nanoparticle depot for intraperitoneal …

143
Purdue University Purdue e-Pubs Open Access Dissertations eses and Dissertations January 2016 NANOPARTICLE DEPOT FOR INTPERITONEAL CHEMOTHEPY OF OVARIAN CANCER Bo Sun Purdue University Follow this and additional works at: hps://docs.lib.purdue.edu/open_access_dissertations is document has been made available through Purdue e-Pubs, a service of the Purdue University Libraries. Please contact [email protected] for additional information. Recommended Citation Sun, Bo, "NANOPARTICLE DEPOT FOR INTPERITONEAL CHEMOTHEPY OF OVARIAN CANCER" (2016). Open Access Dissertations. 1480. hps://docs.lib.purdue.edu/open_access_dissertations/1480

Upload: others

Post on 26-Feb-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Purdue UniversityPurdue e-Pubs

Open Access Dissertations Theses and Dissertations

January 2016

NANOPARTICLE DEPOT FORINTRAPERITONEAL CHEMOTHERAPY OFOVARIAN CANCERBo SunPurdue University

Follow this and additional works at: https://docs.lib.purdue.edu/open_access_dissertations

This document has been made available through Purdue e-Pubs, a service of the Purdue University Libraries. Please contact [email protected] foradditional information.

Recommended CitationSun, Bo, "NANOPARTICLE DEPOT FOR INTRAPERITONEAL CHEMOTHERAPY OF OVARIAN CANCER" (2016). OpenAccess Dissertations. 1480.https://docs.lib.purdue.edu/open_access_dissertations/1480

Graduate School Form 30 Updated

PURDUE UNIVERSITY GRADUATE SCHOOL

Thesis/Dissertation Acceptance

This is to certify that the thesis/dissertation prepared

By

Entitled

For the degree of

Is approved by the final examining committee:

To the best of my knowledge and as understood by the student in the Thesis/Dissertation Agreement, Publication Delay, and Certification Disclaimer (Graduate School Form 32), this thesis/dissertation adheres to the provisions of Purdue University’s “Policy of Integrity in Research” and the use of copyright material.

Approved by Major Professor(s):

Approved by:

Head of the Departmental Graduate Program Date

Bo Sun

NANOPARTICLE DEPOT FOR INTRAPERITONEAL CHEMOTHERAPY OF OVARIAN CANCER

Doctor of Philosophy

Yoon Yeo

Chair

Tonglei Li

Rodolfo Pinal

Kinam Park

Yoon Yeo

Lynne Taylor 11/25/2015

i

NANOPARTICLE DEPOT FOR INTRAPERITONEAL CHEMOTHERAPY OF

OVARIAN CANCER

A Dissertation

Submitted to the Faculty

of

Purdue University

by

Bo Sun

In Partial Fulfillment of the

Requirements for the Degree

of

Doctor of Philosophy

May 2016

Purdue University

West Lafayette, Indiana

ii

ACKNOWLEDGEMENTS

First and foremost, I would like to express my sincere gratitude to my advisor, Dr.

Yoon Yeo. I do not know where I would be if she did not offer me the opportunity to join

her research team five years ago. Her pure passion to science, insightful suggestion, and

constant support set an example of a true scientist and a knowledgeable mentor. I really

appreciate the great patience and valuable resources that she has provided in the past five

years for me to grow. Additionally, I would like to thank my committee members, Drs.

Kinam Park, Tonglei Li, and Rodolfo Pinal for sharing their knowledge in research and

advice in career with me.

My gratitude goes to all the current members and alumni of Dr. Yeo’s lab. Your

outstanding work established the foundation of this lab, letting me see forward on the

shoulders of giants. Special thanks to Drs. Eun Jung Cho, Kyung-Oh Doh and Hillary

Holback who trained me during my first couple of years in the lab.

I am also very grateful to our collaborators, Benjamin Ramsey, Sandra

Torregrosa-Allen and Bennett Elzey and undergraduate students, Alice Chang, Ji Ae Lee

and Olivia Rivera, who have made great contribution to my work.

Hail Purdue and Boiler up!

iii

TABLE OF CONTENTS

Page

ABSTRACT ....................................................................................................................... ix

CHAPTER 1. INTRODUCTION .................................................................................... 1

1.1 Background ............................................................................................................... 1

1.2 Materials for IP Delivery Systems ............................................................................ 2

1.2.1 Requirements for IP Drug Carriers .................................................................. 2

1.2.2 Biomaterials for IP Drug Delivery .................................................................. 4

1.2.2.1 Natural Polymers ........................................................................................ 4

1.2.2.2 Synthetic Polymers ..................................................................................... 5

1.3 Dosage Forms for IP Drug Delivery ......................................................................... 5

1.3.1 Solutions .......................................................................................................... 6

1.3.2 Micro- or Nanoparticles ................................................................................... 7

1.3.3 Implantable Depots ........................................................................................ 11

1.3.4 Injectable Hydrogels ...................................................................................... 13

1.4 Challenges in IP Chemotherapy .............................................................................. 15

1.4.1 Drug Release in Peritoneal Environment ...................................................... 16

1.4.2 Tumor Specificity and Penetration ................................................................ 18

1.4.3 Tissue Responses to Carrier Materials .......................................................... 21

iv

Page

1.4.3.1 Effects of Carrier Materials on Peritoneal Tissues ................................... 21

1.4.3.2 Effects of Carrier Materials on Tumors .................................................... 22

1.5 Conclusion ............................................................................................................... 23

1.6 References ............................................................................................................... 25

CHAPTER 2. DEVELOPMENT AND CHARACTERIZATION OF

NANOPARTICLE DEPOT FOR INTRAPERITONEAL CHEMOTHERAPY ............. 41

2.1 Introduction ............................................................................................................. 41

2.2 Materials and Methods ............................................................................................ 43

2.2.1 Materials ........................................................................................................ 43

2.2.2 Preparation and Characterization of PTX Nanocrystals (PNC) .................... 44

2.2.3 Synthesis of Gel Precursors (HA-ADH, HA-CHO, CMC-CHO) ................. 45

2.2.4 Synthesis of Hydrophobically Modified Gel Precursor (EtCA-CMC-CHO) 45

2.2.4.1 Synthesis of 5β-cholanic Acid Methyl Ester (MeCA) (Scheme 1) .......... 45

2.2.4.2 Synthesis of Aminoethyl 5β-cholanoamide (EtCA) (Scheme 1) ............. 45

2.2.4.3 Synthesis of EtCA-CMC-CHO Conjugate (Scheme 2) ........................... 46

2.2.5 Preparation of Gel formulations .................................................................... 47

2.2.6 Evaluation of Gel Precursors and Crosslinked Gels ...................................... 48

2.2.6.1 Hydrophilicity of Gel Precursors .............................................................. 48

2.2.6.2 Toxicity of EtCA and EtCA-CMC-CHO ................................................. 48

2.2.6.3 Toxicity of Gels ........................................................................................ 49

2.2.7 PTX Solubility and Stability in Various Media ............................................. 49

v

Page

2.2.7.1 PTX Solubility in PBS, PBS Containing Serum, or PBS Containing

Tween 80 ................................................................................................................. 49

2.2.7.2 PTX Stability in PBS Containing Tween 80 or PBS Containing Serum .. 50

2.2.8 Dissolution Kinetics of PNC ......................................................................... 50

2.2.9 PTX Release Kinetics of HA Gel Formulations ............................................ 52

2.2.9.1 In Tween/PBS under Sink Condition ....................................................... 52

2.2.9.2 In Tween/PBS under Non-Sink Condition ............................................... 53

2.2.9.3 In FBS/PBS or Tween/PBS containing Hyaluronidase under Non-Sink

Condition ................................................................................................................. 54

2.2.10 HPLC Analysis of PTX ............................................................................... 54

2.3 Results and Discussions .......................................................................................... 55

2.3.1 Characterization of PNC and PPT ................................................................. 55

2.3.2 Synthesis and Characterization of EtCA-CMC-CHO ................................... 57

2.3.2.1 Hydrophobicity of Gel Precursors ............................................................ 58

2.3.2.2 Toxicity of EtCA and EtCA-CMC-CHO Conjugate ................................ 58

2.3.3 PTX Solubility and Stability in Various Media ............................................. 60

2.3.3.1 PTX Solubility in PBS, 50-FBS/PBS, or Tween/PBS .............................. 60

2.3.3.2 PNC Stability in PBS Containing Serum ................................................. 61

2.3.4 Dissolution Kinetics of PNC and PPT ........................................................... 62

2.3.4.1 Dissolution Kinetics of PNC and PPT with Dialysis Method .................. 62

2.3.4.2 Limitations of Current Dissolution Kinetics Study Methods and Potential

Alternatives ............................................................................................................. 64

vi

Page

2.3.5 PTX Release Kinetics from HA Gel Formulations ....................................... 65

2.3.5.1 In Tween/PBS under Sink Condition ....................................................... 66

2.3.5.2 In Tween/PBS under Non-Sink Condition ............................................... 66

2.3.5.3 In FBS/PBS or Tween/PBS containing Hyaluronidase under Non-Sink

Condition ................................................................................................................. 68

2.3.6 PTX Release Kinetics from HA-EtCA-CMC Gel ......................................... 70

2.4 Conclusions ............................................................................................................. 71

2.5 References ............................................................................................................... 73

CHAPTER 3. BIOACTIVITY EVALUATION OF NANOPARTICLE DEPOT ........ 79

3.1 Introduction ............................................................................................................. 79

3.2 Materials and Methods ............................................................................................ 80

3.2.1 Materials ........................................................................................................ 80

3.2.2 Determination of IC50 of Taxol, PPT and PNC on SKOV3 Cell Line ......... 80

3.2.3 Cellular Retention and Cytotoxicity of PTX ................................................. 81

3.2.4 Determination of the Maximum Tolerated Doses of Treatments .................. 83

3.2.5 In-vivo Efficacy Studies ................................................................................ 84

3.2.6 Statistical Analysis ......................................................................................... 84

3.3 Results and Discussion ............................................................................................ 85

3.3.1 IC50 of PTX in the Form of Taxol, PPT or PNC ........................................... 85

3.3.2 Cellular Retention and Cytotoxicity of PPT and PNC .................................. 86

3.3.3 Cytotoxicity of PPT-gel and PNC-gel ........................................................... 87

3.3.4 Maximum Tolerated Doses of PTX Treatments ............................................ 89

vii

Page

3.3.5 Anti-tumor Effects of PPT-gel and PNC-gel ................................................. 89

3.4 Conclusion ............................................................................................................... 94

3.5 References ............................................................................................................... 95

CHAPTER 4. ALBUMIN-STABILIZED PACLITAXEL NANOCRYSTALS ........... 97

4.1 Literature Review .................................................................................................... 97

4.1.1 Production of Nanocrystals ............................................................................ 98

4.1.1.1 Bottom-up Technologies ........................................................................ 100

4.1.1.2 Top-down Technologies ......................................................................... 101

4.1.1.3 Combined Technologies ......................................................................... 102

4.1.1.4 Nanocrystal Stabilization ........................................................................ 103

4.1.2 Remaining Challenges in Nanocrystal Development for Parental

Applications ............................................................................................................. 104

4.1.2.1 Instability during Storage ....................................................................... 104

4.1.2.2 Instability during Applications ............................................................... 105

4.1.2.3 Lack of Target Specificity ...................................................................... 107

4.2 Introduction ........................................................................................................... 108

4.3 Materials and Methods .......................................................................................... 109

4.3.1 Materials ...................................................................................................... 109

4.3.2 Preparation of Albumin-stabilized PNC ...................................................... 110

4.3.2.1 Crystallization in Matrix (Cim) .............................................................. 110

4.3.2.2 Nonsolvent and Temperature-Induced Crystallization ........................... 111

4.3.3 Characterization of Nanocrystals ................................................................. 112

viii

Page

4.4 Results and Discussions ........................................................................................ 112

4.4.1 Nanocrystal Morphology ............................................................................. 112

4.4.2 Nanocrystal Size .......................................................................................... 113

4.4.3 Albumin content .......................................................................................... 115

4.4.4 Proposed Role of Albumin in Cim-alb ........................................................ 115

4.5 Conclusion ............................................................................................................. 116

4.6 References ............................................................................................................. 117

CHAPTER 5. CONCLUSION ..................................................................................... 125

VITA ............................................................................................................................... 128

ix

ABSTRACT

Sun, Bo. Ph.D., Purdue University, May 2016. Nanoparticle Depot for Intraperitoneal

Chemotherapy of Ovarian Cancer. Major Professor: Yoon Yeo.

Intraperitoneal (IP) chemotherapy is a promising post-surgical therapy of ovarian

cancer, with the full potential yet to be proven. To facilitate IP chemotherapy of ovarian

cancer, we have developed a nanoparticle depot for IP chemotherapy consisted of

paclitaxel (PTX) nanocrystals (PNC) and hyaluronic acid-based hydrogel (HA gel). PNC

with a size of ~310 nm was produced by nonsolvent and temperature-induced

crystallization. Dissolution kinetics of PNC could be determined by the light scattering

method rather than the dialysis method due to drug reprecipitation caused by diffusion

barrier. PTX release profiles from PNC-gel and PTX precipitate-gel (PPT-gel) were

estimated in both sink- and non-sink conditions, where the latter simulated the peritoneal

environment. In-vitro release kinetics studies did not reveal any difference between PNC-

gel and PPT-gel, partly due to the centrifugation-related artifacts.

In cellular toxicity test and maximum tolerated dose assessment, PNC-gel

provided more efficient killing effect and greater toxicity than PPT-gel, which contained

larger PTX particles, indicating a greater dissolution rate of PNC due to the small size. A

single IP administration of PNC-gel extended the survival of mice with IP tumors

significantly better than the same dose Taxol, due to the local depot effect, whereas PPT-

x

gel was not superior to Taxol in survival extension. While the cell toxicity test and in-

vivo results consistently point to the beneficial effect of particle size reduction, in-vitro

drug release kinetics did not predict the difference between PPT- and PNC-gels,

suggesting the limitation of current release study methods.

For PNC-gel to serve the cancer patients to its full potential, the compatibility

between PNC and HA gel could be optimized by incorporating hydrophobic domains in

the hydrogel and introducing surface stabilizer on PNC to achieve a well-controlled

release. Aminoethyl 5β-cholanoamide (EtCA) was conjugated to one of the gel

precursors, and the in-vitro PTX release was enhanced by the inclusion of EtCA.

However, it was not pursued in the subsequent studies due to the unexpected toxicity of

EtCA. Albumin-stabilized PNC with a sub-200 nm particle size were prepared using a

method involving incipient crystallization in polymer matrix and subsequent surface

stabilization with albumin. The function and quantitation of surface stabilizers need

further investigation. In-vitro dissolution test and bioactivity evaluation of Cim-alb

remains to be performed to test the contribution of small size to enhancing local

availability of PTX.

1

CHAPTER 1. INTRODUCTION1

1.1 Background

The mainstay of current peritoneal malignancy treatment is surgical debulking of

visible tumors and post-surgical chemotherapy to remove residual microscopic tumors [1-

3]. Recently, intraperitoneal (IP) chemotherapy has been pursued in post-surgical

management of peritoneal malignancies, due to the promise of a high local concentration

and a longer half-life of a drug in the peritoneal cavity, which provides a unique

opportunity for the locoregional treatment of the IP malignances [4-6]. Part of the IP-

administered drugs are absorbed to systemic circulation, but it occurs at a slower rate

than those administered intravenously [7-9]; therefore, IP dosage forms can also serve as

a depot for sustained systemic drug delivery. IP chemotherapy has proven significantly

more effective than intravenous (IV) therapy in several clinical studies [1, 3, 10].

Accordingly, the National Cancer Institute issued a clinical alert to recommend IP

chemotherapy for stage III patients with optimally debulked ovarian cancer in 2006 [11].

On the other hand, several challenges remain to be overcome before IP

chemotherapy to make a standard protocol for post-surgical management of peritoneal

1 The content of this chapter has been previously published in Ceeln, W.P. and Levine, E.A., eds.,

Intraperitoneal Cancer Therapy: Principles and Practice. CRC Press/Taylor & Francis Group, Boca Raton,

FL, 2016.

2

malignancies. For example, IP-administered drugs show limited penetration into tumors

[12], thus necessitating the use of high IP doses. The high IP doses in turn account for

increased toxicities such as myelotoxicity, neurotoxicity, nephrotoxicity, nausea,

vomiting, and abdominal pain [11, 13, 14]. Cumulative toxicities reduce options for

subsequent rounds of therapy [15]. Moreover, complications related to IP administration,

such as discomfort due to prolonged infusion, catheter implantation, and peritoneal

adhesion, result in poor quality of life and, thus, high rate of dropout prior to the

completion of planned treatment [1, 14].

While the long list of challenges seems discouraging, this leaves the formulation

scientists with several questions: Can drug delivery systems help overcome any of these

problems? What are the unique requirements for IP drug delivery? What needs to be

done and what has been done to improve drug delivery to the peritoneal cavity? In this

chapter, we intend to address these questions by reviewing recent literature concerning IP

drug delivery. We will discuss experimental approaches to improve the effectiveness of

IP chemotherapy, focusing on the biomaterials used as drug carriers and various dosage

forms that have been reported to date. The chapter will conclude with a discussion of

remaining challenges and future perspectives.

1.2 Materials for IP Delivery Systems

1.2.1 Requirements for IP Drug Carriers

Typical first-line chemotherapeutic agents such as paclitaxel (PTX), docetaxel

(DTX) and cisplatin are low molecular weight drugs (<20 kDa), which are absorbed

3

through the peritoneal capillaries and enter the systemic circulation in a few hours [7-9].

The short residence time not only compromises the effectiveness of local chemotherapy

but also requires frequent or continuous dosing, culminating in complications related to

catheters and infection [16]. For the delivery of low molecular weight drugs, it is

therefore important to attenuate fast systemic absorption and maintain a high local

concentration of a drug. Ideally, the IP drug carriers should provide a sustained drug

release to maintain the local drug concentration within an effective range over several

weeks. The sustained local delivery of chemotherapy is also found beneficial for avoiding

tumor repopulation, which can occur during drug-free cycles in conventional intermittent

therapy [17]. In addition, it is desirable that the carriers are degraded into molecules that

are readily absorbed and cleared from the body by the time the loaded drug is exhausted

so that surgical removal of empty carriers may not be necessary.

While the primary goal of the IP delivery system is to remain in the peritoneal

cavity and provide a local reservoir of a drug for a prolonged period, such an effort often

faces a challenge due to the sensitivity of the peritoneal cavity to foreign materials. The

peritoneal cavity is responsible for protecting the body from breaches in the integrity of

the gut; therefore, it is armed with powerful innate and adaptive immune mechanisms

[18]. When confronted by an insult, peritoneal mesothelial cells, polymorphonuclear

neutrophils, and the resident peritoneal-associated lymphoid tissues interact with one

another via chemical signaling to produce inflammatory responses to the foreign

materials [18]. Due to this sensitivity, some biomaterials typically considered

biocompatible are found to induce significant inflammatory responses such as peritoneal

4

adhesions [19, 20]. Therefore, in designing an IP drug delivery system, it is necessary to

apply more stringent criteria for the selection of biomaterials for formulations.

1.2.2 Biomaterials for IP Drug Delivery

A list of polymers used for other biomedical applications is a good starting point

for selection of drug carrier materials. In particular, biomaterials used for peritoneal

adhesion prevention are great candidates for IP drug delivery. Several natural and

synthetic polymers have been used clinically and experimentally as physical barrier

devices, as reviewed elsewhere in detail [21].

1.2.2.1 Natural Polymers

Polysaccharides such as hyaluronic acid (HA) [22-26], cellulose derivatives [26-

28], dextran [29-31], and chitosan [20, 32, 33] have been explored for IP application. The

popularity of these polysaccharides stems from the biocompatibility proven in various

biomedical applications. The polysaccharides are chemically modified into reactive

precursors, which can form crosslinkable hydrogels upon application [20, 34, 35]. For

example, HA is modified into two types of precursors - one with adipic dihydrazide and

the other oxidized to have aldehydes, which instantly form a hydrogel upon contact [25].

The polysaccharide-based hydrogels are enzymatically degraded; therefore, the

degradation rate can be controlled by combining polymers with different enzyme

susceptibility. For example, the degradation rate of HA gel in the peritoneal cavity was

extended by replacing one of the gel precursors with cellulose derivatives, which were

5

not degraded by human enzymes [35]. For the delivery of hydrophobic drugs, it is likely

necessary to balance the hydrophobicity and hydrophilicity of the polymer [36]. For this

purpose, it is conceivable to modify the polymer with hydrophobic moieties to increase

the compatibility between drugs and polymers [37, 38]. Chitosan has also been widely

explored as a local depot of chemotherapeutic agents, where various stimuli (light, pH,

temperature) are employed as a trigger to form a hydrogel in-situ [39].

1.2.2.2 Synthetic Polymers

Synthetic polymers used for the prevention of peritoneal adhesion and IP

chemotherapy include polylactic acid (PLA), poly(lactide-co-glycolide) (PLGA),

polyethylene glycol (PEG), poly(Ɛ-caprolactone) (PCL) and their block co-polymers [26,

40-44]. Most of these polymers are commercially available at reasonable prices. In

particular, PLGA is widely used as a drug carrier [45] or a device [46, 47] because of its

track record in the approved products and the well-known biodegradability and

biocompatibility [21]. An advantage of synthetic polymers over natural polymers is that it

is relatively easier to control the molecular weight, monomer composition, and structure

of the polymer; therefore, there is greater flexibility in delivering various types of drugs

[43, 44].

1.3 Dosage Forms for IP Drug Delivery

The most common form of IP chemotherapy is the repurposed IV solutions. To

increase the drug retention in the peritoneal cavity, viscous polymer solutions, micro- or

6

nanoparticle formulations, implantable polymeric depots, and hydrogel-based systems

have been explored [21, 34, 48, 49].

1.3.1 Solutions

PTX is poorly water-soluble and, thus, requires a solubility enhancer. An equal

parts mixture of ethanol and Cremophor EL (polyethoxylated castor oil) is used to

solubilize PTX in Taxol® [50, 51]. Alternative solubilization strategies are pursued to

reduce toxicities related to Cremophor EL. For example, PTX and randomly methylated-

β-cyclodextrin (RAME-β-CD) form water-soluble inclusion complexes, which does not

precipitate upon dilution and stay stable after 24 h storage at ambient temperature or 2 h

at 41.5 °C [52]. When used for hyperthermic peritoneal perfusion, PTX/RAME-β-CD

complexes showed 40-fold higher plasma concentration than Taxol in a rat model [53]

and delayed the growth of peritoneal carcinomatosis [54]. The authors argued that

PTX/RAME-β-CD complexes had a greater ability to penetrate into IP tumors than Taxol,

which entrapped PTX in surfactant micelles and thus limited direct tumor exposure of the

drug [53].

A viscous solution of hydroxyethyl starch was used for the IP delivery of PTX [55]

and DTX [56]. Sprague Dawley rats were administered IP with the taxane compounds

using 6% hydroxyethyl starch (hetastarch) or 1.5% dextrose peritoneal dialysis solution

as a carrier. Fluid clearance and mean taxane concentrations in plasma were lower when

the drugs were delivered with hetastarch solution than with the peritoneal dialysis

solution. Importantly, the total amount of drug remaining in the peritoneal cavity was

7

significantly higher with hetastarch solution [55, 56]. These studies demonstrate that

hetastarch solution helped retain taxane compounds in the peritoneal cavity and reduce

systemic exposure to the drugs.

1.3.2 Micro- or Nanoparticles

A tumor penetrating microparticles (TPM) loaded with PTX was designed for IP

drug delivery [49]. The TPM system consisted of two types of particles. One was priming

TPM, which encapsulated PTX in a low molecular weight PLGA (LA:GA=50:50) and

released the drug rapidly to “prime” the tumors: i.e., to expand the interstitial space via

apoptosis induction and enhance the penetration of the second type of particles into

tumors. The second component called sustaining TPM encapsulated PTX in a high

molecular weight PLGA (LA:GA=75:25) and, thus, provided sustained drug release to

kill tumor cells. TPMs were able to retain in the peritoneal cavity for a longer time,

achieve greater therapeutic effect and lower toxicity than Taxol [49]. The tumor-priming

technology was later used to promote the delivery of survivin siRNA, which targeted a

gene encoding survivin, an anti-apoptotic protein associated with metastases and poor

prognosis of patients with gastric and colorectal cancers [57]. Here, siRNA was

encapsulated in PEGylated cationic liposomes (PCat) were administered IP after TPM

treatment of IP tumors. The combination of PTX-loaded TPM and PCat-siSurvivin was

more effective than each treatment in suppressing tumor growth due to the synergy of the

two treatment: TPM enhancing the penetration of PCat-siSurvivin into peritoneal tumors

8

and PCat-siSurvivin reducing survivin expression and augmenting TPM-induced anti-

proliferation and apoptosis [57].

