drug delivery of paclitaxel for an intraperitoneal chemotherapy drug

196
Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug delivery of paclitaxel for an Drug delivery of paclitaxel for an intraperitoneal chemotherapy intraperitoneal chemotherapy Wim Bouquet Pharmacist Thesis submitted to obtain the degree of Doctor in Pharmaceutical Sciences 2009 Promoters: Prof. Dr. J. P. Remon and Prof. Dr. C. Vervaet Laboratory of Pharmaceutical Technology

Upload: others

Post on 10-Feb-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Drug delivery of paclitaxel for an

intraperitoneal chemotherapy

Drug delivery of paclitaxel for an Drug delivery of paclitaxel for an

intraperitoneal chemotherapyintraperitoneal chemotherapy

Wim Bouquet

Pharmacist

Thesis submitted to obtain the degree of

Doctor in Pharmaceutical Sciences

2009

Promoters: Prof. Dr. J. P. Remon and Prof. Dr. C. Vervaet

Laboratory of Pharmaceutical Technology

Page 2: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 3: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 4: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Faculty of Pharmaceutical Sciences

Drug delivery of paclitaxel for an intraperitoneal chemotherapy

Wim Bouquet

Pharmacist

Thesis submitted to obtain the degree of Doctor in Pharmaceutical Sciences

2009

Promoters:

Prof. Dr. J.P. Remon Prof. Dr. C. Vervaet

Laboratory of Pharmaceutical Technology

Page 5: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 6: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

The author and the promoters give the authorization to consult and to copy parts of this thesis

for personal use only. Any other use is limited by the Laws of Copyright, especially

concerning the obligation to refer to the source whenever results are cited from this thesis.

Ghent, March 1st, 2009

The Promoters The author

Prof. Dr. Jean Paul Remon Prof. Dr. Chris Vervaet Wim Bouquet

Page 7: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 8: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

“The books of the great scientists are gathering dust on the shelves of learned libraries. And

rightly so. The scientist addresses an infinitesimal audience of fellow composers. His message

is not devoid of universality but it’s universality is disemobdied and anonymous. While the

artist’s communication is linked forever with it’s original form, that of the scientist is

modified, amplified, fused with the ideas and results of others, and melts into the stream of

knowledge and ideas which forms our culture. The scientist has in common with the artist

only this: that he can find no better retreat from the world than his work and also no stronger

link with his world than his work.”

Max Delbrück, 1969 (Nobelprize acceptance speech)

Page 9: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 10: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

DANKWOORD_____________________________________

Nu mijn doctoraat geschreven is, houd ik er aan om de mensen te bedanken die mij

geholpen hebben tijdens dit doctoraat. Iemand helpen kan onder verschillende vormen

gebeuren: door effectief mee te helpen en de handen uit de mouwen te steken of gewoon om

te luisteren… . Aan iedereen die mij de afgelopen 6 jaar geholpen en gesteund heeft: Dank u

wel !!!!

Daarenboven wens ik ook een aantal mensen in het bijzonder te bedanken. Eerst en

vooral wil ik mijn beide promotoren, Prof J.P. Remon en Prof C. Vervaet, bedanken voor de

kans die ze me gegeven hebben om te doctoreren en een fantastische tijd te beleven. Alhoewel

mijn doctoraat hen soms wat vreemd was, hebben ze nooit nagelaten om mij enthousiast te

steunen en te helpen waar ze konden.

Voorts wil ik graag de verschillende laboratoria bedanken waar ik tijdens mijn

doctoraat gewerkt heb of mee samengewerkt heb:

- Het Department of Surgical Oncology: Dr. Wim Ceelen, Valentijn Van Parijs,

Isabelle Debergh en Nancy Vandamme.

- Het Laboratorium van Experimentele Cancerologie: Prof. M. Bracke , Prof M.

Mareel, Dr. Tom Boterberg, Daan Vandekerckhove, Martine De Mil, Arlette

Verspeelt en Georges De Bruyne en alle doctoraatstudenten (Ancy, Barbara,

Lara, Liesbeth, Maria, Olivier, Sofie, Veerle,…).

- Het Laboratorium voor NMR en Structuuranalyse: Prof. J. Martins, Dr. Bernd

Fritzinger en Katelijne Gheysen,

- Het Laboratorium voor Medicinale Chemie: Prof. S. Van Calenbergh en Dr. Ulrik

Hillaert.

- Het Department of Pediatric Hematology and Oncology: Dr. Katrien Swerts.

Page 11: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

- Het Laboratorium voor Radiofarmacie: Prof. F. De Vos, Phillippe Joye en alle

doctoraatstudenten.

- Medisip: Prof S. Staelens, Prof S. Vandenberghe, Steven Deleye en iedereen die

geholpen heeft bij het opnemen van de PET en MRI scans.

Verder wil ik nog de collega’s op het Laboratorium voor Farmaceutische Technologie

bedanken voor de afgelopen 6 jaar. Hier wil ik ook enkele mensen in het bijzonder bedanken:

Bruno en Katharine voor alle administratie, Ana, Christine en Thomas Q. voor het helpen bij

de scans, Els A. voor de hulp bij de statistiek van dit doctoraat, Brenda en Eveline voor de

hulp in mijn begindagen en Lieselotte voor het wisselen van practica zodat ik mijn doctoraat

op tijd kon indienen. Mijn thesisstudenten (Ilse, Josefien, Mado en Katrien) wil ik ook

bedanken voor hun hulp en inzet.

Daar de boog niet altijd gespannen kan zijn wil ik ook graag mijn

vrienden/collega’s/ex-collega’s bedanken met wie ik de afgelopen jaren samen in de

minivoetbal zat, samen mee gaan fietsen ben, samen een pintje gedronken heb en mee

gebabbeld heb (van vrouwen over voetbal tot wetenschap): Arne, Annelies DQ, Bart, Domi,

Frederik, Kevin, Ko, Michael, Peter, Ruben, Stefanie, Thomas DB, Thomas Q., Timo en

Ulrik. Mijn vrienden uit Poperinge wil ik ook bedanken. Hoewel we elkaar soms te weinig

zien door te drukke agenda’s, weet ik mij altijd gesteund door hen: Annelies en Wannes, Bert

en Lieselot, Peter en Nele, Pieter en Elke en Tim en Annelies.

Mijn ouders, mijn broers, mijn zus en de rest van mijn familie wil ik ook bedanken

voor hun hulp en steun de afgelopen jaren ondanks dat mijn doctoraat waarschijnlijk een

redelijk abstract gegeven was voor hen.

Por último, mas não menos importante, eu quero agradecer à minha namorada: Ana,

obrigado por tudo e por todos os dias!

Page 12: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 13: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 14: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

TABLE OF CONTENTS_____________________________ Table of contents i

List of Abbreviations ix

Chapter 1

SITUATION AND OBJECTIVES 1

1.1. INTRODUCTION 1

1.2. OBJECTIVES 2

1.3. REFERENCES 4

Chapter 2

INTRODUCTION 6

2.1. PACLITAXEL 6

2.1.1. History 6

2.1.2. Chemistry and metabolisation 8

2.1.3. Method of action and indications 9

2.1.4. Formulation 11

2.1.5. Alternative formulation strategies 12

2.2. PERITONEAL CARCINOMATOSIS 17

2.2.1. Peritoneum and Peritoneal Cancer 17

2.2.2. Treatment of peritoneal cancer 20

2.2.3. Rationale for hyperthermic intraperitoneal chemotherapy 22

2.3. CYCLODEXTRINS 26

2.3.1. Structure and properties 26

2.3.2. β-Cyclodextrins and their derivatives 27

i

Page 15: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.3.3. β-Cyclodextrins and paclitaxel 29

2.4. REFERENCES 31

Chapter 3:

FORMULATION AND STABILITY OF PACLITAXEL/β-

CYCLODEXTRIN COMPLEXES 41

3.1. Abstract 41

3.2. INTRODUCTION 42

3.3. MATERIALS AND METHODS 44

3.3.1. Materials 44

3.3.2.Preparation of the inclusion complexes 44

3.3.3. Paclitaxel determination 45

3.3.4. Determination of inclusion efficiency 45

3.3.5. Characterisation of Pac/RAME-β-CD complex 46

3.3.5.1. Thermal analysis 46

3.3.5.2. 1H-NMR 46

3.3.6. Physical stability testing 46

3.3.7. Analysis of the freeze dried product 47

3.3.8. Statistical Analysis 47

3.4. RESULTS AND DISCUSSION 48

3.4.1. Solubilisation of paclitaxel 48

3.4.1.1. Selection of the chemically modified β-cyclodextrins 48

3.4.1.2. Characterization of Pac/RAME-β-CD complex 49

3.4.2. Determination of physical stability 51

ii

Page 16: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

3.5. CONCLUSION 56

3.6. REFERENCES 57

Chapter 4:

IN VITRO CYTOTOXICITY OF PACLITAXEL/

β−CYCLODEXTRIN COMPLEXES FOR HIPEC 60

4.1. ABSTRACT 60

4.2. INTRODUCTION 61

4.3. MATERIALS AND METHODS 62

4.3.1. Materials 62

4.3.2. Preparation of the inclusion complexes 63

4.3.3. Determination of the administered paclitaxel concentrations 63

4.3.4. Cell Culture 64

4.3.5. Cytotoxicity Assays 65

4.3.5.1. MTT assay 65

4.3.5.2. SRB assay 66

4.3.6. Statistical Analysis 66

4.4. RESULTS 68

4.4.1. Cytotoxicity of Cremophor EL® and RAME-β-CD 68

4.4.2. Cytotoxicity of Taxol® and paclitaxel/RAME-β-CD complexes 70

4.4.2.1. CC531s cell line 70

4.4.2.2. CaCo-2 cell line 72

4.5. DISCUSSION 75

4.6. CONCLUSION 78

iii

Page 17: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

4.7. REFERENCE LIST 79

Chapter 5:

PDMP ANALOGUE RESENSITIZES MDR CELLS FOR

PACLITAXEL/β-CYCLODEXTRIN COMPLEX 84

5.1. ABSTRACT 84

5.2. INTRODUCTION 85

5.3. MATERIALS AND METHODS 87

5.3.1. Materials 87

5.3.2. Preparation of the inclusion complexes 87

5.3.3. Determination of paclitaxel concentration 88

5.3.4. Cell Culture 88

5.3.5. Cytotoxicity Assays 89

5.3.5.1. MTT assay 90

5.3.5.2. Sulphorhodamine B (SRB) assay 90

5.3.6. MDR Analysis 91

5.3.7. Statistical Analysis 91

5.4. RESULTS 92

5.4.1 Comparison of cytotoxicity of Pac/RAME-β-CD and Taxol® 92

5.4.2 Modulation of multidrug resistance 95

5.5. DISCUSSION 98

5.6. CONCLUSION 100

5.7. REFERENCES 102

iv

Page 18: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6:

IN VIVO TOXICITY AND BIOAVAILIBILITY OF TAXOL®

AND A PACLITAXEL/β-CYCLODEXTRIN FORMULATION

IN A RAT MODEL DURING HIPEC 106

6.1. ABSTRACT 106

6.2. INTRODUCTION 107

6.3. MATERIALS AND METHODS 108

6.3.1. Materials 108

6.3.2. Preparation of the inclusion complexes 109

6.3.3. Determination of the paclitaxel concentration in the perfusate 109

6.3.4. Determination of paclitaxel plasma concentration 110

6.3.5. HIPEC procedure 111

6.3.6. Maximum tolerated dose and toxicity study 112

6.3.7. Statistical Analysis 114

6.4. RESULTS 114

6.4.1 Compatibility of paclitaxel formulations in different tubings 114

6.4.2. Maximum Tolerated Dose 115

6.4.3. Blood parameters 116

6.4.4. Bioavailability study 118

6.5. DISCUSSION 120

6.6. CONCLUSION 123

6.7. REFERENCES 124

v

Page 19: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 7:

PREMILINARY EVALUATION OF THE TUMOUR GROWTH

DELAY IN A RAT MODEL FOLLOWING HIPEC WITH TWO

PACLITAXEL FORMULATIONS 129

7.1. ABSTRACT 129

7.2. INTRODUCTION 130

7.3. MATERIALS AND METHODS 131

7.3.1. Materials 131

7.3.2. Preparation of the inclusion complexes 132

7.3.3. HIPEC procedure 132

7.3.4. Tumor model 134

7.3.5. Tumour imaging 135

7.3.5.1. Determination of tumor activity via PET 135

7.3.6.2. Determination of tumor volume via MRI 136

7.3.7. Statistical Analysis 136

7.4. RESULTS 137

7.4.1 Treatment characteristics 137

7.4.2. Tumour Growth Delay 137

7.4.2.1. PET imaging 138

7.4.2.2. MRI 139

7.5. DISCUSSION 141

7.6. CONCLUSION 143

7.7. REFERENCES 144

vi

Page 20: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

vii

Chapter 8:

GENERAL CONCLUSION 147

Chapter 9:

FUTURE PERSPECTIVES 149

Summary 151

Samenvatting 155

Curriculum vitae 159

Page 21: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

viii

Page 22: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

LIST OF ABBREVIATIONS__________________________

ALT Alanine amino transferase

ANOVA Analysis of variance

β-CD β-cyclodextrin

DIME-β-CD 2,6-dimethylated-β-cyclodextrin

DSC Differential scanning calorimetry

FDG 2-(fluorine-18)-fluoro-2-deoxy-D-glucose

GCS Glucosylceramide synthetase

GGT γ-glutamyl transferase

HIPEC Hyperthermic intraperitoneal chemotherapy

HP-β-CD Hydroxypropyl-β-cyclodextrin

HPLC High performance liquid chromatography

HPMC Hydroxypropylmethylcellulose

IC50 50 % inhibitory drug concentration

IP Intraperitoneal

IV Intravenous

MDR Multidrug resistance

MRI Magnetic resonance imaging

MTD Maximum tolerated dose

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

bromide

NB-DNJ N-butyldeoxynojirimycin.HCl

NMR Nuclear magnetic resonance

Pac Paclitaxel

PBS Phosphate buffered saline

PC Peritoneal carcinomatosis

PDMP D-threo-(1R,2R)-1-phenyl-2-decanoylamino-3-

morpholino-1-propanol

PET Positron emission tomography

P-gp P-glycoprotein

QOL Quality of life

RAME-β-CD Randomly-methylated-β-cyclodextrin

TDS Total degree of substitution

ix

Page 23: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

x

TGD Tumour Growth Delay

SBE-β-CD Sulphobutyl-ether-β-CD

SRB Sulphorhodamine B

UV/VIS Ultraviolet/visible

Page 24: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 25: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 26: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 1 Situation and objectives 1

SITUATION AND OBJECTIVES

1.1. INTRODUCTION

Peritoneal carcinomatosis (PC) is a frequent clinical phenomenon in the course of

gastrointestinal and ovarian cancer. It is present in 40 to 80 % of the patients with a fatal

colorectal cancer (Koppe et al., 2006) and in a majority of the patients diagnosed with ovarian

cancer (Tan et al., 2006). Peritoneal spread is responsible for a low quality of life and a high

morbidity and mortality (McQuellon et al., 2001). Standard treatment for PC consists of

systemic 5-fluorouracil-based chemotherapy and offers a median survival of 5.2 to 12.6

months (Koppe et al., 2006). This treatment is often merely palliative. Peritoneal spread is

mostly confined to the peritoneal cavity, which makes it very interesting for locoregional

treatment. In recent years, cytoreductive surgery combined with hyperthermic intraperitoneal

chemotherapy has been introduced as an alternative treatment, which is already showing

promising results (Verwaal et al., 2008). The rationale for intraperitoneal chemotherapy is

based on the peritoneal-plasma barrier which allows higher concentrations of the cytotoxic

drug to be administered with a limited systemic toxicity (Witkamp et al., 2001). Based on the

linear correlation between dose and effect for most chemotherapeutic drugs, it can be

expected that the antitumour activity will increase. Intraperitoneal chemotherapy is often

combined with hyperthermia based on the synergism between hyperthermia and cytostatic

agents (Sugarbaker, 2007). Hyperthermia can also enhance drug penetration in the tumour

(Witkamp et al., 2001). Paclitaxel, which is considered as one of the most important drugs of

11

Page 27: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

the chemotherapeutic arsenal, is an interesting molecule for intraperitoneal therapy. The

absorption of paclitaxel from the peritoneum is limited due to its large molecular weight. Its

predominantly hepatic metabolisation will further decrease the systemic exposure for the

patient. The use of paclitaxel is however hampered by its poor aqueous solubility (0.34 µg/ml

or 0.4 µM, Sharma et al., 1995) and the currently used vehicle (Cremophor EL®/ethanol,

50/50 (v/v) ratio) to solubilise paclitaxel is associated with severe hypersensitivity reactions

and leaching of plasticisers from PVC infusion sets normally used in hospitals (Gelderblom et

al., 2001). One of the possible alternatives for dissolving paclitaxel in water are β-

cyclodextrins. These cyclic oligosaccharides consist of 7 glucopyranose units. Their 3D

structure is characterized by a hydrophobic cavity and a hydrophilic exterior which enhances

the aqueous solubility of hydrophobic drugs via the formation of inclusion complexes. Two

reports in literature have demonstrated the feasibility of making paclitaxel/β-cyclodextrin

complexes without loss of activity for paclitaxel. However, they either used large amounts of

ethanol (Szente el al., 1999) or toxic organic solvents (methanol and tert-butyl alcohol) during

production of the complexes (Sharma et al., 1995).

1.2. OBJECTIVES

The objective of this study was to develop a safe paclitaxel formulation suitable for

hyperthermic intraperitoneal chemotherapy (HIPEC). To achieve this objective the following

topics were addressed in the research project:

- Formulation and in-vitro characterisation of a co-solvent-free and tensioactive-

free paclitaxel formulation using chemically modified β-cyclodextrins to design stable

water soluble inclusion complexes of paclitaxel.

Chapter 1 Situation and objectives 2

Page 28: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 1 Situation and objectives 3

- In vitro evaluation of the cytotoxicity of the paclitaxel inclusion complexes

against colon cancer cell lines. The efficiency of this novel paclitaxel formulation was

compared to the commercially available Taxol® formulation. In addition the toxicity

of the formulation excipients of both formulations was also evaluated.

- In vivo evaluation of the maximum tolerated dose, general toxicity and

bioavailability following HIPEC with the paclitaxel/cyclodextrin inclusion complexes

and Taxol® in a rat model for intraperitoneal cancer originating from a colorectal

cancer.

- In vivo evaluation of the tumour growth delay via magnetic resonance imaging

(MRI) and positron emission tomography (PET) following HIPEC with the

paclitaxel/cyclodextrin inclusion complexes and Taxol® in a rat model for

intraperitoneal cancer originating from a colorectal cancer.

Page 29: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

1.3. REFERENCES

Gelderblom H., Verweij J., Nooter K., Sparreboom A., 2001. Cremophor EL: the drawbacks

and advantages of vehicle selection for drug formulation. Eur. J. Cancer, 37, 1590-

1598.

Koppe M.J., Boerman O.C., Oyen W.J.G., Bleichrodt R.P., 2006. Peritoneal carcinomatosis of

colorectal origin-Incidence and current treatment strategies. Ann. Surg., 243, 212-222.

McQuellon R.P., Loggie B.W., Fleming R.A., Russel G.B., Lehman A.B., Rambo T.D., 2001.

Quality of life after intraperitoneal hyperthermic chemotherapy (IPHC) for peritoneal

carcinomatosis. E.J.S.O., 27, 65-73.

Sharma U.S., Balasubramanian S.V., Straubinger R.M., 1995. Pharmaceutical and physical

properties of paclitaxel (Taxol) complexes with cyclodextrins. J. Pharm. Sci., 84,

1223-1230.

Sugarbaker, P.H., 2007. Laboratory and clinical basis for hyperthermia as a component of

intracavitary chemotherapy. Int. J. Hyperthermia, 23, 431-442.

Szente L., Vikmon M., Szemán J., Otta K., 1999. Methyl-b-cyclodextrin/paclitaxel aqueous

solutions: a tool to test Cremophor-free paclitaxel. STP Pharm. Sci., 9, 243-247.

Tan D.S.P., Agarwal R., Kaye S.B., 2006. Mechanisms of transcoelomic metastasis in ovarian

cancer. Lancet Oncol., 7, 925-934.

Chapter 1 Situation and objectives 4

Page 30: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 1 Situation and objectives 5

Verwaal V.J., Bruin S., Boot H., van Slooten G., van Tinteren H., 2008. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus

sytemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer.

Ann. Surg. Oncol., 15, 2426-2432.

Witkamp A.J., de Bree E., Van Goethem A.R., Zoetmulder F.A.N., 2001. Rationale and

techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer

Treat. Rev., 27, 365-374.

Page 31: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 32: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 2 Introduction 6

INTRODUCTION

“Progression of gastrointestinal or ovarian cancer to the peritoneal surfaces remains a

dreaded clinical condition. It remains a perplexing challenge with pitfalls in both diagnosis

and treatment that continue to vex the oncologist. Recently, pharmacologic studies, an

aggressive surgical approach, and concentration of patients in peritoneal surface malignancy

treatment centers have begun to generate a small optimism regarding the management of these

patients.”

Paul H. Sugarbaker, Washington Cancer Institute

In “Peritoneal Carcinomatosis: a multidisciplinary approach (Springer, Berlin, 2006)”

As the treatment of peritoneal cancer is based on a multidisciplinary approach, the

different domains covered in this research project will be briefly introduced in this chapter.

2.1. PACLITAXEL

2.1.1. History

The first report about paclitaxel dates back to the beginning of 1900’s when a British

official in the Indian subcontinent had noted that parts of Taxus baccata were used to treat

cancer.

22

Page 33: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Some 60 years later, in 1962, a program of the National Cancer Institute (USA)

evaluating domestic plants for their anticancer activity revealed that crude extracts of the

Pacific (or Western) yew (Taxus Brevifolia, Fig. 1) showed cytotoxic activity against several

tumours. It took another 10 years before the active compound paclitaxel was isolated and fully

characterized by Wani et al. (1971). In 1979, Horwitz and colleagues elucidated the unique

mechanism of paclitaxel (Schiff et al., 1979), discovering that paclitaxel promoted the

polymerization of tubulin. This disturbance of the dynamic balance causes cells to go into

apoptosis.

Fig. 1: Taxus Brevifolia

Initially, a major problem was the scarcity of the crude plant material and

consequently of paclitaxel, but finally a semi-synthetic process using 10-deacetylbaccatin III

as precursor allowed to produce sufficient amounts of paclitaxel and the first phase I trials

were initiated in 1983. Its poor water solubility further hampered the development of a

commercially viable formulation, until a collaboration between the National Cancer Institute

and Bristol-Myers-Squibb® resulted in a formulation (Taxol®) where paclitaxel was dissolved

in a 50/50 (v/v) mixture of Cremophor EL® and ethanol. This concentrate (6 mg/ml) is diluted

to the appropriate concentration and administered via an infusion set to the patient. Finally, in

1992 and 1994 the Food and Drug Administration (FDA) approved Taxol ® for the treatment

Chapter 2 Introduction 7

Page 34: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

of ovarian and breast cancer, respectively. Since then the interest in paclitaxel and other

taxanes has only increased. They are considered as the most important additions to the

chemotherapeutic arsenal in the late 20th century.

2.1.2. Chemistry and metabolisation

Paclitaxel, together with docetaxel are the most important representatives of the

taxanes. These molecules have a taxane ring in common, which is in fact a diterpene carbon

skeleton (Fig. 2). Paclitaxel is most stable in a pH range of 4 to 8 (Huizing et al., 1995). In

alkaline medium it degrades rapidly due to hydrolysis of the ester function (Volk et al., 1997).

C

OHN C

H

HC C

O

O

OHHO

O

CO

OO C

O

CH3

OH

O

O C

O

CH3

Fig. 2: Chemical structure of paclitaxel

Whereas early phase I studies suggested that the pharmacokinetic behaviour was linear

and could be described by a simple two-compartment model (Wiernik et al., 1987), later

studies identified nonlinear pharmacokinetics, with peak plasma concentrations and drug

exposure increasing disproportionally at higher doses. In plasma, paclitaxel is highly protein-

bound and has a large volume of distribution. It is metabolized in the liver and eliminated via

Chapter 2 Introduction 8

Page 35: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

the biliary tract. Renal clearance and urine excretion are minimal (Rowinsky and Donehower,

1995). Sparreboom and colleagues suggested that the non-linear pharmacokinetic behaviour

of Taxol® is due to Cremophor EL®, a non-ionic tensioactive excipient used for the

solubilisation of paclitaxel (Gelderblom et al., 2002). Because paclitaxel has a higher affinity

for Cremophor EL® than for plasma and human serum albumin, the free paclitaxel fraction in

blood will be affected when higher amounts of Cremophor EL® are administered

(Sparreboom et al., 1999).

2.1.3. Method of action and indications

At the Albert Einstein College of Medicine, paclitaxel’s unique mechanism of action

was discovered in 1979 (Schiff et al., 1979). They found that paclitaxel binds to the

microtubule rather than to the tubulin dimers. The dynamic equilibrium between microtubules

and tubulin dimers is important to eukaryotic cells and is essential for mitosis, maintenance of

cell shape, intracellular transport, cell signalling and division. Microtubuli are formed due to

the polymerization of α1- and β1-tubulin, under the influence of guanosine triphosphate

(GTP) and microtubule-associated proteins (MAPs) (Fig. 3). The taxane binding site of β-

tubulin in the microtubule is the cellular target for paclitaxel (Downing et al., 1999).

Paclitaxel is able to shift the equilibrium between microtubuli and tubulin to the assembly of

microtubuli (even in the absence of GTP and MAPs). Paclitaxel forms very stable and

dysfunctional microtubules and is, consequently, known as a microtubule-stabilizing agent.

Hereby, paclitaxel blocks the normal microtubule dynamics during mitosis. This causes the

cell to go into cell arrest and apoptosis. Cells treated with paclitaxel typically accumulate in

the G2 and M phase of the cell cycle. Other antimicrotubule drugs (i.e. vincristine,

vinblastine, colchicine and podophyllotoxin) induce the disassembly of microtubules. Because

Chapter 2 Introduction 9

Page 36: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

cancer cells have a much faster cell division than non-cancerous cells, they are more affected

by this microtubule stabilizing effect of paclitaxel.

Fig. 3: Binding of paclitaxel to the interior surface of the microtubule at the taxane-binding site (McGrogan et al., 2008)

Taxanes are used in monotherapy (ovarian, breast and non-small-cell lung cancer) as

well as in combination therapy (combined with cisplatin for ovarian cancer, combined with

doxorubicin for breast cancer and combined with cisplatin or carboplatin for non-small-cell

lung cancer). Paclitaxel is regulatory approved for the treatment of patients with ovarian

cancer after failure of first-line or subsequent chemotherapy. In addition, combined with a

platinum compound it is indicated for first-line therapy of ovarian cancer in patients with

suboptimally debulked stage III or IV ovarian cancer. Recently, it also received approval for

the treatment of first-line metastatic breast cancer when used in combination with gemicitabin

(yielding response rates between 25 and 69 % (Paridaens et al., 2000)), for second-line

Chapter 2 Introduction 10

Page 37: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

treatment of Kaposi’s sarcoma and in combination with cisplatin as a primary treatment of

non-small-cell lung cancer (Rowinsky and Calvo, 2006). Further clinical trials are exploring

other possible indications.

Resistance to paclitaxel has been reported due to mutations in α en β tubulin, differing

β-tubulin isotype compositions, P-glycoprotein (P-gp) overexpression or increased

microtubule dynamics associated with altered MAP expression (McGrogan et al., 2008).

Side-effects of taxane therapy include myelosuppression, neuropathy, myalgia, fatigue,

alopecia, diarrhoea, mucosal toxicity and skin and nail changes. Hypersensitivity reactions are

frequently encountered during administration of Taxol® (i.e. a paclitaxel formulation using an

ethanol/Cremopher EL® (50/50, v/v) mixture as solvent for paclitaxel) and are treated via

premedication with steroids and histamine antagonists.

2.1.4. Formulation

Due to its chemical structure, paclitaxel has a very low aqueous solubility (0.4 µM or

0.34 µg/ml, Sharma et al., 1995) making the development of an intravenous formulation

challenging. Bristol Myers and Squibb® (BMS) developed, in collaboration with the National

Cancer Institute (USA), the current Taxol® formulation. This formulation for intravenous (IV)

administration contains 6 mg paclitaxel per ml, dissolved in a 50/50 (v/v) mixture of

polyoxyethylated castor oil (Cremophor EL®) and ethanol. This concentrate is administrated

via infusion after dilution in either 5 % dextrose or 0.9 % sodium chloride to a concentration

of 0.3-1.2 mg/ml. Typically doses of 135-175 mg paclitaxel per m² body surface are IV

administered over 3 or 24 hrs.

Cremophor EL® is implicated as the potential cause of anaphylactic reactions in

humans and is believed to be the cause of the hypersensitivity reactions often encountered

Chapter 2 Introduction 11

Page 38: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

after Taxol® administration (Hennenfent and Govindan, 2006). In approximately 40 % of the

patients minor reactions like rash and flushing are observed despite premedication and almost

3 % of the patients have potentially life-threatening adverse effects (Rowinsky et al., 1993;

ten Tije et al., 2003). As Cremophor EL® also extracts diethylhexylphtalate (DEHP) used as

plasticizer in polyvinylchloride (PVC) containers, contact between Taxol® and PVC should be

avoided. Due to these problems encountered by the use of Cremophor EL®, a lot of research

efforts are focused on alternative approaches to formulate paclitaxel.

2.1.5. Alternative formulation strategies

The aim of developing an alternative paclitaxel formulation is to eliminate the

drawbacks of the current formulation and to achieve reduced toxicity, easier administration,

greater patient convenience and maximization of paclitaxel activity. Next to reformulation of

paclitaxel, taxane analogues are also being developed. An example of this is docetaxel, which

has been developed by Sanofi-Aventis® in an attempt to find a more water soluble paclitaxel

analogue. Other microtubule polymerizing agents have been synthesized and are currently in

early clinical development (Ferlini et al., 2008). This approach will not be discussed in detail

as it is beyond the scope of this research project.

In an excellent review Singla et al. (2002) defined several reformulation strategies for

paclitaxel: co-solvency, emulsification, micellisation, liposome formation, non-liposomal

drug carrier systems, cyclodextrins and local drug delivery. In the meanwhile, some of these

alternatives have been tested in clinical trials or have been approved. In this introductory

chapter only those alternatives for Taxol® are discussed which are already in an advanced

stage of development.

Chapter 2 Introduction 12

Page 39: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Co-solvency

One approach to replace Cremophor EL® in the Taxol® formulation is the use of other

co-solvents. Commonly used co-solvents for the dissolution of hydrophobic drugs include

ethanol, dimethylsulphoxide, polyethylene glycol, polysorbate and propylene glycol (Adams

et al., 1993). A 75 % (v/v) polyethylene glycol 400 solution in water was able to dissolve 16

mg/ml of paclitaxel but upon dilution for infusion, precipitation was observed (Adams et al.,

1993). The vehicle used for Taxotere® consists of ethanol and polysorbate 80 to dissolve

docetaxel (Bissery et al., 1991). A serious drawback of the co-solvency formulation approach

is the frequent precipitation upon dilution when the drug is administered (Singla et al., 2002).

Emulsions

Emulsions and microemulsions of the oil in water (O/W) type have received a lot of

attention (Tarr et al., 1987; Wheeler et al., 1994; Date and Nagarsenker, 2008; Nornoo and

Chow, 2008). Tocosol® is a Cremophor EL®-free, vitamin E-based paclitaxel emulsion, which

uses α-tocopherol PEG succinate as surfactant. Preclinical data showed that its maximum

tolerated dose (MTD) was three times higher compared to Taxol®. Phase I and II trials

indicated that Tocosol® was well tolerated and had promising antitumoral activity against

bladder, non-small-cell lung cancer and ovarian cancer (Hennenfent and Govindan, 2006;

Rowinsky and Calvo, 2006).

Micellisation

During micellisation a hydrophobic drug is physically entrapped in a biodegradable

drug carrier micelle. Two representatives of this approach are Genexol PM® and Pacliex®.

Genexol PM® is a polymeric-micellar paclitaxel formulation consisting of a

biodegradable block copolymer which includes monomethoxy poly(ethylene glycol)-block-

Chapter 2 Introduction 13

Page 40: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

poly(D,L-lactide). Phase I data revealed an MTD of 390 mg paclitaxel/m² following 3 hrs

intravenous infusion every 3 weeks, whereas paclitaxel doses are typically between 135 – 175

mg/m² administered during 3 to 24 hrs intravenous infusion. In a multicenter phase II trial, it

showed significant efficacy in patients with metastatic breast cancer. Further trials are being

conducted (Lee et al., 2008).

Pacliex® is a mixed micelles preparation consisting of paclitaxel and a surfactant

system based on an equimolar mixture of two isoforms of an amphiphilic synthetic derivative

of retinoic acid. The in vitro and in vivo cytotoxicity of Pacliex® was similar to Taxol®

(Hassan et al., 2005)

Liposomes

Liposomes are interesting carrier systems with the ability to encapsulate both

lipophilic and hydrophilic drugs as they consist of one or more aqueous compartments

contained within a lipid membrane bilayer. Because of the presence of both lipophilic and

hydrophilic compartments they can accommodate therapeutic agents with diverse

physicochemical properties. However, the disadvantages associated with liposomes are a low

entrapment efficiency, limited drug stability, drug leakage and poor storage stability.

Liposomal-encapsulated paclitaxel with promising antitumor activity in mice models has

already been described (Cabanes et al., 1998). Phase I clinical trials determined an MTD of

175 mg/m² (which is similar to Taxol®). Although neuropathy, myalgia and alopecia were not

observed, premedication (anti-histamins and steroids) was required to suppress liposome

infusion reactions (Treat et al., 2001).

Chapter 2 Introduction 14

Page 41: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Non-liposomal drug carrier systems

Non-liposomal drug carrier systems are formulated as biodegradable polymeric micro-

or nanocapsules, whereby nanospheres are especially interesting as they allow intraveneous

and intramuscular administration. One example is the Paclimer® delivery system, which is a

biopolymer-based formulation designed for site-specific and controlled delivery of paclitaxel.

It has been evaluated in preclinical studies for its activity against prostate, ovarian and non-

small-cell lung cancer (Harper et al., 1999; Lapidus et al., 2004; Armstrong et al., 2006) but it

remains unknown if this will result into clinical therapeutic benefits.

