183
University of Groningen Delivery of biologicals van Dijk, Fransien IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2018 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): van Dijk, F. (2018). Delivery of biologicals: Sustained release of cell-specific proteins in fibrosis. [Groningen]: Rijksuniversiteit Groningen. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 10-04-2020

University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

University of Groningen

Delivery of biologicalsvan Dijk, Fransien

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):van Dijk, F. (2018). Delivery of biologicals: Sustained release of cell-specific proteins in fibrosis.[Groningen]: Rijksuniversiteit Groningen.

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 10-04-2020

Page 2: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Delivery of biologicals

Sustained release of cell-specific proteins in fibrosis

Fransien van Dijk

Page 3: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The work described in this thesis was performed at the Groningen Research Institute of

Pharmacy, department of Pharmacokinetics, Toxicology & Targeting and department of

Pharmaceutical Technology and Biopharmacy, University of Groningen, The Netherlands. The

research project was financially supported by a grant from the Netherlands Institute of

Regenerative Medicine (NIRM) and a grant from NanoNext.NL (program 03.10).

Printing of this thesis was financially supported by the University of Groningen, LinXis BV,

InnoCore Pharmaceuticals and BiOrion Technologies BV.

Delivery of biologicals

Sustained release of cell-specific proteins in fibrosis

© Copyright 2018 by Fransien van Dijk. All rights reserved. No part of this thesis may be

reproduced or transmitted in any form or by any means without prior permission of the

author.

ISBN (printed version): 978-94-034-0509-4

ISBN (electronic version): 978-94-034-0508-7

Cover design: Jakko de Jong & Fransien van Dijk

Lay-out: Fransien van Dijk

Printing: Ipskamp Printing

Page 4: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Delivery of biologicals

Sustained release of cell-specific proteins in fibrosis

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de rector magnificus prof. dr. E. Sterken

en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op

vrijdag 23 maart 2018 om 11:00 uur

door

Fransien van Dijk

geboren op 21 oktober 1989 te Groningen

Page 5: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Promotores

Prof. dr. K. Poelstra

Prof. dr. P. Olinga

Copromotores

Dr. L. Beljaars

Dr. W.L.J. Hinrichs

Beoordelingscommissie

Prof. dr. K.N. Faber

Prof. dr. R. Gosens

Prof. dr. J. Trebicka

Page 6: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Paranymphs

Eduard Post

Hester Hoving

Page 7: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 8: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Dit proefschrift is opgedragen aan mijn Oma Sien

‘’Anyone who stops learning is old, whether at twenty or eighty.

Anyone who keeps learning stays young.’’ - Henry Ford

Page 9: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 10: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Table of contents

Chapter 1 Introduction and aim of the thesis 10

Chapter 2 Targeted therapies in liver fibrosis: combining the best parts of 16

platelet-derived growth factor BB and interferon gamma

Chapter 3 Steering therapeutic proteins: the mechanism of action of mimetic 48

interferon gamma delivered to the disease-induced PDGFβ-receptor

Chapter 4 Polymeric microspheres for the sustained release of a protein-based 74

drug carrier targeting the PDGFβ-receptor in the fibrotic kidney

Chapter 5 Pharmacokinetics of a sustained release formulation of 104

PDGFβ-receptor directed carrier proteins to target the fibrotic liver

Chapter 6 The antifibrotic potential of a sustained release formulation of a 128

PDGFβ-receptor targeted rho kinase inhibitor

Chapter 7 Summary and general discussion 154

Chapter 8 Nederlandse samenvatting 172

Dankwoord 177

Curriculum vitae 181

List of publications 182

Page 11: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

1

Page 12: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Introduction and aim of the thesis

Page 13: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 1

12

Introduction

Many new molecular entities currently developed and introduced to the market are

biological, protein-based therapeutics1,2. These therapeutic proteins are increasingly used in

the treatment of a broad spectrum of disorders, including chronic conditions varying from

cancer and diabetes to rheumatoid arthritis3. A severe, chronic condition that cannot be fully

treated yet, and was therefore the focus of this thesis, is fibrosis, characterized by an

excessive production of extracellular matrix (ECM) proteins by activated fibroblasts4.

Therapeutic proteins are particularly interesting candidates for future therapies, because they

often have highly specific and complex functions that cannot be easily mimicked by small-

molecule drugs, and are generally well tolerated1. Clinical application of many experimental

compounds is however often still limited despite these promising characteristics, mostly

related to low exposure to target cells and induction of severe side effects due to ubiquitous

receptor expressions3. An approach to overcome these limitations is to increase their local

concentration at the target cells via cell-selective delivery to disease-specific receptors. It is

widely known that upon activation, fibroblasts highly and specifically express the platelet-

derived growth factor receptor beta (PDGFβR). This receptor provides an excellent target for

the delivery of potential antifibrotic proteins5,6.

Not only the low exposure to target cells impedes the clinical application of many biologicals,

but also the administration route is considered a challenge. Generally, protein therapeutics

are administered parenterally, leading to unfavorable high fluctuations in the plasma

concentration7. Subcutaneously injectable sustained release drug delivery systems assuring

the gradual and prolonged release of a protein therapeutic following a single injection are a

patient-friendly alternative8,9. One such delivery system is a polymeric microsphere

formulation, that allows flexible dosing of the drug9. These microspheres are particularly

attractive as they provide physical protection of the drug from surrounding cells and tissues9.

The in this thesis applied polymers provide diffusion-controlled release of encapsulated

therapeutic proteins, instead of degradation-mediated release, which results in a less harsh

environment for these proteins10. This is important for complex protein constructs like the

cell-specific drug carriers applied in the present thesis.

Page 14: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Introduction and aim of the thesis

13

Aim of the thesis

The aim of this thesis is to evaluate the different PDGFβ-receptor directed constructs that

were developed in the past years, and to gain more insight in the uptake and signaling

mechanism at the cellular level of our most promising lead compound: the PDGFβ-receptor

targeted antifibrotic cytokine interferon gamma (Fibroferon). Additionally, we aim to develop

a sustained release formulation for PDGFβ-receptor targeted therapeutic proteins. Chapter 2

outlines the onset of this thesis regarding the cell-specific delivery of therapeutic proteins. It

provides a review of recent protein-based strategies to deliver compounds to the PDGFβ-

receptor. In the studies described in chapter 3 the signaling pathway of the potent compound

Fibroferon is examined. In an attempt to bring the clinical application of such biologicals one

step closer, we performed the studies described in chapter 4. In these studies, we developed

and tested a sustained release formulation based on polymeric microspheres for our model

protein pPB-HSA, which is a highly versatile PDGFβ-receptor targeted drug carrier composed

of multiple PDGFβR-recognizing peptides (pPB) coupled to human serum albumin (HSA). The

concept of combining cell-specific constructs with a sophisticated polymeric microsphere

formulation was further explored in vitro and in vivo in studies described in chapter 5. In

particular, the in vivo pharmacokinetic release profile of pPB-HSA loaded microspheres was

studied. Finally, the antifibrotic effectivity of microspheres containing the drug carrier

coupled to the active antifibrotic compound Y27632, suitable for use in mice, was tested in

studies described in chapter 6. Chapter 7 provides a summary and a general discussion of the

results described in this thesis, as well as future directions for protein-based drugs and an

appropriate sustained release formulation.

1

Page 15: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 1

14

References

1. Leader B, Baca QJ, Golan DE. Protein therapeutics: A summary and pharmacological

classification. Nat Rev Drug Discov. 2008;7(1):21-39.

2. Kinch MS. An overview of FDA-approved biologics medicines. Drug Discov Today.

2015;20(4):393-398.

3. Pisal DS, Kosloski MP, Balu-Iyer SV. Delivery of therapeutic proteins. J Pharm Sci.

2010;99(6):2557-2575.

4. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology. 2008;134(6):1655-

1669.

5. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

6. van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted therapies in liver fibrosis: Combining

the best parts of platelet-derived growth factor BB and interferon gamma. Front Med

(Lausanne). 2015;2:72.

7. Wu F, Jin T. Polymer-based sustained-release dosage forms for protein drugs,

challenges, and recent advances. AAPS PharmSciTech. 2008;9(4):1218-1229.

8. Prajapati VD, Jani GK, Kapadia JR. Current knowledge on biodegradable microspheres in

drug delivery. Expert Opin Drug Deliv. 2015;12(8):1283-1299.

9. Vaishya R, Khurana V, Patel S, Mitra AK. Long-term delivery of protein therapeutics.

Expert Opin Drug Deliv. 2015;12(3):415-440.

10. Teekamp N, Van Dijk F, Broesder A, et al. Polymeric microspheres for the sustained

release of a protein-based drug carrier targeting the PDGFbeta-receptor in the fibrotic

kidney. Int J Pharm. 2017;534(1-2):229-236.

Page 16: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 17: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

2

Page 18: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

F. van Dijka,b, P. Olingab, K. Poelstraa and L. Beljaarsa

aGroningen Research Institute of Pharmacy, Department of Pharmacokinetics,

Toxicology and Targeting, University of Groningen, Groningen, The Netherlands, bGroningen Research Institute of Pharmacy, Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen, Groningen, The

Netherlands.

Published in Frontiers in Medicine (Lausanne), 2015

Targeted therapies in liver fibrosis:

combining the best parts of

platelet derived growth factor BB

and interferon gamma

Page 19: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

18

Abstract

Cytokines, growth factors and other locally produced mediators play key roles in the

regulation of disease progression. During liver fibrosis, these mediators orchestrate the

balance between pro- and antifibrotic activities as exerted by the hepatic cells. Two important

players in this respect are the profibrotic mediator platelet derived growth factor BB (PDGF-

BB) and the antifibrotic cytokine interferon gamma (IFNγ). PDGF-BB, produced by many

resident and infiltrating cells, causes extensive proliferation, migration and contraction of

hepatic stellate cells (HSCs) and myofibroblasts. These cells are the extracellular matrix

producing hepatic cells and they highly express the PDGFβ-receptor. On the other hand, IFNγ

is produced by natural killer cells in fibrotic livers and is endowed with pro-inflammatory,

antiviral and antifibrotic activities. This cytokine attracted much attention as a possible

therapeutic compound in fibrosis. However, clinical trials yielded disappointing results

because of low efficacy and adverse effects, most likely related to the dual role of IFNγ in

fibrosis. In our studies, we targeted the antifibrotic IFNγ to the liver myofibroblasts. For that,

we altered the cell binding properties of IFNγ, by delivery of the IFNγ-nuclear localization

sequence to the highly expressed PDGFβ-receptor using a PDGFβ-receptor recognizing

peptide, thereby creating a construct referred to as “Fibroferon” (i.e. fibroblast-targeted

interferon γ). In recent years, we demonstrated that HSC-specific delivery of IFNγ increased

its antifibrotic potency and improved its general safety profile in vivo, making Fibroferon

highly suitable for the treatment of (fibrotic) diseases associated with elevated PDGFβ-

receptor expression. The present review summarizes the knowledge on these two key

mediators, PDGF-BB and IFNγ, and outlines how we used this knowledge to create the cell-

specific antifibrotic compound Fibroferon containing parts of both of these mediators.

Page 20: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

19

Introduction

Nowadays the most prevalent causes of chronic injury to the liver are chronic hepatitis B and

C viral (HBV and HCV, respectively) infections, alcohol abuse and metabolic problems related

to obesity. Irrespective of the etiology of the disease, all types of chronic damage can finally

lead to the induction of a common pathological response in the liver. In this common

pathological response the same cells and mediators play a role in the progression from

chronic injury to fibrosis, cirrhosis and eventually to tumor formation. At the moment,

hepatocellular carcinoma is among the top 3 of most lethal carcinomas and its incidence is

still increasing1,2.

Fibrogenesis is characterized by excessive fibrotic matrix deposition, and by activation of

macrophages and myofibroblasts. Within the liver, the major pool of matrix producing

myofibroblasts originates from activated and transformed hepatic stellate cells (HSCs),

although other fibroblast sources such as the portal fibroblast or fibrocytes also have been

identified3-5. Clearly not only myofibroblasts and resident macrophages (Kupffer cells) are

involved, but all other resident cell types contribute each in their own way to the fibrotic

pathology. Additionally, infiltrating immune cells, both of the innate and acquired immune

system, such as B cells, T cells, natural killer (NK) cells, NKT cells, neutrophils and monocytes

contribute to the disease progression. Their specific roles in the disease are becoming more

and more clear4,6,7. Although multiple cells are involved in hepatic fibrosis, crucial players are

the fibroblasts, i.e. in the liver the activated HSCs and myofibroblasts, and macrophages, i.e.

the Kupffer cells together with infiltrated monocytes. The fibroblasts are considered the

master producers of all the fibrotic matrix proteins, while the macrophages are the master

regulators4,5,8.

Fibroblasts and macrophages communicate with each other via production and release of

various growth factors, cytokines, and chemokines. The two most prominent profibrotic

growth factors are platelet derived growth factor BB (PDGF-BB) and transforming growth

factor beta (TGFβ). While PDGF-BB stimulates fibroblast proliferation and migration, TGFβ is

more involved in activation, transformation and matrix production. In our studies, we chose

the PDGFβ-receptor (PDGFβR) as our target receptor for the delivery of antifibrotic

compounds9. The PDGFβR was preferred, since the TGFβ-receptor was not only expressed on

HSCs and myofibroblasts, but also hepatocytes and macrophages express this receptor10-12.

Next to the high production of profibrotic factors in diseased livers, various antifibrotic factors

are produced and released as well13. One of the major antifibrotic factors is interferon gamma

(IFNγ). This cytokine is excreted by infiltrating T cells and NK(T) cells6,14. However, as disease

2

Page 21: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

20

progression continues, the antifibrotic factors cannot effectively counterbalance all

profibrotic stimuli, leading to disturbance of the homeostatic balance and thus worsening of

the disease.

In this review, we will give an overview of the characteristics and pathological roles of two

mediators in fibrosis, i.e. PDGF and IFNγ, and summarize their receptor expressions and

counterbalancing activities in various cell types. After that, recent studies on the structure,

the receptor interaction, the in vitro and in vivo characteristics of the cell-specific therapeutic

compound Fibroferon (previously called BipPB-PEG-IFNγ mimetic, mimγ-BipPB or BOT191),

that contains the most useful parts of both these two abovementioned mediators, are

summarized.

Platelet derived growth factor

PDGF, one of first growth factors characterized, was discovered in the early 1970s as a key

mediator involved in vascular pathologies such as atherosclerosis15. Studies at that time

proved that this growth factor, released from platelets, promoted vascular smooth muscle

cell migration and proliferation. Later on, it became clear that PDGF was a potent mitogen for

all mesenchymal cells, including fibroblasts and myofibroblasts. In later decades, PDGF was

also identified as the most potent mitogen for HSCs in fibrotic livers16. The distribution of the

PDGF-receptor correlated closely with the localization of activated mesenchymal cells

predominantly present in collagen lesions within the liver17.

PDGF belongs to the large cysteine-knot superfamily. This family is characterized by the

presence of eight conserved cysteine residues, forming a typical cysteine-knot structure.

Within this large family, PDGF is structurally and functionally mostly related to the vascular

endothelial growth factor (VEGF) family. The structure of the PDGF-family is highly conserved

throughout the animal species18,19. Four different chains of PDGF-molecules are known: the

traditional A- and B-chains and the more recently discovered C- and D- chains. The PDGF-

molecule is present in a dimeric form and four homodimeric (AA, BB, CC and DD) forms and

one heterodimeric (AB) form are described. PDGF-A (211 aa, 24 kDa) and PDGF-B (241 aa, 27

kDa) are each other’s paralogs. Both proteins are processed mainly intracellularly and

secreted in the dimeric active form. On the other hand, PDGF-C (345 aa, 39 kDa) and PDGF-D

(370 aa, 43 kDa), also each other’s paralogs, are secreted in a dimeric latent form. These

PDGF-isoforms contain a so-called CUB (Complement C1r/C1s, Uegf, Bmp1) domain at the N-

terminal region of these proteins. This CUB domain, mainly found in developmentally-

Page 22: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

21

regulated extracellular and plasma membrane-associated proteins, causes retention of PDGF-

CC and -DD in the extracellular matrix. For interaction with the PDGFR, extracellular activation

of the C- and D-chain is necessary and it requires proteolytic cleavage of the CUB-domain by

respectively tissue plasminogen activator or urokinase plasminogen activator 18,20. All five

dimeric PDGF-forms can be found in the extracellular space but also in intracellular

compartments, such as the endoplasmic reticulum and Golgi. There are even reports that

state that the C-chain can be present in the nucleus21.

PDGF exerts its effects via binding to one of the two structurally related PDGF-receptors, the

PDGF alpha receptor (PDGFαR; Mw = 123 kDa) and beta receptor (PDGFβR; Mw = 124 kDa)

that both belong to the class III receptor tyrosine kinases. Similar to the growth factor itself,

also the receptor exists in homo- (αα and ββ) and hetero- (αβ) dimeric forms, all containing

an extracellular part to which a particular PDGF-isoform binds, and an intracellular signaling

part. PDGF-A and C chains bind to the α-receptor, while B and D chains bind to the β-

receptor. Both receptors are predominantly expressed on the cell membrane with minor

expression in the lysosomes and even nucleus is reported17,19. In addition, PDGFαR was

detected as a soluble receptor in the extracellular space22.

Biological effects

PDGF has a clear role during embryonic development18, but after that, in normal healthy

tissues the activity and expression of the PDGF-system is very limited. During pathological

conditions, however, the PDGF-system is activated again and diseases that are associated

with overactivity of the PDGF-system can be divided in three groups: tumors, vascular

diseases and fibrotic diseases. In fibrotic livers, several inflammatory and fibrotic mediators,

such as interleukin-1β, tumor necrosis factor alpha (TNFα), TGFβ and fibroblast growth factor

(FGF) are released that can increase PDGF and its receptor expression. In addition, bacterial

endotoxin (lipopolysaccharide), which is released in the serum after intestinal leakage and

associated with liver fibrosis induction, can upregulate PDGFR-expressions in the liver23. The

upregulation of the PDGFβ-receptor in human cirrhotic livers was previously shown by Bansal

et al.24.

Various isoforms of PDGF are reported to be present in the fibrotic liver, and all these

isoforms have mitogenic and chemoattractive effects on mesenchymal cells. PDGF-BB is the

major stimulus for mesenchymal cell proliferation, and the PDGFβ-receptor is primarily

involved in this cellular effect of PDGF. Also, activation of the β-receptor stimulates

2

Page 23: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

22

chemotaxis, while activation of the α-receptor is associated with either stimulation or

inhibition of chemotaxis, depending on the mesenchymal cell type. PDGF is known for its

effects on intracellular actin reorganization resulting in stimulated cell migration, chemotaxis,

and contraction. There is, however, a difference between both PDGF-receptors regarding

their effects on the actin filament system. Whereas both receptors stimulate edge ruffling

and loss of stress fibers, only the β-receptor mediates the formation of circular actin

structures on the dorsal surface of the cell. Gu et al. nicely showed that these circular

structures are involved in rapid recruitment, internalization and membrane relocation of

surface integrins and in this way these circular structures may serve as indicators of cellular

transition from static to motile states25. Together with TGFβ, PDGF-BB also induces fibroblast

activation and differentiation, resulting in induction of α-smooth muscle actin (α-SMA)

expression in these cells19,26. Figure 1 gives an overview of the PDGF-BB producing and

responsive cells and its key functions.

The PDGF-isoform that is most prominent in liver fibrosis is PDGF-BB16,17. PDGF-BB is secreted

by both resident and infiltrating cells of the liver, such as platelets, infiltrating monocytes and

macrophages, activated Kupffer cells (M2-dominant type), hepatocytes, cholangiocytes, HSCs

and myofibroblasts, and it affects primarily HSCs and myofibroblasts. These latter cells display

a highly induced PDGFβ-receptor expression in mice and humans, and this expression can be

related to the proliferative and migratory activities of these fibroblasts in fibrosis. Of note,

activated HSCs and myofibroblasts produce PDGF-BB and thus create an autocrine profibrotic

loop without interference of other cells.

Although PDGF-production increases profoundly during the disease and thus local

concentrations are high, the amounts present in the circulation are generally very low. While

concentrations of only 17.5 ± 3.1 ng/ml were detectable in whole human blood, the amount

in plasma was undetectable27. The low concentrations in the circulation were due to a very

short plasma half-life of less than two minutes and due to a high binding affinity to several

proteins in plasma and extracellular spaces27. Examples of PDGF-binding proteins are its

soluble receptor, α2 macroglobulin, and PDGF-associated protein (PAP). Additionally, various

extracellular matrix proteins are able to bind PDGF to form a local storage pool. This implies

that the biological activities of PDGF can be modulated by binding to one of these

molecules28,29.

Page 24: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

23

Figure 1. Overview of PDGF-BB and IFNγ-producing cells and cells that respond to these mediators of

fibrogenesis, illustrating the versatility of both mediators.

PDGF-receptor expression

PDGFαR and PDGFβR have overlapping, but also distinct expression patterns and physiological

roles. Generally, PDGFαR signaling is important in embryonic development since it controls

gastrulation and the embryonic development of several organs such as lung, intestine, skin,

testis, kidneys, bones, and neuroprotective tissues. Some cell types only express the PDGFαR,

such as platelets and liver sinusoidal endothelial cells. PDGFβR signaling is recognized as an

essential regulator of early hematopoiesis and blood vessel formation. The classical target

cells for PDGF, fibroblasts and smooth muscle cells, express both α- and β-receptors, although

the expression of the PDGFβR is generally at a much higher level. In fibrotic livers, PDGFβR is

mainly expressed on the cell membrane of activated and transformed HSCs or myofibroblasts

in the collagenous fibrotic bands17,24,30. Borkham-Kamphorst et al. showed that the PDGFR in

the endothelium and hepatocytes are more of the α-type30. In addition, PDGFαR was stained

outside the area of CCl4-induced hepatocellular fibrosis and they speculated that this

increased expression could be related to hepatocyte survival. Furthermore, they found that

the upregulation of PDGFαR was accompanied by PDGF-A and PDGF-C staining in

regenerative hepatocytes after CCl4 intoxication.

How PDGF exactly binds to its receptor is subject of various studies. Insight into this

interaction can lead to the generation of PDGF-receptor antagonizing drugs or to drug

targeting compounds. Already in early 1990s, Clements et al. identified arginine27 and

isoleucine30 as the PDGF-B chain residues that mediate the binding of the growth factor to

2

Page 25: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

24

the β-receptor in rat and human cells31. Binding to the α-receptor requires another more

cationic part of the molecule (arginine159-lysine160-lysine161-lysine162), and both in the

PDGF-A and -B chain this sequence is present32. More recently, the interaction between the

growth factor and the receptor was studied in more detail with modern techniques like

crystallography, partly confirming the abovementioned regions in PDGF, but also pointing to

additional points of interaction with its receptors33,34.

PDGF-receptor signaling

After binding of PDGF to one of the receptor chains, dimerization of the receptor is induced

and this receptor dimer is then stabilized by the ligand. The subsequent interaction between

the cytoplasmic domains of the two receptor chains induces phosphorylation of intracellular

tyrosines leading to conformational changes in the PDGF-receptor protein and thus to

activation of the kinase domains of the receptor. Activation of src-homology-2 (SH2) or

phosphotyrosine-binding (PTB) domains will lead to activation of one of the downstream

cytoplasmic pathways. Although more pathways are described, activation of the MAPK/ERK

pathway, the AKT/PKB pathway, and the PKC pathway are the major routes19,35. The

MAPK/ERK pathway is also known as the Ras-Raf-MEK-ERK pathway, because the GTP-binding

protein Ras can activate mitogen-activated protein kinases (MAPK) such as extracellular

signal-regulated kinase (ERK). Phosphorylated ERK-1 and 2 form activated ERK dimers and

they translocate to the nucleus, where they can activate cellular processes linked to

stimulation of cell proliferation and chemotaxis. Secondly, phosphoinositol 3-kinase can also

be recruited to the activated PDGF-receptor leading to stimulation of cell proliferation and

migration via the AKT/PKB pathway. AKT however regulates multiple other biological

processes including glycogen metabolism and cell survival, and it is linked to promotion of

tumor growth. Activation of the third pathway involves phospholipase C-γ (PLC) recruitment

after stimulation of the PDGFR. This causes mobilization of cellular Ca2+ storage pools and

leads to activation of the intracellular signaling molecule protein kinase C (PKC). This route is

also associated with increased cell growth and motility.

