Top Banner
Research Collection Doctoral Thesis Novel Applications of Metallic Nanoparticles for Cancer Treatments Author(s): Herzog, Antoine F. Publication Date: 2018 Permanent Link: https://doi.org/10.3929/ethz-b-000325544 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library
140

Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

Jul 03, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

Research Collection

Doctoral Thesis

Novel Applications of Metallic Nanoparticles for CancerTreatments

Author(s): Herzog, Antoine F.

Publication Date: 2018

Permanent Link: https://doi.org/10.3929/ethz-b-000325544

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Page 2: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

DISS. ETH No. 25466

NOVEL APPLICATIONS OF METALLIC

NANOPARTICLES FOR CANCER TREATMENTS

A thesis submitted to attain the degree of

DOCTOR OF SCIENCES of ETH Zurich

(Dr. sc. ETH Zurich)

presented by

ANTOINE FLORENT HERZOG

MSc. Chemistry, ETH Zürich

born on 10.09.1990

citizen of France,

accepted on the recommendation of

Prof. Dr. Wendelin J. Stark, examiner

Prof. Dr. Pablo Rivera-Fuentes, co-examiner

Prof. Dr. Beatrice Beck-Schimmer, co-examiner

2018

Page 3: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

2

To my family

Be the change that you wish to see in the world.

Mahatma Gandhi

Page 4: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

3

Acknowledgments

First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD thesis as well as

my master thesis and research projects. Wendelin is certainly a unique professor in his way of

thinking, conducting research and his ability to build a great team of people. As a result of his

influence and the ones of my colleagues, the time I spent at the Functional Materials Laboratory

changed me in many aspects with long-lasting effects. As caricatural as at it may sound, my

stay at FML opened my eyes on the endless opportunities life reserves and reinforced my will

not to be a passive witness of my life.

I would like to mention and thank all the group members from my years at the Functional

Materials Laboratory: Julian Koch, Konstantin Schulz-Schönhagen, Lukas Langenegger,

Michele Gregorini, Nadine Lobsiger, Nikita Kobert, Olivier Gröninger, Philipp Antkowiak,

Phillipe Bechtold, Simon Doswald, Urs Lustenberger, Weida Chen, Xavier Kohll, Dr. Carlos

Mora, Dr. Christoph Kellenberger, Dr. Christoph Schumacher, Dr. Corinne Hofer, Dr. Daniela

Paunsecu-Bluhm, Dr. Dirk Mohn, Dr. Elia Schneider, Dr. Gediminas Mikutis, Dr. Jonas Halter,

Dr. Mario Stucki, Dr. Martin Zeltner, Dr. Michael Loepfe, Dr. Michela Puddu, Dr. Nicholas

Cohrs, Dr. Oleksander Stepuk, Dr. Philipp Stoessel, Dr. Samuel Hess and Dr. Vladimir Zlateski.

I express my gratitude to Dr. Elia Schneider for his support regarding the synthesis of caged

gemcitabine and reviewing my manuscript, to Dr. Martin Zeltner for his extensive support for

the projects involving magnetic nanoparticles and to Dr. Carlos Mora for the supervision of one

of my research projects and my master thesis. I would especially like to thank them for the

always uncomplicated and pleasant collaborations. I would also like to thank Prof. Dr. Robert

Grass for always giving great advice whenever I would come with a question.

From the Institute for Chemical- and Bioengineering of ETH Zürich, I would like to thank

Bianca Wittmer, Luca Andres and Urs Krebs. I would like to give special thanks to Elias Halabi

for running HPLC-MS measurements when it was much needed and Andrin Bear for the nice

collaboration. From the University Hospital Zürich, I would like to thank Prof. Dr. Beatrice

Beck-Schimmer and Anja Zabel for the fruitful collaboration on the magnetic nanoparticles

projects.

Finally, I would like to thank my parents, my brother and my girlfriend. They are always here

for me, whenever I need them and I am very grateful for their support throughout all those

years.

Page 5: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

4

Page 6: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

5

Table of Contents

Acknowledgments 3

Résumé 11

Summary 14

1. Cancer: Origin, Burden and Therapy 18

1.1 Origin of Cancer and its Societal Impact ................................................................. 21

1.1.1 Cancer Biology and Aetiology ..................................................................... 21

1.1.1.1 Hallmarks of Cancer..................................................................................... 21

1.1.1.1.1 Hallmark 1: Sustaining Proliferative Signalling ................................................................... 22

1.1.1.1.2 Hallmark 2: Evading Growth Suppressors ........................................................................... 22

1.1.1.1.3 Hallmark 3: Activating Invasion and Metastasis .................................................................. 22

1.1.1.1.4 Hallmark 4: Enabling Replicative Immortality..................................................................... 23

1.1.1.1.5 Hallmark 5: Inducing Angiogenesis ..................................................................................... 23

1.1.1.1.6 Hallmark 6: Resisting Cell Death ......................................................................................... 23

1.1.1.2 Carcinogenesis ............................................................................................. 23

1.1.1.2.1 Genotoxic Carcinogenesis .................................................................................................... 24

1.1.1.2.1.1 Oxidative Damages ....................................................................................................... 24

1.1.1.2.1.2 Photolesions ................................................................................................................... 24

1.1.1.2.1.3 DNA Alkylation ............................................................................................................ 24

1.1.1.2.2 Non-genotoxic Carcinogenesis ............................................................................................. 25

1.1.1.2.2.1 Hormone-mediated Carcinogenesis ............................................................................... 25

1.1.1.2.2.2 Receptor-mediated Carcinogenesis ............................................................................... 25

1.1.1.2.2.3 Carcinogenesis Mediated by Epigenetic Changes ......................................................... 25

1.1.1.2.3 Viral carcinogenesis .............................................................................................................. 26

1.1.2 Current Cancer Treatments Strategies.......................................................... 26

1.1.2.1 Surgery ......................................................................................................... 26

1.1.2.2 Radiation Therapy ........................................................................................ 27

1.1.2.3 Chemotherapy .............................................................................................. 28

1.1.2.3.1 Alkylating Agents ................................................................................................................. 28

1.1.2.3.2 Antimetabolites ..................................................................................................................... 29

1.1.2.3.2.1 Antifolates ..................................................................................................................... 29

1.1.2.3.2.2 Base Analogues ............................................................................................................. 29

1.1.2.3.3 Anti-microtubules agents ...................................................................................................... 29

1.1.2.3.4 Topoisomerase inhibitors ...................................................................................................... 30

1.1.2.4 Targeted Therapy ......................................................................................... 30

Page 7: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

6

1.1.2.4.1 Tyrosine Kinase Inhibitors .................................................................................................... 30

1.1.2.4.2 Angiogenesis Inhibitors ........................................................................................................ 31

1.1.2.4.3 Epidermal Growth Factor Receptor (EGFR) Inhibitors........................................................ 31

1.1.3 Psychological Impact and Financial Burden of Cancer ............................... 31

1.2 Nanomedicine: a Widely Encompassing Concept ................................................... 32

1.2.1 Drug Delivery ............................................................................................... 32

1.2.2 Nanoparticles with a Therapeutic Action ..................................................... 33

1.3 Interactions between Nanoparticles and In Vivo Elements ..................................... 34

1.3.1 Interaction with Blood Components ............................................................ 34

1.3.1.1 Nanoparticle-Protein Interactions ................................................................ 34

1.3.1.2 Nanoparticle-Cell Interactions ..................................................................... 35

1.3.2 Interaction with Organs Influencing Circulation Time ................................ 36

1.3.2.1 Kidneys......................................................................................................... 36

1.3.2.2 Liver ............................................................................................................. 36

1.3.2.3 Spleen ........................................................................................................... 37

1.4 Conclusion ................................................................................................................ 37

2. Hydrogen as a Bio-orthogonal Trigger for Spatiotemporally Controlled Caged

Prodrug Activation 39

2.1 Introduction .............................................................................................................. 40

2.2 Experimental ............................................................................................................ 41

2.2.1 Synthesis of Caged Coumarin (Compound 2a) ............................................ 41

2.2.2 Synthesis of Caged Coumarin (Compound 1a) ............................................ 41

2.2.3 Screening of Catalyst Concentrations .......................................................... 42

2.2.4 Transmission Electron Microscopy Measurements ..................................... 42

2.2.5 Kinetics of Deprotection of Caged Coumarin .............................................. 42

2.2.6 Interval Kinetics of Deprotection of Caged Coumarin ................................ 43

2.2.7 EC50 Experiments ......................................................................................... 43

2.2.8 Extracellular Prodrug Activation in Cancer Cells ........................................ 44

2.2.9 Zeta-Potential Measurement ........................................................................ 44

2.3 Results and Discussion ............................................................................................. 45

2.3.1 EC50 Experiments ......................................................................................... 45

2.3.2 Kinetics of the Uncaging Reaction............................................................... 46

2.3.3 Toxicity of the Nanoparticles ....................................................................... 48

Page 8: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

7

2.3.4 Extracellular Caged Prodrug Activation ................................................. 49

2.4 Conclusion ................................................................................................................ 50

3. Preliminary Pre-Clinical Trials for the Gemcitabine Caged Prodrug System 52

3.1 Introduction .............................................................................................................. 53

3.2 Experimental ............................................................................................................ 54

3.2.1 Formulations Preparation ............................................................................. 54

3.2.2 Dynamic Light Scattering Measurements .................................................... 54

3.2.3 Nebulization Assays ..................................................................................... 54

3.2.4 Tolerability Profiles of Formulation 2 and of Platinum Nanoparticles

Solutions 55

3.2.4.1 Animals ........................................................................................................ 55

3.2.4.2 Administration Protocol ............................................................................... 55

3.2.4.3 Clinical Follow-up........................................................................................ 56

3.2.4.4 Terminal Procedures .................................................................................... 56

3.2.5 Feasibility Test in Xenograft Mice Model ................................................... 56

3.2.5.1 Animals ........................................................................................................ 56

3.2.5.2 Cells .............................................................................................................. 56

3.2.5.3 Preparation of Cells to Be Implanted ........................................................... 57

3.2.5.4 Implementation of Tumour Cells ................................................................. 57

3.2.5.5 Tumour Measurements and Clinical Follow-up........................................... 57

3.2.5.6 Administration Protocol ............................................................................... 57

3.2.5.7 Terminal Procedures .................................................................................... 58

3.2.5.8 Necropsy and Lung Collection ..................................................................... 58

3.3 Results and Discussion ............................................................................................. 58

3.3.1 Influence of Surfactant on the Average Droplet Size .................................. 58

3.3.2 Freeze-thaw Stability.................................................................................... 59

3.3.3 Nebulization Properties of the Selected Formulations ................................. 60

3.3.4 Tolerability Profile of Formulation 2 and Platinum Nanoparticles Solutions

61

3.3.5 Feasibility Test in Xenograft Mice Model ................................................... 62

3.4 Conclusion ................................................................................................................ 63

Page 9: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

8

4. Effective Tumour Cells Removal from Patient Blood using Magnetic Nanoparticles.

65

4.1 Introduction .............................................................................................................. 66

4.2 Materials and Methods ............................................................................................. 68

4.2.1 Synthesis of C/Co-PhEtOH .......................................................................... 68

4.2.2 Synthesis of C/Co-PhEtO-Na+ ...................................................................... 69

4.2.3 Synthesis of C/Co@polyglycidin ................................................................. 69

4.2.4 Synthesis of C/Co@polyglycidyl-COOH .................................................... 70

4.2.5 Synthesis of C/Co@polyglycidyl-COO-EpCAM and C/Co@polyglycidin-

COO-IgG 70

4.2.6 Characterization of Antifouling Efficiency .................................................. 71

4.2.7 Characterization of Separation Efficiency ................................................... 72

4.2.8 Cell Line Experiments .................................................................................. 72

4.2.9 Scanning Electron Microscopy of anti-EpCAM Nanoparticles incubated

with HT-29 Cells .............................................................................................................. 72

4.2.10 Influence of Magnetic Nanoparticles Treatment on Lymphocytes

Enumeration in Healthy Subjects Spiked Blood .............................................................. 72

4.2.11 Removal of Circulating Tumour Cells (CTC) from Healthy Subjects Spiked

Blood 73

4.2.12 Removal of Circulating Tumour Cells (CTCs) from Cancer Patients ......... 74

4.3 Results and Discussion ............................................................................................. 74

4.3.1 Technical Aspects of the Newly Designed Nanoparticles ........................... 74

4.3.2 First Tests in Human Blood from Healthy Subjects Spiked with Cancer

Cells 77

4.3.3 Specificity, Safety Aspects and Reproducibility .......................................... 77

4.3.4 First Tests with Blood Samples from Cancer Patients ................................. 80

4.4 Conclusion ................................................................................................................ 80

5. Functionalization Reaction Study and Lymphocytes’ Capture as a Demonstration

of Highly Abundant Cells Removal 82

5.1 Introduction .............................................................................................................. 83

5.2 Materials and Methods ............................................................................................. 85

5.2.1 Synthesis of C/Co-PhEtOH .......................................................................... 85

5.2.2 Synthesis of C/Co-PhEtO-Na+ ...................................................................... 85

Page 10: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

9

5.2.3 Example of a Standard Synthesis of C/Co@polyglycidin ........................... 86

5.2.4 Synthesis of C/Co@polyglycidyl-COOH .................................................... 86

5.2.5 Synthesis of C/Co@polyglycidyl-COO-Campath and C/Co@polyglycidin-

COO-IgG 86

5.2.6 Removal of Lymphocytes from Healthy Subjects ....................................... 87

5.2.7 Removal of Lymphocytes from Cancer Patients.......................................... 88

5.3 Results and Discussion ............................................................................................. 89

5.3.1 Influence of Various Reaction Parameters on Polymer Length ................... 89

5.3.2 Reproducibility of Polymerization Step ....................................................... 90

5.3.3 B- and T-Lymphocytes Removal from Healthy Subject Blood Samples .... 91

5.3.4 B- and T-Lymphocytes Removal from Cancer Patient Blood Samples ....... 92

5.4 Conclusion ................................................................................................................ 93

6. Conclusion and Outlook 95

A.1 Supplementary Information Chapter 2 ..................................................................... 98

Screening of Hydrogen Concentrations ........................................................................... 98

Screening of Oxygen Concentrations ............................................................................... 99

Toxicity Study of Platinum Nanoparticles and Hydrogen ............................................. 100

Kinetics of Deprotection of Caged Gemcitabine ........................................................... 101

Re-use of Platinum Nanoparticles after Recycling ........................................................ 101

Extracellular Prodrug Activation in Normal Cells ......................................................... 102

Screening of Catalyst Concentrations ............................................................................ 105

Screening of Hydrogen Concentrations ......................................................................... 106

Kinetic of the Uncaging of Caged Gemcitabine ............................................................ 107

Screening of Oxygen Concentrations ............................................................................. 108

Toxicity of Platinum Nanoparticles and Hydrogen ........................................................ 109

Extracellular Prodrug Activation in Normal Cells ......................................................... 110

Reuse of Platinum Nanoparticles after Recycling .......................................................... 111

A.2 Supplementary Information Chapter 3 ................................................................... 112

Macroscopic Observation of the Lungs after Tolerability Profile Experiments ............ 112

A.3 Supplementary Information Chapter 4 ................................................................... 113

Particle Size Distribution................................................................................................ 113

Elemental Analysis ......................................................................................................... 113

Page 11: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

10

Infrared Spectroscopy..................................................................................................... 113

Rotational Thromboelastometry Measurements ............................................................ 113

Removal Efficiency of pSPM-functinalized Nanoparticles ........................................... 114

Particles Size Distribution .............................................................................................. 114

Materials Reproducibility ............................................................................................... 115

Influence of Magnetic Nanoparticles Treatment on other Blood Cells.......................... 116

Rotational Thromboelastometry Measurements ............................................................ 116

Infrared Spectroscopy..................................................................................................... 117

A.4 Supplementary Information Chapter 5 ................................................................... 119

Elemental Analysis after the Various Synthesis Steps ................................................... 119

Calculations of the Number of Polymer Units (example for standard synthesis) .......... 119

7. References 120

8. Curriculum Vitae 136

Page 12: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

11

Résumé

Cette thèse décrit deux types de technologies, impliquant des nanoparticules et pouvant trouver

des applications dans le domaine du traitement du cancer. La première technologie est un

nouveau système de pré-médicament qui pourrait éventuellement réduire les effets secondaires

des chimiothérapies du cancer du poumon et/ou en augmenter l’efficacité. La deuxième

technologie implique un changement d’enduit (« coating ») par rapport à des nanoparticules

magnétiques précédemment développées. Les nouvelles nanoparticules permettent une

séparation spécifique et efficace de certaines cellules présentes dans le sang et pourraient être

utilisées pour ôter du sang les cellules tumorales circulantes, à l’origine des métastases, mais

également pour isoler les lymphocytes qui sont impliqués dans de nombreuses maladies comme

les lymphomes ou les maladies auto-immunes.

Le chapitre 1 consiste en une introduction générale aux domaines du cancer et de la

nanomédecine. Premièrement, des faits généraux et des définitions sont décrits. Ceci est suivi

par la présentation des traits caractéristiques (« hallmarks ») du cancer. Ils permettent

notamment de comprendre les différences centrales entre les cellules cancéreuses et normales.

Afin de mieux discerner les origines moléculaires du cancer, une introduction à la biologie de

la carcinogénèse est inclue et une vue d’ensemble des traitements principaux (chirurgie,

radiothérapie, chimiothérapie et thérapie ciblée) du cancer est établie. Aussi, un examen succin

des conséquences sociales et financières du cancer pour les patients et leurs familles est

introduit. Dans la seconde partie, une vue d’ensemble de la nanomédicine est présentée. Cela

inclue la description des systèmes classiques de nanoparticules par l’administration de

médicament (« drug delivery ») ainsi que différentes nanoparticules ayant des effets

thérapeutiques. Cela est complété par quelques éléments clés afin de comprendre les

interactions nanoparticules-protéines, nanoparticules-cellules ainsi que les interactions avec les

organes influençant le temps de circulation (par exemple les reins, le foie et la rate).

Le chapitre 2 présente le concept d'un nouveau système de pré-médicaments en cage. Il utilise

la gemcitabine, une chimiothérapie du cancer du poumon approuvée par la FDA, qui est « mise

en cage » par un groupe protecteur. La réaction de déprotection nécessite l'utilisation de

nanoparticules de platine et est provoquée par l'administration d'hydrogène. Sur une lignée de

cellules de cancer du poumon, il est démontré que le précurseur en cage est beaucoup moins

cytotoxique que son homologue non protégé, que la réaction de déprotection est rapide et peut

être contrôlée en manipulant l'approvisionnement en hydrogène. Dans une autre expérience in

Page 13: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

12

vitro utilisant une lignée cellulaire de cancer du poumon, il a été démontré que le pré-

médicament, une fois déprotégé, retrouve entièrement la toxicité initiale. Cette expérience est

répétée avec des cellules épithéliales pulmonaires normales où l'on observe que le pré-

médicament en cage n'est pas toxique. De plus, une expérience de recyclage du catalyseur est

effectuée, montrant que l'efficacité catalytique est largement conservée pendant au moins 10

cycles.

Au chapitre 3, des données pré-cliniques préliminaires sont rapportées pour le système de pré-

médicaments en cage. Le chapitre commence par une description du développement d'une

formulation d'émulsion nécessaire à l'administration intratrachéale. À savoir, l'influence de la

concentration en tensioactif sur la taille des gouttelettes et l'influence de diverses températures

de congélation sur la stabilité à la congélation / décongélation est divulguée. Une sélection de

5 formulations est testée pour la formation d'aérosols en utilisant un MicroSprayer. La

meilleure formulation est ensuite administrée une fois par semaine aux souris pendant un mois.

En outre, deux solutions de nanoparticules de platine sont administrées à des souris qui sont

ensuite suivies pendant un mois. Ni l'émulsion, ni les solutions de nanoparticules de platine ne

génèrent de toxicité. Des souris xénogreffées préalablement inoculées avec des cellules

cancéreuses du poumon exprimant la luciférase sont administrées avec l’émulsion (véhicule),

la gemcitabine en cage et la gemcitabine normale. La croissance tumorale semblait être la plus

lente pour la gemcitabine normale, mais la plupart des souris de ce groupe sont décédées de

causes non liées au cancer. Il est supposé que cela est dû à la toxicité de la gemcitabine envers

les cellules normales, mais cela devra être confirmée par les expériences histologiques et

immunohistochimiques qui sont actuellement en cours sur les poumons des souris sacrifiées.

Le chapitre 4 décrit des nanoparticules de cobalt recouvertes de carbone et fonctionnalisées

par une couche de polyglycidol conjuguées à des anticorps anti-EpCAM. Tout d'abord, il est

démontré qu'il existe une relation entre la séparabilité magnétique, les propriétés anti-fouling

et l'efficacité d'élimination des cellules tumorales circulantes (CTC). L'élimination des CTC est

testée avec du sang de personnes en bonne santé dans lequel des cellules cancéreuses sont

ajoutées. La reproductibilité de la synthèse des nanoparticules et de l'efficacité de l'élimination

est rapportée pour trois lots de nanoparticules indépendants. De telles nanoparticules

magnétiques démontrent une efficacité d'élimination élevée associée à une spécificité élevée

puisque les autres cellules sanguines, telles que les lymphocytes, restent intactes pendant la

procédure d'isolement magnétique des CTC. Le chapitre est complété par les données obtenues

sur des échantillons de patients atteints de différents cancers.

Page 14: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

13

Au chapitre 5, l’étape de polymérisation du processus de production des nanoparticules est

étudiée dans le cadre d’une anticipation de la mise à l’échelle du processus qui sera nécessaire

pour mener des essais sur des animaux. Pour illustrer la possibilité d'utiliser de telles

nanoparticules pour d'autres applications où des quantités plus importantes de cellules doivent

être isolées, un anticorps Campath a été conjugué au polyglycidol. Une procédure rappelant

celle utilisée pour l'élimination des CTC est utilisée pour éliminer les lymphocytes du sang des

patients sains et cancéreux. Cette preuve de concept est une première indication que les

nanoparticules magnétiques fonctionnalisées avec du polyglycidol pourraient être utilisées dans

des maladies impliquant les lymphocytes telles que les lymphomes ou les maladies auto-

immunes. Cependant, pour être plus pertinents sur le plan thérapeutique, des anticorps ciblant

uniquement des sous-types spécifiques de lymphocytes doivent être utilisés.

Le chapitre 6 est une conclusion générale de ce travail et offre une perspective sur les futures

recherches possibles à la fois pour le système de pré-médicaments en cage et les nanoparticules

magnétiques.

Page 15: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

14

Summary

This thesis describes two types of technologies, involving nanoparticles, that could have

applications in the field of cancer treatments. The first one is a new caged prodrug system that

could eventually result in reduced side effects for lung cancer chemotherapeutics and/or

increased efficacy. The second technology involves a coating change of previously developed

magnetic nanoparticles. The new nanoparticles allow the specific and efficient separation of

cells from whole blood and could find applications in the removal of circulating tumour cells

at the origin of metastasis, but also the isolation of lymphocytes that are involved in numerous

diseases such lymphomas and autoimmune diseases.

In Chapter 1, a general introduction to the field of cancer and nanomedicine. First, general

facts and definitions about cancer are reported. This is followed by presenting the hallmarks of

cancer, which help understand the central differences between cancer and normal cells. To

better discern the molecular origins of cancer, an introduction to the biology of carcinogenesis

is included. An overview of the main cancer treatments (surgery, radiotherapy, chemotherapy

and targeted therapy) is established. Also, a brief examination of the potential social and

financial consequences of cancer for patients and their family is reported. In the second part, an

overview of nanomedicine is presented. It includes a description of the classical types of drug

delivery systems and various nanoparticles having a therapeutic action. This is complemented

by some key elements to understand nanoparticle-protein interactions, nanoparticle-cell

interactions as well as interactions with organs influencing the circulation time (e.g. kidneys,

liver and spleen).

In Chapter 2 the concept of a new caged prodrug system is introduced. It uses gemcitabine, a

FDA-approved lung cancer chemotherapeutic, which is caged with a protecting group. The

uncaging reaction requires the use of platinum nanoparticles and is prompted upon

administration of hydrogen. On a lung cancer cell line, it is shown that the caged prodrug is

much less cytotoxic than its uncaged counterpart and that the uncaging reaction is rapid and can

be controlled by manipulating the hydrogen supply. In a further in vitro experiment using a lung

cancer cell line, it is demonstrated that the prodrug, once uncaged, fully recovers the initial

toxicity. This experiment is repeated with normal lung epithelial cells where it is observed that

the caged prodrug is not toxic. Additionally, a catalyst recycling is done showing that the

catalytic efficiency is largely retained for at least 10 cycles.

Page 16: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

15

In Chapter 3, preliminary pre-clinical data are reported for the caged prodrug system. The

chapter starts by a description of the development of an emulsion formulation that is needed for

intratracheal administration. Namely, the influence of the surfactant concentration on the

droplet size and the influence of various freezing temperatures on the freeze/thaw stability is

disclosed. A selection of 5 formulations is tested for aerosol formation by using a

MicroSprayer. The best formulation is weekly administered to mice over the time course of

one month. Also, two platinum nanoparticles solutions are acutely administered to mice which

are then followed for a month. Neither the emulsion formulation, nor the platinum nanoparticles

solutions generate toxicity. Xenograft mice previously inoculated with luciferase-expressing

lung cancer cells are administered with the emulsion formulation (vehicle), the caged

gemcitabine and the uncaged gemcitabine. The tumour growth seemed to be the slowest for the

uncaged gemcitabine but most mice from this group died from non-cancer related causes. It is

hypothesized that this is due to the toxicity of uncaged gemcitabine towards normal cells but it

will have to be confirmed by the histological and immunohistochemical experiments that are

currently being done on the lungs of the sacrificed mice.

In Chapter 4 polyglycidol-functionalized carbon-coated cobalt nanoparticles conjugated with

anti-EpCAM antibodies are described. First, it is shown that there is a relationship between the

magnetic separability, the antifouling properties and the circulating tumour cells (CTCs)

removal efficiency. The CTCs removal is tested by spiking healthy human blood with cancer

cells. The reproducibility of both the nanoparticles synthesis and removal efficiency is reported

for three independent nanoparticles batches. Such magnetic nanoparticles achieved a high

removal efficiency combined with high specificity since other blood cells such as lymphocytes

remain mainly untouched during magnetic CTCs isolation procedure. The chapter is augmented

by first cancer patients’ data for patients with different cancer types.

In Chapter 5 the polymerization step of the production process of the nanoparticles is

investigated in an anticipation of the process scale-up that will be required to conduct animal

trials. To illustrate the feasibility of using such nanoparticles for other applications where larger

quantities of cells need to be isolated, a Campath antibody was conjugated to the polyglycidol

coating. A procedure reminiscent of that used for CTCs removal is employed to eliminate

lymphocytes from the blood of both healthy and cancer patients. This proof-of-concept is a first

hint that the polyglycidol-functionalized magnetic nanoparticles could be used in

lymphocytes-involving diseases such a lymphomas or autoimmune diseases. However, to be

Page 17: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

16

more therapeutically relevant, alternative antibodies that target only specific lymphocytes

subtypes should be used.

Finally, Chapter 6 gives a general conclusion of this work and offers an outlook on possible

future investigations for both the caged prodrug system and the magnetic nanoparticles.

Page 18: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

17

Page 19: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

18

Cancer: Origin, Burden and Therapy

Page 20: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

19

According to the World Health Organization, cancer is the second leading cause of death

worldwide with around 8.8 million deaths being directly attributed to cancer in 2015.1 The

number of patients diagnosed with cancer is expected to increase to reach, in 2025, a staggering

worldwide yearly rate of 20 million people newly diagnosed with cancer; most people being

diagnosed in financially rich countries (Figure 1-1).2,3 Already in 2010, it was estimated that

the global economic cost of cancer was around US$ 1.16 trillion (i.e. over 2% of the global

GDP).2,3 Therefore, it is no understatement than saying that cancer is one of humanity’s most

pressing issues.

Figure 0-1. Incidence and mortality of cancer for the various geographical regions. Data are given per

100 000 people. Reproduced from Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C,

Rebelo M, Parkin DM, Forman D, Bray, F. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality

Worldwide: IARC CancerBase No. 11[Internet]. Lyon, France: International Agency for Research on

Cancer; 2013. Available from: http://globocan.iarc.fr, accessed on 19/08/2018.

Page 21: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

20

As a consequence of the importance it carries, significant research efforts have been undertaken

to better understand cancer. For instance, it is now known that the incidence of the various types

of cancers strongly depends on environmental and genetic factors, with over 90% of cancer

being attributed to environmental factors such as tobacco and alcohol consumption

(Figure 1-2).4,5 This implies that behavioural changes can strongly reduce the chances of

developing a cancer and also explains the differences in incidence for the various cancer types

at different regional locations.3

To better discern the reasons why cancer arises and how it can be defeated, the first part of this

introductory chapter consists in a brief overview of the biology and aetiology of cancer as well

as the main treatments available. In the second part, the main types of cancer nanomedicines

and their interactions with biological systems will be evoked.

Figure 0-2. The role of genes and environment in the development of cancer. A. The percentage

contribution of genetic and environmental factors to cancer. B. Family risk ratios for selected cancers.

The numbers represent familial risk ratios, defined as the risk to a given type of relative of an affected

individual divided by the population prevalence. C. Percentage contribution of each environmental

factor. Adapted from Pharmaceutical Research, Vol. 25, Anand et al., Cancer is a Preventable Disease

that Requires Major Lifestyle Changes, 2097-2116, Copyright (2008), with permission from Springer.

Page 22: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

21

1.1 Origin of Cancer and its Societal Impact

1.1.1 Cancer Biology and Aetiology

As defined by the American National Cancer Institute, cancer is a collection of diseases

involving abnormal and uncontrolled cell growth with the possibility for these cells to invade

surrounding tissues.6 Additionally, cancer is an ever-evolving disease, because cancer cells

have an intrinsic genetic instability due to higher replication errors rates as compared to normal

cells, that can ultimately lead to highly aggressive mutant cells.7 To rationalize and better

apprehend cancer, oncology researchers described specific hallmarks that define cancer. These

hallmarks are key to understand how cancer cells differ from normal cells. Hallmarks are also

useful to explain how many cancer therapies work as well as carcinogenesis. Hanahan and

Weinberg described 6 hallmarks (Figure 1-3) of cancer which are succinctly presented below.8,9

The two seminal papers they wrote cumulate over 50000 citations and de facto constitute the

prevailing literature authority and served as the basis for the next section.

