applied Polymer Science, 2011, 121, 3175-3184.

4. C.-H. Huang, K. Nwe, A. Al Zaki, M. W. Brechbiel and A. Tsourkas, ACS Nano, 2012,

6, 9416-9424.

5. X. Wang, D. Niu, Q. Wu, S. Bao, T. Su, X. Liu, S. Zhang and Q. Wang, Biomaterials,

2015, 53, 349-357.

6. D. Bates, S. Abraham, M. Campbell, I. Zehbe and L. Curiel, PLoS ONE, 2014, 9,

e97220.

7. H. Duan, M. Kuang, X. Wang, Y. A. Wang, H. Mao and S. Nie, The Journal of Physical

Chemistry C, 2008, 112, 8127-8131.

8. G. M. Nicolle, É. Tóth, H. Schmitt-Willich, B. Radüchel and A. E. Merbach, Chemistry

– A European Journal, 2002, 8, 1040-1048.

9. M. Benmelouka, A. Borel, L. Moriggi, L. Helm and A. E. Merbach, The Journal of

Physical Chemistry B, 2007, 111, 832-840.

10. M. K. Jaiswal, M. De, S. S. Chou, S. Vasavada, R. Bleher, P. V. Prasad, D. Bahadur

and V. P. Dravid, ACS Applied Materials & Interfaces, 2014, 6, 6237-6247.

11. L. An, Y. Yu, X. Li, W. Liu, H. Yang, D. Wu and S. Yang, Materials Research Bulletin,

2014, 49, 285-290.

12. A. Mandal, S. Sekar, M. Kanagavel, N. Chandrasekaran, A. Mukherjee and T. P. Sastry,

Biochimica et Biophysica Acta (BBA) - General Subjects, 2013, 1830, 4628-4633.

13. H. Yang, Y. Zhuang, Y. Sun, A. Dai, X. Shi, D. Wu, F. Li, H. Hu and S. Yang,

Biomaterials, 2011, 32, 4584-4593.

14. T. Kawaguchi, A. Yoshino, M. Hasegawa, T. Hanaichi, S. Maruno and N. Adachi,

Journal of materials science. Materials in medicine, 2002, 13, 113-117.

Page 147: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

126

Chapter 7

Conclusions and future scope

7.1 Conclusions

The nanoparticle platforms have emerged as promising drug delivery vehicles for

carrying a variety of cargo in cancer therapeutics. Among a vast variety of nanoparticulate

systems, very few could be actively used in biomedical applications because of their poor

aqueous stability, off-site toxicity, poor biocompatibility and toxic degradation profile. Fe3O4

nanoparticles have been thoroughly and critically evaluated for biomedical applications.

Recent works have also established dendrimers as promising nanocarriers as they show

improved properties and efficiencies when compared to the classical polymers. To improve the

aqueous stability of Fe3O4 nanoparticles, a variety of organic molecules are used to engineer

their surface which also plays an important role in linking to the cargo molecules. Towards this

end, we evaluated surface functionalised Fe3O4 nanoparticles for their ability to carry and

deliver doxorubicin to cancer cells. These DOX-loaded nanoparticles (loading efficiency, 90%,

w/w) were stable, biocompatible with good specific absorption rate. They exhibited a pH-

dependent release pattern and released DOX in appreciable amounts in mild acidic

environment. As most of the DOX molecules were bound to the surface of the nanoparticles,

their release was uncontrolled as they were vulnerable to the change in pH in their immediate

environment. This could lead to undesirable, off-site release of DOX. To circumvent this

potential drawback, the Fe3O4 nanoparticles were surface modified with PAMAM dendrimers.

Page 148: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

127

The modification of Fe3O4 nanoparticles by different generations of PAMAM (G 3, 5

and 6) improved their material properties which were thoroughly characterised. Their

capability to carry both cationic (DOX) as well as anionic (EGCG) drugs was evaluated. It was

seen that the drug loading capacity of PAMAM-Fe3O4 increased with increase in the generation

of the dendrimer. It could be explained by the fact that as the generation of dendrimer is

increased, the surface functional groups also increase along with the size of their internal

cavities. The cargo molecules are known to be bound either by the surface groups or

encapsulated in the cavities. Thus, the results indicated that higher generations of dendrimer

were more efficient in carrying as well as releasing the drug molecules as compared to the

lower generations. Overall, these nanosystems showed no loss of cell proliferation activity with

the mouse fibroblast (L929), human cervical carcinoma (HeLa), oral carcinoma (KB), prostate

cancer (PC-3) and human breast cancer (MCF-7) cells. But it was noted that the higher

generations of the PAMAM dendrimer showed noticeable amount of toxicity towards cells and

was not entirely biocompatible. PAMAM is the most studied and widely used dendrimer in

various biomedical applications due to its excellent physico-chemical properties and tailorable

architecture. It application in clinical settings is limited by its compromised biocompatibility,

which is a limiting factor and thus raises a demand of improved biocompatibility. Thus, we

synthesised a biocompatible peptide dendrimer that had amide interiors and amine exteriors

(chemically similar to PAMAM). The structural characterization of peptide dendrimer was

carried out by various sophisticated techniques as nuclear magnetic resonance (NMR), FTIR

and X-ray photoelectron spectroscopy (XPS). Since amino acids were used as branching units,

the peptide dendrimer was found to be biocompatible even at higher concentrations (20 mg/ml).

When the performances of as-prepared peptide dendrimer were compared to PAMAM, it was

seen that PAMAM was still more efficient in conjugating with Fe3O4 nanoparticles. The KR2–

IO nanoparticles exhibited similar drug loading efficiency as the PAMAM–IO, however, their

drug release capacity was significantly improved. The comparative evaluation of these

dendritic nanoparticles for drug delivery, magnetic hyperthermia and combinatorial therapy

established that peptide dendrimer have an edge over the PAMAM dendrimers. Also the

physico-chemical properties of peptide dendrimer were minimally compromised as compared

to its PAMAM counterpart.

After validating the in vitro efficacy of PAMAM–IO and KR2–IO nanoparticles, their

in vivo investigation in a subcutaneous syngeneic murine melanoma model was performed. It

was seen that both the dendritic nanoparticles were haemocompatible and had no adverse

Page 149: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

128

effects on the normal functioning of the body. Both the dendritic nanoparticles showed a

tendency to localise in lungs, liver and spleen which was responsible for their rapid clearance

from the systemic circulation. These Fe3O4-based nanosystems also have the advantage of

being responsive to external magnetic field while circulating in the bloodstream. This was

exploited as magnetically targeting these nanoparticles to the tumour site. Approximately 60-

fold increase in the accumulation of iron in the tumour region was observed when compared to

the tumour of mice which were not exposed to the magnetic field. This high localisation of

DOX-loaded nanoparticles led to high concentrations of DOX in the tumour and thus the rate

of tumour regression was faster than other mice groups. It was also seen that lower number of

doses were sufficient to suppress the tumour growth in combination with magnetic field than

that required without the magnetic field. Though the tumour was completely supressed by 20th

day, it reappeared after 35 days of treatment. This necessitated the revision of parameters for

improved therapeutic efficacy of these nanoparticles.

Furthermore, the dendrimer functionalized Fe3O4 nanoparticles were also seen to be

MR active and showed high transverse relaxivities. Various physico-chemical parameters

potentiate and affect their MR contrast properties. A decreasing trend in the relaxivity values

of the nanoparticles was observed when the temperature was increased from ambient (25 °C)

to physiological (37 °C) and then to hyperthermia (45 °C) temperatures. Despite this decreasing

behaviour, the relaxivity of KR2–IO in simulated body fluid at 45 °C show optimally high value

of 165 mM-1s-1. In vitro T2 weighted MR imaging of dendrimer functionalized Fe3O4 treated

(for various treatment times) HeLa cells showed increased contrast when compared to the

untreated cells. Owing to the promising drug delivery performances, high magnetisation, high

specific absorption rate and shorter transverse relaxation times, the dendrimer functionalized

Fe3O4 nanoparticles not only prove to be suitable delivery vectors, but can also act as anchors

for passive targeting and contrast agents for MR imaging and multimodal cancer theranostics.