Intracellular routing of PDGF and its receptor

Much is known about the intracellular signaling and biological effects of PDGFR. However, the

cellular kinetics of the PDGFR itself are less well studied and also the fate of the growth factor

and its receptor after binding of the ligand at the cell membrane leading to induction of

Page 26: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

25

intracellular signaling is poorly described. It is described that the PDGFβR is localized in

specialized structures of the cell membrane referred to as caveolae36-38. The morphology of

these structures is generated by the protein caveolin and they are enriched in

glycosphingolipids and cholesterol39. Sorkin et al. showed that induction of the tyrosine kinase

activity of the PDGFβR promotes ligand-induced internalization and subsequently degradation

of the ligand-receptor complex in endosomal and lysosomal compartments of the cell. They

showed that the receptors began to migrate out of caveolae after one hour of exposure to

PDGF and that the internalized PDGFβR retained kinase activity and thus could participate in

signaling considerable time after internalization36. The receptor can recycle back to the cell

membrane waiting for its next signal. The fate of the growth factor after dissociation from the

receptor in the endosomes is degradation in the lysosomal compartment, but other

intracellular routes are possible40 .

Interferon gamma

The involvement of interferons (IFNs) in the defense response against viral infections was first

described in 1957. Isaacs and Lindenmann reported that influenza-virus infected cells

produced a secreted factor, i.e. interferon, that caused a virus-resistant state in previously

uninfected cells41. The biochemical characterization of this activity was pursued for a long

time without success, and it was only in the late 1970’s that the interferon protein sequence

was ascertained42,43. Later, it appeared that the biological activities of interferons were much

broader and antifibrotic, antitumor, immunoregulatory and pro-inflammatory activities were

also assigned to the IFN-family44.

Interferons, belonging to the interferon/interleukin-10 cytokine family, can be divided in two

groups (type I and II). They are classified according to their receptor specificity and sequence

homology14. The type I interferons, also described as viral IFNs, include IFNα (of which

currently 17 subtypes are described), IFNβ, IFNω and IFNτ. These type I factors are produced

by almost all cell types. The type II interferons consist only of IFNγ, which shows structurally

little to no homology with type I IFN, and this protein is also described as immune IFN45. In

contrast to these type I IFNs, expression and secretion of IFNγ is not the result of the infection

itself, but occurs by activated immune cells after recognition of infected cells46. Therefore,

although IFNγ possesses antiviral activity, it functions primarily as an immune modulator

responsible for pathogen clearance, rather than an antiviral compound47,48.

2

Page 27: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

26

IFNγ is a non-covalently bound, homodimeric glycoprotein with subunits consisting of 146

amino acids (17.1 kDa)49,50, which are connected in an antiparallel fashion45. Intracellular

processing of this cytokine includes intensive glycosylation, giving rise to a mature form of the

molecule that exhibits a predominant molecular mass of 50 kDa51. Murine and human IFNγ

show approximately only 40% sequence homology at the protein level (www.uniprot.org).

The main cell types that produce IFNγ upon endogenous or exogenous stimulation in the

fibrotic liver include T lymphocytes and NK(T) cells, B cells, and other antigen presenting

cells52-58. NK cells were shown to be involved in the resolution of fibrosis in both mice and

humans by stimulating the production of IFNγ, but also via killing of activated HSCs59-61. The

production of IFNγ in turn increased the number of NK cells60. Emerging evidence suggests

that NK cells and IFNγ are very important in the negative regulation of liver fibrosis6. This was

demonstrated by the amelioration of liver fibrosis in vivo, after activation of NK cells by poly

I:C or treatment of rodents with IFNγ60,62-64.

Biological effects

Within the liver, IFNγ exerts a variety of sometimes even opposing effects. For example, IFNγ

is known to be a potent activator of macrophages, by stimulating them to kill phagocytosed

microbes and cancer cells and inducing the production of TNFα and interleukin-1265. IFNγ

induces an inflammatory (‘M1’) activation state of the hepatic macrophages (comprising

resident Kupffer cells and infiltrating monocytes and macrophages)66. The pro-inflammatory

factors produced via this route can activate the HSC to become a profibrotic myofibroblast. In

addition, IFNγ induces the expression of both MHC-I and -II in several cells44, promotes the

differentiation of T- and B lymphocytes and it activates neutrophils, all contributing to

promotion of fibrosis. It was also reported that IFNγ inhibits the proliferation of vascular

endothelial cells65. On the other hand, IFNγ has direct antifibrotic effects when it interacts

with HSCs. Increased production of IFNγ, as secreted by NK cells in fibrotic livers, is

accompanied by an increase in TNF-related apoptosis-inducing ligand (TRAIL) expression in NK

cells, which causes both induction of apoptosis and cell cycle arrest in HSCs60,64,67. The

interaction between the stimulatory receptor NKG2D on NK cells and its ligand retinoic acid

early inducible 1 (RAE1), expressed on activated hepatic stellate cells, leads to killing of

activated hepatic stellate cells by NK cells and thus to resolution of fibrosis60. A summary of

cells that produce and respond to IFNγ and its main functions is presented in figure 1.

In addition, IFNγ leads to reduced α-SMA, HSC proliferation, and collagen levels in

fibroblasts24,62. IFNγ antagonizes the profibrotic activities of TGFβ, most probably by

Page 28: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

27

intracellular induction of the expression of the antagonistic Smad7. Smad7 causes impairment

of the TGFβ-response, via blocking of the interaction of the transcription factor Smad3 with

the TGFβ-receptor68.

IFNγ-receptor expression

Like many other cytokines, the receptor for IFNγ consists of multiple distinct subunits: the α

chain (IFNγR1; 54.4 kDa), necessary for ligand binding and processing, and the β subunit

(IFNγR2; 37.8 kDa), needed to induce a biological response by triggering the intracellular

signal transduction cascade14,69. The complete receptor complex contains two IFNγR1 and

two IFNγR2 subunits and these four parts associate after ligand binding. In more detail, IFNγ

binds to one IFNγR1 and induces rapid dimerization of two IFNγR1 chains, thereby forming a

site that is recognized by the extracellular domain of IFNγR245. The IFNγR2 chain is generally

the limiting factor in IFNγ responsiveness, as the IFNγR1 chain is usually in excess45,70. The

IFNγR2 chain is constitutively expressed, but its expression level is tightly regulated according

to the state of cellular differentiation or activation. Of note, the binding of human and murine

IFN to their receptors is strictly species specific. They only induce an effect in species-matched

cells. Moreover, the interaction of IFNγ with its receptor is not inhibited by type I

interferons45.

IFNγR1 and R2 are ubiquitously expressed on the cell membrane of virtual all types of cells,

but most predominantly on cells of the immune system. In particular macrophages are

strongly positive for the IFNγR. Within the fibrotic liver, expression on infiltrated macrophages

and Kupffer cells is high, while other cells like HSCs and hepatocytes weakly express the

receptor 71. Bansal et al. previously showed the ubiquitous and increased expression of the

IFNγR1 in human cirrhotic livers as compared to healthy livers72. Although expression of the

IFNγR at the plasma membrane varies widely between tissues (200-25.000 sites/cell), no

direct correlation between the expression level and the magnitude of IFN-induced responses

in cells was found45.

IFNγ-receptor signaling: the classical model

In the case of IFNγ, a conformational change in the receptor cytoplasmic domain is induced

upon binding of IFNγ to the extracellular domains of two IFNγR1 chains. This causes

movement of the inactive Janus activated kinase 2 (Jak2) from IFNγR2 to IFNγR1 and

2

Page 29: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

28

subsequent autophosphorylation and activation73. After transphosphorylation and activation

of Jak1 by Jak2, the recruitment of signal transducer and activator of transcription 1 alpha

(Stat1α) is induced by phosphorylation of tyrosine440 residues of both IFNγR1 subunits,

forming a homodimeric complex of pStat1α69,74-76. This Stat1 pair dissociates from the

receptor upon phosphorylation at the tyrosine701-residue, followed by active nuclear

translocation via its nuclear localization sequence (NLS)73,74. In the nucleus, dimeric Stat1α

binds to the IFNγ-activated sequence (GAS element) of the IFNγ-promotor, thereby

determining specific gene activation of fibrosis-related genes like procollagen I & III and TGFβ,

but also transcription factors like interferon regulatory factor-1 (IRF-1)77-79. The cellular

response of IFNγ occurs within 15 to 30 minutes after treatment80. Binding of dimeric Stat1α

to the GAS element is regarded as validation of the classical model of cytokine signaling81.

To a lesser extent, IFNγ-signaling also produces heterodimeric complexes like Stat1:Stat2 and

heterotrimeric complexes such as Stat1:Stat1:IRF-9 and Stat1:Stat2:IRF-977,82-84. These

complexes are able to bind to IFN-stimulated response element (ISRE) promotor regions in

target genes to regulate transcription of for example inducible nitric oxide synthase (iNOS),

IRF-2 and IFNβ14.

IFNγ-receptor signaling: the non-canonical model

Several reasons led to the recognition that the classical model of cytokine signaling is an

oversimplified model. It was shown that upon IFNγ-stimulation besides the Jak/Stat pathway,

also other pathways were activated or interfered with the Jak/Stat signaling. Examples include

the activation of MAPK, phosphoinositide 3-kinase, Ca2+/calmodulin kinase II and nuclear

factor-κB85. Moreover, it was shown in multiple cell lines that besides activated Stat, also Jak

kinases were involved in the epigenetics of gene activation, indicating that Stat1 is not the

only key player in the IFNγ-signaling86. In addition to this, many ligands activate the same Stat

transcription factors, but have completely different effects in cells, tissues or organs87. In the

classical model of signaling of IFNγ, it is suggested that the Stat proteins possess intrinsic

NLSs, which are responsible for the nuclear translocation of Stat1 and subsequent IFNγ-

related gene activation88. Of note, in this classical model, the internalization of the ligand-

receptor complex does not play an active role in the signaling process87. Different to what was

assumed in the classical model, it appeared that ligand, receptor and activated Jaks and Stats

were directly involved in nuclear events leading to specific gene activation. Therefore, the

non-canonical model of IFNγ-signaling was developed, which shows large resemblance to that

of steroid hormone/steroid receptor signaling81.

Page 30: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

29

In this model, IFNγ binds to the extracellular domain of the receptor and moves to the

cytoplasmic side of the IFNγR1 domain, after which endocytosis of this complex is induced.

The movement of Jak2 to the R1 domain results in autoactivation of the Jaks. This causes

phosphorylation of R1 and eventually the recruitment and activation of Stat1α. The complex

of IFNγ, the IFNγR1, Stat1α, Jak1 and 2 is actively transported to the nucleus, a process which

is under the influence of the NLS of IFNγ14,81,87. The non-canonical model of IFNγ-signaling

formed the conceptual framework for the development of IFNγ-mimetics81.

Intracellular routing of IFNγ and its receptor

In contrast to the numerous publications on the signal transduction and biological effects of

IFNγ similar to the PDGFβ-receptor, there are relatively little data available on the trafficking

of the IFNγ-receptor. The interferon receptor has been localized in clathrin-coated pits (CCPs),

lipid rafts, and caveolae39.

CCPs are specialized regions in the plasma membrane that are engaged in the efficient

internalization of receptors from the cell surface. It is known that most transmembrane

receptors are endocytosed via clathrin-dependent endocytosis which involves CCPs89. Both

IFNγ and its receptor were shown to be associated with CCPs90,91. In general, receptors are

recruited into CCPs through direct binding of their cytoplasmic binding motifs to the adaptor-

related protein complex 2 (AP-2).

It is suggested that other endocytotic pathways also may participate in the endocytosis of the

IFNγ-receptor39. Lipid rafts are microdomains in the plasma membrane, which are enriched in

certain lipids, cholesterol and proteins, caused by redistribution within the lateral plane of

cellular membranes. It is hypothesized that rafts exist in a separate phase that diffuses

dynamically in a sea of poorly ordered lipids in the plasma membrane. By dynamic diffusion,

proteins can be in- or excluded in these rafts91. The IFNγ-receptor was shown to be present in

lipid rafts. Moreover, the downstream signaling molecules Jak1 and 2 and Stat1 were also

associated with lipid rafts in the plasma membrane92-94. Caveolae are considered a subtype of

lipid rafts39. It was shown that IFNγ and its receptor also localize in caveolae, albeit to a lesser

extent than in CCPs95.

When the ligand-receptor complex is internalized via endocytosis, it enters a cellular

compartment with acidic pH. Within this compartment, the ligand dissociates from the

receptor. Free IFNγ can then be trafficked to the lysosome, where it is degraded. In many

cells, such as fibroblasts and macrophages, the receptor eventually recycles back to the cell

2

Page 31: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

30

surface. In most cells, the intracellular pool is approximately 2-4 times larger as compared to

the receptors expressed at the cell surface96-100.

Mimetic IFNγ

It was previously shown that the N-terminal side of IFNγ plays an important role in the

recognition of the extracellular domain of its receptor, whereas the C-terminus is involved in

endocytosis by binding to the membrane proximal region of the cytoplasmic part of the

IFNγR1 subunit101. Therefore, it is suggested that the N-terminal region of IFNγ is involved in

species specific recognition and binding, while the C-terminal region may be involved in

binding of internalized IFNγ to the cytoplasmic receptor domain101. This led to the

development of a truncated peptide, which contains the C-terminal region of IFNγ, namely

mimetic IFNγ (IFNγ95-132 AA)101. It appeared that this mimetic peptide possesses similar

biological activities as its full length equivalent, despite the fact that it is not able to bind to

the extracellular domain of its receptor102. It was for instance shown that mimetic IFNγ

possesses IFNγ-agonistic activity without toxicity, such as the induction of MHCII expression

on macrophages101. Furthermore, mimetic IFNγ displayed antiviral activity in tissue cultures

and in mice103. In order to be able to deliver mimetic IFNγ into the cytoplasm, the peptide was

modified with a single N-terminal ϵ-palmitoyl-lysine residue. This was shown to be suitable to

allow penetration across the cell plasma membrane and subsequent cytoplasmic

delivery87,104, but the use of this palmitoyl-residue is unfavorable for in vivo use, because it

induces non-specific uptake in any cell it encounters. Similar to IFNγ, mimetic IFNγ forms a

complex with IFNγR1, Stat1α and Jak1 and 2 and translocates to the nucleus as directed by its

NLS14,81,87. Advantages of mimetic proteins in general are their versatility, the high biological

activity and specificity, and their low toxicity103. There are already several peptide mimetics

on the market to treat a broad variety of diseases. One example is the tripeptide mimetic

boceprevir for the treatment of hepatitis C viral infections105. The therapeutic applications of

mimetic IFN are nowadays explored106.

Antifibrotic compounds

Although in the past decade some progress was made, the development of antifibrotic drugs

for the treatment of liver cirrhosis is still in its infancy. Major steps forwards in the treatment

of liver diseases were made at the level of eradication of the inciting stimulus. The high

Page 32: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

31

successes obtained with anti-HCV drugs that effectively clear the virus and thus reverse the

chronic pathology, resulted also in cases in which regression of the fibrosis in the livers of

these patients was ascertained107. This contributed to the believe that liver fibrosis even in

advanced state is reversible and treatable. Parallel to this, experimental therapeutic

interventions directly aiming at fibrogenic key cells and processes emerged4,108. In 2005,

Pinzani et al. showed an impressive list with all possible compounds under (pre)clinical

investigation for the treatment of liver fibrosis109. Later on, more reviews with promising

antifibrotic drugs appeared110-112. Currently 370 clinical trials are listed at

www.clinicaltrials.gov, illustrating the upcoming incidence of clinical trials ongoing in liver

cirrhosis, and showing that the pharmaceutical industry is interested to invest in drugs for the

treatment of liver cirrhosis, despite the expected long-term trials to assess efficacy in this

chronic, slow progressing disease. Of note, about 90% of the clinical trials are still dealing with

diagnostics improvements, treatment of cirrhosis-associated complications, or with the

application and evaluation of direct antiviral agents. This means that only a minority of the

clinical studies really tests novel antifibrotic compounds108. Remarkably, there are hardly any

completed or ongoing clinical trials in patients with liver cirrhosis testing compounds that

target PDGF and IFNγ pathways. Only in 2002, a study was started in which IFNγ1b was tested

for safety and efficacy in HCV patients with liver fibrosis or cirrhosis. However, although IFNγ

was well tolerated, the drug was abandoned and not approved because of lack of antifibrotic

activity113,114. With regard to intervention at the PDGF level, several experimental approaches

aiming at PDGF were described in literature. Examples include inhibition of the PDGF-B

chain115,116, inhibition of the PDGFβ-receptor expressions117,118, and inhibition of the PDGFβ-

receptor signaling119,120. However, none of these compounds were found in the clinical trial

database on liver fibrosis or cirrhosis.

New targets for direct-acting antifibrotic compounds were identified in the past decades, due

to a better understanding of the pathological process at molecular level. This includes the

specific roles of the different (hepatic) cell types, the mediators involved in the

communication between these cells and their intracellular signaling pathways, and the

unravelment of the complex process of fibrotic extracellular matrix deposition, during which

matrix degrading matrix metalloproteinases (MMPs) and inhibitors of these

metalloproteinases (TIMPs) parallel the enhanced production of matrix components. This

basic knowledge contributed to the generation of various antifibrotic compounds that can be

tested in patients with liver cirrhosis. One of the current promising ongoing clinical trials deals

with the application of mesenchymal stem cells to treat liver cirrhosis121. Mesenchymal stem

cells have shown to have the ability to effectively reduce liver fibrosis and improve liver

function. The effects are based on various activities of these mesenchymal stem cells. First,

2

Page 33: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

32

these stem cells can differentiate into hepatocytes, allowing the replacement of damaged

hepatocytes, and they can promote regeneration of residual hepatocytes. In addition,

mesenchymal stem cells also affect HSCs and possess immunomodulatory properties, all

adding to their antifibrotic potential. However, some limitations need to be considered. Over

the past few years, concerns have been raised about its long-term effectiveness, the potential

tumorigenic risk, and the lack of standardized protocols for mesenchymal stem cell

transplantation.

The development of cell-specific antifibrotic drugs is another approach that seems

promising122-124. Fibroferon, based upon the structure of both PDGF-BB and IFNγ, is one of the

latest compounds in this context. Fibroferon is an IFNγ-derived mimetic peptide that is

specifically delivered to activated HSCs and myofibroblasts. We used the highly upregulated

PDGFβR as a target receptor for the HSC/myofibroblast-directed drug targeting approach in

fibrosis. We were the first to be able to deliver drugs to HSCs using a series of modified

proteins, and one of these drug-carriers was directed at the PDGFβR9,125,126. Nowadays,

several groups have developed or applied cell-specific proteins to HSCs124,127-129, recently

reviewed by Poelstra et al.122. In the following paragraphs we will delineate our approach and

show, as depicted in figure 2, how this has led to the development of the promising

antifibrotic PDGFβR-targeted interferon, referred to as Fibroferon.

PDGFβ-receptor recognizing peptide

We designed a cyclic peptide (pPB) that mimics the binding site of PDGF-BB to its endogenous

receptor. Similar to PDGF-BB itself, the arginine and isoleucine residues and their adjacent

amino acids represent the PDGFβ-receptor binding moieties in the pPB peptide (C*SRNLIDC*,

where the C* denotes the cyclizing cysteine residues)31. The peptide was designed in a cyclic

conformation, because earlier observations revealed that cyclic peptides have stronger

interactions with their receptors and have increased stability in plasma as compared to their

linear analogues130,131.

At first, a number of pPB peptides were coupled to human serum albumin (HSA) yielding the

HSC-specific carrier pPB-HSA (see Fig. 2)9. The PDGFβ-receptor requires a dimeric interaction

for ligand binding and receptor activation132, and therefore multiple pPB moieties (on average

10 molecules of pPB per 1 molecule HSA at different distances from each other) were

necessary to obtain optimal receptor binding.

Page 34: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

33

Figure 2. Design of constructs directed at the PDGFβ-receptor in time, finally leading to the development of

Fibroferon. The first PDGFβR-targeted product was the carrier pPB-HSA in which several (on average n=10)

groups of pPB were randomly attached to human serum albumin (HSA), in order to allow multivalent interaction

with the PDGFR. Several other constructs, comprising a therapeutic entity (drug or protein) coupled to this

carrier, were subsequently developed and tested in vivo. In the final step, the structure was simplified by

omitting the albumin protein core and applying low molecular weight PEG to couple truncated IFNγ to BipPB, to

create divalent interaction of truncated IFNγ with the PDGFβR.

It was confirmed in vitro that pPB-HSA bound to the PDGFβ-receptor on fibroblast cell lines

and on primary rat HSCs. This binding was inhibited by PDGF-BB but not by PDGF-AA.

Coupling of pPB-peptides to a macromolecule like HSA was necessary for a competitive effect

with PDGF-BB, since free pPB did not displace PDGF-BB from its receptor. PDGFβR-induced

effects were not found after incubation of fibroblasts with pPB-HSA. Based on these

observations, it was hypothesized that coupling of pPB to a macromolecule such as HSA

enabled a multivalent interaction with the PDGFβR, but no receptor activation was induced by

pPB-based molecules. In vivo, pPB-HSA was shown to accumulate in HSCs in fibrotic human,

mouse and rat livers9; cells expressing the highest PDGFβR. These results formed the basis for

the development of HSC-selective targeting of drugs applying pPB-HSA as drug-carrier and for

example the antifibrotic agents IFNγ133 and 15d-prostaglandin-J2134 were evaluated. This is

particularly relevant for IFNγ, which has a therapeutic application in a wide variety of

2

Page 35: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

34

immunological, viral and neoplastic diseases, and where high percentages, up to 100%, of

patients suffer from general side effects such as headache, malaise and fever44. A

fundamental question is however whether a cytokine can be delivered to another target

receptor, without losing the biological effects elicited by its endogenous receptor.

pPB-based delivery of IFNγ to HSCs for the treatment of liver fibrosis

We further developed HSC-selective compounds containing the cyclic peptide pPB and

covalently conjugated this peptide to the antifibrotic compound IFNγ (see Fig. 2). This led to

various compounds either with (pPB-PEG-IFNγ and pPB-HSA-PEG-IFNγ) or without (pPB-IFNγ)

polyethylene glycol (PEG) linker in between24,133. The choice for a distinct PEG-linker was

based on previous experiments, showing that PEGylation of IFNγ with different sizes of PEG-

molecules (5, 10 and 20 kDa) improved the pharmacokinetic profile, liver uptake and

antifibrotic effects of IFNγ, with the smallest PEG molecule as most favorable72. Additionally,

improved conformational flexibility of the pPB peptide using a PEG-spacer allowed improved

presentation of pPB to the PDGFβ-receptor72. Our conjugates were shown to bind to the

PDGFβ-receptor on fibroblasts and HSCs and inhibited their activation in vitro, as shown by a

reduction in collagen type I and α-SMA expression. In addition, the conjugates were shown to

inhibit PDGF-induced cell proliferation. A remarkable finding in these studies was that the

specific binding to the PDGFβ-receptor and subsequent biological effects of the pPB-modified

IFNγ-constructs was not species-specific anymore in contrast to IFNγ. This was demonstrated

by the absence of both binding and an antifibrotic effect of mouse IFNγ in human cell

cultures, whereas mouse IFNγ conjugated to pPB did bind to human cells and showed

biological activity24,133. It was therefore proposed that following binding to the PDGFβ-

receptor, the complex was internalized, degraded and IFNγ (or its signaling moiety) was

released into the cytoplasm leading to IFNγ-mediated effects135.