1.1.1.1 Hallmarks of Cancer

Figure 0-3. Scheme summarizing all six hallmarks described by Hanahan and Weinberg. Reprinted

from Cell, Vol. 144, Hanahan and Weinberg, Hallmarks of Cancer: The Next Generation, 646-674,

Copyright (2011), with permission from Elsevier.

Page 23: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

22

1.1.1.1.1 Hallmark 1: Sustaining Proliferative Signalling

For normal cells to grow and divide, they need to be stimulated by growth factors. These growth

factors are typically proteins or hormones that act as signalling molecules between cells. Cancer

cells have the capacity to grow without such factors and thus enter the otherwise highly

controlled process of cell division.9 Cancer cells can achieve this via several mechanisms:

- Autocrine signalling, the expression of growth factors by the cell itself,

- Perpetual activation of pathways normally triggered by growth factor binding,

- Destruction of entities responsible of negative feedback controls.

1.1.1.1.2 Hallmark 2: Evading Growth Suppressors

To avoid the growth and division of abnormally functioning cells, tumour suppressor genes can

express growth suppressor proteins that act through several mechanisms to inhibit cell division

or trigger apoptosis. For instance, when cells are in contact with other cells, growth suppressor

proteins repress genes necessary for the cell cycle to continue. This inhibits cell division and

avoids forming cell clusters. Another example of the role of growth suppressor proteins is the

triggering apoptosis in the event of DNA damages that are beyond repair. In the case of cancer

cells, growth suppressors’ expression is altered resulting in either ineffective proteins or in an

insufficient number of proteins.

1.1.1.1.3 Hallmark 3: Activating Invasion and Metastasis

Most normal cells are confined to the tissue from which they originated. The localization of

such cells is maintained by cell-to-cell adhesion molecules (e.g. E-cadherin) and by attachment

to the extracellular matrix (ECM). In the case of cancer cells, some of them acquire traits that

make them able to detach from the tumour, find access to the circulatory system and eventually

later form metastases in other organs. To do so, several mechanisms have been hypothesized

but the main one is epithelial-mesenchymal transition (EMT).10–13 During the EMT, traits such

as the expression of transcription factors repressing the E-cadherin expression or the expression

of matrix-degrading enzymes are acquired thereby “freeing” the cell from its surrounding. Of

note, similar events can be caused by cells from the tumour microenvironment. For instance,

macrophages are able to release matrix-degrading enzymes.14 However, cells in the

mesenchymal state can detach but are not able to invade (i.e. to duplicate). To do so they must

convert back to an epithelial character via the reverse process called mesenchymal-epithelial

transition (MET).9 Only a fraction of cells are able to undergo this transition in the new tumour

Page 24: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

23

microenvironment where the tumour is located after extravasation from the circulatory system.

This explains why only a fraction of the disseminated cells can form macrometastases.15

1.1.1.1.4 Hallmark 4: Enabling Replicative Immortality

Normal cells only undergo a limited number of growth and division cycles. One well-known

factor controlling how many cycles a cell can go through is the length of telomeres. Telomeres

are DNA sequences found at the end of the DNA strand and, during each cycle of the DNA

replication, part of the telomere sequence is consumed. Cells stop growing and dividing once

the sequence is too short to protect the DNA strand. Most cancer cells have high expression of

telomerases, the DNA polymerases responsible of incorporating the telomere sequence,

activity, thereby making cells immortal.

1.1.1.1.5 Hallmark 5: Inducing Angiogenesis

Once adult, angiogenesis, the growth of new blood vessels, is stopped with the exception of

processes such as wound healing or the female cycling. Cancer tissues permanently grow and

require increasing supply of oxygen and nutrients. This growing demand can only be met by

producing new vasculature. Cancer cells thus influence angiogenesis by up-regulating

angiogenesis inducers (e.g. vascular endothelial growth factor or VEGF) and down-regulating

angiogenesis inhibitors (e.g. thrombospondin-1 or TSP-1).

1.1.1.1.6 Hallmark 6: Resisting Cell Death

Normal cells use apoptosis as a mean to avoid complications, for instance, after a cell is

damaged or infected. In other words, when a cell is not working normally, apoptosis is

triggered. The induction of apoptosis can be intracellular (intrinsic program) or extracellular

(extrinsic program). Both initiate a cascade of proteolysis by activating proteases called caspase

9 and caspase 8, respectively. These proteolysis cascades ultimately result into the disassembly

of the cell which is then consumed by neighbouring cells or phagocytes. Cancer cells can bypass

apoptosis by modifying abnormality sensors (e.g. p53). The abnormalities not being detected,

the apoptosis mechanisms are not triggered.

1.1.1.2 Carcinogenesis

Normal cells can transform into cancer cells via many mechanisms that can be classified into

two main categories: genotoxic (mutated DNA) or non-genotoxic carcinogenesis (non-mutated

DNA). Both categories involve the abnormal expression of proteins controlled by tumour

suppressor genes and oncogenes. To put it simply, proto-oncogenes are genes that, when not

properly functioning, cause cancer and are then called oncogenes. An example of a proto-

Page 25: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

24

oncogene is the myc gene that regulates the transcription of transcription factors that ultimately

induce cell proliferations. Tumour suppressor genes on the other hand are genes that express

proteins that prevent abnormal cell growth. A well-known example is the TP53 gene which

expresses p53, a protein that act as a DNA damage sensor and triggers cell cycle arrest or

apoptosis if necessary.

1.1.1.2.1 Genotoxic Carcinogenesis

In the case of genotoxic carcinogenesis, the abnormal protein expression is due to a DNA

mutation. The origins of such mutations are diverse but can be summarized as follows: DNA is

damaged and if the enzymes or enzyme complexes responsible of DNA repair mechanisms

cannot fix the DNA, the normal DNA polymerases are switched out and translesion (TLS) DNA

polymerases are used instead. Such TLS polymerases have a tolerance for DNA lesions and can

introduce DNA mutations. The mutations can either be single base changes or frame shift-

causing modifications such bases deletions or insertions.

1.1.1.2.1.1 Oxidative Damages

Reactive oxygen species, generated for instance by smoking or a chronic stressed state, can

oxidise the DNA bases at several locations. An example of such a damage is the formation of

8-Oxogunanine (8-oxoG) that leads to a transversion (GC base pair is replaced by AT). Indeed,

8-oxoG can form a base pair with adenine and during replication an adenine is introduced on

the complementary strand in fine resulting into a mutation. However, the base excision repair

(BER) mechanism can repair oxidative damages as well as alkylated bases and AP-sites.

1.1.1.2.1.2 Photolesions

DNA photolesions are a well-known consequence of prolonged sun exposure. For instance,

UV-B (280-315 nm) can generate a dimerization of two thymine nucleobases. The nucleotide

excision repair (NER) mechanism usually repairs such types of helix-distorting DNA lesions

(this also holds true for Pt-induced DNA crosslinks).

1.1.1.2.1.3 DNA Alkylation

DNA alkylation is due to xenobiotics (i.e. a substance normally not found within the organism)

and their potential for DNA damage is diverse among patients due to differences in metabolism.

For instance, aflatoxin, that can be found in contaminated grains or seeds, can be transformed

by cytochrome P450 into an epoxide able to react with DNA to form an adduct that can lead to

the promotion of depurination. Such DNA reactive epoxides can be quenched by glutathione in

a reaction catalysed by glutathione S-transferase (GST). As a consequence of GST

Page 26: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

25

polymorphisms, patients are at various cancer risks depending on how effective are their GST

at detoxificating xenobiotics. This type of mechanisms explains how genetic factors can

influence cancer risk.

1.1.1.2.2 Non-genotoxic Carcinogenesis

Non-genotoxic carcinogenesis does not involve DNA mutations directly. Instead, for receptor-

mediated and hormone-mediated carcinogenesis, the mitosis rate is increased thus correlating

with an increased rate of mistake introduction during DNA replication. This can then lead to

mutations. In the case of carcinogenesis originating from epigenetic changes, it is the DNA

transcription that is altered, not the DNA sequence itself.

1.1.1.2.2.1 Hormone-mediated Carcinogenesis

Hormones are a type of signalling molecules that can trigger signal transduction when binding

to receptors on the surface (non-genomic endocrine modulation) or inside cells (genomic

endocrine modulation). A classic example is the oestrogen receptor (ER). Upon genomic

activation, the ER translocates to the nucleus where it activates DNA transcription and leads to

proliferation. Upon non-genomic activation, the ERK and Akt signalling pathways are activated

thereby leading to the prevention of apoptosis. The ER can for instance be stimulated by

xenobiotics such as bisphenol A, phytoestrogens and pesticides (e.g. DDT).

1.1.1.2.2.2 Receptor-mediated Carcinogenesis

Similarly to hormone receptors, some other receptors can trigger signalling pathways that can

lead to tumour initiation or promotion by for instance inhibiting expression of key tumour

suppressor genes such as p53. A typical example is the aryl hydrocarbon receptor (AhR) which

can be activated upon binding of some aromatic hydrocarbons such as dioxin, a commercial

by-product that notably created a food scandal in Belgium in 1999 after being found in livestock

(e.g. chicken, pork).

1.1.1.2.2.3 Carcinogenesis Mediated by Epigenetic Changes

DNA transcription is partially controlled by DNA methylation and histone (i.e. proteins around

which DNA is coiled) modifications. DNA methylation is a natural process used to control

which part of the DNA a specific cell should express. As a result, when DNA is

hypomethylated, its transcription is activated and deactivated when hypermethylated. Histones

regulate chromatin condensation and the extent of condensation is controlled by the histone

acetyl transferase (DNA expansion) and the histone deacetylase (DNA condensation) as well

as by the level of methylation. The DNA methylation can obviously be influenced by some

Page 27: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

26

alkylating agents but also indirectly by heavy metals such as Cd, which interfere with DNA

methyltransferases (DNMT), the enzymes that regulate DNA methylation.

1.1.1.2.3 Viral carcinogenesis

12% of human cancer originate from viral infections.16 For instance, human papilloma virus

(HPV), Hepatitis B and Epstein-Barr virus (EBV) are all DNA viruses that, after infecting a

cell, transfer part of their DNA in the host cell DNA thereafter leading to cell proliferation.

Vaccines were developed for viruses such as HPV thereby preventing the apparition of various

cancers (e.g. cervical cancer, vaginal cancer).

1.1.2 Current Cancer Treatments Strategies

1.1.2.1 Surgery

The treatment of cancer originated with surgery. A papyrus dating back from ancient Egypt was

found already describing surgical procedures to remove breast tumours.17 However, for a long

time surgery was synonym of high risks and inconvenience until anaesthesia and the use of

carboxylic soap (antiseptic agent) appeared in 1846 and 1867, respectively. In the 1970s,

techniques such as computed tomography (CT) scanning allowed surgeons to evaluate more

precisely the tumours’ locations. Also, notably with the advent of minimally invasive

techniques such as laparoscopy (see Figure 1-4), mortality during surgical operations has

drastically fallen for many cancers18,19 and modern surgery now also aims at preserving the

form and function of the body parts being operated. Therefore, many techniques such as breast

reconstruction, breast preservation or endoprosthetic reconstruction have been developed.20,21

The positive impact of surgery on the disease course is nowadays further strengthened by

adjuvant therapy. That is, the use of radiotherapy and chemotherapy after surgery. A notable

case is breast-conservation surgery where radiotherapy is applied to maintain local control.22

More recently, neoadjuvant (i.e. the administration of adjuvant therapy before surgery) started

being used to facilitate surgery and/or allow surgery of previously inoperable tumours.23 In the

future, surgery efficiency might be further increased by intraoperative molecular imaging and

the refinement of robot-assisted surgery.24 However, an important drawback of surgery is its

Page 28: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

27

potential to release cancer cells in the patient’s systemic circulation thus presumably increasing

the risk of metastatic disease and relapse.25,26

1.1.2.2 Radiation Therapy

In the 1950s, linear accelerators (LINAC) started to be used in medical settings to treat cancer.

Upon exposition to radiations, radicals are formed (e.g. hydroxyl radicals from water and

peroxy radicals from molecular oxygen). Such radicals have the potential to damage DNA and

in the case of peroxy radicals for instance, these damages cannot be repaired and trigger

apoptosis. Nowadays, radiation therapy is frequently used as an adjuvant therapy and proved

to be an efficient tool for local control.27 Because the efficiency is influenced by the selection

and delineation of the target volume, new technologies combining radiotherapy and magnetic

resonance imaging (MRI) are being developed.28 Beyond the classical photon radiotherapy,

methods having the potential to be more specific using protons or heavier atoms have

flourished. Such methods have a reverse depth dose profile (see Figure 1-5) with most energy

deposited in the Bragg peak. By modifying the beam energy, the depth of the Bragg peak is

altered. Proton radiotherapy could therefore be used for cases where vital organs/tissues are

Figure 0-4. Schematic representation of a laparascopic setup. Image reproduced from the “Medical

gallery of Blausen Medical 2014”, WikiJournal of Medicine 1 (2).

Page 29: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

28

located near the tumour.27 However, both types of radiotherapies have several drawbacks. First,

there is an heterogeneity in the patients’ responses29 and there can be an increased risk of

developing a secondary cancer30. Also, geographical misses can occur due to many factors such

as weight loss (change in the radiation penetration), changes in tumour volume/location,

breathing cycles or organ filling.27

1.1.2.3 Chemotherapy

Chemotherapy is the use of mainly small cytotoxic molecules to target dividing cells. The

various classes of agents are affecting different processes necessary for cell division. Because

each dose of chemotherapy only kills a fraction of tumour cells, a certain number of treatment

cycles is necessary. Interestingly, already very early in the history of the development of

chemotherapy, it became clear that tumours have a propensity to develop resistance to

chemotherapeutics.31 It was later found that by combining chemotherapies, tumour cells

developed less resistance and this still forms the basis of current strategies in chemotherapy

treatments.31 Chemotherapy is mainly used as adjuvant therapy in combination with surgery for

instance or to treat patients with metastases. Due to its unspecific nature, chemotherapy has

consequent side-effects.

1.1.2.3.1 Alkylating Agents

Alkylating agents are small molecules that alkylate guanine bases of DNA. This alkylation leads

to DNA cross-linking which results into induction of apoptosis. Cancer cells tend to be more

sensitive to such type of agents because they tend to divide more rapidly than normal cells and

Figure 0-5. Depth dose profile of a photon versus proton beam. Reproduced from Nature Reviews

Cancer, Vol. 16, Baumann et al., Radiation oncology in the era of precise medicine, 234-249, Copyright

(2016), with permission from Nature.

Page 30: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

29

the DNA error-correcting mechanisms are less efficient than in normal cells. This type of

molecules marked the beginning of the development of chemotherapy. This after the discovery

in the 1940s that mustard gas victims had strong myelosuppression (i.e. a reduction in the

production of blood cells). In 1942, scientists at Yale University administered nitrogen mustard,

a molecule derived from mustard gas, to a non-Hodgkin’s lymphoma patient and observed a

brief remission.32 Molecules such as cyclophosphamide were then developed in order to

improve chemical stability and allow oral administration.31 Of note, a number of alkylating-like

agents such as cisplatin or carboplatin are still being widely used. They do not alkylate DNA

but coordinate to DNA and induce apoptosis.

1.1.2.3.2 Antimetabolites

Antimetabolites constitute a class of chemotherapeutics that work by preventing effective DNA

and RNA synthesis which in turn interferes with cell division and growth. Unlike alkylating

agents, such drugs only intervene in cells being in the S phase of the cell cycle when DNA is

being replicated. There are several subclasses of antimetabolite drugs and the two main ones

will be discussed.

1.1.2.3.2.1 Antifolates

The synthesis of thymidine requires tetrahydrofolic acid which is obtained by reduction of

dihydrofolic acid by dihydrofolate reductase (DHFR), itself synthesized by reduction of folic

acid. Antifolates drugs such as methotrexate block DHFR thereby preventing the production of

thymidines which in turn lack for DNA synthesis.

1.1.2.3.2.2 Base Analogues

Base analogues such as 5-fluorouracil or gemcitabine can work via different mechanisms. One

example of mechanism of action of gemcitabine is via its incorporation into DNA. This creates

an error that cannot be repaired by the base-excision repair mechanism. This irreparable error

leads to inhibition of DNA synthesis ultimately to cell death.33

1.1.2.3.3 Anti-microtubules agents

Microtubules are dynamic tubular structures composed of tubulins. Beyond forming part of the

cytoskeleton, they are involved in the cell division process during which they pull apart the

chromosomes. Anti-microtubules agents, or mitotic inhibitors, disrupt the structure of the

microtubules thereby triggering cell cycle arrest and ultimately apoptosis. There are two main

types of anti-microtubules agents, vinca alkaloids and taxanes. Vinca alkaloids are originally

obtained from a plant, Madagascar periwinkle. Belonging to vinca alkaloids, vincristine and

Page 31: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

30

vinblastine work by inhibiting the assembly of tubulin into microtubules. On the other hand,

taxanes such as paclitaxel and docetaxel act by preventing the microtubules disassembly which

is necessary for chromosomes to achieve their metaphase spindle configuration.

1.1.2.3.4 Topoisomerase inhibitors

Topoisomerases I and II are enzymes influencing the DNA topology. The action of these

enzymes is necessary to ensure successful replication and transcription. Two well-known anti-

cancer drugs, irinotecan and etoposide are topoisomerase I and II inhibitors, respectively.

Irinotecan works by intercalating between a DNA cleavage complex and topoisomerase I

thereby inactivating the enzyme. Etoposide forms a complex with topoisomerase II. This

prevents the re-ligation step and the DNA strand remains broken. Such type of errors ultimately

promote apoptosis.

1.1.2.4 Targeted Therapy

Targeted therapy focuses on molecular abnormalities specific to cancer to selectively prevent

the growth of cancer cells. Although it raised large expectations, high failure rates were

observed (drugs working in only 10-20% of patients) for the first generation targeted

therapies.34 This can be explained by a significant intra-tumour and inter-tumour

heterogeneity.35 To address this, came the need to define, via biomarkers, the molecular

subtypes of the various cancers. Another recurrent issue with targeted therapy is the propensity

of tumours to become resistant. This is most likely explained by the fact that a small proportion

of resistant cells are already present in the tumour at the beginning of the treatment and, under

the selective pressure of the therapy, these cells start taking over the other tumour cells.36

Moreover, it has been demonstrated that cells can transiently resist, thereby preventing total

eradication and indicating that a combination of drugs is needed.37

1.1.2.4.1 Tyrosine Kinase Inhibitors

Tyrosine kinases are responsible for the activation of many proteins by phosphorylation.

Tyrosine kinase inhibitors (TKIs) can work either by competing with adenosine triphosphate,

the substrate or both or act in an allosteric manner.38 Unlike classical chemotherapeutics, TKIs

do not kill cells but rather interfere in signalling pathways that are key to cancer growth. They

can, for instance, interfere with kinases that are constitutively activated in cancer cells which

are otherwise regulated in normal cells.39

Page 32: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

31

1.1.2.4.2 Angiogenesis Inhibitors

Tumours require the formation of new blood vessels (angiogenesis) to sustain their growth and

cannot grow to a size larger than 1-2 mm3 without angiogenesis.40 Therefore, by preventing

angiogenesis, drugs have the potential to contain tumour growth. Angiogenesis inhibitors use

various mechanisms but many target the vascular endothelial growth factor (VEGF); a key

signalling protein for angiogenesis.41

1.1.2.4.3 Epidermal Growth Factor Receptor (EGFR) Inhibitors

The epidermal growth factor receptor (EGFR) is an important transmembrane protein that is

activated by extracellular ligands. Upon activation of EGFR, a cascade of signals is triggered

in the cell leading to the activation of pathways resulting in DNA synthesis and cell

proliferation.42 In cancer cells, EGFR tend to be overexpressed or dysregulated and drugs have

been developed that are either directed to the EGFR extracellular domain or to its tyrosine

kinase domain.43

1.1.3 Psychological Impact and Financial Burden of Cancer

Beyond the physical impairments that can result from cancer treatment, the psychological and

social outcomes of cancer are central aspects that are often overlooked.44 For instance, cancer

is frequently associated with anxiety and depression that can be caused by the treatment (e.g.

altered body image), the release of chemicals (e.g. for pancreatic and lung cancers) or the

prolonged presence in a hospital setting.45 Moreover, patients report a decline in cognitive

functions such as memory and attention/concentration impairments as well as a reduced

processing speed.46 Reminiscences of such cognitive impairments can sometimes be observed

even years after treatment arrest.47 Also, due to the shock of cancer diagnosis and the resulting

mental stress, some patients contract post-traumatic stress disorders.48 A non-negligible fraction

of cancer patients report sexual dysfunction during or following treatment, a factor known to

potentially highly affect quality of life.49 Less expectedly, some patients declare having post-

traumatic positive outcomes and notably “improved relationships”, “better outlook on life” and

enhanced coping skills.44

Financial toxicity is described as an additional side-effect of cancer.50 This is especially in

countries like the U.S. where a significant fraction of the treatments costs are covered by the

patients or the patients’ families. This can lead to deleterious coping strategies such as the use

of family savings or patients that do not take all medications and skip medical appointments.51

Page 33: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

32

1.2 Nanomedicine: a Widely Encompassing Concept

Nanomedicine is a hard to define word given the fact that it englobes a wide variety of

technologies having not much more in common than a given size range. Indeed, cancer

nanomedicines englobe liposomes and polymeric micelles, belonging to the subfield of drug

delivery, but also, nanoparticles used for radiotherapy, chemodynamic therapy or magnetic

hyperthermia.

1.2.1 Drug Delivery

The field of nanomedicine is very largely dominated by drug delivery technologies. As early as

1995, a liposomal formulation was approved by the FDA for doxorubicin.52 The rationale

behind using nanoparticulate formulations is to improve the pharmacodynamics and

biodistribution.53 However, until recently no marketed liposomal therapeutic agent increased

the overall survival when compared to the non-liposomal parent drug.54 In 2017, a first example

of a liposome formulation improving overall survival was demonstrated for Vyxeos, liposome-

encapsulated daunorubicin and cytarabine to treat acute myeloid leukemia.55 Another important

category of nanoparticles used for drug delivery purposes are polymeric micelles. A wide

variety of polymers have been used by researchers. For instance poly(ethylene glycol),

polypeptides (e.g. poly(L-lysine) or poly(L-glutamic acid)), polyethylenimine or

poly(lactic-co-glycolic acid) all have been exploited in polymeric micelles.56–58

To improve the specificity of the drug delivery systems, stimuli-responsive nanocarriers have

been developed.59 Such nanocarriers can take advantage of the fact that the chemical

environment of tumours is different than in normal tissues and/or use an external stimulus to

trigger the release of the active entity. For instance, systems targeting the decreased pH and

more reducing properties of the TME were developed.60,61 Also, technologies based on external

stimuli such as heat, light or a magnetic field have flourished.59,62 Additionally, antibodies and

protein fragments have been conjugated on the surface of various drug delivery systems to

improve the cellular uptake selectivity (i.e. targeted delivery). However, clinical translation

turned out to be arduous. Indeed, large discrepancies between rodent and human models

(accumulation being much lower in humans than in rodents) were observed.63 Moreover, an

Page 34: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

33

improved internalization does not necessarily translate into improved clinical efficiency since

factors, other than just accumulation, might be limiting to the drug action.63 Despite the many

limitations, numerous nanoparticulate drug delivery systems are nowadays marketed or in late

clinical trials.53

1.2.2 Nanoparticles with a Therapeutic Action

Although more confidential than drug delivery, the subfield of nanomedicine using

nanoparticles to generate or improve a therapeutic action has witnessed a plethora of research

directions. For example, nanoparticles, especially gold nanoparticles, were used for their

radiosensitising potential. Such nanoparticles, via the emission of secondary electrons, can

increase free radicals production, thereby improving the efficiency of radiotherapy treatments.64

Nanoparticles used for magnetic hyperthermia constitute another broad field of research with

already at least one marketed product.53 Magnetic hyperthermia relies on the fact that magnetic

nanoparticles, such as iron oxide, when placed in a magnetic field dissipate heat and locally

increase the temperature. This may enhance the results of radio- and chemotherapy.65 More

recently, nanoparticles used for chemodynamic therapy emerged.66 Iron-based nanoparticles

were used to generate radicals via H2O2-dependent Fenton reaction, thereby exploiting the fact

that tumour cells are overproducing H2O2.67 Even though there is a wealthy literature of

nanomedicines having a therapeutic action, they had a marginal impact in the clinic as

compared to their drug delivery counterparts.

It is worth noting that the field of nanomedicine is ever-evolving as new knowledge is acquired

and an interesting perspective is the exploitation of synergistic interactions between

nanoparticles and biological entities. For instance, it has been observed that nanoparticles below

around 50 nm diameter tend to accumulate in the lymph nodes, a secondary immune system

organ where antigen-presenting cells, such as dendritic cells, present antigens to T-cells.68 To

take advantage of this, researchers developed nanoparticles that can deliver tumour antigens

directly to the dendritic cells which can in turn activate other leukocytes.69 Such type of

technologies might provide sturdy applications of nanoparticles by avoiding having to

outcompete the immune system which, as a result of evolution, has set up a profusion of

strategies to target nano- and microscale exogenous entities.

Page 35: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

34

1.3 Interactions between Nanoparticles and In Vivo Elements

1.3.1 Interaction with Blood Components

1.3.1.1 Nanoparticle-Protein Interactions

Proteins account for over 90% of the dry weight of human blood plasma and when entering

blood circulation nanoparticles are rapidly covered by proteins.70 This process, often named

biofouling, is largely described in the literature since over a decade.71,72 Akin to any adsorption

process, biofouling is determined by the frequency of protein collision as well as the adsorption

and desorption rates. Key equations describing this phenomenon are reported in the review from

Lane et al.70 A visual representation summarizing the principles of biofouling can be found on

Figure 1-6. Of note, the equilibrium dissociation constant (Kd) is defined as the ratio of the

desorption rate over the adsorption rate and the latter is also proportional to the frequency of

collision. Therefore, it can be predicted that proteins displaying a strong interaction and/or a

high frequency of collision will have a low Kd. That is, a low probability of desorbing from the

Figure 0-6. (a) Schematic illustration of the interparticle distance, d, defined as the distance from the

nanoparticle surface to the protein surface. (b) Plot of a typical potential energy curve between a

nanoparticle and a protein molecule. (c) Potential energy plot of a particle with a deep primary energy

well for adsorption and a small barrier for protein adsorption, leading to surface fouling that is

thermodynamically stable. (d) Potential energy plot of a nanoparticle with a small primary well and a

high-energy barrier for protein adsorption, leading to surface fouling that is thermodynamically

unstable. Reprinted from Annu. Rev. Phys. Chem., Vol. 66, Lane et al., Physical Chemistry of

Nanomedicine: Understanding the Complex Behaviors of Nanoparticles in Vivo, 521-547, Copyright

(2015), with permission from Annual Reviews.

Page 36: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

35

surface. This first layer defines the so-called hard corona (i.e. slowly exchanging proteins) in

opposition to the soft corona formed by proteins loosely bound to the hard corona.73,74 Since

the corona is what cells actually “see”, it is known that its composition influences the biological

course of the nanoparticles.75 In other terms, nanoparticles with different surface properties will

have different protein coronas and ultimately different interactions with the various types of

blood cells. For instance, if there are many opsonins in the corona of nanoparticles, they tend

to be rapidly eliminated by the immune system.70,75

1.3.1.2 Nanoparticle-Cell Interactions

Due to the lower concentration and larger size of cells, cell-nanoparticle interactions are

happening at slower timescales as compared to protein-nanoparticle interactions. Beyond the

classical types of intermolecular interactions responsible of the affinity between proteins and

nanoparticles, there are some cell-specific aspects to consider. First, in some cases the

nanoparticle-cell interaction will lead to cellular uptake; a feature typically wanted for drug

delivery applications. In order to be internalized, there are some active processes for which the

affinity of the nanoparticle for the cell receptors has to be high enough to overcome the

energetic cost of wrapping the cellular membrane around the nanoparticle (Figure 1-7).70

Alternatively, internalization can occur via diffusion through the lipid bilayer for nanoparticles

that are small enough and exhibit rather lipophilic properties. Interestingly, it is not fully known

whether, even in the case of targeted nanoparticles, the interaction between the cell and the

nanoparticles indeed occurs via the targeting protein present on the surface of the nanoparticles

are rather via the proteins present in the corona.70 Moreover, successful internalization does not

necessarily translate into effective therapeutic action. In fact it was found that many

nanoparticles are trapped into endocytic vesicles.76 This potentially explains why, despite

reaching more efficiently the required cells, most targeted drug delivery system did not result

in an improved overall survival when compared to their standard treatment counterpart.63

Page 37: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

36

1.3.2 Interaction with Organs Influencing Circulation Time

1.3.2.1 Kidneys

Blood is filtered by the kidneys through the glomerulus which is composed of three consecutive

elements with different filtration capacities. The first one, the endothelium, has fenestrae of

around 60 nm while the last element only lets entities with a small (<5-6 nm) hydrodynamic

diameters pass through.75,77 The entities that are not retained during glomerular filtration are

directly being reabsorbed into the blood circulation.

1.3.2.2 Liver

The liver receives blood from the gut (via the portal vein) and the heart (via the hepatic artery).

It has important immunological functions since this is where Kupffer cells, a type of

Figure 0-7. (a) Multivalent interactions, by forming multiple ligand-receptor bond pairs as opposed

to a single bond, significantly increase the affinity of nanoparticles to the cell membrane. (b)

Nanoparticles with multiple affinity ligands increase the flux of receptors toward the particle, leading

to more binding events to gain the energy required to wrap the membrane around the particle. (c) A

free nanoparticle with affinity ligands (1) can contact the cell surface at which it binds multiple cell

surface receptors to create energetically favorable conditions for membrane wrapping and

endocytosis (2 and 3). Reprinted from Annu. Rev. Phys. Chem., Vol. 66, Lane et al., Physical

Chemistry of Nanomedicine: Understanding the Complex Behaviors of Nanoparticles in Vivo, 521-

547, Copyright (2015), with permission from Annual Reviews.