7.2 Future scope

The studies presented in this report demonstrate the potential of peptide dendrimer-

coated Fe3O4 nanoparticles in drug delivery, hyperthermia, combinatorial therapy and in vitro

MR imaging studies against PAMAM coated nanoparticles. This also opens up avenues of

using peptide dendrimer-coated Fe3O4 nanoparticles in a variety of other biomedical

applications.

Page 150: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

129

(a) Higher generations of this peptide dendrimers could be evaluated for their biomedical

applications. It is expected that higher generations will prove better vehicles as compared

to the lower generations.

(b) These dendritic nanoparticles could be used in combining chemotherapy and MDT with

magnetic hyperthermia for tumour model in small animals and if possible taken up further

to clinical settings.

(c) Attachment of a targeting ligand or antibody to these formulations might add on to the

specificity and improved efficacy of these formulations in addition to their magnetic

targeting.

(d) Initial MR imaging studies reveal their potential for in vivo tumour imaging in animal

models.

(e) These dendrimer-coated nanoparticles could also be used as platforms for a variety of

other biomedical applications such as cell sorting and separation, immunodetection,

electrochemical sensing towards early detection of cancer etc.

Page 151: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

This chapter has been published in J. Nanopharma. Drug Del., 2013, 1, 365-375.

Appendix I

Thermally-activated delivery of curcumin using magnetic

liposomes

AI.1 Introduction

Chemotherapy involves the use of chemicals to damage DNA, RNA and proteins in order

to trigger cell cycle arrest or apoptosis in cancer cells. However, such agents generally induce

apoptosis in both cancer and normal cells1. Hence, it is highly desirable to develop a drug delivery

system that has maximum chemotherapeutic efficiency and minimal side effects. An approach

toward the development of such a drug delivery system is the combining of a chemical that

selectively induce apoptosis in targeted cancer cells, with a delivery vehicle that can release the

drug to the targeted tumour in a controlled manner. Curcumin (diferuloylmethane, a natural,

hydrophobic polyphenol, and the primary constituent of the rhizome of turmeric) is one of the few

agents that selectively induce apoptosis in highly proliferative cells. Cellular apoptosis induced by

curcumin is significantly higher in cancer cells than in non-cancerous cells. Curcumin first

attracted attention for its antioxidant, anti-inflammatory2, antimicrobial3, 4 and antineoplastic

activities5, 6. The efficacy of curcumin as a therapeutic agent in cancer was reported later5, 7, 8.

Curcumin has since been reported to increase apoptotic death, control cell proliferation and down-

Page 152: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

131

regulate the oncogenic phenotype by controlling the signalling cascades involved in the cell cycle9,

10. However, the low aqueous solubility, poor chemical stability and lipophilic nature of curcumin

have limited its oral bioavailability and delivery efficiency to targeted cancer sites and have, thus,

impaired its anticancer therapeutic potential.

Certain types of polymer nanoparticles have been used in the encapsulation of curcumin in

order to enhance its chemical stability11-15; poly(lactic-co-glycolic) (PLGA) is the most studied

among these polymer nanoparticles. PLGA is a biodegradable polymer which generates lactic and

glycolic acid as degradation products, which are metabolised by cells through the Krebs cycle.

Although PLGA nanospheres as vehicles can enhance the delivery of curcumin when compared to

curcumin alone, the amphiphilic nature of the polymer results in low levels of encapsulation, which

results in a need for the administration of higher dosages in order to achieve pharmaceutical

activity at the targeted site14, 16-18. Saccharide-based nanoparticle vehicles have also been used to

deliver curcumin for oral administration in order to enhance the release efficiency. However, the

fast water solubility of saccharide compromises the chemical stability of the drug system19. In

short, the delivery vehicles mentioned above have failed to address the current issues that are

associated with the curcumin delivery.

Liposomes, which are bilayer vesicles of lipid molecules, have been regarded as promising

alternatives to polymeric systems20-22. The amphiphilic nature of lipid molecules causes them to

form a closed vesicular structure in aqueous solutions, with the apolar regions being oriented away

from the aqueous phase and the polar regions being in contact with the water. The hydrophilic

surface of liposomes renders them highly water soluble, while the amphiphilic nature of the lipid

bilayer facilitates the anchoring of both hydrophilic and lipophilic molecules. A liposomal carrier

that is made up of soy phosphatidylcholine and 1,1-diphenyl-2-picrylhydrazyl has been reported

to enhance the antioxidant (therapeutic) effects of the encapsulated curcumin23. However, it

remains to be elucidated whether the liposome carrier affects the anticancer properties of

curcumin. Therefore, an objective of the present work was to investigate the curcumin-

encapsulation efficiency, curcumin-releasing profile and the anticancer therapeutic effect of

curcumin when delivered by liposomes. Another advantage of liposomes is that they can release

encapsulated drugs under external stimuli such as a changing pH or temperature, or ultrasound

waves, allowing control over the release profile and the pharmacokinetics of the drug molecules.

Among these stimuli, a changing temperature is clinically the most feasible because it is virtually

Page 153: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

132

impossible to control the pH values in the body, and ultrasound has the potential to damage the

phospholipid membranes of healthy cells while destroying the liposomes. In fact, hyperthermia

treatment is already used in some forms of cancer treatment, especially in solid tumours, in order

to kill cancer cells directly or to make them more sensitive to radiation and certain anticancer

drugs24, 25. There are a number of techniques to deliver heat remotely, which include infrared

sources, focused microwaves, magnetic fields and infusions of warmed liquids26-28. In the case of

magnetic hyperthermia, nanoparticles can be subjected to an alternating magnetic field, which

induces electron flow in the nanomaterial, thereby producing localised heat. To use magnetic

hyperthermia to control the release of curcumin from liposomes was another objective of this work.

Superparamagnetic Fe3O4 nanoparticles have been employed in hyperthermia treatment24,

29, 30, and as drug delivery vectors31, 32 such as curcumin delivery17, 18. When compared with other

ferrite systems (iron, cobalt, manganese and nickel), superparamagnetic Fe3O4 nanoparticles have

shown greater potential due to their better aqueous stability and biocompatibility33. These

nanomaterials also exhibit a relatively large surface-area-to-volume ratio due to their small size.

The downside of superparamagnetic Fe3O4 nanoparticles is that they have a high surface energy,

which leads to a strong tendency to agglomerate in colloidal suspensions via van der Waals forces

and magnetic dipole–dipole interactions. This agglomeration necessitates colloidal stability of

these nanoparticles in aqueous suspensions for the biomedical applications mentioned above.

Surface engineering of Fe3O4 nanoparticles for biomedical applications has also been performed

using a variety of organic molecules34, 35, in addition to polymers36, 37 and polysaccharides38, 39.

This results in bioactive surfaces that can be used to anchor different molecules of interest, which

will enhance their colloidal and surface chemistry35. Small organic molecules of biological origin

have been considered to be promising candidates for nanoparticle surface engineering because

their biocompatible degradation products minimise the risk of toxicity to biological systems.

Saccharides and oligosaccharides, for example, readily degrade into sugar monomers, which are

actively metabolised by cells. Due to their colloidal solubility, amino acids40, monosaccharides38,

and oligosaccharides39 are three of the most frequently used small organic molecules.

Oligosaccharides such as dextran and β-cyclodextrin, have been reported to significantly improve

the colloidal stability of nanoparticles41, 42. Monosaccharides are also well known for their ease of

conjugation with various biomolecules such as proteins, which forms peptidoglycans and

proteoglycans, or with lipids, which forms glycolipids and aminoglycans. Given the above

Page 154: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

133

properties, dextrin was selected as the surface engineering moiety for the Fe3O4 nanoparticles in

the present study.

