148
Research Collection Doctoral Thesis Preparation of drug nanocrystals stabilized by functionalized polymeric coatings Author(s): Fuhrmann, Kathrin Publication Date: 2014 Permanent Link: https://doi.org/10.3929/ethz-a-010175706 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

Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

  • Upload
    others

  • View
    9

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

Research Collection

Doctoral Thesis

Preparation of drug nanocrystals stabilized by functionalizedpolymeric coatings

Author(s): Fuhrmann, Kathrin

Publication Date: 2014

Permanent Link: https://doi.org/10.3929/ethz-a-010175706

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 ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

DISS. ETH NO. 21903

PREPARATION OF DRUG NANOCRYSTALS

STABILIZED BY FUNCTIONALIZED

POLYMERIC COATINGS

Kathrin Fuhrmann

Page 3: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted
Page 4: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

1

DISS. ETH NO. 21903

Preparation of Drug Nanocrystals Stabilized by

Functionalized Polymeric Coatings

A thesis submitted to attain the degree of

DOCTOR OF SCIENCES of ETH ZURICH

(Dr. sc. ETH Zurich)

presented by

KATHRIN FUHRMANN

Pharmacist, Freie Universität Berlin

born on 20.02.1981

citizen of Germany

accepted on the recommendation of

Prof. Jean-Christophe Leroux

Prof. Marc A. Gauthier

Prof. Bruno Alfred Gander

2014

Page 5: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

2

Page 6: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

3

Table of Contents

I. Summaries .............................................................................................................. 5I.1. Summary .......................................................................................................................... 7I.2. Zusammenfassung ........................................................................................................... 9I.3. List of Abbreviations ..................................................................................................... 11

II. Introduction ........................................................................................................ 13II.1. General Background on Drug Nanocrystals for Chemotherapy ................................... 15II.2. Targeting of Injectable Drug Nanocrystals .................................................................. 25

Introduction ......................................................................................................................... 25In Vitro ................................................................................................................................ 27In Vivo ................................................................................................................................. 32Stabilizer: To Shed or Not To Shed ..................................................................................... 36Altering Dissolution Profiles (by Means Other than Size) .................................................. 39Outlook ................................................................................................................................ 41

III. Results and Discussion ..................................................................................... 43III.1. PEG Nanocages as Non-Sheddable Stabilizers for Drug Nanocrystals ...................... 45

Introduction ......................................................................................................................... 45Experimental Section .......................................................................................................... 47Results and Discussion ........................................................................................................ 54Conclusion .......................................................................................................................... 69

III.2. Modular Design of Redox-Responsive Stabilizers for Nanocrystals .......................... 71Introduction ......................................................................................................................... 71Experimental Section .......................................................................................................... 73Results and Discussion ........................................................................................................ 78Conclusion .......................................................................................................................... 90

IV. Conclusion and Outlook .................................................................................. 91

V. Supplementary Information ............................................................................ 101

VI. Curriculum Vitae and Scientific Contributions .......................................... 117

VII. Acknowledgments ......................................................................................... 121

VIII. List of References ........................................................................................ 125

Page 7: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

4

Page 8: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

5

I. Summaries

Page 9: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

6

Page 10: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

7

I.1. Summary

The poor water-solubility of many newly discovered drugs causes problems for

several routes of administration. In the case of intravenous administration, the

dissolved drug should not precipitate lest it causes an embolism. One elegant

approach to overcome solubility problems is to formulate poorly water soluble drugs

as nanocrystals. Due to their small size, nanocrystals dissolve faster than the

corresponding bulk solid drug, can have a higher saturation solubility, and can be

administered intravenously. Nanocrystals are almost entirely composed of drug, with

only a small amount of stabilizing excipient. This is beneficial for parenteral use

because adverse side effects, such as pain and immune reactions, have been reported

for a variety of cosolvents and solubilizers. Unfortunately, the stabilizer cannot be

completely eliminated from the formulation as nanocrystals tend to aggregate due to

their high surface energy. Polymers and/or surfactants are added to provide steric

and/or electrostatic barriers to aggregation. Nevertheless, modification of the

stabilizer offers new opportunities for nanocrystal targeting and release in specific

tissues, which would be inaccessible to non-stabilized nanocrystals. This concept

could prove especially useful in the treatment of cancer, where most drugs are poorly

water-soluble.

Cancer is a disease in which the uncontrolled division and growth of cells results in

tumor formation and potential invasion of other locations throughout the body. The

prevalence for this disease is high, and lung and breast cancers are the worldwide

leading causes of cancer mortality. Cancer treatments have a high impact on the

patient’s well-being, because the pharmacological action of the toxic anticancer drugs

is often untargeted and therefore affects all dividing cells. Patients as well as

survivors suffer from effects of the disease, but also from complications caused by the

treatment, such as hair loss, nausea, and neurological pain. These adverse side effects

could be avoided if a drug formulation is made to only target and release the drug at

the tumor site.

In the work presented herein, stabilizers for nanocrystals of paclitaxel (poorly

soluble chemotherapeutic drug) were chemically modified to examine new

opportunities for improving stability, targeting, and stimuli-responsive release. The

stabilizer platform consisted of diblock copolymers of methoxy(polyethylene glycol)-

Page 11: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

8

b-(-propargyl--valerolactone-co--caprolactone). These polymers stabilized

paclitaxel nanocrystals produced by wet milling. Moreover, they could be cross-

linked with different diazido compounds via the copper-catalyzed azide-alkyne

Huisgen cycloaddition reaction to form nanocages on the surface of nanocrystals. The

nanocages improved the size stability for the nanocrystals, and they were also less

shed from the surface of the nanocrystals.

In a second study, the stabilizers were modified with different alkane thiols by

radical thiol−yne addition. A library of polymers with different lipophilicities was

obtained and tested as stabilizers for paclitaxel nanocrystals. In addition, the acquired

thioether bonds in the polyester block were sensitive to oxidation, which reversed

their affinity for the hydrophobic nanocrystals’ surface. This could be beneficial

where drug release/uptake in response to a stimulus, e.g., presence of reactive oxygen

species in the tumor, is desired.

In summary, the proposed strategies for increased nanocrystal stabilization and

oxidation responsive release may be useful for improving chemotherapy and reducing

adverse side effects. Such methods can also be applicable for modifications of other

intravenously administered nanoformulations where a non-covalent attachment of the

coating and/or location-specific shedding of stabilizers is desired. Thus, in the long

term, this work can help improve the properties of drug nanocrystals for anticancer

therapy.

Page 12: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

9

I.2. Zusammenfassung

Die schlechte Wasserlöslichkeit vieler neu entdeckten Arzneistoffe stellt ein

Problem für mehrere Applikationsarten dar. Im Falle einer intravenösen Gabe darf der

Wirkstoff nicht in der Blutbahn ausfallen, da er sonst eine Embolie verursacht. Eine

elegante Art diese Löslichkeitsprobleme zu überwinden, ist die Formulierung dieser

Klasse von Arzneistoffen als Nanokristalle. Wegen ihrer geringen Grösse lösen diese

sich schneller auf als der entsprechende grobe Arzneistoff, sie können eine höhere

Sättigungslöslichkeit haben und sie können auch intravenös gegeben werden.

Nanokristalle bestehen hauptsächlich aus reinem Arzneistoff und nur einem kleinen

Anteil an Hilfsstoff. Dies ist von Vorteil für den parenteralen Gebrauch, weil

unerwünschte Nebenwirkungen, wie Schmerzen und Immunreaktionen, für vielerlei

Lösungsmittel und Lösungsvermittler bekannt sind. Allerdings haben Nanokristalle

auf Grund ihrer hohen Oberflächenenergie die Tendenz zu aggregieren. Deshalb

werden Polymere und/oder Tenside hinzugefügt, welche als sterische und/oder

elektrostatische Barrieren fungieren. Durch Modifizieren dieser Polymere ergeben

sich neue Möglichkeiten für Nanokristall Targeting und Freigabe in spezifischen

Geweben. Dies könnte sich als besonders nützlich in der Behandlung von Krebs

erweisen, wo die meisten Arzneistoffe schlecht wasserlöslich sind.

Krebs ist eine Krankheit, bei welcher die unkontrollierte Zellteilung und -wachstum

zur Tumorbildung und möglicher Invasion in anderen Teilen des Körpers führen. Die

Prävalenz für diese Krankheit ist hoch, und Lungen- und Brustkrebs sind die weltweit

führenden Ursachen für Krebssterblichkeit. Krebsbehandlungen haben eine hohe

Auswirkung auf das Wohlbefinden eines Patienten, weil die pharmakologische

Wirkung des toxischen Chemotherapeutikums häufig ungerichtet ist und deshalb alle

sich teilenden Zellen betrifft. Patienten sowie Überlebende leiden unter den

Auswirkungen der Krankheit, aber auch an den Komplikationen die durch die

Behandlung entstehen, wie z.B. Haarausfall, Übelkeit und neurologische Schmerzen.

Diese unerwünschten Arzneimittelwirkungen könnten vermieden werden, wenn eine

Wirkstoffformulierung so gemacht ist, dass sie den Wirkstoff gezielt zum Tumor

bringt und dort freisetzt.

In dieser Arbeit wurden Polymere für Paclitaxel Nanokristalle (schlecht

wasserlöslicher chemotherapeutischer Arzneistoff) chemisch modifiziert, um neue

Page 13: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

10

Mittel für verbesserte Größenstabilität, gewebespezifischer Pharmakotherapie und

bedarfsgesteuerter Freisetzung zu finden. Diese Polymere basierten auf einem

Diblock Kopolymer, Methoxy-polyethylenglycol-b-(-propargyl--valerolacton-

co-caprolacton), und konnten Paclitaxel Nanokristalle stabilisieren, welche durch

Nassvermahlung hergestellt wurden. Weiterhin konnten sie mit Hilfe verschiedener

Diazidverbindungen in einer Kupfer katalysierten Azid-Alkin Huisgen Zykloaddition

vernetzt werden, wodurch Nanokäfige auf der Oberfläche der Nanokristalle

entstanden. Die Nanokäfige verbesserten die Grössenstabilität der Nanokristalle und

neigten weniger dazu von deren Oberfläche abgestreift zu werden.

In einer zweiten Studie wurden diese Polymere mit verschiedenen Alkanthiolen

durch radikale Thiol-Alkin Reaktion modifiziert. Eine Sammlung neuer Polymere mit

unterschiedlichen Lipophilien wurde erhalten und als Stabilisierer für Paclitaxel

Nanokristalle getestet. Ausserdem waren die entstandenen Thioetherbindungen

empfindlich gegenüber Oxidation, welches ihre Affinität für die Oberfläche der

Nanokristalle umkehrte. Dies könnte dann von Vorteil sein wenn die

Wirkstofffreisetzung/-aufnahme in Abhängigkeit von einem Stimulus, z.B. Präsenz

von reaktiven Sauerstoff Spezies im Tumor, gewünscht ist.

Zusammenfassend kann gesagt werden, dass die vorgeschlagenen Strategien für

erhöhte Nanokristall Stabilisation und Oxidationsreaktionsfähigkeit durch

Polymermodifikation ein nützliches Mittel für die Verbesserung der Chemotherapie

mit weniger unerwünschten Arzneimittelwirkungen werden kann. Solche Methoden

könnten auch auf die Modifizierung anderer intravenös gegebener

Nanoformulierungen übertragen werden, da wo nicht-kovalentes Verknüpfen

und/oder ortsspezifisches Abstreifen des Polymers gewünscht ist. Daher kann diese

Arbeit in Zukunft dazu beitragen, die Eigenschaften von Nanokristallen für die

Chemotherapie zu verbessern.

Page 14: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

11

I.3. List of Abbreviations

BSA Bovine serum albumin

CMC Critical micelle concentration

COSY Correlation spectroscopy (two-dimensional NMR)

DLS Dynamic light scattering

DMF Dimethylformamide

DMSO Dimethylsulfoxide

VL -Valerolactone

CL -Caprolactone

EPR Enhanced permeation and retention

FDA Food and drug administration

FTIR Fourier transform infrared (spectroscopy)

HMPA Hexamethylphosphoramide

HPLC High performance liquid chromatography

HPCD Hydroxypropyl--cyclodextrin

i.v. Intravenous

ICP-OES Inductively coupled plasma optic emission spectroscopy

LDA Lithium diisopropylamide

MALDI-TOF MS Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry

mCPBA m-Chloroperoxybenzoic acid

mPEG Methoxy poly(ethylene glycol)

mPEG-b-[PVL-co-CL]

Methoxy poly(ethylene glycol)-b-[-propargyl--valerolactone-co--caprolactone]

mPEG-b-PSO Methoxy poly(ethylene glycol)-b-poly(styrene oxide)

MPS Mononuclear phagocyte system

MWCO Molecular weight cutoff

nab-Paclitaxel Nanoparticle albumin-bound paclitaxel

NC Nanocrystal

Page 15: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

12

NMP 1-Methyl-2-pyrrolidone

NMR Nuclear magnetic resonance (spectroscopy)

P-gp P-glycoprotein

PCL Poly(caprolactone)

PEG Poly(ethylene glycol)

PTX Paclitaxel

PVL Propargyl--valerolactone

ROP Ring-opening polymerization

ROS Reactive oxygen species

SEC Size exclusion chromatography

TEM Transmission electron microscopy

THF Tetrahydrofuran

TPGS D-α-tocopheryl poly(ethylene glycol 1000) succinate

Page 16: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

13

II. Introduction

Page 17: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

14

Page 18: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

15

II.1. General Background on Drug Nanocrystals for

Chemotherapy

What are drug nanocrystals?

Most drugs emerging from high throughput screenings are poorly water-soluble,1

which can be a disadvantage for their administration to a patient. For instance, a

drug’s oral bioavailability depends on its solubility (in addition to permeability), but

the same also applies to topical administration of a drug, because only dissolved drug

can be absorbed. Low or erratic bioavailabilty can have consequences for the safety

and efficacy of a drug. To overcome this problem crystalline drugs can, for example,

be converted to amorphous material.2 Alternatively drugs can be modified into

prodrugs,3 a process that involves considerable manufacturing efforts and risk of

lowered stability or change in efficacy. Furthermore, drugs can be formulated in a

liquid dosage form,4 which, however, can make it less appealing to the patient

because of the potentially required cold storage conditions, increased single dose

volume, and possibly unpleasant taste. In the case of the intravenous (i.v.)

administration route, an aqueous drug solution is the preferred type of pharmaceutical

formulation, however, cosolvents and solubilizers used to dissolve lipophilic

substances can pose problems. These can be systemic immune reactions

(hypersensitivity reactions), organ toxicity, and pain if the amount is high.5-8 When

poorly water soluble drugs are formulated as i.v. suspensions or emulsions one has to

bear in mind that particles larger than ca. 5 µm can potentially clog blood capillaries

and cause embolism. Therefore, an attractive approach for administering a drug in its

solid state is to engineer them as drug nanocrystals. These crystals are composed

almost entirely of drug, which differentiates them from other nanomedicines such as

liposomes,9, 10 micelles,11 or polymeric nanoparticles12 since only a small amount of

excipient is needed for stabilization. Typically the drug content ranges between 50

and 90%,13, 14 and the material is crystalline as the name states. In the 1990s, a wet

media milling process from the paint and photographic industry was adapted by

Liversidge and colleagues from Sterling Drug Inc./Eastman Kodak for reducing the

particle size of poorly soluble drugs in order to increase their oral bioavailability. This

process was patented as NanoCrystal® Technology, and currently there are five

Page 19: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

16

products on the market (Emend®, Invega® Sustenna®, Megace® ES, Rapamune®, and

TriCor®)15 with another product (Panzem® NCD)16, 17 in the pipeline (Table 1). The

particularity of nanocrystals is that smaller particle size results in faster dissolution,18

according to the Noyes-Whitney equation (Equation 1):

dc

dt

A (CS CX )

h (1)

because the greater the specific surface area A of a solid, the faster is the dissolution

velocity dc/dt, (CS is the saturation solubility of the solute, CX is the solute

concentration in the medium, and h is the diffusion barrier). Furthermore, nanosizing

(reducing the radius r and increasing surface energy/interfacial tension ) can

increase the saturation solubility CS19 according to the Ostwald-Freundlich equation

(Equation 2):

logCS

C

2V

2.303RT r (2)

where C∞ is the solubility of very large particles, the interfacial tension between

the solid surface and the surrounding medium, V the molar volume of the compound,

R the gas constant, T the absolute temperature, and the density of the solid.

Particle size reduction generally improves the bioavailability of orally administered

drugs by increasing their solubility and dissolution rate. Furthermore, the fed/fasted

variability can also be attenuated. This benefit of nanocrystals becomes clear in the

example Emend® (aprepitant, anti-emetic). The conventional formulation showed

greater bioavailability in the presence of food but the fasted state is rather difficult to

obtain for a patient who suffers from nausea/vomiting.13, 20

Most nanocrystal formulations are transformed into tablets, which is a convenient

oral dosage form with high patient compliance. Nanocrystals are also amenable to

additional modifications, including the conjugation of targeting ligands or

environment-sensitive triggers to the stabilizer for increasing the bioavailability of the

drug specifically at the site of disease. Advantages and disadvantages of drug

nanocrystals as formulations for parenteral use specifically will be highlighted in

Chapter II.2.

Page 20: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

17

Table 1. Products on the market produced by nanosizing technologies.13, 15, 21

Product Company Drug compound Nanosizing technology

Indication Administration route

Therapeutic benefit Year of FDA approval

Emend® Merck Aprepitant Wet milling Nausea, vomiting Oral Increased bioavailabi-lity, no food effects

2003

Invega® Sustenna®

Janssen Paliperidone palmitate

Wet milling Schizophrenia Intramuscular injection

Reformulated from tablets for sustained release

2009

Megace® ES Strativa/ PAR Pharmaceuticals

Megestrol acetate Wet milling Anorexia, chachexia

Oral Increased bioavailabi-lity, no food effects

2005

Rapamune® Pfizer/Wyeth Sirolimus Wet milling Prophylaxis of organ rejection (immunosuppress-ant)

Oral Reformulated from solution, better storage conditions and higher bioavailability

2000

TriCor® Abbot/Abbvie Fenofibrate Wet milling Hypercholesterol-emia

Oral Increased bioavailabi-lity, no food effects

2004

Triglide® Skye Pharma/ Shionogi

Fenofibrate High pressure homogeniza-tion

Hypercholesterol-emia

Oral Increased bioavailabi-lity, no food effects

2005

Page 21: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

18

How are drug nanocrystals produced?

Nanocrystals can be produced by two major routes: (1) by decreasing size of the

starting material using various comminuting techniques (“top-down”) and (2) by

building new crystals from a solution via precipitation in an antisolvent (“bottom-

up”). In the “top-down” approach coarse drug powder is fractured by wet milling,22

sonication,23 high pressure homogenization,19 or even laser fragmentation24 in a wet

environment, and usually in presence of a surfactant to prevent aggregation of newly

formed nanocrystals.25, 26 Wet milling in particular is established for industrial scale

production and a number of products manufactured by this procedure are on the

market.13, 27 In this process, the coarse drug powder is suspended in aqueous media

and introduced into a milling chamber containing beads. These beads and often also

the milling chamber are of very hard material such as yttrium-stabilized zirconium

oxide to limit wear which could contaminate the product.22, 28 “Bottom-up”

approaches have been mostly used on laboratory scale, sometimes resulting in

amorphous material.29, 30 Many variations exist, but essentially they all have in

common a precipitation in an anti-solvent accompanied by sonication and/or stirring.

Due to a wider range of parameters that need to be controlled they have not yet

resulted in a commercialized pharmaceutical product. For further details on the

fabrication techniques the reader is directed to the thorough recent reviews by

Merisko-Liversidge and Liversidge 13 and by Chan and Kwok.31

How are nanocrystals stabilized?

The high surface energy of nanocrystals causes them to aggregate and form larger

particles. To prevent aggregation, a surfactant or polymer is added to the medium. It

physically adsorbs to the surface of the nanocrystals, thereby stabilizing them by

steric hindrance and/or electrostatic repulsion.22, 28 The stabilizer is in equilibrium

between its adsorbed and free state. A seemingly endless list of factors have been

studied for their influence on the preparation of stable nanosuspensions, including

intensity of grinding energy, drug content during milling, bead size, logP,

morphology, melting point, solubility, density, and mechanical properties of the drug

substance.22, 26, 28, 32, 33 Nevertheless, the choice of stabilizer remains often empirical,

although it seems that the affinity of the stabilizer to hydrophobic surfaces, which is

related to the hydrophobicity of the drug material itself, plays a crucial role.26

Commonly used stabilizers for nanocrystals include different types of poloxamers,

Page 22: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

19

povidone, cellulose derivatives, and D-α-tocopheryl poly(ethylene glycol 1000)

succinate (TPGS).28, 34-36 The use of block copolymers containing hydrophilic flexible

polymers such as poly(ethylene glycol) (PEG) may also increase the circulation time

of i.v. injected nanoparticles by reducing opsonization and uptake by the mononuclear

phagocyte system (MPS).37, 38 Prominent examples of PEG-containing stabilizers are

poloxamers (a poly(propylene glycol) block flanked by two PEG chains) and TPGS.

Different attempts have been made to enhance adsorption and/or anchoring in order to

improve colloidal stability. These include, for example, the covalent cross-linking of

the surfactant network on the crystal surface to form nanocages (Chapter III.1),39 and

electrostatic cross-linking by layer-by-layer assembly of oppositely-charged polyions

in multiple layers around the crystal.23, 40, 41 Charged polymers can be of natural

source like chitosan (polycation) and alginic acid (polyanion), or synthetic such as

poly(allylamine hydrochloride) (polycation) and sodium poly(styrene sulfonate)

(polyanion).41

What is paclitaxel?

Paclitaxel is a poorly soluble drug with high potency in the treatment of cancer of

various types. Its long use in the clinic has shown that it is safe and effective,

however, there are issues related to the currently used formulations. Thus, it was

chosen as a model drug for the work in this doctoral thesis.

Paclitaxel was the first drug originating from the taxane family, a group of

microtubule-stabilizing agents that inhibit mitosis, and it is widely used in the

chemotherapy of cancer.42 Microtubules are essential during mitosis, a process during

which duplicated chromosomes of a cell are separated into two identical sets before it

divides into two daughter cells. Cancer cells constantly divide and thus the

importance of the microtubules in mitosis and cell division makes them a significant

target for anticancer drugs.42, 43 Paclitaxel is a natural component, which was

originally (about 40 years ago) isolated from the bark of the Pacific yew tree (Taxus

brevifolia). Due to its structural complexity (Figure 1) and the limited presence of

Pacific yew trees, it is now produced semisynthetically from 10-deacetylbaccatin III

from the European yew tree (Taxus baccata).44 Other representatives of the taxanes

include docetaxel and cabazitaxel.43 Paclitaxel is used in cancer chemotherapy to treat

ovarian cancer, breast cancer, small and non-small cell lung cancer, colon cancer, and

Kaposi’s sarcoma.42, 45 However, an important issue is its poor water solubility, which

Page 23: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

20

requires the use of ethanol and Cremophor EL (macrogolglycerol ricinoleate).

Cremophor EL has been associated with hypersensitivity reactions.5

Figure 1. Chemical structure of paclitaxel

Adverse effects of taxanes generally include neurological and myeloid toxicities.

Neurological toxicity can be potentially severe and is a dose-limiting side effect that

manifests itself as a painful and debilitating peripheral axonal neuropathy for which

there are currently no effective symptomatic treatments.43 Symptoms tend to

disappear some months after the end of treatment, though in some cases patients

retain considerable sequelae several years after therapy.43 This toxicity is not yet

understood at a mechanistic level, however, it is apparently related to the relative

abundance of tubulin in neurons and the importance of an intact, functional

microtubule cytoskeleton for adequate nerve conduction.46 Myeloid toxicity is

frequently observed, with neutropenia as the most frequent and/or severe side effect

occurring in combination regimens, but is usually manageable.43

A novel surfactant-free taxane formulation which addresses the problems associated

with cosolvent induced hypersensitivity reactions is the nanoparticle albumin-bound

paclitaxel (nab-paclitaxel, Abraxane®), which is mainly amorphous.47, 48 Since its

approval in the US in 2005 (Europe in 2008) its use in the clinic allows the

administration of higher doses of drug without steroid or antihistamine prophylaxis

for hypersensitivity reactions. In a Phase III trial comparing Abraxane® and solvent

Page 24: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

21

based paclitaxel formulation in the treatment of patients with metastatic breast cancer,

nab-paclitaxel was associated with a better outcome as well as with a lower rate of

severe neutropenia but a slightly higher rate of sensory neuropathy, which mostly

improved after treatment interruption.49 Abraxane® has also shown activity in other

contexts including melanoma, small cell lung cancer, and gynecological tumours.50, 51

Nevertheless, it still causes strong discomforting and dose limiting side effects such as

alopecia, fatigue, neutropenia, sensory neuropathy, and others.49, 52, 53 The importance

of these side effects will be discussed in the next section.

Why nanocrystals made of paclitaxel?

The issue with new oncology drugs is their high attrition rate during Phase II and III

trials.54, 55 Specifically 90% of all new oncology drugs do not obtain market

approval.56 Analyzing the cause for these drop outs, it has been stated that poor

pharmacokinetic properties or bioavailability were only of minor influence, whereas

lack of efficacy and low margins of safety were the major obstacles.54 Thus, paclitaxel

as well as microtubule stabilizing agents in general will likely continue to be

important drugs in the treatment of cancer, even as more selective approaches are

developed.42

“Old” drugs have the advantage that they have already been proven to be safe and

effective. Nevertheless, they can be reformulated to increase the therapeutic index and

improve the compliance of patients. Systemic drug administration, unless very

selective for the target tissue, commonly affects all cells. In particular, targeting of a

drug to the tumor in cancer therapy is of paramount importance because side effects

such as neutropenia,57 neuropathy,46 hair loss,58 and mucositis59 are caused by the vast

distribution of the drug to other tissues. Clearly, the quality of life and psychosocial

burden of cancer have an effect on therapeutic outcome because patients prefer

treatments with less harsh side effects that do not disrupt their everyday life.58, 60

A potential avenue for improving therapy with paclitaxel is its formulation as

nanocrystals. Owing to their small size, they can take advantage of the so-called

“passive targeting” via the enhanced permeation and retention (EPR) effect.61 This

term refers to the leaky vasculature combined with often dysfunctional lymphatic

clearance as a result of the tumor’s fast and chaotic growth.62 Nanoparticles which are

sufficiently small (less than 400 nm) can extravasate through the leaky endothelium

and accumulate in tumoral tissues.63 Nanocrystals are, in theory, the ideal formulation

Page 25: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

22

approach for exploiting the EPR effect after i.v. injection because of their high drug

content paired with the poor solubility of the starting material. Furthermore, the

biodistribution could be favored to the tumor tissue if the nanocrystals carry a

targeting ligand and/or a trigger for selective drug release/uptake at the tumor site.

These features can potentially be included in the nanocrystal formulation by alteration

of the polymeric stabilizer. While an increase in tolerability of a formulation only

allows administration of higher doses, a targeted formulation would bring more drug

to the disease site, which effectively could reduce the overall dose to be given. This is

an opportunity to change the biodistribution of the drug, with more drug acting at the

site of disease and less causing adverse side effects because of systemic exposure.

What is the approach within this thesis?