A nanocrystal (NC) form (sub-micron drug particles stabilized by surfactants

and/or polymers) of PTX has been used in conjunction with hyperthermia for IP

chemotherapy [58]. PTX NCs were produced using Pluronic F127 as a stabilizer and

administered to rats bearing peritoneal tumors via IP perfusion at 41.5°C for 45 min. The

PTX NCs showed similar anti-tumor activity as Taxol with relatively low apparent

toxicity [58]. Unlike Taxol, the blood level of PTX continued to increase even after the

discontinuation of NC HIPEC, which suggests the long-term residence of NCs in the

peritoneal cavity due to their mucoadhesive properties [58].

Polymeric micelles have been used for delivery of multiple drugs to IP tumors

[42]. A combination of three drugs with distinct mechanisms of action-PTX as a

cytotoxic drug, cyclopamine (CYP) as an inhibitor of hedgehog signaling to reverse

taxane resistance, and gossypol (GSP) as a proapoptotic compound-was encapsulated in

poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) micelles for co-delivery

to tumors. Simultaneous encapsulation of three drugs in a single polymeric micelle was

possible as they have similarly poor water solubility [42]. The 3-drug loaded micelles

were able to disaggregate three dimensional spheroids of ES-2 human ovarian cancer

cells, whereas micelles with a single-drug or 2-drug combination showed negligible

effects on tumor spheroids. In mouse models of IP xenograft, the IP administered 3-drug

micelles reduced tumor burden to a greater extent than PTX alone and vehicles controls

[42]. Following debulking and induction of apoptosis of IP tumors with the 3-drug

micelles, secondary micelles with a near-infrared (NIR) fluorescence probe, DiR, was

9

systemically administered to visualize and help remove residual tumors (Fig. 1) [59].

Here the secondary micelles were conjugated with a peptide having high affinity for

apoptotic tissues (externalized phosphatidylserine) so that the secondary micelles could

specifically visualize apoptotic tumors [59]. A similar approach was used for systemic

administration of drug combinations and visualization of residual tumors [60, 61].

Figure 1. Schematic illustration of two-step strategy for neoadjuvant therapy, apoptosis-

targeted optical imaging and intraoperative surgical guidance, enabled by a tandem of

PEG-b-PCL micelles. (Reprinted from Cho, H. and Kwon, G.S., PLos One, 9,e89968,

2014, per Creative Commons Attribution (CC BY) license.)

While the particle systems in the above examples are intended for local drug

delivery, they also provide a useful tool for lymphatic or systemic drug delivery. Particles

administrated in the peritoneal cavity are drained into lymphatic circulation (Fig. 2) [48,

62]. Within the lymphatic system, particles smaller than 50 nm pass through lymph nodes

and reach systemic circulation via thoracic lymph ducts [62]. Kohane et al reported that

even larger PLGA nanoparticles (265 nm) entered systemic circulation from the

peritoneal cavity in less than 2 days, ending up in the spleen and the liver [40]. On the

other hand, larger particles (>500 nm) are trapped in lymph nodes. Therefore, depending

10

on the particle size, nanoparticles can serve as a sustained systemic delivery system or for

targeting cancer cells spreading via lymphatics.

Conversely, microparticles (1-100 µm) are retained longer in the peritoneal cavity

and, thus, have a greater potential to enhance local availability of a drug in the peritoneal

cavity and attenuate systemic drug absorption than nanoparticles [48, 49] (Fig. 2).

However, the long-term residence of microparticles in the peritoneal cavity can cause

inflammatory tissue responses and peritoneal adhesions [40], which may offset the

benefits of the localized medicine. For this reason, a combination of nanoparticles and

hydrogels was proposed as an alternative option. Here, nanoparticles served as a

sustained drug delivery system and hydrogels prevented premature clearance of

nanoparticles from the peritoneal cavity [22]. Additionally, the hydrogel prevented the

retained polymeric nanoparticles from causing adhesions [22].

11

Figure 2. A model of kinetic processes during IP administration of nano- or

microparticles. Nanoparticles are drained through the lymphatic system, reaching

systemic circulation or trapped in the lymph nodes depending on the size. Microparticles

tend to stay in the peritoneal cavity. Free drug released from particles are absorbed

directly to IP tissues or systemic circulation. Consequently, microparticles can maintain a

higher drug concentration in the peritoneal cavity than nanoparticles, whereas

nanoparticles yield a higher rate and extent of systemic absorption than microparticles.

(Modified from Tsai, M. et al., Pharm. Res., 24, 1691, 2007.)

1.3.3 Implantable Depots

Chitosan-phospholipid implantable formulations have been developed for

sustained and localized delivery of taxane compounds to ovarian tumors in the peritoneal

cavity [63-65]. The films were composed of chitosan and egg phosphatidylcholine (ePC)

(Chitosan-ePC film) blended with PLA-b-PEG [66], PLA-b-PEG/PLA [67], or PLGA

nanoparticles [68] containing PTX. PTX release from the nanoparticle/chitosan-ePC

films was evaluated in lysozyme-containing PBS [66], cell culture medium [68], or

ascitic fluid [67]. PTX was released over three months with no significant initial burst

Nano- or microparticles

Free drug

Peritoneal cavity Plasma

Free drug

Lymph node

Tissue

Lymphatic drainage Nanoparticles

Free drug

Absorption through peritoneumD

ose

rec

ove

red

Pla

sma

leve

l

Time Time

Nanoparticles

Microparticles Nanoparticles

Microparticles

[Drug]-time profile in peritoneal lavage sample

[Drug]-time profile in Plasma

Elimination

12

release as the chitosan-ePC matrix swelled and degraded by lysozyme [66-68]. Animals

implanted with chitosan-ePC films IP showed no signs of fibrous encapsulation or

inflammation around the implant area over 2-4 weeks, indicating good biocompatibility

of the implants [68]. The maximum tolerable dose (MTD) of PTX was 280 mg/kg/week

when delivered with the nanoparticle/chitosan-ePC film, much higher than Taxol with the

MTD of 20 mg/kg/week [64]. The PTX/nanoparticle/chitosan-ePC film significantly

enhanced the anti-tumor efficacy as compared to Taxol at total dose of 60 mg/kg [64, 65].

It is noticeable that continuous sustained delivery of PTX by PTX/nanoparticle/chitosan-

ePC film (60 mg/kg) was superior to intermittent Taxol administration (20 mg/kg q7d 3

schedule) at an equivalent dose and administration route (IP) in suppressing tumor

growth and extending animal survival [65]. Moreover, intermittent Taxol administration

resulted in a significant increase of tumor proliferation indices as compared with non-

treated controls, which indicates potential repopulation of tumors during treatment-free

intervals [65]. In contrast, PTX/nanoparticle/chitosan-ePC film is thought to increase

tumor responsiveness to the treatment by continuously exposing the tumors to high

concentration of PTX by localizing the drug source in the peritoneal cavity [65]. In

addition, PTX/nanoparticle/chitosan-ePC film did not induce the expression of multidrug

resistance gene (MDR1) in-vivo, whereas intermittent Taxol administration caused

significant MDR1 expression [69]. These results demonstrate the benefits of local

sustained drug delivery to IP tumors.

13

1.3.4 Injectable Hydrogels

The implantable films are an effective way of localizing chemotherapy in the

peritoneal cavity, but the fact that it requires surgical implantation poses challenges in

administration and patient compliance [70]. Therefore, the implantable film was replaced

with an injectable depot (PoLigel), composed of a water-soluble chitosan derivative, ePC,

and a fatty acid analog [70]. This mixture formed a gel at physiological temperature via

hydrophobic interaction between water-soluble chitosan and acyl chains of ePC,

reinforced by their interactions with a fatty acid analog [70]. Biocompatibility of the

PoLigel depended on the fatty acid analog–lauric aldehyde was better tolerated than

lauric chloride when injected subcutaneously in mice and observed over 4 weeks [71].

PoLigel was used for sustained and localized delivery of DTX to murine models of

SKOV3 ovarian cancer [63, 72]. DTX loaded in PoLigel (DTX/PoLigel) showed

constant release kinetics over 2 weeks with a minimal initial burst release in 0.01M PBS

(pH 7.4) containing lysozyme and albumin [63]. DTX/PoLigel IP administrated to mice

maintained constant DTX levels in plasma and peritoneal tissues over 2 weeks without

causing significant toxicity or inflammation [63]. Tumor burden was reduced by >70% in

DTX/PoLigel-treated group as compared with saline control [63]. Similar to the

PTX/nanoparticle/chitosan-ePC film [65], DTX/PoLigel achieved a greater anti-tumor

effect than intermittent Taxol injections, which was partly attributed to antiangiogenic

effect of local DTX, continuously released by PoLigel [73]. In addition to local tumors,

the IP administrated DTX/PoLigel was capable of delivering DTX to subcutaneous

tumors distant from the peritoneal cavity [72]. This result demonstrates that IP sustained

14

and localized delivery platform can treat not only local tumors but also metastasis distal

to the site of administration via the systemically absorbed drug. The PoLigel was also

used for co-delivery of DTX and cepharanthine, where the latter inhibited multidrug

resistance (MDR) due to drug efflux transporters and helped manage refractory ovarian

cancer with the MDR phenotype [74].

Another type of injectable gel used for IP chemotherapy is a thermosensitive

hydrogel based on PLGA-b-PEG-b-PLGA tri-block copolymers [75]. These polymers are

soluble in water, form a free-flowing solution at low temperature (e.g., 4°C), but

spontaneously gels at body temperature to create a water-insoluble gel called ReGel® [75].

ReGel® (23w/w%) injected subcutaneously into rats maintained the gel structure for 2

weeks but degraded into a viscous liquid and gradually resorbed in the next 2 weeks. The

hydrophobic segments (PLGA) formed hydrophobic regions in the gel, in which poorly

water-soluble drugs could be encapsulated. ReGel® loaded with PTX (OncoGel™)

released ~40% of the loaded PTX in the first 10 days and continuously released the

remaining payload in the next 40 days. In subcutaneous injection and the FDA Modified

Biocompatibility Tests, ReGel caused no signs of inflammation and significant toxicities,

confirming its biocompatibility [75]. OncoGel has been evaluated in local therapy of

solid tumors through preclinical and clinical studies and considered a promising adjuvant

therapy for ovarian cancer [76]. ReGel® was recently evaluated as a carrier of a three-

drug combination (PTX and two protein inhibitors) for IP chemotherapy [77]. The three-

drug loaded ReGel (Triogel) IP injected as a free-flowing solution formed a depot at body

temperature and released drugs at an equal rate according to gel erosion. The IP

administered Triogel showed greater antitumor efficacy and lower systemic toxicity than

15

IP or IV administrated PEG-b-PLA micelles with the same payloads in ES-2 ovarian

cancer xengraft model [77]. These results demonstrated the potential of Triogel as a

multi-drug carrier for IP chemotherapy of ovarian cancer.

While the PoLigel or ReGel form gels through non-covalent interactions between

polymer chains, polymers with functional groups that can react in-situ and form covalent

crosslinking to make hydrogels are also used for IP drug delivery [78]. The HA

derivatives that formed in-situ hydrogels via hydrazone bond (Section 2.2.1 ) were

evaluated for the prevention of post-surgical adhesion as a physical adhesion barrier

and/or drug carrier, showing excellent biocompatibility in the peritoneal cavity and

effectiveness in adhesion prevention [22, 34, 79, 80]. Accordingly, the in-situ

crosslinkable HA hydrogels have also been evaluated as an IP drug delivery system for

local therapy of cancers in the peritoneal cavity [36, 81].

1.4 Challenges in IP Chemotherapy

The above examples illustrate that various drug delivery systems deliver standard

anti-cancer drugs to the peritoneal cavity and help manage peritoneal malignancies by

providing sustained drug release and maintaining high local drug concentration. However,

several challenges remain to be addressed before IP chemotherapy becomes a standard

therapeutic option in the clinic.

16

1.4.1 Drug Release in Peritoneal Environment

The volume of peritoneal fluid in a healthy adult is about 50 mL with a turnover

rate of 4-5 mL/h, whereas blood volume is ~5 L [82]. The protein content of peritoneal

fluid is 25% of that in the blood [83]. Although malignancies may raise the protein level

and accelerate the turnover in the ascites, the relatively small volume of the peritoneal

fluid poses challenges to the IP delivery of poorly water-soluble drugs. Our previous

effort to deliver PTX with HA-based hydrogel provides an example of such a challenge

[36]. Here, PTX was mixed in HA hydrogel precursor solutions in the form of Taxol or

concentrated DMSO solution and administered IP to tumor-bearing mice so that the

precursors could form a hydrogel in-situ [36]. Upon dilution in the gel precursor solutions,

the concentrated PTX/DMSO solution started to form micrometer-scale precipitates

(PPTs), whereas Taxol maintained the micelle size (14 nm) [36]. The large particle size

of the former and HA hydrogel helped maintain PTX in the peritoneal cavity over 2

weeks. However, the prolonged retention of PPT-hydrogel did not translate to an

improved anti-tumor effect, due to the large size of PPTs and, thus, limited dissolution of

PTX [36]. This result indicates that hydrogel as a delivery medium helps localize drug in

the peritoneal cavity but does not prevent precipitation of poorly water-soluble drug,

limiting its availability to the local tumors.

A potential solution to this problem may be to reduce the particle size to facilitate

drug dissolution; however, it should not be as small as Taxol micelles as they are not

retained in the hydrogel [36]. Therefore, the challenge is to produce drug particles with

an optimal size, large enough to remain in the gel but small enough to allow continuous

17

and unhindered drug dissolution. There are several methods to produce drug particles in a

specific size range [84]. For example, nanocrystallization is a technique to produce

crystalline particles of poorly water-soluble drugs in the nanometer range (i.e., NCs). Due

to the size and, thus, the high surface area to volume ratio, NCs can increase the

dissolution rate of drug particles [85]. The unique advantage of NCs is that they are

mainly composed of drug molecules and create little concern for the safety of excipients

[86]. A minimal amount of surface stabilizers are however needed to prevent aggregation

of NCs during their lifetime [87]. For example, ionic surfactants such as sodium cholate,

sodium deoxycholate, and sodium lauryl sulfate are used to stabilize NCs via electrostatic

repulsion. Alternatively, NCs are stabilized with amphiphilic polymers that establish a

steric barrier against aggregation [86]. Another strategy is to use proteins as stabilizers

[88]. Serum proteins, such as human serum albumin (HSA), can serve as a stabilizer due

to their ability to adsorb onto hydrophobic surfaces and therefore provides steric

hindrance to NC aggregation and growth [89, 90]. Moreover, serum proteins can interact

with cell membrane to facilitate cellular uptake of anticancer drugs in tumors [91]. We

have recently used PTX NCs stabilized with HSA for IP delivery with hydrogels (Fig. 3)

and observed that the NC-hydrogel hybrid system had a superior anti-tumor activity than

Taxol at an equivalent total dose in a murine model of IP tumors.

18

Figure 3. Schematic illustration of an injectable depot system, composed of an in-situ

crosslinkable hydrogel and albumin-stabilized PTX NCs.

1.4.2 Tumor Specificity and Penetration

Reduced blood flow, increased interstitial fluid pressure, and high collagen

density interfere with drug transport into solid tumors [92]. In addition, tumor hyaluronan

[93] and stromal cells [94] in the tumor extracellular matrix aggravate the difficulty in

drug penetration into tumors. While the tumor penetration is a general issue in

chemotherapy of solid tumors [95], IP delivery faces a greater challenge than IV or

intratumoral administrations in tumor penetration because IP administrated drugs mostly

approach the tumor tissue from the periphery rather than the interior of the tumors (Fig.

4). In-vitro studies using multicellular layers [96-99] and tumor spheroids [100, 101]

have shown that limited drug penetration results in constant tumor exposure to a sub-

optimal level of drug, making an important mechanism for tumor resistance against

chemotherapy. An approach to address this challenge is to loosen up the tumor matrix

prior to chemotherapy to alleviate the drug penetration barrier [102, 103]. This strategy

involved two-step chemotherapy with an interval, where the first treatment with pro-

apoptotic agent like PTX induced reduction in cell density of solid tumors and allowed

In-situ cross-linkable hydrogel

PTX NCs

Paclitaxel Albumin

19

the subsequent dose to reach the interior of the tumors. This study provided a proof of

concept for the development of TPM for IP delivery of anticancer drugs or siRNA to

peritoneal malignances (Section 3.2) [49, 57]. Manipulating tumor microenvironment is

another way of improving delivery and intratumoral distribution of a drug or a

nanoparticulate drug delivery system [104]. For example, local mild hyperthermia was

applied to tumors to improve vasculature permeability, perfusion, and interstitial fluid

flow [105]. Local hyperthermia at 41°C increased tumor vasculature permeability and

allowed liposomes (~85 nm) to extravasate into the interstitial space (Fig. 5) [105]. This

effect may explain the increasing popularity and positive clinical outcomes of IP

chemotherapy combined with hyperthermia (hyperthermic intraperitoneal chemotherapy,

HIPEC) [106-113]. Recently, several new approaches have been proposed to improve

drug delivery to the interior of solid tumors [114, 115]. These approaches facilitate drug

penetration into tumors by enhancing interactions between drug carriers and tumor

stroma [116], preventing intracellular sequestration of a drug [117], or addressing

hypoxic region of tumors prior to standard chemotherapy [118]. Although these studies

have not been presented in the context of IP chemotherapy, it is worthwhile to explore

their applicability in the treatment of peritoneal malignances.

20

Figure 4. Schematic illustration of poor penetration of IP administered drug. Drug

approaching tumors from the peritoneal cavity has a limited depth of penetration, which

aggravates with increasing carrier size.

Nanoparticles

Released drug

Organ stroma

Basement membrane

Cancer cells

Organ epithelium

21

Figure 5. (a) Liposome (red) extravasation through tumor vasculature (green) with or

without mild hyperthermia for 1 h. (b) Quantification of liposome extravasation through

tumor vasculature in 4 tumor models under local mild hyperthermia at 41 °C for 1 h. NEF,

normalized extravascular fluorescence. Bar, 200 μm. Reprinted with permission from Li,

L., Koning, G.A., et al. J. Control. Release, 130-137, Copyright 2013 Elsevier.)

1.4.3 Tissue Responses to Carrier Materials

1.4.3.1 Effects of Carrier Materials on Peritoneal Tissues

As mentioned in section 2.1, careful selection of biomaterials as a drug carrier is

particularly important for IP application because of the high sensitivity of the peritoneal

cavity to foreign insults. Biomaterials generally considered biocompatible or wildly used

22

in other biomedical applications have caused inflammatory responses and peritoneal

adhesion upon IP application. For example, PLGA microparticles (5 µm) induced

adhesions 2 weeks after IP injection in mice [40]. This problem worsened with increasing

molecular weight of the polymer [40]. A photo-crosslinkable chitosan derivative, which

showed no attractive interactions or proliferative effect on mesothelial cells and

macrophages in-vitro, caused extensive and persistent peritoneal adhesions in animals

receiving it as a hydrogel in the peritoneal cavity [20]. It was later learned that the parent

chitosan had the same effect in-vivo [20]. Both chitosans had significant pro-

inflammatory properties, which might have been tolerated in other locations but not in the

peritoneal cavity.

1.4.3.2 Effects of Carrier Materials on Tumors

Drug carriers, once they are proven biocompatible, are often used under an

assumption that they have no other roles than delivering the payload to the body. Our

recent studies suggest otherwise. We have delivered platinum into the peritoneal cavity

using HA nanoparticles and hydrogels IP in mice for local chemotherapy of ovarian

cancer. We find that they do not show a greater anti-tumor efficacy than platinum

solution but rather cause a slight increase in tumor burdens at later time points, which

suggests a potential involvement of empty carriers and degradation products in the

growth of residual tumors. This hypothesis is not groundless, given various biological

roles of HA. HA is an indispensable extracellular matrix macromolecule for cell

migration, differentiation and proliferation [119]. Endogenous HA is implicated in the

invasion and growth of several tumor cells [119-122]. CD44, a well-known receptor of

23

HA, is found at the surface of various tumor cells and positively correlated with poor

prognosis of ovarian cancer patients [123]. HA concentration in tumor stroma is

considered an essential indicator of tumor aggressiveness and overall survival [124, 125].

However, the role of exogenous HA in tumor invasion and growth remains controversial.

Picaud et al reported that HA-carboxymethyl cellulose (CMC) membrane had no effect

on the proliferation of tumor cells in-vitro and in-vivo [126]. Similarly, HA-based anti-

adhesion membrane was found to have no influence on metastasis of colon cancer in a

human xenograft/nude mouse model [127]. On the other hand, Tan et al reported that HA

solution promoted proliferation and motility of colorectal tumor cell lines and increased

peritoneal tumor load as compared with non-treated control group, confirming its positive

role on metastatic potential of colorectal tumors [128]. Although the exact effect of HA-

based biomaterials on IP tumors and its role remain to be investigated, the mixed results

beg a question – if the drug carrier is indeed biologically inert and does no harm after

when it remains in the body after complete exhaustion of the payload. This means that in

designing a new drug carrier one should consider not only the biocompatibility of the

material but also its biological effects on residual tumors, which may not be readily

predicted from routine toxicity testing.

1.5 Conclusion

The premise of IP chemotherapy in the treatment of malignant diseases confined

in the peritoneal cavity lies in the theoretical potential for increased exposure of the

tumors to anti-cancer drugs and improved toxicity to local tumors [12]. Although the

24

proof of concept has been demonstrated in several clinical trials, current practice of IP

chemotherapy leaves plenty of room for improvement in delivery methods. Solutions,

micro- or nanoparticles, implantable depots, and in-situ crosslinkable hydrogels have

been evaluated in the context of IP chemotherapy, achieving varying levels of success in

preclinical studies. Due to the unique biological environment of the peritoneal cavity,

several challenges remain to be overcome before the IP drug delivery systems can benefit

patients to the full potential. Despite the needs and gravity of the challenges, there are

surprisingly few players in the field of IP drug delivery systems. It means that this is a

prime time to explore opportunities in IP drug delivery.

25

1.6 References

1. Armstrong, D.K., et al., Intraperitoneal Cisplatin and Paclitaxel in Ovarian

Cancer. New Eng J Med, 2006. 354(1): p. 34-43.

2. Ozols, R.F., et al., Phase III trial of carboplatin and paclitaxel compared with

cisplatin and paclitaxel in patients with optimally resected stage III ovarian

cancer: a Gynecologic Oncology Group study. J Clin Oncol, 2003. 21(17): p.

3194-3200.

3. Alberts, D.S., et al., Intraperitoneal Cisplatin plus Intravenous

Cyclophosphamide versus Intravenous Cisplatin plus Intravenous

Cyclophosphamide for Stage III Ovarian Cancer. New Eng J Med, 1996. 335(26):

p. 1950-1955.

4. Markman, M., Intraperitoneal drug delivery of antineoplastics. Drugs, 2001.

61(8): p. 1057-1065.

5. Markman, M., et al., Phase I trial of intraperitoneal taxol: a Gynecoloic

Oncology Group study. J Clin Oncol, 1992. 10(9): p. 1485-1491.

6. Dedrick, R.L., et al., Pharmacokinetic rationale for peritoneal drug

administration in the treatment of ovarian cancer. Cancer Treat Rep, 1978. 62(1):

p. 1-11.

7. Marchettini, P., et al., Docetaxel: pharmacokinetics and tissue levels after

intraperitoneal and intravenous administration in a rat model. Cancer Chemother

Pharmacol, 2002. 49(6): p. 499-503.

26

8. Nemes, K.B., et al., Oral, intraperitoneal and intravenous pharmacokinetics of

deramciclane and its N-desmethyl metabolite in the rat. J Pharm Pharmacol, 2000.

52(1): p. 47-51.

9. Krasner, C.N., et al., Case 11-2006. New Eng J Med, 2006. 354(15): p. 1615-

1625.

