179
INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY DEVICE FOR THE TREATMENT OF EPILEPSY Sébastien Hyacinthe Bauquier http://orcid.org/0000-0001-7355-5590 Submitted in total fulfilment of the requirements of the degree of Doctor of Philosophy August 2018 Faculty of Veterinary and Agricultural Sciences & Faculty of Medicine, Dentistry and Health Sciences The University of Melbourne

INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

INITIAL DEVELOPMENT OF AN

IMPLANTABLE DRUG DELIVERY DEVICE

FOR THE TREATMENT OF EPILEPSY

Sébastien Hyacinthe Bauquier

http://orcid.org/0000-0001-7355-5590

Submitted in total fulfilment of the requirements of the

degree of Doctor of Philosophy

August 2018

Faculty of Veterinary and Agricultural Sciences &

Faculty of Medicine, Dentistry and Health Sciences

The University of Melbourne

Page 2: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

ii

Abstract

Brief abstract

Epilepsy is a chronic neurological condition, characterized by recurrent seizures.

Treatment with conventional anti-epileptic drugs (AED) results in only 33% of patients

having no seizure recurrence after a prolonged period. Some evidence suggests that the

lack of effectiveness of AED penetration into the brain parenchyma could be one of the

mechanisms for resistance to treatment. Furthermore, AED side effects often prevent

large increases in their dose. It was for these reasons that a new alternative approach to

therapy, intracranial implantation of polymer-based drug delivery systems aimed at

improving the bioavailability of AED, was investigated in this PhD research project.

Genetic Absent Epilepsy Rats from Strasbourg (GAERS) is a strain of rat that has 100%

of the animals with recurrent generalised non-convulsive seizures. This animal model

has become the gold standard to study the mechanisms underlying absence epilepsy.

After validating an automated spike and wave discharges (SWDs) detection algorithm

applied to GAERS’ electroencephalograms (EEG) and demonstrating some effects of

enrofloxacin on the GAERS’ epileptic activity, in vitro characterisation and in vivo

testing of the antiepileptic efficacy of Poly(D,L-lactide-co-glycolide) (PLGA) polymers

loaded with anticonvulsants, either phenytoin or lacosamide, were performed. PLGA is

a biodegradable copolymer that it is very well tolerated by the brain and can be used as

a passive release substrate.

In prospective randomized masked experiments, GAERS underwent surgery for

implantation of skull electrodes, skull electrodes and blank polymers, or skull electrodes

and AED loaded polymers. The polymers were implanted bilaterally and subdurally on

the surface of the cortex. Electroencephalogram recordings were started at day 7 post-

surgery and continued for eight weeks. The number of SWDs and mean duration of one

SWD were compared week-by-week between the groups. Although temporary changes

were seen, phenytoin loaded PLGA polymer sheets did not decrease seizure activity in

GAERS. However lacosamide loaded PLGA polymer sheets affected seizure activity in

GAERS by decreasing the mean duration of SWDs for a sustained period of up to 7

Page 3: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

iii

weeks. This provided proof of concept that intracranial implantation of polymer-based

drug delivery systems could be used in the treatment of epilepsy.

The last chapter of this thesis evaluated the in vivo biocompatibility of Polypyrrole

(PPy) implanted subdurally and unilaterally on the surface of the motor cortex in

GAERS. Polypyrrole offers the advantage of being electrically conductive which could

allow a controlled release of the anti-epileptic medications. Due to the absence of

evidence of toxic injury or immune mediated inflammation, it was concluded that PPy

offers good histocompatibility with central nervous system cells. Furthermore, the

comparison of immunohistochemical scores reveals that the amount of neuronal death

and gliosis was significantly less in the PPy side than in the sham surgery side of the

cortices (p values of 0.005 and 0.002 respectively) implying that the application of PPy

could protect the CNS tissue after surgery.

With improvements in polymer technologies and episodic release offering potentially

much longer lasting release durations, intracranial polymer-based drug delivery systems

may provide an effective therapeutic strategy for chronic epilepsy. While the

development of such an antiepileptic device is worthwhile but still years away, the

development of an implantable device combining the potential neuroprotective effect of

PPy and an anti-inflammatory drug like dexamethasone would certainly be quicker to

develop.

Page 4: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

iv

Declaration

I, Sébastien Hyacinthe Bauquier, hereby certified that:

The thesis comprises only my original work towards the PhD except where

indicated in the Preface,

Due acknowledgement has been made in the text to all other material used,

The thesis is fewer than 100 000 words in length, exclusive of tables, maps,

bibliographies and appendices.

Date: 13th

of December 2017

Signature:

Page 5: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

v

Acknowledgments

First and foremost I would like to thank Professor Mark Cook for welcoming me into

his team of exceptional scientists and engineers and for giving me the opportunity to

complete this PhD under his trusting supervision. Mark is a brilliant and supportive

supervisor who gave me the freedom to perform various experiments without

objections. Mark is a professional role model for me and I am extremely grateful to

have had the opportunity to work with him.

I would like to thank my other supervisors, Dr Sam Long for introducing me to

Professor Cook and for being at the origin this project, and Professor Ted Whittem for

his mentoring. Ted has and continues to provide me with insightful guidance and

support throughout my time at the University of Melbourne. Ted’s office and home

doors have always been open and Ted has always been very generous with his time.

I would like to thank Professor Andrew Fisher for his role as advisory committee chair.

Andrew’s calm and confident attitude and his valuable contributions were very much

valued.

I would like to thank all my co-authors Ray Boston, Yu Chen, Amy Halliday, Jonathan

Jiang, Alan Lai, Karen McLean, Simon Moulton, Yi Sui, Sara Vogrin, Gordon Wallace

and Zhilian Yue. The projects reported in this thesis are very much multi-disciplinary

and I couldn’t have completed my PhD without the help of all my co-authors. I would

like to especially acknowledge Alan and Jonathan who were my everyday supporters at

work.

I would like to thank my parents Aline and Gérard for their unconditional love, support

and encouragement, and for their sacrifices that allowed me to get where I am today. To

my late father who always believed in me when all others were giving up: I miss you!

I would like to thank my wife Jennifer and our two children Julien and Miles; I love you

so infinity much! Jenni, you remind me every day what generosity looks like. Petit

Bonhomme and Milo, you are so different and still best friends; the world would be a

much better place if we could all be as non-judgemental and tolerant of each other’s

differences as you are. Please don’t change!

Page 6: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

vi

Preface The format of the thesis is “Thesis with Publications”. The published manuscripts

include multiple co-authors. Below are their affiliations and a chapter by chapter short

description of the authors’ contributions.

Affiliations of co-authors

Ray C. Boston

Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia.

Yu Chen

Intelligent Polymer Research Institute and ARC Centre of Excellence for

Electromaterials Science, AIIM Facility, Innovation Campus, University of

Wollongong, Wollongong, New South Wales 2522, Australia.

Mark J. Cook

Department of Clinical Neurosciences, St Vincent's Hospital, Fitzroy, Victoria 3065,

Australia. Faculty of Medicine, University of Melbourne, Parkville, Victoria 3010,

Australia. Intelligent Polymer Research Institute and ARC Centre of Excellence for

Electromaterials Science, AIIM Facility, Innovation Campus, University of

Wollongong, Wollongong, New South Wales 2522, Australia.

Amy J. Halliday,

Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

Jonathan L. Jiang

Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

Alan Lai

Page 7: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

vii

Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

Karen J. McLean

Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

Simon E. Moulton

ARC Centre of Excellence for Electromaterials Science, Bioengineering Program

Leader – Iverson Health Innovation Research Institute, Biomedical Engineering,

Faculty of Science, Engineering and Technology, Swinburne University of Technology,

Hawthorn, VIC 3122, Australia.

Yi Sui

Department of Neurology, Shenyang First People's Hospital, Shenyang 110041, China.

Sara Vogrin

Faculty of Medicine, University of Melbourne, Parkville, Victoria 3010, Australia.

Gordon Wallace

Intelligent Polymer Research Institute and ARC Centre of Excellence for

Electromaterials Science, AIIM Facility, Innovation Campus, University of

Wollongong, Wollongong, New South Wales 2522, Australia

Zhilian Yue

Intelligent Polymer Research Institute and ARC Centre of Excellence for

Electromaterials Science, AIIM Facility, Innovation Campus, University of

Wollongong, Wollongong, New South Wales 2522, Australia

List of co-authors for Chapter 2: Chapter with publication

EVALUATION OF AN AUTOMATED SPIKE AND WAVE COMPLEXES

DETECTION ALGORITHM APPLIED TO GAERS EEGS.

Page 8: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

viii

Sebastien H. Bauquier, Alan Lai, Jonathan L. Jiang, Yi Sui, Mark J. Cook

Neuroscience Bulletin. 2015;31(5):601-10.

All the work described in chapter 2 was performed at Centre for Clinical Neurosciences

and Neurological Research (St. Vincent’s Hospital Melbourne, P.O. Box 2900, Fitzroy,

Victoria 3065, Australia). The first author contributed 70% of the work. The

establishment of the algorithm was mainly the work of the first two authors, and the

manual marking of the EEGs was done by both Sebastien Bauquier and Jonathan Jiang.

List of co-authors for Chapter 3: Chapter with publication

CLONIC SEIZURES INDUCED BY ORAL ADMINISTRATION OF

ENROFLOXACIN IN GAERS

Sebastien H. Bauquier, Jonathan L. Jiang, Alan Lai and Mark J. Cook

Comparative Medicine, 2016;66(3):220-4.

All the work described in chapter 3 was performed at Centre for Clinical Neurosciences

and Neurological Research (St. Vincent’s Hospital Melbourne, P.O. Box 2900, Fitzroy,

Victoria 3065, Australia). The first author contributed 80% of the work.

Contributions for Chapter 4: Unpublished material not submitted for publication

ANTI-EPILEPTIC EFFECTS OF PHENYTOIN LOADED POLYMERS

IMPLANTED SUB-DURALLY IN GAERS RATS

The work described in chapter 4 was performed at Intelligent Polymer Research

Institute and ARC Centre of Excellence for Electromaterials Science (AIIM Facility,

Innovation Campus, University of Wollongong, Wollongong, New South Wales 2522,

Australia) and at Centre for Clinical Neurosciences and Neurological Research, (St.

Vincent’s Hospital Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia).

Sebastien Bauquier contributed 70% of the work. The in vitro experiments were mainly

Page 9: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

ix

performed at the University of Wollongong in collaboration with Sebastien Bauquier

and the in vivo experiments were mainly performed by Sebastien Bauquier.

This work also required the use of the facilities and the assistance of Mr. Tony Romeo

at the University of Wollongong Electron Microscopy Centre.

List of co-authors for Chapter 5: Chapter with publication

ANTI-EPILEPTIC EFFECTS OF LACOSAMIDE LOADED POLYMERS

IMPLANTED SUB-DURALLY IN GAERS RATS

Sebastien H. Bauquier, Jonathan L. Jiang, Zhilian Yue, Alan Lai, Yu Chen, Simon

E. Moulton, Karen J. McLean, Sara Vogrin, Amy J. Halliday, Gordon Wallace,

Mark J. Cook

International Journal of Polymer Science. 2016; (2016) Article ID 65949602016

The work described in chapter 5 was performed at Intelligent Polymer Research

Institute and ARC Centre of Excellence for Electromaterials Science (AIIM Facility,

Innovation Campus, University of Wollongong, Wollongong, New South Wales 2522,

Australia) and at Centre for Clinical Neurosciences and Neurological Research, (St.

Vincent’s Hospital Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia). The

first author contributed 70% of the work. The in vitro experiments were mainly

performed by the authors from the University of Wollongong in collaboration with the

other authors including the first author and the in vivo experiments were mainly

performed by the first author.

The authors also acknowledge the use of the facilities and the assistance of Mr. Tony

Romeo at the University of Wollongong Electron Microscopy Centre.

List of co-authors for Chapter 6: Chapter with publication

EVALUATION OF THE BIOCOMPATIBILITY OF PPY ACTIVE RELEASE

POLYMERS IMPLANTED SUB-DURALLY IN GAERS

Page 10: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

x

Sebastien H. Bauquier, Karen J. McLean, Jonathan L. Jiang, Zhilian Yue,

Raymond C. Boston, Alan Lai, Yu Chen, Simon E. Moulton, Amy J. Halliday,

Gordon Wallace, Mark J. Cook

Macromolecular Bioscience, 2017 May;17(5)

The work described in chapter 6 was performed at Centre for Clinical Neurosciences

and Neurological Research, (St. Vincent’s Hospital Melbourne, P.O. Box 2900, Fitzroy,

Victoria 3065, Australia) in collaboration with the Intelligent Polymer Research

Institute and ARC Centre of Excellence for Electromaterials Science (AIIM Facility,

Innovation Campus, University of Wollongong, Wollongong, New South Wales 2522,

Australia). The first author contributed 70% of the work. The immuno-histologic

preparations were performed in collaboration with Karen McLean and the grading of the

immuno-histologic changes was performed by both the first author and Jonathan Jiang.

Page 11: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xi

Funding

The candidate gratefully acknowledges the sponsorship of the Victorian Government

through its Science Technology and Innovation Initiative, administered by the

Department of Industry, Innovation, and Regional Development, and of NHMRC. The

candidate also wishes to acknowledge funding from the Australian Research Council

(ARC) Centre of Excellence Scheme, the use of facilities at the University of

Wollongong Electron Microscopy Centre, support of the Australian National

Fabrication Facility (ANFF) – Materials Node.

Page 12: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xii

Presentations arising from work undertaken as part of this thesis

S.H. Bauquier, J.L Jiang, Z. Yue, A. Lai , Yu Chen, S. Moulton, S Long, G. Wallace,

M.J. Cook. Anti-epileptic effects of lacosamide loaded polymers implanted sub-durally

in GAERS rats. 4th Asia-Pacific Symposium on Nanobionics, Melbourne VIC Australia,

14-15 November 2013.

S.H. Bauquier, J.L. Jiang, Z. Yue, A. Lai , Y. Chen, S. Moulton, S. Vogrin, G.

Wallace, M.J. Cook. Anti-epileptic effects of laser cut lacosamide loaded polymers

implanted sub-durally in GAERS rats. 28th

Annual Scientific Meetings of the Epilepsy

Society of Australia, Melbourne VIC Australia, 5-7 November 2014.

S.H. Bauquier, K.J. McLean, J.L. Jiang, Z. Yue, A. Lai, Y Chen, S.E. Moulton, S.

Vogrin, A.J. Halliday, G. Wallace, M.J. Cook. Evaluation of the biocompatibility of

polypyrrole polymers implanted sub-durally in GAERS. 29th

Annual Scientific Meetings

of the Epilepsy Society of Australia, Adelaide SA Australia, 29 November- 3 December

2015.

Page 13: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xiii

Contents Abstract ........................................................................................................................................ ii

Declaration .................................................................................................................................. iv

Acknowledgments ....................................................................................................................... v

Preface ......................................................................................................................................... vi

Affiliations of co-authors ..................................................................................................... vi

List of co-authors for Chapter 2: Chapter with publication ................................................... vii

List of co-authors for Chapter 3: Chapter with publication .................................................. viii

Contributions for Chapter 4: Unpublished material not submitted for publication ............... viii

List of co-authors for Chapter 5: Chapter with publication .................................................... ix

List of co-authors for Chapter 6: Chapter with publication .................................................... ix

Funding ............................................................................................................................... xi

Presentations arising from work undertaken as part of this thesis ....................................... xii

List of tables .............................................................................................................................. xvi

List of figures ........................................................................................................................... xvii

List of appendices ..................................................................................................................... xix

Abbreviations ............................................................................................................................. xx

Chapter 1 – A new approach to the treatment of epilepsy. ..................................................... 1

1.1. Epilepsy................................................................................................................. 1

1.1.1. What is epilepsy? ............................................................................................... 1

1.1.2. Seizure types and classifications......................................................................... 1

1.1.3. Pathophysiology of epilepsy ............................................................................... 2

1.1.4. Diagnosis of epilepsy ......................................................................................... 2

1.2. Conventional treatments of epilepsy .......................................................................... 3

1.2.1. Anti-epileptic drug (AED) therapy ..................................................................... 3

1.2.2. Epilepsy neurosurgery ........................................................................................ 6

1.2.3. Vagus nerve Stimulation (VNS) ......................................................................... 6

1.2.4. Responsive neurostimulation (RNS) ................................................................... 6

1.2.5. Consequences of epilepsy on patient life style .................................................... 8

1.2.5.1. Severe impact of epilepsy on the patient life style ............................................ 8

1.2.5.2. Seizures prediction is becoming reality ........................................................... 8

1.3. AED loaded polymer implant as a novel treatment for epilepsy .................................. 8

1.3.1. The blood brain barrier, an obstacle for drug delivery to the brain parenchyma . 13

Page 14: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xiv

1.3.2. Polymer helping AEDs to cross the blood brain barrier .................................... 14

1.3.3. Polymer allowing AEDs delivery to the brain parenchyma while bypassing the

blood brain barrier ........................................................................................................... 15

1.3.3.1. Implantable polymers delivering AEDs into the cerebrospinal fluid .............. 15

1.3.3.2. Implantable polymers delivering AEDs into/onto brain parenchyma ............. 15

1.3.3.3. Injectable polymers delivering AEDs into/onto brain parenchyma ................ 16

1.4. Initial development of an implantable drug delivery device for the treatment of

epilepsy .............................................................................................................................. 17

1.4.1. Animal model used: GAERS ............................................................................ 18

1.4.2. Antiepileptic drugs used: Phenytoin and Lacosamide ....................................... 18

1.4.3. Polymers used: PLGA and PPy ........................................................................ 19

1.4.4. Proposed steps for the initial development of an implantable drug delivery device

for the treatment of epilepsy ............................................................................................ 21

Chapter 2 - Evaluation of an automated spike and wave complexes detection algorithm

applied to GAERS EEGs .......................................................................................................... 23

2.1 Outline .................................................................................................................... 23

2.2 Manuscript .............................................................................................................. 23

Chapter 3 - Seizures induced by oral administration of enrofloxacin in GAERS. ................. 41

3.1. Outline .................................................................................................................... 41

3.2. Manuscript .............................................................................................................. 42

Chapter 4 – Anti-epileptic effects of Phenytoin loaded polymers implanted subdurally in

GAERS ....................................................................................................................................... 55

4.1. Introduction ............................................................................................................. 55

4.2. Materials and methods ............................................................................................. 55

4.2.1. PLGA polymer mat production ........................................................................ 56

4.2.2. Morphology Study ........................................................................................... 56

4.2.3. Determination of drug loading in polymer mats ................................................ 56

4.2.4. Silicone sheets production ................................................................................ 57

4.2.5. In vitro drug release ......................................................................................... 57

4.2.6. Animals ........................................................................................................... 57

4.2.7. Primary outcome: epileptic activity .................................................................. 60

4.2.8. Secondary outcome: Post-operative health monitoring ...................................... 60

4.2.9. Statistical analysis ............................................................................................ 60

4.3. Results ..................................................................................................................... 61

Page 15: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xv

4.4. Discussion ............................................................................................................... 66

Chapter 5 – Anti-epileptic effects of lacosamide loaded polymers implanted subdurally in

GAERS ........................................................................................................................................ 69

5.1 Outline .................................................................................................................... 69

5.2 Manuscript .............................................................................................................. 69

Chapter 6 – Evaluation of the biocompatibility of polypyrrole polymers implanted sub-

durally in GAERS ..................................................................................................................... 98

6.1 Outline .................................................................................................................... 98

6.2 Manuscript .............................................................................................................. 98

Chapter 7 – Conclusion ........................................................................................................... 124

7.1 Development of the PLGA Polymers ..................................................................... 124

7.2 Development of the PPy polymers ......................................................................... 127

7.3 What are the challenges in the development of an implantable drug delivery device for

the treatment of epilepsy? .................................................................................................. 129

7.4 What is next? ......................................................................................................... 130

References ................................................................................................................................ 132

Appendices ............................................................................................................................... 148

Page 16: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xvi

List of tables Chapter 1

Table 1. 1 Main Mechanisms of action of common AEDs. ........................................................ 5

Table 1. 2 Anti-epileptic drugs delivered using polymer-based devices (in vitro experiments). .. 9

Table 1. 3 Anti-epileptic drugs delivered using polymer-based devices (in vivo experiments). . 11

Chapter 2

Table 2. 1 Median number and mean duration of spike-and-wave discharges (SWDs). ............ 36

Table 2. 2 Sensitivity, specificity, positive predictive value (PPV), and negative predicted value

(NPV) ..................................................................................................................................... 36

Chapter 5

Table 5. 1 Post-operative health monitoring chart. ................................................................... 81

Chapter 6

Table 6. 1 Post-operative health monitoring chart. ................................................................. 107

Page 17: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xvii

List of figures

Chapter 1

Figure 1. 1- Summary of success and failure rates of the conventional anti-epileptic treatments. 7

Figure 1. 2 Poly-lactide-co-glycolide polymer sheets produced by coaxial electrospinning

method.................................................................................................................................... 20

Figure 1. 3 Polypyrrole polymer sheets produced by electro-polymerization method ............... 21

Chapter 2

Figure 2. 1 Experimental protocol ........................................................................................... 28

Figure 2. 2 Example of rats EEGs when active, sleeping or seizing. ........................................ 29

Figure 2. 3 Selection the EEG channel and determination of amplitude threshold for SWD

detection ................................................................................................................................. 31

Figure 2. 4 Flowchart of the semi-automated algorithm ........................................................... 32

Figure 2. 5 The process of automatic marking. ........................................................................ 33

Figure 2. 6 Performance metrics .............................................................................................. 35

Chapter 3

Figure 3. 1 Experimental protocol ........................................................................................... 46

Figure 3. 2 Box-and-whisker plots of the epileptic activity before enrofloxacin treatment (n = 5)

and afterward (posttreatment, n = 5). ....................................................................................... 49

Chapter 4

Figure 4. 1 Polymers and electrodes implantation sites, and timeline of the experiment. .......... 59

Figure 4. 2 Scanning electron micrographs of coaxially electrospun polymer mats. ................. 61

Figure 4. 3 In vitro release data are presented for the phenytoin polymer (A) ........................... 62

Figure 4. 4 Comparison of the epileptic activity monitoring results (not including the silicone

group) ..................................................................................................................................... 63

Figure 4. 5 Comparison of the epileptic activity monitoring results from group silicone versus

group blank polymer. .............................................................................................................. 65

Page 18: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xviii

Chapter 5

Figure 5. 1 Polymers and electrodes implantation sites, and timeline of the experiment. .......... 79

Figure 5. 2 Electron micrographs of coaxially electrospun polymer mats and in vitro release

data. ........................................................................................................................................ 83

Figure 5. 3 Post-operative debilitation scores .......................................................................... 84

Figure 5. 4 Comparison of the epileptic activity monitoring results.......................................... 86

Figure 5. 5 Example of EEG recordings .................................................................................. 87

Chapter 6

Figure 6. 1 Scanning Electron Microscope (SEM) images of the polypyrrole thick film doped

with dextran sulfate (top surface A and B, cross-section C). .................................................. 112

Figure 6. 2 Samples of parallel series of coronal sections cut throughout rats’ cortex ............. 114

Figure 6. 3 Summary of the immunohistochemical scores (NeuN, GFAP and ED1) ............... 115

Page 19: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xix

List of appendices

Appendix 1– Updated Table 1.2 and Table 1.3 ...................................................................... 148

Appendix 2- In vivo evaluation of the anti-epileptic effects of laser cut lacosamide loaded

polymers implanted sub-durally in GAERS rats .................................................................... 153

Page 20: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xx

Abbreviations

α-MSH Neuropeptide α-melanocyte stimulating hormone

aCSF Artificial cerebrospinal fluid

AED Antiepileptic drug

AMPA α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

ANG Angiopep

BBB Blood brain barrier

BCSFB Blood-CSF barrier

BID “bis in die” - Twice a day

CEC Triblock copolymers synthesized by non-catalyzed ring opening

polymerization of caprolactone in the presence of PEG

CNS Central nervous system

CSF Cerebrospinal fluid

D Day

DAPI 4',6-diamidino-2-phenylindole

DMF Dimethylformamide

DPX Mountant for histology made of distyrene, a plasticizer, and

xylene

DS Dextran sulfate

EEG Electroencephalogram

EOE End of experiment

ERHNPs Electro-responsive hydrogel nanoparticles

EVA Ethylene vinylacetate

FAD Fatty acid dimer

FESEM Field emission scanning electron microscope

GαM Goat anti mouse

Page 21: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xxi

GAERS Genetic absence epilepsy rats from Strasbourg

GABA Gamma-aminobutyric acid

GFAP Glial fibrillary acidic protein

H&E Hematoxylin and Eosin

HPLC High performance liquid chromatography

HPMC Hydroxypropyl methylcellulose

IgG Immunoglobulin G

IHC Immunohistochemical

IP Intraperitoneal

LPSPs Lipid protein sugar particles

MK-801 Methyl-lO,ll-dihydro-SH-dibenzo[a,d]cyclohepten-5,10-imine

maleate

NA Not available

NBF Neutral buffered formalin

NeuN Neuron-specific nuclear protein

NMDA N-methyl-D-aspartate

NPs Nanoparticles

NPV Negative predictive value (NPV)

PBLG Polybenzyl l-glutamate

PCL Polycaprolactone

PDLLA Poly(D,L-Lactide)

PECA Polyethylcyanoacrylate

PEO Poly(ethylene oxide)

PEG Polyethyleneglycol

PHBV Poly(3-hydroxybutyrate-co-hydroxyvalerate)

PHT Phenytoin

Page 22: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

xxii

PLA Poly(D,L-lactide)

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

PNP N,N-dimethylacrylamide-based pegylated polymeric

nanoparticles

PPV Positive predictive value

PPy Polypyrrole

PTZ Pentylenetetrazole

PVA Polyvinyl acetate

PVP Polyvinyl pyrrolidone

Py Pyrrole

RNS Responsive neurostimulation

RT Room temperature

SA Sebacic acid

SQ Subcutaneous

STTP Sodium tripolyphosphate

SWD Spike-and-wave discharges

TRH Thyrotropin-releasing hormone

WE Working electrode

WK Week

Page 23: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 1 -

Chapter 1 – A new approach to the treatment of epilepsy.

1.1. Epilepsy

1.1.1. What is epilepsy?

The international league against epilepsy (ILAE) proposed in 2005 a conceptual definition of

epileptic seizures and epilepsy, and more recently in 2014 a practical clinical definition of

epilepsy.(Fisher, Vickrey et al. 2000, Fisher, van Emde Boas et al. 2005, Fisher, Acevedo et

al. 2014) Following those guidelines, epilepsy is defined as a “disease” of the brain

characterised by a predisposition to have “transient occurrence of signs and/or symptoms due

to abnormal excessive or synchronous neuronal activity in the brain” (i.e. seizures).(Fisher,

van Emde Boas et al. 2005, Fisher, Acevedo et al. 2014) To be diagnosed with epilepsy the

patient must have had “at least two unprovoked seizures occurring more than 24 hours

apart”, or “one unprovoked seizure and a probability of further seizures similar to the

general recurrence risk (at least 60%) after two unprovoked seizures, occurring over the next

10 years”, or had a “diagnosis of an epilepsy syndrome”.(Fisher, Acevedo et al. 2014)

The incidence of epilepsy in most developed countries is between 50 and 100 cases per

100,000 population per year and the prevalence is approximately 5 to 8 cases per 1,000.

Although it is estimated that up to 5% of a population will experience non-febrile seizures at

some point in life.(Shorvon 1996, Sander 2003)

1.1.2. Seizure types and classifications

During an epileptic seizure, the abnormal function of the neuronal cells can affect one limited

area of the brain (inducing a partial seizure) or the whole brain (inducing a primarily

generalised seizure).

If during a partial seizure the awareness, memory and consciousness of the patient is

preserved the seizure is referred as a simple partial seizure, but if the patient cannot recall the

event the seizure is referred as a complex partial seizure. The effects of a partial seizure will

be influence by the part of the brain that is affected.

A primarily generalised seizure starts with a bilateral spread of electrical discharges followed

by generalised movements and unconsciousness. There are three main types of primarily

Page 24: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 2 -

generalised seizures: 1) tonic-clonic seizures (or grand mal seizure) characterised by tonic

stiffening of the patient followed by clonic jerking movements, 2) absence seizures when the

patient stays immobile and seems disconnected from its surrounding for a few seconds to a

few minutes, and 3) myoclonic seizures characterised by jerking movements of the patient

that last only a few seconds.

1.1.3. Pathophysiology of epilepsy

Brain injuries and developmental abnormalities are two main categories of causes of seizures

although 50% of seizures remain idiopathic. The causes of seizures are age dependent with

for example children being likely to develop seizures due to birth trauma and congenital

abnormalities, whereas middle age patients are likely to develop seizures due to alcohol and

medications side effects, and elderly patients are likely to develop seizures due to sequels of

strokes and brain tumours.

There is a genetic component that is of particular importance for primarily generalised

seizures (tonic-clonic, absence and myoclonic seizures). Multiple gene abnormalities may be

involved that will affect the excitability of the brain predisposing the patient to for example

chemical imbalances resulting in a higher likelihood of seizure occurrence.(Engel and Pedley

2008)

1.1.4. Diagnosis of epilepsy

An anamnesis can provide strong suggestions of an epileptic activity especially when a clear

description of the events is coming from an observer.(Malmgren, Reuber et al. 2012,

Tolaymat, Nayak et al. 2015) Although not always feasible, performing an inpatient video

recorded electroencephalogram (EEG) will allow confirmation of the diagnosis.(Malmgren,

Reuber et al. 2012, Tolaymat, Nayak et al. 2015) Finally, magnetic resonance imaging (MRI)

is the current gold standard for non-invasive structural evaluation of the brain and is used to

diagnose epilepsy in patients with a clear brain lesion (e.g. contusion, tumour, stroke,

dysplasia, etc.…).(Urbach 2013, Tolaymat, Nayak et al. 2015)

Page 25: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 3 -

1.2. Conventional treatments of epilepsy

1.2.1. Anti-epileptic drug (AED) therapy

The diversity of the aetiology of epilepsy is largely responsible for the challenge that faces a

clinician when prescribing an anti-epileptic drug (AED). A long term AED treatment is

usually not prescribed after a single epileptic seizure event but after determination of the

epileptic syndrome that the patient may suffer from. The different epileptic syndromes are

associated with different treatment options and the choice of the first line AED will be done

in consideration of the safety profile of the AED, the co-morbidities that the patient may

suffer from and the concomitant medications prescribed to the patient.(Miller and Goodkin

2014) Changing or adding another AED is usually done if the first line AED at high dose

does not control seizures or if it induces severe side effects. Due to the poor understanding of

the pathophysiology of epilepsy, AEDs aim at abolishing symptoms but very little progress is

made in curing the underlying cause of epilepsy. Considering the great range of AEDs

commercialised (more than 24 presently), the success rate of AED treatments remains low

with only 50% the patients being seizure free after a first line AED and 66% ultimately

controlled after changes in the AED therapy.(Miller and Goodkin 2014) Even though the

quality of life of patients may have progressed with the apparition of newer AEDs, the

percentage of AED-resistant epilepsy has not significantly decreased.(Sankaraneni and

Lachhwani 2015)

As for anaesthetic drugs, the mechanism of action of antiepileptic drugs on the CNS remains

mostly unknown. There is evidence that certain AEDs interact with certain receptors and

based on those interactions some mechanisms of actions are hypothesised. Some AEDs

interact with voltage-gated cation channel (Na+, Ca

++ and/or K

+) reducing trans-membrane

cation gradient and inhibiting the spread of seizure activity within the motor cortex. Some

other AEDs enhance the effects of the inhibitory neurotransmitter gamma-amino butyric acid

(GABA) and/or weaken the effects of the excitatory neurotransmitter glutamate. Finally,

other potential mechanisms of action have been reported such as inhibition of carbonic

anhydrase, although its implication in the anti-epileptic action of AED remains controversial.

The speculated characteristics and mechanisms of action of common AEDs are presented in

table 1.1.(Rho, Donevan et al. 1994, Perucca 1997, Gurbanova, Aker et al. 2006, Taylor,

Page 26: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 4 -

Angelotti et al. 2007, Chadwick, Shukralla et al. 2009, Hofler and Trinka 2013, Vajda and

Eadie 2014, Sankaraneni and Lachhwani 2015, Calandre, Rico-Villademoros et al. 2016,

Chong and Lerman 2016, Vickery, Tillery et al. 2017). The spectrum of activity relates to the

type of seizure the particular AED has been reported to treat. Broad spectrum AEDs are used

for a wide variety of seizures whereas narrow spectrum AEDs are used for specific types of

seizures (e.g. partial, focal, or absence, myoclonic seizures).

Page 27: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 5 -

Table 1. 1 Main Mechanisms of action of common AEDs.

Drug name Generation Spectrum

Interaction with voltage-gated cation channel reducing trans-

membrane cation gradient

Enhancement of

the effects of the inhibitory

neurotransmitter

GABA

Inhibition of the

effects of the excitatory

neurotransmitter

Glutamate

Others / Comments

Na+

Channel

Ca++

channel

K+

channel

Carbamazepine First Narrow ✔ ✔ ✔

Diazepam First Broad ✔ ✔

Effects on Na+ and Ca++ channels are not typical, allosteric

Modulation of GABA receptor,

Ethosuximide First Specific absence seizure

Block thalamic "T" type Ca++ channel

Ezogabine

Narrow

Discontinued in several country

Felbamate Second Broad

Activation of GABA receptor, NMDA receptor blockade

Gabapentine Second Narrow

Binds α2−δ subunit of L-type voltage-regulated Ca++ channels

Lacosamide Third Broad ✔

Lamotrigine Second Specific absence seizure ✔ ✔

Presynaptic inhibition of neurotransmission (e.g., GLU, ASP)

Levitracetam Second Broad

Binds to synaptic vesicle protein 2A

Oxcarbazepine Second Narrow ✔

Similar mechanism of action than carbamazepine

Perampanel Third Narrow

NMDA receptor antagonist, AMPA-receptor non-competitive

antagonist

Phenobarbitol First Broad

Prolong the opening of the Chloride anion channel within the GABA receptor

Phenytoin First Narrow ✔ ✔

Pregabaline Second Narrow

Binds alpha2delta subunit of L-type voltage-gated Ca++

channels

Tiagabine

Narrow

Inhibition of GABA reuptake from synaptic cleft

Topiramate Second Broad ✔ ✔

Activation of GABA receptor, inhibition of AMPA, carbonic

anhydrase inhibitor

Valproic acide First Broad ✔

Reduced release and/or effects of excitatory amino acids

(NMDA?), modulation of dopaminergic and serotoninergic transmission

Vigabatrin Third Narrow

Irreversible inhibition GABA-transaminase

Zonisamide Second Broad ✔ ✔

Carbonic anhydrase inhibitor

AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; GABA, Gamma-aminobutyric acid; NMDA, N-methyl-D-aspartate.

Page 28: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 6 -

1.2.2. Epilepsy neurosurgery

When the patient’s epilepsy is refractory to AED therapy, surgical excision of brain tissue

responsible for initiation of the seizure sometimes can be achieved. However this procedure

can only be performed on selected patients (35%) depending on the size and the location of

the epileptic foci.(Kwan and Sperling 2009, de Flon, Kumlien et al. 2010, Miller and

Goodkin 2014) The success of inducing long-lasting seizure remission from epilepsy surgery

ranges from a low of 25% for patients exhibiting extra-hippocampal seizure origin to 90% in

patient with hippocampal sclerosis.(Kwan and Sperling 2009, Miller and Goodkin 2014)

Such surgery also comes with potential psychiatric (e.g. changes in cognitive functions),

neurologic (e.g. stroke in up to 4% of the patients) and medical complications (e.g. local

infection, venous thrombosis or pulmonary embolism in up to 12% of the patients).(Kwan

and Sperling 2009, Miller and Goodkin 2014)

1.2.3. Vagus nerve Stimulation (VNS)

When a patient suffers from an AED refractory epilepsy and is not a suitable candidate for

epilepsy neurosurgery, vagus nerve stimulation can reduce the incidence of seizures by 50%

although very few patients become seizure free after this treatment.(Uthman, Reichl et al.