In this doctoral work, paclitaxel nanocrystals were prepared by wet milling (Figures

2A and 2B) and stabilized with newly designed polymeric surfactants to prevent

aggregation (Figure 2C). In addition, it was the scope of this thesis to exploit

strategies for the improved targeting/accumulation and release of drug at the tumor

site by means of stabilizer modification. Novel biodegradable block copolymers were

developed for this task, which generally consisted of PEG (steric stabilization and

stealth properties) and a polyester (hydrophobic interaction with crystal surface,

possibility for postpolymerization modification). The polymers were modified with

cross-linking agents or reactive groups in order to improve their anchoring on the

particles’ surfaces and/or make their affinity responsive to stimuli such as oxidation.

In this context reactive oxygen species (ROS) are particularly interesting triggers as

they are associated with chronic inflammation and cancer.64, 65 Responsiveness to

ROS could eventually be exploited to promote deposition of the nanocrystals in the

tumor. Furthermore these polymeric stabilizers offer the possibility of more stably

attaching targeting moieties by use of heterotelechelic PEG derivatives, which may

favor accumulation or uptake of nanocrystals at specified locations. Nonetheless, it

remains critical that the surfactant−targeting agent construct is not shed rapidly after

administration due to changes in the equilibrium of adsorbed and desorbed stabilizer

upon dilution in the bloodstream.

Page 26: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

23

Figure 2. Wet milling of paclitaxel nanocrystals. (A) Zirconium oxide beads are

agitated in a vessel containing coarse drug powder and polymeric stabilizer solution.

(B) Attrition between beads breaks powder particles into drug nanocrystals. (C)

Diblock copolymer adsorbs to nanocrystal surface (yellow: mPEG, red: polyester).

In chapter III.1, different stabilizers are cross-linked around paclitaxel nanocrystals

by click chemistry to form polymeric “nanocages”. These nanocages function as

sterically stabilizing barriers to particle−particle interactions and prevent aggregation

as observed by size measurements over time by dynamic light scattering (DLS) and

transmission electron microscopy (TEM). Dialysis experiments combined with TEM

revealed the presence of nanocages. Centrifugation experiments showed that these

were less shed from the nanocrystal surface than comparable non-cross-linked

stabilizers. The development of these nanocages contributes an important general

approach for the preparation of poorly sheddable stabilizing coatings for nanocrystals,

and potentially also for other classes of nanoparticles for which covalent attachment

of the stabilizer is inappropriate (e.g., a drug) or impossible (e.g., chemically inert

material).

Chapter III.2 presents a modular and systematic strategy for preparing ROS

sensitive polymeric stabilizers with different affinity for drug nanocrystals both

before and after oxidation. Using the thiol−yne addition reaction, a library of ten

redox-responsive polymer stabilizers was prepared from two parent block

copolymers. The stabilizing potential for paclitaxel nanocrystals as well as the

influence of oxidation on size after exposure to ROS was studied by DLS. TEM as

well as dissolution tests before and after treatment with ROS were carried out. The

versatility of the presented postpolymerization modification approach makes it a

Page 27: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

24

potential platform for preparing triggered-sheddable stabilizing coatings for

nanoparticles.

Page 28: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

25

II.2. Targeting of Injectable Drug Nanocrystals1

Kathrin Fuhrmann, Marc A. Gauthier, and Jean-Christophe Leroux

Introduction

“Nano” drug delivery carriers are in many respects established technologies for

improving the therapeutic index of chemotherapeutic drugs and overcoming critical

formulation challenges of poorly water-soluble compounds. The latter are, for

instance, difficult to administer intravenously (i.v.) because of their potential

aggregation in the bloodstream, which can lead to embolism and accumulation in the

lungs. A variety of anticancer drug nanocarriers based on, for example, liposomes,66

micelles,67 polymeric nanoparticles,68 and so forth, are in clinical trials, and some

have reached the market. For example, liposomal doxorubicin (Doxil® or Myocet®)

has now been used for more than 15 years in the treatment of myeloma, breast cancer,

ovarian cancer, and AIDS-related Kaposi's sarcoma (see refs in 69). In comparison to

conventional drug formulations, nanocarriers can be advantageous because of the

lesser use of solubilizing agents or cosolvents (excipients), which are often a source

of side-effects such as hypersensitivity reactions (e.g., Cremophor EL® in the

paclitaxel formulation Taxol®).70 Furthermore, nanocarriers can reduce side-effects of

the drug by targeting them specifically to sites of disease rather than to healthy tissues

(e.g., cardiotoxicity of free doxorubicin).71 Two important remaining technological

challenges of nanocarriers, however, are the need to formulate drugs on a case-by-

case basis (i.e., due to the specific chemistry of each drug) and the difficulty

associated with transporting large amounts of drug to the site of the tumor (i.e., in part

because of moderate to poor drug loadings).

One of the most valuable “nano” opportunities in this field is to address these

challenges by creating nanocarriers composed of the drug itself.72-76 This strategy

reduces the use of “non-drug” material within the formulation compared to the other

nanocarriers above, and, if successfully targeted to the site of disease, should deposit

a significant amount of drug at this location. To this end, several drugs have been

processed into colloidal dispersions known as "nanocrystals" (NCs),25 and have been

1 Published in Mol. Pharm. 2014, 11, 1762–1771

Page 29: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

26

examined in cells, animal models, and in humans. In an excellent recent contribution,

Gao et al.21 describe the state-of-the-art of the in vivo performance of drug NCs. The

NC platform is particularly attractive because production can be achieved by a variety

of bottom–up and top–down approaches more-or-less irrespectively of the physical-

chemical properties of the drug.13, 31 Many techniques can lead to products with

reduced particle size, including sophisticated ones such as laser fragmentation24, 77 or

supercritical fluids with enhanced mass transfer,78, 79 but the most commonly used

techniques in industry are high pressure homogenization, and wet milling.80, 81 For

example, wet milling can produce unimodal NCs with mean diameters in the ca. ~200

nm range with little batch-to-batch variability.82 This process is suitable for many

different classes of compounds and there currently exist a variety of oral NC

formulations produced by wet milling on the market, including Rapamune®

(sirolimus), Emend® (aprepitant), TriCor® (fenofibrate), and Megace® ES (megestrol

acetate).73, 83

Opportunities That “Nano” Brings to Targeted NC Delivery. Because of their

adjustable sub- ~400 nm size, intravenously injected NCs can, in principle,

extravasate from the blood through the leaky endothelium and accumulate in tumoral

tissue via the enhanced permeation and retention effect.63 In addition to this passive

targeting phenomenon, the stabilizers used to mask the high-energy drug surfaces

created during the size-reduction process can be functionalized with

targeting/internalizing ligands to promote active tumor accumulation or uptake,

respectively. These stabilizers, for example polymers or surfactants, typically stabilize

NCs by adsorbing to the surface of the NCs and providing steric (e.g., poloxamers,

cellulose derivatives)28 or electrostatic (e.g., sodium dodecyl sulfate)84 barriers to

aggregation.25, 26, 82, 85 Steric stabilization is most efficient in a good solvent (for the

stabilizer) and a minimum layer thickness is required,86 while the comparable

guidelines for electrostatic repulsion are that the absolute value of zeta potential

should be at least 30 mV.26 As only a small amount of stabilizing agent is typically

required to mask NCs and prevent their aggregation, drug content of typically 50 to

90 wt% has been reported,40, 87 which is dramatically higher than for other nanocarrier

systems.

Challenges That “Nano” Brings to Targeted NC Delivery. NCs exhibit a

characteristic nonlinear increase of kinetic solubility upon miniaturization that is

described by the Ostwald–Freundlich equation.88 This phenomenon, which only

Page 30: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

27

becomes evident when particles reach the submicrometer size range, dramatically

increases their rate of dissolution and is generally exploited for the “untargeted”

administration of insoluble drugs, with little need for solubilizing agents or

cosolvents.21 However, enhanced dissolution complicates targeted NC delivery due to

off-target drug delivery, insufficient circulation time for passive targeting, and

potential shedding of the stabilizing agent/targeting agents used for active targeting.

In addition, passive targeting via the enhanced permeation and retention effect is

increasingly becoming a subject of debate. 89

Overall, despite the fact that drug NCs have been studied and used clinically for

nearly three decades, enhancing NC uptake through specific interactions in vitro, and

targeting in vivo remain elusive objectives. This noncomprehensive review highlights

the opportunities offered by NCs as well as the important challenges that remain for

achieving targeted delivery. For this purpose, selected studies on drug NC

performance, irrespective of disease treated, in cell culture models and in vivo after

parenteral administration are presented, and current and future avenues of research for

enhancing their therapeutic potential are discussed. It should be noted that another

advanced nanoparticulate system, namely Abraxane®, is also presented in this review.

Abraxane® is an injectable paclitaxel formulation produced by high-pressure

homogenization in the presence of human serum albumin and used for the treatment

of metastatic breast cancer.90 Although not a (nano)crystalline material per se,48 the

clinical use of this product and its multiple physical–chemical similarities to other

NCs supports its inclusion in this discussion, for comparative purposes.

In Vitro

Cell-based assays are often used prior to in vivo experimentation to validate the

performance of drug NCs in comparison to other formulations. In the case of

anticancer drugs, the cytotoxicity of NCs compared to that of the free drug in solution

or within other nanocarriers is used as a parameter for establishing activity. For drugs

with limited cytotoxicity, uptake is either measured directly or via specific assays

associated with their mechanism of action. Unfortunately, due to the specific

conditions used in these assays, many studies involving drug NCs have yielded

disparate results, which complicate generalizations and extrapolation of in vitro

findings on NCs to the in vivo setting.

Page 31: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

28

One fundamental, but sometimes forgotten, characteristic of cell culture assays is

that they are performed within a closed system, of finite volume. As a consequence,

the rate of drug dissolution from the NC, and its resulting consequence on therapeutic

efficacy, can cease to depend on the dose administered if it is above the saturation

solubility of the drug. In addition, the relationship between incubation time and

cytotoxicity will depend on the dissolution rate of the NC. For instance, when the

contact time with cells is short, rapidly dissolving NCs of cytotoxic drugs should have

a comparable effect on cell viability as the free drug in solution, and more slowly

dissolving NCs should be less cytotoxic.91 That is, when uptake is rapid compared to

NC dissolution, structural or chemical parameters associated with the NC may play a

role in performance. In support of this, Shegokar et al.92 have shown that the in vitro

uptake after 2 h of ca. 450-nm NCs formed of the antiretroviral drug nevirapine by

macrophages was sensitive to the nature of the stabilizer, indicating that the NCs are

still intact and have not shed their stabilizing coating within this time frame. The

authors notably observed that the NCs coated with poly(ethylene glycol) were less

taken up by cells in comparison to NCs coated with dextran or albumin, in accordance

with the stealth-like behavior previously reported for this polymer. NC endocytosis

appears to be clathrin and caveolae mediated, as observed for 240-nm NCs of

anticancer drug camptothecin with needle-like morphology.93

The situation changes when longer incubation times with NCs are used, given that

dissolution may occur early in the overall incubation process. For example, 240-nm

NCs of camptothecin showed similar cytotoxicity to that of the solution after 72 h of

incubation.93 Indeed, rapid dissolution has been observed in a number of reports. For

instance, 125-nm tamoxifen NCs coated with three bilayers of

poly(dimethyldiallylamide ammonium chloride) and poly(styrenesulfonate) were

~50% solubilized within 2 h under sink conditions.40 In vitro, Ben Zirar et al.94 have

evaluated the viability of both K562 and U937 cells after 48–72 h incubation with

melarsoprol either as a free drug solution or as 300–600 nm poloxamer-stabilized

NCs (Figure 1A). Under these incubation conditions, differences between the NC and

the free drug, when statistically significant, were generally small. Vergara et al.95

have assessed the cytotoxicity of ~150-nm paclitaxel NCs stabilized by electrostatic

layer-by-layer (LbL) assembly of alginic acid and chitosan. Interestingly, the authors

observed that cell viability ceased to decrease when the dose of paclitaxel was

increased beyond ca. 5–10 ng·mL–1. At first glance, this result appeared to indicate

Page 32: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

29

that saturation (and thus prevention of NC dissolution) was occurring in the culture

medium, despite the fact that these concentrations were well below the saturation

solubility of paclitaxel in water (300 ng·mL–1).96 However, only marginal differences

were observed between the NC and freely soluble drug (Figure 1B), suggesting that

dissolution had occurred well within this timeframe in cell culture medium. The

authors attributed this phenomenon to the poor effectiveness of paclitaxel in OVCAR-

3 cells due to the expression of the multidrug resistance transporter MDR1. This study

points to the necessity of performing adequate control experiments with freely soluble

drug. More recently, the uptake and intracellular trafficking of larger (ca. 300–900

nm) NCs of the antiretroviral drug ritonavir has been examined in macrophages.97

Testing NCs of drugs with limited cytotoxicity allowed the authors to evaluate uptake

at higher concentrations (100 µM). The tested NCs were stabilized with a mixture of

poloxamer 188, 1,2-distearoyl-phosphatidyl ethanolamine-methyl-poly(ethylene

glycol) (2 kDa), and 1,2-dioleoyl-3-trimethylammonium propane. The authors

demonstrated by electron microscopy that these NCs loaded into macrophages and

remained mostly intact (68%) 24 h postuptake (Figure 2A). In addition, the NCs

appeared to aggregate with time within the cells and possessed rougher edges.

Sustained release of drug from the macrophages, which serve as drug NC reservoirs

in this example, was observed for a variety of antiretroviral drug NC loaded

macrophages for a prolonged period of time extending over a period of two weeks

and longer.98 In an extension of this work, the uptake of 21 different NCs (ca. 200–

400 nm) of four antiretroviral drugs has been evaluated in macrophages under

comparable conditions (100 µM).99 The authors observed that drug type, surfactant

coating, and NC shape had substantive effects on NC uptake, release, and

antiretroviral response. NCs with rounded and irregular edges showed diminished cell

uptake, while rod-like NCs with smooth and regular edges were taken up more

rapidly, and the loaded macrophages slowly released the drug in a period of days.

Page 33: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

30

Figure 3. Drug solution-like behavior of NCs in vitro. (A) Viability of both K562

and U937 cells after 48–72 h incubation with melarsoprol either as a free drug

solution, as 300–600 nm poloxamer-stabilized NCs, or as a drug–hydroxypropyl--

cyclodextrin (HPCD) complex (*: p < 0.01 versus free melarsoprol). Redrawn from

Ben Zirar et al.,94 with permission from Elsevier (B) Cell viability of OCVAR-3 cells

decreases as a function of paclitaxel concentration for both free paclitaxel, and

paclitaxel NCs up to ca. 5–10 ng·mL–1 (24 h incubation time), after which it is

unaffected. This phenomenon was attributed to the expression of the multidrug

resistance transporter MDR1, rather than saturation of the medium with paclitaxel.

Little differences are observed in comparison to the free drug. Redrawn from Vergara

et al.,95 with permission from Elsevier.

Page 34: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

31

Figure 4. Particle-like behavior and enhanced NC uptake in vitro. (A) Electron

micrographs of ritonavir NCs prior to macrophage uptake, within macrophages, and

after release from macrophages into the surrounding medium (24 h after uptake).

Adapted from Kadiu et al.,97 with permission from Future Medicine. (B) Paclitaxel

NCs targeting the folate receptor are more cytotoxic than untargeted ones in a human

folate-receptor-positive oral carcinoma cell line. The difference between the targeted

and untargeted NCs disappeared when excess free folic acid was added to compete for

the cell-surface receptor (*: p < 0.01 versus in presence of excess free folic acid).

Redrawn from Liu et al.,91 with permission from John Wiley and Sons.

Despite the apparent ability to maintain, in certain cases, the integrity of NCs in the

presence of cells over a certain period of time, to the extent of our knowledge, few

attempts have been made to modify the surface of the NCs with targeting ligands to

improve cellular uptake in vitro. Liu et al.91 have shown that ca. 150-nm paclitaxel

NCs coated with poloxamer 407 bearing 10% folic acid as targeting ligand were

significantly more cytotoxic than the comparable nontargeted NCs at short incubation

times (2 h; Figure 2B). This effect was abolished in a competition assay with free

folic acid potentially indicating that drug uptake was associated with folate-mediated

receptor-mediated endocytosis. The cytotoxicity of the untargeted NCs was not

affected by addition of folic acid. One caveat, however, is that cytotoxicity is an

indirect measurement of drug uptake that cannot distinguish the folate-mediated

uptake of free versus that of NC-associated paclitaxel. More recently, Bui et al.100

have prepared fluorescent and biotinylated squalene–gemcitabine (prodrug) NCs and

have observed increased cell uptake and improved anticancer efficiency in three

cancer cell lines.

Page 35: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

32

Overall, several cell culture studies support that the drug NCs can remain intact for

a certain time and indeed behave like nanoparticles rather than freely dissolved drug.

The dominant factors in vitro for maintaining NC integrity are size and concentration

in the medium. Depending on the in vivo application foreseen, in vitro experiments

might sometimes benefit from being performed under more dilute conditions. This

would avoid saturating the medium with drug, which prevents NC dissolution.

Dissolution profiles in the absence of cells and under sink conditions are indeed not

always performed in the literature. Imaging of NCs within cells may also provide

more insight into how the performance of NCs can be rationally altered.

In Vivo

Quite often, NCs display pharmacokinetic profiles that are very similar to the drug

solution when administered i.v.30, 101-103 This is generally a consequence of their rapid

dissolution under in vivo sink conditions. In Mouton et al.’s101 report on an early

clinical study in humans, the authors compare 200–300 nm itraconazole NCs to an

itraconazole–hydroxypropyl--cyclodextrin complex. The NCs exhibited a higher

mean maximum plasma concentration at the end of infusion than those receiving the

cyclodextrin formulation (Figure 3). The authors speculated that this difference may

be explained by assuming that NCs were not yet dissolved, and were consequently

confined in the circulatory system, and unavailable for diffusion and distribution to

the peripheral tissues. However, after this time point, the differences between the two

formulations with regards to the other pharmacokinetic parameters was less

pronounced or was not significant at all. In addition, the authors also mentioned that

other (unpublished) data with several animal species showed that the drug NCs were

specifically trapped in Kupffer cells in the liver and in the macrophages of the spleen

and that pharmacokinetic changes were related to the size of the NCs (most

pronounced for NCs 340 nm). The similar plasma concentration profiles and high

drug concentrations early in the liver obtained for 200-nm NCs of an antitumor p-

terphenyl derivative versus the drug solution could also be explained by fast

dissolution and NC instability.102 In a previous study this NC formulation exhibited

complete dissolution within 2 h compared to less than 10% for the bulk drug.104

Sharma et al.103 have observed that the sub-150-nm NCs of the investigational

anticancer compound SN 30191, stabilized with poloxamer 407 and poly(ethylene

Page 36: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

33

glycol)-15-hydroxystearate, were rapidly cleared from the blood of mice and

accumulated in the kidney, liver, and heart. The authors postulated that drug

accumulation in these tissues could be due to rapid dissolution of the NCs in the

blood, which facilitated distribution in highly perfused tissues. Unfortunately, as the

free soluble formulation of SN 30191 was four times less tolerated than the NCs,

comparison between the two was not possible. Sigfridsson et al.30 have compared

100–150 nm (amorphous drug) nanoparticles, 300–400 nm NCs, and the solution of

the investigational antipsychotic drug AZ68. Both nanoparticles and NCs were

stabilized with poly(vinylpyrrolidone) and a combination of small-molecule

surfactants. When administered i.v. to rats, no significant difference among the three

formulations was observed in terms of their plasma profiles.

Figure 5. Drug solution-like behavior of NCs in vivo. Multiple-dose study

comparing itraconazole NCs to an itraconazole–hydroxypropyl--cyclodextrin

(HPCD) complex administered i.v. to humans. Two hundred milligramm doses were

given every 24 h except on days 1 and 2, when the dose was given every 12 h. Note

that samples were collected just before and 1 h after each infusion for the first 5 days.

Redrawn from Mouton et al.,101 with permission from American Society for

Microbiology.

Differences between the NC and other formulations begin to manifest themselves

when the particle size is large. Ganta et al.105 have prepared 130−700 nm poloxamer

188-stabilized NCs of asulacrine and, when administered i.v. to mice, observed

preferential accumulation in the liver compared to the free drug in solution. The

authors rationalized this result to stem from uptake of the NCs by the mononuclear

Page 37: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

34

phagocyte system, which removed them from the systemic circulation. From the

phagocytes, the drug was released over a period of a couple of hours, a timeframe that

was consistent with the in vitro dissolution profiles of the NCs in 1% polysorbate 80

solution. This is in line with the observation of enhanced liver accumulation and

retention of radioactive 450-nm nevirapine NCs in the liver compared to the drug in

solution (Figure 4A).92 Gao et al.106 have investigated the effects of particle size on

the pharmacokinetics and tissue distribution of two oridonin NCs with markedly

different size (ca. 100 and 900 nm) following i.v. injection in rabbits. In vitro,

complete dissolution occurred within 10 min and 2 h, for the smaller and larger NCs,

respectively. In vivo, the smaller NCs behaved similarly to the drug in solution,

whereas the larger NCs accumulated to a greater extent in the liver, spleen, and lungs.

Based on these findings, the authors suggested that the larger NCs were subjected to

mononuclear phagocyte system uptake. Indeed, Rabinow et al.107 have demonstrated,

seven days postinjection, that 600-nm itraconazole NCs stabilized with poloxamer

188 were taken up intact by the spleen in rats by histological analysis (Figure 4B).

Unfortunately, organ toxicity resulting from NC accumulation in the liver and spleen

was so far not assessed. Nevertheless, such toxicity would be drug dependent and

would have to be evaluated for each composition. In addition, continuous

accumulation and organ toxicity, as observed for biopersistent nanoparticles, such as

asbestos and carbon nanotubes, is unlikely since drug NCs dissolve with time and are

eliminated metabolically or by renal excretion.108, 109

Page 38: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

35

Figure 6. Particle-like behavior of NCs in vivo. (A) Gamma scintigrams depicting

biodistribution of bare radiolabeled nevirapine 450-nm NCs and NCs surface-coated

with albumin and dextran at 1 h and 24 h in rat compared to the drug in solution.

Reproduced from Shegokar et al.,92 with permission from Elsevier. (B) Histological

analysis of rat spleen by transmission electron microscopy shows the presence of

itraconazole NCs within macrophages. Adapted from Rabinow et al.,107 with

permission from Elsevier.

In contrast to the aforementioned i.v. injection, however, 200-nm rilpivirine NCs

administered as a single intramuscular or subcutaneous injection achieve stable

sustained plasma concentration profiles detectable up to three months in dogs.36 With

respect to NC size, the 200-nm NCs displayed improved early release (higher Cmax) in

dogs, when compared with 400 or 800 nm particles. For instance, 40 × 150-nm

paclitaxel NCs stabilized with D--tocopheryl poly(ethylene glycol) 1000 succinate

(TPGS) exhibited greater antitumor efficacy than Taxol® at equivalent dose in a drug

resistant NCI/ADR-RES xenograft mouse model.34 Although it was hypothesized that

the improved activity of the NCs could be attributed to the inhibition of efflux pump

(permeability glycoprotein 1; P-gp) function by TPGS, as reported elsewhere,110 the

slow dissolution of the NCs observed in vitro (<20% in 24 h) might have contributed

to a better drug distribution to the tumor via the EPR effect. Unfortunately, in this

study the pharmacokinetics and biodistribution of the drug were not assessed.34 Zhang

et al.93 observed a greater antitumor efficacy for 240-nm uncoated NCs of

camptothecin in an MCF-7 tumor xenograft mouse model compared to the salt

solution of camptothecin in a mixture of propylene glycol and saline. They attributed

Page 39: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

36

this to the EPR effect and the higher resistance of NCs against hydrolysis. This was

further supported by biodistribution data showing higher camptothecin deposition in

the tumor and reduced drug hydrolysis. However, the pharmacokinetic profiles were

difficult to interpret as the NCs displayed in rats a lower area under the plasma

concentration versus time curve than the drug solution and a comparable mean

residence time.111 Based on previous data,93 this may be indicative of retention of the

uncoated needle shape NCs in the lungs but could also result from their aggregation

and embolization in the lung capillaries. Hollis et al. developed 200-nm hybrid NCs

consisting of partially radiolabeled paclitaxel and the fluorescent dye FPI-749.89 Both

NCs and paclitaxel solution, injected i.v. into HT-29 tumor xenograft bearing mice,

accumulated less than 1% at the site of the tumor as determined by scintillation

counting. Additionally, repeated injections of both formulations gave no significant

difference in treatment efficacy at the defined end point.

While not a NC per se, Karmali et al.112 have modified Abraxane®, an amorphous

paclitaxel nanoformulation stabilized with albumin (130 nm),113 with tumor-targeting

peptides and observed a change in the biodistribution 3 h postadministration versus

the untargeted formulation. Indeed, at this time point, the targeted NC colocalized

with its target while untargeted form did not. Unfortunately, targeted Abraxane® only

showed a small effect in inhibiting MDA-MB-435 tumor growth in comparison to its

unmodified form. This may attest to some dissolution prior to efficient targeting of

the NC, and should be further investigated.

Stabilizer: To Shed or Not To Shed

One important limitation in the design of therapeutic drug NCs is the desire not to

chemically modify the NC itself with the stabilizing agent. As such, stabilizers cannot

be covalently anchored onto the NC surface, and non-specific interactions must thus

be exploited for adsorption. Independently of the dissolution of the NC that leads to

desorption of the stabilizer (vide infra), high dilution conditions encountered either in

vitro or in vivo will inevitably lead to loss of the stabilizing agent as well as any

appended targeting/internalizing agents. For instance, Deng et al.114 have shown

(through NC size increase) that poloxamer 407 desorbs from paclitaxel NCs upon

dilution. Another observation was that increasing the stabilizer-to-drug ratio resulted

in poorer NC stability. Supported by evidence that higher concentrations of stabilizer

Page 40: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

37

led to the formation of micelles in addition to stabilizing NC coatings, the authors

hypothesized that stabilizers deposited as unimers (i.e., below the CMC) may have

higher affinity to NCs than stabilizers deposited as multimers (i.e., above the CMC),

whose deposition process was in competition with micellization. This result could

potentially attest to a different organization of the stabilizer on the surface of the NC.

Indeed, owing to the complexity of systematically altering the structure of

macromolecular stabilizing agents, few studies have attempted to rationally modulate

interactions between the stabilizer and the NC for preventing desorption. Our group

has recently presented a modular and systematic strategy for optimizing the affinity of

polymeric stabilizers for NCs based on the postpolymerization modification of

polymer precursors (containing -propargyl-δ-valerolactone)115 by thiol–yne click

chemistry.87 In this approach, two parent block copolymers of methoxy poly(ethylene

glycol)-b-(-propargyl-δ-valerolactone-co-ε-caprolactone) were used to create a

library of 10 different stabilizers in which the hydrophobic polyester block was

modified with alkanes of different length and structure. All stabilizers had equivalent

numbers of monomeric units and polydispersity indices to the parent stabilizers.

Under the production conditions used, all stabilizers produced dense polymer brushes

on the surface of the NC, and size-stability assays were found to strongly depend on

the structure of the hydrophobic block.