10. Markman, M., et al., Phase III Trial of Standard-Dose Intravenous Cisplatin Plus

Paclitaxel Versus Moderately High-Dose Carboplatin Followed by Intravenous

Paclitaxel and Intraperitoneal Cisplatin in Small-Volume Stage III Ovarian

Carcinoma: An Intergroup Study of the Gynecologic Oncology Group,

Southwestern Oncology Group, and Eastern Cooperative Oncology Group. J

Clinl Oncol, 2001. 19(4): p. 1001-1007.

11. NCI, NCI Clinical Announcement on Intraperitoneal Therapy for Ovarian Cancer.

2006: p. http://ctep.cancer.gov/highlights/docs/clin_annc_010506.pdf.

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

The Lancet Oncol, 2003. 4(5): p. 277-283.

13. Zeimet, A., et al., Pros and Cons of Intraperitoneal Chemotherapy in the

Treatment of Epithelial Ovarian Cancer. Anticancer Res, 2009. 29(7): p. 2803-

2808.

14. Wenzel, L.B., et al., Health-Related Quality of Life During and After

Intraperitoneal Versus Intravenous Chemotherapy for Optimally Debulked

Ovarian Cancer: A Gynecologic Oncology Group Study. J Clin Oncol, 2007.

25(4): p. 437-443.

27

15. Armstrong, D.K., Relapsed Ovarian Cancer: Challenges and Management

Strategies for a Chronic Disease. Oncologist, 2002. 7(suppl 5): p. 20-28.

16. Poveda, A., et al., Update in the management of ovarian and cervical carcinoma.

Clin Transl Oncol, 2007. 9(7): p. 443-451.

17. Vassileva, V., et al., Effects of sustained and intermittent paclitaxel therapy on

tumor repopulation in ovarian cancer. Mol Cancer Ther, 2008. 7(3): p. 630-637.

18. Hall, J.C., et al., The pathobiology of peritonitis. Gastroenterology, 1998. 114(1):

p. 185-196.

19. Dufrane, D., et al., The influence of implantation site on the biocompatibility and

survival of alginate encapsulated pig islets in rats. Biomaterials, 2006. 27(17): p.

3201-3208.

20. Yeo, Y., et al., Peritoneal application of chitosan and UV-cross-linkable chitosan.

J Biomed Mater Res A, 2006. 78A(4): p. 668-675.

21. Yeo, Y. et al., Polymers in the prevention of peritoneal adhesions. Eur J Pharm

Biopharm, 2008. 68(1): p. 57-66.

22. Yeo, Y., et al., In situ cross-linkable hyaluronan hydrogels containing polymeric

nanoparticles for preventing postsurgical adhesions. Ann Surg, 2007. 245(5): p.

819-824.

23. Burns, J.W., et al., Prevention of tissue injury and postsurgical adhesions by

precoating tissues with hyaluronic acid solutions. J Surg Res, 1995. 59(6): p. 644-

652.

24. Rodgers, K.E., et al., Reduction of adhesion formation with hyaluronic acid after

peritoneal surgery in rabbits. Fertil Steril, 1997. 67(3): p. 553-558.

28

25. Bulpitt, P., et al., New strategy for chemical modification of hyaluronic acid:

preparation of functionalized derivatives and their use in the formation of novel

biocompatible hydrogels. J Biomed Mater Res, 1999. 47(2): p. 152-169.

26. Park, S.N., et al., Preparation and characterization of biodegradable anti-

adhesive membrane for peritoneal wound healing. J Mater Sci Mater Med, 2007.

18(3): p. 475-482.

27. Leach, R.E., et al., Reduction of postsurgical adhesion formation in the rabbit

uterine horn model with use of hyaluronate/carboxymethylcellulose gel. Fertil

Steril, 1998. 69(3): p. 415-418.

28. Caicco, M.J., et al., Characterization of hyaluronan–methylcellulose hydrogels

for cell delivery to the injured spinal cord. J Biomed Mater Res A, 2012: p. 1472-

1477.

29. Hudson, S.P., et al., Injectable in situ cross-linking hydrogels for local antifungal

therapy. Biomaterials, 2010. 31(6): p. 1444-1452.

30. Ito, T., et al., Dextran-based in situ cross-linked injectable hydrogels to prevent

peritoneal adhesions. Biomaterials, 2007. 28(23): p. 3418-3426.

31. Patenaude, M., et al., Injectable, Mixed Natural-Synthetic Polymer Hydrogels

with Modular Properties. Biomacromolecules, 2012. 13(2): p. 369-378.

32. Risbud, M., et al, Chitosan-polyvinyl pyrrolidone hydrogel does not activate

macrophages: potentials for transplantation applications. Cell Transplant, 2001.

10(2): p. 195-202.

33. Prasitsilp, M., et al., Cellular responses to chitosan in vitro: the importance of

deacetylation. J Mater Sci Mater Med, 2000. 11(12): p. 773-778.

29

34. Yeo, Y., et al., In situ cross-linkable hyaluronic acid hydrogels prevent post-

operative abdominal adhesions in a rabbit model. Biomaterials, 2006. 27(27): p.

4698-4705.

35. Ito, T., et al., The prevention of peritoneal adhesions by in situ cross-linking

hydrogels of hyaluronic acid and cellulose derivatives. Biomaterials, 2007. 28(6):

p. 975-983.

36. Bajaj, G., et al., Hyaluronic acid-based hydrogel for regional delivery of

paclitaxel to intraperitoneal tumors. J Control Release, 2012. 158(3): p. 386-392.

37. Ilevbare, G.A., et al., Impact of Polymers on Crystal Growth Rate of Structurally

Diverse Compounds from Aqueous Solution. Mol Pharm, 2013. 10(6): p. 2381-

2393.

38. Kwon, S., et al., Physicochemical characteristics of self-assembled nanoparticles

based on glycol chitosan bearing 5 beta-cholanic acid. Langmuir, 2003. 19: p.

188-193.

39. Ta, H.T., et al., Injectable chitosan hydrogels for localised cancer therapy. J

Control Release, 2008. 126(3): p. 205-216.

40. Kohane, D.S., et al., Biodegradable polymeric microspheres and nanospheres for

drug delivery in the peritoneum. J Biomed Mater Res A, 2006. 77(2): p. 351-361.

41. Shin, H.C., et al., Pharmacokinetic study of 3-in-1 poly(ethylene glycol)-block-

poly(D, L-lactic acid) micelles carrying paclitaxel, 17-allylamino-17-

demethoxygeldanamycin, and rapamycin. J Control Release, 2012. 163(1): p. 93-

99.

30

42. Cho, H., et al., Poly(ethylene glycol)-block-poly(epsilon-caprolactone) micelles

for combination drug delivery: evaluation of paclitaxel, cyclopamine and

gossypol in intraperitoneal xenograft models of ovarian cancer. J Control Release,

2013. 166(1): p. 1-9.

43. Santovena, A., et al., Pharmacokinetics analysis of sustained release hGH

biodegradable implantable tablets using a mouse model of human ovarian cancer.

Int J Pharm, 2010. 388(1-2): p. 175-180.

44. Tang, Q., et al., Preparation of anti-tumor nanoparticle and its inhibition to

peritoneal dissemination of colon cancer. PLoS One, 2014. 9(6):p. e98455.

45. Wang, F., et al., A mechanistic model of controlled drug release from polymer

millirods: effects of excipients and complex binding. J Control Release, 2007.

119(1): p. 111-20.

46. Middleton, J.C., et al., Synthetic biodegradable polymers as orthopedic devices.

Biomaterials, 2000. 21(23): p. 2335-2346.

47. Wu, L. et al., In vitro degradation of three-dimensional porous poly(d,l-lactide-

co-glycolide) scaffolds for tissue engineering. Biomaterials, 2004. 25(27): p.

5821-5830.

48. Tsai, M., et al., Effects of Carrier on Disposition and Antitumor Activity of

Intraperitoneal Paclitaxel. Pharm Res, 2007. 24(9): p. 1691-1701.

49. Lu, Z., et al., Tumor-Penetrating Microparticles for Intraperitoneal Therapy of

Ovarian Cancer. J Pharmacol Exp Ther, 2008. 327(3): p. 673-682.

50. Trissel, L.A., et al., Pharmaceutical Properties of Paclitaxel and Their Effects on

Preparation and Administration. Pharmacotherapy, 1997. 17(2): p. 133-139.

31

51. Panchagnula, R., et al., Pharmaceutical aspects of paclitaxel. Int J Pharm, 1998.

172(1–2): p. 1-15.

52. Bouquet, W., et al., Paclitaxel/β-cyclodextrin complexes for hyperthermic

peritoneal perfusion – Formulation and stability. Eur J Pharm and Biopharm,

2007. 66(3): p. 391-397.

53. Bouquet, W., et al., In vivo toxicity and bioavailability of Taxol and a

paclitaxel/beta-cyclodextrin formulation in a rat model during HIPEC. Ann Surg

Oncol, 2010. 17(9): p. 2510-2517.

54. Bouquet, W., et al., Antitumour efficacy of two paclitaxel formulations for

hyperthermic intraperitoneal chemotherapy (HIPEC) in an in vivo rat model.

Pharm Res, 2011. 28(7): p. 1653-1660.

55. Mohamed, F., et al., Pharmacokinetics and tissue distribution of intraperitoneal

paclitaxel with different carrier solutions. Cancer Chemother Pharmacol, 2003.

52(5): p. 405-410.

56. Mohamed, F., et al., Pharmacokinetics and tissue distribution of intraperitoneal

docetaxel with different carrier solutions. J Surg Res, 2003. 113(1): p. 114-120.

57. Wang, J., et al., Tumor priming enhances siRNA delivery and transfection in

intraperitoneal tumors. J Control Release, 2014. 178(0): p. 79-85.

58. De Smet, L., et al., Development of a Nanocrystalline Paclitaxel Formulation for

Hipec Treatment. Pharm Res, 2012. 29(9): p: 2398-2406.

59. Cho, H., et al., Polymeric Micelles for Apoptosis-Targeted Optical Imaging of

Cancer and Intraoperative Surgical Guidance. PLoS One, 2014. 9(2): p. e89968.

32

60. Cho, H. et al., Polymeric Micelles for Neoadjuvant Cancer Therapy and Tumor-

Primed Optical Imaging. ACS Nano, 2011. 5(11): p. 8721-8729.

61. Cho, H., et al., In vivo cancer imaging by poly(ethylene glycol)-b-poly(ɛ-

caprolactone) micelles containing a near-infrared probe. Nanomedicine, 2012.

8(2): p. 228-236.

62. Hirano, K. et al., Lymphatic transport of liposome-encapsulated agents: effects of

liposome size following intraperitoneal administration. J Pharm Sci, 1985. 74(9):

p. 915-921.

63. Zahedi, P., et al., Chitosan–phospholipid blend for sustained and localized

delivery of docetaxel to the peritoneal cavity. Int J Pharm, 2009. 377(1–2): p. 76-

84.

64. Vassileva, V., et al., Novel biocompatible intraperitoneal drug delivery system

increases tolerability and therapeutic efficacy of paclitaxel in a human ovarian

cancer xenograft model. Cancer Chemother Pharmacol, 2007. 60(6): p. 907-914.

65. Vassileva, V., et al., Efficacy assessment of sustained intraperitoneal paclitaxel

therapy in a murine model of ovarian cancer using bioluminescent imaging. Br J

Cancer, 2008. 99(12): p. 2037-2043.

66. Grant, J., et al., Hybrid films from blends of chitosan and egg phosphatidylcholine

for localized delivery of paclitaxel. J Pharm Sci, 2005. 94(7): p. 1512-1527.

67. Lim Soo, P., et al., Drug release mechanism of paclitaxel from a chitosan–lipid

implant system: Effect of swelling, degradation and morphology. Eur J Pharm

Biopharm, 2008. 69(1): p. 149-157.

33

68. Ho, E.A., et al., In vitro and in vivo characterization of a novel biocompatible

polymer–lipid implant system for the sustained delivery of paclitaxel. J Control

Release, 2005. 104(1): p. 181-191.

69. Ho, E.A., et al., Impact of intraperitoneal, sustained delivery of paclitaxel on the

expression of P-glycoprotein in ovarian tumors. J Control Release, 2007. 117(1):

p. 20-27.

70. Grant, J., et al., Influence of molecular organization and interactions on drug

release for an injectable polymer-lipid blend. Int J Pharm, 2008. 360(1-2): p. 83-

90.

71. De Souza, R., et al., Biocompatibility of injectable chitosan–phospholipid implant

systems. Biomaterials, 2009. 30(23–24): p. 3818-3824.

72. Zahedi, P., et al., An injectable depot system for sustained intraperitoneal

chemotherapy of ovarian cancer results in favorable drug distribution at the

whole body, peritoneal and intratumoral levels. J Control Release, 2012. 158(3):

p. 379-385.

73. De Souza, R., et al., Continuous Docetaxel Chemotherapy Improves Therapeutic

Efficacy in Murine Models of Ovarian Cancer. Mol Cancer Ther, 2010. 9(6): p.

1820-1830.

74. Zahedi, P., et al., Combination Drug Delivery Strategy for the Treatment of

Multidrug Resistant Ovarian Cancer. Mol Pharm, 2010. 8(1): p. 260-269.

75. Zentner, G.M., et al., Biodegradable block copolymers for delivery of proteins

and water-insoluble drugs. J Control Release, 2001. 72(1–3): p. 203-215.

34

76. Elstad, N.L. et al., OncoGel (ReGel/paclitaxel)--clinical applications for a novel

paclitaxel delivery system. Adv Drug Deliv Rev, 2009. 61(10): p. 785-794.

77. Cho, H. et al., Thermosensitive poly-(d,l-lactide-co-glycolide)-block-poly(ethylene

glycol)-block-poly-(d,l-lactide-co-glycolide) hydrogels for multi-drug delivery. J

Drug Target, 2014. 22(7): p. 669-677.

78. Deligkaris, K., et al., Hydrogel-based devices for biomedical applications. Sens

Actuators B, 2010. 147(2): p. 765-774.

79. Yeo, Y., et al., Prevention of peritoneal adhesions with an in situ cross-linkable

hyaluronan hydrogel delivering budesonide. J Control Release, 2007. 120(3): p.

178-185.

80. Yeo, Y., et al., Peritoneal adhesion prevention with an in situ cross-linkable

hyaluronan gel containing tissue-type plasminogen activator in a rabbit repeated-

injury model. Biomaterials, 2007. 28(25): p. 3704-3713.

81. Emoto, S., et al., Intraperitoneal administration of cisplatin via an in situ cross-

linkable hyaluronic acid-based hydrogel for peritoneal dissemination of gastric

cancer. Surg Today, 2013: p. 1-8.

82. Sherwood, L., Human physiology : from cells to systems 2007, Australia; Belmont,

CA: Thomson/Brooks/Cole.

83. Watson, M.S., Oxford handbook of palliative care 2009, Oxford; New York:

Oxford University Press.

84. D'Addio, S.M. et al., Controlling drug nanoparticle formation by rapid

precipitation. Adv Drug Deliv Rev, 2011. 63(6): p. 417-426.

35

85. Patravale, V.B., et al., Nanosuspensions: a promising drug delivery strategy. J

Pharm Pharmacol, 2004. 56(7): p. 827-840.

86. Sun, B. et al., Nanocrystals for the parenteral delivery of poorly water-soluble

drugs. Curr Opin Solid State Mater Sci, 2012. 16(6): p: 295-301.

87. Van Eerdenbrugh, B., et al., Top-down production of drug nanocrystals:

Nanosuspension stabilization, miniaturization and transformation into solid

products. Int J Pharm, 2008. 364(1): p. 64-75.

88. Lu, Y., et al., Development and evaluation of transferrin-stabilized paclitaxel

nanocrystal formulation. J Control Release, 2013.

89. Jeyachandran, Y.L., et al., Quantitative and Qualitative Evaluation of

Adsorption/Desorption of Bovine Serum Albumin on Hydrophilic and

Hydrophobic Surfaces. Langmuir, 2009. 25(19): p. 11614-11620.

90. Seo, J., et al., Facile internalization of paclitaxel on titania nanoparticles in

human lung carcinoma cells after adsorption of serum proteins. J Nanopart Res,

2012. 14(10): p. 1-8.

91. Kratz, F. et al., Clinical impact of serum proteins on drug delivery. J Control

Release, 2012. 161(2): p. 429-445.

92. Torosean, S., et al., Nanoparticle uptake in tumors is mediated by the interplay of

vascular and collagen density with interstitial pressure. Nanomedicine, 2013.

9(2): p. 151-158.

93. Kohli, A.G., et al., Improving the distribution of Doxil(R) in the tumor matrix by

depletion of tumor hyaluronan. J Control Release, 2014. 20(14): p. 322-328.

36

94. Johansson, A. et al., Remodeling of tumor stroma and response to therapy.

Cancers, 2012. 4(2): p. 340-353.

95. Minchinton, A.I. et al., Drug penetration in solid tumours. Nat Rev Cancer, 2006.

6(8): p. 583-592.

96. Grantab, R., S. et al., The penetration of anticancer drugs through tumor tissue as

a function of cellular adhesion and packing density of tumor cells. Cancer Res,

2006. 66(2): p. 1033-1039.

97. Tannock, I.F., et al., Limited penetration of anticancer drugs through tumor tissue:

a potential cause of resistance of solid tumors to chemotherapy. Clin Cancer Res,

2002. 8(3): p. 878-884.

98. Kyle, A.H., et al., Limited tissue penetration of taxanes: a mechanism for

resistance in solid tumors. Clin Cancer Res, 2007. 13(9): p. 2804-2810.

99. Lee, J.H., et al., The distribution and retention of paclitaxel and doxorubicin in

multicellular layer cultures. Oncol Rep, 2012. 27(4): p. 995-1002.

100. Nicholson, K.M., et al., Influence of drug exposure parameters on the activity of

paclitaxel in multicellular spheroids. Eur J Cancer, 1997. 33(8): p. 1291-1298.

101. Ho, W.Y., et al., Development of multicellular tumor spheroid (MCTS) culture

from breast cancer cell and a high throughput screening method using the MTT

assay. PLoS One, 2012. 7(9): p. e44640.

102. Kuh, H.J., et al., Determinants of paclitaxel penetration and accumulation in

human solid tumor. J Pharmacol Exp Ther, 1999. 290(2): p. 871-880.

37

103. Jang, S.H., et al., Enhancement of paclitaxel delivery to solid tumors by

apoptosis-inducing pretreatment: effect of treatment schedule. J Pharmacol Exp

Ther, 2001. 296(3): p. 1035-1042.

104. Ishida, T. et al., Alteration of tumor microenvironment for improved delivery and

intratumor distribution of nanocarriers. Biol Pharm Bull, 2013. 36(5): p. 692-697.

105. Li, L., et al., Improved intratumoral nanoparticle extravasation and penetration

by mild hyperthermia. J Control Release, 2013. 167(2): p. 130-137.

106. Helm, J.H., et al., Cytoreductive Surgery and Hyperthermic Intraperitoneal

Chemotherapy for Malignant Peritoneal Mesothelioma: A Systematic Review and

Meta-analysis. Ann Surg Oncol, 2015. 22(5): p. 1686-1693.

107. Ihemelandu, C., L. et al., Iterative Cytoreductive Surgery and Hyperthermic

Intraperitoneal Chemotherapy for Recurrent or Progressive Diffuse Malignant

Peritoneal Mesothelioma: Clinicopathologic Characteristics and Survival

Outcome. Ann Surg Oncol, 2015. 22(5): p. 1680-1685.

108. Teo, M.C., et al., Colorectal peritoneal carcinomatosis treated with cytoreductive

surgery and hyperthermic intraperitoneal chemotherapy: The experience of a

tertiary Asian center. Asian J Surg, 2014. 21(14): p. 1-9.

109. Suidan, R.S., et al., A comparison of primary intraperitoneal chemotherapy to

consolidation intraperitoneal chemotherapy in optimally resected advanced

ovarian cancer. Gynecol Oncol, 2014. 17(14): p. 468-472.

110. Jarvinen, P., et al., Comparison of serial debulking and cytoreductive surgery with

hyperthermic intraperitoneal chemotherapy in pseudomyxoma peritonei of

appendiceal origin. Int J Colorectal Dis, 2014. 29(8): p. 999-1007.

38

111. Sammartino, P., et al., Long-term results after proactive management for

locoregional control in patients with colonic cancer at high risk of peritoneal

metastases. Int J Colorectal Dis, 2014. 29(9): p. 1081-1089.

112. Safra, T., et al., Cytoreduction surgery with hyperthermic intraperitoneal

chemotherapy in recurrent ovarian cancer improves progression-free survival,

especially in BRCA-positive patients-A case-control study. J Surg Oncol, 2014.

24(10): p. 23688.

113. Cui, H.B., et al., Effect of neoadjuvant chemotherapy combined with hyperthermic

intraperitoneal perfusion chemotherapy on advanced gastric cancer. Exp Ther

Med, 2014. 7(5): p. 1083-1088.

114. Saggar, J.K., et al., The tumor microenvironment and strategies to improve drug

distribution. Front Oncol, 2013. 3: p: 154.

115. Choi, I.K., et al., Strategies to increase drug penetration in solid tumors. Front

Oncol, 2013. 3: p: 193.

116. Sagnella, S.M., et al., Dextran-based Doxorubicin nanocarriers with improved

tumor penetration. Biomacromolecules, 2014. 15(1): p. 262-275.

117. Patel, K.J., et al., Use of the proton pump inhibitor pantoprazole to modify the

distribution and activity of doxorubicin: a potential strategy to improve the

therapy of solid tumors. Clin Cancer Res, 2013. 19(24): p. 6766-6776.

118. Saggar, J.K. et al., Activity of the hypoxia-activated pro-drug TH-302 in hypoxic

and perivascular regions of solid tumors and its potential to enhance therapeutic

effects of chemotherapy. Int J Cancer, 2014. 134(11): p. 2726-2734.

39

119. Zhang, L., et al, Hyaluronan on the surface of tumor cells is correlated with

metastatic behavior. Cancer Res, 1995. 55(2): p. 428-433.

120. Kimata, K., et al., Increased synthesis of hyaluronic acid by mouse mammary

carcinoma cell variants with high metastatic potential. Cancer Res, 1983. 43(3): p.

1347-1354.

121. Turley, E.A. et al., Glycosaminoglycan production by murine melanoma variants

in vivo and in vitro. Cancer Res, 1985. 45(10): p. 5098-5105.

122. McBride, W.H. et al., Hyaluronidase-sensitive halos around adherent cells. Their

role in blocking lymphocyte-mediated cytolysis. J Exp Med, 1979. 149(2): p. 507-

515.

123. Kayastha, S., et al., Expression of the hyaluronan receptor, CD44S, in epithelial

ovarian cancer is an independent predictor of survival. Clin Cancer Res, 1999.

5(5): p. 1073-1076.

124. Anttila, M.A., et al., High levels of stromal hyaluronan predict poor disease

outcome in epithelial ovarian cancer. Cancer Res, 2000. 60(1): p. 150-155.

125. Hiltunen, E.L., et al., Elevated hyaluronan concentration without hyaluronidase

activation in malignant epithelial ovarian tumors. Cancer Res, 2002. 62(22): p.

6410-6413.

126. Picaud, L., et al., Evaluation of the effects of hyaluronic acid-carboxymethyl

cellulose barrier on ovarian tumor progression. J Ovarian Res, 2014. 7(1): p. 40.

127. Hubbard, S.C. et al., Effects of a hyaluronan-based membrane (Seprafilm) on

intraperitoneally disseminated human colon cancer cell growth in a nude mouse

model. Dis Colon Rectum, 2002. 45(3): p. 334-341.

40

128. Tan, B., et al., Sodium hyaluronate enhances colorectal tumour cell metastatic

potential in vitro and in vivo. Br J Surg, 2001. 88(2): p. 246-250.

41

CHAPTER 2. DEVELOPMENT AND CHARACTERIZATION OF NANOPARTICLE DEPOT FOR INTRAPERITONEAL CHEMOTHERAPY2

2.1 Introduction

The potential advantages of IP chemotherapy have been discussed thoroughly.

However, frequently mentioned problems are the complications related to IP infusion,

including abdominal pain, intolerance to a high level of drug, and discomfort related to

the catheter implantation [1]. We speculate that these problems mainly stem from the

difficulty in controlling drug release. Small molecule drugs such as paclitaxel (PTX) or

docetaxel were cleared from the peritoneal cavity in less than one day [2-4]. The short IP

retention time requires frequent or continuous dosing, necessitating the use of large

volume of treatment and indwelling catheters. For IP chemotherapy to provide the

anticipated benefits, it is critical that the formulation control the drug release and avoid

burst initial release, followed by immediate absorption to the systemic circulation.