Page 38: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

37

macrophage accounting for 80-90% of the total body macrophages, reside.75 Once internalized,

the nanoparticles are phagocytosed by the Kupffer cells. Additionally, the liver produces bile

and biliary excretion is the only way to excrete non-biodegradable nanoparticles which are

larger than 5-6 nm (smaller particles can be excreted by the kidneys).75

1.3.2.3 Spleen

The spleen is a lymphatic organ having roles in lymphocytes maturation as well as pathogens

filtration.75 The efferent lymph vessels connect the spleen to the lymphatic circulation. This has

direct consequences for nanoparticles that need to be administered more than once. Indeed, in

the case of prolonged retention in the spleen, B-cells trigger antibodies secretion which may

result in the opsonization of the nanoparticles at the second injection, ultimately leading to their

faster clearance.75 This resistance mechanism is called accelerated blood clearance (ABC) and

has been observed for different types of nanoparticles.78

1.4 Conclusion

In this first chapter we touched various aspects of cancers. In addition to contextualizing the

disease by describing how it affects patients and their families, we introduced key components

of the biology and aetiology of cancer. This helped us to understand the risks of contracting

cancer and how those risks can be alleviated. It also provided us with the possibility to better

capture how current treatments function. A brief overview of nanomedicines and of the

interactions between nanoparticles and various biological components was presented.

The next four chapters are dedicated to two technologies related to the field of nanomedicine;

albeit in a less classical way. Chapters 2 and 3 report a caged prodrugs system relying on

catalytic nanoparticles to convert a prodrug into its drug counterpart with the former focusing

on in vitro data and the latter on in vivo. Chapters 4 and 5 describe magnetic nanoparticles to

physically remove cells from the blood. In chapter 4, the nanoparticles are applied to remove

circulating tumour cells. Chapter 5 focuses on first aspects towards scaling up the production

in view of future pre-clinical trials and discloses the application of the nanoparticles to remove

lymphocytes as further illustration of the therapeutic potential of such magnetic particles.

Page 39: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

38

Page 40: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

39

Hydrogen as a Bio-orthogonal Trigger for

Spatiotemporally Controlled Caged Prodrug Activation

Manuscript published in part as:

A. F. Herzog, E. M. Schneider, W. J. Stark,

Helvetica Chimica Acta, 2018, 10.1002/hlca.201800134

Author contributions: concept/design by AFH and WJS. Experiments planned/conducted by

AFH with support of EMS. Data Analysis/interpretation by AFH and WJS. Drafting article:

AFH. Critical revision of article by AFH, EMS and WJS. Approval of article by AFH, EMS

and WJS.

Page 41: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

40

2.1 Introduction

In the last decades, extensive efforts have been made to improve the prognosis of lung

cancer. Indeed, lung cancer remains a staggering cause of human deaths and accounted for

around 1.6 millions of deaths in 2012 alone, thereby making it the leading cause of cancer

deaths worldwide.79 Stage IV patients, as well as patients having undergone surgery, are being

given chemotherapy; most of which are untargeted and have significant negative side effects.80–

82

To reduce the side effects of cancer treatments the field of caged prodrugs has emerged.83,84

The so-called cage is a chemical protecting group and the resulting caged prodrug is a less

potent version of its uncaged analogue. Unlike classical prodrugs, which are converted to the

drug under physiological conditions, caged prodrugs require the use of an exogenous catalyst

to remove the protecting group. Seminal works were based on ruthenium and palladium metals

to catalytically cleave allyl carbamates.85,86 Recently, gold was successfully utilized in vivo to

carry out similar allyl carbamate cleavages.87,88 Most of these bio-orthogonal bond cleavage

reactions rely on heterogeneous catalysis, thus possibly conferring spatial control. Within recent

months, ruthenium complexes have been utilized as caging agents rather than as catalysts. The

uncaging of the reported complexes was achieved upon light exposure.89,90 This strategy gives

a temporal control over the uncaging; however, it is limited by the shallow depth of light tissue

penetration.91 Consequently, applications further than cutaneous or peri-cutaneous applications

remain out of reach.

To combine the advantages of both research directions (heterogeneous catalytic uncaging

and light-triggered metal complexes decomposition) and to avoid the shortcomings of the latter,

we developed a catalytic uncaging prompted upon activation via an exogenous bio-orthogonal

trigger. Herein, we disclose an example of a temporally- and spatially-controlled

caging/uncaging bio-orthogonal system. The method uses platinum, a metallic catalyst that has

not been exploited so far in the domain of caged prodrugs. The protecting group p-

nitrobenzyloxycarbonyl (pNZ) is used as the cage while hydrogen serves as the trigger for the

uncaging (Figure 2-1A).

For this in vitro proof-of-concept, we focused upon one specific possible clinical application -

the treatment of lung cancer. Hence, the A549 cell line (human lung carcinoma) and

gemcitabine were selected. The latter being an antimetabolite which, in combination with

Page 42: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

41

carboplatin, form the gemcarbo regimen, a clinically used treatment for non-small cell lung

cancer.81 With the objective of a later in vivo application, hydrogen-containing gas mixtures

inspired by those encountered in deep diving breathing gas (e.g. hydrox composition: 96%

hydrogen and 4% oxygen) were employed.92 The nanoparticles used could be applied via an

aerosol (inhalation), thereby making the administration of the catalyst minimally invasive.93

This is in contrast to the injection required for most of the aforementioned systems. Moreover,

it has been reported that particles remained confined to the lungs of mice administered with

ultrafine particles aerosols.94–96 This should thus confer a spatial control to our system.

The absence of significant cytotoxicity of the materials and of hydrogen are here reported

for conditions relevant to the in vitro proof-of-concept of the working principle of our system.

2.2 Experimental

2.2.1 Synthesis of Caged Coumarin (Compound 2a)

7-amino-4-methylcoumarin (200 mg, 1.14 mmol) was dissolved in 5 mL of dry DCM under N2

atmosphere. Pyridine (100 µL, 1.24 mmol) was added and the solution was cooled to 0°C.

Subsequently, 4-Nitrobenzyl chloroformate (365 mg, 1.7 mmol) was added and the reaction

stirred overnight. 20 mL of 0.5 M HCl was added which lead to the precipitation of the product.

The precipitate was filtered off and washed using Et2O. After drying, 212 mg of product was

obtained (53% yield).

1H NMR (200 MHz, DMSO-d6) δ 10.45 (s, 2H), 8.50 (s, 1H), 8.36 – 8.22 (m, 4H), 7.73 (d, J

= 8.6 Hz, 6H), 7.57 (d, J = 2.0 Hz, 2H), 7.44 (dd, J = 8.7, 2.1 Hz, 2H), 6.26 (d, J = 1.4 Hz,

2H), 5.36 (s, 4H), 2.40 (d, J = 1.2 Hz, 6H). 13C NMR (50 MHz, DMSO-d6) δ 153.64, 124.12.

HRMS (m/z) [M + H]+ calculated for C18H14N2O6, 355.0930; found: 355.0923.

2.2.2 Synthesis of Caged Coumarin (Compound 1a)

Gemcitabine hydrochloride (299 mg, 1 mmol) was dissolved in dry DMA (4 mL) with NaHCO3

(252 mg, 3 mmol) under N2 atmosphere. The mixture was then cooled to 4°C in an ice bath. 4-

Nitrobenzyl chloroformate (324 mg, 1.5 mmol) was subsequently added to the mixture. The

mixture was stirred overnight at room temperature. The solution was filtered and the filtrate

was concentrated in vacuo. The filtrate was added in its liquid form to a silica column (66 g).

The column was run with 500 mL of a mixture of hexane/ethyl acetate (20:80), followed by

500 mL of ethyl acetate. After drying, 93 mg of product was obtained (21% yield).

Page 43: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

42

1H NMR (200 MHz, DMSO-d6) δ 11.15 (s, 1H), 8.26 (dd, J = 8.1, 4.2 Hz, 3H), 7.69 (d, J =

8.5 Hz, 2H), 7.09 (d, J = 7.6 Hz, 1H), 6.32 (d, J = 6.4 Hz, 1H), 6.17 (t, J = 7.5 Hz, 1H), 5.33

(d, J = 10.7 Hz, 3H), 4.19 (tt, J = 13.0, 7.4 Hz, 1H), 3.98 – 3.72 (m, 2H), 3.75 – 3.54 (m, 1H),

2.09 (s, 1H). 13C NMR (50 MHz, DMSO-d6) δ 147.59, 144.18, 128.73, 124.07, 59.25.

HRMS (m/z) [M + Na]+ calculated for C17H16F2N4O8, 465.0834; found: 465.0827.

2.2.3 Screening of Catalyst Concentrations

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum solution (125 ppm, 250 ppm or 500

ppm; 3 nm diameter; Sigma-Aldrich) was added and 10 L of Milli-Q water was added to

control wells. The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The

chamber was placed in the incubator, connected to a gas bottle outside the incubator and to an

outlet. The gas chamber was flushed for 0.5 min with hydrogen gas at 0.07 bar and closed. The

plate was incubated at 37°C for 0, 5, 10, 15, 20, 25, 30, 40, 50, 60, 80, 100,120 min in the gas

chamber. After each time point, the plate was taken out of the gas chamber and fluorescence

emission was measured using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em:

460 nm). Values were normalized by the mean value of the positive controls wells.

2.2.4 Transmission Electron Microscopy Measurements

An aqueous solution of platinum nanoparticles (3 nm diameter, Sigma-Aldrich) was prepared

at a concentration of 5 ppm. 1.5 µL of the solution was deposited on carbon-coated copper

grids. After overnight drying, the samples were measured on a Tecnai F30 (FEI, 300 kV).

2.2.5 Kinetics of Deprotection of Caged Coumarin

In a 96-well plate, wells were filled with 90 L of fresh MEM or F-12K medium containing

caged 7-amino-4-methylcoumarin (110 M) and DMSO (2% v/v). All cell culture media were

supplemented with 10% Foetal Bovine Serum. To treated wells, 10 L of a platinum

nanoparticles solution (1000 ppm, 3 nm diameter, Sigma-Aldrich) was added and 10 L of

Milli-Q water was added to control wells. The plate was placed in a custom-designed gas

chamber (see Scheme A1-1). The chamber was placed in the incubator, connected to a gas

bottle outside the incubator and to an outlet. The gas chamber was flushed for 0.5 min with

Page 44: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

43

hydrogen gas at 0.07 bar and closed. The plate was incubated at 37°C for 0, 5, 10, 15, 20, 25,

30, 100 min in the gas chamber. After each time point, the plate was taken out of the gas

chamber and fluorescence emission was measured using a microplate reader (Tecan, Infinite

200 PRO; ex: 340 nm; em: 460 nm). Values were normalized by the mean value of the positive

controls wells.

2.2.6 Interval Kinetics of Deprotection of Caged Coumarin

In a 96-well plate, wells were filled with 100 L of fresh MEM medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). All cell culture media were

supplemented with 10% Foetal Bovine Serum. To treated wells, 10 L of a platinum

nanoparticles solution (250 ppm or 500 ppm, 3 nm diameter, Sigma-Aldrich) was added and 10

L of Milli-Q water was added to control wells. The plate was placed in a custom-designed gas

chamber (see Scheme A1-1). The chamber was placed in the incubator, connected to a gas

bottle outside the incubator and to an outlet. The gas chamber was flushed for 0.5 min with

hydrogen gas at 0.07 bar and closed. The plate was incubated at 37°C for 0.5 min in the gas

chamber. The plate was taken out of the gas chamber and left open for 2 min in order to

equilibrate with the ambient atmosphere of the room. Fluorescence emission was measured

using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). The plate was

placed back in the incubator for 2 min before measuring fluorescence again. Four such cycles

were made, followed by two cycles with 1 min incubation in the gas chamber and 2 min of

incubation directly in the incubator. Two last cycles were done with 4 min incubation with

hydrogen and 2 and 4 min, respectively, of incubation directly in the incubator. Values were

normalized by the mean value of the positive controls wells.

2.2.7 EC50 Experiments

A549 cells were seeded in 96-well plates at a concentration of 2,000 cells/well and incubated

overnight at 37°C before treatment. The supernatant of each well was then replaced with 100

L of fresh F-12K medium containing protected gemcitabine (concentration ranging from 104

M to 10-4 M) and DMSO (2% v/v), control wells were replaced with 100 L of fresh F-12K

medium and DMSO (2% v/v). All cell culture media were supplemented with 10% Foetal

Bovine Serum. Five biological replicates for each condition were included in the plate. Control

and treated plates were incubated at 37°C for 72 h before PrestoBlue™ cell viability reagent

(10% v/v) was added to each well and the plate incubated at 37°C for 2 h. Fluorescence emission

Page 45: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

44

was measured using a microplate reader (Tecan, Infinite 200 PRO; ex: 560 nm; em: 590 nm).

Values were normalized by the mean value of the positive controls wells.

2.2.8 Extracellular Prodrug Activation in Cancer Cells

A549 cells were seeded in 96-well plates at a concentration of 4,000 cells/well and incubated

overnight at 37°C before treatment. The supernatant of each well was then replaced with 100

L of fresh F-12K medium containing protected gemcitabine or gemcitabine (0.05 M) and

DMSO (2% v/v), control wells were replaced with 100 L of fresh F-12K medium and DMSO

(2% v/v). All cell culture media were supplemented with 10% Foetal Bovine Serum. Plates

were incubated at 37°C for 1 h before 10 L of a platinum nanoparticles solution (100 ppm, 3

nm diameter, Sigma-Aldrich) was added to each well and 10 L of Milli-Q water was added to

control wells. Six biological replicates for each condition were included in the plates. Plates

were incubated at 37°C for 1 h before placing them in a custom-designed gas chamber (see

Scheme A1-1). Control plates were left untouched in the incubator. The chamber was placed in

the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas chamber

was flushed for 1 min with hydrogen gas at 0.07 bar and closed. Plates were incubated at 37°C

for 2 h in the gas chamber before placing them back in the incubator with the control plates.

Control and treated plates were incubated at 37°C for 72 h before PrestoBlue™ cell viability

reagent (10% v/v) was added to each well and the plate incubated at 37°C for 2 h. Fluorescence

emission was measured using a microplate reader (Tecan, Infinite 200 PRO; ex: 560 nm; em:

590 nm). Values were normalized by the mean value of the positive controls wells.

2.2.9 Zeta-Potential Measurement

100 µL of a platinum nanoparticles solution (1000 ppm, 3 nm diameter, Sigma-Aldrich) was

added to a 1.5 mL Eppendorf tube containing 1000 µL of 0.9% saline solution. The Eppendorf

tube was centrifuged for 1.5h at 22130g (Eppendorf, Centrifuge 5424 R). The supernatant was

discarded and replaced by 1000 µL saline solution. The nanoparticles were redispersed with an

ultrasonic horn (Hielsher, UP50H; 30 kHz) before being transferred to a Disposable Capillary

Cell (DTS1070, Malvern) and measured using a Zetasizer Nano (Malvern).

Page 46: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

45

2.3 Results and Discussion

2.3.1 EC50 Experiments

Inspired by research in the field of solid-phase peptide synthesis, the caged prodrug was

generated by introducing a p-nitrobenzyloxycarbonyl protecting group via chloroformate

chemistry (Figure 2-1A and Supporting Information).97–99 Both molecules were dissolved in

DMSO and then diluted in F-12K medium, which was used as the cell culture medium for the

cancer cells. The dose-response behaviour of both the uncaged and caged gemcitabine were

investigated using the A549 cell line. After 72h of incubation at 37°C, viability was evaluated

Figure 0-1 A) Caging and uncaging reactions for gemcitabine and 7-amino-4-methylcoumarin (model

compound). In their uncaged forms, gemcitabine and 7-amino-4-methylcoumarin are active and

fluorescent, respectively. Once caged the former is inactive and the latter is not fluorescent. Upon

uncaging with platinum nanoparticles and hydrogen, they return to their initial state. B) Transmission

electron micrographs of the platinum nanoparticles. The red symbols delimit the distance between two

crystalline layers. C) Photographs of reaction mixtures under a 360 nm UV-lamp. The cuvettes 1-4

correspond to the four possible reaction conditions (+/- hydrogen and +/- Pt nanoparticles). When both

hydrogen and Pt nanoparticles are present, the fluorescence is restored to a level similar to that of the

control with 7-amino-4-methylcoumarin.

Page 47: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

46

using a PrestoBlue™ assay and a plate reader (Figure 2-2). As anticipated, gemcitabine and its

caged counterpart cause notably different responses. The caged prodrug is much less cytotoxic

than gemcitabine with an EC50 difference spanning over several orders of magnitude.

Gemcitabine is fully active in a sub-micromolar range while such a level of cytotoxicity cannot

be reached, even at over millimolar concentrations, for the caged molecule.

2.3.2 Kinetics of the Uncaging Reaction

Having confirmed the first hypothesis of our system, the catalysis of the drug activation

was studied. This was achieved with a model reaction using the fluorophore 7-amino-4-

methylcoumarin as a drug surrogate (Figure 2-1A). The fluorescence of the caged coumarin is

quenched upon protection of the amino group. The fluorescence is recovered upon uncaging

(Figure 2-1C). For our application, small nanoparticles exhibiting a high surface-to-volume

ratio and a potential ability to cross the cellular membrane are needed. Therefore, small (3 nm

diameter) platinum nanoparticles were used as catalysts. To fully characterize the reaction,

catalyst concentrations ranging from 12.5 to 50 ppm were screened (Figure A1-1) and gas

mixtures containing various hydrogen levels were investigated (Figure A1-2). Kinetic

Figure 0-2. Dose-response curve of gemcitabine (1a) and caged gemcitabine (1b). A A549 human lung

adenocarcinoma cell line was used. Solutions were prepared in Ham’s F-12K (Kaighn’s) Medium (F-

12K). After 72h of incubation at 37°C, the viability was evaluated using the PrestoBlue™ assay and a

plate reader (n = 5; 𝜆ex/em = 560/590 nm).

Page 48: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

47

measurements were done in two frequently encountered cell culture media; namely, F-12K and

MEM. A custom-designed gas chamber accommodating 96-well plates was built (Scheme A1-

1); this allowed the concomitant use, within a standard incubator, of both non-standard gas

mixtures (containing from 3.2% to 100% of hydrogen) and of standard incubator atmosphere

(air supplemented with 5% CO2). The reactions were conducted without stirring or bubbling

and the hydrogen supply was thus diffusion-controlled. The rapid reaction rates found (τ1/2 <

10 min, Figure 2-3) make this reaction propitious for our application. To ensure that the kinetic

of the uncaging of caged gemcitabine remains akin to that of the caged fluorophore, further

kinetic experiments were carried out with caged gemcitabine. Caged prodrug to drug

conversion was followed by HPLC-MS and a comparable reaction half-life was observed

(Figure A1-3).

To investigate the hypothesis of using hydrogen as an on/off switch, an interval kinetics

experiment was designed (Figure 2-4). The model reaction with 7-amino-4-methylcoumarin

was used and after each hydrogen interval, the plate was left open at ambient conditions to let

it equilibrate before measuring fluorescence intensity. This alternative regimen is reminiscent

to that of a breathing cycle with normal air, and one cycle with a gas mixture containing

hydrogen. Intervals of 30 seconds incubation are enough to trigger the uncaging and thus

Figure 0-3. Kinetics of the uncaging of caged prodrug surrogate 2b. Solutions of 99 µM of caged

prodrug surrogate 2b were prepared in F-12K and Minimum Essential Medium Eagle (MEM) cell

media. A concentration of platinum nanoparticles of 100 ppm and pure hydrogen were employed.

Page 49: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

48

support the idea of breathing two alternating gas mixtures. The reaction is virtually stopped

during the hydrogen-free intervals.

2.3.3 Toxicity of the Nanoparticles

It is well known that the tumour microenvironment is hypoxic and patients median percentage

of oxygen in lung tumours have been reported between 1.9 and 2.2% while normal tissues had

an average of 5.6%.100 The influence of the oxygen concentration on the uncaging reaction was

studied (Figure A1-4). The presence of oxygen decreases both the reaction yield and kinetics.

Such a phenomenon could thus be at the origin of a partial selectivity between cancer and

normal tissues because of the lower oxygen tension in cancer tissues. The toxicity of the

platinum nanoparticles and hydrogen were investigated (Figure A1-5). As expected, the cells

treated with hydrogen display a biologically insignificant decrease in viability. Cells were

treated with three different platinum nanoparticles concentrations. Interestingly, cells treated

with the two lowest platinum concentrations exhibit a small increase in viability. This supports

the idea of a beneficial reducing environment in the presence of Pt nanoparticles.94,101–104

Additionally, the zeta-potential of the Pt nanoparticles measured in physiological saline

solution was -0.08±0.27 mV. According to the results of Cho et al. such nanoparticles should

Figure 0-4. Interval kinetics of the uncaging of caged prodrug surrogate 2b. A solution of 99 µM of

caged prodrug surrogate 2b was prepared in F-12K medium. For both experiments a plate reader was

used (n = 5; 𝜆ex/em = 340/460 nm).

Page 50: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

49

thus be non-hemolytic and have a low acute lung inflammogenic potential.105 It also

corroborates the results from Onizawa et al. who found no pathological changes in mice that

were intranasally administered with platinum nanoparticles.94 Having proved that hydrogen

supply can act as an on/off switch and that neither hydrogen, nor platinum are toxic in vitro at

the required concentrations, we combined all these elements into a single experiment.

2.3.4 Extracellular Caged Prodrug Activation

To prove that the activity is fully restored after the uncaging, an experiment was designed

where cells were administered with caged gemcitabine and were treated with a 2x2 matrix of

conditions: presence/absence of hydrogen and presence/absence of a catalyst. Positive controls

were performed with Milli-Q water and negative controls with uncaged gemcitabine. Two

plates were made and incubated: one within the hydrogen-containing gas chamber and the other

freely standing in the incubator. After 2h, the plate was taken out and both plates were incubated

for 72h before a PrestoBlue™ assay was used to evaluate the viability (Figure 2-5). The addition

of caged gemcitabine slightly lowers the viability compared to the positive control with water.

In a similar experiment with non-cancerous lung cells (Figure A1-6), this was not observed.

The difference could potentially be explained by a constitutive reduction of the nitro moiety of

the protecting group by cancer cells.106,107 Interestingly, the cells treated with caged gemcitabine

where platinum and hydrogen were applied exhibited a statistically significant lower viability

compared to the gemcitabine control (p-value < 0.01, assumption of homoscedascity, two-tailed

unpaired t-test). This may be caused by an improved cellular uptake of the caged gemcitabine,

due to the increased lipophilicity compared to its uncaged counterpart, followed by an

intracellular uncaging. It has been shown on mice that using elevated gemcitabine

concentrations lead to a more marked tumour growth inhibition; however, this was at the

expense of considerable side effects.108 We believe that through use of our system, this could

be alleviated. Indeed, the observation that at equimolar concentrations the drug activity is fully

recovered upon uncaging, suggests that higher drug doses could be used while keeping side

effects to a level similar to that encountered today.

Page 51: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

50

2.4 Conclusion

In summary, we have shown in vitro the advantageous application of hydrogen gas for

selective uncaging of a drug thus conveying spatiotemporal control. The fast kinetics enable

the fascinating use of hydrogen as an on/off switch locally triggering the bio-orthogonal bond

cleavage reaction. The catalyst can be administered in a minimally invasive manner

(inhalation). The toxicity of here used hydrogen and platinum particles concentrations under

realistic conditions has been reported as non-inhibiting such applications. Overall, the system

described expands upon the growing repertoire of bond cleavage reactions in medicinal

chemistry. These early results only constitute a preliminary step and need to be followed by

studies in higher level models such as in xenografted mice. However, if similar results are

obtained in vivo, we believe this system could provide a strategy to reduce side effects,

particularly for lung cancer chemotherapeutics.

Figure 0-5. Extracellular caged prodrug activation. A A549 human lung adenocarcinoma cell line was

used. Solutions of gemcitabine (1a) and caged gemcitabine (1b) were prepared at a concentration of

0.05 µM in the F-12K cell medium. A concentration of platinum nanoparticles of 10 ppm was used. A

custom-designed gas chamber was used for incubation with hydrogen (2h). After 72h of incubation in

normal cell culture atmosphere, the viability was evaluated using the PrestoBlue™ assay and a plate

reader (n = 6; 𝜆ex/em = 560/590 nm). ٭٭٭ ,٭٭ ,٭ = p < 10-7 (assumption of homoscedascity, two-tailed

unpaired t-test).

Page 52: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

51

Page 53: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

52

Preliminary Pre-Clinical Trials for the Gemcitabine

Caged Prodrug System

Manuscript in preparation:

A. F. Herzog, E. M. Schneider, W. J. Stark,

Author contributions: concept/design by AFH and WJS. Experiments planned/conducted by

AFH with support of EMS. Data Analysis/interpretation by AFH and WJS. Drafting article:

AFH. Critical revision of article by AFH and WJS. Approval of article by AFH, EMS and WJS.

Page 54: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

53

3.1 Introduction

Clinical translation is a long and risky process with new drug approval taking an average

of 12 years with an average success rate from pre-clinical to phase III trials as low as 11%.109

In the US, the regulatory process begins with an Investigational New Drug (IND) application.

For the IND approval, it is necessary to document information about the drug production but,

most importantly, data from animal studies are required describing the drug’s safety and

efficacy.110

The vast majority of pre-clinical studies (over 80% of the studies published in 2016 in

the 8 most important journals) use cell-line derived models and xenograft mice in particular.111

In such models, the mice used are immune-compromised and human cancer cells are introduced

directly in the target organ or inoculated systemically. Although standard, xenograft models

have serious disadvantages such as the lack of a competent immune system, a much faster

tumour growth and an altered tumour microenvironment as compared to naturally developing

tumours.111 These limitations most likely reduce the clinical translation predictive power and

in many cases reproduction of key experiments in more complex models such as

environmentally-induced and genetically engineered mouse models is recommended.111

However, xenograft models can provide useful information regarding the toxicity of the various

components of the caged prodrug system and a lung cancer xenograft model was selected where

bioluminescent A549 cells were injected in the caudal vein of immuno-deficient mice.112 As a

result, such a model displays a cancer cell biology that closely resemble the one of the in vitro

experiments. Therefore, this has the potential to allow us to develop the treatment methodology

(e.g. how hydrogen is supplied) in a model for which it is known that the uncaged drug is active.

In other words, this reduces the number of unknowns and facilitates the development process.

Gagnadoux et al. administered aqueous aerozolized gemcitabine solutions to a similar

mice model.108 Their work served as a basis when planning the in vivo experiments reported

here. However, due to the insufficient water solubility of the caged prodrug, a formulation had

to be developed. In early tests, a proportion of ethanol and DMSO was added to ensure

solubility of the caged prodrug. However, if acute administration did not engender

complications, during chronic administration a large fraction of mice experienced strong lung

inflammation leading to death in some cases. To overcome this issue, it was decided to develop

emulsion formulations. In an attempt to facilitate future clinical translation, only materials

approved by the FDA for intratracheal or inhalation were used.113

Page 55: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

54

Preliminary formulation tests, using peanut oil, TWEEN® 80, propylene glycol and a

saline solution are here described. Additionally, toxicity profiles of the vehicle and platinum

nanoparticles solutions are disclosed. Finally, first data are reported for an experiment where

the lung cancer xenograft mice were administered with both caged gemcitabine and

gemcitabine.

3.2 Experimental

3.2.1 Formulations Preparation

The desired amounts of peanut oil and TWEEN® 80 were weighted and vortexed for 10 seconds

(Vortex-Genie 2, Scientific Industries). When applicable, the drug/prodrug was added to the

peanut oil/TWEEN® 80 mixture. The solution was vortexed for 1 minute, placed in an

ultrasonic bath (Sonorex Digitec DT 103 H, Bandelin) and vortexed for an additional minute.

A saline solution (0.9% NaCl) or a saline solution and propylene glycol were added. The

resulting solution was vortexed for 10 seconds before using an ultrasonic horn (UP50H,

Hielscher) for 30 seconds or until formation of a deep white emulsion.

3.2.2 Dynamic Light Scattering Measurements

Solutions were prepared by dissolving 10 µL of 990 µL of milli-Q water and vortexing 10

seconds (Vortex-Genie 2, Scientific Industries). The solutions were transferred to disposable

cuvettes (ZEN0040, Malvern) before the average size of the nanoparticles was determined by

dynamic light scattering (Zetasizer Nano ZS, Malvern).

3.2.3 Nebulization Assays

The following formulations of vehicle were nebulized using a IA-1C MicroSprayer® (Penn-

Century, USA) connected to a FMJ-250 high-pressure syringe (Penn-Century, USA). Right

before nebulisation assays, the formulations were vortexed for 30 seconds, sonicated for 1

minute and vortexed for additional 30 seconds to obtain stable emulsions.

Formulation 1: 0.3% TWEEN® 80, 20% peanut oil, 25% propylene glycol, rest 0.9% NaCl

Formulation 2: 0.3% TWEEN® 80, 20% peanut oil, rest 0.9% NaCl

Formulation 3: 1% TWEEN® 80, 20% peanut oil, 25% propylene glycol, rest 0.9% NaCl

Formulation 4: 1% TWEEN® 80, 20% peanut oil, rest 0.9% NaCl

Formulation 5: 0.3% TWEEN® 80, 20% peanut oil, 5% propylene glycol, rest 0.9% NaCl

Page 56: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

55

3.2.4 Tolerability Profiles of Formulation 2 and of Platinum Nanoparticles Solutions

3.2.4.1 Animals

Experiments were carried out using 9 female NOD/SCID mice (Janvier Labs, France), 9-10

weeks old on the day of the administration. Mice were housed in social groups of 3-6

individuals in polysulfone individually ventilated cages (IVC) (floor area = 501 cm²). An

external ventilation unit supplied the cages individually with fresh HEPA (High Efficiency-

Particulate Air)-filtered air (75 air volume replacements/hour). A positive pressure ventilation

was maintained in each cage. Cages were equipped to provide food and sterile water ad libitum.