In addition to altering the chemical structure of the stabilizer to promote

interactions with the NCs, another approach is to cross-link the stabilizer around the

NC and thus reduce shedding via physical entrapment. For instance, Kim and Lee116

have electrostatically cross-linked chitosan on the surface of paclitaxel with

tripolyphosphate, but have not evaluated the size stability of the NCs, nor the decrease

of shedding achieved after cross-linking. Other electrostatically cross-linked

stabilizers produced via the LbL deposition of polyelectrolytes are discussed in the

following section. Our group has more recently designed block copolymer stabilizers

that could be cross-linked directly on the surface of paclitaxel NCs by copper-

catalyzed 1,3-dipolar cycloaddition to form nanocage–NC constructs.39 Size-stability

analysis showed that nanocages acted as sterically stabilizing barriers to NC–NC

interactions and aggregation, which in turn imparted better size-stability to the NCs in

comparison to the non-cross-linked coating. By dosing the amount of polymer

released from nanocage–NC constructs, it was shown that the nanocages were 3–4-

Page 41: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

38

fold less shed from the NCs than comparable non-cross-linked stabilizers. In addition,

transmission electron microscopy of the nanocages after complete dissolution of the

drug NC revealed the intactness of the nanocage, demonstrating a successful cross-

linking reaction (Figure 5A).

It should be noted, however, that shedding of the stabilizer may in fact be beneficial

under certain circumstances. For instance, Liu et al.34 have sought to exploit the

shedding phenomenon by stabilizing 40 × 150 nm paclitaxel NC rods with TPGS. The

rationale of this study was that the tocopheryl-functionalized stabilizer may inhibit P-

gp upon shedding, which may permit an enhanced treatment of multi-drug resistant

cells. Indeed, the authors observed that in NCI/ADR-RES cells, which overexpress P-

gp and are resistant to paclitaxel, NCs stabilized with TPGS exhibited a significantly

enhanced antiproliferative effect than free paclitaxel or paclitaxel NCs stabilized with

poloxamer 407 (Figure 5B). The authors also observed that as the amount of TPGS

increased compared to drug, the antiproliferative effect increased for both TPGS-

stabilized NCs and the physical mixture, indicating that TPGS modulated drug

resistance transporters. Interestingly, however, at low TPGS concentrations, TPGS-

stabilized NCs were more cytotoxic than the mixture, whereas at high surfactant

concentrations they were comparable. These observations indicate that additional

mechanistic investigations are warranted. In another example, our group has created

amphiphilic block copolymer stabilizers that are spontaneously shed in response to a

stimulus.87 More specifically, the hydrophobic block of the stabilizer, responsible for

physisorption on the investigated paclitaxel NCs, contained thioether groups that

became substantially more hydrophilic in the presence of reactive oxygen species,

thus driving the stabilizer from the NC and provoking its destabilization. Stabilizer

shedding in areas of oxidative stress in the body, which are associated with a variety

of diseases,117, 118 may provide a means of provoking selective aggregation or promote

uptake at these target locations, but should be tested in vivo.

Page 42: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

39

Figure 7. Controlling or exploiting stabilizer shedding. (A) Preventing NC

stabilizers from shedding by cross-linking. Transmission electron microscopy images

of paclitaxel NCs before (I) and after cross-linking (II). Following dissolution of the

NCs from (I) and (II), an aggregate structure was observed for the non-cross-linked

NCs (III), whereas intact polymeric coatings were observed for the correspondingly

cross-linked NCs, in the form of discrete spheroids (IV). Reproduced from Fuhrmann

et al.,39 with permission from American Chemical Society. (B) Stabilizer shedding

promotes activity of NCs in vitro. Effects of paclitaxel/TPGS NCs (10 µM) with

different amounts of TPGS, in comparison to a physical mixture of paclitaxel and

TPGS demonstrating that the “shed” stabilizer potentiates the activity of paclitaxel.

Redrawn from Liu et al.,34 with permission from American Chemical Society.

Altering Dissolution Profiles (by Means Other than Size)

It is clear from the examples above that size plays a key role in the dissolution

characteristics of drug NCs, which alters their performance. Based on in vivo

evidence, NCs with sizes above ca. 300–400 nm (depending on the specific drug in

question) persist for a sufficiently long time that they could, in principle, accumulate

passively within tumors via the EPR effect. However, particles of this size may be

subject to greater uptake by the mononuclear phagocyte system and would poorly

diffuse in the extracellular tumoral matrix. Smaller NCs may have greater abilities to

penetrate tumors, but their targeting is more challenging because dissolution must be

delayed. Several approaches have been examined for this purpose.

Page 43: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

40

Drug NCs can be reprocessed following miniaturization to alter dissolution kinetics.

For instance, paclitaxel NCs could be renanozised by an incubation–sonication

technique.114 In this technique, the authors incubated NCs at 37 ºC for a certain period

during which time NC size increased via ripening processes. This was then followed

by sonication to break the growing NCs into smaller ones. The authors observed that

the renanosized NCs displayed significantly greater size stability, which they

attributed to the disruption of the preferred growth pattern of the NCs. These

interesting findings should be pursued with analysis of dissolution kinetics under sink

conditions and a more in-depth characterization of this phenomenon. Lu et al. also

produced very stable 300-nm NCs of paclitaxel by adsorption of transferrin. The NCs

did not exhibit a size change during the 3 months study period. The increased stability

compared to the bare NCs was also reflected in a slightly slower drug release during

dissolution experiments.119

One of the most investigated systematic approaches for altering microparticle120, 121

and, more recently, NC dissolution kinetics via stabilizing coatings produced by LbL

assembly of polyeletrolytes. In this technique, the hydrophobic drug NC is first

covered by an anchoring layer typically composed of a small molecule amphiphile

and a polymer, and is followed by the sequential deposition of multiple layers of

charged polyelectrolytes. Model experiments have shown these coatings are

semipermeable (i.e., permeable to small molecules smaller than specific cut-offs),122

which points to the importance of well characterizing the coating. Despite this

semipermeability, an effect on dissolution rate of NCs, albeit a small one, has been

observed. For instance, the rate of drug release from 300-nm paclitaxel NCs was

independent of the thickness of the stabilizing coating, when this coating was thinner

than 3.5 bilayers of poly-L-lysine and sodium heparin (Figure 6).123 However, when

the number of bilayers increased from 4 to 12, a slight decrease in the drug release

rate was observed. Agarwal et al.40 have observed a marginal difference in the rate of

dissolution of 125-nm tamoxifen NCs stabilized with either 0.5 or 3 bilayers of

poly(dimethyldiallylamide ammonium chloride) /poly(styrenesulfonate). One

particularly interesting feature of the LbL approach is the large parameter space

available for constructing these stabilizing coatings including: polyelectrotyte

type/architecture/molecular weight, addition of salts and other additives, anchoring

layer chemistry. Future work should focus on assessing how these parameters inflence

Page 44: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

41

the semipermeability of LbL-assembled coatings, which may provide the means to

rationally alter NC dissolution.

Figure 8. Altered NC dissolution via the stabilizing coating. Paclitaxel release from

300 nm NCs coated with (poly-L-lysine/heparin)n shells. Redrawn from Shutava et

al.,123 with permission from Royal Society of Chemistry.

Outlook

The studies presented above suggest that drug NCs may in the future play an

important role in targeting significant amounts of drug to sites of disease. NC size can

for all intents and purposes be selected using (universal) preparation approaches – NC

persistence in vitro/in vivo strongly correlates with size – and cellular uptake and

tumor accumulation can be promoted by modifying the surface chemistry of the

coating. While these aspects have been individually demonstrated in the examples

discussed herein, future research should question how to overcome the challenges

associated with assembling these individual properties into efficient and optimized

therapeutics. For instance, as small (sub-100 nm) nanocarriers are known to penetrate

even poorly permeable tumors,124, 125 could such an effect also be achieved in practice

with NCs? Can the rate of drug release of these smaller, dissolution prone NCs be

controlled by their stabilizing coating? Can stimuli-responsive stabilizers provide a

means for selective NC destabilization and accumulation at sites of disease? In

addition, other forms of targeting strategies in vivo could be foreseen. For instance, it

has been recently reported that composite nanoparticles of a gemcitabine prodrug and

magnetite yielded enhanced tumor accumulation and therapeutic activity via magnet-

assisted targeting.126 Finally, other opportunities for enhancing the targeting potential

Page 45: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

42

of NCs should be explored and which are not part of the NC construct itself. Sugahara

et al.127 have indeed recently demonstrated that tumor-penetrating peptides,

coadministered with Abraxane®, increased vascular and tissue permeability leading to

a 12-fold increase of the tumor accumulation versus in the absence of peptide. This

area is ripe for discovery and answering these questions will require creative new

hypotheses to be tested.

Page 46: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

43

III. Results and Discussion

Page 47: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

44

Page 48: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

45

III.1. PEG Nanocages as Non-Sheddable Stabilizers for Drug

Nanocrystals2

Kathrin Fuhrmann, Jessica D. Schulz, Marc A. Gauthier, and

Jean-Christophe Leroux

Introduction

Many modern and potent anticancer drugs are poorly water soluble, which can

create formulation problems when the drug is intended to be injected by the

intravenous (i.v.) route.128 Drug solubility may be altered on a number of levels

including modification of the drug’s structure (e.g., change of crystalline form)1 and

by formulation approaches.129 For example, paclitaxel (PTX), a potent antitumor

agent, is typically formulated in a vehicle composed of polyethoxylated castor oil and

dehydrated ethanol (Cremophor EL; Taxol).130 However, owing to the required dose

of Cremophor EL necessary to deliver a sufficient amount of PTX, a number of

undesirable side effects have been observed.5 In addition to the desire of eliminating

large amounts of excipient, modern formulation strategies are also designed to

increase the drug’s therapeutic index by exploiting passive131, 132 and more recently

active targeting. For instance, PTX has been formulated as a water-soluble prodrug,133

as a polymer nanoconjugate,134 conjugated to the natural lipid squalene,135 in

liposomes,136 with carbon nanotubes,137 as cyclodextrin complexes,138 and in

micelles.139 Formulations that have been successfully applied or tested in the clinic

are Abraxane, an albumin-bound nanoparticle system,113 and Opaxio, a poly(L-

glutamic acid)-based PTX conjugate (clinical phase III underway).140 Nanosized

systems have the potential to accumulate at cancer sites through the enhanced

permeation and retention (EPR) effect, and can possess targeting agents to promote

active uptake by target cells or deposition at specific tumoral sites.

Current limitations include insufficient solubilization capacity,141 potential

excipient-related toxicity,5, 142 instability upon dilution in the bloodstream,143, 144

manufacturing difficulties, and drug stability issues during processing.145 One

2 Published in ACS Nano 2012, 6, 1667-1676. J.D. Schulz synthesized a starting compound.

Page 49: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

46

validated and robust approach for addressing these issues is to process the drug

powder into colloidal dispersions known as “nanocrystals” (NCs).25, 77, 80, 81 Wet

milling can produce uniformly sized drug NCs with mean diameters of less than 200

nm, and little batch-to-batch variability.82 This top-down process is suitable for many

different classes of compounds and there currently exist several oral formulations

produced by wet milling on the market.83 One key feature of NCs is the minimal use

of excipients compared to other formulation approaches, which implies both high

drug content and diminished excipient-related toxicity. For the i.v. route, NCs are

promising drug carriers because their very high loading can lead to higher deposition

of drug in cancer cells upon delivery.91 Indeed, there is increasing evidence that drug

NCs can accumulate in tumors via the EPR effect.34

NCs are generally stabilized with surfactants (or “stabilizers”), whose functional

role is to prevent aggregation between the high-energy crystal surfaces produced

during the size-reduction process. Consequently, much research is now directed

towards optimizing the coating.72 For example, Agarwal et al. have produced stable

NCs of hydrophobic drugs by sonication and layer-by-layer assembly of oppositely-

charged polyelectrolytes leading to electrostatic repulsion between NCs.40 Similarly,

Abraxane is an injectable NC formulation of PTX produced by high-pressure

homogenization in the presence of human serum albumin, which acts as a steric

stabilizer for the NCs.146 In addition, the stabilizer provides a means of anchoring

targeting moieties to the NCs. For example, Liu et al. have modified poloxamer 407

with folic acid and have shown enhanced cellular uptake of targeted PTX NCs, in

comparison to untargeted ones.91 A recent study by Sugahara et al. has investigated

the efficacy of PTX NCs when coadministered with a tumor-penetrating peptide and

of Abraxane decorated with the latter.127 The authors have shown that these

approaches could slightly enhance the antitumoral effect.112 However, a critical

challenge in achieving active targeting is the possibility that the targeting agents are

shed along with the stabilizer upon high dilution. For instance, Deng et al. have

indirectly, shown that poloxamer 407 can desorb from PTX NCs upon mild heating or

dilution.114 The authors also found that the conditions under which the NCs are

prepared can lead to different affinities between the polymer and the drug.

The objective of this study was to prepare non- or poorly-sheddable biodegradable

stabilizers for NCs that are maintained in place independently of specific stabilizer–

drug interactions. It was hypothesized that this could be accomplished by chemical

Page 50: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

47

cross-linking of the stabilizer around the NC, thereby maintaining the drug in place by

a combination of physical entrapment and physisorption. Conceptually, the cross-

linked polymer coating forms a polymeric “nanocage” in which the NC is trapped.

Earlier work on nanocages has been reported by Turner et al.147 These were prepared

in a multistep process involving shell cross-linking of micellar structures, core

digestion and functionalization with lipids to improve subsequent drug loading. In

contrast to their method, the specific challenge addressed herein lies in performing

cross-linking reactions near the surface of PTX NCs, due to the presence of high-

reactive crystal surfaces and to reactions taking place in heterogeneous phase. As

illustrated in Figure 9, herein is reported the preparation of PTX NCs by wet milling

with amphiphilic biodegradable and cross-linkable copolymers, and conditions for

successfully cross-linking them to form a nanocage–NC construct. Evidence for the

lower sheddability of the nanocage in comparison to non-cross-linked stabilizers is

given. This approach is advantageous because it can easily be adapted to the grafting

of targeting ligands. In addition, as the stabilizing coating is maintained in place

more-or-less independently of specific drug–polymer interactions, it should be

amenable to other hydrophobic drugs.

Figure 9. Schematic representation of diblock-copolymer self-assembly on the

surface of drug NCs and nanocage formation following cross-linking.

Experimental Section

Materials

PTX was obtained from Bioxel Pharma Inc. (Sainte-Foy, QC, Canada) and

docetaxel from ScinoPharm Taiwan, Ltd. (Tainan County, Taiwan). Sepharose CL-

4B, Sephadex G10, lithium diisopropylamide (LDA), -valerolactone, propargyl

bromide, hexamethylphosphoramide (HMPA), -caprolactone, 2,6-bis(4-

Page 51: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

48

azidobenzylidene)-4-methylcyclohexanone (97%, containing 35 – 45% water),

sodium azide, copper(II) sulfate, albumin from bovine serum (BSA; ≥ 96%), and

methoxy poly(ethylene glycol) (mPEG; 2000 g/mol) were purchased from Sigma-

Aldrich (Buchs, Switzerland) and used as received. Hydrogen chloride (1 N solution

in diethyl ether), m-chloroperoxybenzoic acid (mCPBA, 70 – 75%), and 1-methyl-2-

pyrrolidone were purchased from Chemie Brunschwig AG (Basel, Switzerland). 1,11-

Diazido-3,6,9-trioxaundecane was obtained from Santa Cruz Biotechnology (Santa

Cruz, CA), sodium L-(+)-ascorbate from Axon Lab AG (Baden-Dättwil, Switzerland),

and deuterium oxide from ReseaChem GmbH (Burgdorf, Switzerland). Ultra pure

water was prepared by a Barnstead Nanopure system (Thermo Fisher Scientific,

Reinach, Switzerland). Dry solvents were taken from a solvent purification system

(LC Technology Solutions Inc., Seabrook, NH). -valerolactone and -caprolactone

were distilled over calcium hydride under inert atmosphere before use. All other

solvents were of highest purity and bought from Sigma-Aldrich (Buchs, Switzerland).

Synthesis of -propargyl-δ-valerolactone (1)

The synthesis of 1 was adapted from Parrish et al.115 LDA (6.49 mL, 11.69 mmol,

1.1 eq.) was dissolved in THF and cooled to –78 °C. A solution of δ-valerolactone

(0.96 mL, 10.63 mmol, 1 eq.) in THF was added dropwise over 1 h and then stirred

for another 30 min. Propargyl bromide (1.14 mL, 12.75 mmol, 1.2 eq.) and HMPA

(2.22 mL, 12.75 mmol, 1.2 eq.) were added dropwise over 10 min. The brown

reaction mixture was warmed to approximately –42 °C and the temperature was

maintained while stirring for 3 h. After this period, 2.5 mL of a saturated solution of

ammonium chloride was added and the reaction was allowed to warm to room

temperature. Volatiles were removed by rotary evaporation and the residual yellow oil

was dissolved in diethyl ether (40 mL), which was washed three times with brine (40

mL). The organic phase was dried over MgSO4 and concentrated by rotary

evaporation. After column chromatography (30% ethyl acetate in hexane, Rf 0.35) and

distillation under reduced pressure (160 °C, 10 mbar) a colorless viscous liquid was

obtained (0.675 g, 4.9 mmol, 46% yield). The density of 1 was measured by weighing

pipetted volumes (n = 10) and determined to be 1.10 g/mL. 1H NMR (400 MHz,

CDCl3, δ) 4.37 – 4.27 (m, 2H, CH2O), 2.74 – 2.61 (m, 2H, COCHCH2C≡C), 2.53 –

2.46 (m, 1H, COCHCH2), 2.33 – 2.24 (m, 1H, CHCH2CH2), 2.01 (t, J = 2.6 Hz, 1H,

Page 52: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

49

C≡CH), 1.98 – 1.87 (m, 2H, OCH2CH2CH2), 1.77 – 1.67 (m, 1H, COCHCH2CH2)

(for an annotated spectrum see Supplementary Figure S 1).

Synthesis of -azido--caprolactone (2)

The first step of the synthesis of 2 was adapted from Lenoir et al.148 2-

chlorocyclohexanone (1.98 g, 15 mmol, 1 eq.) was dissolved in 20 mL

dichloromethane to which mCPBA (4.1 g, 16.5 mmol, 1.1 eq.) was added. The

reaction was stirred for 96 h at room temperature then stopped by cooling to –20 °C

for 1 h. The precipitated mCPBA was removed by filtration to yield a pale viscous

liquid, which was purified by column chromatography (35% ethyl acetate in hexane,

Rf 0.32) to yield -chloro--caprolactone (1.033 g, 7 mmol, 45% yield). 1H NMR

(400 MHz, CDCl3, δ) 4.80 (dd, J = 7.9, 2.7 Hz, 1H, COCHCl ), 4.63 (ddd, J = 12.8,

7.8, 1.8 Hz, 1H CH2CH2O), 4.23 (ddd, J = 12.7, 7.4, 1.8 Hz, 1H, CH2CH2O), 2.23 –

1.77 (m, 6H, ClCHCH2CH2CH2) (1H NMR spectrum in Supplementary Figure S 2).

In a second step, -chloro--caprolactone (500 mg, 3.4 mmol, 1 eq.) was added to a

solution of sodium azide (1.1 g, 16.9 mmol, 5 eq.) in DMSO (30 mL) and stirred at 50

°C for 48 h. The turbid yellow mixture was diluted with water (150 mL) and extracted

three times against 50% ethyl acetate in hexane (40 mL). The organic phases were

combined, dried over sodium sulfate, and concentrated by rotary evaporation under

reduced pressure to yield 2 as a viscous pale liquid (0.299 g, 1.9 mmol, 60% yield). 1H NMR (400 MHz, CDCl3, δ) 4.45 – 4.40 (m, 1H, COCHN3), 4.18 – 4.12 (m, 2H,

CH2CH2O), 2.09 – 1.67 (m, 6H, N3CHCH2CH2CH2) (Supplementary Figure S 3). The

density of 2 (1.154 g/mL) was measured by weighing pipetted volumes (n = 6).

General polymerization procedure and synthesis of mPEG-b-(CL-co-1) (3)

The polymerization procedure was adapted from Kim et al.149 In a typical

experiment, mPEG (800 mg, 0.4 mmol, 1 eq.) was dried by azeotropic distillation

with dry toluene (50 mL) under a flow of nitrogen. The flask was sealed and the

mPEG was dissolved in dry dichloromethane (2 mL). Dry -caprolactone (132.8 μL,

1.2 mmol, 3 eq.) and 1 (150.5 μL, 1.2 mmol, 3 eq.) were then added using gastight

syringes. The polymerization was initiated by the addition of HCl in ether (1.2 mL,

1.2 mmol, 3 eq.) and stirred at room temperature for 24 h. The reaction mixture was

then precipitated twice in cold ether, the solvent evaporated in vacuo, the residue

Page 53: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

50

taken up in water, and the polymer recovered by lyophilization (0.867 g, 80% yield).

The fully annotated NMR spectrum of 3 can be found in Supplementary Figure S 4.

This polymer contained on average two units of 1 per polymer chain. SEC (DMF): Mn

= 2,200 g/mol, Mw/Mn = 1.11.

Synthesis of mPEG-b-(CL-co-1) (4)

According to the general procedure described above, mPEG (1.50 g, 0.75 mmol, 1

eq.) was dried by azeotropic distillation with toluene and dissolved in dry

dichloromethane. Then, dry -caprolactone (166 µL, 1.5 mmol, 2 eq.) and 1 (376.4

µL, 3 mmol, 4 eq.) were introduced and the polymerization was started by the

addition of HCl in ether (1.5 mL, 1.5 mmol, 2 eq.). After 24 h, the polymer was

precipitated with diethyl ether, affording 4 as a white solid. The fully annotated NMR

spectrum can be found in Supplementary Figure S 5. This polymer contains on

average four units of 1 per polymer chain. SEC (DMF): Mn = 2,700 g/mol, Mw/Mn =

1.11.

Synthesis mPEG-b-(CL-co-2) (5)

According to the general procedure described above, mPEG (500 mg, 0.25 mmol, 1

eq.) was dried and dissolved in dichloromethane. Dry -caprolactone (83 µL, 0.75

mmol, 3 eq.) and 2 (134.4 µL, 1 mmol, 4 eq.) were introduced and the polymerization

started by the addition of HCl in ether (0.75 mL, 0.75 mmol, 3 eq.). After 24 h, the

polymer was precipitated twice in cold n-hexane to yield 5 as a white solid. The fully

annotated NMR spectrum can be found in Supplementary Figure S 6. This polymer

contains on average four units of 2 per polymer chain. SEC (DMF): Mn = 2,000

g/mol, Mw/Mn = 1.18.

NC preparation

PTX NCs were produced by wet milling. In a typical experiment, 2 mL of a 5%

(w/w) polymer solution in ultrapure water was filtered (0.2 μm pore size) into a 20

mL cylindrical glass vessel containing 10 mg of PTX and 4 mL of zirconium oxide

beads (~ 14.7 g, 0.3 mm in diameter, Union Process, Akron, OH). The vessel was

sealed with a plastic cap and then placed horizontally on a Ratek BTR5 blood tube

Page 54: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

51

roller (with custom modified motor, Labortechnik Fröbel GmbH, Lindau, Germany)

and rolled at 220 rpm for 48 h in a cold room (6 °C). After milling, the beads were

separated from the suspension by filtration through polyamide sieve fabric (30 μm

pores, VWR, Dietikon, Switzerland), and the residue washed four times with 2 mL of

ultra pure water. The suspension was centrifuged at 12000 × g for 6 min to remove

larger aggregates. The supernatant was then subjected to SEC with Sepharose CL-4B

to remove excess polymer, typically yielding a suspension containing 0.55 mg/mL or

0.74 mg/mL PTX for polymers 3 or 4, respectively.

Cross-linking in solution

In a proof of principle experiment, a 1.8 mM solution of 3 was reacted with 7 at two

different ratios (1/1 and 1/10 azide/alkyne) in a solution containing 1.8 mM copper

sulfate and 9 mM sodium ascorbate for 20 min. The reaction mixture was then frozen

in liquid nitrogen, lyophilized and dissolved in CDCl3 for 1H NMR analysis and

subsequent FTIR analysis.

Cross-linking on NCs

For cross-linking in the presence of NCs, the procedure above was slightly

modified. As a general example, 7 (0.159 mg, 0.65 μmol, 2.5/1 molar equivalent

azide/alkyne) in ethanol was transferred to a round bottom flask and the solvent

removed under a stream of nitrogen. Then, 3 mL of NC suspension (~ 0.55 mg/mL

PTX, ~ 0.262 μmol alkyne) was added followed by 10 μL of a solution containing

100 mM sodium ascorbate and 20 mM copper sulfate, final concentrations 333 µM

and 67 µM, respectively. Owing to the different solubility characteristics of 5 and 6,

they were dissolved in water and NMP, respectively, and added after the catalyst. The

flask was placed on a rotary shaker (~ 100 rpm) at room temperature for 24 h. After

this period, the cross-linked NCs were separated from catalyst and excess cross-linker

by SEC using Sephadex G10. The fraction with the highest concentration (~ 0.5

mg/mL PTX) was used for measuring particle size with time by DLS at both 6 ºC and

20 ºC. For 1H NMR and FTIR analyses, the fractions were pooled, frozen in liquid

nitrogen and lyophilized to concentrate the NCs. Then, deuterated methanol was

added to the lyophilized powder, which was then sonicated at 45 ºC to form a

homogenous suspension that was then analyzed. A summary of all cross-linking

conditions examined herein is given in Table 2.

Page 55: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

52

Imaging of NCs and the polymer coating after PTX removal

For transmission electron microscopy (TEM), samples (4 µL) of the NC

suspension, prepared at a concentration of ~ 0.5 mg/mL PTX, were adsorbed to glow

discharged carbon-coated copper grids for 1 min. After two washings with water to

remove excess NC, they were negatively stained with 2% (w/v) uranyl acetate for 1

min and air-dried after blotting with filter paper. The specimens were examined in a

Philips CM12 (tungsten cathode) transmission electron microscope (FEI, Hillsboro,

OR) at 100 kV, and images were recorded with a Gatan CCD 794 camera (Gatan Inc.,

Pleasanton, CA).

To dissolve the drug within nanocages, the NCs (3 mL in water, ~ 0.5 mg/mL) were

added to a 5 mL Spectra/Por Float-A-Lyzer G2 (MWCO 100 kDa, Sigma-Aldrich,

Buchs, Switzerland), and, after addition of 870 µL ethanol, were dialyzed against 2.5

L 25% ethanol/water mixture. The drug concentration inside the dialysis bag was

measured before medium change every day by HPLC. After complete removal of

drug, the contents inside the dialysis bag were concentrated with Spectra/Gel

Absorbent (VWR, International AG, Dietikon, Switzerland) to about 0.5 mL (~ 0.4

mg/mL of polymer) and analyzed by TEM. The presence of mPEG was tested by

adding CoSCN to the concentrate.150 Samples were prepared according to the protocol

for NC suspension, but without the extra washing steps.

Particle volume and polymer coverage calculations

From TEM images, the average dimensions of the NCs were measured (n = 138).

Using equation 3, the number of polymer chains (nPolymer) per NC could be calculated

nPolymer NA V (wtPolymer /wtPTX ) / Mw Polymer (3)

where NA is Avogadro’s constant, V is the average volume of a NC (assumed to be

a cylinder), is the density (1.4035 g/mL for PTX dihydrate), (wtPolymer/wtPTX) is the

weight ratio of drug to polymer after purification determined by 1H NMR

spectroscopy, and Mw Polymer is the weight average molecular weight of the polymer.