After being surface engineered with small organic molecules, magnetic nanoparticles can

be further modified by various macromolecules43, polymers44 and biomolecules45 in order to

improve their recognition in biological systems and to lower their immunogenicity46. In this

respect, lipids47, 48 have an advantage over other macromolecules because they can aid in cellular

uptake of nanoparticles by facilitating transport across the phospholipid membranes of cells.

Fabrication of magnetic liposomes (MLs) by coating magnetic nanoparticles with a lipid bilayer

has been explored for the possible delivery of a number of drugs, except for curcumin45. In

summary, the objectives of this chapter are to fabricate and characterise an ML formulation, to

evaluate its curcumin encapsulation and delivery performance, and finally, to assess the anticancer

effects of the ML/curcumin formulation within cancer cells.

AI.2 Experimental Techniques

AI.2.1 Synthesis of Fe3O4 Nanoparticles and Magnetic Liposomes

To synthesise the Fe3O4 nanoparticles, 4.44 g of ferric chloride and 1.732 g of ferrous

chloride were dissolved in 80 ml of ultrapure water, and the temperature was increased to 60 °C

under a nitrogen atmosphere with mechanical stirring at 1000 rpm. The temperature was then held

at 70 °C for 30 min, which was followed by the addition of 30 ml of ammonia solution and the

maintenance of the temperature at 70 °C for another 30 min. Finally, 10 ml of an aqueous solution

of dextrin (0.07 g/ml) was added to the reaction mixture and the temperature was raised to 90–95

°C under reflux and maintained for 90 min with continuous stirring. A black precipitate of dextrin-

coated Fe3O4 nanoparticles (Dx–Fe3O4) was obtained and was washed thoroughly with ultrapure

water. During each wash step, the precipitate was separated from the supernatant through the use

of a permanent magnet.

The MLs were prepared by a thin-film hydration technique that was reported previously45.

In a typical synthesis, 200 mg of soy-PC was dissolved in a solvent mixture of

chloroform/methanol (2:1 v/v). The solvent was evaporated under vacuum (120 mbar) at 35 °C in

a rotary evaporator in order to form a thin and uniform lipid film on the walls of the round-

Page 155: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

134

bottomed flask. In order to optimise the lipid-to-nanoparticle ratio, the lipid film was hydrated

with varying amounts of Dx–Fe3O4 nanoparticles (20, 40, 60, 80, 100, 150 and 200 µg) using a

water bath-type sonicator for approximately 30 min. The temperature was maintained below 40

°C until the lipid film was transferred to the aqueous suspension, yielding the MLs. Control

liposomes were synthesised by hydrating the lipid film in PBS (pH 7.34). A lipid-to-nanoparticle

ratio of 10:3 was used for the drug delivery vesicle because it showed the maximum encapsulation

efficiency of the Dx–Fe3O4 nanoparticles and the most stable aqueous suspension.

AI.2.2 Analysis of Drug Loading and Release

The absorbance spectra of curcumin were studied in order to investigate the loading

efficiency of curcumin. In a typical loading experiment, 200 mg of soy-PC was dissolved in a

solvent of chloroform/methanol (2:1 v/v), which contained varying amounts (20, 40, 60, 80, and

100 µl) of curcumin (stock solution - 1 mg/ml in chloroform) and had ML-to-curcumin ratios of

10, 5, 3.33, 2.5 and 2. The solvent was evaporated under vacuum (120 mbar) at 35 °C in a rotary

evaporator in order to form a thin and uniform lipid film on the walls of the round-bottomed flask.

The lipid film was then hydrated with a previously optimised amount (lipid-to-nanoparticle

ratio=10:3) of Dx–Fe3O4 nanoparticles under ultrasonication for approximately 30 min. The

temperature was kept below 40 °C until the lipid film was transferred to the aqueous suspension

to yield the drug-loaded MLs. The absorbance spectrum of the supernatant (after magnetic

sedimentation of the curcumin-loaded MLs) was recorded with a UV-Vis spectrophotometer in

order to determine the amount of the drug loaded into the MLs. The curcumin-loaded MLs were

re-suspended in water and the loading efficiency (w/w%) was calculated as follows:

% 𝐿𝑜𝑎𝑑𝑖𝑛𝑔 𝑒𝑓𝑓𝑖𝑐𝑖𝑒𝑛𝑐𝑦 = 𝐴𝐶𝑢𝑟− 𝐴𝑆𝑢𝑝

𝐴𝐶𝑢𝑟 × 100 (Eqn. AI.1)

where ACur is the absorbance of pure curcumin and ASup is the absorbance of the supernatant.

The drug release study was carried out at temperatures of 37 °C and 45 °C. The amount of

curcumin released from loaded MLs was quantified on the basis of the loading efficiency. The

curcumin-loaded MLs (10 mg) were suspended in PBS at pH 7.3 and placed in a dialysis bag.

Dialysis was performed with 200 ml of PBS (pH 7.3) under continuous stirring at a physiological

temperature (37 °C) and at a hyperthermic temperature (45 °C). At different time intervals, aliquots

Page 156: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

135

were withdrawn and replaced with fresh PBS. The fluorescence intensity of these aliquots was

analysed at λex= 425 nm and λem=540 nm. Drug release was calculated from the fluorescence

intensity of the aliquots according to a standard curve prepared under similar conditions. The

thermos-sensitivity of these curcumin-loaded MLs was evaluated by the calcein release assay,

which utilizes the fluorescence property of calcein49. At a high concentration, calcein shows self-

quenching, resulting in a decrease in fluorescence, and an increase, which is observed when calcein

is released from the liposomal carrier into the surrounding medium. To establish the effect of pH

sensitivity on the release of the drug by the curcumin-loaded MLs, similar experiments were

performed with a sodium acetate buffer (pH 4.8) as a stimulus at 37 °C. The results showed that

negligible amounts of the drug were released from the ML system at the decreased pH, thereby

establishing the independence of pH sensitivity on the curcumin-loaded MLs.

Time-dependent calorimetric measurements to evaluate the heating ability of the Dx-Fe3O4

suspensions were performed using a radio frequency generator. A total of 1 ml (10 mg/ml of iron)

of the Dx-Fe3O4 colloidal suspension was placed in an AC magnetic field (7.64, 8.82, 9.41 and

10.0 kA/m) with a fixed frequency of 425 kHz and with arrangements to minimise heat loss. The

specific absorption ratio (SAR) was calculated using eqn. 3.2.

AI.2.4 Evaluation of Biocompatibility and Therapeutics

The biocompatibility of the MLs was established with a mouse fibroblast cell line (L929)

and cervical cancer cell lines (HeLa). The toxicity of the curcumin-loaded MLs was evaluated with

cervical cancer cell lines (HeLa) by an SRB colorimetric assay50. In order to establish the potential

of these carriers in delivering curcumin, a dose-dependent study was undertaken to evaluate the

50% inhibitory concentration(IC50) values of free curcumin and the curcumin-loaded MLs over 48

h. The cells were seeded in 96-well plates at a cell density of 2×104 cells per well and incubated

in a tissue culture medium for 24 h at 37 °C in a 5% CO2 environment. After 24 h, different

concentrations of the MLs (2, 1, 0.5, 0.25, 0.125, 0.0625 and 0.03125 mg/ml) and the curcumin-

loaded MLs (10, 8, 4, 2, 1, 0.5, 0.25, 0.125, 0.0625 and 0.03125 mg/ml) were mixed with the

growth media, and the cells were incubated for an additional 24 h(MLs) or 48 h (curcumin-loaded

MLs). After 24 h (MLs) or 48 h (curcumin-loaded MLs), the cells were carefully washed with PBS

(pH 7.3), and an SRB assay was performed in order to determine cell viability. For the assay, cells

Page 157: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

136

were fixed with cold 10% trichloroacetic acid (at 4 °C) and stained with 0.4% SRB (in 1% acetic

acid). After one hour of incubation in the dark, the unbound dye was thoroughly washed with 1%

acetic acid, and the cell-bound dye was later extracted with 10 mM Tris buffer (pH 10.5).