2004, Ardesch, Buschman et al. 2007, Aihua, Lu et al. 2014, Orosz, McCormick et al. 2014)

In consequence it is used as an adjunct therapy to AEDs. Similar to a cardiac pacemaker, a

subclavicular implanted pacer sends intermittent electrical impulse to the vagus nerve. The

mechanism of action is unclear although vagus nerve stimulations could disrupt several

thalamic and brains stem nuclei synaptic transmissions leading to desynchronization of the

thalamocortical connections.(Miller and Goodkin 2013) Most common side effects of the

treatment include hoarseness and coughing during stimulations.(Uthman, Reichl et al. 2004,

Ardesch, Buschman et al. 2007). Also, a long term follow up study has shown that 44% of

the patients wished to have the implant disconnected, when the batteries need to be replaced,

due to lack of efficacy.(Uthman, Reichl et al. 2004)

1.2.4. Responsive neurostimulation (RNS)

Only approved by the FDA in 2013, responsive neurostimulation devices send imperceptible

electrical stimuli to seizure onset areas with abnormal electrographic patterns. As of 2017, only

1000 of those devices were implanted and moderate-to-low-quality evidence supports the

efficacy and safety of RNS.(Boon, De Cock et al. 2018)

Page 29: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 7 -

Figure 1.1 provides a summary of success and failure rates of the conventional anti-epileptic

treatments.

Figure 1. 1- Summary of success and failure rates of the conventional anti-epileptic treatments.

Out of 100 patients diagnosed with epilepsy, 50 will have their seizures controlled after a first line

anti-epileptic drug (AED) treatment and another 16 after refinement of the AED treatment. Out of the

34 patients that remain epileptic approximately 12 will be eligible to undergo epilepsy surgery with 7

of whom will be treated successfully. Out of the 27 patient that are still AED resistant, non-eligible

for surgery or in whom the surgery failed, 3 will be seizure free as a response to the vagal nerve

stimulation. In conclusion, out of the 100 patients diagnosed with epilepsy, 24 will remain epileptic

after all the conventional treatments.

Page 30: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 8 -

1.2.5. Consequences of epilepsy on patient life style

1.2.5.1. Severe impact of epilepsy on the patient life style

A study by Fisher et al. (2000) from the Barrow Neurological Institute and the University of

Arizona has shown that patients with epilepsy are less educated, more likely to be

unemployed and single, and come from lower income households when compared to the US

census bureau norms.(Fisher, Vickrey et al. 2000) Those patients also have a great fear of

experiencing another seizure and show ongoing psychological problems. Their lifestyle has

important limitations such as the inability to drive and limited socialisation. Those patients

have effected cognitive functions which decreased their ability to remember and

concentrate.(Fisher, Vickrey et al. 2000) They are also afraid of missing medication and of its

consequences for their safety and of those around them.(Fisher, Vickrey et al. 2000)

1.2.5.2. Seizures prediction is becoming reality

Being able to predict when a seizure will occur would greatly improve patient safety and

would increase their independence. In a recent multicentre study including 15 drug-resistant

epileptic patients, Cook et al. (2013) has shown that “intracranial electroencephalography

monitoring is feasible in ambulatory patients with drug-resistant epilepsy” and have

established proof of concept that seizure prediction is possible, although larger scale studies

will be needed to confirm it.(Cook, O'Brien et al. 2013) The prediction of the seizure

likelihood could also lead to new therapeutic strategies including acute AED administration

when a seizure is likely to occur. Also, delivering the AED directly over its site of action

would increase its efficiency when the time window for drug delivery can be as short as a few

seconds.

1.3. AED loaded polymer implant as a novel treatment for epilepsy

A review of the literature investigating AEDs administration using polymer-based drugs was

published by Halliday et al. (2012). Up to date summaries are presented in table 1.2 and

table1.3 for in vitro and in vivo experiments respectively.

Page 31: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 9 -

Table 1. 2 Anti-epileptic drugs delivered using polymer-based devices (in vitro experiments).

Drug Polymer Device type In vitro release time Release profile Paper

Acetazolamide Eudragit Injectable 10 hours (EOE) however most within 4 hours Biphasic (Duarte, Roy et al. 2007)

Acetazolamide Eudragit Injectable 7 hrs (EOE) Linear (Haznedar and Dortunc 2004)

Adenosine EVA Implantable 17 days (EOE) however most within 4 days Triphasic (Boison, Scheurer et al. 1999)

Adenosine Silk Implantable 14 days (EOE) Biphasic (Wilz, Pritchard et al. 2008)

Adenosine Silk Implantable 14 days Linear (Pritchard, Szybala et al. 2010)

Adenosine Silk Implantable 21 days Triphasic (Szybala, Pritchard et al. 2009)

Carbamazepine Chitosan Injectable 3 hours (EOE) however most within 2 hours Linear (Gavini, Hegge et al. 2006)

Carbamazepine Chitosan, HPMC Injectable 5 hours (EOE) Biphasic (Filipovic-Grcic, Perissutti et al. 2003)

Carbamazepine Eudragit, HPMC Injectable Between 30 and 60 minutes Biphasic (Dong, Maincent et al. 2007)

Carbamazepine Gelucire Injectable 10 minutes (EOE) Linear (Passerini, Perissutti et al. 2002)

Carbamazepine PECA Injectable 4 hours (EOE) Biphasic (Fresta, Cavallaro et al. 1996)

Carbamazepine PLGA Injectable 24 hours (EOE) Biphasic (Barakat and Radwan 2006)

Carbamazepine Precifac Injectable 12 hrs (EOE) Biphasic (Barakat and Yassin 2006)

Carbamazepine PVP/PVA Injectable 1 hr (EOE) Linear (Patterson, James et al. 2008)

Clonazepam PCL/PEG Implantable Up to 250 days (EOE) Biphasic (Cho, Han et al. 1999)

Clonazepam PBLG/PEO Injectable 8 days (EOE) Linear (Jeong, Cheon et al. 1998)

Clonazepam CEC Injectable 100 hours (EOE) NA (Ryu, Jeong et al. 2000)

Clonazepam PLGA Injectable 24 hrs (EOE) Linear (Montanari, Cilurzo et al. 2001)

Clonazepam PLGA Injectable 8 to 9 days (EOE) Biphasic (Benelli, Conti et al. 1998)

Clonazepam PLGA Injectable 7 days (EOE) Biphasic (Nah, Paek et al. 1998)

CNF PLGA (microspheres) Injectable 70 days Biphasic (Nkansah, Tzeng et al. 2008)

CNF PLGA (nanospheres) Injectable Most after 14 days Triphasic (Nkansah, Tzeng et al. 2008)

Diazepam EVA Implantable 21 days (EOE) Linear (Haik-Creguer, Dunbar et al. 1998)

Diazepam PHBV (microspheres) Injectable 30 days (EOE) Triphasic (Chen and Davis 2002)

Diazepam (Diazepam/Gelatine) in PHBV

microspheres Injectable 30 days (EOE) Biphasic (Chen and Davis 2002)

Diazepam PHBV/Gelatine (microcapsules) Injectable 30 days (EOE) Linear (Chen and Davis 2002)

Diazepam PLA Injectable NA NA (Bodmeier and McGinity 1987)

Diazepam PDLLA Injectable 8 days, most after 1 day Biphasic (Giunchedi, Conti et al. 1998)

Diazepam PLGA Injectable Most within 10 h Linear (Bohrey, Chourasiya et al. 2016)

Ethosuxamide PECA Injectable 4 hrs (EOE), most after 3 hours Biphasic (Fresta, Cavallaro et al. 1996)

Ethosuxamide PLGA Injectable 24 hrs (EOE) Linear (Montanari, Cilurzo et al. 2001)

GABA EVA Implantable 24 to 72 hours Biphasic (Kokaia, Aebischer et al. 1994)

GABA PNP Injectable 6 hrs (EOE) Linear (Yurtdas Kirimlioglu, Menceloglu et al. 2016)

Gabapentine Albumine nanoparticles coated with

polysorbate Injectable 24 hours (EOE) Biphasic (Wilson, Lavanya et al. 2014)

Page 32: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 10 -

Table 1.2. Anti-epileptic drugs delivered using polymer-based devices (in vitro experiments) (cont’d).

Drug Polymer Device type In vitro release time Release profile Paper

Lacosamide PLGA Injectable/

implantable Up to 250 hours (EOE) NA (Chen, Gu et al. 2017)

Levetiracetam PLGA Implantable Most within 3 days NA (Halliday, Campbell et al. 2013)

Lorazepam PLGA Injectable 24 hours (EOE) Biphasic (Sharma, Maheshwari et al. 2014)

MK-801 EVA Implantable 60 days (EOE) however most within 30 days Biphasic (Smith, Cordery et al. 1995)

Muscimol LPSPs Injectable 5 days however most within 20 hours Biphasic (Kohane, Holmes et al. 2002)

Noradrenaline EVA Implantable Within 24 hours Biphasic (Kokaia, Aebischer et al. 1994)

Oxcarbazepine Chitosan Injectable NA NA (Rane, Mashru et al. 2007)

Phenobarbital PLGA Injectable NA NA (Barichello, Morishita et al. 1999)

Phenobarbitone PCL Injectable NA NA (Berrabah, Andre et al. 1994)

Phenytoin EVA Implantable 105 days (EOE) Biphasic (Tamargo, Rossell et al. 2002)

Phenytoin PCL Injectable 22 days (EOE) however mostly within 12 days Triphasic (Li, Li et al. 2007)

Phenytoin PECA Injectable 4 hrs (EOE) Biphasic (Fresta, Cavallaro et al. 1996)

Phenytoin PCL Injectable 30 days (EOE) however mostly within 4 days Triphasic (Jiang, Yue et al. 2015)

Phenytoin ANG-ERHNPs Injectable 24 hours (EOE) Triphasic (Ying, Wang et al. 2014)

Phenytoin ANG-ERHNP Injectable 4 hours (EOE) (with electrical field stimulation) NA (Wang, Ying et al. 2016)

Pregabalin Alginate/HPMC Injectable 12 hours (EOE) Biphasic (Madan, Adokar et al. 2015)

Primidone PCL Injectable 10 hours (EOE) however most within 1 hours Triphasic (Ferranti, Marchais et al. 1999)

Valproate Hexadecanol Injectable 3 hours Biphasic (Giannola, De Caro et al. 1993)

Valproate PLGA Injectable NA NA (Barichello, Morishita et al. 1999)

ANG, Angiopep; CEC, Triblock copolymers synthesized by non-catalyzed ring opening polymerization of caprolactone in the presence of PEG;

CNF, Ciliary neurotrophic factor; EOE, End of experiment; ERHNPs, Electro-responsive hydrogel nanoparticles; EVA, Ethylene vinylacetate;

GABA, Gamma amino butyric acid; HPMC, Hydroxypropyl methylcellulose; LPSPs, Lipid protein sugar particles; MK-801, Methyl-lO,ll-

dihydro-SH-dibenzo[a,d]cyclohepten-5,10-imine maleate; NA, Not available; PBLG, Polybenzyl l-glutamate; PCL, polycaprolactone; PDLLA,

Poly(D,L-Lactide); PECA, Polyethylcyanoacrylate; PEO, Poly(ethylene oxide); PEG, Polyethyleneglycol; PHBV, Poly(3-hydroxybutyrate-co-

hydroxyvalerate); PLA, Poly(dl-lactide); PLGA, Poly-lactide-co-glycolide; PNP, dimethylacrylamide-based pegylated polymeric nanoparticles;

PVA, Polyvinyl acetate ; PVP, Polyvinyl pyrrolidone ; SA, Sebacic acid.

Page 33: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 11 -

Table 1. 3 Anti-epileptic drugs delivered using polymer-based devices (in vivo experiments).

Drug Polymer Device type Animal model Delivery In vivo results Paper

Adenosine EVA Implantable Kindled rats Rod, unilateral implantation in ventricle

1 week seizure reduction (Boison, Scheurer et al. 1999)

Adenosine Silk Implantable Kindled rats Rod, unilateral implantation

within the infrahippocampal cleft

Dose-dependent delay in kindling acquisition for 11 days, and reduce after

discharge duration

(Wilz, Pritchard et al. 2008)

Adenosine Silk Implantable Kindled rats Rod, unilateral implantation

within the infrahippocampal cleft

Protection from generalized seizures over

a period of 10 days of suppressed seizures, delay in epileptogenesis

(Szybala, Pritchard et al.

2009)

Carbamazepine Chitosan Injectable Sheep Powder Intranasal Three hours increased drug concentration in the serum when compared to the nasal

administration of the pure drug

(Gavini, Hegge et al. 2006)

Carbamazepine Eudragit, HPMC Injectable Rabbits Oral (powder in capsules) Ten hours increased plasma levels (Dong, Maincent et al. 2007)

Clonazepam PCL/PEG Implantable Rabbits IP pellet

Constant plasma concentration of

clonazepam for 45 days after initial burst effect.

(Cho, Han et al. 1999)

GABA EVA Implantable Kindled rats

Bilateral implantation into the

mesencephalon dorsal to the

substanti nigra

At 48 h, generalized convulsions were

prevented

(Kokaia, Aebischer et al.

1994)

GABA PNP Injectable Rats, PTZ-induced

acute seizure model IP nanoparticules Shorter seizure and decrease in mortality

(Yurtdas Kirimlioglu,

Menceloglu et al. 2016)

Gabapentine

Albumin

nanoparticles

coated with polysorbate

Injectable Rats, PTZ and electrochock induced

convulsion

IP nanoparticules Increase gabapentin brain concentration,

decrease duration of all phases of convulsion

(Wilson, Lavanya et al.

2014)

Levetiracetam PLGA Implantable

Rats, ,hippocampal

tetanus toxin model of mesial temporal lobe

epilepsy

Bilateral implantation above motor and somatosensory cortices

Trend towards a reduction in seizure frequency

(Halliday, Campbell et al. 2013)

Lorazepam PLGA Injectable Rats Intranasal Increased brain concentration for up to 8

hours

(Sharma, Maheshwari et al.

2014)

Lorazepam PLGA Injectable Sheep nasal mucosa

Ex vivo, biphasic controlled release 58% (24

hours)

(Sharma, Maheshwari et al.

2014)

MK-801 EVA Implantable Unknown

Implanted over brain area for 28

to 65 days followed by in vitro

release studies

Still releasing drug after 60 days in vivo

(EOE). The in vitro release profiles appears

to predict accurately the in vivo release.

(Smith, Cordery et al. 1995)

Page 34: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 12 -

Table 1.3 Anti-epileptic drugs delivered using polymer-based devices (in vivo experiments) (cont’d).

Drug Polymer Device type

Animal model Delivery In vivo results Paper

Muscimol LPSPs Injectable Rats, hippocampal injection

of pilocarpine

Unilateral injection into the

hyppocampus (i.e. seizure focus)

Prevents seizures formation for up to 120

min (EOE)

(Kohane, Holmes et al.

2002)

Noradrenaline EVA Implantable Rats, interventricular treatment with 6-

hydroxydopamine

Bilateral implanation of matrices

and electrode in the hypocampus No change in kindling acquisition

(Kokaia, Aebischer et al.

1994)

Phenytoin EVA Implantable Cobalt-induced rat model of epilepsy

Unilateral implantation in cortical seizure focus

reduction in seizure activity for 6 days (EOE)

(Tamargo, Rossell et al. 2002)

Phenytoin EVA Implantable Rat Two weeks in vitro release study following 365 days intracortical implantation

NA (Tamargo, Rossell et al.

2002)

Phenytoin PCL Injectable Rats, hippocampal injection of tetanus toxin

Unilateral implantation into cortical seizure focus

Significant reduction in epileptic events for 3 days with no observed clinical side effects.

(Jiang, Yue et al. 2015)

Phenytoin ANG-ERHNPs

Injectable Amygdala-kindled seizures in rats

IP nanoparticles

PHT high concentrations in the hippo-campus, amygdale, cerebellum, and brainstem regions, decrease in the kindling-

stimulation-induced seizures

(Ying, Wang et al. 2014)

Phenytoin ANG-ERHNP Injectable

Mices, electrical- (maximal

electrical shock) and chemical-induced (pentylenetetrazole and

pilocarpine) seizure models

IP nanoparticles lowered the effective therapeutic doses of PHT and demonstrated the improved anti-seizure effects

(Wang, Ying et al. 2016)

Piperine Chitosan/STPP NPs

Injectable Mices, PTZ-induced kindling model

IP nanoparticles Enhance the neuroprotection and ameliorate the astrocytes activation

(Anissian, Ghasemi-Kasman et al. 2017)

TRH FAD/SA Implantable Kindled rats Unilateral implantation in amygdala

Reduced seizure duration for 50 days (EOE) (Kubek, Liang et al. 1998)

Valproate PCL Implantable Rats, necocortical injected

tetanus toxin model

Implanted above the cortical

seizure focus

Decrease in epileptiform potential however

the model failed to induce consistent seizures.

(Rassner, Hebel et al.

2015)

ANG, Angiopep; EOE, End of experiment; ERHNPs, Electro-responsive hydrogel nanoparticles; EVA, Ethylene vinylacetate; FAD, Fatty acid

dimer; GABA, Gamma amino butyric acid; HPMC, Hydroxypropyl methylcellulose; IP, Intraperitoneal; LPSPs, Lipid protein sugar particles; MK-

801, Methyl-lO,ll-dihydro-SH-dibenzo[a,d]cyclohepten-5,10-imine maleate; NA, Not available; NPs, Nanoparticles; PCL, polycaprolactone; PEG,

Polyethyleneglycol; PLGA, Poly-lactide-co-glycolide; PNP, dimethylacrylamide-based pegylated polymeric nanoparticles; PTZ,

Pentylenetetrazole; SA, Sebacic acid; STTP, Sodium tripolyphosphate; TRH, Thyrotropin-releasing hormone.

Page 35: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 13 -

1.3.1. The blood brain barrier, an obstacle for drug delivery to the brain

parenchyma

The central nervous system (CNS) is a delicate organ that can only maintain its normal

function within a narrow homeostatic range that is achieved through a strict control of all

exchanges between the blood and the neural tissue.(Serlin, Shelef et al. 2015) Three key

elements play a major role in that control: 1) the blood brain barrier (BBB) with an extremely

specific endothelial cells layer, 2) the blood-CSF barrier (BCSFB) with a cerebro-spinal fluid

(CSF) secreting choroid plexus epithelium and 3) the blood-subarachnoid CSF barrier (i.e.

arachnoid epithelium).(Reese and Karnovsky 1967, Abbott, Ronnback et al. 2006, Serlin,

Shelef et al. 2015). Those barriers play an important protective role for the brain but also

prevent easy access for drugs targeting the CNS.

The specialised endothelial cells of the BBB have continuous tight junctions, no fenestrae,

efflux transporter expression and poor pinocytotic uptake.(Correale and Villa 2009, Abbott,

Patabendige et al. 2010, Hartz and Bauer 2011, Daneman 2012, Lajoie and Shusta 2015) To

be able to diffuse freely across the BBB, a molecule must have a low molecular weight (<400

Da) and be lipid soluble (forms less than 8 hydrogen bonds).(Pardridge 2012) Larger drug

molecules and even smaller ones often do not comply with those characteristic making them

unable to reach the brain parenchyma.(Lipinski 2000) However, the drug could cross the

BBB through carrier-mediated transport systems (e.g. large neutral amino-acid transporter

type 1 in the case of the two following examples) if a particular endogenous substrate has a

similar structure to the drug (e.g. Gabapentine) or if the drug can be conjugated with a

particular substrate (e.g. with the thiol group of cysteine).(Killian, Hermeling et al. 2007,

Pardridge 2012) Larger molecules can also be engineered using a molecular Trojan horse to

access receptor mediated transport systems within the BBB.(Lajoie and Shusta 2015)

Although feasible for some drug molecules, those alternatives to cross the BBB remain

inaccessible for most.

Page 36: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 14 -

1.3.2. Polymer helping AEDs to cross the blood brain barrier

The encapsulation of some AEDS in biodegradable polymers has also allowed better BBB

penetration. Albumin nanoparticles coated with polysorbate 80 and loaded with gabapentin

improved gabapentin brain concentration by up to 3 times when compared to the free drug

administration, after intraperitoneal (IP) administration.(Wilson, Lavanya et al. 2014)

Furthermore, the IP administration of gabapentin encapsulated nanoparticles significantly

reduced the duration of all phases of convulsion in a model of Pentylenetetrazole (PTZ)

induced convulsions.(Wilson, Lavanya et al. 2014) The encapsulation of phenytoin (PHT) in

Angiopep-Conjugated Electro-Responsive Hydrogel Nanoparticles (ANG-ERHNP)

administered intraperitoneally lowered the effective therapeutic doses of PHT and

demonstrated an improved anti-seizure effect when compared with free PHT solution in

several models of epilepsy (i.e. amygdala kindling model, maximal electroshock and PTZ

induced convulsion models)(Ying, Wang et al. 2014, Wang, Ying et al. 2016). Those

nanoparticles are of a particular interest as rapid PHT release could be achieved following the

application of an electrical current.(Ying, Wang et al. 2014) Other IP administrations of

encapsulated AEDs in nanoparticles (i.e. piperine and GABA) have demonstrated positive

results in in vitro release study and/or in in vivo models of epilepsy. (Yurtdas Kirimlioglu,

Menceloglu et al. 2016, Anissian, Ghasemi-Kasman et al. 2017)

Polymer based formulation of AEDs for intranasal or oral administration has also been

studied. (Gavini, Hegge et al. 2006, Dong, Maincent et al. 2007, Sharma, Maheshwari et al.

2014) Using gamma scintigraphy studies, Sharma et al. (2014) found that the brain/blood

concentration ratio of lorazepam was higher when the rats were administered intranasally the

Poly(D,L-lactide-co-glycolide (PLGA) loaded with lorazepam nanoparticles compared to the

free lorazepam formulation.(Sharma, Maheshwari et al. 2014) Also, the pregabalin oral gel

formulation enabled controlled release of the AED for up to 12 hours and the

carbamazepine/Eudragit oral formulation increase plasma concentration of carbamazepine for

up to 10 hours. (Dong, Maincent et al. 2007, Madan, Adokar et al. 2015)

Page 37: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 15 -

1.3.3. Polymer allowing AEDs delivery to the brain parenchyma while

bypassing the blood brain barrier

If crossing the BBB is proven difficult, there are ways to bypass it. These include 1)

administration of AEDs into the CSF and 2) direct topical application of the AEDs onto/into

the brain parenchyma. The administration of drugs directly into/onto the brain parenchyma

and into the CSF is invasive and cannot be repeated frequently unless a cannula is

permanently implanted or a mini-osmotic pump is used. These techniques permanently

compromise the isolation of the brain parenchyma, leaving patients prone to infection and

injury and are not recommended for use in humans. In consequence, polymer devices that can

store and release the drugs over time were implanted onto/into the brain parenchyma and in

the ventricles. This approach has shown some successes in the treatment of animal models of

several neurological disorders such as Parkinson’s disease, Huntington’s disease and

Alzheimer’s disease.(Halliday and Cook 2009) The approach also offers the advantages of

achieving high concentration of drug in the brain with minimal systemic toxicity and would

eliminate the need of the patient for strict compliance to regular drug intake. However in the

field of epilepsy, the report of such techniques remains rare.

1.3.3.1. Implantable polymers delivering AEDs into the cerebrospinal fluid

The administration of drug into the CSF is often as efficient as intravenous drug

administration. The BCSFB produce a constant and rapid flow of CSF allowing molecules to

be eliminated from the brain and into the blood stream. The result is a poor molecular

transport from CSF to brain parenchyma and an efficient molecular transport from CSF to

blood.(Pardridge 2012) Nevertheless, Boison et al. (1999) have demonstrated that ethylene

vinylacetate (EVA) rods loaded with adenosine implanted in the ventricles of kindled rats

induced a 75% reduction of grade 5 seizures over 7 days and a reduction of the amplitude and

duration of after-discharges.(Boison, Scheurer et al. 1999)

1.3.3.2. Implantable polymers delivering AEDs into/onto brain parenchyma

Monolithic or implantable polymers (e.g. rods and sheets) are solid devices that can be

implanted onto the brain parenchyma or into the brain parenchyma adjacent to deeper seizure

focuses. Out of 22 in vivo experiments investigating polymer-based drug delivery devices

Page 38: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 16 -

(table 1.3.) for the treatment of epilepsy, 12 used implantable polymers, including the first

original study.

To the best of the author’s knowledge, the first prototypes were studies by Kokaia et al. in

1994. In that study, EVA polymer matrices were used to deliver GABA into the

mesencephalon of rats subjected to amygdala kindling as a model of epilepsy.(Kokaia,

Aebischer et al. 1994) The implantation of the GABA loaded polymer significantly reduced

the severity of seizures at 48 h post-surgery when compared to the control polymer. The

authors concluded that the GABA loaded polymer was not only inhibiting epileptogenesis but

also dampened seizure generalisation in already established kindling epilepsy.

The kindled rats model was also used in 3 other publications evaluating the antiepileptic

efficacy of implantable polymer loaded with adenosine or thyroid releasing hormone (TRH).

In 2 studies by Wiltz et al. (2008) and Szybala et al. (2009), adenosine loaded silk polymers

implanted within the infrahippocampal cleft protected rats from general seizure for a period

of up to 11 days. (Wilz, Pritchard et al. 2008, Szybala, Pritchard et al. 2009) In the third

study, by Kubeck et al. (1998), utilizing a kindling model of temporal lobe epilepsy,

biodegradable polyanhydride copolymers loaded with TRH and implanted into the seizure

focus (i.e. amygdala) achieved a reduction in clonus duration for a period of 50 to 60

days.(Kubek, Liang et al. 1998)

Finally, Tamargo et al. (2002) demonstrated a reduction in seizure activity for 6 days using

EVA polymer loaded with phenytoin in a cobalt-induced rat model of epilepsy.(Tamargo,

Rossell et al. 2002)

1.3.3.3. Injectable polymers delivering AEDs into/onto brain parenchyma

Although implantation of monolithic polymers deeper into the brain parenchyma is feasible,

there is a significant risk of disrupting superficial layers of the cortex inducing permanent

post-implantation neurological deficits. Injectable polymers are particles of different sizes

(i.e. nano or micro-particles) that can be reconstituted in solution and injected at a precise

location into the brain parenchyma. Injectable polymers allow a controlled delivery of AEDs

adjacent to deep seizure focuses that cannot be surgically resected due to multiple

reasons.(Halliday, Moulton et al. 2012).

Page 39: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 17 -

To the best of the author’s knowledge only 2 studies have been published reporting the use of

injectable polymers for in vivo delivery of AEDs into the brain parenchyma. The first by

Kohane et al. (2002) studied the antiepileptic effects of muscimol delivered adjacent to the

hippocampal seizure focus using lipid protein sugar particles (LPSPs).(Kohane, Holmes et al.

2002) Rats were pre-treated with saline loaded LPSPs or muscimol loaded LPSP, and

seizures were induced with hippocampal injections of pilocarpine. That study demonstrated

that muscimol loaded LPSPs prevented seizure formation during the entire experimentation

period of 120 minutes.

A more recent study by Jiang et al. (2015) studied the antiepileptic effects of phenytoin

loaded polycaprolactone (PCL) microspheres.(Jiang, Yue et al. 2015) Phenytoin-PCL or

blank-PCL microspheres were injected into the hippocampus of a rat tetanus toxin model of

temporal lobe epilepsy. The results demonstrated that phenytoin-PCL microspheres induced a

significant reduction in epileptic events for 3 days with no observed clinical side effects.

1.4. Initial development of an implantable drug delivery device for the

treatment of epilepsy

As previously discussed, to the best of the author’s knowledge there is no report

demonstrating that polymer devices loaded with commonly used AEDs and implanted onto or

into the brain parenchyma can decrease seizure activity for a sustained period. Most reports

of positive effects lasted from a few hours to a few days. The implantation of a polymer-

based drug delivery device onto or into the brain parenchyma requires an invasive

neurosurgery. The repetition of such a procedure can only exacerbate the associated risks

encountered by the patient such as infection, stroke, brain tissue damage and haemorrhage, all

of which can endanger the life of the patient or lead to long term neurological deficits. In

consequence, the AED/polymer device will not only need to be biocompatible and

efficacious but also capable of controlled and long term release of the AED.

The present thesis aims to report the initial development of polymer based biodegradable

implantable drug delivery devices loaded with commonly used AEDs and tested in an

animal model of absence epilepsy.

Page 40: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 18 -

1.4.1. Animal model used: GAERS

Genetic Absent Epilepsy Rats from Strasbourg (GAERS) is a strain of rats that 100% of the

animals present with recurrent generalized non-convulsive seizures.(Marescaux, Vergnes et

al. 1992) This animal model has become the gold standard to study the mechanisms

underlying absence epilepsy. GAERS’ EEGs present typical and very consistent spike and

wave discharges (SWDs) of 0.3 to 1 mV in amplitude and 7 to 11 SWDs per second in spike

rate.(Marescaux, Vergnes et al. 1992, Danober, Deransart et al. 1998) Typical absence

seizures originate from abnormal thalamocortical networks including the thalamocortical

neurons of sensory thalamic nuclei and their main inhibitory input (GABAergic

neurons).(Marescaux, Vergnes et al. 1992, Danober, Deransart et al. 1998) With age the

number, frequency and duration of SWDs increase, to achieve maximum number at the age

of 6 months and full maturity in frequency and duration at the age of 18 months.(Marescaux,

Vergnes et al. 1992)

1.4.2. Antiepileptic drugs used: Phenytoin and Lacosamide

Phenytoin anticonvulsant properties were described for the first time by Putnam and Merritt

in animals in 1937 and a year later in human patients.(Putnam and Merritt 1937, Merritt and

Putnam 1984) After eighty years, the antiepileptic mechanism of action of phenytoin remains

a mystery although phenytoin can be classified as a sodium and calcium channel blocker,

reducing membrane cations’ gradient and inhibiting the spread of seizure activity within its

supposedly primary site of action, the motor cortex.(Keppel Hesselink and Kopsky 2017) The

fact that the structure of phenytoin has two phenyl rings at the C5 position of an hydantoin

molecule seems responsible for phenytoin’s anticonvulsant properties without inducing

sedation.(Keppel Hesselink and Kopsky 2017) The solubility of phenytoin (71 mg/L in

aqueous solution) suggests that the elution of the phenytoin from the PLGA polymer should

be slow, prolonging the release of the AED from the polymer.

Phenytoin may be an “old “drug but it remains as efficacious as new generation AEDs.(Kwan

and Brodie 2000, Kellinghaus, Berning et al. 2014) Phenytoin has a narrow epileptic

spectrum and has been shown to aggravate absence epilepsy when administered systemically.

Nevertheless, it decreases or abolishes brain epileptic activity in two rats model (including

GAERS) when administered directly onto the cortex.(Gurbanova, Aker et al. 2006)

Page 41: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 19 -

Lacosamide is a third generation broad spectrum AED that was only approved in 2008 in the

European Union and USA, and in 2009 in Australia. It was first administered as an adjunct

AED in patient with partial–onset seizure however because of its availability as an

intravenous formulation its use for the treatment of status epilepticus is increasing.(Bauer,

Willems et al. 2017) Studies report the rate of successful treatment of status epilepticus is up

to 100 % in adults treated with lacosamide.(Hofler, Unterberger et al. 2011) In a study

comparing lacosamide and phenytoin, both drugs had similar success rates (33% and 40%

respectively) in treating status epilepticus after failure of benzodiazepines and

levetiracetam.(Kellinghaus, Berning et al. 2014) However lacosamide was associated with

significantly fewer side effects.

Lacosamide is a functionalised amino acid that stabilizes neuronal membranes through the

enhancement of slow inactivation of voltage-gated sodium channel.(Hofler and Trinka 2013,

Bauer, Willems et al. 2017) The solubility of lacosamide (465 mg/L in aqueous solution) is

superior to that of phenytoin which could potentially contribute to a relatively faster elution

of lacosamide from the polymers.

1.4.3. Polymers used: PLGA and PPy

The first polymers to be studied for AEDs delivery (i.e. EVA) were non-biodegradable. The

intra-cranial implantation of non-biodegradable polymer may require the removal of the

polymer, submitting the patient to another neurosurgery and its associated risks. Newer

biodegradable polymers that offer good brain biocompatibility and that are intended for

intracranial implantation are often made of PLGA, PCL, or polylactic acid (PLA) in the form

of implantable sheets or injectable microspheres.(Halliday and Cook 2009, Halliday, Moulton

et al. 2012, Halliday, Campbell et al. 2013, Jiang, Yue et al. 2015, Rassner, Hebel et al. 2015)

In the present thesis two kinds of biodegradable polymer will be used: 1) PLGA polymer

sheets used as devices that continuously deliver AED and 2) Polypyrrole (PPy) polymer

sheets used as devices that can be triggered to deliver AED.

1.4.3.1. PLGA polymer sheets

PLGA copolymers (i.e. lactide and glycolide) are physically strong, biocompatible and

biodegradable. The degradation products of PLGA are glycolic acid and lactic acid, both of

which can be eliminated from the body as carbon dioxide and water.(Bohrey, Chourasiya et

al. 2016) PLGA polymers have been extensively used and are approved by the United States

Page 42: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 20 -

Drug Administration.(Halliday and Cook 2009, Halliday, Moulton et al. 2012, Bohrey,

Chourasiya et al. 2016, Wang, Qu et al. 2017) The structure of the polymer includes a

hydrophilic surface and a hydrophobic core enabling solubilization in aqueous solution and

drug encapsulation that is later released in a continuous way. The rate of the drug delivery

can be altered by modifying the drug concentration and the ratio of lactide to

glycolide.(Makadia and Siegel 2011)

The PLGA sheets were produced using a coaxial electrospinning method (see Chapter 4 and 5

of the present thesis for detail descriptions). Briefly, electrospinning is a relatively simple and

versatile fibre production method consisting of drawing a polymer through a needle onto a

collecting plate using electrostatic forces.(Halliday, Moulton et al. 2012) In a coaxial

electrospining method an outer fluid is drawn with an inner fluid simultaneously (Figure 1.2

A) In our case, the result is a three dimensional porous sheets made of randomly oriented

fibres (Figure 1.2 B).

Figure 1. 2 Poly-lactide-co-glycolide polymer sheets produced by coaxial electrospinning method

(A) Illustration of the co-axial electrospinning process. (B) Scanning electron micrographs of PLGA

coaxially electrospun polymer mats. Scale bar represents 10 µm.

1.4.3.2. PPy polymer sheets

There are more than 25 PPy based conducting polymers used in direct contact with biological

tissues reported in the literature and they all have a conjugated structure of alternating

Page 43: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 21 -

carbon–carbon double bonds (figure 1.3). (Ateh, Navsaria et al. 2006) Upon intermittent

application of an electrical current, the ionic and hydrophobic properties of these polymers

vary resulting in actuation of the polymers.(Wallace 2009) In cases where drugs are

impregnated within the polymers, the expansion and/or contraction of the structure may be

responsible for some drug release.(Hutchison, Lewis et al. 2000, Wallace and Wallace 2003)

The mechanism of drug release has been reported for a wide range of incorporated drugs such

as dexamethasone, fos-phenytoin and the anti-inflammatory neuropeptide α-melanocyte

stimulating hormone (α-MSH).(Zhong and Bellamkonda 2005, Moulton, Imisides et al. 2008,

Muller, Yue et al. 2016)

Synchronizing the AED release from a polymer with a seizure advisory system could be

technically feasible when using a conductive polymer such as PPy. PPy sheets have been

shown in vitro to be well tolerated by biological tissues, including brain tissues, and they

seem to be as compatible as Teflon® when implanted into the cerebral cortex.(George,

Lyckman et al. 2005, Ateh, Navsaria et al. 2006) Nevertheless in vivo biocompatibility needs

first to be proven. The PPy sheets were produced using an electro-polymerization method

(Figure 1.3). The pyrrole is oxidized before being polymerized. The PPy polymer is then

peeled off the anode. Details of the method used are provided in chapter 6.

Figure 1. 3 Polypyrrole polymer sheets produced by electro-polymerization method

1.4.4. Proposed steps for the initial development of an implantable drug

delivery device for the treatment of epilepsy

Page 44: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 22 -

The following steps are proposed in this research project to investigate the application of

polymers as an implantable drug delivery device for the treatment of epileptic seizures.

1) Establishment and evaluation of an automated spike and wave complexes detection

algorithm applied to GAERS EEGs: EEG analysis is time consuming and subjective

and the goal of the study described in chapter 2 is to develop a time efficient and

accurate method for analysing days of EEG recordings.

2) Influence of enrofloxacin on GAERS epileptic activity: Craniotomies are invasive

surgeries that require supportive treatment such as prophylactic antibiotic

administration and the goal of chapter 3 is to evaluate whether enrofloxacin affects

the GAERS epileptic activity as it has been reported in other species.

3) Testing of the anti-epileptic effects of phenytoin loaded PLGA polymers implanted

sub-durally in GAERS.

4) Testing of the anti-epileptic effects of lacosamide loaded PLGA polymers implanted

sub-durally in GAERS.

5) Evaluation of the in vivo biocompatibility of PPy polymers implanted sub-durally in

GAERS.