Cyclodextrins

Cyclodextrins have also been used in combination with paclitaxel, but as this

formulation strategy is the focus of this research project, cyclodextrins are discussed in detail

in a separate part of the introduction.

Local drug delivery

Local drug delivery of paclitaxel consists of the implantation of drug releasing stents

or biodegradable devices loaded with paclitaxel. These devices create a high local

concentration of paclitaxel in an attempt to eradicate residual cancer cells.

Macromolecular drug carrier conjugates

The conjugation of paclitaxel with macromolecular drug carriers is already applied in

several commercially available formulations: Xyotax®, Abraxane® and Taxoprexin®. Because

of their large size, these conjugates will preferably extravasate through the permeable

pathological vasculature and thereby accumulate in malignant tissue. This process is known as

the enhanced permeability and retention (EPR) effect and reduces the systemic toxicity of

Chapter 2 Introduction 15

Page 42: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

anticancer drugs. After administration of these formulations conversion from an inactive

prodrug to active paclitaxel occurs predominantly in the cell, thus reducing systemic toxicity

and allowing a higher concentration to be administered.

Xyotax® (Opaxio®) contains a poly-L-glutamate paclitaxel conjugate as paclitaxel is

covalently bound at the 2´hydroxyl group to the carboxylate of L-glutamic acid. Conjugated

paclitaxel is inactive but inside the cell paclitaxel is released. Although the results of different

phase III clinical trials in patients with non-small-cell lung cancer were disappointing (Cella

et al., 2005; Langer et al., 2005; O’Brien et al., 2005), analysis of the data unexpectedly

indicated a significantly higher survival rate for women in comparison to men. Hence, new

trials were started to evaluate its efficacy against non-small-cell lung cancer in women.

Abraxane® consists of paclitaxel bound to human serum albumine. It is a nanoparticle

colloidal suspension prepared by high-pressure homogenization of paclitaxel in the presence

of human serum albumin. In contrast to Taxol®, it is characterised by an almost linear

pharmacokinetic behaviour and a rapid distribution into tissue. It is approved by FDA for

metastatic breast cancer in patients who have failed first line treatment. Approval was

supported by the absence of hypersensitivity reactions in patients receiving no premedication

(Ferlini et al., 2008).

In Taxoprexin® paclitaxel is bound to docosahexaenoic acid. The conjugation with a

fatty acid is based on the observation that tumours have a higher fatty acid uptake, which

could enhance the drug targeting. As this formulation still contains Cremophor EL® (albeit in

a reduced concentration) premedication is still mandatory. It has been evaluated for the

treatment of different cancers (non-small-cell lung cancer, melanoma, advanced gastric and

oesophageal adenocarcinoma), the best results being obtained for melanoma treatment

(Hennenfent and Govindan, 2006).

Chapter 2 Introduction 16

Page 43: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.2. PERITONEAL CARCINOMATOSIS

2.2.1. Peritoneum and Peritoneal Cancer

The peritoneum (Fig. 4) is a serous membrane that lines the peritoneal cavity and is

composed of a layer of mesothelium and a thin layer of connective tissue. Its total area is

about the same as the skin surface. There are two types of peritoneum: the visceral

peritoneum which covers the intra-abdominal organs and mesenteries, and the parietal

peritoneum which lines the abdominal wall, pelvis, anterior surfaces of retroperitoneal organs

and inferior surface of the diaphragm. The space between the visceral and parietal peritoneum

is filled with peritoneal fluid. This fluid, together with the peritoneum, reduces the friction

and hereby improves the free movement of the abdominal viscera. Other functions of the

peritoneum include resisting or localizing infection and the storage of fat. The flow of the

peritoneal fluid is directed from the pelvis to the paracolic gutters and driven by the breathing

rate (Raptopoulos and Gourtsoyiannis, 2001).

LIVER

PANCREAS

STOMACH

COLON

PERITONEUM

SMALL  INTESTINE

Fig. 4: Abdominal cavity showing peritoneum (blue)

Chapter 2 Introduction 17

Page 44: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

The peritoneum forms a barrier between the abdominal cavity and the blood stream.

Therefore, it is also referred to as the peritoneal-plasma barrier and it consists of peritoneal

fluid, mesothelium, basal membrane, interstitium with bundles of collagen fibrils, blood

vessels and lymphatic vessels. Drugs with a molecular weight of less than 2000 Da can be

absorbed by diffusion through the parietal and visceral peritoneum, while larger molecules are

absorbed by the lymphatic drainage at the diaphragmatic peritoneum (Sugarbaker, 1996). This

peritoneal-plasma barrier reduces the amount of drug that is absorbed in the blood, allowing

higher concentrations to be administered in the abdominal cavity. Another advantage of

intraperitoneal administration of drugs is that a part of the peritoneum is drained with the

portal vein system. Hence drugs absorbed via this vein system are immediately transported to

the liver for metabolisation, reducing systemic exposure.

In general, peritoneal carcinomatosis (PC) (Fig. 5) is considered as independent

metastatic deposits on the visceral and parietal peritoneum, originating from a primary organ

of origin in the abdominal cavity. PC is a nice example of the “seed and soil” hypothesis of

Paget, which states that the metastatic capability of tumour cells (“the seed”) is related to

finding a good environment (‘the soil’) for implantation.

In general, the course of PC includes the following steps: release and transport of

cancer cells, adhesion to the mesothelial layer and the invasion of the peritoneum and

subperitoneal tissue (Ceelen et al., 2006). The etiology of PC is diverse. One possible cause is

the shedding of free cells from serosal penetration of the primary tumour. Other possible

causes are leakage of malignant cells from venous blood backflow or transected lymphatic

vessels and direct dissemination as a result of surgical trauma.

Chapter 2 Introduction 18

Page 45: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Fig. 5: Peritoneal cancer and its histological environment (de Bree et al., 2002)

PC is a common evolution in the course of digestive and ovarian cancers (Table 1).

For instance, the peritoneal surface is an important failure site for patients with colorectal

cancer. Recurrent disease on the peritoneal surface is involved in up to 44 % of patients with

recurrent colorectal cancer and in 40 to 80 % of patients with a fatal colorectal cancer (Koppe

et al., 2006). For gastric, ovarian and appendiceal cancer 50, 75 and 31 % of the patients,

respectively, have peritoneal disease at exploration (Stewart et al., 2005).

Primary tumor Incidence in US and Europe (cases/year)

% of patients with peritoneal disease at

exploration

Median survival after diagnosis of carcinomatosis

(months) Colorectal cancer Gastric cancer Ovarian cancer Peritoneal mesothelioma Appendiceal cancer

130,000 22,000 27,000 1500 500

10-15

50 75

100 31

5.2

3.1

36 12-17

NA

Table 1: Common indications for intraperitoneal hyperthermic chemoperfusion (NA, not available) (Stewart et al., 2005)

Chapter 2 Introduction 19

Page 46: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.2.2. Treatment of peritoneal cancer

Based on the data described above, it is clear that PC is a rather common phenomenon

with important clinical consequences and poor prognosis. Therefore, PC merits attention to

find new and improved treatment modalities to help these patients. This introduction about the

treatment modalities for PC will focus on PC from non-gynaecologic origin and specifically

from colorectal origin. Until recently, PC was considered a terminal condition and the only

treatment strategy for PC consisted of conventional systemic chemotherapy, possibly in

combination with cytoreductive surgery of macroscopic tumour nodules. In this case systemic

chemotherapy is considered as palliative and provides only a limited improvement of survival.

The median survival after systemic 5-fluorouracil-based chemotherapy for patients with PC of

colorectal origin varied from 5.2 to 12.6 months (Chu et al., 1989; Verwaal et al., 2003).

Aggressive cytoreductive surgery followed by (hyperthermic) intraperitoneal

chemotherapy is a novel therapeutic approach for patients where the peritoneum is the only

site of metastasis. To date, this technique is used in approximately 30 medical centers

worldwide (Koppe et al., 2006). Several factors are determining the prognosis after

intraperitoneal chemotherapy as survival is correlated with gender, age, clinical performance

status, PC stage and completeness of resection. Obviously, patients without residual

macroscopic tumours after resection have a higher survival rate as the follow-up study of

Verwaal et al. (2008) indicated a 5-year survival in 45 % of patients with complete tumour

resection. Another important parameter to implement a treatment is the quality of life (QOL)

after surgery. Typically for hyperthermic intraperitoneal chemotherapy (HIPEC), QOL is

reduced in the first 3 months after surgery but afterwards QOL improved and 1 year post-

operation 58 % of the patients had a normal patient performance status rating (McQuellon et

al., 2001). Due to the aggressiveness of the treatment, there are high morbidity, mortality (9.3

Chapter 2 Introduction 20

Page 47: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

% according to Elias et al., 2001) and failure rates. Therefore, it is important to select only

those patients who will benefit the most from HIPEC.

In 2007, the consensus statement about cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy in the management of peritoneal surface malignancies of

colonic origin was issued (Fig. 6), providing a tool to select the patients (Esquivel et al.,

2007).

Fig. 6: Consensus scheme for the management of peritoneal cancer from colonic origin

(Esquivel et al., 2008)

Chapter 2 Introduction 21

Page 48: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Although there is consensus in the selection of patients, there are still significant

differences in the HIPEC procedure used by the different medical centers: open versus closed

circulation (to allow recirculation of the perfusion fluid), open versus closed abdominal

cavity. Other variables include the choice of drug, perfusion duration, temperature of

perfusion liquid and post-operative administration of intraperitoneal chemotherapy. The

consensus statement stated that for patients with PC of colonic origin HIPEC should be

performed with mitomycin C (15 -35 mg/m²) at a temperature of 39-42 °C for 60-120 min.

Whether an open or closed technique and post-operative chemotherapy of 5-fluorouracil (5-

FU) is used is the choice of the surgeon.

2.2.3. Rationale for hyperthermic intraperitoneal chemotherapy

In 1980, HIPEC (Fig. 7) was applied for the first time in a clinical setting by Spratt et

al. for the treatment of a patient with pseudomyxoma peritonei. Since then, the interest for this

novel treatment of PC has only grown. HIPEC is an interesting approach for this type of

cancer as peritoneal spread is mostly confined to the abdominal cavity. Therefore, regional

therapy via HIPEC could improve the cure rate of PC patients as the cancer cells are in direct

contact with the cytotoxic drug. Another interesting advantage of antitumour therapy via

HIPEC is the peritoneal plasma barrier. This barrier reduces the movement of large molecular

drugs from the peritoneal cavity to the systemic compartment. For these high molecular

weight drugs, the peritoneal-plasma partitioning is inverse to the square root of their

molecular weight (Stewart et al., 2005). This offers an advantage to high molecular weight

anticancer drugs such as paclitaxel. As diffusion of these molecules across the peritoneal

membrane is controlled by the peritoneal plasma barrier higher doses can be administered

intraperitoneally (IP) compared to intravenous (IV) administration which is beneficial as in

Chapter 2 Introduction 22

Page 49: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

some cases the effective IV dose exceeds the toxic IV dose. Hence, the peritoneal-plasma

barrier offers an important pharmacokinetic advantage to IP therapy: high drug concentrations

are in direct contact with the tumour cells and a reduced systemic toxicity. This advantage is

expressed by the ratio between the IP and IV drug concentrations.

In addition to a high molecular weight a drug with optimal properties for HIPEC

should be metabolised by the liver in order to limit the drug concentration in the systemic

blood flow since part of the peritoneal blood flow is connected with the portal blood flow. As

an IP/IV concentration ratio of 1000 has been reported for paclitaxel (Markman, 2003), as the

IP clearance of paclitaxel is low (resulting in prolonged biological relevant IV concentrations)

and as paclitaxel is predominantly metabolised in the liver, this drug is a promising candidate

for application via HIPEC. Also, the non-vesicant properties of paclitaxel are advantageous

for IP delivery of drugs.

TEMP. PROBE

STAGED CLAMPS

REGULATEDVACUUM

FILTERED CLOSEDRESERVOIR

ROLLERPUMP

PRESSORSTAT

HEAT EXCHANGER

Fig. 7: Schematic representation of HIPEC

Chapter 2 Introduction 23

Page 50: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

A last important criterion for drugs suitable for HIPEC is the enhancement of their

effectiveness by (mild) hyperthermia. The use of hyperthermia dates back to ancient times:

“Where drugs do not cure, iron does; where iron does not cure, heat does; where real heat

does not cure, cure is impossible” (Hippocrates, 470-377 BC). In modern medicine,

hyperthermia alone has shown in vitro cytotoxic activity (Crile, 1963), due to its effect on

DNA repair, induction of apoptosis and inhibition of angiogenesis (Witkamp et al., 2001).

Increasing the temperature also enhances drug penetration into the tumour due to membrane

protein denaturation. The inactivation of tumour cells starts at 40-41 °C and is time and

temperature dependent (Ceelen et al., 2000). Hyperthermia has also shown to improve the

cytotoxic activity of various drugs such as mitomycin C, doxorubicin, cisplatin and

oxaliplatin (Glehen et al., 2004). Most medical centers use HIPEC temperatures between 41

and 43 °C because the synergism between drug and heat is at its maximum in this temperature

range and because at temperatures higher than 43 °C bowel toxicity increases (Urano et al.,

1999). Although paclitaxel is heat stable, and paclitaxel and heat have a common target as

heat also disrupts the microtubule system, the results about the cytotoxic effect when

combining heat and paclitaxel have been less consistent and in vitro as well as in vivo studies

have been inconclusive. These conflicting results will be discussed in detail in Chapter 5.

Possible drawbacks of IP chemotherapy are limited penetration in the tumour tissue

and surface exposure. The surface exposure can be enhanced by increasing the intraperitoneal

fluid distribution. Therefore, peritoneal expansion is often applied to optimise the surface

exposure in the open abdomen technique (Fig. 8). The open technique can be performed via

the coliseum technique developed by Sugarbaker et al. (1999), where a retractor ring, attached

to the skin of the abdomen, is placed in the laparotomy wound. This ring is covered with a

plastic sheet, which has a small opening allowing the surgeon to stir the abdominal organs in

order to better distribute heat and drug. Yonemura introduced a peritoneal access device to be

Chapter 2 Introduction 24

Page 51: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

fitted into the laparotomy wound (Yonemura et al., 1991) (Fig. 8). Disadvantages of these

open techniques are heat loss and more importantly leakage, creating a hazardous situation for

the operating personnel. Next to the open abdominal IP techniques, Fujimoto and Koga

independently developed a technique with a temporary closure of the abdomen (Fujimoto et

al., 1988; Koga et al., 1988). This closed technique (Fig. 8) not only prevents drug leakage

and heat loss, but also increases the pressure which may improve drug penetration (Tsiftis et

al., 1999). Another difference between perfusion techniques is the use of open or closed

perfusion models: the closed model recirculates the perfusate, whereas the open model

continuously uses new perfusate. Closed perfusion offers easier control of the system whereas

there is no risk of recirculation of tumour cells when using the open system.

Fig. 8: HIPEC with 3 different peritoneal expansion techniques: A. use of an peritoneal cavity expander, B. Coliseum technique and C. closed abdomen (Witkamp et al., 2001)

Chapter 2 Introduction 25

Page 52: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.3. CYCLODEXTRINS

As previously mentioned, the aqueous solubility of paclitaxel is very low (0.4 µM or

0.34 mg/ml) and several strategies have been investigated to formulate a suitable carrier for

this drug molecule in order to prepare an aqueous formulation suitable for IV or IP

administration. One of these options is the use of cyclodextrins to improve the solubility of

paclitaxel. Therefore, cyclodextrins will be discussed in detail as this formulation strategy was

used in this research project.

2.3.1. Structure and properties

Cyclodextrins (CDs) are cyclic oligosaccharides, consisting of α-1,4 linked

glucopyranose units. They are produced via degradation of starch (mostly corn) using

glucosyltransferase. Apart from their pharmaceutical use, they are being used in agriculture,

food industry, fragrances and others. They were first described by Villiers in 1891 and in

1953 Freudenberg et al. were the first to be granted a patent on the use of CDs in drug

formulations. The nomenclature of CDs depends on the amount of glucose residues

(minimum 6 residues). The most commonly used are α, β, γ-CDs consisting of 6, 7 and 8

glucose residues, respectively. Their 3D structure (Fig. 9) has a conical shape with primary

hydroxyl groups at the top of the cone and secondary hydroxyl groups located at the bottom.

The interest in CD for pharmaceutical applications is based on its dual properties: they are

water soluble because of their hydrophilic exterior, but they have a hydrophobic interior

cavity. This allows them to form water soluble complexes with less soluble molecules via

incorporation of hydrophobic moieties in the hydrophobic interior cavity (thus improving the

solubility and bioavailability of the complexed drug). Hence their extensive use to solubilise

Chapter 2 Introduction 26

Page 53: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

and stabilise poorly soluble drugs (Szeltli, 1994; Loftsson and Brewster, 1996). β-CDs are the

most interesting for pharmaceutical applications as its cavity is large enough to interact with

aromatic rings. Increasing the glucose residues does not necessarily enlarge the cavity as the

cavity collapses and twists to form a helix shape when the amount of glucose units further

increases.

Fig. 9: 3D-structure of β-cyclodextrin

2.3.2. β-Cyclodextrins and their derivatives

Despite their higher number of hydroxyl groups in comparison with α-CD the

solubility of β-CDs is lower compared to α-CDs. This is due to the formation of internal

hydrogen bonds between the secondary hydroxyl groups. Breaking these hydrogen bonds by

substituting the hydroxyl groups with other chemical groups increases the solubility of β-CD.

In addition substituted CDs are more amorphous, enhancing the water solubility. Substitution

also modifies the shape of the cone, altering the complexation capability.

Chemical modification results from the introduction of new functional groups at the 2-

, 3- and 6-hydroxyl groups. For β-CD there are 21 possible substitution positions and 221-1

different combinations possible. Therefore, the European Pharmacopoeia and the United

Chapter 2 Introduction 27

Page 54: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

States Pharmacopoeia specify a range of molar substitution (the number substituents per

anhydroglucose unit ) as the most important characterisation criterion. Manufacturers also use

the total degree of substitution (TDS) as characterisation parameter. This is the average

number of substituents per cyclodextrin molecule (for β-CD the maximum TDS is 21).

Hydroxypropyl-β-CD (HP-β−CD) and sulphobutylether-β-CD are FDA approved for

human use (Davis and Brewster, 2004), HP-β−CD being the most commonly used CD for

oral, buccal, intravenous, rectal and ophthalmic applications. The best example is Sporanox®

(itraconazole), which was developed by Janssen Pharmaceutica. Both the oral and intravenous

solutions of Sporanox® are formulated with HP-β−CD, producing solutions of 10 mg/ml for a

molecule that has an aqueous solubility at neutral pH of 1 ng/ml. Other molecules include

cisapride (Propulsid®) and mitomycin (Mitozytrex®) (Loftsson and Duchêne, 2007).

Randomly-methylated-β-CD (RAME-β-CD), has been used to formulate chloramphenicol

(Clorocil®) and β-oestradiol (Aerodiol®) (Loftsson and Duchêne, 2007). The most recent

chemically modified β−CD to come to the market is sulphobutyl-ether-β-CD (SBEβCD)

(Captisol®, Cydex). It has been used by Pfizer to formulate an antifungal agent (voriconazole,

Vfend®) and an antipsychotic agent (ziprasidone, Geodon®) (Loftsson and Duchêne, 2007).

At present, CDs are further investigated for future applications. This not only includes

new applications of existing CDs but also comprises the development of new cyclodextrin

derivatives. Today, CDs are mainly used for low-molecular drugs but new applications for

CDs could improve the formulation and drug delivery of proteins, peptides and

oligonucleotides.

Chapter 2 Introduction 28

Page 55: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.3.3. β-Cyclodextrins and paclitaxel

As discussed previously, β-CDs are the most interesting CDs for pharmaceutical

application as they are capable of including aromatic groups in their hydrophobic cavities.

This makes them also an interesting option for the solubilisation of paclitaxel via the

formation of water soluble complexes. Theoretically one paclitaxel molecule can interact with

3 cyclodextrins molecules based on the accessible aromatic groups in its molecular structure

(Fig. 2). A variety of chemically modified β-CDs has already been investigated for their

solubilising capacity towards paclitaxel (Sharma et al., 1995; Szente, 1999).

Sharma et al. (1995) extensively investigated β- and γ-CDs for complex formation

with paclitaxel, using 2 similar methods to form the complexes. Paclitaxel was either

dissolved in methanol or in tert-butyl alcohol. After the addition of the appropriate amount of

CD the solution was lyophilized. The lyophilized powder was reconstituted in distilled water

to evaluate the water solubility of the Pac inclusion complexes: 2,6-dimethylated-β-

cyclodextrin (DIME-β-CD) induced the largest increase of paclitaxel solubility (about 3

orders of magnitude), whereas Pac solubility only increased 2 orders of magnitude in

combination with HP-β-CD. 50 % DIME-β-CD solutions containing less than 14 mM

paclitaxel showed no precipitation upon dilution in buffer. The Pac/CD formulations also

retained their cytotoxic activity. The authors however concluded that the importance of CDs

for IV paclitaxel administration was marginal because the required amount of CDs for

solubilisation was close to the maximum tolerated dose of the CDs (Sharma et al., 1995).

Szente et al.(1999) confirmed the superior complexing activity of methylated-β-CDs

for paclitaxel. These β-CDs (at a concentration of 200 mg/ml) were also used to prepare an

aqueous ethanol solution of paclitaxel (4 mg/ml). These solutions were stable for 6 months

Chapter 2 Introduction 29

Page 56: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

without drug precipitation and showed no precipitation upon dilution. Furthermore, these

solutions had a similar cytotoxic activity compared to Taxol®.

Based on the promising literature data about the interaction of paclitxel and β-CDs this

type of CD was used in this research project to formulate a water soluble, co-solvent-free and

tensioactive free paclitaxel formulation.

Chapter 2 Introduction 30

Page 57: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2.4. REFERENCES

Adams J.D., Flora K.P., Goldspiel B.R., Wilson J.W., Arbuck S.G., Finley R., 1993. Taxol: A

history of pharmaceutical development and current pharmaceutical concerns. J. Natl.

Cancer Inst., 15, 141-147.

Armstrong D.K., Fleming G.F., Markman M., Bailey H.H., 2006. A phase I trial of

intraperitoneal sustained-release paclitaxel microspheres (Paclimer) in recurrent ovarian

cancer: a Gynecologic Oncology Group study. Gynecol. Oncol., 103, 391-396.

Bissery M.C., Guenard D., Guerrite-Voegelein F., Lavelle F., 1991. Experimental antitumor

activity of taxotere (RP56976, NSC628503) a taxol analogue. Cancer Res., 51, 4845.

Cabanes A., Briggs K.E., Gokhale P.C., 1998. Comparative in vivo studies with paclitaxel

and liposome encapsulated paclitaxel. Int. J. Oncol., 12, 1035-1040.

Ceelen W.P., Hesse U., de Hemptinne B., Pattyn P., 2000. Hyperthermic intratperitoneal

chemoperfusion in the treatment of locally advanced intra-abdominal cancer. Br. J.

Surg., 87, 1006-1015.

Ceelen W., 2006. Sugery for peritoneal carcinomatosis from colorectal origin: techniques and

limitations. Acta Chir. Belg., 106, 276-282.

Chapter 2 Introduction 31

Page 58: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Cella D., Cashy J., 2005. Symptom relief and quality of life (QOL) in PS2 patients with non-

small cell lung cancer (NSCLC) subsequent to treatment with paclitaxel poliglumex

(Xyotax ™): Phase 3 trial results. Lung Cancer, 49, S277-S278.

Chu D.Z.J., Lang N.P., Thompson C., Osteen P.K., Westbrook K.C., 1989. Peritoneal

carcinomatosis in nongynecologic malignancy: a prospective study of prognostic

factors. Cancer, 64, 364-367.

Crile G., 1963. The effect of heat and radiation on cancers implanted in the feet of mice.

Cancer Res., 1963, 23, 372-380.

Date A.A., Nagarsenker M.S., 2008. Parenteral microemulsions: An overview. Int. J. Pharm.,

355, 19-30.

Davis M.E., Brewster M.E., 2004. Cyclodextrin-based pharmaceutics: past, present and

future. Nat. Rev. Drug Discov., 3, 1023-1035.

de Bree E., Witkamp A.J., Zoetmulder F.A.N., 2002. Intraperitoneal Chemotherapy for

colorectal cancer. J. Surg. Oncol., 79, 46-61.

Downing K.H., Nogales E., 1999. Crystallographic structure of tubulin: implications for

dynamic and drug binding. Cell Struct. Funct., 24, 269-275.

Chapter 2 Introduction 32

Page 59: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Elias D., Blot F., El Otmany A., Antoun S., Lasser P., Boige V., Rougier P., Ducreux M.,

2001. Curative treatment of peritoneal carcinomatosis arising from colorectal cancer by

complete resection and intraperitoneal chemotherapy. Cancer, 92, 71-76.

Esquivel J., Sticca R., Sugarbaker P., et al., 2007. Cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy in the management of peritoneal surface malignancies

of colonic orignin: a consensus statement. Ann. Surg. Oncol., 14, 128-133.

Ferlini C., Gallo D., Scambia G., 2008. New taxanes in development. Expert Opin. Investig.

Drugs, 17, 335-347.

Freudenberg K., Cramer F., Plieninger, H., 1953. Verfahren zur Herstellung von

Einschlussverbindungen physiologisch wirksamer organischer Verbindungen. German

Pat., 895, 769.

Fujimoto S., Shrestha R., Kokobun M., Ohta M., Takahashi M., Kobayashi K., Kiuchi S.,

Okui K., Miyoshi T., Arimizu N., Takamizawa H., 1988. Intraperitoneal hyperthermic

perfusion combined with surgery effective for gastric cancer patients with peritoneal

seeding. Ann. Surg., 208, 36-40.

Gelderblom H., Verweij J., van Zomeren D.M., Buijs D., Ouwens L., Nooter K., Stoter G.,

Sparreboom A., 2002. Influence of Cremophor EL on the bioavailability of

intraperitoneal paclitaxel. Clin. Canc. Res., 8, 1237-1241.

Chapter 2 Introduction 33

Page 60: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Glehen O., Mohamed F., Gilly F.N., 2004. Peritoneal carcinomatosis from digestive tract

cancer: new management by cytoreductive surgery and intraperitoneal

chemohyperthermia. Lancet Oncol., 5, 219-228.

Harper E., Dang W.B., Lapidus R.G., Garver R.I., 1999. Enhanced efficacy of a novel

controlled release paclitaxel formulation (Paclimer delivery system) for local-regional

therapy of lung cancer tumor nodules in mice. Clin. Cancer Res., 5, 4242- 4248.

Hennenfent K.L., Govindan R., 2006. Novel formulations of taxanes: a review. Old wine in a

new bottle?. Ann. Oncol., 17, 735-749.

Hassan S., Dhar S., Sandström M., Arsenau D., Budnikova M., Lokot I., Lovanov N.,

Karlsson M.O., Larsson R., Lindhagen E., 2005. Cytotoxic activity of a new paclitaxel

formulation, Pacliex, in vitro and in vivo. Cancer Chemother. Pharmacol., 55, 47-54.

Huizing M.T., Misser V.H. Sewberath, Pieters R.C., Huinink W.W. ten Bokkel, Veenhof

C.H.N., Vermorken J.B., Pinedo H.M., Beijnen, J.H., 1995. Taxanes: a new class of

antitumor agents. Cancer Invest., 13, 381-404.

Koga S., Hamazoe R., Maeta M., Shimizu N., Murakami A., Wakatsuki T., 1988.

Prophylactic therapy for peritoneal recurrence of gastric cancer by continuous

hyperthermic peritoneal perfusion with mitomycin-C. Cancer, 61, 232-237.

Koppe M.J., Boerman O.C., Oyen W.J.G., Bleichrodt R.P., 2006. Peritoneal carcinomatosis of

colorectal origin. Ann. Surg., 243, 212-222.

Chapter 2 Introduction 34

Page 61: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Langer C., O’Byrne K., Ross H., Socinski M., Szczesna A., Dediu M., Orlov S., Smakal M.,

2005. Xyotax ™/carboplatin vs paclitaxel/carboplatin for the treatment of PS2 patients

with chemotherapy-naïve advanced non-small cell lung cancer (NSCLC): the stellar 3

phase III study. Lung Cancer, 49, S36.

Lapidus R.G., Dang W.B., Rosen D.M., Gady A.M., Zabelinka Y., O’Meally R., DeWeese

T.L., Denmeade S.R., 2004. Anti-tumor effect of combination therapy with intratumoral

controlled-release paclitaxel (Paclimer microspheres) and radiation. Prostate, 58, 291-

298.

Lee, K.S., Chung H.C., Im S.A., Park Y.H., Kim C.S., Kim S.-B., Rha S.Y., Ro J., 2008.

Multicenter phase II trial of Genexol-PM, a Cremophor-free, polymeric micelle

formulation of paclitaxel, in patients with metastatic breast cancer. Breast Cancer Res.

Treat., 108, 241-250.

Loftsson T., Brewster M., 1996. Pharmaceutical applications of cyclodextrins. 1. Drug

solubilization and stabilization. J. Pharm. Sci., 85, 1016-1025.

Loftsson T., Duchêne D., 2007. Cyclodextrins and their pharmaceutical applications. Int. J.

Pharm., 329, 1-11.

Markman M., 2003. Intraperitoneal antineoplastic drug delivery: rationale and results. Lancet

Oncol., 4, 277-283.

Chapter 2 Introduction 35

Page 62: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

McGrogan B.T., Gilmartin B., Carney D.N., McCann A., 2008. Taxanes, microtubules and

chemoresistant breast cancer. Biochim. et Biophys. Acta, 1785, 96-132.

McQuellon R.P., Loggie B.W., Fleming R.A., Russel G.B., Lehman A.B., Rambo T.D., 2001.

Quality of life after intraperitoneal hyperthermic chemotherapy (IPHC) for peritoneal

carcinomatosis. E.J.S.O., 27, 65-73.

Nornoo A.O., Chow D. S.-L., 2008. Cremophor-free intravenous micoremulsions for

paclitaxel II. Stability, in vitro release and pharmacokinetics. Int. J. Pharm., 349, 117-

123.

O’Brien M., Sandler A., Popovich A., Ganchev H., Bogdanova N. Miziara J., 2005. Xyotax™

vs gemcitabine or vinorelbine for the treatment of performance status (PS) 2 patients

with chemotherapy-naïve advanced non-small cell lung cancer (NSCLC): the stellar 4

phase III study. Lung Cancer, 49, S37.

Paridaens R., Biganzoli L., Bruning P., Klijn J.G.M., Gamucci T., Houston S., Coleman R.,

Schachter J., Van Vreckem A., Sylvester R., Awada A., Wildiers J., Piccart M., 2000.

Paclitaxel versus doxorubicin as first-line single-agent chemotherapy for metastatic

breast cancer: a European Organization for Research and Treatment of Cancer

randomized study with cross-over. J. Clin. Oncol., 18, 724-733.

Raptopoulos V., Gourtsoyiannis N., 2001. Peritoneal Carcinomatosis. Eur. Radiol., 11, 2195-

2206.

Chapter 2 Introduction 36

Page 63: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Rowinsky E.K., Eisenhauer E.A., Chaudry V., Arbuck S.G., Donehower R.C., 1993. Clinical

toxicities encountered with paclitaxel (Taxol). Semin. Oncol., 20, 1-15.

Rowinsky E.K., Donehower R.C., 1995. Paclitaxel (Taxol). N. Engl. J. Med., 332, 1004-1014.

Rowinsky E.K., Calvo E., 2006. Novel agents that target tublin and related elements. Semin.

Oncol., 33, 421-435.

Schiff P.B., Fant J., Horwitz S.B., 1979. Promotion of microtubule assembly in vitro by taxol.

Nature, 277, 665-667.

Sharma U.S., Balasubramanian S.V., Straubinger R.M., 1995. Pharmaceutical and physical

properties of paclitaxel (Taxol) complexes with cyclodextrins. J. Pharm. Sci., 84, 1223-

1230.

Singla, A.K., Garg A., Aggarwal D., 2002. Paclitaxel and its formulations. Int. J. Pharm., 235,

179-192.

Sparreboom A., van Zuylen L., Brouwer E., Loos W.J., de Bruijn P., Gelderblom H., Pillay

M., Nooter K., Stoter G., Verweij J., 1999. Cremophor EL-mediated alteration of

paclitaxel distribution in human blood: clinical pharmacokinetic implications. Cancer

Res., 59, 1454-1457.

Spratt J.S., Adcock R.A., Muskovin M., Sherrill W., McKeown, 1980. Clinical delivery

system for intraperitoneal hyperthermic chemotherapy. Cancer Res., 40, 256-260.

Chapter 2 Introduction 37

Page 64: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Stewart, J.H., Shen P., Levine E.A., 2005. Intraperitoneal hyperthermic chemotherapy for

peritoneal surface malignancy: current status and future directions. Ann. Surg. Oncol.,

12, 765-777.

Sugarbaker P.H., 1996. Pertioneal carcinomatosis: principles of management. Kluwer

Academic Publishers, Boston.

Sugarbaker P.H., 1999. Management of peritoneal-surface malignancy: the surgeon’s role.

Langenbecks Arch. Surg., 384, 576-587.

Szeltli, J., 1994. Medical applications of cyclodextrins. Med. Res. Rev., 14, 353-386.

Szente L., Vikmon M., Szemán, Otta K., 1999. Methyl-β-cyclodextrin/paclitaxel aqueous

solutions: a tool to test Cremophor-free paclitaxel. STP Pharm. Sci., 9, 243-247.

Tarr, B.D., Sambadan, T.G., Yalkowski, S.H., 1987. A new parenteral emulsion for the

administration of Taxol. Pharm. Res., 4, 162-165.

ten Tije A.J., Verweij J., Loos W.J., Sparreboom A., 2003. Pharmacological effects of

formulation vehicles: implications for cancer chemotherapy. Clin. Pharmacokinet., 42,

665-685.

Chapter 2 Introduction 38

Page 65: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Treat J., Damjanov N., Huang C et al., 2001. Liposomal-encapsulated chemotherapy:

preliminary results of a phase I study of a novel liposomal paclitaxel. Oncology, 15, 44-

48.

Tsiftis D., de Bree E., Romanos J., et al., 1999. Peritoneal expansion by artificially produced

ascites durng perfusion chemotherapy. Arch. Surg., 134, 545-549.