Considering this need, we previously used a hyaluronic acid (HA)-based hydrogel

(gel) as a carrier of PTX provided in the form of micrometer-scale precipitates (PPT) to

improve their IP retention and control the drug release [5]. The gel remained in the

peritoneal cavity and maintained the high level of local PTX concentration for 2 weeks.

2 The content of this chapter is adapted with permission from Sara A. Abouelmagd, Bo Sun, Alice C. Chang, et al., Release kinetics study of poorly water-soluble drugs from nanoparticles: Are we doing it right? Mol. Pharm. 2015, 12(3), p 997-1003. Copyright (2015) American Chemical Society.

42

However, when tested in a murine model of IP SKOV-3 human ovarian cancer, the PPT-

gel had no more benefit than Taxol-gel, which did not retain PTX as well as PPT-gel did.

One of the possible explanations was that the dissolution of PPT was too slow to provide

an effective local drug concentration.

In the present study, we aim to overcome this challenge by designing a drug

delivery system that can localize PTX in the peritoneal cavity and provide sustained, yet

uninterrupted PTX release. PTX is used as a model drug because it is a first-line therapy

of ovarian cancer for both intravenous and intraperitoneal administration [6-9]. Besides,

PTX is a well-known poorly water-soluble drug, previously found to be challenging to

control the release in the peritoneal cavity with a limited amount of fluid.

PTX is formulated as nanocrystals (NCs), crystalline particles of poorly soluble

drugs in the nanometer range [10]. Due to the small size of NC and thus, the high surface

area to volume ratio, NC can increase the dissolution rate of drug particles [11].

Typically, NCs are produced by breaking down large particles (“top-down”) or

crystallizing from drug solutions (“bottom-up”) of the drug itself with an optional aid

with surfactants or polymeric stabilizers on the surface [10, 12]. Here, we produce PNC

by nonsolvent and temperature-induced crystallization [13] and evaluate its in-vitro

dissolution in comparison with PPT.

PNCs are then embedded in HA gel. IP-administered small molecule drugs could

be cleared out from peritoneal cavity in a few hours which compromised the

effectiveness of local chemotherapy [14-16]. In situ forming gel has been developed as a

standalone drug delivery system to handle the short residence time of chemotherapeutic

43

agents [5, 17-21]. HA gel is degradable in-vivo by prevalent hyaluronidase and highly

biocompatible [22-25]. In this study, we anticipate that HA gel will provide a local

reservoir of PNC and prolong the retention of PTX in the peritoneal cavity to provide

sustained local delivery of chemotherapy to ovarian tumors.

In order to predict PTX dissolution within the peritoneal cavity, it is necessary to

develop a dissolution method that accounts for biological and physicochemical factors

relevant to in-vivo conditions. Several attempts have been made to estimate the PTX

release profiles from gel formulations in conditions mimicking the peritoneal cavity with

a limited fluid volume and amphiphilic components.

2.2 Materials and Methods

2.2.1 Materials

Hyaluronic acid (HA, 20 kDa and 500 kDa) was purchased from Lifecore

Biomedical, LLC (Chaska, MN, USA). Paclitaxel (PTX) was a gift of Samyang Genex

Corp (Seoul, Korea). Carbamazepine was purchased from Enzo life sciences (Plymouth

Meeting, PA, USA). Cremophor ELP was a gift from BASF (New York, NY, USA).

Carboxyl methylcellulose sodium (CMC, 250 kDa) was purchased from Sigma-Aldrich.

Cell culture medium and supplements were purchased from Invitrogen (Carlsbad, CA,

USA). All other reagents were purchased from Sigma-Aldrich (St. Louis, MO, USA).

44

2.2.2 Preparation and Characterization of PTX Nanocrystals (PNC)

PTX nanocrystals (PNC) were prepared according to the published method [13].

Briefly, 4 mg/mL PTX/ethanol solution was added to 20 mL of deionized (DI) water and

stirred at 42 rcf for 10 min in a round-bottom flask immerged in a sonication bath filled

with ice water. The formed PNC was captured on a 100 nm polycarbonate membrane and

resuspended in DI water. Taxol and PTX precipitates (PPT) were prepared as described

previously [5]. Taxol was prepared by dluting “Taxol concentrate” (6 mg PTX dissolved

in 1 mL of 1:1 mixture of Cremophor ELP and ethanol). PPT was prepared by the

addition of 30 mg/mL of PTX-dimethyl sulfoxide (DMSO) solution to DI water without

low temperature, vigorous stirring and sonication. Particle size of PNC was measured

with a Zetasizer Nano-ZS90 (Malvern instruments, Westborough, MA, USA). The zeta

potential of PNC and PPT were measured with a Zeta sizer Nano-ZS90 in 1 mM

phosphate buffer (pH 7.4). The morphology of lyophilized PNC and PPT was visualized

with a FEI Nova nanoSEM field emission scanning electron microscopy (Hillsboro, OR,

USA). Freeze dried PNC or PPT was sputter-coated with platinum for 1 min and

observed with a high resolution through-the-lens detector under 5kV accelerating voltage

and spot size 3. PPT suspended in DI water was observed with Axio Imager 2 microscope

with polarized light (Carl Zeiss Microscopy GmbH, Germany). X-ray powder diffraction

(XPRD) patterns of lyophilized PNC and PPT were obtained on a Siemens D5000 X-ray

diffractometer with a Cu Kα radiation source (40kV, 40 mA). All the measurements were

conducted at room temperature over an angle (2θ) range of 5-40° with a step size of 0.02°

and a scan rate of 4°/min.

45

2.2.3 Synthesis of Gel Precursors (HA-ADH, HA-CHO, CMC-CHO)

In-situ crosslinkable HA derivatives, HA-adipic acid dihydrazide (HA-ADH) and

HA-aldehyde (HA-CHO), and a crosslinkable HA gel were prepared as described

previously [5, 26]. Briefly, HA-ADH was synthesized by conjugating adipic dihydrazide

to carboxyl groups in HA which was catalyzed by 1-ethyl-3-carbodiimide (EDC) and 1-

hydroxybenzotriazole (HOBt) at pH 6.8 and room temperature. HA-CHO was produced

by oxidizing HA with sodium periodate. HA-ADH and HA-CHO were purified by

dialysis, freeze dried and stored at 4°C until use. Carboxyl methylcellulose-aldehyde

(CMC-CHO) was synthesized in the same approach as HA-CHO.

2.2.4 Synthesis of Hydrophobically Modified Gel Precursor (EtCA-CMC-CHO)

2.2.4.1 Synthesis of 5β-cholanic Acid Methyl Ester (MeCA) (Scheme 1)

500 mg of 5β-cholanic acid (CA) (1.4 mmol) was dissolved in 50 mL of methanol

with 180 µL concentrated hydrochloride acid (4.9 µmol) at 60°C. The solution was

stirred for 6h under reflux. The solution was cooled to room temperature and added

dropwise into cold deionized water to obtain precipitates, which were further filtrated and

washed with cold methanol for 3 times and cold deionized water for 3 times. The product

was dried under vacuum at room temperature to obtain MeCA.

2.2.4.2 Synthesis of Aminoethyl 5β-cholanoamide (EtCA) (Scheme 1)

450 mg of MeCA (1.2 mmol) was dissolved in 8 mL of ethylene diamine (119.6

mmol) under stirring and reflux for 6h at 130 °C. The solution was cooled to room

46

temperature and added dropwise into cold deionized water to obtain precipitates, which

were further filtrated and washed with cold deionized water for 5 times. The final product

(EtCA) was dried under vacuum at room temperature and its chemical structure was

characterized with 1H NMR in CD3OD at a concentration of 10 mg/mL.

Scheme 1. Synthesis scheme of aminoethyl 5β-cholanoamide.

2.2.4.3 Synthesis of EtCA-CMC-CHO Conjugate (Scheme 2)

41 mg of CMC-CHO (100 µmol) was dissolved in 37 mL of methanol/water

(2v/1v) containing 20.6 mg of N, N’-Dicyclohexylcarbodiimide (DCC, 100 µmol) and

11.5 mg of N-Hydroxysuccinimide (NHS, 100 µmol) under stirring at room temperature

for 1h. After 4 or 12 mg of EtCA (10 or 30 µmol) in 1 mL of methanol was slowly added,

the reaction mixture was stirred for 12h. The resulting solution was dialyzed against

methanol/water (2v/1v) for 2 days and deionized water for another 2 days. EtCA0.1 or 0.3-

CMC-CHO was lyophilized and stored at -20°C until use.

47

Scheme 2. Synthesis scheme of EtCA-CMC-CHO conjugate.

2.2.5 Preparation of Gel formulations

Gels were prepared by suspending PNC or PPT in solutions of gel precursors (one

ADH derivative and one CHO derivative) in phosphate buffer saline (PBS, pH 7.4) and

extruding them through a common outlet using a double-barreled syringe. HA gel was

made of HA-ADH and HA-CHO; HA-CMC gel was made of HA-ADH and CMC-CHO.

The polymer concentration in gel was 40 mg/mL. When EtCA0.1-CMC-CHO was used,

37.5% (w/w) of HA-CHO was replaced with EtCA0.1-CMC-CHO. HA-EtCA0.1-CMC gel

was made of 40 mg/mL HA-ADH, 25 mg/mL CMC-CHO and 15 mg/mL EtCA0.1-CMC-

CHO.

48

2.2.6 Evaluation of Gel Precursors and Crosslinked Gels

2.2.6.1 Hydrophilicity of Gel Precursors

Contact angle is defined as the angle formed by the intersection of the liquid-solid

interface and the liquid-vapor interface [27]. The larger a contact angle is, the more

hydrophobic the surface is. The hydrophilicity of a specific material can be evaluated by

measuring the contact angle of water droplet placed on the thin film made of the material.

Four gel precursors, HA-CHO, CMC-CHO and EtCA0.1 or 0.3-CMC-CHO, were

dissolved in deionized water at 10 mg/mL. The solutions were casted on glass slides

respectively and dried at ambient temperature for 12h to form thin films. The contact

angle of water droplet on each polymeric film was measured with a contact angle

goniometer.

2.2.6.2 Toxicity of EtCA and EtCA-CMC-CHO

Toxicity of CA, MeCA, EtCA and EtCA0.1-CMC-CHO were studied with

NIH/3T3 fibroblast cells (ATCC). NIH/3T3 cells were maintained in Dulbecco's

Modified Eagle's Medium (DMEM) medium supplemented with 10% bovine calf serum

(BCS) and 1% penicillin-streptomycin. Cells were plated in a 96-well plate at a density of

8,000 cells per well with 0.2 mL of complete medium. After 24h incubation, CA, MeCA,

EtCA (in methanol) or EtCA0.1-CMC-CHO (in PBS) was added to each well at varied

concentrations and incubated with cells for 2 days. A control group was treated with an

equal volume of methanol or PBS. The cell viability was evaluated with the MTT (3-(4,

5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide) assay. The absorbance of the

49

solubilized formazan was measured with a SpectraMax M3 microplate reader (Molecular

Devices, Sunnyvale, CA, USA) at a wavelength of 562 nm. The measured absorbance

was normalized to the absorbance of the control group.

2.2.6.3 Toxicity of Gels

Toxicity of gels was evaluated with NIH/3T3 cells using Transwells. Cells were

plated in a 12-well plate at a density of 17,500 cells per wells with 1.5 mL DMEM

complete medium (cell density equal to 5,000 cells per well in 96-well plate). CMC-

CHO, HA-CMC gel or HA-EtCA0.1-CMC gel was added to the apical side of a Transwell

polycarbonate membrane insert (pore size = 0.4 µm), placed in the 12-well plate, and

incubated with cells for 2 days. Cell viability was evaluated with MTT assay as described

above.

2.2.7 PTX Solubility and Stability in Various Media

2.2.7.1 PTX Solubility in PBS, PBS Containing Serum, or PBS Containing Tween 80

PTX solubility in PBS, PBS containing 0.2 v/v% Tween 80 (Tween/PBS), and

PBS containing 50 v/v% FBS (50-FBS/PBS) were determined by incubating excess PTX

(0.6 – 2.4 mg) in 1 mL of each medium at 37 °C for 7 or 24 h with agitation. Samples

were centrifuged at 9300 rcf for 20 min to separate a supernatant. PTX dissolved in PBS

and Tween/PBS was analyzed with HPLC as described in Section 2.2.4. PTX dissolved

in 50-FBS/PBS was first extracted with ethyl acetate and reconstituted in the HPLC

mobile phase prior to HPLC analysis.

50

2.2.7.2 PTX Stability in PBS Containing Tween 80 or PBS Containing Serum

The stability of a drug was an important issue if it would be tested or delivered for

an extended period of time. PTX stability was first tested in a fully dissolved state (PTX

concentration below the solubility limit). 1.5 µg/mL of PTX solution in Tween/PBS was

prepared by diluting 10 mg/mL PTX-DMSO solution with Tween/PBS (final DMSO

concentration: 0.015%). The solution was divided into several 1 mL aliquots, and the

initial PTX concentration was determined with 7 of them. The remaining aliquots were

incubated at 37°C, and 3 aliquots were taken at predetermined time points and kept at -

80°C until HPLC analysis. The frozen samples were thawed and filtered through 0.45 µm

filters for HPLC analysis. The concentration of intact PTX at each time point was divided

by the original PTX concentration (1.5 µg/mL) and expressed as the percentage of

original PTX.

PTX stability in 50-FBS/PBS was tested in its solid form of PNC and PPT at a

concentration of 100 µg/mL (above solubility limit) and compared to that of Taxol. These

PTX formulations were incubated in 50-FBS/PBS at 37 °C for 10 days. PTX remained in

the suspensions was extracted with ethyl acetate and analyzed with HPLC.

2.2.8 Dissolution Kinetics of PNC

PTX dissolution was first tested with the dialysis method. Three milliliters of PBS

containing 200 µg PNC was injected into a dialysis cassette (MWCO= 3.5 kDa). The

cassette was placed in 200 mL PBS. Therefore, the initial PTX concentrations in cassette

and in the system were 66.7 µg/mL and 1 µg/mL, respectively. The cassette was

51

incubated in PBS at 37 °C on an orbital shaker for 10 days. At predetermined time point,

5 mL of the release medium was sampled and replaced with 5 mL of fresh PBS. PTX in

the release medium was extracted with ethyl acetate and analyzed as described in Section

2.2.4. At the completion of this study, dialysis cassettes were rinsed with PBS to collect

the remaining PTX. PTX leftover was analyzed in the same way as release samples.

To determine how quickly PTX precipitated in PBS at 37 °C, PTX solution in

PBS at a concentration of 20 µg/mL was prepared by diluting 1 mg/mL PTX stock

solution in Tween/PBS with PBS (Tween80 concentration in the diluted solution was

below the critical micelle concentration 0.0015%), aliquoted by 1 mL, and incubated at

37°C with shaking. At predetermined time points, 3 aliquots were taken and centrifuged

at 2095 rcf for 5 min. The supernatants were additionally centrifuged at 9300 rcf for 20

min and filtered with 0.45 µm PVDF syringe filters and directly analyzed with HPLC.

Dissolution kinetics of PPT and PNC was also tested in PBS containing 0.2%

Tween 80 (PBST) following a method based on light scattering [28]. PPT suspension was

prepared by diluting 30 mg/mL PTX in DMSO in PBS by 100 times. PNC suspension (1

mg/mL) was pre-diluted in 0.2% PBST by 10 times. Final dilution was performed

directly in a cuvette by adding 20 µL of pre-diluted PNC (equivalent to 2 µg of PTX) or

6.7 or 20 µL of PPT suspension (equivalent to 2 or 6 µg of PTX) to 1 mL of 0.2% PBST.

Their derived count rates were measured with a Zetasizer Nano-ZS90 from the time of

final dilution. Each measurement was done for 3 runs at 3s with a 30-sec interval and an

attenuator setting of 11. All measurements were performed at 25 °C.

52

2.2.9 PTX Release Kinetics of HA Gel Formulations

PTX release from gel formulations was studied in two ways: one in the sink

condition (total drug concentration being less than one third of the saturation solubility

[29]) and the other in a condition violating the sink condition to simulate the peritoneal

environment which has limited fluid available for drug dissolution. PNC-gel and PPT-gel

referred to PNC loaded in HA gel and PPT loaded in HA gel, respectively.

2.2.9.1 In Tween/PBS under Sink Condition

For the release kinetics study in the sink condition, 0.1 mL of PNC-gel or PPT-gel

containing 22 µg PTX was formed as described above and placed in a tube containing 20

mL of Tween/PBS (n=3). This condition made the initial PTX concentration in the

release medium 1.1 µg/mL, one third of the PTX solubility in Tween/PBS (3.3 µg/mL)

[30]. Thirty six identical samples were incubated at 37 °C on an orbital shaker. At

predetermined time points, 3 samples per group were taken and centrifuged at 2095 rcf or

10 min to separate the gels. The collected gels were freeze dried and incubated with 5 mL

mixture of acetonitrile/water (50/50) (ACN/water) at room temperature for 24 h. The

swollen gels were ground with a mortar and a pestle to release PTX into ACN/water. The

crushed gel suspension was filtered and analyzed with HPLC to determine the PTX

content.

53

2.2.9.2 In Tween/PBS under Non-Sink Condition

For a non-sink condition, 0.3 mL of PNC-gel or PPT-gel containing 200 µg PTX

was placed in a tube containing 20 mL of Tween/PBS (n=3). The initial PTX

concentration in the release medium was therefore 10 µg/mL, 3 times higher than the

PTX solubility in Tween/PBS [30]. All the samples were incubated at 37°C on an orbital

shaker. At predetermined time points, 3 mL release medium were sampled and

centrifuged at 9300 rcf for 10 min to separate a supernatant. Two milliliters of the

supernatant was taken for HPLC analysis, and the remaining 1 mL was combined with 2

mL of fresh Tween/PBS and returned to the sample tube for further incubation. At

completion of the study, the remaining mass in the sample container was freeze-dried and

reconstituted with 10 mL of acetonitrile to determine the PTX leftover.

PTX release from HA-EtCA0.1-CMC gel was also tested in Tween/PBS under

non-sink condition with minor differences. HA-EtCA0.1-CMC gel containing PPT was

formed as described in Section 2.2.5 by replacing 37.5% (w/w) of HA-CHO with

EtCA0.1-CMC-CHO. 0.3 mL of PPT-(HA-EtCA0.1-CMC) gel containing 200 µg of PPT

was placed in 20 mL of Tween/PBS. All the samples were incubated at 37 °C on an

orbital shaker. At predetermined time points, 5 mL release medium were sampled and

centrifuged at 2095 rcf for 10 min to separate a supernatant. Two milliliters of the

supernatant was taken for HPLC analysis, and the remaining 3 mL was combined with 2

mL of fresh Tween/PBS and returned to the sample container for further incubation. At

completion of this study, the remaining mass in the sample container was freeze-dried

and reconstituted with 10 mL of acetonitrile to determine the PTX leftover.

54

2.2.9.3 In FBS/PBS or Tween/PBS containing Hyaluronidase under Non-Sink

Condition

Hyaluronidase, a prevalent enzyme in the body, was added in the release medium

to simulate the degradation of HA gel in the peritoneal cavity. 0.3 mL of PNC-gel or

PPT-gel containing 200 µg PTX was placed in 20 mL of 25 v/v% FBS (25-FBS/PBS) or

Tween/PBS containing hyaluronidase (10 U/mL). The initial PTX concentration in the

release medium was therefore 10 µg/mL. All the samples were incubated at 37°C on an

orbital shaker. At predetermined time points, 5 mL release medium were sampled and

centrifuged at 2095 rcf for 10 min to separate a supernatant. Two milliliters of the

supernatant was taken for HPLC analysis, and the remaining 3 mL was combined with 2

mL of fresh 25-FBS/PBS and returned to the sample container for further incubation. At

the completion of the study, the remaining mass in the sample container was freeze-dried

and reconstituted with 10 mL of acetonitrile to determine the PTX leftover.

2.2.10 HPLC Analysis of PTX

PTX in PBS or PTX in Tween/PBS solution was directly analyzed with HPLC

after filtration with 0.45 µm PVDF syringe filter. PTX in PBS containing 50 or 25 v/v%

FBS was extracted with ethyl acetate prior to HPLC analysis. Briefly, 1 mL of PTX

solution in FBS medium with 10 µg of carbamazepine as an internal standard was mixed

with 3 mL ethyl acetate and shaken on a rotating shaker for 40 min. The mixture was then

centrifuged at 2095 rcf for 15 min to separate an organic layer, which was transferred to a

new glass vial and dried under vacuum. The dried sample was resuspended in the HPLC

55

mobile phase, filtered through 0.45 µm syringe filter, and analyzed by HPLC. A

calibration curve was drawn with PTX solutions in FBS medium in known

concentrations, treated in the same manner as the sample solutions. PTX was analyzed

with HPLC equipped with UV detector (1100 series, Agilent Technologies, Palo Alto,

CA) and an Ascentis C18 column (25 cm × 4.6 mm, particle size 5 µm) (Supelco, St.

Louis, MO, USA). The mobile phase was a mixture of acetonitrile and water (50:50) run

in the isocratic mode at a flow rate of 1 mL/min. PTX was detected at 227 nm.

2.3 Results and Discussions

2.3.1 Characterization of PNC and PPT

The properties of PNC and PPT are summarized in Table 1. PNC showed a rod-

shape with a length of 310 ± 86 nm and a width of 65 ± 10 nm under SEM (Fig. 6A). The

average diameter of PNC measured by Dynamic Light Scattering (DLS) was 258.0 ±

28.1 nm (average and standard deviation of 8 independently prepared batches) with

polydispersity index values ranging from 0.045 to 0.223, which indicated mid-range

polydispersity. On the contrary, PPT were needle-shaped crystals with a length of 11.5 ±

2.2 µm and a width of 2.0 ± 0.4 µm (Fig. 6B). Polarized light microscopy detected

clusters of PTX crystals, confirming the broad size distribution of PPT. Both PNC and

PPT showed weakly negative charges (PNC: -5.51 ± 0.42 mV; PPT: -3.08 ± 0.98 mV) at

pH 7.4. PNC and PPT exhibited sharp peaks typical of crystalline solids (Fig. 6C). The

crystal pattern of PNC was consistent with the result in the literature [13].

56

Figure 6. (A) Scanning electron micrograph of PNC. (B) Polarized light micrograph of PPT. (C) XPRD pattern of PNC and PPT.

Table 1. Summary of PNC and PPT properties PNC PPT Particle size Length1: 310 ± 86 nm;

Width1: 65 ± 10 nm z-average2: 258 ± 28.1 nm

Length3: 11.5 ± 2.2 µm; Width3: 2.0 ± 0.4 µm

Zeta potential4 -5.51 ± 0.42 mV -3.08 ± 0.98 mV 1Estimated by SEM; based on 50 measurements with ImageJ 2Measured by a zeta sizer (DLS); average and standard deviation of 8 independently prepared batches 3Estimated by light microscopy; based on 50 measurements with ImageJ 4Measured by a zeta sizer; average and standard deviation of 5 independently prepared batches

57

2.3.2 Synthesis and Characterization of EtCA-CMC-CHO

In an attempt to increase compatibility between PNC and the hydrogel carrier, 5β-

cholanic acid was introduced to one of the gel precursors. 5β-cholanic acid has been

conjugated to HA as EtCA to make an amphiphilic HA conjugate that formed self-

assembly nanoparticles for cancer therapy in the literature [31]. The EtCA synthesis was

adapted to produce EtCA-CMC-CHO. The chemical structure of EtCA was confirmed

with 1H NMR as shown in Fig. 7. The characteristic peaks of EtCA appeared from the

ethylene group (δ = 3.3 ppm [2H, –NHCH2–], δ = 2.7 ppm [2H,–CH2NH2]). EtCA0.1 or 0.3-

CMC-CHO was poorly soluble in water, which confirmed the conjugation of EtCA to

CMC-CHO. However, due to the low solubility in D2O and CD3OD, the chemical

structure of EtCA0.1 or 0.3-CMC-CHO was not successfully characterized with 1H NMR.

Figure 7. 1H NMR spectrum of EtCA (300 MHz, CD3OD). δ=3.30 (m, 2H), 2.86 (m, 2H), 0.97-0.94 (m, 6H), 0.68 (s, 3H).