Dissolution test

Release of PTX from NCs was tested under sink conditions in 5% BSA solution,

which is expected to be the main solubilizing component for PTX in vivo.151 In a

Page 56: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

53

preliminary experiment, the saturation solubility of PTX powder in the test conditions

was found to be 0.01 mg/mL. A 1 mL Spectra/Por Float-A-Lyzer G2 (MWCO 100

kDa, Sigma-Aldrich, Buchs, Switzerland) was filled by mixing 20 mM phosphate

buffer (pH 7.4) containing 10% BSA with NCs in water to yield a final volume of 1

mL containing 0.1 mg/mL PTX in 10 mM phosphate buffer (pH 7.4) and 5% BSA.

The dialysis device was placed in a 50 mL centrifugation tube containing 45 mL of 10

mM phosphate buffer (pH 7.4) with 5% BSA, on a rotary shaker ( 400 rpm) in an

incubator at 37 °C. Under these conditions, the maximum PTX concentration was 5

times below its saturation solubility. At selected time points, 30 μL aliquots were

taken from inside the dialysis device and prepared for HPLC analysis by addition of

60 L internal standard and 100 µL methanol. The samples were stored at –20 ºC

overnight to precipitate albumin. The precipitate was removed by centrifugation and

the supernatant was filtered through polyamide (0.2 µm pore size) and analyzed.

Polymer shedding test

Dispersions of NCs with either non-cross-linked or cross-linked coatings (4 cross-

linked with 7) were diluted to a PTX concentration of 0.4 mg/mL in water. The

dispersions were then repeatedly centrifuged at 20,000 × g for 45 min at 14 °C until

the supernatant contained a stable concentration of PTX (measured by UV-Vis

spectroscopy at 230 nm). This procedure lead to removal of ~95% of the initially

present PTX. The supernatant was lyophilized and residual mPEG content was

analyzed by 1H NMR spectroscopy in D2O using DMSO as internal standard for

quantitative analysis.

Statistical analysis

Differences in groups were calculated by one-way ANOVA (normal distribution

was assumed) followed by a Tukey post hoc test. A value of p < 0.05 was considered

significant.

Equipment 1H NMR spectra were recorded on a Bruker Av400 spectrometer (Bruker BioSpin,

Fällanden, Switzerland) operating at 400 MHz for protons. Analytical size-exclusion

chromatography (SEC) measurements were performed in 0.01 M LiBr in DMF using

Page 57: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

54

a Viscotek TDAmax system (Viscotek, Houston, TX) equipped with a differential

refractive index detector. Molecular weights are given relative to narrow PEG

standards (PSS Polymer Standards Service GmbH, Mainz, Germany). Separation was

achieved using two Viscotek columns (CLM 3047) in series at a flow rate of 1.0

mL/min at 45 °C. Particle hydrodynamic diameter was determined by dynamic light

scattering (DLS) using a DelsaNano C Particle Analyzer (Beckman Coulter, Brea,

CA). The cumulants result calculated by the software was used to report the

hydrodynamic diameter of the NCs. PTX concentration was determined by HPLC

analysis, using an autosampler and pump system (CTC, Thermo Fisher) equipped

with a reversed phase column (Hypersil Gold column, 1 × 100 mm, Thermo Fisher)

heated to 30 °C (HotDog column oven) and an Accela PDA detector (Thermo Fisher

Scientific, Reinach, Switzerland). Gradient elution was performed starting with a mix

of 55% methanol in water (both containing 0.1% formic acid) rising to 85% methanol

within 15 min. PTX was detected at 227 nm. A solution of docetaxel in methanol was

added as internal standard. FTIR spectra were obtained using ATR geometry on a

Spectrum 65 infrared spectrophotometer (Perkin Elmer, Schwerzenbach,

Switzerland). After centrifugation of NC dispersions (180 min, at 19,000 × g), the

copper concentration in the supernatant was measured by ICP-OES relative to

aqueous CuSO4 standards (ULTIMA 2 ICP-OES, HORIBA Jobin Yvon Gmbh,

Unterhaching, Germany).

Results and Discussion

Polymer design and synthesis

A number of differing polymeric agents have been examined as steric stabilizers for

drug NCs. The currently used systems are maintained on the surface of NCs through

physisorption,25, 114 an equilibrium process which can lead to the reversible desorption

of the stabilizer upon dilution or heating.114 To examine the hypothesis that a non-

sheddable stabilizing nanocage can be made, a cross-linkable amphiphilic polymer

was prepared. The structure of the stabilizers and cross-linking agents examined in

this study are illustrated in Scheme 1. The stabilizing copolymer was designed to

contain mPEG as hydrophilic polymer segment to act as a steric stabilizing agent for

the NCs and thereby prevent NC agglomeration by masking their high-energy

surfaces. In addition, this polymer should decrease opsonization and convey “stealth-

like” properties to the NCs.37, 128, 152 Also, a variety of heterotelechelic variants of this

Page 58: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

55

polymer are commercially available, which opens the opportunity for future

functionalization of the hydrophilic corona of stabilized NCs. Poly(epsilon

caprolactone) was selected in favor of other polyesters because of its hydrophobicity

and slow degradability. 153 It was rationalized that these properties would minimize

polymer degradation during the milling process and favor the hydrophobic

interactions with the drug NC. In addition, there are medical devices fabricated with

poly(epsilon caprolactone) which have FDA approval (suture materials like Coated

Monoderm or Monocryl (poliglecaprone 25)). The hydrophobic block was prepared

by statistical cationic ring-opening copolymerization (ROP) of ε-caprolactone with

functional lactone monomers.149 A short hydrophobic segment was selected (ca. 5 – 7

units) so that the overall strength of the interaction between the polymer and the drug

was moderate to low. As a consequence, findings obtained using this polymer may be

relatively independent of specifically strong (or weak) polymer–drug interactions.

Several recent reports have investigated other ROP approaches for preparing

functional polyesters.115, 154 Cationic ROP is advantageous for producing polymers for

pharmaceutical applications owing to the absence of commonly used tin containing

catalysts.115 ε-Caprolactone was copolymerized with variable ratios of 1 to produce 3

and 4. These polymers had monomodal and narrow molecular weight distributions

(Figure 10) and contained on average two and four alkynyl groups per polymer chain

for 3 and 4, respectively, as determined by 1H NMR spectroscopy (Supplementary

Figures S 4 and S 5). An azido analog of 3 and 4 was prepared in the same fashion as

above by replacing 1 with 2 in the polymerization reaction. The resulting polymer, 5,

also had a narrow and monomodal molecular weight distribution and contained on

average four azido units per polymer (Figure 10 and Supplementary Figure S 6).

Precipitation with ether or hexane was sufficient for removing unreacted monomer

and hydrophobic oligomers not connected to mPEG (i.e., initiated by trace water in

the reaction vessel) as evidenced by analytical SEC (Figure 10).

Page 59: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

56

Scheme 1. (a) Synthesis of polymers 3, 4, and 5 from functional monomers 1 or 2

and-caprolactone (CL) from an mPEG macroinitiator; (b) structure of small

molecule cross-linking agents 2,6-bis(4-azidobenzylidene)-4-methylcyclohexanone

(6) and 1,11-diazido-3,6,9-trioxaundecane (7).

Figure 10. SEC traces of 3 (Mn = 2191 g/mol, Mw/Mn = 1.112), 4 (Mn = 2723 g/mol,

Mw/Mn = 1.112), and 5 (Mn = 2020 g/mol, Mw/Mn = 1.182) in DMF.

Preparation of cross-linkable NCs

PTX was milled in the presence of 3 and the size reduction monitored with time

(Figure 11). A stable and reproducible particle size of about 200 nm was achievable

by milling for 48 h, which was the smallest size achievable with the current

experimental setup. This time was substantially shorter than the seven days reported

for poloxamer 407,155 with similar milling equipment. The size and shape of particles

obtained was consistent with values reported for paclitaxel and other drug NCs

Page 60: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

57

produced by wet milling.85, 155, 156 The characteristic needle-like structure of paclitaxel

NCs was always observed using this process (Figures 12A and 12B). Owing to their

similarity, comparable particle sizes were obtained using 3 and 4. Briefly centrifuging

the crude suspension of NCs to remove larger aggregates had only a small effect on

mean particle size and polydispersity (Figure 13) confirming a narrow distribution of

particle sizes. The suspensions were purified from excess polymer by SEC, yielding a

suspension with a typical concentration of 0.55 or 0.74 mg/mL PTX, when using 3 or

4, respectively. The recovered yields depended on the initial particle size after milling

and the centrifugation step, which removed less drug in case of the smaller particles.

Owing to the biodegradable nature of the hydrophobic block, the stability of the

polymer during the milling process was assessed by analytical SEC. In a drug-free

run, a slight decrease of polymer molecular weight was observed after 96 h of milling,

which is twice as long as the time used for NC preparation (Figure 14).

Figure 11. Optimization of wet milling of PTX with 3. A milling duration between

42−54 h was found to be ideal for size-reduction and narrow particle size distribution.

Page 61: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

58

Figure 12. TEM images of NCs stabilized with 3 before (A) and after cross-linking

(B) with 7 (2.5/1 azide/alkyne). Following dissolution of the NCs from A and B in

25% ethanol, an aggregate structure was observed for the non-cross-linked NCs (C)

while intact polymeric coatings were observed for the correspondingly cross-linked

NCs, in the form of discrete spheroids (D).

Figure 13. Purification of NCs from large aggregates by centrifugation after milling

with 3. A reduction of both average NC size and polydispersity was observed

following centrifugation for 6 min at 12,000 g (n = 3).

Page 62: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

59

Figure 14. SEC traces of 3 before and after milling for 96 h at 6 °C under the same

conditions as for NC preparation (without PTX). Polymer length was slightly reduced

due to hydrolysis.

To quantify the amount of polymer per drug NC, 1H NMR spectra of the NCs

stabilized with 3 and 4 were recorded in CDCl3, a good solvent for both polymer and

drug (Figure 15). By comparing the signals from the aromatic protons of PTX (8.12

ppm) and the signal of PEG (3.64 ppm), a molar polymer/drug ratio of 0.134:1

(approximately 1:2 w/w) was obtained. Thus the drug content of the PTX NCs was

approximately 67 wt %, a value which is comparable or higher to that reported for

other drug/stabilizer systems.114, 155 Drug content was comparable when either 3 or 4

were used as stabilizers. TEM analysis of the stabilized NCs showed the characteristic

needle-like shape of PTX and lack of aggregation (Figures 12A and 12B). From these

and additional images, the average length and diameter of the NCs were 206 × 26 nm

(n = 138), respectively. Considering the weight ratio of polymer to drug, and

assuming the drug NCs to be cylinders, one can estimate that approximately 1.35 ×

104 polymer chains are adsorbed per NC. This equates to 1.34 nm2/polymer chain or

an ethylene glycol monomer density of 34/nm2, which both point to the polymer

adopting a brush-like conformation.157, 158 This type of conformation is advantageous

for preventing clearance by the mononuclear phagocyte system.

Page 63: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

60

Figure 15. Polymer/drug ratios were calculated by 1H NMR spectroscopy in CDCl3

using the characteristic peaks for PTX (8.12 ppm) and mPEG (3.64 ppm). NCs

prepared with 3 (A) and 4 (B) gave similar polymer/drug ratios.

Cross-linking and purification of cross-linked NCs

a) General considerations

In a proof of principle experiment, 3 was cross-linked with 7 in solution and in the

absence of NCs (Table 2). The reaction was monitored by 1H NMR and FTIR

spectroscopy to verify the consumption of alkynyl and azido peaks and the

appearance of the triazole product (Figures 16 and 17). At a stoichiometric azide to

alkyne ratio, both reacting groups were completely consumed within 20 min,

indicating that the alkynyl groups on 3 were readily available for reaction in the

absence of NCs.

In general, for cross-linking the NCs, a lower concentration of copper sulfate and

sodium ascorbate was used compared to the reaction in solution above because

insoluble copper(I) salts caused rapid destabilization and aggregation of the NCs.

Cross-linking performed at 6 °C also resulted in the formation of large aggregates

indicating that the reaction should be carried out at room temperature. An optimal

cross-linking time of 24 h was found for the NCs (no further evolution of their 1H

NMR spectra after this reaction duration). After the reaction, the salts were removed

Page 64: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

61

from the cross-linked NCs by SEC. For this purpose, Sephadex G10 had to be

employed because the interactions between cross-linked NCs and Sepharose CL-4B

lead to column blockage or very low recovery of the NCs. Inductively coupled plasma

optical emission spectroscopy (ICP-OES) analysis revealed less than 1 ppm residual

copper after purification. This result could be improved by increasing the column

length or by performing a second purification by SEC.

Table 2. Conditions of cross-linking experiments

Polymer (mM)

Cross-linker Azide/alkyne molar ratio

CuSO4 (mM)

Ascorbate (mM)

Time (h)

In solution

3 (1.8) 7 1/1 1.8 9 0.3

3 (1.8) 7 1/10

On NCs

3 (0.087) 7 2.5/1

0.067 0.333 24

3 (0.087) 7 10/1

3 (0.087) 7 15/1

4 (0.114) 5 2/1

4 (0.114) 6 2/1

4 (0.114) 7 15/1

Page 65: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

62

Figure 16. 1H NMR spectra in CDCl3 of 3 cross-linked with 7 at azide/alkyne ratios

of 1:10 (A) and 1:1 (B). The signal for the acetylene proton at 2.01 ppm disappeared

while at 8.12 ppm a signal of the resulting triazole appeared.

Figure 17. FTIR spectra of 3 cross-linked with 7. The starting compounds exhibit the

characteristic bands for the alkynyl group (≡C–H stretch at 3270 cm-1) and azido

group (N3 at 2100 cm-1), which are consumed after the reaction. The broad new peak

between 3700−3200 cm-1 is due to residual sodium ascorbate, which was not removed

from the reaction mixture.

Page 66: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

63

b) Analysis of the extent of cross-linking

Following purification, analysis of the extent of cross-linking was complicated by

reduced possibilities for dissolving the nanocage. For instance, after reaction with

excess 7 (10/1 azide/alkyne), purification, and lyophilization, the NCs were no longer

fully soluble in chloroform, indicating successful interpolymer chain cross-linking.

Sonication of the cross-linked polymer coating while heating in deuterated methanol

lead to a homogeneous suspension which could be analyzed by 1H NMR spectroscopy

(Figure 18). From this spectrum, a decrease of the intensity of the alkynyl protons and

the appearance of a peak for the triazole groups could be observed, though both were

slightly shifted in comparison to those observed for the polymers in solution, which

were analyzed in deuterated chloroform. Quantitative disappearance of the alkynyl

group (2.59 ppm) was observed for 3 when cross-linked with 10 eq. or more of 7.

FTIR spectroscopy of the NCs before and after cross-linking also qualitatively

revealed the decrease of the band associated with the alkynyl group (3270 cm–1)

indicating reaction (Figure 19A). The absence of the band for the azido group (2100

cm–1) indicated both successful removal of excess diazido reagent during the

purification step and argues in favour of bivalent reaction of 7 with the polymer (i.e.,

reaction of both rather than a single azide of 7 with the alkynyl groups). In contrast,

NCs prepared with 4 had lower conversion of the alkynyl group (85%, 65%, and 75%

for 5, 6, and 7, respectively) and showed residual azide by FTIR spectroscopy (Figure

19B). As evidenced by TEM (Figure 12B), inter-NC cross-linking was not observed.

Figure 18. 1H NMR spectra of NCs stabilized with 3 before (A) and after (B) cross-

linking with 7 (azide/alkyne ratio 10/1). Signals belonging to the acetylene proton at

2.59 ppm were reduced and a peak for the resulting triazole at 7.78 ppm appeared. A

break on the x-axis between 7.6 ppm and 2.7 ppm was added for clarity.

Page 67: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

64

Figure 19. FTIR spectra of NCs stabilized with 3 (A) or 4 (B) before cross-linking, in

a physical mix with 7, and following cross-linking. After cross-linking, signals from

the alkyne decreased or disappeared, while the azide band completely disappeared,

providing a strong indication of cross-linking. Only for the NCs cross-linked with 5

was residual azide observed, possibly due to lower reactivity of the polymeric tetra-

azide.

To verify that the polymer coating was indeed cross-linked, NCs were dialyzed

against an ethanol/water mixture to remove the drug from the polymer network. The

dialysis membrane was selected so that the polymer and dissolved drug molecules

could freely pass through the pores, while the cross-linked polymer, because of its

higher molecular weight, remained trapped inside. Following reaction with CoSCN,

residual mPEG was detected in the dialysis bag containing the cross-linked NCs,

while none was detected for the non-cross-linked analogs (Figure 20). TEM imaging

revealed the presence of intact polymeric spheroids for the cross-linked samples

(Figure 12D), proof for intermolecular cross-linking, but showed dense aggregates,

probably residual polyester, for the non-cross-linked analogs (Figure 12C). These

experiments demonstrated that the nanocage remained structurally intact even

following complete removal of the drug. Although different, this system is

reminiscent of hollow cage-like structures, as prepared by Turner and Wooley, which

were obtained after core degradation of shell cross-linked particles.159

Page 68: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

65

Figure 20. Addition of cobalt thiocyanate to the concentrate of the polymer after

dialysis of NCs revealed the presence of mPEG in the case of the cross-linked coating

(D) compared to the non-cross-linked coating (C). (A) and (B) are negative and

positive controls for the absence and presence of mPEG, respectively.

Sheddability of the polymer nanocages

Owing to its adequate water solubility, the influence of cross-linking excess on

particle-size stability was examined using 7 and NCs stabilized with 3. Azide/alkyne

ratios of 2.5/1, 10/1, and 15/1 were examined, as listed in Table 2. NCs stored at 6 °C

showed very little or no change of size over a period of three weeks (Figure 21),

which was comparable to findings from the literature.85 At room temperature, an

increase of particle size with time was observed for all samples. As seen in Figure 22,

cross-linking led to enhanced size-stability in comparison to NCs with non-cross-

linked stabilizers. Owing to the relatively short hydrophobic anchoring moieties on 3

and 4, strong interaction between these and the PTX NCs is unlikely. Consequently,

while the cross-linked coating is likely to prevent NC growth by aggregation, it may

not efficiently prevent crystal growth by Ostwald ripening.160, 161 This is consistent

with the lack of crystal growth at 6 ºC (Figure 21), where the solubility of PTX is

lower than at 20 ºC, thereby slowing ripening. Future experiments involving more

polymer layers, longer hydrophobic segments and/or hydrophobic segments capable

of tightly packing on the surface of the NC may provide insight as to whether the

nanocages can fully prevent Ostwald ripening. For now, storage at 6 ºC was found to

be sufficient for preventing this process.

Page 69: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

66

Figure 21. Results from size measurements over time of non-cross-linked and cross-

linked NCs coated with 3 (A) (n = 2−6) or 4 (B) (n = 6−13), which were stored at 6

°C.

Figure 22. Evolution of NC size over time. (A) Non-cross-linked and cross-linked

NCs stabilized with 3 and cross-linked with 7. On day six the size of non-cross-linked

NCs was significantly different from those cross-linked with 10 and 15 eq. of 7 (p <

0.05; n = 6). (B) Non-cross-linked and cross-linked NCs stabilized with 4 and cross-

linked with 5−7. On day 3, 6, and 20 the size of non-cross-linked NC was

significantly different from those which were cross-linked with 5 and 7, on day 13

only non-cross-linked and cross-linked with 7 were different (p < 0.05; n = 6−13).

Values represent means – SD.

It was observed that the size stability of the cross-linked NCs improved with

increased feed azide/alkyne ratios up to 10/1, at which point differences were no

longer observed. This indicated that maximal conversion of the alkynyl group was

achieved at this ratio. Overall, these results indicated that cross-linking is an

Page 70: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

67

important parameter in slowing crystal growth, and argued in favor of nanocage

desorption being reduced in comparison to the non-cross-linked control. Increasing

the number of alkynyl groups on the polymer (i.e., NCs stabilized with 4, bearing four

cross-linkable groups per chain) did not influence the results significantly in

comparison to those obtained with 3 (Figure 22B).

The relative sheddability of the polymer coatings before and after cross-linking was

analyzed by quantifying by 1H NMR the amount of dissolved mPEG present in the

supernatant of freshly centrifuged NC dispersions (Figure 23). In comparison to non-

cross-linked NCs, the cross-linked analogs showed between 3–4-fold less mPEG in

the supernatant, as determined for two different batches of NCs. This experiment

demonstrated that cross-linking the surfactant to form a nanocage favors retention of

the latter on the surface of the NC.

Figure 23. 1H NMR spectra in D2O of the supernatant of centrifuged NCs with non-

cross-linked (A) and cross-linked polymer coating (B).

NC–nanocage interactions

Two additional cross-linking agents were selected based on their different physico-

chemical characteristics, which could potentially influence the nature of the

interactions between the polymer nanocages and the NCs. 5 is a polymeric cross-

linking agent with a structure analogous to 3 and 4, but with four azido groups in its

hydrophobic segment. These could promote greater inter-chain cross-linking.

However, owing to its amphiphilic nature, it was only added in small amounts as

competition with 4 for the surface of the NC prior to cross-linking was anticipated. 5

displayed a similar stabilizing effect to 7, though with less cross-linker added. In

Page 71: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

68

addition, the FTIR spectrum of this NC revealed the presence of residual azido

groups, indicating incomplete conversion of this group on the NC. 6 is a hydrophobic

cross-linking agent that should preferentially partition to the surface of NCs, thereby

not only favoring cross-linking but also interaction between the hydrophobic

segments of the nanocage and the NC. However, only low concentrations could be

used, since the compound, owing to its poor water solubility, precipitated from its

organic solution upon mixing with water. When the nature of the cross-linking agent

was altered to more hydrophobic 6, no statistical difference in size was observed in

comparison to the non-cross-linked NCs (Figure 22B). These results indicate that

specific drug–nanocage interactions do not play a large role on size stability of the

NCs at least within the range of modifications examined (i.e., two hydrophobic

anchoring groups and three cross-linking agents). Due to its unknown toxicity and

small molecular weight, this compound was uniquely used for gaining a better

understanding of the nanocage system and was not intended for in vivo use.

Drug dissolution under sink conditions

NCs of PTX have previously been shown to possess sufficiently long circulation

times to benefit from passive accumulation in tumours.91, 162 The dissolution kinetics

of the NCs were evaluated herein under sink conditions to assess whether the lack of

desorption of the polymer nanocage was an influencial factor on the latter. The

dissolution characteristics of all NCs, cross-linked and non-cross-linked, were overall

statistically the same (Figure 24). Under the present experimental conditions, the

polymer nanocage was expected to remain intact over the entire time frame of the

experiment and maintain residual drug particle within its hydrophobic core due to a

combination of physical entrapment and physisorption. From these results it was

found that about 50% of the NCs dissolved in more than 3 h. This time frame would

be sufficient for high deposition of drug in the tumor tissue if a targeting ligand were

attached to the nanocage.127 In addition to the high surface coverage with mPEG, the

high aspect ratio of the NCs (Figures 12A and 12B) might increase circulation time,

as has been observed in other studies.163

Page 72: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

69

Figure 24. Drug dissolution from NCs. Values represent means SD (n = 4 – 5). It

was noteworthy that free drug (control) released very rapidly from the dialysis device,

indicating that diffusion of the drug was not limited by the molecular weight cutoff of

the membrane

Conclusion

This work demonstrated successful cross-linking of polymeric stabilizers around

PTX NCs to form polymeric nanocages. These retained the particulate drug through a

combination of physical entrapment and physisorption. The nanocages were found to

act as sterically stabilizing barriers to particle–particle interactions and aggregation.

These were also shown to be shed to a lesser extent than non-cross-linked coatings,

thereby providing a means for enhanced retention of targeting agents on NCs. These

findings provide crucial tools for preparing non-sheddable stabilizing coatings for

NCs and potentially other classes of nanoparticles under circumstances where

covalent attachment between the coating and the particle is not possible or desired. In

addition, the proposed strategy should be applicable to other polyesters to thereby

modulate the rate of degradation of the nanocage. In theory, it should be possible to

attach targeting ligands to the nanocage by use of heterotelechelic PEG derivatives

and thus access active targeting or internalization of NCs.

Page 73: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

70

Page 74: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

71

III.2. Modular Design of Redox-Responsive Stabilizers for

Nanocrystals3

Kathrin Fuhrmann, Anna Połomska, Carmen Aeberli, Bastien Castagner,

Marc A. Gauthier, and Jean-Christophe Leroux

Introduction

Many key features of nanoparticles, such as size stability, propensity to

aggregation, photonic properties, dissolution profiles, interactions with biomolecules,

and circulation lifetime intrinsically depend on the efficacy of stabilizing agents to

mask their surface.37, 157 Correspondingly, much work has been devoted to the

development of stabilizing agents for drug nanocrystals (NCs), quantum dots, metal

nanoparticles, etc.164-166 Interestingly, developing methods for spatio-temporally de-

stabilizing nanoparticles has received much less attention,167-170 despite the fact that

this property may have applications in imaging and drug delivery. Triggered de-

stabilization can, in principle, be achieved by developing functional stabilizers whose

properties change in response to a local endogenous stimulus. However, designing

such a system requires that the dynamic non-covalent interactions between the

stabilizer in both the original and “triggered” state be carefully balanced to the surface

chemistry of the nanoparticle, which can vary broadly. This poses several synthetic

and developmental challenges that have impeded the design of tailored responsive

stabilizers.

This study presents a simple strategy for systematically adjusting the affinity of

polymeric stabilizers for nanoparticles, and which simultaneously introduces chemical

groups that are sensitive to endogenous oxidants, such as reactive oxygen species

(ROS). ROS are strongly associated with chronic inflammation and cancer65 and,

owing to their short lifetime, their action is limited to these locations. ROS are

notably associated with tumor tissues where hypoxic stress causes ROS-mediated

signaling.171 This endogenous stimulus is increasingly being investigated as a means

of triggering the response of smart polymeric systems. Examples include

3 Published in ACS Nano 2013, 7, 8243-8250. A. Połomska contributed to compound analysis.

Page 75: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

72

nanoparticles made of polymers bearing (aryl) boronic esters,172, 173 of poly(propylene

sulfide),174-176 or of cross-linked oligo(proline).177 In the current study, paclitaxel

(PTX) NCs were selected as model for demonstrating how ROS-sensitive polymeric

stabilizers can be systematically and rationally tuned to achieve the best compromise

between stability prior to oxidation, and subsequent responsiveness upon exposure to

ROS. PTX NCs have already been examined for their therapeutic potential in treating

cancer and thus represent a pharmaceutically relevant system. Notably, the size of

PTX NCs is expected to strongly depend on their stabilizing coating, and triggered

de-stabilization may be of interest for promoting cellular uptake.170, 178

A library of ten redox-responsive amphiphilic block copolymer stabilizers was

prepared from two parent block copolymers (methoxy polyethylene glycol-b-[-

propargyl--valerolactone-co--caprolactone] (mPEG-b-[PVL-co-CL]); Figure

25A) by postpolymerization modification using the thiol–yne reaction.

Postpolymerization modification is a powerful tool for systematically altering the

functionality of polymers without influencing chain length or chain length

distribution.179 The radical thiol–yne reaction is well established as a mild, efficient,

and functional group tolerant reaction which conforms to the criteria of “click”

chemistry.180 This reaction was used to systematically alter the polarity of the

polyester block by grafting different hydrophobic thiols, while the two thioether

linkages produced at each PVL repeat unit render this block sensitive to oxidation by

ROS (Figure 25). Oxidation of the thioether to a sulfoxide or sulfone significantly

alters the polarity of the hydrophobic block,181 which may in turn cause desorption

from the surface of the NC. The influence of the nature of the hydrophobic thiol agent

grafted to the polymer on its stabilizing potential for PTX NCs is reported herein as

well as the response of stabilized PTX NCs towards oxidizing conditions.