Urano M., Kuroda M., Nishimura Y., 1999. Invited review for the clinical application of

thermochemotherapy given at mild temperatures. Int. J. Hyperthermia, 15, 79-107.

Verwaal V.J., van Ruth S., de Bree E., van Slooten G.W., van Tinteren H., Boot H.,

Zoetmulder F.A.N., 2003. Randomized trial of cytoreduction an hyperthermic

intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in

patient with peritoneal carcinomatosis of colorectal cancer. J. Clin. Oncol., 21, 3737-

3743.

Verwaal V.J., Bruin S., Boot H., van Slooten G., van Tinteren H., 2008. 8-year follow-up of

randomized trial: cytoreduction and hyperhtermic intraperitoneal chemotherapy versus

systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer.

Ann. Surg. Oncol., 15, 2426-2432.

Villiers, A., 1891. Sur la fermentation de la fecule par l’action du ferment butyrique. C. R.

Hebd. Secances Acad. Sci., 112, 536.

Chapter 2 Introduction 39

Page 66: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 2 Introduction 40

Volk K.J., Hill S.E., Kerns E.H., Lee M.S., 1997. Profiling degradants of paclitaxel using

liquid chromatography-mass spectrometry and liquid chromatography-tandem mass

spectrometry substructural techniques. J. Chromatography B, 696, 99-115.

Wani M.C., Taylor H.L., Wall M.E., Coggan P., McPhail A.T., 1971. Plant antitumor agents.

VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from

Taxus Brevifolia. J. Am. Chem. Soc., 93, 2325-2327.

Wheeler, J.J., Wong, K.F., Ansell, S.M., Masin, D., Ball, M.B., 1994. Polyethylene glycol

modified phospholipids stabilize emulsions prepared from triacylglycerol. J. Pharm.

Sci., 83, 1558-1564.

Wiernik P.H., Schwartz E.L., Straumann J.J., et al, 1987. Phase I clinical and pharmacokinetic

study of Taxol. Cancer Res, 47, 2486-2493.

Witkamp A.J., de Bree E., Van Goethem A.R., Zoetmulder F.A.N., 2001. Rationale and

techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer Treat.

Rev., 27, 365-374.

Yonemura Y., Fujimura T., Fushida S., Takegawa S., Kamata T., Katayama K., Kosaka T.,

Yamaguchi A., Miwa K., Miyazaki I., 1991. Hyperthermochemotherapy combined with

cytoreductive surgery for the treatment of gastric cancer with peritoneal dissemination.

World J. Surg., 15, 530-536.

Page 67: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 68: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 41

FORMULATION AND PHYSICAL STABILITY OF

PACLITAXEL/β-CYCLODEXTRIN COMPLEXES

3.1. ABSTRACT

Due to its low aqueous solubility paclitaxel (Pac) is currently formulated in a

Cremophor EL®/ethanol mixture. However, the vehicle of this formulation causes several

side-effects. Our objective was to formulate a tensioactive-free and co-solvent-free paclitaxel

solution, which can be used for a hyperthermic intraperitoneal chemoperfusion procedure

(HIPEC). The potential of chemically modified β-cyclodextrins (β-CDs) to form complexes

with paclitaxel was investigated as a means to increase the aqueous solubility of paclitaxel.

Methylated β-CDs (randomly-methylated and 2,6-dimethylated) showed the best ability to

solubilise paclitaxel compared to sulphobutyl-ether- and hydroxypropyl-β-CD. The minimal

ratio of paclitaxel versus randomly-methylated-β-cyclodextrin (RAME-β-CD) yielding 100 %

inclusion efficiency was 1/20 (mol/mol). Pac/RAME-β-CD inclusion complexes prepared via

freeze drying were stable for at least 6 months when stored at 4 °C. A 5 mg/ml paclitaxel

solution was formulated using Pac/RAME-β-CD-complexes. Upon dilution of these solutions,

no precipitation was seen. After 24 hrs storage at room temperature or 2 hrs at HIPEC

conditions (41 °C) the 1/40 (mol/mol) ratio showed the highest stability at paclitaxel

concentrations of 0.1 and 0.5 mg/ml. When hydroxypropyl methylcellulose (HPMC) was

added to the reconstitution medium, the stability significantly increased, offering the

opportunity to reduce the amount of RAME-β-CDs in the formulation.

33

Page 69: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

3.2. INTRODUCTION

Taxol®, containing paclitaxel as a microtubule stabilizing agent, was approved by the

FDA for the treatment of breast and ovarian cancer in 1992 and 1994, respectively

(Panchagnula, 1998). Unlike most antineoplastic agents, paclitaxel does not depolymerize the

microtubules, but it promotes the polymerization of tubulin. The formed microtubules are

extremely stable and inhibit the normal microtubule dynamics, which are essential for the cell

cycle, thus causing cell death (Rowinsky and Donehower, 1995).

Since paclitaxel has a low aqueous solubility (0.4 µM or 0.34 µg/ml, Sharma et al.,

1995), the drug is currently formulated in a 1:1 (v/v) Cremophor EL®/ethanol mixture,

available as Taxol®. Amongst multiple problems caused by this vehicle, in particular by

Cremophor EL®, the most important side effects include hypersensitivity reactions,

nephrotoxicity and neurotoxicity (Singla et al., 2002). Furthermore, the leaching of the

plasticizer diethylhexylphthalate (DEHP) from polyvinylchloride (PVC) infusion sets into the

Cremophor EL®/ethanol formulation should be considered as a another severe problem.

Therefore, much research is performed to reformulate paclitaxel using the following

approaches: co-solvency, emulsification, micellisation, liposome formation, non-liposomal

lipid carriers (microspheres, nanocapsules), cyclodextrins and local drug delivery devices

(Singla et al., 2002).

β-Cyclodextrins (β-CDs) are cyclic oligosaccharides which consist of 7 covalently

linked glucopyranose units. β-CDs have a cone-like structure, combining a hydrophilic

exterior with a hydrophobic interior which can encapsulate hydrophobic drug molecules or

parts of these molecules. The resulting inclusion complexes increase the aqueous solubility of

the drug. β-CDs are chemically modified to enhance their solubility and their complexing

activity.

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 42

Page 70: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Hyperthermic intraperitoneal chemoperfusion (HIPEC) after debulking surgery is a

novel strategy in the treatment or prevention of peritoneal carcinomatosis. Using this

technique the peritoneum is perfused with a solution (40-43 °C) containing a cytostatic agent.

The rationale for this treatment lies in the combination of the tumoricidal properties of

intraperitoneal hyperthermia (Shiu and Fortner, 1980) and various chemotherapeutic drugs. In

addition, hyperthermia is known to enhance the antitumoral effect of several cytostatic drugs:

doxorubicin (Jacquet et al., 1998), gemcitabine (Pestieau et al., 1998), cisplatin (Bartlett et al.,

1997), tumor necrosis factor (Bartlett et al., 1997), mitomycin C (Storm, 1989). By applying

the drug locally, there is a pharmacokinetic advantage as the clearance of the drug from the

peritoneal cavity is delayed and also higher concentrations of the cytotoxic drug can be

applied. Randomized clinical trials are available to support HIPEC in the treatment and

prevention of peritoneal carcinomatosis following resection of pathological gastric cancer

(Ceelen et al., 2000). Phase I data showed a ≥ 1,000-fold exposure to paclitaxel in the

peritoneal cavity compared with the systemic compartment (Markman, 1995). The dose-

limiting toxicity of intraperitoneal administered paclitaxel was found to be abdominal pain

which could be due to the drug, the vehicle (ethanol/Cremophor EL®-mixture in case of

Taxol®) or both. The latter seems the most plausible, as in recent years it has become clear

that Cremophor EL® is not an inert vehicle, but exerts a range of biological effects, some of

which have important clinical implications (Gelderblom et al., 2001).

The purpose of this study was to formulate a co-solvent- and tensioactive-free

paclitaxel solution, based on chemically modified β-CDs, which can be used for a HIPEC

procedure.

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 43

Page 71: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

3.3. MATERIALS AND METHODS

3.3.1. Materials

Randomly-methylated-β-cyclodextrin (RAME-β-CD) with a total degree of

substitution (TDS) of 13 and 2,6- dimethyl-β-cyclodextrin (DIME-β-CD) with a TDS of 14

were purchased from Cyclolab (Budapest, Hungary). The following cyclodextrins were

donated: sulphobutyl-ether-β-cyclodextrin (SBE-β-CD), TDS 6.5 (Captisol®, Cydex,

Overland Park, USA); hydroxypropyl-β-cyclodextrin (HP-β-CD), TDS 3 (Encapsin®,

Johnson & Johnson, Beerse, Belgium); hydroxypropyl-β-cyclodextrin (HP-β-CD), TDS 4.5

(Kleptose HPB®, Roquette, Lestrem, France); hydroxypropyl-β-cyclodextrin (HP-β-CD),

TDS 5.2 (Cavitron 82005®, Cerestar, Vilvoorde, Belgium); hydroxypropyl-β-cyclodextrin

(HP-β-CD), TDS 6.1 (Cavitron 82006®, Cerestar). Hydroxypropyl methylcellulose (HPMC)

(Metolose® 60SH-4000, Shin-Etsu, Tokyo, Japan) was also donated. Paclitaxel was purchased

from Acros Organics (Geel, Belgium).

3.3.2.Preparation of the inclusion complexes

First the required amount of paclitaxel was dissolved in absolute ethanol (Merck,

Overijse, Belgium), in a ratio of 1 to 60 (w/w). β-Cyclodextrins were added to the solution to

obtain Pac/β-cyclodextrin ratios varying from 1/60 to 1/20 (mol/mol) and the solution was

placed in an ultrasonic bath (Sonis 3 GT, Iskra-Pio, Šentjernej, Slovenia) for 5 min. Next,

phosphate buffered saline (PBS) (10 PBS tablets (Sigma, Bornem, Belgium) in 2 l

demineralised water) was added in a ratio of 1/2 (w/w) versus the CD-fraction. Next, the

solution was placed in an ultrasonic bath for 1 min and afterwards stirred with a magnetic

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 44

Page 72: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

stirrer for 5 min. After evaporating most of the co-solvent from the solution under reduced

pressure by a rotavap, the solution was freeze dried for 24 hrs at -50 °C and at 1 mbar, after

being frozen at -70 °C using solid carbondioxide. After freeze drying, a white amorphous

powder (Alcaro et al., 2002) was obtained.

3.3.3. Paclitaxel determination

The analysis method was based on a method described by Lee et al. (1999). The

HPLC-system consisted of a pump (L-6000, Merck-Hitachi, Tokyo, Japan), an integrator (D-

2000, Merck-Hitachi, Tokyo, Japan), an injector (Vici, Valco Instruments, Houston, USA)

with a loop of 25 µL and a UV/VIS detector (UV 2000, Spectra-systems, Darmstadt,

Germany). Detection was performed at a wavelength of 227 nm. Chromatographic separation

was achieved with a guard column (Lichrospher® 100-RP-18, 4*4 mm (5µm), Merck,

Darmstadt, Germany) and an analytical column (Lichrospher® 100-RP-18, 125*4 mm (5µm),

Merck, Darmstadt, Germany). Before use, the mobile phase consisting of acetonitrile

(Biosolve, Valkenswaard, The Netherlands) and 0.1% (v/v) phosphoric acid in water (Acros

Organics, Geel, Belgium) (42:58, v/v) was degassed by ultrasonication under vacuum. A

calibration curve was validated (inter- and intravariability and precision) for a concentration

ranging from 1 to 100 µg paclitaxel/ml.

3.3.4. Determination of inclusion efficiency

In order to evaluate the inclusion procedure and to measure the amount of paclitaxel

that was complexed with β-cyclodextrins, an amount of freeze dried material, equivalent to 1

mg paclitaxel, was dispersed either in 10 ml PBS or in 10 ml mobile phase used for HPLC-

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 45

Page 73: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 46

analysis. The concentration of dissolved paclitaxel in both samples was determined with the

HPLC-method. The mobile phase dissolved free as well as complexed paclitaxel, whereas

only complexed paclitaxel dissolved in PBS (aqueous solubility of paclitaxel: 0.34 µg/ml).

Therefore, the concentration in the mobile phase was regarded as the 100% value and the

concentration in PBS was expressed relative to the concentration in the mobile phase to

calculate the inclusion efficiency. Thus, any increase in aqueous solubility was considered to

be due to the interaction and formation of complexes between paclitaxel and β-CDs.

3.3.5. Characterisation of Pac/RAME-β-CD complex

3.3.5.1. Thermal analysis

Thermal analysis of the samples was performed via differential scanning calorimetry

(2920 Modulated DSC, TA Instruments, Leatherhead, UK). Samples (approximately 10 mg)

were heated in sealed aluminum pans at 10 °C/min to 400 °C under N2 purge. Both

complexes and physical mixtures of paclitaxel and RAME-β-CDs (molar ratios ranging from

1/20 to 1/60) were analysed.

ead.

3.3.5.2. 1H-NMR

1H NMR spectra of complexes in 1/20 and 1/40 (mol/mol) ratios were obtained in

D2O solution at 25 °C at 700 MHz using a Bruker AVANCE II spectrometer equipped with a

5mm TXI Z-gradient probe h

3.3.6. Physical stability testing

Page 74: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

The short term physical stability of paclitaxel complexes in PBS was tested at room

temperature (25 °C, during 24 h) and at HIPEC temperature (41 °C, during 2 h) for 0.1 and

0.5 mg/ml paclitaxel solutions with or without HPMC. In the experiments where HPMC was

used, the polymer was added to the reconstitution medium in a concentration of 0.1% (w/v).

The solutions were stored in sealed glass vials (pharmaceutical grade) exposed to daylight.

Long term physical stability of a 5 mg/ml solution, stored in the dark, was tested at 4° C for 2

weeks. For all tests, samples, taken at set intervals, were filtered through a 0.22 µm filter

(Spartan 30/0.2 RC (Schleicher & Schuell, Dassel, Germany)) and analysed by HPLC

(following appropriate dilution using PBS). When precipitation was observed in the 5 mg/ml

paclitaxel solution, the samples were centrifuged at 450g for 10 min and aliquots of the

supernatans were analysed by HPLC after filtration using a 0.2 µm filter (Spartan 30/0.2 RC).

3.3.7. Analysis of the freeze dried product

The water and ethanol content of the starting material and the freeze dried products

was determined. The products (200 mg) were analysed for their water content using a Karl

Fisher titrator (Mettler Toledo DL 35, Beerse, Belgium), while their ethanol concentration

was determined using a head-space gas chromatography flame ionization detection (GC-FID)

method. About 20 mg material was dissolved in 0.5 ml H2O, next 0.5 ml H2O (HPLC grade)

and 0.5 ml of an internal standard solution (t-butanol in H2O, 40mg/100 ml) was added before

injection into the GC-FID system.

3.3.8. Statistical Analysis

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 47

Page 75: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Results were analysed with SPSS 14.0 statistical software. For the 24 hrs stability

experiment the effect of the ratio was evaluated using a one-way ANOVA and a Scheffé post

hoc test. The stability data at HIPEC conditions were evaluated using a two-way ANOVA

with pairwise comparisons within each factor, using a Bonferroni correction. For all results

the homogeneity of variances was analysed with the Levene’s test and the normality of the

residuals was checked for by a Kolmogorov-Smirnov test.

3.4. RESULTS AND DISCUSSION

3.4.1. Solubilisation of paclitaxel

3.4.1.1. Selection of the chemically modified β-cyclodextrins

In this research project, different chemically modified β-cyclodextrins were evaluated

in order to obtain a tensioactive- and co-solvent-free paclitaxel formulation based on these β-

cyclodextrins. By reformulating the commercially available Taxol® solution, it was the

intention to reduce the dose-limiting toxicity of Cremophor EL® by replacing it with β-

cyclodextrins, being fully aware that these compounds could also exert toxic effects,

especially renal toxicity. This will be closely monitored during future in vitro and in vivo

experiments.

RAME-β-CD, DIME-β-CD, SBE-β-CD and the different HP-β-CDs were investigated

for their paclitaxel solubilising effect. In a first screening experiment at a fixed cyclodextrin

concentration (1/80 ratio, w/w) the highest amount of paclitaxel in PBS was recovered using

methylated-β-cyclodextrins (approximately 90 % inclusion efficiency). SBE-β-CD and HP-β-

CD had a very low affinity for paclitaxel and induced only a small increase of paclitaxel

solubility (about 2% inclusion efficiency). The degree of substitution of HP-β-CD had a

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 48

Page 76: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

minor effect on paclitaxel inclusion. These results confirmed previous literature reports:

methylated-β-cyclodextrins provided a better enhancement of the aqueous solubility of

paclitaxel compared to HP-β-CD (Sharma et al., 1995; Szente et al., 1999).

As both methylated-β-cyclodextrins produced similar results, RAME-β-CD was

chosen for futher research because of economical reasons (lower cost). Using the inclusion

procedure via freeze drying the amount of RAME-β-CD could be reduced to a molar ratio of

1/20 and still complete inclusion of paclitaxel was obtained. After reconstitution in PBS, all

ratios (1/20 to 1/60, mol/mol) yielded visually clear solutions at a concentration of 1 mg/ml

paclitaxel. At a lower ratio (<1/20), not all paclitaxel molecules (about 60 % inclusion

efficiency) were complexed by RAME-β-CD and no visually clear solutions could be

obtained. The water content of the freeze dried powder varied between 1.4 and 2.2 %,

whereas the original RAME-β-CD material had an average water content of 5.4 %. Due to the

large volumes perfused through the peritoneal cavity during HIPEC and because ethanol is

known to enhance the sedative effects of anaesthetics it is imperative that the ethanol content

(used as a co-solvent during production of the freeze dried material) is kept to a minimum.

The ethanol content of the freeze dried material varied between 2.5 and 3.7 %. At these low

concentrations, no problems are expected during HIPEC.

3.4.1.2. Characterization of Pac/RAME-β-CD complex

Differential Scanning Calorimetry (DSC) was used to identify the complexes in the

freeze dried powder. In contrast to a physical mixture of paclitaxel and RAME-β-CD, no

endothermal melting peak of paclitaxel (at 225 °C) was observed in the freeze dried material,

indicating that inclusion complexes have been formed during processing (Fig. 1). This was

most obvious at the lowest molecular ratio (1/20) as at higher ratios (> 1/40 molar ratio) the

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 49

Page 77: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

sensitivity for the paclitaxel signal was rather low due to the large amount of cyclodextrins in

the formulation.

A

B

C

D

Fig 1.: DSC thermograms of Pac (A), RAME-β-CD (B), a physical mixture (C) of Pac and RAME-β-CD and an inclusion complex (1/20 molar ratio) (D) of Pac and RAME-β-CD. The frame indicates the zone of interest for evaluation of complex formation between Pac and RAME-β-CD.

1H NMR spectroscopy was used to further support complex formation. Given the very

low solubility of paclitaxel in D2O, the fact that a set of clearly visible resonances, distinctive

for the phenylgroups present in paclitaxel (between 7.5 and 8.5 ppm), was observed when

evaluating the freeze dried powder can only be explained by complexation between drug and

CD as uncomplexed paclitaxel (pure and in a physical mixture) did not produce a spectrum

(Fig. 2).

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 50

Page 78: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Paclitaxel

Fig. 2.: 700 MHz 1H NMR spectrum of the 1/20 (mol/mol) inclusion complex. Clearly distinguishable paclitaxel resonances, from the phenylgroups, appear between 7.5 and 8.5 ppm.

3.4.2. Determination of physical stability

To test the physical stability of paclitaxel solutions, the freeze dried material was

reconstituted with PBS. Stability is expressed as the concentration of paclitaxel dissolved in

the solution at the time of sampling compared to the concentration obtained immediately after

reconstitution. As it is the objective of this study to formulate an alternative for Taxol®, which

is available in a concentrated solution of 6 mg paclitaxel/ml, a maximum concentration of 5

mg/ml was chosen for the Pac/RAME-β-CD-formulation. This concentration could be

obtained at all ratios, but using lower molar ratios (< 1/40) precipitation occurred within 48

hrs. The higher ratios (≥ 1/40) remained stable for 2 weeks: after storage at 4 °C and out of

the light, they contained ≥ 95 % of paclitaxel. The improved physical stability of solutions

containing higher amounts of RAME-β-CD is in accordance with data reported in literature

(Szente et al., 1999): cyclodextrins not only enhanced solubility, but also the physical stability

of solutions containing paclitaxel. Precipitation upon dilution can be a major problem when

drugs with a poor aqueous solubility are administered intravenously via an infusion set.

Throughout all experiments precipitation upon dilution did not occur with this

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 51

Page 79: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

paclitaxel/RAME-β-CD formulation at all molar ratios. These observations are in good

agreement with Sharma et al. (Sharma et al., 1995), who reported no precipitation upon

dilution of paclitaxel solutions which have a concentration below 12 mg/ml in 50% DIME-β-

CD, whereas higher paclitaxel concentrations precipitated when diluted. Concentrations

above 12 mg/ml are of no interest for a HIPEC procedure as lower concentrations are

administered due to the large volume used during intraperitoneal perfusion. At all molar ratios

the freeze dried material remained stable for at least 6 months when stored at 4 °C.

As it is the objective to use this RAME-β-CD/Pac formulation for an experimental

HIPEC procedure in rats, the stability of these solutions was evaluated at room temperature

(25 °C, 24 hrs) and at HIPEC conditions (41 °C, 2 hrs). The physical stability results at room

temperature will be an indication whether the solution should be prepared immediately before

administration or can be prepared in advance. Two paclitaxel concentrations (0.1 and 0.5

mg/ml) were selected, taken in consideration the body surface area of a rat (0.03-0.04 m²), the

intraperitoneal dose (± 170 mg/m²) and the volume used for the HIPEC procedure (± 80 ml).

The physical stability of a 0.1 mg/ml paclitaxel solution after 24 hrs storage at room

temperature was poor (Table 1). Only the formulation with a 1/40 molar ratio had a stability

above 50 % (69.5 ± 20.4 %). A lower ratio significantly reduced the physical stability of the

complex, whereas a higher ratio did not result in a significant improvement of stability. At a

paclitaxel concentration of 0.5 mg/ml the physical stability was higher and ranged from 34.4

to 97.0 % (Table 1). However, due to the large variation on these data, it was impossible to

perform any statistical analysis, even after transformation. Taken both concentrations into

consideration the 1/40 molar ratio was the most stable. The reason for this optimum in

drug/RAME-β-CD ratio remains unclear, but might be due to a molecular interaction of

paclitaxel (Balasubramanian et al., 1994). Depending on the drug concentration and the

polarity of the solvent paclitaxel interacts via intermolecular hydrogen bonds, forming a stack

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 52

Page 80: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

of drug molecules in solution. These stacks influence the physical stability because they act as

nuclei which promote aggregation and finally precipitation of paclitaxel. Since precipitation

was the primary cause of instability of these Pac/RAME-β-CD solutions, we assume that this

self-association may be influenced by the β-cyclodextrin concentration in the solution. A

variable degree of self-association could explain the high variability in paclitaxel stability.

Without HPMC With HPMC 0.1 mg/ml 0.1 mg/ml Pac/RAME-β-CD ratio (mol/mol) Stability (%) Stability (%)

1/60 29.2 ± 21.9 97.0 ± 1.1 1/52 13.7 ± 5.1 96.9 ± 1.6 1/46 43.4 ± 22.0 96.8 ± 0.9 1/40 69.6 ± 20.4 96.4 ± 1.0 1/33 14.0 ± 8.7 95.5 ± 0.1 1/26 5.2 ± 3.6 95.1 ± 0.8 1/20 2.9 ± 1.3 94.3 ± 1.1 0.5 mg/ml 0.5 mg/ml Pac/RAME-β-CD ratio (mol/mol) Stability (%) Stability (%)

1/60 95.4 ± 3.5 96.8 ± 14.2 1/52 78.8 ± 31.5 99.0 ± 2.3 1/46 97.0 ± 1.6 99.0 ± 6.2 1/40 94.8 ± 3.6 90.4 ± 10.2 1/33 66.3 ± 53.6 93.9 ± 4.7 1/26 35.1 ± 55.9 93.7 ± 2.9 1/20 34.5 ± 43.5 98.5 ± 4.9

Table 1: Physical stability ± S.D. (n = 3) of Pac/RAME-β-CD solutions, with or without

HPMCa added, stored for 24 hrs in sealed vials at room temperature (25 °C) Pac: paclitaxel; RAME-β-CD: randomly-methylated-β-cyclodextrins; HPMC: hydroxypropylmethyl-cellulose (aMetolose® 60SH-4000).

Stability data at HIPEC conditions showed no interaction between molar ratio and

time, hence both variables were evaluated individually (Fig. 3). At both drug concentrations,

the physical stability at a 1/20 molar ratio was significantly lower compared to the other

evaluated ratios (1/40 and 1/60). For all ratios a significant decrease of stability in function of

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 53

Page 81: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

time was measured, but – similar to the stability data at room temperature – the highest drug

concentrations after 2 hrs at HIPEC conditions were detected for the 1/40 molar ratio.

0.1 mg/ml 0.5 mg/ml

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

A

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

A

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

B

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

B

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

C

020406080

100120

0 30 60 90 120

t (min)

Stab

ility

(%)

C Fig. 3: Physical stability ± S.D. (n = 3) at hyperthermic conditions (41 °C) of aqueous

paclitaxel solutions ( without and with HPMCa added to the reconstitution medium) formulated using different Pac/RAME-β-CD ratios (mol/mol) a. 1/60; b. 1/40; c. 1/20. a Metolose® 60SH-4000.

As it has been reported that HPMC significantly increased the complexation behaviour

of cylcodextrins (even at a HPMC concentration as low as 0.1%, w/v) (Jug and Becirevic,

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 54

Page 82: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2004; Loftsson and Frioriksdottir, 1998), it was evaluated if HPMC might improve the

stability of the complex and reduce the variability of the stability data. Adding 0.1 % (w/v)

HPMC to the medium used to dissolve the freeze dried Pac/RAME-β-CD powder resulted

after 24 hrs at room temperature in paclitaxel concentrations higher than 90 % of the initial

values (Table 1). Even after 10 days no precipitation was seen and more than 70 % of the

drug was recovered (data not shown). In contrast to the solutions without HPMC, there were

no significant differences in stability between the different Pac/RAME-β-CD ratios, offering

the opportunity to lower the amount of cyclodextrins in the formulation. Adding HPMC also

reduced the variability of the stability data (Table 1). Similar to the stability testing without

HPMC, an effect of the drug/CD ratio was seen after 2 hrs at HIPEC conditions (Fig. 3),

independent of the paclitaxel concentration: 1/20 performed significantly worse than 1/40 and

1/60 (although its stability at a concentration of 0.1 mg/ml was also higher than 90 %).

Physical stability of the HPMC-containing formulations was time-dependent in case of

0.1 mg/ml solutions: drug concentrations at each time point which was significantly lower

compared to initial concentration, but the concentrations between 30 and 120 min were not

significantly different. Adding HPMC to the formulation significantly improved stability at 41

°C (except for 0.5 mg/ml solution at a 1/60 ratio), although HPMC could not enterily prevent

the precipitation of paclitaxel at a 1/20 molar ratio. These experiments showed that it was not

necessary to add the hydrophilic polymers during the complexation process, but that adding

the polymer to the reconstitution medium also had a beneficial effect. The exact mechanism

behind the stabilising effect of HPMC is unknown. However, Ribeiro et al. (2005) proposed a

mechanism in which the polymer was able to stabilise the binary drug/CD complex by

hydrogen bonds and van der Waals interactions, thus forming a stable ternary complex.

Although the preparation method in this study (manufacturing binary drug/CD complexes

which were dissolved in reconstitution medium containing HPMC) was different from the

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 55

Page 83: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 56

method used by Ribeiro et al. (2005) (immediate preparation of ternary complexes)

interaction between the drug/CD complexes and HPMC is very likely since the Pac/CD

complexes completely dissolved in the dissolution medium. In addition to the stabilizing

effect, we believe that the binding of the polymer to the binary complex also prevented

precipitation since solutions without HPMC formed a precipitate and HPMC-containing

solutions remained clear (even after 10 days). The limited loss of paclitaxel in solutions with

HPMC was due to chemical degradation, evidenced by the appearance of additional peaks in

the HPLC chromatogram. Based on these observations we assume that the interactions

between the binary complex and the polymer not only stabilised but also prevented the binary

complexes to aggregate and precipitate because of the steric hindrance caused by the polymer.

3.5. CONCLUSION

Complexation of paclitaxel and RAME-β-CDs using a freeze-drying procedure

allowed to formulate tensioactive-free paclitaxel solutions suitable for HIPEC. Both the freeze

dried product and paclitaxel solutions (0.1, 0.5 and 5 mg/ml) were stable during a sufficient

period of time. Addition of HPMC to the reconstitution medium significantly improved the

stability of the Pac/RAME-β-CD formulation, allowing to reduce the amount of RAME-β-CD

in the formulation. This formulation of Pac/RAME-β-CD will be used for further in vitro and

in vivo experiments to evaluate its efficiency and toxicity.

Page 84: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

3.6. REFERENCES

Alcaro S., Ventura C.A., Paolino D., Battaglia D., Ortuso F., Cattel L., Puglisi G., Fresta M.,

2002. Preparation, characterization, molecular modeling and in vitro activity of

paclitaxel-cyclodextrin complexes. Bioorg. Med. Chem. Lett., 12, 1637-1641.

Balasubramanian S.V., Alderfer J.L., Straubinger R.M., 1994. Solvent- and concentration-

dependent molecular interactions of taxol (paclitaxel). J. Pharm. Sci., 9, 243-247.

Bartlett D.L., Ma G., Alexander H.R., Libutti S.K., Fraker D.L., 1997. Isolated limb

reperfusion with tumor nexrosis factor and melphalan in patients with extremity

melanoma after failure of isolated limb perfusion with chemotherapeutics. Cancer, 80,

2084-2090.

Ceelen, W.P., Hesse U., de Hemptinne B., Pattyn P., 2000. Hyperthermic intraperitoneal

chemoperfusion in the treatment of locally advanced intra-abdominal cancer. Br. J.

Surg., 87, 1006-1015.

Gelderblom H., Verweij J., Nooter K., Sparreboom A., 2001. Cremophor EL: the drawbacks

and advantages of vehicle selection for drug formulation. Eur. J. Cancer, 37, 1590-

1598.

Jacquet P., Averbach A., Stuart O.A., Chang D., Sugarbaker P.H., 1998. Hyperthermic

intraperitoneal doxorubicin: pharmacokinetics, metabolism, and tissue distribution in a

rat model. Cancer Chemother. Pharmacol., 41, 147-154.

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 57

Page 85: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Jug M., Becirevic-Lacan M., 2004. Multicomponent complexes of piroxicam with

cyclodextrins and hyrdroxypropyl methylcellulose. Drug Dev. Ind. Pharm., 30, 1051-

1060.

Lee S.-H., Yoo S.D., Lee K.-H., 1999. Rapid and sensitive determination of paclitaxel in

mouse plasma by high-performance liquid chromatography. J. Chromatogr. B, 724,

357-363.

Loftsson T., Frioriksdottir H., 1998. The effect of water-soluble polymers on the aqueous

solubility and complexing abilities of b-cyclodextrin. Int. J. Pharm., 163, 115-121.

Markman M., 1995. Intraperitoneal paclitaxel in the management of ovarian cancer. Semin.

Oncol., 22 (Suppl. 12), 86-87.

Panchagnula R., 1998. Pharmaceutical aspects of paclitaxel. Int. J. Pharm., 172, 1-15.

Pestieau S.R., Stuart O.A., Chang D., Jacquet P., Sugarbaker P.H., 1998. Pharmacokinetics of

intraperitoneal gemcitabine in a rat model. Tumori, 84, 706-711.

Ribeiro L., Carvalho R.A., Ferreira D.C., Veiga F.J.B., 2005. Multicomponent complex

formation between vinpocetine, cyclodextrins, tartaric acid and water-soluble

polymers monitored by NMR and solubility studies. Eur. J. Pharm. Sci., 24, 1-13.

Rowinsky E.K., Donehower R.C., 1995. Paclitaxel (Taxol). N. Eng. J. Med., 332, 1004-1014.

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 58

Page 86: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 3 Formulation and stability of paclitaxel/β-cyclodextrin complexes 59

Sharma U.S., Balasubramanian S.V., Straubinger R.M., 1995. Pharmaceutical and physical

properties of paclitaxel (Taxol) complexes with cyclodextrins. J. Pharm. Sci., 84,

1223-1230.

Shiu M.H., Fortner J.G., 1980. Intraperitoneal hyperthermic treatment of implanted peritoneal

cancer in rats. Cancer Res., 40, 4081-4084.

Singla A.K., Garg A., Aggarwal D., 2002. Paclitaxel and its formulations. Int. J. Pharm., 235,

179-192.

Storm F.K., 1989. Clinical hyperthermia and chemotherapy. Radiol. Clin. North Am., 27,

621-627.

Szente L., Vikmon M., Szemán J., Otta K., 1999. Methyl-b-cyclodextrin/paclitaxel aqueous

solutions: a tool to test Cremophor-free paclitaxel. STP Pharm. Sci., 9, 243-247.

Page 87: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 88: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 60

IN VITRO CYTOTOXICITY OF PACLITAXEL/

β−CYCLODEXTRIN COMPLEXES FOR HIPEC

4.1. ABSTRACT

Hyperthermic intraperitoneal chemotherapy (HIPEC) is a promising strategy in the

treatment of peritoneal carcinomatosis. To perform HIPEC, a tensioactive- and co-solvent-

free paclitaxel formulation consisting of water-soluble paclitaxel/randomly-methylated-β-

cyclodextrin (Pac/RAME-β-CD) complexes was previously developed. Using MTT and SRB

assays the cytotoxic activity of this formulation versus Taxol®, was evaluated as well as the

cytotoxicity of the different formulation excipients (RAME-β-CD and Cremophor EL®). The

possible synergistic effect of heat and paclitaxel-based chemotherapy during HIPEC was also

evaluated in vitro. The cytotoxicity assays revealed differences in viability of 40 and 50 %

between Cremophor EL® and RAME-β-CD treated cells for the CaCo-2 human and the

CC531s rat colon cancer line, respectively, in favour of RAME-β-CD. Despite the higher

cytotoxicity of Cremophor EL®, Pac/RAME-β-CD complexes and Taxol® were equipotent.

Using the MTT and SRB assays the average difference in viability between both cell lines

was below 10 % and IC50 values showed no significant difference. Hyperthermia after drug

administration (41 °C during 1 hr) had no effect on cell viability. These results indicated that

it was possible to reformulate paclitaxel with a less cytotoxic vehicle while maintaining the

cytotoxic activity of the formulation and that there was no synergism between paclitaxel and

heat for in vitro cytotoxicity.