Page 45: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 23 -

Chapter 2 - Evaluation of an automated spike and wave

complexes detection algorithm applied to GAERS EEGs

2.1 Outline

The assessment of the anti-epileptic efficacy of novel treatments in animal model relies on

the evaluation of typically many hours of electroencephalogram (EEG) recordings. Manual

evaluation of those recordings is time consuming and is very subjective while the

development of automatic seizures detection methods can make those analyses quicker and

more reproducible. The aim of the study presented in chapter 2 was to develop and evaluate

an automated SWD algorithm applied to GAERS’ EEGs.

The algorithm used to automatically mark the onset and termination of SWDs was

implemented using MATLAB® (The MathWorks, Inc., Natick, Massachusetts, USA) and

was based on the well-defined, consistent and described morphology of the

GAERS’SWD.(Marescaux, Vergnes et al. 1992, Rudolf, Bihoreau et al. 2004) A previously

published rigorous assessment method of performance was used.(Navakatikyan, Colditz et al.

2006) A total of 6 hours and 20 minutes of EEG recording were analysed and by comparing

manual (reference) versus automatic (test) markings, the study demonstrated that the tested

algorithm sensitivity, specificity, positive predictive value and negative predictive value were

all over 94%, which is comparable to published methodologies based on analysing changes in

the EEG in the frequency domain.(Sitnikova, Hramov et al. 2009, Van Hese, Martens et al.

2009, Ovchinnikov, Luttjohann et al. 2010) The algorithm described in this chapter analyses

the changes in the EEG in the time domain mimicking manual marking while decreasing

subjectivity. In consequence, it is a good performing alternative to scientists and researchers

without engineering background.

The use of this algorithm allowed substantial time savings and improved reproducibility

when performing EEG analysis during the in vivo experiments of the following chapters.

2.2 Manuscript This chapter is presented as the author-accepted version of a peer-reviewed manuscript

published in Neuroscience Bulletin: Neurosci Bull. 2015 Oct;31(5):601-10. The figures

Page 46: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 24 -

and tables’ numbers and titles of the author-accepted version of the manuscript may have

been changed to follow the formatting of the thesis.

Page 47: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 25 -

Evaluation of an automated spike-and-wave complex detection algorithm in the EEG

from a rat model of absence epilepsy

Sebastien H. Bauquier1, Alan Lai

2, Jonathan L. Jiang

3,4, Yi Sui

5, Mark J. Cook

3,4

1 Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee,

VIC, Australia.

2 Department of Electrical and Electronic Engineering, The University of Melbourne,

Parkville, VIC, Australia.

3 Department of Clinical Neurosciences, St Vincent's Hospital, Fitzroy, Vic, Australia.

4

Faculty of Medicine, The University of Melbourne, Parkville, VIC, Australia.

5 Department of Neurology, Shenyang First People's Hospital, Shenyang, China.

Abstract

The aim of this prospective blinded study was to evaluate an automated algorithm for spike-and-wave

discharge (SWD) detection applied to EEGs from genetic absence epilepsy rats from Strasbourg

(GAERS). Five GAERS underwent four sessions of 20-min EEG recording. Each EEG was manually

analyzed for SWDs longer than one second by two investigators and automatically using an algorithm

developed in MATLAB®. The sensitivity, specificity, positive predictive value (PPV), and negative

predictive value (NPV) were calculated for the manual (reference) versus the automatic (test)

methods. The results showed that the algorithm had specificity, sensitivity, PPV and NPV >94%,

comparable to published methods that are based on analyzing EEG changes in the frequency domain.

This provides a good alternative as a method designed to mimic human manual marking in the time

domain.

Keywords: GAERS; seizure detection; epilepsy.

Introduction

Epilepsy is a chronic neurological condition characterized by recurrent seizures. Its incidence

in most developed countries is between 50 and 100 cases per 100 000 per year[1, 2]

. As

treatment with conventional anti-epileptic drugs provides adequate seizure control in only 2/3

of patients, more translational research in this field is urgently needed[1, 2]

. The efficacy

Page 48: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 26 -

assessment of novel treatments in animal models typically relies on the evaluation of many

hours of electroencephalogram (EEG) recordings. Manual evaluation of these recordings is

time-consuming and very subjective, while the development of automatic seizure-detection

methods can make these analyses quicker and more reproducible.

Genetic absence epilepsy rats from Strasbourg (GAERS) is a strain in which 100% of

the animals exhibit recurrent generalized non-convulsive seizures[3]

; and it has become the

gold standard for studying the mechanisms of absence epilepsy This model is characterized

by the development of well-defined and consistent spike-and-wave discharges (SWDs) even

though their duration and numbers vary between colonies[3, 4]

. Spontaneous SWDs (7–11 Hz,

330-1 000 μV, 0.5–75 s) start and end abruptly in a normal background EEG[3]

.

The algorithm to automatically mark the onset and termination of SWDs was

implemented using MATLAB® and Signal Processing ToolboxTM

R2010b (The MathWorks,

Inc., Natick, Massachusetts, USA). Many groups have published methods that automatically

detect seizures, but they are predominantly designed for studies in humans[5-9]

. The

morphology of SWDs in GAERS is noticeably more consistent than seizures in humans, so

we hypothesized that an algorithm based on the definition of SWD (referred to as the ‘SWD

detection algorithm’) would mark the onset and termination of SWD accurately.

The aim of the present study was to determine whether the SWD detection algorithm

can successfully replace the manual marking of EEG recording with high accuracy. A time-

domain-based assessment of algorithm performance was used to compare the SWD detection

algorithm with human markers[10]

.

MATERIALS AND METHODS

The study was designed as a prospective controlled masked experiment. All experiments

were approved by St Vincent’s Hospital (Melbourne) Animal Ethics Committee and

conducted in accordance with the Australian Code of Practice for the Care and Use of

Animals for Scientific Purposes (2004).

Page 49: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 27 -

Animals

Six-month-old female GAERS, obtained from the University of Melbourne (Parkville,

Victoria, Australia), were used in experiments on novel techniques for the delivery of anti-

epileptic drugs. The first five animals randomly allocated to the control group in that study

were also used in the present study. These animals underwent surgery for the implantation of

EEG recording electrodes and their epileptic activity was evaluated for 8 weeks. They were

housed individually under a 12-h light/dark regime with ad libitum access to food and water.

Recording Electrode Implantation

Immediately before surgery, the rats were weighed and anaesthetized with an intraperitoneal

injection of ketamine (75 mg/kg) and xylazine (10 mg/kg). Following the induction of

anesthesia, each rat was placed in a stereotaxic apparatus and given isoflurane (0.5 to 1% in

oxygen, 1 L/min) via a nose-cone, along with subcutaneous carprofen (5 mg/kg) for pain

relief and 0.9% sodium chloride (2 mL) for cardiovascular support. Then the EEG recording

electrodes were implanted as follows: after preparation of the scalp, a single incision was

made down the midline, the skull cleared of tissue, and the exposed bone dried with 3%

hydrogen peroxide. Four extradural electrodes, consisting of small jeweler’s screws, were

implanted cranial to the interaural line (two on each side of the sagittal suture) and one caudal

to the interaural line to the right of the sagittal suture (Fig. 2.1A). The electrodes were then

connected to an adaptor secured with dental cement. The skin was sutured leaving only part

of the dental cement exposed and each animal was placed on a heating pad for recovery. Post-

operative treatment included subcutaneous buprenorphine (0.03 mg/kg, twice/day), saline (2

mL, once/day) and carprofen (5 mg/kg, once/day) for up to 3 days.

Electroencephalographic Recording

Beginning on day 7 or 8 after surgery, rats were monitored for at least 60 min (30 min for

recovery from anesthesia/acclimation and 30 min for EEG recording), on at least 2 days per

week for the following 7 weeks. For the purpose of developing of the semi-automatic

detection algorithm, only the first two and the last two recordings were analyzed (Fig. 2.1C)

and only the first 20 min of each recording were studied. At each monitoring session, rats

were briefly anesthetized with isoflurane (4% in oxygen, 2 L/min) in an induction cage, and

Page 50: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 28 -

shielded cables were used to connect the recording electrodes to the EEG acquisition system

(TDT processors; Tucker-Davis Technologies, Alachua, FL) and high-impedance head-stages

driven by custom-designed software. The EEGs were sampled at 3051.76 Hz.

Figure 2. 1 Experimental protocol

(A) Schematic diagram illustrating the positioning of epidural recording screw electrodes (1-4) and

the reference electrode (R). (B) Comparisons were made between the manual EEG recording

evaluations of the two researchers, and between the manual evaluations and the semi-automated SWD

detection algorithm. (C) Experimental design: 20-min EEG recording began at day 7 or 8 after surgery

and 2 days per week for the following 7 weeks; for the purpose of developing of the semi-automatic

detection algorithm only the first two and the last two recordings were analyzed.

The rats were allowed to fully recover from anesthesia before recording. During the

recording sessions, their behavior was observed and the EEGs were visualized using a

custom-designed MATLAB® program. If the rats were perceived as being asleep and after

confirmation of no seizure activity on the EEG, noise stimuli of 94 and 98 dB were delivered

Page 51: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 29 -

(Fig. 2.2). At the end of each recording session, the rats were briefly anaesthetized with

isoflurane (4% in oxygen, 2 L/min) for disconnection from the shielded cable.

Figure 2. 2 Example of rats EEGs when active, sleeping or seizing.

During a recording session, the EEGs were visualized using a custom-designed MATLAB® program.

The visualization of the EEGs allowed confirmation of the status of the rat: active (A), sleeping (B),

or seizing (C). If the rats were perceived as being asleep and seizure activity on the EEG was

confirmed, a noise stimulus of 94 to 98dB was applied by knocking on the Plexiglas cage. The three

examples were recorded at the same scale. Calibration 1 s, 1 mV.

Evaluation of EEG Recordings

Preparation of raw data The EEG data were transcribed using a graphical interface

developed in MATLAB®

. They were first band-pass filtered between 3 and 30 Hz using a

second-order Butterworth filter, and then filtered in both the forward and reverse directions to

Page 52: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 30 -

avoid introducing a delay in the signal, which effectively doubled the filter order to fourth

order. During the second step, all recorded EEG channels were displayed for the observer to

select the one with the best signal for SWD detection (i.e., largest SWD amplitude relative to

background EEG amplitude) and without signal dropout (i.e., poor physical connection

resulting in occasional flat lines) (Fig. 2.3A). The selected channel was used for manual and

semi-automated marking.

Manual analysis of EEG data EEG recordings were independently analyzed manually by

two experienced researchers. While reviewing the recorded EEG, the researcher marked the

beginning and the end of all identified SWDs that were more than one second in duration

(Fig. 2.3 B) and an Excel data sheet reporting the start and end times of all the SWDs was

generated. After completing the manual marking, each researcher applied the semi-automated

SWD detection algorithm to the selected channel of EEG recording.

Semi-automated SWD detection algorithm The semi-automated SWD detection algorithm

was developed within the same graphical interface as that noted above. This algorithm

required human intervention to select the best EEG channel and to set an amplitude threshold

for SWD detection. In the latter step, non-parametric thresholds were generated by evaluating

the empirical cumulative distribution function of the voltage magnitude (from 90% to 100%

at 0.5% intervals) and were plotted against the filtered EEG data, so that the observer could

select, by visual inspection, a percentile threshold that would best differentiate the SWD

activity from the background EEG (Fig. 2.3).

Page 53: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 31 -

Figure 2. 3 Selection the EEG channel and determination of amplitude threshold for SWD

detection

EEG analysis using a semi-automated SWD detection requiring human intervention to select the best

EEG channel/electrode and set an amplitude threshold for SWD detection. (A) Example of EEG

recording: in this example, channel four was selected by the investigator as having the best signal-to-

noise ratio, so subsequent analysis was done using this channel. (B) Example of manual marking: the

EEG was analyzed using a 10-s window and the start and end of the SWD were noted. (C) Example of

threshold selection: the traces show the EEG of the selected channel (channel 4 in this example) with

superimposed automatically-calculated thresholds. To be selected, the threshold red line had to be just

above the baseline EEG signal. In this example a threshold of 0.71 mV was selected (middle panel).

Once the threshold was selected, the commands of the program were to: (1) construct a

binary array corresponding to whether the filtered EEG amplitude was above the user-set

threshold (i.e., 0 for below and 1 for above); (2) evaluate the derivative of the binary array to

identify spike rises and spike falls; (3) iterate through the derivative array, data-point by data-

point, and alternatively search for SWD onsets and terminations; and (4) when the end of the

Page 54: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 32 -

data set was reached, to evaluate the number of SWDs (number of SWD onsets) and their

durations (time between onset and termination) (Fig. 2.4).

Figure 2. 4 Flowchart of the semi-automated algorithm

Page 55: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 33 -

The first sample of the 1-s observation window was classified as a SWD onset if it met

the following criteria (Fig. 2.5): (1) the first data point of the window corresponded to a spike

rise; (2) the number of spike rises within the window was between 5 and 13; and (3) the

interspike intervals were between 40 and 300 ms. The first data point of the 1-s observation

window was classified as a SWD termination if it met the following criteria (Fig. 2.5): (1) the

first sample of the window corresponded to a spike fall; and (2) apart from the first sample,

all other samples in the window were below the threshold amplitude. So, a SWD event had to

be at least 1 s in duration and two SWD events were considered separate if they were >1 s

apart.

Figure 2. 5 The process of automatic marking.

Horizontal red line, the threshold level. The left green arrow indicates the first 1-s observation

window classified as a SWD onset (yellow crosses represent spike rises going above the threshold

level), and the right green arrow indicates the first 1-s observation window classified as a SWD

termination (yellow cross represents a spike fall). See text for detailed criteria of classification.

Data Analysis and Performance Metrics

Page 56: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 34 -

Comparisons were made between the manual evaluations of EEG recordings by the

researchers and evaluation by the semi-automated SWD detection algorithm. A previously-

published, rigorous method of assessing performance was used (Fig. 2.6), where only the

time intersection of an automatically detected event with one that was manually marked as a

positive SWD was considered a true positive time window[10]

. Likewise, only when the

automatic time intersection of the absence of an event coindided with a manually marked

negative SWD was considered a true negative time window. When a time window was

evaluated to be SWD-positive by the detection algorithm, but not by the reviewer, it was

considered to be a false-positive. When the detection algorithm evaluated a time window as

SWD-negative, but the reviewer evaluated it as SWD-positive, it was considered to be a

false-negative. Then, the performance metrics of sensitivity, specificity, positive predictive

value (PPV), and negative predictive value (NPV) were calculated using equations 1 to 4.

The performance metrics were assessed to a precision of 0.1 s.

(1)

(2)

(3)

(4)

iveFalseNegatveTruePositi

veTruePositiySensitivit

iveFalsePositveTrueNegati

veTrueNegatiySpecificit

iveFalsePositveTruePositi

veTruePositilueedictiveVaPositivePr

iveFalseNegatveTrueNegati

veTrueNegatilueedictiveVaNegativePr

Page 57: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 35 -

Figure 2. 6 Performance metrics

SWD negative, EEG not showing spike-and-wave discharges (SWDs) as evaluated by the researcher

(human) or the algorithm (auto); SWD positive, EEG showing SWDs as evaluated by the researcher

(human) and the algorithm (auto); true negative, neither human nor auto detected SWDs; false

negative, human-detected SWDs whereas auto did not; true positive, both human and auto detected

SWDs; false positive, human did not detect SWDs whereas auto did.

Comparisons were also made between the two researchers using the same approach

(with one being considered the detection algorithm and the other as the reviewer). In total,

three comparisons were made for each EEG recording (Fig. 2.1B).

Data were analyzed with Microsoft Excel 2010 and graphic representations generated

with GraphPad Prism 6. The computer used to run the detection algorithm was a desktop PC

(twelve Intel® Xenon® CPUs X560 at 3.47 GHz) running Windows 7, 64-bit).

Page 58: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 36 -

RESULTS

The first recording from one of the rats was missing, so only 19 EEG recordings were

evaluated, representing a total of 6 h and 20 min of recording. The median number of SWDs

over 20 min ranged from 40 to 46 and the mean duration of SWDs from 8.9 to 11.4 s for each

evaluator and method, with extreme values resulting from the two manual markings (Table

1). The calculated sensitivity, specificity, PPV, and NPV was >91% for all (Table 2).

Table 2. 1 Median number and mean duration of spike-and-wave discharges (SWDs).

Investigator A

Automatic A

Investigator B

Automatic B

Median

number of

SWDs

40 (30-56)

41 (29-58)

46 (37-63)

42 (29-54)

Mean

duration of

SWD (s)

11.4 (±2.9)

10.7 (±2.9)

8.9 (±2.8)

10.2 (±2.8)

Automatic A and B represent the semi-automated algorithm run by investigators A and B. Median

(min-max), mean (± SD).

Table 2. 2 Sensitivity, specificity, positive predictive value (PPV), and negative predicted value

(NPV)

Investigator A versus

Automatic A

Investigator B versus

Automatic B

Investigator A versus

investigator B

Sensitivity 0.96 ± 0.02

0.95 ± 0.03

0.91 ± 0.05

Specificity 0.96 ± 0.03

0.97 ± 0.02

0.98 ± 0.02

PPV 0.94 ± 0.04 0.94 ± 0.05 0.97 ± 0.03

NPV

0.97 ± 0.01

0.97 ± 0.02

0.95 ± 0.03

Automatic A and B represent the semi-automated algorithm run by investigators A and B. Mean ±

SD.

Page 59: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 37 -

DISCUSSION

The algorithm was demonstrated to have good sensitivity, specificity, PPV, and NPV for

detecting SWDs with durations ≥1 s. The study was not randomized, as the EEG data were

always analyzed manually by the investigators first and then by automatic detection. This was

done so that the investigators’ manual analysis would not be biased by the automatic

detection results.

The characteristics of the SWDs of GAERS used to build the algorithm were slightly

modified from Maurescaux et al. (1992)[3]

. Indeed only SWDs of ≥1 sec duration were

detected and the reported SWD frequency range of 7 to 11 Hz was extended to include SWDs

ranging from 5 to 13 Hz. The larger frequency range provided tolerance as, for example,

spikes can sometimes be below threshold resulting in a lower spike count.

Twenty-minute recording was used for the analysis as this was the duration reported in

the original paper describing the EEG of GAERS (Marescaux et al., 1992). Comparisons

were made between the manual EEG recording evaluations by the two researchers to

establish a baseline range of sensitivity, specificity, PPV and NPV. Comparison between the

manual and that investigator’s evaluation using the semi-automated SWD detection algorithm

resulted in a higher sensitivity when compared to baseline (95 to 96% versus 91%) while

retaining a high specificity (96–97%). These results are comparable to the performance of

other high-quality detection algorithms for SWDs in rodents[11-13]

. In these three publications,

the analyses were based on transforming the EEG from the time domain to the frequency

domain to quantify changes. The methodology reported by Ovchinnikov et al. 2010, only

detect the onset of SWDs (evaluating the number of leading edges without taking the length

of the detector event into consideration) and is based on wavelet analysis[11]

. The

methodology reported by Van Hese et al. 2009 is based on spectral and variant analysis and

that of Sitnikova et al. 2009 on spectral and wavelet analysis[12, 13]

. In contrast, the

methodology described in the present study analyzes the EEG in the time domain, mimicking

manual marking, while decreasing the subjectivity and allowing the results of the EEG

analysis to be more reproducible. In consequence, it provides a good alternative for

researchers. Nelson et al. 2011developed an SWD detection system in GAERS based on

time-series analysis, gauging changes in amplitude and/or frequency[14]

. This system only

Page 60: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 38 -

allowed the detection of the start of the SWD in order to trigger therapy. Subsequent EEG

analyses were performed manually.

The median number of SWDs over 20 min and the mean duration of one SWD were

similar in the different evaluations, with the results of the manual recordings showing the

extreme values. This could indicate that the semi-automated algorithm provides more

accurate results than manual marking. The frequency of SWDs (~2/min) was slightly higher

than that described in the original paper (1.5/min) [3]

. This is easily explained by the fact that

the seizure activity in GAERS is colony- and age-dependent[3]

.

Although spontaneous SWDs start and end abruptly on a normal background EEG and

are quite easy to isolate, the EEG patterns seen during sleep make it more difficult to

differentiate the beginning and end of an SWD. During the present experiments, rats were

stimulated when seen to be sleeping, to improve seizure detection.

Although not all of the manual analyses were timed, it took the researchers on average

15 min to analyze one EEG recording. In comparison, it took the computer 95–100 sec to

analyze one EEG recording using the algorithm. If we consider that some studies can include

>400 EEG recordings (3 groups of 6 rats undergoing 3 recordings a week for 8 weeks), the

use of the algorithm can avoid 88 h of laborious work. The computer used in this study was

certainly more powerful than average, but subsequent analysis using a laptop computer

resulted in similar durations.

The system requires manual intervention to select the best EEG channel/electrode and

set an amplitude threshold for SWDs. These steps may cause inter-individual variability in

seizure detection; however, the use of the present algorithm is still an improvement in inter-

individual variation compared to manual marking. Also, the manual intervention is relatively

short and allows control of the quality of the EEG recording.

Another limitation of the study is the absence of an automatic artefact-rejection step.

Nonetheless, the performance of the algorithm was as expected from such a system and the

manual channel-selection allowed rejection of recordings that included too many artefacts.

Page 61: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 39 -

The semi-automatic SWD detector algorithm described here allowed analysis of the

EEG of GAERs with sensitivity, specificity, PPV, and NPV >94% compared to manual

analysis. The use of this algorithm would reduce the time necessary to analyze such data and

the subjectivity of the results. as well as providing a good performance alternative for

researchers without an engineering background.

Acknowledgements

The authors gratefully acknowledge the sponsorship of the Victoria Government through its

Science Technology and Innovation Initiative administered by the Department of Industry,

Innovation and Regional Development. The authors thank the Australian Research Council

for continuing financial support.

REFERENCES

[1] Sander JW. The epidemiology of epilepsy revisited. Curr Opin Neurol 2003, 16: 165-170.

[2] Shorvon SD. The epidemiology and treatment of chronic and refractory epilepsy. Epilepsia 1996,

37 Suppl 2: S1-S3.

[3] Marescaux C, Vergnes M, Depaulis A. Genetic absence epilepsy in rats from Strasbourg--a

review. J Neural Transm Suppl 1992, 35: 37-69.

[4] Rudolf G, Bihoreau MT, Godfrey RF, Wilder SP, Cox RD, Lathrop M, et al. Polygenic control of

idiopathic generalized epilepsy phenotypes in the genetic absence rats from Strasbourg (GAERS).

Epilepsia 2004, 45: 301-308.

[5] Greene BR, Faul S, Marnane WP, Lightbody G, Korotchikova I, Boylan GB. A comparison of

quantitative EEG features for neonatal seizure detection. Clin Neurophysiol 2008, 119: 1248-1261.

[6] Osorio I, Frei MG, Wilkinson SB. Real-time automated detection and quantitative analysis of

seizures and short-term prediction of clinical onset. Epilepsia 1998, 39: 615-627.

[7] Paivinen N, Lammi S, Pitkanen A, Nissinen J, Penttonen M, Gronfors T. Epileptic seizure

detection: a nonlinear viewpoint. Comput Methods Programs Biomed 2005, 79: 151-159.

Page 62: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 40 -

[8] Saab ME, Gotman J. A system to detect the onset of epileptic seizures in scalp EEG. Clin

Neurophysiol 2005, 116: 427-442.

[9] Wilson SB, Scheuer ML, Emerson RG, Gabor AJ. Seizure detection: evaluation of the Reveal

algorithm. Clin Neurophysiol 2004, 115: 2280-2291.

[10] Navakatikyan MA, Colditz PB, Burke CJ, Inder TE, Richmond J, Williams CE. Seizure detection

algorithm for neonates based on wave-sequence analysis. Clin Neurophysiol 2006, 117: 1190-1203.

[11] Ovchinnikov A, Luttjohann A, Hramov A, van Luijtelaar G. An algorithm for real-time detection

of spike-wave discharges in rodents. J Neurosci Methods 2010, 194: 172-178.

[12] Sitnikova E, Hramov AE, Koronovsky AA, van Luijtelaar G. Sleep spindles and spike-wave

discharges in EEG: Their generic features, similarities and distinctions disclosed with Fourier

transform and continuous wavelet analysis. J Neurosci Methods 2009, 180: 304-316.

[13] Van Hese P, Martens JP, Waterschoot L, Boon P, Lemahieu I. Automatic detection of spike and

wave discharges in the EEG of genetic absence epilepsy rats from Strasbourg. IEEE Trans Biomed

Eng 2009, 56: 706-717.

[14] Nelson TS, Suhr CL, Freestone DR, Lai A, Halliday AJ, McLean KJ, et al. Closed-loop seizure

control with very high frequency electrical stimulation at seizure onset in the GAERS model of

absence epilepsy. Int J Neural Syst 2011, 21: 163-173.

Page 63: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 41 -

Chapter 3 - Seizures induced by oral administration of

enrofloxacin in GAERS.

3.1. Outline

Antibiotics have known adverse effects on the nervous system such as antibiotic induced

seizures that have not been intensively investigated. The literature mainly describes case

reports of antibiotic induced seizures in predisposed patients.(Lerner, Smith et al. 1967,

Arcieri, Becker et al. 1989, Grondahl and Langmoen 1993, Lode 1999, Kim, Kim et al. 2013,

Bhattacharyya, Darby et al. 2014, Tsubouchi, Ikematsu et al. 2014) Fluoroquinolones are one

of the antibiotic classes most commonly associated with seizures and are thought to provoke

seizures through inhibitory effects on GABA transmission.(Bhattacharyya, Darby et al. 2014)

Enrofloxacin is a broad spectrum potent fluoroquinolone that can be administered orally and

that is routinely used post-operatively in rats.(Adams 2001)

GAERS express typical absence seizures originating from abnormal thalamocortical

networks including the thalamocortical neurons of sensory thalamic nuclei and their main

inhibitory input (GABAergic neurons).(Noebels and Jasper 2012) Consequently it was

hypothesized that GAERS could be predisposed to fluoroquinolone induced seizures, and that

administration of enrofloxacin would increase their seizure activity. To test this hypothesis,

five GAERS were monitored for spike and wave discharges (SWD) activity before and after

addition of enrofloxacin into their drinking water.

Although no effect of enrofloxacin on GAERS’ SWD activity was demonstrated with

statistical significance, its administration coincided with the appearance of tonic seizures,

which is abnormal in this model of epilepsy. The incidence of tonic seizures (n=2) remained

uncommon when compared to the number of non-convulsive seizures recorded (n>500)

during the same recording period, however two rats out of five were affected representing

40% of the sample population. Also, considering that the alteration of the blood brain barrier

is a risk factor for antibiotic induce seizure and that certain quinolones have synergistic

inhibition with non-steroidal anti-inflammatory drugs at the GABA receptor, the

administration of enrofloxacin peri-operatively when performing craniotomies could result in

Page 64: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 42 -

a higher incidence and more severe clonic seizures and should be avoided.(Lode 1999,

Bhattacharyya, Darby et al. 2014)

To the best of the authors’ knowledge, this is the first study looking at the effect of

enrofloxacin in GAERS. The results also allowed the author to refine the experimental

protocol and provided evidence to change the standard operative procedures of the institution

for the in vivo studies presented in the following chapters.

3.2. Manuscript

This chapter is presented as the author-accepted version of a peer-reviewed manuscript

published in Comparative Medicine, 2016;66(3):220-4. The figures’ numbers and titles of

the author-accepted version of the manuscript may have been changed to follow the

formatting of the thesis.

Page 65: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 43 -

Clonic Seizures in GAERS Rats after Oral Administration of Enrofloxacin

Sebastien H Bauquier,1,

* Jonathan L Jiang,2,3

Alan Lai,4 and Mark J Cook

2,3

1Translational Research and Animal Clinical Trial Study (TRACTS) Group, Faculty of

Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, Victoria,

Australia;

2Department of Clinical Neurosciences, St Vincent’s Hospital, Fitzroy, Victoria, Australia;

3Faculty of Medicine, The University of Melbourne, Parkville, Victoria, Australia;

4Department of Electrical and Electronic Engineering, The University of Melbourne,

Parkville, Victoria, Australia.

*Corresponding author. Email: [email protected].

Enrofloxacin induces clonic seizures in GAERS

The aim of this study was to evaluate the effect of oral enrofloxacin on the epileptic status of

Genetic Absence Epilepsy Rats from Strasbourg (GAERS). Five adult female GAERS rats,

with implanted extradural electrodes for EEG monitoring, were declared free of clonic

seizures after an 8-wk observation period. Enrofloxacin was then added to their drinking

water (42.5 mg in 750 mL), and rats were observed for another 3 days. The number of spike-

and-wave discharges and mean duration of a single discharge did not differ before and after

treatment, but 2 of the 5 rats developed clonic seizures after treatment. Enrofloxacin should

be used with caution in GAERS rats because it might induce clonic seizures.

GABA, γ-aminobutyric acid; GAERS, genetic absence epilepsy in rat from Strasbourg;

SWD, spike-and-wave discharge

Page 66: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 44 -

The potential adverse effects of antibiotics on the nervous system include antibiotic-induced

seizures that have not been investigated intensively. With a few exceptions, the literature

includes only case reports of antibiotic-induced seizures in predisposed patients.2,5,7-

10,15Fluoroquinolones are one of the antibiotic classes most commonly associated with

seizures and are thought to provoke seizures through inhibitory effects on gamma-

aminobutyric acid (GABA) transmission.5 Enrofloxacin is a fluoroquinolone antimicrobial

drug that is rapidly bactericidal against a wide variety of clinically important bacterial

organisms. Enrofloxacin is potent, well tolerated by animals, and can be administered

through a variety of routes (orally by tablet and drinking water, subcutaneously, and

intramuscularly).1

The GAERS (Genetic Absence Epilepsy in Rat from Strasbourg) strain is a genetic Wistar rat

model of absence epilepsy that displays electroclinical, behavioral, and pharmacologic

features of absence seizures and has become a ‘gold standard’ to study the mechanisms

underlying absence epilepsy.6,12

The EEG of GAERS presents typical spike-and-wave

discharges (SWD) of 0.3 to 1 mV in amplitude and 7 to 11 SWD per second in spike rate.6,12

Typical absence seizures originate from abnormal thalamocortical networks with cellular

elements that include pyramidal cells and interneurons of different cortical layers, the

thalamocortical neurons of sensory thalamic nuclei and their main inhibitory input

(GABAergic neurons of the nucleus reticularis thalami).13

Consequently and for the purpose of a pilot study, we hypothesized that GAERS could be

predisposed to fluoroquinolone-induced seizures and that oral administration of enrofloxacin

would increase their seizure activity. During that pilot study, some rats developed clonic

seizures. To our knowledge, no description of antibiotic-induced clonic seizure in GAERS

has been published, the aim of the present case study was to document clonic seizures

induced by oral administration of enrofloxacin in GAERS.

Materials and Methods

The pilot study was designed as a prospective, blinded experiment. The experiments were

approved by St Vincent’s Hospital (Melbourne) Animal Ethics Committee and conducted in

Page 67: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 45 -

accordance with the Australian Code of Practice for the Care and Use of Animals for

Scientific Purposes (2004).3

Female GAERS rats (Rattus norvegicus; age, 6 mo) with a clean colony pathogen status

(tested for 17 different virus, 18 bacterias and fungi, and 13 ectoparasites and endoparasites)

were obtained from the Royal Melbourne Hospital’s Biologic Research Facilities, The

University of Melbourne (Parkville, Victoria, Australia), to take part in an 8-wk experiment

investigating novel techniques to deliver antiepileptic drugs. The first 5 rats randomly

allocated to the control group were also used in the present pilot study.

The GAERS were housed individually in high-top rat cages (RB2, Wiretainers, Melbourne,

Victoria, Australia) with recycled paper bedding (Fibrecycle, Yatala, Queensland, Australia).

The temperature and humidity of the room were kept at 22 °C and 55%, respectively. The rats

were maintained under a 12:12-h light:dark cycle with free access to food (Barastoc GR2

pellets, Ridley, Melbourne, Victoria, Australia) and tap water.

All 5 rats underwent surgery for the implantation of EEG recording electrodes (Figure 3.1A).

Immediately before surgery, the rats were weighed and anesthetized with an intraperitoneal

injection of ketamine (75 mg/kg; ketamine hydrochloride 100 mg/mL, Ceva Animal Health,

Glenorie, New South Wales, Australia) and xylazine (10 mg/kg; xylazine hydrochloride 20

mg/mL, Troy Laboratories, Glendenning, New South Wales, Australia). After the induction

of anesthesia, each rat was placed in a stereotaxic apparatus (Kopf, Stoelting, Wood Dale, IL)

and given isoflurane (0.5% to 1% in oxygen, 1 L/min; Isoflo, Abbott Australasia, Botany,

New South Wales, Australia) by nose-cone, subcutaneous carprofen (5 mg/kg; 50 mg/mL,

Parnell Australia, Alexandria, New South Wales, Australia) for pain relief, and 0.9% sodium

chloride (2 mL; Baxter, Old Toongabble, New South Wales, Australia) for cardiovascular

support. The EEG recording electrodes were implanted as follows: after aseptic preparation

of the scalp, a single incision was made down the midline, the skull cleared of tissue, and the

exposed bone dried with 3% hydrogen peroxide (Sanofi, Victoria, Queensland, Australia).

Four extradural electrodes, consisting of small jewelers’ screws (diameter, 3.2 mm; Plastics

One, Roanoke VA), were implanted cranial to the interaural line (2 on each side of the

Page 68: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 46 -

Figure 3. 1 Experimental protocol

(A) Schematic diagram illustrating the positioning of the epidural recording screw electrodes (1

through 4) and reference electrode (R). (B) Experimental design. EEG recording (30-min session

twice weekly) began on day 7 or 8 after surgery and continued for the following 7 wk. On day 61, the

drinking water was replaced with enrofloxacin-containing water (1.7 mL of enrofloxacin 25 mg/mL in

750 mL of drinking water), and additional EEG recordings were performed on days 62 and 64. The

enrofloxacin treatment was stopped on day 65. (C) The clonic seizure of rat 5 started on day 64 after

308 s of EEG recording and lasted for 40 s. The intensity of the clonic seizure was classified as stage

4 (clonic seizure in a seating position).11

The EEG recordings presented SWD that were similar in

amplitude (0.3 to 0.7 mV) and frequency (average, 7 SWD per s) to that observed during absence

seizure.

sagittal suture), and one screw was implanted caudal to the interaural line to the right of the

sagittal suture. The electrodes were connected to an adaptor secured with dental repair

material (Cold-curing acrylic, Vertex, Zeist, The Netherlands). The skin was sutured (3-0

Monosyn, B Braun, Rubi, Spain), leaving only part of the dental cement exposed, and each

rat was placed on a heating pad for recovery from anaesthesia. Postoperative treatment

Page 69: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 47 -

included subcutaneous buprenorphine (0.03 mg/kg BID; Temgesic, 0.324 mg/mL

buprenorphine hydrochloride, Reckitt Benckiser Healthcare, Hull, United Kingdom), 0.9%

sodium chloride (2 mL daily), and carprofen (5 mg/kg, daily) as needed for a maximum of 3

d.

Beginning at day 7 or 8 after surgery, rats were monitored for at least 60 min (30 min for

recovery from anaesthesia and acclimation, 30 min for EEG recording) daily and on at least 2

d each week for the following 7 wk. At day 61, regular drinking water was replaced with

water containing enrofloxacin (42.5 mg in 750 mL of drinking water; Baytril, 25 mg/mL,

Bayer Australia, Pymble, New South Wales, Australia), and additional EEG recordings were

obtained on days 62 and 64 (Figure 3.1A). Each EEG recording session began 4 to 6 h after

the start of the light cycle. Treatment with enrofloxacin stopped on day 65. The rats were

weighed (sensitivity, ± 1 g; model KD-200, Tanika, Kowloon, Hong Kong) on days 61, 62,

and 64.

For each monitoring session, rats were anesthetized in an induction cage by using isoflurane

in oxygen for 3 min to connect the recording electrodes through shielded cables to the EEG

acquisition system,4 which consisted of TDT processors (Tucker-Davis Technologies,

Alachua FL) and high-impedance head stages driven by TDT’s programmable software. The

EEG was sampled at 3051.76 Hz, and recording began only after the rats had returned to

normal behavior (moving around without ataxia, eating, cleaning itself) and presented

absence seizures. During the recording sessions, the rat’s behaviour was observed, and the

EEG was visualized by using a custom-designed MATLAB program.4 When clonic seizures

occurred, their intensity was classified according to the modified Racine scale.11

At the end of

a recording session, the rats were briefly anesthetized with isoflurane (4% in oxygen, 2

L/min) to be disconnected from the shielded cable.