Page 76: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

73

Figure 25.(A) Synthesis of a library of ten oxidation-sensitive polymeric stabilizers

by thiol–yne postpolymerization modification; (B) Oxidation of the stabilizer by

endogenous oxidants (e.g., ROS) triggers desorption from the surface of NCs, leading

to de-stabilization.

Experimental Section

Materials

PTX was obtained from Bioxel Pharma Inc. (Sainte-Foy, QC, Canada) and

docetaxel from ScinoPharm Taiwan, Ltd. (Tainan County, Taiwan). Lithium

diisopropylamide, -valerolactone (VL), propargyl bromide,

hexamethylphosphoramide, -caprolactone, ethane-1-thiol, butane-1-thiol, octane-1-

thiol, benzyl mercaptane, thiocholesterol, 2,2-dimethoxy-2-phenylacetophenone

(99%), pyrene (99%), iron sulfate heptahydrate (> 99%, FeSO4), albumin from bovine

serum (BSA; ≥ 96 %), and methoxy poly(ethylene glycol) (mPEG; 2 kDa) were

purchased from Sigma-Aldrich (Buchs, Switzerland) and used as received. Hydrogen

chloride (1 N solution in diethyl ether), was purchased from Chemie Brunschwig AG

(Basel, Switzerland) and hydrogen peroxide (30% solution, Perhydrol) from Merck

(Altdorf, Switzerland). mPEG-b-CL and mPEG-b-PSO were bought from Advanced

Polymer Materials (Montreal, QC, Canada). Ultra pure water was obtained from a

Barnstead Nanopure system (Thermo Fisher Scientific, Reinach, Switzerland). Dry

Page 77: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

74

solvents were taken from a solvent purification system (LC Technology Solutions

Inc., Seabrook, NH). VL and CL were distilled over calcium hydride under inert

atmosphere before use. All other solvents were of highest purity and bought from

Sigma-Aldrich (Buchs, Switzerland).

Equipment. 1H NMR spectra were recorded on a Bruker Av400 spectrometer (Bruker BioSpin,

Fällanden, Switzerland) operating at 400 MHz for protons. Analytical size-exclusion

chromatography (SEC) measurements were performed in THF using a Viscotek

TDAmax system (Viscotek, Houston, TX) equipped with a differential refractive

index detector. Molecular weights are given relative to narrow PEG standards (PSS

Polymer Standards Service GmbH, Mainz, Germany). Separation was achieved using

two Viscotek columns (GMHHRM) in series at a flow rate of 1.0 mL.min–1 at 45 °C.

Particle hydrodynamic diameter was determined by dynamic light scattering using a

DelsaNano C Particle Analyzer (Beckman Coulter, Brea, CA). The cumulants result

calculated by the software was used to report the hydrodynamic diameter of the NCs.

PTX concentration was determined by HPLC analysis, using an autosampler and

pump system (Ultimate 3000, Dionex, Thermo Fisher) equipped with a reversed

phase column (Accucore C18 column, 2.6 µm particle size, 100 × 2.1 mm, Thermo

Fisher) held in a column oven at 30 °C and a diode array detector (Thermo Fisher

Scientific, Reinach, Switzerland). Gradient elution was performed starting with a mix

of 40 % acetonitrile in water rising to 70% acetonitrile within 8 min. PTX was

detected at 230 nm. A solution of docetaxel in acetonitrile was added as internal

standard. FTIR spectra were obtained using ATR geometry on a Spectrum 65 infrared

spectrophotometer (Perkin Elmer, Schwerzenbach, Switzerland).

Synthesis of polymer precursor 3

PVL was synthesized as described elsewhere.39 In a typical experiment, mPEG

(800 mg, 0.4 mmol, 1 eq.) was dried by azeotropic distillation with dry toluene (50

mL) under a flow of nitrogen. The flask was sealed and the mPEG was dissolved in

dry dichloromethane (2 mL). Dry CL (132.8 μL, 1.2 mmol, 3 eq.) and PVL (150.5

μL, 1.2 mmol, 3 eq.) were then added using gastight syringes. The polymerization

was initiated by the addition of HCl in ether (1.2 mL, 1.2 mmol, 3 eq.) and stirred at

Page 78: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

75

room temperature for 24 h. The reaction mixture was then precipitated twice in cold

ether, the solvent evaporated in vacuo, the residue taken up in water, and the polymer

recovered by lyophilization (0.867 g, 80% yield). The synthesis of 13 followed the

protocol above with the following quantities: mPEG (1.50 g, 0.75 mmol, 1 eq.), PVL

(564.6 µL, 4.5 mmol, 6 eq.), HCl in ether (1.5 mL, 1.5 mmol, 2 eq.). The fully

annotated NMR spectrum of 3 and 13 and the compositions of the polymers can be

found in Supplementary Figures S 4 and S 7 and Table 3, respectively.

Postpolymerization modification by radical thiol–yne addition.

In a general procedure, 3 (30 mg, 0.0115 mmol, 1 eq.) was dissolved in 100 μL of

stabilizer-free tetrahydrofuran. A fresh stock solution of 2,2-dimethoxy-2-

phenylacetophenone (0.89 mg, 0.0035 mmol, 5 mol% of thiol-compound) in

stabilizer-free tetrahydrofuran THF was added (8.9 μL) to the polymer solution in a

quartz cuvette. After addition of the thiol compound (6 or 12 eq. for modification of 3

and 13, respectively), the solution was subjected to UV light at 365 nm for 30 min.

The solution was then precipitated twice with about 15 volume parts of cold hexane,

recovered by centrifugation (unless otherwise stated, see Supporting Information),

and dried under a flow of nitrogen. The polymer was dissolved/dispersed in water and

lyophilized. Fully annotated 1H NMR spectra of all polymers and can be found in

Supplementary Figures S 8–S 17.

Preparation of PTX NCs.

PTX NCs were produced by wet milling. In a typical experiment, 2 mL of a 0.5%

(w/w) polymer solution in ultra pure water was filtered (0.2 μm pore size) into a 20

mL cylindrical glass vessel containing 10 mg of PTX and 4 mL of zirconium oxide

beads (~ 14.7 g, 0.3 mm in diameter, Union Process, Akron, OH). When the stabilizer

was not soluble in water, an aqueous solution was first prepared by micellization

following dialysis from an ethanol/water or tetrahydrofuran/water solution. The vessel

was closed with a plastic cap and then placed horizontally on a Ratek BTR5 blood

tube roller (with custom modified motor, Labortechnik Fröbel GmbH, Lindau,

Germany) and rolled at 220 rpm in a cold room (6 °C) for 18 – 48 h. After milling,

the beads were separated from the suspension by filtration through polyamide sieve

fabric (30 μm pores, VWR, Dietikon, Switzerland), and the residue washed four times

Page 79: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

76

with 2 mL of ultra pure water. The suspension was centrifuged at 12,000 × g for 6

min to remove larger aggregates.

Imaging of stabilized PTX NCs.

NCs were imaged by TEM. 4 μL of a NC suspension was adsorbed to a glow

discharged carbon-coated copper grid for 1 min. After two washings with water, NCs

were negatively stained with 2% (w/v) uranyl acetate for 1 min and air-dried after

blotting with filter paper. The specimens were examined in a Philips CM12 (tungsten

cathode) transmission electron microscope (FEI, Hillsboro, OR) at 100 kV and

images were recorded with a Gatan CCD 794 camera (Gatan Inc., Pleasanton, CA).

Drug to stabilizer ratio.

NC dispersions were centrifuged at 20,000 × g for 90 min at 15 °C. Afterwards, the

supernatant was carefully and quantitatively removed and the pellet was dried by

lyophilization. The dried pellet was dissolved in deuterated chloroform for 1H NMR

spectroscopy. Integration of the aromatic proton signal of PTX at 8.12 ppm versus the

proton signal of PEG at 3.64 ppm provided an estimate of the drug to stabilizer ratio.

NC volume and density of the stabilizer coating.

Average dimensions of NCs were measured in TEM images. The number of

polymer chains (nPolymer) per NC can be calculated with equation (3) below.

nPolymer NA V (wtPolymer /wtPTX ) / Mw Polymer (3)

where NA is Avogadro’s constant, V is the average volume of a NC (assumed to be

a cylinder), is the density (1.4035 g/mL for PTX dihydrate), (wtPolymer/wtPTX) is the

weight ratio of drug to polymer after centrifugation determined by 1H NMR

spectroscopy, and Mw Polymer is the weight average molecular weight of the polymer as

calculated by 1H NMR spectroscopy.

Dissolution test

Release of PTX from NCs was tested under sink conditions in a 5% BSA solution,

which is expected to be the main solubilizing component for PTX in vivo.151 In a

preliminary experiment, the saturation solubility of PTX powder in the test conditions

was found to be 0.01 mg.mL–1. A 1 mL Spectra/Por Float-A-Lyzer G2 (MWCO 100

Page 80: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

77

kDa, Sigma-Aldrich, Buchs, Switzerland) was filled by mixing 20 mM phosphate

buffer (pH 7.4) containing 10% BSA with NCs in water to yield a final volume of 1

mL containing 0.1 mg/mL PTX in 10 mM phosphate buffer (pH 7.4) and 5% BSA.

The dialysis device was placed in a 50 mL centrifugation tube containing 45 mL of 10

mM phosphate buffer (pH 7.4) with 5% BSA, on a rotary shaker ( 400 rpm) in an

incubator at 37 °C. Under these conditions, the maximum PTX concentration was 5

times below its saturation solubility. At selected time points, 30 μL aliquots were

taken from inside the dialysis device and prepared for HPLC analysis by addition of

30 L internal standard (docetaxel in acetonitrile) and vortexed. After addition of 200

L 0.1 M ZnSO4 solution and 500 L acetonitrile the samples were vortexed for 1

min. Following two centrifugation steps of 10 min at 15,000 × g 200 L of the

supernatant was mixed with 100 L water and filtered through polyamide (0.2 µm

pore size) and analyzed by HPLC as described under “Equipment”.

Sensitivity to ROS, dissolution and polymer shedding

NC suspensions were diluted with water to a PTX content of 0.3 mg/mL. In the

case of NCs formulated with 16 this step was neglected. The NC dilution (0.5 mL)

was mixed with Fenton’s reagent (H2O2/FeSO4 final concentrations of either 1

mM/0.1 mM or 0.1 mM/0.01 mM) directly in a UV cuvette and size was monitored at

25 °C for 12 h. To determine polymer shedding after oxidation, diluted NC

suspensions (0.2 and 0.3 mg/mL PTX for polymers 16 and 14/15 respectively) were

incubated with 1 mM H2O2/0.1 mM FeSO4 for 2 h at room temperature, then they

were treated and analyzed as described under “Drug to stabilizer ratio”. In a further

experiment, Fenton’s reagent (H2O2/FeSO4 final concentration in the mixture 1

mM/0.1 mM) was added to a NC suspension at the beginning of a dissolution test, and

then the experiment was conducted as described in the previous section. For the NMR

characterization of the shed polymer, a NC suspension stabilized with polymer 14 was

incubated with 1 mM H2O2 and 0.1 mM FeSO4 for 4 h. The particles were then

centrifuged at 20,000 × g for 90 min. The supernatant containing shed polymer was

collected, lyophylized, and later analyzed by 1H NMR spectroscopy.

Page 81: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

78

Results and Discussion

Stabilizer design and synthesis

As illustrated in Figure 25A, a block copolymer structure was chosen as template

for the preparation of redox-responsive polymeric stabilizers. Two parent copolymers

bearing alkynyl groups in the hydrophobic block were prepared by cationic ring-

opening polymerization of mixtures of PVL and CL from a mPEG initiator (3 and

13 in Table 3). This polymerization approach is advantageous for producing polymers

for pharmaceutical applications owing to the absence of transition metal catalysts.149

mPEG was selected as hydrophilic polymer segment to prevent NC agglomeration by

masking their high energy surfaces and to convey “stealth-like” properties to the

NCs.38, 128, 152 The number of repeat units in the polyester block was targeted to be low

(ca., 4–5) so that affinity to the NC surface would not be too strong to prevent

desorption upon oxidation, and so that individual oxidation events within this short

block would stand a chance of significantly influencing the overall polarity of the

chain (i.e., for responsiveness). 3 and 13 possessed narrow molecular weight

distributions (Figure 26A), and analysis of their 1H NMR spectra (Figure 26B,

Supplementary Figures S 4 and S 7) allowed for the accurate determination of the

final composition of the short hydrophobic segment (Table 3).

Page 82: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

79

Table 3.Characteristics of stabilizers examined in this study

Composition R Mn, SEC

(kDa)a Mw/Mn

Thiol–ynecoupling

(%)b 3 PVL2 CL3 2.6 1.1 –

8 PVL2 CL3 Et 2.8 1.3 100

9 PVL2 CL3 Bu 2.8 1.3 100

10 PVL2 CL3 Oct 3.3 1.5 100

11 PVL2 CL3 Bn 3.6 1.8 90

12 PVL2 CL3 Chol 3.9 1.9 100

13 PVL4 2.8 1.1 –

14 PVL4 Et 2.8 1.4 100

15 PVL4 Bu 3.1 1.4 100

16 PVL4 Oct 3.8 1.8 100 c

17 PVL4 Bn 3.6 2.4 95

18 PVL4 Chol 2.9 1.3 100

19 CL5 2.6 1.1 –

20 PSO9 2.7 1.1 –

a: in THF; b: by 1H NMR spectroscopy; c: by MALDI-TOF MS Abbreviations: ethyl

(Et), n-butyl (Bu), n-octyl (Oct), benzyl (Bn), and cholesteryl (Chol).

Page 83: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

80

Figure 26. Polymer library synthesis by postpolymerization modification. (A) SEC

traces of parent polymers 3 and 13 in THF; (B) Representative 1H NMR spectra of 13

and 14 demonstrating quantitative functionalization by thiol–yne coupling.

Assignments based on the 1H–1H correlation spectrum of 14 (Supplementary

Figure S 18).

Postpolymerization modification of 3 and 13 by radical thiol–yne coupling using

various alkyl, aromatic, and multi-cyclic thiols led to the straightforward and efficient

preparation of the library of polymeric stabilizers 8-12, 14-18 (Table 3) with

systematically different number and nature of side chains (Figure 25A). Quantitative

grafting of all thiols except benzyl mercaptane was achieved in less than 30 min as

determined by 1H NMR spectroscopy (Figure 26B). For 11 and 17, small peaks at 5.4

Page 84: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

81

and 5.9 ppm corresponding to the mono-substituted vinyl sulfide were observed

(Supplementary Figures S 11 and S 16). For 16, which bore eight octyl units,

integration of the 1H NMR spectrum did not match expectations, though peaks

associated with mono-substituted vinyl sulfide were not observed (Supplementary

Figure S 15). Consequently, matrix-assisted laser desorption ionization time-of-flight

mass spectrometry was used to verify complete functionalization of the polymer. The

latter showed a multiple peak distribution with mass differences of about 430.29,

which corresponds to one -valerolactone monomer conjugated with two octanethiols.

In addition, FTIR spectroscopy revealed complete disappearance of the characteristic

stretching vibration of the alkynyl group at 3270 cm–1 following thiol–yne

functionalization (Figure 27). Analytical size-exclusion chromatography in THF

showed in most cases a mono-modal molecular weight distribution, though as the

hydrophobicity (and bulk) of the polyester block increased, the chromatograms

became less well defined, likely due to potential enthalpic interaction with the

stationary phase of the column (Figure 28).

Figure 27. FTIR spectra of parent polymers 3 (A) and 13 (B) and their respective

thiol–yne adducts showing the disappearance of the alkynyl peak at 3270 cm–1.

Figure 28. SEC traces of polymers 8-12 (A) and 14-18 (B) after thiol–yne coupling

and control polymers 19, 20 (C).

Page 85: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

82

NC production and stability

PTX NCs were produced by wet-milling in the presence of 8-12, 14-18 as well as

with mPEG-b-CL (19) or mPEG-b-poly(styrene oxide) (mPEG-b-PSO, 20) which

are un-branched aliphatic and aromatic controls, respectively. Overall, fourteen

polymers were investigated for their ability to sterically stabilize PTX NCs. With the

exception of 12, 17, and 18, wet-milling of PTX for 18 – 48 h led to the production of

NCs with mean diameters around 200 nm, as determined by dynamic light scattering,

which is in accordance with requirements for systemic administration. The size and

shape of particles obtained by wet-milling were consistent with values reported for

PTX NCs elsewhere.85, 156 NCs were subjected to a short centrifugation step to

remove larger aggregates, and then PTX concentration was measured by high-

performance liquid chromatography (Table 4). PTX recovery was lower with

increasing hydrophobicity of the polymer, which indicates inefficient milling in cases

where self aggregation is favored versus the interaction of the polymer with the NC

surface. While the polymers bearing the pendant cholesteryl and benzyl units were

poorly soluble and inefficient stabilizers for PTX NCs, the ability to quantitatively

introduce complex and bulky groups such as these by the thiol–yne reaction

demonstrates the versatility of the chemistry employed. Inefficient stabilization of

NCs observed using 12, 17, and 18 may be related the presence of branching within

the hydrophobic segment. Indeed polymer functionality can be broadly tuned using

the extensive list of commercially available thiol agents.

Page 86: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

83

Table 4. Characteristics of stabilized PTX NCs examined in this study

Stab. R PTX conc. (mg/mL) a

Size (nm) a

PDI b PTX

content (wt%)

L/2Rg c

8 Et 0.82 180 0.234 77 0.40

9 Bu 0.60 285 0.255 79 0.43

10 Oct 0.29 189 0.211 77 0.43

11 Bn 0.42 298 0.219 78 0.45

14 Et 0.77 232 0.206 76 0.42

15 Bu 0.54 193 0.218 73 0.37

16 Oct 0.19 176 0.133 69 0.32

19 – 0.91 136 0.137 82 0.41

20 – 0.64 229 0.190 74 0.41

a: after centrifugation; b: polydispersity index; c: L is the average mean distance

between mPEG chains; Rg is the radius of gyration of “free” mPEG in aqueous solution; L/2Rg >1 mPEG is well-separated and in mushroom conformation, <1 mPEG in brush conformation, <0.5–0.7 enhanced protein resistance regime.158

The characteristic needle-like shape of PTX NCs was observed by transmission

electron microscopy (TEM) for all stabilizers (examples in Figure 29). Under the

simplification that the NCs are cylindrical, the average specific surface area of the

NCs was determined from these images. This, in combination with drug content

determined by 1H NMR spectroscopy, allowed for the calculation of the amount of

polymer per surface area of the NC which can be expressed as L, the average mean

distance between mPEG chains. According to the geometric model of Pasche et al.,158

the ratio L/2Rg (Rg being the radius of gyration for free mPEG) was calculated to

provide an indication of the packing structure of the stabilizer on the NC (Table 4).

An L/2Rg higher than 1 suggests that mPEG is well-separated and in mushroom

conformation, while an L/2Rg below 1 indicates brush conformation. mPEG modified

surfaces with L/2Rg below 0.5–0.7 have been reported to efficiently reduce protein

adsorption.158 Based on this model, all stabilizers on the surface of PTX NCs were

within a dense brush regime (L/2Rg ranges from 0.32–0.45, see Table 4), suggesting

Page 87: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

84

intimate contact and interaction between neighboring polymer chains. While stabilizer

surface density for all polymers was roughly the same, a trend towards increased

stabilizer density was observed from 14 to 16 (increasing side-chain length).

Figure 29. Representative transmission electron micrographs of PTX NCs stabilized

with (A) 9, (B) 10, (C) 14, and (D) 15. Scale bars represent 200 nm.

Following purification, the stabilized NCs were stored at room temperature (Figure

30) and their size over time was monitored by dynamic light scattering. Compared to

the branched polymers, the un-branched aliphatic control 19 was the least efficient

stabilizer, pointing to the importance of multiple interactions and intimate contact

within the hydrophobic domain. Polymers 8-11 (i.e., with four branches) provided

improved size stability compared to 19 (Figure 30C). Increasing the number of

branches to eight (polymers 14-16, Figure 30B) led to significant stabilization of the

NCs over a period of over 21 days. At 6 °C, little or no evolution in size was observed

for any of the stabilized NCs (Figure 31), suggesting that the size increase observed at

room temperature was due to Ostwald ripening rather than aggregation. It should be

noted that the influence of mass transport, which is affected by initial NC size and

concentration, on NC stability cannot be discounted, the relative similarity of these

parameters between samples suggest that their effect is secondary to stabilizer

chemistry in Figure 30.

Page 88: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

85

Figure 30. Size-stability of PTX NCs stabilized with (A) 8-11, (B) 14-16, (C) 19, and

20 at room temperature. Values represent mean ± SD (n = 3–6).

Figure 31. Size stability of NCs milled with different polymers and stored at 6 °C.

Values represent mean ± SD, n = 3–6.

Response of PTX NCs to reactive oxygen species

Stabilizers 8-12 and 14-18 contain on average four and eight thioether bonds per

polymer chain, respectively. Oxidation of these groups produces a sulfoxide and

ultimately a sulfone.181 This process is expected to impart a large change of polarity to

the hydrophobic block of the stabilizer. For instance, while the thioether side-chain of

methionine in proteins is one of the most hydrophobic, its sulfoxide/sulfone form is

more hydrophilic and bulky, which is used in practice for protein secondary structure

characterization by breaking -sheet assembly.182, 183 As PTX NCs stabilized with 14-

16 demonstrated excellent size stability (Figure 30B), their response to ROS was

investigated. These NC suspensions are characterized by a high drug content (69–

82%, see Table 4), which implies that the ROS-responsiveness of only a minor

component (i.e., the stabilizer) has the potential to trigger the destabilization of a

correspondingly large amount of drug. To examine the sensitivity of 14 and 15, a low

concentration of ROS was first examined (100 M H2O2, 10 M FeSO4). As shown

in Figure 32A and B this concentration did not influence NC stability. However,

exposure to an increased oxidizing environment (1 mM H2O2, 0.1 mM FeSO4) led to

the rapid de-stabilization of NCs prepared with 14 and 15. Oxidation of the thioether

Page 89: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

86

was monitored by 1H NMR spectroscopy analysis of the fraction of 14 that was shed

from the NC. Examination of the 1H-1H correlation spectrum revealed the appearance

of a new signal consistent with an ethylsulfoxide group, at lower field than the

corresponding ethylthioether (Figure 33), as expected from prior observations from

the literature.184 Overlapping signals in the 1D 1H NMR spectrum does not allow for

precise quantification as only a low level of oxidation was observed. Semi-

quantitative analysis suggests that only a single, or very few oxidation events within

the very short hydrophobic block were able to drive 14 from the surface of the PTX

NCs. Thus a greater responsiveness to oxidation in comparison to other thioether-

based materials was observed. For the latter, multiple oxidation events are required to

significantly alter the polarity of the individual polymer chains. Responsiveness was

manifested by a rapid increase in size beginning well within the first hour (Figure 32),

which then leveled off thereafter. The size distribution of the NCs also increased to a

certain extent after exposure to ROS. TEM images of NCs before and after exposure

to ROS suggest that the increased size observed by light scattering could be the result

of aggregation, due to exposure of hydrophobic PTX areas (Figures 34 and 35).

Bearing in mind that this is a closed system, oxidation of 14 produced a shedding of

about 20% of the stabilizer from the NC (polymer to drug ratio (w/w) decreased from

0.311 ± 0.014 to 0.251 ± 0.006), suggesting that more shedding may be observed

within an open system such as the body. NCs stabilized with 16, bearing eight

pendant octyl chains in its hydrophobic block, showed a much slower and less

pronounced response to ROS, likely because the longer hydrophobic alkane chain

(octane) provided a stronger anchor to the surface of the NC than the shorter ones

(i.e., ethane or butane). These combined results suggest that the change of polarity of

the stabilizer’s hydrophobic block due to low level of oxidation event is sufficient to

induce desorption from the surface of the NC when the pendant groups are short and

when the concentration of oxidant is sufficiently high. Exposure to ROS did not

significantly influence the release kinetics of PTX from stabilized NCs, which points

to the absence of deleterious effects of the oxidation reaction on the NC itself (Figure

36).

Page 90: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

87

Figure 32. Size-stability of PTX NCs stabilized with 14 (A), 15 (B), 16 (C) after

exposure to oxidizing agent (oxidant concentration: 1: 0.1 mM H2O2/0.01 mM

FeSO4 or 10: 1 mM H2O2/0.1 mM FeSO4). Data represent mean ± SD, n = 3–4.

Figure 33. Oxidation of 14. 500 MHz 1H-1H COSY NMR in CDCl3 of the polymer

shed from nanoparticles following exposure to 1mM H2O2 and 0.1 mM FeSO4 for 4 h.

The particles were centrifuged at 20,000 x g for 90 min and the supernatant

containing shed polymer was collected and lyophylized. The spectrum exhibits a new

peak at 1.31 ppm that correlates with a signal at 2.74 ppm, which is consistent with an

ethylsulfoxide moiety.185

Page 91: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

88

Figure 34. Representative TEM overview and zoomed images of NCs stabilized with

14 before (A) and after (B) exposure to 1 mM H2O2/0.1 mM FeSO4 for 4 h,

demonstrating signs of aggregation. The specimens were examined in a FEI Morgagni

268 (tungsten cathode) transmission electron microscope (FEI Company,

Netherlands) at 100 kV and images were recorded with a Keen View camera (Soft

Imaging System, Germany).

Figure 35. Representative size distribution of NCs stabilized with 14 (A, B) and 15

(C) before and after exposure to 1 mM H2O2/0.1 mM FeSO4 for 4 h and 12 h,

demonstrating growth of the particles and increase in polydispersity index.

Page 92: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

89

Figure 36. Dissolution profiles of NCs stabilized with 14 (A) and 15 (B) before and

after exposure to an oxidizing environment (1 mM H2O2/0.1 mM FeSO4), a solution

of PTX (A) served as control for the free diffusion of dissolved drug through the

dialysis device.

Sensitivity and responsiveness compared to other ROS-sensitive systems

In contrast to other ROS-sensitive drug delivery systems reported in the literature,

the stabilized NCs presented herein represent the first example of a core-shell system

in which the redox-sensitivity of the shell is designed to release the “insensitive” bulk.

As a consequence, ROS-sensitive stabilized NCs stand to respond faster and have the

potential to be more sensitive to oxidation than bulk-type systems, for which the

oxidation process is slow due to diffusion phenomena, swelling, erosion, etc. Indeed,

other bulk thioether-based systems have shown to respond over a period of a couple

days to very high concentrations of H2O2 (5 – 10 vol%).176 For instance, Mahmoud et

al. have reported the release of a dye from thioether-containing particles to occur over

1 day upon exposure to 100 mM H2O2.186 Indeed, it should be noted that Allen et al.

have recently shown rapid (<10 min) dye release from thioether-containing particles

in response to 25 – 2000 ppm sodium hypochlorite or ROS generated by enzymatic

processing of H2O2.175 This study provides an indication that thioether bonds can be

very sensitive to other endogenous and potentially more reactive ROS than H2O2, and

that existing oxidants can be potentiated with enzymes. Interestingly, Almutairi and

co-workers have recently reported that polymers bearing aryl boronic ester protecting

groups were substantially more sensitive than thioethers to oxidation. Indeed,

concentrations of H2O2 as low as 100 μM led to polymer backbone degradation and

release of about 50% of cargo within 24 h.172 The system reported in our work is

sensitive to oxidant concentrations that are higher than that in the aforementioned

Page 93: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

90

studies. However, the reaction was relatively fast (in under 2 h) making the system

suitable to target regions with high/moderate ROS concentrations with lower chances

of off target effects (due to oxidation in low ROS concentration regions).