44

Page 89: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

4.2. INTRODUCTION

Peritoneal carcinomatosis (PC) is characterised by metastatic implants on the

peritoneal surface of patients suffering in most cases from intra-abdominal cancer, i.e.

stomach, colon, pancreas or ovarium cancer. For example, after colon cancer resection

approximately 25-35 % of the patients will develop peritoneal carcinomatosis (Minsky et al.,

1988). Due to the ascites, which frequently accompanies peritoneal cancer, the patients have a

very low quality of life combined with a high morbidity and mortality (McQuellon et al.,

2001). PC remains difficult to cure because even extended surgery can never completely

remove all cancer cells embedded in the peritoneum. The median survival of these patients is

approximately one year when treated intravenously with 5-fluorouracil-based chemotherapy

and palliative surgery (Machover, 1997). Hyperthermic intraperitoneal chemotherapy

(HIPEC) after debulking surgery is a strategy in the treatment of this type of cancer. This

treatment does not only allow the use of a higher dosage of the drug because of the

peritoneum-plasma barrier, it also combines the tumoricidal properties of hyperthermia and

various chemotherapeutic drugs. Research has shown a synergism between hyperthermia and

some cytotoxic drugs, including doxorubicin (Jacquet et al., 1998), gemcitabin (Pestieau et

al., 1998), cisplatin (Bartlett et al., 1998), mitomycin C (Teicher et al., 1981) and

immunomodulators like TNF (Bartlett et al., 1997). Randomized clinical trials also supported

the use of HIPEC in the treatment and prevention of PC following resection of pT3 or pT4

gastric cancer (Ceelen et al., 2000).

Paclitaxel (Pac), one of the most potent cancer drugs of recent years, is a good

candidate for HIPEC due to the limited absorption from the peritoneal cavity, its high first-

pass effect in the liver and its activity against ovarian cancer (Witkamp et al., 2001).

However, a major problem of paclitaxel is its low aqueous solubility. This problem is

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 61

Page 90: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

currently overcome by dissolving paclitaxel in a Cremophor EL®/ethanol-mixture (1/1, v/v)

which is commercially available as Taxol®. Since the Cremophor EL® fraction in this

formulation can cause hypersensitivity reactions, patients have to be pre-treated with

corticosteroids and antihistamines when receiving an intravenous administration of Taxol®

(Gelderblom et al., 2001; van Zuylen et al., 2001). Considering that high doses of Cremophor

EL® and ethanol would be administered during a HIPEC procedure (due to the large volume

of perfusion liquid used (1.5 L/m² body surface area) and the high intraperitoneal paclitaxel

concentration required), there was a need for a co-solvent- and tensioactive-free paclitaxel

formulation suitable for HIPEC. Therefore a paclitaxel formulation was developed using

randomly-methylated-β-cyclodextrins (RAME-β-CD), resulting in a new co-solvent- and

tensioactive-free formulation consisting of Pac/RAME-β-CD complexes (Bouquet et al.,

2007). The absence of ethanol is noteworthy because paclitaxel/CD complexes were already

developed, but ethanol was still used in that formulation (Szente et al., 1999).

The current paper investigates the in vitro efficiency of this newly developed

formulation versus Taxol® using a human (CaCo-2) and a rat colon (CC531s) cancer cell line.

In addition, the cytotoxicity of the excipients used in both formulations is compared and the

possible synergism between paclitaxel and heat during application of HIPEC is determined.

4.3. MATERIALS AND METHODS

4.3.1. Materials

Randomly-methylated-β-cyclodextrin (RAME-β-CD) with a total degree of

substitution (TDS) of 13 was purchased from Cyclolab (Budapest, Hungary), paclitaxel (Pac)

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 62

Page 91: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

from Acros Organics (Geel, Belgium), Taxol® from Bristol-Myers Squibb (Brussels,

Belgium) and Cremophor EL® from Alpha Pharma (Waregem, Belgium).

4.3.2. Preparation of the inclusion complexes

The required amount of paclitaxel was dissolved in absolute ethanol (Merck, Overijse,

Belgium), in a ratio of 1 to 60 (w/w). Randomly-methylated-β-cyclodextrin (RAME-β-CD)

was added to the solution to obtain Pac/RAME-β-CD ratios of 1/60, 1/40 and 1/20 (mol/mol)

and the solution was placed in an ultrasonic bath (Sonis 3 GT, Iskra-Pio, Šentjernej, Slovenia)

for 5 min. Next, phosphate buffered saline (PBS, 0.01M, pH 7.4) (1 PBS tablet (Sigma,

Bornem, Belgium) in 200 ml demineralised water) was added in a ratio of 1/2 (w/w) versus

the RAME-β-CD fraction. Next, the solution was placed in an ultrasonic bath for 1 min and

afterwards stirred with a magnetic stirrer for 5 min. After evaporation of most of the co-

solvent under reduced pressure, the solution was frozen at -70 °C using solid carbon dioxide

and freeze dried for 24 h at -50 °C and 1 mbar. After freeze drying, a white amorphous

powder was obtained (Alcaro et al., 2002).

4.3.3. Determination of the administered paclitaxel concentrations

The HPLC-system consisted of a pump (L-6000, Merck-Hitachi, Tokyo, Japan), an

integrator (D-2000, Merck-Hitachi, Tokyo, Japan), an injector (Vici, Valco Instruments,

Houston, USA) with a 25 µL loop and a UV/VIS detector (UV 2000, Spectra-systems,

Darmstadt, Germany). Detection was performed at a wavelength of 227 nm. Chromatographic

separation was achieved with a guard column (Lichrospher® 100-RP-18, 4*4 mm (5µm),

Merck, Darmstadt, Germany) and an analytical column (Lichrospher® 100-RP-18, 125*4 mm

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 63

Page 92: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

(5µm), Merck, Darmstadt, Germany). Before use, the mobile phase consisting of acetonitrile

(Biosolve, Valkenswaard, The Netherlands) and 0.1 % (v/v) phosphoric acid in water (Acros

Organics, Geel, Belgium) (42:58, v/v) was degassed by ultrasonication under vacuum. A

calibration curve was validated for a concentration ranging from 1 to 100 µg paclitaxel/ml.

4.3.4. Cell Culture

The human colon adenocarcinoma cell line, CaCo-2, was obtained from the Institut

National de la Santé et de la Recherche Médicale (Unité 55 Hopital St-Antoine, Paris,

France). The rat colon cancer cell line, CC531s, was obtained from the Laboratory of

Experimental Oncology (University Antwerp, Belgium). Both cell lines were maintained in T

25 culture flasks (Sarstedt, Newton, NC, USA) at 37 °C in a humidified atmosphere

containing 10 % CO2. Dulbecco’s modified medium (DMEM; Invitrogen, Merelbeke,

Belgium), supplemented with 10 % foetal bovine serum (FBS; Invitrogen, Merelbeke,

Belgium), 100 IU/ml penicillin (Invitrogen, Merelbeke, Belgium), 100 µg/ml streptomycin

(Invitrogen, Merelbeke, Belgium) and 2.5 µg/ml fungizone (Bristol Myers Squibb, Brussels,

Belgium), was used as growth medium for both cell lines. For subculturing, cells were rinsed

with Moscona’s solution (8.0 g NaCl; 0.3 g KCl; 0.05 g Na2HPO4.H2O; 0.025 g KH2PO4;

1.0 g NaHCO3 and 2.0 g dextrose in 950 mL distilled water) and were finally detached with

trypsin/EDTA (Invitrogen, Merelbeke, Belgium). The viability of the cells was determined by

their ability to exclude 0.4 % trypan blue dye (Sigma, Bornem, Belgium). All cells were free

of mycoplasma as tested via DAPI staining.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 64

Page 93: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

4.3.5. Cytotoxicity Assays

The cytotoxicity of Taxol® and Pac/RAME-β-CD formulations (1/20, 1/40 and 1/60

mol/mol ratio or 1/30, 1/60 and 1/90 w/w ratio) was tested at paclitaxel concentrations of

0.01, 0.1, 1, 5 and 10 µg/ml (corresponding to 0.012, 0.12, 1.17, 5.86 and 11.72 µM,

respectively). In case of Taxol® these paclitaxel concentrations corresponded to a Cremophor

EL® concentration ranging from 0.880 to 880 µg/ml. In case of Pac/RAME-β-CD complexes

the paclitaxel concentration corresponded to cyclodextrin concentrations varying between 0.3

and 900 µg/ml depending on the molar ratios. The cytotoxicity of RAME-β-CD and

Cremophor EL was also tested separately (without drug) using 10 concentrations: 0, 0.01, 0.1,

0.5, 1, 1.5, 2, 2.5, 3, 3.5 mg/ml.

To evaluate the cytotoxicity, cells were seeded in 96-well plates (Sarstedt, Newton,

NC, USA) at a concentration of 15x10³ and 60x10³ cells/ml for the CC531s and CaCo-2 cell

line, respectively. After 24hrs, 20 µl medium was replaced by 20 µl drug (or excipient)

solution to obtain the required drug (or excipient) concentration. Eight wells per concentration

were used and all experiments (MTT and SRB) were repeated (n = 2). After drug

administration, the plates were incubated for 1hr at normothermic (37 °C) or hyperthermic (41

°C) conditions. Next the cells were incubated for 96 hrs at 37 °C and 10 % CO2. Hereafter,

the cytotoxicity of the formulations was determined via MTT and SRB assays.

4.3.5.1. MTT assay

One hundred microlitres medium was replaced by 100 µl MTT-reagent (3-(4,5-

dimethylthiazol-2-yl)-2,5-difenyl-tetrazolium bromide, at a concentration of 1 mg/ml in PBS-

D+). The reagent was mixed and incubated in dark for 2 hrs at 37 °C. Afterwards all medium

was removed and 200 µl DMSO (Acros Organics, Geel, Belgium) was added to dissolve the

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 65

Page 94: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

formed formazan. After incubation of the plates for 1 hr at 37 °C and mixing, the optical

density was measured at 490 nm using an ELISA reader (Molecular Devices, Sunnyvale,

USA).

4.3.5.2. Sulphorhodamine B (SRB) assay

The SRB test was initiated by fixing the cells via addition of 50 µl of 50 %

trichloracetic acid (Sigma, Bornem, Belgium) to the incubation medium and incubating the

plates for 1 hr at 2 °C. Afterwards, the wells were rinsed five times with water and dried. The

cells were stained with 200 µl SRB (0.4 % in 1 % acetic acid) (Sigma, Bornem, Belgium) for

30 min and rinsed 4 times in 1 % glacial acetic acid (Novolab, Geraardsbergen, Belgium).

After drying the 96 well plate, 200 µl 10 mM Tris buffer (pH 10.5) was added per well to

release the dye. After mixing, the optical density was measured at 490 nm with an ELISA

reader (Molecular Devices, Sunnyvale, USA).

For both assays, cell viability of each well was expressed using the following

equation:

Cell viability (%) = (Absorbancetest cell / Absorbancecontrol cells) x 100

4.3.6. Statistical Analysis

For each individual experiment the 50 % inhibitory drug concentration (IC50) was

determined using probit analysis. To determine the difference in cytotoxicity between RAME-

β-CD and Cremophor EL® the mean cell viability per excipient concentration was determined

(i.e. average of 8 wells). Next, the mean cell viability of RAME-β-CD at a specific

concentration was subtracted from the mean cell viability of Cremophor EL® at the same

concentration. This was done at all 9 concentrations of the formulation excipients (excluding

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 66

Page 95: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

the blank), thus creating a new set of 9 variables. Of this group (n = 9) the overall average and

its 95 % confidence interval was determined. When the cytotoxicity experiments were

repeated (n = 2, performed on different days) the average difference in viability of all

experiments was 6.6 ± 5.1 % and 6.8 ± 6.3 % for the CC531s and CaCo-2 cell line,

respectively. As this percentage was caused by (biological) variation of the experiment (i.e.

the MTT and SRB test), a 10 % difference in cytotoxicity was set as cut-off value to

determine the equivalence in cytotoxicity between RAME-β-CD and Cremophor EL®. When

the difference of the overall average in cell viability between both excipients was less than 10

% (absolute value), their cytotoxic activity was considered equipotent. The same protocol was

followed when comparing normo- versus hyperthermic conditions. In this case the viability at

normothermic conditions was subtracted from the viability at hyperthermic conditions. The

equivalence of the cytotoxic activity of the paclitaxel/RAME-β-CD complex and Taxol®

formulation was analysed using a similar procedure: 5 concentrations (excluding the blank)

were used to determine the difference in overall cytotoxicity of Taxol® versus paclitaxel/

RAME-β-CD complexes. Repeated experiments revealed a biological variation of 6.4 ± 6.1 %

and 8.1 ± 6.7 % for the CC531s and CaCo-2 cell line, respectively. Hence, a 10 % difference

was again chosen as cut-off value to determine equipotency of both formulations. All other

statistical analyses were performed using a one-way ANOVA test (two-sided) and p-values of

less than 5 % were considered statistically significant. Statistical Program for Social Scientists

(SPSS 14.0) was used to analyse the results.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 67

Page 96: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 68

4.4. RESULTS

4.4.1. Cytotoxicity of Cremophor EL® and RAME-β-CD

In a first series of experiments, the cytotoxicity of the formulation excipients was

investigated, in a concentration that ranged from 0.01 to 3.5 mg/ml. The viability graphs (Fig.

1) showed that the cell viability of RAME-β-CD and Cremophor EL® was comparable at the

lowest concentrations (0.01 and 0.1 mg/ml) but at approximately 0.5 mg/ml a sharp drop in

viability was observed in case of Cremophor EL®, reaching a minimum viability at 1.5

mg/ml. At hyper- and normothermic conditions, the viability graphs of RAME-β-CD

indicated a viability of more than 50 % for both cell lines and both staining methods at the

highest concentration (3.5 mg/ml) tested (Fig. 1B, 1C and 1D), except for the SRB staining of

the CaCo-2 cell line with a viability of 50 % at a concentration of 3 mg/ml (Fig. 1A). This

was confirmed by the determination of the IC50 values.

CaCo-2, SRB

0

25

50

75

100

125

0 0,5 1 1,5 2 2,5 3 3,Concentration (mg/ml)

Viab

ility

(%)

5

A

CaCo-2, MTT

0

25

50

75

100

125

0 0,5 1 1,5 2 2,5 3 3Concentration (mg/ml)

Viab

ility

(%)

,5

B

Page 97: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 69

CC531s, SRB

0

25

50

75

100

125

0 0,5 1 1,5 2 2,5 3Concentration (mg/ml)

Viab

ility

(%)

3,5

C

CC531s, MTT

0

25

50

75

100

125

0 0,5 1 1,5 2 2,5 3 3Concentration (mg/ml)

Viab

ility

(%)

,5

D

Fig. 1: Viability graphics (determined via SRB and MTT cytotoxic assays) of the CaCo-2 and CC531s cell line after addition of different concentrations of Cremophor EL and RAME-β-CD at normothermic (37 °C) and hyperthermic (41 °C) conditions (n = 8 wells per concentration, error bars represent SD). ▲ Cremophor EL® 37 °C ■ Cremophor EL® 41 °C ▲ RAME-β-CD 37 °C ■ RAME-β-CD 41 °C. The replicate experiments (n = 2) of the SRB and MTT test are presented as individual data sets, not as mean value.

Compared to RAME-β-CD, the IC50 values for Cremophor EL® at hyper- and

normothermic conditions were much lower for both cell lines. The IC50 values of the SRB

tests for Cremophor EL® were between 0.1 and 0.5 mg/ml for both cell lines. The MTT test

resulted in IC50 values between 0.5 and 1 mg/ml and between 0.3 and 0.7 mg/ml for the

CaCo-2 and CC531s cell line, respectively. No statistical analysis was performed between

RAME-β-CD and Cremophor EL® as the majority of IC50 values for RAME-β-CD were

above the highest concentration tested and could therefore not be included in the statistical

test. The large differences in cytotoxicity were further confirmed by determining the

equivalence of both excipients. Here, the minimum difference in cytotoxicity between both

excipients at 37 °C for both assays was 50.0 and 42.2 % for the CC531s and CaCo-2 cell line,

respectively. Hyperthermia for 1 hr at 41 °C did not increase the cytotoxicity of either

excipient. In case of CaCo-2 cells the largest difference in cytotoxicity between both

temperatures was -7.4 % for the SRB test, whereas these differences ranged between -2.8 and

1.2 % for the MTT test. Similar results were found when investigating the effect of heat using

Page 98: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

the CC531s cell line: the absolute differences between RAME-β-CD and Cremophor EL®

were smaller than 10 % (except for one MTT experiment of RAME-β-CD: 15.7 %). Heat was

unable to create differences larger than the biological variation of the experiment.

4.4.2. Cytotoxicity of Taxol® and paclitaxel/RAME-β-CD complexes

In a next step the cytotoxic activity of the Pac/RAME-β-CD formulations and Taxol®

was evaluated. The procedure, which was used to evaluate the cytotoxicity of the formulations

on the cancer cell lines, was based on the HIPEC procedure intended during in future in vivo

work. Three molar Pac/RAME-β-CD ratios (1/20, 1/40 and 1/60) were tested because a

difference in stability as a function of the Pac/RAME-β-CD ratio was revealed in a previous

study (Bouquet et al., 2007) which might affect the cytotoxicity of these inclusion complexes.

In addition, the highest ratio (1/90, w/w or 1/60, mol/mol) was included as this ratio is similar

to the Pac/Cremophor EL® ratio used in Taxol® (1/88, w/w).

4.4.2.1. CC531s cell line

The viability data of the CC531s cell line revealed small differences in cytotoxic

activity between Taxol® and the different Pac/RAME-β-CD formulations at 37 °C (Fig. 2A):

the average difference in viability varied between -5.0 and 2.3 % for the SRB test (Table 1A)

and between -6.5 and 4.0 % for the MTT test (Table 1B). At 41 °C, the differences in cell

viability between Taxol® and the Pac/RAME-β-CD complexes remained below 10 % (Table

1C and 1D). At both temperatures, the different types of formulation were equivalent (Fig. 2A

and 2B). Heat was unable to significantly increase or decrease the differences in cell viability

between Taxol® and the Pac/RAME-β-CD complexes (p-value of 0.376 and 0.963 for the

SRB and MTT test, respectively). This showed that both formulations were similarly affected

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 70

Page 99: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

by heat. The effect of heat was also studied for each formulation individually. For Taxol®, the

differences in cell viability (Fig. 2C) between the different temperatures (37 vs. 41 °C) were

small: all below 10 % and on average: -0.9 ± 5.6 % and -1.1 ± 2.9 % for the MTT and SRB

test, respectively.

A

B

C

D

-30 -20 -10 10

Fig. 2: Mean (n = 5 concentrations and 8 wells per concentration ) and 95 % confidence interval of the difference in cell viability (determined via SRB (♦) and MTT (■) cytotoxicity assays, n = 2) between Taxol® and Pac/RAME-β-CD at normo- (A) and hyperthermic conditions (B) and between normo- and hyperthermic conditions for Taxol® (C) and Pac/RAME-β-CD (D) for the CC531s cell line. 1/60 (mol/mol) ratio, 1/40 (mol/mol) ratio and 1/20 (mol/mol) ratio.

For Pac/RAME-β-CD (not considering the different ratios) the average difference in

viability (Fig. 2D) between normothermic and hyperthermic treatment was 0.7 ± 4.7 % and

0 20 30

Difference at 37 °C between Taxol® and Pac/RAME-β-CD (%)10 20 -30 -20 -10 0 30

Difference at 41 °C between Taxol® and Pac/RAME-β-CD (%)

-30 -20 -10 0 10 20 30

Difference for Taxol® between 37 °C and 41 °C (%)10 20 -30 -20 -10 0 30

Difference for Pac/RAME-β-CD between 37 °C and 41 °C (%)

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 71

Page 100: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

0.8 ± 1.5 % for the MTT and SRB test, respectively. These observations showed that heat did

not improve the activity of both formulations.

The IC50 values obtained after evaluation of both types of formulation at 37 °C did

not show a difference: all values were smaller or equal than the lowest paclitaxel

concentration (0.01 µg/ml) tested, except for two MTT tests of Taxol® (IC50: 0.03 and 0.05

µg/ml) and for one MTT test of the complexes (IC50: 0.06 µg/ml). The IC50 values at 41 °C

were similar to the values obtained at 37 °C: most IC50 values (± 80 %) were smaller than the

lowest concentration tested (0.01 µg/ml) and could not be taken into account for statistical

analysis to compare between both temperatures.

Cell line Cell assay

Temp. (°C)

Min. meanDifference

(%)

Confidence Interval

(%)

Max. mean Difference

(%)

Confidence Interval

(%) A SRB - 5.0 [- 14.5; 4.5] 2.3 [- 0.4; 4.9] B MTT 37 - 6.5 [- 16.1; 3.2] 4.0 [-16.3; 24.4] C SRB - 2.7 [- 10.8; 5.3] 8.6 [- 11.9; 29.1] D

CC531s

MTT 41 - 4.8 [-13.0; 3.4] - 1.6 [- 8.7; 5.6] E SRB - 9.2 [- 21.2; 2.9] 3.3 [0.5; 6.1] F MTT 37 - 8.5 [- 11.6; -5.5] 3.3 [- 3.1; 9.7] G SRB -10.0 [-14.0; -6.1] 3.7 [-2.2; 9.5] H

CaCo-2

MTT 41 -7.6 [-20.2; 4.9] 7.1 [-2.5; 16.6] Table 1: Minimum and maximum mean (n = 5 concentrations and 8 wells per concentration)

difference in cell viability (determined via MTT and SRB cytotoxicity assays, n = 2) between Pac/RAME-β-CD (all three ratios) and Taxol® at 37 and 41 °C.

4.4.2.2. CaCo-2 cell line

The results (Fig. 3) of the CaCo-2 cell line were similar to the CC531s cell line. All

Pac/RAME-β-CD formulations were equivalent to Taxol® at both temperatures (Fig 3A and

3B). The difference in viability at 37 °C ranged between -9.2 and 3.3 % for SRB (Table 1E)

and between -8.5 and 3.3 % for MTT (Table 1F). The difference in viability at 41 °C for the

SRB test ranged from -10.0 to 3.7 % (Table 1G) and from -7.6 to 7.1 % for the MTT assay

(Table 1H). The differences between Taxol® and Pac/RAME-β-CD were not significantly

different when comparing both temperatures (p-values of 0.885 and 0.383 for SRB and MTT

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 72

Page 101: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

test, respectively). When investigating the influence of temperature on either the activity of

Taxol® or the Pac/RAME-β-CD formulations, the differences were small. For Taxol® (Fig.

3C), the SRB test showed an average difference of 2.4 ± 3.5 % and the MTT test a difference

of -4.0 ± 4.0 %. For the inclusion complexes (Fig. 3D) the average difference was 1.4 ± 7.9

% and -0.9 ± 5.6 % for the SRB and MTT test, respectively.

A

B

C

D

-30 -20 -10 10

Fig. 3: Mean (n = 5 concentrations and 8 wells per concentration) and 95 % confidence intervals of the difference in cell viability (determined via SRB (♦) and MTT (■) cytotoxicity assays, n = 2) between Taxol® and Pac/RAME-β-CD at normo- (A) and hyperthermic conditions (B) and between normo- and hyperthermic conditions for Taxol® (C) and Pac/RAME-β-CD (D) for the CaCo-2 cell line. 1/60 (mol/mol) ratio, 1/40 (mol/mol) ratio and 1/20 (mol/mol) ratio.

The IC50 values of the MTT at 37 °C (Table 2) ranged from 2.76 to 8.59 µg/ml and

from 3.12 to 10.64 µg/ml for Taxol® and Pac/RAME-β-CD formulations, respectively. There

was no significant difference between these groups (p = 0.958). At 41 °C, the values for the

0 20 30

Difference at 37 °C between Taxol® and Pac/RAME-β-CD (%)0 10 20 -30 -20 -10 30

Difference at 41 °C between Taxol® and Pac/RAME-β-CD (%)

-30 -20 -10 0 10 20 30

Difference for Taxol® between 37 °C and 41 °C (%)10 20 -30 -20 -10 0 30

Difference for Pac/RAME-β-CD between 37 °C and 41 °C (%)

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 73

Page 102: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

MTT ranged from 4.77 to 7.72 µg/ml and from 4.29 to 12.90 µg/ml for Taxol® and

Pac/RAME-β-CD formulations, respectively (Table 2). There was also no significant

difference (p = 0.179) between these IC50 values. When comparing the data of Taxol® at 37

°C versus the data at 41 °C to evaluate the influence of heat, no significant difference between

both groups (p = 0.672) could be detected for MTT. The IC50 values of the MTT test also did

not differ significantly (p = 0.227) between both temperatures for the Pac/RAME-β-CD

formulations.

Formulation Temperature (°C)

Cell assay Minimum IC50 (µg/ml)

Maximum IC50 (µg/ml)

MTT 2.76 8.59 37 SRB < 0.01 0.57 MTT 4.77 7.72

Taxol® 41 SRB < 0.01 0.35

MTT 3.12 10.64 37 SRB 0.03 0.42 MTT 4.29 12.90 Pac/RAME-β-CD

41 SRB < 0.01 0.58 Table 2: Minimum and maximum IC50 values (determined via MTT and SRB cytotoxic

assays) of Pac/RAME-β-CD complexes (taking all three ratios into consideration) and Taxol® at 37 °C and 41 °C for the CaCo-2 cell line.

Considering the SRB test, the IC50 values were smaller compared to the MTT test

(some even below the lowest concentration tested, i.e. <0.01 µg/ml). At 37 °C the SRB test

had a maximum IC50 value of 0.57 and 0.42 µg/ml for Taxol® and the Pac/RAME-β-CD

formulations, respectively (Table 2). There was no statistical significant difference between

both formulations (p = 0.450). Similar results were seen at 41 °C: no significant difference

between both formulations (p = 0.906) with maximum IC50 values of 0.35 and 0.58 µg/ml for

Taxol® and Pac/RAME-β-CD, respectively. In function of temperature, no significant

differences between the IC50 values were detected for Taxol® and the Pac/RAME-β-CD

formulations (p-values of 0.573 and 0.791, respectively).

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 74

Page 103: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

4.5. DISCUSSION

Paclitaxel is an essential cytotoxic agent used in the management of different types of

cancer. However, the adverse effects observed after administration of Taxol® are a

considerable drawback for the use of paclitaxel. It has become clear that Cremophor EL®

(essential in the formulation in order to solubilise paclitaxel) is not a physiological inert

compound as numerous pharmacokinetic and pharmacodynamic effects of Cemophor EL®

have been reported in literature (Gelderblom et al., 2001; van Zuylen et al., 2001). Due to

these adverse effects, the development of new paclitaxel formulations, containing a suitable

carrier to improve the aqueous solubility, is essential.

A paclitaxel formulation intended for a HIPEC procedure in the treatment of

peritoneal cancer of colorectal origin was developed using RAME-β-CD (Bouquet et al.,

2007). The difference with a similar approach, used by Szente et al. (1999), was that our

formulation did not contain ethanol. Exclusion of ethanol is essential because of the

synergism with volatile anaesthetics, which may lead to increased fatalities in a future in vivo

model. RAME-β-CD significantly improved the solubility of paclitaxel and the in vitro

stability of the formulation was sufficient to achieve suitable paclitaxel concentrations

(Bouquet et al., 2007). In this study we evaluated the cytotoxicity of this Pac/RAME-β-CD

formulation and compared the cytotoxic activity of RAME-β-CD versus Cremophor EL®.

Based on the results of the MTT and SRB assays it is obvious that Cremophor EL® is

a stronger cytotoxic agent in comparison to RAME-β-CD with average differences in viability

above 40 %. This was also reflected in the IC50 values. The values for RAME-β-CD were in

most cases above 3.5 mg/ml (highest concentration tested), whereas the IC50 values for

Cremophor EL® were all below or equal to 1 mg/ml. This confirmed a study (Szente et al.,

1999) which concluded that methylated-beta-cyclodextrins did not exhibit cytotoxicity on

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 75

Page 104: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

PC3 cancer cell lines in contrast to Cremophor EL®. However, it has been reported that a

concentration of 1-2 mg/ml methylated-beta-cyclodextrins showed cytotoxic effects on human

breast and ovarian adenocarcinoma cell lines (Grosse et al., 1998) which is in contrast to our

data.

The presented data clearly illustrated that Cremophor EL® is more cytotoxic than

RAME-β-CD, although in literature there have been conflicting reports about the cytotoxic

activity of Cremophor EL®. Liebmann et al. (1994) observed that a Cremophor EL®

concentration of 1.35 mg/ml was able to block a portion of the cells in the G1-phase, making

it impossible for paclitaxel to exert its effect during mitosis and because of this effect lower

paclitaxel concentrations (0.043 µg/ml or 50 nM) were as effective as higher concentrations.

In contrast, Cordes and Plasswilm (1998) reported a cytotoxic effect of the diluent used in

Taxol® and Fjällskog et al. (1994) discovered that paclitaxel in Cremophor EL®/ethanol was

more potent against doxorubicin-resistant cell lines than paclitaxel dissolved in ethanol alone,

suggesting an additional cytotoxic effect of Cremophor EL®. The latter was confirmed by

Reinecke et al. (1997), who observed a higher cytotoxicity of Taxol® than paclitaxel dissolved

in DMSO and suggested that this was caused by the additional growth inhibitory effect of

Cremophor EL®. Although it is difficult to compare our data with other literature reports

because of differences in cell lines and administered concentrations, our data showed a clear

cytotoxic effect of Cremophor EL® on both cell lines at a concentration of approximately 1

mg/ml whereas RAME-β-CD is much less cytotoxic with IC50 values above 3.5 mg/ml.

Exposure to heat did not affect cytotoxicity.

When comparing Taxol® versus the Pac/RAME-β-CD complexes, there was no

relevant difference between both formulations since the differences in viability were below 10

%. Although the formulation excipients have a large difference in cytoxicity, both types of

paclitaxel formulations were equipotent, even for the Pac/RAME-β-CD formulation with the

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 76

Page 105: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

lowest ratio (1/20, mol/mol). This shows that the different in vitro stability observed for the

different molar ratios reported in previous research (Bouquet et al., 2007) did not have a

negative effect on the cytotoxicity of the inclusion complexes: all three ratios yielded similar

results in cytotoxicity. For both cell lines, this similarity in cytotoxicity of Taxol® and

Pac/RAME-β-CD was confirmed by the IC50 values. This is in accordance with other

researchers who did not observe a loss in activity of paclitaxel formulated in β-cyclodextrins

(Szente et al., 1999). Our research has proven that although the formulation excipients are

very different in cytotoxicity, the Pac/RAME-β-CD formulation is as cytotoxic as Taxol®. We

hope that these promising in vitro results can be confirmed in vivo: producing a paclitaxel

formulation for intraperitoneal chemotherapy and more specific for HIPEC, that is equipotent

to Taxol® but less toxic. If this new formulation proves to be effective, it could also be

evaluated against a peritoneal cancer of other origin, ovarian cancer being the most relevant.

After investigating the cytotoxicity of the excipients and both formulations, one

question remained unanswered: does hyperthermia have a beneficial effect on the cytotoxicity

of the investigated paclitaxel formulations? Because paclitaxel and hyperthermia both act on

the microtubuli, it is questioned if they have a synergistic or antagonistic effect. Our results

showed no enhancement of the cytotoxic activity of all investigated paclitaxel formulations at

higher temperature using the rat CC531s and the human CaCo-2 cell line, indicating no

synergistic effect of heat nor a loss of activity due to instability of the formulation at higher

temperatures. Rietbroek et al. also reported a lack of thermal enhancement for paclitaxel and

docetaxel (Rietbroek et al., 1997). This was confirmed for docetaxel (Dumontet et al., 1998).

In contrast, Cividalli et al. reported an effect between paclitaxel and hyperthermia in a mouse

tumour model (Cividalli et al., 1999) and Othman et al. confirmed that there was an

hyperthermic enhancement of the apoptotic and antiproliferative activity of paclitaxel

(Othman et al., 2001). More recently, results were published in which no enhancement

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 77

Page 106: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 78

between hyperthermia and paclitaxel was seen, but there was an enhancement for docetaxel

(Faheez et al., 2003). A possible explanation for these contradictory results is the numerous

factors involved in this process. Recent research illustrated that depending on the cell line, the

applied temperature and the number of days after treatment different results were obtained

(Michalakis et al., 2007). At present, there is no consensus on hyperthermia and taxanes, but

our data suggested that there was no thermal enhancement of the cytotoxic activity of

paclitaxel, independent of the type of formulation, on the CaCo-2 and CC531s cell line.

4.6. CONCLUSION

A newly developed Pac/RAME-β-CD formulation has shown a similar cytotoxic

activity as Taxol® against a rat (CC531s) and a human (CaCo-2) colon cancer cell line, as was

confirmed via two cytotoxicity assays (MMT and SRB). For the Pac/RAME-β-CD

formulation there was no difference in cytotoxicity between the different ratios of RAME-β-

CD used. Importantly, a clear difference in cytotoxicity between Cremophor EL and RAME-

β-CD was detected; the latter being the less toxic. There was no effect of applying

hyperthermia (41 °C during 1 hr) on the survival rate of the cells.

Page 107: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

4.7. REFERENCE LIST

Alcaro S., Ventura C.A., Paolino D., Battaglia F., Ortuso F., Cattel L., Puglisi G., Fresta M.,

2002. Preparation, characterization, molecular modeling and in vitro activity of

paclitaxel-cyclodextrin complexes. Bioorg. Med. Chem., 12, 1637–1641.

Bartlett D.L., Ma G., Alexander H.R., Libutti S.K., Fraker D.L., 1997. Isolated limb

reperfusion with tumor necrosis factor and melphalan in patients with extremity

melanoma after failure of isolated limb perfusion with chemotherapeutics. Cancer, 80,

2084–2090.

Bartlett D.L., Buell J.F., Libutti S.K., Reed E., Lee K.B., Figg W.D., Venzon D.J., Alexander

H.R., 1998. A phase I trial of continuous hyperthermic peritoneal perfusion with

tumor necrosis factor and cisplatin in the treatment of peritoneal carcinomatosis.

Cancer, 83, 1251-1261.

Bouquet W., Ceelen W., Fritzinger B., Pattyn P., Peeters M., Remon J.P., Vervaet C., 2007.