Irradiated food (RM1 from SDS Dietex) and irradiated poplar wood bedding (MP 16/4 from

Anibed) were used. Cages and water bottles were cleaned and decontaminated using hydrogen

peroxide once a week. Ventilated cages were maintained in a specific pathogen-free (SPF)

animal facility under controlled temperature (22±2°C), hygrometry (55±10%), and photoperiod

(12h light/12h dark, light on at 8:00 am) conditions. Mice were numbered by marking their tail

with indelible markers. Mice were allowed to acclimate to their new environment for at least 5

days prior to the first day of the experiments.

3.2.4.2 Administration Protocol

On the first dosing day (D0), mice were randomly allocated to 3 groups (with 3 mice per group)

and administered as follows:

- Formulation 2 was administered once weekly aerosol for 4 weeks on D0, D7, D14 and

D21.

- Platinum nanoparticles solution (10 ppm, sterile-filtered) was administered as a single

aerosol administration on D0.

- Platinum nanoparticles solution (100 ppm, sterile-filtered) was administered as a single

aerosol administration on D0.

Aerosol administration were performed using a IA-1C MicrosSprayer® (Penn-Century, USA)

connected to a FMJ-250 high-pressure syringe (Penn-Century, USA) with an administration

volume of 25 μL/mouse, under isoflurane anaesthesia. Briefly, the mice were anaesthetised with

isoflurane and anaesthesia was maintained all along the procedure via a mask on the nose. Each

mouse was placed on its back on a platform hanging by its incisors to clearly visualize the

background of the throat. The MicroSprayer® was introduced into the animal’s trachea using a

laryngoscope to slowly inject 25 µL of the respective solutions. The mice were observed until

they recovered and displayed normal activity.

Page 57: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

56

3.2.4.3 Clinical Follow-up

From the first day of administration and until the sacrifice of the animals (on day 28), mice

were clinically observed daily for their general health status and weighed twice a week, and

then daily if substantial body weight loss was observed.

3.2.4.4 Terminal Procedures

Animals were sacrificed by sodium pentobarbital overdose, given by intraperitoneal (i.p.)

injection 28 days after the first administration (D28). An autopsy was performed with a detailed

macroscopic observation of the lung.

3.2.5 Feasibility Test in Xenograft Mice Model

3.2.5.1 Animals

Experiments were carried out using 21 female NOD/SCID mice (Janvier Labs, France), 7-8

weeks old on the day of the bioluminescent A549 cell implantation. From the 21 mice, 18 mice

were randomized on the first dosing day. Mice were housed in social groups of 3-6 individuals

in polysulfone individually ventilated cages (IVC) (floor area = 501 cm²). An external

ventilation unit supplied the cages individually with fresh HEPA (High Efficiency-Particulate

Air)-filtered air (75 air volume replacements/hour). A positive pressure ventilation was

maintained in each cage. Cages were equipped to provide food and sterile water ad libitum.

Irradiated food (RM1 from SDS Dietex) and irradiated poplar wood bedding (MP 16/4 from

Anibed) were used. Cages and water bottles were cleaned and decontaminated using hydrogen

peroxide once a week. Ventilated cages were maintained in a specific pathogen-free (SPF)

animal facility under controlled temperature (22±2°C), hygrometry (55±10%), and photoperiod

(12h light/12h dark, light on at 8:00 am) conditions. Mice were numbered by marking their tail

with indelible markers. Mice were allowed to acclimate to their new environment for at least 5

days prior to the first day of the experiments.

3.2.5.2 Cells

Human NSCLC A549 (ATCC® CCL-185™) cells were genetically modified to express

luciferase. Cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM High Glucose

+ GlutaMax™-I; ref 61965-026; Gibco, Life Technologies) supplemented with 10% foetal

bovine serum (FBS; ref 10270-098; Gibco, Life Technologies) and 1% cell culture

contamination preventive (ZellShield; ref 13-0150; Minerva-Biolabs). Cells were maintained

in an incubator with 5% CO2 at 37°C. Cells were routinely passaged two to three times per

week using 0.05% Trypsin 0.53 mM EDTA (ref 25300-056; Gibco).

Page 58: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

57

3.2.5.3 Preparation of Cells to Be Implanted

On the day of cell inoculation, cells were harvested by trypsinizing with 0.25% Trypsin-EDTA.

Cells were then washed with Dulbecco’s phosphate-buffered saline (D-PBS, ref. 14190-094,

Gibco, Life Technologies), centrifuged at 150 x g for 5 minutes at 4°C and suspended in serum-

free culture media at a concentration of 1x104 cells/µL, allowing for an inoculation of 1x106

cells/100 μL per mouse. Cells were kept on ice until the intravenous injection in mice.

3.2.5.4 Implementation of Tumour Cells

Each mouse received an intravenous injection of a suspension of 1x106 tumour cells in the

caudal vein, with an administration volume of 100 μL per mouse, on day 0 (D0). Cells were

carefully resuspended just prior to injection.

3.2.5.5 Tumour Measurements and Clinical Follow-up

After A549 cell injection and until the sacrifice of the animals, mice were clinically observed

daily for their general health status and weighed twice a week, and then daily if substantial body

weight loss was observed. Mice were monitored for 8 weeks after A549 cell injection. Tumour

growth was assessed on days 2, 10, 23, 37 and 52 (D2, D10, D23, D37 and D52) after cell

inoculation by non-invasive in vivo bioluminescence imaging. Each mouse was injected

intraperitoneally (i.p.) with 150 mg/kg D-luciferin (XenoLight D-Luciferin Potassium Salt, Ref

122799, Perkin Elmer) and anesthetized with isoflurane. Mice were positioned in the imaging

chamber (PhotonImager, Biospace Lab) and anaesthesia was maintained during the acquisition

via a mask on the nose. Analysis of bioluminescence data was performed using M3Vision

Software (Biospace Lab).

3.2.5.6 Administration Protocol

According to the bioluminescence imaging data obtained on D2 and D10, 15 out of the 21

implanted mice were randomly allocated to 3 groups (with 5 mice per group), in such a manner

that the tumours bioluminescence were homogeneous between the different groups. Mice not

allocated to a treatment group were sacrificed. Treatment was initiated 14 days after cell

implantation (D14) to let the tumour grow. Each mouse received once weekly aerosol

administration of protected gemcitabine, non-protected gemcitabine or vehicle for 4 weeks, i.e.

on D14, D21, D28 and D35, as follows:

1. Vehicle (0 mg/kg, aerosol)

2. Protected gemcitabine (5.9 mg/kg, aerosol)

3. (Non-protected) Gemcitabine (4 mg/kg, aerosol)

Page 59: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

58

Aerosol administrations were performed using a IA-1C MicroSprayer® (Penn-Century, USA)

connected to a FMJ-250 high-pressure syringe (Penn-Century, USA) with an administration

volume of 25 μL/mouse, under isoflurane anaesthesia. Briefly, the mice were anaesthetized

with isoflurane and anaesthesia was maintained all along the procedure via a mask on the nose.

Each mouse was placed on its back on a platform hanging by its incisors to clearly visualize the

background of the throat. The MicroSprayer® was introduced into the animal’s trachea using a

laryngoscope to slowly inject 25 µL of the respective solutions. The mice were observed until

they recovered and displayed normal activity.

3.2.5.7 Terminal Procedures

Animals were sacrificed by sodium pentobarbital overdose, given by intraperitoneal (i.p.)

injection 28 days after the first administration (D28).

3.2.5.8 Necropsy and Lung Collection

A necropsy of mice, even those found dead, was performed. The lungs were carefully observed

and the presence of tumour nodules was evaluated. After the sacrifice of each mouse, the lungs

were fixed by intratracheal perfusion of 4% formaldehyde solution, collected, and a picture was

taken. Lungs were placed in 4% formaldehyde solution for a minimum of 24 hours. All samples

were kept at room temperature.

3.3 Results and Discussion

3.3.1 Influence of Surfactant on the Average Droplet Size

After screening various surfactants and co-solvents, several stable formulations were prepared

with peanut oil, TWEEN® 80 as the surfactant, propylene glycol as the co-solvent and normal

saline solution (0.9 w/v% of NaCl). To test the influence of surfactant concentration on the

droplet size and stability, formulations composed of 0 to 5 wt. % of TWEEN® 80 were prepared

in 20 wt. % peanut oil, 25 wt. % propylene glycol and a corresponding saline solution

concentration of 50 to 55 wt. %. Formulations having less than 0.3 wt. % of TWEEN® 80 were

only stable for few minutes and were thus discarded from further tests. Above the 0.3 wt. %

threshold level, the average droplet size, as measured by dynamic light scattering (DLS), was

not significantly influenced by the surfactant concentration and was comprised between 560

nm and 840 nm (Figure 3-1). The small average droplet size ensures that the formulations are

compatible with the MicroSprayer® aerolizer utilized for the mice study. Of note, formulations

with higher TWEEN® 80 concentrations were stable at room temperature for weeks while

Page 60: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

59

formulations containing 0.3 wt. % were stable for few hours only. However, few hours stability

is long enough to carry out the mice administration procedure with a comfortable margin.

3.3.2 Freeze-thaw Stability

For the mice study it is necessary to store the drug-containing formulations over long period of

time (at least several weeks). To reduce the risk of degradation, it was decided to freeze the

formulations. It is well known that freeze-thaw stability represents a significant hurdle for

oil-in-water emulsions.114 Because the crystal size influences the freeze-thaw stability, samples

were placed in either a -20°C or -80°C freezer. As can be seen on Figure 3-2, the emulsions

remained relatively stable after one freezing/thawing cycle. However, after two cycles

significant phase separation could be observed. Therefore, it was decided to prepare aliquots

for the various administration steps of the in vivo study and to apply ultrasonication briefly

before administration.

Figure 0-1. Influence of surfactant concentration on the average droplet size. The average droplet size

was measured by dynamic light scattering (DLS) for 4 solutions containing 0.3 to 5 wt. % of

TWEEN® 80 in 20 wt. % peanut oil, 25 wt. % propylene glycol and a corresponding saline solution

concentration of 50 to 54.7 wt. %.

Page 61: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

60

3.3.3 Nebulization Properties of the Selected Formulations

A pre-selection of five formulations were passed through an aerolizer. Formulations were pre-

selected based on the following criteria: a low TWEEN® 80 content, a high peanut oil content

and varying degrees of propylene glycol content. The maximal peanut oil and propylene glycol

concentrations values were taken from the FDA database for inactive ingredients.113 It was

found that none of the formulations generated an aerosol but rather a thin jet instead. The

formulation giving best results was formulation 2 and was therefore selected for the mice

experiments. Remarkably, the formulation selected was the one containing the lowest

TWEEN® 80 concentration and no propylene glycol.

Figure 0-2. Influence of freezing temperature on the freeze-thaw stability. Two freezing/thawing cycles

were made at both -20°C and -80°C. After thawing, a short vortexing step was made before the average

droplet size was measured with a dynamic light scattering device.

Table 0-1. Table indicating the compositions of the various formulations and the macroscopic

observations after passing through a MicroSprayer® aerolizer.

Page 62: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

61

3.3.4 Tolerability Profile of Formulation 2 and Platinum Nanoparticles Solutions

Tolerability profiles were determined to ensure that no toxicity originates from neither the

vehicle (formulation 2) itself, nor from platinum nanoparticles. Three groups of 3 mice were

made. Mice from group 1 were chronically administered (once a week over one month) with

formulation 2. Mice from group 2 and 3 were acutely (on D0) administered with platinum

aqueous dispersions of 10 and 100 ppm, respectively. Mice from all 3 groups were regularly

clinically followed and were weighted twice a week during one month. The mice were then

sacrificed and their lungs macroscopically observed (Figure A2-1). No toxicity signs could be

observed for the lungs. Furthermore, the bodyweight curves (Figure 3-3) indicated normal

growth. In conclusion, neither formulation 2, nor the platinum nanoparticles solutions should

be toxic at the here used concentrations over a one-month period.

Figure 0-3. Tolerability profiles of formulation 2 and platinum nanoparticles solutions. Mice from

group 1 were administered weekly over one month with formulation 2. Mice from group 2 and 3 were

administered once on D0 and were clinically followed during one month.

Page 63: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

62

3.3.5 Feasibility Test in Xenograft Mice Model

Before testing the uncaging reaction in vivo, it was decided to first study the caged prodrug in

vivo toxicity and study its influence on the growth of tumours as compared to both the vehicle

and gemcitabine. This was done by weekly administering the various formulations to mice

previously inoculated with luciferase-expressing A549 cells. Bioluminescence was recorded

biweekly, mice weight was measured twice a week and mice were clinically observed daily

(Figure 3-4).

Due to the restricted number of animals of the current study, the statistical power is not

significant enough to draw definitive conclusions. However, several comments can already be

made from those preliminary data. First, caged gemcitabine did not, at least at a macroscopic

level, trigger any toxicity. This confirms what was observed in vitro with normal lung epithelial

cells. Second, the gemcitabine-containing formulation triggered significant toxicity as is

exemplified by the fact that many mice from this group prematurely died despite the slower

tumour growth. At the time of writing, histological and immunohistochemical studies are being

undertook on the lungs obtained after sacrificing the animals at the end of the study. The

information that will be gathered might provide more insights into the causes of death for mice

belonging to the gemcitabine group and indicate whether caged gemcitabine actually generated

some toxicity that was not detectable at the macroscopic level. If not, this would permit to either

go to higher drug concentration and/or have more frequent drug administrations. Another aspect

that was assessed during these experiments was whether the reducing environment, generally

present in tumours, was sufficient to trigger the uncaging reaction. This was not the case but it

has to be noted that tumours in such models are much less hypoxic that their human or

environmentally induced and genetically engineered mice models counterparts.63,111

Further studies are required to determine whether and with which conditions can the

prodrug be activated in vivo. Also, more therapeutically realistic, but costlier, models could be

envisaged to more precisely determine the therapeutic potential of this caged prodrug system.

Page 64: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

63

3.4 Conclusion

In this preliminary pre-clinical study, a formulation suitable for intratracheal lung

administration of caged gemcitabine was developed. Moreover, it could be shown that neither

the vehicle, the caged prodrug nor the platinum nanoparticles introduce toxicity. The

gemcitabine-containing formulation lead to significant toxicity and an important fraction of the

mice prematurely died from non-cancer related causes. The reducing environment of the tumour

was not sufficient to uncage the prodrug and new experiments are required to establish an in

vivo uncaging protocol. Overall, the results were encouraging but request further experiments.

For instance, once a prodrug uncaging protocol is found, it would have to be determined

whether the uncaging indeed translates into an elevated cytotoxicity and whether a higher drug

concentration and/or a more frequent administration could be used while keeping side-effect at

a level similar to that of gemcitabine. Also, more complex and more expensive mouse models

mimicking more closely human cancer could be used in the perspective of phase I clinical trials.

Figure 0-4. A) Growth of tumours as measured by the tumour bioluminescence signal. Of note, the

dashed lines are only indicative since the average does not correspond to the same number of animals

as in the beginning of the experiments. At t = 2 d, there were 6 animals in the vehicle and protected

gemcitabine groups and 5 animals in the gemcitabine group (one animal was removed from the analysis

because at t= 10 d, it displayed a bioluminescence signal two orders of magnitude higher than the other

animals within the same group). B) Kaplan-Meier curves for the time period during which

bioluminescence signals were regularly acquired.

Page 65: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

64

Page 66: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

65

Effective Tumour Cells Removal from Patient Blood

using Magnetic Nanoparticles.

Manuscript submitted:

A. F. Herzog, M. Zeltner, A. Zabel, N. Schläpfer, A. Siebenhüner, W. J. Stark,

B. Beck-Schimmer

Author contributions: concept/design by AFH, MZ, NS, WJS and BBS. Experiments

planned/conducted by AFH, MZ and AJ. Data Analysis/interpretation by AFH, MZ, AZ, NS,

WJS and BBS. Drafting article by AFH, MZ, AZ and BBS. Critical revision of article by AFH,

MZ, AZ, NS, AS, WJS and BBS. Approval of article by AFH, MZ, AZ, NS, AS, WJS and BBS.

Page 67: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

66

4.1 Introduction

Metastatic disease is responsible for over 90% of cancer deaths.115–118 Cells that detach

from the primary tumour and join the vascular system are also known as circulating tumour

cells (CTCs). They are thought to be involved in metastasis formation, and a direct correlation

between the CTCs number and patient prognosis has been found.119–122 Apart from the

continous release of CTCs from the primary tumour, a large increase in CTCs may be recorded

during and after cancer surgery,123–127 which further puts cancer patients at risk for recurrent

relapses as well as a decreased overall survival.25,26,128 Therefore, perioperative removal of

CTCs could potentially provide a way to improve long-term prognosis. However, targeted

elimination of blood cells is a challenging task, particularly with focus on CTCs, which often

present in low concentrations (down to 1 CTCs per 107 leukocytes or per 1010 erythrocyte).129

Here, we show a way to magnetically remove CTCs from blood. It relies on highly magnetic

and antifouling carbon-coated cobalt nanoparticles functionalised with polyglycidol and

epithelial-selective anti-epithelial cell adhesion molecule (EpCAM) antibodies. By using these

magnetic particles, our approach allows for the reproducible and specific removal of CTCs-

spiked blood. Moreover, first translation into a clinical scenario was possible, eliminating CTCs

from blood obtained from cancer patients. When evaluating safety aspects, such as possible

interference of the magnetic particles with the blood coagulation system, no major deviations

were observed. Our system is flexible in that the nanoparticles can be functionalized with other

antibodies, thereby potentially allowing further applications, as well as the possibility to adapt

to the discoveries in the field of CTCs biomarkers. We anticipate the development of such a

platform to translate into the feasibility of filtering the entire blood of patients, thus

hypothetically increasing patients’ prognosis, especially for the ones undergoing tumour

resection.

Although the significance of circulating tumour cells (CTCs) has been discussed for a long

time, their detection in the peripheral blood of cancer patients has received increasing interest

in the last decade. Reviews summarizing prognostic studies in breast, prostate, colon and lung

cancer highlighted the importance of CTCs as prognostic factors for tumour progression and

overall survival.118,130 The following examples further support these findings: For

adenocarcinoma of the breast, several trials suggested that patients tested “positive” for CTCs

had a shorter progression-free and overall survival.119,131 Also, in patients suffering from

urothelial cancer the presence of CTCs in the blood indicated poor outcome.132 Similarly, for

Page 68: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

67

the entity of gastric and gastro-oesophageal junction adenocarcinoma, CTCs were associated

with worse progression-free survival as well as impaired overall survival.133

Circulating tumour cells are cancer cells, which detach from the primary tumour and find

access to the blood. This way, they are carried around the body to other locations and, as they

leave the blood system and find access to the extravascular space, possibly lead to cancer

metastasis.134 Various technologies were developed for CTCs detection and isolation. Namely,

immunocytological-based approaches (e.g. immunocytochemistry, CellSearch system, flow

cytometry), molecular RNA-based technologies (e.g. qPCR, FISH) as well as functional assays

were reported.130,135 Among those technologies, the CellSearch system is the only detection

system to be approved by the FDA for tumour entities such as breast, prostate and colorectal

cancer, all of them of epithelial origin and therefore defined as carcinomas. Consequently,

CellSearch serves as the gold standard for CTCs enumeration. Akin to many of the methods

available for the detection of CTCs, the technology is taking advantage from the fact that CTCs

of epithelial origin express epithelial cell adhesion molecule (EpCAM) as a cell surface marker.

Many different EpCAM antibodies directed against this epitope are available, which allow a

specific interaction with it. This way CTCs of epithelial origin can be identified. This implies

that to detect CTCs from other tumour entities such as sarcoma or melanoma other markers are

needed for CTCs enrichment.136 Of note is that most of the malignancies are solid tumours.137–

139

While the diagnostic field of CTCs detection in the blood has been extensively

explored140–142, there is still no technique available to therapeutically remove CTCs in vivo from

a complex biological fluid, such as the blood, in a specific and efficient way without interacting

with the single blood components. The objective of the present work is to provide means and

methods to enable the efficient removal of CTCs from peripheral blood. This was done using

highly magnetic carbon-coated cobalt (C/Co) nanoparticles functionalized with polyglycidol

and conjugated with anti-EpCAM antibodies (Figure 4-1 and Figure 4-2a).

Page 69: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

68

4.2 Materials and Methods

4.2.1 Synthesis of C/Co-PhEtOH

Following previously described synthesis143, 10 g carbon-coated cobalt nanoparticles (C/Co)

were added to 400 mL of distilled water. The nanoparticles were dispersed by using an

ultrasonication bath (10 min, Sonorex Digitec DT 103 H, Bandelin). In a round-bottom flask,

4-aminophenethyl alcohol (1.2 g, 8.76 mmol) was mixed with 30 mL of distilled water and

dissolved by addition of 10 mL hydrochloric acid (HCl conc./37% fuming). The solution of 4-

aminophenyl alcohol was added to the dispersed particles before being placed for an additional

5 min in an ultrasonication bath. The nanoparticle dispersion was then placed in an ice bath

before a sodium nitrite solution (1.2 g, 17.4 mmol in 10 mL of distilled water) was added

dropwise. Instantaneous evolution of nitrogen gas (N2) could be observed. The reaction vessel

Figure 0-1. Overview of working principles. Human blood is sampled from a cancer patient (step 1) and a

dispersion of anti-EpCAM-functionalized nanoparticles is added (step 2). After gentle mixing, the nanoparticles

bind to CTCs and are then recovered by magnetic separation (step 3). In practice, this is done within a few minutes

by passing the sample through a magnetic beads-containing column. The filtrate is then subject to CTCs

enumeration (step 4). In later clinical applications, the purified blood could be reinjected to the patient after

extracorporeal purification.

Page 70: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

69

was then placed in an ultrasonication bath for 2 h. The nanoparticles were thoroughly washed

with distilled water (3 x 100 mL), EtOH (3 x 100 mL) and acetone (3 x 100 mL). At every step,

the particles were dispersed in an ultrasonication bath for 3 min and separated by application

of a permanent magnet (magnetic decantation). After washing, the particles were dried

overnight in a vacuum oven at 50°C.

4.2.2 Synthesis of C/Co-PhEtO-Na+

Following previously described syntheses144,145, 10 g of C/Co-PhEtOH were added to 20 mL of

a sodium methoxide solution (2 M in dry methanol). After being dispersed with an ultrasonic

bath for 2 min, the solution was stirred overnight at 65 °C. The particles were washed with dry

methanol (8 x 10 mL). At every step, the particles were dispersed in an ultrasonication bath for

3 min and separated by application of a permanent magnet (magnetic decantation). After

washing, the particles were dried overnight in a vacuum oven at 50 °C.

Elemental microanalysis:

[C] =5.13 %, [H] = 0.24%, [N] = 0.1%, [S] =0%

4.2.3 Synthesis of C/Co@polyglycidin

Following previously described synthesis146, 500 mg C/Co-PhEtO-Na+ were added to 20 mL of

dry toluene. After being dispersed with an ultrasonic bath for 2 h, the reaction mixture was

degassed by bubbling nitrogen through for 30 min. After installation of a reflux condenser, the

reaction mixture was heated up to 140 °C under inert conditions. Once the reaction mixture

reached 140 °C, 10 mL (+/-)-glycidol (+/- -oxiran-2-ylmethanol) was slowly added with a

syringe pump (1.3 mL/h). The reaction mixture was then stirred for 16 h before being down to

room temperature. The nanoparticles were thoroughly washed with toluene (100 mL), MeOH

(100 mL) and distilled water (100 mL). At every step, the particles were dispersed in an

ultrasonication bath for 3 min and separated by application of a permanent magnet (magnetic

decantation). The washing process with water was repeated until no foam generation could be

observed anymore. After washing, the particles were dried overnight in a vacuum oven at 50

°C.

Elemental microanalysis:

[C] = 10.3 %, [H] = 1.2 %, [N] = 0.07 %, [S] = 0 %

ΔC = 5.17 % = 4.31 mmol/g , ΔH = 1.08 % = 9.6 mmol/g, ΔN =-0.03 % = -0.02 mmol/g, ΔS

=0 %

Page 71: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

70

Calculated amount of polyglycidol: 1.44 mmol/g nanoparticles.

Calculated average chain length: 15 units per starter.

Infrared Spectroscopy:

Peak list: 2873 cm-1, 2356 cm-1, 1469 cm-1, 1328 cm-1, 1068 cm-1, 923 cm-1, 862 cm-1

4.2.4 Synthesis of C/Co@polyglycidyl-COOH

Following previously described synthesis147, 300 mg C/Co@polyglycidin were added to 15 mL

of dry dimethylformamide. After being dispersed with an ultrasonic bath for 3 min, succinic

anhydride (150 mg, 1.3 mmol) was added. The solution was placed for 10 min in an

ultrasonication bath before N,N-dimethylpyridin-4-amine (DMAP, 180 mg, 1.5 mmol) and

triethylamine (1.5 mL, 10.8 mmol) were added. The reaction mixture was degassed by bubbling

nitrogen through for 30 min. The reaction mixture was heated up to 70 °C and stirred overnight

under inert conditions. The nanoparticles were thoroughly washed with distilled water (3 x 100

mL). At every step, the particles were dispersed in an ultrasonication bath for 3 min and

separated by application of a permanent magnet (magnetic decantation).

Elemental Analysis:

[C] = 14.3 %, [H] = 1.4 %, [N] = 0.26 %, [S] = 0 %;

ΔC = 4 % = 3.3 mmol/g, ΔH = 0.2 % = 2 mmol/g, ΔN =0.19 % = 0.1 mmol/g, ΔS =0 %

Infrared Spectroscopy:

Peak list: 2939 cm-1, 2873 cm-1, 2675 cm-1, 2360 cm-1, 2356 cm-1, 1725 cm-1, 1560 cm-1,

1406 cm-1, 1244 cm-1, 1168 cm-1, 1068 cm-1, 838cm-1

4.2.5 Synthesis of C/Co@polyglycidyl-COO-EpCAM and C/Co@polyglycidin-

COO-IgG

1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and N-hydroxysulfosuccinimide

(sulfo-NHS) were left at room temperature until equilibration. Each compound was dissolved

in activation buffer (OceanNanotech) at concentrations of 4 mg/mL and 2 mg/mL, respectively.

The solutions were homogenised by vortexing for 10 sec. C/Co@polyglycidyl-COOH

nanoparticles solutions (5 mg/mL in activation buffer) were prepared. After being dispersed

Page 72: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

71

with an ultrasonic bath for 2 min, 200 µL of the solution were added to 1.5 mL Eppendorf tubes

containing 100 µL of activation buffer. The EDC and sulfo-NHS solutions were mixed in a 1:1

volume ratio and 10 µL were added to each nanoparticles-containing Eppendorf tube. To ensure

homogeneous dispersion, the solutions were vortexed for 10 sec, followed by 20 sec in an

ultrasonication bath. The activation step was done by shaking the Eppendorf tubes at 1200 rpm

for 10 min at 25°C in a thermomixer (ThermoMixer Comfort, Eppendorf). 100 µL of an

antibody solution (anti-EpCAM or IgG isotype control; 1 mg/mL) were added to the Eppendorf

tubes. To ensure homogeneous dispersion, the solutions were vortexed for 10 sec followed by

20 sec in an ultrasonication bath. The Eppendorf tubes were then shaken at 1200 rpm for 4 h at

25°C in a thermomixer. To quench the reaction, 10 µL of quenching buffer (OceanNanotech)

were added to each Eppendorf tube. To ensure homogeneous dispersion, the solutions were

vortexed for 10 sec followed by 20 sec in an ultrasonication bath. The quenching reaction was

done by shaking the Eppendorf tubes at 1200 rpm for 30 min at 25 °C in the thermomixer. The

particles were washed by placing the reaction vessel in a pre-cooled (4 °C) SuperMag separator

(OceanNanotech). To ensure complete separation, the magnet was placed in a fridge for 1.5 h

before the supernatant was discarded and replaced with 420 µL of fresh cold PBS (pH 7.4, Life

Technologies). The nanoparticles were dispersed by vortexing for 10 sec followed by 20 sec in

an ultrasonication bath. The Eppendorf tubes were placed back in the fridge and the washing

procedure was repeated 3 times. Once the washing process completed, the solutions were

aliquoted (30 µL aliquots) and stored at -20 °C until use.

4.2.6 Characterization of Antifouling Efficiency

Various nanoparticles batches were added to PBS (concentration of 2 mg/mL). Dispersion was

achieved by using an ultrasonic horn (3x30 sec on ice, UP50H, Hielscher).

Tetramethylrhodamine-conjugated bovine serum albumin (rhodamine-BSA) was dissolved in

PBS (concentration of 0.4 mg/mL), and the rhodamine-BSA solution was then diluted 3 times

to reach a concentration range where linear behaviour is observed for the antifouling tests. For

each nanoparticle batch 500 µL of the rhodamine-BSA and 500 µL of a nanoparticle solution

were added to an Eppendorf tubes. Particles having no polymer coating were used as the

negative control while milli-Q water was used as the positive control. To ensure homogeneous

dispersion, the solutions were vortexed for 10 sec followed by 30 sec in an ultrasonication bath.

To ensure optimal contact between the proteins and the nanoparticles surface, the samples were

shaken for 90 min at 1000 rpm at 25 °C (Thermomixer Comfort, Eppendorf). The particles were

washed by placing the Eppendorf tubes for 1 h in a SuperMag separator. Supernatant aliquots

Page 73: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

72

(5x100 µL) of each sample were made and transferred to a 96-well plate before fluorescence

was measured (ex: 540 nm, em: 620 nm; Spark 10M; Tecan).

4.2.7 Characterization of Separation Efficiency

Solutions of nanoparticles were prepared at a concentration of 2 mg/mL in PBS. Four mL of

the solutions were transferred to 5 mL glass vials. The vials were placed for 10 min in an

ultrasonic bath. The separation was started by placing one glass vial on each side of a permanent

magnet (1.3T, Webcraft AG). The separation was recorded using two CMOS sensors (sensor

1: 12 MP, 1.25 µm, f/2.2; sensor 2: 12 MP, 1.0 µm, f/2.6; Xiaomi A1, Xiaomi Inc.). The

quantification was done using an image processing program (ImageJ, NIH).