Conclusion

This study demonstrates that postpolymerization modification via the thiol–yne

reaction is a powerful tool for rapidly, rationally, and systematically preparing ROS-

responsive biodegradable polymeric stabilizers for NCs. Achieving a fine balance

between stabilization of the NCs and observing a tangible and rapid response upon

oxidation was possible using this approach. The prepared NCs were sensitive to

oxidation and, in the case of PTX, this phenomenon may be self-amplifying due to the

known triggering of ROS production in PTX-treated cells.187, 188 This paves the way

for the design of ROS-sensitive systems based on nanoparticles and drug NCs, and

offers many opportunities in the biotechnological field for location-specific shedding

of stabilizers, which can be used for imaging 189-191 or for improving cellular uptake. 178, 192 While hydrophobic thiols were used herein for affinity with the hydrophobic

surface of PTX NCs, the extensive list of other commercially available (and

hydrophilic) thiols render this a universal approach for preparing libraries of

polymeric surfactants with precisely the same number of repeat units for the

reversible stabilization of drug NCs and potentially other classes of nanoparticles such

as quantum dots and gold nanoparticles, for which aggregation strongly influences

their photonic properties. 193

Page 94: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

91

IV. Conclusion and Outlook

Page 95: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

92

Page 96: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

93

What was the reason for developing nanocrystals with functional stabilizers?

The successful therapy of cancer remains challenging because of the similarity of

cancer cells to those of the host, the difficulty associated with accessing them, and

evolving drug resistance mechanisms. In the US, the most underlying cause of death

in women, aged 40 to 79 years, and in men, aged 60 to 79 years, is cancer, whereas

about 15 years ago it was heart disease.194 In Europe, the cancer prevalence (the

number of patients diagnosed with cancer and survivors of cancer) was close to 13

million in 2010.195 This is related to the age distribution shifting toward the elderly

due to increased life expectancy and reduced mortality. Lung and breast cancers are

worldwide leading causes of cancer mortality.194 These figures will probably continue

to increase because the incidence of all epithelial cancer entities rises steeply with

age.195 By 2040, approximately one in four people in the United Kingdom aged 65

and over will be cancer survivors.196 Survivors of cancer suffer not only from effects

caused by the cancer and its treatment, but also from myriad treatment

complications.197 Some effects may quickly resolve (e.g., hair loss or nausea), while

others are long-term or permanent (e.g., infertility or neurological pain)43, 198

Understandably, adherence to life-saving cancer therapies remains a challenge.197

Should treatments be more selective to cancerous tissues, improved quality of life for

patients during therapy and beyond could ensue. Nanocrystals of poorly soluble

chemotherapeutic drugs, paclitaxel in particular, could be an avenue for improved

chemotherapy with less adverse effects. Different from conventional formulations,

nanocrystals correspond to small size drug systems almost entirely composed of drug,

and stabilized by a low amount of excipient.13, 14 Patients thus benefit from

formulations devoid of excessive amounts of cosolvents and excipients (e.g.,

surfactants), which can cause, for example, hypersensitivity reactions.5, 142

Furthermore, because of their small size, they are ideal for i.v. administration and

current ongoing research is attempting to promote their accumulation at the tumor site

by passive or active targeting. The more drug is selectively deposited at the site of

disease, the less adverse effects in healthy tissues (Figure 37A).

Page 97: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

94

Figure 37. Rationale of the presented work. (A) Systemic exposure to a drug can

cause adverse effects which are, in chemotherapy, often severe. The tumor’s unique

physiology, however, offers possibilities for selective accumulation of drug carriers.

This includes leaky vasculature (paired with poor lymphatic drainage in the core of

the tumor) for passive targeting via the EPR effect and/or special ligands for active

targeting. (B) Nanocrystals are prone to aggregation due to their high surface energy.

Steric stabilization with a polymeric stabilizer can be enhanced by cross-linking and

formation of nanocages. Furthermore this non-sheddable nanocage can be modified

with a targeting ligand for tumor targeting. (C) The preferential release of the

nanocrystal can be achieved by tailoring the stabilizer to be reactive to the tumors

microenvironment, in this case the presence of ROS. Quick shedding can potentially

facilitate uptake by tumor cells and eventually accelerate drug dissolution.

There are two known mechanisms for concentrating a nanosized drug carrier in a

tumor: by passive targeting via the EPR effect and/or active targeting (e.g., as

antibodies or DARPins).199-201 In certain cases the selective targeting of nanocarriers

to specific cells (i.e., tumor cells expressing certain receptors) in the body is

achievable using targeting ligands. 202 However, passive targeting, which has

nonetheless been studied excessively in animal models, has met with varied

Page 98: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

95

outcomes. For example, Yuan et al. concluded from injecting sterically stabilized

liposomes that the cutoff size (for exclusion from the tumor) lies somewhere above

~400 nm, because liposomes of 600 nm could not penetrate the tumor vessel wall.63

This cutoff size, however, depends on the type of tumor and its own physiology. For

example, the cutoff of polymeric micelles in hypervascular tumor tissue was 100 nm,

in contrast to a hypovascular tumor tissue that had a cutoff larger than 50 nm. The

coadministration of transforming growth factor signaling inhibitor enabled larger

micelles of 70 nm to accumulate in the tumor tissue, because this factor transiently

decreased the pericyte coverage of the endothelium in the neovasculature of

tumors.124 In humans, these less accessible tumors have been treated by using

angiotensin II to elevate blood pressure, which increased the diffusion of the colloid

into the tumor.203 Overall, the EPR effect in humans remains a highly heterogenous

phenomenon with variation from tumor model to tumor model and from patient to

patient.204, 205 For example, PEGylated liposomal doxorubicin failed to improve

overall survival for patients compared to conventional doxorubicin in metastatic

breast cancer.206 This is perhaps related to the fact that the tumor deposition of

liposomes in this type of tumors may not be sufficient. On the opposite, the good

response of Kaposi’s sarcoma and malignant pleural mesothelioma to this liposomal

formulation are attributed to accumulation via the EPR effect.207, 208 In the latter case,

tumor accumulation was emphasized by scintigraphic images showing in 80% of the

cases a significantly higher uptake of radio-labeled liposomal doxorubicin in the

tumor than in soft tissue.208 Based on these examples it appears that passive drug

targeting is possible, although many factors interplay and its success cannot be

predicted. Key parameters seem to include successful shielding from MPS uptake by

PEGylation for long circulation times and small size of the carrier. Therefore, within

this doctoral thesis, a method was sought out to stabilize nanocrystals with a PEG

copolymer to enable long circulation times and increased size stability for passive

accumulation via EPR effect. In addition, new approaches for a non-sheddable

coating to facilitate potential active targeting were developed as well as a coating for

fast release of the drug in response to a stimulus.

The potential change in biodistribution of a drug by using a nanocrystal formulation

can increase the therapeutic benefit for patients.

Page 99: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

96

What was shown within this work?

An outline of the work achieved in this thesis is given in Figure 37. This work was

directed towards the study of whether a certain design of polymer coating can slow

down the dissolution of nanocrystals. Although this could not be achieved within this

work, findings showed that coatings can be designed to be either less sheddable from

the surface or reactive to cause nanocrystal destabilization in response to a stimulus.

As model system, nanocrystals of the drug paclitaxel were produced by wet milling.

Special emphasis was put on the design of a stabilizer to prevent aggregation. A non-

covalently attached coating on the nanocrystal surface can, in principle, permit active

targeting of the whole construct. This purpose can be achieved via ligand attachment

to the polymer without compromising the drug molecule’s integrity. Therefore,

functional monomers carrying alkyne moieties (-propargyl--valerolactone) were

synthesized and copolymerized with -caprolactone from a methoxy-PEG

macroinitiator to obtain custom polymeric stabilizers. These stabilizers were cross-

linked (Figure 37B) with different diazido compounds by a copper-catalyzed 1,3-

dipolar cycloaddition (Figure 18). In addition to increased size stability for the

nanocrystals, the formed nanocages resisted dissolution, and were less prone to be

shed from the surface of the nanocrystal. In a second study, the stabilizers were used

to prepare a library of new polymers by systematic grafting of different alkane thiols

to the polymer by radical thiol−yne addition. These additions produced polyester

blocks with different polarities, which were tested for optimal nanocrystal size

stabilization. Furthermore, oxidation of their thioether bonds changed the stabilizer’s

lipophilicity and affinity for the nanocrystal surface (Figure 37C), which in certain

cases destabilized the system as seen by an increase in nanocrystal size (Figures 32

and 34). Although not tested within this doctoral thesis, the fast shedding from the

crystal surface can, in principle, be useful for fast release of the drug and increased

uptake by tumoral cells. Non-sheddable nanocages and ROS responsive stabilizers

could contribute to increased drug uptake in the tumor by passive targeting via the

EPR effect.

How does this relate to other work?

Within this doctoral thesis, special emphasis was put on the design of a stabilizer,

which was amenable for modifications to improve distribution of the drug to the

Page 100: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

97

tumor. Traditional stabilizers in use for parenteral nanocrystal formulations are

usually members of the poloxamer type.36, 103 However, these can desorb from the

surface upon dilution, as seen for poloxamer 407-stabilized nanocrystals.114 This can

cause nanocrystal destabilization with aggregation and increase in size, which is a

disadvantage in the use of nanosuspensions for anticancer therapy. Other groups have

evaluated stabilizer-free fluorescent paclitaxel nanocrystals but only observed

minimal accumulation via the EPR effect.89, 162 These “hybrid” (precipitate of

paclitaxel and dye) nanocrystals could be tracked via fluorescence imaging. When

injected into mice bearing MCF-7 tumor xenografts, the nanoparticle-incorporated

dye was retained within the tumor and surrounding tissue for several days compared

to the rather fast clearance (ca. one day) of the free dye.162 Fluorescence images

showed dye deposition in tumor and lung (possibly due to the accumulation of

nanocrystals) and kidney (excretion pathway for the hydrophilic free dye). However,

a further study using hybrid nanocrystals containing radiolabeled paclitaxel revealed

that less than 1% drug accumulated in the tumor (HT-29 xenograft in mice).89 Close

to 40% of injected drug was found in the liver. The lack of stabilizer might have

caused the nanocrystals to aggregate and/or to be recognized by MPS after injection.

Therefore, the high liver and the low tumor accumulation possibly resulted from low

size stability and uptake by MPS. Masking the surface of paclitaxel nanocrystals with

transferrin increased size stability over time but could not improve anticancer

efficiency compared to the conventional dissolved formulation.119 Indeed, it is a

challenging task to secure surfactants to the surface of nanocrystals, especially during

the administration and blood circulation stages.89 The approach presented in this

thesis can provide the means for addressing this issue.

Modifications to standard polymers are possible, but often limited in the number of

introducible functional groups. The commonly used poloxamers only offer

modification at the two terminal hydroxyl groups,209 which is a restriction to testing a

range of introduced modifications. The custom polymerization of functional

monomers has the advantage of including functional moieties of desired number.

The nanocrystals within this work were prepared with a coating susceptible to

cross-linking via click-chemistry to form a non-sheddable nanocage. This nanocage is

expected to not only increase colloidal stability by introducing steric hindrance, but

also to reduce opsonization and uptake by MPS. Furthermore, similar stabilizers were

modified by the postpolymerization approach with thiol bearing organic compounds

Page 101: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

98

(of increasing length and hydrophobicity) and investigated in a systematic fashion for

their stabilizing capabilities. In some cases, relatively stable nanocrystals (about 20%

size increase over 3 weeks at room temperature) were obtained. The increased

colloidal stability is an important stepping stone for passive accumulation via the EPR

effect. On the other hand, the reactivity of the thioether containing polymers to

predominant ROS presence in the tumor tissue could trigger oxidation-induced

shedding of the stabilizer. After removal of the polymer coating the nanocrystals

could potentially be taken up to a higher extent by tumoral cells. Increased cell uptake

after PEG shedding from the surface has been shown for other nanoformulations.210

Both, the non-sheddable nanocage and the ROS-reactive stabilizer, are new

approaches of how to make the most of passive targeting.

What more could be done?

This doctoral thesis covered the thorough in vitro characterization involving size

stability, nanocrystal drug content and integrity after milling, polymer

characterization before and after cross-linking, sheddability, and oxidation. However,

in order to fulfill requirements for pharmaceutical application, these formulations

should be compared to benchmark formulations from the literature and more in vitro

characterization should be included, such as size stability measurements at elevated

temperatures, for longer durations, and at higher concentrations; interaction with cells

and cell uptake; and more investigation into the degradability of the polymeric

nanocage and the adsorption strength of the designed polymers in general. The in vivo

characterization of the pharmacokinetics and biodistribution of the formulated

nanocrystals is the next step to perform. The relevance of a non-sheddable coating for

tumor targeting with a conjugated ligand relies on a sufficiently long circulation time

of the nanocrystals. In theory, the PEG coating should protect the particles from rapid

clearance by the MPS. Adjustments in length and density of PEG on the crystal

surface can potentially help in achieving an optimal circulation time. Furthermore, the

dissolution velocity of paclitaxel nanocrystals enclosed by nanocages appeared to be

independent of the extent of cross-linking tested. Optimization in terms of coating

thickness can potentially prevent premature dissolution. Possible ways for increasing

coating thickness could include layer-by-layer assembly,23, 40 the adsorption and

cross-linking of more polymer layers, or the choice of a different stabilizer which

inherently covers the nanocrystal surface to a higher degree. Although the preparation

Page 102: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

99

of a thicker coating on the nanocrystal might make the system more complex and less

elegant, it is nevertheless vital for a sufficient reduction in dissolution velocity to

enable a longer circulation time of the drug nanocrystal. Once a sufficiently long

circulation time for the nanocrystals has been achieved, further experiments with a

targeting ligand attached to the nanocage could be designed. New polymers amenable

for coupling to targeting ligands can be obtained by use of a heterotelechelic PEG as

macroinitiator for the polymerization of -propargyl--valerolactone and -

caprolactone. This polymer could be added at, for example, 5 to 10 wt. % of the

regular polymer amount during milling. After cross-linking and purification, a

targeting ligand could be attached through simple one step reactions such as the

reaction of a thiol group with a maleimide. This targeted nanocrystal formulation

could first be tested in vitro in cell culture for uptake and cytotoxic activity and then

in vivo for toxicity and efficacy in tumor-bearing mice.

Similar experiments should be designed for nanocrystals carrying the ROS sensitive

polymer. Here it would be important to assess the production of ROS in the tumor and

try to correlate it with the performance of the nanocrystal system. Additionally, the

selectivity of the ROS reactivity for the desired target tissue and the elimination of the

oxidized polymer should be studied. Further polymer modification may be necessary

as the conditions tested herein do not reflect the in vivo situation (high ROS

concentrations, closed system).

As discussed above, the diblock copolymers described in this work are intended to

be administered intravenously. PEG is known to be safe for i.v. administration and is

used in a variety of commercialized products.142, 211 However the coupled polyester

block has not been studied so far. Care needs to be taken that residual free alkyne

groups of -propargyl--valerolactone do not react in vivo with thiol-containing

molecules, which are vastly present as proteins such as enzymes or covering cell

surfaces. Indeed the successful cross-linking reaction with azides and

postpolymerization modification with thiols should in theory consume all available

alkyne groups. Otherwise a quenching reaction with a small alkane thiol (e.g.,

mercaptoethanol) could be included. The biodegradable nature of the polyester should

allow fast degradation into smaller fragments, which are expected to be readily

excretable through the kidneys. Studies need to be undertaken to characterize the

elimination and safety profile of these novel block copolymers.

Page 103: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

100

The nanocrystal formulations presented herein have not been compared to other

benchmark formulations and a thorough evaluation is needed to see whether this

process is developable. To satisfy pharmaceutical quality aspects these formulations

need to show their potency, selectivity and safety. The milling process and subsequent

cross-linking would need to be done under sterile conditions for i.v. purposes. The

residual content, such as wear from milling or catalyst from polymer synthesis and

cross-linking, must be removed or reduced to below authorized thresholds. Many

parameters in the preparation would have to be optimized for a reliable production on

larger scale, such as milling apparatus, milling media composition, drug-to-polymer

ratio and concentration for input, separation of the nanocrystals from milling media

and from excess polymer, cross-linking conditions and purification, and so forth.

In conclusion, new routes for controlled nanocrystal size stabilization were

developed and studied by creating cross-linkable and ROS sensitive polymers. The

knowledge obtained from these studies could potentially help in the development of a

safe nanocrystal formulation for improved, because selective, chemotherapy of

cancer.

Page 104: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

101

V. Supplementary Information

Page 105: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

102

Page 106: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

103

Figure S 1. 1H NMR spectra of -propargyl--valerolactone (1) in CDCl3 (A) and in

MeOD (B). The signals at 2.01 and 4.25 ppm confirm the presence of an acetylene

proton and an intact lactone ring, respectively. The acetylene peak was found to be

very solvent sensitive and moved to 2.33 ppm in MeOD.

Page 107: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

104

Figure S 2. 1H NMR spectrum of -chloro--caprolactone. The proton signals at 4.63

and 4.23 ppm indicated oxidation of cyclohexanone to the lactone, while the signal at

4.80 ppm showed the incorporation of chlorine.

Figure S 3. 1H NMR spectrum of -azido--caprolactone (2). The proton signals at

4.40 and 4.12 ppm indicated incorporation of the azide and the lactone integrity,

respectively. The peak at 2.61 belongs to residual DMSO.

Page 108: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

105

Figure S 4. 1H NMR spectrum of 3. The signal at 2.01 ppm showed successful

incorporation of -propargyl--valerolactone. Polymer composition was calculated

from signals at 2.01 ppm for -propargyl--valerolactone units and 1.31 ppm for -

caprolactone units using the signal from the methoxy group of mPEG at 3.38 ppm as

reference.

Page 109: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

106

Figure S 5. 1H NMR spectrum 4. The proton signal at 2.01 ppm showed successful

incorporation of -propargyl--valerolactone units. Polymer composition was

calculated as for 3.

Page 110: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

107

Figure S 6. 1H NMR spectrum of 5. The proton signal at 3.85 ppm indicated

successful incorporation of -azido--caprolactone units. Polymer composition was

calculated from signals at 3.85 ppm for -azido--caprolactone units (from which the 13C satellite from mPEG was removed) and 2.28 ppm for -caprolactone units using

the signal from the methoxy group of PEG at 3.38 ppm as reference.

Page 111: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

108

Figure S 7. 1H NMR spectrum of polymer 13.

Page 112: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

109

Figure S 8. 1H NMR spectrum of polymer 8.

Figure S 9. 1H NMR spectrum of polymer 9.

Page 113: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

110

Figure S 10. 1H NMR spectrum of polymer 10.

Figure S 11. 1H NMR spectrum of polymer 11.

Page 114: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

111

Figure S 12. 1H NMR spectrum of polymer 12. The polymer was purified by

extensive dialysis against 63% THF.

Page 115: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

112

Figure S 13. 1H NMR spectrum of polymer 14.

Figure S 14. 1H NMR spectrum of polymer 15.

Page 116: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

113

Figure S 15. 1H NMR spectrum of polymer 16. The polymer was purified by

extensive dialysis against 63% THF.

Page 117: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

114

Figure S 16. 1H NMR spectrum of polymer 17.

Page 118: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

115

Figure S 17. 1H NMR spectrum of polymer 18. The polymer was purified by

extensive dialysis against 63% THF.

Page 119: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

116

Figure S 18. 1H–1H correlation spectrum of 14.

Page 120: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

117

VI. Curriculum Vitae and Scientific Contributions

Page 121: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

118

Page 122: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

119

Kathrin FUHRMANN née Lüling born 20 Feburary 1981 in Hagen, Germany German citizen Doctoral Studies 09/2008−08/2013 Research project: Preparation of Drug Nanocrystals Stabilized by

Functionalized Polymeric Coatings, under supervision of Prof. Jean-Christophe Leroux, Institute of Pharmaceutical Sciences (IPW), ETH Zurich, Switzerland and co-supervised by Marc A. Gauthier, Énergie Matériaux Télécommunications Research Centre (INRS-EMT), Varennes, Canada

Undergraduate Studies and professional experience 07/2008

Licensure as pharmacist

12/2007−05/2008

Pharmacy internship at „Apotheke im Schweizer Viertel“ in Berlin, Germany

05/2007−10/2007 Research internship at the laboratory of Prof. Jean-Christophe Leroux, Faculty of Pharmacy, Université de Montréal, QC, Canada Research project: Preparation of Drug Nanocrystals by Femtosecond Laser Ablation

02/2006−03/2006 Research internship at the laboratory of Prof. Fritz Pragst, Institut f. Rechtsmedizin, Charité Berlin, Germany Research project: Untersuchung von Rohtabak auf seine enthaltenen Alkaloide

04/2003−04/2007 Undergraduate studies in pharmacy, Freie Universität Berlin, Germany

Original publications:

K Fuhrmann, A Połomska, C Aeberli, B Castagner, MA Gauthier, JC Leroux (2013) Modular Design of Redox-Responsive Stabilizers for Nanocrystals. ACS Nano, 7, 8243-8250. K Fuhrmann, JD Schulz, MA Gauthier, JC Leroux (2012) PEG Nanocages as Non-sheddable Stabilizers for Drug Nanocrystals. ACS Nano 6, 1667-1676. K Fuhrmann, MA Gauthier, JC Leroux (2010) Crosslinkable Polymers for Nanocrystal Stabilization. J. Control. Release 148, e12-e13. S Kenth, JP Sylvestre, K Fuhrmann, M Meunier, JC Leroux, (2011) Fabrication of Paclitaxel Nanocrystals by Femtosecond Laser Ablation and Fragmentation. J. Pharm. Sci. 100, 1022-1030.

Page 123: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

120

Oral presentations K Fuhrmann, MA Gauthier, JC Leroux (2012), PEG Nanocages as Non-sheddable Stabilizers for Drug Nanocrystals, Swiss Galenic Meeting, ETH Zürich, Switzerland, K Fuhrmann, MA Gauthier, JC Leroux (2011), Stabilization of Drug Nanocrystals by Functionalized Polymeric Coatings, PharmSciFair, Prague, Czech Republic K Fuhrmann, MA Gauthier, JC Leroux (2011), Drug Nanocrystals Stabilized by Functionalized Polymeric Coatings, Doktorandentag (Doctoral Students Day), ETH Zürich, Switzerland K Fuhrmann, MA Gauthier, JC Leroux (2010), Functionalized Polymers for Nanocrystal Stabilization, Zürich-Geneva-Basle Meeting, University of Geneva, Switzerland K Lüling, JC Leroux (2009), Preparation of Drug Nanocrystals Stabilized by Polymeric Coatings, Zürich-Geneva Meeting, ETH Zürich, Switzerland Poster presentations K Fuhrmann, MA Gauthier, JC Leroux (2010), Cross-linkable Polymers for Nanocrystal Stabilization, Swiss Pharma Science Day, Bern, Switzerland K Fuhrmann, MA Gauthier, JC Leroux (2010), Cross-linking Polymers for Nanocrystal Stabilization, 3rd International NanoBio Conference 2010, Zurich, Switzerland K Fuhrmann, MA Gauthier, JC Leroux (2010), Cross-linkable Polymers for Nanocrystal Stabilization, 11th European Symposium on Controlled Drug Delivery, (ESCDD), Egmond aan Zee, Netherlands

Page 124: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

121

VII. Acknowledgments

Page 125: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

122

Page 126: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

123

First of all, I would like to thank Prof. Jean-Christophe Leroux for the unique

opportunity to do doctoral studies in his research group. I am very thankful for his

great scientific input, constant availability, honesty, and fairness.

My sincerest thanks go to Prof. Marc A. Gauthier for his extensive scientific input,

great support in writing, and neverending motivation.

I thank Prof. Bruno Gander for accepting to be co-examiner of this doctoral thesis.

Furthermore, I am grateful for all his help with everyday matters and orientation in

the organization of ETH not only during the beginning but throughout my doctoral

studies.

I would like to thank the institution ETH for its innovativeness, management and

structure, and motivated employees. I enjoyed conducting research here.

Many thanks also go to Prof. Werner (Pharmaceutical analytics group, IPW) for the

extensive use of the FTIR instrument whenever needed.

I thank every former and present member of the Drug Formulation and Delivery

Group for the time spent inside and outside of the lab. Special thanks go to Dr. Paola

Luciani, Dr. Bastien Castagner, Dr. Nuria Bayo-Puxan, Dr. Marie-Hélène Dufresne,

Dr. Arnaud Felber, Dr. Vincent Forster, Dr. Gregor Fuhrmann, Dr. Davide Brambilla,

Dr. Soo Hyeon Lee, Jessica Schulz, Lorine Brülisauer, Athanasia Dasargyri, Estelle

Durantie, Yuhui Gong, Mattias Ivarsson, Monica Langfritz, Elena Moroz, Anna

Połomska, Maurizio Roveri, and Mi Liu for their advice and help.

Finally, I thank my friends and family for their support.

Page 127: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

124

Page 128: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

125

VIII. List of References

Page 129: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

126

Page 130: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

127

1. Lipinski, C. A. Drug-like Properties and the Causes of Poor Solubility and Poor Permeability. J. Pharmacol. Toxicol. Methods 2000, 44, 235-249.

2. Bhugra, C.; Pikal, M. J. Role of Thermodynamic, Molecular, and Kinetic Factors in Crystallization from the Amorphous State. J. Pharm. Sci. 2008, 97, 1329-1349.

3. Stella, V. J.; Nti-Addae, K. W. Prodrug Strategies to Overcome Poor Water Solubility. Adv. Drug Del. Rev. 2007, 59, 677-694.

4. Kawakami, K. Modification of Physicochemical Characteristics of Active Pharmaceutical Ingredients and Application of Supersaturatable Dosage Forms for Improving Bioavailability of Poorly Absorbed Drugs. Adv. Drug Del. Rev. 2012, 64, 480-495.

5. Gelderblom, H.; Verweij, J.; Nooter, K.; Sparreboom, A. Cremophor EL: The Drawbacks and Advantages of Vehicle Selection for Drug Formulation. Eur. J. Cancer 2001, 37, 1590-1598.

6. Weiszhár, Z.; Czúcz, J.; Révész, C.; Rosivall, L.; Szebeni, J.; Rozsnyay, Z. Complement Activation by Polyethoxylated Pharmaceutical Surfactants: Cremophor-EL, Tween-80 and Tween-20. Eur. J. Pharm. Sci. 2012, 45, 492-498.

7. Morshed, K. M.; Jain, S. K.; McMartin, K. E. Propylene Glycol-Mediated Cell Injury in a Primary Culture of Human Proximal Tubule Cells. Toxicol. Sci. 1998, 46, 410-417.

8. Petrou, E.; Iakovou, I.; Boutsikou, M.; Girasis, C.; Mavrogeni, S.; Pavlides, G. Acute Epigastric and Low Back Pain during Amiodarone Infusion; Is It the Drug or the Vehicle to Blame? Heart & Lung 2014, 43, 60-61.

9. Drummond, D. C.; Noble, C. O.; Hayes, M. E.; Park, J. W.; Kirpotin, D. B. Pharmacokinetics and In Vivo Drug Release Rates in Liposomal Nanocarrier Development. J. Pharm. Sci. 2008, 97, 4696-4740.