Paclitaxel/β-cyclodextrin complexes for hyperthermic peritoneal perfusion –

Formulation and stability. Eur. J. Pharm. Biopharm., 66, 391–397.

Ceelen W.P., Hesse U., de Hemptinne B., Pattyn P., 2000. Hyperthermic intraperitoneal

chemoperfusion in the treatment of locally advanced intra-abdominal cancer. Br. J.

Surg., 87, 1006–1015.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 79

Page 108: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Cividalli A., Cruciani G., Lividi E., Pasqualetti P., Tirindelli Danesi D., 1999. Hyperthermia

enhances the response of paclitaxel and radiation in a mouse adenocarcinoma. Int. J.

Radiat. Oncol. Biol. Phys., 44, 407–412.

Cordes N., Plasswilm L., 1998. Cell line and schedule-dependent cytotoxicity of paclitaxel

(Taxol®): Role of the solvent cremophor EL/ethanol. Anticancer Res., 18, 1851–1857.

Dumontet C., Bodin F., Michal Y., 1998. Potential interactions between antitubulin agents

and temperature: implications for modulation of multidrug resistance. Clin. Cancer

Res., 4, 1563–1566.

Faheez M., Marchettini P., Stuart A., Urano M., Sugarbaker P.H., 2003. Thermal

enhancement of new chemotherapeutic agents at moderate hyperthermia. Ann. Surg.

Oncol., 10, 463–468.

Fjällskog M.L., Frii L., Bergh J., 1994. Paclitaxel-induced cytotoxicity- the effects of

Cremophor EL (castor oil) on two human breast cancer cell lines with acquired

multidrug resistant phenotype and induced expression of the permeability

glycoprotein. Eur. J. Cancer, 30A, 687–690.

Gelderblom H., Verweij J., Nooter K., Sparreboom A., 2001. Cremophor EL: the drawbacks

and advantages of vehicle selection for drug formulation. Eur. J. Cancer, 37, 1590–

1598.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 80

Page 109: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Grosse P.Y., Bressolle F., Pinguet F., 1998. Antiproliferative effect of methyl-β-cyclodextrin

in vitro and in human tumour xenografted athymic nude mice. Br. J. Cancer, 78,

1165–1169.

Jacquet P., Averbach A., Stuart O.A., Chang D., Sugarbaker P.H., 1998. Hyperthermic

intraperitoneal doxorubicin: pharmacokinetics, metabolism, and tissue distribution in a

rat model. Cancer Chemother. Pharmacol., 41, 147–154.

Liebmann J., Cook J.A., Lipschultz C., Teague D., Fisher J., Mitchell J.B., 1994. The

influence of Cremophor EL on the cell cycle effects of paclitaxel (Taxol®) in human

tumor cell lines. Cancer Chemother. Pharmacol., 33, 331–339.

Machover D.A., 1997. A comprehensive review of 5-fluoracil and leucovorin in patients with

metastatic colorectal carinoma. Cancer, 80, 1179–1187.

McQuellon R.P., Loggie B.W., Fleming R.A., Russel G.B., Lehman A.B., Rambo T.D., 2001.

Quality of life after intraperitoneal hyperthermic chemotherapy (IPHC) for peritoneal

carcinomatosis. Eur. J. Surg. Oncol., 27, 65–73.

Michalakis J., Georgatos S.D., de Bree E., Polioudaki H., Romanos J., Georgoulias V., Tsiftis

D.D., Theodoropoulos P.A., 2007. Short-term exposure of cancer cells to micromolar

doses of paclitaxel, with or without hyperthermia, induces long-term inhibition of cell

proliferation and cell death in vitro. Ann. Surg. Oncol., 14, 1220-1228.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 81

Page 110: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Minsky B.D., Mies C., Rich T.A., Recht A., Chaffey J.T., 1988. Potentially curative surgery

of colon cancer: patterns of failure and survival. J. Clin. Oncol., 6, 106–118.

Othman T., Goto S., Lee J.B., Taimura A., Matsumoto T., Kosaka M., 2001. Hypertermic

enhancement of the apoptotic and antiproliferative activities of paclitaxel.

Pharmacology, 62, 208–212.

Pestieau S.R., Stuart O.A., Chang D., Jacquet P., Sugarbaker P.H., 1998. Pharmacokinetics of

intraperitoneal gemcitabine in a rat model. Tumori, 84, 706–711.

Reinecke P., Corvin J., Gabbert H.E., Gerharz C.D., 1997. Antiproliferative effects of

paclitaxel (Taxol®) on human renal clear cell carcinomas in vitro. Eur. J. Cancer, 33,

1122–1129.

Rietbroek R.C., Katschinski D.M., Reijers M.H.E., Robins H.I., Geerdink A., Tutsch K.,

D’Oleire F., Haveman J., 1997. Lack of thermal enhancement for taxanes in vitro. Int.

J. Hyperthermia, 13, 525–533.

Szente L., Vikmon M., Szemán J., Otta K., 1999. Methyl-β-cyclodextrin/paclitaxel aqueous

solutions: a tool to test Cremophor-free paclitaxel. STP Pharm. Sci., 9, 243–247.

Teicher B.A., Kowal C.D., Kennedy K.A., Sartorelli A.C., 1981. Enhancement by

hyperthermia of the in vitro cytotoxicity of mitomycin C toward hypoxic tumor cells.

Cancer Res., 41, 1096–1099.

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 82

Page 111: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 4 In vitro cytotoxicity of paclitaxel/β-cyclodextrin complexes for HIPEC 83

van Zuylen L., Verweij J., Sparreboom A., 2001. Role of formulation vehicles in taxane

pharmacology. Invest. New Drugs, 19, 125–141.

Witkamp A.J., de Bree E., Van Goethem A.R., Zoetmulder F.A.N., 2001. Rationale and

techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer

Treat. Rev., 27, 365–374.

Page 112: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 84

PDMP ANALOGUE RESENSITIZES MDR CELLS

FOR PACLITAXEL/β-CYCLODEXTRIN

COMPLEX

5.1. ABSTRACT

As multi drug resistant (MDR) cells have higher levels of glucosylceramide, the

addition of ceramide or glucosylceramide synthase (GCS) inhibitors is a possible strategy for

resensitization of MDR cells. A previously developed paclitaxel/randomly methylated-β-

cyclodextrin (Pac/RAME-β-CD) formulation was evaluated versus Taxol® for its cytotoxic

potency using MTT and SRB assays. The different formulation excipients (RAME-β-CD and

Cremophor EL®) were also evaluated. The results showed that Cremophor EL® contributed to

the cytotoxic activity of Taxol®, whereas Pac/RAME-β-CD produced similar results as

paclitaxel dissolved in DMSO. Cell viability after exposure to Pac/RAME-β-CD was not

concentration dependent due to the presence of P-glycoprotein (P-gp) efflux pumps in the

CC531s cell line which induced the MDR.

A recently developed D-threo-(1R,2R)-1-phenyl-2-decanoylamino-3-morpholino-1-

propanol (PDMP) analogue and GCS inhibitor (HPPP) was tested for its ability to resensitize

the MDR cells. Addition of 5 µM HPPP was sufficient to generate a concentration-dependent

decrease of cell viability after administration of Pac/RAME-β-CD complexes. The addition of

C6-ceramide at a concentration of 10 µM produced similar results, whereas addition of

verapamil (P-gp inhibitor) and NB-DNJ (selective GCS inhibitor) showed no effect on the

cytotoxic activity of Pac/RAME-β-CD. These results indicated that Cremophor EL®

55

Page 113: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

contributed to the activity of Taxol® and that the addition of HPPP (non-selective GCS

inhibitor) resensitized MDR cells. This resensitization, however, was not related with the

inhibition of P-gp pumps.

5.2. INTRODUCTION

Multidrug resistance (MDR) is a major obstacle in the clinical application of various

cytotoxic drugs. It is characterized by the ability of tumours to exhibit simultaneous resistance

to structurally and functionally unrelated chemotherapeutic agents. The best known and

studied mechanism of MDR is the overexpression of a transmembrane protein, P-glycoprotein

(P-gp) which is encoded by the MDR1 gene (Ueda et al., 1986). P-gp is a member of the

ATP-dependent membrane transport proteins and causes the efflux of chemotherapeutics

(including anthracyclines, taxanes, vinca-alkaloids) resulting in a reduced intracellular drug

concentration and thus reduced antitumour activity (Nobili et al., 2006).

In 1996, Lavie et al. demonstrated that MDR cells also have higher glucosylceramide

levels, indicating a correlation between MDR and (glucosyl)ceramide metabolism. Ceramide

is a lipid second messenger involved in several cell regulating processes, such as

differentiation, proliferation and apoptosis (Kolesnick et al., 1998). Because of its role in

apoptosis, this sphingolipid has attracted a lot of attention as a possible target for cancer

therapy. There is a growing body of evidence that combining paclitaxel and ceramide

increases the cytotoxic potency of paclitaxel (Myrick et al., 1999; Mehta et al., 2000; Charles

et al., 2001; Qiu et al., 2006), even in multidrug resistant cells (Devalapally et al., 2007; van

Vlerken et al., 2007; Desai et al., 2008; Swanton et al., 2008). Hence increasing cellular

ceramide levels via co-administration of ceramide or via inhibition of ceramide metabolism

(via addition of glucosylceramide synthase inhibitors) has been proposed as another way to

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 85

Page 114: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

increase the cytotoxic activity of anticancer drugs (Lavie et al., 1997; Lucci et al., 1999; Liu

et al., 2001). The inhibitors of glucosylceramide synthase (GCS) can be divided into two

classes: D-threo-(1R,2R)-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) and

N-butyldeoxynojirimycin (NB-DNJ) and their respective analogues.

Paclitaxel (Pac) is a powerful chemotherapeutic that has a special place in the anti-

cancer therapy because of its unique mechanism which consists of the stabilization of the

microtubuli. Unfortunately, the clinical application of paclitaxel is hampered by its poor

solubility in water. Therefore, it is formulated in a Cremophor EL®/ethanol (1/1, v/v) solution,

which is commercially available as Taxol®. However, this formulation causes several

problems, the most important being hypersensitivity reactions (Singla et al., 2002), and

formulation development of paclitaxel has focused on alternative excipients to improve the

aqueous solubility of paclitaxel, one of these alternatives being cyclodextrins.

Cyclodextrins are cyclic oligosaccharides, which increase the solubility and

bioavailability of numerous hydrophobic compounds. In previous research (Bouquet et al.,

2007), randomly-methylated-β-cyclodextrins (RAME-β-CD) have been used to form

complexes with Pac, which after reconstitution in phosphate buffered saline render a

paclitaxel solution that can be used in hyperthermic intraperitoneal chemotherapy (HIPEC).

This formulation showed a similar activity as Taxol® against two colon cancer cell lines

(CC531s and CaCo-2) during in vitro cytotoxicity tests (Bouquet et al., 2009). In this research

we have further investigated the activity of our formulation against the CC531s cell line, a rat

colon cancer cell line. After the detection of a P-gp-positive subpopulation in the CC531s cell

line, co-administration of Pac and HPPP, a PDMP analogue (Hillaert et al., 2006) was

investigated as a way to enhance the cytotoxic activity of the Pac/RAME-β-CD formulation.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 86

Page 115: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

5.3. MATERIALS AND METHODS

5.3.1. Materials

Randomly-methylated-β-cyclodextrin (RAME-β-CD) with a total degree of

substitution (TDS) of 13 was purchased from Cyclolab® (Budapest, Hungary), paclitaxel

(Pac) from Acros Organics (Geel, Belgium), Taxol® from Bristol-Myers Squibb (Brussels,

Belgium), Cremophor EL® from Alpha Pharma (Waregem, Belgium), R(+)-verapamil.HCL

(Verapamil) from Sigma-Aldrich (Bornem, Belgium), N-butyldeoxynojirimycin.HCl (NB-

DNJ) from Tebu-Bio (Boechout, Belgium), C6-ceramide from Sigma-Aldrich (Bornem,

Belgium). N-((E,2R,3R)-3-Hydroxy-5-phenyl-1-(pyrrolidin-1-yl)pent-4-en-2-yl)palmitate

(C31H52O2N2) (HPPP, Fig. 1) was synthesised at the Laboratory of Medicinal Chemistry,

Ghent University (Hillaert et al, 2006).

Fig.1: Molecular structure of HPPP

5.3.2. Preparation of the inclusion complexes

The required amount of paclitaxel was dissolved in absolute ethanol (Merck, Overijse,

Belgium), in a 1/60 (w/w) ratio. RAME-β-CD was added to the solution to obtain a

Pac/RAME-β-CD ratio of 1/40 (mol/mol) and the solution was placed in an ultrasonic bath

(Sonis 3 GT, Iskra-Pio, Šentjernej, Slovenia) for 5 min. Next, phosphate buffered saline (PBS,

0.01M, pH 7.4) (Sigma, Bornem, Belgium) was added in a 1/2 (w/w) ratio versus the RAME-

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 87

Page 116: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

β-CD fraction. This solution was placed in an ultrasonic bath for 1 min and afterwards stirred

with a magnetic stirrer for 5 min. After evaporation of most of the co-solvent under reduced

pressure, the solution was frozen at -70°C using solid carbon dioxide and freeze dried for 24

hrs at -50°C and 1 mbar. After freeze drying, a water-soluble powder was obtained.

5.3.3. Determination of paclitaxel concentration

To determine the Pac concentration in the formulations used during the cytotoxicity

studies the following HPLC-system was used: a pump (L-6000, Merck-Hitachi, Tokyo,

Japan), an integrator (D-2000, Merck-Hitachi, Tokyo, Japan), an auto sampler (L-7200,

Merck-Hitachi, Tokyo, Japan) with a 25 µL loop and a UV/VIS detector (L-4200, Merck-

Hitachi, Tokyo, Japan). Detection was performed at a wavelength of 227 nm.

Chromatographic separation was achieved with a guard column (Lichrospher® 100-RP-18,

4*4 mm (5µm), Merck, Darmstadt, Germany) and an analytical column (Lichrospher® 100-

RP-18, 125*4 mm (5µm), Merck, Darmstadt, Germany). The mobile phase consisting of

acetonitrile (Biosolve, Valkenswaard, The Netherlands) and 0.1% (v/v) phosphoric acid in

water (Acros Organics, Geel, Belgium) (42:58, v/v) was degassed by ultrasonication under

vacuum. A calibration curve was validated for a concentration range from 1 to 100 µg

paclitaxel/ml.

5.3.4. Cell Culture

The rat colon cancer cell line, CC531s, was obtained from the Laboratory of

Experimental Oncology (University Antwerp, Belgium). The cell line was maintained in T 25

culture flasks (Sarstedt, Newton, NC, USA) at 37°C in a humidified atmosphere containing

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 88

Page 117: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

10% CO2. Dulbecco’s modified medium (DMEM; Invitrogen, Merelbeke, Belgium),

supplemented with 10% foetal bovine serum (FBS; Invitrogen, Merelbeke, Belgium), 100

IU/ml penicillin (Invitrogen, Merelbeke, Belgium), 100 µg/ml streptomycin (Invitrogen,

Merelbeke, Belgium) and 2.5 µg/ml fungizone (Bristol Myers Squibb, Brussels, Belgium),

was used as growth medium. All cells were tested free of mycoplasma with DAPI staining.

5.3.5. Cytotoxicity Assays

The cytotoxicity of Taxol® and the Pac/RAME-β-CD formulation (1/40 mol/mol ratio

or 1/60 w/w ratio) was tested using the following paclitaxel concentrations: 0.1, 1, 5, 10, 20

µg/ml (corresponding with a range of 0.12 to 23.44 µM). Considering the Pac concentration

range, this corresponded to a Cremophor EL® concentration ranging from 8.8 to 1760 µg/ml

and to a RAME-β-CD concentration varying between 6.0 and 1200 µg/ml. The cytotoxicity of

RAME-β-CD and Cremophor EL was also tested (without Pac) using 10 concentrations: 0,

0.01, 0.1, 0.5, 1, 1.5, 2, 2.5, 3 and 3.5 mg/ml. When a additional compound (C6-ceramide,

HPPP, verapamil) was added to test its synergism on cytotoxicity, Pac concentrations of 1, 5,

10 and 20 µg/ml were used.

To evaluate the cytotoxicity, cells were seeded in 96-well plates (Sarstedt, Newton,

NC, USA) at a concentration of 15x10³ CC531s cells/ml. After 72 hrs, 20 µl medium was

replaced by 20 µl drug (or excipient) solution to obtain the required drug (or excipient)

concentration. Eight wells per concentration were used and all experiments (MTT and SRB)

were performed in duplicate. Next, the cells were incubated for 48 hrs at 37°C and 10% CO2.

The addition of heat was investigated by placing the cells at 41 ° for 1 hr after administration

before placing them back at 37 °C.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 89

Page 118: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Since C6-ceramide and HPPP were dissolved in DMSO before addition to the drug

solution, each experiment included a DMSO blank which contained the DMSO concentration

used to dissolve the investigated molecule. No toxicity was detected at all DMSO

concentrations used. The cytotoxicity was determined via MTT and SRB assays.

5.3.5.1. MTT assay

One hundred µl medium was replaced by 100 µl MTT-reagent (3-(4,5-

dimethylthiazol-2-yl)-2,5-difenyl-tetrazolium bromide, at a concentration of 1 mg/ml in PBS-

D+). The reagent was mixed and incubated in dark for 2 hrs at 37°C. Afterwards all medium

was removed and 200 µl DMSO (Acros Organics, Geel, Belgium) was added to dissolve the

formed formazan. After incubation of the plates for 1 hr at 37°C and mixing, the optical

density was measured at 490 nm using an ELISA reader (Molecular Devices, Sunnyvale,

USA).

5.3.5.2. Sulphorhodamine B (SRB) assay

Cells were fixed by adding 50 µl of 50% trichloracetic acid (Sigma, Bornem,

Belgium) to the incubation medium and incubating the plates for 1 hr at 2°C. Afterwards, the

wells were rinsed five times with water and dried. The cells were stained with 200 µl SRB

(0.4% in 1% acetic acid) (Sigma-Aldrich, Bornem, Belgium) for 30 min and rinsed 4 times in

1% glacial acetic acid (Novolab, Geraardsbergen, Belgium). After drying the 96 well plate,

200 µl 10 mM Tris buffer (pH 10.5) was added per well to release the dye. After mixing, the

optical density was measured at 490 nm with an ELISA reader (Molecular Devices,

Sunnyvale, USA).

For both assays, cell viability of each well was expressed using the following

equation: Cell viability (%) = (Absorbancetest cells / Absorbancecontrol cells) x 100

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 90

Page 119: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

5.3.6. MDR Analysis

MDR analysis was performed as previously described by Swerts et al. (2004). Flow

cytometric detection of P-gp activity was performed using a rhodamine 123 (rho 123) efflux

assay. Cells were incubated for 1 hr at 37°C with 200 ng/ml rho123 (Sigma-Aldrich, Bornem,

Belgium) with or without 10 µM of the P-gp inhibitor R(+)-verapamil.HCL (Sigma-Aldrich,

Bornem, Belgium). The cells were washed twice and resuspended in medium without rho 123

in the presence or absence of verapamil. After incubation of 1 hr at 37°C, the intracellular

rho123 concentration was measured with a flow cytometer (FACSort, BD Biosystems,

Erembodegem, Belgium). At least 10,000 events were counted and analyzed with the Cell

Quest software (BD Biosystems, Erembodegem, Belgium). From this data, the retention ratio

was calculated by dividing the median fluorescence of the cells incubated with verapamil by

the median fluorescence of the cells incubated without verapamil. A test result was considered

positive if the cells exhibited rho 123 efflux and if the intracellular rho 123 fluorescence was

enhanced in the presence of verapamil by at least 10 % (resulting in a rho 123 retention ratio

(RR) of at least 1.10) (Wuchter et al., 2000). For testing the influence of the different added

compounds on P-gp activity, the same procedure was followed as described above, only

verapamil was replaced by the compound under investigation.

5.3.7. Statistical Analysis

For each individual experiment the 50% inhibitory drug concentration (IC50) was

determined using probit analysis. To determine the difference in cytotoxicity between RAME-

β-CD and Cremophor EL® the mean cell viability per excipient concentration was determined

(i.e. average of 8 wells). Next, the mean cell viability of RAME-β-CD at a specific

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 91

Page 120: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

concentration was subtracted from the mean cell viability of Cremophor EL® at the same

concentration. This was done at all 9 concentrations of the formulation excipients (excluding

the blank), thus creating a new set of 9 variables. Of this group (n=9) the overall average and

its 95% confidence interval was determined. When the difference of the overall average in

cell viability between both excipients was less than 10% (absolute value), their cytotoxic

activity was considered equipotent (similar to the method used by Bouquet et al., 2009).

The equivalence of cytotoxic activity between paclitaxel/RAME-β-CD complexes

and Taxol® was analysed using a similar procedure: 5 concentrations (excluding the blank)

were used to determine the difference in overall cytotoxicity of Taxol® versus

paclitaxel/RAME-β-CD complexes, using a 10% difference as cut-off value to determine

equipotency of both formulations. This evaluation method indicates if both formulations are

equipotent over a fixed concentration range. Statistical Program for Social Scientists (SPSS

15.0) was used to analyse the results.

5.4. RESULTS

5.4.1 Comparison of cytotoxicity of Pac/RAME-β-CD and Taxol®

Both the MTT and SRB tests (Fig. 2) indicated a difference in viability of the CC531s

cells after exposure to Taxol® and Pac/RAME-β-CD, mainly at concentrations above 5 µg/ml.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 92

Page 121: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 93

SRB

0

25

50

75

100

125

0 2 4 6 8 10 12 14 16 18 20

Concentration (µg/ml)

Viab

ility

(%)

A

MTT

0

25

50

75

100

125

0 2 4 6 8 10 12 14 16 18 20

Concentration (µg/ml)

Via

bilit

y (%

)

B

Fig. 2: Viability graphics (determined via SRB and MTT cytotoxic assays) of the CC531s cell line after addition of different concentrations of Taxol®, Pac/RAME-β-CD and Pac dissolved in DMSO (Pac/DMSO) at 37°C (n = 8 wells per concentration, error bars represent standard deviations). The replicate experiments (n=2) of the SRB and MTT test are presented as individual data sets. ■ Pac/RAME-β-CD, ■ Taxol®, ■ Pac/DMSO

Whereas both formulations showed a similar response in the lower concentration

range (< 5µg/ml), an increasing difference was observed at higher concentrations. At 20

µg/ml Taxol® reduced the viability to about 10 % whereas the viability after treatment with

Pac/RAME-β-CD remained above 50 %. This was also reflected in the IC50 values (Table 1).

IC50 (µg/ml) after treatment with Cell assay Conc (µg/ml) Taxol® Pac/RAME-β-CD Pac/DMSO

4.11 > 20.00 > 20.00 MTT 0.10-20.00 3.85 > 20.00 > 20.00 1.86 > 20.00 > 20.00 SRB 0.10-20.00 2.03 8.15 > 20.00

Table 1: IC50 values (determined via MTT and SRB cytotoxic assays) of Taxol®,

Pac/RAME-β-CD and Pac dissolved in DMSO (Pac/DMSO). The cell viability was determined at 37 °C using the CC531s cell line. The replicate experiments (n = 2) of the SRB and MTT assay are presented as individual data sets.

For the MTT and SRB tests the IC50 values of Taxol® ranged between 1.86 and 4.11

µg/ml, whereas the IC50 values for Pac/RAME-β-CD were above 20 µg/ml (except for one

SRB staining: 8.15 µg/ml). Hyperthermia had no influence on these results as the IC50 values

Page 122: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 94

of Pac/RAME-β-CD remained above 20 µg/ml (except for one SRB staining: 13.36 µg/ml )

and IC50 of Taxol® ranged between 0.37 and 3.52 µg/ml for the MTT and SRB assays (Table

2).

IC50 (µg/ml) after treatment with Cell assay Conc (µg/ml) Taxol® Pac/RAME-β-CD Pac/DMSO

3.31 > 20.00 > 20.00 MTT 0.10-20.00 0.37 > 20.00 > 20.00 3.52 13,36 > 20.00 SRB 0.10-20.00 1.52 > 20.00 > 20.00

Table 2: IC50 values (determined via MTT and SRB cytotoxic assays) of Taxol®,

Pac/RAME-β-CD and Pac dissolved in DMSO (Pac/DMSO). The cell viability was determined at 41 °C using the CC531s cell line. The replicate experiments (n = 2) of the SRB and MTT assay are presented as individual data sets.

Pac dissolved in DMSO (Pac/DMSO) was also tested in the same concentration range

(0.10 – 20. µg/ml) (Fig 2). This resulted in similar viability graphs as for the Pac/RAME-β-

CD complex and IC50 values of Pac/DMSO were above 20 µg/ml for both cell assays and at

both temperatures (Table 1, Table 2). In contrast to Taxol®, both Pac/RAME-β-CD and

Pac/DMSO did not induce a concentration-dependent decrease of viability. Although the

cytotoxicity of both formulation excipients (RAME-β-CD and Cremophor EL®) was

previously reported, their effect on the CC531s cell line was also determined using the test

procedure applied in this study. Here, the previously reported results (Bouquet et al., 2008)

were confirmed as the IC50 values of RAME-β-CD and Cremophor EL® were > 3.5 mg/ml

and ≤ 0.5 mg/ml, respectively.

As the cytotoxicity of Pac/RAME-β-CD was concentration-independent, the CC531s

cells were examined for P-gp activity with a Rho123 efflux assay. The flowcytometric results

yielded a retention ratio of 5.44, indicating that the cells were P-gp positive (Fig. 3). To rule

out the possibility that this resistance against paclitaxel was recently acquired by the CC531s

cells and therefore unobserved in the previous study, the Pac/RAME-β-CD formulation and

Page 123: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Taxol® were tested using the same test setup as reported by Bouquet et al. (2009) (i.e. lower

cell concentration and longer contact period between formulation and cells). These results

(data not shown) were consistent with those results published by Bouquet et al. (2009):

Taxol® and Pac/RAME-β-CD were equipotent as a similar cytotoxicity was observed over the

entire paclitaxel concentration range.

Fig. 3: Flowcytometric assay of Rhodamine 123 efflux in the CC531s cell line. Cells

incubated without verapamil, Cells incubated with verapamil.

5.4.2 Modulation of multidrug resistance

To increase the activity of the Pac/RAME-β-CD formulation in order to obtain a

similar efficiency compared to Taxol®, the effect of the addition of different compounds to

Pac/RAME-β-CD was investigated. Taxol® was included as a reference in each experiment

investigating the effect of a specific compound: its average IC50 values of all MTT and SRB

tests (n = 32) were 12.59 ± 4.66 and 7.47 ± 3.75 µg/ml, respectively. Increasing

concentrations of HPPP (0.5, 5 and 50 µM) significantly decreased IC50 values of the

Pac/RAME-β-CD formulation in a concentration-dependent manner (Table 3). Addition of 5

µM HPPP significantly reduced the IC50 of the inclusion complex to approximately 9 µg/ml

(MTT assay), which is similar to the one reported for Taxol®. The cytotoxic effect of HPPP

alone was limited at 5µM: cell viability was approximately 80 % and IC50 values were

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 95

Page 124: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

between 22.10 and 26.00 µM for the MTT test. However, as increasing the HPPP

concentration to 50 µM resulted in IC50 values below the lowest paclitaxel concentration

tested, it is obvious that at this concentration the cytotoxicity of HPPP was predominant. The

results obtained for the SRB assay confirmed the MTT results (Table 3).

IC50 (µg/ml) determined via Compound Conc (µM) MTT MTT SRB SRB 0.00 > 20.00 > 20.00 > 20.00 > 20.00 0.50 > 20.00 > 20.00 > 20.00 20.00 5.00 9.00 9.75 1.55 2.81 HPPP

50.00 < 1.00 < 1.00 < 1.00 < 1.00 0.00 > 20.00 > 20.00 > 20.00 20.00 0.50 > 20.00 > 20.00 > 20.00 > 20.00 5.00 > 20.00 > 20.00 > 20.00 > 20.00 Verapamil

50.00 > 20.00 > 20.00 > 20.00 19.32 0.00 > 20.00 > 20.00 > 20.00 > 20.00 0.50 > 20.00 > 20.00 16.35 > 20.00 5.00 > 20.00 > 20.00 16.01 > 20.00 NB-DNJ

50.00 > 20.00 > 20.00 17.83 > 20.00 0.00 > 20.00 > 20.00 > 20.00 > 20.00 10.00 < 1.00 9.26 < 1.00 1.64 30.00 < 1.00 < 1.00 < 1.00 < 1.00 C6-Ceramide

50.00 < 1.00 < 1.00 < 1.00 < 1.00

Table 3: IC50 values (determined via MTT and SRB cytotoxic assays) of Pac/RAME-β-CD formulation combined with different compounds to evaluate their effect on its cytotoxic activity. The cell viability was determined at 37 °C using the CC531s cell line in a Pac concentration range of 1.00 -20.00 µg/ml. The replicate experiments (n = 2) of the SRB and MTT assay are presented as individual data sets.

Figure 4 shows the average difference in viability between Pac/RAME-β-CD and

Taxol® over the entire paclitaxel concentration range (1-20 µg/ml). Without HPPP added to

the Pac/RAME-β-CD formulation or when 0.5 µM HPPP was added, the difference between

both formulations was above 20 %. However, this was reduced to less than 10 % (indicating

equipotent formulations) when 5 µM HPPP was used, demonstrating the effect of HPPP to

enhance the activity of paclitaxel. At a concentration of 50 µM HPPP Pac/RAME-β-CD was

more cytotoxic than Taxol®, but this was caused by the cytotoxic activity of HPPP itself.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 96

Page 125: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

-80

-60

-40

-20

0

20

40

60

80

0.00 0.50 5.00 50.00

Added concentration (µM)

Diff

eren

ce in

via

bilit

y (%

)

-80

-60

-40

-20

0

20

40

60

80

0.00 0.50 5.00 50.00

Added concentration (µM)

Diff

eren

ce in

via

bilit

y (%

)

Fig. 4: Mean (n = 5 concentrations and 8 wells per concentration) and standard deviation of the difference in cell viability (determined via MTT assay, n = 2) between Taxol® and Pac/RAME-β-CD. Cell viability at 37°C was tested over a concentration range of 1-20 µg paclitaxel/ml using a CC531s cell line. Different concentrations (0, 0.5, 5 and 50 µM) of HPPP , Verapamil and NB-DNJ were combined with Pac/RAME-β-CD to evaluate their effect on paclitaxel cytotoxicity. The replicate experiments (n = 2) of the MTT assay are presented as individual data sets.

In addition to HPPP, two other compounds (verapamil and NB-DNJ) were added to

the Pac/RAME-β-CD formulation. Used at the same concentrations as HPPP, both did not

increase the cytotoxicity of Pac/RAME-β-CD as neither compound lowered the IC50 values

of Pac/RAME-β-CD (Table 3): verapamil and NB-DNJ had individual IC50 values above 50

µM. Even a concentration of 500 µM NB-DNJ (a concentration capable of blocking all GCS

activity) did not induce an effect. Combining Pac/RAME-β-CD with 0.5 and 5 µM C6-

ceramide showed no effect on cell viability, but at concentrations of 10, 30 and 50 µM

Pac/RAME-β-CD became more potent compared to Taxol® (Table 3). The IC50 values of C6-

ceramide were about 50 µM for the MTT tests. In general, SRB data confirmed the MTT test

(Table 3).

A last series of experiments investigated the influence of the different compounds

(verapamil, NB-DNJ, HPPP and C6-ceramide) on the activity of the P-gp efflux pump.

Verapamil and NB-DNJ were tested at the highest concentration (50 µM), whereas HPPP and

C6-ceramide were tested at a concentration of 5 and 10 µM, respectively. Only verapamil

resulted in a retention ratio higher than 1.10: 23.07 at a concentration of 50 µM verapamil.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 97

Page 126: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

5.5. DISCUSSION

Although Bouquet et al. (2009) previously concluded that Taxol® and the Pac/RAME-

β-CD formulation had a similar cytotoxic activity towards colon cancer cell lines of human

and rat origin, the procedure previously used to evaluate the cytotoxicity of paclitaxel (over a

concentration range from 0.01 to 10.00 µg/ml) might not be optimal to detect differences

between potent cytotoxic agents as a low cell concentration (15 x 10³ cells per ml) and a long

exposure time (96 hrs) of the cells to paclitaxel were used. Therefore this study further

investigated the activity of Pac/RAME-β-CD complexes and Taxol® against the rat colon

cancer cell line (CC531s) using a higher cell concentration at the time of administration and a

shorter time of exposure to the drug (48 hrs). As a result of the different procedure a

significant difference in cytotixicity between both formulations was detected: the

concentration-dependent decrease of viability remained after exposure to Taxol®, whereas the

cytotoxic effect of Pac/RAME-β-CD was not concentration dependent. At the highest

paclitaxel concentration the viability was above 50% compared to a viability of 10 % for

Taxol®.

These observations can be correlated with the IC50 value of Cremophor EL®

(<0.5mg/ml) and the paclitaxel/Cremophor EL® ratio (1/88, w/w) in Taxol®: at a paclitaxel

concentration of 10 and 20 µg/ml (corresponding to Cremophor EL® concentrations of 0.9

and 1.8 mg/ml), Cremophor EL® significantly contributed to the cytotoxic effect of Taxol®.

As a result, the difference between Taxol® and Pac/RAME-β-CD only became apparent at

these higher concentrations since RAME-β-CD (the excipient used to formulate the inclusion

complexes) did not induce cell death based on its IC50 value (≥ 3.5 mg/ml) and the

Pac/RAME-β-CD ratio (1/60, w/w) in the inclusion complexes. Other researchers also

suggested that Cremophor EL® contributed to the cytotoxic effect of Taxol® (Fjällskogg et al.,

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 98

Page 127: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

1994; Reinecke et al., 2000). In contrast, Engblom et al. (1999) claimed that Cremophor EL®

had no influence on cell viability, the maximum paclitaxel concentration tested in that study

however was only 3 nM (equivalent to 0.22 µg/ml Cremophor EL®).

Our results clearly indicated that the Pac/RAME-β-CD formulation has an antitumour

effect which only depended on paclitaxel because its effect was similar to Pac dissolved in

DMSO and RAME-β-CD itself was not cytotoxic at the applied concentrations. The

concentration-independent effect on viability observed with Pac/RAME-β-CD and

Pac/DMSO was due to P-gp protein efflux pumps present in the CC531s cells, reducing the

sensitivity of the cancer cells towards paclitaxel.