58

2.3.2.1 Hydrophobicity of Gel Precursors

Contact angles of water droplet on the thin films of different gel precursors were

measured to compare their hydrophobicity. The contact angles of EtCA0.1-CMC-CHO

and EtCA0.3-CMC-CHO were similar but much higher than those of HA-CHO or CMC-

CHO, indicating the hydrophobicity imparted by EtCA (Table 2). EtCA0.1-CMC-CHO

was chosen for further studies and used as a partial replacement of HA-CHO, because it

was easier to dissolve in water than EtCA0.3-CMC-CHO.

Table 2. Contact angles of water droplet on gel precursor films. n=3 HA-CHO CMC-CHO EtCA0.1-CMC-

CHO EtCA0.3-CMC-CHO

Contact angle ( ) 33.0±2.0 38.8±1.4 63.2±7.3 67.2±2.7

2.3.2.2 Toxicity of EtCA and EtCA-CMC-CHO Conjugate

Quite unexpectedly, EtCA showed high toxicity on NIH/3T3 cells at all tested

concentrations (2-71 µg/mL) (Fig. 8A). CA or MeCA, precursors of EtCA, did not show

comparable toxicity, which indicated that the toxicity came from ethylene diamine

conjugation. Accordingly, EtCA0.1-CMC-CHO was toxic to NIH/3T3 cells at

concentrations ≥ 100µg/mL (equivalent to EtCA 9 µg/mL or above with a theoretical

EtCA content in EtCA0.1-CMC-CHO of 9.5%) (Fig. 8B and 8C). Toxicity of EtCA0.1-

CMC-CHO was further evaluated after gel formation. HA-EtCA0.1-CMC gel was placed

on the apical side of the insert and immersed in the culture medium, letting degradants or

molecules dissociated from the gel approach the cells through the pores of the membrane

insert. After 2 days incubation, cells treated with HA-EtCA0.1-CMC gel (equivalent to

67.5 µg/mL EtCA) were all dead (Table 3). CMC-CHO showed no toxicity on NIH/3T3

cells, and cells treated with HA-CMC gel maintained about 72% cell viability. Due to the

59

significant toxicity, EtCA0.1-CMC-HA gel containing 1.7% w/w of EtCA was not further

pursued in animal studies. Nevertheless, in-vitro release kinetics from PNC-loaded

EtCA0.1-CMC-HA gel was studied to examine the effect of hydrophobic moiety in the gel

on controlling the PTX release.

Figure 8. Toxicity of EtCA and EtCA0.1-CMC-CHO.

Table 3. Toxicity of gel precursors and gels with NIH/3T3 cells Samples CMC-CHO CMC-HA gel EtCA0.1-CMC-

HA gel Control

Sample preparation*

1.5 mg CMC-CHO in 0.2 mL PBS

0.2 mL gel consisting of 4 mg HA-ADH, 2.5 mg HA-CHO and 1.5 mg CMC-CHO

0.2 mL gel consisted of 4mg HA-ADH, 2.5 mg HA-CHO and 1.5 mg EtCA0.1-CMC-CHO

0.2 mL PBS

Cell viability% # 106.4 ± 4.9 71.6 ± 3.2 0 100% * Each sample (0.2 mL total) was placed in the apical side of a Transwell and supplemented with 0.3 mL DMEM prior to placement in the 12-well plate containing 1.5 mL medium and 17,500 cells. #: cell viability expressed as average ± standard deviation after treated with 3 independently and identically prepared gels.

25 50 100

250

500

750

A B C

60

2.3.3 PTX Solubility and Stability in Various Media

2.3.3.1 PTX Solubility in PBS, 50-FBS/PBS, or Tween/PBS

Prior to testing PNC dissolution kinetics and PTX release kinetics from gel

formulations, PTX solubility was evaluated in PBS, Tween/PBS, and 50-FBS/PBS after

incubation at 37 °C for 7 or 24h. PTX solubility in PBS was measured at ~0.2 µg/mL

with no specific trend according to the incubation time, although the values were variable

due to the sensitivity limitation of HPLC (Fig. 9A). PTX solubility in Tween/PBS was

measured to be 3.3 µg/mL irrespective of the incubation time (Fig. 9B). PTX solubility in

50-FBS/PBS was measured at 35 µg/mL after 7h incubation at 37 °C, much higher than

those in PBS or Tween/PBS, which confirmed the solubilizing effect of serum proteins

(Fig. 9C). Notably, PTX concentration measured after 24h incubation was 25 µg/mL,

28.6% lower than that after 7h. This difference is attributable to the instability of PTX in

serum, reported in our previous study [5] as well as in others. 38.5% of paclitaxel was

hydrolyzed to 7-epitaxol in cell culture medium containing FBS within 10h [32] and

35 % of paclitaxel would degrade in 24h at 37 °C in human plasma [33]. This indicates

that, if PTX release kinetics studies are performed in serum-containing medium and the

medium is not sampled and analyzed frequently, one may not recover 100% of PTX from

the formulation due to the degradation of released PTX. On the other hand, PTX was

relatively more stable in Tween/PBS as compared to 50-FBS/PBS, maintaining 94% of

the initial concentration for 1 day (Fig. 9D). This means that, as long as the medium is

sampled at least once a day, PTX stability in Tween/PBS is less likely to be a problem.

61

Figure 9. PTX solubility in (A) PBS, (B) Tween/PBS, or (C) 50-FBS/PBS. Blue square: measured after 7h; red triangle: measured after 24h. (D) Stability of 1.5 µg/mL PTX in Tween/PBS at 37 °C.

2.3.3.2 PNC Stability in PBS Containing Serum

PTX stability in serum-containing medium improved when it was incubated as

solid particles. PTX was least stable as Taxol and more stable as solid (PNC and PPT) in

25-FBS/PBS (Fig. 10). Between PNC and PPT, the recovery of PNC was relatively low,

likely due to the faster initial dissolution of PNC due to the small size. The improved

stability justifies the use of solid crystal forms for a long-term delivery of PTX.

A B

C D

62

Figure 10. Stability of PTX formulations (0.1 mg/mL) in 25-FBS/PBS for 10 days (n=6). *: p<0.05.

2.3.4 Dissolution Kinetics of PNC and PPT

2.3.4.1 Dissolution Kinetics of PNC and PPT with Dialysis Method

PTX dissolution kinetics from PNC or PPT was evaluated using the dialysis

method and PBS as a release medium. Here, the PNC equivalent to 200 µg PTX was

suspended in 3 mL of PBS, put in a dialysis cassette, and incubated in 200 mL of PBS.

Fig. 11A shows that PTX dissolution from the PNC was very slow, reaching less than

10% cumulative release in 10 days. Given that the PTX concentration in the dialysis

cassette was 66.7 µg/mL, far exceeding the solubility (0.2 µg/mL), and the dialysis

membrane could delay PTX diffusion into the release medium, we suspected that PTX

might have precipitated in the dialysis cassette. Indeed a significant fraction of PTX

remained in the dialysis cassette after 10 days. To estimate how quickly PTX precipitated

in a dialysis cassette, we prepared a 20 µg/mL PTX solution in PBS (<1/3 of the initial

concentration in a dialysis cassette) and sampled the solution at different time points to

quantify the dissolved PTX. PTX rapidly precipitated out in less than 30 min, leaving

Taxol PPT NC0

50

100

150

% P

TX

rec

ov

ery

**

*

P

63

PTX in solution only to the solubility level (Fig. 11B). This result suggests that even

though the small size of PNC had increased the dissolution rate of PTX, the dissolved

PTX might have undergone reprecipitation in the dialysis cassette. In other words, PTX

detected in the release medium did not necessarily reflect the dissolution of PNC but that

of PTX precipitates entrapped in the cassette. Since only the dissolved PTX could pass

the membrane and became diluted in the release medium, the PTX concentration in the

release medium was <0.1 µg/mL, much below the solubility limit, at any time point (Fig.

11C).

Drug dissolution or release kinetics of a particulate formulation is typically

studied by separating dissolution medium from the formulation by high speed

centrifugation or a dialysis bag and measuring the drug concentration in the sampled

medium. Neither is appropriate for estimating PTX dissolution from PNC or PPT:

centrifugation does not completely separate small particles from the suspension and

accelerates particle aggregation, and the dialysis method bears the risk of underestimating

drug release/dissolution due to drug adsorption to the membrane and/or reprecipitation of

dissolved drug in the bag [30].

Given these challenges, we resorted to an in-situ analytical technique based on

light scattering [28]. As expected, the derived count rate of PNC decreased from the

initial value of 214.3 kcps to 22.0 kcps (count rate of PBST) in 10 min, indicating rapid

dissolution of PNC in PBST. On the other hand, PPT showed an average count rate of

23.0 kcps from the beginning and did not change for 4 min, likely due to the small

number of PPT particles per volume and/or their instantaneous sedimentation. The count

rate was not much different (26 kcps) even when the concentration of PPT suspension

64

was tripled (6 µg/mL, above the PTX solubility in 0.2% PBST) (Fig. 11D). This indicates

that light scattering method was not suitable for large particles.

Figure 11. (A) Dissolution of PNC in PBS. (B) Kinetics of PTX precipitation in PBS. (C) PTX concentration in the sampled medium at each time point. Symbols indicate each replicate. (D) Derived count rate of PNC and PPT suspended in 0.2% PBST.

2.3.4.2 Limitations of Current Dissolution Kinetics Study Methods and Potential

Alternatives

Our release kinetics demonstrated a critical limitation of dialysis as a sampling

method in testing dissolution kinetics of particle formulations of PTX. Dialysis method is

often preferred to centrifugation because centrifugation makes it difficult to resuspend

nanoparticles for further incubation. However, the dialysis membrane itself could delay

A B

C D

65

the drug diffusion and involve drug adsorption, especially for poorly water-soluble drugs

like PTX, causing underestimation of dissolution kinetics.

Given these challenges, it is worthwhile to note several in-situ analytical

techniques developed to measure the dissolved drug directly and non-invasively. An

ultraviolet/visible (UV/Vis) fiber optic probe is a good example of real-time method in

dissolution test [34-36]. However, this technique is only applicable when excipients do

not interfere with analysis. Moreover, the utility of UV/Vis probe in the release kinetics

study of nanoparticle is still limited because of the light absorbing potential of

nanoparticles [37]. Electrochemical analytical methods were also explored but their

application was restricted to electroactive drugs [38, 39]. A method based on the light

scattering of micro/nano particles was implemented in determination of the dissolution

rate of nanoparticles [40, 41]. A linear decrease of scattering intensity is expected if

particles dissolves in the release medium with time. As indicated by our results, this

approach was not suitable for large particles with inadequate count rates and high

tendency of sedimentation. An alternative method could be using hydrogel to contain

drug particles, thereby separating them from receptor medium [42].

2.3.5 PTX Release Kinetics from HA Gel Formulations

Due to the limitations of the dialysis method mentioned above, gel formulations

were directly exposed to release media, and the released PTX was sampled by

centrifugation.

66

2.3.5.1 In Tween/PBS under Sink Condition

PNC and PPT were loaded in the HA gel, and the PTX release was tested in-vitro

using Tween/PBS as release medium. We first attempted to compare the dissolution rates

under a sink condition as defined by the United States Pharmacopeia (the volume of

medium at least three times that required to form a saturated solution of a drug [29]).

Since the highest PTX concentration (1.1 µg/mL) in the release medium fell far below the

limit of quantitation (3.8 µg/mL) which was calculated per the ICH guideline, the

released amount was indirectly determined by measuring the PTX amount remaining in

the gel at each time point and deducting it from the initial PTX amount. With this

method, up to 80% of PTX was found to be released from the gels in 7 days, with no

apparent difference between the two formulations (Fig. 12A).

2.3.5.2 In Tween/PBS under Non-Sink Condition

We next tested the dissolution rate in a non-sink condition to reflect the limited

fluid volume in the peritoneal cavity. The volume of peritoneal fluid in a healthy adult is

approximately 50 mL with a protein content 75% lower than that of the blood [44, 45].

The turnover rate of peritoneal fluid is 4-5 mL/h [44]. Although malignancies may

increase the protein level and accelerate the volume and the turnover rate of the ascites,

the small volume of the peritoneal fluid (compared to blood) is likely to challenge the

sink condition assumption. Therefore, we used 20 mL of release medium (Tween/PBS)

for PPT-gel and PNC-gel equivalent to 200 µg of PTX to create a release condition that

intentionally violated a sink condition. The release medium was directly analyzed at each

time point to determine the cumulative drug release. Under this non-sink condition,

67

32.6% and 30.6% of the total PTX were released from PPT-gel and PNC-gel,

respectively, in 12 days (Fig. 12B). 66.0% (PPT-gel) and 59.7% of PTX (PNC-gel) were

recovered in the remaining gel (Fig. 12C). The relatively slow release (compared to the

sink condition) suggests that the released PTX might have undergone reprecipitation in

the medium as the PTX concentration reached the saturation solubility. We expected that

the two gels would show different release kinetics at least initially but did not observe

any difference, most likely due to rapid reprecipitation that might have offset the effect of

particle size difference.

Figure 12. (A) In-vitro PTX release kinetics from HA-gel under sink condition (initial PTX concentration=1.1 µg/mL). (B) In-vitro PTX release kinetics from HA-gel under non-sink condition (initial PTX concentration=10 µg/mL). (C) Total PTX recovery from Tween/PBS medium. Data are expressed as averages and standard deviations of three independently and identically prepared samples.

The lack of difference in in-vitro release kinetics between the two gels may be

attributable to several reasons. First, both methods may involve the centrifugation-

induced artifacts. In the sink condition method, it is possible that the remaining PTX at

each time point might have been underestimated (i.e., drug release overestimated) due to

A B C

68

the centrifugation, which would have pressurized the gel and caused artificial release of

PPT and PNC loosely associated with the gel. In the non-sink method, the centrifugation

force applied to separate the released PTX from the gels might have contributed to

underestimation by accelerating the aggregation of reprecipitates. Second, the

invasiveness of Tween80 might also have masked the potential difference between two

gels. In this regard, it is worthwhile to reconsider the utility of in-vitro release kinetics

studies in predicting in-vivo outcomes. The release kinetics is largely affected by the

sampling method, concentration gradient between the formulation and release medium

(i.e., whether it satisfies the sink condition or not) and the choice of release medium [30].

However, the sampling method involves the aforementioned artifacts, and the sink

condition assumption is not necessarily applicable to drug release in the peritoneal cavity

due to the limited fluid volume and delay in systemic absorption of the dissolved drug.

Tween/PBS is widely used as a dissolution/release medium for poorly water-soluble drug

formulations to ensure the solubility [46-51]; however, its physiological relevance is not

clearly established, and the active role of Tween80 in drug release is seldom considered

in data interpretation.

2.3.5.3 In FBS/PBS or Tween/PBS containing Hyaluronidase under Non-Sink

Condition

In-vitro PTX release kinetics was studied with a medium simulating the peritoneal

cavity environment, which contains amphiphilic solutes (plasma proteins or Tween80)

and hyaluronidase: 25-FBS/PBS with 10 U/mL HAase or Tween/PBS with 10 U/mL

HAase. We were curious if HAse decelerated PTX release by degrading the gels and

69

facilitating the particle aggregation. The total PTX concentration in the media was kept at

10 µg/mL, above the PTX solubility in Tween/PBS (3.3 µg/mL). PTX solubility in 25-

FBS/PBS was not measured, but given that PTX solubility in 50-FBS/PBS was 35

µg/mL, so PTX solubility in 25-FBS/PBS was likely to be lower than 35 µg/mL. Thus,

both tests were done under a non-sink condition.

HA gel gradually degraded in 6 days in the presence of HAase. PPT-gel and

PNC-gel continuously released 35.2% and 29.5% over 10 days in 25-FBS/PBS,

respectively, similar to those tested without HAse (Fig. 13A). This indicates that the PTX

reprecipitation dominated the potential effect of HAse. The PTX recovery from PNC or

PPT-gel was much lower (< 60%) than that in Tween/PBS, probably due to the poor

stability of PTX in serum containing medium and inadequate sampling frequency after

the first day of the release kinetics study (Fig. 13B).

Similarly, PPT-gel and PNC-gel released 32.7% and 29.6% over 10 days in

Tween/PBS, respectively (Fig. 13C). Total PTX recovery from PNC or PPT-gel (~70%)

was higher than that in 25-FBS/PBS reflecting the stability profile (Fig. 13D). On the

other hand, this was lower than that in Tween/PBS without HAse (above 90%, Fig. 12C).

This difference is not due to the effect of HAse but more likely due to the sampling

condition. Here, the samples were centrifuged at 2095 rcf (as opposed to 9300 rcf in

section 1.2.9.2), which might have caused incomplete separation of supernatant and

subsequent loss of samples during each replacement with fresh medium.

70

Figure 13. (A) In-vitro PTX release kinetics performed in 25-FBS/PBS with 10U/mL HAase. (B) Total PTX recovery from 25-FBS/PBS medium (n=3). (C) In-vitro PTX release kinetics performed in Tween/PBS with 10U/mL HAase. (D) Total PTX recovery from Tween/PBS medium (n=3).

2.3.6 PTX Release Kinetics from HA-EtCA-CMC Gel

PTX release from HA-EtCA0.1-CMC gel was tested under non-sink condition.

Although reprecipitation of PTX is likely to have played a dominant role in PTX release

kinetics from the gel formulations in the non-sink condition, we speculated that the

segregation of PNC within the gel might further decelerate drug release during

incubation. We hypothesized that the addition of hydrophobic moieties like 5β-cholanic

PPT-gel

PNC-g

el

PPT-gel

PNC-g

el

A B

C D

71

acid would create domains for PNC to dwell in and prevent segregation of PNC in the gel

during the delivery period. As expected, HA- EtCA0.1-CMC gel increased to 31.8%, as

compared to 19.5% from HA gel in 10 days (Fig. 14A). Total PTX recovery was also

improved from 30.5% in HA gel to 45.2% in EtCA0.1-CMC-HA gel (Fig. 14B). This

result suggested that PTX release was facilitated by incorporating hydrophobic domains

to the gel.

Figure 14. (A) In-vitro PTX release from PNC containing HA gel or EtCA0.1-CMC-HA gel. (B) Total PTX recovery from Tween/PBS medium (n=3).

2.4 Conclusions

We have developed a nanoparticle depot for IP chemotherapy consisted of PNC

and HA gel. PNC with a size of ~310 nm was produced by nonsolvent and temperature

induced crystallization. To improve compatibility between hydrophobic PNC and

0.1

A B

72

hydrophilic HA gel, a hydrophobic moiety, EtCA, was conjugated on CMC-CHO and

employed as a partial replacement of HA-CHO. Dissolution kinetics of PNC could not be

determined by the dialysis method due to drug reprecipitation caused by diffusion barrier,

while fast dissolution of PNC was observed with light scattering method. PTX release

profiles from PNC-gel and PPT-gel were estimated in both sink- and non-sink conditions,

where the latter simulated the peritoneal environment. In-vitro release kinetics studies did

not reveal any difference between PNC-gel and PPT-gel, partly due to the centrifugation-

related artifacts. Despite the technical limitations, in-vitro release kinetics studies

detected enhancement of PTX release due to the inclusion of EtCA in the gel, indicating

that hydrophobic domain in the gel helped prevent segregation of PNC. However, due to

the unexpected toxicity of EtCA-CMC-CHO, EtCA-CMC-HA gel was not further

pursued in subsequent studies.

73

2.5 References

1. Markman, M., et al., Intraperitoneal Chemotherapy of Ovarian Cancer: A

Review, With a Focus on Practical Aspects of Treatment. J Clin Oncol, 2006.

24(6): p. 988-994.

2. Mohamed, F., et al., Pharmacokinetics and tissue distribution of intraperitoneal

paclitaxel with different carrier solutions. Cancer Chemother Pharmacol, 2003.

52(5): p. 405-410.

3. Mohamed, F., et al., Pharmacokinetics and tissue distribution of intraperitoneal

docetaxel with different carrier solutions. J Surg Res, 2003. 113(1): p. 114-120.

4. Tsai, M., et al., Effects of Carrier on Disposition and Antitumor Activity of

Intraperitoneal Paclitaxel. Pharm Res, 2007. 24(9): p. 1691-1701.

5. Bajaj, G., et al., Hyaluronic acid-based hydrogel for regional delivery of

paclitaxel to intraperitoneal tumors. J Control Release, 2012. 158(3): p. 386-392.

6. Armstrong, D.K., et al., Intraperitoneal Cisplatin and Paclitaxel in Ovarian

Cancer. New Eng J Med, 2006. 354(1): p. 34-43.

7. Hofstra, L.S., et al., Kinetic Modeling and Efficacy of Intraperitoneal Paclitaxel

Combined with Intravenous Cyclophosphamide and Carboplatin as First-Line

Treatment in Ovarian Cancer. Gynecol Oncol, 2002. 85(3): p. 517-523.

8. Chambers, S.K., et al., Phase I Trial of Intraperitoneal Pemetrexed, Cisplatin,

and Paclitaxel in Optimally Debulked Ovarian Cancer. Clin Cancer Res, 2012.

18(9): p. 2668-2678.

74

9. de Bree, E., et al., Intraperitoneal chemotherapy with taxanes for ovarian cancer

with peritoneal dissemination. Eur J Surg Oncol, 2006. 32(6): p. 666-670.

10. Sun, B., et al., Nanocrystals for the parenteral delivery of poorly water-soluble

drugs. Curr Opin Solid State Mater Sci, 2012. 16(6): p. 295-301.

11. Patravale, V.B., et al., Nanosuspensions: a promising drug delivery strategy. J

Pharm Pharmacol, 2004. 56(7): p. 827-840.

12. Müller, R.H., et al., State of the art of nanocrystals – Special features, production,

nanotoxicology aspects and intracellular delivery. Eur J Pharm and Biopharm,

2011. 78(1): p. 1-9.

13. Zhao, R., et al., Hybrid Nanocrystals: Achieving Concurrent Therapeutic and

Bioimaging Functionalities toward Solid Tumors. Mol Pharm, 2011. 8(5): p.

1985-1991.

14. Marchettini, P., et al., Docetaxel: pharmacokinetics and tissue levels after

intraperitoneal and intravenous administration in a rat model. Cancer Chemother

Pharmacol, 2002. 49(6): p. 499-503.

15. Nemes, K.B., et al., Oral, intraperitoneal and intravenous pharmacokinetics of

deramciclane and its N-desmethyl metabolite in the rat. J Pharm Pharmacol,

2000. 52(1): p. 47-51.

16. Krasner, C.N., et al., Case 11-2006. New Eng J Med, 2006. 354(15): p. 1615-

1625.

17. Zahedi, P., et al., Combination Drug Delivery Strategy for the Treatment of

Multidrug Resistant Ovarian Cancer. Mol Pharm, 2010. 8(1): p. 260-269.

75

18. De Souza, R., et al., Biocompatibility of injectable chitosan–phospholipid implant

systems. Biomaterials, 2009. 30(23–24): p. 3818-3824.

19. Zahedi, P., et al., Chitosan–phospholipid blend for sustained and localized

delivery of docetaxel to the peritoneal cavity. Int J Pharm, 2009. 377(1–2): p. 76-

84.

20. Yu, J., et al., The antitumor effect of a thermosensitive polymeric hydrogel

containing paclitaxel in a peritoneal carcinomatosis model. Invest New Drugs,

2012. 30(1): p. 1-7.

21. Wang, Y., et al., 5-FU-hydrogel inhibits colorectal peritoneal carcinomatosis and

tumor growth in mice. BMC Cancer, 2010. 10(402): p. 1471-2407.

22. Yeo, Y., et al., Prevention of peritoneal adhesions with an in situ cross-linkable

hyaluronan hydrogel delivering budesonide. J Control Release, 2007. 120(3): p.

178-185.

23. Yeo, Y., et al., Peritoneal adhesion prevention with an in situ cross-linkable

hyaluronan gel containing tissue-type plasminogen activator in a rabbit repeated-

injury model. Biomaterials, 2007. 28(25): p. 3704-3713.

24. Yeo, Y., et al., In situ cross-linkable hyaluronic acid hydrogels prevent post-

operative abdominal adhesions in a rabbit model. Biomaterials, 2006. 27(27): p.

4698-4705.

25. Yeo, Y., et al., In situ cross-linkable hyaluronan hydrogels containing polymeric

nanoparticles for preventing postsurgical adhesions. Ann Surg, 2007. 245(5): p.

819-824.