10. Yang, T.; Choi, M.-K.; Cui, F.-D.; Kim, J. S.; Chung, S.-J.; Shim, C.-K.; Kim, D.-D. Preparation and Evaluation of Paclitaxel-loaded PEGylated Immunoliposome. J. Control. Release 2007, 120, 169-177.

11. Gaucher, G.; Dufresne, M.-H.; Sant, V. P.; Kang, N.; Maysinger, D.; Leroux, J.-C. Block Copolymer Micelles: Preparation, Characterization and Application in Drug Delivery. J. Control. Release 2005, 109, 169-188.

12. Musumeci, T.; Ventura, C. A.; Giannone, I.; Ruozi, B.; Montenegro, L.; Pignatello, R.; Puglisi, G. PLA/PLGA Nanoparticles for Sustained Release of Docetaxel. Int. J. Pharm. 2006, 325, 172-179.

13. Merisko-Liversidge, E.; Liversidge, G. G. Nanosizing for Oral and Parenteral Drug Delivery: A Perspective on Formulating Poorly-water Soluble Compounds Using Wet Media Milling Technology. Adv. Drug Del. Rev. 2011, 63, 427-440.

14. Sun, B.; Yeo, Y. Nanocrystals for the Parenteral Delivery of Poorly Water-soluble Drugs. Curr. Opin. Solid State Mater. Sci. 2012, 16, 295-301.

Page 131: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

128

15. Möschwitzer, J. P. Drug Nanocrystals in the Commercial Pharmaceutical Development Process. Int. J. Pharm. 2013, 453, 142-156.

16. Harrison, M.; Hahn, N.; Pili, R.; Oh, W.; Hammers, H.; Sweeney, C.; Kim, K.; Perlman, S.; Arnott, J.; Sidor, C., et al. A Phase II Study of 2-Methoxyestradiol (2ME2) NanoCrystal® Dispersion (NCD) in Patients with Taxane-refractory, Metastatic Castrate-resistant Prostate Cancer (CRPC). Invest. New Drugs 2011, 29, 1465-1474.

17. Tevaarwerk, A. J.; Holen, K. D.; Alberti, D. B.; Sidor, C.; Arnott, J.; Quon, C.; Wilding, G.; Liu, G. Phase I Trial of 2-Methoxyestradiol NanoCrystal Dispersion in Advanced Solid Malignancies. Clin. Cancer. Res. 2009, 15, 1460-1465.

18. Butcher, J.; Garg, S.; Kim, D.; Sharma, P. A Modified Approach to Predict Dissolution and Absorption of Polydisperse Powders. Pharm. Res. 2008, 25, 2309-2311.

19. Müller, R. H.; Peters, K. Nanosuspensions for the Formulation of Poorly Soluble Drugs: I. Preparation by a Size-reduction Technique. Int. J. Pharm. 1998, 160, 229-237.

20. Wu, Y.; Loper, A.; Landis, E.; Hettrick, L.; Novak, L.; Lynn, K.; Chen, C.; Thompson, K.; Higgins, R.; Batra, U., et al. The Role of Biopharmaceutics in the Development of a Clinical Nanoparticle Formulation of MK-0869: A Beagle Dog Model Predicts Improved Bioavailability and Diminished Food Effect on Absorption in Human. Int. J. Pharm. 2004, 285, 135-146.

21. Gao, L.; Liu, G.; Ma, J.; Wang, X.; Zhou, L.; Li, X. Drug Nanocrystals: In Vivo Performances. J. Control. Release 2012, 160, 418-430.

22. Cerdeira, A. M.; Gander, B.; Mazzotti, M. Role of Milling Parameters and Particle Stabilization on Nanogrinding of Drug Substances of Similar Mechanical Properties. Chem. Eng. Technol. 2011, 34, 1427-1438.

23. Lvov, Y. M.; Pattekari, P.; Zhang, X.; Torchilin, V. Converting Poorly Soluble Materials into Stable Aqueous Nanocolloids. Langmuir 2011, 27, 1212-1217.

24. Kenth, S.; Sylvestre, J.-P.; Fuhrmann, K.; Meunier, M.; Leroux, J.-C. Fabrication of Paclitaxel Nanocrystals by Femtosecond Laser Ablation and Fragmentation. J. Pharm. Sci. 2011, 100, 1022-1030.

25. Rabinow, B. E. Nanosuspensions in Drug Delivery. Nat. Rev. Drug Discov. 2004, 3, 785-796.

26. Peltonen, L.; Hirvonen, J. Pharmaceutical Nanocrystals by Nanomilling: Critical Process Parameters, Particle Fracturing and Stabilization Methods. J. Pharm. Pharmacol. 2010, 62, 1569-1579.

27. Gao, L.; Liu, G.; Ma, J.; Wang, X.; Zhou, L.; Li, X.; Wang, F. Application of Drug Nanocrystal Technologies on Oral Drug Delivery of Poorly Soluble Drugs. Pharm. Res. 2013, 30, 307-324.

Page 132: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

129

28. Van Eerdenbrugh, B.; Vermant, J.; Martens, J. A.; Froyen, L.; Van Humbeeck, J.; Augustijns, P.; Van den Mooter, G. A Screening Study of Surface Stabilization During the Production of Drug Nanocrystals. J. Pharm. Sci. 2009, 98, 2091-2103.

29. Dhumal, R. S.; Biradar, S. V.; Yamamura, S.; Paradkar, A. R.; York, P. Preparation of Amorphous Cefuroxime Axetil Nanoparticles by Sonoprecipitation for Enhancement of Bioavailability. Eur. J. Pharm. Biopharm. 2008, 70, 109-115.

30. Sigfridsson, K.; Forssén, S.; Holländer, P.; Skantze, U.; de Verdier, J. A Formulation Comparison, Using a Solution and Different Nanosuspensions of a Poorly Soluble Compound. Eur. J. Pharm. Biopharm. 2007, 67, 540-547.

31. Chan, H.-K.; Kwok, P. C. L. Production Methods for Nanodrug Particles Using the Bottom-up Approach. Adv. Drug Del. Rev. 2011, 63, 406-416.

32. Ghosh, I.; Schenck, D.; Bose, S.; Liu, F.; Motto, M. Identification of Critical Process Parameters and Its Interplay with Nanosuspension Formulation Prepared by Top Down Media Milling Technology – A QbD Perspective. Pharm. Dev. Technol. 2013, 18, 719-729.

33. George, M.; Ghosh, I. Identifying the Correlation between Drug/Stabilizer Properties and Critical Quality Attributes (CQAs) of Nanosuspension Formulation Prepared by Wet Media Milling Technology. Eur. J. Pharm. Sci. 2013, 48, 142-152.

34. Liu, Y.; Huang, L.; Liu, F. Paclitaxel Nanocrystals for Overcoming Multidrug Resistance in Cancer. Mol. Pharm. 2010, 7, 863-869.

35. Smet, L.; Colin, P.; Ceelen, W.; Bracke, M.; Bocxlaer, J.; Remon, J.; Vervaet, C. Development of a Nanocrystalline Paclitaxel Formulation for Hipec Treatment. Pharm. Res. 2012, 29, 2398-2406.

36. Baert, L.; van ‘t Klooster, G.; Dries, W.; François, M.; Wouters, A.; Basstanie, E.; Iterbeke, K.; Stappers, F.; Stevens, P.; Schueller, L., et al. Development of a Long-acting Injectable Formulation with Nanoparticles of Rilpivirine (TMC278) for HIV Treatment. Eur. J. Pharm. Biopharm. 2009, 72, 502-508.

37. Alexis, F.; Pridgen, E.; Molnar, L. K.; Farokhzad, O. C. Factors Affecting the Clearance and Biodistribution of Polymeric Nanoparticles. Mol. Pharm. 2008, 5, 505-515.

38. 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.

39. Fuhrmann, K.; Schulz, J. D.; Gauthier, M. A.; Leroux, J.-C. PEG Nanocages as Non-sheddable Stabilizers for Drug Nanocrystals. ACS Nano 2012, 6, 1667-1676.

40. Agarwal, A.; Lvov, Y.; Sawant, R.; Torchilin, V. Stable Nanocolloids of Poorly Soluble Drugs with High Drug Content Prepared Using the Combination of Sonication and Layer-by-layer Technology. J. Control. Release 2008, 128, 255-260.

41. Becker, A. L.; Johnston, A. P. R.; Caruso, F. Layer-By-Layer-Assembled Capsules and Films for Therapeutic Delivery. Small 2010, 6, 1836-1852.

Page 133: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

130

42. Jordan, M. A.; Wilson, L. Microtubules as a Target for Anticancer Drugs. Nat. Rev. Cancer 2004, 4, 253-265.

43. Dumontet, C.; Jordan, M. A. Microtubule-binding Agents: A Dynamic Field of Cancer Therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790-803.

44. Kingston, D. G. I. The Shape of Things to Come: Structural and Synthetic Studies of Taxol and Related Compounds. Phytochemistry 2007, 68, 1844-1854.

45. Zhang, Z.; Mei, L.; Feng, S.-S. Paclitaxel Drug Delivery Systems. Expert Opin. Drug Deliv. 2013, 10, 325-340.

46. Windebank, A. J.; Grisold, W. Chemotherapy-induced Neuropathy. J. Peripher. Nerv. Syst. 2008, 13, 27-46.

47. Green, M. R.; Manikhas, G. M.; Orlov, S.; Afanasyev, B.; Makhson, A. M.; Bhar, P.; Hawkins, M. J. Abraxane®, a Novel Cremophor®-free, Albumin-bound Particle Form of Paclitaxel for the Treatment of Advanced Non-small-cell Lung Cancer. Ann. Oncol. 2006, 17, 1263-1268.

48. Desai, N. P.; Tao, C.; Yang, A.; Louie, L.; Soon-shiong, P., Formulations of Pharmacological Agents, Methods for the Preparation Thereof and Methods for the Use Thereof, United States Patent 8137684 B2, 2012

49. Gradishar, W. J.; Tjulandin, S.; Davidson, N.; Shaw, H.; Desai, N.; Bhar, P.; Hawkins, M.; O'Shaughnessy, J. Phase III Trial of Nanoparticle Albumin-Bound Paclitaxel Compared With Polyethylated Castor Oil-Based Paclitaxel in Women With Breast Cancer. J. Clin. Oncol. 2005, 23, 7794-7803.

50. Teneriello, M. G.; Tseng, P. C.; Crozier, M.; Encarnacion, C.; Hancock, K.; Messing, M. J.; Boehm, K. A.; Williams, A.; Asmar, L. Phase II Evaluation of Nanoparticle Albumin–Bound Paclitaxel in Platinum-Sensitive Patients With Recurrent Ovarian, Peritoneal, or Fallopian Tube Cancer. J. Clin. Oncol. 2009, 27, 1426-1431.

51. Stinchcombe, T.; Socinski, M.; Walko, C.; O’Neil, B.; Collichio, F.; Ivanova, A.; Mu, H.; Hawkins, M.; Goldberg, R.; Lindley, C., et al. Phase I and Pharmacokinetic Trial of Carboplatin and Albumin-bound Paclitaxel, ABI-007 (Abraxane®) on Three Treatment Schedules in Patients with Solid Tumors. Cancer Chemother. Pharmacol. 2007, 60, 759-766.

52. Ibrahim, N. K.; Samuels, B.; Page, R.; Doval, D.; Patel, K. M.; Rao, S. C.; Nair, M. K.; Bhar, P.; Desai, N.; Hortobagyi, G. N. Multicenter Phase II Trial of ABI-007, an Albumin-Bound Paclitaxel, in Women With Metastatic Breast Cancer. J. Clin. Oncol. 2005, 23, 6019-6026.

53. Hersh, E. M.; O'Day, S. J.; Ribas, A.; Samlowski, W. E.; Gordon, M. S.; Shechter, D. E.; Clawson, A. A.; Gonzalez, R. A Phase 2 Clinical Trial of nab-Paclitaxel in Previously Treated and Chemotherapy-naive Patients with Metastatic Melanoma. Cancer 2010, 116, 155-163.

Page 134: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

131

54. Kola, I.; Landis, J. Can the Pharmaceutical Industry Reduce Attrition Rates? Nat. Rev. Drug Discov. 2004, 3, 711-716.

55. Paul, S. M.; Mytelka, D. S.; Dunwiddie, C. T.; Persinger, C. C.; Munos, B. H.; Lindborg, S. R.; Schacht, A. L. How to Improve R&D Productivity: The Pharmaceutical Industry's Grand Challenge. Nat. Rev. Drug Discov. 2010, 9, 203-214.

56. Kelloff, G. J.; Sigman, C. C. Cancer Biomarkers: Selecting the Right Drug for the Right Patient. Nat. Rev. Drug Discov. 2012, 11, 201-214.

57. Crawford, J.; Dale, D. C.; Lyman, G. H. Chemotherapy-induced Neutropenia. Cancer 2004, 100, 228-237.

58. Paus, R.; Haslam, I. S.; Sharov, A. A.; Botchkarev, V. A. Pathobiology of Chemotherapy-induced Hair Loss. Lancet Oncol. 2013, 14, e50-e59.

59. Rubenstein, E. B.; Peterson, D. E.; Schubert, M.; Keefe, D.; McGuire, D.; Epstein, J.; Elting, L. S.; Fox, P. C.; Cooksley, C.; Sonis, S. T. Clinical Practice Guidelines for the Prevention and Treatment of Cancer Therapy–induced Oral and Gastrointestinal Mucositis. Cancer 2004, 100, 2026-2046.

60. Fallowfield, L.; McGurk, R.; Dixon, M. Same Gain, Less Pain: Potential Patient Preferences for Adjuvant Treatment in Premenopausal Women with Early Breast Cancer. Eur. J. Cancer 2004, 40, 2403-2410.

61. Matsumura, Y.; Maeda, H. A New Concept for Macromolecular Therapeutics in Cancer Chemotherapy: Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Res. 1986, 46, 6387-6392.

62. Brown, J. M.; Giaccia, A. J. The Unique Physiology of Solid Tumors: Opportunities (and Problems) for Cancer Therapy. Cancer Res. 1998, 58, 1408-1416.

63. Yuan, F.; Dellian, M.; Fukumura, D.; Leunig, M.; Berk, D. A.; Torchilin, V. P.; Jain, R. K. Vascular Permeability in a Human Tumor Xenograft: Molecular Size Dependence and Cutoff Size. Cancer Res. 1995, 55, 3752-3756.

64. Pelicano, H.; Carney, D.; Huang, P. ROS Stress in Cancer Cells and Therapeutic Implications. Drug Resist. Update 2004, 7, 97-110.

65. Fang, J.; Seki, T.; Maeda, H. Therapeutic Strategies by Modulating Oxygen Stress in Cancer and Inflammation. Adv. Drug Del. Rev. 2009, 61, 290-302.

66. Papahadjopoulos, D.; Allen, T. M.; Gabizon, A.; Mayhew, E.; Matthay, K.; Huang, S. K.; Lee, K. D.; Woodle, M. C.; Lasic, D. D.; Redemann, C. Sterically Stabilized Liposomes: Improvements in Pharmacokinetics and Antitumor Therapeutic Efficacy. P. Natl. Acad. Sci. USA 1991, 88, 11460-11464.

67. Kang, N.; Perron, M.-È.; Prud'homme, R. E.; Zhang, Y.; Gaucher, G.; Leroux, J.-C. Stereocomplex Block Copolymer Micelles: Core−Shell Nanostructures with Enhanced Stability. Nano Lett. 2005, 5, 315-319.

Page 135: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

132

68. Soppimath, K. S.; Aminabhavi, T. M.; Kulkarni, A. R.; Rudzinski, W. E. Biodegradable Polymeric Nanoparticles as Drug Delivery Devices. J. Control. Release 2001, 70, 1-20.

69. Barenholz, Y. Doxil® — The First FDA-approved Nano-drug: Lessons Learned. J. Control. Release 2012, 160, 117-134.

70. Singla, A. K.; Garg, A.; Aggarwal, D. Paclitaxel and Its Formulations. Int. J. Pharm. 2002, 235, 179-192.

71. Olson, R. D.; Mushlin, P. S. Doxorubicin Cardiotoxicity: Analysis of Prevailing Hypotheses. The FASEB Journal 1990, 4, 3076-3086.

72. Wu, L.; Zhang, J.; Watanabe, W. Physical and Chemical Stability of Drug Nanoparticles. Adv. Drug Del. Rev. 2011, 63, 456-469.

73. Junghanns, J.-U. A. H.; Müller, R. H. Nanocrystal Technology, Drug Delivery and Clinical Applications Int. J. Nanomed. 2008, 3, 295-309.

74. Muller, R. H.; Keck, C. M. Challenges and Solutions for the Delivery of Biotech Drugs – a Review of Drug Nanocrystal Technology and Lipid Nanoparticles. J. Biotechnol. 2004, 113, 151-170.

75. Choi, J.-Y.; Yoo, J. Y.; Kwak, H.-S.; Uk Nam, B.; Lee, J. Role of Polymeric Stabilizers for Drug Nanocrystal Dispersions. Curr. Appl. Phys. 2005, 5, 472-474.

76. Keck, C. M.; Muller, R. H. Drug Nanocrystals of Poorly Soluble Drugs Produced by High Pressure Homogenisation. Eur. J. Pharm. Biopharm. 2006, 62, 3-16.

77. Sylvestre, J.-P.; Tang, M.-C.; Furtos, A.; Leclair, G.; Meunier, M.; Leroux, J.-C. Nanonization of Megestrol Acetate by Laser Fragmentation in Aqueous Milieu. J. Control. Release 2011, 149, 273-280.

78. Chattopadhyay, P.; Gupta, R. B. Production of Antibiotic Nanoparticles Using Supercritical CO2 as Antisolvent with Enhanced Mass Transfer. Ind. Eng. Chem. Res. 2001, 40, 3530-3539.

79. Chattopadhyay, P.; Gupta, R. B. Production of Griseofulvin Nanoparticles Using Supercritical CO2 Antisolvent with Enhanced Mass Transfer. Int. J. Pharm. 2001, 228, 19-31.

80. Date, A. A.; Patravale, V. B. Current Strategies for Engineering Drug Nanoparticles. Curr. Opin. Colloid Interface Sci. 2004, 9, 222-235.

81. Merisko-Liversidge, E. M.; Liversidge, G. G. Drug Nanoparticles: Formulating Poorly Water-Soluble Compounds. Toxicol. Pathol. 2008, 36, 43-48.

82. Merisko-Liversidge, E.; Liversidge, G. G.; Cooper, E. R. Nanosizing: A Formulation Approach for Poorly-water-soluble Compounds. Eur. J. Pharm. Sci. 2003, 18, 113-120.

Page 136: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

133

83. Van Eerdenbrugh, B.; Van den Mooter, G.; Augustijns, P. Top-down Production of Drug Nanocrystals: Nanosuspension Stabilization, Miniaturization and Transformation Into Solid Products. Int. J. Pharm. 2008, 364, 64-75.

84. Cerdeira, A. M.; Werner, I. A.; Mazzotti, M.; Gander, B. Simultaneous quantification of polymeric and surface active stabilizers of nanosuspensions by using near-infrared spectroscopy. Drug Dev. Ind. Pharm. 2012, 38, 1360-1370.

85. Cerdeira, A. M.; Mazzotti, M.; Gander, B. Miconazole Nanosuspensions: Influence of Formulation Variables on Particle Size Reduction and Physical Stability. Int. J. Pharm. 2010, 396, 210-218.

86. Ploehn, H. J.; Russel, W. B., Interactions Between Colloidal Particles and Soluble Polymers. In Advances in Chemical Engineering; James, W., Eds.; Academic Press: San Diego, 1990, pp 137-228.

87. Fuhrmann, K.; Połomska, A.; Aeberli, C.; Castagner, B.; Gauthier, M. A.; Leroux, J.-C. Modular Design of Redox-Responsive Stabilizers for Nanocrystals. ACS Nano 2013, 7, 8243-8250.

88. Shchekin, A. K.; Rusanov, A. I. Generalization of the Gibbs–Kelvin–Köhler and Ostwald–Freundlich Equations for a Liquid Film on a Soluble Nanoparticle. J. Chem. Phys. 2008, 129, 154116.

89. Hollis, C. P.; Weiss, H. L.; Leggas, M.; Evers, B. M.; Gemeinhart, R. A.; Li, T. Biodistribution and Bioimaging Studies of Hybrid Paclitaxel Nanocrystals: Lessons Learned of the EPR Effect and Image-guided Drug Delivery. J. Control. Release 2013, 172, 12-21.

90. Moreno-Aspitia, A.; Perez, E. A. North Central Cancer Treatment Group N0531: Phase II Trial of Weekly Albumin-Bound Paclitaxel (ABI-007; Abraxane®) in Combination with Gemcitabine in Patients with Metastatic Breast Cancer. Clin. Breast Cancer 2005, 6, 361-364.

91. Liu, F.; Park, J.-Y.; Zhang, Y.; Conwell, C.; Liu, Y.; Bathula, S. R.; Huang, L. Targeted Cancer Therapy with Novel High Drug-loading Nanocrystals. J. Pharm. Sci. 2010, 99, 3542-3551.

92. Shegokar, R.; Singh, K. K. Surface Modified Nevirapine Nanosuspensions for Viral Reservoir Targeting: In Vitro and In Vivo Evaluation. Int. J. Pharm. 2011, 421, 341-352.

93. Zhang, H.; Hollis, C. P.; Zhang, Q.; Li, T. Preparation and Antitumor Study of Camptothecin Nanocrystals. Int. J. Pharm. 2011, 415, 293-300.

94. Ben Zirar, S.; Astier, A.; Muchow, M.; Gibaud, S. Comparison of Nanosuspensions and Hydroxypropyl-β-cyclodextrin Complex of Melarsoprol: Pharmacokinetics and Tissue Distribution in Mice. Eur. J. Pharm. Biopharm. 2008, 70, 649-656.

95. Vergara, D.; Bellomo, C.; Zhang, X.; Vergaro, V.; Tinelli, A.; Lorusso, V.; Rinaldi, R.; Lvov, Y. M.; Leporatti, S.; Maffia, M. Lapatinib/Paclitaxel

Page 137: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

134

Polyelectrolyte Nanocapsules for Overcoming Multidrug Resistance in Ovarian Cancer. Nanomedicine: NBM 2012, 8, 891-899.

96. Cho, Y. W.; Lee, J.; Lee, S. C.; Huh, K. M.; Park, K. Hydrotropic Agents for Study of In Vitro Paclitaxel Release from Polymeric Micelles. J. Control. Release 2004, 97, 249-257.

97. Kadiu, I.; Nowacek, A.; McMillan, J.; Gendelman, H. E. Macrophage Endocytic Trafficking of Antiretroviral Nanoparticles. Nanomedicine 2011, 6, 975-994.

98. Nowacek, A.; McMillan, J.; Miller, R.; Anderson, A.; Rabinow, B.; Gendelman, H. Nanoformulated Antiretroviral Drug Combinations Extend Drug Release and Antiretroviral Responses in HIV-1-Infected Macrophages: Implications for NeuroAIDS Therapeutics. J Neuroimmune Pharmacol 2010, 5, 592-601.

99. Nowacek, A. S.; Balkundi, S.; McMillan, J.; Roy, U.; Martinez-Skinner, A.; Mosley, R. L.; Kanmogne, G.; Kabanov, A. V.; Bronich, T.; Gendelman, H. E. Analyses of Nanoformulated Antiretroviral Drug Charge, Size, Shape and Content for Uptake, Drug Release and Antiviral Activities in Human Monocyte-derived Macrophages. J. Control. Release 2011, 150, 204-211.

100. Bui, D. T.; Nicolas, J.; Maksimenko, A.; Desmaele, D.; Couvreur, P. Multifunctional Squalene-based Prodrug Nanoparticles for Targeted Cancer Therapy. Chem. Commun. 2014, 50, 5336-5338.

101. Mouton, J. W.; van Peer, A.; de Beule, K.; Van Vliet, A.; Donnelly, J. P.; Soons, P. A. Pharmacokinetics of Itraconazole and Hydroxyitraconazole in Healthy Subjects After Single and Multiple Doses of a Novel Formulation. Antimicrob. Agents Chemother. 2006, 50, 4096-4102.

102. Tian, X.; Li, H.; Zhang, D.; Shen, J.; Jia, L.; Zheng, D.; Liu, G.; Hao, L.; Shen, Y.; Zhang, Q. Parenteral nanosuspension of a novel synthesized antitumor candidate: Investigation of tissue biodistributions and plasma pharmacokinetics. Colloids Surf. Physicochem. Eng. Aspects 2013, 436, 868-872.

103. Sharma, P.; Zujovic, Z. D.; Bowmaker, G. A.; Denny, W. A.; Garg, S. Evaluation of a Crystalline Nanosuspension: Polymorphism, Process Induced Transformation and In Vivo Studies. Int. J. Pharm. 2011, 408, 138-151.

104. Tian, X.; Li, H.; Zhang, D.; Liu, G.; Jia, L.; Zheng, D.; Shen, J.; Shen, Y.; Zhang, Q. Nanosuspension for parenteral delivery of a p-terphenyl derivative: Preparation, characteristics and pharmacokinetic studies. Colloids Surf. B. Biointerfaces 2013, 108, 29-33.

105. Ganta, S.; Paxton, J. W.; Baguley, B. C.; Garg, S. Formulation and Pharmacokinetic Evaluation of an Asulacrine Nanocrystalline Suspension for Intravenous Delivery. Int. J. Pharm. 2009, 367, 179-186.

106. Gao, L.; Zhang, D.; Chen, M.; Duan, C.; Dai, W.; Jia, L.; Zhao, W. Studies on Pharmacokinetics and Tissue Distribution of Oridonin Nanosuspensions. Int. J. Pharm. 2008, 355, 321-327.

Page 138: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

135

107. Rabinow, B.; Kipp, J.; Papadopoulos, P.; Wong, J.; Glosson, J.; Gass, J.; Sun, C.-S.; Wielgos, T.; White, R.; Cook, C., et al. Itraconazole IV Nanosuspension Enhances Efficacy through Altered Pharmacokinetics in the Rat. Int. J. Pharm. 2007, 339, 251-260.

108. Shegokar, R.; Müller, R. H. Nanocrystals: Industrially feasible multifunctional formulation technology for poorly soluble actives. Int. J. Pharm. 2010, 399, 129-139.

109. Sanchez, V. C.; Pietruska, J. R.; Miselis, N. R.; Hurt, R. H.; Kane, A. B. Biopersistence and potential adverse health impacts of fibrous nanomaterials: what have we learned from asbestos? WIREs Nanomed Nanobiotechnol 2009, 1, 511-529.

110. Dintaman, J. M.; Silverman, J. A. Inhibition of P-Glycoprotein by D-α-Tocopheryl Polyethylene Glycol 1000 Succinate (TPGS). Pharm. Res. 1999, 16, 1550-1556.

111. Zhang, H.; Wang, X.; Dai, W.; Gemeinhart, R. A.; Zhang, Q.; Li, T. Pharmacokinetics and Treatment Efficacy of Camptothecin Nanocrystals on Lung Metastasis. Mol. Pharm. 2013, 11, 226-233.

112. Karmali, P. P.; Kotamraju, V. R.; Kastantin, M.; Black, M.; Missirlis, D.; Tirrell, M.; Ruoslahti, E. Targeting of Albumin-embedded Paclitaxel Nanoparticles to Tumors. Nanomedicine: NBM 2009, 5, 73-82.