Since the discovery of increased levels of glucosylceramide in multidrug resistant cells

(Lavie et al., 1996), ceramide and GCS inhibitors have gained attention as potential

therapeutics in cancer management. Paclitaxel stresses the cell which induces the production

of ceramide (Radin, 2003), but MDR cells decrease the levels of ceramide by transforming it

to glucosylceramide by up-regulation of GCS and thus escaping apoptosis (Lavie et al., 1997).

Combining Pac treatment with ceramide or a GCS inhibitor would increase intracellular

ceramide levels, causing even MDR cells to go into apoptosis. In light of this view HPPP, a

GCS inhibitor and PDMP analogue, was added to the Pac/RAME-β-CD solution. Addition of

a non-cytotoxic concentration of 5 µM increased the cytotoxic potency of the Pac/RAME-β-

CD formulation. Other researchers reported similar results with PDMP (Bieberich et al.,

1999). Our research confirmed that a selective inhibition of GCS with NB-DNJ was not

effective for improving paclitaxel activity (Bieberich et al., 1999; Veldman et al., 2003;

Norris-Cervetto et al., 2004), which indicated - as suggested by Norris-Cervetto et al. (2004) -

that the chemosensitization by PDMP is not solely caused by inhibition of GCS. Other

researchers have recently shown other possible intracellular interactions of paclitaxel and

ceramide (Qiu et al., 2006; Swanton et al., 2008). In addition, co-administration of 10 µM C6-

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 99

Page 128: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 100

ceramide to a Pac/RAME-β-CD formulation enhanced the cytotoxic activity of the

Pac/RAME-β-CD formulation, showing that both exogenous ceramide addition or non-

selective GCS inhibition are effective methods to improve Pac cytotoxicity.

MDR modulators like verapamil, that inhibit the P-gp efflux pump, have an effect on

GCS activity and expression (Bleicher et al., 2002). Therefore, it was investigated if GCS

inhibition by HPPP and NB-DNJ had an effect on the activity of the P-gp efflux pump.

According to Liu et al. (2001) GCS targeting did not modify P-gp expression or Rho123

efflux. However, Gouazé et al. (2005) found that blocking of GCS down-regulated P-gp. Our

results indicated that only verapamil had an effect on the efflux of Rho123 as HPPP, NB-

DNJ, C6-ceramide had no effect on the retention ratio of Rho123. This indicated that HPPP

did not affect P-gp activity.

5.6. CONCLUSION

The tumoricidal activity of a novel co-solvent-free and tensioactive-free cyclodextrin-

based paclitaxel formulation was only related to paclitaxel as the formulation excipient

(RAME-β-CD) played no role in the cytotoxicity of the formulation. In contrast, the observed

effect of Taxol® was in part due to the cytotoxicity of Cremophor EL® which is used to

solubilise paclitaxel. The cytotoxic activity of the Pac/RAME-β-CD formulation was not

concentration dependent as the P-gp efflux pumps in the colon cancer cell line induced

multidrug resistance (MDR). Combining the Pac/RAME-β-CD complexes with HPPP, a

novel PDMP analogue which inhibits glucosylceramide transferase, resensitized the MDR

cells for paclitaxel. The mechanism for this higher efficiency in MDR cells is not related to an

inhibition of the activity of the P-gp efflux pumps, but to its non-selective inhibition of GCS.

Page 129: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

5.7. REFERENCES

Bieberich E., Freischütz B., Suzuki M., Yu R.K., 1999. Differential effects of glycolipid

biosynthesis inhibitors on ceramide-induced cell death in neuroblastoma cells. J.

Neurochem., 72, 1040-1049.

Bleicher R.J., Cabot M.C., 2002. Glucosylceramide synthase and apoptosis. Biochimica et

Biophysica Acta, 1585, 172-178.

Bouquet W., Ceelen W., Fritzinger B., Pattyn P., Peeters M., Remon J.P., Vervaet C., 2007.

Paclitaxel/β-cyclodextrin complexes for hyperthermic peritoneal perfusion –

Formulation and stability. Eur. J. Pharm. Biopharm., 66, 391–397.

Bouquet W., Boterberg T., Ceelen W., Pattyn P., Peeters M., Bracke M., Remon J.P., Vervaet

C., 2009. In vitro cytotoxicity of paclitaxel/beta-cyclodextrin complexes for HIPEC.

Int. J. Pharm., 367, 148-154.

Charles A.G., Han T.-Y., Liu Y. Y., Hansen N., Giuliano A. E., Cabot M . C., 2001. Taxol-

induced ceramide generation and apoptosis in human breast cancer cells. Cancer

Chemother. Pharmacol., 47, 444-450.

Desai A., Vyas T., Amiji M., 2008. Cytotoxicity and apoptosis enhancement in brain tumor

cells upon coadministration of paclitaxel and ceramide in nanoemulsion formulations.

J. Pharm. Sci, 97, 2745-2756.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 101

Page 130: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Devalapally H., Duan Z., Seiden M. V., Amiji M. M., 2007. Paclitaxel and ceramide co-

administration in biodegradable polymeric nanoparticulate delivery system to

overcome drug resistance in ovarian cancer. Int. J. Cancer, 121, 1830-1838.

Engblom P., Pulkkinen J.O., Rantanen V., Hirvonen H., Kulmala J., Grènman R., Grènman

S., 1999. Effects of paclitaxel with or without Cremophor EL on cellular clonogenic

survival and apoptosis. Eur. J. Canc., 35, 284-288.

Fjällskog M.-L., Frii L., Bergh J., 1994. Paclitaxel-induced cytotoxicity-the effects of

Cremophor EL (castor oil) on two human breast cancer cell lines with acquired

multidrug resistant phenotype and induced expression of the permeability

glycoprotein. Eur. J. Canc., 30A, 687-690.

Gouazé V., Liu Y.-Y., Prickett C.S., Yu J.Y., Giuliano A.E., Cabot M.C., 2005.

Glucosylceramide synthase blockade down-regulates P-glycoprotein and resensitizes

multidrug-resistant breast cancer cells to anticancer drugs. Cancer Res., 65, 3861-

3867.

Hillaert U., Boldin-Adamsky S., Rozenski J., Busson R., Futerman A.H., Van Calenbergh S.,

2006. Synthesis and biological evaluation of novel PDMP analogues. Bioorg. Med.

Chem., 14, 5273-5284.

Kolesnick R.N., Krönke M., 1998. Regulation of ceramide production and apoptosis. Annu.

Rev. Physiol., 60, 643-665.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 102

Page 131: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Lavie Y., Cao H., Bursten S.L., Giuliano A.E., Cabot M.C., 1996. Accumulation of

glucosylceramides in multidrug-resistant cancer cells. J. Biol. Chem., 271, 19530-

19536.

Lavie Y., Cao H., Volner A., Lucci A., Han T.-Y., Geffen V., Giuliano A.E., Cabot M.C.,

1997. Agents that reverse multidrug resistance, tamoxifen, verapamil, and

cyclosporine A, block glycosphingolipid metabolism by inhibiting ceramide

glycosylation in human cancer cells. J. Biol. Chem., 272, 1682-1687.

Liu Y.-Y., Han T.-Y., Giuliano A.E., Cabot M.C., 2001. Ceramide glycosylation potentiates

cellular multidrug resistance. FASEB J., 15, 719-730.

Lucci A., Han T.-Y., Liu Y.-Y., Giuliano A.E., Cabot M.C., 1999. Modification of ceramide

metabolism increases cancer cell sensitivity to cytotoxics. Int. J. Oncol., 15, 541-546.

Mehta S., Blackinton D, Omar I., Kouttab N., Myrick D., Klostergaard J., Wanebo H., 2000.

Combined cytotoxic action of paclitaxel and ceramide against the human Tu138 head

and neck squamous carcinoma cell line. Cancer Chemother. Pharmacol., 46, 85-92.

Myrick D., Blackinton D., Klostergaard J., Kouttab N., Maizel A., Wanebo H., Mehta S.

1999. Paclitaxel-induced apoptosis in Jurkat, a leukemic T cell line, is enhanced by

ceramide. Leuk. Res., 23, 569-578.

Nobili S., Landini I., Giglioni B., Mini E., 2006. Pharmacological strategies for overcoming

multidrug resistance. Current Drug Targets, 7, 861-879.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 103

Page 132: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Norris-Cervetto E., Callaghan R., Platt F.M., Dwek R.A., Butters T.D., 2004. Inhibition of

glucosylceramide synthase does not reverse drug resistance in cancer cells. J. Biol.

Chem., 279, 40412-40418.

Qiu L., Zhou C., Sun Y., Di W., Scheffler E., Healey S., Wanebo H., Kouttab N., Chu W.,

Wan Y., 2006. Paclitaxel and ceramide synergistically induce cell death with transient

activation of EGFR and ERK pathway in pancreatic cancer cells. Oncology Reports,

16, 907-913.

Radin N.S., 2003. Designing anticancer drugs via the Achilles heel: ceramide, allylic ketones,

and mitochondria. Bioorg. Med. Chem., 11, 2123-2142.

Reinecke P., Knopf C., Schmitz M., Schneider E.M., Gabbert H.E., Gerharz C.D., 2000.

Growth inhibitory effects of paclitaxel on human epithelioid sarcoma in vitro. Cancer,

88, 1614-1622.

Singla A.K., Garg A., Aggarwal D., 2002. Paclitaxel and its formulations. Int. J. Pharm., 235,

179-192.

Swanton C., Marani M., Pardo O., Warne P.H., Kelly G., Sahai E., Elustondo F., Chang J.,

Temple J., Ahmed A.A., Brenton J.D., Downward J., Nicke B. 2008. Regulators of

mitotic arrest and ceramide metabolism are determinants of sensitivity to paclitaxel

and other chemotherapeutic drugs. Cancer Cell, 11, 498-512.

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 104

Page 133: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 5 PDMP analogue resensitizes MDR cells for paclitaxel/β-cyclodextrin complex 105

Swerts K., De Moerloose B., Dhooge C., Noens L., Laureys G., Benoit Y., Philippe J., 2004.

Comparison of two functional flow cytometric assays to assess P-gp activity in acute

leukemia. Leuk. Lymphoma, 45, 2221-2228.

Ueda K., Cornwell M.M., Gottesman M.M., Pastan I., Roninson I.B., Ling V., Riordan J.R.,

1986. The mdr1 gene, responsible for multidrug-resistance, codes for P-glycoprotein.

Biochem. Biophys. Res. Commun., 141, 956-962.

van Vlerken L.E., Duan Z., Seiden M. V., Amiji M. M., 2007. Modulation of intracellular

ceramide using polymeric nanoparticles to overcom multidrug resistance in cancer.

Cancer Res., 67, 4843-4850.

Veldman R.J., Mita A., Cuvillier O., Garcia V., Klappe K., Jeffrey A.M., Campbell J.D.,

Carpentier S., Kok J.W., Levade T., 2003. The absence of functional glucosylceramide

synthase does not sensitize melanoma cells for anticancer drugs. FASEB J., 17, 1144-

1146.

Wuchter C., Karawajew L., Ruppert V., Schrappe M., Buchner T., Schoch C., Haferlach T.,

Harbott J.N., Ratei R., Dorken B., Ludwig W.D., 2000. Clinical significance of P-

glycoprotein expression and function for response to induction chemotherapy, relapse

rate and overall survival in acute leukemia. Haematologica, 85, 711-721.

Page 134: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

106

IN VIVO TOXICITY AND BIOAVAILIBILITY OF

TAXOL® AND A PACLITAXEL/β-

CYCLODEXTRIN FORMULATION IN A RAT

MODEL DURING HIPEC

6.1. ABSTRACT

The spread of gastrointestinal or ovarian cancer to the peritoneum is a clinical

condition associated with a high mortality and high morbidity. In recent years, hyperthermic

intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery has emerged as a

promising strategy in the management of peritoneal carcinomatosis (PC). A paclitaxel (Pac)

formulation using randomly-methylated-β-cyclodextrins (RAME-β-CDs) to form a water

soluble complex was previously developed for HIPEC application. In the current study, the in

vivo toxicity and bioavailability of this formulation was compared in an in vivo rat model in

comparison to Taxol®. The maximum tolerated dose (MTD) of both formulations as well as

of β-cyclodextrins was determined. Blood parameters (red blood cell and white blood cell

count, creatinine, ALT and GGT) were evaluated over a 20 day period. The bioavailability of

both formulations was determined under normo- (37 °C) and hyperthermic (41 °C)

conditions.

Following HIPEC, both formulations had a similar MTD: 0.24 mg paclitaxel per ml.

Red blood cell count decreased to a minimum after 10 days and was not fully recovered after

20 days for both formulations. White blood cell monitoring showed an inflammatory state

66

Page 135: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

following therapy with Pac/RAME-β-CD formulation at day 10 and 15. Liver and kidney

parameters did not change significantly during the 20 day period. Bioavailability data of the

inclusion complexes showed a 40-fold increase of the area under the curve (AUC) of the

plasma concentrations for the Pac/RAME-β-CD formulation in comparison to Taxol®.

Hyperthermia yielded no significant differences in bioavailability data. These results showed

that both formulations had a similar toxicity profile but differed in bioavailability, establishing

the bioavailability enhancing effect of cyclodextrins.

6.2. INTRODUCTION

Peritoneal carcinomatosis (PC) is present in 40 to 80 % of the patients with a terminal

colorectal cancer (Koppe et al., 2006) and in a majority of patients diagnosed with ovarian

cancer (Tan et al., 2006). Patients with PC have a low quality of life combined with a high

morbidity and mortality (McQuellon et al., 2001). Systemic 5-fluorouracil-based

chemotherapy offers a median survival of 5.2 to 12.6 months for patients diagnosed with PC

(Koppe et al., 2006). In the last decennium, hyperthermic intraperitoneal chemotherapy

(HIPEC) after debulking surgery was introduced as a curative approach for PC. An 8-year

follow-up of a randomized trial in patients with PC of colorectal origin has recently shown a

significant increase in the disease-specific survival from 12.6 to 22.2 months and a 5-year

survival of 45 % for patients treated with HIPEC following complete cytoreduction (R-1)

(Verwaal et al., 2008). One of the rationales behind HIPEC is the presence of the peritoneum-

plasma barrier which permits a higher exposure of peritoneal carcinoma nodes to

chemotherapy: phase I studies have shown up to 1000-fold higher intraperitoneal (IP) levels

compared to plasma levels after IP administration of paclitaxel (Markman et al., 2003). The

combination of IP chemotherapy and hyperthermia during HIPEC is based on the synergism

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

107

Page 136: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

of both and the direct cytotoxic effect of hyperthermia at temperatures above 41 - 42 °C

(Wust et al., 2002). Hyperthermia can also increase drug uptake into the tumour tissue due to

improved cell permeability (Witkamp et al., 2001).

Paclitaxel (Pac), which binds and stabilizes the microtubuli, is a promising molecule

for intraperitoneal chemotherapy. Due to its large molecular weight, absorption from the

peritoneum is reduced. Another advantage is that Pac is rapidly cleared by the liver (Witkamp

et al., 2001). However, local toxic effects like abdominal pain and life-threatening

hypersensitivity reactions of the current Taxol® formulation have limited the use of paclitaxel

and prompted the need for newer and safer paclitaxel formulations (Markman et al., 2003;

Gelderblom et al., 2001). In previous research (Bouquet et al., 2007), a co-solvent- and

tensioactive-free paclitaxel formulation was developed using randomly-methylated-β-

cyclodextrins (RAME-β-CD) to improve the water solubility of paclitaxel. These Pac/RAME-

β-CD complexes were dissolved in phosphate buffered saline solution with 0.1 %

hydroxypropylmethylcellulose (HPMC) to produce stable solutions which can be used for

HIPEC applications. In the current study, the Pac/RAME-β-CD formulation and Taxol® were

used for a HIPEC procedure in rats to determine the maximum tolerated dose (MTD), general

toxicity and bioavailability.

6.3. MATERIALS AND METHODS

6.3.1. Materials

Randomly-methylated-β-cyclodextrin (RAME-β-CD) with a total degree of

substitution (TDS) of 13 was purchased from Cyclolab (Budapest, Hungary), paclitaxel (Pac)

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

108

Page 137: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

from Acros Organics (Geel, Belgium), Taxol® from Bristol-Myers Squibb (Brussels,

Belgium).

6.3.2. Preparation of the inclusion complexes

The required amount of paclitaxel was dissolved in absolute ethanol (Merck, Overijse,

Belgium), in a ratio of 1 to 60 (w/w). RAME-β-CD was added to the solution in a 1/20

(mol/mol) Pac/RAME-β-CD ratio and the solution was placed in an ultrasonic bath (Sonis 3

GT, Iskra-Pio, Šentjernej, Slovenia) for 5 min. Next, phosphate buffered saline (PBS) (Sigma,

Bornem, Belgium) was added in a ratio of 1/2 (w/w) versus the RAME-β-CD fraction. Next,

the solution was placed in an ultrasonic bath for 1 min and afterwards stirred with a magnetic

stirrer for 5 min. After evaporation of most of the co-solvent under reduced pressure, the

solution was frozen at -70 °C using solid carbon dioxide and freeze dried for 24 hrs at -50 °C

and 1 mbar. After freeze drying, a powder, suitable for reconstitution, was obtained. The

reconstitution medium consisted of phosphate buffered saline with 0.1 %

hydroxypropylmethylcellulose (HPMC) (Metolose® 60SH-4000, Shin-Etsu, Tokyo, Japan).

6.3.3. Determination of the paclitaxel concentration in the perfusate

To ensure that an accurate amount of paclitaxel was delivered during HIPEC the

compatibility of both formulations with different tubings was evaluated. Taxol® was tested in

combination with Marprene® (Watson-Marlow, Zwijnaarde, Belgium), Pt-cured silicone

(Pumpsil®, Watson-Marlow, Zwijnaarde, Belgium) , Tygon® (VWR, Leuven, Belgium) and

Tygothane® (VWR, Leuven, Belgium), while Pac/RAME-β-CD was only tested in

combination with Marprene® tubing. The paclitaxel solution (concentration: 0.24 mg/ml) was

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

109

Page 138: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

circulated at 41 °C through the tubing using a roller pump (Watson-Marlow, flow rate: 30

ml/min). Samples of the perfusate were collected at 0, 15, 30, 45 min. The samples were

filtered through Spartan 30/0.2 RC filters (Whatman, Brussels, Belgium) and following

appropriate dilution using PBS analysed by HPLC. The HPLC-system (Merck-Hitachi,

Tokyo, Japan) consisted of a pump (L-6000,), an integrator (D-2000), an autosampler (L-

7200) with a 25 µL loop and a UV/VIS detector (L-4200). Detection was performed at a

wavelength of 227 nm. Chromatographic separation was achieved with a guard column

(Lichrospher® 100-RP-18, 4*4 mm (5µm), Merck, Darmstadt, Germany) and an analytical

column (Lichrospher® 100-RP-18, 125*4 mm (5µm), Merck, Darmstadt, Germany). The

mobile phase consisting of acetonitrile (Biosolve, Valkenswaard, The Netherlands) and 0.1%

(v/v) phosphoric acid in water (Acros Organics, Geel, Belgium) (42:58, v/v) was degassed by

ultrasonication under vacuum. A calibration curve was validated for a concentration ranging

from 1 to 100 µg paclitaxel/ml. Stability is expressed in a percentage relative to the initial

concentration at 0 min.

6.3.4. Determination of paclitaxel plasma concentration

For the determination of Pac plasma concentration during HIPEC, blood samples were

collected at 0, 5, 10, 20, 40, 60 and 90 min in heparin containing tubes via a catheter in the

jugular vein. The plasma samples were analysed at the Department of Pharmacy and

Pharmacology (Netherlands Cancer Institute) according to a validated HPLC-MS/MS method

(Stokvis et al., 2004).

Briefly, 13C6-paclitaxel was added to the samples as an internal standard. Paclitaxel

was extracted from the samples by a liquid-liquid extraction using tert-butyl methyl ether.

After evaporation of the solvent, the sample was injected onto the analytical column. The

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

110

Page 139: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

analytical column was a Zorbax Extend C18 column (150 x 2.1 mm i.d., particle size 5 µm)

(Agilent Technologies, Palo Alto, USA), followed by chromatographic analysis using an

eluent consisting of 10 mM ammonium hydroxide-methanol (30:70, v/v) with a flow rate of

0.2 ml/min. Positive ionization electrospray tandem mass spectrometry was performed for

selective and sensitive detection with an API 3000 triple quadrupole MS (Sciex, Tornhill,

Canada). The resulting multiple reaction monitoring (MRM) chromatograms were used for

quantification using Analyst 1.2. software (Sciex). The validated range for paclitaxel was

from 0.25-1000 ng/ml using 200 µl plasma. AUC, Tmax and Cmax were calculated from the

plasma concentration values with the MW-Pharm 3.0® software (Mediware® 1987-1991,

Utrecht, The Netherlands).

6.3.5. HIPEC procedure

This experiment was approved by the ethical committee for animal testing of the

Faculty of Medicine, Ghent University (ECD 03/23). Adult Wag/Rij rats (Harlan, Horst, The

Netherlands) (body weight ≥ 270 g) were housed under the following conditions: ad libitum

access to food and water, 12-hour day/night cycle and 24 °C as room temperature. The

animals were anaesthetized with isoflurane (Forene®, Abbott, Louvain-La-Neuve, Belgium)

and were subcutaneously injected with buprenorfine hydrochloride (0.3 mg/kg) (Temgesic®,

Shering-Plough, Brussels, Belgium) except for the bioavailability study where no Temgesic®

was used. An incision was made in the abdomen. Inlet and outlet tubing (Marprene® and

Pumpsil® for Pac/RAME-β-CD and Taxol®, respectively) were placed in the peritoneal cavity

for perfusion with the cytostatic solution during 45 min. The paclitaxel concentration in the

solution was monitored with the method described above at 0,15, 30 and 45 min. A roller

pump (Watson-Marlow®, Zwijnaarde, Belgium) circulated the perfusate at flow rate of 30

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

111

Page 140: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

ml/min through a heat exchanger, ensuring a temperature of 37 or 41 °C for normo- and

hyperthermic conditions, respectively. During perfusion, the body and perfusate temperature

were closely monitored by thermosensors (ELLAB®, Roedovre, Denmark) and data were

collected using E-Val® 2.10 software (ELLAB®, Roedovre, Denmark). After HIPEC, the

solution was removed from the peritoneum and the incision was sutured.

Illustration 1: HIPEC in a ratmodel.

6.3.6. Maximum tolerated dose and toxicity study

The maximum tolerated dose (MTD) of RAME-β-CD and both Pac formulations

(Taxol® and Pac/RAME-β-CD complex) was defined as the highest non-lethal dose with a

maximum reduction in body weight of 10% 2 weeks after HIPEC treatment. The test

procedure was based on the Organisation for Economic Co-operation and Development

(OECD) guidelines for testing of chemicals. Briefly, to determine MTD the concentration of

the test substance (RAME-β-CD, Taxol® and Pac/RAME-β-CD) was gradually increased

using one rat per concentration.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

112

Page 141: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

The influence of both formulations on blood parameters (i.e. red blood cell (RBC)

count, white blood cell (WBC) count, creatinine, alanine amino transferase (ALT) and γ-

glutamyl transferase (GGT) concentration) was determined at the Laboratory of Clinical

Biology (Ghent University Hospital). One ml blood was collected from the tail vein of the rat

at 0, 5, 10, 15, 20 days post intervention. For RBC and WBC count, blood was collected in

K2EDTA tubes (Vacutest®, Milian, Geneva). For RBC count 4.0 µL blood was analysed with

flow cytometry (Sysmex® XE-2100-L, Norderstedt, Germany). WBC count was performed

with 18 µL blood, pretreated with Stromatotlyser-FB® (Sysmex®, Norderstedt, Germany) for

RBC lysis. For differentiation of WBC, 18 µL samples were pretreated with Stromatolyser 4-

DL® and Stromatolyser-DS®. Hereafter, all analyses of WBC were performed with

fluorescent flow cytometry (Sysmex® XE-2100-L, Norderstedt, Germany).

Creatinine, ALT and GGT concentrations were measured in serum. Creatinine

determination was done via a colorimetric analysis based on the reaction of creatinine and

picrine acid in an alkaline solution to form a creatinine-picrine acid complex. The absorption

was measured with a Cobas 6000® c501 (Roche, Vilvoorde, Belgium) analyser. The ALT

concentration was linked to its ability to catalyse the following reaction: α-ketoglutarate and

L-alanine reacted to form L-glutamate and pyruvate. The pyruvate fraction was reduced by

lactate dehydrogenase to lactate and NAD+. The absorption of the NAD+ was measured in the

Cobas 6000® c501 analyser and allowed to measure the activity of ALT. The activity of GGT

was based on its ability to catalyse the formation of 5-amino-2-nitrobenzoate from the

reaction between L-γ-glutamyl-3-carboxy-4-nitroanilide and glycylglycine. The increase in

absorption due to the formation of 5-amino-2-nitrobenzoate is measured with a Cobas 6000®

c501 analyser.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

113

Page 142: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

6.3.7. Statistical Analysis

Statistical Program for Social Scientists (SPSS 16.0) was used to analyse the results,

using 0.05 as significance level. A repeated univariate analysis of variances was performed to

investigate the time effect and the interaction between time and formulation (and temperature)

on body weight, blood parameters or plasma concentration. Mauchly’s test of sphericity was

used to test the homogeneity of the covariance matrices. In case this assumption was violated

the Greenhouse-Geisser correction factor was used. A Bonferroni-corrected post-hoc analysis

was performed to investigate all possible pairwise comparisons between the repeated

measures in each treatment group and between the treatment groups at each time point. All

other statistical analyses were performed using a two-sided one-way ANOVA.

6.4. RESULTS

6.4.1 Compatibility of paclitaxel formulations in different tubings

After 45 min perfusion of the Pac/RAME-β-CD formulation under hyperthermic

conditions using Marprene® tubing, 89 ± 1 % (n = 3) of the drug remained in the perfusate. In

contrast, hyperthermic perfusion of Taxol® in Marprene® tubing only resulted in a residual

paclitaxel fraction of 40 ± 2 % (n = 3). Using Pt-cured silicone (Pumpsil®), Tygon® and

Tygothane® tubings for perfusion of Taxol® under the conditions encountered during the

HIPEC procedure (41 °C, 45 min) resulted in a residual paclitaxel concentration equivalent to

65 ± 7 %, 46 ± 13 % and 52 ± 4 % (n = 3), respectively, compared to the initial paclitaxel

concentration. Based on these results Marprene® was used during HIPEC for delivery of the

Pac/RAME-β-CD solution and Pumpsil® for Taxol®.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

114

Page 143: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

115

6.4.2. Maximum Tolerated Dose

The maximum tolerated dose (MTD) of all compounds was determined using the

HIPEC settings (41 °C, 45 min). MTD of RAME-β-CD (dissoloved in PBS) was set at a

concentration of 10 mg/ml as no mortality was observed at this concentration and the weight

loss was less than 10 % 2 weeks post intervention (Table 1).

Compound Concentration (mg/ml) Mortality (%) Weight (%)*

10.00 0 97 ± 2 % RAME-β-CD 15.00 50 ND 15.00 0 98 ± 3 % RAME-β-CD + 0.1 %

HPMC (w/w) 20.00 50 ND

Formulation Pac concentration (mg/ml) Mortality (%) Weight (%)*

0.24 0 100 ± 7 % Taxol®

0.30 50 ND 0.24 0 95 ± 2 % Pac/RAME-β-CD 0.30 25 ND

Table 1: Determination of maximum tolerable dose (MTD) of randomly-methylated-β-

cyclodextrin (RAME-β-CD), RAME-β-CD + 0.1 % hydroxypropylmethylcellulose (HPMC), Taxol® and paclitaxel/RAME-β-CD (Pac/RAME-β-CD) complexes (1/20, mol/mol) in a hyperthermic peritoneal perfusion procedure in a rat (n = 4). * weight of a rat 14 days post treatment, expressed as a percentage of the initial body weight, ND: not determined

As Pac/RAME-β-CD complexes are dissolved in PBS supplemented with 0.1 % (w/v)

HPMC prior to HIPEC, the MTD of RAME-β-CD dissolved in this medium was also

determined. This experiment indicated that the addition of HPMC allowed to increase the

RAME-β-CD concentration to 15 mg/ml RAME-β-CD without encountering mortality. At a

RAME-β-CD concentration of 20 mg/ml 50% mortality (n = 4) was observed (Table 1). No

significant difference (p = 0.279) was seen between the weight curves of rats treated with a 10

mg/ml RAME-β-CD solution in PBS or 15 mg/ml RAME-β-CD solution in PBS

supplemented with 0.1 % HPMC.

Page 144: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

The study to determine the MTD of both Pac-formulations was started at a Pac

concentration of 0.06 mg/ml and was stepwise increased with increments of 0.06 mg/ml.

After treatment with Taxol® the mortality was 50 % (n = 4) at a Pac concentration of 0.3

mg/ml, whereas the mortality after treatment with the Pac/RAME-β-CD complexes at the

same concentration was 25 % (n = 4) (Table 1). Hence, 0.24 mg/ml was considered the MTD

for both formulations. At this concentration, there was no significant difference (p = 0.194)

between both treatments based on the weight curves of the rats.

6.4.3. Blood parameters

For both Pac formulations, the RBC count decreased significantly in function of time

(p < 0.05) (Fig. 1). There was also a marginal significant difference (p = 0.046) between both

paclitaxel formulations: pairwise comparison of the different time points only revealed a

significant difference at day 10 and 15 (p-values < 0.05). The levels of lymfocytes were not

significantly different over time and between both formulations (p = 0.241 and p = 0.275,

respectively) (Fig. 1). The neutrocytes showed an interaction between time and the two

formulations. Therefore, no general conclusions for both formulations could be drawn and

both formulations were analysed with a Bonferroni post hoc test at each time point. HIPEC

treatment with Taxol® had no significant effect on the neutrocyte count over the 20-day

period (all p-values ≥ 0.05), whereas a significant difference was observed in case of

Pac/RAME-β-CD complexes at day 10 and 15 compared to day 0 (p-values < 0.05).

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

116

Page 145: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Neutrocytes

Fig. 1: White blood cell count (neutrocytes and lymfocytes), red blood cell count and serum

creatinine concentration (mg/dl) in rats (n = 4) after HIPEC with Pac/RAME-β-CD and Taxol® using a paclitaxel concentration of 0.24 mg/ml. day 0; ■ day 5; ■ day 10; ■ day 15 and ■ day 20.

Serum samples were evaluated for their creatinine, ALT and GGT concentration.

Creatinine levels showed no significant difference between Pac/RAME-β-CD and Taxol® (p =

0.906) over the 20 day period. There was also no significant time effect (p = 0.615) for both

formulations. Since GGT concentrations of about half of the samples of both formulations

were below the detection limit of 3 U/L, the results of GGT were not statistically analysed.

There was a marginal significant (p = 0.041) decrease of ALT in time, but more importantly

no significant difference between the formulations was observed (p = 0.983).

0,00E+00 2,00E+03 4,00E+03 6,00E+03 8,00E+03 1,00E+04 1,20E+04 1,40E+04

Taxol Pac/RAME-β-CD

Num

ber o

f cel

ls/µ

l Lymfocytes

1,00E+04

Num

ber o

f cel

ls/µ

l 8,00E+03

6,00E+03

4,00E+03

2,00E+03

0,00E+00Pac/RAME-β-CDTaxol

Red Blood Cells

0,00E+00

2,00E+06

4,00E+06

6,00E+06

8,00E+06

1,00E+07

Taxol Pac/RAME-β-CD

Num

ber o

f cel

ls/µ

l

Creatinine 1,60E+001,40E+001,20E+001,00E+00

mg/

dl

8,00E-016,00E-014,00E-012,00E-010,00E+00

Taxol Pac/RAME-β-CD

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

117

Page 146: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

118

6.4.4. Bioavailability study

HIPEC was performed using a paclitaxel concentration of 0.24 mg/ml (previously

determined as MTD). After 45 min the paclitaxel concentration in the perfusate amounted to

62 ± 4 % and 61 ± 2 % (n = 3) of the initial concentration when the Pac/RAME-β-CD

solution was perfused using the in-vivo settings at normo- and hyperthermic conditions,

respectively. Similar results were obtained after perfusion of Taxol®: 61 ± 7 % and 61 ± 8 %

(n = 6) for normo- and hyperthermic conditions, respectively.

1

100

10000

0 15 30 45 60 75 9Time (min)

Pacl

itaxe

l (ng

/ml)

0

Fig 2: Paclitaxel plasma concentration (ng/ml) in rats (n = 6) after intraperitoneal perfusion

using different solutions at a paclitaxel concentration of 0.24 mg/ml and at normothermic (37 °C) and hyperthermic (41 °C) conditions: Taxol, normothermic,

Taxol, hyperthermic, ▲Pac/RAME-β-CD, normothermic and ■ Pac/RAME-β-CD, hyperthermic.

Plasma concentrations of paclitaxel were monitored during 90 min (i.e. 45 min during

and 45 min after HIPEC). The average plasma concentrations (Fig. 2) showed large

differences between Taxol® and Pac/RAME-β-CD and statistical analysis of the individual

plasma concentrations indicated a significant interaction between three factors (time,

formulation and temperature) (p-value < 0.05); therefore the simple effect of each factor was

investigated at a specific level of another factor via a Bonferroni post hoc test. This analysis

confirmed that at both temperatures (37 and 41 °C) the plasma values of Pac/RAME-β-CD

Page 147: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

119

were significantly higher than those of Taxol® (both p-values equal to 0.000). Investigating

the effect of temperature on both formulations showed that temperature had no significant

influence (p = 0.993) on Taxol®, whereas there was a marginal significant effect for

Pac/RAME-β-CD (p = 0.040).

The pharmacokinetic parameters (AUC0-90min, Cmax, ratio between AUCIP and

AUCplasma during perfusion) after HIPEC are summarized in Table 2. Intraperitoneal

perfusion with Pac/RAME-β-CD complexes resulted at both temperatures in a significantly

higher Cmax and AUC0-90min compared to Taxol® (p-values < 0.05). One-way ANOVA

showed no significant difference of AUC and Cmax between normo- and hyperthermic

treatment with Taxol® (p-values of 0.638, 0.617 and 0.341, respectively) and Pac/RAME-β-

CD complexes (p-values of 0.757, 0.076 and 0.073, respectively). Hyperthermia also had no

significant effect on the ratio of intraperitoneal and plasma AUC’s after administration of both

formulations (p-values of 0.654 and 0.185 for Taxol® and Pac/RAME-β-CD, respectively).