4.2.8 Cell Line Experiments

The EpCAM-expressing human colon cancer cell line HT-29 (HTB38™, ATCC) was cultured

in RPMI 1640 medium, supplemented with glutamax (Gibco), 10% foetal bovine serum (FBS,

Gibco) and a mixture of penicillin/streptomycin (final concentration 100 U/ml/ 100 µg/ml,

Gibco). The cells were incubated at 37°C in 5% CO2. Once cells reached confluence, they were

detached with accutase (Gibco) and resuspended in blood at a concentration of 5x105 cells/mL.

Cell membrane of the HT-29 cells was stained with the PKH26GL labelling kit (Sigma-Aldrich)

before the tumour cells were added to PBS or whole blood. Passages between 5 and 40 were

chosen for the experiments.

4.2.9 Scanning Electron Microscopy of anti-EpCAM Nanoparticles incubated with

HT-29 Cells

HT-29 cells were cultured for 48h in 12-well plates where carbon-coated sapphire disks were

deposited. 200 L of an anti-EpCAM nanoparticles solution (2.38 mg/mL in PBS) were added

to each well. The sapphire disks were then freeze substituted (-90°C to room temperature) in

dry ethanol containing 1% uranyl acetate. Room temperature samples were washed 3 times with

dry ethanol and critical point dried with liquid CO2 (Autosamdri 931, Tousimis). The samples

were coated with 7 nm of Au (CCU-010, Safematic) and imaged by SEM (Leo 1530, Zeiss).

4.2.10 Influence of Magnetic Nanoparticles Treatment on Lymphocytes Enumeration in

Healthy Subjects Spiked Blood

After informed patient consent (ethics approval: KEK-ZH-Nr. 2012-0274), up to 20 mL blood

samples were drawn from healthy subjects into heparin tubes (BD Biosciences). Eppendorf

Page 74: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

73

tubes containing either an anti-EpCAM nanoparticles solution (2.38 mg/mL in PBS) or an IgG-

isotype control nanoparticles solution (2.38 mg/mL in PBS) were sonicated (5x1 min with 1

min breaks in between) in an ultrasonication bath (Sonorex Digital 10P, Bandelin) on ice-cold

water. Once sonicated, the 25 L of the nanoparticles were added to 475 L of blood. In

addition to the IgG-isotype control, a control was made with 25 L PBS. The samples were

incubated for 2 min on an orbital shaker (16 mot/min, WS 10, Edmund Bühler) before being

passed through a column magnet system (MS columns, MACS, Miltenyi Biotec). Each column

was washed twice with 500 µL of PBS. Lymphocytes were labelled by transferring a third of

the filtrates to Eppendorf tubes and incubating with 1 L of Human CD19 APC-conjugated

(SJ25-C1, ThermoFisher) and 1 L of Human CD3d Alexa Fluor 48-conjugated (7D6,

ThermoFisher) for 30 min at 4°C. Samples were then prepared for flow cytometry analysis.

Red blood cells were lysed by adding 10 mL of a red blood cells lysis buffer (Biolegend, USA).

The samples were centrifuged for 10 min at 400 g (centrifuge 5810R, Eppendorf). The

supernatant was discarded and the pellet resuspended with 200 µL of PBS. The cells were fixed

with 200 µL of a 4% formalin solution. 25 µL of counting beads solution (concentration of

5.2x104 counting beads per 50 µL, CountBright Absolute, Life Technologies) were added to

each sample before analysing them using a flow cytometer (BD Canto II, BD Biociences).

Acquisition was carried out with the BD FACSDiva Software (BD Biosciences). Forward and

side scatter area as well as signal height were recorded. Lymphocytes were detected using either

side scatter area or forward scatter area vs either APC-A or Alexa Fluor 488-A. FACS data

were processed using FlowJo V10.0.8.

4.2.11 Removal of Circulating Tumour Cells (CTC) from Healthy Subjects Spiked Blood

After informed patient consent (ethics approval: KEK-ZH-Nr. 2012-0274), up to 20 mL blood

samples were drawn from healthy subjects into heparin tubes (BD Biosciences) and spiked with

labelled HT-29 cells (see above) to reach a concentration of 5x105 cells/mL. Eppendorf tubes

containing either an anti-EpCAM nanoparticles solution (2.38 mg/mL in PBS) or an IgG-

isotype control nanoparticles solution (2.38 mg/mL in PBS) were sonicated (5x1 min with 1

min breaks in between) in an ultrasonication bath (Sonorex Digital 10P, Bandelin) on ice-cold

water. Once sonicated, the 25 L of the nanoparticles were added to 475 L of cancer cells-

containing blood. In addition to the IgG-isotype control, a control was made with 25 L PBS.

The samples were incubated for 2 min on an orbital shaker (16 mot/min, WS 10, Edmund

Bühler) before being passed through a column magnet system (MS columns, MACS, Miltenyi

Page 75: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

74

Biotec). Each column was washed twice with 500 µL of PBS. A third of the filtrates were

transferred to Eppendorf tubes. Samples were then prepared for flow cytometry analysis. Red

blood cells were lysed by adding 10 mL of red blood cells lysis buffer lysis buffer (Biolegend,

USA). The samples were centrifuged for 10 min at 400 g (centrifuge 5810R, Eppendorf). The

supernatant was discarded and the pellet resuspended with 200 µL of PBS. The cells were fixed

with 200 µL of a 4% formalin solution. 25 µL of counting beads solution (concentration of

5.2x104 counting beads per 50 µL, CountBright Absolute, Life Technologies) were added to

each sample before analysing them using a flow cytometer (BD Canto II, BD Biociences).

Acquisition was carried out with the BD FACSDiva Software (BD Biosciences). Forward and

side scatter area as well as signal height were recorded. CTCs were detected using forward

scatter area vs the PE-. FACS data were processed using FlowJo V10.0.8.

4.2.12 Removal of Circulating Tumour Cells (CTCs) from Cancer Patients

After informed patient consent (ethics approval: KEK-ZH-Nr. 2016-01140), up to 25 mL blood

samples were drawn from cancer patients into either CellSave (Menarini-Silicon Biosystems),

heparin or EDTA tubes. After confirming the presence of circulating tumour cells with the

CellSearch system (Menarini-Silicon Biosystems), 7.5 mL CTCs-containing blood samples

were treated with either 375 µL of an anti-EpCAM nanoparticles solution (2.38 mg/mL in PBS)

or with 375 µL of PBS. Before being added to the blood samples, the nanoparticles solutions

were sonicated (5x1 min with 1 min breaks in between) in an ultrasonication bath (Sonorex

Digital 10P, Bandelin) on ice-cold water. The samples were incubated for 5 min on an orbital

shaker (16 mot/min, WS 10, Edmund Bühler) and passed through a column magnet system (LS

columns, MACS, Miltenyi Biotec). The filtrate was analysed with the CellSearch system

(Menarini-Silicon Biosystems) and evaluated by trained experts with CellSpotter.

4.3 Results and Discussion

4.3.1 Technical Aspects of the Newly Designed Nanoparticles

As a prerequisite for a successful translation into a clinical scenario, efficiency is highly

desirable, which allows blood purification of the patient’s blood within a short period of time.

This can be reached by using particles that are rapidly separated from the blood, allowing high

filtration rates. Magnetic separability is influenced by the metallic core’s magnetic properties

and, to a notable extent, by the agglomeration properties. In order to obtain efficient

separability, a paradigm shift was necessary for the design of the here described nanoparticles.

Page 76: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

75

Indeed, generally the focus lies on engineering nanoparticles as stable as possible. This is

because, for most applications, agglomeration is an undesirable outcome. As an example of

this, recently, C/Co nanoparticles were developed for later drug delivery applications.148 The

nanoparticles were functionalised with poly(3-sulfopropyl methacrylate potassium salt)

(pSPM), which affords ultra-stable particles. However, in our preliminary tests, the high

stability lead to a curtailed separability and poor performances for filtration applications (Figure

S1, green line). To overcome this problem, polyglycidol was selected as an alternative polymer

to pSPM because electrosteric repulsions are much lower than for pSPM, yet retain interesting

antifouling properties. Beyond the adequate separability properties, polyglycidol has further

advantages over pSPM. It is biologically inert and is approved by the FDA.149–151 The

preparation of the nanoparticles involves established syntheses that provide for a robust

manufacture (Figure 4-2a and Table A3-1). Additionally, the assessment of the degree of

Figure 0-2. Nanoparticles synthesis and characterisation. a, Overview of the synthesis of antibody-

functionalized C/Co nanoparticles. b, Infrared spectra of the C/Co nanoparticles coated with polyglycidol

and identical particles modified with a carboxylic group. The introduction of the carboxylic can be

monitored by the introduction of a peak around 1735 cm-1. c, Transmission electron micrographs of the

anti-EpCAM-functionalised C/Co nanoparticles. d, Transmission electron micrographs of the HT-29

cancer cells after incubation with anti-EpCAM-functionalised C/Co nanoparticles. The blue area delimits

a HT-29 cell, the red area nanoparticles bound on the cell surface.

Page 77: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

76

polymerisation as well as the number of carboxyl moieties incorporated can easily be

determined. The former is obtained by elemental microanalysis while the introduction of the

carboxyl group gives a clear infrared (IR) peak (Table A3-1, Figure 4-2b and Figures A3-4 to

A3-6). The resulting nanoparticles were further characterised by transmission electron

microscopy (Figure 4-2c). In an additional characterisation experiment where the nanoparticles

were incubated with cancer cells, nanoparticles aggregates could be observed on the surface of

the cells (Figure 4-2d). Assuming that colloidal stability is directly influenced by the extent of

polymer coating, three nanoparticles batches, with varied polymer lengths, were manufactured.

The batches had coating with average polymer lengths of 0 (no polymer spacer), 16 and 48

polymer units, respectively. An experiment was designed to obtain qualitative information

about the relative magnetic separability of the various batches. The nanoparticles were

dispersed in phosphate-buffered saline (PBS) and collected using a permanent magnet. The

separation was recorded using a camera and quantified using an image processing software. As

hypothesised, the larger the polymer chain, the slower the separation (Figure 4-3a, Figure A3-

1).

Removal of CTCs implies using nanoparticles that do not display extensive unspecific

protein adsorption (i.e. designing nanoparticles with good antifouling properties). As a

consequence, a protocol using rhodamine-labelled albumin, the most abundant plasma

Figure 0-3. Polymer coating optimisation. a, Qualitative information about the polymer length influence on

the magnetic separability. Nanoparticles solutions were placed next to a permanent magnet. The separation

was recorded using a camera and quantification was done using an image processing software. b, Polymer

length influence on the protein adsorption. Rhodamine-modified bovine serum albumin (BSA) was incubated

in PBS with the nanoparticles. After magnetic separation, the fluorescence of the supernatant was measured

with a plate reader (n = 5 technical replicates; 𝜆ex/em = 540/620 nm). c, Polymer length influence on the CTC

removal. Healthy subjects’ blood was spiked with HT-29 cells. Anti-EpCAM-functionalised C/Co nanoparticles

were added to samples which were passed through a magnetic column system. The enumeration of the HT-29

cells in the filtrate was assessed by flow cytometry (n = 3 biological replicates).

Page 78: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

77

protein152, was established. After incubation, the nanoparticles were collected and the

fluorescence of the supernatant measured. The protein adsorption was back-calculated using a

standard curve. As expected, antifouling properties were directly influenced by the degree of

polymerisation. Particles functionalised with no spacer had a 6.8-fold higher bovine serum

albumin (BSA) adsorption on their surface as compared to particles having 48 polyglycidol

units (Figure 4-3b). Particles with an intermediate polymer chain length had an adsorption level

comprised between the two aforementioned extremes.

4.3.2 First Tests in Human Blood from Healthy Subjects Spiked with Cancer Cells

After the magnetic separability and antifouling tests, CTCs removal efficiency of the three types

of nanoparticles was assessed in blood samples from healthy subjects. Nanoparticles, coated

with anti-EpCAM antibodies, were supplemented to fresh human blood previously spiked with

HT-29 cells, a human colorectal adenocarcinoma cell line. After mixing, the samples were

passed through magnetic microbeads-containing columns for positive selection. Erythrocytes

present in the filtrate were then lysed and CTC enumeration was obtained by flow cytometry.

Interestingly, the removal efficiency followed a divergent pattern as compared to the antifouling

properties, and an optimum was found for particles functionalised with an average of 16 units

per polyglycidol chain (Figure 4-3c). This could indicate that two competing factors are at play.

On one hand, the removal efficiency increases with improved antifouling properties (over 7-

fold higher efficiency for particles with 16 units as compared to particles with no polymer

coating). On the other hand, particles with superior antifouling properties dreadfully performed

(over 28-fold higher efficiency for particles with 16 units as compared to particles with 48

units). This suggests that, in our case, separability plays a more substantial role for cell removal

efficiency over antifouling properties. A plausible explanation is that, for more stable particles,

the forces obtained by placing the separation columns in a permanent magnet are not enough to

overcome the electrosteric repulsion forces and the forces resulting from the flow through the

magnetic column. Hence, the antifouling properties might only be necessary to ensure that

nanoparticles are moderately covered by other blood components and thereby playing only a

secondary role.

4.3.3 Specificity, Safety Aspects and Reproducibility

When eliminating CTCs from blood, the removal is expected be specific through the EpCAM

epitope-antibody interaction. That is, the CTCs removal should not originate from unspecific

binding of the tumour cells on the nanoparticles’ surface. To study this, nanoparticles were

Page 79: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

78

conjugated with either the IgG isotype as control or the anti-EpCAM antibodies. Both groups

of nanoparticles were subject to the protocol for CTCs removal from healthy subjects’ blood

spiked with cancer cells (HT-29 cells). When using IgG isotype nanoparticles between 83%

and 91.4% of the spiked cells remained in the blood samples, hence demonstrating the

specificity of the nanoparticles (Figure 4-4a and Figure 4-4b).

Another aspect of specificity concerns elimination of CTCs without interacting with other blood

cells. Therefore, experiments were performed to test the hypothesis that EpCAM-functionalised

nanoparticles do not eliminate important immunological blood components such as B- and T-

lymphocytes. Blood was exposed to anti-EpCAM and purified using the magnetic column

system. Lymphocytes counts were assessed in the filtrate via flow cytometry. The level of B-

and T-lymphocytes after treatment with anti-EpCAM nanoparticles did not differ significantly

from control blood that had run over the column (Figure 4-4c and Figure A3-3).

To explore the possible interaction of the nanoparticles with the coagulation system, rotational

thromboelastometry (ROTEM®) measurements were performed. ROTEM® is a clinically

well-established viscoelastic method to test haemostasis in whole blood, which allows not only

the definition of diagnostic, but also therapeutic processes as a reliable point-of-care tool. It

provides detailed information about clot formation such as clotting time (CT; start of reaction

until clots starts forming) or maximum clot firmness (MCF), reflecting the strength of the clot.

For the anti-EpCAM nanoparticles, all ROTEM® data were found to be within the reference

values not only for the EXTEM with activation of the clotting cascade via tissue factor, but also

for the INTEM with contact activation (Table A3-2)153 ROTEM®

The safest way to avoid in vivo complications, is to prevent nanoparticles from entering into

the patient’s bloodstream. This can be obtained by efficient magnetic separation followed by

monitoring before blood reinjection into the patient’s circulatory system. To achieve this, we

recently co-developed a method to detect nanoparticles in blood down to sub-picomolar

concentrations.154 The method can be setup in line with our previously developed

extracorporeal continuous blood purification device.155

Reproducibility being one of the key requirements of the FDA’s manufacturing process

validation, and indeed for the clinical application itself as well, several batches of the optimised

nanoparticles were synthesised. The synthesis was particularly robust; as exemplified by the

limited batch to batch variation in the extent of carboxyl moiety introduction, less than 2.5%,

as measured by elemental microanalysis (Table A3-1). The batches were later functionalised

Page 80: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

79

with either anti-EpCAM or IgG isotype control antibodies and CTCs removal from spiked

blood experiments were repeated. All batches yielded similar results, with over 98% CTCs

removal, thereby confirming the biological reproducibility beyond the chemical reproducibility

(Figure 4-4b).

Figure 0-4. CTCs removal from spiked blood and from cancer patient blood samples. a, Flow

cytometry data performed on filtrates obtained after CTCs removal procedure in healthy subject’s

spiked blood. The gate indicates the region where the spiked HT-29 cells appear. b, Specific

elimination of CTCs with particles functionalised with anti-EpCAM. Also, this graph displays batch

to batch reproducibility of CTC removal. The CTC removal procedure was reproduced three times

with each of the three optimised anti-EpCAM nanoparticles batches. c, Table showing B- and T-

lymphocyte count after treatment with anti-EpCAM coated magnetic nanoparticles. d, CTCs

removal from cancer patient blood samples. Blood samples were taken from cancer patients with

metastatic disease. CTCs enumeration was carried out on a CellSearch device for samples with

and without treatment with nanoparticles.

Page 81: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

80

4.3.4 First Tests with Blood Samples from Cancer Patients

In a subsequent step, the optimised nanoparticles were used to remove CTC from blood samples

obtained from cancer patients. This included blood from patients suffering from prostate

(patient no. 1, 2 and 3), pancreatic (patient no. 4 and 6) and colon cancer (patient no. 5). The

protocol consisted in a direct 15 times scale up of the spiked blood experiments (7.5 mL vs. 0.5

mL). All cancer patients had suffered from metastatic disease, and blood was drawn before they

received standard of care chemotherapy. In blood of two cancer patients with colon cancer,

results were not conclusive. Of the remaining six patients, the average removal efficiency was

over 68% (Figure 4-4d). These preliminary results obtained are encouraging, especially given

the fact that patients had very different initial CTCs counts and their CTCs most likely exhibit

various EpCAM-expressing profiles.

4.4 Conclusion

In summary, carbon-coated cobalt (C/Co) nanoparticles were covalently functionalised with a

polyglycidol coating and anti-EpCAM antibodies. Such nanoparticles exhibited a high

magnetisation and the antifouling properties necessary to provide the desired ability to remove

CTCs from peripheral blood with a remarkable efficiency, both from blood of healthy subjects

spiked with tumour cells as well as from cancer patients. Of highlight is extraction of CTCs of

different tumour entities as well as elimination of tumour cells of various concentrations.

Additionally, the nanoparticles exhibited adequate specificity towards EpCAM-expressing

cancer cells without elimination of other blood cells such as lymphocytes. Moreover, the

coagulation system, a major component of the blood, does not seem to be impaired, neither

towards thrombosis or thrombolysis. Finally, the material could be manufactured reproducibly

using robust syntheses procedures. These preliminary findings constitute a first step towards

whole blood filtration for CTCs removal. If confirmed by further preclinical and clinical data,

this might provide a new tool in the field of cancer surgery.

Page 82: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

81

Page 83: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

82

Functionalization Reaction Study and Lymphocytes’

Capture as a Demonstration of Highly Abundant Cells

Removal

Manuscript in preparation:

A. F. Herzog, M. Zeltner, A. Zabel, N. Schläpfer, A. Siebenhüner, W. J. Stark,

B. Beck-Schimmer

Author contributions: concept/design by AFH, MZ, NS, WJS and BBS. Experiments

planned/conducted by AFH, MZ and AJ. Data Analysis/interpretation by AFH, MZ, AZ, NS,

WJS and BBS. Drafting article by AFH, MZ, AZ and BBS. Critical revision of article by AFH,

MZ, AZ, NS, AS, WJS and BBS. Approval of article by AFH, MZ, AZ, NS, AS, WJS and BBS.

Page 84: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

83

5.1 Introduction

Extracorporeal blood purification using magnetic nanoparticles falls into the class III of

the FDA’s medical devices classification.156 Such devices must undergo the most stringent

process of medical devices approval. The development costs are typically comprised between

$10 and $20 million dollars and take an average of 3 to 7 years until reaching the market.156

Similarly to new drugs, for a class III medical device to be approved, it is required to

demonstrate its safety and efficacy.156

In the perspective of pre-clinical and clinical trials, consequent quantities of the carbon-

coated cobalt nanoparticles, described in Chapter 4, are needed and production at the upper

gram scale is required. The current limiting step for scaling up is the polyglycidol

polymerization. Therefore, the influence of various reaction parameters was studied in order to

ease the scale up process.

In the previous chapter, the use of such magnetic nanoparticles to remove rare cells was

reported. To demonstrate that such nanoparticles can further be used to remove highly abundant

cells, the polyglycidol-functionalized magnetic nanoparticles were conjugated with Campath,

a monoclonal antibody binding to the CD52 protein present on the surface of mature

lymphocytes.157 Normal lymphocytes counts are in the range of several millions cells per

millilitre blood.158 This is at least 4 to 6 orders of magnitude more than circulating tumour cells.

Moreover, abnormal lymphocyte counts or abnormal lymphocytes behaviour are encountered

in numerous diseases. Therefore, the use of lymphocyte-removing nanoparticles could find

applications for diseases such as the ones exemplified below.

Autoimmune diseases are a broad class of conditions where the immune system

recognizes self-antigens as foreign and mounts an immune response against self-entities.

Autoimmune diseases include heavy conditions such as multiple sclerosis or rheumatoid

arthritis. There are no cure for autoimmune diseases and, up to now, treatments where mainly

aiming at improving symptoms. For instance, the burden of several autoimmune diseases could

be alleviated by administering patients with rituximab; an antibody reducing the number of B-

cells which are notably responsible of presenting antigens to T-cells.159,160 However, anti-

rituximab immune responses can decrease the effectiveness of the drug.159,161 This serious issue

could be bypassed by achieving B-cell depletion via magnetic nanoparticles-based

extracorporeal blood purification with which, due to the shorter exposition time, the likeliness

of mounting an adaptive immune response would be diminished.

Page 85: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

84

Allergies, also called type I hypersensitivities, form a class of conditions where the

immune system mounts an inappropriate immune response when presented to an actually

inoffensive antigen. Removal of especially T-lymphocytes could weaken the symptoms

observed during chronic allergic inflammation (i.e. when the allergen exposure is continuous)

or mitigate the extent of late-phase allergy, which occurs few hours after early-phase reaction

(i.e. acute reaction).162

Lymphomas constitute a group of cancers involving lymphocytes. There are over 30

types of lymphomas and currently, to establish a diagnostic, a lymph node biopsy is required to

observe the cells under a microscope.163 By using magnetic nanoparticles to isolate

lymphocytes circulating in the blood, it would provide a mean to obtain a liquid biopsy that

would be much less invasive and would benefit both the surgeon and the patient.

This chapter thus contains two parts. First, in view of the future need to scale up the

nanoparticles’ production process, the influence of various reaction variables for the

polymerization step are reported as well as its reproducibility. Also, this chapter constitutes an

illustration of the possibility to use polyglycidol-functionalized carbon-coated cobalt

nanoparticles to remove the highly abundant B- and T-lymphocytes from the blood of healthy

and cancer patients.

Figure 0-1. Schematic overview of the extracorporeal lymphocytes’ removal and examples of diseases

for which polyglycidol-functionalized C/Co nanoparticles could find applications.

Page 86: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

85

5.2 Materials and Methods

5.2.1 Synthesis of C/Co-PhEtOH

To a 500 mL Schott® flask containing 200 mL of distilled water, 10 g carbon-coated cobalt

nanoparticles (C/Co) were added. Nanoparticles were dispersed by placing the Schott® flask

for 10 min in an ultrasonication bath (Sonorex Digitec DT 103 H, Bandelin). Concomitantly,

in 20 mL of distilled water, 2-(4-aminophenyl)ethanol (1.5 g, 10.95 mmol) was added. A

hydrochloric acid solution (5 mL, HCl conc. / 37% fuming) was used to dissolve 4-aminophenyl

alcohol before addition to the nanoparticles solution. A sodium nitrite (NaNO2, 1.5 g, 21.8

mmol) was prepared in 5 mL of distilled water. Before the dropwise addition of the sodium

nitrite solution, the nanoparticles solution was cooled down with an ice bath. The nanoparticles

solution was left reacting for 2 h by placing back the Schott® flask in the ultrasonication bath.

Once the reaction over, the nanoparticles were washed several times by magnetic decantation.

The general washing procedure was: nanoparticles dispersion by placing the container for 3

min in the ultrasonication bath and subsequent application of a magnetic field with a permanent

magnet. After discarding the supernatant, fresh solvent was added and the washing procedure

repeated. The following solvents were used: distilled water (3 x 100 mL), EtOH (3 x 100 mL)

and acetone (3 x 100 mL). The particles were then dried overnight in a vacuum oven at 50°C.

5.2.2 Synthesis of C/Co-PhEtO-Na+

To a 50 mL two-neck round-bottom flask, 10 g of C/Co-PhEtOH nanoparticles and a magnetic

stirrer were added. A reflux condenser was installed and the installation was placed under inert

atmosphere. A commercial sodium methoxide solution (20 mL, 2 M in dry methanol, Fluka)

was added. Nanoparticles were dispersed by placing for 30 s an ultrasonic horn (UP50H,

Hielscher) in the over-pressurized installation. The solution was left reacting by stirring

overnight at 65°C. The general washing procedure was: nanoparticles dispersion by placing the

ultrasonic horn for 30 s in the solution and subsequent application of a magnetic field with a

permanent magnet. After discarding the supernatant with a syringe, fresh solvent was added

and the washing procedure repeated. The following solvents were used: dry methanol (8 x 100

mL). Five needles were placed in the septum be the contained was placed overnight in a vacuum

oven at 50°C to dry the nanoparticles.

Page 87: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

86

5.2.3 Example of a Standard Synthesis of C/Co@polyglycidin

To a 100 mL three-neck round-bottom flask, 500 mg C/Co-PhEtO-Na+ were added to 20 mL of

dry toluene. A reflux condenser and a high shear mixer (1050 FME-P Geradschleifer, Kress)

were installed while the last opening was closed with a septum for injection. The particles were

dispersed by placing the high shear mixer on maximal speed (25000 rpm). The reaction mixture

was degassed by bubbling nitrogen through for 30 minutes and temperature was then increased

to 140°C under inert conditions. Once the reaction mixture reached 140°C, the shear speed was

reduced to 16500 rpm and 10 mL (+/-)-glycidol (+/- -Oxiran-2-ylmethanol) was slowly added

with a syringe pump (1.3 mL/h, Model 100, KD Scientific). From the time of injection, the

reaction mixture was stirred for 16 h before being cooled down to room temperature. The

nanoparticles were washed 7 times with toluene distilled water (100 mL). At every step, the

particles were dispersed in an ultrasonic bath (Sonorex Digitec DT 103 H, Bandelin) for 3 min

and separated by application of a permanent magnet (magnetic decantation). After washing, the

particles were dried overnight in a vacuum oven at 50°C.

5.2.4 Synthesis of C/Co@polyglycidyl-COOH

300 mg C/Co@polyglycidin were added to 15 mL of dry dimethylformamide. After being

dispersed with an ultrasonic bath (3 min, Sonorex Digitec DT 103 H, Bandelin), succinic

anhydride (150 mg, 1.3 mmol) was added. The solution was placed for 10 min in an

ultrasonication bath before N,N-Dimethylpyridin-4-amine (180 mg, 1.5 mmol) and

triethylamine (1.5 mL, 10.8 mmol) were added. The reaction mixture was degassed by bubbling

nitrogen through for 30 minutes. The reaction mixture was heated up to 70°C and stirred

overnight under inert conditions. The nanoparticles were thoroughly washed with distilled

water (3 x 100 mL). At every step, the particles were dispersed in an ultrasonication bath for 3

min and separated by application of a permanent magnet (magnetic decantation).

5.2.5 Synthesis of C/Co@polyglycidyl-COO-Campath and C/Co@polyglycidin-COO-

IgG

1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and N-hydroxysulfosuccinimide

(sulfo-NHS) were left at room temperature until equilibration. Each compound was dissolved

in activation buffer (OceanNanotech) at concentrations of 4 mg/mL and 2 mg/mL, respectively.

The solutions were homogenized by vortexing for 10 s. C/Co@polyglycidyl-COOH

nanoparticles solutions (5 mg/mL in activation buffer) were prepared. After being dispersed

with an ultrasonic bath (2 min, Sonorex Digitec DT 103 H, Bandelin), 200 µL the solution were

Page 88: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

87

added to 1.5 mL Eppendorf tubes containing 100 µL of activation buffer. The EDC and sulfo-

NHS solutions were mixed in a 1:1 volume ratio and 10 µL were added to each nanoparticles-

containing Eppendorf tube. To ensure homogeneous dispersion, the solutions were vortexed for

10 seconds followed by 20 seconds in an ultrasonication bath (Sonorex Digitec DT 103 H,

Bandelin). The activation step was done by shaking the Eppendorf tubes at 1200 rpm for 10

min at 25°C in a ThermoMixer (ThermoMixer Comfort, Eppendorf). 100 µL, 200 µL or 300

µL of an antibody solution (Campath or IgG isotype control; 30 mg/mL and 1 mg/mL,

respectively) was added to the Eppendorf tubes. To ensure homogeneous dispersion, the

solutions were vortexed for 10 seconds followed by 20 seconds in an ultrasonication bath

(Sonorex Digitec DT 103 H, Bandelin). The Eppendorf tubes were then shaken at 1200 rpm for

4 h at 25°C in a ThermoMixer (ThermoMixer Comfort, Eppendorf). To quench the reaction, 10

µL of quenching buffer (OceanNanotech) were added to each Eppendorf tube. To ensure

homogeneous dispersion, the solutions were vortexed for 10 seconds followed by 20 seconds

in an ultrasonication bath (Sonorex Digitec DT 103 H, Bandelin). The quenching reaction was

done by shaking the Eppendorf tubes at 1200 rpm for 30 min at 25°C in a ThermoMixer

(ThermoMixer Comfort, Eppendorf). The particles were washed by placing the reaction vessel

in a pre-cooled (4°C) SuperMag separator (OceanNanotech).To ensure complete separation,

the magnet was placed in a fridge for 1.5 h before the supernatant was discarded and replaced

with 420 µL of fresh cold PBS (pH 7.4, Life Technologies). The nanoparticles were dispersed

by vortexing for 10 seconds followed by 20 seconds in an ultrasonication bath (Sonorex Digitec

DT 103 H, Bandelin). The Eppendorf tubes were placed back in the fridge and the washing

procedure was repeated 3 times. Once the washing process completed, the solutions were

aliquoted (30 µL aliquots) and stored at -20°C until use.