The targeted constructs (pPB-PEG-IFNγ, pPB-HSA-PEG-IFNγ and pPB-IFNγ) co-localized with

desmin-positive PDGFβR-expressing HSCs in the liver. In addition, they strongly inhibited

fibrogenesis and induced fibrolysis in a two week treatment study in 8 weeks old CCl4 mice, as

demonstrated by attenuated collagen type I and α-SMA deposition. Additionally, the balance

between collagen-degrading MMP-13 and its endogenous inhibitor TIMP-1 was shown to be

shifted towards a fibrolytic state, characterized by a reduced TIMP-1 expression. This effect

was not seen in the animals treated with untargeted IFNγ. Furthermore, in contrast to

untargeted IFNγ, the targeted IFNγ-conjugates did not induce unwanted IFNγ-related side

effects. In these mice treated with targeted IFNγ systemic inflammation, hyperthermia,

Page 36: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

35

elevated plasma triglyceride levels and neurotropic effects, as demonstrated by amongst

others a reduced release of pro-inflammatory cytokines (IL-1β) were absent. Moreover,

targeted IFNγ lowered MHC-II expression in the brain as compared to native IFNγ24,133, which

makes this compound attractive for long-term treatment.

Redirection of IFNγ to myofibroblasts in the fibrotic liver proves to be a promising approach.

We pursued this by creating a smaller, more simplified construct to facilitate clinical

application (see Fig. 2)24,133. This minimized structure combines three basic elements, i.e.

IFNγ-activity, PEG and a PDGFβR-binding peptide106. In this truncated IFNγ-structure (i.e.

mimetic IFNγ), the extracellular IFNγ-receptor binding sequence was removed, yet the NLS

accommodating the biological effects of IFNγ was retained. In this way, interaction with the

ubiquitously expressed IFNγ-receptor at the membrane of the cells was prevented, leaving

only an interaction with the intracellular IFNγR136,137. This mimetic IFNγ-structure was

redirected to the PDGFβ-receptor by coupling it to PDGFβR-recognizing peptides. We also

minimized this entity by incorporating only two cyclic PDGFβ-receptor binding peptides

(BipPB) at a specific distinct distance from each other to allow dimeric ligand-receptor

interaction. The PEG-linker was minimized to only 2 kDa and BipPB, PEG and mimetic IFNγ

were subsequently coupled in a 1:1:1 ratio, yielding an 8.8 kDa construct (referred to as

mimγ-BipPB, BOT191, or Fibroferon). Similar to pPB-PEG-IFNγ, Fibroferon markedly inhibited

both early and advanced CCl4 induced liver fibrosis in mice and significantly reduced IFNγ-

related side effects106. The advantages of this targeted mimetic IFNγ-conjugate are the in vivo

stability, the cell selectivity, dissociation of effects, the loss of species specificity, the minimal

size and less complexity, making it a better-defined product. This all provides Fibroferon with

clear advantages above the therapeutic use of native IFNγ as an antifibrotic compound.

pPB-based HSC-selective drug carriers for the treatment of other diseases

In addition to the demonstrated antifibrotic properties of re-directed truncated IFNγ in a

mouse liver fibrosis model, antifibrotic effects of pPB-PEG-IFNγ were also found in the

unilateral ureteral obstruction (UUO) mouse model for renal fibrosis. In this model, pPB-PEG-

IFNγ specifically accumulated in PDGFβR-overexpressing interstitial myofibroblasts in the

kidney. Treatment of mice with pPB-PEG-IFNγ significantly attenuated collagen I, fibronectin

and α-SMA mRNA levels and protein expression in fibrotic kidneys similar to the liver fibrosis

studies. Compared to vehicle treatment, pPB-PEG-IFNγ protected the tubular morphology,

significantly attenuated interstitial T-cell infiltration and reduced the formation of lymphatic

vessels, without affecting the peritubular capillary density. In addition, pPB-PEG-IFNγ reduced

2

Page 37: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

36

IFNγ-mediated side effects, as shown by reduced MHC-II mRNA expression in the brain135,

which was applied as a marker for off-target effects on macrophages.

pPB-HSA-IFNγ was also shown to be effective as antitumor agent138, because the PDGFβ-

receptor is also upregulated in various types of cancer139,140. The PDGFβ-receptor was

immunohistochemically shown to be abundantly expressed in stromal cells and pericytes of

neoplastic human diseases, like colorectal, pancreatic and breast cancer19. Stromal cells,

including tumor-associated fibroblasts, endothelial cells, vascular pericytes and infiltrating

inflammatory cells, are increasingly recognized as key players in the induction of growth and

progression of tumors141,142. Activation and migration of fibroblasts and tube formation in

endothelial cells induced by fibroblasts was inhibited by pPB-HSA-IFNγ, suggesting an effect

on angiogenic processes. In C26 colon carcinoma tumor-bearing mice, pPB-HSA-IFNγ

specifically accumulated into PDGFβ-receptor expressing tumor stromal fibroblasts and

pericytes and inhibited the tumor growth. Treatment of these mice with pPB-HSA-IFNγ was

associated with a significant inhibition of angiogenesis, as reflected by reduced α-SMA and

CD31 stainings138.

Other drugs were also delivered to the PDGFβ-receptor using the cyclic peptide pPB as a

targeting device. For example, the anticancer agent doxorubicin was coupled to pPB-HSA via

an acid-sensitive hydrazine linkage for delivery to the PDGFβ-receptor on stromal cells. The

targeted construct was shown to significantly reduce tumor growth with high response rate,

as compared to the untargeted doxorubicin. Therefore, the delivery of anticancer drugs to

these stromal cells, nurturing the neighboring tumor cells, is a novel and maybe promising

approach to treat cancers effectively143.

These studies suggests that the PDGFβ-receptor targeted constructs not only exert

therapeutic effects in liver fibrosis24,106,133, but also in other diseases associated with high

PDGFβ-receptor expression, such as kidney fibrosis135 and some types of cancer138,143.

Conclusion

The ever increasing knowledge on molecular mechanisms and mediators that orchestrate the

progression of liver fibrosis can be used to design new therapeutic compounds against this

lethal disease. The fundamental processes that underlie this disease are similar in many other

fibrotic and sclerotic diseases, including stromal cancers, which broadens the scope of

applications of such novel compounds. In the past decades, the key cells and key mediators

that stimulate and inhibit liver fibrogenesis have been identified and we used this knowledge

Page 38: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

37

to create a cell-specific chimeric compound which combines the best of two important

mediators: the high disease-induced receptor expression of one mediator (PDGFR) is used to

deliver another mediator (IFNγ), endowed with antifibrotic activities, to the HSC, yielding

Fibroferon. Further studies on the intracellular trafficking of this compound that induces the

activation of the IFNγ-signaling cascade via binding to the PDGFR are in progress. This

chimeric molecule might be one of the next generation antifibrotic therapeutics. These

studies also illustrate the surprising adaptability of biological compounds that are the natural

inhibitors of this chronic disease.

2

Page 39: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

38

References

1. Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence and epidemiology of

hepatocellular carcinoma: A global and regional perspective. Oncologist. 2010;15 Suppl

4:5-13.

2. Bruix J, Han KH, Gores G, Llovet JM, Mazzaferro V. Liver cancer: Approaching a

personalized care. J Hepatol. 2015;62(1 Suppl):S144-56.

3. Friedman SL. Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the

liver. Physiol Rev. 2007;88(1):125-172.

4. Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: Concept to

treatment. J Hepatol. 2015;62(1S):S15-S24.

5. Wynn TA, Barron L. Macrophages: Master regulators of inflammation and fibrosis. Semin

Liver Dis. 2010;30(3):245-257.

6. Jeong WI, Park O, Suh YG, et al. Suppression of innate immunity (natural killer

cell/interferon-gamma) in the advanced stages of liver fibrosis in mice. Hepatology.

2011;53(4):1342-1351.

7. Seki E, Schwabe RF. Hepatic inflammation and fibrosis: Functional links and key

pathways. Hepatology. 2015;61(3):1066-1079.

8. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: Immune

regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181-194.

9. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

10. Baer HU, Friess H, Abou-Shady M, et al. Transforming growth factor betas and their

receptors in human liver cirrhosis. Eur J Gastroenterol Hepatol. 1998;10(12):1031-1039.

11. Bedossa P, Peltier E, Terris B, Franco D, Poynard T. Transforming growth factor-beta 1

(TGF-beta 1) and TGF-beta 1 receptors in normal, cirrhotic, and neoplastic human livers.

Hepatology. 1995;21(3):760-766.

12. Gratchev A, Kzhyshkowska J, Kannookadan S, et al. Activation of a TGF-beta-specific

multistep gene expression program in mature macrophages requires glucocorticoid-

mediated surface expression of TGF-beta receptor II. J Immunol. 2008;180(10):6553-

6565.

13. Tilg H, Kaser A, Moschen AR. How to modulate inflammatory cytokines in liver diseases.

Liver Int. 2006;26(9):1029-1039.

14. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: An overview of signals,

mechanisms and functions. J Leukoc Biol. 2004;75(2):163-189.

Page 40: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

39

15. Paul D, Lipton A, Klinger I. Serum factor requirements of normal and simian virus 40-

transformed 3T3 mouse fibroplasts. Proc Natl Acad Sci U S A. 1971;68(3):645-652.

16. Pinzani M, Gesualdo L, Sabbah GM, Abboud HE. Effects of platelet-derived growth factor

and other polypeptide mitogens on DNA synthesis and growth of cultured rat liver fat-

storing cells. J Clin Invest. 1989;84(6):1786-1793.

17. Pinzani M, Milani S, Herbst H, et al. Expression of platelet-derived growth factor and its

receptors in normal human liver and during active hepatic fibrogenesis. Am J Pathol.

1996;148(3):785-800.

18. Betsholtz C. Biology of platelet-derived growth factors in development. Birth Defects Res

C Embryo Today. 2003;69(4):272-285.

19. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived

growth factor. Physiol Rev. 1999;79(4):1283-1316.

20. Reigstad LJ, Varhaug JE, Lillehaug JR. Structural and functional specificities of PDGF-C and

PDGF-D, the novel members of the platelet-derived growth factors family. FEBS J.

2005;272(22):5723-5741.

21. Reigstad LJ, Martinez A, Varhaug JE, Lillehaug JR. Nuclear localisation of endogenous

SUMO-1-modified PDGF-C in human thyroid tissue and cell lines. Exp Cell Res.

2006;312(6):782-795.

22. Tiesman J, Hart CE. Identification of a soluble receptor for platelet-derived growth factor

in cell-conditioned medium and human plasma. J Biol Chem. 1993;268(13):9621-9628.

23. Kim TH, Moon JH, Savard CE, Kuver R, Lee SP. Effects of lipopolysaccharide on platelet-

derived growth factor isoform and receptor expression in cultured rat common bile duct

fibroblasts and cholangiocytes. J Gastroenterol Hepatol. 2009;24(7):1218-1225.

24. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted

interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology. 2011;54(2):586-596.

25. Gu Z, Noss EH, Hsu VW, Brenner MB. Integrins traffic rapidly via circular dorsal ruffles

and macropinocytosis during stimulated cell migration. J Cell Biol. 2011;193(1):61-70.

26. Liu Z, Liu H, Jiang J, et al. PDGF-BB and bFGF ameliorate radiation-induced intestinal

progenitor/stem cell apoptosis via akt/p53 signaling in mice. Am J Physiol Gastrointest

Liver Physiol. 2014;307(11):G1033-43.

27. Bowen-Pope DF, Malpass TW, Foster DM, Ross R. Platelet-derived growth factor in vivo:

Levels, activity, and rate of clearance. Blood. 1984;64(2):458-469.

28. Zafiropoulos A, Fthenou E, Chatzinikolaou G, Tzanakakis GN. Glycosaminoglycans and

PDGF signaling in mesenchymal cells. Connect Tissue Res. 2008;49(3):153-156.

29. Gonias SL, Carmichael A, Mettenburg JM, Roadcap DW, Irvin WP, Webb DJ. Identical or

overlapping sequences in the primary structure of human alpha(2)-macroglobulin are

2

Page 41: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

40

responsible for the binding of nerve growth factor-beta, platelet-derived growth factor-

BB, and transforming growth factor-beta. J Biol Chem. 2000;275(8):5826-5831.

30. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

31. Clements JM, Bawden LJ, Bloxidge RE, et al. Two PDGF-B chain residues, arginine 27 and

isoleucine 30, mediate receptor binding and activation. EMBO J. 1991;10(13):4113-4120.

32. Fenstermaker RA, Poptic E, Bonfield TL, et al. A cationic region of the platelet-derived

growth factor (PDGF) A-chain (Arg159-Lys160-Lys161) is required for receptor binding

and mitogenic activity of the PDGF-AA homodimer. J Biol Chem. 1993;268(14):10482-

10489.

33. Chen PH, Chen X, He X. Platelet-derived growth factors and their receptors: Structural

and functional perspectives. Biochim Biophys Acta. 2013;1834(10):2176-2186.

34. Yang Y, Yuzawa S, Schlessinger J. Contacts between membrane proximal regions of the

PDGF receptor ectodomain are required for receptor activation but not for receptor

dimerization. Proc Natl Acad Sci U S A. 2008;105(22):7681-7686.

35. Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci.

2002;7:d1720-6.

36. Sorkin A, Eriksson A, Heldin CH, Westermark B, Claesson-Welsh L. Pool of ligand-bound

platelet-derived growth factor beta-receptors remain activated and tyrosine

phosphorylated after internalization. J Cell Physiol. 1993;156(2):373-382.

37. Liu P, Ying Y, Ko YG, Anderson RG. Localization of platelet-derived growth factor-

stimulated phosphorylation cascade to caveolae. J Biol Chem. 1996;271(17):10299-

10303.

38. Liu P, Ying Y, Anderson RG. Platelet-derived growth factor activates mitogen-activated

protein kinase in isolated caveolae. Proc Natl Acad Sci U S A. 1997;94(25):13666-13670.

39. Claudinon J, Monier MN, Lamaze C. Interfering with interferon receptor sorting and

trafficking: Impact on signaling. Biochimie. 2007;89(6-7):735-743.

40. Gill GN. A pit stop at the ER. Science. 2002;295(5560):1654-1655.

41. Isaacs A, Lindenmann J. Virus interference. I. the interferon. by A. isaacs and J.

lindenmann, 1957. J Interferon Res. 1987;7(5):429-438.

42. Rubinstein M, Rubinstein S, Familletti PC, Miller RS, Waldman AA, Pestka S. Human

leukocyte interferon: Production, purification to homogeneity, and initial

characterization. Proc Natl Acad Sci U S A. 1979;76(2):640-644.

43. Zoon KC, Smith ME, Bridgen PJ, zur Nedden D, Anfinsen CB. Purification and partial

characterization of human lymphoblast interferon. Proc Natl Acad Sci U S A.

1979;76(11):5601-5605.

Page 42: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

41

44. Younes HM, Amsden BG. Interferon-gamma therapy: Evaluation of routes of

administration and delivery systems. J Pharm Sci. 2002;91(1):2-17.

45. Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: A paradigm for cytokine

receptor signaling. Annu Rev Immunol. 1997;15:563-591.

46. Young HA, Hardy KJ. Interferon-gamma: Producer cells, activation stimuli, and molecular

genetic regulation. Pharmacol Ther. 1990;45(1):137-151.

47. Farrar MA, Schreiber RD. The molecular cell biology of interferon-gamma and its

receptor. Annu Rev Immunol. 1993;11:571-611.

48. Billiau A. Interferon-gamma: Biology and role in pathogenesis. Adv Immunol. 1996;62:61-

130.

49. Gray PW, Leung DW, Pennica D, et al. Expression of human immune interferon cDNA in

E. coli and monkey cells. Nature. 1982;295(5849):503-508.

50. Gray PW, Goeddel DV. Cloning and expression of murine immune interferon cDNA. Proc

Natl Acad Sci U S A. 1983;80(19):5842-5846.

51. Kelker HC, Le J, Rubin BY, Yip YK, Nagler C, Vilcek J. Three molecular weight forms of

natural human interferon-gamma revealed by immunoprecipitation with monoclonal

antibody. J Biol Chem. 1984;259(7):4301-4304.

52. Carnaud C, Lee D, Donnars O, et al. Cutting edge: Cross-talk between cells of the innate

immune system: NKT cells rapidly activate NK cells. J Immunol. 1999;163(9):4647-4650.

53. Frucht DM, Fukao T, Bogdan C, Schindler H, O'Shea JJ, Koyasu S. IFN-gamma production

by antigen-presenting cells: Mechanisms emerge. Trends Immunol. 2001;22(10):556-

560.

54. Gessani S, Belardelli F. IFN-gamma expression in macrophages and its possible biological

significance. Cytokine Growth Factor Rev. 1998;9(2):117-123.

55. Yoshimoto T, Takeda K, Tanaka T, et al. IL-12 up-regulates IL-18 receptor expression on T

cells, Th1 cells, and B cells: Synergism with IL-18 for IFN-gamma production. J Immunol.

1998;161(7):3400-3407.

56. Flaishon L, Hershkoviz R, Lantner F, et al. Autocrine secretion of interferon gamma

negatively regulates homing of immature B cells. J Exp Med. 2000;192(9):1381-1388.

57. Harris DP, Haynes L, Sayles PC, et al. Reciprocal regulation of polarized cytokine

production by effector B and T cells. Nat Immunol. 2000;1(6):475-482.

58. Baron S, Tyring SK, Fleischmann WR,Jr, et al. The interferons. mechanisms of action and

clinical applications. JAMA. 1991;266(10):1375-1383.

59. Muhanna N, Abu Tair L, Doron S, et al. Amelioration of hepatic fibrosis by NK cell

activation. Gut. 2011;60(1):90-98.

60. Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer cells ameliorate liver

fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-

2

Page 43: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

42

related apoptosis-inducing ligand-dependent manners. Gastroenterology.

2006;130(2):435-452.

61. Melhem A, Muhanna N, Bishara A, et al. Anti-fibrotic activity of NK cells in experimental

liver injury through killing of activated HSC. J Hepatol. 2006;45(1):60-71.

62. Baroni GS, D'Ambrosio L, Curto P, et al. Interferon gamma decreases hepatic stellate cell

activation and extracellular matrix deposition in rat liver fibrosis. Hepatology.

1996;23(5):1189-1199.

63. Rockey DC, Chung JJ. Interferon gamma inhibits lipocyte activation and extracellular

matrix mRNA expression during experimental liver injury: Implications for treatment of

hepatic fibrosis. J Investig Med. 1994;42(4):660-670.

64. Jeong WI, Park O, Radaeva S, Gao B. STAT1 inhibits liver fibrosis in mice by inhibiting

stellate cell proliferation and stimulating NK cell cytotoxicity. Hepatology.

2006;44(6):1441-1451.

65. Arai KI, Lee F, Miyajima A, Miyatake S, Arai N, Yokota T. Cytokines: Coordinators of

immune and inflammatory responses. Annu Rev Biochem. 1990;59:783-836.

66. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization.

Front Biosci. 2008;13:453-461.

67. Jeong WI, Park O, Gao B. Abrogation of the antifibrotic effects of natural killer

cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis.

Gastroenterology. 2008;134(1):248-258.

68. Ulloa L, Doody J, Massague J. Inhibition of transforming growth factor-beta/SMAD

signalling by the interferon-gamma/STAT pathway. Nature. 1999;397(6721):710-713.

69. Greenlund AC, Farrar MA, Viviano BL, Schreiber RD. Ligand-induced IFN gamma receptor

tyrosine phosphorylation couples the receptor to its signal transduction system (p91).

EMBO J. 1994;13(7):1591-1600.

70. Bernabei P, Coccia EM, Rigamonti L, et al. Interferon-gamma receptor 2 expression as

the deciding factor in human T, B, and myeloid cell proliferation or death. J Leukoc Biol.

2001;70(6):950-960.

71. Uhlen M, Fagerberg L, Hallstrom BM, et al. Proteomics. tissue-based map of the human

proteome. Science. 2015;347(6220):1260419.

72. Bansal R, Post E, Proost JH, de Jager-Krikken A, Poelstra K, Prakash J. PEGylation

improves pharmacokinetic profile, liver uptake and efficacy of interferon gamma in liver

fibrosis. J Control Release. 2011;154(3):233-240.

73. Briscoe J, Rogers NC, Witthuhn BA, et al. Kinase-negative mutants of JAK1 can sustain

interferon-gamma-inducible gene expression but not an antiviral state. EMBO J.

1996;15(4):799-809.

Page 44: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

43

74. Greenlund AC, Morales MO, Viviano BL, Yan H, Krolewski J, Schreiber RD. Stat

recruitment by tyrosine-phosphorylated cytokine receptors: An ordered reversible

affinity-driven process. Immunity. 1995;2(6):677-687.

75. Heim MH, Kerr IM, Stark GR, Darnell JE,Jr. Contribution of STAT SH2 groups to specific

interferon signaling by the jak-STAT pathway. Science. 1995;267(5202):1347-1349.

76. Igarashi K, Garotta G, Ozmen L, et al. Interferon-gamma induces tyrosine

phosphorylation of interferon-gamma receptor and regulated association of protein

tyrosine kinases, Jak1 and Jak2, with its receptor. J Biol Chem. 1994;269(20):14333-

14336.

77. Darnell JE,Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in

response to IFNs and other extracellular signaling proteins. Science.

1994;264(5164):1415-1421.

78. Schindler C, Darnell JE,Jr. Transcriptional responses to polypeptide ligands: The JAK-STAT

pathway. Annu Rev Biochem. 1995;64:621-651.

79. Ramana CV, Chatterjee-Kishore M, Nguyen H, Stark GR. Complex roles of Stat1 in

regulating gene expression. Oncogene. 2000;19(21):2619-2627.

80. Kerr IM, Stark GR. The control of interferon-inducible gene expression. FEBS Lett.

1991;285(2):194-198.

81. Johnson HM, Noon-Song EN, Dabelic R, Ahmed CM. IFN signaling: How a non-canonical

model led to the development of IFN mimetics. Front Immunol. 2013;4:202.

82. Matsumoto M, Tanaka N, Harada H, et al. Activation of the transcription factor ISGF3 by

interferon-gamma. Biol Chem. 1999;380(6):699-703.

83. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to

interferons. Annu Rev Biochem. 1998;67:227-264.

84. Bluyssen AR, Durbin JE, Levy DE. ISGF3 gamma p48, a specificity switch for interferon

activated transcription factors. Cytokine Growth Factor Rev. 1996;7(1):11-17.

85. Gough DJ, Levy DE, Johnstone RW, Clarke CJ. IFNgamma signaling-does it mean JAK-

STAT? Cytokine Growth Factor Rev. 2008;19(5-6):383-394.

86. Dawson MA, Bannister AJ, Gottgens B, et al. JAK2 phosphorylates histone H3Y41 and

excludes HP1alpha from chromatin. Nature. 2009;461(7265):819-822.

87. Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy

of poxvirus infection and multiple sclerosis: An example of occam's broom? JAKSTAT.

2013;2(4):e26227.

88. Reich NC, Liu L. Tracking STAT nuclear traffic. Nat Rev Immunol. 2006;6(8):602-612.

89. Brodsky FM, Chen CY, Knuehl C, Towler MC, Wakeham DE. Biological basket weaving:

Formation and function of clathrin-coated vesicles. Annu Rev Cell Dev Biol. 2001;17:517-

568.

2

Page 45: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

44

90. Filgueira L, Groscurth P, Aguet M. Binding and internalization of gold-labeled IFN-gamma

by human raji cells. J Immunol. 1989;142(10):3436-3439.