To evaluate the effects of the enrofloxacin treatment on the GAERS SWD, the median

number of SWD, duration of a single SWD, and cumulative duration of SWD during 60 min

were compared between weeks 8 and 9 (Figure 3.1B) by using the Wilcoxon signed ranks test

(IBM SPSS Statistics, version 20, IBM Corp, Armonk, NY USA). The significance level was

set at 5%. The researcher performing EEG analysis was blinded regarding treatment and used

a GAERS-specific automated SWD detection algorithm.4

Page 70: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 48 -

Results

All 5 rats were declared free of clonic seizures during the first 8 wk of EEG recordings. On

day 61, the median weight of the rats was 189 g (range, 185 to 204 g). The median weight

gains were 0 g (range, –1 to 2 g) and –0.2 g (range, –1 to 1 g) on days 62 and 64,

respectively, when compared with day 61. Although the duration of SWD showed an

increasing trend (p=0.34), the number of SWD, duration of a single SWD, and cumulative

duration of SWD during 60 min did not differ significantly (Figure 3.2). However, 2 rats each

manifested a single episode of clonic seizures. The first observed episode occurred on day 62

(24 h after the start of treatment) and lasted 31 s, whereas the second episode occurred on day

64 (72 h after the start of treatment) and lasted 40 s. During both episodes, the affected rats

had convulsions interpreted as clonic seizures while they were in a sitting position. The first

signs were trembling of the whiskers, shortly followed by spasms and jerking of the neck

muscles and some flexion of the legs. Both seizures were classified as intensity stage 4

seizures.

The EEG recordings obtained during the clonic seizures presented SWD of similar amplitude

(0.3 to 0.7 mV) and frequency (7 SWD per s) to those observed during absence seizures

(Figure 3.1C).

Page 71: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 49 -

Figure 3. 2 Box-and-whisker plots of the epileptic activity before enrofloxacin treatment (n = 5)

and afterward (posttreatment, n = 5).

(A) Number of SWD. (B) Median duration of a single SWD. (C) Total duration of SWD during 60

min of recording time each week. ° represent a yellow card zone outlier (distance between the outlier

and the interquartile range box superior to 1.5 times the interquartile range box length) and * represent

Page 72: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 50 -

a Red card zone outlier (distance between the outlier and the interquartile range box superior to 3

times the interquartile range box length).

Discussion

The GAERS strain is a genetic rat model of absence epilepsy of which all rats present

recurrent generalized nonconvulsive seizures accompanied with behavioural arrest, staring,

and sometimes twitching of the vibrissae.12

Although the small number of rats in the present

study precluded definitive determination of the effect of enrofloxacin on SWD activity, the

administration of the antibiotic coincided with the appearance of clonic seizures, which are

abnormal in this model of epilepsy.12

To our knowledge, the neurologic circuits and biochemical substrates of absence seizures and

clonic seizures are similar. However, why some similar patterns of abnormal brain activity

result in absence compared with clonic compared with tonic–clonic seizures is unknown. The

incidence of clonic seizures (n = 2) remained uncommon when compared with the number of

nonconvulsive seizures recorded (n > 500) during the same recording period, but 2 of the 5

rats were affected, representing 40% of the sample population.

In addition, considering that alteration of the blood–brain barrier is a risk factor for antibiotic-

induced seizures (by allowing for increased concentration of the antibiotic in the brain) and

that certain quinolones have synergistic inhibition with NSAID at the GABA receptor, the

administration of enrofloxacin perioperatively when performing craniotomies might result in

a higher incidence and more severe clonic seizures than those observed in the present

study.5,10

The 5 GAERS rats used in this pilot experiment were control animals in a separate study

investigating novel techniques to deliver antiepileptic drugs. Because the recording period for

the antiepileptic drug experiment was 8 wk, enrofloxacin couldn’t be administered earlier. In

addition, GAERS epileptic activity is age-dependent; therefore, the SWD activity that

occurred during week 9 (after the start of enrofloxacin treatment) was compared only with

week 8 (pretreatment) data.12

Specifically, the number and duration of SWD increase with

age, with the maximal number of SWD at the age of 6 mo.12

Page 73: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 51 -

One limitation of the current study is that the water consumption was not measured.

However, rats were weighed daily, and all rats maintained appropriate weight during the

antibiotic treatment, implying a normal hydration status.

When combined with a nonseizure-modulating dose of theophylline, ciprofloxacin and

norfloxacin increased seizure activity in amygdaloid-kindled rats.16

However, when given as

a sole agent in that model, enrofloxacin decreased seizure activity.16

In another experiment,

ciprofloxacin (≥ 50 mg/L) showed moderate to marked epileptogenic effects when tested on

slices of rat hippocampus, which have a low threshold for the development of epileptiform

activity.7

The GABAA receptor plays a major role in the epileptogenic activity of fluoroquinolones, and

(in vitro) the N-methyl-D-aspartate receptor is directly involved in this process as a target for

the excitatory effect of fluoroquinolones.5,10,14

The substituent at the C7 position of

fluoroquinolones appears to be an important predictor of GABA binding and seems to be, at

least partially, responsible for the differences in seizure incidence among fluoroquinolones.10

To our knowledge, the C7 substituent of enrofloxacin has not been studied for its

epileptogenic activity.

The EEG of GAERS rats presents typical SWD that originate from abnormal thalamocortical

networks, including its GABAergic neurons.6,13

In contrast, kindling is a local process

whereby an electrical stimulus typically delivered to a limbic brain structure evokes an

electrographic after-discharge capable of spreading and initiating a generalized convulsive

seizure.16

The kindled rat model of epilepsy doesn’t involve the GABAergic neurons in the

initiation of the seizure, and this might be the reason for the different effects of enrofloxacin

between the GAERS and kindled rat models. In addition, amygdala-kindled events are focal

as compared with the primarily generalized nature of the seizures in GAERS rats;

consequently, these processes have different mechanisms from the pharmacologic

perspective.

Isoflurane was used briefly when connecting and disconnect the recording apparatus. In

previous experiments, we found that some rats showed stress behavior (for example,

vocalization) during handling for connection and disconnection; consequently we opted for

brief general anaesthesia or sedation to increase animal wellbeing. Rats recovered quickly

Page 74: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 52 -

from the anaesthetic episodes, and it is unlikely that, after the 30-min acclimation period,

isoflurane would still be present to interfere with epileptic activity.

Finally, no postmortem brain histologic analyses were performed on the rats and, although

improbable, it cannot be excluded that the appearance of the clonic seizures coincided with

the development of an intracranial pathology unrelated to administration of enrofloxacin.

Similarly, it cannot be excluded that the isoflurane anaesthesia affected the appearance of

SWD; however the 30-min recovery period prior to recording likely reduced isoflurane

effects to being negligible.

Within the described experimental conditions, enrofloxacin did not appear to change the

duration or frequency of SWD in GAERS rats. However, this antibiotic should be

administered with caution because it might induce clonic seizures in GAERS rats, especially

during the perioperative period, when aggravating factors such as disruption of the blood–

brain barrier and coadministration of NSAID may be present.

Acknowledgment

We gratefully acknowledge the sponsorship of the Victorian Government through its Science

Technology and Innovation Initiative, administered by the Department of Industry,

Innovation, and Regional Development, and of NHMRC (grant no. 619614). We thank the

Australian Research Council (ARC) for continuing financial support (ARC grant no.

DP0987344).

References

1. Adams HR. 2001. Veterinary pharmacology and therapeutics, 8th ed. Ames (IA): Iowa State

University Press.

2. Arcieri GM, Becker N, Esposito B, Griffith E, Heyd A, Neumann C, O’Brien B, Schacht P. 1989.

Safety of intravenous ciprofloxacin. A review. Am J Med 87 Suppl 1:S92–S97. PubMed

doi:10.1016/0002-9343(89)90032-6

Page 75: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 53 -

3. Australian Government National Health and Medical Research Council, 2004. Asutralian Code of

Practice for the care and use of animals for scientific purposes 7Th

edition. Available at :

https://www.nhmrc.gov.au/_files_nhmrc/publications/attachments/ea16_animal_code.pdf

4. Bauquier SH, Lai A, Jiang JL, Sui Y, Cook MJ. 2015. Evaluation of an automated spike-and-wave

complex detection algorithm in the EEG from a rat model of absence epilepsy. Neurosci Bull.31:601–

610. PubMed doi:10.1007/s12264-015-1553-5

5. Bhattacharyya S, Darby R, Berkowitz AL. 2014. Antibiotic–induced neurotoxicity. Curr Infect Dis

Rep 16:448. PubMed doi:10.1007/s11908-014-0448-3

6. Danober L, Deransart C, Depaulis A, Vergnes M, Marescaux C. 1998. Pathophysiological

mechanisms of genetic absence epilepsy in the rat. Prog Neurobiol 55:27–57. PubMed

doi:10.1016/S0301-0082(97)00091-9

7. Grondahl TO, Langmoen IA. 1993. Epileptogenic effect of antibiotic drugs. J Neurosurg 78:938–

943. PubMed doi:10.3171/jns.1993.78.6.0938

8. Kim A, Kim JE, Paek YM, Hong KS, Cho YJ, Cho JY, Park HK, Koo HK, Song P. 2013.

Cefepime-induced nonconvulsive status epilepticus (NCSE). J Epilepsy Res 3:39–41. PubMed

doi:10.14581/jer.13008

9. Lerner PI, Smith H, Weinstein L. 1967. Penicillin neurotoxicity. Ann N Y Acad Sci 145:310–318.

PubMed doi:10.1111/j.1749-6632.1967.tb50228.x

10. Lode H. 1999. Potential interactions of the extended-spectrum fluoroquinolones with the CNS.

Drug Saf 21:123–135. PubMed doi:10.2165/00002018-199921020-00005

11. Luttjohann A, Fabene PF, van Luijtelaar G. 2009. A revised Racine’s scale for PTZ-induced

seizures in rats. Physiol Behav 98:579–586. PubMed doi:10.1016/j.physbeh.2009.09.005

12. Marescaux C, Vergnes M, Depaulis A. 1992. Genetic absence epilepsy in rats from Strasbourg—a

review. J Neural Transm Suppl 35:37–69. PubMed

13. Noebels J, Avoli M, Rogawski M, Olsen R, Delgado-Escueta A, editors. 2012. Jasper’s basic

mechanisms of the epilepsies,4th ed. New York (NY): Oxford University Press.

doi:10.1093/med/9780199746545.001.0001

14. Schmuck G, Schurmann A, Schluter G. 1998. Determination of the excitatory potencies of

fluoroquinolones in the central nervous system by an in vitro model. Antimicrob Agents Chemother

42:1831–1836. PubMed

Page 76: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 54 -

15. Tsubouchi K, Ikematsu Y, Hashisako M, Harada E, Miyagi H, Fujisawa N. 2014. Convulsive

seizures with a therapeutic dose of isoniazid. Intern Med 53:239–242. PubMed

doi:10.2169/internalmedicine.53.1303

16. Vancutsem PM, Schwark WS. 1992. Effects of fluoroquinolone antimicrobials alone and in

conjunction with theophylline on seizures in amygdaloid-kindled rats. Mechanistic and

pharmacokinetic study. Epilepsy Res 13:59–71. PubMed doi:10.1016/0920-1211(92)90008-H

Page 77: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 55 -

Chapter 4 – Anti-epileptic effects of Phenytoin loaded polymers

implanted subdurally in GAERS

4.1. Introduction

Before undergoing human clinical trials, polymer-based drug delivery devices need to go

through thorough investigations reviewing product characterisation, in vivo long term

efficacy, and biocompatibility and safety animal studies. In the following investigation,

PLGA sheets loaded with the commonly used AED phenytoin were developed and their

ability to decrease seizure activity was investigated in the GAERs. As mentioned in chapter

1, phenytoin has been used for the treatment of epilepsy for almost 80 years and PLGA

polymers have been approved by the United State Drug Administration after extensive

investigations of its safety profile.

The aim of the study presented in this chapter is to provide some in vitro product

characterizations and to investigate the effect of subdural implantation of biodegradable

PLGA polymers loaded with phenytoin on seizure activity in the GAERS animal model of

epilepsy. The hypothesis was that subdural implantation of AED-loaded PLGA polymers can

decrease seizure activity of the GAERS. The novelty of the study would reside in 1) the in

vitro release profile of phenytoin from PLGA polymer and 2) the demonstration that focal

delivery of AEDs can achieve sustained anti-epileptic effect in an animal model of

generalized epilepsy.

4.2. Materials and methods

The in vitro experiments were designed to characterise the structure of the polymers and the

in vitro release profile of phenytoin. Those experiments were performed at the Intelligent

Polymer Research Institute and ARC Centre of Excellence for Electromaterials Science

(Wollongong).

The in vivo study was designed as a randomized controlled masked experiment. PLGA

polymer loaded with the AEDs phenytoin was investigated. All experiments were approved

Page 78: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 56 -

by St Vincent’s Hospital (Melbourne) Animal Ethics Committee and conducted in

accordance with the Australian Code of Practice for the Care and Use of Animals for

Scientific Purposes (2004).

4.2.1. PLGA polymer mat production

Phenytoin-laden polymer mats were produced using a coaxial electrospinning method (Viry

et al., 2012), with the core composed of phenytoin and 75:25 PLGA (lactide/glycolide =

75:25), and the shell composed of 85:15 PLGA (lactide/glycolide = 85:15). The core solution

was prepared at 17% w/v 75:25 PLGA in dimethylformamide (DMF), to which phenytoin

was added to a final concentration of 10% w/w relative to the polymer. The shell solution

was prepared at 20% w/v 85:15 PLGA in a binary solvent system comprising

dichloromethane and DMF (dichloromethane/DMF = 7/3). Coaxial electrospinning was

conducted using a nano-electrospinning system (NANON-01A, MECC CO. Ltd) at an

applied DC voltage of 23 kV. A coaxial spinneret with a diameter of 0.2 mm for the core and

0.8 mm for the sheath nozzles was connected to the core and shell solutions and a feed rate of

0.3 mL h-1

and 1.2 mL h-1

was applied for the core and shell solutions respectively. Fibre

mats were collected on a grounded plate collector that was set 15 cm away from the spinneret

tip. The drug-free, coaxially spun polymer mats were also prepared using the above

procedure. The polymer mats containing drug are denoted as ‘phenytoin-loaded polymers’

whilst the drug-free mats are denoted as ‘blank polymers’. All the samples were finally dried

in a vacuum oven at 40 oC for 72 h to remove residual solvent and were stored at -20

oC prior

to subsequent physiochemical characterization and animal studies.

4.2.2. Morphology Study

The morphology of the electrospun mats (with and without drug) was examined using a field

emission scanning electron microscope (FESEM, JEOL JSM-7500FA). The samples were

sputter-coated with gold prior to FESEM to avoid sample charging.

4.2.3. Determination of drug loading in polymer mats

An extraction method was used to determine the drug loading in the electrospun mats.

Briefly, each sample (1 cm × 1 cm) was weighed and placed into 1 mL methanol for 12 hours

after which the methanol was removed and replenished with 1 mL of fresh methanol. This

Page 79: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 57 -

extraction procedure was repeated four times with each methanol sample allowed to

evaporate to leave residual drug behind which was reconstituted using artificial cerebrospinal

fluid (aCSF). Each aCSF sample was then analysed for drug using high performance liquid

chromatography (HPLC) (see below). The fourth reconstituted sample showed no presence of

drug indicating that the entire drug had been extracted from the electrojetted sample.

4.2.4. Silicone sheets production

Silicone sheets were prepared using implant grade liquid silicone rubber (Cochlear Ltd), LSR

30, which comprises a polymer precursor base and a crosslinker. The polymer precursor base

and crosslinker were dissolved in hexane at a ratio of 10:1, cast in a Teflon mould at room

temperature, and cured by crosslinking at 110 oC for 30 min and then at 200

oC for 4 h. The

silicone sheets (approximately 1 mm sick) were finally extracted with hexane for 72 h to

remove any residual oligomers, and dried in vacuo at 60 oC overnight.

4.2.5. In vitro drug release

The phenytoin-loaded polymers (1 cm × 1 cm) were suspended in 1 mL of aCSF, and the

release experiment was conducted at 37 oC in a shaker water bath (Julabo Pty. Ltd.). The

aCSF contained NaCl (0.866% w/v), KCl (0.224% w/v), CaCl2-2H2O; (0.0206% w/v) and

MgCl2-6H2O (0.0164% w/v) in 1 mM phosphate buffer (pH 7.4).(Halliday, Campbell et al.

2013) For each sample, the release medium solution was collected and replenished with fresh

aCSF at various time points, and stored at -20 oC prior to HPLC analysis being undertaken.

The eluted samples were analysed by HPLC using a modified method on an Agilent 1260

Infinity HPLC system.(Lin, Hsieh et al. 2010) An Atlantis T3 C18 column (250 mm × 4.6

mm, 5 µm) was employed as the analytic column and set at 40 oC. The mobile phase was

composed of water, acetonitrile and methanol (65:26.2:8.8, v/v/v), the injection volume was

10 L with the flow rate of 0.8 mL/min. The eluting phenytoin was detected using a UV-vis

detector set at a wavelength of 203 nm (max of phenytoin). To convert the UV-vis

absorbance to drug concentration a standard curve was established by plotting the UV-vis

peak areas against respective concentrations of standard solutions.

4.2.6. Animals

Adult female GAERS were obtained from the University of Melbourne (Parkville, Victoria,

Australia) and housed individually in 12-hour light/dark cycles with ad libitum access to food

Page 80: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 58 -

and water. The rats were randomly allocated to a control group (no implant; n=6), blank

polymer group (bilateral implantation of blank PLGA polymers not containing AED; n=6) or

treatment group (bilateral implantation of phenytoin loaded PLGA polymers; n=6). The

randomization was performed using the random function in Microsoft Excel 2007. Later, a

silicone group (bilateral implantation of silicone sheets; n=4) was added to test the effects of

the products resulting from the polymer biodegradation. The group attribution list was kept

concealed from the researchers performing the EEG analysis.

4.2.6.1 Implantation surgery

Immediately prior to surgery, rats were weighted and anaesthetized with an intra-peritoneal

injection of ketamine (75 mg/kg) and xylazine (10 mg/kg). Following anaesthesia induction

rats were placed in a stereotaxic apparatus, and administered isoflurane (0.5 to 1% in oxygen,

1 L/min) via a nose-cone, and were administered subcutaneous (SQ) carprofen (5 mg/kg) for

pain relief and 0.9% sodium chloride (2 mL) for cardiovascular support.

Rats from the control group underwent surgery for EEG recording electrode implantation

whereas surgeries for the rats from the blank polymer group, the treatment group and the

silicone group also included bilateral craniotomies for subdural placement of two identical

implants. Over the scalp of all rats, the hair was clipped and the skin aseptically prepared. A

single incision was made down the midline, the skull cleared of tissue and the exposed bone

dried with 3% hydrogen peroxide. Five extradural electrodes, consisting of small jeweller’s

screws, were implanted caudal to the intended polymer implantation sites through burr holes.

Four were implanted cranially to the interaural line (two on each side of the sagittal suture)

and one caudally to the interaural line on the right side of the sagittal suture (figure 4.1 A and

B). The electrodes were then connected to an adaptor and secured with dental cement.

Page 81: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 59 -

Figure 4. 1 Polymers and electrodes implantation sites, and timeline of the experiment.

Schematic diagram illustrating the positioning of epidural recording screw electrodes (1-4), reference

electrode (R) and polymer mats (A); Picture of the actual surgery (B); Timeline of the experimental

design (C): For a minimum of three days post-surgery the rats were monitored for weight loss once a

day, and for mobility and grooming twice a day. Thirty minute electroencephalogram recordings

began at day 7 or 8 post surgery and for the following 7 weeks for 2 days per week during experiment

one, and for 3 days per week during experiment two. For each rat, the mean desired value (number of

SWDs, duration of each SWD or cumulative duration of SWDs over 20 min) were calculated for eight

recording blocks, each block being 1 week of duration.

Implants measuring 3 mm by 4 mm were cut from the polymer sheets described above.

Placements of the implants were performed after 5 mm by 4 mm craniotomies were created

bilaterally at the level of the coronal suture (over the motor cortices) and after incising the

dura to expose the brain surface (Figure 4.1A and B). Following implant placements, the dura

was flapped-back over the polymer and the skull removed from craniotomy sites replaced.

The craniotomy sites were then sealed with an alginate-based hydrogel, the entire surgical

site covered with dental cement and the skin sutured leaving exposed only part of the dental

cement. The animals were placed on heat pads for recovery. Post-operative treatment

included SQ buprenorphine every twelve hours (0.03mg/kg, twice a day), saline (2mL, once a

day) and carprophen SID (5mg/kg, once a day) for up to 3 days.

Page 82: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 60 -

4.2.6.2 Electroencephalograph recording and analysis

At day 7 or 8 post surgery and at least 2 days per week for the following 7 weeks, rats were

monitored for one hour (30 to 40 min anaesthesia recovery/acclimation time, 20 min

recording time) (Figure 4.1C). At each monitoring session, rats were briefly anaesthetized in

an induction cage with isoflurane (4% in oxygen, 2 L/min), and shielded cables were used to

connect the recording electrodes to the EEG acquisition system, which consisted of TDT

processors and high impedance head stages driven by custom-designed software (Tucker

Davis Technologies, USA). The rats were allowed to recover from the anaesthetic before

recordings began. The EEGs were visualized using a custom-designed MATLAB program

(The MathWorks, Inc., U.S.A.). During EEG recording, if the rats were perceived as being

asleep, and after confirmation of no seizure activity on the EEG, a noise stimulus between 94

and 98 decibels was applied. At the end of a recording session, the rats were again briefly

anaesthetized with isoflurane (4% in oxygen, 2 L/min) to be disconnected from the shielded

cable.

4.2.7. Primary outcome: epileptic activity

For each rat, the median value and interquartile range for number of SWDs, duration of one

SWD and cumulative duration of SWDs over 30 min were calculated for eight recording

blocks, each block representing 1 week of recording. The first block only included one

recording whereas the following 7 blocks included 2 recordings (Figure 4.1C).

4.2.8. Secondary outcome: Post-operative health monitoring

For a minimum of three days post-surgery and until full recovery from the surgery, the rats

were monitored for weight loss once a day, and for mobility and grooming twice a day.

4.2.9. Statistical analysis

Data were analysed using commercially available software (IBM SPSS Statistic 22; Stata

13.0; StataCorp, 2013). The number of SWDs, mean duration of each SWD and cumulative

duration of SWDs were compared between the three groups using individual Kruskal-Wallis

tests for each block of the study. When significant values were found, post-hoc pairwise

comparisons were conducted for this block, comparing the control versus AED polymer, and

Page 83: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 61 -

blank polymer versus AED polymer conditions using the Mann-Whitney U test. Significance

level was set at 5%.

4.3. Results The FESEM images obtained from the PLGA mats with and without Phenytoin (Figure 2A

and 2B respectively) show a smooth and regular morphology characteristic of PLGA

electrospun fibres and indicated that there was no difference between the fibre morphology

and diameter range. Measured by visual inspection of the FESEM images, the fibre diameter

ranged between 2 µm and 5 µm.

Figure 4. 2 Scanning electron micrographs of coaxially electrospun polymer mats.

Blank PLGA polymer mat (A), Phenytoin loaded PLGA polymer mat (B). Scale bar represents 10

µm.

The in vitro release results for the PLGA-phenytoin mats are presented in figure 4.3 and show

linear release of phenytoin over a 20-day time with the release not demonstrating substantial

plateau off. A total of approximately 11% of the incorporated phenytoin has been released by

day 20.

PLGA

PLGA

PLGA-PhenytoinA B

C D

Page 84: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 62 -

Figure 4. 3 In vitro release data are presented for the phenytoin polymer (A)

Bars are representing standard deviation.

Epileptic activity monitoring results for the groups control, blank polymer and phenytoin

polymer are presented in figure 4.4. On week 1, 2 and 3 the mean numbers of SWD did not

significantly differ among the groups, however the mean duration of each SWD and the

cumulative time spent having SWD was significantly lower in phenytoin polymer group

compared to the control group (p= 0.006, 0.016, 0.004 and p=0.006, 0.25, 0.25 respectively).

During that same period the mean duration of individual SWD and the cumulative time spent

having SWD were not significantly different in the phenytoin polymer group compared to the

blank polymer group. For week 4 to 8, no significant differences were seen between any of

the groups on any of the variables tested.

0

2

4

6

8

10

12

0 5 10 15 20 25

Cu

mu

lati

ve R

ele

ase %

Time (days)

Page 85: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 63 -

Figure 4. 4 Comparison of the epileptic activity monitoring results (not including the silicone

group)

Epileptic activity monitoring results were obtained for groups control (n=6), blank polymer (n=6) and

phenytoin polymer (n=6). SWD: Spike and wave discharge; IQR: Interquartile range; Results are

reported as median; * Phenytoin polymer group significantly different when compared to control

group (p<0.05).

Page 86: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 64 -

As the rats from the silicone group were on average 2 months older and not randomly

allocated, their epileptic activity monitoring results are presented in a different figure (figure

5). The silicone group showed a similar decreased in seizure activity immediately following

surgery when compared to the blank polymer group however those results were not tested for

statistical significance.

No post-operative complication was detected in any of the rats and they were all fully

recovered within 3 days.

Page 87: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 65 -

Figure 4. 5 Comparison of the epileptic activity monitoring results from group silicone versus

group blank polymer.

Epileptic activity monitoring results obtained for groups blank polymer (n=6) and silicone sheet

(n=4). SWD: Spike and wave discharge; IQR: Interquartile range; Results are reported as median. The

silicone group showed a similar decreased in seizure activity following surgery when compared to the

blank polymer group however those results were not tested for statistical significance.

Page 88: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 66 -

4.4. Discussion

Drug release from electrospun polymeric structures typically follows zero order

kinetics.(Viry, Moulton et al. 2012) Zero order kinetics implies a homogeneous drug

distribution and a release profile governed by the wetting properties of the material and

encapsulation of hydrophilic and neutrally charged drugs of low molecular weight can be

problematic for these types of structures.(Ishihara and Mizushima 2010, Jyothi, Prasanna et

al. 2010, Kluge, Mazzotti et al. 2010, Tiwari, Tzezana et al. 2010) The interaction of these

types of drug molecules with the polymer is usually very poor and their rate of diffusion is

often faster than the rate of polymer erosion. This fast rate of diffusion has detrimental effects

on drug release from electrospun polymer structures. Several factors could be responsible for

variations in release, namely the solubility of the drug in the mat, the morphology of the

fibres within the mat and the distribution of the drug throughout the fibres (i.e., the degree of

drug encapsulation within the core of the coaxial spun mats). The low percentage of released

phenytoin at day 20 (Figure 4.3) is most likely, in part, the result of its solubility. As

discussed above the less soluble a drug is in aqueous solutions (when all other parameters are

constant between the mats such as polymer type, polymer concentration and fabrication

conditions) the more likely it is for this drug to elute from the structure at a slower rate. The

solubility of phenytoin in aqueous solution is 71 mg/L therefore suggesting that the elution of

the phenytoin should be slow which is what is reflected in the release profile presented in

Figure 4.3.

Biodegradable PLGA polymer sheets containing a large amount of phenytoin were implanted

above the motor cortices of GAERS rats. In this strain of rats, 100% of the animals present

recurrent generalized non-convulsive seizures characterized by bilateral and synchronous

SWD accompanied with behavioural arrest, staring and sometimes twitching of the vibrissae.

(Marescaux, Vergnes et al. 1992) Although depth EEG recordings and lesion experiments

show that SWD in GAERs depend on cortical and thalamic structures with a possible

rhythmic triggering by the lateral thalamus, more recent studies indicate a seizure initiation

site within the peri-oral region of the somatosensory cortex (S1po) as well as the

somatosensory cortex forelimb region (S1FL) (Marescaux, Vergnes et al. 1992, Manning,

Richards et al. 2004, Gurbanova, Aker et al. 2006) Neurophysiological, behavioural,

Page 89: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 67 -

pharmacological and genetic studies have demonstrated that spontaneous SWD in GAERS

fulfil all the requirements for an experimental model of absence epilepsy.(Marescaux,

Vergnes et al. 1992) Twenty-minute recordings were used during the experiment as this is the

duration used by the original paper describing GAERS EEGs.(Marescaux, Vergnes et al.

1992)

Although spontaneous SWD start and end abruptly on a normal background EEG and are

quite easy to isolate, the EEG patterns seen during sleep make it more difficult to differentiate

start and stop of SWD. During the experiment, rats were stimulated when seen sleeping to

improve seizure detection. This intervention could be reduced by inverting the light cycle of

the rats. The EEG recordings of those diurnal animals would then be recorded during the time

of maximal activity reducing sleep time EEG interferences. SWDs usually occur at a mean

frequency of 1.5 per min when the animals are in a state of quiet wakefulness and have

duration of 0.5 to 75 s. (Marescaux, Vergnes et al. 1992)

During the first 3 weeks of the experiment, the phenytoin polymer group had shorter duration

of SWDs when compared to the control group however there were no significant differences

when compared to the blank polymer group. In consequence, the phenytoin was not

responsible for this effect, which could be attributed to brain injuries resulting from the

surgery or the PLGA polymers and their degradation products. The phenytoin was chosen for

its lipophilic properties allowing easy drug loading within the polymer and its proven

efficacy in GAERS when applied directly over S1po and S1FL.(Gurbanova, Aker et al. 2006)

However, the mats were placed over the motor cortex and the delivery of phenytoin even

only 2 mm cranially to S1po and S1Fl resulted in no changes in duration and numbers of

SWDs. The minimal amount of drug release observed in the in vitro experiment may also

have been a contributing factor to the negative results.

The presence of an anti-epileptic effect after local administration and the absence of effect

after systemic injections may be due to phenytoin’s different selectivity of sodium channels

in the cortex and other brain areas and have already been discussed elsewhere.(Gurbanova,

Aker et al. 2006)

Page 90: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 68 -

To better understand the effect seen during the first 3 weeks, silicone sheets were implanted

in four older GEARS from the same colony. Although the SWD activity in GAERS is age

dependent, preventing statistical comparison with the previous groups (blank polymer and

phenytoin loaded polymer groups), the post-surgical transient decrease in SWD’s duration

was observed again. We could conclude that the surgery itself, and not the PLGA sheets, was

responsible for the decreased duration of SWDs, which is in agreement with previous

publications reporting that PLGA-based implants are very well tolerated by the brain in

animal models of other neurological disorders.(McRae, Hjorth et al. 1991, McRae and

Dahlstrom 1994, Gouhier, Chalon et al. 2000, Menei, Pean et al. 2000, Pean, Menei et al.

2000, Arica, Kas et al. 2005)

We have demonstrated that phenytoin loaded PLGA polymer can provide constant delivery

of phenytoin for at least 20 days (end of experiment). However, we haven’t been able to

demonstrate that phenytoin loaded PLGA polymer sheets implanted on the surface of the

cortex could decrease seizure activity in GAERS for a sustained period. In the next chapter

we will endeavour to improve the experimental protocol and test a more soluble and broad

spectrum AED (i.e.lacosamide).

Page 91: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 69 -

Chapter 5 – Anti-epileptic effects of lacosamide loaded polymers

implanted subdurally in GAERS

5.1 Outline

Following the negative in vivo results obtained in chapter 4, a more water soluble and broad

spectrum AED was selected and modifications in the experimental protocol were made. The

daylight cycle of the rats was inverted in order to record EEGs during a period of more

intense activity and in consequence reduce the artefacts produce by sleep EEG pattern. Only

three groups of rats were selected: control group (no implant; n=6), blank polymer group

(bilateral implantation of PLGA polymers not containing AED; n=6) or treatment group

(bilateral implantation of lacosamide loaded PLGA polymers; n=6). The surgical technique

was refined, the piece of the dura exposed following craniotomy was removed and the skull

was repositioned directly above the polymer. The EEG recording time at each recording

session was extended to 30 minutes and the EEG sessions occurred 3 days a week instead of

two. Finally, for a minimum of three days post-surgery and until full recovery from the

surgery, the rats were monitored for weight loss once a day, and for mobility and grooming

twice a day, and scores were attributed accordingly.

The aim of the study in chapter 5 remains similar to one presented in chapter 4: to provide

some in vitro product characterization and to investigate the effect of subdural implantation

of biodegradable PLGA polymers loaded with lacosamide on seizure activity in the GAERS

animal model of epilepsy. The hypothesis was that subdural implantation of AEDs loaded

PLGA polymers can decrease seizure activity of the GAERS.

5.2 Manuscript

This chapter is presented as the author-accepted version of a peer-reviewed manuscript

published in International Journal of Polymer Science, Volume 2016 (2016), Article ID

6594960, 10 pages, ttp://dx.doi.org/10.1155/2016/6594960. The figures’ numbers and titles

of the author-accepted version of the manuscript may have been changed to follow the

formatting of the thesis.

Page 92: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 70 -

Anti-epileptic Effects of Lacosamide Loaded Polymers Implanted Sub-durally in

GAERS Rats

Sebastien H. Bauquier a, *

, Jonathan L. Jiang b, Zhilian Yue

c, Alan Lai

b, Yu Chen

c, Simon E.

Moulton c,d*

, Karen J. McLean b, Sara Vogrin

b, Amy J. Halliday

b, Gordon Wallace

c, Mark

J. Cook b

a Faculty of Veterinary Science, University of Melbourne, 250 Princes Hwy, Werribee,

Victoria 3030, Australia

b Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s Hospital

Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

c Intelligent Polymer Research Institute and ARC Centre of Excellence for Electromaterials

Science, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, New

South Wales 2522, Australia

d Faculty of Science, Engineering and Technology, Swinburne University of Technology,

Hawthorn, Victoria, 3122, Australia

* Corresponding authors Tel.: +61 3 9731 2311; fax: +613 9731 2377 (S.H. Bauquier)

Tel: +61 3 9214 5651 (S.E. Moulton)

E-mail address [email protected] (S.H. Bauquier)

[email protected] (S.E. Moulton)

Page 93: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 71 -

ABSTRACT

The current experiment investigated the ability of coaxial electrospun Poly(D,L-lactide-co-glycolide)

(PLGA) biodegradable polymer implants loaded with the anti-epileptic drugs (AED) lacosamide to

reduce seizures following implantation above the motor cortex in the Genetic Absence Epilepsy Rat

from Strasbourg (GAERS).

In this prospective, randomized, masked experiments, GAERS underwent surgery for implantation of

skull electrodes (n=6), skull electrodes and blank polymers (n=6), or skull electrodes and lacosamide

loaded polymers (n=6). Thirty-minute electroencephalogram (EEG) recordings were started at day 7

post-surgery and continued for eight weeks. EEGs were analyzed using an automated detection

algorithm. The number of SWDs and mean duration of one SWD were compared week-by-week

between the three groups using Kruskal-Wallis tests and when significant values were found, post-hoc

pairwise comparisons were conducted using the Mann-Whitney U test. Significance level was set at

5%.

There was no difference in the number of SWD between any of the groups. However, the mean

duration of one SWD was significantly lower in the lacosamide polymer group for up to 7 weeks

when compared to the control group (0.004 <p< 0.038). The mean duration of one seizure was also

lower at week 3, 5, 6 and 7 when compared to the blank polymer group (p= 0.016, 0.037, 0.025 and

0.025 respectively).

We have demonstrated that AED loaded PLGA polymer sheets implanted on the surface of the cortex

could affect seizure activity in GAERS by decreasing the mean duration of the SWDs events for a

sustained period of up to 7 weeks.

Keywords: Absent seizure; Antiepileptic Drug Delivery; Biodegradable; Epilepsy; PLGA;

SWD.

Page 94: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 72 -

Introduction

Epilepsy is a chronic neurological condition characterized by recurrent seizures. The

incidence of epilepsy in most developed countries is between 50 and 100 cases per 100,000

population per year although it is estimated that up to 5% of a population will experience

non-febrile seizures at some point in life (Shorvon 1996, Sander 2003). Individuals with

medically untreatable epilepsy often have impaired ability to work or function socially (e.g.

inability to drive, difficulty at attending school, losing jobs and friends, anxiety regarding the

possibility of having seizure in potentially hazardous conditions) (Swarztrauber, Dewar et al.

2003). Treatment with conventional anti-epileptic drugs (AEDs; e.g.: phenytoin and

lacosamide administered orally) results in only 33% of the patients having no seizure

recurrence (Shorvon 1996, Sander 2003). Alternatively, neuro-stimulation based therapy has

also shown to reduce seizure activity but has typical reductions of seizure frequency of

approximately 40% acutely and 50–69% after several years (Fisher and Velasco 2014).

Surgical resection of the seizure focus can be performed in the case of focal seizures,

however this procedure can only be applied on selected patients depending on the localization

of the epileptic foci (Kwan and Sperling 2009). Indeed, the success of inducing long-lasting

seizure remission from epilepsy surgery range from a low of 25% for patients exhibiting

extrahippocampal seizure origin to 70% in appropriately selected candidates (Kwan and

Sperling 2009).