76

26. Bulpitt, P., et al New strategy for chemical modification of hyaluronic acid:

preparation of functionalized derivatives and their use in the formation of novel

biocompatible hydrogels. J Biomed Mater Res, 1999. 47(2): p. 152-169.

27. Yuan, Y., et al., Contact Angle and Wetting Properties, in Surface Science

Techniques, G. Bracco and B. Holst, Editors. 2013, Springer Berlin Heidelberg. p.

3-34.

28. Anhalt, K., et al., Development of a new method to assess nanocrystal dissolution

based on light scattering. Pharm Res, 2012. 29(10): p. 2887-2901.

29. The United States Pharmacopeia: The National Formulary (USP37/NF32).

General Chapters: <1092> The Dissolution Procedure: Development and

Validation2014, Rockville, MD: The United States Pharmacopeial Convention,

Inc.

30. Abouelmagd, S.A., et al., Release Kinetics Study of Poorly Water-Soluble Drugs

from Nanoparticles: Are We Doing It Right? Mol Pharm, 2015. 12(3): p. 997-

1003.

31. Choi, K.Y., et al., Self-assembled hyaluronic acid nanoparticles as a potential

drug carrier for cancer therapy: synthesis, characterization, and in vivo

biodistribution. J Mater Chem, 2009. 19(24): p. 4102-4107.

32. Ringel, I., et al., Taxol is converted to 7-epitaxol, a biologically active isomer, in

cell culture medium. J Pharmacol and Exp Ther, 1987. 242(2): p. 692-698.

33. Willey, T.A., et al., High-performance liquid chromatographic procedure for the

quantitative determination of paclitaxel (Taxol®) in human plasma. J Chromatogr

B, 1993. 621(2): p. 231-238.

77

34. Aldridge, P.K., et al., A robotic dissolution system with on-line fiber-optic UV

analysis. J Pharm Sci, 1995. 84(8): p. 909-914.

35. Chen, C.-S., et al., A Drug Dissolution Monitor Employing Multiple Fiber Optic

Probes and a UV/Visible Diode Array Spectrophotometer. Pharm Res, 1994.

11(7): p. 979-983.

36. Alonzo, D., et al., Understanding the Behavior of Amorphous Pharmaceutical

Systems during Dissolution. Pharm Res, 2010. 27(4): p. 608-618.

37. Van Eerdenbrugh, B., et al., Influence of Particle Size on the Ultraviolet Spectrum

of Particulate-Containing Solutions: Implications for In-Situ Concentration

Monitoring Using UV/Vis Fiber-Optic Probes. Pharm Res, 2011. 28(7): p. 1643-

1652.

38. Rosenblatt, K.M., et al., Drug release from differently structured

monoolein/poloxamer nanodispersions studied with differential pulse

polarography and ultrafiltration at low pressure. J Pharm Sci, 2007. 96(6): p.

1564-1575.

39. Charalampopoulos, N., et al., Differential pulse polarography: a suitable

technique for monitoring drug release from polymeric nanoparticle dispersions.

Anal Chim Acta, 2003. 491(1): p. 57-62.

40. Crisp, M.T., et al., Turbidimetric measurement and prediction of dissolution rates

of poorly soluble drug nanocrystals. J Control Release, 2007. 117(3): p. 351-359.

41. Anhalt, K., et al., Development of a New Method to Assess Nanocrystal

Dissolution Based on Light Scattering. Pharm Res, 2012. 29(10): p. 2887-2901.

78

42. Peschka, R., et al., A simple in vitro model to study the release kinetics of

liposome encapsulated material. J Control Release, 1998. 56(1-3): p. 41-51.

43. The United States Pharmacopeia: The National Formulary 574

(USP37/NF32); The United States Pharmacopeial Convention, Inc.: 575

Rockville, MD, 2014.

44. Sherwood, L., Human physiology : from cells to systems 2007, Australia;

Belmont, CA: Thomson/Brooks/Cole.

45. Watson, M.S., Oxford handbook of palliative care 2009, Oxford; New York:

Oxford University Press.

46. Kilfoyle, B.E., et al., Development of paclitaxel-TyroSpheres for topical skin

treatment. J Control Release, 2012. 163(1): p. 18-24.

47. Yang, T., et al., Enhanced solubility and stability of PEGylated liposomal

paclitaxel: In vitro and in vivo evaluation. Int J Pharm, 2007. 338(1–2): p. 317-

326.

48. Cha, E.-J., et al., Stabilized polymeric micelles by electrostatic interactions for

drug delivery system. Eur J Pharm Sci, 2009. 38(4): p. 341-346.

49. Gullotti, E., et al., Beyond the imaging: Limitations of cellular uptake study in the

evaluation of nanoparticles. J Control Release, 2012. 164(2): p. 170-176.

50. Amoozgar, Z., et al., Low Molecular-Weight Chitosan as a pH-Sensitive Stealth

Coating for Tumor-Specific Drug Delivery. Mol Pharm, 2012. 9(5): p. 1262-1270.

51. Gullotti, E., et al., Polydopamine-Based Surface Modification for the

Development of Peritumorally Activatable Nanoparticles. Pharm Res, 2013.

30(8): p. 1956-1967.

79

CHAPTER 3. BIOACTIVITY EVALUATION OF NANOPARTICLE DEPOT

3.1 Introduction

Although the difference in in-vitro release profile between PNC-gel and PPT-gel

was not identified, it is still possible that PNC-gel can demonstrate potential advantages

over PPT-gel in a more biologically relevant experimental setting or in-vivo because the

in-vitro release kinetics does not always correlate well with the in-vivo outcomes. In this

chapter, the half maximal inhibitory concentrations (IC50) of PTX in the form of PNC or

PPT were measured with different cell viability assays on SKOV3 human ovarian cancer

cells. Low IC50 value could be attributed to fast drug release as well as sensitivity of the

cells to the drug. In addition, Maximum tolerated doses (MTD) of PPT-gel and PNC-gel

were estimated using healthy mice to determine the PTX dose in in-vivo efficacy study

and observe the difference in drug release rate in-vivo. In anti-tumor efficacy study, PNC

was administered IP to nude mice bearing peritoneal ovarian tumor xenografts with the

HA gel as a carrier to retain the drug in the peritoneal cavity. Progression of tumor

burden after single treatment of PNC-gel was monitored over 10 weeks via non-invasive

whole body imaging, and the outcome was compared with that of PPT-gel. We expect

that PNC can show higher cytotoxicity than PPT in both in-vitro and in-vivo bioactivity

studies

80

3.2 Materials and Methods

3.2.1 Materials

Hyaluronic acid (HA, 20 kDa and 500 kDa) was purchased from Lifecore

Biomedical, LLC (Chaska, MN, USA). Paclitaxel (PTX) was a gift of Samyang Genex

Corp (Seoul, Korea). Carbamazepine was purchased from Enzo life sciences (Plymouth

Meeting, PA, USA). Cremophor ELP was a gift from BASF (New York, NY, USA). D-

Luciferin potassium salt was purchased from Gold Biotechnology (St. Louis, MO, USA).

Geneticin® selective antibiotic (G418 sulfate, 50 mg/mL) was purchased from Life

technologies (Grand Island, NY, USA). Cell culture medium and supplements were

purchased from Invitrogen (Carlsbad, CA, USA). Micro BCA Protein Assay Kit was

purchased from Life technologies (Grand Island, NY, USA). CellTiter-Glo® 2.0 Assay

Kit was purchased from Promega (Madison, WI, USA). All other reagents were

purchased from Sigma-Aldrich (St. Louis, MO, USA).

3.2.2 Determination of IC50 of Taxol, PPT and PNC on SKOV3 Cell Line

IC50 of PNC on SKOV3 cell line was determined with luminescent cell viability

assay (CellTiter-Glo® 2.0 Assay) which determines the number of viable cells by

quantitating the amount of ATP from metabolically active cells. SKOV3 human ovarian

cancer cells (ATCC) were maintained in RPMI-1640 medium supplemented with 10%

fetal bovine serum (FBS) and 1% penicillin-streptomycin. Cells were plated in a 96-well

plate at a density of 5,000 cells per well with 0.2 mL of complete medium. After 24h

81

incubation, Taxol, PPT or PNC suspended in 22 µL PBS was added to each well and the

final PTX concentration ranged from 0.05 to 100 nM. Control groups were treated with

22 µL PBS or Cremophor ELP in PBS at the same concentrations as Taxol treated group.

Cells were exposed to treatments for 24h and allowed to recover for 2 days after

replacing the treatments with fresh medium. One hundred microliters of CellTiter-Glo

Reagent was added to each well after removal of 100 µL of medium on the top. The plate

was placed on an orbital shaker for 2 minutes to induce cell lysis and incubated at room

temperature for 10 minutes to stabilize the luminescent signal. The luminescence of each

treatment group was measured with a SpectraMax M3 microplate reader (Molecular

Devices, Sunnyvale, CA, USA) and normalized to the signal of PBS control group.

3.2.3 Cellular Retention and Cytotoxicity of PTX

SKOV3 human ovarian cancer cells (ATCC) were maintained in RPMI-1640

medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-

streptomycin. Cells were plated in a 12-well plate at a density of 200,000 cells per well

with 0.8 mL of complete medium. After 24h incubation, PNC or PPT suspended in 88 µL

PBS was added to each well to make the final PTX concentration 6 µg/mL (7 µM). A

control group was treated with 88 µL PBS. Cells were incubated with the treatments for

3h and washed twice with 0.5 mL complete medium after removal of the treatments. The

treated cells were incubated for two days in fresh medium and evaluated with the MTT

(3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide) assay. The absorbance

of the solubilized formazan was measured with a SpectraMax M3 microplate reader

82

(Molecular Devices, Sunnyvale, CA, USA) at a wavelength of 562 nm. The measured

absorbance was normalized to the absorbance of the control group.

To determine PTX retained in the cells after each treatment, SKOV3 cells were

treated in the same way as described above. Immediately after removing treatments, 1

mL of sterile water was added to each well, frozen and thawed once to lyse the cells. The

cell lysate was analyzed with HPLC (see section 2.2.5) and Micro BCA assay to

determine the concentrations of PTX and protein, respectively.

PTX in the cell lysate was analyzed after ethyl acetate extraction. Briefly, 2 mL of

the cell lysate was spiked with 10 µg/mL carbamazepine as an internal standard, mixed

with 6 mL ethyl acetate, and agitated on a rotating shaker for 40 min. The mixture was

then centrifuged at 3724 rcf for 15 min to separate an organic layer, which was

transferred to a new glass vial and dried under vacuum. The dried sample was dissolved

in HPLC mobile phase, filtered through 0.45 µm syringe filter, and analyzed with HPLC.

A calibration curve was drawn with PTX in FBS medium in known concentrations,

treated in the same manner as the sample solutions. PTX was analyzed with HPLC

equipped with UV detector (1100 series, Agilent Technologies, Palo Alto, CA) and an

Ascentis C18 column (25 cm × 4.6 mm, particle size 5 µm) (Supelco, St. Louis, MO,

USA). The mobile phase was a mixture of acetonitrile and water (50:50), run in the

isocratic mode at a flow rate of 1 mL/min. PTX was detected at 227 nm.

In a separate experiment, SKOV3 cells were plated in a 12-well plate at a density

of 60,000 cells per well with 1.5 mL of complete medium. After 24h incubation, a

Transwell insert was suspended in each well as a container of PTX treatment. The

Transwell membrane was perforated with fifteen 21-gauge needle holes to facilitate the

83

transport of released PTX (either as free molecules or particles) to the underlying cell

layer. 0.1 mL of PNC-gel, PPT-gel, PNC, or PPT suspension was placed in each well

along with 0.4 mL PBS, making the final PTX concentration in the well 30 µg/mL (35

µM). A control group was treated with 0.1 mL of PBS. Cells were incubated with the

treatments for 3h, 24h or 48h, removed of the treatments, washed twice, and incubated

for additional two days in complete medium prior to the MTT assay. The solubility of

PTX in the culture medium containing 10% FBS was determined in the same method as

PBS containing 50 v/v% FBS described in Chapter 2.

3.2.4 Determination of the Maximum Tolerated Doses of Treatments

All animal procedures were approved by Purdue Animal Care and Use

Committee, in conformity with the NIH guidelines for the care and use of laboratory

animals. The MTD of each treatment was determined according to the method published

by the National Cancer Institute’s Developmental Therapeutics Program [1]. Healthy

female Balb/c wild-type mice (8-10 week old, ~20g, Harlan Laboratories, Indianapolis,

IN, USA) were randomly assigned to Taxol, PPT-gel, and PNC-gel groups and given a

single IP injection of each formulation at different dose levels (one mouse per dose). The

mice were observed over a period of 2 weeks after the injection. The highest dose

tolerated without >20% weight loss or other signs of significant toxicity was designated

as the MTD of each treatment. This experiment was repeated at least three times to

confirm the reproducibility.

84

3.2.5 In-vivo Efficacy Studies

A mouse model of IP tumor was prepared as described in our previous study [2].

Luciferase-expressing SKOV3 cells (SKOV3-luc, donated by Prof. Glen Kwon at

University of Wisconsin-Madison) [3] were maintained in a complete RPMI-1640

medium containing 500 µg/mL G418 sulfate. 107 cells were suspended in 1 mL RPMI-

1640 medium and IP injected to a female Balb/c nude mouse (8-10 week old, ~20g,

Harlan Laboratories). Tumor growth was monitored every week by measuring the

bioluminescence with the IVIS Lumina II whole body imaging system (Caliper Life

Science, Hopkinton, MA, USA) [4]. When the radiance of tumors reached 105-106

p/s/cm2/sr, which usually took one or two weeks, animals were evenly assigned to 5

treatment groups (n=3 per group per study): PBS, HA gel, Taxol, PNC-gel and PPT-gel

(equivalent to 30 mg/kg of PTX). One milliliter of each treatment was IP injected through

a catheter, and the skin was sealed with GLUture topical tissue adhesive (Abbott Park,

IL, USA). Bioluminescence of IP tumor was measured weekly over 10 weeks. Animals

were observed every 3 days for weight change and the signs of pain until they reached the

criteria of sacrifice, which include the loss of appetite and/or body weight, bloated

abdomen and signs of respiratory distress, according to the approved animal procedures.

3.2.6 Statistical Analysis

All in-vitro data were analyzed using GraphPad Prism 6 (La Jolla, CA), with

unpaired t test to determine the difference of means between two groups. The comparison

of survival curves was conducted with Log-rank (Mantel-Cox) test built in GraphPad

85

Prism 6. A value of p< 0.05 was considered statistically significant. For the tumor growth

study, analysis of variance (ANOVA) was performed using IBM SPSS Statistics 23

(Armonk, NY, USA) to test the differences between the treatment groups in the change of

tumor burden over time.

3.3 Results and Discussion

3.3.1 IC50 of PTX in the Form of Taxol, PPT or PNC

The growth profile of a certain cell line plays an important role in the

determination of IC50 of a treatment. PTX interferes with the normal depolymerization of

microtubules during cell proliferation [5, 6]. The population doubling time of SKOV3

cell line was approximately 35h, which means that SKOV3 cells need at least 1.5 days to

show their response to PTX. These indicate that it is necessary to leave SKOV3 cells in

fresh medium for at least 1.5 days to observe the cytotoxic effect of PTX. However, this

recovery time cannot be too long because challenged cells may become viable as before

if the treatment discontinues, resulting in underestimation of PTX cytotoxicity. On the

other hand, complete medium contains 10% FBS, which can solubilize PTX; thus,

relatively long incubation time of PNC or PPT in the culture medium may diminish the

potential difference in the release rate between PPT and PNC. Given these constraints,

cells were exposed to each treatment for only one day, a time frame speculated to be

short enough to observe differential PTX release from PPT and PNC, and incubated for 2

more days in treatment-free medium to develop responses to the treatments. This

experiment was repeated twice, and IC50 value was obtained with nonlinear regression

86

using GraphPad Prism 6. Although the apparent IC50 values increased in the order of

Taxol, PNC, and PPT, reflecting the expected difference in dissolution rate of PTX, they

did not show statistical difference, probably due to the variation of cell response to PTX

(Fig. 15). Therefore, alternative study was carried out to directly quantitate the amount of

PTX retained with cells.

Figure 15. IC50 of PTX on SKOV3 cell line in the form of Taxol, PPT and PNC (n=3).

3.3.2 Cellular Retention and Cytotoxicity of PPT and PNC

The cytotoxicity of PPT and PNC were compared as an indirect indicator of drug

dissolution profiles. SKOV3 cells were incubated with PPT and PNC for 3h, and the level

of PTX retained with cells were determined. Much less PTX was detected in the SKOV3

cells treated with PNC than those with PPT (Fig. 16A). The relatively high level of PTX

remaining in PPT-treated cells may be explained by the large size of PPT, which was not

readily removed from cells by simple washing. Surprisingly, despite the low level of PTX

retained with the cells, PNC showed higher cytotoxicity than PPT (Fig. 16B). The cell

IC5

0 o

f P

TX

(n

M)

Taxo

l

PP

T

PN

C

0

5

1 0

1 5

2 0

87

fraction affected by the unit amount of PTX was 3.3 times greater for PNC than PPT

(Fig. 16C), which indicates that PTX provided as PNC was much more efficient in

killing SKOV-3 cells than PPT. These results suggest that PNC made PTX better

available than PPT via the small size that facilitated the drug dissolution. In addition, the

small size may have helped PNC to enter cells. A recent study supports that drug

nanocrystals could be endocytosed as solid particles [7].

3.3.3 Cytotoxicity of PPT-gel and PNC-gel

Acknowledging aforementioned limitations of in-vitro release kinetics tests, we

measured the cytotoxicity of PPT-gel and PNC-gel varying the exposure time to predict

their in-vivo effects (Fig.16D). The gel was contained in a perforated Transwell insert to

avoid direct contact with the cell layer that might limit oxygen supply. Free particles

were supplied in the same manner to mimic a situation where a degrading gel was no

longer able to retain the particles. Therefore, the tests with gels and free particles

represented the initial and later phase of delivery, respectively. With 3h exposure, PNC-

gel was found to be more toxic than PPT-gel, indicating faster dissolution of PNC than

PPT. This difference disappeared upon longer incubation (24h and 48h), which may be

explained by the reprecipitation of released PTX exceeding the saturation solubility of

PTX in the culture medium (1.4 µg/mL), as predicted from the release kinetics results. In

contrast, free PNC showed consistently high cytotoxicity as compared to PPT, indicating

that at least part of PNC were endocytosed by the cells as solid particles before they

underwent dissolution and reprecipitation in the medium. This result suggests that PNC-

88

gel would achieve greater anti-tumor effect than PPT-gel in-vivo, as the endocytosis of

released PNC offset the effect of reprecipitation of dissolved PTX.

Figure 16. (A) PTX retention normalized with total protein content. Micro BCA assay

was performed to determine the total protein content in the cell lysate as an estimate of

cell population. The PTX to protein ratio reflects PTX retained by each cell. (B)

Cytotoxicity of PPT and PNC (equvilant to 7 µM PTX) to SKOV3 cells after 3h of direct

exposure followed by incubation in drug-free medium. (C) Fraction of cells affected by

unit amount of PTX. Data are expressed as averages and standard deviations of three

measurements of a representative batch. Cytotoxicity of (D) PNC-gel and PPT-gel and

(E) PPT and PNC (equivalent to 35 µM PTX) to SKOV3 cells after 3h, 24h or 48 h

exposure followed by incubation in drug-free medium. Data are expressed as averages

and standard deviations of three measurements of a representative batch. *: p<0.05 by t-

test.

PT

X (

g)/

pro

tein

(

g)

PP

T

PN

C

0 .0 0

0 .0 5

0 .1 0

0 .1 5

Ce

ll v

iab

ilit

y (

%)

PP

T

PN

C

0

2 0

4 0

6 0

De

ad

ce

lls

(%

) /

PT

X (

g)

PP

T

PN

C

0

2

4

6

8

A B C

**

*

3h

24h

48h

0

2 0

4 0

6 0

8 0

1 0 0

Ce

ll v

iab

ilit

y (

%)

P N C -g e l

P P T -g e l

3h

24h

48h

0

2 0

4 0

6 0

8 0

1 0 0

Ce

ll v

iab

ilit

y (

%)

P P T

P N C

D E

*

**

*

89

3.3.4 Maximum Tolerated Doses of PTX Treatments

MTDs of PNC-gel, PPT-gel, and Taxol were determined using healthy Balb/c

mice for two purposes: (i) to determine the maximum PTX dose to administer in the anti-

tumor efficacy study and (ii) to observe the difference in drug release rate in-vivo.

Specifically, we hypothesized that the gel more rapidly releasing PTX would have a

lower MTD value. Mice did not survive a single administration at dose higher than 60

mg/kg. On the other hand, PPT-gel and PNC-gel were tolerated much higher doses: 120

mg/kg and 90 mg/kg, respectively. This result first confirms the benefit of the gel

formulations free of Cremophor EL, which is associated with several adverse effects such

as severe anaphylactoid hypersensitivity reactions, abnormal lipoprotein pattern and

peripheral neuropathy [8]. Interestingly, PNC-gel showed lower MTD than PPT-gel,

despite the identical composition. The difference between PNC-gel and PPT-gel suggests

that PNC-gel might have released a greater amount of PTX than PPT-gel in the given

time. This result is consistent with the cellular PTX retention and cytotoxicity results.

3.3.5 Anti-tumor Effects of PPT-gel and PNC-gel

The anti-tumor efficacy of PPT- and PNC-gels was studied using a mouse model

of IP tumor. Once the tumor reached a certain size as indicated by the bioluminescence

signals, treatments (Taxol, PPT-gel and PNC-gel) equivalent to 30 mg/kg PTX as well as

vehicle controls were administered IP once. A single administration regimen was chosen

to compare the duration of the therapeutic effects of the treatments (Fig. 17A). The tumor

burden was monitored up to 14 weeks until the animals reach a humane endpoint (Fig.

90

17B). Animals treated with PBS or HA gel vehicle reached the endpoint in <7 weeks

with median survival periods of 37 days. All animals receiving PTX-treatments survived

longer than the vehicle control groups with notable difference according to the treatment.

Tumor growth in animals treated with Taxol was initially delayed but resumed after 3

weeks to reach the endpoint in <9 weeks with a median survival time of 56 days. This

result may be explained by the rapid clearance of PTX from the peritoneal cavity [2].

Animals treated with PPT-gel showed a similar survival curve (median survival time of

51 days) as Taxol-treated ones. On the other hand, mice treated with PNC-gel showed a

significant extension of the survival period (67 days), which clearly contrasted with

Taxol-treated groups (p<0.05: PNC-gel vs. Taxol, Log rank Mantel-Cox test) (Fig. 18A).

Both PPT- and PNC-gel-treated animals showed minimal increase in tumor signals in

surviving animals until 7 weeks post-treatment (Fig. 18B). However, it was difficult to

obtain meaningful statistical analysis from averages of the bioluminescence values, due

to the attrition of animals over the course of survival period and large variations in tumor

growth. Thus, the change of tumor burden (tumor burden at each time point-initial tumor

burden) was plotted with respect to the survival time for each mouse, and the area under

the curve over time (AUC/time) was calculated as an average tumor burden for the mouse

during the survival period. The mice treated with PNC-gel or PPT-gel showed the lowest

median AUC/time value among the treatment (Fig. 19). The AUC/time value of Taxol

group was not significantly different from PBS or HA gel control groups. Both PPT-gel

and PNC-gel showed significantly lower AUC/time values than Taxol group, but the

difference between PNC-gel and Taxol groups was more significant than that of PPT-gel

and Taxol groups (p: 0.0012 vs. 0.0306).

91

Figure 17. (A) Treatment schedule. (B) Representative whole body bioluminescence

imaging of animals administered with different treatments. The most representative

animal in each group is presented.

a

b

\

\

Weekly monitoring of tumor growth

0 1w 2w 3w 4w 5w 6-14w

Tumor injection

Single IP administration(PBS, HA gel, Taxol, PPT-gel, PNC-gel at 30 mg/kg)

-1 or 2w

B

A

92

Figure 18. (A) Kaplan-Meier analysis for survival time post tumor cells inoculation. n=9

per group. *: p<0.05, PNC-gel vs. Taxol by Log-rank Mantel-Cox test. (B)

Bioluminescence of IP tumors in animals treated with PBS, HA gel, Taxol, PPT-gel, and

PNC-gel. Data are expressed as median with ranges of surviving animals. The box

extends from the 25th to 75th percentiles, the line in the middle of the box indicates the

median, and the whiskers go down to the smallest value and up to the largest.