91. Sharma P, Varma R, Sarasij RC, et al. Nanoscale organization of multiple GPI-anchored

proteins in living cell membranes. Cell. 2004;116(4):577-589.

92. Takaoka A, Mitani Y, Suemori H, et al. Cross talk between interferon-gamma and -

alpha/beta signaling components in caveolar membrane domains. Science.

2000;288(5475):2357-2360.

93. Truchet S, Wietzerbin J, Debey P. Mouse oocytes and preimplantation embryos bear the

two sub-units of interferon-gamma receptor. Mol Reprod Dev. 2001;60(3):319-330.

94. Subramaniam PS, Johnson HM. Lipid microdomains are required sites for the selective

endocytosis and nuclear translocation of IFN-gamma, its receptor chain IFN-gamma

receptor-1, and the phosphorylation and nuclear translocation of STAT1alpha. J

Immunol. 2002;169(4):1959-1969.

95. Sadir R, Lambert A, Lortat-Jacob H, Morel G. Caveolae and clathrin-coated vesicles: Two

possible internalization pathways for IFN-gamma and IFN-gamma receptor. Cytokine.

2001;14(1):19-26.

96. Farrar MA, Fernandez-Luna J, Schreiber RD. Identification of two regions within the

cytoplasmic domain of the human interferon-gamma receptor required for function. J

Biol Chem. 1991;266(29):19626-19635.

97. Hershey GK, Schreiber RD. Biosynthetic analysis of the human interferon-gamma

receptor. identification of N-linked glycosylation intermediates. J Biol Chem.

1989;264(20):11981-11988.

98. Anderson P, Yip YK, Vilcek J. Human interferon-gamma is internalized and degraded by

cultured fibroblasts. J Biol Chem. 1983;258(10):6497-6502.

99. Celada A, Schreiber RD. Internalization and degradation of receptor-bound interferon-

gamma by murine macrophages. demonstration of receptor recycling. J Immunol.

1987;139(1):147-153.

100. Finbloom DS. Internalization and degradation of human recombinant interferon-gamma

in the human histiocytic lymphoma cell line, U937: Relationship to fc receptor

enhancement and antiproliferation. Clin Immunol Immunopathol. 1988;47(1):93-105.

101. Szente BE, Johnson HM. Binding of IFN gamma and its C-terminal peptide to a

cytoplasmic domain of its receptor that is essential for function. Biochem Biophys Res

Commun. 1994;201(1):215-221.

102. Ahmed CM, Johnson HM. IFN-gamma and its receptor subunit IFNGR1 are recruited to

the IFN-gamma-activated sequence element at the promoter site of IFN-gamma-

activated genes: Evidence of transactivational activity in IFNGR1. J Immunol.

2006;177(1):315-321.

Page 46: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

45

103. Mujtaba MG, Patel CB, Patel RA, et al. The gamma interferon (IFN-gamma) mimetic

peptide IFN-gamma (95-133) prevents encephalomyocarditis virus infection both in

tissue culture and in mice. Clin Vaccine Immunol. 2006;13(8):944-952.

104. Thiam K, Loing E, Delanoye A, et al. Unrestricted agonist activity on murine and human

cells of a lipopeptide derived from IFN-gamma. Biochem Biophys Res Commun.

1998;253(3):639-647.

105. Albericio F, Kruger HG. Therapeutic peptides. Future Med Chem. 2012;4(12):1527-1531.

106. Bansal R, Prakash J, De Ruiter M, Poelstra K. Interferon gamma peptidomimetic targeted

to hepatic stellate cells ameliorates acute and chronic liver fibrosis in vivo. J Control

Release. 2014;179:18-24.

107. Ramachandran P, Iredale JP, Fallowfield JA. Resolution of liver fibrosis: Basic mechanisms

and clinical relevance. Semin Liver Dis. 2015;35(2):119-131.

108. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest. 2013;123(5):1887-

1901.

109. Pinzani M, Rombouts K, Colagrande S. Fibrosis in chronic liver diseases: Diagnosis and

management. J Hepatol. 2005;42 Suppl(1):S22-36.

110. Mehal WZ, Schuppan D. Antifibrotic therapies in the liver. Semin Liver Dis.

2015;35(2):184-198.

111. Hauff P, Gottwald U, Ocker M. Early to phase II drugs currently under investigation for

the treatment of liver fibrosis. Expert Opin Investig Drugs. 2015;24(3):309-327.

112. Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrotic diseases: Nearing

the starting line. Sci Transl Med. 2013;5(167):167sr1.

113. Muir AJ, Sylvestre PB, Rockey DC. Interferon gamma-1b for the treatment of fibrosis in

chronic hepatitis C infection. J Viral Hepat. 2006;13(5):322-328.

114. Soza A, Heller T, Ghany M, et al. Pilot study of interferon gamma for chronic hepatitis C. J

Hepatol. 2005;43(1):67-71.

115. Borkham-Kamphorst E, Stoll D, Gressner AM, Weiskirchen R. Antisense strategy against

PDGF B-chain proves effective in preventing experimental liver fibrogenesis. Biochem

Biophys Res Commun. 2004;321(2):413-423.

116. Ogawa S, Ochi T, Shimada H, et al. Anti-PDGF-B monoclonal antibody reduces liver

fibrosis development. Hepatol Res. 2010;40(11):1128-1141.

117. Chen SW, Zhang XR, Wang CZ, Chen WZ, Xie WF, Chen YX. RNA interference targeting

the platelet-derived growth factor receptor beta subunit ameliorates experimental

hepatic fibrosis in rats. Liver Int. 2008;28(10):1446-1457.

118. Chen SW, Chen YX, Zhang XR, Qian H, Chen WZ, Xie WF. Targeted inhibition of platelet-

derived growth factor receptor-beta subunit in hepatic stellate cells ameliorates hepatic

fibrosis in rats. Gene Ther. 2008;15(21):1424-1435.

2

Page 47: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 2

46

119. Neef M, Ledermann M, Saegesser H, et al. Oral imatinib treatment reduces early

fibrogenesis but does not prevent progression in the long term. J Hepatol.

2006;44(1):167-175.

120. Hong F, Chou H, Fiel MI, Friedman SL. Antifibrotic activity of sorafenib in experimental

hepatic fibrosis: Refinement of inhibitory targets, dosing, and window of efficacy in vivo.

Dig Dis Sci. 2013;58(1):257-264.

121. Berardis S, Dwisthi Sattwika P, Najimi M, Sokal EM. Use of mesenchymal stem cells to

treat liver fibrosis: Current situation and future prospects. World J Gastroenterol.

2015;21(3):742-758.

122. Poelstra K, Beljaars L, Melgert BN. Cell-specific delivery of biologicals: Problems, pitfalls

and possibilities of antifibrotic compounds in the liver. Drug Discov Today. 2013;18(23-

24):1237-1242.

123. Jimenez Calvente C, Sehgal A, Popov Y, et al. Specific hepatic delivery of procollagen

alpha1(I) siRNA in lipid-like nanoparticles resolves liver fibrosis. Hepatology. 2015.

124. Elrick LJ, Leel V, Blaylock MG, et al. Generation of a monoclonal human single chain

antibody fragment to hepatic stellate cells--a potential mechanism for targeting liver

anti-fibrotic therapeutics. J Hepatol. 2005;42(6):888-896.

125. Beljaars L, Molema G, Weert B, et al. Albumin modified with mannose 6-phosphate: A

potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic

stellate cells. Hepatology. 1999;29(5):1486-1493.

126. Beljaars L, Molema G, Schuppan D, et al. Successful targeting to rat hepatic stellate cells

using albumin modified with cyclic peptides that recognize the collagen type VI receptor.

J Biol Chem. 2000;275(17):12743-12751.

127. Yang N, Singh S, Mahato RI. Targeted TFO delivery to hepatic stellate cells. J Control

Release. 2011;155(2):326-330.

128. Li F, Li QH, Wang JY, Zhan CY, Xie C, Lu WY. Effects of interferon-gamma liposomes

targeted to platelet-derived growth factor receptor-beta on hepatic fibrosis in rats. J

Control Release. 2012;159(2):261-270.

129. Bartneck M, Warzecha KT, Tacke F. Therapeutic targeting of liver inflammation and

fibrosis by nanomedicine. Hepatobiliary Surg Nutr. 2014;3(6):364-376.

130. Koivunen E, Wang B, Ruoslahti E. Phage libraries displaying cyclic peptides with different

ring sizes: Ligand specificities of the RGD-directed integrins. Biotechnology (N Y).

1995;13(3):265-270.

131. Marcelino J, McDevitt CA. Attachment of articular cartilage chondrocytes to the tissue

form of type VI collagen. Biochim Biophys Acta. 1995;1249(2):180-188.

Page 48: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Targeted therapies in liver fibrosis

47

132. Fretto LJ, Snape AJ, Tomlinson JE, et al. Mechanism of platelet-derived growth factor

(PDGF) AA, AB, and BB binding to alpha and beta PDGF receptor. J Biol Chem.

1993;268(5):3625-3631.

133. Bansal R, Prakash J, de Ruijter M, Beljaars L, Poelstra K. Peptide-modified albumin carrier

explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver

fibrosis. Mol Pharm. 2011;8(5):1899-1909.

134. Hagens WI, Mattos A, Greupink R, et al. Targeting 15d-prostaglandin J2 to hepatic

stellate cells: Two options evaluated. Pharm Res. 2007;24(3):566-574.

135. Poosti F, Bansal R, Yazdani S, et al. Selective delivery of IFN-gamma to renal interstitial

myofibroblasts: A novel strategy for the treatment of renal fibrosis. FASEB J.

2015;29(3):1029-1042.

136. Subramaniam PS, Flowers LO, Haider SM, Johnson HM. Signal transduction mechanism

of a peptide mimetic of interferon-gamma. Biochemistry. 2004;43(18):5445-5454.

137. Szente BE, Soos JM, Johnson HW. The C-terminus of IFN gamma is sufficient for

intracellular function. Biochem Biophys Res Commun. 1994;203(3):1645-1654.

138. Bansal R, Tomar T, Ostman A, Poelstra K, Prakash J. Selective targeting of interferon

gamma to stromal fibroblasts and pericytes as a novel therapeutic approach to inhibit

angiogenesis and tumor growth. Mol Cancer Ther. 2012;11(11):2419-2428.

139. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and

medicine. Genes Dev. 2008;22(10):1276-1312.

140. Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth

Factor Rev. 2004;15(4):255-273.

141. Angeli F, Koumakis G, Chen MC, Kumar S, Delinassios JG. Role of stromal fibroblasts in

cancer: Promoting or impeding? Tumour Biol. 2009;30(3):109-120.

142. Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in

cancer. Nat Rev Cancer. 2004;4(11):839-849.

143. Prakash J, de Jong E, Post E, Gouw AS, Beljaars L, Poelstra K. A novel approach to deliver

anticancer drugs to key cell types in tumors using a PDGF receptor-binding cyclic peptide

containing carrier. J Control Release. 2010;145(2):91-101.

2

2

Page 49: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

3

Page 50: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

F. van Dijka,b, L. Beljaarsa, E. Posta, C. Draijera, X. Hua, K. Luotoc, P. Olingab and K.

Poelstraa

aGroningen Research Institute of Pharmacy, Department of Pharmacokinetics,

Toxicology and Targeting, University of Groningen, Groningen, The Netherlands, bGroningen Research Institute of Pharmacy, Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen, Groningen, The

Netherlands, cBiOrion Technologies BV, MediTech Center UMCG, Groningen,

The Netherlands.

Manuscript submitted

Steering therapeutic proteins:

the mechanism of action of

mimetic interferon gamma delivered to

the disease-induced PDGFβ-receptor

Page 51: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

50

Abstract

Therapeutic proteins including cytokines are promising agents in treating numerous diseases,

and their clinical use could be drastically improved with cell-selective delivery. We explored

this concept for the antifibrotic cytokine interferon gamma (IFNγ) in attenuating liver fibrosis,

and designed the chimeric construct Fibroferon that targets the IFNγ-signaling peptide,

lacking the IFNγ-receptor binding moiety, to the fibrosis-induced platelet-derived growth

factor beta receptor (PDGFβR) on myofibroblasts. We now aim to elucidate this mechanism of

action and hypothesize that steering the IFNγ-activity domain to the PDGFβR does not affect

PDGFβR-signaling, but triggers the IFNγ-receptor signaling in the same cell instead. Specific

PDGFR-binding of Fibroferon was shown, without activating its signaling pathway or blocking

PDGF-BB effects. Instead, activation of the intracellular IFNγR-signaling in fibroblasts

occurred, hallmarked by pStat1-activation and translocation into the nucleus. Unlike IFNγ,

Fibroferon did not activate macrophages in vitro, since these cells lack the PDGFR.

Furthermore, we demonstrated clathrin-dependent endocytosis of Fibroferon, followed by

translocation and nuclear uptake. In conclusion, we demonstrated the cell-selective binding

of Fibroferon to the PDGFR, yet it switches to the intracellular routing of IFNγ. Interestingly,

Fibroferon escapes from endosomal/lysosomal pathways and enters the nucleus. Herewith

we confirmed the possibility of redirecting cytokines, representing a stride towards their

therapeutic use.

Page 52: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

51

Introduction

Therapeutic proteins are becoming increasingly popular in the treatment of many diseases.

The first recombinant protein that gained approval by the US Food and Drug Administration

(FDA) was human insulin in the early 1980s, and ever since numerous therapeutic proteins

have emerged in the market. Already, more than 130 different protein-based entities like

monoclonal antibodies, modified enzymes and natural interferons have been approved by the

FDA for clinical use, and many more are in development1,2. Such proteins are particularly

attractive as therapeutic agents, because they are highly potent and they display exquisite

affinity and specificity for their targets; something that cannot be easily mimicked by small-

molecule drugs2.

Despite their promising therapeutic effects, the efficacy and safety of many therapeutic

proteins is still limited, mostly related to low exposure to target cells, due to rapid renal

clearance and a short circulation half-life, and to severe systemic side effects attributed to

ubiquitous receptor expressions or development of immunogenicity2,3. Many attempts are

being made to overcome these challenges, by either manipulation of the protein itself, or by

changing the formulation3. A common way to modify a therapeutic protein is for example by

covalent coupling of polyethylene glycol (PEG) or by fusion to other proteins with longer in

vivo half-lives. Our strategy to improve the therapeutic applicability of potent proteins is to

target the protein of interest to key pathogenic cells by redirecting them to disease-induced

receptors with homing devices.

We previously developed several therapeutic proteins targeting the fibrotic liver, where

excessive amounts of extracellular matrix proteins are produced by mainly activated hepatic

stellate cells (HSCs)4,5. The platelet-derived growth factor β receptor (PDGFβR) is abundantly

and specifically expressed on activated HSCs during fibrogenesis6, which makes it an attractive

target for cell-specific delivery of antifibrotic compounds such as the potent antifibrotic

cytokine interferon γ (IFNγ). We established cell-specific targeting of a series of proteins that

all share the same targeting moiety, i.e. the cyclic PDGFβ-receptor recognizing peptide (pPB)7,

as reviewed recently by Van Dijk et al.8, including a minimized targeted IFNγ construct

referred to as ‘Fibroferon’: fibroblast-targeted interferon γ (previously called BipPB-PEG-IFNγ

mimetic, mimγ-BipPB, or BOT191). In this construct, the nuclear localization sequence of IFNγ

containing the agonistic activity domain is retained, while the extracellular IFNγ-receptor

binding moiety is lacking9. This truncated IFNγ-structure was targeted to the PDGFβ-receptor

by coupling it to the bicyclic PDGFβ-receptor recognizing peptide (BipPB) via a 2 kDa

polyethylene glycol (PEG) linker, allowing a dimeric receptor interaction. We previously

demonstrated an improved in vivo antifibrotic effect and reduced IFNγ-related side effects of

3

Page 53: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

52

Fibroferon in mice suffering from CCl4-induced liver fibrosis10 and renal fibrosis, thereby

tackling the two main problems associated with clinical use of IFNγ11.

When native IFNγ binds to its endogenous receptor it induces a cascade of events eventually

leading to the recruitment and phosphorylation of the signal transducer and activator of

transcription 1 alpha (Stat1α). The recently developed non-canonical model of IFNγ-receptor

signaling describes active transport of a complex consisting of amongst others Stat1α, IFNγ

and its receptor into the nucleus9,12. It is described that both IFNγ and its receptor are

associated within clathrin-coated pits, and they are also reported to be present in lipid rafts

and caveolae, albeit to a lesser extent13. However, our chimeric protein only contains the

activity domain of IFNγ, lacking the extracellular receptor binding domain, and instead is

designed to bind to the PDGFβ-receptor. The endogenous ligand of the PDGFβ-receptor is

PDGF-BB, a dimeric molecule produced by various cell types, including platelets, macrophages

and hepatocytes, which stimulates profibrogenic processes like proliferation, migration and

contraction of HSCs14. The signaling cascade downstream of the PDGFβ-receptor involves

multiple pathways, in which the MAPK/ERK pathway and Akt/PKB pathway are the major

routes15,16. In order to reveal the mechanism of action of Fibroferon and thus answer the key

question how truncated IFNγ delivered to the PDGFβ-receptor exerts its effects, we analyzed

receptor binding and intracellular routing as well as activation of signaling pathways in

fibroblasts. Our studies show that redirection of the signaling moiety of IFNγ to the PDGFβR

leads to activation of the IFNγR. Moreover, we demonstrated escape from the endosomal

pathway and nuclear translocation of our chimeric peptide.

Materials and methods

Synthesis and characterization of Fibroferon

Fibroferon was custom-prepared (Eurogentec, Liège, Belgium) as described before10. The final

product was analyzed by silver staining, western blot, electrospray mass spectrometry (ES-

MS) and ultraperformance liquid chromatography (UPLC). UPLC was performed on an

analytical Acclaim RSLC C18 (2.2 μm, 120 Å, 2.1 x 100 mm) column (Thermo Scientific).

Separation was achieved by elution with 0.1% TFA in H2O and 0.1% TFA in ACN. The flow rate

was kept at 0.4 ml/min during a 7.5 min run (λ = 220 nm).

Page 54: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

53

Silver staining

Samples (10 μg protein) were applied on a 15% SDS polyacrylamide gel. The gel was fixed in

10% acetic acid in H2O/MeOH = 1/1 for 1 h, washed 3 x 30 min in 25% EtOH in H2O, and

incubated 1 min in H2O containing 200 μg/ml Na2S2O3. After washing the gel incubated 20min

in H2O containing 2 mg/ml AgNO3 and 1.5‰ formaldehyde, was washed again and developed

in H2O containing 4 ng/ml Na2S2O3, 0.5‰ formaldehyde and 60 mg/ml Na2CO3. The reaction

was stopped after washing, by incubation in 12% acetic acid in H2O/MeOH = 4/5 for 15 min.

Western blot analysis

Samples (10 μg protein, 25 μl cell lysate or 100 μg tissue homogenate) were applied on a SDS

polyacrylamide gel (10 or 12%) and transferred to a 0.2 μm polyvinylidene difluoride

membrane. Membranes were blocked for 1 h in 5% nonfat dry milk in tris-buffered

saline/0.1% Tween-20 (ELK/TBS-T) and incubated overnight at 4 °C with the primary antibody

in 5% ELK/TBS-T. The appropriate HRP-conjugated secondary antibodies were applied in 5%

ELK/TBS-T for 2 h after washing in TBS-T. Bands were visualized with enhanced

chemiluminescence (VisiGlo Plus HRP, Amresco) and quantified with GeneSnap (Syngene,

Synoptics, Cambridge, UK). The used antibodies are listed in table 1.

Cell cultures

Human LX2 hepatic stellate cells were kindly provided by prof. Scott Friedman (Mount Sinai

Hospital, New York). Murine NIH/3T3 fibroblasts and RAW264.7 macrophages were obtained

from the American Type Culture Collection (Rockville, MD). Cells were cultured in Dulbecco’s

modified Eagle’s medium (DMEM, Invitrogen, Carlsbad, CA) containing 10% FCS,

supplemented with 100 U/ml penicillin and 100 μg/ml streptomycin (3T3s and LX2s), 2 mM L-

glutamine (3T3s and RAWs) and 10 μg/ml gentamicin (RAWs).

In vitro studies

Macrophage activation. RAW264.7 cells were stimulated with Fibroferon (2.5, 5, 25, 50, 500

ng/ml) or IFNγ (5, 10, 50, 100, 1000 ng/ml) for 48 h, detached with lidocaine/EDTA and

washed with PBS containing 2% FCS and 5 mM EDTA (PFE). After 30 min incubation with an

anti-MHCII-APC/Cy7 antibody (1:100, Biolegend) at 4 °C and washing in PFE, the cells were

3

Page 55: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

54

resuspended and studied using the BD FACSArrayTM bioanalyzer (BD Biosciences). Data were

analyzed using FlowJo software (Tree Start, Ashland, USA).

Binding and uptake. Compounds were labeled with Alexa Fluor Succinimidyl Ester label

according to manufacturer’s instructions (ThermoFisher Scientific). Cells plated on coverslips

were stimulated for 24 h with TGFβ (5 ng/ml, Peprotech, Rocky Hill, NJ) and incubated with

Alexa-labeled Fibroferon or IFNγ (both 10 μg/ml) for 30, 60 or 120 min at 37 °C. Binding was

inhibited by 20 min preincubation with 4 mM trapidil, by 10 min preincubation with 25 μM

chlorpromazine or 5 μg/ml filipin or by 30 min preincubation with 100 mM NaN3. Binding and

uptake were analyzed with fluorescence microscopy (Leica Microsystems) or confocal

microscopy (Leica Microsystems, SP8). In addition, quantification of the cellular accumulation

up to 4 h at 4 and 37 °C was performed by flow cytometry analysis.

Binding to the PDGFβR. Fibroblasts were washed with PBS-based binding buffer containing

0.9 mM CaCl2, 0.49 mM MgSO4 and 1 mg/ml BSA, and incubated with Fibroferon (0.2, 0.5,

0.9, 1.9, 3.8, 7.5 and 15 μM) for 30 min at 4 °C, followed by 60 min incubation with 50.000

CPM I125PDGF-BB (Perkin Elmer). Cells were washed, lysed overnight in 20 mM Tris-HCl pH

7.5, 1% Triton-X1000 and 10% glycerol and measured with Packard Riastar gamma counter

(Perkin Elmer).

Signaling and effect. For PDGFβ-receptor mediated signaling, cells were stimulated with

PDGF-BB (50 ng/ml, Peprotech, Rocky Hill, NJ), Fibroferon (1 µg/ml) or IFNγ (2 µg/ml,

Peprotech) for 10 and 60 min at 37 °C. For IFNγ-receptor mediated signaling, cells were

starved 24 h followed by stimulation with TGFβ for 24 h, prior to incubation with Fibroferon

(1 and 10 µg/ml) or IFNγ (2 µg/ml) for 10 and 60 min at 37 °C. Cells were harvested in sample

buffer for WB and stored at -80 °C until analysis.

Cellular fractionation

Cells were stimulated with Fibroferon (1 μg/ml) or IFNγ (2 μg/ml) for 10 or 120 min (37 °C) in

duplicates and after this kept at 4 °C. For total cell lysates, pellets were resuspended in 50 μl

cell extraction buffer (ThermoFisher) containing 1 mM phenylmethane sulfonyl fluoride and

protease inhibitor, and incubated for 30 min, with vortexing at 10 min intervals. After

centrifugation (13.000 rpm, 10 min) the supernatants were aliquoted. For cytoplasmic

fractions, pellets were resuspended in 250 μl hypotonic buffer containing 20 mM Tris-HCl (pH

7.4), 10 mM NaCl and 3 mM MgCl2 and incubated for 15 min. After addition of 12.5 μl 10%

NP40, samples were vortexed 10 s, centrifuged (3.000 rpm, 10 min) and aliquoted. For nuclear

fractions, the pellets were resuspended in 250 μl hypotonic buffer, incubated for 15 min, and

Page 56: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

55

were centrifuged (3.000 rpm, 10 min). Pellets were processed as described for total cell

lysates, and were collected after centrifugation (14.000 g, 30 min). All fractions were diluted

in sample buffer, boiled for 5 min at 95 °C and stored at -20 °C until analysis. To confirm purity

of the different fractions, all fractions were tested for the presence of nuclear and

cytoplasmic components using topoisomerase-IIβ and α-tubulin as markers for nuclear and

cytoplasmic proteins, respectively.