Formulation Temperature

(°C)

AUC0-90min

(mg/ml)*min

Cmax

(mg/ml)

AUC ratio*

Taxol® 37 2.80 ± 0.83 0.05 ± 0.01 10441 ± 4249

Taxol® 41 3.03 ± 0.82 0.05 ± 0.02 9542 ± 2155

Pac/RAME-β-CD 37 122 ± 66 1.91 ± 1.00 276 ± 59

Pac/RAME-β-CD 41 134 ± 72 2.73 ± 0.54 177 ± 181

Table 2: Pharmacokinetic parameters (mean ± standard deviation, n=6) after intraperitoneal perfusion for 90 min with paclitaxel solutions (using a paclitaxel concentration of 0.24 mg/ml) at normothermic (37 °C) and hyperthermic (41 °C) conditions. *AUC ratio of intraperitoneal concentration and plasma concentration over a 40 min period.

Page 148: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

6.5. DISCUSSION

For IV administration of Taxol® it is recommended to use polyvinylchloride (PVC)-

free infusion sets due to the leaching of diethylhexylphthalate (used as plasticiser for PVC),

which is primarily caused by Cremophor EL® (Allwood et al., 1996). The low recovery of

paclitaxel after perfusion of Taxol® in Marprene® tubing was probably also due to an

interaction of the formulation with the tubing as a white deposit was formed inside the tubing.

This adsorbed layer did not dissolve in water, but was soluble in DMSO. HPLC analysis

confirmed the presence of Pac in this DMSO solution. The Pac/RAME-β-CD formulation had

a better compatibility with Marprene® tubing at normo- and hyperthermic conditions since the

recovery of paclitaxel was comparable to the in vitro stability data of Pac/RAME-β-CD

complexes (± 80 %) reported by Bouquet et al. (2007). To ensure that a sufficient dose of

paclitaxel was delivered during HIPEC perfusion of Taxol®, tubings composed of different

materials were tested. A Pt-cured silicone tubing (Pumpsil®) yielded the best results.

Although this was slightly lower than the stability of Pac/RAME-β-CD, the in vivo stability

of both formulations was similar (as indicated by the paclitaxel concentration in the

perfusate). The current results stress the importance of selecting the appropriate material to

deliver Taxol® in a HIPEC setting in order to ensure that the appropriate dose is delivered to

the patient.

Before bioavailability testing, the MTD of both paclitaxel formulations after HIPEC

was determined. In addition the safety of RAME-β-CD was also evaluated as to our

knowledge, the MTD of RAME-β-CD for HIPEC administration has not been documented.

As the MTD of a RAME-β-CD solution increased from 10 to 15 mg/ml when 0.1 % (w/w)

HPMC was added to the aqueos solution, it is obvious that HPMC is not only beneficial for

the aqueous stability of Pac/RAME-β-CD complexes (Bouquet et al., 2007). The MTD of

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

120

Page 149: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Taxol® and the Pac/RAME-β-CD formulation in the HIPEC setting was set at a paclitaxel

concentration of 0.24 mg/ml (equivalent to 0.28 mM). Taking the body surface area (BSA) of

a rat (BSA (cm²) = 9.1 x g0.66, Baker et al., 1979) and the total perfusate volume (100 ml) into

account, a maximum dose of approximately 65 mg/m² can be tolerated by the rats when

HIPEC is performed with these formulations. To our knowledge, this is the first determination

of the MTD of paclitaxel for HIPEC in rats, but this dose was lower than the dose (175

mg/m²) administered to humans in a clinical pharmacokinetic trial of HIPEC therapy (de Bree

et al., 2008). Paclitaxel has also been administered IP in clinical trials via instillation therapy.

In these trials doses ranged between 20 and 175 mg/m² (de Bree et al., 2006). The

intraperitoneal MTD determined in rats (0.28 mM) clearly underlined the pharmacokinetic

advantage of HIPEC as only concentrations in the nanomolar range were obtained after IV

therapy (de Bree et al., 2006).

Although specific cyclodextrins (2-hydroxypropyl-β-cyclodextrin and

sulphobutylether β-cyclodextrin (Rajewski et al., 1995; Gould and Scott, 2005) have been

investigated extensively and are considered safe for parental use, there have been concerns

about the safety (nephrotoxicity and hemolysis) of other cyclodextrins used for parental

delivery (Frank et al., 1976; Irie and Uekama, 1997; Loftsson and Duchêne, 2007). Therefore

blood parameters were evaluated after IP administration of RAME-β-CD-containing

paclitaxel formulations as absorption of RAME-β-CD following IP administration could

hamper its use for HIPEC application. Although no histopathological study of the kidneys

was done creatinine levels indicated no modification of the kidney function. The initial

decrease in RBC count (to a minimum value at day 10) observed for both formulations is

probably linked to the surgical procedure. The normal RBC level however was not restored at

day 20 for both formulations. The marginal but significant difference in RBC count between

both formulations can be due to the significantly higher plasma concentrations of paclitaxel

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

121

Page 150: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

after HIPEC with Pac/RAME-β-CD complexes as paclitaxel, like most cytostatic agents, has

an influence on RBC count The constant serum ALT levels indicated that no liver toxicity

was encountered.

Based on the in vitro toxicity data reported by Bouquet et al. (2009) it was surprising

that Taxol® and Pac/RAME-β-CD had a similar MTD as in vitro experiments had shown that

Cremophor EL® was more cytotoxic than RAME-β-CD against different colon cancer cell

lines. The similarity in MTD can be linked to the difference in paclitaxel plasma

concentration observed after HIPEC therapy using both formulations. It has previously been

described that Cremophor EL® significantly reduced the bioavailability of paclitaxel after IP

injection by the micellar entrapment of the drug (Gelderblom et al., 2002). Even IV, this

entrapment had an influence on the blood/plasma distribution ratio (Sparreboom et al., 1999).

A beneficial effect of this slow release of paclitaxel from the micelles is a reduced toxicity,

but the entrapment of paclitaxel in micelles also suppressed its permeation into tumor tissue

(Yokogawa et al., 2004). Poor tumor penetration is a possible drawback of IP therapy

(Dedrick and Flessner, 1997), eliminating the advantage of placing the tumour in direct

contact with high concentrations of the cytostatic drug. In this respect the inclusion complex

of paclitaxel has a pharmacokinetic advantage as cyclodextrins are known to enhance the

solubility and bioavailability of poorly soluble drugs (Davis and Brewster, 2004). Moreover,

they are known as permeability enhancers (Rajewski and Stella, 1996). The high plasma

levels observed in this study clearly showed that cyclodextrins enhance the bioavailability and

permeability of IP administered drugs. In contrast, IP administration of paclitaxel nano- and

microparticles yielded a similar or slower absorption in comparison to Taxol® (Tsai et al.,

2007). What impact the different bioavailability has on the therapeutic efficiency of both

formulations will be investigated via a tumor growth delay study.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

122

Page 151: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

123

The use of hyperthermia is not only based on the direct cytotoxic effect of heat; it can

also enhance the efficacy and tumour penetration of cytostatic drugs (Witkamp et al., 2001).

However, in this study no significant influence of temperature on the bioavailability data was

observed, indicating that there is no effect of temperature on the permeation of paclitaxel

through the peritoneum. As previous in vitro toxicity data showed no influence of

hyperthermia on the cytotoxic activity of Pac/RAME-β-CD and Taxol® (Bouquet et al.,

2009), a possible additive effect of heat on tumour growth delay will have to come from its

direct cytotoxic action.

6.6. CONCLUSION

Evaluation of the toxicity of Taxol® and Pac/RAME-β-CD complexes during HIPEC

therapy in a rat model indicated that the maximum tolerated dose of both formulation was

similar (0.24 mg paclitaxel/ml), despite the different in-vitro toxicity of the excipients used in

the dosage forms (Cremophor EL® vs. RAME-β-CD). Blood parameters showed

inflammation in rats treated with Pac/RAME-β-CD in comparison to Taxol®. The

bioavailability of paclitaxel after HIPEC at normo- and hyperthermic conditions using

Pac/RAME-β-CD complexes was significantly higher in comparison to Taxol®. Hyperthermia

had no significant influence on the bioavailability of both formulations.

Page 152: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

6.7. REFERENCES

Allwood M.C., Martin H., 1996. The extraction of diethylhexylphtalate (DEHP) from

polyvinyl chloride components of intravenous infusion containers and administration

sets by paclitaxel injection. Int. J. Pharm., 127, 65-71.

Baker H.J., Lindsey J.R., Weisbroth S.H., 1979. Laboratory Rat, Volume I: Biology and

Disease. New York, Academic Press.

Bouquet W., Ceelen W., Fritzinger B., Pattyn P., Peeters M., Remon J.P., Vervaet C., 2007.

Paclitaxel/β-cyclodextrin complexes for hyperthermic peritoneal perfusion –

Formulation and stability. Eur. J. Pharm. Biopharm., 66, 391–397.

Bouquet W., Boterberg T., Ceelen W., Pattyn P., Peeters M., Bracke M., Remon J.P., Vervaet

C., 2009. In vitro cytotoxicity of paclitaxel/beta-cyclodextrin complexes for HIPEC.

Int. J. Pharm., 367, 148-154.

Davis M.E., Brewster M.E., 2004. Cyclodextrin-based pharmaceutics: past, present and

future. Nat. Rev. Drug. Discov., 3, 1023-1035.

de Bree E., Theodoropoulos P.A., Rosing H., Michalakis J., Romanos J., Beijnen J.H., Tsiftsis

D.D., 2006. Treatment of ovarian cancer using intraperitoneal chemotherapy with

taxanes: From laboratory bench to bedside. Cancer Treat. Rev., 32, 471-482.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

124

Page 153: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

de Bree E., Rosing H., Filis D., Romanos J., Melisssourgaki M., Daskalakis M., Pilatou M.,

Sanidas E., Taflampas P., Kalbakis K., Beijnen J.H., Tsiftsis D.D., 2008.

Cytoreductive surgery and intraoperative hyperthermic intraperitoneal chemotherapy

with paclitaxel: a clinical and pharmacokinetic study. Ann. Surg. Oncol., 15, 1183-

1192.

Dedrick R.L., Flessner M.F., 1997. Pharmacokinetic problems in peritoneal drug

administration: tissue penetration and surface exposure. J. Natl. Cancer Inst., 89, 480-

487.

Frank D.W., Gray J.E., Weaver R.N., 1976. Cyclodextrin nephrosis in the rat. Am. J. Pathol.,

83, 367-374.

Gelderblom H., Verweij J., Nooter K., Sparreboom A., 2001. Cremophor EL: the drawbacks

and advantages of vehicle selection for drug formulation. Eur. J. Cancer, 37, 1590-

1598.

Gelderblom H., Verweij J., van Zomeren D.M., Buijs D., Ouwens L., Nooter K., Stoter G.,

Sparreboom A., 2002. Influence of Cremophor EL on the bioavailability of

intraperitoneal paclitaxel. Clin. Canc. Res., 8, 1237-1241.

Gould S., Scott R.C., 2005. 2-Hydroxypropyl-b-cyclodextrin (HP-b-CD):a toxicology review.

Food Chem. Toxicol., 43, 1451-1459.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

125

Page 154: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Irie T., Uekama K., 1997. Pharmaceutical applications of cyclodextrins. III. Toxicological

issues and safety evaluation. J. Pharm. Sci., 86, 147-162.

Koppe M.J., Boerman O.C., Oyen W.J.G., Bleichrodt R.P., 2006. Peritoneal carcinomatosis of

colorectal origin-Incidence and current treatment strategies. Ann. Surg., 243, 212-222.

Loftsson T., Duchêne D., 2007. Cyclodextrins and their pharmaceutical applications. Int. J.

Pharm., 329, 1-11.

Markman M., 2003. Intraperitoneal antineoplastic drug delivery: rationale and results. Lancet

Oncol., 4, 277-283.

McQuellon R.P., Loggie B.W., Fleming R.A., Russel G.B., Lehman A.B., Rambo T.D., 2001.

Quality of life after intraperitoneal hyperthermic chemotherapy (IPHC) for peritoneal

carcinomatosis. E.J.S.O., 27, 65-73.

Rajewski R.A., Traiger G., Bresnahan J., Jaberaboansari P., Stella V.J.,1995. Preliminary

safety evaluation of parenterally administered sulphoalkyl ether β-cyclodextrin

derivatives. J. Pharm. Sci., 84, 927-932.

Rajewski R.A., Stella V.J., 1996. Pharmaceutical applications of cyclodextrins. 2. In vivo

drug delivery. J. Pharm. Sci., 85, 1142-1169.

Sparreboom A., van Zuylen L., Brouwer E., Loos W.J, de Bruijn P., Gelderblom H., Pillay

M., Nooter K., Stoter G., Verweij J., 1999. Cremophor EL-mediated alteration of

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

126

Page 155: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

paclitaxel disitribution in human blood: clinical pharmacokinetic implications. Cancer

Res., 59, 1454-1457.

Stokvis E., Ouwehand M., Nan L.G.A.H., Kemper E.M., van Tellingen O., Rosing H.,

Beijnen J.H., 2004. A simple and sensitive assay for the quantitative analysis of

paclitaxel in human and Mouse plasma and brain tumor tissue using coupled liquid

chromatography and tandem mass spectrometry. J. Mass Spectrom., 39, 1506-1512.

Tan D.S.P., Agarwal R., Kaye S.B., 2006. Mechanisms of transcoelomic metastasis in ovarian

cancer. Lancet Oncol., 7, 925-934.

Tsai M., Lu Z., Wang J., Yeh T.-K., Wientjes M.G., Au J.L.-S., 2007. Effects of carrier on

disposition and antitumor activity of intraperitoneal paclitaxel. Pharm. Res., 24, 1691-

1701.

Verwaal V.J., Bruin S., Boot H., van Slooten G., van Tinteren H., 2008. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus

sytemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer.

Ann. Surg. Oncol., 15, 2426-2432.

Witkamp A.J., de Bree E., Van Goethem A.R., Zoetmulder F.A.N., 2001. Rationale and

techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer

Treat. Rev., 27, 365-374.

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

127

Page 156: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 6 In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β−cyclodextrin formulation in a rat model during HIPEC

128

Wust P., Hildebrandt B., Sreenivasa G., Rau B., Gellerman J., Riess H., Felix R., Schlag

P.M., 2002. Hyperthermia in combined treatment of cancer. Lancet Oncol., 3, 487-

497.

Yokogawa K., Jin M., Furui N., Yamazaki M., Yoshihara H., Nomura M., Furukawa H.,

Ishizaki J., Fushida S., Miwa K., Miyamoto K., 2004. Disposition kinetics of taxanes

after intraperitoneal administration in rats and influence of surfactant vehicles. J.

Pharm. Pharmacol., 56, 629-634.

Page 157: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug
Page 158: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

129

PREMILINARY EVALUATION OF THE TUMOUR

GROWTH DELAY IN A RAT MODEL

FOLLOWING HIPEC WITH TWO PACLITAXEL

FORMULATIONS

7.1. ABSTRACT

Peritoneal carcinomatosis (PC) remains a dreaded clinical syndrome, common to the

evolution of gastrointestinal and ovarian cancers. In the last years, a new treatment modality

consisting of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC)

has given new hope to PC patients. For this treatment, a cyclodextrin-based paclitaxel

formulation was developed and previously evaluated for bioavailability and maximum

tolerated dose in healthy rats. In the current investigation, the feasibility of a tumour growth

delay study in rats with a PC of colorectal origin via positron emission tomography (PET) and

magnetic resonance imaging (MRI) was evaluated. Based on the preliminary results (3 rats

per treatment group), a study protocol was established. From the limited information available

in the current data set, some trends were emerging: (a), hyperthermia does not have a

beneficial effect on the activity of paclitaxel, (b) the cyclodextrin-based formulation

performed better and was the only formulation so far capable of producing a significant

decrease in tumour volume 2 days post treatment. Based on these preliminary data a study

using 6 rats per treatment group will be initiated to generate conclusive results about the effect

77

Page 159: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

of Taxol® and Pac/RAME-β-CD complexes on tumour growth delay following HIPEC at

normo- and hyperthermic conditions.

7.2. INTRODUCTION

Peritoneal carcinomatosis (PC) is in most cases a secondary cancer. For example, PC

is present in up to 80 % of patients with terminal colorectal cancer (Koppe et al., 2006) and

peritoneal spread is diagnosed in the majority of ovarian cancer patients (Tan et al., 2006).

Pseudomyxoma peritonei, a specific type of PC (Smeenk et al., 2007), is mostly caused by the

spread of an appendiceal tumour to the peritoneum. PC is associated with poor prognosis and

quality of life, and often treatment is limited to palliative care with patients having a median

survival of 6 months (Chu et al., 1989).

However, the combination of cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy (HIPEC) has shown promising results for the treatment of PC (Verwaal et al.,

2003). Recently, an 8-year follow-up study in patients with PC of colorectal origin has

indicated a significant increase in survival after HIPEC (Verwaal et al., 2008): 5-year survival

in 45 % of fully resected patients. This treatment procedure in combination with complete

cytoreduction offers new hope to PC patients.

HIPEC combines the cytotoxic actions of hyperthermia and chemotherapy, offering a

synergistic antitumoral effect in some cases (oxaliplatin, mitomycin, doxorubicin and

cisplatin) (Glehen et al., 2004) and resulting in higher drug concentration at the cancer

nodules compared to intravenous administration.

In previous research (Chapter 6), the maximum tolerated dose (MTD), following

HIPEC, was determined at 0.24 mg/ml or 0.28 mM paclitaxel in a rat in vivo model (Wag/Rij

rats). This largely exceeded the paclitaxel concentration obtained in humans after IV

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

130

Page 160: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

administration which are in the nanomolar range. A similar observation was made following

intraperitoneal and intravenous administration of paclitaxel to humans during a Phase I

clinical trial as the IP concentration was 1000-fold higher than the IV concentration

(Markman, 2003).

For the accurate assessment of peritoneal cancers, laparotomy and laparoscopy are still

considered as the golden standards. However, the invasive nature of these procedures requires

more animals and increases surgical stress to the animals. Therefore, this study monitors the

growth of a macroscopic tumour (induced in the peritoneal cavity of rats) in function of time

using MRI (to obtain anatomical data) and PET (to obtain information about the metabolic

status of the tumour tissue) for repeated evaluation of the tumour in the same test animals.

Following the development (Bouquet et al., 2007), in vitro (Bouquet el al., 2009) and in vivo

toxicity testing and bioavailability study (Chapter 5), the tumour growth delay (TGD) in a rat

model for colon cancer was determined following (normo- and hyperthermic) intraperitoneal

administration of different paclitaxel formulations. This chapter describes the preliminary

evaluation of tumour response following IP administration of paclitaxel using magnetic

resonance imaging (MRI) and positron emission tomography (PET) to visualize the tumour.

The information obtained during this study will form the basis of an extensive TGD study.

7.3. MATERIALS AND METHODS

7.3.1. Materials

Randomly-methylated-β-cyclodextrin (RAME-β-CD) with a total degree of

substitution (TDS) of 13 was purchased from Cyclolab (Budapest, Hungary), paclitaxel (Pac)

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

131

Page 161: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

from Acros Organics (Geel, Belgium), Taxol® from Bristol-Myers Squibb (Brussels,

Belgium).

7.3.2. Preparation of the inclusion complexes

The required amount of paclitaxel was dissolved in absolute ethanol (Merck, Overijse,

Belgium), in a ratio of 1 to 60 (w/w). Randomly-methylated-β-cyclodextrin (RAME-β-CD)

was added to the solution in a 1/20 (mol/mol) Pac/RAME-β-CD ratio and the solution was

placed in an ultrasonic bath (Sonis 3 GT, Iskra-Pio, Šentjernej, Slovenia) for 5 min. Next,

phosphate buffered saline (PBS) (Sigma, Bornem, Belgium) was added in a ratio of 1/2 (w/w)

versus the RAME-β-CD fraction. Next, the solution was placed in an ultrasonic bath for 1 min

and afterwards stirred with a magnetic stirrer for 5 min. After evaporation of most of the co-

solvent under reduced pressure, the solution was frozen at -70°C using solid carbon dioxide

and freeze dried for 24 h at -50°C and 1 mbar. After freeze drying, a powder for reconstitution

was obtained. The reconstitution medium consisted of phosphate buffered saline with 0.1 %

hydroxypropylmethylcellulose (HPMC) (Metolose® 60SH-4000, Shin-Etsu, Tokyo, Japan).

7.3.3. HIPEC procedure

This study was approved by the ethical committee for animal tests of Faculty of

Medicine (Ghent University, ECD 03/23). Adult Wag/Rij rats (Harlan, Horst, The

Netherlands) of at least 270 g body weight were kept under the following conditions: access at

libitum to food and water, 12-hour day-night cycle and 24°C room temperature. The animals

were anaesthetized with isoflurane (Forene®, Abbot, Louvain-La-Neuve, Belgium). An

incision was made in the abdomen and in- and outlet tubings (Marprene® and Pumpsil®

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

132

Page 162: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

(Watson-Marlow, Zwijnaarde, Belgium) for Pac/RAME-β-CD and Taxol® administration,

respectively) were placed in the peritoneal cavity for perfusion with the cytostatic solution

during 45 min. A roller pump (Watson-Marlow®, Zwijnaarde, Belgium) circulated the

perfusate at a flow rate of 30 ml/min through a heat exchanger, ensuring a temperature of

37°C (normotherm) or 41 °C (hypertherm). During the perfusion body and perfusate

temperature were closely monitored by thermosensors (ELLAB®, Roedovre, Denmark) and

data was collected using E-Val® 2.10 software (ELLAB®, Roedovre, Denmark). After the

HIPEC procedure, the solution was removed from the peritoneum and the incision was

sutured.

Illustration 1: HIPEC in a ratmodel

The Pac concentration in the perfusion solution used for the HIPEC procedure was

monitored (at 0, 15, 30 and 45 min) using a validated HPLC-UV/VIS method. All samples

were filtered through a 0.22 µm filter (Spartan 30/0.2 RC) (Schleicher & Schuell, Dassel,

Germany) and analysed by HPLC (following appropriate dilution using PBS). The HPLC-

system (Merck-Hitachi, Tokyo, Japan) consisted of a pump (L-6000), an integrator (D-2000),

an autosampler (L-7200) with a 25 µL loop and a UV/VIS detector (L-4200). Detection was

performed at a wavelength of 227 nm. Chromatographic separation was achieved with a guard

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

133

Page 163: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

column (Lichrospher® 100-RP-18, 4*4 mm (5µm), Merck, Darmstadt, Germany) and an

analytical column (Lichrospher® 100-RP-18, 125*4 mm (5µm)). The mobile phase consisting

of acetonitrile (Biosolve, Valkenswaard, The Netherlands) and 0.1% (v/v) phosphoric acid in

water (Acros Organics, Geel, Belgium) (42:58, v/v) was degassed by ultrasonication under

vacuum. A calibration curve was validated for a concentration ranging from 1 to 100 µg

paclitaxel/ml.

7.3.4. Tumor model

A CC531s rat colon adenocarcinoma cell line was obtained from the Laboratory of

Experimental Oncology (University Antwerp, Belgium). The cell line was grown in culture

flasks (Sarstedt, Newton, NC, USA) at 37 °C in a humidified atmospehere with 5 % CO2

using RPMI 1640 medium, buffered with HEPES (20 mM) (Invitrogen Corporation®, Gibco®,

Ghent, Belgium) and supplemented with 10 % fetal calf serum, 4 mM L-glutamine, 50 U/ml

penicillin and 50 µg/ml streptomycin. Confluent cells were detached with trypsin/EDTA

(Invitrogen, Zwijnaarde, Belgium) and vitality was evaluated with trypan blue (Sigma). Cell

suspensions with a cell concentration of 2,000,000 cells in 0.2 mL PBS were injected

subcutaneously in the upper hind leg of Wag/Rij rats to induce a tumour. After 4 weeks,

tumors of about 1 cm size were prelevated and tissue samples (5 x 5 mm, with 2-3 mm

thickness) were transplanted on the parietal peritoneum (covering the abdominal muscle) of a

new rat.

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

134

Page 164: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

7.3.5. Tumour imaging

Tumour characteristics were evaluated using PET and MRI imaging 6, 9 and 14 days

after implantation of the tumour. HIPEC treatment with paclitaxel was performed 7 days after

implantation. For both imaging techniques rats were anaesthetized with xylazine (Xyl-M 2 %,

VMD, Arendonk, Belgium) and ketamine (Ketamine 100, CEVA Santé Animale, Brussel,

Belgium) using a concentration of 10 mg/kg and 90 mg/kg, respectively. Animals were

divided into 6 groups which were treated with different HIPEC procedures (normo- vs.

hypertherm) and different formulations (Taxol® vs. Pac/RAME-β-CD complexes): (a)

Taxol®-37 °C, (b) Taxol®-41 °C, (c) Pac/RAME-β-CD-37 °C, (d) Pac/RAME-β-CD-41 °C,

(e) blank control group which received no treatment, (f) HIPEC control group which received

HIPEC treatment at 41.5°C using phosphate buffered saline as perfusion solution.

7.3.5.1. Determination of tumor activity via PET

Tumor growth in the peritoneal cavity of rats was monitored by positron emission

tomography (PET) imaging after the administration of 2-(fluorine-18)-fluoro-2-deoxy-D-

glucose (FDG) using a Philips® PET Allegro (Philips Medical Systems®, Cleveland, USA)

(Illustration 2). The spatial resolution of this system ranges from 5-6 mm FWHM (Full-Width

Half-Maximum). The Allegro scanner (Suleman et al, 2004) is a whole-body 3D PET scanner

using 4x6x20 mm³ gadolinium oxyorthosilicate (GSO) crystals. The physical diameter is 82.0

cm and the axial field of view is 18 cm without interplane septa. The transaxial field of view

(FOV) of the system is 576 mm and the axial FOV is 180 mm. The injected acitivity ranged

from 200 to 700 µCi and all images were appropriately normalized. During PET acquisition,

the emission time was 15 minutes. To get quantitatively comparable values of the signal

intensities from different acquisitions, an appropriate decay correction and image analysis was

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

135

Page 165: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

applied, using MATLAB® (Mathworks, MA, USA) as image processing toolbox. The latter

method consists of creating a virtual slice presenting maximum values along the axis

perpendicular to the coronal plane (maximum intensity projection). These images were

filtered using a mean filter of 13 x 13 kernel width. Regions of interest (ROI) around heart

and the tumor were drawn to calculate the ratio of the maxima within these ROI’s.

7.3.6.2. Determination of tumor volume via MRI

Complementary anatomical information about the tumour was acquired with a

Siemens® Trio 3T MRI (Erlangen, Germany) using a T1-weighted 3D FLASH sequence. The

rats were placed head first and prone in a (wrist) coil to measure the tumor volume. The MRI

Trio is a clinical whole-body scanner with a bore size of 60 cm. The highest homogeneity is

0.3 ppm Vrms (Volts root mean square) over 40 DSV (diameter spherical volume). The

maximum gradient strength is 40 mT/m and has a slew rate of 200 T/m/sec. The wrist coil is a

circulary polarized no-tune transmit/receive coil normally used for joint examinations of

wrist, hand and fingers. The aforementioned sequence involved a flip angle of 10°, a

repetition time (TR) of 13 ms and echo time (TE) of 4.9 ms to obtain a voxel size of 0.19 x

0.19 x 0.4 mm³. In order to easily locate the tumor, the rat was palpated and a vitamin B12

pellet was attached to the skin. Tumor volume was defined by creating a volume of interest

(VOI) consisting of a stack of planar ROI’s (using PMOD software, PMOD Technologies,

Adliswil, Switserland).

7.3.7. Statistical Analysis

Statistical Program for Social Scientists (SPSS 16.0) was used to analyse the results.

Data of day 6 were used as reference (100 %) in the PET and MRI study. The different

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

136

Page 166: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

treatment groups were compared with each other on day 9 and day 14 using a one-way-

ANOVA. A significance level of 0.05 was used. Bonferroni post-hoc analysis was performed

for pairwise comparisons between the treatment groups. Differences between the day 9 and 14

time points for each treatment group was tested using a paired samples t-test with a

significance level of 0.05.

7.4. RESULTS

7.4.1 Treatment characteristics

All treatments were well tolerated: no mortalities occurred and 2 weeks post treatment

all rats had recovered to at least 95 % of their starting weight.

After a 45 min HIPEC procedure with Taxol®, 77 ± 12 % and 70 ± 9 % (n = 3) of the

paclitaxel concentration remained in the perfusate at normo- and hyperthermic conditions,

respectively. Paclitaxel concentrations at the end of the HIPEC procedure with Pac/RAME-β-

CD were 74 ± 5 % and 58 ± 9 % (n=3) of the initial concentration at normo- and hyperthermic

conditions, respectively.

7.4.2. Tumour Growth Delay

Tumour data obtained at day 6 (one day prior to treatment) were considered as

reference values and the measurements at day 9 and 14 post implantation (i.e. 2 and 7 days

after HIPEC treatment) were expressed as a percentage of these values.

Tumours were evaluated via MRI and PET on day 9 and 14 as tumour activity in the

blank control group (i.e. without treatment) decreased at day 21 due to necrosis. PET data

indicated that the tumour activity had increased to a maximum of 167 ± 22 % (n=3) at day 14

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

137

Page 167: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

but had dropped to 113 ± 37 % after 21 days. The same evolution was seen on MRI: a tumour

volume of 153 ± 19 % at day 14 and 130 ± 100 % at day 21. The large variation at day 21 was

due to a tumour that continued to increase in volume in one rat. In general, results of PET and

MRI confirmed each other, showing a similar evolution of the tumour.

7.4.2.1. PET imaging

Fig. 1: Axial view of a rat with peritoneal cancer (black arrow); head and tail of rat are at the bottom and top of image, respectively, and the heart is indicated by the white arrow.

At day 9 no significant differences were detected between the average FDG activity

measured in the different treatment groups (Fig. 1). Although the tumour activity at day 9

dropped for some formulations below 100 % (i.e. blank HIPEC procedure, Taxol®-37 °C and

Pac/RAME-β-CD-37 °C) while the activity increased in the other groups, these observations

were not significant due to the large variability (standard deviations between 20 and 40 %).

On day 14 the tumour activity in rats without treatment had increased to 167 ± 22 %

(Fig. 2), but again no significant differences were observed between the 6 groups (p-values ≥

0.05).

None of the 6 treatments showed a significant difference between day 9 and 14 (p-

values ≥ 0.05). On both days there was also no significant difference between normo- and

hyperthermic administration of both formulations (p-values of 1).

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

138

Page 168: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

PET

0

50

100

150

200

250

Blank HIPEC Taxol® 41°C Pac/RAMEB41°C

Taxol® 37°C Pac/RAMEB37°C

Tum

or A

ctiv

ity (%

)

Fig. 2: Tumour activity (%), measured via PET imaging, 2 ( ) and 7 ( ) days post treatment in rats without paclitaxel treatment (Blank, HIPEC) and in rats having different paclitaxel treatments: Taxol®-37 °C, Taxol®-41 °C, Pac/RAME-β-CD-37 °C and PAC/RAME-β-CD-41 °C) (n = 3). Blank = no treatment; HIPEC = hyperthermic perfusion with saline.

7.4.2.2. MRI

Fig.3: Radial MRI image of rat with peritoneal cancer (white arrow) (abdomen at bottom of

image).

The tumour volume in the blank control group increased to 140 and 153 % on day 9

and 14, respectively. At day 9, all treatment groups had a reduction in tumour volume but

only two groups (Pac/RAME-β-CD 37 °C and 41°C) differed significantly from the blank

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

139

Page 169: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

control group (p-values of 0.044 and 0.022, respectively). The other groups had a non-

significant reduction of tumour volume (Fig. 4).

On day 14 no significant differences between the different treatments were detected,

although treatment with Pac/RAME-β-CD formulation at 37 and 41 °C resulted in tumour

reduction. Remarkably, the smallest tumour volume was observed for the HIPEC control

group (69 ± 30 % )(Fig. 4).

Despite the initial tumour reduction on day 9 after hyper- and normothermic Taxol®

treatment the tumour volume had increased at day 14: 141 and 108 %, respectively. However,

only for rats treated with Taxol® at hyperthermic conditions (41 °C) the increase of tumour

volume at day 14 was significantly different (p = 0.018) compared to day 9. For both

formulations no significant difference was detected in function of temperature (p-values of 1).

MRI

0

50

100

150

200

Blank HIPEC Taxol® 41°C Pac/RAMEB41°C

Taxol® 37°C Pac/RAMEB37°C

Tum

or V

olum

e (%

)

Fig.4: Tumour volume (%), measured via MRI scan, 2 ( ) and 7 ( ) days post treatment in rats without paclitaxel treatment (Blank, HIPEC) and in rats having different paclitaxel treatments: Taxol-37 °C, Taxol-41 °C, Pac/RAME-β-CD-37 °C and PAC/RAME-β-CD-41 °C) (n = 3). Blank = no treatment; HIPEC = hyperthermic perfusion with saline.

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

140

Page 170: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

7.5. DISCUSSION

Since this preliminary study was intended to determine the feasibility of a tumour

growth delay study using PET and MRI as monitoring tools, a number of essential aspects

were evaluated in this chapter:

- Tolerance of rats with an implanted intraperitoneal tumour towards the surgical

HIPEC procedure and administered paclitaxel dose. The indicators (mortality and

recovery of weight) confirmed the tolerance of the rats for the HIPEC procedure.

- Stability of paclitaxel solutions during HIPEC procedure. Since at the end of

the HIPEC procedure a large fraction of the initial paclitaxel concentration was

recovered in the perfusate, a consistent dose of paclitaxel will be intraperitoneally

delivered to the rats.

- Time points for PET and MRI analysis to monitor tumour growth. As

autonecrosis of the peritoneal tumour was observed 21 days after implantation,

evaluation of tumour growth was set at day 9 and 14 (i.e. 2 and 7 days after HIPEC).

- Number of test subjects required per treatment group. Based on the variability

of the data obtained during this preliminary study it is predicted that increasing the

number of rats per group to 6 will provide sufficient power (> 80 %, Table 1) for

statistical analysis (except for analysis of PET data at day 9 as one value obtained

during this preliminary study is probably an outlier). This indicated that using PET

and MRI as evaluation tools allowed to reduce the number of test animals during a

TGD study since the same animal can be used to determine the TGD.