Absorbance was recorded at 560 nm using a Thermo Scientific Multiskan EX multiplate reader.

The relative cell viability was calculated according to eqn. 3.3.

AI.3 Results and Discussion

AI.3.1 Characterisation of the Synthesised Nanoparticles

The characterisation of the prepared nanomaterials was undertaken as described in section

2.2.5. The crystalline structure and crystallite size of the Dx–Fe3O4 nanoparticles were investigated

by powder XRD (Figure AI.1a). The corresponding diffraction planes of the indices showed good

agreement with the reported values for magnetite (JCPDS Card No. 19-0629, a = 8.3967 Å). The

XRD pattern revealed the formation of single-phase magnetite, which has an inverse spinel

structure with a crystallite size of ∼8.48 nm, as calculated by the Scherrer formula. A high degree

of crystallinity of the nanoparticles was indicated by the presence of the sharp and intense peaks

of the Dx–Fe3O4 nanoparticles.

Figure AI.1 (a) XRD pattern of Dx–Fe3O4, TEM micrographs of (b) Dx–Fe3O4 (inset shows the

selected area diffraction pattern of Dx–Fe3O4 and (c) magnetic liposomes (inset shows magnified

image of MLs)

Figure AI.1b shows the TEM micrograph of the Dx–Fe3O4 nanoparticles. The Dx–Fe3O4

nanoparticles were mostly spherical and smaller than 15 nm in diameter. The inset in figure AI.1b

shows the electron diffraction pattern of a selected area. The pattern, which has been indexed with

Page 158: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

137

the inverse spinel magnetite crystal structure, is consistent with the XRD results. Figure AI.1c

shows the morphology of the MLs. The inner and outer diameters of the MLs averaged ∼100 and

∼200 nm, respectively, with a bilayer thickness of 50 nm. The MLs were stable and well dispersed

in the aqueous solution (the inset shows an image of an ML at a scale of 50 nm). The bilayers were

formed uniformly. The intact liposome is spherical and has varying amounts of Fe3O4 encapsulated

in its hydrophilic core.

Figure AI.2a shows the FTIR spectra of pure dextrin and the Dx–Fe3O4 nanoparticles. The

spectrum of dextrin is well resolved and contains a few broad bands in addition to narrow bands.

The very broad peak at 3297 cm−1 is characteristic of the aromatic sp2 C–H stretch and is attributed

to the pyranose ring vibrations of dextrin. Characteristic peaks of the α-D-glucose units of the

polysaccharide were visible at 1203 cm−1, which occur due to the in-plane C–H and O–H

vibrations51. The multiple bands that appear in the region between 1150 and 930 cm−1 coincide

with the in-plane C–H bending vibrations of the pyranose ring and the C–O stretching vibrations.

The vibrational bands present at 1150 and 1077 cm−1 are attributed to valent vibrations of the C–

O–C bond of the glycosidic bridge, whereas, the peak at 1025 cm−1 is due to the substantial chain

flexibility of dextrin around the glycosidic bonds. The vibrational bands of Dx–Fe3O4 are relatively

broad and are fewer compared with those of pure dextrin. The peaks that are present in the

spectrum of dextrin at 1150 cm−1, 1077 cm−1 and 1025 cm−1 were also present in the spectrum of

Dx–Fe3O4, indicating the successful coating of dextrin on the surface of the nanoparticles. Other

peaks are interpreted as follows: The small and narrow bands around 3000 cm−1 are due to the

presence of H-bonded –OH groups from water molecules, which are physically adsorbed on the

surface of the Dx–Fe3O4 that is suspended in an aqueous medium. A new peak at 3009 cm−1 is

visible in the O–H stretch region. This may be the result of the formation of hydrogen bonds

between the free –OH groups on dextrin and the oxygen from the Fe–O core of the magnetite

nanoparticles, which results in the formation of an iron-oxy-hydroxide monodentate coordination

complex. The peaks at 2934 and2986 cm−1 present in the dextrin spectrum are retained in the Dx–

Fe3O4 spectrum, indicating that the C–H bonds were unaltered during the conjugation and did not

play a role in the coating process. The appearance of the peak at1660 cm−1 indicates the retention

of the cyclic ring structure of dextrin after its conjugation with the Fe3O4 nanoparticles. As seen in

figure AI.2a, the peaks at 1365 and 1417 cm−1 in dextrin shift to 1400 and 1535 cm−1 in the Dx–

Fe3O4 spectrum. This might be due to the formation of O–H bonds in the iron-oxy-hydroxide

Page 159: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

138

complex and the overlap with C–H bending vibrational bands of the dextrin molecules. The

appearance of the peak at 864 cm−1 (boat conformation) and the disappearance of the peak at 930

cm−1 (chair conformation) could be attributed to the loss of the chair conformation or to a

conformational change to the boat conformation of the glucopyranose units after interaction with

the surface of the Fe3O4. The peak at 575 cm−1 can be attributed to the stretching vibrational modes

of Fe–O.

Figure AI.2 FTIR spectra of (a) dextrin and Dx–Fe3O4 nanoparticles (b) MLs and Cur-MLs

Figure AI.2b shows the FTIR spectra of the soy-PC, MLs, curcumin and curcumin-loaded

MLs. The spectrum of soy-PC reveals characteristic vibrational bands consistent with previously

reported studies52. The peak at 1739 cm−1 is due to C=O vibrations of the fatty acid chains, and the

bands at 1643, 1460 and 1232 cm−1 are due to C–NH3 symmetric scissoring of the choline entity

of lipid, C–H scissoring and P=O vibrations of the phosphatidyl group, respectively. The shift in

the vibrational bands at 1739 cm−1 to 1737 cm−1 in the spectrum of the MLs is indicative of the

interaction of the C=O groups of soy-PC with the Dx–Fe3O4 nanoparticles by either electrostatic

or van der Waals interactions. The bands at 1643 and 1460 cm−1 do not exhibit any shifts, pointing

to the non-participation of the –NH3 groups of the fatty acid in any type of bond formation with

the Dx–Fe3O4 nanoparticles. The band representing the P=O of the choline group of soy-PC (1232

cm−1) exhibits a minor shift (1234 cm−1), indicating the interaction of the group with the free –OH

groups present on the Dx–Fe3O4 nanoparticles. The overlap of soy-PC vibrations on dextrin masks

the dextrin frequencies in the spectrum of the MLs.

Page 160: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

139

The FTIR spectrum of curcumin-loaded MLs shows sharp, intense characteristic peaks of

soy-PC due to the high ratio of ML to curcumin. Characteristic peaks of curcumin are also present

in the FTIR spectrum of curcumin-loaded MLs, which are in good agreement with the vibration

spectrum of curcumin, which was reported by Mohan et al53. According to their interpretations,

the band at 3502 cm−1 is due to phenolic –OH stretching vibrations, the peak at 1427 cm−1 is

attributed to olefinic bending vibration of the C–H bound to the benzene ring of curcumin, the

peak at 1272 cm−1 due to an enol vibration (C=O), the band at 958 cm−1 due to benzoate trans –

CH vibrations, and the vibrations at 709 cm−1 due to the cis-CH of an aromatic ring. These results

further indicate the successful loading of curcumin into the MLs. The successful conjugation of

curcumin to the MLs was indicated by the shift of some peaks in the FTIR spectrum of curcumin-

loaded MLs, as discussed as follows. A shift observed in the band at 1737 cm−1 in MLs to 1741

cm−1 in the curcumin-loaded MLs is attributed to the attachment of the drug to the –COO groups

of the fatty acid. Other bands shifted from 1643 to 1651 cm−1, 1460 to 1457 cm−1, 1234 and 1232

to 1238 cm−1 and 1056 to 1067 cm−1 in the spectrum of the curcumin-loaded MLs. The shift in

these bands indicates that the –CH3 side chains and the free –OH groups may be involved in the

conjugation of curcumin to the MLs, confirming the drug loading into the MLs. These results

confirm the conjugation of dextrin onto the Dx–Fe3O4 nanoparticles, encapsulation by the lipid

bilayer and the subsequent conjugation of curcumin.