Animal experiments

Male balb/c mice (Envigo, Horst, The Netherlands) received ad libitum normal diet with 12 h

light/dark cycle. All experimental protocols were approved by the Animal Ethical Committee

of the University of Groningen (the Netherlands). Liver fibrosis was induced by intraperitoneal

injections of increasing doses of CCl4 as described before17. In week 8, mice (n=3) received an

intravenous injection of 10 mg/kg PDGFβR-targeted mimetic IFNγ 30 min prior to sacrifice,

after which blood and different organs were collected.

Immunohistochemical staining

Liver cryosections at 4 μm of thickness (CryoStar NX70 cryostat, ThermoFisher Scientific,

Waltham, USA) were dried and fixed with acetone. Sections were rehydrated in PBS and

incubated with a primary antibody, applied either 1 h at RT (anti-pPB, anti-desmin and anti-

CD68) or overnight at 4 °C (anti-IFNγR and anti-PDGFβR). Next, sections incubated 30 min

with the appropriate HRP-, AP- or FITC-conjugated secondary antibodies, and if necessary

visualized with ImmPACT NovaRED and VECTASTAIN ABC-AP (both Vector, Burlingame, USA),

respectively. Photomicrographs were captured at 400x magnification (Aperio, Burlingame,

USA). Fluorescence stainings were obtained with fluorescence microscopy (Leica

Microsystems). The used antibodies are listed in table 1.

Statistical analyses

At least 3 individual experiments were performed to measure in vitro effects. Data are

presented as mean ± standard error mean (SEM). The graphs and statistical analyses were

performed using Graphpad Prism version 6.00 (GraphPad Prism Software, Inc., La Jolla, CA,

USA). Differences between groups were assessed by Friedman test followed by Dunn’s

multiple comparison test, unless stated otherwise in the corresponding figure legends. The

differences were considered significant at p<0.05.

3

Page 57: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

56

Table 1. Primary antibodies used for the western blot and immunohistochemical analyses.

Results

Characterization of Fibroferon

To characterize specific parts in the molecular structure of Fibroferon, illustrated in figure 1A,

we performed a silver staining and western blot analysis for pPB. All elements of Fibroferon

were connected, as reflected by the molecular weight of Fibroferon, which was smaller as

compared to recombinant IFNγ (15.6 kDa), but larger than mimetic IFNγ (4.7 kDa), and the

presence of pPB (Figs. 1B and C). The molecular weight as determined by electrospray mass

spectrometry (ES-MS) revealed a mass of 9344 Da (Fig. 1D), which matched with a structure

of 2 cyclic peptides (~1 kDa each), conjugated with PEG (2 kDa) and truncated IFNγ (IFNγ95-132:

AKFEVNNPQVQRQAFNELIRVVHQLLPESSLRKRKRSR, 4.7 kDa). The purity of the synthesized

lyophilized compound was 98%, as measured with ultra-performance liquid chromatography

(UPLC) (Fig. 1E). Furthermore, we assessed the stability of the protein at room temperature

when dissolved in PBS, by measuring the UPLC profiles at t=7, 14 and 21 days. The UPLC data

Antibody Source Dilution

Polyclonal rabbit anti-pPB Charles

Rivers

1:1000 (WB)

1:250 + 5% NMS (staining)

Monoclonal rabbit anti-PDGFβR Cell Signaling 1:1000 (WB)

1:50 (staining)

Monoclonal mouse anti-GAPDH Sigma-

Aldrich

1:20000

Polyclonal goat anti-desmin Santa Cruz 1:100

Monoclonal rat-anti-CD68 Serotec 1:500

Polyclonal rabbit anti-IFNγR1 antibodies-

online.com

1:1000 (WB)

1:20 (staining)

Polyclonal rabbit anti-phospho-p44/42 MAPK Cell Signaling 1:1000

Monoclonal rabbit anti-p44/42 MAPK Cell Signaling 1:1000

Polyclonal rabbit anti-phospho-Akt Cell Signaling 1:1000

Polyclonal rabbit anti-Akt Cell Signaling 1:1000

Monoclonal rabbit anti-phospho-Stat1 (Y701) Cell Signaling 1:1000

Polyclonal rabbit anti-Stat1 Cell Signaling 1:1000

Monoclonal rabbit topoisomerase-IIβ Abcam 1:1000

Monoclonal mouse α-tubulin Merck

Millipore

1:1000

Page 58: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

57

confirmed stability at least up to 21 days, as demonstrated by the constant retention factor

(Fig. S1A). This was confirmed by ES-MS, that showed no change in the molecular structure of

Fibroferon in time (Fig. S1B).

Figure 1. Characterization of Fibroferon. (A) Schematic representation of Fibroferon. (B) Silver staining and (C)

western blot analysis for pPB on full length IFNγ (I), Fibroferon (II) and mimetic IFNγ (III) to demonstrate the

presence of all key elements in the structure of Fibroferon. M denotes molecular weight marker. (D) ES-MS data

showing a molecular mass of 9344 Da for Fibroferon. (E) UPLC data demonstrating a purity of 98% for the

Fibroferon construct.

Effect of Fibroferon on macrophages

Since the therapeutic application of IFNγ is limited due to severe side effects, we designed

Fibroferon in such a way that extracellular binding to the ubiquitously expressed IFNγ-

receptor is prevented. We observed that Fibroferon with increasing concentrations did not

induce MHCII-expression in macrophages after 48 hours of stimulation, unlike IFNγ in the

equimolar range (Fig. 2A). This is in agreement with the absence of IFNγ-related side effects

seen in vivo10. RAW264.7 macrophages do express the IFNγR but lack the PDGFβ-receptor, in

contrast to the positive controls i.e. 3T3 fibroblasts and LX2 hepatic stellate cells (Fig. 2B).

3

Page 59: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

58

Figure 2. Effect of Fibroferon on macrophages. (A) Analysis of MHCII-expression in RAW macrophages using flow

cytometry. Data show no MHCII-induction 48 h after stimulation with increasing concentrations of Fibroferon, in

contrast to IFNγ (n=3). (B) Western blot analysis of the PDGFβR-expression in cell lysates of 3T3 fibroblasts (high

expression), LX2 hepatic stellate cells (medium expression) and RAW macrophages (no expression).

Targeting to activated hepatic stellate cells in vivo

Although the therapeutic effects of Fibroferon were seen in different models10,11, the in vivo

localization of this compound was unknown. Using a specific antibody against the pPB moiety,

we are now able to localize PDGFβ-receptor targeted mimetic IFNγ in vivo, and found

predominant staining in the fibrotic areas of CCl4-exposed livers (Fig. 3A). In addition, double-

stainings revealed colocalization of the construct with the HSC-marker desmin and not with

the macrophage-marker CD68 (Fig. 3B).

Figure 3. Localization of Fibroferon in livers of mice with CCl4-induced liver fibrosis. (A) Immunohistochemical

staining for pPB (red) in livers of mice treated with (upper panel) and without (lower panel) PDGFβ-receptor

targeted IFNγ. (B) Immunohistochemical double stainings for pPB (red) plus the HSC marker desmin (blue), and

pPB (red) plus CD68 (blue), as marker for liver macrophages. It can be seen that pPB co-localizes with desmin

and not with CD68 (arrows). Magnification 400x.

Page 60: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

59

Receptor expressions in the human liver

To illustrate the translation to the human situation, we analyzed IFNγR- and PDGFβR-

expression in human normal and fibrotic livers by western blot (Figs. 4A and C) and

immunohistochemistry (Figs. 4B and D). IFNγR-expression was found in normal human livers

on many different cell types and its expression was strongly enhanced on multiple cell types

in cirrhotic livers. PDGFβR-expression was virtually absent in normal livers and very high on

fibroblasts only in the collagenous bands in cirrhotic livers, confirming earlier reports5.

Figure 4. IFNγR- and PDGFβR-expressions in normal and cirrhotic human livers. (A) Western blot analysis for the

IFNγ-receptor in healthy (n=5) and cirrhotic livers (n=7). (B) Immunohistochemical staining for IFNγR in normal

and cirrhotic livers. High expression was observed in macrophages (indicated by asterisks) and immune cells

(indicated by arrow). Next to this, cells in cirrhotic bands as well as hepatocytes stained positive for IFNγR (400x).

(C) Western blot analysis of the PDGFβR-expression levels in normal and cirrhotic human livers. (D)

Immunohistochemical staining to detect PDGFβR-expression. Strong staining can be seen in fibrotic bands,

indicated by F (200x). Statistics were performed using the Mann-Whitney test.

Binding of Fibroferon to the PDGFβ-receptor

Since the PDGFβ-receptor is the designated target receptor, we first assessed the binding of

Fibroferon (Alexa555-labeled) to the PDGFβ-receptor on fibroblasts (Fig. 5A). Cells stimulated

with TGFβ, characterized by an increased PDGFβR-expression (Fig. 5B), displayed a high

binding of Fibroferon (Fig. 5A). The binding of Fibroferon was blocked to a great extent by the

3

Page 61: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

60

PDGFR-antagonist trapidil (Fig. 5A). Specific binding of Fibroferon to the PDGFβR was

confirmed in a radioactive assay, in which increasing concentrations of Fibroferon reduced

the cellular binding of 125I-labeled PDGF-BB (Fig. 5C). The IC50 of Fibroferon in these

experiments was calculated to be 1.80 ± 0.78 nmol/ml.

Figure 5. In vitro binding of Fibroferon to the PDGFβR. (A) Representative fluorescent photographs depicting

specific binding and uptake of Alexa555-labeled Fibroferon (red) in fibroblasts (200x). The fluorescent signal was

increased in TGFβ-stimulated cells, and significantly reduced upon trapidil preincubation. Nuclei were

counterstained with DAPI (blue). (B) Immunofluorescent photographs showing the TGFβ-induced PDGFβ-

receptor expression (green) in fibroblasts (400x). (C) Radioactive binding assay using I125

-PDGF-BB plus increasing

concentrations of Fibroferon (n=4). It is shown that Fibroferon inhibits the binding of I125

-PDGF-BB to cells. The

half maximal inhibitory concentration (IC50) of Fibroferon was 1.80 ± 0.76 nmol/ml. All experiments were done in

mouse fibroblasts (NIH/3T3).

Activation of the PDGFβ-receptor

The two major signaling pathways downstream of the PDGFβ-receptor, i.e. the

phosphorylation of both ERK1,2 and Akt, were used as hallmark to determine whether

activation of this profibrotic signaling cascade occurred after binding of Fibroferon to the

PDGFR. The phosphorylation of ERK1,2 was significantly induced in fibroblasts after 10

minutes stimulation with PDGF-BB, whereas this response remained absent when cells were

stimulated with Fibroferon or IFNγ up to 60 minutes (Fig. 6A). Similarly, no phosphorylation of

Akt was seen upon stimulation with Fibroferon or IFNγ for 10 or 60 minutes (Fig. 6B). So,

despite binding to Fibroferon to the PDGFβ-receptor on myofibroblasts, it did not induce its

downstream signaling cascade. Fibroferon also did not attenuate PDGF-BB-induced pERK

Page 62: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

61

activation at various time points and concentrations (Fig. 6C), indicating no significant

antagonistic activity on the PDGFR.

Figure 6. In vitro signaling of Fibroferon after binding to the PDGFβR. Representative bands and quantitative

analysis of the western blots for (A) pERK1,2 and ERK1,2 and (B) pAkt and Akt on mouse fibroblasts (NIH/3T3)

stimulated with PBS (I), PDGF-BB (II), Fibroferon (III) or IFNγ (IV). Note the absence of activation of both pERK1,2

and pAkt after 10 and 60 min Fibroferon stimulation (n=4). Statistics with 2way ANOVA followed by Dunnett’s

multiple comparison test. (C) pERK1,2 and ERK1,2 on fibroblasts stimulated with PDGF-BB (II), co-stimulated with

Fibroferon at increasing concentrations (lanes V, VI and VII, corresponding with the graph below) (n=3).

Activation of the IFNγ-signaling pathway

Phosphorylation of Stat1 was used as marker for the

activation of the IFNγ-receptor. We observed

significant induction of phosphorylation of Stat1 by

Fibroferon, albeit to a lesser extent than IFNγ (Fig. 7).

Fibroferon did not induce any pStat1 signaling in RAW

macrophages in contrast to native IFNγ (data not

shown), which is in line with our data on MHCII

expression by Fibroferon and IFNγ in these cells.

Figure 7. Signaling of the IFNγR. Western blot analysis for pStat1 and Stat1 on fibroblasts stimulated with TGFβ

(I), IFNγ (II) or Fibroferon (III, IV) (the latter three preincubated with TGFβ). Induction of pStat1 can be seen by

IFNγ and Fibroferon (n=3). Statistics with 2way ANOVA followed by Dunnett’s multiple comparison test.

3

Page 63: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

62

Active internalization via clathrin-dependent endocytosis

The binding of Fibroferon to the PDGFβ-receptor and the subsequent activation of the

signaling pathway downstream of the IFNγ-receptor raised the question whether Fibroferon

was internalized and followed the intracellular routing of IFNγ in these cells. Therefore, we

assessed uptake characteristics of Alexa-labeled Fibroferon and IFNγ in mouse fibroblasts

using flow cytometry. Fibroblasts internalized both Fibroferon and IFNγ in a temperature

dependent way, demonstrating active uptake (Figs. 8A and B). The uptake of IFNγ by 100% of

the cell population took approximately 30 min, compared to 60 min for the uptake of

Fibroferon. Although all cells were positive for IFNγ or Fibroferon at respectively 30 and 60

minutes after incubation, the uptake of both compounds was still increasing in time,

ultimately reaching similar values as reflected by their median fluorescence intensities (MFI)

(Figs. 8A and B). This indicates that the uptake was not saturated yet. Active uptake was

confirmed by blocking the ATP-dependent uptake processes with NaN3, which significantly

reduced the uptake of Fibroferon and IFNγ (Fig. S2). In addition, we found that the uptake of

both Fibroferon and IFNγ was clathrin dependent, as the uptake was inhibited by an inhibitor

of clathrin-dependent endocytosis (chlorpromazine), and not with an inhibitor of caveolin

(filipin) (Figs. 8C and D).

Figure 8. Kinetics of binding and uptake of Fibroferon and IFNγ. Analysis of the uptake of (A) Fibroferon-A647 and

(B) IFNγ-A555 at 4 and 37 °C in fibroblasts, as determined by the percentage positive cells and the median

fluorescence intensity (MFI) using flow cytometry (n=3). Statistics were performed using Wilcoxon’s test,

comparing areas under the curve. (C) and (D) Effect of chlorpromazine and filipin on uptake of Fibroferon-A647

(C) and IFNγ-A555 (D) (n=5).

Page 64: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

63

Intracellular localization of Fibroferon

The intracellular routing of proteins was studied in more detail using fluorescence and

confocal microscopy. Both Fibroferon and IFNγ were at early time points localized in the early

and late endosomes, while at later time points (predominantly after 1-2 hours of incubation)

localization of both compounds could be observed at the nuclear membrane (Fig. 9A),

suggesting similar intracellular routing in fibroblasts. Moreover, using flow cytometry analysis

we showed that after simultaneous addition of IFNγ and Fibroferon, both compounds were

internalized by the same cells (Fig. 9B). Using confocal microscopy, we demonstrated a

nuclear localization of Fibroferon and IFNγ (Fig. 10A). In addition, cellular fractionation studies

demonstrated the translocation of pStat1 to the nucleus after 10 and 120 minutes of

incubation with Fibroferon or IFNγ (Fig. 10B). Interestingly, pStat1 was found only in the

nuclear fraction (marked by topoisomerase-IIβ), and not in the cytoplasmic fraction (marked

by α-tubulin) when induced by Fibroferon, whereas IFNγ induced pStat expression both in the

cytoplasmic and the nuclear fraction. These data support the non-canonical model of

receptor signaling as presented by Johnson et al.9, which describes this IFNγ-induced nuclear

translocation of pStat1.

Figure 9. Intracellular localization of Fibroferon and IFNγ. (A) Fluorescence photographs demonstrating

localization of Fibroferon-A555 (red) or IFNγ-A488 (green) after 60min in the endosomes and at the nuclear

membrane after 120 min incubation (400x). Nuclei were counterstained with DAPI (blue). (B) Flow cytometry

analysis of fibroblasts treated with either Fibroferon-A647, IFNγ-A555 or the combination of both compounds. It

is shown that Fibroferon and IFNγ are internalized by exactly the same cells, as represented by the double-

positive fibroblasts (orange) after co-incubation of both compounds.

3

Page 65: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

64

Figure 10. Nuclear translocation of Fibroferon and IFNγ. (A) Confocal microscopic photograph of Fibroferon-A555

(red) and IFNγ-A488 (green) after 120 min incubation of fibroblasts (630x). Nuclear localization can be seen in

both cases. A movie of this is presented in the supplementary data. (B) Representative bands and (C)

quantitative analysis of the western blots for pStat1, topoisomerase-IIβ (marker for nuclear proteins) and α-

tubulin (marker for cytoplasmic proteins) on nuclear and cytoplasmic fractions of fibroblasts stimulated with

Fibroferon or IFNγ (n=3) at different time points. All experiments were done in mouse fibroblasts (NIH/3T3).

Discussion

Many potent therapeutic proteins have been developed, tested in clinical trials and emerged

in the market. Unfortunately, many potent proteins never reach the clinic, often mainly due

to low exposure to the target cells and the induction of adverse effects by triggering the

immune system2. Since higher dosing is usually not possible due to side effects, many

attempts are made to improve the kinetic properties of the protein by for example changing

parts of the protein structure3. Our approach focusses on the attachment of homing devices

to the protein of interest to target a specific receptor, thereby aiming at cell-specific delivery

of the protein. We therefore developed a chimeric construct called Fibroferon, in which the

signaling moiety of the antifibrotic cytokine interferon gamma IFNγ is coupled to a bi-cyclic

PDGFβ-receptor recognizing peptide (BipPB) in order to deliver the compound specifically to

Page 66: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

65

the PDGFβ-receptor expressed on activated fibroblasts in the fibrotic liver. Treatment of mice

suffering from CCl4-induced liver fibrosis with this redirected IFNγ has been shown to improve

its therapeutic efficacy compared to full length IFNγ, and markedly reduced IFNγ-related side

effects10, but the mechanism of action of Fibroferon was unclear. It remained intriguing how

after binding of the compound to the PDGFβ-receptor this construct induced IFNγ-related

effects. In the present study we therefore studied receptor binding, intracellular routing and

nuclear uptake, as well as activation of signaling pathways of Fibroferon in myofibroblasts,

relative to IFNγ, and found that delivery of IFNγ to the PDGFβR leads to activation of the IFNγ-

signaling pathway and accumulation of pStat1 and the compound itself in the nucleus.

The profibrotic growth factor PDGF-BB plays a pivotal role in fibrotic diseases and

correspondingly the PDGFβ-receptor is abundantly expressed on cell membranes of liver cells

involved in fibrotic processes. Therefore, our group developed a series of PDGFβ-receptor

targeted constructs including Fibroferon, all based on the same targeting moiety, i.e. pPB, as

recently reviewed8. pPB is a PDGF-BB-derived peptide that binds to the PDGFβ-receptor.

Several studies established specific targeting of these constructs to cells with disease-induced

PDGFβR expression, such as the myofibroblasts in liver fibrosis and other cells associated with

high PDGFβ-receptor expression during diseases, like certain types of cancer and kidney

fibrosis7,10,17-20. Significant therapeutic effects of Fibroferon were demonstrated in animals

with liver and renal fibrosis10,11. In the present study, we were able to show delivery of

Fibroferon to liver fibroblasts in vivo and we demonstrated in vitro that Fibroferon binds to

the PDGFβ-receptor. However, it did not activate the profibrotic signaling cascade, as shown

by absence of pERK and pAkt upregulation. Fibroferon did not act as an antagonist of PDGF-

BB, since PDGF-BB mediated signaling was not impeded in the presence of increasing

concentrations of Fibroferon. In vivo antifibrotic effects of this compound are apparently not

due to blocking of the PDGFβR.

In the Fibroferon molecule the IFNγ-part that harbors the nuclear localization sequence is

incorporated, to prevent the extracellular interaction with the ubiquitously expressed IFNγ-

receptor. This is demonstrated by its inability to induce MHCII expression in macrophages or

activate a signaling cascade, unlike IFNγ. Previously prepared PDGFβR-directed full length

IFNγ-constructs17 still were able to bind to the IFNγR and thus still activated MHCII (data not

shown). Since RAW macrophages do not express the PDGFβR, Fibroferon cannot interact with

these cells. Fibroferon does bind to cells expressing the PDGFβR, and then activates the

intracellular domain of IFNγR, leading to activation of this signaling cascade, as hallmarked by

the phosphorylation of Stat1 in PDGFβ-receptor expressing fibroblasts. Therefore, we propose

that after interaction with the PDGFβR, Fibroferon is internalized and flips to an intracellular

part of the IFNγR.

3

Page 67: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

66

We found that Fibroferon was mainly internalized via clathrin-dependent endocytosis. The

IFNγ-receptor is generally associated with clathrin-coated pits and mainly endocytosed via

clathrin-dependent endocytosis21,22, inhibitable by RNAi-mediated silencing of clathrin23. The

PDGFβ-receptor on the other hand is specifically localized in distinct membrane regions

enriched in caveolin, known as caveolae24,25. Apparently, the interaction of Fibroferon with

the PDGFβ-receptor permits the twisting of Fibroferon to the intracellular part of the IFNγ-

receptor, inducing clathrin-dependent endocytosis.

After translocation from the plasma membrane to early endosomes, receptors can either

recycle to the cell surface, or exit from this pathway and move to other intracellular

destinations, i.e. in most cases towards the late endosomes and eventually lysosomes or to

new domains on the cell surface26. The localization of Fibroferon and IFNγ 60 minutes after

addition to cells, fits with the localization in late endosomes. Escape from this endosomal

pathway to ensure cytosolic delivery and possibly nuclear translocation of the content is

uncommon, but it is described before27. One example includes peptides and oligonucleotides

covalently attached to a peptide that is able to cross lipid membranes, which led to efficient

delivery to the cytosol and nucleus28,29. Other mechanisms for endosomal escape have been

proposed, including pore formation in the endosomal membrane and fusion into the lipid

bilayer of endosomes27. Which exact processes are involved in the endosomal escape and

nuclear translocation of both IFNγ and Fibroferon is an enigma and an intriguing question to

be further studied.

Johnson et al. described a non-canonical model of IFNγ-receptor signaling which involves

nuclear translocation of the receptor complex9. Upon binding of IFNγ to the extracellular

domain of the receptor, a series of events is induced in conjunction with endocytosis,

resulting in the active nuclear transport of a complex consisting of IFNγ, the IFNγR1, Stat1α,

Jak1 and Jak2. They stated that this process is under the influence of the intrinsic polycationic

nuclear localization sequence (NLS) in the C-terminus of IFNγ9. The present study is in

agreement with this. Fibroferon contains this NLS and nuclear localization of both Fibroferon

and IFNγ was demonstrated, as well as pStat1 translocation to the nucleus. Also other

research groups reported the ability of proteins, notably cytokines, such as all members of the

IL-1 family including IL-1α, IL-33 and IL-3730, to translocate to the nucleus facilitated by a

nuclear localization sequence. They possibly affect the gene expression there. Now we

demonstrated this for our redirected chimeric cytokine. Generally, molecules smaller than

approximately 45 kDa are allowed to diffuse passively from cytosol to nucleus31.