- Resolution of MRI and PET scans. A sufficient resolution for accurate

visualisation of the tumour was obtained using a wrist coil MRI designed for human

applications. Tumour volume was determined using a human PET, which already

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

141

Page 171: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

provided sufficient resolution. However, future experiments will be performed using a

PET for small animals (µPET), enhancing the resolution.

Power in function of the number of test subjects

n = 3 n = 6 PET at day 9 0.203 0.490 PET at day 14 0.489 0.918 MRI at day 9 0.821 0.998 MRI at day 14 0.563 0.956

Table 1: Post hoc calculated (n=3) and estimated (n=6) (based on the same variation) power

of ANOVA analysis in function of the number of animals per treatment group.

Although only a limited number of significant differences were detected in this

preliminary study due to the small number of test subjects and the large variability (resulting

in a low power of the ANOVA, Table 1) some trends were emerging:

- hyperthermic treatment with PBS (i.e. blank HIPEC procedure) reduced the

tumour activity and volume which confirmed the well known cytotoxic activity of heat

(Wust et al., 2002; Sugarbaker, 2007).

- no synergism between the cytotoxicity of paclitaxel and hyperthermia was

observed. This could not be statiscally confirmed due to the low power of this study,

but is in accordance with previous results which showed that heat had no effect on the

in vitro cytotoxicity of paclitaxel (Bouquet et al., 2009) nor on the bioavailability of

paclitaxel after HIPEC using Taxol and Pac/RAME-β-CD complexes (Chapter 6).

- Pac/RAME-β-CD inclusion complexes had a better efficiency compared to

Taxol®. MRI scans indicated that Pac/RAME-β-CD was the only formulation which

induced at both temperatures a significant decrease in tumour volume at day 9. In

contrast to Taxol®, one week post treatment the tumour size was still below its initial

size after Pac/RAME-β-CD treatment. PET images were less conclusive, but also

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

142

Page 172: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

143

indicated a slightly better result for the Pac/RAME-β-CD formulation. The difference

in efficiency between Pac/RAME-β-CD and Taxol® is probably linked to the different

bioavailability of both formulations after HIPEC (Chapter 6).

Extension of the TGD study to a larger number of animals will provide an answer to

the question if sustained release of paclitaxel is beneficial for its tumoricidal activity.

Sparreboom and co-workers (Sparreboom et al., 1999; Gelderblom et al., 2001; van Zuylen et

al., 2001; Gelderblom et al., 2002) claimed that the Cremophor EL® fraction of Taxol® was

responsible for a sustained release of paclitaxel and that this reduced the side-effects and

provided long term exposure of the cancer to chemotherapy. However, a short treatment with

a high paclitaxel dose might be more effective, especially in a PC setting where it is the

intention to eradicate microscopic free-flowing or residual cancer cells to eliminate the

recurrence of peritoneal spread.

7.6. CONCLUSION

This preliminary study has shown the feasibility of MRI and PET to evaluate tumour

growth delay after HIPEC with paclitaxel solutions and allowed to establish the study

protocol for a future in-depth TGD study after HIPEC in a rat model.

Page 173: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

7.7. REFERENCES

Bouquet W., Ceelen W., Fritzinger B., Pattyn P., Peeters M., Remon J.P., Vervaet C., 2007.

Paclitaxel/β-cyclodextrin complexes for hyperthermic peritoneal perfusion –

Formulation and stability. Eur. J. Pharm. Biopharm., 66, 391–397.

Bouquet W., Boterberg T., Ceelen W., Pattyn P., Peeters M., Bracke M., Remon J.P., Vervaet

C., 2009. In vitro cytotoxicity of paclitaxel/beta-cyclodextrin complexes for HIPEC.

Int. J. Pharm., 367, 148-154.

Chu D.Z., Lang N.P., Thompson C., Osteen P.K., Westbrook K.C., 1989. Peritoneal

carcinomatosis in nongynecologic malignancy. A prospective study of prognostic

factors. Cancer, 63, 364-367.

Glehen O., Mohamed F., Gilly F.N., 2004. Peritoneal carcinomatosis from digestive tract

cancer: new management by cytoreductive surgery and intraperitoneal

chemohyperthermia. Lancet Oncol., 5, 219-228.

Koppe M.J., Boerman O.C., Oyen W.J.G., Bleichrodt R.P., 2006. Peritoneal carcinomatosis of

colorectal origin-Incidence and current treatment strategies. Ann. Surg., 243, 212-222.

Markman M., 2003. Intraperitoneal antineoplastic drug delivery: rationale and results. Lancet

Oncol., 4, 277-283.

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

144

Page 174: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Smeenk R.M., Verwaal V.J., Zoetmulder F.A.N., 2007. Pseudomyxoma peritonei. Cancer

Treat. Rev., 33, 138-145.

Sugarbaker, P.H., 2007. Laboratory and clinical basis for hyperthermia as a component of

intracavitary chemotherapy. Int. J. Hypertheramia, 23, 431-442.

Suleman S., Karp J.S., 2004. Imaging characteristics of a 3-Dimensional GSO whole-body

PET camera. J. Nucl. Med., 45, 1040-1049.

Tan D.S.P., Agarwal R., Kaye S.B., 2006. Mechanisms of transcoelomic metastasis in ovarian

cancer. Lancet Oncol., 7, 925-934.

Verwaal V.J., van Ruth S., de Bree E., van Slooten G.W., van Tinteren H., Boot H.,

Zoetmulder F.A.N., 2003. Randomized trial of cytoreduction and hyperthermic

intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in

patients with peritoneal carcinomatosis of colorectal cancer. J. Clin. Oncol., 21, 3737-

3743.

Verwaal V.J., Bruin S., Boot H., van Slooten G., van Tinteren H., 2008. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus

sytemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer.

Ann. Surg. Oncol., 15, 2426-2432.

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

145

Page 175: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 7 Preliminary evaluation of the tumour growth delay in a rat model following HIPEC with two paclitaxel formulations.

146

Wust P., Hildebrandt B., Sreenivasa G., Rau B., Gellermann J., Riess H., Felix R., Schlag

P.M., 2002. Hyperthermia in combined treatment of cancer. Lancet Oncol., 3, 487-

497.

Page 176: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 8 General Conclusion 147

GENERAL CONCLUSION

The objective of this doctoral research project was to develop an alternative paclitaxel

formulation suitable for HIPEC. Randomly-methylated-β-cyclodextrins (RAME-β-CD) were

chosen as alternative excipients and produced a stable water soluble complex with paclitaxel.

Dissolution of this complex was rapid and rendered a tensioactive- and co-solvent-free

paclitaxel solution. Due to the addition of HPMC, this solution was sufficiently stable during

HIPEC conditions. The activity of the cyclodextrin-based formulation was only based on

paclitaxel, whereas the cytotoxicity of Taxol® was also due to Cremophor EL® (used as

excipient to solubilise paclitaxel). This phenomenon was responsible for the observed activity

of Taxol® against multidrug resistant (MDR) cells, while the P-gp efflux pumps in the MDR

cells resulted in a concentration-independent response towards paclitaxel administered via a

cyclodextrin inclusion complex. Addition of a novel PDMP analogue restored the

cytotoxicactivity of the Pac/RAME-β-CD formulation.

Following HIPEC the Pac/RAME-β-CD formulation had a similar maximum tolerated

dose as Taxol®. Bioavailability data showed a larger drug penetration through the peritoneum

in case of the Pac/RAME-β-CD formulation. A preliminary tumour growth delay study

indicated that MRI and PET could be used to monitor the activity and volume of an

intraperitoneal tumour in function of time. This research is part of a larger framework in the

treatment of PC, which will only be successful thanks to further enhancements in surgical

techniques, better screening and selection of patients, and improvement of the diagnostic tools

88

Page 177: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 8 General Conclusion 148

(i.e. MRI, PET/CT,…). Only the combination of these fields will lead to an improvement of

the management of patients with PC.

Page 178: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 9 Future Perspectives 149

FUTURE PERSPECTIVES

A number of aspects have been investigated in this research project towards the

successful treatment of peritoneal carcinomatosis via hyperthermic peritoneal chemotherapy

using a water soluble paclitaxel formulation. Nevertheless, further research is essential to

optimize this treatment strategy and to broaden its applications:

- Evaluation of different paclitaxel formulations using the rat model for HIPEC as well

as using PET and MRI to monitor tumor growth delay In recent years, several

strategies have been explored to find safer paclitaxel formulations. Some of these new

formulations (Abraxane®, Pacliex®, Xyotax®, Genexol PM®) and an O/W emulsion

(formulated at the Laboratory of Pharmaceutical Technology) will be included in

future studies to evaluate their toxicity, bioavailability and toxicity against peritoneal

tumours.

- Evaluation of drug penetration in tumour tissue using different formulations for

HIPEC. To strengthen the body of scientific evidence regarding drug distribution in

the peritoneal tissue, these data can be correlated with histology data to evaluate

apoptosis.

- Develop an animal model to evaluate the efficiency of the different paclitaxel

formulations against peritoneal carcinomatosis following metastasis of an ovarian

cancer. Extending the application of this HIPEC procedure to different types of

peritoneal cancer would expand the clinical relevance of this research.

99

Page 179: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Chapter 9 Future Perspectives 150

- Elucidating the mechanism of the stabilising effect of HPMC on drug/cyclodextrin

complexes with NMR spectroscopy. This aspect is currently investigated in

collaboration with the Laboratory of NMR and Structure analysis of Ghent University.

Page 180: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

SUMMARY________________________________________

Patients diagnosed with peritoneal carcinomatosis have a median survival of 5.2 to

12.6 months. This limited survival has prompted a search for a more effective treatment of

this type of cancer, which is a common event in the course of gastrointestinal and ovarian

cancer. Recently, a combination of an aggressive surgical approach and hyperthermic

intraperitoneal chemotherapy (HIPEC) has generated new hope for these patients, but

significant research efforts are still needed to optimize this treatment strategy which is the

focus of this research project.

The current situation and overall objective of this research project are addressed in

Chapter 1: development of a novel and safer paclitaxel (Pac) formulation, which is suitable

for HIPEC, using β-cyclodextrins (β-CD) to solubilise paclitaxel. After an in-vitro

characterisation of the Pac/β-CD formulation and evaluation of its activity against different

colon cancer cell lines, its maximum tolerated dose, toxicity, bioavailability and tumour

growth delay was evaluated in an animal model for HIPEC treatment of peritoneal cancer of

colorectal origin.

Chapter 2 gives an overview of the main topics of this research project i.e. paclitaxel,

peritoneal carcinomatosis and cyclodextrins. After a brief description of the history, chemistry

and metabolisation and the method of action of paclitaxel, the advantages and disadvantages

of the different formulation types of paclitaxel are reviewed, including the currently dosage

forms marketed and the alternative formulation approaches. Treatment of peritoneal

carcinomatosis, which is currently limited to systemic chemotherapy, via the cytoreductive

Summary 151

Page 181: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) is highlighted and the

rationale for HIPEC is also elucidated. In the final part of the introduction, a general overview

of cyclodextrins and their pharmaceutical applications is presented as in this project the

formation of inclusion complexes between cyclodextrins and paclitaxel was used as a strategy

to design a novel water soluble paclitaxel formulation.

Chapter 3 describes the study that was executed to design a cyclodextrin-based

paclitaxel formulation. The complexation efficiency of chemically modified β-CDs with

paclitaxel was investigated, randomly-methylated-β-cyclodextrin (RAME-β-CD) yielded the

best results. After characterization of the inclusion complex, its aqueous stability (at room

temperature as well as at normo- (37 °C) and hyperthermic (41 °C) conditions) was evaluated.

The complex with a molar Pac/RAME-β-CD ratio of 1/40 showed the best stability. Addition

of 0.1 % (w/v) hydroxypropylmethylcellulose (HPMC) to the aqueous medium significantly

improved the stability and allowed to reduce the amount of RAME-β-CD to a 1/20 molar

ratio.

In Chapter 4, the in vitro efficiency of this new formulation was compared to the

commercially available Taxol® formulation. Both formulations were equipotent (≤ 10 %

difference in viability results) against a human (CaCo-2) and a rat (CC531s) cell line. The

formulation excipients, however, showed significant differences in IC50 values: ≥3.5 mg/ml

and 0.5 mg/ml for RAME-β-CD and Cremophor EL®, respectively. Hyperthermia (1 hr at 41

°C) had no effect on the cytotoxic activity of both formulations. This study indicated that it

was possible to reformulate paclitaxel in a less cytotoxic vehicle while maintaining the

cytotoxic activity of the formulation.

Using a different test setup (Chapter 5) (higher cell concentration and reduction of

time between administration and detection) to evaluate the cytotoxicity of Taxol® and the

Pac/RAME-β-CD formulation against the CC531s colon cancer cell line significant

Summary 152

Page 182: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

differences were observed between Taxol® and Pac/RAME-β-CD, especially at higher

concentrations (5 - 20 µg/ml) as the Cremophor® fraction also contributed to the cytotoxicity

of Taxol®. In contrast, RAME-β-CD was not cytotoxic and Pac/RAME-β-CD showed a

similar cytotoxicity as Pac dissolved in dimethyl sulphoxide (DMSO). The reduction in

viability of CC531s cells was concentration-independent after administration of Pac/RAME-

β-CD due to the presence of P-glycoprotein (P-gp) efflux pumps, making the cells resistant to

paclitaxel. The Pac/RAME-β-CD formulation was combined with HPPP, a glucosyl ceramide

synthase (GCS) inhibitor, to improve the sensitivity of P-gp positive cells towards paclitaxel.

At an added concentration of 5 µM this novel compound worked synergistically and restored

the activity of Pac/RAME-β-CD against these multidrug resistant (MDR) cells. The action of

HPPP was based on a non-selective inhibition of GCS.

Chapter 6 describes the in vivo evaluation of the Pac/RAME-β-CD formulation

following HIPEC. The maximum tolerated dose (MTD) of RAME-β-CD was 10 mg/ml; after

addition of hydroxypropylmethycellulose (HPMC) to the medium the MTD increased to 15

mg/ml. Pac/RAME-β-CD had an MTD of 0.24 mg paclitaxel/ml, which was similar to the

MTD of Taxol®. The influence of HIPEC using both formulations on blood parameters (red

blood cell (RBC) and white blood cell (WBC) count, creatinine, alanine aminotransferase

(ALT) and γ-glutamyltransferase (GGT) levels) was monitored. Pac/RAME-β-CD reduced

the RBC count to a larger extent than Taxol® and also resulted in an elevated neutrocyte

count. Both observations can be linked to the inflammatory state of the animal.

Bioavailability data, however, showed significant differences between both formulations:

compared to Taxol®, Cmax and AUC of Pac/RAME-β-CD were 50- and 40-fold higher,

respectively. Hyperthermia (41 °C) during the perfusion did not alter these observations.

In Chapter 7 a tumour growth delay (TGD) study was performed to evaluate the

feasibility of positron emission tomography (PET) and magnetic resonance imaging (MRI)

Summary 153

Page 183: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Summary 154

for monitoring the growth of an intraperitoneal tumour in function of time. This preliminary

study using 3 animals per treatment group allowed to establish the study protocol that will be

followed in a future TGD study using larger number of animals to accurately determine the

effect of formulation and hyperthermia on the activity of paclitaxel formulations following

HIPEC.

Page 184: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

SAMENVATTING__________________________________

Patiënten, bij wie peritoneale carcinomatose wordt vastgesteld, hebben gemiddeld nog

5.2 à 12.6 maanden te leven. Deze beperkte overleving onderstreept de nood aan een

zoektocht naar een nieuwe en meer effectieve behandeling van dit type kanker, die zeer

frequent voorkomt bij patiënten met een gastro-intestinale kanker of een ovariumkanker.

Recent heeft een combinatie van een agressieve cytoreductieve chirurgie en een hypertherme

intraperitoneale chemotherapie (HIPEC) nieuwe hoop gegenereerd bij deze patiënten. Er is

wel nog veel onderzoek nodig om deze therapie te optimaliseren; ook naar de selectie van

patiënten toe.

De situering en de doelstelling van deze studie wordt besproken in Hoofdstuk 1. Deze

bestaat uit de ontwikkeling van een nieuwe en veiligere paclitaxel formulatie op basis van β-

cyclodextrines, die kan toegepast worden in een HIPEC. Na een in vitro karakterisatie van de

Pac/RAME-β-CD formulatie en een evaluatie op verschillende coloncarcinoom cellijnen, is

het finale doel om deze formulatie in vivo te onderzoeken op een HIPEC diermodel met

peritoneale carcinomatose van colorectale oorsprong. Deze in vivo evaluatie bestudeert de

maximum tolereerbare dosis, toxiciteit, biologische beschikbaarheid en tenslotte de tumor

groei vertraging.

Hoofdstuk 2 geeft een overzicht van de 3 belangrijkste onderwerpen van dit

onderzoeksproject. Dit zijn paclitaxel, peritoneal carcinomatose en cyclodextrines. Na een

korte beschrijving van de geschiedenis, werking, chemische eigenschappen en metabolisatie

van paclitaxel, wordt er verder aandacht besteed aan de verschillende formulatietypes van

Samenvatting 155

Page 185: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

paclitaxel, zowel de huidige als de alternatieve formulaties. Iedere aanpak wordt besproken en

geëvalueerd aan de hand van zijn voor- en nadelen. In het tweede deel van de introductie

komen peritoneale carcinomatose en het peritoneum aan bod. De behandeling van dit type

kanker, die voorlopig meestal beperkt is tot systemische chemotherapie, wordt samen met de

cytoreductieve chirurgie en HIPEC besproken. De redenering/motivering achter deze nieuwe

therapie wordt in dit stuk uit de doeken gedaan. In het derde deel van de introductie wordt een

algemeen overzicht van de cyclodextrines en hun farmaceutisch toepassingen gegeven daar

deze als alternatieve formulatie excipientia gekozen zijn voor dit onderzoek.

Hoofdstuk 3 beschrijft de formulatiestudie die uitgevoerd werd om een paclitaxel

formulatie op basis van cyclodextrines te vinden. Verschillende types van chemisch

gemodificeerde β-cyclodextrines zijn getest geweest op hun complexatie efficiëntie met

paclitaxel. Random-gemethyleerde-β-cyclodextrine (RAME-β-CD) leverde de beste

complexatie resultaten op. Vervolgens, na de karakterisatie van het complex, werden de

verschillende stabiliteiten (24 u. op kamertemperatuur en 2 u. in normo (37°)- en hypertherme

(41 °C) omstandigheden), van deze nieuwe formulatie geëvalueerd. Het complex met een

molaire ratio van 1/40 (Pac/RAME-β-CD) vertoonde de beste stabiliteit. Additie van 0.1 %

(w/v) hydroxypropylmethylcellulose (HPMC) verhoogde significant de stabiliteit en liet toe

om de hoeveelheid RAME-β-CD te reduceren tot een molaire ratio van 1/20.

In Hoofdstuk 4 werd de in vitro efficiëntie van deze nieuwe formulatie vergeleken met

de huidige formulatie, Taxol®. Beide formulaties waren equipotent (verschil in

leefbaarheidsresultaten ≤ 10 %) voor twee coloncarcinoom cellijnen (CaCo-2 (humaan) en

CC531s (rat)). De formulatie excipientia, daarentegen, vertoonden wel een significant verschil

in cytotoxiciteit met IC50 waarden van ≥ 3.5 mg/ml en ± 0.5 mg/ml voor respectievelijk

RAME-β-CD en Cremophor EL®. De additie van hyperthermie (1 uur op 41 °C) had geen

effect op de cytotoxiciteit van beide formulaties. Deze studie gaf aan dat het mogelijk was om

Samenvatting 156

Page 186: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

paclitaxel te herformuleren in een minder toxisch vehiculum maar met behoud van de

cytotoxische activiteit.

Een verderzetting van dit in vitro onderzoek in Hoofdstuk 5 gebeurde aan de hand van

een gewijzigde test set-up (hogere celconcentratie en een kleiner tijdsinterval tussen

toediening en detectie). Deze nieuwe test set-up bracht significante verschillen in de

leefbaarheidsresultaten van beide formulaties aan het licht, vooral in de hogere concentraties

(5 - 20 µg/ml) daar waar de Cremophor EL® fractie bijdroeg tot de cytotoxiciteit van Taxol®.

In tegenstelling hiermee was RAME-β-CD veel minder cytotoxisch en was de cytotoxiciteit

van Pac/RAME-β-CD te vergelijken met Pac opgelost in dimethylsulphoxide (DMSO). Het

uitblijven van een concentratie dependente daling voor zowel Pac/DMSO als Pac/RAME-β-

CD in levensvatbaarheid werd uiteindelijk verklaard door de aanwezigheid van P-

glycoproteïne (P-gp) efflux pompen, die verantwoordelijk zijn voor de resistentie bij

kankercellen aan paclitaxel. Omdat de complexen een activiteit vertonen die enkel gebaseerd

is op paclitaxel, werd deze formulatie gebruikt voor het uittesten van de additie van een

glucosylceramide synthase (GCS) inhibitor, HPPP. Deze nieuwe molecule, een analoog van

D-threo-(1R,2R)-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), werkte

synergistisch (na de additie van 5 µM van HPPP) en herstelde de activiteit van Pac/RAME-β-

CD. Dit synergisme was gebaseerd op een niet-selectieve inhibitie van GCS.

Hoofdstuk 6 beschrijft de in vivo evaluatie van Pac/RAME-β-CD. RAME-β-CD had

een maximum tolereerbare dosis (MTD) van 10 mg/ml; additie van HPMC aan het

oplosmedium verhoogde de MTD tot 15 mg/ml. Pac/RAME-β-CD en Taxol® hadden beiden

een MTD van 0.24 mg paclitaxel/ml. In een volgende stap werd voor beide formulaties de

invloed van een HIPEC behandeling op de bloedparameters (aantal rode en witte bloedcellen,

creatinine, alanine aminotransferase (ALT) en γ-glutamyl transferase (GGT) concentratie) van

de dieren gevolgd. Pac/RAME-β-CD veroorzaakte een iets sterkere daling in aantal rode

Samenvatting 157

Page 187: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Samenvatting 158

bloedcellen alsook een verhoogd niveau aan neutrocyten. Biologische beschikbaarheidsdata

brachten veel grotere en significantere verschillen aan het licht tussen de 2 formulaties. De

maximum concentratie (Cmax) en oppervlakte onder de curve (AUC) van de Pac/RAME-β-CD

formulatie nam met een factor 50 en 40, respectievelijk, toe in vergelijking met Taxol®. Het

verhogen van de temperatuur van het perfusaat van 37 naar 41 °C had geen invloed op de

bekomen resultaten (voor beide formulaties).

Tenslotte, in hoofdstuk 7, werd een preliminaire studie uitgevoerd om de

uitvoerbaarheid van een tumor groei vertraging (TGD) studie met behulp van positron emissie

tomografie (PET) en magnetische resonantie beeldvorming (MRI) te bestuderen. Aan de hand

van deze preliminaire studie op 3 dieren per behandelingsgroep liet toe om een studie protocol

vast te leggen. Deze studie heeft ook aangetoond dat een toename tot 6 dieren per groep

voldoende moet zijn om statistisch significante conclusies te trekken.

Page 188: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

CURRICULUM VITAE______________________________

1. PERSONAL FACTS

NAME: BOUQUET-GEERARDYN

FIRST NAMES: Wim, Joseph Nicole

DATE OF BIRTH: December 12th , 1979

PLACE OF BIRTH: Geel, Belgium

PROFFESIONAL ADRES: Ghent University

Faculty of Pharmaceutical Sciences

Laboratory of Pharmaceutical Technology

Harelbekestraat 72

B-9000 Gent

Tel: +32 9 264 8056

[email protected]

NATIONALITY: Belgian

MARITAL STATE: Not married

NATIVE LANGUAGE: Dutch

OTHER LANGUAGES: English, French, German, Portuguese

Curriculum vitae 159

Page 189: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

2. DEGREES

High School St.-Stanislascollege (Poperinge)

1991-1992: Type: Latijn

1992-1995: Type: Latijn Wiskunde-Wetenschappen

1995-1997: Type: Latijn Wetenschappen

University Ghent University

1997-1999: Candidate in the farmaceutical sciences (passed)

1999-2002: Pharmacist (passed)

2006-2007: Laboratory Animal Science: Experiment Leader (passed)

3. COURSES, WORKSHOPS, SYMPOSIA AND SCIENTIFIC MEETINGS

2002 - Merck-Hitachi HPLC users’ meeting, 12 december 2002, Haasrode, Belgium

- Autumn Meeting of the Belgian-Dutch Biopharmaceutical Society, 13 december

2002, Leuven, Belgium

2003: - GMP and Validation (specialisationcourse of Pharmaceutical Sciences, Ghent

University) (examination – passed)

- 38th Congress of the European Society for Surgical Research, 28-31 may 2003,

Ghent, Belgium

2004: - Bioanalyse (2° lic. pharmacist, Universiteit Gent) (examination – passed)

- Innovating therapies for digestive tumors (Post-university courses Hepato-

Curriculum vitae 160

Page 190: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Gastro-enterology, Ghent University), 22 january 2004, Ghent, Belgium

- 12th International Cyclodextrin Symposium, 16 – 19 may, Montpellier, France

- AAPS Annual Meeting and Exposition, 7 – 11 november, Baltimore, USA

2005: - 40th Congress of the European Society for Surgical Research, 25 – 28 may,

Konya, Turkey

- Spring Meeting of the Belgian-Dutch Biopharmaceutical Society, 20 may, Ghent,

Belgium

- AAPS Annual Meeting and Exposition, 6 – 10 november, 2005, Nashville, USA

2006: - Cyclus Oncology (Post-university courses for pharmacists, Ghent), IPSA

- 41st Congress of the European Society for Surgical Research, 17 – 20 may,

Rostock, Germany

- Spring Meeting of the Belgian-Dutch Biopharmaceutical Society, 22 may,

Beerse, Belgium

- Basic course in laboratory animal sciences, partim 1 & 2, 2 – 13 october 2006,

Ghent University, Faculty of Veterinary medicine, Ghent, Belgium

- Portuguese I (CEF: A1), University Language Centre, October – December 2007,

Ghent, Belgium

- AAPS Annual Meeting and Exposition, 29 october – 2 november, 2006, San

Antonio, USA

2007 - Drug Delivery for Oncology Therapeutics (Course No. 6099), APV, 21 – 22

May, 2007, Basel, Switserland.

- Forum der Farmaceutische Wetenschappen, Belgisch Genootschap der

Farmaceutische Wetenschappen, 11 – 12 October, 2007, Spa, Belgium

2008 - Hands-on implementation of Process Analytical Technology (PAT), 25-26

August, 2008, Ghent, Belgium.

Curriculum vitae 161

Page 191: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

- Design of Experiments “Pharma Application” (course by Umetrics), 16-17

December, 2008, Ghent, Belgium.

2009 - Multivariate Data Analysis Basic Course (by Umetrics), 6-7 January, 2009,

Ghent, Belgium.

4. WORKING EXPERIENCE

Juli 2002: Self-employed substitute-pharmacist, C.V.B.A. Vooruit,

Nieuwevaart 151, 9000 Ghent, Belgium

Oct. 2002 – present: Fulltime PhD scholar reseacher, Faculty of Pharmaceutical

Sciences, Laboratorium of Pharmaceutical Technology, Ghent

University

Activities:

- Researchproject (Drug delivery of paclitaxel for an

intracavitairy chemotherapy in a ratmodel with peritoneal

carcinomatosis)

- Guidance of practical courses Artsenijbereidkunde (1° lic.

pharmacist) and Farmaceutische Technologie (3° lic.

pharmacist) (in total 100h).

- Guidance of researchthesis (2° lic pharmacist):

• Paclitaxel-cyclodextrin complexes: complexation and

determination of concentration (Ilse Naert, 2002-2003).

Curriculum vitae 162

Page 192: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

• Stability and in vitro toxicity of a paclitaxel/β-

cyclodextrin formulation (Josefien Van Laere, 2003-

2004).

• In vitro evaluation of a surfactant-free paclitaxel

formulation for a hyperthermic peritoneal perfusion

(Mado Vandewoestyne, 2004-2005).

• Stability and in vitro evaluation of a surfactant-free

paclitaxel formulation for a hyperthermic peritoneal

perfusion (Katrien Huyghe, 2005-2006)

5 PUBLICATIONS

1. Bouquet W., Ceelen W., Fritzinger B., Pattyn P., Peeters M., Remon J. P., Vervaet C.,

2007. Paclitaxel / β-cyclodextrin complexes for hyperthermic peritoneal perfusion –

formulation and stability. Eur. J. of Pharm. Biopharm, 66, 391-397.

2. Bouquet W., Boterberg T., Ceelen W., Pattyn P., Peeters M., Bracke M., Remon J.P.,

Vervaet C., 2009. In vitro cytotoxcity of paclitaxel/β-cyclodextrin complexes for HIPEC.

Int. J. Pharm, 367, 148-154.

3. Bouquet W., Hillaert U., Boterberg T., Ceelen W., Swerts K., Pattyn P., Bracke M., Van

Calenbergh S., Remon J.P., Vervaet C., 2009. PDMP analogue resensitizes MDR cells for

paclitaxel/β-cyclodextrin formulation. (submitted).

4. Bouquet W., Ceelen W., Adrieans E., De Vos F., Pattyn P., Peeters M., Remon J.P.,

Vervaet C., 2009. In vivo toxicity and bioavailability of Taxol® and a paclitaxel/β-

cyclodextrin formulation in a rat model during HIPEC. (submitted).

Curriculum vitae 163

Page 193: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

6 PROJECTS

1. Bijzonder Onderzoeksfonds (BOF) project (2 tot 4 jaar) (2009): Hypertherme

intraperitoneale drug delivery van paclitaxel formulaties in een diermodel voor

peritoneale carcinomatose. (accepted).

7. ORAL PRESENTATIONS DURING SCIENTIFIC MEETINGS

1. ‘Paclitaxel solubilisation by means of cyclodextrins for hyperthermic peritoneal

perfusion’ – 12th Forum of Pharmaceutical Sciences, Blankenberge, Belgium (may,

2004).

2. ‘In vitro evaluation of a surfactant-free paclitaxel formulation for hyperthermic peritoneal

perfusion’ – Spring Meeting of the Belgian-Dutch Biopharmaceutical Society, Ghent,

Belgium (may, 2005).

3. ‘Development and in vitro evaluation of a surfactant-free paclitaxel formulation for

hyperthermic peritoneal perfusion’ – 41st Congress of the European Society for Surgical

Research, Rostock, Germany (may, 2006).

4. ‘Development and in vitro evaluation of a surfactant-free paclitaxel formulation for

hyperthermic peritoneal perfusion’ - Forum der Farmaceutische Wetenschappen, Belgisch

Genootschap der Farmaceutische Wetenschappen, 11 – 12 October, 2007, Spa, Belgium.

Curriculum vitae 164

Page 194: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

8. POSTERS DURING SCIENTIFIC MEETINGS

1. Bouquet, W., Ceelen, W., Pattyn, P., Peeters, M., Remon, J.P., Vervaet, C. – Paclitaxel

solubilisation by means of cyclodextrins for hyperthermic peritoneal perfusion. – 12th

International Cyclodextrin Symposium, Montpellier, France (may, 2004)

2. Bouquet, W., Ceelen, W., Vervaet, C., Pattyn, P., Peeters, M., Remon, J.P. – Paclitaxel

beta-cyclodextrins complexes for hyperthermic peritoneal. – AAPS Annual Meeting and

Exposition, Baltimore, USA (november, 2004)

3. Bouquet, W., Boterberg, T., Ceelen, W., Vervaet, C., Bracke, M., Peeters, M. – Taxol

versus paclitaxel beta-cyclocextin complexes for hyperthermic peritoneal perfusion: an in

vitro study. – 40th Congress of the European Society for Surgical Research, Konya,

Turkye (may, 2005)

4. Bouquet, W., Boterberg, T., Ceelen, W., Vervaet, C., Bracke, M., Peeters, M., Pattyn, P.,

Remon, J.P. – The effect of HPMC on the stability and in vitro cytotoxicity of

paclitaxel/beta-cyclodextrin inclusion complexes. – AAPS Annual Meeting and

Exposition, Nashville, USA (november, 2005)

5. Bouquet, W., Boterberg, T., Ceelen, W., Vervaet, C., Bracke, M., Peeters, M., Pattyn, P.,

Remon, J.P. – In vitro evaluation of a surfactant-free paclitaxel formulation for

hyperthermic peritoneal perfusion. AAPS Annual Meeting and Exposition, Nashville,

USA (november, 2005)

6. Bouquet, W., Boterberg, T., Ceelen, W., Vervaet, C., Bracke, M., Peeters, M., Pattyn, P.,

Remon, J.P. – The effect of HPMC on the stability and in vitro cytotoxicity of

paclitaxel/beta-cyclodextrin inclusion complexes. Spring Meeting of the Belgian-Dutch

Biopharmaceutical Society (may, 2006), Beerse, Belgium

Curriculum vitae 165

Page 195: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug

Curriculum vitae 166

7. Bouquet, W., Ceelen, W., Vervaet, C., Peeters, M., Pattyn, P., Remon, J.P. – The

influence of HPMC on the stability of paclitaxel/randomly-methylated-beta-cyclodextrin

inclusion complexes. AAPS Annual Meeting and Exposition, San Antonio, USA (october,

2006)

8. Bouquet, W., Boterberg, T., Ceelen, W., Hillaert, U., Van Calenbergh, S., Bracke, M.,

Vervaet, C., Remon, J.P. – Enhancement of cytotoxic activity of paclitaxel in P-

glycoprotein positive cells via the addition of ceramide analogues. AAPS Annual Meeting

and Exposition, San Antonio, USA (october, 2006)

9. Bouquet, W., Ceelen W., De Vos F., Pattyn P., Peeters M., Remon J.P., Vervaet C. –

Hyperthermic intraperitoneal chemotherapy of Taxol and a novel paclitaxel/b-

cyclodextrin formulation in a rat model : toxicity and bioavailability study. APV Annual

Meeting, Barcelona, Spain (april, 2008)

9. AWARDS

1. Poster Prize Award (may, 2005): Bouquet, W., Boterberg, T., Ceelen, W., Vervaet, C.,

Bracke, M., Peeters, M. – Taxol versus paclitaxel beta-cyclocextin complexes for

hyperthermic peritoneal perfusion: an in vitro study. – 40th Congress of the European

Society for Surgical Research, Konya, Turkey (25 – 28 may, 2005)

Page 196: Drug delivery of paclitaxel for an intraperitoneal chemotherapy Drug