0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8

0

5 .01 0 6

1 .01 0 7

1 .51 0 7

2 .01 0 7

T im e (w e e k s )

Av

g R

ad

ian

ce

[p

/s/c

m²/

sr] P B S c o n tro l

H A g e l c o n tro l

T a x o l

P P T g e l

P N C g e l

c

d

0 5 0 1 0 0

0

5 0

1 0 0

T im e (d a y s )

Pe

rc

en

t s

urv

iva

l

P B S c o n tro l

H A g e l c o n tro l

T a x o l

P P T g e l

P N C g e l

*

Overall survival

Tumor radiance in surviving animalsB

A

93

Figure 19. AUC of tumor burden change over survival time of animals treated with PBS,

HA gel, Taxol, PNC-gel and PPT-gel. *: p<0.05, PPT-gel vs. Taxol, **: p<0.05, PNC-gel

vs. Taxol by Tukey’s test.

Taken together, the in-vivo studies demonstrated that a single dose PNC-gel was

significantly better in delaying tumor progression as compared to Taxol, while PPT-gel

was not different from Taxol in the median survival time. This result is consistent with

the in-vitro cellular toxicity test and MTD values, which indicated greater dissolution and

cellular uptake of PNC than PPT. Based on the depot effect of HA gel, PNC could have a

prolonged effect on IP tumors than Taxol. It remains to be investigated how the

prolonged local delivery improved the efficacy of PTX. A potential mechanism may

involve the tumor priming effect by initial PTX exposure, which suppresses stroma

expansion [9], thereby facilitating the transport of subsequently released PTX. A

remaining challenge is to improve the specificity of local delivery of PTX. The current

PNC-gel does not have a mechanism to distinguish tumors vs. normal tissues; thus, some

toxicity to adjacent normal tissues is likely unavoidable (although no abnormalities were

PB

S c

on

tro

l

HA

gel co

ntr

ol

Taxo

l

PP

T-g

el

PN

C-g

el

-4 .01 0 6

-2 .01 0 6

0

2 .01 0 6

4 .01 0 6

6 .01 0 6

AU

C/t

ime

AUC/time

* **

94

observed on macroscopic level in this study). This may be achieved by surface

modification of PNC with cell-specific ligands.

3.4 Conclusion

PTX-gel formulation comprising PNC and HA gel was developed for IP

chemotherapy of ovarian cancer. In cellular toxicity test and MTD assessment, PNC-gel

provided more efficient killing effect and greater toxicity than PPT-gel, which contained

larger PTX particles, indicating a greater dissolution rate of PNC due to the small size. A

single IP administration of PNC-gel extended the survival of mice with IP tumors

significantly better than the same dose Taxol, due to the local depot effect, whereas PPT-

gel was not superior to Taxol in survival extension. While the cell toxicity test and in-

vivo results consistently point to the beneficial effect of particle size reduction, in-vitro

drug release kinetics did not predict the difference between PPT- and PNC-gels,

suggesting the limitation of current release study methods.

95

3.5 References

1. Lengyel, E., et al., Expression of latent matrix metalloproteinase 9 (mmp-9)

predicts survival in advanced ovarian cancer, Gynecol. Oncol. 2001. 82(2): p.

291-298.

2. Bajaj, G., et al., Hyaluronic acid-based hydrogel for regional delivery of

paclitaxel to intraperitoneal tumors. J Control Release, 2012. 158(3): p. 386-392.

3. Cho, H., et al., Poly(ethylene glycol)-block-poly(ε-caprolactone) micelles for

combination drug delivery: Evaluation of paclitaxel, cyclopamine and gossypol in

intraperitoneal xenograft models of ovarian cancer. J Control Release, 2013.

166(1): p. 1-9.

4. Cho, E.J., et al., Intraperitoneal delivery of platinum with in-situ crosslinkable

hyaluronic acid gel for local therapy of ovarian cancer. Biomaterials, 2015. 37: p.

312-319.

5. Horwitz, S.B., Taxol (paclitaxel): mechanisms of action. Ann Oncol, 1994. 5(6):

p. S3-6.

6. http://physics.cancer.gov/docs/bioresource/ovary/NCI-PBCF-HTB77_SK-OV-

3_SOP-508.pdf.

7. Chen, Y. et al., Cellular Uptake Mechanism of Paclitaxel Nanocrystals

Determined by Confocal Imaging and Kinetic Measurement. AAPS J, 2015.

17(5): p. 1126-1134.

8. Gelderblom, H., et al., Cremophor EL: the drawbacks and advantages of vehicle

selection for drug formulation. Eur J Cancer, 2001. 37(13): p. 1590-1598.

96

9. Lu, Z., et al., Tumor-Penetrating Microparticles for Intraperitoneal Therapy of

Ovarian Cancer. J Pharmacol Exp Ther, 2008. 327(3): p. 673-682.

97

CHAPTER 4. ALBUMIN-STABILIZED PACLITAXEL NANOCRYSTALS

4.1 Literature Review3

Approximately 40% of active pharmaceutical ingredients in the discovery stage

have poor water-solubility [1]. In order to attain adequate bioavailability of poorly

soluble drugs, special formulation strategies are employed to increase their dissolution in

aqueous medium [2]. Traditionally, organic solvents are used as co-solvents [3] or a part

of emulsion [4] to formulate the poorly soluble drugs as aqueous dosage forms.

Alternatively, the drug is fitted in cyclodextrins, which have a hydrophobic interior and a

hydrophilic exterior, and made soluble in water [5]. Another way of enhancing the

solubility of poorly soluble drugs is to produce nanoparticulate formulations, such as

liposomes [6], micelles [7], nanoemulsion [8], solid lipid nanoparticles [9], and

polymeric nanoparticles [10]. However, relatively low drug loading efficiency, concerns

for the safety of excipients, and complicated manufacturing process are noted as potential

disadvantages of these strategies.

Nanocrystallization is a technique to produce crystalline particles of poorly

soluble drugs in the nanometer range (i.e., nanocrystals). Due to the size and, thus, the

high surface area to volume ratio, nanocrystals can increase the saturation solubility of a

3 Reprinted from Current Opinion in Solid State & Materials Science, 16(6), Bo Sun, Yoon Yeo,

Nanocrystals for the parenteral delivery of poorly water-soluble drugs, p 295-301, Copyright (2015), with

permission from Elsevier.

98

drug and the dissolution rate of drug particles [11]. Nanocrystals have gained increasing

interest in the pharmaceutical industry because of the simple structures and compositions.

They have been explored for a variety of therapeutic applications including oral [12],

dermal [13], pulmonary [14], systemic administration [15], as well as targeted drug

delivery [16] and intraperitoneal chemotherapy [17]. The objective of this section is to

review the production of nanocrystals and discuss the remaining challenges in the

development of nanocrystal products.

4.1.1 Production of Nanocrystals

Nanocrystals of poorly soluble drugs can be created by “top-down” or “bottom-

up” technologies (Fig. 20), or combinations of the two [18]. Nanocrystals are produced

from the drug itself, with surfactants or polymeric stabilizers on the surface; thus, the

drug content in nanocrystals approaches 100% [18]. The production of nanocrystals is

relatively easy to scale up and transfer to industry as compared to other formulations on

the market, such as liposomes [19, 20] and albumin-based nanoparticles [21]. Several

nanocrystal products, produced by wet milling and high-pressure homogenization, have

been approved by the US Food and Drug Administration as oral products (Table 4).

99

Figure 20. Schematics of (A) bottom-up and (B) top-down production of nanocrystals.

Aqueous dispersion of nanocrystals can be further proceeded into sterile products or other

dosage forms.

Suspension of crystalline or amorphous

particles

Homogenization or wet millingVarious techniques of

controlled precipitation

Aqueous suspension of

nanocrystals

(Nanosuspension)

Sterilized liquid

suspension or

lyophilized powder

Solid or semi-solid

products

Top-downBottom-up

Drug solution in a

water-miscible

solvent

Aqueous buffer

with a stabilizer

100

Table 4. Examples of nanocrystal products for oral administration approved by the FDA

[22, 23].

Trade name (drug) Manufacturing

techniques

Indications Company

Rapamune® (Sirolimus) Top-down, wet milling Immunosuppres

sive

Wyeth

Pharmaceuticals

Emend® (Aprepitant) Top-down, wet milling Antiemetic Merck & Co.

Tricor® (Fenofibrate) Top-down, wet milling Hypercholestero

lemia

Abbott

Laboratories

Triglide® (Fenofibrate) Top-down, high-pressure

homogenization

Hypercholestero

lemia

Skye Pharma

Megace ES® (Megestrol

acetate)

Top-down, wet milling Antianorexic Par

Pharmaceutical

Avinza® (Morphine sulfate) Top-down, wet milling Psychostimulant

drug

King

Pharmaceuticals

Focalin® XR (Dexmethyl-

phenidate HCl)

Top-down, wet milling Attention deficit

hyperactivity

disorder

Novartis

Ritalin® LA

(Methylphenidate HCl)

Top-down, wet milling Attention deficit

hyperactivity

disorder

Novartis

Zanaflex CapsulesTM

(Tizanidine HCl)

Top-down, wet milling Muscle relaxant Acorda

4.1.1.1 Bottom-up Technologies

The bottom-up approach refers to methods that create small drug particles from

drug molecules dissolved in an organic solution. Small drug particles are formed as drug

molecules precipitate from solution in the presence of an agent and/or a condition that

induces nucleation of the molecules. For example, a non-solvent, which is miscible with

the solvent but does not dissolve the drug, is used to induce the nanocrystal formation, in

conjunction with various methods to mix the drug solution with non-solvents such as

rotation, liquid jets, or multi-inlet vortex mixing [24]. Alternatively, supercritical fluid,

101

ultrasonic waves, or controlled solvent evaporation are employed to induce drug

precipitation. These technologies are discussed in detail in a recent review article [24].

Particles produced by the bottom-up approach can be crystalline or amorphous.

Amorphous nanoparticles produced by a technique called NanomorphTM achieve higher

saturation solubility and faster dissolution rate than nanocrystals [25, 26]. However, they

are prone to partial or complete re-crystallization, which may lead to decreased

bioavailability. Due to the stability and consistent performance, nanocrystals are usually

favored over amorphous particles. On the other hand, the production of nanocrystals

within a desired size range depends critically on precise control of the precipitation and

prevention of the crystal growth during the production [18]. The complexity of the

process control and potential risk of residual organic solvents have discouraged the

development of commercial products [22]. Recently, spray-drying [27] and freeze-drying

[28, 29] processes have been used to achieve continuous control of the crystallization at

large scales.

4.1.1.2 Top-down Technologies

Top-down approach is based on two basic size reduction methods: wet milling

[30] and high-pressure homogenization [31]. The wet milling process applies shear stress

on large drug particles by grinding an aqueous suspension that contains a drug and a

surface stabilizer using beads or pearls in a milling chamber [32]. The outcome of the

milling process is determined by the hardness of the drug, energy input, milling time, and

stabilizer concentration [32]. Microfluidization and piston-gap homogenization are

examples of high-pressure homogenization [18]. Microfluidization is based on the jet

102

stream principle, where the size diminution is achieved by collision of two fluid streams

of particle suspension in a Y-type chamber under high pressure [22, 31]. The piston-gap

homogenizer forces a particle suspension to pass a small gap (~5 µm) under pressure.

The high shear forces, turbulent flow, and cavitation generated during this passage can

reduce the particle size to the nanometer range [22, 31]. The performance of this process

depends on the number of cycles, power density, and temperature [31]. These techniques

are widely used in industry.

Compared to bottom-up methods, top-down methods require higher energy

consumption and a longer operation time. The risk of contamination due to the erosion of

milling beads is also a disadvantage of wet milling [33]. Moreover, the high-energy

process may induce phase transition of a drug, which may compromise the in-vivo

performance of the products [34].

4.1.1.3 Combined Technologies

A pre-treatment step (bottom-up) and particle size reduction step (top-down) may

be combined. For example, precipitates are first obtained from anti-solvent precipitation,

spray-drying, or lyophilization (pre-treatment step), followed by high-pressure

homogenization (particle-size reduction) [18, 22]. One of the roles of the high-pressure

homogenization step is to anneal the initial precipitates, which are often

thermodynamically unstable, into an ordered crystal structure [22]. Two well-established

techniques, NANOEDGETM and smartcrystals®, have been discussed elsewhere in detail

[18, 22].

103

4.1.1.4 Nanocrystal Stabilization

Due to the high surface energy generated by nanonization, surface stabilizers are

needed to prevent aggregation and precipitation of nanocrystals. Examples of stabilizing

systems are summarized in a recent review article [23]. For drug nanocrystals with no

surface charge, anionic surfactants such as sodium cholate, sodium deoxycholate, and

sodium lauryl sulfate are often used to keep the particles separated via electrostatic

repulsion. Another way of stabilizing nanocrystals is to apply polymeric stabilizers on

their surface and establish a steric barrier against aggregation. Polymers used for this

purpose are derivatives of cellulose, polyvinyl alcohol, polyvinyl pyrrolidone,

Polysorbates (polyoxyethylene sorbitan fatty acid esters), and Pluronics (or Poloxamers,

triblock-copolymers of polyoxyethylene and polyoxypropylene) [35, 36]. Some of the

stabilizers, such as arginine, amphiphilic amino acid copolymers, and vitamin E

polyethylene glycol succinate (TPGS), are biologically active and provide additional

functions to the nanocrystals [30]. For example, TPGS, an effective P-glycoprotein

inhibitor [37], enables paclitaxel (PTX) nanocrystals to overcome multidrug resistance

[38]. The effectiveness of PTX nanocrystals stabilized with TPGS was demonstrated in a

nude mouse model bearing multidrug-resistant NCI/ADR-RES human ovarian cancer

cells [38].

The effectiveness of a nanocrystal stabilizer depends on its affinity for a drug, the

concentration, and the stabilizer to drug ratio in suspension. Relatively hydrophobic

stabilizers have higher affinity for drug crystals and a greater stabilizing effect [39].

There is a positive correlation between particle size and the hydrophilic lipophilic balance

(HLB) value of a non-ionic surfactant in bottom-up approaches; thus, the HLB value can

104

be a useful guideline for the selection of a stabilizer [40]. Typically, a surfactant with a

low HLB value (lipophilic surfactant) is a good stabilizer of hydrophobic nanocrystals.

Adequate surface coverage by stabilizers, irrespective of their mechanisms, is critical to

the stabilization of nanocrystals. However, it does not necessarily mean that the stability

increases in proportion to the concentration of a stabilizer. When the concentration of a

surfactant exceeds the critical micelle concentration (CMC), the excessive surfactant has

a negative effect on the stability of the nanocrystal suspension (nanosuspension) because

micelle formation begins to compete with adsorption to the nanocrystal surface [41-43].

4.1.2 Remaining Challenges in Nanocrystal Development for Parental Applications

4.1.2.1 Instability during Storage

Ostwald ripening refers to a phenomenon that small particles gradually dissolve

and redeposit on the surface of larger particles over time (Fig. 21) [44]. It occurs when

the particle size in a dispersion system is heterogeneous and the dispersed phase (drug)

has a limited solubility in the medium (water) [45], which, unfortunately, are the

conditions frequently encountered in pharmaceutical suspensions [46, 47]. Ostwald

ripening leads to the particle size growth and physical instability of a dispersion system

during storage; therefore, there is a strong need for preventing this process.

105

Figure 21. Schematic representation of Ostwald ripening in nanosuspension.

Surfactants and polymers are commonly used as stabilizers to delay detachment

and attachment of drug molecules at the surface of dispersed particles [45]. Polymers are

believed to be more effective than small molecular-weight surfactants because they tend

to adhere to the nanocrystal surface less dynamically than surfactants [48]. Another way

of preventing particle size growth is to produce uniform nanocrystals, thus eliminating

one of the conditions for Ostwald ripening. Optimizing the process parameters, such as

the number of high-pressure homogenization cycles [49], milling time [34, 50] and

milling speed [51], can eliminate large particles and obtain narrow size distribution.

Processing nanosuspensions into solid products is another way of avoiding dynamic

changes in the medium and thus Ostwald ripening [23, 52].

4.1.2.2 Instability during Applications

Due to the high surface area to volume ratio, complete dissolution of nanocrystals

can occur quickly (in less than an hour) as long as a sink condition is maintained (Table.

5). On the other hand, in locations with a limited volume of fluid, such as the peritoneal

cavity, stomach, or the lungs, nanocrystals may be exposed to a non-sink condition for an

Nanocrystals with different

particle sizes

Drug molecules gradually

transfer from small particles

to large particles.

Large particles grow

continuously at the expense of

small particles and dominate

the system.

106

extended period of time. This may limit the dissolution rate of the nanocrystals and

provide an opportunity for sustained drug release [16]. However, with the gradual surface

erosion and loss of stabilizing agents, the nanocrystals may become increasingly unstable

and, thus, undergo agglomeration and Ostwald ripening over time. When this occurs,

drug dissolution slows down significantly, to an extent that the drug is no longer

bioavailable. We have experienced a consequence of particle agglomeration in an animal

model of IP tumors [53]: we produced PTX particles with precipitation followed by

sonication and administered the particles IP into mice bearing ovarian tumors in the

peritoneal cavity, using an in-situ crosslinkable hyaluronic acid-based hydrogel as a

carrier [53]. The PTX precipitates delivered with the hydrogel were best retained in the

peritoneal cavity as compared to other formulations, which included multiple injections

of Taxol, a bolus injection of Taxol, PTX precipitates alone, and Taxol delivered with the

hydrogel. Despite the prolonged IP retention, the anti-tumor effect of hydrogel-embedded

PTX precipitates was not significantly different from the others, which were cleared from

the peritoneal cavity much earlier. One of the possible explanations is the agglomeration

of PTX precipitates, which led to incomplete dissolution of PTX [53].

107

Table 5. Examples of nanocrystal dissolution in a sink condition.

Drug Particle size (nm) Dissolution rate Reference

Oridonin 322.7 98% dissolved in 24 min [41]

Asulacrine d(v; 0.5)* 133 ± 20 42% dissolved in 6 h [15]

Celecoxib d(v; 0.5) 360 91.8% dissolved in 50 min [54]

Meloxicam d(v; 0.5) 530 ± 110 100% dissolved in 10 min [55]

Artemisinin 100-360 75.9% dissolved in 4 h [56]

Nitrendipine 209 ± 9 90% dissolved in 2 min [57]

Quercetin 213.6 ± 29.3 73.2% dissolved in 20 min [58]

Itraconazole ~300 85% dissolved in 90 min [12]

Camptothecin 200-700 50% dissolved in 2 h [16]

*Note: size of the particles for which 50% of the same volume contains particles smaller

than d (v; 0.5).

4.1.2.3 Lack of Target Specificity

The surface of nanocrystals is decorated with specific ligands for target-specific

delivery. However, continuous surface erosion poses a challenge to the longevity of the

targeting effect. In this regard, it is worthwhile to note a recent approach to produce a co-

crystal of a drug and functional molecules [59]. Here, Li et al. produced hybrid crystals

by co-crystallization of PTX and fluorescent dyes, where guest dye molecules were

integrated in PTX nanocrystals [59]. Since the dye was embedded throughout the matrix,

the PTX-dye hybrid nanocrystals could be located via real-time imaging during their

lifetime in the body. The same principle is applicable to producing drug-ligand hybrid

nanocrystals with the prolonged target specificity.

108

4.2 Introduction

While the design of nanoparticulate formulations has become increasingly

sophisticated, structurally and conceptually simple nanocrystals have a unique advantage

with respect to the development of commercial products. Nanocrystals can greatly

enhance the dissolution rate of poorly soluble drugs. The large contact area of

nanocrystals can allow for a greater interaction with tissue or cell surfaces and enhance

drug absorption. Several oral nanocrystal products are available on the market and dermal

and IV products are actively explored. However, the potential of nanocrystals has not

been thoroughly investigated for different applications such as targeted or local drug

delivery. For example, nanocrystals can be combined with implantable delivery systems

to attain a higher local concentration for a prolonged period of time. This requires

nanocrystals to maintain the small size during long-term delivery without using toxic

surfactants as surface stabilizers. Current methods of nanocrystal production do not

adequately address the challenges in development of such products, and new approaches

to engineer nanocrystals are strongly awaited.

To overcome the limitations in current nanocrystal production for local therapy,

we developed a method to make PTX nanocrystals smaller than 200 nm with albumin as

a surface stabilizer. Albumin was chosen because of its ability of adsorbing onto

hydrophobic surfaces and therefore provides steric hindrance to nanocrystal aggregation

and growth [60, 61]. Moreover, serum proteins have the ability to bind certain membrane

proteins which may facilitate targeting of anticancer drugs to the tumors [62, 63]. This

method involves crystallization in a matrix of surfactant, such as Pluronic F127. The

109

presence of polymer matrix limits the growth of crystals; thus, this method can form

relatively small particle size (<200 nm). The incipient nanocrystals (iCim) are harvested

by sonication and hydration and stabilized with surface modifiers such as albumin.

Pluronic F127 is replaced by the surface modifiers with higher affinity for nanocrystals

and mostly absent in the final product. Due to the small size and the lack of polymeric

surfactant, it is expected to provide faster dissolution of a drug and greater safety than

other nanocrystal formulations.

4.3 Materials and Methods

4.3.1 Materials

Paclitaxel (PTX) was a gift of Samyang Genex Corp (Seoul, Korea). Pluronic

F127 was a gift from BASF (New York, NY, USA). Albumin from human serum (alb, 66

kDa, ≥ 96%) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Reagents for

sodium dodecyl sulfate polyacrylamide (SDS-PAGE) gel electrophoresis were purchased

from Bio-Rad (Hercules, CA, USA). Micro BCA Protein Assay Kit was purchased from

Life technologies (Grand Island, NY, USA). All other reagents were purchased from

Sigma-Aldrich (St. Louis, MO, USA).

110

4.3.2 Preparation of Albumin-stabilized PNC

4.3.2.1 Crystallization in Matrix (Cim)

Crystallization in Matrix consists of two steps: first, incipient crystallization in

matrix (iCim), adapted from the literature [64]; second, surface stabilization of iCim.

Incipient Crystalliation in Matrix (iCim): PTX was crystallized in the polymeric

matrix and recovered by sonication and hydration (Fig. 22). Briefly, 6 mg PTX and 24

mg Pluronic F127 (1/4, w/w) were fully dissolved in 3 mL of chloroform in a round

bottom flask. Chloroform was evaporated with rotary evaporator at 40 ℃ for 2 hours to

form a thin film on the wall of the flask. Six milliliters of DI water was added to hydrate

the film under stirring at room temperature for 1hour. The hydrated suspension was

sonicated with ice-water bath to help reduce the particle size. The settings of the probe

sonicator were: power = 40%, time = 15 min and pulse = 1s/1s.

Surface Stabilization of iCim: iCim was stabilized with albumin. iCim suspension

(PTX concentration = 1 mg/mL) was mixed with an equal volume of aqueous albumin

aqueous solution (8 mg/mL) and incubated at room temperature for 3 hours on a rotating

rocker. The mixture was centrifuged at 4 ℃ at 104 rpm (9300 rcf) for 10 min to remove

Pluronic F127 and unadsorbed albumin. The pellet was re-suspended with 0.5 mg/mL

albumin solution, and the surface stabilized nanocrystals were collected by centrifugation

at 4 ℃ at 104 rpm (9300 rcf) for 10 min. This nanocrystal formulation was called Cim-

alb.

111

4.3.2.2 Nonsolvent and Temperature-Induced Crystallization

Control nanocrystals were prepared by the bottom-up method based on

nonsolvent and temperature-induced crystallization [59] (Section 2.2.2) and referred to as

PNC. To further stabilize PNC, PNC suspension (1 mg/mL) was mixed with an equal

volume of aqueous albumin solution (2 mg/mL) and incubated at room temperature for

1.5 hours on a rotating rocker. The albumin-coated PNC (PNC-alb) was collected by

centrifugation at 4 ℃ at 104 rpm (9300 rcf) for 15 min and washed with deionized (DI)

water twice.

Figure 22. Diagram of preparation of Cim-alb.