Macromolecules such as the complex of IFNγ, its receptor and signaling molecules, as

suggested to occur for IFNγ by Johnson et al.9, are transported in a selective, energy-

dependent manner through nuclear import proteins such as importin α and β32. Unravelment

Page 68: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

67

of these intracellular pathways may open fascinating ways to deliver proteins to the nucleus

in designated target cells.

Advantages of the use of chimeric peptide mimetics like Fibroferon as therapeutic proteins

include their versatility, high biological activity, high specificity for their targets, and low

toxicity9. There are already several peptide mimetics on the market, such as boceprevir for

the treatment of hepatitis C. An example of a cytokine-based chimeric protein is DAB389-IL2,

in which the catalytic and translocation domains of diphtheria toxin are fused to full length

IL233, which has been FDA-approved for treatment of human cutaneous T-cell lymphoma

patients. However, there are currently no clinical trials ongoing that test chimeric proteins

where the receptor-binding part of one cytokine is combined with the signaling part of

another cytokine.

In conclusion, we were able to deliver the IFNγ-signaling moiety to the PDGFβ-receptor and

obtained induction of the IFNγ-signaling pathway in fibroblasts. In addition, we found escape

from the endosomal pathway and nuclear localization of this chimeric construct. The

unraveling of the signaling and intracellular routing of Fibroferon and its associated

antifibrotic effects10 show the feasibility of steering cytokines to disease-specific target

receptors on pathogenic cells. This might form the basis of novel targeted therapies for the

treatment of chronic diseases like liver fibrosis, and in general increase the use of cytokines

for therapeutic purposes.

Acknowledgements

The authors thank dr. Inge Zuhorn and prof. dr. Sven van IJzendoorn for their valuable

suggestions on the uptake and trafficking studies and Pharmacy students Marjolein Bouwhuis

and Niek Breg for their practical contributions. Part of the work was performed at the UMCG

Imaging and Microscopy Center (UMIC). This study was performed using a grant of the

Netherlands Institute of Regenerative Medicine (NIRM) and a grant from NanoNext.NL

(program 03.10).

3

Page 69: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

68

References

1. Kinch MS. An overview of FDA-approved biologics medicines. Drug Discov Today.

2015;20(4):393-398.

2. Leader B, Baca QJ, Golan DE. Protein therapeutics: A summary and pharmacological

classification. Nat Rev Drug Discov. 2008;7(1):21-39.

3. Pisal DS, Kosloski MP, Balu-Iyer SV. Delivery of therapeutic proteins. J Pharm Sci.

2010;99(6):2557-2575.

4. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209-218.

5. Friedman SL. Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the

liver. Physiol Rev. 2008;88(1):125-172.

6. Wong L, Yamasaki G, Johnson RJ, Friedman SL. Induction of beta-platelet-derived growth

factor receptor in rat hepatic lipocytes during cellular activation in vivo and in culture. J

Clin Invest. 1994;94(4):1563-1569.

7. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

8. van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted therapies in liver fibrosis: Combining

the best parts of platelet-derived growth factor BB and interferon gamma. Front Med

(Lausanne). 2015;2:72.

9. Johnson HM, Noon-Song EN, Dabelic R, Ahmed CM. IFN signaling: How a non-canonical

model led to the development of IFN mimetics. Front Immunol. 2013;4:202.

10. Bansal R, Prakash J, De Ruiter M, Poelstra K. Interferon gamma peptidomimetic targeted

to hepatic stellate cells ameliorates acute and chronic liver fibrosis in vivo. J Control

Release. 2014;179:18-24.

11. Poosti F, Bansal R, Yazdani S, et al. IFNγ peptidomimetic targeted to interstitial

myofibroblasts attenuates renal fibrosis in unilateral ureteral obstruction in mice.

Oncotarget. 2016;Accepted.

12. Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy

of poxvirus infection and multiple sclerosis: An example of occam's broom? JAKSTAT.

2013;2(4):e26227.

13. Claudinon J, Monier MN, Lamaze C. Interfering with interferon receptor sorting and

trafficking: Impact on signaling. Biochimie. 2007;89(6-7):735-743.

14. Ross R, Raines EW, Bowen-Pope DF. The biology of platelet-derived growth factor. Cell.

1986;46(2):155-169.

15. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived

growth factor. Physiol Rev. 1999;79(4):1283-1316.

Page 70: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

69

16. Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci.

2002;7:d1720-6.

17. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted

interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology. 2011;54(2):586-596.

18. Poosti F, Bansal R, Yazdani S, et al. Selective delivery of IFN-gamma to renal interstitial

myofibroblasts: A novel strategy for the treatment of renal fibrosis. FASEB J.

2015;29(3):1029-1042.

19. Prakash J, de Jong E, Post E, Gouw AS, Beljaars L, Poelstra K. A novel approach to deliver

anticancer drugs to key cell types in tumors using a PDGF receptor-binding cyclic peptide

containing carrier. J Control Release. 2010;145(2):91-101.

20. Bansal R, Tomar T, Ostman A, Poelstra K, Prakash J. Selective targeting of interferon

gamma to stromal fibroblasts and pericytes as a novel therapeutic approach to inhibit

angiogenesis and tumor growth. Mol Cancer Ther. 2012;11(11):2419-2428.

21. Brodsky FM, Chen CY, Knuehl C, Towler MC, Wakeham DE. Biological basket weaving:

Formation and function of clathrin-coated vesicles. Annu Rev Cell Dev Biol. 2001;17:517-

568.

22. Filgueira L, Groscurth P, Aguet M. Binding and internalization of gold-labeled IFN-gamma

by human raji cells. J Immunol. 1989;142(10):3436-3439.

23. Marchetti M, Monier MN, Fradagrada A, et al. Stat-mediated signaling induced by type I

and type II interferons (IFNs) is differentially controlled through lipid microdomain

association and clathrin-dependent endocytosis of IFN receptors. Mol Biol Cell.

2006;17(7):2896-2909.

24. Sorkin A, Eriksson A, Heldin CH, Westermark B, Claesson-Welsh L. Pool of ligand-bound

platelet-derived growth factor beta-receptors remain activated and tyrosine

phosphorylated after internalization. J Cell Physiol. 1993;156(2):373-382.

25. Liu P, Ying Y, Ko YG, Anderson RG. Localization of platelet-derived growth factor-

stimulated phosphorylation cascade to caveolae. J Biol Chem. 1996;271(17):10299-

10303.

26. Morelon E, Dautry-Varsat A. Endocytosis of the common cytokine receptor gammac

chain. identification of sequences involved in internalization and degradation. J Biol

Chem. 1998;273(34):22044-22051.

27. Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of

biologicals. J Control Release. 2011;151(3):220-228.

28. Damante G, Pellizzari L, Esposito G, et al. A molecular code dictates sequence-specific

DNA recognition by homeodomains. EMBO J. 1996;15(18):4992-5000.

3

Page 71: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

70

29. Derossi D, Calvet S, Trembleau A, Brunissen A, Chassaing G, Prochiantz A. Cell

internalization of the third helix of the antennapedia homeodomain is receptor-

independent. J Biol Chem. 1996;271(30):18188-18193.

30. Ross R, Grimmel J, Goedicke S, et al. Analysis of nuclear localization of interleukin-1

family cytokines by flow cytometry. J Immunol Methods. 2013;387(1-2):219-227.

31. Jans DA, Hubner S. Regulation of protein transport to the nucleus: Central role of

phosphorylation. Physiol Rev. 1996;76(3):651-685.

32. Subramaniam PS, Larkin J,3rd, Mujtaba MG, Walter MR, Johnson HM. The COOH-

terminal nuclear localization sequence of interferon gamma regulates STAT1 alpha

nuclear translocation at an intracellular site. J Cell Sci. 2000;113 ( Pt 15)(Pt 15):2771-

2781.

33. Duvic M, Cather J, Maize J, Frankel AE. DAB389IL2 diphtheria fusion toxin produces

clinical responses in tumor stage cutaneous T cell lymphoma. Am J Hematol.

1998;58(1):87-90.

Page 72: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Steering therapeutic proteins

71

Supplementary data

Figure S1. Stability of Fibroferon. (A) Stability of Fibroferon 7, 14 and 21 days after dissolving it in PBS was

determined with UPLC, which showed a constant retention factor (Rf) over the entire period of the experiment,

indicating stability over longer periods. (B) Additionally, the stability of Fibroferon was confirmed by ES-MS.

Figure S2. Uptake inhibition of Fibroferon. (A) Flow cytometry analysis revealed the inhibition in uptake of

Fibroferon-A647 and IFNγ-A555 upon NaN3 preincubation, as reflected by the significantly decreased MFI (n=3).

Statistics with paired t test.

3

Page 73: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 3

72

Still of supplementary movie 1. Nuclear translocation of Fibroferon and IFNγ. Movie of confocal microscopic

photographs of Fibroferon-A555 (red) and IFNγ-A488 (green) after 120 min incubation of fibroblasts (630x),

demonstrating nuclear localization of both compounds. The experiment was done in mouse fibroblasts

(NIH/3T3).

Page 74: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 75: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

4

Page 76: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

N. Teekampa,*, F. van Dijka,b,*, A. Broesdera, M. Eversa, J. Zuidemac, R. Steendamc,

E. Postb, J.L. Hillebrandsd, H.W. Frijlinka, K. Poelstrab, L. Beljaarsb, P. Olingaa and

W.L.J. Hinrichsa

* The authors contributed equally

aGroningen Research Institute of Pharmacy, Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen, Groningen, The

Netherlands, bGroningen Research Institute of Pharmacy, Department of

Pharmacokinetics, Toxicology and Targeting, University of Groningen,

Groningen, The Netherlands, cInnoCore Pharmaceuticals, Groningen, The

Netherlands, dDepartment of Pathology and Medical Biology, University of

Groningen, University Medical Center Groningen, Groningen, The Netherlands.

Published in International Journal of Pharmaceutics, 2017

Polymeric microspheres for the sustained

release of a protein-based drug carrier

targeting the PDGFβ-receptor

in the fibrotic kidney

Page 77: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

76

Abstract

Injectable sustained release drug delivery systems are an attractive alternative for the

intravenous delivery of therapeutic proteins. In particular, for chronic diseases such as

fibrosis, this approach could improve therapy by reducing the administration frequency while

avoiding large variations in plasma levels. In fibrotic tissues the platelet-derived growth factor

receptor beta (PDGFR) is highly upregulated, which provides a target for site-specific

delivery of drugs. Our aim was to develop an injectable sustained release formulation for the

subcutaneous delivery of the PDGFR-targeted drug carrier protein pPB-HSA, which is

composed of multiple PDGFR-recognizing moieties (pPB) attached to human serum albumin

(HSA). We used blends of biodegradable multi-block copolymers with different swelling

degree to optimize the release rate using the model protein HSA from microspheres

produced via a water-in-oil-in-water double emulsion evaporation process. The optimized

formulation containing pPB-HSA, showed complete release in vitro within 14 days. After

subcutaneous administration to mice suffering from renal fibrosis pPB-HSA was released from

the microspheres and distributed into plasma for at least 7 days after administration.

Furthermore, we demonstrated an enhanced accumulation of pPB-HSA in the fibrotic kidney.

Altogether, we show that subcutaneously administered polymeric microspheres present a

suitable sustained release drug delivery system for the controlled systemic delivery for

proteins such as pPB-HSA.

Page 78: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

77

Introduction

Organ fibrosis is a progressive and chronic condition that is hallmarked by excessive

deposition of extracellular matrix (ECM) proteins by myofibroblasts. Ultimately, functional

cells are replaced by redundant ECM proteins, which causes scarring of the affected organ,

leading to impaired organ function with a high mortality of up to 45% in the developed

world1. Currently, the therapeutic options to treat advanced fibrosis are very limited,

demonstrating the necessity for the development of innovative therapies to attenuate

fibrosis.

A promising strategy in the development of a novel antifibrotic therapy is the targeting of

therapeutic proteins to key pathogenic cells, mainly myofibroblasts. These cells highly and

specifically express the platelet-derived growth factor beta receptor (PDGFβR)2,3, which

makes it an excellent target for the delivery of potential antifibrotic compounds. The drug

carrier protein pPB-HSA is composed of multiple PDGFβR-recognizing peptide moieties (pPB)

coupled to human serum albumin (HSA)4, and binds to the PDGFβR without activating the

downstream intracellular signaling pathway. The potential of pPB-HSA as a carrier protein was

demonstrated previously by the potent antifibrotic effect when small molecules such as

doxorubicin or proteins like interferon gamma were coupled to it4-7. It is hypothesized that

after binding of the pPB-moiety to the PDGFβR, the whole construct is internalized and the

antifibrotic compound is released from the construct after lysosomal degradation6,7.

The most common route of administration for proteins such as pPB-HSA is an (intravenous)

injection, as this provides the most efficient delivery by ensuring complete bioavailability.

However, the fast elimination of proteins causes large variations in plasma levels. Moreover,

such rapid elimination makes frequent injections necessary, which creates a high burden to

the patient. To overcome these issues, subcutaneously or intramuscularly injectable sustained

release drug delivery systems providing sustained release are increasingly used. Advantages

of this type of drug delivery system are that the administration frequency is reduced and that

a constant protein plasma level can be achieved. Biodegradable polymers are excellent

matrices for such drug delivery systems; they offer a versatile platform for a multiplicity of

release profiles and dosage forms8,9.

A frequently applied and FDA approved biodegradable polymer for sustained release is poly

(lactic-co-glycolic acid) (PLGA), which degrades in the body by hydrolysis10. However, the use

of PLGA for protein delivery may lead to changes in protein structure and incomplete release

related to, amongst others, its hydrophobicity and its acidic degradation products10,11.

Moreover, the release from PLGA matrices is influenced by many factors, which results in

unpredictable and often unfavorable, multi-phasic release profiles12. As an alternative, phase-

4

Page 79: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

78

separated multi-block copolymers composed of amorphous hydrophilic poly(ε-caprolactone)

− poly (ethylene glycol) − poly(ε-caprolactone) (PCL-PEG-PCL) blocks combined with semi-

crystalline poly(L-lactic acid) (PLLA) blocks can be used. In contrast to PLGA, the hydrophilic

nature of these polymers allows continuous release by diffusion, caused by controlled

swelling of PEG in the amorphous blocks by water uptake13. By varying the size of the PEG

blocks, the ratio of the blocks within the copolymer or by blending different copolymers, the

release of proteins can be customized to the desired characteristics needed for a specific

protein14.

In this study, we used these [PCL-PEG-PCL]-b-[PLLA] multi-block copolymers to prepare

microspheres, thereby aiming for the sustained release of the drug carrier pPB-HSA. As an in

vivo proof of concept, we assessed the release of pPB-HSA from the microspheres up to 7

days after subcutaneous administration of pPB-HSA containing microspheres in mice suffering

from kidney fibrosis.

Materials and methods

Polymer synthesis and characterization

The prepolymers PLLA and PCL-PEG-PCL were synthesized using procedures similar to

described in13. The PLLA prepolymer with a target molecular weight of 4000 g/mol was

prepared of 1001 g (13.89 mol) anhydrous L-lactide (Corbion, Gorinchem, The Netherlands),

using anhydrous 1,4-butanediol (22.7 g, 251.9 mmol, Thermo Fisher Scientific, Waltham, MA,

USA) to initiate the ring-opening polymerization and stannous octoate (Sigma Aldrich,

Zwijndrecht, The Netherlands) as a catalyst at a catalyst/monomer molar ratio of 5.38 × 10-

5/1. The mixture was magnetically stirred for 136 h at 140 ˚C and subsequently cooled to

room temperature.

The [PCL-PEG1000- PCL] prepolymer with a target molecular weight of 2000 g/mol and the

[PCL-PEG3000-PCL] prepolymer with a target molecular weight of 4000 g/mol were synthesized

in a similar way using 250 g (2.19 mol) CL (Thermo Fisher Scientific), 250 g (250 mmol) PEG1000

(Thermo Fisher Scientific) and molar catalyst/monomer ratio of 7.94 × 10-5 /1 for [PCL-

PEG1000-PCL] and 69.6 g (0.61 mol) CL, 229 g (76.33 mmol) PEG3000 (Thermo Fisher Scientific)

and molar catalyst/monomer ratio of 3.03 × 10-4 /1 for [PCL-PEG3000-PCL]. The mixture was

magnetically stirred at 160 ˚C for 24 h ([PCL-PEG1000-PCL]) or 12 days ([PCL-PEG3000-PCL]) and

subsequently cooled to room temperature.

Page 80: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

79

The [PLLA] was then chain-extended with [PCL-PEG1000 or 3000-PCL] using 1,4-

butanediisocyanate to prepare x[PCL-PEG1000 or 3000-PCL]-y[PLLA] multi-block copolymer where

x/y is the [PCL-PEG1000 or 3000-PCL]/[PLLA] weight ratio, being 50/50 with PCL-PEG1000 (referred

to as polymer A in this paper) or 30/70 with PCL-PEG3000 (referred to as polymer B in this

paper) (Table S1). [PLLA] and [PCL-PEG1000 or 3000-PCL] were dissolved in dry 1,4-dioxane (80 ˚C,

30 wt-% solution). 1,4-Butanediisocyanate (Actu-all Chemicals BV, Oss, The Netherlands) was

added and the reaction mixture was mechanically stirred for 20 h. Subsequently, the reaction

mixture was frozen and freeze-dried at 30 ˚C shelf temperature to remove 1,4-dioxane.

The synthesized multi-block copolymers 50[PCL-PEG1000-PCL]-50[PLLA]/polymer A and 30[PCL-

PEG3000-PCL]-70[PLLA]/polymer B were analyzed for chemical composition, intrinsic viscosity

and residual 1,4-dioxane content (Table S2). The actual composition of the multi-block

copolymers, as determined by 1H-NMR from LA/PEG and CL/PEG molar ratios, was in

agreement with the target composition. The residual 1,4-dioxane contents were well below

600 ppm indicating effective removal of the solvent by freeze-drying. A schematic

representation of composition of the multi-block copolymers is displayed in figure 1.

Figure 1. Schematic representation of the composition of the two semi crystalline multi-block copolymers used in

this study. The amorphous and crystalline blocks are randomly distributed. PLLA, poly(L-lactic acid); PCL, poly(ε-

caprolactone); PEG, poly(ethylene glycol).

Synthesis of pPB-HSA

The cyclic peptide pPB was covalently coupled to HSA as described previously15. In brief, 15

µmol N--maleimidobutyryl-oxysuccinimide ester in dry dimethylformamide (DMF) was added

to 0.75 µmol HSA (purified from Cealb®, Sanquin, Amsterdam, The Netherlands) in PBS (10

mM, pH 7.2 in all experiments). After dialysis for 2 d against PBS using a 10 kDa cut-off dialysis

membrane (Thermo Fisher Scientific), 15 µmol of N-succinimidyl S-acetylthioacetate (SATA)-

modified pPB (C*SRNLIDC*, 20 mg/ml in dry DMF, Ansynth Service BV, Roosendaal, The

Netherlands) and activation solution (1.0 mmol hydroxylamine and 12.7 µmol EDTA in PBS)

4

Page 81: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

80

were added. This mixture was allowed to react overnight at room temperature and was

extensively dialyzed afterwards. Next, the product was purified using chromatographic

methods and then freeze dried and stored at -20 ˚C. MALDI-TOF MS analysis showed an

average molecular weight of 73.4 kDa, corresponding to 7 pPB units coupled per HSA

molecule.

Microsphere production

Microspheres were produced using a water-in-oil-in-water double emulsion

extraction/evaporation method. The polymers (1 g in total) were dissolved in

dichloromethane (DCM) in the desired ratio (Table 1) to obtain a 15 wt-% solution, which was

subsequently filtered (PTFE, 0.2 μm). Next, to make the primary emulsion, PBS (control) or a

solution of 80 mg/mL protein (HSA or pPB-HSA and HSA in a 3:2 ratio) in PBS was added to the

polymer solution to obtain 5 wt-% theoretical protein load and homogenized for 40 s at

13,500 rpm using a turrax mixer (Heidolph Diax 600, Salm & Kipp, Breukelen, The

Netherlands). To make the secondary emulsion, the primary emulsion was added in 40 s to an

aqueous solution containing 4 wt-% poly vinyl alcohol (Mw: 13-23 kDa, 87-89% hydrolyzed,

Sigma Aldrich) and 5 wt-% NaCl solution (1:100 v/v ratio) under stirring (19,000 rpm) also

using a turrax mixer. Consecutively, the secondary emulsion was mixed for an additional 20 s

under these conditions and then stirred at 200 rpm with a magnetic stirrer for 3 h to

evaporate DCM. Next, the hardened microspheres were collected by filtration and washed

with 750 mL of 0.05% Tween 80 in Millipore water and 750 mL of Millipore water. Finally, the

microspheres were freeze dried.

Page 82: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

81

Table 1. Characteristics of HSA microspheres of different polymer ratios. X10, X50 and X90 represent the volume

percentages of particles (10%, 50% and 90% undersize, respectively).

† For clarity reasons, individual standard deviations are not displayed. The maximum standard

deviations are: 0.3 µm for X10, 1.7 µm for X50 and 1.6 µm for X90.

Microsphere size analysis

Laser diffraction (Helos/BF, Sympatec GmbH, Clausthal-Zellerfeld, Germany) was performed

with a 100 mm lens (range: 0.5/0.9-175 m). In brief, 12-15 mg of microspheres were

dispersed in duplo in 1 mL of Millipore water by ultra-sonication. Next, 0.2 mL of the

suspension was dispersed in 40 mL Millipore water in a 50 mL quartz cuvette. Three single

measurements of 10 s were recorded with a 50 s pause in between. The last step was

repeated with another 0.2 mL of suspension, resulting in a total of 12 single measurements

per microsphere batch. The particle size distribution was calculated according to the

Fraunhofer diffraction theory. The span of the particle size distribution was calculated using

Eq. 1,

𝑆𝑝𝑎𝑛 = 𝑋10−𝑋90

𝑋50 Equation 1

where X10, X50 and X90 represent the volume percentages of particles (10%, 50% and 90%

undersize, respectively).

Ratio polymer A:

polymer B

Particle size†

X10 X50 X90

Span

Encapsulation

efficiency (%)

100:0 3.6 22.4 50.4 2.1 91

90:10 2.8 18.6 55.6 2.8 85

70:30 4.7 27.8 57.7 1.9 39

50:50 6.7 26.4 52.0 1.8 76

30:70 5.7 27.4 72.4 2.4 92

10:90 3.1 20.4 51.0 2.4 67

0:100 2.3 13.6 46.9 3.3 86

4

Page 83: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

82

Protein content of microspheres

Microsphere samples of 5 mg were accurately weighed in triplicate in 4.0 mL glass vials. Next,

0.4 mL of dimethyl sulfoxide was added and the samples were placed at 37 °C. After 3 h, 3.6

mL 0.5 wt-% sodium dodecyl sulfate (SDS) in 0.05 N NaOH was added, and the polymer was

allowed to dissolve overnight. Next, the protein content was determined with the

bicinchoninic acid (BCA) assay. The BCA reagent mixture was prepared by mixing 4 wt-%

aqueous copper (II) sulfate solution with BCA reagent A (Thermo Fisher Scientific) in a 1:50

volume ratio. Samples of 25 µL were added in triplicate to a 96-wells plate. After addition of

200 µL BCA reagent to the wells, the plate was incubated for 2 h at 37 ˚C. The absorbance was

measured at 562 nm after the plate was cooled to room temperature (Synergy HT Microplate

Reader, BioTek Instruments, Winooski, VT, USA). Protein concentrations were calculated

using an 8-point calibration curve.