5.2.6 Removal of Lymphocytes from Healthy Subjects

After informed patient consent (ethics approval: KEK-ZH-Nr. 2012-0274), up to 20 mL blood

samples were drawn from healthy subjects into heparin tubes (BD Biosciences). Eppendorf

tubes containing either a Campath nanoparticles solution (2.38 mg/mL in PBS) or an IgG-

isotype control nanoparticles solution (2.38 mg/mL in PBS) were sonicated (5x1 min with 1

min breaks in between) in an ultrasonication bath (Sonorex Digital 10P, Bandelin) on ice-cold

water. Once sonicated, the 25 L of the nanoparticles were added to 250 L of blood. In

addition to the IgG-isotype control, a control was made with 25 L PBS. The samples were

incubated for 2 min on an orbital shaker (16 mot/min, WS 10, Edmund Bühler) before being

passed through a column magnet system (MS columns, MACS, Miltenyi Biotec). Each column

Page 89: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

88

was washed twice with 500 µL and 750 µL of PBS. Lymphocytes were labelled by transferring

a third of the filtrates to Eppendorf tubes and incubating with 1 L of Human CD19 APC-

conjugated (SJ25-C1, ThermoFisher; final concentration: 0.2 mg/mL) and 1 L of Human

CD3d Alexa Fluor 48-conjugated (AB, ThermoFisher; final concentration: 0.2 mg/mL) for 30

min at 4°C. Samples were then prepared for flow cytometry analysis. Red blood cells were

lysed by adding 10 mL of a red blood cells lysis buffer (Biolegend, USA). The samples were

centrifuged for 10 min at 400 g (centrifuge 5810R, Eppendorf). The supernatant was discarded

and the pellet resuspended with 200 µL of PBS. The cells were fixed with 200 µL of a 4%

formalin solution. 25 µL of counting beads solution (concentration of 5.2x105 counting beads

per 50 µL, CountBright Absolute, Life Technologies) were added to each sample before

analysing them using a flow cytometer (BD Canto II, BD Biociences). Acquisition was carried

out with the BD FACSDiva Software (BD Biosciences). Forward and side scatter area as well

as signal height were recorded. Lymphocytes were detected using either side scatter area or

forward scatter area vs either APC-A or Alexa Fluor 488-A. FACS data were processed using

FlowJo V10.0.8.

5.2.7 Removal of Lymphocytes from Cancer Patients

After informed patient consent (ethics approval: KEK-ZH-Nr. 2016-01140), up to 25 mL blood

samples were drawn from cancer patients into heparin tubes (BD Biosciences). Eppendorf tubes

containing either a Campath nanoparticles solution (2.38 mg/mL in PBS) or an IgG-isotype

control nanoparticles solution (2.38 mg/mL in PBS) were sonicated (5x1 min with 1 min breaks

in between) in an ultrasonication bath (Sonorex Digital 10P, Bandelin) on ice-cold water. Once

sonicated, the 25 L of the nanoparticles were added to 250 L of blood. In addition to the IgG-

isotype control, a control was made with 25 L PBS. The samples were incubated for 2 min on

an orbital shaker (16 mot/min, WS 10, Edmund Bühler) before being passed through a column

magnet system (MS columns, MACS, Miltenyi Biotec). Each column was washed twice with

500 µL and 750 µL of PBS. Lymphocytes were labelled by transferring a third of the filtrates

to Eppendorf tubes and incubating with 1 L of Human CD19 APC-conjugated (SJ25-C1,

ThermoFisher; final concentration: 0.2 mg/mL) and 1 L of Human CD3d Alexa Fluor 48-

conjugated (AB, ThermoFisher; final concentration: 0.2 mg/mL) for 30 min at 4°C. Samples

were then prepared for flow cytometry analysis. Red blood cells were lysed by adding 10 mL

of a red blood cells lysis buffer (Biolegend, USA). The samples were centrifuged for 10 min at

400 g (centrifuge 5810R, Eppendorf). The supernatant was discarded and the pellet resuspended

Page 90: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

89

with 200 µL of PBS. The cells were fixed with 200 µL of a 4% formalin solution. 25 µL of

counting beads solution (concentration of 5.2x105 counting beads per 50 µL, CountBright

Absolute, Life Technologies) were added to each sample before analysing them using a flow

cytometer (BD Canto II, BD Biociences). Acquisition was carried out with the BD FACSDiva

Software (BD Biosciences). Forward and side scatter area as well as signal height were

recorded. Lymphocytes were detected using either side scatter area or forward scatter area vs

either APC-A or Alexa Fluor 488-A. FACS data were processed using FlowJo V10.0.8.

5.3 Results and Discussion

5.3.1 Influence of Various Reaction Parameters on Polymer Length

It has been reported that one of the limiting factors to achieve successful clinical translation of

nanomedicines, was the limited reproducibility and/or scalability.53 Empirically, it has been

found that, for such polyglycidol-functionalized carbon-coated cobalt nanoparticles, the

polymerization is the most delicate step. Therefore, the influence of three key reaction

parameters of the polymerization reaction have been tested by reproducing the procedure and

changing one variable at a time in ranges near the values obtained for the batch displaying

optimal properties in Chapter 4. The first parameter to be changed was the speed of mixing. To

obtain results that are more easily transferrable to industrial processes, the magnetic stirrer used

in chapter 4 was replaced by a high shear mixer. This provides a more controlled mixing as

compared to what is achieved with a traditional chemistry setup (i.e. magnetic stirrer combined

with a magnetic plate) but it also facilitates the scale-up process. It was found that increasing

the shear speed only resulted in minor increase of the number of polymer units introduced

(Figure 5-2A). Second, the excess of glycidol value was modified while keeping the rate of

addition constant. As can be seen on Figure 5-2B, going to larger volumes did not result in

much change with respect to the number of polymer units. Lastly, the reaction temperature was

modified. Temperature appeared to be a key factor in controlling the extent of polymer

functionalization (Figure 5-2C). Moreover, for the values reported here, a linear correlation was

found between the reaction temperature and the number of polymer units. This could therefore

provide an easy mean to control the amount of polyglycidol introduced by simply altering the

temperature.

Page 91: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

90

5.3.2 Reproducibility of Polymerization Step

Materials reproducibility is one of the key requirements from the U.S. FDA. Polyglycidol

functionalization was thus repeated three times with identical reaction conditions and the

batches characterized by elemental analysis. As can be seen on Figure 5-3A, the carbon content

was comprised within a narrow range for all three batches thus indicating the introduction of

similar amounts of polymer as calculated (Appendix A4 and Figure 5-3D). On Figure 5-3C, a

decrease in nitrogen content is observed for the third batch. However, since the nitrogen content

is very low, the differences measured are not significant and are more likely to be due to a

residual error of the instrument or the introduction of air in the sample. Moreover, the

polymerization step does not introduce any nitrogen and the starting material was the same for

all three batches. Of note, one of the limitations from elemental analysis is that it does not

provide any information about the structure of the polymer layer introduced which, in the case

of polyglycidol, can differ greatly (i.e. ratio of linear vs branched polymer). However, in their

Figure 0-2. Influence of various reaction parameters on the number of polymer units introduced. A)

Influence of the speed of mixing. B) Influence of the glycidol volume excess. C) Influence of temperature.

Page 92: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

91

draft report of the guidance for industry, the U.S. FDA does not require the description of the

polymer structure but rather the “coating properties”.164

5.3.3 B- and T-Lymphocytes Removal from Healthy Subject Blood Samples

As a demonstration of the possibility of using polyglycidol-functionalized carbon-coated cobalt

nanoparticles for lymphocytes removal, the nanoparticles were functionalized with Campath

(also called Alemtuzumab and Lemtrada). This antibody is used in the clinic for the treatment

of multiple sclerosis.165 Three batches were synthesized with varying excesses of antibody

solutions volumes (100 µL, 200 µL and 300 µL, respectively) to determine whether this would

influence the conjugation and, ultimately, the removal efficiency. To test the latter, blood from

a healthy subject was sampled and magnetic nanoparticles were added before passing the

resulting samples through a magnetic column system. The B- and T-lymphocytes counts were

then assessed by flow cytometry. Additionally, the lymphocytes removal experiments were

repeated at different time points, always with blood from the same subject, in order to get

Figure 0-3. Elemental analysis of three batches produced with the same reaction conditions. A) Carbon

content B) Hydrogen content C) Nitrogen content D) Carboxyl functionalities content calculated from

the carbon content (see Appendix A4 for a description of such calculations).

Page 93: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

92

insights about the storability of such nanoparticles. As can be seen on Figure 5-4, all batches

reduced the number of both B- and T-lymphocytes although Batch 3 turned out to be less

efficient than the two first ones having a small excess. Moreover, the nanoparticles retained

their efficiency even after at least 2 months of storage at 5°C. This is an important feature in

the view of an eventual clinical translation. However, after 6 months of storage, the efficiency

was drastically reduced. The variability between the results obtained after 1 and 2 months of

storage could presumably be due to the fact that the CD52 expression might slightly differ at

various time points even for a single subject. Of note, the removal efficiency was considerable

for both B- and T-lymphocytes although there is an order of magnitude difference between the

two types of cells.

5.3.4 B- and T-Lymphocytes Removal from Cancer Patient Blood Samples

To explore the robustness of the lymphocytes removal procedure, blood samples from cancer

patients were used to repeat the experiments done with healthy subjects samples. Although this

is not therapeutically relevant for the cancers involved to remove a large fraction of the

lymphocytes, it provides insights on the efficiency of the nanoparticles for widely differing

lymphocytes levels and in a disease setting. Indeed, cancer patients exhibit lymphocytes counts

that can be several times lower than healthy subjects. Batch 2 from the experiments with healthy

subject blood was used and yielded efficient lymphocytes removal from the blood samples of

cancer patients (Figure 5-5). However, if the results are encouraging and show that the

Figure 0-4. Removal of B- and T-lymphocytes after treatment of blood samples from healthy subjects.

Campath-functionalized C/Co nanoparticles were added to samples which were passed through a

magnetic column system before measuring lymphocytes counts by flow cytometry. Batches with slight

synthesis changes were produced and stored at 5°C.

Page 94: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

93

nanoparticles are efficient in several contexts, the study would require to be extended to more

relevant types of cancers such as lymphomas. Indeed, the isolation of lymphocytes in the case

of lymphoma could provide a mean of obtaining a liquid biopsy and this could be an alternative

to lymph node biopsy which is the current diagnosis standard.

5.4 Conclusion

In conclusion, we provided insights into the polymerization step, a central aspect for the

successful scale up of the production of polyglycidol-functionalized carbon-coated cobalt

nanoparticles. We also reported data about the reproducibility of the synthesis. In addition, we

disclosed the successful use of Campath-conjugated nanoparticles to remove lymphocytes from

the blood of patients. In this proof-of-principle, the focus was not on specifically removing

either B- or T-lymphocytes, as would require some of the mentioned therapeutic applications,

but rather to show that such nanoparticles are not only suitable to isolate rare cells but abundant

cells as well. However, already the Campath-nanoparticles could find applications in field of

diagnosis where they could allow surgeons to obtain a liquid biopsy, necessary for the diagnosis

of lymphoma, in a less-invasive manner as what is currently done. Finally, by conjugating the

nanoparticles with antibodies that are specific to a given type of lymphocytes, therapeutically

relevant applications could be developed. For instance, by magnetically removing B-cells only,

one could potentially circumvent the resistance mechanism encountered for B-cell depleting

drugs in the treatment of autoimmune diseases.

Figure 0-5. Removal of B- and T-lymphocytes after treatment of blood samples from cancer patients.

Campath-functionalized C/Co nanoparticles were added to samples which were passed through a

magnetic column system before measuring lymphocytes counts by flow cytometry. Patients had widely

varying lymphocytes counts notably with three patients having very low B-lymphocytes counts.

Page 95: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

94

Page 96: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

95

Conclusion and Outlook

This thesis covered two concepts of cancer treatments using nanoparticles. First,

platinum nanoparticles were used as part of a caged prodrug system with applications for lung

cancer. The caged prodrug is a less potent form of a chemotherapeutic that is deactivated by

introducing a chemical protecting that can later be removed in situ thanks to the catalytic

properties of nanoparticles. This system could potentially lead to either a reduction in side-

effects or allow the use of higher drug concentrations and/or more frequent drug administration.

A second concept was presented where polyglycidol-functionalized magnetic nanoparticles

were conjugated with antibodies to remove cells from the blood in a targeted manner. By

removing circulating tumour cells, this could translate into a tool having the potential to reduce

the appearance of metastases, the main cause of cancer deaths. Also, such nanoparticles were

used to remove lymphocytes and could therefore find use in applications such as lymphoma

diagnosis by providing a way to obtain a liquid biopsy and, additionally, in the treatment of

autoimmune diseases thereby avoiding resistance mechanisms of current B-cell depletion

treatments.

In Chapter 2, we could show that the introduction of a protecting group, a “cage”, on

gemcitabine resulted in a caged prodrug that was in vitro much less cytotoxic as compared to

the uncaged counterpart. The system was further composed of platinum nanoparticles and

hydrogen in order to uncage the prodrug in an efficient and timely manner. It could be shown

that the prodrug could recover desired cytoxicity.

Preliminary pre-clinical data for the caged prodrug system were then presented. A

formulation for intratracheal administration was developed and administered to mice which did

not display any subsequent toxicity. Also, at least at a macroscopic level, aqueous platinum

nanoparticles solutions did not trigger toxicity in the mice models. Formulations containing

caged gemcitabine, gemcitabine or the vehicle itself were administered to mice previously

inoculated with bioluminescent A549 cells. In this experiment it was observed that caged

gemcitabine was not toxic to the mice and that it did not provide a significant advantage with

respect to tumour growth as compared to the vehicle. Gemcitabine on the other hand triggered

significant complications and mice prematurely died from non-cancer related causes.

Page 97: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

96

Polyglycidol-functionalized carbon-coated cobalt nanoparticles were used as another

nanomedicine application. First, they were functionalized with an anti-EpCAM antibody to

target circulating tumour cells present in the blood. We could establish a relationship between

the antifouling and separability properties of the nanoparticles and their efficiency to remove

CTCs. Experiments with spiked blood from healthy subjects demonstrated the high efficiency,

selectivity and reproducibility of the CTCs removal procedure. The work was extended by

showing that the nanoparticles do not generate blood coagulation and applying the procedure

to blood samples originating from cancer patients.

In Chapter 5, the polymerization step of the nanoparticles’ functionalization was

studied. This provided key insights that will be necessary for the scale up of the nanoparticles

production in prevision of pre-clinical animal studies. Furthermore, it was shown that the

synthesis is reproducible, thereby fulfilling one of the central requirements of the U.S. FDA for

regulatory approval. The chapter was augmented by demonstrating the use of such

nanoparticles for the removal of lymphocytes from the blood. An interesting perspective given

the vast field of applications ranging from lymphoma diagnosis to autoimmune treatments.

Both technologies require some further development before entering clinical studies.

For the caged prodrug system, a protocol for the in vivo uncaging reaction has to be developed

and thereafter similar mice experiments have to be repeated to determine the feasibility of such

a system to reduce the side-effects of lung chemotherapy and/or improve its efficacy by

increasing the dosage and frequency of chemotherapy administrations. The technology could

be extended to other free amino-containing chemotherapeutics and eventually other triggering

systems could be developed. Regarding the magnetic nanoparticles for specific cell isolation

from blood, the next step will be the scale up of the material production in order to conduct

animal safety tests. There are also some new development ideas such as evolving antibodies

that are specific to the antigen receptors that recognize self-antigens on the surface of immune

cells. This would allow one to selectively remove the pathogenic cells while not reducing the

immune protection of the patients. Additionally, nanoparticles could be developed for which

the captured cells could be released upon the action of a biorthogonal trigger. This could find

applications and facilitating a new trend in research consisting in training immune cells for a

specific target in vitro before reintroducing them in the patient’s circulatory system.

Page 98: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

97

Appendix

Page 99: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

98

A.1 Supplementary Information Chapter 2

Additional Materials and Methods

Screening of Hydrogen Concentrations

Experiment with 3.2% hydrogen

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with hydrogen gas at 0.07 bar and closed. The gas mixture is

further composed of 5% of CO2 and 91.8% of nitrogen. The plate was incubated at 37°C for 0,

0.25, 1, 2, 3 hours in the gas chamber. After each time point, the plate was taken out of the gas

chamber and fluorescence emission was measured using a microplate reader (Tecan, Infinite

200 PRO; ex: 340 nm; em: 460 nm). Values were normalized by the mean value of the positive

controls wells.

Experiment with 20% hydrogen

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with hydrogen gas at 0.07 bar and closed. The gas mixture is

further composed of 5% of CO2 and 75% of nitrogen. The plate was incubated at 37°C for 0, 5,

20, 35, 50, 65, 100, 130, 160, 190 min in the gas chamber. After each time point, the plate was

taken out of the gas chamber and fluorescence emission was measured using a microplate reader

(Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). Values were normalized by the mean value

of the positive controls wells.

Page 100: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

99

Experiment with 100% hydrogen

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with hydrogen gas at 0.07 bar and closed. The plate was

incubated at 37°C for 0, 5, 10, 15, 20, 25, 30, 150 min in the gas chamber. After each time

point, the plate was taken out of the gas chamber and fluorescence emission was measured

using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). Values were

normalized by the mean value of the positive controls wells.

Screening of Oxygen Concentrations

Experiment with 0% oxygen

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with the gas mixture at 0.07 bar and closed. The gas mixture

is further composed of 90% of hydrogen and 10% of nitrogen. The plate was incubated at 37°C

for 0, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 90 minutes in the gas chamber. After each

time point, the plate was taken out of the gas chamber and fluorescence emission was measured

using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). Values were

normalized by the mean value of the positive controls wells.

Experiment with 1% oxygen

Page 101: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

100

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with the gas mixture at 0.07 bar and closed. The gas mixture

is further composed of 90% of hydrogen and 9% of nitrogen. The plate was incubated at 37°C

for 0, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 90 minutes in the gas chamber. After each

time point, the plate was taken out of the gas chamber and fluorescence emission was measured

using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). Values were

normalized by the mean value of the positive controls wells.

Experiment with 2.1% oxygen

In a 96-well plate, wells were filled with 90 L of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (110 M) and DMSO (2% v/v). Positive controls wells were filled

with 90 L of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (110 µM) and

DMSO (2% v/v). To treated wells, 10 L of a platinum nanoparticles solution (1000 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with the gas mixture at 0.07 bar and closed. The gas mixture

is further composed of 90% of hydrogen and 7.9% of nitrogen. The plate was incubated at 37°C

for 0, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 90 minutes in the gas chamber. After each

time point, the plate was taken out of the gas chamber and fluorescence emission was measured

using a microplate reader (Tecan, Infinite 200 PRO; ex: 340 nm; em: 460 nm). Values were

normalized by the mean value of the positive controls wells.

Toxicity Study of Platinum Nanoparticles and Hydrogen

A549 cells were seeded in 96-well plates at a concentration of 4,000 cells/well and incubated

overnight at 37°C before treatment. The supernatant of each well was then replaced with 100

L of fresh F-12K medium and DMSO (2% v/v). All cell culture media were supplemented

Page 102: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

101

with 10% Foetal Bovine Serum. Plates were incubated at 37°C for 1 h before 10 L of a

platinum nanoparticles solution (1, 5 or 10 ppm; 3 nm diameter; Sigma-Aldrich) was added to

each well and 10 L of Milli-Q water was added to control wells. Six biological replicates for

each condition were included in the plates. Plates were incubated at 37°C for 1 h before placing

them in a custom-designed gas chamber (see Scheme A1-1). A control plate was left untouched

in the incubator. The chamber was placed in the incubator, connected to a gas bottle outside the

incubator and to an outlet. The gas chamber was flushed for 1 min with hydrogen gas at 0.07

bar and closed. Plates were incubated at 37°C for 2 h in the gas chamber before placing them

back in the incubator with the control plate. Control and treated plates were incubated at 37°C

for 72 h before PrestoBlue cell viability reagent (10% v/v) was added to each well and the plate

incubated at 37°C for 2 h. Fluorescence emission was measured using a microplate reader

(Tecan, Infinite 200 PRO; ex: 540 nm; em: 590 nm). Values were normalized by the mean value

of the positive controls wells.

Kinetics of Deprotection of Caged Gemcitabine

In a 96-well plate, wells were filled with 100 L of fresh F-12K medium containing caged

gemcitabine (0.1 M) and DMSO (2% v/v). All cell culture media were supplemented with

10% Foetal Bovine Serum. To treated wells, 10 L of a platinum concentration (200 ppm, 3

nm diameter, Sigma-Aldrich) was added and 10 L of Milli-Q water was added to control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 1 min with hydrogen gas at 0.07 bar and closed. The plate was

incubated at 37°C for 0, 15, 30, 120 min in the gas chamber before placing them back in the

incubator with the control plates. After each iteration, the plate was taken out of the gas chamber

and the content of 6 wells were pooled together in an Eppendorf tube. The plate was placed

back in the gas chamber and back in the incubator, flushed for 1 min with hydrogen gas at 0.07

bar and closed until the next sampling. The Eppendorf tubes collected were centrifuged at

21130 g for 1 h and the supernatant was transferred to MS sample vials before measurement

with an HPLC-MS (HPLC: Agilent 1260 Infinity, MS: Agilent 6130).

Re-use of Platinum Nanoparticles after Recycling

In a 24-well plate, wells were filled with 1.5 mL of fresh F-12K medium containing caged 7-

amino-4-methylcoumarin (100 µM) and DMSO (2% v/v). Positive controls wells were filled

Page 103: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

102

with 1.5 mL of fresh F-12K medium containing caged 7-amino-4-methylcoumarin (100 µM)

and DMSO (2% v/v). All cell culture media were supplemented with 10% Foetal Bovine Serum.

To treated wells, 150 µL of a platinum nanoparticles solution (1000 ppm, 3 nm diameter,

Sigma-Aldrich) was added and 150 µL of Milli-Q water was added to negative control wells.

The plate was placed in a custom-designed gas chamber (see Scheme A1-1). The chamber was

placed in the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas

chamber was flushed for 0.5 min with hydrogen gas at 0.07 bar and closed. The plate was

incubated at 37°C for 40 min in the gas chamber. Fluorescence emission was measured using a

microplate reader (Tecan, Spark 10M; ex: 340 nm; em: 460 nm). Values were normalized by

the mean value of the positive controls wells. The wells content was transferred to 1.5 mL

Eppendorf tubes and were centrifuged for 1.5h at 22130g (Eppendorf, Centrifuge 5424 R). The

supernatant was discarded and replaced by 1.5 mL of fresh solution. The nanoparticles were

redispersed with an ultrasonic horn (Hielsher, UP50H; 30 kHz). The content of the Eppendorf

tubes was transferred to the 24-well plate before it was placed back in the incubator and another

cycle was done. Ten such cycles were made.

Extracellular Prodrug Activation in Normal Cells

The surface of 96-well plates was modified by adding to each well 50 L of a 0.02 mg/mL

poly-L-lysine solution. The plates were incubated overnight at 37°C and were washed twice

with 60 L of Milli-Q water. Human Pulmonary Alveolar Epithelial (HPAEpiC) cells were

seeded in 96-well plates at a concentration of 4,000 cells/well and incubated overnight at 37°C

before treatment. The supernatant of each well was then replaced with 100 L of fresh

HPAEpiC medium containing protected gemcitabine or gemcitabine (0.05 M) and DMSO

(2% v/v), control wells were replaced with 100 L of fresh HPAEpiC medium and DMSO (2%

v/v). All cell culture media were supplemented with 10% Foetal Bovine Serum. Plates were

incubated at 37°C for 1 h before 10 L of a platinum nanoparticles solution (100 ppm, 3 nm

diameter, Sigma-Aldrich) was added to each well and 10 L of Milli-Q water was added to

control wells. Five biological replicates for each condition were included in the plates. Plates

were incubated at 37°C for 1 h before placing them in a custom-designed gas chamber (see

Scheme A1-1). Control plates were left untouched in the incubator. The chamber was placed in

the incubator, connected to a gas bottle outside the incubator and to an outlet. The gas chamber

was flushed for 1 min with hydrogen gas at 0.07 bar and closed. Plates were incubated at 37°C

for 2 h in the gas chamber before placing them back in the incubator with the control plates.

Page 104: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

103

Control and treated plates were incubated at 37°C for 48 h before PrestoBlue cell viability

reagent (10% v/v) was added to each well and the plate incubated at 37°C for 2 h. Fluorescence

emission was measured using a microplate reader (Tecan, Spark 10M; ex: 560 nm; em: 590

nm). Values were normalized by the mean value of the positive controls wells.

Page 105: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

104

Scheme A1-1. Plan of the custom-designed box. The box is made out of aluminium. It is sealed thanks to a toric

joint and screws that maintain the cover tight. One can place a typical well plate inside.

Page 106: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

105

Additional Results and Discussion

Screening of Catalyst Concentrations

To determine the catalyst concentration required for the uncaging reaction to take place, a

screening of several platinum concentrations was done (Figure A1-1). Above a concentration

of 25 ppm, the uncaging is particularly fast with the reaction reaching completion within about

20 minutes. At a concentration of 12.5 ppm, the maximum is reached after two hours.

Additionally, such a reaction rate is obtained without stirring, nor bubbling and is thus

particularly fast for a reaction that is only diffusion-controlled. In order to reduce the risk of

platinum-induced toxicity, the concentration for cell experiments was reduced to 10 ppm. It is

noteworthy that hydrogen without platinum (0 ppm Pt curve on Figure A1-1) does not lead to

any conversion. This feature translates into a background that is particularly low.

Figure A1-1. Kinetics of deprotection of prodrug surrogate 2a for various catalyst concentrations. A

solution of 99 µM of caged coumarin was prepared in Ham’s F-12K (Kaighn’s) Medium (F-12K). Pt

nanoparticles concentrations of 0, 12.5, 25, and 50 ppm and pure hydrogen were used. Fluorescence

was measure with a plate reader (n = 5; 𝜆ex/em = 340/460 nm).

Page 107: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

106

Screening of Hydrogen Concentrations

To study the influence of the hydrogen concentration present in the gas mixture, a screening of

3 hydrogen concentrations was done (3.2%, 20% and 100%, Figure A1-2). The kinetics is

extensively affected by the hydrogen level and the reaction half-life is shifted from about 10

minutes for 100% hydrogen to about an hour for 20% hydrogen while for 3.2% hydrogen,

completion was not reached within the timeframe of the experiment. Among the three options,

we opted for the fast kinetics and carried out an interval kinetics experiment (Figure 2-4) that

mimics alternating breathing regimen in order to cope with the absence of oxygen in the gas

mixture.

Figure A1-2. Kinetics of deprotection of prodrug surrogate 2a for various hydrogen concentrations. A

solution of 99 µM of caged coumarin was prepared in Ham’s F-12K (Kaighn’s) Medium (F-12K)

supplemented with 2% DMSO. Hydrogen concentrations of 3.2, 20 and 100% and a Pt nanoparticles

concentration of 100 ppm were used. Fluorescence was measured with a plate reader (n = 5;

𝜆ex/em = 340/460 nm).

Page 108: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

107

Kinetic of the Uncaging of Caged Gemcitabine

The kinetics of the uncaging reaction was done with caged gemcitabine (Figure A1-3). The

nanoparticles were removed by ultracentrifugation before measuring the supernatant via HPLC-

MS. The reaction turned out to be as fast as with the caged fluorophore. Namely, a half-life of

about 10 minutes was found for the uncaging reaction of caged gemcitabine.

Figure A1-3. Kinetics of deprotection of caged gemcitabine. A solution of 0.1 µM of caged coumarin

was prepared in Ham’s F-12K (Kaighn’s) Medium (F-12K) supplemented with 2% DMSO and 10%

FBS. Pt nanoparticles solutions with concentrations of 20 ppm and pure hydrogen were used. The

reaction was followed by HPLC-MS.

Page 109: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

108

Screening of Oxygen Concentrations

To determine the influence of oxygen on the uncaging reaction to take place, a screening of

oxygen concentrations was done (Figure A1-4). A reduction in kinetics can be observed when

going from 0 to 1% of oxygen. When increasing the oxygen fraction to 2.1%, the extent of

reaction is reduced. As mentioned in the manuscript, it has been reported that the median

percentage of oxygen in lung tumours were 1.9 and 2.2% of oxygen while normal tissues had

an average of 5.6%. The exact values cannot be reproduced as such due to gas supplier’s internal

regulations implying that no gas mixtures can be manufactured with oxygen concentration

higher than 2.1% if the hydrogen concentration is above 5%. However, the trends are expected

to be similar with higher oxygen concentrations. Namely, the higher the oxygen concentration,

the slower and less extensive the uncaging reaction.

Figure A1-4. Kinetics of deprotection of prodrug surrogate 2a for various oxygen concentrations. A

solution of 99 µM of caged coumarin was prepared in Ham’s F-12K (Kaighn’s) Medium (F-12K)

supplemented with 2% DMSO. Oxygen concentrations of 0, 1 and 2.1% and a Pt nanoparticles

concentration of 100 ppm were used. A hydrogen concentration of 90% was used. Fluorescence was

measured with a plate reader (n = 5; 𝜆ex/em = 340/460 nm).

Page 110: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

109

Toxicity of Platinum Nanoparticles and Hydrogen

The toxicity of hydrogen and of platinum was studied (Figure A1-5). The addition of platinum

did not hinder the growth of the cells and even seemed to slightly increase it for the two lowest

concentrations. The impact of hydrogen administration even over an extended period of time

(2h) did not seem to impair cellular growth. When both were combined, the viability remained

close to the positive control (no Pt, no H2).