The mechanisms by which resistance to AEDs treatment develops are not fully understood,

some evidence suggests that this may be due to a lack of effective penetration into the brain

parenchyma, however the drug side effects prevent large increase in the posology(Loscher

2005). Alternative therapies aiming at improving the availability of AEDs such as the

Page 95: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 73 -

intracranial implantation of polymer-based drug delivery systems are being investigated

(Halliday, Moulton et al. 2012, Halliday, Campbell et al. 2013). This targeted drug delivery

approach has shown some success in the treatment of animal models of several neurological

disorders such as Parkinson’s disease, Huntington’s disease and Alzheimer’s disease

(Halliday and Cook 2009). Also, Halliday et al. (2013) used Levetiracetam-loaded

biodegradable polymer implants in the tetanus toxin model of temporal lobe epilepsy in rats;

the results of this study indicated that drug-eluting polymer implants represent a promising

evolving treatment option for intractable epilepsy, however important limitations of the study

were that the effects could only be seen for a week and only a single group of control animals

were investigated. These animals received an injection of tetanus toxin and a sham

craniotomy, without the implantation of a polymer sheet (Halliday, Campbell et al. 2013).

Poly(D,L-lactide-co-glycolide) (PLGA) is the most commonly used biodegradable polymer as

it is highly biocompatible, easily engineered and has been approved for drug delivery

purposes by the United States Food and Drug Administration (Halliday, Campbell et al.

2011). It has been used in numerous applications including bone and skin tissue engineering,

ocular treatment, vaccine, cancer therapy, nerve regeneration and many more (Allahyari and

Mohit 2015, Cao, Chen et al. 2015, Groynom, Shoffstall et al. 2015, Yu, Zhao et al. 2015,

Zhang, Song et al. 2015). PLGA polymers have also been successfully used for intracranial

drug delivery in animal models of neurological disorders, showing no evidence of toxic

injury or immune-mediated inflammation when implanted subdurraly above the motor cortex

in rats (Halliday, Campbell et al. 2011).

Genetic Absent Epilepsy Rats from Strasbourg (GAERS) is a strain of rats where 100% of

the animals present with recurrent generalized non-convulsive seizures (Marescaux, Vergnes

Page 96: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 74 -

et al. 1992). This animal model has become the gold standard to study the mechanisms

underlying absence epilepsy (Marescaux, Vergnes et al. 1992). In the present investigation,

PLGA sheets loaded with the commonly used AED lacosamide were developed and their

ability to decrease seizure activity was investigated in the GAERs. Lacosamide stabilizes

neuronal membranes through enhancing slow inactivation of voltage-gated sodium channel

and is effective in different rodent seizure models including generalized seizure (Beyreuther,

Freitag et al. 2007).

The aim of the study is to investigate the effect of sub-dural implantation of biodegradable

polymers (PLGA) loaded with lacosamide on seizure activity in an animal model of epilepsy

(GAERS); the hypothesis is sub-dural implantation of lacosamide loaded PLGA polymers

can decrease seizure activity of the GAERS. Our results demonstrated that focal delivery of

lacosamide can achieve partial sustained anti-epileptic effect in an animal model of

generalized epilepsy.

MATERIALS AND METHODS

The study was designed as a randomized controlled masked experiments. The experiment

was approved by St Vincent’s Hospital (Melbourne) Animal Ethics Committee and

conducted in accordance with the Australian Code of Practice for the Care and Use of

Animals for Scientific Purposes (2004).

Page 97: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 75 -

1. PLGA polymer mat production

Lacosamide-laden polymer mats were produced using a coaxial electrospinning method

(Viry, Moulton et al. 2012), with the core composed of lacosamide and 75:25 PLGA

(lactide/glycolide = 75:25), and the shell composed of 85:15 PLGA (lactide/glycolide =

85:15). The core solution was prepared at 17% w/v 75:25 PLGA in dimethylformamide

(DMF), to which lacosamide was added to give a range of final concentration of 2.5, 12.5 or

20% w/w relative to the polymer. The shell solution was prepared at 20% w/v 85:15 PLGA in

a binary solvent system comprising dichloromethane and DMF (dichloromethane/DMF =

7/3). Coaxial electrospinning was conducted using a Nano-electrospinning system (NANON-

01A, MECC CO. Ltd) at an applied DC voltage of 23 kV. A coaxial spinneret with a

diameter of 0.2 mm for the core and 0.8 mm for the sheath nozzles was connected to

the core and shell solutions and a feed rate of 5 µL min-1

and 20 µL min-1

for the core and

shell solutions respectively. Fibre mats were collected on a grounded plate collector that was

set 15 cm away from the spinneret tip. The drug-free, coaxially spun polymer mats were also

prepared using the above procedure.

Depending on lacosamide loading in the core, the as-prepared, coaxially electrospun polymer

mats were denoted as PLGA-2.5%, PLGA-12.5% and PLGA-20%. The drug-free mats are

denoted as blank polymers. All the samples were finally dried in a vacuum oven at 40 oC for

72 h to remove residual solvent and were stored at -20 oC prior to subsequent physiochemical

characterization and animal studies.

Page 98: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 76 -

2. Morphology study

The morphology of the electrospun mats (with and without drug) was examined using a field

emission scanning electron microscope (FESEM, JEOL JSM-7500FA). The samples were

sputter-coated with gold prior to FESEM to avoid sample charging.

3. Determination of drug loading in polymer mats

An extraction method was used to determine the drug loading in the electrospun mats.

Briefly, each sample (1 cm × 1 cm) was weighed and placed into 1 mL methanol for 12 hours

after which the methanol was removed and replenished with 1 mL of fresh methanol. This

extraction procedure was repeated four times with each methanol sample allowed to

evaporate to leave residual drug behind which was reconstituted in methanol (1 mL), diluted

20 times with the HPLC mobile phase (see below) and filtered through a 0.2 µm syringe

filter. The fourth reconstituted sample showed no presence of drug indicating that the entire

drug had been extracted from the electrojetted sample.

4. In vitro drug release

The lacosamide-loaded polymers (1 cm × 1 cm) were suspended in 1 mL of aCSF, and the

release experiment was conducted at 37 oC in a shaker water bath (Julabo Pty. Ltd.). The

aCSF contained NaCl (0.866% w/v), KCl (0.224% w/v), CaCl2-2H2O; (0.0206% w/v) and

MgCl2-6H2O (0.0164% w/v) in 1 mM phosphate buffer (pH 7.4) (Halliday, Campbell et al.

2013). For each sample, the release medium solution was collected and replenished with

fresh aCSF at various time points, and stored at -20 oC prior to HPLC analysis being

undertaken. The eluted samples were analyzed by HPLC using a modified method on an

Agilent 1260 Infinity HPLC system (Lin, Hsieh et al. 2010). An Atlantis

T3 C18 column

(250 mm × 4.6 mm, 5 µm) was employed as the analytic column and set at 40 oC. The mobile

Page 99: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 77 -

phase was composed of water, acetonitrile and methanol (65:26.2:8.8, v/v/v), the injection

volume was 10 L with the flow rate of 0.8 mL/min. The eluting lacosamide was detected

using a UV-vis detector set at a wavelength of 230 nm (max of lacosamide). To convert the

UV-vis absorbance to drug concentration a standard curve was established by plotting in

triplicate the UV-vis peak areas against respective concentrations of standard solutions (10,

20, 50, 100, 200, 500uM Lacosamide).

5. Animals

Adult female GAERS were obtained from the University of Melbourne (Parkville, Victoria,

Australia) and housed individually in inverted 12-hour light/dark cycles (the light was turn

off between 6 am and 6 pm) with ad libitum access to food and water. Six-month-old rats

were randomly allocated to a control group (no implant; n=6), blank polymer group (bilateral

implantation of blank PLGA polymers not containing lacosamide; n=6) or treatment group

(bilateral implantation of lacosamide loaded PLGA polymers; n=6). The randomization was

performed using the random function in Microsoft Excel 2007. The group attribution list was

kept concealed from the researchers performing the EEG analysis.

5.1.Implantation surgery

Immediately prior to surgery, rats were weighted and anaesthetized using a balanced

anaesthesia protocol including an intra-peritoneal injection of ketamine (75 mg/kg) and

xylazine (10 mg/kg). Following anaesthesia induction rats were placed in a stereotaxic

apparatus, and administered isoflurane (0.5 to 1% in oxygen, 1 L/min) via a nose-cone as

needed, and were administered subcutaneous (SQ) carprofen (5 mg/kg) for pain relief and

0.9% sodium chloride (2 mL) for cardiovascular support.

Page 100: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 78 -

Rats from the control group underwent surgery for EEG recording electrode implantation

whereas surgeries for the rats from the blank polymer group, the treatment group and the

silicone group also included bilateral craniotomies for subdural placement of two identical

implants. Over the scalp of all rats, the hair was clipped and the skin aseptically prepared. A

single incision was made down the midline, the skull cleared of tissue and the exposed bone

dried with 3% hydrogen peroxide. Five extradural electrodes, consisting of small jeweller’s

screws, were implanted caudal to the intended polymer implantation sites through burr holes.

Four were implanted cranially to the interaural line (two on each side of the sagittal suture)

and one caudally to the interaural line on the right side of the sagittal suture (Figure 1A and

B). The electrodes were then connected to an adaptor and secured with dental cement.

Implants measuring 3 mm by 4 mm were cut from the polymer sheets described above.

Placements of the implants were performed after 5 mm by 4 mm craniotomies were created

bilaterally at the level of the coronal suture (over the motor cortices) and after excising the

dura to expose the brain surface (Figure 1A and B). Following implant placements, the skull

removed from craniotomy sites was replaced. The craniotomy sites were then sealed with an

alginate-based hydrogel, the entire surgical site covered with dental cement and the skin

sutured leaving exposed only part of the dental cement. The animals were placed on heat pads

for recovery. Post-operative treatment included SQ buprenorphine every twelve hours

(0.03mg/kg, twice a day), saline (2mL, once a day) and carprophen SID (5mg/kg, once a day)

for up to 3 days.

Page 101: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 79 -

Figure 5. 1 Polymers and electrodes implantation sites, and timeline of the experiment.

Schematic diagram illustrating the positioning of epidural recording screw electrodes (1-4), reference

electrode ® and polymer (A); Picture of the actual surgery (B); Timeline of the study design: For a

minimum of three days post-surgery the rats were monitored for weight loss once a day, and for

mobility and grooming twice a day. Thirty minutes electroencephalogram recording began at day 7 or

8 post surgery and for the following 7 weeks on 3 days per week. For each rat, the mean desired value

(number of SWDs, duration of one SWD or cumulative duration of SWDs over 30 min) were

calculated for eight recording blocks, each block being 1 week of duration.

5.2.Electroencephalograph recording and analysis

At day 7 or 8 post surgery and at least 3 days per week for the following 7 weeks, rats were

monitored for one hour (half an hour anaesthesia recovery/acclimation time, half an hour

recording time) (Figure 1C). At each monitoring sessions, rats were briefly anaesthetized in

an induction cage with isoflurane (4% in oxygen, 2 L/min, for 2 to 3 minutes), and shielded

Page 102: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 80 -

cables were used to connect the recording electrodes to the EEG acquisition system, which

consisted of TDT processors and high impedance head stages driven by custom-designed

software (Tucker Davis Technologies, USA). The rats were allowed to recover from the

anesthetic before recordings began. The EEGs were visualized using a custom-designed

MATLAB program (The MathWorks, Inc., U.S.A.). During EEG recording, if the rats were

perceived as being asleep, and after confirmation of no seizure activity on the EEG, a noise

stimulus between 94 and 98 decibels was applied. At the end of a recording session, the rats

were again briefly anaesthetized with isoflurane (4% in oxygen, 2 L/min) to be disconnected

from the shielded cable. The researcher performing EEG analysis was masked to the

treatment and used a GAERS specific automated spike-and-wave discharges (SWDs)

detection algorithm.(Bauquier, Lai et al. 2015)

6. Primary outcome: Epileptic activity

For each rat, the median value and interquartile range for number of SWDs, duration of one

SWD and cumulative duration of SWDs over 30 min were calculated for eight recording

blocks, each block representing 1 week of recording. The first block only included one

recording at day 6 or 7 post-surgery (total of 30 minutes of EEG recording) whereas the

following 7 blocks included 3 recordings per week (total of 90 minutes of EEG recording per

week) (Figure 1C).

7. Secondary outcome: Post-operative health monitoring

For a minimum of three days post-surgery and until full recovery from the surgery, the rats

were monitored for weight loss once a day, and for mobility and grooming twice a day. Those

observations were given scores (Table 5.1). For each day a debilitation score was calculated

by cumulating the highest grooming and mobility scores with the weight loss score (Table 1).

Page 103: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 81 -

A debilitation score of 0, 1 to 3, 4 to 6 and 7 to 9 meant that the rat health was not affected,

mildly affected, moderately affected, severely affected by the surgery/polymer implantation

respectively. The highest debilitation score of each rat over the post-operative period is

reported.

Table 5. 1 Post-operative health monitoring chart.

For a minimum of three days post-surgery and until full recovery from the surgery, the rats were

monitored for weight loss once a day, and for mobility and grooming twice a day. For each day a

debilitation score was calculated by cumulating the highest grooming and mobility scores with the

weight loss score. A debilitation score of 0, 1 to 3, 4 to 6 and 7 to 9 meant that the rat health was not

affected, middle affected, moderately affected, severely affected by the surgery/polymer implantation

respectively.

Score

Percentage of weight loss

No weight loss

0

less than 10% weight loss

1

10% to less than 20% weight

loss

2

20% or more weight loss

3

Mobility Score

Rat moving normally

0

Rat ataxic moving at a normal

speed

1

Rat ataxic and moving slowly

2

Rat recumbent

3

Grooming Score

No decrease in grooming

activity

0

Middle decrease in grooming

activity

1

Moderate decrease in grooming

activity

2

No grooming activity

3

Total score = debilitation

score

0-9

Page 104: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 82 -

8. Statistical analysis

Data analysis was performed using a commercially available software (IBM SPSS Statistic

22; Stata 13.0; StataCorp, 2013). The number of SWDs, mean duration of one SWD and

cumulative duration of SWDs were compared between the three groups using individual

Kruskal-Wallis tests for each block of the study. When significant values were found, post-

hoc pairwise comparisons were conducted for this block, comparing the control versus

lacosamide polymer, and blank polymer versus lacosamide polymer conditions using the

Mann-Whitney U test. The debilitation scores of group lacosamide were compared to group

control and group blank polymer using a 2-tailed Mann-Whitney exact test. Significance

level was set at 5%.

RESULTS

The FESEM images obtained from the PLGA mats without lacosamide (figure 2A) show a

smooth and regular morphology characteristic of PLGA electrospun fibres. However when

lacosamide was incorporated into the PLGA the fibres within the mats demonstrated a

mixture of porous and non-porous morphology when examined under FESEM (figure 2B).

Measured by visual inspection of the FESEM images, the fibre diameter ranged between 2

µm and 5 µm. These observations of the PLGA-12.5% samples shown in figure 2C were also

observed in the other PLGA-2.5% and PLGA-20.0% samples.

The in vitro release results for the PLGA-lacosamide mats are presented in figure 2C. The

polymer containing 20% lacosamide showed a cumulative drug release above 40% after

only7 days. The polymers containing 2.5 and 12.5 % lacosamide both had a reduction in

release after around 42 days (6 weeks) however the total drug release was higher for 12.5%

Page 105: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 83 -

lacosamide polymer. In consequence, the polymer containing 12.5% lacosamide was chosen

to be implanted for the in vivo experiment.

Figure 5. 2 Electron micrographs of coaxially electrospun polymer mats and in vitro release

data.

Scanning electron micrographs of coaxially electrospun polymer mats of PLGA Blank (A) and

PLGA-12.5% lacosamide (B) (Scale bar represents 10 µm). In vitro release data of lacosamide

polymers with final core lacosamide concentration of 2.5, 12.5 and 20% w/w relative to the

polymer (C). Bars are representing standard deviation.

Page 106: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 84 -

The results for the postoperative debilitation score are presented in figure 3. One rat from the

blank polymer group and 3 rats from the lacosamide had a debilitation score that were

classified as moderate. One rat from the blank polymer group and 3 rats from the lacosamide

group had a debilitation score that were classified as severe. The debilitation scores were

significantly increased for the lacosamide polymer group when compared to the control and

blank polymer groups (p=0.002 and p=0.041 respectively).

Figure 5. 3 Post-operative debilitation scores

Post-operative debilitation scores obtained for groups control (n=6), blank polymer (n=6) and

lacosamide polymer (n=6). Rats from the lacosamide polymer group had debilitation scores that were

higher when compared to the 2 other groups (*: p=0.002; **: p=0.041). The box indicates the

interquartile range (25th to 75th percentile), and the whiskers indicate the range.

Page 107: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 85 -

The results of the measurements performed to evaluate the epileptic activity for the groups

control, blank polymer and lacosamide polymer are presented in figure 4. The difference in

the number of SWDs between the groups was not statistically significant. However, the mean

duration of one SWD was significantly lower in the lacosamide polymer group for up to 7

weeks when compared to the control group (p=0.037, 0.004, 0.01, 0.025, 0.037 and 0.016 for

week 1, 2, 3, 4, 6, and 7 respectively). The mean duration of one seizure was also lower at

week 3, 5, 6 and 7 when compared to the blank polymer group (p= 0.016, 0.037, 0.025 and

0.025 respectively). The cumulative duration of SWDs of the lacosamide group was

significantly lower when compared to the control group at week 1 and 5 (p=0.010 and

p=0.055 respectively) and when compared to the blank polymer group at week 3 and 5

(p=0.010 and p=0.055 respectively). Examples of EEG recordings are presented on figure 5.

Page 108: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 86 -

Figure 5. 4 Comparison of the epileptic activity monitoring results

Comparison of the measurements obtained to evaluate the epileptic activity of groups control (n=6),

blank polymer (n=6) and lacosamide polymer (n=6). SWD: Spike and wave discharge; IQR:

Interquartile range; Results are reported as median. * and

+ represent the time points at which the

results from the lacosamide polymer group were significantly different to the control and blank

polymer groups respectively (p<0.05).

Page 109: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 87 -

Figure 5. 5 Example of EEG recordings

Example of EEG recording from 3 rats belonging to group control (A), blank polymer (B) and

lacosamide polymer (C). Figures D and E are amplifications of spike and wave discharges observed

during the recording shown and figure A and C respectively.

Page 110: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 88 -

DISCUSSION

Drug release from electrospun polymeric structures typically follows zero order kinetics

(Viry, Moulton et al. 2012). Zero order kinetics implies a homogeneous drug distribution and

a release profile governed by the wetting properties of the material and encapsulation of

hydrophilic and neutrally charged drugs of low molecular weight can be problematic for

these types of structures (Ishihara and Mizushima 2010, Jyothi, Prasanna et al. 2010, Kluge,

Mazzotti et al. 2010, Tiwari, Tzezana et al. 2010). The interaction of these types of drug

molecules with the polymer is usually very poor and their rate of diffusion is often faster than

the rate of polymer erosion. This fast rate of diffusion has detrimental effects on drug release

from electrospun polymer structures.

Several factors are responsible for the variations in the release profiles of AEDs, namely the

solubility of the drug in the mat, the morphology of the fibres within the mat and the

distribution of the drug throughout the fibres (i.e., the degree of drug encapsulation within the

core of the coaxial spun mats). The solubility of lacosamide (465 mg/L) contributes to the

relatively fast elution of that drug from the polymer. Also, the morphology of the fibers

within the electrospun mats shows a porous nature for the PLGA-Lacosamide structures

which increases the rate at which the release media (aCSF) can infuse into the internal region

of the fibers and promote the elution of the drug.

The initial rapid drug release observed in the in vitro experiment for the polymer mats tested

with a final core lacosamide concentration of 12.5 % (figure 2C) coincided with the post-

surgical debilitation seen in most animals of the lacosamide polymer group. It is unlikely that

the surgery alone was responsible for these adverse events as this debilitation was not

Page 111: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 89 -

observed to such extent in the blank polymer group. Debilitated rats were treated with fluid

therapy and analgesic administration and they all recovered well. Looking at the results

retrospectively, one could argue that implanting the PLGA-2.5% may have been a better

choice as the initial release of drug is not as abrupt and the constant lacosamide release seem

to last longer. Indeed the lack of effect of lacosamide after 7 weeks correlate with the almost

absence of release of the lacosamide from the polymer. Variations in the release profiles

observed in figure 5.2.C from the samples prepared with varying amounts of Lacosamide

indicates that the interaction of the drug with the polymer (and hence its propensity to be

released from the structures) is influenced by the drug loading. It has previously been shown

that the amount of drug loaded into electrospun fibres and drug-polymer-electrospinning

solvent interactions has an effect of the release profiles (Chew, Wen et al. 2005, Zeng, Yang

et al. 2005, Cui, Li et al. 2006, Xie 2009).

Biodegradable PLGA polymer sheets containing a large amount of lacosamide were

implanted above the motor cortices of GAERS rats. In the GAERS model of epilepsy, the rats

present recurrent generalized non-convulsive seizures characterized by bilateral and

synchronous SWD accompanied with behavioral arrest, staring and sometimes twitching of

the vibrissae (Marescaux, Vergnes et al. 1992). Furthermore, the GAERS rats were at least 6

months old, time at which 100% of the GAERS should present SWDs and at which the

numbers of SWDs are at their maximum.(Marescaux, Vergnes et al. 1992) Although depth

EEG recordings and lesion experiments show that SWD in GAERs depend on cortical and

thalamic structures with a possible rhythmic triggering by the lateral thalamus, more recent

studies indicate a seizure initiation site within the peri-oral region of the somatosensory

cortex (S1po) as well as the somatosensory cortex forelimb region (S1FL) (Marescaux,

Page 112: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 90 -

Vergnes et al. 1992, Manning, Richards et al. 2004, Gurbanova, Aker et al. 2006).

Neurophysiological, behavioral, pharmacological and genetic studies have demonstrated that

spontaneous SWD in GAERS fulfill all the requirements for an experimental model of

absence epilepsy (Marescaux, Vergnes et al. 1992). Although twenty-minute recording were

used by the original paper describing GAERS EEGs, to try to improve the performance of the

EEG analysis 30 min recording were used during the present experiment (Marescaux,

Vergnes et al. 1992).

Although spontaneous SWD start and end abruptly on a normal background EEG and are

quite easy to isolate, the EEG patterns seen during sleep make it more difficult to differentiate

start and stop of SWD(Bauquier, Lai et al. 2015). In previous experiments, rats were

stimulated when seen sleeping to improve seizure detection(Bauquier, Lai et al. 2015). This

intervention was reduced during the present experiment by inverting the light cycle of the

rats. The EEG recordings of those diurnal animals were then recorded during the time of

maximal activity reducing sleep time EEG interferences. SWDs usually occur at a mean

frequency of 1.5 per min when the animals are in a state of quiet wakefulness and have

duration ranging from 0.5 to 75 s (Marescaux, Vergnes et al. 1992).

Isoflurane was used for a short period to connect and disconnect the recording apparatus

before and after each recording. The authors found in previous experiments that during that

setup without the use of anaesthesia some rats were showing stress behaviour (like crying)

and in consequence the authors opted for a short general anaesthesia/sedation to improve

animal welfare. Isoflurane was chosen for its low blood:gas solubility (1.4) allowing quick

elimination. Indeed, rats recovered quickly from those short anaesthesia episodes and,

Page 113: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 91 -

although it cannot be excluded, it is unlikely that after 30 minutes isoflurane could still be

interfering with their epileptic activity (Bauquier, Lai et al. 2016).

Implantation of the lacosamide sheets led to shorter seizures for up to seven weeks after

implantation compared to rats that did not receive the implant.

During the first 2 weeks of the experiment, the blank polymer group also demonstrated

decrease duration of SWDs when compared to the control group and to better understand the

effect seen, silicone sheets were implanted in four older GAERS from the same colony

(unpublished data). Although the SWD activity in GAERS is age dependent, preventing

statistical comparison with the present experiment, the post-surgical transient decrease in

SWD’s duration was observed again. We could assume that the PLGA sheets were not

themselves responsible for the decreased duration of SWDs seen in the control blank polymer

group but could be attributed to brain injuries resulting from the surgery. This assumption is

in agreement with previous publication reporting that PLGA-based implants are very well

tolerated by the brain in animal models of other neurological disorders (McRae, Hjorth et al.

1991, McRae and Dahlstrom 1994, Gouhier, Chalon et al. 2000, Menei, Pean et al. 2000,

Pean, Menei et al. 2000, Arica, Kas et al. 2005).

Only one EEG recording per rat was performed during the first week at day 6 or 7 while there

were three recordings per week performed during the following seven weeks. This study

design allowed time for good surgical wound healing before some traction could be applied

to the electrodes ‘adaptor.

The lacosamide was chosen for its lipophilic properties allowing easy drug loading within the

polymer and its proven efficacy in treating absence epilepsy (Sodemann, Moller et al. 2014).

Page 114: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 92 -

Knowing that the polymer mats were implanted over the motor cortex, and that previous

investigations have shown that substances released from intraparenchymally implanted

polymers are able to penetrate around 3 mm, it is possible that the lacosamide released from

the sheets in our study may not have reached the seizure triggering focus in high enough

concentration to stop seizure from happening (Krewson, Klarman et al. 1995, Krewson,

Dause et al. 1996, Saltzman, Mak et al. 1999, Bensadoun, Pereira de Almeida et al. 2003).

However, lacosamide reduces the ability of epileptic neurons to endure extended firing burst

by enhancing slow inactivation of voltage-gated sodium channel (Kellinghaus 2009). This

would explain the unchanged number of SWDs but significant decrease in SWDs duration.

The total duration of SWDs during the recording SWD reflects the arithmetic product of the

number of seizures and the mean duration of SWDs. Another AED alternative could have

been the use of valproic acid which is one of the drugs of choice for the treatment of absence

seizure. Its use could have provided additional support for the validity of the novel delivery

method. Lastly, now that it has been shown that a lacosamide loaded PLGA polymer sheets

implanted on the surface of the cortex could affect the duration of the individual SWDs in

GAERS, performing dose-response experiments in order to determine optimal concentrations

of the drug in PLGA would be required to effectively improve the treatment and the novel

delivery method.

From the statistical perspective, Bender and Lange (2001) recommended that data of

exploratory studies be analyzed without multiplicity adjustment (Bender and Lange 2001).

However, the lack of adjustment for multiple comparisons as well as the pilot nature of our

study means that caution needs to be exercised when interpreting the results of this

exploratory study. A larger study is needed to confirm these findings.

Page 115: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 93 -

Although temporary side-effects were seen, we have demonstrated that lacosamide loaded

PLGA polymer sheets implanted on the surface of the cortex could affect seizure activity in

GAERS by decreasing the mean duration of the SWDs events for a sustained period of up to

7 weeks. With improvements in polymer technologies and episodic release offering

potentially much longer lasting release durations, intracranial polymer-based drug delivery

systems may provide an effective therapeutic strategy for chronic epilepsy.

Conflict of interest/disclosures

The authors declare that they have no financial or other conflicts of interest in relation to this

research and its publication.

Acknowledgements

The authors gratefully acknowledge the sponsorship of the Victorian Government through its

Science Technology and Innovation Initiative administered by the Department of Industry,

Innovation and Regional Development. The authors thank the Australian Research Council

(ARC) for continuing financial support. GGW thanks the ARC for his Laureate Fellowships.

The authors also acknowledge use of the facilities and the assistance of Mr. Tony Romeo at

the UOW Electron Microscopy Centre. The authors are grateful for access to materials

synthesis and characterization equipment made available through the Australian National

Fabrication Facility (ANFF).

Page 116: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 94 -

REFERENCES

[1] Sander JW. The epidemiology of epilepsy revisited. Curr Opin Neurol 2003, 16: 165-170.

[2] Shorvon SD. The epidemiology and treatment of chronic and refractory epilepsy. Epilepsia 1996,

37 Suppl 2: S1-S3.

[3] Swarztrauber K, Dewar S, Engel J, Jr. Patient attitudes about treatments for intractable epilepsy.

Epilepsy Behav 2003, 4: 19-25.

[4] Fisher RS, Velasco AL. Electrical brain stimulation for epilepsy. Nat Rev Neurol 2014, 10: 261-

270.

[5] Kwan P, Sperling MR. Refractory seizures: try additional antiepileptic drugs (after two have

failed) or go directly to early surgery evaluation? Epilepsia 2009, 50 Suppl 8: 57-62.

[6] Loscher W. Mechanisms of drug resistance. Epileptic Disord 2005, 7 Suppl 1: S3-9.

[7] Halliday AJ, Moulton SE, Wallace GG, Cook MJ. Novel methods of antiepileptic drug delivery --

polymer-based implants. Adv Drug Deliv Rev 2012, 64: 953-964.

[8] Halliday AJ, Campbell TE, Nelson TS, McLean KJ, Wallace GG, Cook MJ. Levetiracetam-loaded

biodegradable polymer implants in the tetanus toxin model of temporal lobe epilepsy in rats. J Clin

Neurosci 2013, 20: 148-152.

[9] Halliday AJ, Cook MJ. Polymer-based drug delivery devices for neurological disorders. CNS

Neurol Disord Drug Targets 2009, 8: 205-221.

[10] Halliday AJ, Campbell TE, Razal JM, McLean KJ, Nelson TS, Cook MJ, et al. In vivo

biocompatibility and in vitro characterization of poly-lactide-co-glycolide structures containing

levetiracetam, for the treatment of epilepsy. J Biomed Mater Res A 2011.

[11] Groynom R, Shoffstall E, Wu LS, Kramer RH, Lavik EB. Controlled release of photoswitch

drugs by degradable polymer microspheres. J Drug Target 2015, 23: 710-715.

[12] Allahyari M, Mohit E. Peptide/protein vaccine delivery system based on PLGA particles. Hum

Vaccin Immunother 2015: 0.

[13] Zhang F, Song Q, Huang X, Li F, Wang K, Tang Y, et al. A Novel High Mechanical Property

PLGA Composite Matrix Loaded with Nanodiamond-Phospholipid Compound for Bone Tissue

Engineering. ACS Appl Mater Interfaces 2015.

[14] Cao H, Chen MM, Liu Y, Liu YY, Huang YQ, Wang JH, et al. Fish collagen-based scaffold

containing PLGA microspheres for controlled growth factor delivery in skin tissue engineering.

Colloids Surf B Biointerfaces 2015, 136: 1098-1106.

[15] Yu K, Zhao J, Zhang Z, Gao Y, Zhou Y, Teng L, et al. Enhanced delivery of Paclitaxel using

electrostatically-conjugated Herceptin-bearing PEI/PLGA nanoparticles against HER-positive breast

cancer cells. Int J Pharm 2015.

Page 117: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 95 -

[16] Marescaux C, Vergnes M, Depaulis A. Genetic absence epilepsy in rats from Strasbourg--a

review. J Neural Transm Suppl 1992, 35: 37-69.

[17] Beyreuther BK, Freitag J, Heers C, Krebsfanger N, Scharfenecker U, Stohr T. Lacosamide: A

review of preclinical properties. Cns Drug Reviews 2007, 13: 21-42.

[18] Viry L, Moulton SE, Romeo T, Suhr C, Mawad D, Cook M, et al. Emulsion-coaxial

electrospinning: designing novel architectures for sustained release of highly soluble low molecular

weight drugs. Journal of Materials Chemistry 2012, 22: 11347-11353.

[19] Lin PC, Hsieh YH, Liao FF, Chen SH. Determination of free and total levels of phenytoin in

human plasma from patients with epilepsy by MEKC: an adequate alternative to HPLC.

Electrophoresis 2010, 31: 1572-1582.

[20] Bauquier SH, Lai A, Jiang JL, cook MJ. Evaluation of an automated spike-and-wave complex

detection algorithm in the EEG from a rat model of absence epilepsy. Neuroscience Bulletin 2015.

[21] Ishihara T, Mizushima T. Techniques for efficient entrapment of pharmaceuticals in

biodegradable solid micro/nanoparticles. Expert Opin Drug Deliv 2010, 7: 565-575.

[22] Jyothi NV, Prasanna PM, Sakarkar SN, Prabha KS, Ramaiah PS, Srawan GY.

Microencapsulation techniques, factors influencing encapsulation efficiency. J Microencapsul 2010,

27: 187-197.

[23] Kluge J, Mazzotti M, Muhrer G. Solubility of Ketoprofen in colloidal PLGA. Int J Pharm 2010,

399: 163-172.

[24] Tiwari SK, Tzezana R, Zussman E, Venkatraman SS. Optimizing partition-controlled drug

release from electrospun core-shell fibers. Int J Pharm 2010, 392: 209-217.

[25] Chew SY, Wen J, Yim EK, Leong KW. Sustained release of proteins from electrospun

biodegradable fibers. Biomacromolecules 2005, 6: 2017-2024.

[26] Cui W, Li X, Zhu X, Yu G, Zhou S, Weng J. Investigation of drug release and matrix

degradation of electrospun poly(DL-lactide) fibers with paracetanol inoculation. Biomacromolecules

2006, 7: 1623-1629.

[27] Zeng J, Yang L, Liang Q, Zhang X, Guan H, Xu X, et al. Influence of the drug compatibility

with polymer solution on the release kinetics of electrospun fiber formulation. J Control Release

2005, 105: 43-51.

[28] Xie ZB-D, G. . Electrospun Poly(D,L-lactide) Fibers for Drug Delivery: The Influence of

Cosolvent and the Mechanism of Drug Release. . J. Appl Polym. Sci. 2009, 115: 8.

[29] Manning JP, Richards DA, Leresche N, Crunelli V, Bowery NG. Cortical-area specific block of

genetically determined absence seizures by ethosuximide. Neuroscience 2004, 123: 5-9.

Page 118: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 96 -

[30] Gurbanova AA, Aker R, Berkman K, Onat FY, van Rijn CM, van Luijtelaar G. Effect of

systemic and intracortical administration of phenytoin in two genetic models of absence epilepsy. Br J

Pharmacol 2006, 148: 1076-1082.

[31] Bauquier SH, Lai A, Jiang JL, cook MJ. Clonic seizures induced by oral administration of

enrofloxacin in GAERS. Comparative medicine 2015, In press.

[32] McRae A, Hjorth S, Mason DW, Dillon L, Tice TR. Microencapsulated dopamine (DA)-induced

restitution of function in 6-OHDA-denervated rat striatum in vivo: comparison between two

microsphere excipients. J Neural Transplant Plast 1991, 2: 165-173.

[33] McRae A, Dahlstrom A. Transmitter-loaded polymeric microspheres induce regrowth of

dopaminergic nerve terminals in striata of rats with 6-OH-DA induced parkinsonism. Neurochem Int

1994, 25: 27-33.

[34] Gouhier C, Chalon S, Venier-Julienne MC, Bodard S, Benoit J, Besnard J, et al. Neuroprotection

of nerve growth factor-loaded microspheres on the D2 dopaminergic receptor positive-striatal

neurones in quinolinic acid-lesioned rats: a quantitative autoradiographic assessment with

iodobenzamide. Neurosci Lett 2000, 288: 71-75.

[35] Menei P, Pean JM, Nerriere-Daguin V, Jollivet C, Brachet P, Benoit JP. Intracerebral

implantation of NGF-releasing biodegradable microspheres protects striatum against excitotoxic

damage. Exp Neurol 2000, 161: 259-272.

[36] Pean JM, Menei P, Morel O, Montero-Menei CN, Benoit JP. Intraseptal implantation of NGF-

releasing microspheres promote the survival of axotomized cholinergic neurons. Biomaterials 2000,

21: 2097-2101.

[37] Arica B, Kas HS, Moghdam A, Akalan N, Hincal AA. Carbidopa/levodopa-loaded biodegradable

microspheres: in vivo evaluation on experimental Parkinsonism in rats. J Control Release 2005, 102:

689-697.

[38] Sodemann U, Moller HS, Blaabjerg M, Beier CP. Successful treatment of refractory absence

status epilepticus with lacosamide. J Neurol 2014.

[39] Krewson CE, Dause R, Mak M, Saltzman WM. Stabilization of nerve growth factor in controlled

release polymers and in tissue. J Biomater Sci Polym Ed 1996, 8: 103-117.

[40] Krewson CE, Klarman ML, Saltzman WM. Distribution of nerve growth factor following direct

delivery to brain interstitium. Brain Res 1995, 680: 196-206.

[41] Saltzman WM, Mak MW, Mahoney MJ, Duenas ET, Cleland JL. Intracranial delivery of

recombinant nerve growth factor: release kinetics and protein distribution for three delivery systems.

Pharm Res 1999, 16: 232-240.