TGA-DTA analysis was conducted to determine the decomposition profile of the

formulations and the relative mass of dextrin (Figure AI.3a) and liposome (Figure AI.3b) coatings.

The thermal profile of the Dx–Fe3O4 nanoparticles revealed that the degradation of the

nanoparticles occurred in three primary steps. Initially, a weight loss of 3.5% occurred, which

corresponded to the endothermic DTA peak at 80 °C. This loss could be attributed to the water

molecules which were superficially adsorbed by the dextrin molecules. A gradual 5.1% loss in the

weight of the nanoparticles was then observed in the temperature range of 100–430 °C, which

corresponded to two DTA peaks at 285 and 400 °C, respectively. Dextrin molecules underwent a

decomposition step at 320 °C. Thus, this observed decrease in weight could be due to the surface-

conjugated dextrin molecules54. The third weight loss step of 0.4% was observed at 570 °C, which

had a corresponding sharp exothermic DTA peak. This decrease occurs due to the loss of FeO

molecules during degradation of magnetite to maghemite.

Page 161: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

140

Figure AI.3 Thermal degradation profiles of (a) Dx–Fe3O4 and (b) magnetic liposomes

The thermogram of MLs indicated that weight loss occurred in three distinct steps. In the

first step, an initial weight loss of approximately 3.7% occurred, which corresponded to a small

endothermic DTA peak at 60 °C due to the removal of physically absorbed water. In the second

step, a steep decline in the curve was observed. This drop in the TGA plot represents a weight loss

of approximately 9.0% and has a corresponding broad exothermic DTA peak at 280 °C, again due

to the removal of the organic dextrin and lipid molecules by thermal decomposition from the

surface of the Fe3O4 nanoparticles. This large and well-defined weight loss may be due to the

larger surface concentration and high molecular weight of the molecules, which were made up of

carbon, oxygen, hydrogen and phosphorus. In the third step, a negligible weight loss of

approximately 0.3% occurs. There is a corresponding exothermic DTA peak at 600 °C due to the

removal of iron (II) oxide (FeO) during the phase transformation of magnetite to maghemite.

Hence, the total weight loss percentage was approximately 13.7%. This mass loss represents the

weight percentage of organic coatings on the surface of the Fe3O4 nanoparticles and the

encapsulating liposome bilayer.

The colloidal stability and dispersion of the nanoparticles is associated with the electric

charge of the particle surface. Figure AI.4 shows the zeta potential of the Dx–Fe3O4 and MLs at

different pH values, indicating that the conjugation of the dextrin molecules onto the surface of

the Fe3O4 nanoparticles creates a highly negative surface charge with an isoelectric point at pH 5.

These zeta potential values confirm the presence of negatively charged dextrin groups on the

surface of the Fe3O4 nanoparticles. The addition of the lipid bilayer to the Dx–Fe3O4 lowered the

surface charge of the MLs and shifted the isoelectric point to pH 4.5. At pH 7.0, the MLs showed

a negative surface charge of approximately −30 mV, which is highly desirable, as it ensures high

Page 162: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

141

aqueous stability of the MLs in a physiological environment before reaching the targeted cancer

cells. The dynamic light scattering (DLS) measurements on the MLs (Inset of Figure AI.4) showed

that the MLs had a mean hydrodynamic diameter of 1.2 µm, which can be attributed to the presence

of the water that is associated with the hydrophilic heads of the lipid layer.

Figure AI.4 Zeta potential of Dx–Fe3O4 and MLs as a function of pH. Inset shows the

hydrodynamic diameter of the MLs obtained from DLS measurements (1.2 µm)

Figure AI.5a shows the field-dependent magnetisation (M versus H) plot of the Dx–Fe3O4

nanoparticles and the MLs at room temperature. Both exhibited superparamagnetic behaviour, that

is, zero magnetic hysteresis and remanence. The maximum magnetisation of the Dx–Fe3O4 and

the MLs was 54.2 emu/g and 24.3 emu/g, respectively, at a field of 20 kOe. The observed

magnetisation of Dx–Fe3O4 is comparable to that (approximately 60 emu/g) of bare Fe3O4

nanoparticles that have been reported in previous work55. Dextrin chains with more than 10 glucose

units tend to adopt a helical structure in an aqueous solution that contains cavities that easily form

an inclusion complex with low molecular weight compounds such as Fe3O4 nanoparticles and

ions56. These helical cavities may interfere with the domain alignment of the Fe3O4 nanoparticles

with the applied magnetic field, thus lowering the value of its magnetisation in comparison to bare

nanoparticles. The strong magnetic response of these aqueous-stable nanoparticles could be

exploited for various applications such as magnetic targeting, hyperthermia treatment, bio-sensing

and magnetic resonance imaging. The substantial drop in the magnetisation values of the MLs is

attributed to the high molecular weight of the non-magnetic soy-PC bi-layer. This bilayer masks

Page 163: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

142

the domain alignment and restricts the response of the Dx–Fe3O4 nanoparticles that are trapped

within the bilayers to the applied magnetic field.

Figure AI.5 (a) Field-dependent magnetisation (M vs. H) plot of Dx–Fe3O4 and MLs at room

temperature (b) Time-dependent specific absorption measurements of Dx–Fe3O4 nanoparticles

(Inset depicts the dependence of heat generation on the applied ACMF)

Figure AI.5b shows the time-dependent SAR of the Dx–Fe3O4 nanoparticles in response to

the application of varying ACMFs. The calorimetric measurement was used to determine the

heating rate of the Dx–Fe3O4 suspension. The SAR values of the nanoparticles were 9.51, 19.50,

22.03 and 32.90 W/g of iron with an applied field (H) of 7.64, 8.82, 9.41 and 10 kA/m,

respectively. Figure AI.5b illustrates that with the increment of the applied field, the time required

to reach a temperature of 45 °C from an initial ambient temperature was reduced, which is

consistent with the relationship between heat generation and the applied ACMF (the inset of Figure

AI.5b).

AI.3.2 Encapsulation (Loading) of Curcumin

The absorbance spectra of the pure curcumin and the supernatant (after magnetic

sedimentation) that was obtained from the MLs are given in Figure AI.6a, which shows that the

absorbance of the supernatant decreased with an increase in the ML-to-curcumin ratio. This

decrease that is observed in absorbance is attributed to the conjugation of the curcumin molecules

with the phosphatidyl choline groups, due to the increase in the concentration of the latter relative

to that of the former. In other words, the decrease of absorbance indicates an increased

encapsulation of curcumin. The hydrophobic curcumin and the lipophilic ends of soy-PC in an

Page 164: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

143

aqueous environment interact with each other in a way that facilitates the encapsulation of the drug

within the lipid bilayer. This been reported to be strongly dependent on the ratio of phosphatidyl

choline to curcumin during the thin-film formation step, because a single molecule of curcumin

binds with six molecules of phosphatidylcholine57, 58. In the present work, a maximum

encapsulation efficiency of 97.6% was achieved when the ratio of phosphatidyl choline to

curcumin was 1:0.6, with the curcumin-loaded MLs showing a high degree of homogeneity and

stability in aqueous suspensions. The encapsulation efficacy observed was higher than values

reported for similar nanocarrier systems14, 59.