Figure A1-5. Influence of platinum and of hydrogen on A549 cell viability. The F-12K cell medium was

supplemented with 2% DMSO and 10% FBS. A custom-designed gas chamber was used for incubation

with hydrogen (2h). After 72h of incubation in normal cell culture atmosphere, the viability was evaluated

using the PrestoBlue™ assay and a plate reader (n = 6; 𝜆ex/em = 560/590 nm).

Page 111: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

110

Extracellular Prodrug Activation in Normal Cells

The extracellular prodrug activation experiment was repeated in normal cells (Figure A1-6).

Gemcitabine being an untargeted chemotherapy, the results obtained are similar to the ones

with cancer cells. As is usually the case in the field of caged prodrugs, the selectivity originates

from the spatial restriction of the catalyst. However, the system here introduced is sensitive to

the oxygen level (Figure A1-4) and partial selectivity might thus arise from the different oxygen

tensions that are found in normal and cancer tissues.

Figure A1-6. Extracellular caged prodrug activation. Human pulmonary alveolar epithelial

(HPAEpiC) cells were used. Solutions of gemcitabine (1a) and caged gemcitabine (1b) were prepared

at a concentration of 0.05 µM in the HPAEpiC cell medium. A concentration of platinum nanoparticles

of 10 ppm was used. A custom-designed gas chamber was used for incubation with hydrogen (2h). After

48h of incubation in normal cell culture atmosphere, the viability was evaluated using the PrestoBlue™

assay and a plate reader (n = 5; 𝜆ex/em = 560/590 nm).

Page 112: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

111

Reuse of Platinum Nanoparticles after Recycling

The platinum nanoparticles were used for the uncaging of caged coumarin. The reaction was

done in F-12K medium with 100% hydrogen. After each reaction cycle, the nanoparticles were

recycled by ultracentrifugation. The activity is slowly decreasing over time (Figure A1-7).

Figure A1-7. Reuse of platinum nanoparticles for the deprotection of prodrug surrogate 2a. A solution

of 99 µM of caged coumarin was prepared in Ham’s F-12K (Kaighn’s) Medium (F-12K) supplemented

with 2% DMSO. A hydrogen concentration of 100% and a Pt nanoparticles concentration of 100 ppm

were used. Fluorescence was measured with a plate reader (n = 6; 𝜆ex/em = 340/460 nm).

Page 113: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

112

A.2 Supplementary Information Chapter 3

Macroscopic Observation of the Lungs after Tolerability Profile Experiments

Figure A2-1. Macroscopic observations of the lung after the tolerability profile experiments. Group 1

mice were administered the vehicle. Group 2 mice were administered a 10 ppm aqueous platinum

nanoparticles solution. Group 3 mice were administered a 100 ppm aqueous platinum nanoparticles

solution.

Page 114: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

113

A.3 Supplementary Information Chapter 4

Additional Materials and Methods

Particle Size Distribution

An aqueous solution of carbon-coated cobalt nanoparticles (28 nm diameter, NanoAmor) was

prepared at a concentration of 5 ppm in Milli-Q water. 1.5 µL of the solution was deposited on

carbon-coated copper grids. After overnight drying, the samples were measured on a Tecnai

F30 (FEI, 300 kV). The quantification was done for 428 nanoparticles using an image

processing program (ImageJ, NIH).

Elemental Analysis

Carbon, hydrogen and nitrogen content of the samples were measured by vario MICRO cube

(Elementar). The micro analyzer was calibrated with sulfanilamide. 2 mg of sample were

oxidized/pyrolized over a period of 70 seconds (standard program: 2mg70s). All samples were

double determined.

Infrared Spectroscopy

The nanoparticles were analyzed by FTIR spectroscopy (5% in KBr using a Tensor 27

Spectrometer, Bruker Optics equipped with a diffuse reflectance accessory, DiffusIR™, Pike

Technologies, 200 scans). The measurements were usually done against precursor material (5%

in KBr, as well).

Rotational Thromboelastometry Measurements

Blood from healthy subjects was supplemented with a nanoparticles solution to a final

concentration of 2.38 mg/mL, which had been previously sonicated (5x1 min with 1 min breaks

in between) in an ultrasonication bath (Sonorex Digital 10P, Bandelin) on ice-cold water. A

control group was treated adding only the carrier solution phosphate-buffered saline (PBS). The

samples were incubated for 2 min on an orbital shaker (16 mot/min, WS 10, Edmund Bühler)

before nanoparticles were removed with the help of an external magnet. The blood was then

used to measure EXTEM and INTEM on a rotational thromboelastometry device (ROTEM

sigma, Tem Innovations).

Page 115: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

114

Additional Results and Discussion

Removal Efficiency of pSPM-functinalized Nanoparticles

In early attempts, the previous generation of pSPM particles was used. These were poorly

functioning for CTC removal application. It was empirically determined that this was due to

their low separability within our setup, which was confirmed in the magnetic separability

experiment

Particles Size Distribution

To determine the particle size distribution, a transmission electronic microscope and an image

processing software (ImageJ) were used. From 428 nanoparticles, an average diameter size of

37.2 ± 27.5 nm was determined.

Figure A3-1. Nanoparticle separability. This graph includes the data from the previous generation

pSPM nanoparticles.

Page 116: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

115

Materials Reproducibility

To demonstrate that the overall synthesis protocol is robust, three nanoparticles batches were

produced starting from naked carbon-coated cobalt nanoparticles. All batches exhibited similar

compositions after a 4-step synthesis thereby illustrating the robustness of the procedure.

Figure A3-2. Particle size distribution. A transmission electronic microscope was used to take several

micrographs. The images were analysed using ImageJ and particle size distribution was determined

using 428 nanoparticles.

Table A3-1. Nanoparticles manufacture reproducibility. The compositions of the three manufactured batches were

determined by elemental analysis. Based o the compositions, the amount of carboxyl functionalities could be

determined.

Page 117: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

116

Influence of Magnetic Nanoparticles Treatment on other Blood Cells

To assess the effect of magnetic nanoparticles treatment on other blood cells, filtrates from three

blood experiments using anti-EpCAM functionalised nanoparticles were subject to flow

cytometry analysis. In an overview, no major differences were observed for the different cell

types such as granulocytes, monocytes and lymphocytes.

Rotational Thromboelastometry Measurements

To assess the effect of magnetic nanoparticles treatment blood coagulation, rotational

thromboelastometry measurements were performed with blood from healthy subjects

supplemented with anti-EpCAM nanoparticles (concentration identical to the one used in CTC

removal experiments). A control was made with PBS only. Results represent one of several

experiments from different subjects, which are found within the normal range.

Figure A3-3. Flow cytometry results after treatment of blood with anti-EpCAM-functionalised nanoparticles

focusing on granulocytes, monocytes and lymphocytes.

Page 118: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

117

EXTEM INTEM

CT CFT MCF CT CFT MCF

(s)

______________

(s)

______________

(mm)

______________

(s)

______________

(s)

______________

(mm)

______________

Control 69 95 61 177 72 60

anti-EpCAM

particles

74 116 55 207 71 76

Normal range [1] 38-79 34-159 50-72 100-240 30-110 50-72

Table A3-2. Rotational thrombolelastometry measurement in blood from a healthy subject. The

measurements were made in blood supplemented with either anti-EpCAM nanoparticles or PBS

(negative control). Clotting time (CT), clot formation time (CFT) and maximum clot firmness (MCF) for

the EXTEM channel (activation via tissue factor) and for the INTEM channel (contact activation) are

displayed. Reference: [1] Lang T, Bauters A, Braun SL, Pötzsch B, von Pape KW, Kolde HJ, Lakner M.

Multi-centre investigation on reference ranges for ROTEM thromboelastometry. Blood Coagul

Fibrinolysis. 2005 Jun;16(4):301-10.

Infrared Spectroscopy

Figure A3-4. Infrared Spectrum from anti-EpCAM nanoparticles Batch 1.

Page 119: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

118

Figure A3-5. Infrared Spectrum from anti-EpCAM nanoparticles Batch 2.

Figure A3-6. Infrared Spectrum from anti-EpCAM nanoparticles Batch 3.

Page 120: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

119

A.4 Supplementary Information Chapter 5

Elemental Analysis after the Various Synthesis Steps

Nanoparticles Type Variable

changed

Amount C

(%)

Amount N

(%)

Amount H

(%)

Amount S

(%)

C/Co - 4.11 0.09 0.04 0

C/Co-PhEtO-Na+ - 5.33 0.23 0.11 0

C/Co@polyglycidin Standard 9.55 1.54 0.21 0

C/Co@polyglycidin 5 mL 8.37 0.95 0.09 0

C/Co@polyglycidin 15 ml 10.04 1.29 0.09 0

C/Co@polyglycidin 90°C 6.48 0.69 0.23 0

C/Co@polyglycidin 190°C 12.5 1.77 0.26 0

C/Co@polyglycidin 7700 rpm 8.75 0.98 0.11 0

C/Co@polyglycidin 25000 rpm 10.65 1.56 0.23 0

Calculations of the Number of Polymer Units (example for standard synthesis)

Determination of the molar amount of carbon introduced per theoretical gram of particle:

(𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝐶𝑎𝑓𝑡𝑒𝑟 𝑝𝑜𝑙𝑦𝑚𝑒𝑟𝑖𝑧𝑎𝑡𝑖𝑜𝑛 − 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝐶𝑏𝑒𝑓𝑜𝑟𝑒 𝑝𝑜𝑙𝑦𝑚𝑒𝑟𝑖𝑧𝑎𝑡𝑖𝑜𝑛)

𝐴𝑡𝑜𝑚𝑖𝑐 𝑤𝑒𝑖𝑔ℎ𝑡 𝑜𝑓 𝑐𝑎𝑟𝑏𝑜𝑛

=(0.0955𝑔 − 0.0518𝑔)

12 𝑔/𝑚𝑜𝑙= 3.6 𝑚𝑚𝑜𝑙 𝐶/𝑔

Determination of the amount of polymer repeating units per theoretical gram of particle:

𝑚𝑜𝑙𝑎𝑟 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝐶 𝑖𝑛𝑡𝑟𝑜𝑑𝑢𝑐𝑒𝑑

𝑛𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝐶 𝑎𝑡𝑜𝑚𝑠 𝑐𝑜𝑛𝑡𝑎𝑖𝑛𝑒𝑑 𝑖𝑛 𝑜𝑛𝑒 𝑟𝑒𝑝𝑒𝑎𝑡𝑖𝑛𝑔 𝑢𝑛𝑖𝑡=

3.6 𝑚𝑚𝑜𝑙 𝐶/𝑔

3

= 1.2 𝑚𝑚𝑜𝑙 𝑜𝑓 𝑟𝑒𝑝𝑒𝑎𝑡𝑖𝑛𝑔 𝑢𝑛𝑖𝑡𝑠/𝑔

Page 121: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

120

References

(1) World Health Organization, Cancer Fact Sheets http://www.who.int/news-room/fact-

sheets/detail/cancer (accessed Aug 19, 2018).

(2) Stewart, B. W.; Wild, C. P. World Cancer Report 2014; IARC 2014.

(3) Ferlay, J.; Soerjomataram, I.; Ervik, M.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.;

Parkin, D. M.; Forman, D.; Bray, F. Cancer Incidence and Mortality Worldwide: IARC

CancerBase http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx (accessed Aug 19,

2018).

(4) Anand, P.; Kunnumakara, A. B.; Sundaram, C.; Harikumar, K. B.; Tharakan, S. T.; Lai,

O. S.; Sung, B.; Aggarwal, B. B. Cancer Is a Preventable Disease That Requires Major

Lifestyle Changes. Pharm. Res. 2008, 25, 2097–2116.

(5) Wu, S.; Powers, S.; Zhu, W.; Hannun, Y. A. Substantial Contribution of Extrinsic Risk

Factors to Cancer Development. Nature 2016, 529, 43–47.

(6) National Institue of Health, What is cancer? https://www.cancer.gov/about-

cancer/understanding/what-is-cancer (accessed Aug 19, 2018).

(7) Negrini, S.; Gorgoulis, V. G.; Halazonetis, T. D. Genomic Instability — an Evolving

Hallmark of Cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 220–228.

(8) Hanahan, D.; Weinberg, R. A. The Hallmarks of Cancer. Cell 2000, 100, 57–70.

(9) Hanahan, D.; Weinberg, R. A. Hallmarks of Cancer: The next Generation. Cell 2011,

144, 646–674.

(10) Friedl, P.; Wolf, K. Tube Travel: The Role of Proteases in Individual and Collective

Cancer Cell Invasion. Cancer Res. 2008, 68, 7247–7249.

(11) Friedl, P.; Wolf, K. Plasticity of Cell Migration: A Multiscale Tuning Model. J. Cell

Biol. 2010, 188, 11–19.

(12) Madsen, C. D.; Sahai, E. Cancer Dissemination—Lessons from Leukocytes. Dev. Cell

2010, 19, 13–26.

(13) Sabeh, F.; Shimizu-Hirota, R.; Weiss, S. J. Protease-Dependent versus -Independent

Page 122: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

121

Cancer Cell Invasion Programs: Three-Dimensional Amoeboid Movement Revisited. J.

Cell Biol. 2009, 185, 11–19.

(14) Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix Metalloproteinases: Regulators of the

Tumor Microenvironment. Cell 2010, 141, 52–67.

(15) Yao, D.; Dai, C.; Peng, S. Mechanism of the Mesenchymal-Epithelial Transition and Its

Relationship with Metastatic Tumor Formation. Mol. Cancer Res. 2011, 9, 1608–1620.

(16) Carrillo-Infante, C.; Abbadessa, G.; Bagella, L.; Giordano, A. Viral Infections as a Cause

of Cancer (Review). Int. J. Oncol. 2007, 30, 1521–1528.

(17) Breasted, J. H. The Edwin Smith Surgical Papyrus, 1930, 3.

(18) Hyung, W. J.; Kim, S. S.; Choi, W. H.; Cheong, J. H.; Choi, S. H.; Kim, C. B.; Noh, S.

H. Changes in Treatment Outcomes of Gastric Cancer Surgery over 45 Years at a Single

Institution. Yonsei Med. J. 2008, 49, 409–415.

(19) Osaki, T.; Oyama, T.; Takenoyama, M.; So, T.; Yamashita, T.; Aikawa, M.; Ono, K.;

Yasumoto, K. [Results of Surgical Treatment for Primary Lung Cancer; Time Trends of

Survival and Clinicopathologic Features]. J. UOEH 2001, 23, 277–283.

(20) Veronesi, U.; Banfi, A.; Salvadori, B.; Luini, A.; Saccozzi, R.; Zucali, R.; Marubini, E.;

Del Vecchio, M.; Boracchi, P.; Marchini, S.; et al. Breast Conservation Is the Treatment

of Choice in Small Breast Cancer: Long-Term Results of a Randomized Trial. Eur. J.

Cancer Clin. Oncol. 1990, 26, 668–670.

(21) Myers, G. J. C.; Abudu, A. T.; Carter, S. R.; Tillman, R. M.; Grimer, R. J. Endoprosthetic

Replacement of the Distal Femur for Bone Tumours. J. Bone Joint Surg. Br. 2007, 89–

B, 521–526.

(22) (EBCTCG)., E. B. C. T. C. G. Effect of Radiotherapy after Breast-Conserving Surgery

on 10-Year Recurrence and 15-Year Breast Cancer Death: Meta-Analysis of Individual

Patient Data for 10 801 Women in 17 Randomised Trials. Lancet 2011, 378, 1707–1716.

(23) Wyld, L.; Audisio, R. A.; Poston, G. J. The Evolution of Cancer Surgery and Future

Perspectives. Nat. Rev. Clin. Oncol. 2015, 12, 115–124.

(24) Chi, C.; Du, Y.; Ye, J.; Kou, D.; Qiu, J.; Wang, J.; Tian, J.; Chen, X. Intraoperative

Imaging-Guided Cancer Surgery: From Current Fluorescence Molecular Imaging

Page 123: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

122

Methods to Future Multi-Modality Imaging Technology. Theranostics 2014, 4, 1072–

1084.

(25) Koch, M.; Kienle, P.; Hinz, U.; Antolovic, D.; Schmidt, J.; Herfarth, C.; von Knebel

Doeberitz, M.; Weitz, J. Detection of Hematogenous Tumor Cell Dissemination Predicts

Tumor Relapse in Patients Undergoing Surgical Resection of Colorectal Liver

Metastases. Ann. Surg. 2005, 241, 199–205.

(26) Taniguchi, T.; Makino, M.; Suzuki, K.; Kaibara, N. Prognostic Significance of Reverse

Transcriptase-Polymerase Chain Reaction Measurement of Carcinoembryonic Antigen

MRNA Levels in Tumor Drainage Blood and Peripheral Blood of Patients with

Colorectal Carcinoma. Cancer 2000, 89, 970–976.

(27) Baumann, M.; Krause, M.; Overgaard, J.; Debus, J.; Bentzen, S. M.; Daartz, J.; Richter,

C.; Zips, D.; Bortfeld, T. Radiation Oncology in the Era of Precision Medicine. Nat. Rev.

Cancer 2016, 16, 234–249.

(28) Lagendijk, J. J. W.; Raaymakers, B. W.; Raaijmakers, A. J. E.; Overweg, J.; Brown, K.

J.; Kerkhof, E. M.; van der Put, R. W.; Hårdemark, B.; van Vulpen, M.; van der Heide,

U. A. MRI/Linac Integration. Radiother. Oncol. 2008, 86, 25–29.

(29) Bentzen, S. M.; Overgaard, J. Patient-to-Patient Variability in the Expression of

Radiation-Induced Normal Tissue Injury. Semin. Radiat. Oncol. 1994, 4, 68–80.

(30) Paganetti, H.; Athar, B. S.; Moteabbed, M.; A Adams, J.; Schneider, U.; Yock, T. I.

Assessment of Radiation-Induced Second Cancer Risks in Proton Therapy and IMRT

for Organs inside the Primary Radiation Field. Phys. Med. Biol. 2012, 57, 6047–6061.

(31) Chabner, B. A.; Roberts, T. G. Timeline: Chemotherapy and the War on Cancer. Nat.

Rev. Cancer 2005, 5, 65–72.

(32) Gilman, A. The Initial Clinical Trial of Nitrogen Mustard. Am. J. Surg. 1963, 105, 574–

578.

(33) Mini, E.; Nobili, S.; Caciagli, B.; Landini, I.; Mazzei, T. Cellular Pharmacology of

Gemcitabine. Ann. Oncol. 2006, 17, v7–v12.

(34) Huang, M.; Shen, A.; Ding, J.; Geng, M. Molecularly Targeted Cancer Therapy: Some

Lessons from the Past Decade. Trends Pharmacol. Sci. 2014, 35, 41–50.

Page 124: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

123

(35) Marusyk, A.; Polyak, K. Tumor Heterogeneity: Causes and Consequences. Biochim.

Biophys. Acta - Rev. Cancer 2010, 1805, 105–117.

(36) Turner, N. C.; Reis-Filho, J. S. Genetic Heterogeneity and Cancer Drug Resistance.

Lancet Oncol. 2012, 13, e178–e185.

(37) Sharma, S. V.; Lee, D. Y.; Li, B.; Quinlan, M. P.; Takahashi, F.; Maheswaran, S.;

McDermott, U.; Azizian, N.; Zou, L.; Fischbach, M. A.; et al. A Chromatin-Mediated

Reversible Drug-Tolerant State in Cancer Cell Subpopulations. Cell 2010, 141, 69–80.

(38) Hartmann, J.; Haap, M.; Kopp, H.-G.; Lipp, H.-P. Tyrosine Kinase Inhibitors – A

Review on Pharmacology, Metabolism and Side Effects. Curr. Drug Metab. 2009, 10,

470–481.

(39) Broekman, F.; Giovannetti, E.; Peters, G. J. Tyrosine Kinase Inhibitors: Multi-Targeted

or Single-Targeted? World J. Clin. Oncol. 2011, 2, 80–93.

(40) Hillen, F.; Griffioen, A. W. Tumour Vascularization: Sprouting Angiogenesis and

Beyond. Cancer Metastasis Rev. 2007, 26, 489–502.

(41) El-Kenawi, A. E.; El-Remessy, A. B. Angiogenesis Inhibitors in Cancer Therapy:

Mechanistic Perspective on Classification and Treatment Rationales. Br. J. Pharmacol.

2013, 170, 712–729.

(42) Oda, K.; Matsuoka, Y.; Funahashi, A.; Kitano, H. A Comprehensive Pathway Map of

Epidermal Growth Factor Receptor Signaling. Mol. Syst. Biol. 2005, 1, 2005.0010.

(43) Harari, P. M. Epidermal Growth Factor Receptor Inhibition Strategies in Oncology.

Endocr. Relat. Cancer 2004, 11, 689–708.

(44) Sj Costa, D.; Mercieca-Bebber, R.; Rutherford, C.; Gabb, L.; King, M. T. The Impact of

Cancer on Psychological and Social Outcomes. 2016.

(45) Pitman, A.; Suleman, S.; Hyde, N.; Hodgkiss, A. Depression and Anxiety in Patients

with Cancer. BMJ 2018, 361, k1415.

(46) Lai, J.-S.; Butt, Z.; Wagner, L.; Sweet, J. J.; Beaumont, J. L.; Vardy, J.; Jacobsen, P. B.;

Shapiro, P. J.; Jacobs, S. R.; Cella, D. Evaluating the Dimensionality of Perceived

Cognitive Function. J. Pain Symptom Manage. 2009, 37, 982–995.

Page 125: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

124

(47) Koppelmans, V.; Breteler, M. M. B.; Boogerd, W.; Seynaeve, C.; Schagen, S. B. Late

Effects of Adjuvant Chemotherapy for Adult Onset Non-CNS Cancer; Cognitive

Impairment, Brain Structure and Risk of Dementia. Crit. Rev. Oncol. Hematol. 2013, 88,

87–101.

(48) Kangas, M.; Henry, J. L.; Bryant, R. A. Posttraumatic Stress Disorder Following Cancer:

A Conceptual and Empirical Review. Clin. Psychol. Rev. 2002, 22, 499–524.

(49) Mercadante, S.; Vitrano, V.; Catania, V. Sexual Issues in Early and Late Stage Cancer:

A Review. Support. Care Cancer 2010, 18, 659–665.

(50) Carrera, P. M.; Kantarjian, H. M.; Blinder, V. S. The Financial Burden and Distress of

Patients with Cancer: Understanding and Stepping-up Action on the Financial Toxicity

of Cancer Treatment. CA. Cancer J. Clin. 2018, 68, 153–165.

(51) Zafar, S. Y.; Peppercorn, J. M.; Schrag, D.; Taylor, D. H.; Goetzinger, A. M.; Zhong,

X.; Abernethy, A. P. The Financial Toxicity of Cancer Treatment: A Pilot Study

Assessing out-of-Pocket Expenses and the Insured Cancer Patient’s Experience.

Oncologist 2013, 18, 381–390.

(52) Smith, A. D. Big Moment for Nanotech: Oncology Therapeutics Poised for a Leap.

OncLive. 2013,.

(53) Shi, J.; Kantoff, P. W.; Wooster, R.; Farokhzad, O. C. Cancer Nanomedicine: Progress,

Challenges and Opportunities. Nat. Rev. Cancer 2017, 17, 20–37.

(54) Petersen, G. H.; Alzghari, S. K.; Chee, W.; Sankari, S. S.; La-Beck, N. M. Meta-Analysis

of Clinical and Preclinical Studies Comparing the Anticancer Efficacy of Liposomal

versus Conventional Non-Liposomal Doxorubicin. J. Control. Release 2016, 232, 255–

264.

(55) Lancet, J. E.; Rizzieri, D.; Schiller, G. J.; Stuart, R. K.; Kolitz, J. E.; Solomon, S. R.;

Newell, L. F.; Erba, H. P.; Uy, G. L.; Ryan, R.; et al. Overall Survival (OS) with CPX-

351 versus 7+3 in Older Adults with Newly Diagnosed, Therapy-Related Acute Myeloid

Leukemia (TAML): Subgroup Analysis of a Phase III Study. J. Clin. Oncol. 2017, 35,

7035.

(56) Lv, S.; Tang, Z.; Li, M.; Lin, J.; Song, W.; Liu, H.; Huang, Y.; Zhang, Y.; Chen, X. Co-

Delivery of Doxorubicin and Paclitaxel by PEG-Polypeptide Nanovehicle for the

Page 126: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

125

Treatment of Non-Small Cell Lung Cancer. Biomaterials 2014, 35, 6118–6129.

(57) Ediriwickrema, A.; Zhou, J.; Deng, Y.; Saltzman, W. M. Multi-Layered Nanoparticles

for Combination Gene and Drug Delivery to Tumors. Biomaterials 2014, 35, 9343–9354.

(58) Devulapally, R.; Sekar, N. M.; Sekar, T. V.; Foygel, K.; Massoud, T. F.; Willmann, J.

K.; Paulmurugan, R. Polymer Nanoparticles Mediated Codelivery of AntimiR-10b and

AntimiR-21 for Achieving Triple Negative Breast Cancer Therapy. ACS Nano 2015, 9,

2290–2302.

(59) Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-Responsive Nanocarriers for Drug Delivery.

Nat. Mater. 2013, 12, 991–1003.

(60) Duan, X.; Xiao, J.; Yin, Q.; Zhang, Z.; Yu, H.; Mao, S.; Li, Y. Smart PH-Sensitive and

Temporal-Controlled Polymeric Micelles for Effective Combination Therapy of

Doxorubicin and Disulfiram. ACS Nano 2013, 7, 5858–5869.

(61) Wu, J.; Zhao, L.; Xu, X.; Bertrand, N.; Choi, W. I.; Yameen, B.; Shi, J.; Shah, V.;

Mulvale, M.; MacLean, J. L.; et al. Hydrophobic Cysteine Poly(Disulfide)-Based

Redox-Hypersensitive Nanoparticle Platform for Cancer Theranostics. Angew. Chemie

Int. Ed. 2015, 54, 9218–9223.

(62) Shi, V. J.; Levy, L. L.; Choi, J. N. Cutaneous Manifestations of Nontargeted and

Targeted Chemotherapies. Semin. Oncol. 2016, 43, 419–425.

(63) Björnmalm, M.; Thurecht, K. J.; Michael, M.; Scott, A. M.; Caruso, F. Bridging Bio–

Nano Science and Cancer Nanomedicine. ACS Nano 2017, 11, 9594–9613.

(64) Haume, K.; Rosa, S.; Grellet, S.; Śmiałek, M. A.; Butterworth, K. T.; Solov’yov, A. V.;

Prise, K. M.; Golding, J.; Mason, N. J. Gold Nanoparticles for Cancer Radiotherapy: A

Review. Cancer Nanotechnol. 2016, 7, 8.

(65) Kumar, C. S. S. R.; Mohammad, F. Magnetic Nanomaterials for Hyperthermia-Based

Therapy and Controlled Drug Delivery. Adv. Drug Deliv. Rev. 2011, 63, 789–808.

(66) Zhang, C.; Bu, W.; Ni, D.; Zhang, S.; Li, Q.; Yao, Z.; Zhang, J.; Yao, H.; Wang, Z.; Shi,

J. Synthesis of Iron Nanometallic Glasses and Their Application in Cancer Therapy by

a Localized Fenton Reaction. Angew. Chemie Int. Ed. 2016, 55, 2101–2106.

(67) Lin, L.-S.; Song, J.; Song, L.; Ke, K.; Liu, Y.; Zhou, Z.; Shen, Z.; Li, J.; Yang, Z.; Tang,

Page 127: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

126

W.; et al. Simultaneous Fenton-like Ion Delivery and Glutathione Depletion by MnO 2 -

Based Nanoagent to Enhance Chemodynamic Therapy. Angew. Chemie Int. Ed. 2018,

57, 4902–4906.

(68) Irvine, D. J.; Hanson, M. C.; Rakhra, K.; Tokatlian, T. Synthetic Nanoparticles for

Vaccines and Immunotherapy. Chem. Rev. 2015, 115, 11109–11146.

(69) Cho, N.-H.; Cheong, T.-C.; Min, J. H.; Wu, J. H.; Lee, S. J.; Kim, D.; Yang, J.-S.; Kim,

S.; Kim, Y. K.; Seong, S.-Y. A Multifunctional Core–shell Nanoparticle for Dendritic

Cell-Based Cancer Immunotherapy. Nat. Nanotechnol. 2011, 6, 675–682.

(70) Lane, L. A.; Qian, X.; Smith, A. M.; Nie, S. Physical Chemistry of Nanomedicine:

Understanding the Complex Behaviors of Nanoparticles in Vivo. Annu. Rev. Phys.

Chem. 2015, 66, 521–547.

(71) Lundqvist, M.; Stigler, J.; Elia, G.; Lynch, I.; Cedervall, T.; Dawson, K. A. Nanoparticle

Size and Surface Properties Determine the Protein Corona with Possible Implications for

Biological Impacts. Proc. Natl. Acad. Sci. U. S. A. 2008, 105, 14265–14270.

(72) Cedervall, T.; Lynch, I.; Lindman, S.; Berggård, T.; Thulin, E.; Nilsson, H.; Dawson, K.

A.; Linse, S. Understanding the Nanoparticle-Protein Corona Using Methods to Quantify

Exchange Rates and Affinities of Proteins for Nanoparticles. Proc. Natl. Acad. Sci. U. S.

A. 2007, 104, 2050–2055.

(73) Walczyk, D.; Bombelli, F. B.; Monopoli, M. P.; Lynch, I.; Dawson, K. A. What the Cell

“Sees” in Bionanoscience. J. Am. Chem. Soc. 2010, 132, 5761–5768.

(74) Casals, E.; Pfaller, T.; Duschl, A.; Oostingh, G. J.; Puntes, V. Time Evolution of the

Nanoparticle Protein Corona. ACS Nano 2010, 4, 3623–3632.

(75) Bertrand, N.; Leroux, J. C. The Journey of a Drug-Carrier in the Body: An Anatomo-

Physiological Perspective. J. Control. Release 2012, 161, 152–163.

(76) Iversen, T.-G.; Skotland, T.; Sandvig, K. Endocytosis and Intracellular Transport of

Nanoparticles: Present Knowledge and Need for Future Studies. Nano Today 2011, 6,

176–185.