The protein content was used to calculate the encapsulation efficiency (EE) according to Eq.

2:

𝐸𝐸 = 𝑊𝑒𝑖𝑔ℎ𝑡 𝑜𝑓 𝑒𝑛𝑐𝑎𝑝𝑠𝑢𝑙𝑎𝑡𝑒𝑑 𝑝𝑟𝑜𝑡𝑒𝑖𝑛

𝑊𝑒𝑖𝑔ℎ𝑡 𝑜𝑓 𝑡𝑜𝑡𝑎𝑙 𝑝𝑟𝑜𝑡𝑒𝑖𝑛 𝑢𝑠𝑒𝑑× 100% Equation 2

Scanning Electron Microscopy

Images were obtained using a JSM-6460 microscope (Jeol, Tokio, Japan) at an acceleration

voltage of 10 kV. Samples were fixed on an aluminum sample holder using double sided

adhesive carbon tape. Excessive microspheres were removed using pressurized air. The

samples were sputter coated with 10 nm of gold.

In vitro release

The in vitro release was measured in triplicate by a sample-and-replace method. In brief, 10

mg of microspheres were accurately weighed in a 2.0 mL glass vial and suspended in 1.0 mL

of release buffer (100 mM sodium phosphate, 9.1 mM NaCl, 0.01 wt-% Tween 80, 0.02 wt-%

NaN3, pH 7.4). The vials were placed in a 37 °C shaking water bath. Samples of 0.8 mL were

taken at predetermined time points and replaced by fresh buffer. At the final time point, the

whole volume of buffer was taken to facilitate facile drying of the remaining microspheres for

scanning electron microscopy. Total protein concentration in the release medium was

determined using the BCA assay (section 2.5). Protein concentrations were calculated using a

13-point calibration curve.

Page 84: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

83

pPB-HSA ELISA

The concentration of pPB-HSA in the in vitro release medium samples was determined using

an in-house developed sandwich ELISA. Briefly, the capture antibody rabbit α-pPB (100 μL,

1:1000, custom prepared by Charles Rivers, Den Bosch, The Netherlands) was incubated

overnight in a high protein binding 96-well plate (Corning, New York, NY, USA). After extensive

washing with PBS containing 0.5 wt-‰ Tween-20 (PBS-T), the plate was blocked with 5 wt-%

nonfat dry milk in PBS-T (200 μL) for 1 h and washed again with PBS-T. Next, samples (100 μl)

were incubated for 2 h. The plate was washed again, followed by the addition of the

detection antibody goat α-HSA (100 μL, 1:8000, ICN Biomedicals, Zoetermeer, The

Netherlands) for 1 h and subsequent washed once more. The appropriate HRP-conjugated

secondary antibody was applied for 1 h, and after washing with PBS-T, the substrate

tetramethyl benzidine (100 μL, R&D Systems, Minneapolis, MN, USA) was added. The

absorbance was measured at 450 nm (THERMOmax microplate reader, Molecular Devices,

Sunnyvale, CA, USA) after addition of 50 μL 2 N H2SO4. This protocol was also used to

determine the concentration of pPB-HSA in 50 μl mouse plasma samples.

Modulated Differential Scanning Calorimetry (MDSC)

Samples of 6 to 7 mg of microspheres were analyzed in duplicate in open aluminum pans

using a Q2000 MDSC (TA Instruments). The samples were cooled to -80 °C and kept

isothermal for 5 min to equilibrate. Next, the sample was heated to 150 °C at 2 °C/min and a

modulation amplitude of ± 0.212 °C every 40 s. Differences within duplicates were <0.5 °C for

melting and crystallization temperatures and <1.5 J/g for enthalpies.

Animal experiments

All the experimental protocols for animal studies were approved by the Animal Ethical

Committee of the University of Groningen (The Netherlands). Male C57BL/6 mice, aged 8-10

weeks, were obtained from Envigo (Horst, The Netherlands). Animals received ad libitum

normal diet with a 12 h light/dark cycle. Mice (n=6) were subjected to unilateral ureteral

obstruction (UUO) by a double ligation of the left ureter proximal to the kidney3, and injected

subcutaneously in the neck directly after surgery with 31.5 mg microspheres (dispersed in 500

μl 0.4 w/v% carboxymethyl cellulose (Aqualon high Mw, Ashland)) containing either 5 wt-%

HSA (n=3) or 3 wt-% pPB-HSA/2 wt-% HSA (n=3). The total administered doses were 1.58 mg

HSA for the microspheres containing 5 wt-% HSA and 0.95 mg pPB-HSA/0.63 mg HSA for the

4

Page 85: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

84

microspheres containing 3 wt-% pPB-HSA/2 wt-% HSA. Mice were sacrificed at day 7, after

which blood and different organs were collected and processed for further analysis.

Histochemistry

Cryosections of neck skin tissue were cut at a thickness of 4 μm (CryoStar NX70 cryostat,

Thermo Fisher Scientific), dried and stored at -20 °C until analysis. Paraffin sections for kidney

were cut at a thickness of 4 μm (Leica Reichert-Jung 2040 microtome).

Haematoxylin and eosin staining. The cryosections were dried and fixed for 10 min in

formalin-macrodex (6 wt-% dextran-70 in 0.9 wt-% NaCl containing 3.6 wt-% formaldehyde

and 1 wt-% CaCl2, pH 7.4). After extensive washing in water, the slides were incubated in

haematoxylin solution (Clinipath Pathology, Osborne Park, Australia) for 15 min, washed in

tap water, and incubated 1.5 min in eosin (Clinipath Pathology). Sections were embedded in

DePeX mounting medium (VWR, Amsterdam, The Netherlands) after dehydration in ethanol.

Immunohistochemical stainings. Neck skin cryosections were dried and fixed with acetone.

Kidney paraffin sections were deparaffinized in xylene and ethanol. Sections were then

rehydrated in PBS and incubated 1 h at room temperature with a primary antibody (rabbit α-

HSA (1:1500, ICN Biomedicals), rabbit α-pPB (1:1000, Charles Rivers), goat α-collagen I&III

(both 1:200 + 5% normal mouse serum, Southern Biotech, Birmingham, AL, USA)) or boiled in

10 mM Tris/ 1 mM EDTA (pH 9.0) for 15 minutes prior to overnight incubation with a primary

antibody at 4 °C (rabbit α-PDGFβR (1:50, Cell Signaling, Leiden, The Netherlands)).

Subsequently, sections were incubated 30 min at room temperature with the appropriate

HRP-conjugated secondary antibody. The HRP-conjugated antibodies were visualized with

ImmPACT NovaRED kit (Vector, Burlingame, CA, USA). Hematoxylin counterstaining was

performed. Digital photomicrographs were captured at 400x magnification (Aperio,

Burlingame, CA, USA). Microsphere sizes (median of 100) were determined using Cell D

software (Olympus).

Western blot

Samples (100 μg protein, as determined using Lowry assay) were applied on a SDS

polyacrylamide gel (10%) and transferred to a polyvinylidene difluoride membrane.

Membranes were blocked for 1 h in 5 wt-% nonfat dry milk in tris-buffered saline/0.1%

Tween-20 (ELK/TBS-T) at room temperature, followed by overnight incubation at 4 °C with

the primary antibody in ELK/TBS-T. The primary antibodies used include goat α-HSA (1:1000,

Page 86: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

85

ICN Biomedicals) and mouse α-GAPDH (1:20000, Sigma-Aldrich). The membranes were

extensively washed in TBS-T before application of the appropriate HRP-conjugated secondary

antibodies in ELK/TBS-T for 2 h. After extensive washing of the membranes with subsequently

TBS-T and TBS, bands were visualized with enhanced chemiluminescence and quantified with

GeneSnap (Syngene, Synoptics, Cambridge, UK).

Statistical analyses

At least 3 individual experiments were performed to measure in vitro effects. All the data are

represented as mean ± standard deviation (SD). In vivo data are presented as mean ±

standard error of the mean (SEM). Differences between groups for the ELISA were assessed

by Mann-Whitney U-test. The differences between the groups for the western blot were

assessed by Kruskal-Wallis test followed by Dunn’s multiple comparison test. The graphs and

statistical analyses were performed using Graphpad Prism version 6.0 (GraphPad Prism

Software Inc., La Jolla, CA, USA).

Results

Properties of microspheres of different polymer ratios

Different blend ratios of polymer A and B were used to obtain a microsphere formulation with

the desired release profile, i.e. sustained release with minimal burst and complete release

within 14 days. In these screening experiments, the HSA target loading of the microspheres

was 5%. This protein was later used as filler and untargeted equivalent to pPB-HSA, as the

physicochemical properties of both proteins can be considered similar. The polymer blend

ratios affected the microsphere size distribution and the span of the size distribution (Table

1), as determined with laser diffraction. In particular, microspheres consisting of only polymer

B had a smaller median particle size and a larger size distribution span than microspheres of

other compositions. All formulations had a broad particle size distribution, which is reflected

in the relatively high span values. Such high polydispersity is common for microspheres

produced by water-in-oil-in-water (W/O/W) emulsification by homogenization16. The span

could be decreased at the expense of the particle size, however this negatively affected the

injectability of the microspheres. The encapsulation efficiency (EE) of the model protein HSA

was consistently high (albeit one exception, for unknown reasons), and seemed to be

unaffected by polymer blend composition and particle size.

4

Page 87: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

86

Scanning electron microscopy images revealed the morphology of the microspheres prepared

from all polymer blends as spherical particles with a smooth surface and few pores. A

representative image of lyophilized microspheres composed of a 50:50 polymer blend is

depicted in figure 2. The microspheres retained their spherical shape and smooth surface

during 42 days of release in vitro, but additional pores were formed in time (Fig. S1).

Figure 2. Representative scanning electron microscopy image at a 1000× (panel A) and 5000× (panel B)

magnification of lyophilized microspheres composed of a 50:50 polymer blend of polymer A and B.

Thermal properties of microspheres

The thermal characteristics of the polymer matrix of microspheres composed of different

polymer blends without protein were investigated using MDSC (Fig. 3, Tables 2 and 3). Glass

transitions were observed in the reversing heat flow signal at around -56 ˚C in microspheres

of all polymer ratios except 10:90 and 0:100 (Fig. S2). The peak at 37 ˚C in the total heatflow

thermograms (Fig. 3) can be ascribed to melting of crystalline PEG. With increasing content of

polymer B, the enthalpy of the melting peak at 37 C increased, even though the total PEG

content is decreased (Table 2). This result indicates that PEG3000 is crystalline, but PEG1000

might be too small to crystallize. Furthermore, the cold crystallization temperature Tcc of PLLA

shifted from around 90 C for polymer blends with a high polymer A content to around 85 C

for polymer blends with a low polymer A content (Fig. 3, Table 3). Also, the melting

temperature Tm of PLLA showed a minor decrease with decreasing polymer A content (Table

3). The origin of the shoulder in the PLLA melting peak is unknown, but such thermal events

have been observed before in microspheres prepared from polymers of the same platform

technology17.

Page 88: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

87

Table 2. Theoretical PEG content and thermal characteristics of microspheres of different polymer blends as

determined with MDSC. The relative enthalpy (Hrel) was calculated by dividing the melting enthalpy (J/g) by the

PEG fraction in the copolymer.

Table 3. Theoretical PLLA content and thermal characteristics of microspheres of different polymer blends as

determined by MDSC. The relative enthalpy (Hrel) was calculated by dividing the melting enthalpy (J/g) minus

the crystallization enthalpy (J/g) by the PLLA fraction in the copolymer.

Polymer

ratio

Total PEG

(%)

PEG 1 kDa

(%)

PEG 3 kDa

(%)

Tm (°C)

Hrel

100:0 25 25 0 N.A. N.A.

90:10 24.75 22.5 2.25 37.1 0.37

70:30 24.25 17.5 6.75 37.2 2.04

50:50 23.75 12.5 11.25 37.6 3.04

30:70 23.25 7.5 15.75 37.0 9.12

10:90 22.75 2.5 20.25 37.4 40.97

0:100 22.5 0 22.5 37.7 43.18

Polymer

ratio

Total PLLA

(%)

PLLA

polymer A

(%)

PLLA

polymer B

(%)

Tcc (°C)

Tm (°C)

Hrel

100:0 50 50 0 89.2 133.2 5.0

90:10 52 45 7 90.3 133.1 13.7

70:30 56 35 21 89.4 132.7 15.0

50:50 60 25 35 87.5 132.9 8.5

30:70 64 15 49 85.9 132.6 7.9

10:90 68 5 63 86.0 132.5 6.9

0:100 70 0 70 85.9 132.3 5.4

4

Page 89: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

88

Figure 3. Thermograms of microspheres of different polymer blends. On the y-axis, exothermic is up. The melting

peak of PEG is observed at about 37 °C, the cold crystallization peak of PLLA at 85–90 °C and the melting peak of

PLLA at 132–133 °C.

Release of HSA from microspheres of different polymer ratios

The release of HSA from microspheres containing HSA only and composed of different

polymer ratios during the first 14 days is presented in figure 4. This is the relevant timeframe

for the subsequent in vivo study. Protein release is assumed to be diffusion controlled, as was

found previously using similar phase-separated multi-block copolymers18.

Only a minimal burst release of less than 10% in 3 h was observed in all formulations. The

release profiles of the microspheres composed of the different polymer blends can roughly be

categorized in three sets. Firstly, microspheres with a high content of polymer A (90 or 100

%), showed almost no release of HSA apart from a small burst. Secondly, microspheres

containing 0, 10, 30 and 70 % of polymer A showed an intermediate release rate, with a

cumulative release of 40% to 60% after 14 days. Thirdly, the fastest release rate was observed

from the 50:50 polymer ratio with a cumulative release of 87% after 14 days. Clearly, the

release rates of HSA did not follow the polymer blend composition linearly.

Page 90: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

89

Figure 4. Release of HSA from 5 wt-% HSA microspheres with different polymer ratios.

Reproducibility of the production process

The reproducibility of the production process was assessed using 6 batches of HSA

microspheres with a 50:50 polymer ratio, produced using identical process conditions. The

average median particle size of these batches was 23.4 ± 5.3 µm (span: 2.0) and the average

EE was 82 ± 7%. The normalized release showed the same release rate for all batches with a

cumulative HSA release after 14 days of 88 ± 8% (Table S3, Fig. S3).

Production and characterization of pPB-HSA microspheres

For PDGFβR targeted HSA, a protein load of 3 wt-% was considered to be sufficient for cell

specific delivery. Therefore, the required protein load of pPB-HSA was complemented with

HSA to a total of 5 wt-% protein load to replicate the release rate and EE from the optimized 5

wt-% HSA microspheres with a 50:50 polymer ratio.

The morphology of these pPB-HSA microspheres was comparable with the morphology of

HSA microspheres (Fig. 5A). The median particle size of 24.7 µm was similar to the particle

size of HSA microspheres, while the span of the particle size distribution was slightly lower at

a value of 1.6 (Fig. 5B). The in vitro release rate of pPB-HSA from pPB-HSA microspheres (EE:

83%) showed a similar release profile as HSA from HSA microspheres (Fig. 5C) and had a

cumulative release of 103 ± 5% after 14 days. Evidently, the size difference between HSA and

pPB-HSA (~7 kDa) did not affect the release rate.

4

Page 91: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

90

Figure 5. Morphology, particle size distribution, and in vitro release of pPB-HSA microspheres. (A) Representative

scanning electron microscopy image of pPB-HSA microspheres after freeze drying, 1000× magnification. (B)

Particle size distribution of microspheres with volume percentage on left y-axis (solid line) and cumulative

volume percentage on right y-axis (dashed line). (C) Cumulative in vitro release of pPB-HSA from pPB-HSA

microspheres during 14 days, determined using ELISA and corrected for EE.

Microspheres in vivo

The microspheres composed of the 50:50 polymer blend containing either pPB-HSA or its

untargeted equivalent (HSA) demonstrated an optimal in vitro release profile, and were

selected to be tested in vivo. Before this, we confirmed both in vitro in myofibroblasts and ex

vivo in mouse kidney slices that the microspheres itself did not show any toxicity, as reflected

by the unaffected viability in the presence of microspheres in different dilutions (Fig. S4). We

injected the microspheres subcutaneously in the neck of mice suffering from fibrosis in their

left kidney induced by unilateral ureter obstruction (UUO). Seven days after microsphere

administration, we dissected the skin at the site of injection and performed a haematoxylin

and eosin staining and confirmed the presence of microspheres. The microspheres had a

mean diameter of 25.9 μm (Fig. 6A), which is in accordance with the size found in vitro by

laser diffraction. Immunohistochemical staining for both HSA and pPB revealed that the

subcutaneously located microspheres still contained HSA and/or pPB-HSA (Fig. 6B).

Page 92: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

91

Figure 6. Subcutaneous localization of microspheres in vivo. (A) Haematoxylin and eosin staining of mouse neck

skin at the site of microsphere injection, and quantification of the size of the microspheres. M denotes

microspheres. (B) Immunohistochemical stainings for HSA and pPB of the skin of mice that received

microspheres containing HSA or pPB-HSA.

Sustained release into the systemic circulation of intact pPB-HSA from the microsphere depot

was demonstrated. Plasma concentrations of 15.7±4.0 ng pPB-HSA/ml were measured at day

7 after injection as determined by ELISA (Fig. 7B). In addition, the presence of pPB-HSA

specifically in the fibrotic kidney, which showed high expression of collagens I & III and the

PDGFβR (Fig. 7A), was demonstrated with western blot analysis for HSA (Fig. 7C). Clearly, pPB-

HSA accumulated to a higher extent in the fibrotic kidney as compared to the healthy kidney.

Inherent to the surgical procedure, some untargeted HSA was trapped in the fibrotic kidney

as well, albeit to a lesser extent.

4

Page 93: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

92

Figure 7. In vivo release of pPB-HSA from microspheres, and localization of pPB-HSA in the UUO model. (A)

Immunohistochemical stainings of sham and UUO-operated kidneys for collagen I&III and PDGFβR. (B) ELISA

analysis of the plasma levels of pPB-HSA 7 days after subcutaneous injection with microspheres containing HSA

or pPB-HSA. (C) Representative bands and quantitative analysis of western blot for HSA on kidney homogenates

of mice 7 days after microsphere injection.

Discussion

In this study, we developed sustained release microspheres containing pPB-HSA, a

proteinaceous construct targeted to the PDGFβR expressed in fibrotic tissues including the

kidneys. The release rate was controlled by blending two semi-crystalline multi-block

copolymers yielding a 14-day sustained release formulation. Microspheres composed of a

50:50 polymer blend containing pPB-HSA or HSA were subcutaneously administered to mice

suffering from kidney fibrosis. After 7 days in vivo, the microspheres still released pPB-HSA

and the construct was localized in the fibrotic kidney, thereby showing the therapeutic

potential of these microspheres.

Page 94: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

93

We expected the release rate of proteins from polymer blends to follow a linear trend, i.e. an

increasing release rate with increasing content of the more swellable polymer in the blend.

Surprisingly, this was not the case for the two semi-crystalline multi-block copolymers we

used in this study. Unfortunately, the thermal properties of the 50:50 polymer blend

microspheres did not display a distinguished profile that could provide a definitive answer to

the unconventional trend in release rate. Therefore, to explain the anomalous release from

the 50:50 polymer blend, we hypothesize that the release is governed by the size and content

of the PEG blocks, which will be further described below.

The release from the two semi-crystalline multi-block copolymers was diffusion controlled, as

in the timeframe of the experiments no extensive degradation of the microspheres will have

occurred18. Furthermore, the release rate of proteins from this type of polymers is usually

governed by the content of the PEG blocks which swell by water uptake14,18. A higher PEG

content results in an increased degree of swelling, which causes the release rate of proteins

to increase18. Although differences were small, based on the total PEG content in the polymer

blends in this study, we could expect the highest release rate from microspheres prepared

from 100% polymer A (Table 1). However, properties regarding release rate possibly changed

because of the difference in PEG block length between polymer A and B. Stankovic et al.

showed that diffusion controlled release of high molecular weight proteins such as albumin is

not possible in polymers with PEG block lengths of 1 and 1.5 kDa, even at very high

contents14,18, as the release of albumin was limited to a burst release. Furthermore, Tran et al.

showed that an increase of PEG domain size also accelerates release of proteins19. Thus, the

low release rate from blends with high polymer A content was probably caused by the small

PEG block (1 kDa). It is possible that the size of these PEG blocks was not sufficient to create a

mesh size in the swollen PEG regions that was large enough for diffusion of HSA through the

matrix.

Continuing this hypothesis, one would assume that increasing the size of the PEG block to 3

kDa as in polymer B would facilitate the release of large proteins. Indeed, the release rate of

HSA increased when the content polymer B exceeded 30%, but decreased again at very high

polymer B contents. However, as stated above, the total PEG content also contributes to the

release rate. Thus, our hypothesis for the fast release rate from the 50:50 blend is that for

HSA to be released, 3 kDa PEG blocks are necessary to create pores large enough for

diffusion, but also a certain total PEG content is required to form a hydrated network for

diffusion.

It is known that the production of protein-loaded microspheres by the W/O/W method may

cause protein denaturation or aggregation due to shear and interfacial stresses20. Although

4

Page 95: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

94

the stability of the encapsulated proteins was not tested specifically, the sandwich ELISA for

pPB-HSA required two epitopes for response, i.e. pPB and HSA, and could therefore provide

an indication on preservation of structural integrity. Therefore, the fact that pPB-HSA could

be recovered in release medium and plasma during 14 days in vitro and in plasma after 7 days

in vivo release, indicates that the construct was still intact after microsphere production and

release.

The development of PDGFβR-directed protein constructs is currently advancing towards

clinical application of PDGFβR-directed antifibrotic compounds21. These constructs are cell

selective and very potent21, and therefore perfectly suitable for (patient friendly) sustained

release formulations. As a proof of concept for the sustained release of such a PDGFβR-

directed protein construct, we assessed the protein release and targeting from pPB-HSA/HSA

microspheres in mice suffering from renal fibrosis. Hereby, we aimed to bring the clinical

application of PDGFβR-directed antifibrotic compounds one step closer.

It became apparent from the neck skin stainings (Fig. 6B) that not all protein was released 7

days after administration, which is in accordance with the release profile found in vitro which

showed a release for 14 days. Moreover, since pPB-HSA has a plasma half-life of

approximately 45 min (data not shown), the manifestation of a plasma concentration of pPB-

HSA suggests that release from the microspheres was still ongoing and that at least part of

the pPB-HSA reached the circulation intact. The specific targeting of pPB-HSA in PDGFβR-rich

areas after IV administration has been demonstrated before3,4. Kidney tissue stainings on pPB

were performed in this study, although the concentration of protein construct was most

probably too low to confirm the presence of pPB by immunohistochemical staining.

Nonetheless, our western blot results show that specific localization to fibrotic tissue also

occurs when pPB-HSA is administered as a polymeric sustained release formulation.

Altogether, these results show that subcutaneously administered microspheres composed of

biodegradable hydrophilic multi-block copolymers are suitable sustained release formulations

for the systemic delivery of pPB-HSA. Future research could include pharmacokinetic studies

on the release of pPB-HSA from microspheres and the delivery of an antifibrotic drug coupled

to pPB-HSA by a sustained release formulation.

Acknowledgements

The authors thank Marjolein van der Putten, Niek Breg, Valmira Isufi and Jitske Langeland for

their practical contributions and Floris Grasmeijer for his assistance with the scanning

electron microscopy equipment. This research was performed in the framework of the

Page 96: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

95

Transition II and Peaks 2011 (Transitie II en Pieken 2011) subsidy program of The Northern

Netherlands Provinces alliance (Samenwerkingsverband Noord-Nederland), and financially

supported by the province of Groningen, the municipality of Groningen and The Netherlands

Institute for Regenerative Medicine.