Page 119: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 97 -

[42] Bensadoun JC, Pereira de Almeida L, Fine EG, Tseng JL, Deglon N, Aebischer P. Comparative

study of GDNF delivery systems for the CNS: polymer rods, encapsulated cells, and lentiviral vectors.

J Control Release 2003, 87: 107-115.

[43] Kellinghaus C. Lacosamide as treatment for partial epilepsy: mechanisms of action,

pharmacology, effects, and safety. Ther Clin Risk Manag 2009, 5: 757-766.

[44] Bender R, Lange S. Adjusting for multiple testing--when and how? J Clin Epidemiol 2001, 54:

343-349.

Page 120: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 98 -

Chapter 6 – Evaluation of the biocompatibility of polypyrrole

polymers implanted sub-durally in GAERS

6.1 Outline

In chapter 5 the evidence suggested that passive release AED loaded PLGA polymers can

decrease seizure activity. We will focus in chapters 6 on PPy polymers as they offer the

advantage of being reactive to electrical stimulation and could potentially serve as active

release implantable devices. Upon intermittent application of an electrical current, the ionic

and hydrophobic properties of PPy polymers vary resulting in the expansion and/or

contraction of the structure and this actuation is responsible for the release of impregnated

drug.(Hutchison, Lewis et al. 2000, Wallace and Wallace 2003) The mechanism of drug

release is the same for all polypyrrole despite the chemical composition of the polymer and

has been reported for a wide range of incorporated drugs such as dexamethasone, fos-

phenytoin and the anti-inflammatory neuropeptide α-melanocyte stimulating hormone (α-

MSH).(Zhong and Bellamkonda 2005, Moulton, Imisides et al. 2008, Muller, Yue et al.

2016)

PPy sheets have been shown in vitro to be well tolerated by biological tissues, including brain

tissues, and they seem to be as compatible as Teflon when implanted into the cerebral

cortex.(George, Lyckman et al. 2005, Ateh, Navsaria et al. 2006) Nevertheless thorough

biocompatibility and safety study in an animal model are required during the development of

any implantable medical device. The results of the study presented in chapter 6 showed that

implanted PPy mats on top of the brain cortex of GAERS did not induce obvious

inflammation, trauma, gliosis and neuronal toxicity. Therefore, the results are demonstrating

in vivo that the PPy used offered good histocompatibility with central nervous system cells

and that PPy sheets could be used as a drug delivery device implanted onto the surface of the

brain parenchyma.

6.2 Manuscript

This chapter is presented as the author-accepted version of a peer-reviewed manuscript

published in Macromolecular Bioscience, 2017 May;17(5). doi: 10.1002/mabi.201600334..

Page 121: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 99 -

The figures’ numbers and titles of the author-accepted version of the manuscript may have

been changed to follow the formatting of the thesis.

Page 122: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 100 -

Evaluation of the biocompatibility of polypyrrole implanted sub-durally in

GAERS

Sebastien H. Bauquier a*

, Karen J. McLean b , Jonathan L. Jiang

b, Ray C. Boston

b,

Alan Lai b, Zhilian Yue

c, Simon E. Moulton

‡c, Amy J. Halliday

b, Gordon

Wallace c, Mark J. Cook

b

a Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 250

Princes Hwy, Werribee, Victoria 3030, Australia

b Centre for Clinical Neurosciences and Neurological Research, St. Vincent’s

Hospital Melbourne, P.O. Box 2900, Fitzroy, Victoria 3065, Australia

c Intelligent Polymer Research Institute and ARC Centre of Excellence for

Electromaterials Science, AIIM Facility, Innovation Campus, University of

Wollongong, Wollongong, New South Wales 2522, Australia

*Corresponding author. Tel.: +61 3 9731 2311; fax: +61 3 9731 2366. E-mail address:

[email protected] (S.H. Bauquier).

‡ Current Address: Biomedical Engineering, Faculty of Science, Engineering and

Technology, Swinburne University of Technology, Hawthorn, VIC, 3122, Australia.

Page 123: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 101 -

Abstract

This blinded controlled prospective randomized study investigated the

biocompatibility of polypyrrole (PPy) polymer that will be used for intracranial

triggered release of anti-epileptic drugs (AEDs). Three by three millimetres PPy were

implanted subdurally in six adult female GAERS (Genetic Absence Epilepsy Rat from

Strasbourg). Each rat had a polymer implanted on one side of the cortex and a sham

craniotomy performed on the other side. After a period of 7 weeks, rats were euthanized and

parallel series of coronal sections were cut throughout the implant site. Four series of fifteen

sections were histological (H&E) and immunohistochemically (NeuN, GFAP and ED1)

stained and evaluated by 3 investigators. The results showed that implanted PPy mats did not

induce obvious inflammation, trauma, gliosis and neuronal toxicity. Therefore we conclude

the PPy used offer good histocompatibility with central nervous system cells and that PPy

sheets could be used as intracranial, AED delivery implant.

A brain slice from a rat with a confirmed cortical abscess

Page 124: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 102 -

1. Introduction

The incidence of epilepsy ranges from 50 and 100 cases per 100,000 population per

year in most developed countries though it is estimated that up to 5% of a population

will experience non-febrile seizures at some point in life.[1-3]

Because treatment with

conventional anti-epileptic drugs (AEDs) provides adequate seizure control in only two

third of patients, more novel therapy is urgently needed.[1-3]

Alternative therapies aiming at improving the availability of AEDs such as the

intracranial implantation of polymer-based drug delivery systems are being

investigated.[4, 5]

This targeted drug delivery approach has shown some successes in the

treatment of animal models of several neurological disorders such as Parkinson’s

disease, Huntington’s disease and Alzheimer’s disease.[6]

This approach has been

published once for the treatment of epilepsy.[4]

The results of this study indicated that

drug-eluting poly(D,L-lactide-co-glycolide) (PLGA) polymer implants represents a

promising evolving treatment option for intractable epilepsy. The PLGA polymers used

is a biodegradable copolymer that is very well tolerated by the brain.[7]

However this

polymer can only be used as a passive release vehicle.

Work by Cook et al, 2013 has shown that prediction of seizure likelihood with a long-

term, implanted seizure advisory system in patients with drug-resistant epilepsy is

becoming reality.[8]

Synchronizing the AED release from a polymer with the seizure

advisory system could be technically feasible when using a conductive polymer such as

polypyrrole (PPy) that can change physical properties when being applied electrical

stimulation. The physical changes associated with the mechanism for release is

proposed to involve electrostatic interactions as the application of electrical stimulation

protocol increases the amount of drug release. However, both the ionic and hydrophobic

Page 125: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 103 -

properties of polypyrrole have been shown to vary upon application of electrical

stimulus.[9]

The expansion and contraction (actuation) of polypyrrole is also a well-

documented property of polypyrrole upon oxidation and reduction and these processes

may be involved in the release of incorporated drug.[10, 11]

As these properties change

simultaneously it is not possible to separate them to determine which process is the

driving force for release. These mechanisms of drug release are the same for all

polypyrrole despite the chemical composition of the polymer and has been reported for

a wide range of incorporated drugs such as dexamethasone, fos-phenytoin ] and the anti-

inflammatory neuropeptide α-melanocyte stimulating hormone (α-MSH).[12-14]

PPy sheets have been shown to be well tolerated by the brain tissue in vitro and they

seem to be as compatible as Teflon when implanted into the cerebral cortex.[15]

The aim

of the present study is to test the in vivo biocompatibility of PPys implanted sub-durally

in GAERS (Genetic Absent Epilepsy Rat from Strasbourg) on top of their cerebral

cortex.

2. Experimental Section

The study was designed as blinded controlled prospective randomized experiments.

To evaluate the in vivo biocompatibility of the PPy sheets, the rats underwent surgery

for bilateral craniotomy; a PPy sheet was implanted subdurally on one side of the cortex

and the other side was used as a sham control. After a period of 7 weeks, the rats were

euthanized and parallel series of coronal sections were cut throughout the implant site.

Hematoxylin and Eosin (H&E) staining was used to evaluate inflammation, and general

histological and architectural changes.[16]

Immunohistochemical (IHC) staining for glial

fibrillary acidic protein (GFAP) was used to evaluate possible materials glial reaction

Page 126: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 104 -

(gliosis), whilst the ED1 (CD68) antibody and the neural marker NeuN antibody were

used to evaluate rat macrophages and neuronal toxicity respectively.[16-18]

2.1. Evaluated materials

Pyrrole (Py) was obtained from Merck (Frenchs Forest, NSW, Australia) and distilled

prior to use. The electrolytes used for electropolymerisation were 2 mg/mL dextran

sulphate (DS, MW 9000-20,000, Sigma–Aldrich, Castle Hill NSW, Australia). The PPy

was synthesized from a monomer solution containing 0.2 M pyrrole and the

aforementioned electrolyte. The synthesis method employed was galvanic

polymerisation using a current density of 0.2 mA/cm2 applied for 180 min using a 3

electrode cell containing an auxiliary electrode (stainless steel mesh – 5cm 5cm),

reference electrode (RE: Ag/AgCl - 3.0M NaCl) and a working electrode (WE: polished

stainless steel plate 5cm 5cm). After polymerisation, the WE was rinsed with Milli-Q

water overnight to remove any unreacted monomer and dopant. The PPy film standing

film was obtained by carefully cutting around the edges of the deposited PPy film and

gently removing it from the WE. The free standing film was then laid down on a

KimWipeTM tissue sheet that was pre-wet with Milli-Q water. The films were left on

the tissue paper and placed into a petri dish, covered and left at 2oC until required for

implantation.

The morphology of the as-prepared PPy film was examined using a Field Emission

Scanning Electron Microscope (FESEM, JEOL JSM-7500FA). The thickness of the

sample was determined by analyzing the cross sectional micrographs of the sample (n =

20). The mean static contact angle of the polypyrrole free standing film was measured

using a Dataphysics optical contact angle goniometer. A total of 20 measurements were

Page 127: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 105 -

made on the sample using deionized water droplets of 1 µl for the reported value.

2.2. Animals

Six 2-month-old female GAERS were obtained from the University of Melbourne

(Parkville, Victoria, Australia) and were housed individually in 12 hours light/dark

cycles with ad libitum access to food and water. All experiments were approved by St

Vincent’s Hospital (Melbourne) Animal Ethics Committee and conducted in accordance

with the Australian Code of Practice for the Care and Use of Animals for Scientific

Purposes (2004).

2.3. Animal surgery

Immediately prior to surgery, rats were weighed and anaesthetized with an intra-

peritoneal injection of ketamine (ketamine hydrocloride 100 mg/mL, Ceva Animal

Health, Glenorie NSW, Australia) (75 mg/kg) and xylazine (Ilium xylazine

hydrochloride 20 mg/mL, Troy Laboratories Pty Ltd, Glendenning NSW, Australia)

(10 mg/kg). Following anaesthesia induction, rats were placed in a stereotaxic

apparatus, and administered isoflurane (Isoflo™, Abbott Australasia Pty Ltd, Botany

NSW, Australia) (0.5 to 1% in oxygen, 1 L/min) via a nose-cone, and were

administered subcutaneous (SQ) carprofen (Tergive carprofen 50 mg/mL, Parnell

Australia Pty Ltd, Alexandria NSW, Australia) (5 mg/kg) for pain relief and 0.9%

sodium chloride (Baxter, Old Toongabble NSW, Australia) (2 mL) for cardiovascular

support. Over the scalp of the rats, the hair was clipped and the skin aseptically

prepared. A single incision was made down the midline, the skull cleared of tissue and

the exposed bone dried with 3% hydrogen peroxide (Sanofi, Virginia QLD, Australia).

Page 128: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 106 -

Implants measuring 3 mm by 3 mm were cut from the polymer sheets described above.

Placements of the implants were performed after 5 mm by 4 mm craniotomies were

created bilaterally at the level of the coronal suture (over the motor cortices) and after

excising the dura to expose the brain surface. Only one of the two craniotomy sites was

used to implant the polymer. The site of the polymer implantation was randomly

allocated using the random function in Microsoft Excel 2007. Following implant

placements, although neither the dural flap nor the bone pieces were replaced, an

approximately 20 mm by 10 mm silicone sheet was placed on top of the skull to cover

both craniotomy sites and the scalp was sutured closed (3/0 Monosyn® B Braun, Rubi,

Spain). Silicone sheets were prepared using implant grade liquid silicone rubber

(Cochlear Ltd), LSR 30, which comprises a polymer precursor base and a crosslinker.

The polymer precursor base and crosslinker were dissolved in hexane at a ratio of 10:1,

cast in a Teflon mould at room temperature, and cured by crosslinking at 110 oC for 30

min and then at 200 oC for 4 h. The silicone sheets were finally extracted with hexane

for 72 h to remove any residual oligomers, and dried in vacuo at 60 oC overnight.

The animals were placed on heat pads for recovery. Post-operative treatments included

SQ buprenorphine (Temgesic® 0.324 mg/mL buprenorphine hydrochloride, Reckitt

Benckiser Healthcare Ltd, Hull, UK) every twelve hours (0.03mg/kg, twice a day),

saline (2mL, once a day) and carprofen (5mg/kg, once a day) for up to 3 days.

2.4. Animal post-operative health monitoring

For a minimum of three days post-surgery and until full recovery from the surgery, the

rats were monitored for weight loss, mobility and grooming twice a day (Table 6.1). At

each assessment, a debilitation score was calculated by cumulating weight loss score,

Page 129: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 107 -

mobility scores and grooming score. A debilitation score of 0, 1 to 3, 4 to 6 and 7 to 9

meant that the rat health was not affected, mildly affected, moderately affected, severely

affected by the surgery/polymer implantation respectively.

Table 6. 1 Post-operative health monitoring chart.

Post-operative health monitoring chart

Scores

Percentage of weight loss

No weight loss

0

Less than 10% weight loss

1

Weight loss between 10 and 20%

2

Weight loss of more than 20%

3

Mobility Score

Normal mobility

0

Rat ataxic moving at a normal

speed

1

Rat ataxic and moving slowly

2

Recumbent rat

3

Grooming Score

Normal grooming activity

0

Grooming activity mildly

decreased

1

Grooming activity significantly

decreased

2

No grooming activity

3

Debilitation score

0-9

For a minimum of three days post-surgery and until full recovery from the surgery, the rats

were monitored for weight loss, mobility and grooming twice a day. A debilitation score of 0, 1

to 3, 4 to 6 and 7 to 9 meant that the rat health was not affected, mildly affected, moderately

affected, severely affected by the surgery, respectively.

2.5. Sacrifice and sample collection

Seven weeks after surgery, rats were weighed, anesthetized with isoflurane in oxygen

and euthanized with pentobarbital (Lethabarb, pentobarbitone sodium 325 mg/mL,

Virbac Pty Ltd, Milperra NSW, Australia) (100 mg/kg, intraperitoneal). The brain was

immediately carefully dissected and frozen in liquid nitrogen-cooled isopentane and

Page 130: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 108 -

stored at - 80°C. At a later stage, brains were sectioned using a Leica CM 1850 cryostat.

Parallel series of coronal sections were cut at 10 µm throughout the implant site,

mounted onto chrome-gel-alum subbed microscope slides, air-dried then stored at -80°C

until further processing.

2.6. Histological and immunohistochemical analysis

For each of the histological or IHC staining, the sections from all brains were processed

simultaneously to ensure similarity in the experimental conditions.

2.6.1. H&E staining

H&E staining was performed similarly to previously published protocol.[16]

In brief,

slices were removed from the -80°C freezer and leave to air dry for 10 to 30 min.

They were then placed into room temperature (RT) 10% Neutral Buffered Formalin

(NBF, Sigma–Aldrich, Castle Hill NSW, Australia) for 15 minutes, rinsed with tap

water (3*1 min), dipped in Hematoxylin (Gills 3, Sigma–Aldrich, Castle Hill NSW,

Australia) for 4 minutes, rinsed with tap water for 5 minutes, differentiated by

dipping in ethanol 70% (containing 0.5% HCl) for 20 seconds, rinsed in tap water

(1min) and allowed bluing for 15s (0.1% ammonium hydroxide solution in distilled

water). After another rinse in tap water (3 min), the slides were dipped into ethanol

(50 and 70 %, 1 min each), stained in Eosin 0.6% (Alcoholic, Fisher Scientific,

Pittsburgh, PA) for 30 seconds and dipped in ethanol (95% and 100 % for 3 minutes

and 2 *1minute respectively). Finally the slices were cleared in Xylene (Ajax

Finechem, Taren Point NSW, Australia) for 2* 5 minutes, mounted in DPX (Sigma–

Aldrich, Castle Hill NSW, Australia), and coverslipped.

Page 131: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 109 -

2.6.2. NeuN/GFAP-IHC staining

NeuN/GFAP staining was performed similarly to previously published protocol.[7]

In

brief, immediately after taking the slices out of the freezer, they were placed into room

temperature (RT) 10% NBF for 15 minutes. Sections were then washed in 0.1M

phosphate-buffered saline (PBS, pH=7.4) (3 x 5 min) on a rocking platform and

incubated in 10% normal goat serum (NGS) for 1 h RT in a humidified chamber. The

slices were washed again in PBS (3 x 5 min) on a rocking platform, before overnight

incubations in primary antibodies for neuron-specific nuclear protein (NeuN) and glial

fibrillary acidic protein (GFAP) NeuN/GFAP (1/500 and 1:1000 respectively; NeuN -

mouse monoclonal MAB377 and GFAP – rabbit polyclonal AB5804; both from

Millipore Merck Millipore, Bayswater VIC, Australia) at 4oC in a humid chamber. The

following day slides were washed in PBS (3 x 5 min) on a rocking platform, incubated

in two secondary antibodies (each at 1/1500; goat anti-mouse IgG (H+L) secondary

Antibody, Alexa Fluor® 594 conjugate and goat anti-rabbit IgG (H+L) secondary

antibody, Alexa Fluor® 488 conjugate; both from Life technologies, Scoresby VIC,

Australia) for 1 h RT in a dark humid chamber, and washed again in PBS (3 x 5 min) on

a rocking platform. The slices were then incubated in 4',6-diamidino-2-phenylindole

(DAPI, Sigma–Aldrich, Castle Hill NSW, Australia ; 1/5000) for 5 min in the dark on a

rocking platform and washed again in PBS (3 x 5min) on a rocking platform. Finally the

slices were coverslipped with an aqueous mounting medium (Fluoromont™

, Sigma–

Aldrich, Castle Hill NSW, Australia) and stored in the dark.

Page 132: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 110 -

2.6.3. ED1-IHC staining

The ED1 staining was performed following the protocol than NeuN/GFAP staining

except for the following points: The overnight incubation was performed in primary

antibody for anti-CD68 (ED1) (1/1000; mouse monoclonal from Serotec, Bio-Rad

Laboratories Pty, Gladesville NSW, Australia) at 4oC in a humid chamber and the

secondary antibody used was GαM Alexa Fluor 594 (1/1500). A brain slice from a rat

with a confirmed cortical abscess was also ED1 stained and used as a positive control.

2.6.4. Histological and IHC evaluations

Images were obtained using a Dot Slide Olympus system using an Olympus BX51

microscope (Olympus Corporation, Tokyo, Japan) and captured using Virtual Slide

System Software (VS-ASW-S1 FL 2.5, Olympus Soft Imaging Solution GmbH,

Hamburg, Germany).

Both sides (polymer implantation side and sham surgery side) of 15 cortexes’ parallel

coronal sections cut throughout the surgery site were evaluated independently by three

investigators for each histological and immunohistochemical staining. At the time of the

evaluations, the investigators were unaware of the side of the polymer implantation.

H&E stained sections were evaluated to assess macroscopic consequences of polymer

implantation. The degree of damage was rated following a published scale: minor =

incomplete disruption of the meninges and disruption of molecular cortical layer;

moderate = complete disruption of the meninges and molecular cortical layer; severe =

damage extending beyond the meninges and molecular cortical layer.[7]

Page 133: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 111 -

Immunohistochemically stained sections were subjectively rated from 0 to 3 (0 = no

change in immunoreactivity, 1 = subtle changes in immunoreactivity, 2 = clear changes

in immunoreactivity but only affecting a very limited part of the cortex, 3 = clear

changes in immunoreactivity affecting an extended area of the cortex).

2.7. Statistical methods

Differences in immunoreactivity between the two sides of the cortexes were evaluated

using a multilevel mixed-effects ordered logistic regression using commercially

available statistical software (Stata 11.0; StataCorp, College Station, TX). Graphics

were made using commercially available software (Prism 6 for Windows, GraphPad

Inc., San Diego CA, USA). P-Value of 0.05 was used to distinguish statistically

significant differences. Unless specified, the results are reported as median and range.

3. Results and discussion

The thickness of the PPy mats was determined to be 3.1 ± 0.3 µm (Figure 6.1) with a

contact angle of 67.8 ± 3.9 deg (n = 20) measured for the free standing film.

Page 134: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 112 -

Figure 6. 1 Scanning Electron Microscope (SEM) images of the polypyrrole thick film

doped with dextran sulfate (top surface A and B, cross-section C).

The median of the post-operative health monitoring scores was 0.5 (0 to 4) with only

A

B

C

Page 135: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 113 -

one rat receiving a score above 2 (i.e. score of 4 at the first evaluation of day 2 post-

operatively). The median pre-surgical and pre-euthanasia weight of the 6 rats were 152

g (136 to 156) and 186.5 g (168 to 207) respectively representing a weight gain over

the 7 weeks implantation period of 24 % (21 to 33). Samples of parallel series of

coronal sections representative of the changes seen after H&E, NeuN, GFAP and ED1-

IHC staining are presented in Figure 6.2.

3.1. Histological changes in H&E staining:

The three evaluators were in agreement for the following results: there was no

appreciable differences between the sham and implanted sides of the cortex, and besides

the fact that part of the stratum 1 was missing on some of the sections from both sides

of the cortex (and not limited to the surgical sites), no additional changes occurred that

could have been attributed to the implantation of the polymers (i.e. no evidence of

necrosis, infection or inflammation).

3.2. Neuronal toxicity

The median NeuN-IHC scores were 0 (0 to 3) (average [± standard error] 0.60 ± 0.05)

and 0 (0 to 2) (average score [± standard error] 0.52 ± 0.05) for the sham and implanted

polymer sides of the cortexes respectively (Figure 6.3). Overall, there were subtle

changes in neuronal density in the boundary layer and the surrounding brain tissues in

the two sides of the cortexes and with the sham surgery sides being more affected

(p=0.005).

Page 136: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 114 -

Figure 6. 2 Samples of parallel series of coronal sections cut throughout rats’ cortex

Samples of parallel series of coronal sections that were cut throughout the cortex of a rat with

PPy sheets implanted subdurally for a period of 7 weeks. The rat had a polymer implanted on

one side of the cortex (A, B, C, D) and a sham craniotomy performed on the other side (A’, B’,

C’, D’). Those samples are representative of the changes seen after H&E (A, A’), NeuN (B, B’),

GFAP (C, C’) and ED1 (red)/DAPI (blue) (D, D’) immunohistochemistry (IHC) staining. A

brain slice from a rat with a confirmed cortical abscess was also ED1 (red)/DAPI (blue) stained

(D”) and used as a positive control for ED1 staining. The stratum one is missing from the sham

surgery side of the cortex and changes in IHC staining were minor on both sides of the cortex.

Page 137: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 115 -

Figure 6. 3 Summary of the immunohistochemical scores (NeuN, GFAP and ED1)

Average (and standard error) scores attributed to cortical sections immunohistochemically

(ICHIHC) stained with NeuN, GFAP and ED1. PPy mats were implanted subdurally in six adult

female GAERS rats. Each rat had a polymer implanted on one side of the cortex and a sham

craniotomy performed on the other side. After a period of 7 weeks the rats were euthanized and

parallel series of coronal sections were cut throughout the implant site. Fifteen sections per rat

for each immunohistochemical stain (NeuN, GFAP and ED1), were subjectively rated from 0 to

3 (0 = no change in immunoreactivity, 1 = subtle changes in immunoreactivity, 2 = clear

changes in immunoreactivity but only affecting a very limited part of the cortex, 3 = clear

changes in immunoreactivity affecting an extended area of the cortex) by 3 investigators.

Differences in immunoreactivity between the two sides of the cortex were evaluated using

multilevel mixed-effects ordered logistic regression. *: p=0.005; **: p=0.002.

Page 138: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 116 -

3.3. Glial reaction

The median GFAP-IHC scores were 0 (0 to 2) (average [± standard error] 0.61 ± 0.04)

and 0 (0 to 2) (average score [± standard error] 0.44 ± 0.04) for the sham and implanted

polymer sides of the cortexes respectively (Figure 6.3). Overall, the increases in GFAP-

IHC staining observed were subtle and affected significantly more the sham surgery

sides of the cortexes (p=0.002).

3.4. Presence of macrophages

The median ED1-IHC scores were 0 (0 to 3) (average [± standard error] 0.45 ± 0.05)

and 0 (0 to 2) (average score [± standard error] 0.40 ± 0.04) for the sham and implanted

polymer sides of the cortexes respectively (Figure 6.3). Overall, although ED1-IHC

showed significant amounts of macrophages in the positive control brain tissue, only

very few macrophages were present in rats implanted with PPy sheets. No difference

was observed between the 2 sides of the cortexes (p=0.248).

PPys were implanted subdurally for a period of 7 weeks in adult female GAERS and

using the histological and immunohistological methods (i.e. H&E staining, and NeuN,

GFAP and ED1-IHC staining) described above we demonstrated in vivo

histobiocompatibility of the PPy with central nervous system (CNS) cells. These results

confirmed previously published in vitro work.[15]

All rats recovered well and rapidly following the surgery. They all gain significant

weight during the 7 weeks of the implantation period showing very little impact of the

PPys on the growth and vital functions of GAERS.

Page 139: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 117 -

To be declared histobiocompatible, an implant must: 1) not be toxic to surrounding

tissues and 2) not be recognized as foreign material by the immune system.[7]

In the

presence of acute toxicity, neurons will appear karyopyknotic and in the presence of

chronic toxicity, macrophages/monocytes and astrocytes will be activated by necrotic

cellular debris resulting from neurons necrosis.[19]

Furthermore, as fibroblasts are

absent from within the CNS, the scar tissue will originate from the astrocytes forming

gliotic scarring.[7]

This scarring will also be seen if the implant is recognized as a

foreign material. In this case, astrocytes will undergo gliosis and microglia and

monocytes/macrophages will initiate a local inflammatory reaction resulting in the

formation of multinucleate giant cells (i.e. fusions of multiple macrophages).[7, 20]

The Stratum 1 (or microgyric layer) of the cortex was missing from some of the

sections. This variability in brain implant outline was attributable to mechanical

procedures during sample preparation. Also, no difference between the sheets

implantation side or the sham surgery side of the cortex were seen, the loss of the

stratum 1 represents a limitation of the study. Loss of portions of the boundary layer

adhering to the implant during its removal from the brain for histological procedures

prevents precise examination of interface tissues. This problem could be avoided by

keeping the implant in place during all stages of tissue preparation.[16, 21]

Nevertheless,

on the slices where the stratum 1 is present no major changes were observed (e.g.

Figure 6.2 A, B, C, D).

The IHC scores observed on the PPy implanted side of the cortexes strongly favor the

histobiocompatibility of the PPy. The median IHC scores (0 for all three IHC staining)

Page 140: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 118 -

shows that there were no evidence of neuronal degeneration, glial reaction or of the

presence of macrophages in the majority of the slices rated. However, a previous study

reported middle to moderate acute inflammatory reactions after the subcutaneous

implantation of PPy in rats followed by a resolution of the inflammatory response.[22]

Our study did not analyze the brain tissue immediately after implantation and may have

missed the opportunity to observe an acute inflammatory reaction.

The low average IHC scores (0.40 to 0.52) observed demonstrated that when some IHC

staining changes occurred, they were of minimal extent and of lower or similar

amplitude than the one observed on the sham surgery sides of the cortexes. The

statistically significant differences in IHC scores observed between the PPy

implantation sites demonstrate that it was not the lack of statistical power that allowed

the conclusion that the PPy does not induce more IHC staining changes compared to a

sham surgery. The subjectivity of scale used for grading IHC related changes was taken

into consideration during the planning of the study and it was decided that at least 3

evaluators were to grade every brain section. For each of the 6 rats and for each of the 3

IHC staining, 3 series of 15 sections were graded twice (PPy and sham surgery sides) by

3 investigators making a total 1620 IHC related data points that allowed establishment

of statistical significances. The absence of a control group (rat not undergoing

craniotomy) prevented comparison with normal brain tissue however all evaluator were

trained and experimented in IHC evaluation allowing them to have reasonable

assumption on what a normal brain tissue should look like.

Any surgery will induce tissue trauma resulting in cell death and tissue scarring. The

comparison of NeuN-IHC and GFAP-IHC scores reveals that the amount of neuronal

death and gliosis was significantly less in the PPy side than the sham surgery side of the

Page 141: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 119 -

cortexes (p values of 0.005 and 0.002 respectively) implying that the application of PPy

could protect the CNS tissue after surgery. In the present experiment, neither the dura

nor the skull flaps were replaced. In a pilot study (unpublished data) we found that

during the craniotomy the careful dissection of the dura to maintain a flap intact was

performed at the detriment of the brain tissue situated just below. In consequence the

dura was peeled away and not replaced. The brain tissue also presented some edema

resulting in an expansion of the cortex partially protruding through the craniotomy and

in consequence the skull window was not replaced to avoid more mechanical damages.

Although the PPy may have just been an additional layer of protection, the potential

neuro-protective effects of PPys need to be studied further. Several publications have

demonstrated the benefice of the use of PPy to allow tissue proliferation/regeneration.

Indeed PPy is an advantageous substrate to enhance nerve cell interaction with potential

application as nerve guidance channels to bridge the gap between severed nerve ends.[23]

Furthermore, using an ischemia-reperfusion rat myocardial infarction model researchers

demonstrated that PPy enhanced infiltration of myofibroblasts into the infarct area

highlighting the potential clinical benefit of using this PPy as an injectable scaffold to

repair ischemic myocardium after myocardial infarction.[24]

The softness of the polymer has probably also contributed to the apparent reduction in

local reactive response but the absence of animals implanted with a relatively stiff

implant as a negative control prevents firm conclusion. The PPy’s friable nature

prevented appropriate retrieval of the implant for adequate explant analysis. Also, very

little fibrin was visually detected, on a macroscopic scale, around the implant during

brain dissection. The absence of explant analysis prevents to exclude the possibility of a

formation of a fibrin capsule which could prevent the polymer to release drug into the

Page 142: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 120 -

central nervous system.

Due to the absence of evidence of toxic injury or immune mediated inflammation, the

authors conclude that PPys offer good histocompatibility with CNS cells and that PPy

sheets could potentially be used as intracranial AED delivery implant.

Acknowledgements

The authors gratefully acknowledge the sponsorship of the Victorian

Government through its Science Technology and Innovation Initiative

administered by the Department of Industry, Innovation and Regional

Development. The authors wish to acknowledge funding from the Australian

Research Council (ARC) Centre of Excellence Scheme (CE140100012), the use

of facilities at the University of Wollongong Electron Microscopy Centre,

support of the Australian National Fabrication Facility (ANFF) – Materials

Node. Professor Gordon Wallace acknowledges the support of the ARC through

an ARC Laureate Fellowship (FL110100196).

Keywords: cerebral cortex implants, conductive polymers, toxicity, biocompatibility

Page 143: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 121 -

References

[1] S. H. Bauquier, A. Lai, J. L. Jiang, M. J. cook, Neuroscience Bulletin 2015.

[2] J. W. Sander, Current opinion in neurology 2003, 16, 165.

[3] S. D. Shorvon, Epilepsia 1996, 37 Suppl 2, S1.

[4] A. J. Halliday, T. E. Campbell, T. S. Nelson, K. J. McLean, G. G. Wallace, M. J. Cook, J Clin

Neurosci 2013, 20, 148.

[5] A. J. Halliday, S. E. Moulton, G. G. Wallace, M. J. Cook, Adv Drug Deliv Rev 2012, 64, 953.

[6] A. J. Halliday, M. J. Cook, CNS Neurol Disord Drug Targets 2009, 8, 205.

[7] A. J. Halliday, T. E. Campbell, J. M. Razal, K. J. McLean, T. S. Nelson, M. J. Cook, G. G.

Wallace, J Biomed Mater Res A 2011.

[8] M. J. Cook, T. J. O'Brien, S. F. Berkovic, M. Murphy, A. Morokoff, G. Fabinyi, W. D'Souza, R.

Yerra, J. Archer, L. Litewka, S. Hosking, P. Lightfoot, V. Ruedebusch, W. D. Sheffield, D. Snyder,

K. Leyde, D. Himes, Lancet neurology 2013, 12, 563.

[9] G. G. Wallace, G. G. Wallace, "Conductive electroactive polymers : intelligent materials

systems", 2nd edition, CRC Press, Boca Raton, FL, 2003, p. 237 p.

[10] A. S. Hutchison, T. W. Lewis, S. E. Moulton, G. M. Spinks, G. G. Wallace, Synthetic Met

2000, 113, 121.

[11] T. W. Lewis, S. E. Moulton, G. M. Spinks, G. G. Wallace, Synthetic Met 1997, 85, 1419.

[12] S. E. Moulton, M. D. Imisides, R. L. Shepherd, G. G. Wallace, J Mater Chem 2008, 18, 3608.

[13] Y. Zhong, R. V. Bellamkonda, J Control Release 2005, 106, 309.

[14] R. Muller, Z. Yue, S. Ahmadi, W. Ng, W. M. Grosse, M. J. Cook, G. G. Wallace, S. E.

Moulton, Sensors and Actuators B: Chemical 2016, 236, 732.

[15] P. M. George, A. W. Lyckman, D. A. LaVan, A. Hegde, Y. Leung, R. Avasare, C. Testa, P. M.

Alexander, R. Langer, M. Sur, Biomaterials 2005, 26, 3511.

[16] J. Yang, A. C. Charif, J. E. Puskas, H. Phillips, K. J. Shanahan, J. Garsed, A. Fleischman, K.

Goldman, M. T. Luebbers, S. M. Dombrowski, M. G. Luciano, Journal of the mechanical

behavior of biomedical materials 2015, 45, 83.

[17] L. F. Eng, Journal of neuroimmunology 1985, 8, 203.

[18] V. S. Polikov, P. A. Tresco, W. M. Reichert, Journal of neuroscience methods 2005, 148, 1.

[19] V. Kumar, A. K. Abbas, J. C. Aster, "Robbins and Cotran pathologic basis of disease", Ninth

edition. edition, Elsevier/Saunders, Philadelphia, PA, 2015, p. xvi.

[20] E. Fournier, C. Passirani, C. N. Montero-Menei, J. P. Benoit, Biomaterials 2003, 24, 3311.

[21] S. S. Stensaas, L. J. Stensaas, Acta neuropathologica 1978, 41, 145.

Page 144: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 122 -

[22] Z. Wang, C. Roberge, L. H. Dao, Y. Wan, G. Shi, M. Rouabhia, R. Guidoin, Z. Zhang, J Biomed

Mater Res A 2004, 70, 28.

[23] C. E. Schmidt, V. R. Shastri, J. P. Vacanti, R. Langer, Proceedings of the National Academy

of Sciences of the United States of America 1997, 94, 8948.

[24] S. S. Mihardja, R. E. Sievers, R. J. Lee, Biomaterials 2008, 29, 4205.

Page 145: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 123 -

Alternative therapies aiming at improving the availability of anti-epileptic drugs (AEDs)

such as the intracranial implantation of polymer-based drug delivery systems have shown

some promising results. This blinded controlled prospective randomized study investigated the

biocompatibility of polypyrrole (PPy) polymer that will be used for intracranial triggered

release AED delivery. We conclude the PPy used offer good histocompatibility with cortical

cells.

Sebastien H. Bauquier*, Karen J. McLean, Jonathan L. Jiang, Ray C. Boston, Alan Lai,

Zhilian Yue, Simon E. Moulton, Amy J. Halliday, Gordon Wallace, Mark J. Cook

Corresponding Author*

A brain slice from a rat with a confirmed cortical abscess

Page 146: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 124 -

Chapter 7 – Conclusion

The aim of the thesis was to report the initial development of polymer based

biodegradable implantable drug delivery devices loaded with commonly used AEDs,

and test these in an animal model of absence epilepsy. After validating an automated

spike and wave complex detection algorithm applied to GAERS EEGs and

demonstrating some effects of enrofloxacin on the GAERS epileptic activity, in vitro

characterisation and in vivo testing of the antiepileptic efficacy of PLGA polymer

loaded with either phenytoin or lacosamide were performed. Tables 1.2 and 1.3 of

Chapter 1 were updated to take into account the results presented in Chapters 4 and 5

(Appendix 1).