113. Gardner, E. R.; Dahut, W. L.; Scripture, C. D.; Jones, J.; Aragon-Ching, J. B.; Desai, N.; Hawkins, M. J.; Sparreboom, A.; Figg, W. D. Randomized Crossover Pharmacokinetic Study of Solvent-Based Paclitaxel and nab-Paclitaxel. Clin. Cancer. Res. 2008, 14, 4200-4205.

114. Deng, J.; Huang, L.; Liu, F. Understanding the Structure and Stability of Paclitaxel Nanocrystals. Int. J. Pharm. 2010, 390, 242-249.

115. Parrish, B.; Breitenkamp, R. B.; Emrick, T. PEG- and Peptide-Grafted Aliphatic Polyesters by Click Chemistry. J. Am. Chem. Soc. 2005, 127, 7404-7410.

116. Kim, S.; Lee, J. Folate-targeted Drug-delivery Systems Prepared by Nano-comminution. Drug Dev. Ind. Pharm. 2011, 37, 131-138.

117. Trachootham, D.; Alexandre, J.; Huang, P. Targeting Cancer Cells by ROS-mediated Mechanisms: a Radical Therapeutic Approach? Nat. Rev. Drug Discov. 2009, 8, 579-591.

118. Lin, M. T.; Beal, M. F. Mitochondrial Dysfunction and Oxidative Stress in Neurodegenerative Diseases. Nature 2006, 443, 787-795.

119. Lu, Y.; Wang, Z.-h.; Li, T.; McNally, H.; Park, K.; Sturek, M. Development and Evaluation of Transferrin-stabilized Paclitaxel Nanocrystal Formulation. J. Control. Release 2014, 176, 76-85.

120. Ai, H.; Jones, S. A.; de Villiers, M. M.; Lvov, Y. M. Nano-encapsulation of Furosemide Microcrystals for Controlled Drug Release. J. Control. Release 2003, 86, 59-68.

Page 139: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

136

121. Qiu, X.; Leporatti, S.; Donath, E.; Möhwald, H. Studies on the Drug Release Properties of Polysaccharide Multilayers Encapsulated Ibuprofen Microparticles. Langmuir 2001, 17, 5375-5380.

122. Sukhorukov, G. B.; Brumen, M.; Donath, E.; Möhwald, H. Hollow Polyelectrolyte Shells: Exclusion of Polymers and Donnan Equilibrium. J. Phys. Chem. B 1999, 103, 6434-6440.

123. Shutava, T. G.; Pattekari, P. P.; Arapov, K. A.; Torchilin, V. P.; Lvov, Y. M. Architectural Layer-by-layer Assembly of Drug Nanocapsules with PEGylated Polyelectrolytes. Soft Matter 2012, 8, 9418-9427.

124. Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.; Terada, Y.; Kano, M. R.; Miyazono, K.; Uesaka, M., et al. Accumulation of Sub-100 nm Polymeric Micelles in Poorly Permeable Tumours Depends on Size. Nat. Nanotechnol. 2011, 6, 815-823.

125. Wong, C.; Stylianopoulos, T.; Cui, J.; Martin, J.; Chauhan, V. P.; Jiang, W.; Popović, Z.; Jain, R. K.; Bawendi, M. G.; Fukumura, D. Multistage Nanoparticle Delivery System for Deep Penetration into Tumor Tissue. P. Natl. Acad. Sci. USA 2011, 108, 2426-2431.

126. Arias, J. L.; Reddy, L. H.; Couvreur, P. Magnetoresponsive Squalenoyl Gemcitabine Composite Nanoparticles for Cancer Active Targeting. Langmuir 2008, 24, 7512-7519.

127. Sugahara, K. N.; Teesalu, T.; Karmali, P. P.; Kotamraju, V. R.; Agemy, L.; Greenwald, D. R.; Ruoslahti, E. Coadministration of a Tumor-Penetrating Peptide Enhances the Efficacy of Cancer Drugs. Science 2010, 328, 1031-1035.

128. Aggarwal, P.; Hall, J. B.; McLeland, C. B.; Dobrovolskaia, M. A.; McNeil, S. E. Nanoparticle Interaction with Plasma Proteins as it Relates to Particle Biodistribution, Biocompatibility and Therapeutic Efficacy. Adv. Drug Del. Rev. 2009, 61, 428-437.

129. Lindfors, L.; Forssen, S.; Skantze, P.; Skantze, U.; Zackrisson, A.; Olsson, U. Amorphous Drug Nanosuspensions. 2. Experimental Determination of Bulk Monomer Concentrations. Langmuir 2005, 22, 911-916.

130. Meerum Terwogt, J. M.; Nuijen, B.; Ten Bokkel Huinink, W. W.; Beijnen, J. H. Alternative Formulations of Paclitaxel. Cancer Treat. Rev. 1997, 23, 87-95.

131. Fahr, A.; Liu, X. Drug Delivery Strategies for Poorly Water-soluble Drugs. Expert Opin. Drug Deliv. 2007, 4, 403-416.

132. Chan, J. M.; Zhang, L.; Tong, R.; Ghosh, D.; Gao, W.; Liao, G.; Yuet, K. P.; Gray, D.; Rhee, J.-W.; Cheng, J., et al. Spatiotemporal Controlled Delivery of Nanoparticles to Injured Vasculature. P. Natl. Acad. Sci. USA 2010, 107, 2213-2218.

133. Hayashi, Y.; Skwarczynski, M.; Hamada, Y.; Sohma, Y.; Kimura, T.; Kiso, Y. A Novel Approach of Water-Soluble Paclitaxel Prodrug with No Auxiliary and No Byproduct: Design and Synthesis of Isotaxel. J. Med. Chem. 2003, 46, 3782-3784.

Page 140: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

137

134. Van, S.; Das, S. K.; Wang, X.; Feng, Z.; Jin, Y.; Hou, Z.; Chen, F.; Pham, A.; Jiang, N.; Howell, S. B., et al. Synthesis, Characterization, and Biological Evaluation of Poly(L-γ-glutamyl-glutamine)-paclitaxel Nanoconjugate. Int. J. Nanomed. 2010, 5, 825-837.

135. Dosio, F.; Reddy, L. H.; Ferrero, A.; Stella, B.; Cattel, L.; Couvreur, P. Novel Nanoassemblies Composed of Squalenoyl−Paclitaxel Derivatives: Synthesis, Characterization, and Biological Evaluation. Bioconjugate Chem. 2010, 21, 1349-1361.

136. Zhang, J. A.; Anyarambhatla, G.; Ma, L.; Ugwu, S.; Xuan, T.; Sardone, T.; Ahmad, I. Development and Characterization of a Novel Cremophor® EL Free Liposome-based Paclitaxel (LEP-ETU) Formulation. Eur. J. Pharm. Biopharm. 2005, 59, 177-187.

137. Liu, Z.; Chen, K.; Davis, C.; Sherlock, S.; Cao, Q.; Chen, X.; Dai, H. Drug Delivery with Carbon Nanotubes for In Vivo Cancer Treatment. Cancer Res. 2008, 68, 6652-6660.

138. Brewster, M. E.; Loftsson, T. Cyclodextrins as Pharmaceutical Solubilizers. Adv. Drug Del. Rev. 2007, 59, 645-666.

139. Le Garrec, D.; Gori, S.; Luo, L.; Lessard, D.; Smith, D. C.; Yessine, M.-A.; Ranger, M.; Leroux, J.-C. Poly(N-vinylpyrrolidone)-block-poly(D,L-lactide) as a New Polymeric Solubilizer for Hydrophobic Anticancer Drugs: In Vitro and In Vivo Evaluation. J. Control. Release 2004, 99, 83-101.

140. Li, C.; Wallace, S. Polymer-drug Conjugates: Recent Development in Clinical Oncology. Adv. Drug Del. Rev. 2008, 60, 886-898.

141. Kogan, A.; Garti, N. Microemulsions as Transdermal Drug Delivery Vehicles. Adv. Colloid Interface Sci. 2006, 123-126, 369-385.

142. Strickley, R. G. Solubilizing Excipients in Oral and Injectable Formulations. Pharm. Res. 2004, 21, 201-230.

143. Chiappetta, D. A.; Sosnik, A. Poly(ethylene oxide)-poly(propylene oxide) Block Copolymer Micelles as Drug Delivery Agents: Improved Hydrosolubility, Stability and Bioavailability of Drugs. Eur. J. Pharm. Biopharm. 2007, 66, 303-317.

144. Soussan, E.; Cassel, S.; Blanzat, M.; Rico-Lattes, I. Drug Delivery by Soft Matter: Matrix and Vesicular Carriers. Angew. Chem. Int. Ed. 2009, 48, 274-288.

145. Serajuddin, A. T. M. Solid Dispersion of Poorly Water-soluble Drugs: Early Promises, Subsequent Problems, and Recent Breakthroughs. J. Pharm. Sci. 1999, 88, 1058-1066.

146. Ibrahim, N. K.; Desai, N.; Legha, S.; Soon-Shiong, P.; Theriault, R. L.; Rivera, E.; Esmaeli, B.; Ring, S. E.; Bedikian, A.; Hortobagyi, G. N., et al. Phase I and Pharmacokinetic Study of ABI-007, a Cremophor-free, Protein-stabilized, Nanoparticle Formulation of Paclitaxel. Clin. Cancer. Res. 2002, 8, 1038-1044.

Page 141: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

138

147. Turner, J. L.; Chen, Z.; Wooley, K. L. Regiochemical Functionalization of a Nanoscale Cage-like Structure: Robust Core-shell Nanostructures Crafted as Vessels for Selective Uptake and Release of Small and Large Guests. J. Control. Release 2005, 109, 189-202.

148. Lenoir, S.; Riva, R.; Lou, X.; Detrembleur, C.; Jérôme, R.; Lecomte, P. Ring-Opening Polymerization of α-Chloro-ε-caprolactone and Chemical Modification of Poly(α-chloro-ε-caprolactone) by Atom Transfer Radical Processes. Macromolecules 2004, 37, 4055-4061.

149. Kim, M. S.; Seo, K. S.; Khang, G.; Lee, H. B. Ring-Opening Polymerization of ε-Caprolactone by Poly(ethylene glycol) by an Activated Monomer Mechanism. Macromol. Rapid Commun. 2005, 26, 643-648.

150. Ghebeh, H.; Handa-Corrigan, A.; Butler, M. Development of an Assay for the Measurement of the Surfactant Pluronic F-68 in Mammalian Cell Culture Medium. Anal. Biochem. 1998, 262, 39-44.

151. Letchford, K.; Liggins, R.; Wasan, K. M.; Burt, H. In Vitro Human Plasma Distribution of Nanoparticulate Paclitaxel is Dependent on the Physicochemical Properties of Poly(ethylene glycol)-block-poly(caprolactone) Nanoparticles. Eur. J. Pharm. Biopharm. 2009, 71, 196-206.

152. Torchilin, V. P. Multifunctional Nanocarriers. Adv. Drug Del. Rev. 2006, 58, 1532-1555.

153. Ahmed, F.; Discher, D. E. Self-porating Polymersomes of PEG-PLA and PEG-PCL: Hydrolysis-triggered Controlled Release Vesicles. J. Control. Release 2004, 96, 37-53.

154. Riva, R.; Schmeits, S.; Stoffelbach, F.; Jérôme, C.; Jérôme, R.; Lecomte, P. Combination of Ring-opening Polymerization and "Click" Chemistry towards Functionalization of Aliphatic Polyesters. Chem. Commun. 2005, 42, 5334-5336.

155. Merisko-Liversidge, E.; Sarpotdar, P.; Bruno, J.; Hajj, S.; Wei, L.; Peltier, N.; Rake, J.; Shaw, J. M.; Pugh, S.; Polin, L., et al. Formulation and Antitumor Activity Evaluation of Nanocrystalline Suspensions of Poorly Soluble Anticancer Drugs. Pharm. Res. 1996, 13, 272-278.

156. Van Eerdenbrugh, B.; Vermant, J.; Martens, J. A.; Froyen, L.; Humbeeck, J. V.; Van den Mooter, G.; Augustijns, P. Solubility Increases Associated with Crystalline Drug Nanoparticles: Methodologies and Significance. Mol. Pharm. 2010, 7, 1858-1870.

157. Budijono, S. J.; Russ, B.; Saad, W.; Adamson, D. H.; Prud’homme, R. K. Block Copolymer Surface Coverage on Nanoparticles. Colloids Surf. Physicochem. Eng. Aspects 2010, 360, 105-110.

158. Pasche, S.; De Paul, S. M.; Vörös, J.; Spencer, N. D.; Textor, M. Poly(L-lysine)-graft-poly(ethylene glycol) Assembled Monolayers on Niobium Oxide Surfaces: A Quantitative Study of the Influence of Polymer Interfacial Architecture on Resistance

Page 142: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

139

to Protein Adsorption by ToF-SIMS and in Situ OWLS. Langmuir 2003, 19, 9216-9225.

159. Turner, J. L.; Wooley, K. L. Nanoscale Cage-like Structures Derived from Polyisoprene-Containing Shell Cross-linked Nanoparticle Templates. Nano Lett. 2004, 4, 683-688.

160. Horn, D.; Rieger, J. Organic Nanoparticles in the Aqueous Phase—Theory, Experiment, and Use. Angew. Chem. Int. Ed. 2001, 40, 4330-4361.

161. Lindfors, L.; Skantze, P.; Skantze, U.; Rasmusson, M.; Zackrisson, A.; Olsson, U. Amorphous Drug Nanosuspensions. 1. Inhibition of Ostwald Ripening. Langmuir 2005, 22, 906-910.

162. Zhao, R.; Hollis, C. P.; Zhang, H.; Sun, L.; Gemeinhart, R. A.; Li, T. Hybrid Nanocrystals: Achieving Concurrent Therapeutic and Bioimaging Functionalities Toward Solid Tumors. Mol. Pharm. 2011, 8, 1985–1991.

163. Venkataraman, S.; Hedrick, J. L.; Ong, Z. Y.; Yang, C.; Ee, P. L. R.; Hammond, P. T.; Yang, Y. Y. The Effects of Polymeric Nanostructure Shape on Drug Delivery. Adv. Drug Del. Rev. 2011, 63, 1228-1246.

164. Gaucher, G.; Asahina, K.; Wang, J.; Leroux, J.-C. Effect of Poly(N-vinyl-pyrrolidone)-block-poly(D,L-lactide) as Coating Agent on the Opsonization, Phagocytosis, and Pharmacokinetics of Biodegradable Nanoparticles. Biomacromolecules 2009, 10, 408-416.

165. Cormode, D. P.; Skajaa, G. O.; Delshad, A.; Parker, N.; Jarzyna, P. A.; Calcagno, C.; Galper, M. W.; Skajaa, T.; Briley-Saebo, K. C.; Bell, H. M., et al. A Versatile and Tunable Coating Strategy Allows Control of Nanocrystal Delivery to Cell Types in the Liver. Bioconjugate Chem. 2011, 22, 353-361.

166. Schweiger, C.; Pietzonka, C.; Heverhagen, J.; Kissel, T. Novel Magnetic Iron Oxide Nanoparticles Coated with Poly(ethylene imine)-g-poly(ethylene glycol) for Potential Biomedical Application: Synthesis, Stability, Cytotoxicity and MR Imaging. Int. J. Pharm. 2011, 408, 130-137.

167. Romberg, B.; Hennink, W.; Storm, G. Sheddable Coatings for Long-Circulating Nanoparticles. Pharm. Res. 2008, 25, 55-71.

168. Sankaranarayanan, J.; Mahmoud, E. A.; Kim, G.; Morachis, J. M.; Almutairi, A. Multiresponse Strategies To Modulate Burst Degradation and Release from Nanoparticles. ACS Nano 2010, 4, 5930-5936.

169. Knipe, J. M.; Peters, J. T.; Peppas, N. A. Theranostic Agents for Intracellular Gene Delivery with Spatiotemporal Imaging. Nano Today 2013, 8, 21-38.

170. Motion, J. P. M.; Nguyen, J.; Szoka, F. C. Phosphatase-Triggered Fusogenic Liposomes for Cytoplasmic Delivery of Cell-Impermeable Compounds. Angew. Chem. Int. Ed. 2012, 51, 9047-9051.

Page 143: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

140

171. Preston, T. J.; Abadi, A.; Wilson, L.; Singh, G. Mitochondrial Contributions to Cancer Cell Physiology: Potential for Drug Development. Adv. Drug Del. Rev. 2001, 49, 45-61.

172. de Gracia Lux, C.; Joshi-Barr, S.; Nguyen, T.; Mahmoud, E.; Schopf, E.; Fomina, N.; Almutairi, A. Biocompatible Polymeric Nanoparticles Degrade and Release Cargo in Response to Biologically Relevant Levels of Hydrogen Peroxide. J. Am. Chem. Soc. 2012, 134, 15758-15764.

173. Broaders, K. E.; Grandhe, S.; Fréchet, J. M. J. A Biocompatible Oxidation-Triggered Carrier Polymer with Potential in Therapeutics. J. Am. Chem. Soc. 2010, 133, 756-758.

174. Khutoryanskiy, V. V.; Tirelli, N. Oxidation-responsiveness of Nanomaterials for Targeting Inflammatory Reactions. Pure Appl. Chem. 2008, 80, 1703-1718.

175. Allen, B. L.; Johnson, J. D.; Walker, J. P. Encapsulation and Enzyme-Mediated Release of Molecular Cargo in Polysulfide Nanoparticles. ACS Nano 2011, 5, 5263-5272.

176. Rehor, A.; Hubbell, J. A.; Tirelli, N. Oxidation-Sensitive Polymeric Nanoparticles. Langmuir 2004, 21, 411-417.

177. Yu, S. S.; Koblin, R. L.; Zachman, A. L.; Perrien, D. S.; Hofmeister, L. H.; Giorgio, T. D.; Sung, H.-J. Physiologically Relevant Oxidative Degradation of Oligo(proline) Cross-Linked Polymeric Scaffolds. Biomacromolecules 2011, 12, 4357-4366.

178. Gao, W.; Langer, R.; Farokhzad, O. C. Poly(ethylene glycol) with Observable Shedding. Angew. Chem. Int. Ed. 2010, 49, 6567-6571.

179. Gauthier, M. A.; Gibson, M. I.; Klok, H.-A. Synthesis of Functional Polymers by Post-Polymerization Modification. Angew. Chem. Int. Ed. 2009, 48, 48-58.

180. Hoogenboom, R. Thiol–Yne Chemistry: A Powerful Tool for Creating Highly Functional Materials. Angew. Chem. Int. Ed. 2010, 49, 3415-3417.

181. Napoli, A.; Valentini, M.; Tirelli, N.; Muller, M.; Hubbell, J. A. Oxidation-responsive Polymeric Vesicles. Nat. Mater. 2004, 3, 183-189.

182. Szela, S.; Avtges, P.; Valluzzi, R.; Winkler, S.; Wilson, D.; Kirschner, D.; Kaplan, D. L. Reduction−Oxidation Control of β-Sheet Assembly in Genetically Engineered Silk. Biomacromolecules 2000, 1, 534-542.

183. Dado, G. P.; Gellman, S. H. Redox Control of Secondary Structure in a Designed Peptide. J. Am. Chem. Soc. 1993, 115, 12609-12610.

184. Carampin, P.; Lallana, E.; Laliturai, J.; Carroccio, S. C.; Puglisi, C.; Tirelli, N. Oxidant-Dependent REDOX Responsiveness of Polysulfides. Macromol. Chem. Phys. 2012, 213, 2052-2061.

Page 144: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

141

185. Abraham, R. J.; Byrne, J. J.; Griffiths, L. 1H Chemical Shifts in NMR. Part 27: Proton Chemical Shifts in Sulfoxides and Sulfones and the Magnetic Anisotropy, Electric Field and Steric Effects of the SO Bond. Magn. Reson. Chem. 2008, 46, 667-675.

186. Mahmoud, E. A.; Sankaranarayanan, J.; Morachis, J. M.; Kim, G.; Almutairi, A. Inflammation Responsive Logic Gate Nanoparticles for the Delivery of Proteins. Bioconjugate Chem. 2011, 22, 1416-1421.

187. Alexandre, J.; Batteux, F.; Nicco, C.; Chéreau, C.; Laurent, A.; Guillevin, L.; Weill, B.; Goldwasser, F. Accumulation of Hydrogen Peroxide is an Early and Crucial Step for Paclitaxel-induced Cancer Cell Death Both In Vitro and In Vivo. Int. J. Cancer 2006, 119, 41-48.

188. Alexandre, J.; Hu, Y.; Lu, W.; Pelicano, H.; Huang, P. Novel Action of Paclitaxel against Cancer Cells: Bystander Effect Mediated by Reactive Oxygen Species. Cancer Res. 2007, 67, 3512-3517.

189. Duncan, R.; Gaspar, R. Nanomedicine(s) under the Microscope. Mol. Pharm. 2011, 8, 2101-2141.

190. Basiruddin, S. K.; Saha, A.; Pradhan, N.; Jana, N. R. Advances in Coating Chemistry in Deriving Soluble Functional Nanoparticle. J. Phys. Chem. C 2010, 114, 11009-11017.

191. Yu, S.; Scherer, R.; Ortega, R.; Bell, C.; O'Neil, C.; Hubbell, J.; Giorgio, T. Enzymatic- and Temperature-sensitive Controlled Release of Ultrasmall Superparamagnetic Iron Oxides (USPIOs). J. Nanobiotechnol. 2011, 9, 7.

192. Mailänder, V.; Landfester, K. Interaction of Nanoparticles with Cells. Biomacromolecules 2009, 10, 2379-2400.

193. Dubertret, B.; Skourides, P.; Norris, D. J.; Noireaux, V.; Brivanlou, A. H.; Libchaber, A. In Vivo Imaging of Quantum Dots Encapsulated in Phospholipid Micelles. Science 2002, 298, 1759-1762.

194. Schottenfeld, D.; Beebe-Dimmer, J. L.; Buffler, P. A.; Omenn, G. S. Current Perspective on the Global and United States Cancer Burden Attributable to Lifestyle and Environmental Risk Factors. Annu. Rev. Public Health 2013, 34, 97-117.

195. Gatta, G.; Mallone, S.; van der Zwan, J. M.; Trama, A.; Siesling, S.; Capocaccia, R.; Group, t. E. W. Cancer Prevalence Estimates in Europe at the Beginning of 2000. Ann. Oncol. 2013, 24, 1660-1666.

196. Maddams, J.; Utley, M.; Moller, H. Projections of Cancer Prevalence in the United Kingdom, 2010-2040. Br. J. Cancer 2012, 107, 1195-1202.

197. Mao, J. J.; Bowman, M. A.; Stricker, C. T.; DeMichele, A.; Jacobs, L.; Chan, D.; Armstrong, K. Delivery of Survivorship Care by Primary Care Physicians: The Perspective of Breast Cancer Patients. J. Clin. Oncol. 2009, 27, 933-938.

Page 145: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

142

198. Jefford, M.; Rowland, J.; Grunfeld, E.; Richards, M.; Maher, J.; Glaser, A. Implementing Improved Post-treatment Care for Cancer Survivors in England, with Reflections from Australia, Canada and the USA. Br. J. Cancer 2013, 108, 14-20.

199. Danhier, F.; Feron, O.; Préat, V. To Exploit the Tumor Microenvironment: Passive and Active Tumor Targeting of Nanocarriers for Anti-cancer Drug Delivery. J. Control. Release 2010, 148, 135-146.

200. Hrkach, J.; Von Hoff, D.; Ali, M. M.; Andrianova, E.; Auer, J.; Campbell, T.; De Witt, D.; Figa, M.; Figueiredo, M.; Horhota, A., et al. Preclinical Development and Clinical Translation of a PSMA-Targeted Docetaxel Nanoparticle with a Differentiated Pharmacological Profile. Sci. Transl. Med. 2012, 4, 128ra139.

201. Chen, T.; Öçsoy, I.; Yuan, Q.; Wang, R.; You, M.; Zhao, Z.; Song, E.; Zhang, X.; Tan, W. One-Step Facile Surface Engineering of Hydrophobic Nanocrystals with Designer Molecular Recognition. J. Am. Chem. Soc. 2012, 134, 13164-13167.

202. Ferrari, M. Cancer Nanotechnology: Opportunities and Challenges. Nat. Rev. Cancer 2005, 5, 161-171.

203. Nagamitsu, A.; Greish, K.; Maeda, H. Elevating Blood Pressure as a Strategy to Increase Tumor-targeted Delivery of Macromolecular Drug SMANCS: Cases of Advanced Solid Tumors. Jap. J. Clin. Oncol. 2009, 39, 756-766.

204. Lammers, T.; Kiessling, F.; Hennink, W. E.; Storm, G. Drug Targeting to Tumors: Principles, Pitfalls and (Pre-) Clinical Progress. J. Control. Release 2012, 161, 175-187.

205. Jain, R. K.; Stylianopoulos, T. Delivering Nanomedicine to Solid Tumors. Nat. Rev. Clin. Oncol. 2010, 7, 653-664.

206. O’Brien, M. E. R.; Wigler, N.; Inbar, M.; Rosso, R.; Grischke, E.; Santoro, A.; Catane, R.; Kieback, D. G.; Tomczak, P.; Ackland, S. P., et al. Reduced Cardiotoxicity and Comparable Efficacy in a Phase III Trial of Pegylated Liposomal Doxorubicin HCl (CAELYX™/Doxil®) versus Conventional Doxorubicin for First-line Treatment of Metastatic Breast Cancer. Ann. Oncol. 2004, 15, 440-449.

207. Northfelt, D. W.; Dezube, B. J.; Thommes, J. A.; Miller, B. J.; Fischl, M. A.; Friedman-Kien, A.; Kaplan, L. D.; Du Mond, C.; Mamelok, R. D.; Henry, D. H. Pegylated-liposomal Doxorubicin versus Doxorubicin, Bleomycin, and Vincristine in the Treatment of AIDS-related Kaposi's Sarcoma: Results of a Randomized Phase III Clinical Trial. J. Clin. Oncol. 1998, 16, 2445-2451.

208. Arrieta, O.; Medina, L. A.; Estrada-Lobato, E.; Hernandez-Pedro, N.; Villanueva-Rodriguez, G.; Martinez-Barrera, L.; Macedo, E. O.; Lopez-Rodriguez, V.; Motola-Kuba, D.; Corona-Cruz, J. F. First-line Chemotherapy with Liposomal Doxorubicin plus Cisplatin for Patients with Advanced Malignant Pleural Mesothelioma: Phase II Trial. Br. J. Cancer 2012, 106, 1027-1032.

209. Bae, K. H.; Choi, S. H.; Park, S. Y.; Lee, Y.; Park, T. G. Thermosensitive Pluronic Micelles Stabilized by Shell Cross-Linking with Gold Nanoparticles. Langmuir 2006, 22, 6380-6384.

Page 146: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

143

210. Suresh, A. K.; Weng, Y.; Li, Z.; Zerda, R.; Van Haute, D.; Williams, J. C.; Berlin, J. M. Matrix metalloproteinase-triggered denuding of engineered gold nanoparticles for selective cell uptake. Journal of Materials Chemistry B 2013, 1, 2341-2349.

211. Alconcel, S. N. S.; Baas, A. S.; Maynard, H. D. FDA-approved Poly(ethylene glycol)-protein Conjugate Drugs. Polym. Chem. 2011, 2, 1442-1448.

Page 147: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted

 

   

Page 148: Rights / License: Research Collection In Copyright - Non ...8756/eth-8756-02.pdfpatient’s well-being, because the pharmacological action of the toxic anticancer drugs is often untargeted