PTX and F127 in

chloroform

Evaporate at 40⁰C

for 2 h

A dry film of PTX and

F127 forms on the flask

wall

Hydrate the film at R.T.

for 1 h

Ice-water bath

Probe sonication

for size reduction

-F127

-PTX particles

-PTX nanocrystals

Mixing PNC-

F127 with HSA

solution

Centrifugation

to collect

particles

-Albumin

Cim-alb

112

4.3.3 Characterization of Nanocrystals

The particle sizes of iCim, Cim-alb, PNC, and PNC-alb were measured in DI

water with a Zetasizer Nano-ZS90 (Malvern instruments, Westborough, MA, USA). The

morphology of lyophilized Cim-alb and PNC-alb was visualized with a FEI Nova

nanoSEM field emission scanning electron microscopy (Hillsboro, OR, USA). Cim-alb

and PNC-alb were sputter-coated with platinum for 1 min and observed with a high

resolution through-the-lens detector under 5kV accelerating voltage and spot size 3. The

protein content of Cim-alb or PNC-alb was determined with Micro BCA protein assay or

gel electrophoresis.

4.4 Results and Discussions

4.4.1 Nanocrystal Morphology

PNC-alb showed a rod-shape with a length of ~400 nm and a width of ~150 nm

under SEM (Fig. 23A). Cim-alb showed a similar shape as PNC with a length of ~500

nm and the width of ~80 nm under SEM (Fig. 23B).

113

Figure 23. Scanning electron micrograph of lyophilized (A) PNC-alb and (B) Cim-alb.

Scale bar: 500 nm.

4.4.2 Nanocrystal Size

The size difference visualized in SEM was reflected in DLS measurement. The

average diameter of iCim measured by DLS was 157.1 nm. With surface stabilization

with albumin, Cim-alb had an average diameter of 186.4 nm (Table 6). The initial size

increase was induced by centrifugation, but Cim-alb withstood subsequent centrifugation

with no further size increase. The average diameter of PNC was 301.3 nm. With albumin

stabilization the particle size was 335.5 nm.

A B

114

Table 6. Particle size of albumin-stabilized PNC and intermediate products.

Size (nm)* Polydispersity index range

Min. Max.

iCim (n=8) 157.1 ± 14.6 0.079 0.226

Cim-alb after first centrifugation

(n=9)

188.7 ± 24.6 0.052 0.313

Cim-alb after second centrifugation

(n=2)

186.4 ± 18.1 0.126 0.209

PNC (n=5) 301.3 ± 17.9 0.049 0.362

PNC-alb (n=7) 335.5 ± 19.7 0.152 0.372

*: average diameter and standard deviation of n batches prepared independently and

identically.

The protective effect of albumin was evident when PNC and PNC-alb were

subjected to repeated centrifugation at 4 ℃ at 104 rpm (9300 rcf) for 10 min: bare PNC

aggregated to form ~3 µm particles, while PNC-alb was kept at ~330 nm (Fig. 24). This

observation proved the effectiveness of albumin as a surface stabilizer to PNC.

Figure 24. Particle size of PNC and PNC-alb after repeated centrifugations.

pa

rti

cle

siz

e (

d.

nm

)

Or i

gin

al

1st

cen

trif

ug

at i

on

2n

d c

en

trif

ug

at i

on

3rd

cen

trif

ug

at i

on

4th

cen

trif

ug

at i

on

0

1 0 0 0

2 0 0 0

3 0 0 0

4 0 0 0

5 0 0 0P N C -a lb P N C

115

4.4.3 Albumin content

The albumin contents in Cim-alb and PNC-alb were determined using standard

Micro BCA protein assay. Four percent SDS aqueous solution was used to strip off the

albumin from the nanocrystal surface. The weight ratio between PTX and albumin was

8.21/1 (PTX loading % = 89.1%). However, PTX can interfere with BCA assay because

of the secondary amino group in the structure. SDS-PAGE gel electrophoresis was

recommended to determine the albumin content though the influence of PTX could be

deducted in the BCA assay. The albumin content in iCim-alb was measured based on the

correlation between albumin concentration and band intensity in SDS-PAGE gel

electrophoresis. The weight ratio between PTX and albumin in iCim-alb was 6.3/1 (PTX

loading % =85%) (With courtesy of Joonyoung Park).

4.4.4 Proposed Role of Albumin in Cim-alb

Cim-alb maintained original particle size after repeated centrifugation, whereas

iCim (before albumin protection) could not be resuspended. This indicates albumin was

present on the nanocrystal surface and prevented hydrophobic aggregation of the

particles. The quantity of remaining Pluronic F127 after albumin adsorption has not been

directly measured. Given that albumin and PTX account for 13.5% and 85%,

respectively, the remaining Pluronic F127 is likely to be <2% (With courtesy of

Joonyoung Park). There are two possible scenarios in albumin coverage of the

nanocrystal surface: first, albumin added onto Pluronic F127 surface; second, albumin

replacing Pluronic F127 on nanocrystal surface. Considering the low Pluronic F127

116

content in Cim-alb and improved size stability, second scenario should be the case though

direct evident was still needed.

4.5 Conclusion

Albumin-stabilized nanocrystals with a sub-200 nm particle size were prepared

using a method involving incipient crystallization in polymer matrix and subsequent

surface stabilization with albumin. The presence of polymeric matrix limited the growth

of crystals; thus, PTX could form nanocrystals at ~160 nm. The surface stabilized

nanocrystals (Cim-alb) maintained particle size below 200 nm. Due to the reduced

particle size and the lack of a surfactant, Cim-alb is expected to provide faster PTX

dissolution and greater safety than conventional nanocrystal formulations. The function

and quantitation of surface stabilizers need further investigation. In-vitro dissolution test

and bioactivity evaluation of Cim-alb remains to be performed to test the contribution of

small size to enhancing local availability of PTX.

117

4.6 References

1. Merisko-Liversidge, E.M. et al., Drug Nanoparticles: Formulating Poorly Water-

Soluble Compounds. Toxicol Pathol, 2008. 36(1): p. 43-48.

2. Rabinow, B.E., Nanosuspensions in drug delivery. Nat Rev Drug Discov, 2004.

3(9): p. 785-796.

3. Squillante, I., Emilio et al., Solid Dispersions: Revival with Greater Possibilities

and Applications in Oral Drug Delivery. Crit Rev Ther Drug Carrier Syst, 2003.

20(2&3): p. 34.

4. He, S., et al., A Cremophor-Free Self-Microemulsified Delivery System for

Intravenous Injection of Teniposide: Evaluation In Vitro and In Vivo. AAPS

PharmSciTech, 2012. 13(3): p. 846-852.

5. Bouquet, W., et al., Paclitaxel/β-cyclodextrin complexes for hyperthermic

peritoneal perfusion – Formulation and stability. Eur J Pharm Biopharm, 2007.

66(3): p. 391-397.

6. Chen, H., et al., Lactoferrin-modified procationic liposomes as a novel drug

carrier for brain delivery. Eur J Pharm Sci, 2010. 40(2): p. 94-102.

7. Shao, K., et al., Angiopep-2 modified PE-PEG based polymeric micelles for

amphotericin B delivery targeted to the brain. J Control Release, 2010. 147(1): p.

118-126.

8. Gong, Y., et al., An Excellent Delivery System for Improving the Oral

Bioavailability of Natural Vitamin E in Rats. AAPS PharmSciTech, 2012. 13(3):

p. 961-966.

118

9. Dodiya, S.S., et al., Solid lipid nanoparticles and nanosuspension formulation of

Saquinavir: preparation, characterization, pharmacokinetics and biodistribution

studies. J Microencapsul, 2011. 28(6): p. 515-527.

10. Gullotti, E. et al., Beyond the imaging: Limitations of cellular uptake study in the

evaluation of nanoparticles. J Control Release, 2012 164(2): p 170-176.

11. Patravale, V.B., et al., Nanosuspensions: a promising drug delivery strategy. J

Pharm Pharmacol, 2004. 56(7): p. 827-840.

12. Sun, W., et al., Nanonization of itraconazole by high pressure homogenization:

Stabilizer optimization and effect of particle size on oral absorption. J Pharm Sci,

2011. 100(8): p. 3365-3373.

13. Mitri, K., et al., Lutein nanocrystals as antioxidant formulation for oral and

dermal delivery. Int J Pharm, 2011. 420(1): p. 141-146.

14. Zhang, J., et al., Enhanced bioavailability after oral and pulmonary

administration of baicalein nanocrystal. Int J Pharm, 2011. 420(1): p. 180-188.

15. Ganta, S., et al., Formulation and pharmacokinetic evaluation of an asulacrine

nanocrystalline suspension for intravenous delivery. Int J Pharm, 2009. 367(1–2):

p. 179-186.

16. Zhang, H., et al., Preparation and antitumor study of camptothecin nanocrystals.

Int J Pharm, 2011. 415(1–2): p. 293-300.

17. De Smet, L., et al., Development of a Nanocrystalline Paclitaxel Formulation for

Hipec Treatment. Pharm Res, 2012. 29(9): p. 1-9.

119

18. Müller, R.H., et al., State of the art of nanocrystals – Special features, production,

nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm, 2011.

78(1): p. 1-9.

19. Barenholz, Y., Doxil® — The first FDA-approved nano-drug: Lessons learned. J

Control Release, 2012. 160(2): p. 117-134.

20. Xiong, S., et al., Preparation, therapeutic efficacy and intratumoral localization

of targeted daunorubicin liposomes conjugating folate-PEG-CHEMS. Biomed

Pharmacother, 2011. 65(1): p. 2-8.

21. Montero, A.J., et al., Nab-paclitaxel in the treatment of metastatic breast cancer:

a comprehensive review. Expert Rev Clin Pharmacol, 2011. 4(3): p. 329-334.

22. Shegokar, R. et al., Nanocrystals: Industrially feasible multifunctional

formulation technology for poorly soluble actives. Int J Pharm, 2010. 399(1–2): p.

129-139.

23. Van Eerdenbrugh, B., et al., Top-down production of drug nanocrystals:

Nanosuspension stabilization, miniaturization and transformation into solid

products. Int J Pharm, 2008. 364(1): p. 64-75.

24. Chan, H.-K. et al., Production methods for nanodrug particles using the bottom-

up approach. Adv Drug Deliv Rev, 2011. 63(6): p. 406-416.

25. Lindfors, L., et al., Amorphous Drug Nanosuspensions. 3. Particle Dissolution

and Crystal Growth. Langmuir, 2007. 23(19): p. 9866-9874.

26. Thombre, A.G., et al., In vitro and in vivo characterization of amorphous,

nanocrystalline, and crystalline ziprasidone formulations. Int J Pharm, 2012.

428(1–2): p. 8-17.

120

27. Hu, J., et al., Continuous and scalable process for water-redispersible

nanoformulation of poorly aqueous soluble APIs by antisolvent precipitation and

spray-drying. Int J Pharm, 2011. 404(1–2): p. 198-204.

28. de Waard, H., et al., Preparation of drug nanocrystals by controlled

crystallization: Application of a 3-way nozzle to prevent premature crystallization

for large scale production. Eur J Pharm Sci, 2009. 38(3): p. 224-229.

29. de Waard, H., et al., A novel bottom–up process to produce drug nanocrystals:

Controlled crystallization during freeze-drying. J Control Release, 2008. 128(2):

p. 179-183.

30. Merisko-Liversidge, et al., Nanosizing for oral and parenteral drug delivery: A

perspective on formulating poorly-water soluble compounds using wet media

milling technology. Adv Drug Deliv Rev, 2011. 63(6): p. 427-440.

31. Keck, C.M. et al., Drug nanocrystals of poorly soluble drugs produced by high

pressure homogenisation. Eur J Pharm Biopharm, 2006. 62(1): p. 3-16.

32. Merisko-Liversidge, et al., Nanosizing: a formulation approach for poorly-water-

soluble compounds. Eur J Pharm Sci, 2003. 18(2): p. 113-120.

33. Juhnke, M., et al., Generation of wear during the production of drug

nanosuspensions by wet media milling. Eur J Pharm Biopharm, 2012. 81(1): p.

214-222.

34. Begat, P., et al., The effect of mechanical processing on surface stability of

pharmaceutical powders: Visualization by atomic force microscopy. J Pharm Sci,

2003. 92(3): p. 611-620.

121

35. Raghavan, S.L., et al., Crystallization of hydrocortisone acetate: influence of

polymers. Int J Pharm, 2001. 212(2): p. 213-221.

36. Raghavan, S.L., et al., Formation and stabilisation of triclosan colloidal

suspensions using supersaturated systems. Int J Pharm, 2003. 261(1–2): p. 153-

158.

37. Dintaman, J.M. et al., Inhibition of P-Glycoprotein by D-α-Tocopheryl

Polyethylene Glycol 1000 Succinate (TPGS). Pharm Res, 1999. 16(10): p. 1550-

1556.

38. Liu, Y., et al., Paclitaxel Nanocrystals for Overcoming Multidrug Resistance in

Cancer. Mol Pharm, 2010. 7(3): p. 863-869.

39. Van Eerdenbrugh, B., et al., A screening study of surface stabilization during the

production of drug nanocrystals. J Pharm Sci, 2009. 98(6): p. 2091-2103.

40. Verma, S., et al., A comparative study of top-down and bottom-up approaches for

the preparation of micro/nanosuspensions. Int J Pharm, 2009. 380(1–2): p. 216-

222.

41. Gao, L., et al., Preparation and Characterization of an Oridonin Nanosuspension

for Solubility and Dissolution Velocity Enhancement. Drug Dev Ind Pharm, 2007.

33(12): p. 1332-1339.

42. Deng, J., et al., Understanding the structure and stability of paclitaxel

nanocrystals. Int J Pharm, 2010. 390(2): p. 242-249.

43. Deng, Z., et al., Understanding a relaxation behavior in a nanoparticle

suspension for drug delivery applications. Int J Pharm, 2008. 351(1–2): p. 236-

243.

122

44. Yao, J.H., et al., Theory and simulation of Ostwald ripening. Physical Review B,

1993. 47(21): p. 14110-14125.

45. Verma, S., et al., Physical stability of nanosuspensions: Investigation of the role

of stabilizers on Ostwald ripening. Int J Pharm, 2011. 406(1–2): p. 145-152.

46. Lindfors, L., et al., Amorphous Drug Nanosuspensions. 1. Inhibition of Ostwald

Ripening. Langmuir, 2005. 22(3): p. 906-910.

47. Meinders, M.B.J. et al., The role of interfacial rheological properties on Ostwald

ripening in emulsions. Adv Colloid Interfa Sci, 2004. 108(1): p. 119-126.

48. Walstra, P., Formation of emulsions, in Encyclopedia of Emulsion Technology:

Volume 1 - Basic Theory, P. Becher, Editor 1983, Marcel Dekker: New York. p.

57-128.

49. Gao, Y., et al., Preparation, characterization, pharmacokinetics, and tissue

distribution of curcumin nanosuspension with TPGS as stabilizer. Drug Dev Ind

Pharm, 2010. 36(10): p. 1225-1234.

50. Ali, H.S.M., et al., Hydrocortisone nanosuspensions for ophthalmic delivery: A

comparative study between microfluidic nanoprecipitation and wet milling. J

Control Release, 2011. 149(2): p. 175-181.

51. Singare, D.S., et al., Optimization of formulation and process variable of

nanosuspension: An industrial perspective. Int J Pharm, 2010. 402(1–2): p. 213-

220.

52. Van Eerdenbrugh, B., et al., Drying of crystalline drug nanosuspensions—The

importance of surface hydrophobicity on dissolution behavior upon redispersion.

Eur J Pharm Sci, 2008. 35(1–2): p. 127-135.

123

53. Bajaj, G., et al., Hyaluronic acid-based hydrogel for regional delivery of

paclitaxel to intraperitoneal tumors. J Control Release, 2012. 158(3): p. 386-392.

54. Dolenc, A., et al., Advantages of celecoxib nanosuspension formulation and

transformation into tablets. Int J Pharm, 2009. 376(1–2): p. 204-212.

55. Ambrus, R., et al., Investigation of preparation parameters to improve the

dissolution of poorly water-soluble meloxicam. Int J Pharm, 2009. 381(2): p. 153-

159.

56. Kakran, M., et al., Fabrication of drug nanoparticles by evaporative precipitation

of nanosuspension. Int J Pharm, 2010. 383(1–2): p. 285-292.

57. Xia, D., et al., Preparation of stable nitrendipine nanosuspensions using the

precipitation–ultrasonication method for enhancement of dissolution and oral

bioavailability. Eur J Pharm Sci, 2010. 40(4): p. 325-334.

58. Gao, L., et al., Preparation of a chemically stable quercetin formulation using

nanosuspension technology. Int J Pharm, 2011. 404(1–2): p. 231-237.

59. Zhao, R., et al., Hybrid Nanocrystals: Achieving Concurrent Therapeutic and

Bioimaging Functionalities toward Solid Tumors. Mol Pharm, 2011. 8(5): p.

1985-1991.

60. Jeyachandran, Y.L., et al., Quantitative and Qualitative Evaluation of

Adsorption/Desorption of Bovine Serum Albumin on Hydrophilic and

Hydrophobic Surfaces. Langmuir, 2009. 25(19): p. 11614-11620.

61. Seo, J., et al., Facile internalization of paclitaxel on titania nanoparticles in

human lung carcinoma cells after adsorption of serum proteins. J Nanopart Res,

2012. 14(10): p. 1-8.

124

62. Kratz, F. et al., Clinical impact of serum proteins on drug delivery. J Control

Release, 2012. 161(2): p. 429-45.

63. Zhang, J.-Y., et al., Preparation of the albumin nanoparticle system loaded with

both paclitaxel and sorafenib and its evaluation in vitro and in vivo. J

Microencapsul, 2011. 28(6): p. 528-536.

64. Liu, F., et al., Targeted cancer therapy with novel high drug-loading

nanocrystals. J Pharm Sci, 2010. 99(8): p. 3542-3551.

125

CHAPTER 5. CONCLUSION

PTX was delivered as PNC via an in-situ crosslinkable HA gel, aiming to increase

the retention and chemotherapeutic effect in the peritoneal cavity. Several attempts were

made to evaluate the difference in dissolution rate between PNCs and uncontrolled

precipitates (PPTs) using in-vitro conditions reflecting the peritoneal cavity, such as non-

sink condition and amphiphilic components. The expected difference between PNC and

PPT or gels containing the two was not clearly seen in the in-vitro release studies

(Chapter 2). Nevertheless, PNC was more efficient in killing SKOV3 cells than PPT, and

PNC-gel showed some advantage in anti-tumor efficacy on tumor bearing mice over

PPT-gel. However, the heterogeneity of tumor growth and decreasing number of animals

following the tumor relapse made it difficult to statistically differentiate PNC-gel from

PPT-gel based on bioluminiescence imaging (Chapter 3).

Although the cytotoxicity result and in-vivo anti-tumor effects demonstrate the

PNC-gel demonstrated its effectiveness as an IP chemotherapy delivery system, the small

extent of improvement over PPT-gel suggest that PTX dissolution was still limited in the

non-sink condition of the peritoneal cavity. PNC-gel can be further improved at least in

two aspects. First, it is possible that PNC may have segregated from the hydrogel matrix

and formed aggregates during the prolonged incubation in hydrogel, further delaying the

PTX release. To prevent PNC segregation, HA gel may be modified to improve the

126

compatibility with PNC. To this end, HA gel conjugated with a derivative of 5β-cholanic

acid was synthesized, which would form hydrophobic domains to accommodate

hydrophobic PNC (Chapter 2). The hydrophobically modified hydrogel improved PTX

release, confirming that compatibility between PNC and hydrogel is an important factor

in local delivery of PTX. The current form of the hydrophobically modified hydrogel

will not be pursued due to the unexpected toxicity of the gel precursor (EtCA-CMC-

CHO), but it will be worthwhile to synthesize an alternative hydrophobitized gel

precursor with low toxicity. Second, PNC size may be further reduced to improve

dissolution of PTX. PNC produced with nonsolvent and temperature induced

crystallization method, used for the in-vivo study, and had an average diameter of ~ 260

nm. Albumin-stabilization helped prevent aggregation of PNC but did not produce

smaller PNC. A new method of producing smaller PNC has been developed, based on

crystallization in surfactant matrix and surface stabilization with albumin (Chapter 4).

This method involves forming sub-200 nm PNC with Pluronic F127 and replacing it with

albumin for further stabilization. The function and quantitation of surface stabilizers on

Cim-alb remain to be investigated.

The future delivery systems may be built with hydrophobically modified

hydrogels and/or Cim-alb NCs. Ultimately, their effectiveness in reducing IP tumor

burdens needs to be tested in-vivo, but mechanistic understanding of the contributions of

each component will depend on well-controlled in-vitro studies. This study identified the

limitations of current in-vitro drug release kinetics in predicting the in-vivo outcome of

different formulations. Ideally in-vitro release kinetics should not involve artifacts due to

sampling procedures, such as centrifugation and dialysis, but should be easy to accurately

127

analyze. In this regard, it is worthwhile to note a promising method based on the linearity

between light scattering intensity and the concentration of partially dissolved particles. A

new release/dissolution kinetics test method will be highly beneficial with the advent of

new drug delivery systems, in particular those including poorly water-soluble drugs for

local application to sites with limited body fluid, such as the lungs and the peritoneal

cavity.

127

VITA

128

VITA

Bo Sun, Ph.D.

Education

2011-2016 Ph.D. in Pharmaceutics (Advisor: Dr. Yoon Yeo)

Dept. of Industrial and Physical Pharmacy, College of Pharmacy, Purdue

University, West Lafayette, IN, USA

2008-2011 M.S. in Pharmaceutics (Advisor: Dr. Jianping Zhou)

China Pharmaceutical University, Nanjing, China

2002-2006 B.S. in Pharmaceutics

China Pharmaceutical University, Nanjing, China

Research Experience

8/2011-5/2016 Graduate Research Assistant

Dept. of Industrial and Physical Pharmacy, College of Pharmacy,

Purdue University, West Lafayette, IN, USA

Ph.D. Dissertation: Nanoparticle Depot for Intraperitoneal

Chemotherapy of Ovarian Cancer

9/2008-5/2011 Graduate Student

Dept. of Pharmaceutics, College of Pharmacy, China Pharmaceutical

129

University, Nanjing, China

Master’s Theses: The Study of Selective Cell Penetrating Peptide on

Tumor Cells and Its Combined Nanocarriers

Professional Experience

6/2015-8/2015 Internship at Genentech

Manager: Jane Li, Senior Scientist

Dept. of Small Molecule Analytical Chemistry and Quality Control,

Genentech, South San Francisco, USA.

6/2006-2/2007 Production Management Engineer

Sino-American Tianjin Smith Kline & French Laboratories, Ltd.,

Tianjin, China.

Publications

1. Sun, B., Taha, M.S., Ramsey, B., Torregrosa-Allen, S., Elzey, B.D., Yeo, Y.

Intraperitoneal Chemotherapy of Ovarian Cancer by Hydrogel Depot of Paclitaxel

Nanocrystals. (Manuscript under 2nd review)

2. Sun, B. and Yeo, Y. Biomaterials and Drug Delivery Systems for Intraperitoneal

Chemotherapy. In: Wim Ceelen and Edward Levine (Eds.) Intraperitoneal Cancer

Therapy: Principles and Practice. CRC press (Taylor and Francis). 2015.

3. Abouelmagd, S.A.*, Sun, B.*, Chang, A.C., Ku, Y.J., Yeo, Y. Release Kinetics Study

of Poorly Water-Soluble Drugs from Nanoparticles: Are We Doing It Right? Molecular

Pharmaceutics. (2015) 12(3): 997-1003. (*: co-first authors)

130

4. Cho, E.J.*, Sun, B.*, Doh, K.O., Wilson, E.M., Torregrosa-Allen, S., Elzey, B.D.,

Yeo, Y. Intraperitoneal Delivery of Platinum with in-Situ Crosslinkable Hyaluronic

Acid Gel for Local Therapy of Ovarian Cancer. Biomaterials. (2015) 37: 312-319. (*:

co-first authors)

5. Sun, B., Yeo, Y. Nanocrystals for the parenteral delivery of poorly water-soluble

drugs. Current Opinion in Solid State and Materials Science. (2012) 16(6): 295-301.

Awards and Honors

2015 O'Malley Scholarship, College of Pharmacy, Purdue University

2015 Travel award from the Purdue University Center for Cancer Research Hasson

Graduate Travel Fund

2014 Ronald W. Dollens Graduate Scholarship in the Life Sciences, College of

Pharmacy, Purdue University.

2013 Purdue Research Foundation grant for proposal entitled “Nanoparticle depot for

intraperitoneal chemotherapy of ovarian cancer”.

2005 Scholarship from Suzhou Capsugel Ltd., China

2004 Third-class scholarship at China Pharmaceutical University, Nanjing, China