7.1 Development of the PLGA Polymers

Phenytoin loaded PLGA polymer provided constant delivery of phenytoin for the entire

experimentation period of 20 days. This is the first report of the combination of PLGA

and phenytoin, and the release period observed exceeded that of most other

combinations studied (Appendix 1 Table 1.2).When using EVA polymer loaded with

phenytoin, Tamargo et al. (2002) achieved a release period of 105 days corresponding

to the experimental period.(Tamargo, Rossell et al. 2002) Additional measurements

performed after the polymers were implanted for 365 days allowed establishment of a

calculated release period of 3.5 years. However, that prolonged release period was only

associated with an in vivo demonstrated reduction in seizure activity over a period of 6

days in a cobalt-induced rat model of epilepsy.(Tamargo, Rossell et al. 2002) In that

experiment, the phenytoin loaded EVA polymers were implanted precisely where the

cobalt was applied to the cortex.

In Chapter 4, the researchers haven’t been able to demonstrate that phenytoin loaded

PLGA polymer sheets implanted on the surface of the cortex of GAERS could decrease

seizure activity in GAERS for a sustained period. The phenytoin was chosen for its

lipophilic properties and its proven efficacy in GAERS when applied directly over S1po

and S1FL. (Gurbanova, Aker et al. 2006) The poor water solubility allowed easy drug

loading within the polymer and prolong release of the AED. The minimal amount of

Page 147: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 125 -

phenytoin released in the in vitro experiment may have been a contributing factor to the

negative results. The phenytoin mats were placed over the motor cortex and the delivery

of phenytoin even only 2 mm cranially to S1po and S1Fl could have also contributed to

the absence of changes in duration and numbers of SWDs. Throughout the different

experiment

The in vitro release results for the lacosamide loaded PLGA mats presented in Chapter

5 showed a prolonged release of lacosamide over the entire experimentation period of

100 days with more than 90% of the drug being released within 60 days (i.e. PLGA-

12.5% lacosamide). By comparison, Chen et al. (2017) only reported a release period of

up to 250 hours depending on the nature of the lacosamide loaded PLGA device

used.(Chen, Gu et al. 2017) In that experiment, three PLGA/lacosamide (w/w, 10/1)

solutions prepared with the PLGA concentration varying from 1.5 wt%, 4.5 wt%, to

14.0 wt% were electrospun and resulted in the formation of microspheroids,

microspheres, and microfibers respectively. The microfibers demonstrate the least initial

burst release while the spheroids exhibit the most rapid release characteristics. The

polymer used in Chapter 5 was made using a coaxial electrospinning method meaning

that the core of the polymer was composed of 75:25 PLGA (lactide/glycolide = 75:25),

and had a very similar structure to the one used in the Chen et al. (2017) study; and a

shell composed of 85:15 PLGA (lactide/glycolide = 85:15) which likely allowed a more

prolonged release of the lacosamide. Nevertheless, the Chen et al. (2017) study

confirms the author’s choice to use PLGA fibres rather than microspheres for long term

constant AED delivery. To the best of the author’s knowledge, no other polymer

containing lacosamide has been described in the peer-reviewed scientific literature.

The lacosamide loaded polymer implanted subdurally on the surface of the motor cortex

of GAERS in Chapter 5 affected seizure activity in GAERS by decreasing the mean

duration of the SWDs events for a sustained period of up to 7 weeks. The evidence

suggests that intracranial implantation of polymer-based drug delivery systems could be

used successfully in the treatment of epilepsy. This decrease in seizure duration was

however associated with significant side effects during the post-operative period. Just

before implantation, the PLGA sheets were cut with scissors, potentially leaving the

Page 148: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 126 -

core of the fibres of the implants open to leakage. It was speculated that the high

debilitation scores encountered could have been the result of a significant initial leakage

of lacosamide from the polymer. To test that theory another experiment was designed

using laser cut lacosamide loaded PLGA mats. The hypothesis of that experiment was

that by laser cutting the polymer and consequently sealing the edges of the polymer

mats, the more porous core of the polymer would not be able to release as much

lacosamide immediately after polymer implantation. Because no in vitro study was

performed to characterise those implants, the experiment was not included as a chapter

in the thesis but is presented in Appendix 2. The experimental protocol was similar to

the one described in Chapter 5. The number of seizures, the mean duration of a seizure

and the total duration of seizures were not significantly different except during week

one when they were all significantly lower in the lacosamide polymer group compared

to the control group. None of the rats from the lacosamide polymer group were

moderately or severely affected by the surgery. As it has been demonstrated in Chapter

4, the craniotomy itself can be responsible for a decrease in seizure activity for up to

three weeks following the surgery. In consequence there was no evidence of any

beneficial or negative effects of the lacosamide and it can be assumed that by laser

cutting the polymer and consequently sealing the edges of the polymer mats, the more

porous core of the polymer likely did not release as much lacosamide immediately after

polymer implantation. The speculated reason for the high debilitation scores for the

lacosamide polymer group observed in Chapter 5 is also plausible although additional

experiments would need to be performed to confirm this.

The experiments presented in Chapters 4 and 5 took the common approach of testing a

drug and reporting the effects. Retrospectively, an alternative approach could have been

to first determine the minimum lacosamide intravenous dose that produces the desired

effect in 90% of the GAERS, and to determine the corresponding lacosamide

concentrations achieved in the different parts of the brain as a reference. Once these are

established, lacosamide loaded polymers could be implanted subdurally, and brain

tissue concentrations of lacosamide measured at different time points and compared to

the theoretical target. Based on these comparisons, the structure of the composition of

the polymer sheets could have been optimised and the antiepileptic effects tested. In

Page 149: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 127 -

other words, one major limitation of the studies presented in Chapters 4 and 5 is the

unknown pharmacokinetics and pharmacodynamics of lacosamide within the central

nervous system when administered using polymer devices implanted subdurally.

This alternative approach would not have been suitable for the phenytoin polymers, as

phenytoin has no anti-epileptic effect when administered systemically in GAERS. Also,

as mentioned in the introduction, little is known about the mechanism of action of

AEDs and we do not know with certainty which part of the brain should be targeted.

Consequently, we could potentially have measured lacosamide tissue concentrations in

areas of the brain that have no significance in relation to the anti-epileptic effects of the

drug.

The implantation of the AED loaded polymers comes with significant medical risk such

as meningitis, stroke, brain tissue damage and haemorrhage, and shouldn’t be repeated

frequently. In practice, the patient would probably receive a generic polymer delivering

a certain amount of drug that may or may not be adequate. The AED and the associated

polymer to be implanted may be chosen based on the type of seizures, medical history,

size of the patient and other objective criteria; however the clinician is unlikely to know

for a particular patient the exact amount of drug that will be required for the treatment

of the seizures. The rate at which the AED is released from an implanted passive release

can only be modified by removing or implanting additional devices requiring potentially

several additional invasive procedures.

The lack of flexibility in the AED delivery rate is a major limitation of the PLGA

polymer and is the reason why after establishing proof of concept that intracranial

implantation of polymer-based drug delivery systems could be used successfully in the

treatment of epilepsy, the resources were directed towards active release polymers such

as PPy.

7.2 Development of the PPy polymers

Drugs can be incorporated into the polymer as dopant anions to counterbalance the

positive charge associated with the oxidative polymerization (Chapter 1.4.3.2) or loaded

after polymerization through ion exchange by redox cycling the polymer in appropriate

media.(Uppalapati, Boyd et al. 2016) In reverse, upon intermittent application of an

Page 150: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 128 -

electrical current, the electrostatic interactions between the polymer and the negatively

charged drug change resulting in release of the drug. This advantage of being reactive

to electrical stimulation would allow an endogenous or exogenous electrical current to

control the release of an AED. The electrical firing of the brain during a seizure could

be such endogenous stimulation although the release of the AED would treat and not

prevent the seizure from happening. An exogenous electrical stimulation coming from

an implanted seizure advisory system would offer more flexibility by also providing

treatment during periods of fair or high likelihood of a seizure event and by adjusting

the intensity of the electrical stimulation and therefore the amount of drug being

released. Electrical stimuli can be applied through tissues and therefore the electrodes

delivering the current could be positioned over the dura while the polymer itself could

be placed under the dura. No wire or tubing will permanently compromise the isolation

of the brain as it is the case with a mini-osmotic pump (Chapter 1.3.3). However, one

limitation remains, which is that the nature of the polymer requires the drug to be

charged for incorporation, and therefore a significant number of commonly used AEDS

would not be suitable for administration through this device. For example, phenytoin

could not be loaded into a PPy polymer, although its pro-drug fosphenytoin could. The

effectiveness of fosphenytoin when administered directly onto the brain parenchyma

will need to be studied as it is unknown if the hydrolysis of fosphenytoin to phenytoin

would happen and if other residual products such as formaldehyde could negatively

impact the brain.

The results of the study presented in Chapter 6 showed for the first time that implanted

PPy mats on top of the brain cortex of GAERS did not induce obvious inflammation,

trauma, gliosis and neuronal toxicity. Therefore, the results are demonstrating in vivo

that the PPy used offered good histocompatibility with central nervous system cells and

that PPy sheets could be used as a drug delivery device implanted onto the surface of

the brain parenchyma. These results confirmed those of a previous study where PPy

sheets have been shown to be well tolerated by biological tissues, including brain

tissues.(George, Lyckman et al. 2005, Ateh, Navsaria et al. 2006) Nevertheless, an acute

short lasting inflammatory reaction leading to the formation of a fibrous capsule, around

the polymer, hindering the release of drug could have been missed. The friability of the

polymer did not allow explant analysis and it would limit the ability of the polymer to

Page 151: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 129 -

be linked to any other device. For future experiments, the polymer would benefit from

structural support, such as being fabricated onto a platinum surface.

As mentioned in the discussion of Chapter 6, the comparison of NeuN-IHC and GFAP-

IHC scores reveals that the amount of neuronal death and gliosis was significantly less

in the PPy side than the sham surgery side of the cortex implying that the application of

PPy could protect the CNS tissue after surgery. PPy substrates have been used as nerve

guidance channels to bridge the gap between severed nerve ends and as injectable

scaffold to repair ischemic myocardium after myocardial infarction.(Schmidt, Shastri et

al. 1997, Wang, Roberge et al. 2004) The combination of a “neuro-protector” and an

AED has the potential to help reduce the incidence of complications such as seizures

following craniotomies, brain resections and brain trauma. In those cases, the isolation

of the brain has already been compromised and the benefits resulting from the

implantation of AED/PPy polymer could be superior to the risks. Further studies would

need to be performed to confirm such hypothesis.

7.3 What are the challenges in the development of an implantable drug

delivery device for the treatment of epilepsy?

The challenges in the development of an implantable drug delivery device for the

treatment of epilepsy are numerous and complex. They include but are not limited to:

The nature of the materials used

As mentioned in the introduction (table 1.2), multiple kinds of polymers have been used

to investigate the delivery of antiepileptic drugs. PLGA polymers were chosen because

of their known biocompatibility, their efficacy in drug loading and delivery, and the

experience in its fabrication that the collaborators working on this thesis had. Other

polymers could have been used but it is beyond the scope of the thesis to compare the

characteristic of all kinds of polymer available.

The ability to control/vary/tune drug release profiles

One major limiting factor in the use of PLGA polymers is the inability to adjust the

release of AEDs. PPy was investigated to enable drug delivery on demand. The PPy

drug release will need to be tested and the connection of the PPy polymer to electrodes

Page 152: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 130 -

may prove to be challenging. Because of its friable nature, the PPy polymers may need

to be grown onto a platinum plate to secure such connection.

The effect of the drug on the event and population of animal used in studies

Because seizures can have multiple underlying pathophysiologies one drug/polymer

combination may not have consistent effects. Even if we prove that lacosamide loaded

PPy polymers alleviate seizure in GAERS, there will remain uncertainty to whether or

not such polymers will be efficacious to treat seizure in human. The AED loaded

polymers will likely need to be tested in multiple animal models and still risk having

narrow therapeutic indications.

The location and type of implant

Because the diffusion of AEDs from the polymer remains localised (a few mms of

tissue penetration) the polymer needs to be implanted near the location of the origin of

the seizure. The type of implant (i.e. mats versus injectable) may then depend of the

localisation of the origin of the seizure. Implantable polymers may be preferable for

seizure foci close to the periphery of the cortex and injectable polymers for more in

depth seizure foci.

7.4 What is next?

Coming from a different background and as a PhD candidate, one thing that this journey

taught me is how little is known about the brain and its function, and consequently

about its pathophysiology and treatments. Major initiatives focusing on revolutionizing

the understanding of the brain such as the “White House Neuroscience Initiative” are

primordial for the development of effective therapies for neurological conditions such

as epilepsy. Without significant advancements in our understanding of the brain, the

development of anti-epileptic treatments such as polymer based implantable drug

delivery devices will remain trial and error based. Furthermore, a wide range of

professionals needs to be involved: neurologists for the identification of the patient’s

need, material scientists for the development of the polymer and refinement of the drug

loading and unloading ability, engineers for the integration of the polymer within an

interactive device, neuroscientists and veterinarians for the trials of such devices in

animal models and neurosurgeon to facilitate the transition to human clinical trials.

Page 153: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 131 -

After demonstrating biocompatibility, the next steps in the development of the PPy

implantable delivery device would be to identify an appropriate AED that can be loaded

into the PPy, study the ideal conditions of polymerisation for the maximal drug loading

while still optimizing drug release characteristics for long term efficient drug delivery.

The delivery of AEDs will ideally be triggered by a device predicting the likelihood of

seizure occurrence. Automatic drug administration would then occur only when most

needed decreasing side effects and greatly improving patients’ quality of life.

Once appropriate in vitro release data have been obtained, the anti-epileptic effects of

the device can be tested in different in vivo animal models of epilepsy. In addition to

long term histocompatibility studies, the development of intracranial implantable

devices will require functional studies for biocompatibility to be performed. Cognitive

function, motor functions, memory, and other cortical and limbic functions must not be

significantly compromised by PPy implantation.

The neuro-protective effect of PPy should also be investigated in a brain trauma model.

For that particular application, PPy can be loaded with AEDs but could also be loaded

with other drugs such as anti-inflammatories (e.g. dexamethasone).(Jiang, Sun et al.

2013) While the development of an antiepileptic device is still years away, the

development of an implantable device to be used to decrease post-craniotomy brain

tissue inflammation would certainly be quicker to develop.

Page 154: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 132 -

References

1. Abbott, N. J., A. A. Patabendige, D. E. Dolman, S. R. Yusof and D. J. Begley

(2010). "Structure and function of the blood-brain barrier." Neurobiol Dis

37(1): 13-25.

2. Abbott, N. J., L. Ronnback and E. Hansson (2006). "Astrocyte-endothelial

interactions at the blood-brain barrier." Nat Rev Neurosci 7(1): 41-53.

3. Adams, H. R. (2001). Vet Pharmacol Therap. Ames, Iowa State University

Press.

4. Allahyari, M. and E. Mohit (2015). "Peptide/protein vaccine delivery system

based on PLGA particles." Hum Vaccin Immunother: 0.

5. Anissian, D., M. Ghasemi-Kasman, M. Khalili-Fomeshi, A. Akbari, M.

Hashemian, S. Kazemi and A. A. Moghadamnia (2017). "Piperine-loaded

chitosan-STPP nanoparticles reduce neuronal loss and astrocytes activation in

chemical kindling model of epilepsy." Int J Biol Macromol.

6. Arcieri, G. M., N. Becker, B. Esposito, E. Griffith, A. Heyd, C. Neumann, B.

O'Brien and P. Schacht (1989). "Safety of intravenous ciprofloxacin. A

review." Am J Med 87(5a): 92s-97s.

7. Arica, B., H. S. Kas, A. Moghdam, N. Akalan and A. A. Hincal (2005).

"Carbidopa/levodopa-loaded biodegradable microspheres: in vivo evaluation

on experimental Parkinsonism in rats." J Control Release 102(3): 689-697.

8. Ateh, D. D., H. A. Navsaria and P. Vadgama (2006). "Polypyrrole-based

conducting polymers and interactions with biological tissues." J R Soc

Interface 3(11): 741-752.

9. Barakat, N. S. and M. A. Radwan (2006). "In vitro performance of

carbamazepine loaded to various molecular weights of poly (D,L-lactide-co-

glycolide)." Drug Deliv 13(1): 9-18.

10. Barakat, N. S. and A. E. Yassin (2006). "In vitro characterization of

carbamazepine-loaded precifac lipospheres." Drug Deliv 13(2): 95-104.

11. Barichello, J. M., M. Morishita, K. Takayama and T. Nagai (1999).

"Encapsulation of hydrophilic and lipophilic drugs in PLGA nanoparticles by

the nanoprecipitation method." Drug Dev Ind Pharm 25(4): 471-476.

Page 155: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 133 -

12. Bauer, S., L. M. Willems, E. Paule, C. Petschow, J. P. Zollner, F. Rosenow

and A. Strzelczyk (2017). "The efficacy of lacosamide as monotherapy and

adjunctive therapy in focal epilepsy and its use in status epilepticus: clinical

trial evidence and experience." Ther Adv Neurol Disord 10(2): 103-126.

13. Bauquier, S. H., J. L. Jiang, Z. Yue, A. Lai, Y. Chen, S. E. Moulton, K.

McLean, J., S. Vogrin, A. J. Halliday, G. G. Wallace and M. J. Cook (2016).

"Antiepileptic Effects of Lacosamide Loaded Polymers Implanted Subdurally

in GAERS." Int J Polym Sci: 940.

14. Bauquier, S. H., A. Lai, J. L. Jiang and M. J. cook (2015). "Evaluation of an

automated spike-and-wave complex detection algorithm in the EEG from a

rat model of absence epilepsy." Neurosci Bull.

15. Bauquier, S. H., A. Lai, J. L. Jiang and M. J. cook (2016). "Clonic seizures

induced by oral administration of enrofloxacin in GAERS." Comp med 66(3):

220-224.

16. Bender, R. and S. Lange (2001). "Adjusting for multiple testing--when and

how?" J Clin Epidemiol 54(4): 343-349.

17. Benelli, P., B. Conti, I. Genta, M. Costantini and L. Montanari (1998).

"Clonazepam microencapsulation in poly-D,L-lactide-co-glycolide

microspheres." Journal of Microencapsulation 15(4): 431-443.

18. Bensadoun, J. C., L. Pereira de Almeida, E. G. Fine, J. L. Tseng, N. Deglon

and P. Aebischer (2003). "Comparative study of GDNF delivery systems for

the CNS: polymer rods, encapsulated cells, and lentiviral vectors." J Control

Release 87(1-3): 107-115.

19. Berrabah, M., D. Andre, F. Prevot, A. M. Orecchioni and O. Lafont (1994).

"GC-MS determination of phenobarbitone entrapped in poly-epsilon-

caprolactone nanocapsules." J Pharm Biomed Anal 12(3): 373-378.

20. Beyreuther, B. K., J. Freitag, C. Heers, N. Krebsfanger, U. Scharfenecker and

T. Stohr (2007). "Lacosamide: A review of preclinical properties." CNS Drug

Reviews 13(1): 21-42.

21. Bhattacharyya, S., R. Darby and A. L. Berkowitz (2014). "Antibiotic-induced

neurotoxicity." Curr Infect Dis Rep 16(12): 448.

Page 156: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 134 -

22. Bodmeier, R. and J. W. McGinity (1987). "The preparation and evaluation of

drug-containing poly(dl-lactide) microspheres formed by the solvent

evaporation method." Pharm Res 4(6): 465-471.

23. Bohrey, S., V. Chourasiya and A. Pandey (2016). "Polymeric nanoparticles

containing diazepam: preparation, optimization, characterization, in-vitro

drug release and release kinetic study." Nano Converg 3(1): 3.

24. Boison, D., L. Scheurer, J. L. Tseng, P. Aebischer and H. Mohler (1999).

"Seizure suppression in kindled rats by intraventricular grafting of an

adenosine releasing synthetic polymer." Exp Neurol 160(1): 164-174.

25. Boon, P., E. De Cock, A. Mertens and E. Trinka (2018). "Neurostimulation

for drug-resistant epilepsy: a systematic review of clinical evidence for

efficacy, safety, contraindications and predictors for response." Curr Opin

Neurol 31(2): 198-210.

26. Calandre, E. P., F. Rico-Villademoros and M. Slim (2016). "Alpha2delta

ligands, gabapentin, pregabalin and mirogabalin: a review of their clinical

pharmacology and therapeutic use." Expert Rev Neurother 16(11): 1263-

1277.

27. Cao, H., M. M. Chen, Y. Liu, Y. Y. Liu, Y. Q. Huang, J. H. Wang, J. D. Chen

and Q. Q. Zhang (2015). "Fish collagen-based scaffold containing PLGA

microspheres for controlled growth factor delivery in skin tissue

engineering." Colloids Surf B Biointerfaces 136: 1098-1106.

28. Chadwick, D., A. Shukralla and T. Marson (2009). "Comparing drug

treatments in epilepsy." Ther Adv Neurol Disord 2(3): 181-187.

29. Chen, J. and S. S. Davis (2002). "The release of diazepam from

poly(hydroxybutyrate-hydroxyvalerate) microspheres." J Microencapsul

19(2): 191-201.

30. Chen, Y., Q. Gu, Z. Yue, J. M. Crook, S. E. Moulton, M. J. Cook and G. G.

Wallace (2017). "Development of drug-loaded polymer microcapsules for

treatment of epilepsy." Biomater Sci 5(10): 2159-2168.

31. Chew, S. Y., J. Wen, E. K. Yim and K. W. Leong (2005). "Sustained release

of proteins from electrospun biodegradable fibers." Biomacromolecules 6(4):

2017-2024.

Page 157: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 135 -

32. Cho, C. S., S. Y. Han, J. H. Ha, S. H. Kim and D. Y. Lim (1999).

"Clonazepam release from bioerodible hydrogels based on semi-

interpenetrating polymer networks composed of poly(epsilon-caprolactone)

and poly(ethylene glycol) macromer." Int J Pharm 181(2): 235-242.

33. Chong, D. J. and A. M. Lerman (2016). "Practice Update: Review of

Anticonvulsant Therapy." Curr Neurol Neurosci Rep 16(4): 39.

34. Cook, M. J., T. J. O'Brien, S. F. Berkovic, M. Murphy, A. Morokoff, G.

Fabinyi, W. D'Souza, R. Yerra, J. Archer, L. Litewka, S. Hosking, P.

Lightfoot, V. Ruedebusch, W. D. Sheffield, D. Snyder, K. Leyde and D.

Himes (2013). "Prediction of seizure likelihood with a long-term, implanted

seizure advisory system in patients with drug-resistant epilepsy: a first-in-

man study." Lancet Neurol 12(6): 563-571.

35. Correale, J. and A. Villa (2009). "Cellular elements of the blood-brain

barrier." Neurochem Res 34(12): 2067-2077.

36. Cui, W., X. Li, X. Zhu, G. Yu, S. Zhou and J. Weng (2006). "Investigation of

drug release and matrix degradation of electrospun poly(DL-lactide) fibers

with paracetanol inoculation." Biomacromolecules 7(5): 1623-1629.

37. Daneman, R. (2012). "The blood-brain barrier in health and disease." Ann

Neurol 72(5): 648-672.

38. Danober, L., C. Deransart, A. Depaulis, M. Vergnes and C. Marescaux

(1998). "Pathophysiological mechanisms of genetic absence epilepsy in the

rat." Prog Neurobiol 55(1): 27-57.

39. de Flon, P., E. Kumlien, C. Reuterwall and P. Mattsson (2010). Empirical

evidence of underutilization of referrals for epilepsy surgery evaluation. Eur J

Neurol. England. 17: 619-625.

40. Dong, W. Y., P. Maincent and R. Bodmeier (2007). "In vitro and in vivo

evaluation of carbamazepine-loaded enteric microparticles." Int J Pharm

331(1): 84-92.

41. Duarte, A. R., C. Roy, A. Vega-Gonzalez, C. M. Duarte and P. Subra-

Paternault (2007). "Preparation of acetazolamide composite microparticles by

supercritical anti-solvent techniques." Int J Pharm 332(1-2): 132-139.

Page 158: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 136 -

42. Engel, J. and T. A. Pedley (2008). Epilepsy. [electronic resource] : a

comprehensive textbook, Philadelphia : Lippincott Williams & Wilkins,

c2008.

43. 2nd ed.

44. Ferranti, V., H. Marchais, C. Chabenat, A. M. Orecchioni and O. Lafont

(1999). "Primidone-loaded poly-epsilon-caprolactone nanocapsules:

incorporation efficiency and in vitro release profiles." Int J Pharm 193(1):

107-111.

45. Filipovic-Grcic, J., B. Perissutti, M. Moneghini, D. Voinovich, A. Martinac

and I. Jalsenjak (2003). "Spray-dried carbamazepine-loaded chitosan and

HPMC microspheres: preparation and characterisation." J Pharm Pharmacol

55(7): 921-931.

46. Fisher, R. S., C. Acevedo, A. Arzimanoglou, A. Bogacz, J. H. Cross, C. E.

Elger, J. Engel, Jr., L. Forsgren, J. A. French, M. Glynn, D. C. Hesdorffer, B.

I. Lee, G. W. Mathern, S. L. Moshe, E. Perucca, I. E. Scheffer, T. Tomson,

M. Watanabe and S. Wiebe (2014). "ILAE official report: a practical clinical

definition of epilepsy." Epilepsia 55(4): 475-482.

47. Fisher, R. S., W. van Emde Boas, W. Blume, C. Elger, P. Genton, P. Lee and

J. Engel, Jr. (2005). "Epileptic seizures and epilepsy: definitions proposed by

the International League Against Epilepsy (ILAE) and the International

Bureau for Epilepsy (IBE)." Epilepsia 46(4): 470-472.

48. Fisher, R. S. and A. L. Velasco (2014). "Electrical brain stimulation for

epilepsy." Nat Rev Neurol 10(5): 261-270.

49. Fisher, R. S., B. G. Vickrey, P. Gibson, B. Hermann, P. Penovich, A. Scherer

and S. Walker (2000). "The impact of epilepsy from the patient’s perspective

I. Descriptions and subjective perceptions." Epilepsy Research 41(1): 39-51.

50. Fresta, M., G. Cavallaro, G. Giammona, E. Wehrli and G. Puglisi (1996).

"Preparation and characterization of polyethyl-2-cyanoacrylate nanocapsules

containing antiepileptic drugs." Biomaterials 17(8): 751-758.

51. Gavini, E., A. B. Hegge, G. Rassu, V. Sanna, C. Testa, G. Pirisino, J. Karlsen

and P. Giunchedi (2006). "Nasal administration of carbamazepine using

chitosan microspheres: in vitro/in vivo studies." Int J Pharm 307(1): 9-15.

Page 159: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 137 -

52. George, P. M., A. W. Lyckman, D. A. LaVan, A. Hegde, Y. Leung, R.

Avasare, C. Testa, P. M. Alexander, R. Langer and M. Sur (2005).

"Fabrication and biocompatibility of polypyrrole implants suitable for neural

prosthetics." Biomaterials 26(17): 3511-3519.

53. Giannola, L. I., V. De Caro, V. Di Stefano and M. C. Rizzo (1993). "In vitro

evaluation of cumulative release of valproic acid and vitamin E from

hexadecanol microspheres. Part 2: Antiepileptic agents." Pharmazie 48(12):

917-920.

54. Giunchedi, P., B. Conti, S. Scalia and U. Conte (1998). "In vitro degradation

study of polyester microspheres by a new HPLC method for monomer release

determination." J Control Release 56(1-3): 53-62.

55. Gouhier, C., S. Chalon, M. C. Venier-Julienne, S. Bodard, J. Benoit, J.

Besnard and D. Guilloteau (2000). "Neuroprotection of nerve growth factor-

loaded microspheres on the D2 dopaminergic receptor positive-striatal

neurones in quinolinic acid-lesioned rats: a quantitative autoradiographic

assessment with iodobenzamide." Neurosci Lett 288(1): 71-75.

56. Grondahl, T. O. and I. A. Langmoen (1993). "Epileptogenic effect of

antibiotic drugs." J Neurosurg 78(6): 938-943.

57. Groynom, R., E. Shoffstall, L. S. Wu, R. H. Kramer and E. B. Lavik (2015).

"Controlled release of photoswitch drugs by degradable polymer

microspheres." J Drug Target 23(7-8): 710-715.

58. Gurbanova, A. A., R. Aker, K. Berkman, F. Y. Onat, C. M. van Rijn and G.

van Luijtelaar (2006). "Effect of systemic and intracortical administration of

phenytoin in two genetic models of absence epilepsy." Br J Pharmacol

148(8): 1076-1082.

59. Haik-Creguer, K. L., G. L. Dunbar, B. A. Sabel and U. Schroeder (1998).

"Small drug sample fabrication of controlled release polymers using the

microextrusion method." J Neurosci Methods 80(1): 37-40.

60. Halliday, A. J., T. E. Campbell, T. S. Nelson, K. J. McLean, G. G. Wallace

and M. J. Cook (2013). "Levetiracetam-loaded biodegradable polymer

implants in the tetanus toxin model of temporal lobe epilepsy in rats." J Clin

Neurosci 20(1): 148-152.

Page 160: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 138 -

61. Halliday, A. J., T. E. Campbell, J. M. Razal, K. J. McLean, T. S. Nelson, M.

J. Cook and G. G. Wallace (2011). "In vivo biocompatibility and in vitro

characterization of poly-lactide-co-glycolide structures containing

levetiracetam, for the treatment of epilepsy." J Biomed Mater Res A.

62. Halliday, A. J. and M. J. Cook (2009). "Polymer-based drug delivery devices

for neurological disorders." CNS Neurol Disord Drug Targets 8(3): 205-221.

63. Halliday, A. J., S. E. Moulton, G. G. Wallace and M. J. Cook (2012). "Novel

methods of antiepileptic drug delivery -- polymer-based implants." Adv Drug

Deliv Rev 64(10): 953-964.

64. Hartz, A. M. and B. Bauer (2011). "ABC transporters in the CNS - an

inventory." Curr Pharm Biotechnol 12(4): 656-673.

65. Haznedar, S. and B. Dortunc (2004). "Preparation and in vitro evaluation of

Eudragit microspheres containing acetazolamide." Int J Pharm 269(1): 131-

140.

66. Hofler, J. and E. Trinka (2013). "Lacosamide as a new treatment option in

status epilepticus." Epilepsia 54(3): 393-404.

67. Hofler, J., I. Unterberger, J. Dobesberger, G. Kuchukhidze, G. Walser and E.

Trinka (2011). "Intravenous lacosamide in status epilepticus and seizure

clusters." Epilepsia 52(10): e148-152.

68. Hutchison, A. S., T. W. Lewis, S. E. Moulton, G. M. Spinks and G. G.

Wallace (2000). "Development of polypyrrole-based electromechanical

actuators." Synth Met 113(1-2): 121-127.

69. Ishihara, T. and T. Mizushima (2010). "Techniques for efficient entrapment

of pharmaceuticals in biodegradable solid micro/nanoparticles." Expert Opin

Drug Deliv 7(5): 565-575.

70. Jeong, Y. I., J. B. Cheon, S. H. Kim, J. W. Nah, Y. M. Lee, Y. K. Sung, T.

Akaike and C. S. Cho (1998). "Clonazepam release from core-shell type

nanoparticles in vitro." J Control Release 51(2-3): 169-178.

71. Jiang, J. L., Z. Yue, S. H. Bauquier, A. Lai, Y. Chen, K. J. McLean, A. J.

Halliday, Y. Sui, S. Moulton, G. G. Wallace and M. J. Cook (2015).

"Injectable phenytoin loaded polymeric microspheres for the control of

temporal lobe epilepsy in rats." Restor Neurol Neurosci 33(6): 823-834.

Page 161: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 139 -

72. Jiang, S., Y. Sun, X. Cui, X. Huang, Y. He, S. Ji, W. Shi and D. Ge (2013).

"Enhanced drug loading capacity of polypyrrole nanowire network for

controlled drug release." Synth Met 163(Supplement C): 19-23.

73. Jyothi, N. V., P. M. Prasanna, S. N. Sakarkar, K. S. Prabha, P. S. Ramaiah

and G. Y. Srawan (2010). "Microencapsulation techniques, factors

influencing encapsulation efficiency." J Microencapsul 27(3): 187-197.

74. Kellinghaus, C. (2009). "Lacosamide as treatment for partial epilepsy:

mechanisms of action, pharmacology, effects, and safety." Ther Clin Risk

Manag 5: 757-766.

75. Kellinghaus, C., S. Berning and F. Stogbauer (2014). "Intravenous

lacosamide or phenytoin for treatment of refractory status epilepticus." Acta

Neurol Scand 129(5): 294-299.

76. Keppel Hesselink, J. M. and D. J. Kopsky (2017). "Phenytoin: 80 years

young, from epilepsy to breast cancer, a remarkable molecule with multiple

modes of action." J Neurol 264(8): 1617-1621.

77. Killian, D. M., S. Hermeling and P. J. Chikhale (2007). "Targeting the

cerebrovascular large neutral amino acid transporter (LAT1) isoform using a

novel disulfide-based brain drug delivery system." Drug Deliv 14(1): 25-31.

78. Kim, A., J. E. Kim, Y. M. Paek, K. S. Hong, Y. J. Cho, J. Y. Cho, H. K. Park,

H. K. Koo and P. Song (2013). "Cefepime- Induced Non-Convulsive Status

Epilepticus (NCSE)." J Epilepsy Res 3(1): 39-41.

79. Kluge, J., M. Mazzotti and G. Muhrer (2010). "Solubility of Ketoprofen in

colloidal PLGA." Int J Pharm 399(1-2): 163-172.

80. Kohane, D. S., G. L. Holmes, Y. Chau, D. Zurakowski, R. Langer and B. H.

Cha (2002). "Effectiveness of muscimol-containing microparticles against

pilocarpine-induced focal seizures." Epilepsia 43(12): 1462-1468.

81. Kokaia, M., P. Aebischer, E. Elmer, J. Bengzon, P. Kalen, Z. Kokaia and O.

Lindvall (1994). "Seizure suppression in kindling epilepsy by intracerebral

implants of GABA- but not by noradrenaline-releasing polymer matrices."

Exp Brain Res 100(3): 385-394.

Page 162: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 140 -

82. Krewson, C. E., R. Dause, M. Mak and W. M. Saltzman (1996).

"Stabilization of nerve growth factor in controlled release polymers and in

tissue." J Biomater Sci Polym Ed 8(2): 103-117.

83. Krewson, C. E., M. L. Klarman and W. M. Saltzman (1995). "Distribution of

nerve growth factor following direct delivery to brain interstitium." Brain Res

680(1-2): 196-206.

84. Kubek, M. J., D. Liang, K. E. Byrd and A. J. Domb (1998). "Prolonged

seizure suppression by a single implantable polymeric-TRH microdisk

preparation." Brain Res 809(2): 189-197.

85. Kwan, P. and M. J. Brodie (2000). "Early identification of refractory

epilepsy." N Engl J Med 342(5): 314-319.

86. Kwan, P. and M. R. Sperling (2009). "Refractory seizures: try additional

antiepileptic drugs (after two have failed) or go directly to early surgery

evaluation?" Epilepsia 50 Suppl 8: 57-62.

87. Lajoie, J. M. and E. V. Shusta (2015). "Targeting receptor-mediated transport

for delivery of biologics across the blood-brain barrier." Annu Rev Pharmacol

Toxicol 55: 613-631.

88. Lerner, P. I., H. Smith and L. Weinstein (1967). "Penicillin neurotoxicity."

Ann N Y Acad Sci 145(2): 310-318.

89. Li, Z., Q. Li, S. Simon, N. Guven, K. Borges and B. B. Youan (2007).

"Formulation of spray-dried phenytoin loaded poly(epsilon-caprolactone)

microcarrier intended for brain delivery to treat epilepsy." J Pharm Sci 96(5):

1018-1030.

90. Lin, P. C., Y. H. Hsieh, F. F. Liao and S. H. Chen (2010). "Determination of

free and total levels of phenytoin in human plasma from patients with

epilepsy by MEKC: an adequate alternative to HPLC." Electrophoresis 31(9):

1572-1582.

91. Lipinski, C. A. (2000). "Drug-like properties and the causes of poor solubility

and poor permeability." J Pharmacol Toxicol Methods 44(1): 235-249.

92. Lode, H. (1999). "Potential interactions of the extended-spectrum

fluoroquinolones with the CNS." Drug Saf 21(2): 123-135.

Page 163: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 141 -

93. Loscher, W. (2005). "Mechanisms of drug resistance." Epileptic Disord 7

Suppl 1: S3-9.

94. Madan, J. R., B. R. Adokar and K. Dua (2015). "Development and evaluation

of in situ gel of pregabalin." Int J Pharm Investig 5(4): 226-233.