Figure AI.6 (a) Absorbance spectra of curcumin-loaded MLs against pure curcumin (b) Drug

release profiles of curcumin-MLs at physiological and hyperthermic temperatures

AI.3.3 Release of Curcumin at Elevated Temperatures

Figure AI.6b shows the release profile of the curcumin-loaded MLs under physiological

(37 °C) and elevated (45 °C) temperature in reservoir-sink conditions. The sink (PBS) was spiked

with 0.1% (v/v) chloroform to facilitate the diffusion of hydrophobic curcumin in an aqueous

environment. Under normal physiological conditions, curcumin release was lower than 6%, which

is negligible. In contrast, it was released rapidly under hyperthermia temperatures following an

initial short period (25 min) of slow release. At 45 °C, the curcumin release reached a plateau at

150 min and had a total release percentage of ∼56%. The release of the curcumin molecules could

be attributed to the disruption of the heat sensitive lipid bilayer at elevated temperatures, which

may have weakened and, thus, disrupted the hydrophobic interactions between the curcumin and

the phosphatidyl choline groups in the bilayer, thereby exposing curcumin to the surrounding

Page 165: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

144

environment. The phase transition temperature is an important parameter in determining the

fluidity of the bilayer of liposomes. The fluidity, in turn, affects the release of curcumin from the

MLs. Since the physiological temperature is below the transition temperature of soy-PC, the

release of curcumin in physiological environments is inhibited. In contrast, a hyperthermic

temperature of 45 °C is above the transition temperature and fluidises the bilayer, enhancing the

release of the curcumin60.

Another factor that must be considered in the evaluation of anticancer drug release is the

acidic microenvironment around a tumour. Tumours, due to their hypoxic (low oxygen) conditions

and high lactic acid secretion, tend to cause acidity in their surrounding microenvironment with a

pH value below physiological (7.2–7.4). Hence, a pH value that is lower than 7.0 is typically used

to evaluate the pH sensitivity of cancer drug release. In this work, the effect of pH sensitivity on

the release of the curcumin-loaded MLs was investigated with a sodium acetate buffer (pH 4.8).

The result showed that little curcumin was released from the ML system at the decreased pH value,

thereby establishing the independence of pH sensitivity on the curcumin-loaded MLs. Considering

this result, this work focused on the temperature-stimulated drug release of curcumin-loaded MLs

at pH 7.0.

AI.3.4 Biocompatibility of Magnetic Liposomes and Anticancer Therapeutic

Effect of Curcumin-Loaded Magnetic Liposomes

Figure AI.7a shows the cell viabilities of mouse fibroblast (L929) and cervical cancer

(HeLa) cell lines that were incubated in a growth medium that contained MLs. An SRB assay was

performed in order to quantify the viable cell population, thereby indicating the effect of the MLs

on the growth and phenotype of the L929 and HeLa cells. The results suggested that the MLs alone

are biocompatible and, thus, are safe for in vivo studies. The relative cell viability, which was

calculated from eqn. 3.3, exceeded 100% when the concentration of the nanoparticles was low;

this was attributed to the facilitation of cell growth by the iron released by the cellular degradation

of Fe3O4 61.

Page 166: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

145

Figure AI.7 (a) Percentage of the cell viability of MLs incubated with mouse fibroblasts and

cervical cancer cells for 24 h (b) Dose-dependent evaluation of Curcumin-MLs for determination

of IC50 with HeLa cells

Figure AI.7b illustrates the results of the dose-dependent study that was performed with

HeLa cells in order to evaluate the half maximal inhibitory concentration (IC50) of the curcumin-

loaded MLs. The results indicate that the formulations were able to inhibit cell proliferation by

approximately 65% in HeLa cells. The amount of pure curcumin that reduced the cell population

by half (IC50) was 125 µg/ml over a period of 48 h. The IC50 value of curcumin has been reported

as ranging from 15 to 30 µM with various cancer cell lines14, 62, 63. The dose-response curve fitting

with Origin 8 software showed that the curcumin-loaded MLs hindered cell growth and reduced

the cell population by half (IC50) at a concentration of 2.09 mg/ml (R2=0.982). However, cell

viability was decreased to 70% at a concentration ∼62.5 µg/ml, as indicated by Figure AI.7b. The

IC50 value of 2.09 mg/ml for curcumin-loaded MLs was higher than that (125 µg/ml) of pure

curcumin, which is due to the loading of the Dx–Fe3O4 and the encapsulative lipid bilayers, along

with the ML release profile over 48 h. In a physiological environment, the anticancer therapeutic

effect of pure curcumin is hampered by its low aqueous solubility and poor chemical stability,

which limits its bioavailability at the targeted cancer site. Hence, the MLs would enhance the

therapeutic effect by increasing the delivery efficiency of curcumin when compared with pure

curcumin.

Page 167: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

146

AI.4 Summary

The controlled release of curcumin using the ML system and the therapeutic effect of this

drug formula have been explored for the treatment of cervical cancer. The surface-engineered

magnetic nanoparticles and the dextrin-coated iron oxide (Dx–Fe3O4) were fabricated using a

single-step facile co-precipitation approach. The Dx–Fe3O4 nanoparticles were successfully

encapsulated within the hydrophilic core of liposomes, producing a magnetic liposome (ML)

system. The conjugation of dextrin, lipid and curcumin molecules onto the Fe3O4 nanoparticles

was also achieved. The MLs exerted little or no toxic effects on normal fibroblast cells (L929),

showing potential for being used as a drug carrier to deliver hydrophobic drugs in aqueous

environments without harming normal cells. The release of curcumin molecules can be controlled

by varying the temperature, which can be achieved by applying an ACMF. The fabricated

curcumin-loaded MLs can release curcumin in appreciable amounts (up to 56.3±3.9%) at 45 °C

under the control of magnetic stimulation. More significantly, the curcumin-loaded MLs can

effectively inhibit cancer cell growth and viability and have an IC50 value of 2.09 mg/ml. In short,

the work has demonstrated the ability of MLs to efficiently deliver curcumin and to maintain its

anticancer activity, in an aqueous environment for the treatment of cervical cancer.

AI.5 References

1. T. Vial and J. Descotes, Toxicology, 2003, 185, 229-240.

2. R. Motterlini, R. Foresti, R. Bassi and C. J. Green, Free Radical Biology and Medicine,

2000, 28, 1303-1312.

3. Bhawana, R. K. Basniwal, H. S. Buttar, V. K. Jain and N. Jain, Journal of Agricultural and

Food Chemistry, 2011, 59, 2056-2061.

4. A. J. A. R. S. a. K. K. R. Mari selvam, Journal of Applied Pharmaceutical Science, 2012,

2.

5. R. Wilken, M. S. Veena, M. B. Wang and E. S. Srivatsan, Molecular cancer, 2011, 10, 12.

6. M. Malik, M. Mendoza, M. Payson and W. H. Catherino, Fertility and Sterility, 2009, 91,

2177-2184.

7. C.-L. L. a. J.-K. Lin, Journal of Cancer Molecules, 2008, 4.

Page 168: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

147

8. R. S. Mulik, J. Mönkkönen, R. O. Juvonen, K. R. Mahadik and A. R. Paradkar,

International Journal of Pharmaceutics, 2012, 437, 29-41.

9. G. Sa and T. Das, Cell division, 2008, 3, 14.

10. T. Choudhuri, S. Pal, T. Das and G. Sa, Journal of Biological Chemistry, 2005, 280, 20059-

20068.

11. A. Sahu, U. Bora, N. Kasoju and P. Goswami, Acta Biomater, 2008, 4, 1752-1761.

12. Y. S. Chun, S. Bisht, V. Chenna, D. Pramanik, T. Yoshida, S. M. Hong, R. F. de Wilde, Z.

Zhang, D. L. Huso, M. Zhao, M. A. Rudek, V. Stearns, A. Maitra and S. Sukumar,

Carcinogenesis, 2012, 33, 2242-2249.

13. R. K. Das, N. Kasoju and U. Bora, Nanomedicine, 2010, 6, 153-160.

14. A. MUKERJEE and J. K. VISHWANATHA, Anticancer Research, 2009, 29, 3867-3875.

15. R. Feng, W. Zhu, Z. Song, L. Zhao and G. Zhai, Journal of Nanoparticle Research, 2013,

15, 1-12.

16. P. Anand, H. B. Nair, B. Sung, A. B. Kunnumakkara, V. R. Yadav, R. R. Tekmal and B.

B. Aggarwal, Biochemical Pharmacology, 2010, 79, 330-338.

17. F. Dilnawaz and S. K. Sahoo, European journal of pharmaceutics and biopharmaceutics :

official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V, 2013,

85, 452-462.