(77) Haraldsson, B.; Nyström, J.; Deen, W. M. Properties of the Glomerular Barrier and

Mechanisms of Proteinuria. Physiol. Rev. 2008, 88, 451–487.

Page 128: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

127

(78) Abu Lila, A. S.; Kiwada, H.; Ishida, T. The Accelerated Blood Clearance (ABC)

Phenomenon: Clinical Challenge and Approaches to Manage. J. Control. Release 2013,

172, 38–47.

(79) Bryant, J. L.; Meredith, S. L.; Williams, K. J.; White, A. Targeting Hypoxia in the

Treatment of Small Cell Lung Cancer. Lung Cancer 2014, 86, 126–132.

(80) Wagland, R.; Richardson, A.; Ewings, S.; Armes, J.; Lennan, E.; Hankins, M.; Griffiths,

P. Prevalence of Cancer Chemotherapy-Related Problems, Their Relation to Health-

Related Quality of Life and Associated Supportive Care: A Cross-Sectional Survey.

Support. Care Cancer 2016, 24, 4901–4911.

(81) Systematic Anti-Cancer Therapy Chemotherapy Dataset

http://www.chemodataset.nhs.uk/home (accessed Oct 10, 2017).

(82) Zappa, C.; Mousa, S. A. Non-Small Cell Lung Cancer: Current Treatment and Future

Advances. Transl. lung cancer Res. 2016, 5, 288–300.

(83) Li, J.; Chen, P. R. Development and Application of Bond Cleavage Reactions in

Bioorthogonal Chemistry. Nat. Chem. Biol. 2016, 12, 129–137.

(84) Stark, W. J. Nanoparticles in Biological Systems. Angew. Chemie Int. Ed. 2011, 50,

1242–1258.

(85) Yusop, R. M.; Unciti-Broceta, A.; Johansson, E. M. V.; Sánchez-Martín, R. M.; Bradley,

M. Palladium-Mediated Intracellular Chemistry. Nat. Chem. 2011, 3, 241–245.

(86) Streu, C.; Meggers, E. Ruthenium-Induced Allylcarbamate Cleavage in Living Cells.

Angew. Chemie Int. Ed. 2006, 45, 5645–5648.

(87) Pérez-López, A. M.; Rubio-Ruiz, B.; Sebastián, V.; Hamilton, L.; Adam, C.; Bray, T.

L.; Irusta, S.; Brennan, P. M.; Lloyd-Jones, G. C.; Sieger, D.; et al. Gold-Triggered

Uncaging Chemistry in Living Systems. Angew. Chemie Int. Ed. 2017, 56, 12548–

12552.

(88) Tsubokura, K.; Vong, K. K. H.; Pradipta, A. R.; Ogura, A.; Urano, S.; Tahara, T.;

Nozaki, S.; Onoe, H.; Nakao, Y.; Sibgatullina, R.; et al. In Vivo Gold Complex Catalysis

within Live Mice. Angew. Chemie Int. Ed. 2017, 56, 3579–3584.

(89) Li, A.; Yadav, R.; White, J. K.; Herroon, M. K.; Callahan, B. P.; Podgorski, I.; Turro,

Page 129: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

128

C.; Scott, E. E.; Kodanko, J. J. Illuminating Cytochrome P450 Binding: Ru( Ii )-Caged

Inhibitors of CYP17A1. Chem. Commun. 2017, 53, 3673–3676.

(90) Lameijer, L. N.; Ernst, D.; Hopkins, S. L.; Meijer, M. S.; Askes, S. H. C.; Le Dévédec,

S. E.; Bonnet, S. A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains

Phototoxic in Hypoxic Cancer Cells. Angew. Chemie Int. Ed. 2017, 56, 11549–11553.

(91) Avci, P.; Gupta, A.; Sadasivam, M.; Vecchio, D.; Pam, Z.; Pam, N.; Hamblin, M. R.

Low-Level Laser (Light) Therapy (LLLT) in Skin: Stimulating, Healing, Restoring.

Semin. Cutan. Med. Surg. 2013, 32, 41–52.

(92) Fife, W. P. The Use of Non-Explosive Mixtures of Hydrogen and Oxygen for Diving;

1979.

(93) Grass, R. N.; Limbach, L. K.; Athanassiou, E. K.; Stark, W. J. Exposure of Aerosols and

Nanoparticle Dispersions to in Vitro Cell Cultures: A Review on the Dose Relevance of

Size, Mass, Surface and Concentration. J. Aerosol Sci. 2010, 41, 1123–1142.

(94) Onizawa, S.; Aoshiba, K.; Kajita, M.; Miyamoto, Y.; Nagai, A. Platinum Nanoparticle

Antioxidants Inhibit Pulmonary Inflammation in Mice Exposed to Cigarette Smoke.

Pulm. Pharmacol. Ther. 2009, 22, 340–349.

(95) Wiebert, P.; Sanchez-Crespo, A.; Seitz, J.; Falk, R.; Philipson, K.; Kreyling, W. G.;

Möller, W.; Sommerer, K.; Larsson, S.; Svartengren, M. Negligible Clearance of

Ultrafine Particles Retained in Healthy and Affected Human Lungs. Eur. Respir. J. 2006,

28, 286–290.

(96) Möller, W.; Felten, K.; Sommerer, K.; Scheuch, G.; Meyer, G.; Meyer, P.; Häussinger,

K.; Kreyling, W. G. Deposition, Retention, and Translocation of Ultrafine Particles from

the Central Airways and Lung Periphery. Am. J. Respir. Crit. Care Med. 2008, 177, 426–

432.

(97) Ló Pez, P. E.; Isidro-Llobet, A.; Gracia, C.; Cruz, L. J.; García-Granados, A.; Parra, A.;

Lvarez, M.; Albericio, F. Use of P-Nitrobenzyloxycarbonyl (PNZ) as a Permanent

Protecting Group in the Synthesis of Kahalalide F Analogs. Tetrahedron Lett. 2005, 46,

7737–7741.

(98) Isidro-Llobet, A.; Guasch-Camell, J.; Álvarez, M.; Albericio, F. P-

Nitrobenzyloxycarbonyl (PNZ) as a TemporaryNα-Protecting Group in Orthogonal

Page 130: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

129

Solid-Phase Peptide Synthesis - Avoiding Diketopiperazine and Aspartimide Formation.

European J. Org. Chem. 2005, 14, 3031–3039.

(99) Sun, Y.; Sun, J.; Shi, S.; Jing, Y.; Yin, S.; Chen, Y.; Li, G.; Xu, Y.; He, Z. Synthesis,

Transport and Pharmacokinetics of 5′-Amino Acid Ester Prodrugs of 1-β- d -

Arabinofuranosylcytosine. Mol. Pharm. 2009, 6, 315–325.

(100) McKeown, S. R. Defining Normoxia, Physoxia and Hypoxia in Tumours - Implications

for Treatment Response. Br. J. Radiol. 2014, 87, 1035.

(101) Pedone, D.; Moglianetti, M.; De Luca, E.; Bardi, G.; Pompa, P. P. Platinum

Nanoparticles in Nanobiomedicine. Chem. Soc. Rev. 2017, 46, 4951–4975.

(102) Zheng, W.; Jiang, B.; Hao, Y.; Zhao, Y.; Zhang, W.; Jiang, X. Screening Reactive

Oxygen Species Scavenging Properties of Platinum Nanoparticles on a Microfluidic

Chip. Biofabrication 2014, 6, 045004.

(103) Nomura, M.; Yoshimura, Y.; Kikuiri, T.; Hasegawa, T.; Taniguchi, Y.; Deyama, Y.;

Koshiro, K.; Sano, H.; Suzuki, K.; Inoue, N. Platinum Nanoparticles Suppress

Osteoclastogenesis Through Scavenging of Reactive Oxygen Species Produced in

RAW264.7 Cells. J. Pharmacol. Sci. 2011, 117, 243–252.

(104) Clark, A.; Zhu, A.; Sun, K.; Petty, H. R. Cerium Oxide and Platinum Nanoparticles

Protect Cells from Oxidant-Mediated Apoptosis. J. Nanopart. Res. 2011, 13, 5547–5555.

(105) Cho, W.-S.; Duffin, R.; Thielbeer, F.; Bradley, M.; Megson, I. L.; MacNee, W.; Poland,

C. A.; Tran, C. L.; Donaldson, K. Zeta Potential and Solubility to Toxic Ions as

Mechanisms of Lung Inflammation Caused by Metal/Metal Oxide Nanoparticles.

Toxicol. Sci. 2012, 126, 469–477.

(106) O’Connor, L. J.; Cazares-Körner, C.; Saha, J.; Evans, C. N. G.; Stratford, M. R. L.;

Hammond, E. M.; Conway, S. J. Design, Synthesis and Evaluation of Molecularly

Targeted Hypoxia-Activated Prodrugs. Nat. Protoc. 2016, 11, 781–794.

(107) Hunter, F. W.; Wouters, B. G.; Wilson, W. R. Hypoxia-Activated Prodrugs: Paths

Forward in the Era of Personalised Medicine. Br. J. Cancer 2016, 114, 1071–1077.

(108) Gagnadoux, F.; Pape, A. L.; Lemarié, E.; Lerondel, S.; Valo, I.; Leblond, V.; Racineux,

J.-L.; Urban, T. Aerosol Delivery of Chemotherapy in an Orthotopic Model of Lung

Page 131: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

130

Cancer. Eur. Respir. J. 2005, 26, 657–661.

(109) Van Norman, G. A. Drugs, Devices, and the FDA: Part 1: An Overview of Approval

Processes for Drugs. JACC Basic to Transl. Sci. 2016, 1, 170–179.

(110) Umscheid, C. A.; Margolis, D. J.; Grossman, C. E. Key Concepts of Clinical Trials: A

Narrative Review. Postgrad. Med. 2011, 123, 194–204.

(111) Gengenbacher, N.; Singhal, M.; Augustin, H. G. Preclinical Mouse Solid Tumour

Models: Status Quo, Challenges and Perspectives. Nat. Rev. Cancer 2017, 17, 751–765.

(112) Jung, J. Human Tumor Xenograft Models for Preclinical Assessment of Anticancer Drug

Development. Toxicol. Res. 2014, 30, 1–5.

(113) U. S. Food and Drug Admiministration. Inactive Ingredient Search for Approved Drug

Products https://www.accessdata.fda.gov/scripts/cder/iig/index.cfm.

(114) Degner, B. M.; Chung, C.; Schlegel, V.; Hutkins, R.; McClements, D. J. Factors

Influencing the Freeze-Thaw Stability of Emulsion-Based Foods. Compr. Rev. Food Sci.

Food Saf. 2014, 13, 98–113.

(115) Marshall, J. R.; King, M. R. Surgical Intervention and Circulating Tumor Cell Count: A

Commentary. Transl. Cancer Res. 2016, 5, S126–S128.

(116) Wittekind, C.; Neid, M. Cancer Invasion and Metastasis. Oncology 2005, 69, 14–16.

(117) Paterlini-Brechot, P.; Benali, N. L. Circulating Tumor Cells (CTC) Detection: Clinical

Impact and Future Directions. Cancer Lett. 2007, 253, 180–204.

(118) Krebs, M. G.; Hou, J.-M.; Ward, T. H.; Blackhall, F. H.; Dive, C. Circulating Tumour

Cells: Their Utility in Cancer Management and Predicting Outcomes. Ther. Adv. Med.

Oncol. 2010, 2, 351–365.

(119) Cristofanilli, M.; Budd, G. T.; Ellis, M. J.; Stopeck, A.; Matera, J.; Miller, M. C.; Reuben,

J. M.; Doyle, G. V.; Allard, W. J.; Terstappen, L. W. M. M.; et al. Circulating Tumor

Cells, Disease Progression, and Survival in Metastatic Breast Cancer. N. Engl. J. Med.

2004, 351, 781–791.

(120) de Bono, J. S.; Scher, H. I.; Montgomery, R. B.; Parker, C.; Miller, M. C.; Tissing, H.;

Doyle, G. V; Terstappen, L. W. W. M.; Pienta, K. J.; Raghavan, D. Circulating Tumor

Page 132: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

131

Cells Predict Survival Benefit from Treatment in Metastatic Castration-Resistant

Prostate Cancer. Clin. Cancer Res. 2008, 14, 6302–6309.

(121) Olmos, D.; Arkenau, H.-T.; Ang, J. E.; Ledaki, I.; Attard, G.; Carden, C. P.; Reid, A. H.

M.; A’Hern, R.; Fong, P. C.; Oomen, N. B.; et al. Circulating Tumour Cell (CTC) Counts

as Intermediate End Points in Castration-Resistant Prostate Cancer (CRPC): A Single-

Centre Experience. Ann. Oncol. 2008, 20, 27–33.

(122) Rahbari, N. N.; Aigner, M.; Thorlund, K.; Mollberg, N.; Motschall, E.; Jensen, K.;

Diener, M. K.; Büchler, M. W.; Koch, M.; Weitz, J. Meta-Analysis Shows That

Detection of Circulating Tumor Cells Indicates Poor Prognosis in Patients With

Colorectal Cancer. Gastroenterology 2010, 138, 1714–1726.e13.

(123) Yamaguchi, K.; Takagi, Y.; Aoki, S.; Futamura, M.; Saji, S. Significant Detection of

Circulating Cancer Cells in the Blood by Reverse Transcriptase-Polymerase Chain

Reaction during Colorectal Cancer Resection. Ann. Surg. 2000, 232, 58–65.

(124) Yamashita, J.; Kurusu, Y.; Fujino, N.; Saisyoji, T.; Ogawa, M. Detection of Circulating

Tumor Cells in Patients with Non–small Cell Lung Cancer Undergoing Lobectomy by

Video-Assisted Thoracic Surgery: A Potential Hazard for Intraoperative Hematogenous

Tumor Cell Dissemination. J. Thorac. Cardiovasc. Surg. 2000, 119, 899–905.

(125) Wind, J.; Tuynman, J. B.; Tibbe, A. G. J.; Swennenhuis, J. F.; Richel, D. J.; van Berge

Henegouwen, M. I.; Bemelman, W. A. Circulating Tumour Cells during Laparoscopic

and Open Surgery for Primary Colonic Cancer in Portal and Peripheral Blood. Eur. J.

Surg. Oncol. 2009, 35, 942–950.

(126) Hashimoto, M.; Tanaka, F.; Yoneda, K.; Takuwa, T.; Matsumoto, S.; Okumura, Y.;

Kondo, N.; Tsubota, N.; Tsujimura, T.; Tabata, C.; et al. Significant Increase in

Circulating Tumour Cells in Pulmonary Venous Blood during Surgical Manipulation in

Patients with Primary Lung Cancer. Interact. Cardiovasc. Thorac. Surg. 2014, 18, 775–

783.

(127) Camara, O.; Kavallaris, A.; Nöschel, H.; Rengsberger, M.; Jörke, C.; Pachmann, K.

Seeding of Epithelial Cells into Circulation during Surgery for Breast Cancer: The Fate

of Malignant and Benign Mobilized Cells. World J. Surg. Oncol. 2006, 4, 67.

(128) Dong, Q.; Huang, J.; Zhou, Y.; Li, L.; Bao, G.; Feng, J.; Sha, H. Hematogenous

Page 133: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

132

Dissemination of Lung Cancer Cells during Surgery: Quantitative Detection by Flow

Cytometry and Prognostic Significance. Lung Cancer 2002, 37, 293–301.

(129) Miller, M. C.; Doyle, G. V; Terstappen, L. W. M. M. Significance of Circulating Tumor

Cells Detected by the CellSearch System in Patients with Metastatic Breast Colorectal

and Prostate Cancer. J. Oncol. 2010, 2010, 617421.

(130) Grover, P. K.; Cummins, A. G.; Price, T. J.; Roberts-Thomson, I. C.; Hardingham, J. E.

Circulating Tumour Cells: The Evolving Concept and the Inadequacy of Their

Enrichment by EpCAM-Based Methodology for Basic and Clinical Cancer Research.

Ann. Oncol. 2014, 25, 1506–1516.

(131) Hayes, D. F.; Cristofanilli, M.; Budd, G. T.; Ellis, M. J.; Stopeck, A.; Miller, M. C.;

Matera, J.; Allard, W. J.; Doyle, G. V; Terstappen, L. W. W. M. Circulating Tumor Cells

at Each Follow-up Time Point during Therapy of Metastatic Breast Cancer Patients

Predict Progression-Free and Overall Survival. Clin. Cancer Res. 2006, 12, 4218–4224.

(132) Zhang, Z.; Fan, W.; Deng, Q.; Tang, S.; Wang, P.; Xu, P.; Wang, J.; Yu, M. The

Prognostic and Diagnostic Value of Circulating Tumor Cells in Bladder Cancer and

Upper Tract Urothelial Carcinoma: A Meta-Analysis of 30 Published Studies.

Oncotarget 2017, 8, 59527–59538.

(133) Pernot, S.; Badoual, C.; Terme, M.; Castan, F.; Cazes, A.; Bouche, O.; Bennouna, J.;

Francois, E.; Ghiringhelli, F.; De La Fouchardiere, C.; et al. Dynamic Evaluation of

Circulating Tumour Cells in Patients with Advanced Gastric and Oesogastric Junction

Adenocarcinoma: Prognostic Value and Early Assessment of Therapeutic Effects. Eur.

J. Cancer 2017, 79, 15–22.

(134) Kang, Y.; Pantel, K. Tumor Cell Dissemination: Emerging Biological Insights from

Animal Models and Cancer Patients. Cancer Cell 2013, 23, 573–581.

(135) Alix-Panabières, C.; Pantel, K. Challenges in Circulating Tumour Cell Research. Nat.

Rev. Cancer 2014, 14, 623–631.

(136) Yap, T. A.; Lorente, D.; Omlin, A.; Olmos, D.; de Bono, J. S. Circulating Tumor Cells:

A Multifunctional Biomarker. Clin. Cancer Res. 2014, 20, 2553–2568.

(137) Arnold, M.; Karim-Kos, H. E.; Coebergh, J. W.; Byrnes, G.; Antilla, A.; Ferlay, J.;

Renehan, A. G.; Forman, D.; Soerjomataram, I. Recent Trends in Incidence of Five

Page 134: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

133

Common Cancers in 26 European Countries since 1988: Analysis of the European

Cancer Observatory. Eur. J. Cancer 2015, 51, 1164–1187.

(138) Ferlay, J.; Steliarova-Foucher, E.; Lortet-Tieulent, J.; Rosso, S.; Coebergh, J. W. W.;

Comber, H.; Forman, D.; Bray, F. Cancer Incidence and Mortality Patterns in Europe:

Estimates for 40 Countries in 2012. Eur. J. Cancer 2013, 49, 1374–1403.

(139) Cronin, K. A.; Lake, A. J.; Scott, S.; Sherman, R. L.; Noone, A.-M.; Howlader, N.;

Henley, S. J.; Anderson, R. N.; Firth, A. U.; Ma, J.; et al. Annual Report to the Nation

on the Status of Cancer, Part I: National Cancer Statistics. Cancer 2018, 124, 2785–

2800.

(140) Nagrath, S.; Sequist, L. V.; Maheswaran, S.; Bell, D. W.; Irimia, D.; Ulkus, L.; Smith,

M. R.; Kwak, E. L.; Digumarthy, S.; Muzikansky, A.; et al. Isolation of Rare Circulating

Tumour Cells in Cancer Patients by Microchip Technology. Nature 2007, 450, 1235–

1239.

(141) Hoshino, K.; Huang, Y.-Y.; Lane, N.; Huebschman, M.; Uhr, J. W.; Frenkel, E. P.;

Zhang, X. Microchip-Based Immunomagnetic Detection of Circulating Tumor Cells.

Lab Chip 2011, 11, 3449.

(142) Ahmed, M. G.; Abate, M. F.; Song, Y.; Zhu, Z.; Yan, F.; Xu, Y.; Wang, X.; Li, Q.; Yang,

C. Isolation, Detection, and Antigen-Based Profiling of Circulating Tumor Cells Using

a Size-Dictated Immunocapture Chip. Angew. Chemie Int. Ed. 2017, 56, 10681–10685.

(143) Zeltner, M.; Grass, R. N.; Schaetz, A.; Bubenhofer, S. B.; Luechinger, N. A.; Stark, W.

J. Stable Dispersions of Ferromagnetic Carbon-Coated Metal Nanoparticles: Preparation

via Surface Initiated Atom Transfer Radical Polymerization. J. Mater. Chem. 2012, 22,

12064.

(144) Wang, S.; Zhou, Y.; Yang, S.; Ding, B. Growing Hyperbranched Polyglycerols on

Magnetic Nanoparticles to Resist Nonspecific Adsorption of Proteins. Colloids Surfaces

B Biointerfaces 2008, 67, 122–126.

(145) Khan, M.; Huck, W. T. S. Hyperbranched Polyglycidol on Si/SiO2 Surfaces via Surface-

Initiated Polymerization. Macromolecules 2003, 36, 5088–5093.

(146) Das, P.; Jana, N. R. Highly Colloidally Stable Hyperbranched Polyglycerol Grafted Red

Fluorescent Silicon Nanoparticle as Bioimaging Probe. ACS Appl. Mater. Interfaces

Page 135: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

134

2014, 6, 4301–4309.

(147) Li, Z.; Chau, Y. Synthesis of Linear Polyether Polyol Derivatives As New Materials for

Bioconjugation. Bioconjug. Chem. 2009, 20, 780–789.

(148) Hofer, C. J.; Grass, R. N.; Zeltner, M.; Mora, C. A.; Krumeich, F.; Stark, W. J. Hollow

Carbon Nanobubbles: Synthesis, Chemical Functionalization, and Container-Type

Behavior in Water. Angew. Chemie Int. Ed. 2016, 55, 8761–8765.

(149) Marquardt, F.; Mommer, S.; Lange, J.; Jeschenko, P.; Keul, H.; Möller, M. Homoserine

Lactone as a Structural Key Element for the Synthesis of Multifunctional Polymers.

Polymers (Basel). 2017, 9, 130.

(150) Kainthan, R. K.; Janzen, J.; Levin, E.; Devine, D. V.; Brooks, D. E. Biocompatibility

Testing of Branched and Linear Polyglycidol. Biomacromolecules 2006, 7, 703–709.

(151) Frey, H.; Haag, R. Dendritic Polyglycerol: A New Versatile Biocompatible Material.

Rev. Mol. Biotechnol. 2002, 90, 257–267.

(152) Shen, J.; Ly, K.; Hoang, Y. Cell Culture Medium. In Human Stem Cell Manual; Elsevier,

2012; pp. 53–69.

(153) Lang, T.; Bauters, A.; Braun, S. L.; Pötzsch, B.; von Pape, K.-W.; Kolde, H.-J.; Lakner,

M. Multi-Centre Investigation on Reference Ranges for ROTEM Thromboelastometry.

Blood Coagul. Fibrinolysis 2005, 16, 301–310.

(154) Bougas, L.; Langenegger, L. D.; Mora, C. A.; Zeltner, M.; Stark, W. J.; Wickenbrock,

A.; Blanchard, J. W.; Budker, D. Nondestructive In-Line Sub-Picomolar Detection of

Magnetic Nanoparticles in Flowing Complex Fluids. Sci. Rep. 2018, 8, 3491.

(155) Herrmann, I. K.; Bernabei, R. E.; Urner, M.; Grass, R. N.; Beck-Schimmer, B.; Stark,

W. J. Device for Continuous Extracorporeal Blood Purification Using Target-Specific

Metal Nanomagnets. Nephrol. Dial. Transplant. 2011, 26, 2948–2954.

(156) Van Norman, G. A. Drugs, Devices, and the FDA: Part 2: An Overview of Approval

Processes: FDA Approval of Medical Devices. JACC Basic to Transl. Sci. 2016, 1, 277–

287.

(157) Hu, Y.; Turner, M. J.; Shields, J.; Gale, M. S.; Hutto, E.; Roberts, B. L.; Siders, W. M.;

Kaplan, J. M. Investigation of the Mechanism of Action of Alemtuzumab in a Human

Page 136: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

135

CD52 Transgenic Mouse Model. Immunology 2009, 128, 260–270.

(158) Biggs, J. R.; Zhang, D.-E. Molecular Basis of Lymphoid and Myeloid Diseases. Essent.

Concepts Mol. Pathol. 2010, 189–203.

(159) Looney, R. J. Treating Human Autoimmune Disease by Depleting B Cells. Ann. Rheum.

Dis. 2002, 61, 863–866.

(160) Tsokos, G. C. B Cells, Be Gone — B-Cell Depletion in the Treatment of Rheumatoid

Arthritis. N. Engl. J. Med. 2004, 350, 2546–2548.

(161) Rezvani, A. R.; Maloney, D. G. Rituximab Resistance. Best Pract. Res. Clin. Haematol.

2011, 24, 203–216.

(162) Galli, S. J.; Tsai, M.; Piliponsky, A. M. The Development of Allergic Inflammation.

Nature 2008, 454, 445–454.

(163) Matasar, M. J.; Zelenetz, A. D. Overview of Lymphoma Diagnosis and Management.

Radiol. Clin. North Am. 2008, 46, 175–198.

(164) Drug Products, Including Biological Products, That Contain Nanomaterials - Guidance

for Industry; 2017.

(165) Berger, T.; Elovaara, I.; Fredrikson, S.; McGuigan, C.; Moiola, L.; Myhr, K.-M.; Oreja-

Guevara, C.; Stoliarov, I.; Zettl, U. K. Alemtuzumab Use in Clinical Practice:

Recommendations from European Multiple Sclerosis Experts. CNS Drugs 2017, 31, 33–

50.

Page 137: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

136

Curriculum Vitae

Antoine Florent Herzog

Functional Materials Laboratory

Institute for Chemical and Bioengineering Private Address:

Department of Chemistry and Applied Biosciences Hofwiesenstrasse 378

ETH Zürich, HCI E103 8050 Zürich

8093 Zurich Switzerland

Switzerland

Phone: +41 44 633 37 70

Email: [email protected]

Born: September 10th, 1990 in Mulhouse, France

Citizen of France

Languages: French (native), English (fluent), German (intermediate)

Page 138: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

137

Education

07/2015 – current PhD studies at the Department of Chemistry and Applied Biosciences,

Institute of Chemical- and Bioengineering, Functional Materials

Laboratory, ETH Zurich, Zurich, Switzerland

Advisor: Prof. Dr. Wendelin J. Stark

Title: Novel Applications of Metallic Nanoparticles for Cancer

Treatments

09/2012 – 04/2015 MSc studies in Chemistry, Department of Chemistry and Applied

Biosciences, ETH Zurich, Zurich, Switzerland.

Master thesis: In Vitro Intracellular Metabolite Reduction with an

Exogenous Platinum Nanocatalyst under Hydrogen-Containing

Atmosphere, Prof. Dr. Wendelin J. Stark, Institute for Chemical and

Bioengineering, ETH Zurich.

Research project: Feasibility Study of Living Surface Having the Ability

to Inducibly Express Emerald Green Fluorescent Protein,

Prof. Dr. Wendelin J. Stark, Institute for Chemical and Bioengineering,

ETH Zurich.

09/2009 - 07/2012 BSc studies in Chemistry, Chemistry and Chemical Engineering

Section, EPFL, Lausanne, Switzerland.

09/2011 – 06/2012 Erasmus exchange studies at Imperial College London, London, United

Kingdom.

Research project: On the Stereoselectivity of Photocycloadditions

between Benzene and Ethylene Derivatives, Prof. Dr. Michael

Bearpark, Department of Chemistry, Imperial College London.

09/2005 – 06/2009 High-school, Baccalauréat Scientifique, Section Sport-Étude (judo)

Lycée Louis Pasteur, Strasbourg, France.

Core subjects: Chemistry and Physics

Until 09/2005 Collège Pierre Pflimlin, Section Sport-Étude (judo), Brunstatt, France.

Page 139: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

138

Refereed Journal Articles

3. A. F. Herzog, E. M. Schneider, W. J. Stark, Hydrogen as a bio-orthogonal trigger for

spatiotemporally controlled caged prodrug activation, Helvetica Chimica Acta, 2018,

10.1002/hlca.201800134

2. C. J. Hofer, R. N. Grass, E. M. Schneider, L. Hendriks, A. F. Herzog, M. Zeltner, D.

Günther, W. J. Stark, Water dispersible surface-functionalized platinum/carbon

nanorattles for size-selective catalysis, Chemical Science, 2018, 9, 362.

1. C. A. Mora, A. F. Herzog, R. A. Raso, W. J. Stark, Programmable living material

containing reporter micro-organisms permits quantitative detection of oligosaccharides,

Biomaterials, 2015, 61, 1-9.

Patents

2. A. F. Herzog, W. J. Stark, M. Zeltner, B. Beck-Schimmer, A. Zabel, Bioconjugates of

antibodies and functionalized magnetic nanoparticles, 2018, EP18159763.

1. A. F. Herzog, W. J. Stark, Combination medicament comprising a prodrug and

inhalable catalyst, 2017, EP17176199.

Honours and Awards

09/2011 – 06/2012 Erasmus Scholarship.

Conference Presentations and Proceedings

1. A. F. Herzog, E. M. Schneider, W. J. Stark, In vitro prodrug activation over platinum

nanoparticles for side effect reduction in lung cancer (poster), MRS Fall Meeting 2017,

Boston MA, USA, November 26 – December 1, 2017.

Student supervision

2. Stéphanie Nguengoue (Research Project, 09/2016 – 12/2016): Proof-of-principle of

intracellular polymer/oligomer synthesis for cytotoxicity application.

1. Riccardo Tarchini (Research Project, 02/2016 – 04/2016): Development of an

odour-adsorbing insole for shoes.

Professional experience

Page 140: Rights / License: Research Collection In Copyright - Non ... › bitstream › ... · 3 Acknowledgments First, I would like to thank Prof. Dr. Wendelin J. Stark for mentoring my PhD

139

05/2013 - 11/2013 Internship, Organic Electronics Group, BASF, Basel, Switzerland.

07/2012 - 08/2012 Summer Internship, Cheminformatics Group, Novartis Institute for

Biomedical Research, Basel, Switzerland.