95. Makadia, H. K. and S. J. Siegel (2011). "Poly Lactic-co-Glycolic Acid

(PLGA) as Biodegradable Controlled Drug Delivery Carrier." Polymers

(Basel) 3(3): 1377-1397.

96. Malmgren, K., M. Reuber and R. Appleton (2012). Differential Diagnosis of

Epilepsy. Oxford, Oxford University Press.

97. Manning, J. P., D. A. Richards, N. Leresche, V. Crunelli and N. G. Bowery

(2004). "Cortical-area specific block of genetically determined absence

seizures by ethosuximide." Neuroscience 123(1): 5-9.

98. Marescaux, C., M. Vergnes and A. Depaulis (1992). "Genetic absence

epilepsy in rats from Strasbourg--a review." J Neural Transm Suppl 35: 37-

69.

99. McRae, A. and A. Dahlstrom (1994). "Transmitter-loaded polymeric

microspheres induce regrowth of dopaminergic nerve terminals in striata of

rats with 6-OH-DA induced parkinsonism." Neurochem Int 25(1): 27-33.

100. McRae, A., S. Hjorth, D. W. Mason, L. Dillon and T. R. Tice (1991).

"Microencapsulated dopamine (DA)-induced restitution of function in 6-

OHDA-denervated rat striatum in vivo: comparison between two microsphere

excipients." J Neural Transplant Plast 2(3-4): 165-173.

101. Menei, P., J. M. Pean, V. Nerriere-Daguin, C. Jollivet, P. Brachet and J. P.

Benoit (2000). "Intracerebral implantation of NGF-releasing biodegradable

microspheres protects striatum against excitotoxic damage." Exp Neurol

161(1): 259-272.

102. Merritt, H. H. and T. J. Putnam (1984). "Landmark article Sept 17, 1938:

Sodium diphenyl hydantoinate in the treatment of convulsive disorders. By H.

Houston Merritt and Tracy J. Putnam." Jama 251(8): 1062-1067.

103. Miller, J. W. and H. P. Goodkin (2014). Epilepsy, Chichester, West Sussex :

John Wiley & Sons Ltd., 2014.

Page 164: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 142 -

104. Miller, J. W. and H. P. Goodkin (2014). Epilepsy, Chichester, West Sussex :

John Wiley & Sons Ltd., 2014.

105. Montanari, L., F. Cilurzo, L. Valvo, A. Faucitano, A. Buttafava, A. Groppo, I.

Genta and B. Conti (2001). "Gamma irradiation effects on stability of

poly(lactide-co-glycolide) microspheres containing clonazepam." J Control

Release 75(3): 317-330.

106. Moulton, S. E., M. D. Imisides, R. L. Shepherd and G. G. Wallace (2008).

"Galvanic coupling conducting polymers to biodegradable Mg initiates

autonomously powered drug release." J Mater Chem 18(30): 3608-3613.

107. Muller, R., Z. Yue, S. Ahmadi, W. Ng, W. M. Grosse, M. J. Cook, G. G.

Wallace and S. E. Moulton (2016). "Development and validation of a seizure

initiated drug delivery system for the treatment of epilepsy." Sens Actuators

B: Chem 236: 732-740.

108. Nah, J. W., Y. W. Paek, Y. I. Jeong, D. W. Kim, C. S. Cho, S. H. Kim and M.

Y. Kim (1998). "Clonazepam release from poly(DL-lactide-co-glycolide)

nanoparticles prepared by dialysis method." Arch Pharm Res 21(4): 418-422.

109. Navakatikyan, M. A., P. B. Colditz, C. J. Burke, T. E. Inder, J. Richmond and

C. E. Williams (2006). "Seizure detection algorithm for neonates based on

wave-sequence analysis." Clin Neurophysiol 117(6): 1190-1203.

110. Nkansah, M. K., S. Y. Tzeng, A. M. Holdt and E. B. Lavik (2008).

"Poly(lactic-co-glycolic acid) nanospheres and microspheres for short- and

long-term delivery of bioactive ciliary neurotrophic factor." Biotechnol

Bioeng 100(5): 1010-1019.

111. Noebels, J. L. and H. H. Jasper (2012). Jasper's basic mechanisms of the

epilepsies. New York, Oxford University Press.

112. Ovchinnikov, A., A. Luttjohann, A. Hramov and G. van Luijtelaar (2010). An

algorithm for real-time detection of spike-wave discharges in rodents. J

Neurosci Methods. Netherlands, 2010 Elsevier B.V. 194: 172-178.

113. Pardridge, W. M. (2012). "Drug transport across the blood-brain barrier." J

Cereb Blood Flow Metab 32(11): 1959-1972.

114. Passerini, N., B. Perissutti, M. Moneghini, D. Voinovich, B. Albertini, C.

Cavallari and L. Rodriguez (2002). "Characterization of carbamazepine-

Page 165: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 143 -

Gelucire 50/13 microparticles prepared by a spray-congealing process using

ultrasounds." J Pharm Sci 91(3): 699-707.

115. Patterson, J. E., M. B. James, A. H. Forster and T. Rades (2008). "Melt

extrusion and spray drying of carbamazepine and dipyridamole with

polyvinylpyrrolidone/vinyl acetate copolymers." Drug Dev Ind Pharm 34(1):

95-106.

116. Pean, J. M., P. Menei, O. Morel, C. N. Montero-Menei and J. P. Benoit

(2000). "Intraseptal implantation of NGF-releasing microspheres promote the

survival of axotomized cholinergic neurons." Biomaterials 21(20): 2097-

2101.

117. Perucca, E. (1997). "A pharmacological and clinical review on topiramate, a

new antiepileptic drug." Pharmacol Res 35(4): 241-256.

118. Pritchard, E. M., C. Szybala, D. Boison and D. L. Kaplan (2010). "Silk

fibroin encapsulated powder reservoirs for sustained release of adenosine." J

Control Release 144(2): 159-167.

119. Putnam, T. J. and H. H. Merritt (1937). "EXPERIMENTAL

DETERMINATION OF THE ANTICONVULSANT PROPERTIES OF

SOME PHENYL DERIVATIVES." Science 85(2213): 525-526.

120. Rane, Y. M., R. C. Mashru, M. G. Sankalia, V. B. Sutariya and P. P. Shah

(2007). "Investigations on factors affecting chitosan for dissolution

enhancement of oxcarbazepine by spray dried microcrystal formulation with

an experimental design approach." Drug Dev Ind Pharm 33(9): 1008-1023.

121. Rassner, M. P., J. M. Hebel, D. M. Altenmuller, S. Volz, L. S. Herrmann, T.

J. Feuerstein and T. M. Freiman (2015). "Reduction of epileptiform activity

through local valproate-implants in a rat neocortical epilepsy model." Seizure

30: 6-13.

122. Reese, T. S. and M. J. Karnovsky (1967). "Fine structural localization of a

blood-brain barrier to exogenous peroxidase." J Cell Biol 34(1): 207-217.

123. Rho, J. M., S. D. Donevan and M. A. Rogawski (1994). "Mechanism of

action of the anticonvulsant felbamate: opposing effects on N-methyl-D-

aspartate and gamma-aminobutyric acidA receptors." Ann Neurol 35(2): 229-

234.

Page 166: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 144 -

124. Rudolf, G., M. T. Bihoreau, R. F. Godfrey, S. P. Wilder, R. D. Cox, M.

Lathrop, C. Marescaux and D. Gauguier (2004). "Polygenic control of

idiopathic generalized epilepsy phenotypes in the genetic absence rats from

Strasbourg (GAERS)." Epilepsia 45(4): 301-308.

125. Ryu, J., Y. I. Jeong, I. S. Kim, J. H. Lee, J. W. Nah and S. H. Kim (2000).

"Clonazepam release from core-shell type nanoparticles of poly(epsilon-

caprolactone)/poly(ethylene glycol)/poly(epsilon-caprolactone) triblock

copolymers." Int J Pharm 200(2): 231-242.

126. Saltzman, W. M., M. W. Mak, M. J. Mahoney, E. T. Duenas and J. L.

Cleland (1999). "Intracranial delivery of recombinant nerve growth factor:

release kinetics and protein distribution for three delivery systems." Pharm

Res 16(2): 232-240.

127. Sander, J. W. (2003). "The epidemiology of epilepsy revisited." Curr Opin

Neurol 16(2): 165-170.

128. Sankaraneni, R. and D. Lachhwani (2015). "Antiepileptic drugs--a review."

Pediatr Ann 44(2): e36-42.

129. Schmidt, C. E., V. R. Shastri, J. P. Vacanti and R. Langer (1997).

"Stimulation of neurite outgrowth using an electrically conducting polymer."

Proc Natl Acad Sci U S A 94(17): 8948-8953.

130. Serlin, Y., I. Shelef, B. Knyazer and A. Friedman (2015). "Anatomy and

physiology of the blood-brain barrier." Semin Cell Dev Biol 38: 2-6.

131. Sharma, D., D. Maheshwari, G. Philip, R. Rana, S. Bhatia, M. Singh, R.

Gabrani, S. K. Sharma, J. Ali, R. K. Sharma and S. Dang (2014).

"Formulation and optimization of polymeric nanoparticles for intranasal

delivery of lorazepam using Box-Behnken design: in vitro and in vivo

evaluation." Biomed Res Int 2014: 156010.

132. Shorvon, S. D. (1996). "The epidemiology and treatment of chronic and

refractory epilepsy." Epilepsia 37 Suppl 2: S1-S3.

133. Sitnikova, E., A. E. Hramov, A. A. Koronovsky and G. van Luijtelaar (2009).

Sleep spindles and spike-wave discharges in EEG: Their generic features,

similarities and distinctions disclosed with Fourier transform and continuous

wavelet analysis. J Neurosci Methods. Netherlands. 180: 304-316.

Page 167: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 145 -

134. Smith, A. L., P. M. Cordery and I. D. Thompson (1995). "Manufacture and

release characteristics of Elvax polymers containing glutamate receptor

antagonists." J Neurosci Methods 60(1-2): 211-217.

135. Sodemann, U., H. S. Moller, M. Blaabjerg and C. P. Beier (2014).

"Successful treatment of refractory absence status epilepticus with

lacosamide." J Neurol.

136. Swarztrauber, K., S. Dewar and J. Engel, Jr. (2003). "Patient attitudes about

treatments for intractable epilepsy." Epilepsy Behav 4(1): 19-25.

137. Szybala, C., E. M. Pritchard, T. A. Lusardi, T. Li, A. Wilz, D. L. Kaplan and

D. Boison (2009). "Antiepileptic effects of silk-polymer based adenosine

release in kindled rats." Exp Neurol 219(1): 126-135.

138. Tamargo, R. J., L. A. Rossell, E. H. Kossoff, B. M. Tyler, M. G. Ewend and

J. J. Aryanpur (2002). "The intracerebral administration of phenytoin using

controlled-release polymers reduces experimental seizures in rats." Epilepsy

Res 48(3): 145-155.

139. Taylor, C. P., T. Angelotti and E. Fauman (2007). "Pharmacology and

mechanism of action of pregabalin: the calcium channel alpha2-delta (alpha2-

delta) subunit as a target for antiepileptic drug discovery." Epilepsy Res

73(2): 137-150.

140. Tiwari, S. K., R. Tzezana, E. Zussman and S. S. Venkatraman (2010).

"Optimizing partition-controlled drug release from electrospun core-shell

fibers." Int J Pharm 392(1-2): 209-217.

141. Tolaymat, A., A. Nayak, J. D. Geyer, S. K. Geyer and P. R. Carney (2015).

"Diagnosis and Management of Childhood Epilepsy." Curr Probl Pediatr

Adolesc Health Care 45(1): 3-17.

142. Tsubouchi, K., Y. Ikematsu, M. Hashisako, E. Harada, H. Miyagi and N.

Fujisawa (2014). "Convulsive seizures with a therapeutic dose of isoniazid."

Intern Med 53(3): 239-242.

143. Uppalapati, D., B. J. Boyd, S. Garg, J. Travas-Sejdic and D. Svirskis (2016).

"Conducting polymers with defined micro- or nanostructures for drug

delivery." Biomaterials 111: 149-162.

Page 168: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 146 -

144. Urbach, H. (2013). MRI in epilepsy. [electronic resource], Berlin ; New York

: Springer, c2013.

145. Vajda, F. J. and M. J. Eadie (2014). "The clinical pharmacology of traditional

antiepileptic drugs." Epileptic Disord 16(4): 395-408.

146. Van Hese, P., J. P. Martens, L. Waterschoot, P. Boon and I. Lemahieu (2009).

"Automatic detection of spike and wave discharges in the EEG of genetic

absence epilepsy rats from Strasbourg." IEEE Trans Biomed Eng 56(3): 706-

717.

147. Vickery, P. B., E. E. Tillery and A. P. DeFalco (2017). "Intravenous

Carbamazepine for Adults With Seizures." Ann Pharmacother:

1060028017736785.

148. Viry, L., S. E. Moulton, T. Romeo, C. Suhr, D. Mawad, M. Cook and G. G.

Wallace (2012). "Emulsion-coaxial electrospinning: designing novel

architectures for sustained release of highly soluble low molecular weight

drugs." Journal of Materials Chemistry 22(22): 11347-11353.

149. Wallace, G. G. (2009). Conductive electroactive polymers : intelligent

polymer systems. Boca Raton, CRC Press.

150. Wallace, G. G. and G. G. Wallace (2003). Conductive electroactive polymers

: intelligent materials systems. Boca Raton, FL, CRC Press.

151. Wang, Y., W. Qu and S. H. Choi (2017). "DRUG DELIVERY FDA's

Regulatory Science Program for Generic PLA/PLGA-Based Drug Products."

Am pharmaceut rev 20(4): 52.

152. Wang, Y., X. Ying, L. Chen, Y. Liu, Y. Wang, J. Liang, C. Xu, Y. Guo, S.

Wang, W. Hu, Y. Du and Z. Chen (2016). "Electroresponsive Nanoparticles

Improve Antiseizure Effect of Phenytoin in Generalized Tonic-Clonic

Seizures." Neurotherapeutics 13(3): 603-613.

153. Wang, Z., C. Roberge, L. H. Dao, Y. Wan, G. Shi, M. Rouabhia, R. Guidoin

and Z. Zhang (2004). "In vivo evaluation of a novel electrically conductive

polypyrrole/poly(D,L-lactide) composite and polypyrrole-coated poly(D,L-

lactide-co-glycolide) membranes." J Biomed Mater Res A 70(1): 28-38.

Page 169: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 147 -

154. Wilson, B., Y. Lavanya, S. R. Priyadarshini, M. Ramasamy and J. L. Jenita

(2014). "Albumin nanoparticles for the delivery of gabapentin: preparation,

characterization and pharmacodynamic studies." Int J Pharm 473(1-2): 73-79.

155. Wilz, A., E. M. Pritchard, T. Li, J. Q. Lan, D. L. Kaplan and D. Boison

(2008). "Silk polymer-based adenosine release: therapeutic potential for

epilepsy." Biomaterials 29(26): 3609-3616.

156. Xie, Z. B.-D., G. (2009). "Electrospun Poly(D,L-lactide) Fibers for Drug

Delivery: The Influence of Cosolvent and the Mechanism of Drug Release. ."

J. Appl Polym. Sci. 115(1): 8.

157. Ying, X., Y. Wang, J. Liang, J. Yue, C. Xu, L. Lu, Z. Xu, J. Gao, Y. Du and

Z. Chen (2014). "Angiopep-conjugated electro-responsive hydrogel

nanoparticles: therapeutic potential for epilepsy." Angew Chem Int Ed Engl

53(46): 12436-12440.

158. Yu, K., J. Zhao, Z. Zhang, Y. Gao, Y. Zhou, L. Teng and Y. Li (2015).

"Enhanced delivery of Paclitaxel using electrostatically-conjugated

Herceptin-bearing PEI/PLGA nanoparticles against HER-positive breast

cancer cells." Int J Pharm.

159. Yurtdas Kirimlioglu, G., Y. Menceloglu, K. Erol and Y. Yazan (2016). "In

vitro/in vivo evaluation of gamma-aminobutyric acid-loadedN,N-

dimethylacrylamide-based pegylated polymeric nanoparticles for brain

delivery to treat epilepsy." J Microencapsul 33(7): 625-635.

160. Zeng, J., L. Yang, Q. Liang, X. Zhang, H. Guan, X. Xu, X. Chen and X. Jing

(2005). "Influence of the drug compatibility with polymer solution on the

release kinetics of electrospun fiber formulation." J Control Release 105(1-2):

43-51.

161. Zhang, F., Q. Song, X. Huang, F. Li, K. Wang, Y. Tang, C. Hou and H. Shen

(2015). "A Novel High Mechanical Property PLGA Composite Matrix

Loaded with Nanodiamond-Phospholipid Compound for Bone Tissue

Engineering." ACS Appl Mater Interfaces.

162. Zhong, Y. and R. V. Bellamkonda (2005). "Controlled release of anti-

inflammatory agent alpha-MSH from neural implants." J Control Release

106(3): 309-318.

Page 170: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 148 -

Appendices

Appendix 1– Updated Table 1.2 and Table 1.3

Table 1.2 and 1.3 of chapter 1 were updated to take into account the results presented in

chapter 4 and 5.

Page 171: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 149 -

Updated table 1. 2 Anti-epileptic drugs delivered using polymer-based devices (in vitro experiments).

Drug Polymer Device type In vitro release time Release profile Paper

Acetazolamide Eudragit Injectable 10 hours (EOE) however most within 4 hours Biphasic (Duarte, Roy et al. 2007)

Acetazolamide Eudragit Injectable 7 hrs (EOE) Linear (Haznedar and Dortunc 2004)

Adenosine EVA Implantable 17 days (EOE) however most within 4 days Triphasic (Boison, Scheurer et al. 1999)

Adenosine Silk Implantable 14 days (EOE) Biphasic (Wilz, Pritchard et al. 2008)

Adenosine Silk Implantable 14 days Linear (Pritchard, Szybala et al. 2010)

Adenosine Silk Implantable 21 days Triphasic (Szybala, Pritchard et al. 2009)

Carbamazepine Chitosan Injectable 3 hours (EOE) however most within 2 hours Linear (Gavini, Hegge et al. 2006)

Carbamazepine Chitosan, HPMC Injectable 5 hours (EOE) Biphasic (Filipovic-Grcic, Perissutti et al. 2003)

Carbamazepine Eudragit, HPMC Injectable Between 30 and 60 minutes Biphasic (Dong, Maincent et al. 2007)

Carbamazepine Gelucire Injectable 10 minutes (EOE) Linear (Passerini, Perissutti et al. 2002)

Carbamazepine PECA Injectable 4 hours (EOE) Biphasic (Fresta, Cavallaro et al. 1996)

Carbamazepine PLGA Injectable 24 hours (EOE) Biphasic (Barakat and Radwan 2006)

Carbamazepine Precifac Injectable 12 hrs (EOE) Biphasic (Barakat and Yassin 2006)

Carbamazepine PVP/PVA Injectable 1 hr (EOE) Linear (Patterson, James et al. 2008)

Clonazepam PCL/PEG Implantable Up to 250 days (EOE) Biphasic (Cho, Han et al. 1999)

Clonazepam PBLG/PEO Injectable 8 days (EOE) Linear (Jeong, Cheon et al. 1998)

Clonazepam CEC Injectable 100 hours (EOE) NA (Ryu, Jeong et al. 2000)

Clonazepam PLGA Injectable 24 hrs (EOE) Linear (Montanari, Cilurzo et al. 2001)

Clonazepam PLGA Injectable 8 to 9 days (EOE) Biphasic (Benelli, Conti et al. 1998)

Clonazepam PLGA Injectable 7 days (EOE) Biphasic (Nah, Paek et al. 1998)

CNF PLGA (microspheres) Injectable 70 days Biphasic (Nkansah, Tzeng et al. 2008)

CNF PLGA (nanospheres) Injectable Most after 14 days Triphasic (Nkansah, Tzeng et al. 2008)

Diazepam EVA Implantable 21 days (EOE) Linear (Haik-Creguer, Dunbar et al. 1998)

Diazepam PHBV (microspheres) Injectable 30 days (EOE) Triphasic (Chen and Davis 2002)

Diazepam (Diazepam/Gelatine) in PHBV microspheres

Injectable 30 days (EOE) Biphasic (Chen and Davis 2002)

Diazepam PHBV/Gelatine (microcapsules) Injectable 30 days (EOE) Linear (Chen and Davis 2002)

Diazepam PLA Injectable NA NA (Bodmeier and McGinity 1987)

Diazepam PDLLA Injectable 8 days, most after 1 day Biphasic (Giunchedi, Conti et al. 1998)

Diazepam PLGA Injectable Most within 10 h Linear (Bohrey, Chourasiya et al. 2016)

Ethosuxamide PECA Injectable 4 hrs (EOE), most after 3 hours Biphasic (Fresta, Cavallaro et al. 1996)

Ethosuxamide PLGA Injectable 24 hrs (EOE) Linear (Montanari, Cilurzo et al. 2001)

GABA EVA Implantable 24 to 72 hours Biphasic (Kokaia, Aebischer et al. 1994)

GABA PNP Injectable 6 hrs (EOE) Linear (Yurtdas Kirimlioglu, Menceloglu et

al. 2016)

Gabapentine Albumine nanoparticles coated with polysorbate

Injectable 24 hours (EOE) Biphasic (Wilson, Lavanya et al. 2014)

Page 172: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 150 -

Updated table 1.2. Anti-epileptic drugs delivered using polymer-based devices (in vitro experiments) (cont’d).

Drug Polymer Device type In vitro release time Release profile Paper

Lacosamide PLGA Injectable/

implantable Up to 250 hours (EOE) NA (Chen, Gu et al. 2017)

Lacosamide PLGA Implantable 100 days (EOE) however most within 60 days Multiphasic (Bauquier, Jiang et al. 2016)

Levetiracetam PLGA Implantable Most within 3 days NA (Halliday, Campbell et al. 2013)

Lorazepam PLGA Injectable 24 hours (EOE) Biphasic (Sharma, Maheshwari et al. 2014)

MK-801 EVA Implantable 60 days (EOE) however most within 30 days Biphasic (Smith, Cordery et al. 1995)

Muscimol LPSPs Injectable 5 days however most within 20 hours Biphasic (Kohane, Holmes et al. 2002)

Noradrenaline EVA Implantable Within 24 hours Biphasic (Kokaia, Aebischer et al. 1994)

Oxcarbazepine Chitosan Injectable NA NA (Rane, Mashru et al. 2007)

Phenobarbital PLGA Injectable NA NA (Barichello, Morishita et al. 1999)

Phenobarbitone PCL Injectable NA NA (Berrabah, Andre et al. 1994)

Phenytoin EVA Implantable 105 days (EOE) Biphasic (Tamargo, Rossell et al. 2002)

Phenytoin PCL Injectable 22 days (EOE) however mostly within 12 days Triphasic (Li, Li et al. 2007)

Phenytoin PECA Injectable 4 hrs (EOE) Biphasic (Fresta, Cavallaro et al. 1996)

Phenytoin PCL Injectable 30 days (EOE) however mostly within 4 days Triphasic (Jiang, Yue et al. 2015)

Phenytoin PLGA Implantable 20 days (EOE) Linear (Bauquier, PhD thesis)

Phenytoin ANG-ERHNPs Injectable 24 hours (EOE) Triphasic (Ying, Wang et al. 2014)

Phenytoin ANG-ERHNP Injectable 4 hours (EOE) (with electrical field stimulation) NA (Wang, Ying et al. 2016)

Pregabalin Alginate/HPMC Injectable 12 hours (EOE) Biphasic (Madan, Adokar et al. 2015)

Primidone PCL Injectable 10 hours (EOE) however most within 1 hours Triphasic (Ferranti, Marchais et al. 1999)

Valproate Hexadecanol Injectable 3 hours Biphasic (Giannola, De Caro et al. 1993)

Valproate PLGA Injectable NA NA (Barichello, Morishita et al. 1999)

ANG, Angiopep; CEC, Triblock copolymers synthesized by non-catalyzed ring opening polymerization of caprolactone in the presence of PEG; CNF, Ciliary

neurotrophic factor; EOE, End of experiment; ERHNPs, Electro-responsive hydrogel nanoparticles; EVA, Ethylene vinylacetate; GABA, Gamma amino butyric acid;

HPMC, Hydroxypropyl methylcellulose; LPSPs, Lipid protein sugar particles; MK-801, Methyl-lO,ll-dihydro-SH-dibenzo[a,d]cyclohepten-5,10-imine maleate; NA,

Not available; PBLG, Polybenzyl l-glutamate; PCL, polycaprolactone; PDLLA, Poly(D,L-Lactide); PECA, Polyethylcyanoacrylate; PEO, Poly(ethylene oxide); PEG,

Polyethyleneglycol; PHBV, Poly(3-hydroxybutyrate-co-hydroxyvalerate); PLA, Poly(dl-lactide); PLGA, Poly-lactide-co-glycolide; PNP, dimethylacrylamide-based

pegylated polymeric nanoparticles; PVA, Polyvinyl acetate ; PVP, Polyvinyl pyrrolidone ; SA, Sebacic acid.

Page 173: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 151 -

Updated Table 1. 3 Anti-epileptic drugs delivered using polymer-based devices (in vivo experiments).

Drug Polymer Device type Animal model Delivery In vivo results Paper

Adenosine EVA Implantable Kindled rats Rod, unilateral implantation in

ventricle 1 week seizure reduction (Boison, Scheurer et al. 1999)

Adenosine Silk Implantable Kindled rats Rod, unilateral implantation within the

infrahippocampal cleft

Dose-dependent delay in kindling acquisition for

11 days, and reduce after discharge duration (Wilz, Pritchard et al. 2008)

Adenosine Silk Implantable Kindled rats Rod, unilateral implantation within the

infrahippocampal cleft

Protection from generalized seizures over

a period of 10 days of suppressed seizures, delay

in epileptogenesis

(Szybala, Pritchard et al. 2009)

Carbamazepine Chitosan Injectable Sheep Powder Intranasal

Three hours increased drug concentration in the

serum when compared to the nasal administration

of the pure drug

(Gavini, Hegge et al. 2006)

Carbamazepine Eudragit, HPMC Injectable Rabbits Oral (powder in capsules) Ten hours increased plasma levels (Dong, Maincent et al. 2007)

Clonazepam PCL/PEG Implantable Rabbits IP pellet Constant plasma concentration of clonazepam for

45 days after initial burst effect. (Cho, Han et al. 1999)

GABA EVA Implantable Kindled rats

Bilateral implantation into the

mesencephalon dorsal to the substanti

nigra

At 48 h, generalized convulsions were prevented (Kokaia, Aebischer et al. 1994)

GABA PNP Injectable Rats, PTZ-induced acute

seizure model IP nanoparticules Shorter seizure and decrease in mortality

(Yurtdas Kirimlioglu,

Menceloglu et al. 2016)

Gabapentine

Albumin

nanoparticles

coated with

polysorbate

Injectable

Rats, PTZ and

electrochock induced

convulsion

IP nanoparticules Increase gabapentin brain concentration, decrease

duration of all phases of convulsion (Wilson, Lavanya et al. 2014)

Lacosamide PLGA Implantable GAERS Bilateral implantation above the motor

cortex Decrease in seizure activity for up to 7 weeks (Bauquier, Jiang et al. 2016)

Levetiracetam PLGA Implantable

Rats, ,hippocampal

tetanus toxin model of

mesial temporal lobe

epilepsy

Bilateral implantation above motor

and somatosensory cortices Trend towards a reduction in seizure frequency (Halliday, Campbell et al. 2013)

Lorazepam PLGA Injectable Rats Intranasal Increased brain concentration for up to 8 hours (Sharma, Maheshwari et al. 2014)

Lorazepam PLGA Injectable Sheep nasal mucosa

Ex vivo, biphasic controlled release 58% (24

hours) (Sharma, Maheshwari et al. 2014)

MK-801 EVA Implantable -

Implanted over brain area for 28 to 65

days followed by in vitro release

studies

Still releasing drug after 65 days in vivo (EOE).

The in vitro release profiles appears to predict

accurately the in vivo release.

(Smith, Cordery et al. 1995)

Page 174: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 152 -

Updated table 1.3 Anti-epileptic drugs delivered using polymer-based devices (in vivo experiments) (cont’d).

Drug Polymer Device type Animal model Delivery In vivo results Paper

Muscimol LPSPs Injectable Rats, hippocampal injection of

pilocarpine

Unilateral injection into the

hyppocampus (i.e. seizure focus)

Prevents seizures formation for up to 120 min

(EOE) (Kohane, Holmes et al. 2002)

Noradrenaline EVA Implantable Rats, interventricular treatment

with 6-hydroxydopamine

Bilateral implanation of matrices and

electrode in the hypocampus No change in kindling acquisition (Kokaia, Aebischer et al. 1994)

Phenytoin EVA Implantable Cobalt-induced rat model of

epilepsy

Unilateral implantation in cortical

seizure focus reduction in seizure activity for 6 days (EOE) (Tamargo, Rossell et al. 2002)

Phenytoin EVA Implantable Rat

Two weeks in vitro release study

following 365 days intracortical

implantation

NA (Tamargo, Rossell et al. 2002)

Phenytoin PCL Injectable Rats, hippocampal injection of

tetanus toxin

Unilateral implantation into cortical

seizure focus

Significant reduction in epileptic events for 3 days

with no observed clinical side effects. (Jiang, Yue et al. 2015)

Phenytoin PLGA Implanatble GAERS Bilateral implantation above the

motor cortex No change compare to blank polymer group (Bauquier, PhD Thesis)

Phenytoin ANG-ERHNPs Injectable Amygdala-kindled seizures in rats IP nanoparticles

PHT high concentrations in the hippo-campus,

amygdale, cerebellum, and brainstem regions,

decrease in the kindling-stimulation-induced

seizures

(Ying, Wang et al. 2014)

Phenytoin ANG-ERHNP Injectable

Mices, electrical- (maximal

electrical shock) and chemical-

induced (pentylenetetrazole and

pilocarpine) seizure models

IP nanoparticles

lowered the effective therapeutic doses of PHT

and demonstrated the improved anti-seizure

effects

(Wang, Ying et al. 2016)

Piperine Chitosan/STPP

NPs Injectable

Mices, PTZ-induced kindling

model IP nanoparticles

Enhance the neuroprotection and ameliorate the

astrocytes activation

(Anissian, Ghasemi-Kasman et

al. 2017)

TRH FAD/SA Implantable Kindled rats Unilateral implantation in amygdala Reduced seizure duration for 50-60 days (EOE) (Kubek, Liang et al. 1998)

Valproate PCL Implantable Rats, necocortical injected

tetanus toxin model

Implanted above the cortical seizure

focus

Decrease in epileptiform potential however the

model failed to induce consistent seizures. (Rassner, Hebel et al. 2015)

ANG, Angiopep; EOE, End of experiment; ERHNPs, Electro-responsive hydrogel nanoparticles; EVA, Ethylene vinylacetate; FAD, Fatty acid dimer; GABA,

Gamma amino butyric acid; GAERS:, Genetic absence epilepsy rats from Strasbourg; HPMC, Hydroxypropyl methylcellulose; IP, Intraperitoneal; LPSPs, Lipid

protein sugar particles; MK-801, Methyl-lO,ll-dihydro-SH-dibenzo[a,d]cyclohepten-5,10-imine maleate; NA, Not available; NPs, Nanoparticles; PCL,

polycaprolactone; PEG, Polyethyleneglycol; PLGA, Poly-lactide-co-glycolide; PNP, dimethylacrylamide-based pegylated polymeric nanoparticles; PTZ,

Pentylenetetrazole; SA, Sebacic acid; STTP, Sodium tripolyphosphate; TRH, Thyrotropin-releasing hormone.

Page 175: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 153 -

Appendix 2- In vivo evaluation of the anti-epileptic effects of laser cut

lacosamide loaded polymers implanted sub-durally in GAERS rats

Contribution

The work described in Appendix 2 was performed at the Centre for Clinical

Neurosciences and Neurological Research, (St. Vincent’s Hospital Melbourne, P.O.

Box 2900, Fitzroy, Victoria 3065, Australia). The polymers were provided by the

University of Wollongong and the in vivo experiments were performed by Sébastien

Bauquier.

Introduction

In chapter 5, the authors demonstrated a decrease in seizure activity as a result of the

subdural implantation of lacosamide loaded PLGA in GAERS. Though, the decrease

was associated with a significant debilitation of the rats during the post-operative

period.

Lacosamide-laden polymer mats were produced using a coaxial electrospinning method

with the core of the polymer composed of lacosamide 75:25 PLGA (lactide/glycolide =

75:25), and the shell composed of 85:15 PLGA (lactide/glycolide = 85:15). The shell

was designed to slow the release of the lacosamide from the core of the polymer.

Nevertheless, the PLGA implants were cut with scissors from the PLGA mats,

potentially cutting through the PLGA fibres leaving the core of the polymer open to

leakage. It was speculated that the high debilitation scores encountered could have been

the result of a significant initial leakage of lacosamide from the polymer.

We are now investigating the efficacy of a laser cut lacosamide loaded PLGA mats in

reducing seizure activity in GAERS. The hypothesis is that by laser cutting the polymer

and consequently sealing the cut fibres of the polymer implant, the more porous core of

the polymer would not be able to release as much lacosamide immediately after

polymer implantation.

Page 176: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 154 -

Material and methods:

There was no in vitro study performed. The in vivo experiment followed the same in

vivo experimental protocol than the one described in Chapter 5 except from the fact that

the polymer implants were cut using a laser (CO2 laser, Universal Laser System with

power output of 4.3 W). In brief, in this prospective randomized masked experiments

GAERS underwent surgery for implantation of skull electrodes (control group, n=6),

skull electrodes and blank polymers (blank polymer group, n=6), or skull electrodes and

lacosamide loaded polymers (lacosamide polymer group, n=6). Thirty-minute

electroencephalogram (EEG) recordings were started at day 7 post-surgery and

continued for eight weeks. EEGs were analysed using an automated detection

algorithm. The number of SWD, mean duration of one SWD and cumulative duration of

SWD were compared week-by-week between the lacosamide polymer group and both

the control and blank polymer groups using Kruskal-Wallis tests and when significant

values were found, post-hoc pairwise comparisons were conducted using the Mann-

Whitney U test. Significance level was set at 5%. In addition, for a minimum of three

days post-surgery and until full recovery from the surgery, the rats were monitored for

weight loss once a day, and for mobility and grooming twice a day. Those observations

were given scores (Table 5.1).

Results

The number of SWDs, the mean duration of SWDs and the total duration of SWDs

were not significantly different except during week one when they were all significantly

lower in the lacosamide polymer group compared to the control group (p= 0.01) (Figure

Appendix 2.1). All rats were not affected or middle affected by the surgery during the

recovery period except for one rat from group blank polymer which was moderately

affected at day 3.

Brief discussion

As it has been demonstrated in chapter 4, the craniotomy itself can be responsible for a

decrease in seizure activity for a couple of weeks following the surgery. In consequence

the decrease in seizure activity seen during the first week is not considered a positive

effect resulting from the administration of lacosamide.

Page 177: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 155 -

Figure Appendix 2.1. Comparison of the epileptic activity monitoring results

Comparison of the measurements obtained to evaluate the epileptic activity of groups control

(n=6), blank polymer (n=6) and lacosamide polymer (n=6). SWD: Spike and wave discharge;

IQR: Interquartile range; Results are reported as median. * represents the time point at which

the results from the lacosamide polymer group were significantly different to the control

group (p<0.01).

Page 178: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

- 156 -

The absence of significant decrease in seizure activity in the lacosamide group and of

the post-operative side effects encountered in chapter 5 questions the assumption that

appropriate lacosamide drug release occurred. Consequently, the author believe that the

high debilitation scores encountered in Chapter 5 could have been the result of a

significant initial leakage of lacosamide from the polymer although additional

experiments (such has in vitro release profile and scanning electron micrographs of the

edges of the implants) would need to be performed to confirm this.

Page 179: INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY …

Minerva Access is the Institutional Repository of The University of Melbourne

Author/s:

Bauquier, Sébastien Hyacinthe

Title:

Initial development of an implantable drug delivery device for the treatment of epilepsy

Date:

2018

Persistent Link:

http://hdl.handle.net/11343/214755

File Description:

Thesis_INITIAL DEVELOPMENT OF AN IMPLANTABLE DRUG DELIVERY DEVICE FOR

THE TREATMENT OF EPILEPSY

Terms and Conditions:

Terms and Conditions: Copyright in works deposited in Minerva Access is retained by the

copyright owner. The work may not be altered without permission from the copyright owner.

Readers may only download, print and save electronic copies of whole works for their own

personal non-commercial use. Any use that exceeds these limits requires permission from

the copyright owner. Attribution is essential when quoting or paraphrasing from these works.