18. M. M. Yallapu, M. C. Ebeling, S. Khan, V. Sundram, N. Chauhan, B. K. Gupta, S. E.

Puumala, M. Jaggi and S. C. Chauhan, Molecular Cancer Therapeutics, 2013, 12, 1471-

1480.

19. A. Anitha, S. Maya, N. Deepa, K. P. Chennazhi, S. V. Nair, H. Tamura and R. Jayakumar,

Carbohydrate Polymers, 2011, 83, 452-461.

20. Y. Chen, Q. Wu, Z. Zhang, L. Yuan, X. Liu and L. Zhou, Molecules, 2012, 17, 5972-5987.

21. L. Li, F. S. Braiteh and R. Kurzrock, Cancer, 2005, 104, 1322-1331.

22. J. W. Huanlei Chen, Min Sun, Journal of Liposome Research, 2012, 22, 100-109.

23. P. Basnet, H. Hussain, I. Tho and N. Skalko-Basnet, Journal of Pharmaceutical Sciences,

2012, 101, 598-609.

24. K. H. Bae, M. Park, M. J. Do, N. Lee, J. H. Ryu, G. W. Kim, C. Kim, T. G. Park and T.

Hyeon, ACS Nano, 2012, 6, 5266-5273.

Page 169: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

148

25. N. K. Prasad, K. Rathinasamy, D. Panda and D. Bahadur, Journal of Materials Chemistry,

2007, 17, 5042-5051.

26. S. Shen, F. Kong, X. Guo, L. Wu, H. Shen, M. Xie, X. Wang, Y. Jin and Y. Ge, Nanoscale,

2013, 5, 8056-8066.

27. D. A. M. Iero, T. Isernia, L. Crocco and I. Catapano, 2012.

28. G. Béalle, R. Di Corato, J. Kolosnjaj-Tabi, V. Dupuis, O. Clément, F. Gazeau, C. Wilhelm

and C. Ménager, Langmuir, 2012, 28, 11834-11842.

29. P. Guardia, R. Di Corato, L. Lartigue, C. Wilhelm, A. Espinosa, M. Garcia-Hernandez, F.

Gazeau, L. Manna and T. Pellegrino, ACS Nano, 2012, 6, 3080-3091.

30. P. Pradhan, J. Giri, G. Samanta, H. D. Sarma, K. P. Mishra, J. Bellare, R. Banerjee and D.

Bahadur, Journal of Biomedical Materials Research Part B: Applied Biomaterials, 2007,

81B, 12-22.

31. Q. Quan, J. Xie, H. Gao, M. Yang, F. Zhang, G. Liu, X. Lin, A. Wang, H. S. Eden, S. Lee,

G. Zhang and X. Chen, Molecular Pharmaceutics, 2011, 8, 1669-1676.

32. S. Chandra, K. C. Barick and D. Bahadur, Advanced Drug Delivery Reviews, 2011, 63,

1267-1281.

33. J. Giri, P. Pradhan, V. Somani, H. Chelawat, S. Chhatre, R. Banerjee and D. Bahadur,

Journal of Magnetism and Magnetic Materials, 2008, 320, 724-730.

34. H. Qu, D. Caruntu, H. Liu and C. J. O’Connor, Langmuir, 2011, 27, 2271-2278.

35. M. Mahmoudi, S. Sant, B. Wang, S. Laurent and T. Sen, Advanced Drug Delivery Reviews,

2011, 63, 24-46.

36. A. Quarta, A. Curcio, H. Kakwere and T. Pellegrino, Nanoscale, 2012, 4, 3319-3334.

37. V. Fischer, I. Lieberwirth, G. Jakob, K. Landfester and R. Muñoz-Espí, Advanced

Functional Materials, 2013, 23, 451-466.

38. L. Lartigue, C. Innocenti, T. Kalaivani, A. Awwad, M. d. M. Sanchez Duque, Y. Guari, J.

Larionova, C. Guérin, J.-L. G. Montero, V. Barragan-Montero, P. Arosio, A. Lascialfari,

D. Gatteschi and C. Sangregorio, Journal of the American Chemical Society, 2011, 133,

10459-10472.

39. A. Pfaff, A. Schallon, T. M. Ruhland, A. P. Majewski, H. Schmalz, R. Freitag and A. H.

E. Müller, Biomacromolecules, 2011, 12, 3805-3811.

40. B.-H. Lai, C.-C. Yeh and D.-H. Chen, Process Biochemistry, 2012, 47, 799-805.

Page 170: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

149

41. M. M. Yallapu, S. F. Othman, E. T. Curtis, B. K. Gupta, M. Jaggi and S. C. Chauhan,

Biomaterials, 2011, 32, 1890-1905.

42. A. Moore, E. Marecos, A. Bogdanov and R. Weissleder, Radiology, 2000, 214, 568-574.

43. M. Mikhaylova, D. K. Kim, C. C. Berry, A. Zagorodni, M. Toprak, A. S. G. Curtis and M.

Muhammed, Chemistry of Materials, 2004, 16, 2344-2354.

44. P. Nicolás, M. Saleta, H. Troiani, R. Zysler, V. Lassalle and M. L. Ferreira, Acta

Biomaterialia, 2013, 9, 4754-4762.

45. P. Pradhan, J. Giri, F. Rieken, C. Koch, O. Mykhaylyk, M. Döblinger, R. Banerjee, D.

Bahadur and C. Plank, Journal of Controlled Release, 2010, 142, 108-121.

46. M. Mahmoudi, H. Hofmann, B. Rothen-Rutishauser and A. Petri-Fink, Chemical Reviews,

2011, 112, 2323-2338.

47. X. Ding, K. Cai, Z. Luo, J. Li, Y. Hu and X. Shen, Nanoscale, 2012, 4, 6289-6292.

48. N. Baccile, R. Noiville, L. Stievano and I. V. Bogaert, Physical Chemistry Chemical

Physics, 2013, 15, 1606-1620.

49. J. Bondeson, J. Wijkander and R. Sundler, Biochimica et Biophysica Acta (BBA) -

Biomembranes, 1984, 777, 21-27.

50. V. Vichai and K. Kirtikara, Nat. Protocols, 2006, 1, 1112-1116.

51. M. Ibrahim, M. Alaam, H. El-Haes, A. F. Jalbout and A. d. Leon, Eclética Química, 2006,

31, 15-21.

52. M. Hancer, A. Patist, R. T. Kean and H. S. Muralidhara, Colloids and Surfaces A:

Physicochemical and Engineering Aspects, 2002, 204, 31-41.

53. P. R. K. Mohan, G. Sreelakshmi, C. V. Muraleedharan and R. Joseph, Vibrational

Spectroscopy, 2012, 62, 77-84.

54. I. Orienti, Zuccari, G., Carosio, R., G. Montaldo, P., Drug Delivery, 2009, 16, 389-398.

55. Y.-Q. Zhang, X.-W. Wei and R. Yu, Catal Lett, 2010, 135, 256-262.

56. E. Bertoft, Carbohydrate Research, 1991, 212, 229-244.

57. G. Began, E. Sudharshan, K. Udaya Sankar and A. G. Appu Rao, Journal of Agricultural

and Food Chemistry, 1999, 47, 4992-4997.

58. M. A. Mukesh KUMAR, Surendra Kumar SHARMA, Scientia Pharmaceutica, 2008, 76.

Page 171: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

150

59. S. S. Dhule, P. Penfornis, T. Frazier, R. Walker, J. Feldman, G. Tan, J. He, A. Alb, V. John

and R. Pochampally, Nanomedicine: Nanotechnology, Biology and Medicine, 2012, 8,

440-451.

60. D. Marsh, Biophysical Journal, 1992, 61, 1036-1040.

61. J. Wan, W. Cai, X. Meng and E. Liu, Chemical Communications, 2007, DOI:

10.1039/B712795B, 5004-5006.

62. A. Sahu, N. Kasoju and U. Bora, Biomacromolecules, 2008, 9, 2905-2912.

63. S. Banerjee, P. Prasad, A. Hussain, I. Khan, P. Kondaiah and A. R. Chakravarty, Chemical

Communications, 2012, 48, 7702-7704.

Page 172: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

151

List of Publications

Research Articles

1. Saumya Nigam, K. C. Barick, D. Bahadur, Development of citrate-stabilized Fe3O4

nanoparticles: Conjugation and release of doxorubicin for therapeutic applications, J.

Magn. Magn. Mater., 2011, 323, 237-243. (Citations - 149)

2. S. Chandra, Saumya Nigam, D. Bahadur, Combining unique properties of dendrimers and

magnetic nanoparticles towards cancer theranostics, J. Biomed. Nanotech., 2014, 10, 32-

49. (Citations - 13)

3. Saumya Nigam, S. Chandra, D. F. Newgreen, D. Bahadur, Q. Chen, Poly (ethylene

glycol)-modified PAMAM-Fe3O4-Doxorubicin triads with the potential for improved

therapeutic efficacy: Generation-dependent increased drug loading and retention at neutral

ph and increased release at acidic pH, Langmuir, 2014, 30, 1004-1011. (Citations - 18)

4. Saumya Nigam, A. Kumar, G. A. Thouas, D. Bahadur, Q Chen, Curcumin delivery using

magnetic liposomes, J. Nanopharma. Drug Del., 2013, 1, 365-375. (Citations - 1)

5. Saumya Nigam, S. Chandra, D. Bahadur, Dendrimers based electrochemical biosensors,

Biomed. Res. J., 2015, 2, 21-36. (Citations - 1)

6. Saumya Nigam, D. Bahadur, Dendrimerized magnetic nanoparticles as carriers for the

anti-cancer compound, epigallocatechin gallate, IEEE Trans. Magn., 2016, 52, 1-5.

7. Saumya Nigam, D. Bahadur, Dendritic Fe3O4 nanoparticles for combinatorial therapy:

Peptide dendrimers with enhanced efficiency as alternative platforms for pamam

dendrimers (Under Review)

8. Saumya Nigam, D. Bahadur, Temperature-induced MR contrast properties of dendritic

Fe3O4 nanoparticles (To be communicated)

9. Saumya Nigam, D. Bahadur, Assessment of doxorubicin-loaded dendritic Fe3O4

nanoparticles for magnetic drug targeting in murine melanoma model (Under Review)

Jeotikanta Mohapatra, Saumya Nigam, J. Gupta, A. Mitra, D. Bahadur, M. Aslam,

Enhancement of magnetic heating efficiency in size controlled MFe2O4 (M = Mn, Fe, Co

and Ni) Nanoassemblies, RSC Adv., 2015, 5, 14311-14321. (Citations - 8)

Page 173: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

152

Eswara Vara Prasadarao K, Saumya Nigam, M. Aslam and D. Bahadur, Novel Mg-Al

layered double hydroxide-Fe3O4 magnetic nanohybrids for efficient thermo-chemo therapy

of cervical cancer, New J. Chem., 2016, 40, 423-433. (Citations - 7)

Swati, Saumya Nigam, A. S. Khanna, R.K. Singh Raman, Silane coated magnesium

implants with improved in vitro corrosion resistance and biocompatibility, J. Mater. Sci.

Surf. Engg., 2016, 13, 29.

Presentations at National/International Conferences

1. Saumya Nigam, D. Bahadur, Assessment of Doxorubicin-loaded Dendritic Fe3O4

Nanoparticles for Magnetic Drug Targeting in Murine Melanoma Model, Presented at

IITB-NTU Joint Symposium on Healthcare Technologies 2016, Sept 26 – 27, 2016,

Mumbai, India (Won the Best Poster award)

2. Saumya Nigam, D. Bahadur, Peptide Dendrimer-Fe3O4 Nanoparticles with Enhanced

Efficiency as Platforms for Combinatorial Thermo-Chemo Therapy, Presented at 43rd

Annual Meeting & Exposition of the Controlled Release Society, July 17 – 20, 2016,

Seattle, Washington, U.S.A.

3. Saumya Nigam, D. Bahadur, Peptide dendrimers in non-invasive magnetic resonance

imaging, Presented at International Symposium on Nanotechnology and Cancer

Theranostics, Feb 19 – 21, 2015, Mumbai, India

4. Saumya Nigam, D. Bahadur, Peptide dendrimers in non-invasive magnetic resonance

imaging, Presented at In-house symposium of IITB-Monash Research Academy, Aug 7,

2015, Mumbai, India (Nominated for Best poster award)

5. Saumya Nigam, Q. Z. Chen, D. Bahadur, Comparative therapeutic performances of drug

loaded magnetic dendritic nanoparticles towards various cancer cell lines, Presented at In-

house symposium of IITB-Monash Research Academy, Aug 8, 2014, Mumbai, India

6. Saumya Nigam, Asmita Kumar, Qizhi Chen, D. Bahadur, Therapeutic effect of curcumin

loaded magnetic liposomes on cervical cancer, Presented at 7th International Conference

on Materials for Advanced Technologies, June 30 – July 5, 2013, Singapore, Singapore

7. Saumya Nigam, Qizhi Chen, D. Bahadur, Therapeutic performance of PEG-PAMAM

dendrimers functionalized magnetic nanoparticles towards cervical cancer, 2013 Spring

Page 174: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

153

Meeting of European Materials Research Society, May 27-31, 2013, Strasbourg, France –

Presented by Prof. D. Bahadur

8. Sudeshna Chandra, Saumya Nigam, Akshaya K Swain, Shanta S Naorem, Anand Prakash,

Dipa Dutta, D. Bahadur, Advanced Functional Nanostructures: From Synthesis to

Applications, Presented at Nano India, Feb 19-20, 2013, NIIST, Trivandrum, India

9. Attended one-day workshop on “Fluorescence Steady State and Lifetime Analysis”, on 7th

Dec, 2010 at Mumbai University

Page 175: Dendrimer functionalized magnetic nanoparticles as …...(Saumya Nigam) (IITB: 09411414) (MU: 22667563) Date: 24.10.2016 i Abstract Cancer therapeutics deals with development of preclinical

154

Acknowledgments

I am under no illusion that my PhD work would have been possible without the support of

others, so I am sincerely grateful to a long list of people and the various roles they played during

my PhD. To begin with, I wish to express my utmost gratitude to my parents and friends for always

supporting me and being the steadfast pillars of my life.

I would like to acknowledge the tireless work of my supervisor Prof. D. Bahadur (IIT

Bombay) for his valuable and expert guidance and patience for bearing with me for the last few

years. All these years have been a constant learning process under his supervision and he shall

always be a constant source of inspiration for me. I would also like to thank Dr. Q. Z. Chen (ex-

supervisor; Monash University) for her pivotal suggestions towards my research work during my

tenure at Monash University and later Dr. X. Chen. I would also like to thank my entire research

progress committee for their individual guidance, suggestions and support during my doctoral

studies. Thank you Prof. Rohit Srivastava and Dr. Lizhong He for your scientific guidance and

insightful suggestions.

I am also thankful to the staff members of my department (both at IIT Bombay as well as

Monash University) and the technical staff at SAIF (Sophisticated Analytical Instrument Facility)

for their help in various characterisation facilities and other helps during my work. I would like to

thank Ms. Aradhana Pant, Ms. Pradnya Nikam and Mr. Sachin Tawde at IIT Bombay with a special

mention of Dr. Qizhu Wu of Monash Biomedical Imaging facility for all his efforts and help

towards my MRI studies.

Last but not the least, I would like to extend my gratitude to all of my group members (both at IIT

Bombay and Monash University), past and present, especially, Dr. Sudeshna Chandra, Dr. K. C.

Barick, Dr. Pallab Pradhan and all the co-workers for their insightful and elaborate research

discussions and providing an amiable environment in the lab throughout my PhD.

(Saumya Nigam)