4

Page 97: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

96

References

1. Mehal WZ, Iredale J, Friedman SL. Scraping fibrosis: Expressway to the core of fibrosis.

Nat Med. 2011;17(5):552-553.

2. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

3. Poosti F, Bansal R, Yazdani S, et al. Selective delivery of IFN-gamma to renal interstitial

myofibroblasts: A novel strategy for the treatment of renal fibrosis. FASEB J.

2015;29(3):1029-1042.

4. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

5. Bansal R, Prakash J, de Ruijter M, Beljaars L, Poelstra K. Peptide-modified albumin carrier

explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver

fibrosis. Mol Pharm. 2011;8(5):1899-1909.

6. Hagens WI, Mattos A, Greupink R, et al. Targeting 15d-prostaglandin J2 to hepatic

stellate cells: Two options evaluated. Pharm Res. 2007;24(3):566-574.

7. Prakash J, de Jong E, Post E, Gouw AS, Beljaars L, Poelstra K. A novel approach to deliver

anticancer drugs to key cell types in tumors using a PDGF receptor-binding cyclic peptide

containing carrier. J Control Release. 2010;145(2):91-101.

8. Prajapati VD, Jani GK, Kapadia JR. Current knowledge on biodegradable microspheres in

drug delivery. Expert Opin Drug Deliv. 2015;12(8):1283-1299.

9. Wu F, Jin T. Polymer-based sustained-release dosage forms for protein drugs,

challenges, and recent advances. AAPS PharmSciTech. 2008;9(4):1218-1229.

10. Houchin ML, Topp EM. Chemical degradation of peptides and proteins in PLGA: A review

of reactions and mechanisms. J Pharm Sci. 2008;97(7):2395-2404.

11. van de Weert M, Hennink WE, Jiskoot W. Protein instability in poly(lactic-co-glycolic acid)

microparticles. Pharm Res. 2000;17(10):1159-1167.

12. Fredenberg S, Wahlgren M, Reslow M, Axelsson A. The mechanisms of drug release in

poly(lactic-co-glycolic acid)-based drug delivery systems--a review. Int J Pharm.

2011;415(1-2):34-52.

13. Stankovic M, de Waard H, Steendam R, et al. Low temperature extruded implants based

on novel hydrophilic multiblock copolymer for long-term protein delivery. Eur J Pharm

Sci. 2013;49(4):578-587.

14. Stankovic M, Hiemstra C, de Waard H, et al. Protein release from water-swellable

poly(D,L-lactide-PEG)-b-poly(-caprolactone) implants. Int J Pharm. 2015;480(1-2):73-83.

Page 98: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

97

15. Beljaars L, Molema G, Schuppan D, et al. Successful targeting to rat hepatic stellate cells

using albumin modified with cyclic peptides that recognize the collagen type VI receptor.

J Biol Chem. 2000;275(17):12743-12751.

16. Ye M, Kim S, Park K. Issues in long-term protein delivery using biodegradable

microparticles. J Control Release. 2010;146(2):241-260.

17. Ramazani F, Hiemstra C, Steendam R, et al. Sunitinib microspheres based on [PDLLA-

PEG-PDLLA]-b-PLLA multi-block copolymers for ocular drug delivery. Eur J Pharm

Biopharm. 2015;95(Pt B):368-377.

18. Stankovic M, Tomar J, Hiemstra C, Steendam R, Frijlink HW, Hinrichs WL. Tailored

protein release from biodegradable poly(epsilon-caprolactone-PEG)-b-poly(epsilon-

caprolactone) multiblock-copolymer implants. Eur J Pharm Biopharm. 2014;87(2):329-

337.

19. Tran VT, Karam JP, Garric X, et al. Protein-loaded PLGA-PEG-PLGA microspheres: A tool

for cell therapy. Eur J Pharm Sci. 2012;45(1-2):128-137.

20. Thomas CR, Geer D. Effects of shear on proteins in solution. Biotechnol Lett.

2011;33(3):443-456.

21. van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted therapies in liver fibrosis: Combining

the best parts of platelet-derived growth factor BB and interferon gamma. Front Med

(Lausanne). 2015;2:72.

22. Poosti F, Pham BT, Oosterhuis D, et al. Precision-cut kidney slices (PCKS) to study

development of renal fibrosis and efficacy of drug targeting ex vivo. Dis Model Mech.

2015;8(10):1227-1236.

4

Page 99: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

98

Supplementary materials and methods

Viability assays

MTT assay. Human primary hepatic stellate cells (ScienCell, cultured according manufacturer’s

instruction) grown on 96 wells plates were stimulated with carboxymethylcellulose (CMC) or

empty 50:50 microspheres (0.1, 1 or 2.5 mg/ml) for 24 or 48 h (both n=3) in standard

medium. The medium was removed by aspiration, and 200 μL of 3-(4,5-dimethylthiazol-2-yl)-

2,5-diphenyltetrazolium bromide (MTT) stock solution (500 μg/mL MTT in PBS) was added

per well for 4 h. The solution was aspirated, the precipitate was dissolved in DMSO and the

absorbance was measured at 550 nm (THERMOmax microplate reader, Molecular Devices).

OD values were expressed as percentage relative to vehicle control at 24 h. Differences

between groups were assessed by 2-way ANOVA with Sidak’s multiple comparisons test.

ATP assay. Mouse kidneys (male C57BL/6) were sliced according to standard procedures, as

described previously22. In short, kidneys were sliced in ice-cold carbogen-saturated Krebs-

Henseleit buffer supplemented with 25 mM D-glucose, 25 mM NaHCO3 (both Merck,

Darmstadt, Germany) and 10 mM HEPES (MP Biomedicals, Santa Ana, CA, USA) with a

Krumdieck tissue slicer (Alabama R&D, Munford, AL, USA) at 200-300 μm thickness and 5-6

mg wet weight. Slices were stored in UW until incubation with CMC or empty 50:50

microspheres (0.1, 1 or 2.5 mg/mL) for 24 (n=4) and 48 h (n=2) in oxygenized Williams

Medium E Glutamax-I (Gibco, Paisly, Scotland) supplemented with 25 mM D-Glucose and 50

μg/mL gentamycin. For ATP determination, slices were snap-frozen in 70% ethanol containing

2 mM EDTA (pH 10.9) and stored at -80°C until analysis. Samples were homogenized (Mini-

BeadBeater-8, BioSpec, Bartlesville, OK, USA) and centrifuged (2 minutes, 13,200 rpm). The

supernatant was diluted 10 times in 100 mM Tris-HCl/2 mM EDTA to measure the ATP

content with ATP Bioluminescence Assay Kit CLS II (Roche Diagnostics, Mannheim, Germany)

according to the manufacturer’s protocol. The pellet was incubated for 30 minutes with 200

μL 5M NaOH at 37°C. Next, the samples were diluted 5 times and the protein content was

determined with Lowry (Bio-Rad DC Protein Assay, Munich, Germany). ATP values (pmol)

were corrected for protein content (μg) of the corresponding slice, and expressed as

percentage relative to vehicle control at 24 h. Differences between groups for 24 h were

assessed by Friedman test with Dunn’s multiple comparisons test.

Page 100: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

99

Supplementary data

Table S1. In weights of prepolymers used in synthesis of x[PCL-PEG1000 or 3000-PCL]-y[PLLA] multiblock copolymers.

PCL, poly--caprolactone; PEG, polyethylene glycol; PLLA, poly (L-lactic) acid; BDI, 1,4-butanediisocyanate.

Table S2. Characterization of the synthesized multi-block copolymers for composition, intrinsic viscosity and

residual 1,4-dioxane.

PCL-PEG1000

prepolymer

(wt-%)

PLLA

prepolymer

(wt-%)

PCL-PEG1000

prepolymer

(in weights)

PCL-PEG3000

prepolymer

(in weights)

PLLA

prepolymer

(in weights)

BDI

(in weights)

50[PCL-

PEG1000-

PCL]-

50[PLLA]

50 50

85.17 g

(42.59

mmol)

n.a

83.35 g

(20.53

mmol)

0.95 g

(6.78

mmol)

30[PCL-

PEG3000-

PCL]-

70[PLLA]

30 70 n.a.

99.89 g

(24.91

mmol)

323.9 g

(80.77

mmol)

11.51 g

(82.13

mmol)

Polymer A

50[PCL-PEG1000-PCL]- 50[PLLA]

Polymer B

30[PCL-PEG3000-PCL]- 70[PLLA]

Molar LA/PEG ratio

(1H-NMR) 26.7 (26.6 in-weight) 130.6 (126.8 in-weight)

Molar CL/PEG ratio

(1H-NMR) 8.1 (8.7 in-weight) 7.8 (8.1 in-weight)

Intrinsic viscosity

(dl/g) 0.8 0.7

1,4-dioxane content

(ppm) < 300 < 100

4

Page 101: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

100

Figure S1. Representative scanning electron microscopy image (1,000x magnification) of 5 wt-% HSA

microspheres with a 50:50 polymer ratio after 7 days of release (A) and after 42 days of release (B).

Figure S2. Thermograms (reversing heat flow) of microspheres of different polymer blends, as obtained with

MDSC. On the y-axis, exothermic is up. Arrows indicate glass transitions. Other changes in heat flow occurred at

temperatures at which melting and crystallization events were observed. Therefore, these changes were not

considered as separate events (i.e. glass transitions).

Page 102: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Polymeric microspheres for the sustained release of pPB-HSA

101

Table S3. Characteristics of 50:50 polymer blend microspheres containing 5 wt-% HSA, produced to assess the

reproducibility of the production process.

Batch Median particle

size (μm)

Span Encapsulation

efficiency (%)

Cumulative release

after 14 d (%)

1 29.0 1.8 73 101

2 28.4 2.3 84 91

3 22.4 2.1 91 84

4 26.2 1.9 89 83

5 18.1 2.0 76 82

6 16.4 2.1 76 89

Average ± SD 23.4 ± 5.3 2.0 ± 0.2 82 ± 8 88 ± 7

Figure S3. Cumulative release of HSA from 5% HSA 50:50 polymer blend microspheres (n=6).

4

Page 103: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 4

102

Figure S4. Viability of (A) human myofibroblasts and (B) mouse precision-cut kidney slices after exposure to CMC

(vehicle) and microspheres in different concentrations for 24 and 48h, demonstrating no toxicity to these

biological systems.

Page 104: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 105: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

5

Page 106: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation

F. van Dijka,b,*, N. Teekampa,*, L. Beljaarsb, E. Postb, J. Zuidemac, R. Steendamc,

Y.O. Kimd, H.W. Frijlinka, D. Schuppand,e, K. Poelstrab, W.L.J. Hinrichsa and P.

Olingaa

* The authors contributed equally

aGroningen Research Institute of Pharmacy, Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen, Groningen, The

Netherlands, bGroningen Research Institute of Pharmacy, Department of

Pharmacokinetics, Toxicology and Targeting, University of Groningen,

Groningen, The Netherlands, cInnoCore Pharmaceuticals, Groningen, The

Netherlands, dInstitute of Translational Immunology and Research Center for

Immune Therapy, University Medical Center, Johannes Gutenberg University,

Mainz, Germany, eBeth Israel Deaconess Medical Center, Harvard Medical

School, Boston, MA, USA.

Published in Journal of Controlled Release, 2018

Pharmacokinetics of a sustained release

formulation of PDGFβ-receptor directed

carrier proteins to target the fibrotic liver

Page 107: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

106

Abstract

Liver fibrogenesis is associated with excessive production of extracellular matrix by

myofibroblasts that often leads to cirrhosis and consequently liver dysfunction and death.

Novel protein-based antifibrotic drugs show high specificity and efficacy, but their use in the

treatment of fibrosis causes a high burden for patients, since repetitive and long-term

parenteral administration is required as most proteins and peptides are rapidly cleared from

the circulation. Therefore, we developed biodegradable polymeric microspheres for the

sustained release of proteinaceous drugs. We encapsulated the drug carrier pPB-HSA, which

specifically binds to the PDGFβR that is highly upregulated on activated myofibroblasts, into

microspheres composed of hydrophilic multi-block copolymers composed of poly(L-lactide)

and poly ethylene glycol/poly(ϵ-caprolactone), allowing diffusion-controlled release. Firstly,

we estimated in mice with acute fibrogenesis induced by a single CCl4 injection the half-life of

I125-labeled pPB-HSA at 40 min and confirmed the preferential accumulation in fibrotic tissue.

Subsequently, we determined in the Mdr2-/- mouse model of advanced biliary liver fibrosis

how the subcutaneously injected microspheres released pPB-HSA into both plasma and

fibrotic liver at 24 h after injection, which was maintained for six days. Although the

microspheres still contained protein at day seven, pPB-HSA plasma and liver concentrations

were decreased. This reduction was associated with an antibody response against the human

albumin-based carrier protein, which was prevented by using a mouse albumin-based

equivalent (pPB-MSA). In conclusion, this study shows that our polymeric microspheres are

suitable as sustained release formulation for targeted protein constructs such as pPB-HSA.

These formulations could be applied for the long-term treatment of chronic diseases such as

liver fibrosis.

Page 108: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

107

Introduction

Sustained release drug delivery systems are increasingly used as a patient-friendly alternative

to conventional dosage forms1,2. When controlling the release rate of a drug its therapeutic

actions can be drastically improved by obtaining prolonged release associated with less

fluctuations in plasma concentration. This avoids peak levels and reduces side effects. This

way, the bioavailability, efficacy and safety of drugs can be significantly enhanced2,3. One such

drug delivery system is microspheres, that allows flexible dosing of the drug4. This approach is

particularly interesting for application of potent protein-based therapeutics, as the

administration frequency can be largely reduced as compared to intravenous administration.

Moreover, when encapsulated in polymeric microparticles, the biopharmaceutical can be

effectively protected from degradation induced by biological conditions or enzymes2,5.

In the treatment of chronic diseases such as fibrosis, the application of sustained release

formulations like microspheres for antifibrotic drugs could significantly improve patient

compliance and therapeutic efficacy, especially since the expected treatment would be long-

term. Hepatic fibrosis is a progressive, pathological condition affecting millions of people

worldwide6. Following chronic liver injury, inflammatory and bile ductular cells release a

variety of mediators that provoke the activation of fibroblasts and hepatic stellate cells to

myofibroblasts, which start to produce extracellular matrix (ECM) components, especially

fibrillar collagens in a chronic would healing reaction. In cirrhosis, which represents an

advanced stage of fibrosis, the liver vascular architecture gets progressively distorted and

functional parenchymal cells are ultimately replaced by abundant ECM, which causes liver

failure, and finally decompensated cirrhosis7-9.

For some patients with cirrhosis, liver transplantation or treatment with a new generation of

highly effective antiviral agents against Hepatitis B and C may be a curative treatment,

however there is still an urge for effective antifibrotic treatments to fulfill the needs of all

patients10-12. Many promising new drugs are biological-based, such as growth factors,

cytokines and monoclonal antibodies. A class of therapeutic proteins currently under

development are the fusion proteins, including biologic-based drugs modified with targeting

moieties13,14. Such proteins are particularly interesting as their therapeutic effects can be

increased while avoiding side effects15. The platelet-derived growth factor beta receptor

(PDGFβR) is abundantly expressed on myofibroblasts in fibrotic tissues with high fibrogenic

activity16,17, and therefore its expression was exploited as a potential target for the cell-

specific delivery of antifibrotic drugs. A carrier protein with high affinity for the PDGFβR was

designed, composed of an albumin core with multiple cyclic PDGFβR-recognizing peptides

(pPB) binding the PDGFβR. Previous studies have shown that the carrier, referred to as pPB-

5

Page 109: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

108

HSA, selectively bound to the PDGFβR and accumulated in fibrogenic cells in the fibrotic liver

which highly expressed the PDGFβR. The carrier itself did neither elicit an antifibrotic effect

nor induced proliferation of fibroblasts18.

Because many therapeutic proteins have poor in vivo pharmacokinetic properties as reflected

by relatively short in vivo plasma half-lives, we developed a microsphere formulation

containing pPB-HSA that ensures gradual protein release over a period of 14 days, which

could be suitable for therapeutic application of other large therapeutic proteins as well. A

blend of two biodegradable semi-crystalline multi-block co-polymers, composed of crystalline

blocks of poly(L-lactide) (PLLA) and amorphous blocks of poly ethylene glycol (PEG) and

poly(ϵ-caprolactone) (PCL), was used as a matrix for these microspheres. Whilst mostly

polymers are used that show degradation-controlled release19, e.g. poly (lactic-co-glycolic

acid), these particular polymers provided diffusion-controlled release of proteins, caused by

swelling of the PEG blocks by water uptake20. Disadvantage of degradation-controlled release

is that the degradation products of the polymer are usually incompatible with proteins21.

We previously showed proof of concept of effective release of drug carriers from

microspheres composed of these multi-block copolymers in the unilateral ureter obstruction

model for kidney fibrosis. In that study, we demonstrated the release of pPB-HSA from

subcutaneously injected microspheres into plasma and the subsequent localization of this

drug carrier in the fibrotic kidney 7 days after administration of the microspheres22. Although

we were able to demonstrate protein release after 7 days, the in vivo release characteristics

and the correlation with the in vivo kinetics remained undefined.

In the present study, we therefore further explored the applicability of these microspheres as

a sustained controlled release formulation for biologicals. For this, we examined the in vivo

kinetic behavior of pPB-HSA in two different mouse models for liver fibrosis that display high

and specific PDGFβR-expression, i.e. the acute CCl4 model and the Mdr2-/- model. In the

acute CCl4 model, we used I125-labeled pPB-HSA to determine pharmacokinetic parameters

and tissue distribution of this carrier protein. Subsequently, the in vivo release profile of pPB-

HSA from microspheres was determined in the chronic Mdr2-/- model.

Materials and methods

Pharmacokinetics of I125-labeled pPB-HSA

The experimental protocols for animal studies with the CCl4-model were approved by the

Animal Ethical Committee of the University of Groningen (The Netherlands). Male C57BL/6

Page 110: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

109

mice (20-22 grams) were obtained from Envigo (Horst, The Netherlands). Animals received ad

libitum normal diet with a 12 h light/dark cycle. Mice (n=12) received a single injection of CCl4

(Sigma Aldrich, Zwijndrecht, The Netherlands) diluted in olive oil (0.5 mg/kg) intraperitoneally.

After 24 hours, mice were intravenously injected with tracer amounts of I125-labeled pPB-HSA

(1*105-5*105 counts per minute (CPM) in PBS) and sacrificed after 10, 30 or 60 minutes (n=4

per time point), after which blood and all organs were collected to assess radioactivity18. pPB-

HSA was labeled with 125I as described before18.

Synthesis of polymers and proteins

The two multi-block co-polymers were synthesized as described before from the prepolymers

PLLA and PCL-PEG-PCL22,23. In short, [PLLA] was chain-extended with [PCL-PEG1000-PCL or PCL-

PEG3000-PCL] using 1,4-butanediisocyanate (Actu-all Chemicals BV, Oss, The Netherlands), to

prepare the multi-block co-polymers [PCL-PEG1000-PCL]-[PLLA] (50/50 weight ratio) and [PCL-

PEG3000-PCL]/[PLLA] (30/70 weight ratio). For this, [PLLA] and [PCL-PEG1000-PCL or PCL-PEG3000-

PCL] were dissolved in dry 1,4-dioxane (80 °C, 30 wt-% solution), 1,4-butanediisocyanate was

added and the reaction mixture stirred for 20 h. The reaction mixture was frozen and freeze-

dried at 30 °C shelf temperature to remove 1,4-dioxane.

The proteins pPB-HSA and pPB-MSA were synthesized as described before24. Briefly, N-γ-

maleimidobutyryl-oxysuccinimide ester was added to either human (purified from Cealb®,

Sanquin, Amsterdam, The Netherlands) or mouse serum albumin (Equitech-Bio Inc., Kerville,

TX, USA). Next, N-succinimidyl S-acetylthioacetate (SATA)-modified pPB (C*SRNLIDC*,

Ansynth Service BV, Roosendaal, The Netherlands) and activation solution (containing

hydroxylamine and EDTA) were added. After extensive dialysis, the monomeric product was

freeze-dried and stored at -20 °C.

Production and characterization of microspheres

Microspheres were produced using a similar water-in-oil-in-water double emulsification

evaporation method as described previously22. In brief, the multi-block co-polymers were

dissolved in dichloromethane in a 50:50 weight ratio. For the primary emulsion, PBS (control)

or a solution of 80 mg/mL protein (HSA; or pPB-HSA and HSA, or the mouse equivalents, in a

3:2 ratio) was added to the filtered polymer solution to obtain 5 wt-% theoretical protein load

and homogenized. For the secondary emulsion, the primary emulsion was added to 4 wt-%

5

Page 111: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

110

poly vinyl alcohol (Sigma Aldrich) + 5 wt-% NaCl solution in water under stirring. The hardened

microspheres were collected by filtration, washed and freeze dried.

Microspheres were characterized for morphology by scanning electron microscopy, for

particle size distribution by laser diffraction, and for protein content and in vitro release of

HSA, pPB-HSA or pPB-MSA as described by22. Scanning electron microscopy imaging was

performed at an acceleration voltage of 10 kV (JSM-6460 microscope, Jeol, Tokio, Japan).

Samples were fixed on an aluminum sample holder using double sided adhesive carbon tape

and sputter coated with 10 nm of gold. The particle size distributions of microspheres were

determined with laser diffraction and subsequently the span of the particle size distribution

was calculated using Eq. 1,

𝑆𝑝𝑎𝑛 = 𝑋10−𝑋90

𝑋50 Equation 1

where X10, X50 and X90 represent the volume percentages of particles (10%, 50% and 90%

undersize, respectively). The in vitro release was measured in triplicate by a sample-and-

replace method. Briefly, 10 mg of microspheres were suspended in 1.0 mL release buffer (100

mM sodium phosphate buffer, containing NaCl, Tween 80 and NaN3). Samples of 800 μl were

taken at predetermined time points and replaced by fresh buffer. Protein concentrations

were determined with BCA assay and an in-house developed ELISA for pPB-HSA (see section

ELISA). The protein content of the remaining microspheres was determined using BCA assay

as described before22 and was used to calculate the encapsulation efficiency (EE), which is

defined as the weight of encapsulated protein (i.e. HSA, pPB-HSA or pPB-MSA) divided by the

weight of total protein used.

Pharmacokinetics of pPB-HSA from microspheres

Studies with the Mdr2-/- mouse model were approved by the Animal Ethical Committee of

the State of Rhineland Palatinate. Female FVB mice (n=8) were obtained from Jackson

Laboratory (Jackson Laboratory, Bar Harbor, ME, USA) and FVB Mdr2-/- mice (n=24) (20-28

grams) were bred in homozygosity at the Institute of Translational Immunology at Mainz

University Medical Center. Mdr2-/- mice aged 11-15 weeks display advanced liver fibrosis

with a 4 to 5-fold increased liver collagen content25. Wildtype and Mdr2-/- mice were housed

with a 12 h light/dark cycle with water and ad libitum normal diet. At age 11-15 weeks Mdr2-

/- mice were injected subcutaneously in the neck, a site with sufficient subcutaneous space,

with a suspension of 12.6 wt-% microspheres in 500 μl 0.4% carboxymethyl cellulose (CMC,

Aqualon high Mw, Ashland, pH 7.0-7.4). Microspheres contained either 5 wt-% HSA (n=4), 3

Page 112: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

111

wt-% pPB-HSA + 2 wt-% HSA (n=16), 3 wt-% pPB-MSA + 2 wt-% MSA (n=8), or were empty

(polymer only) (n=4). Groups of mice were sacrificed at 1, 3, 5 or 7 days for pPB-HSA

microspheres (n=4 at each time point) and all other groups at day 7 after microsphere

administration. Blood, liver and off target organs were collected for further analysis.

ELISA

ELISA for pPB-HSA. pPB-HSA levels both in vitro (100 μl release buffer) and in vivo (100 μl

plasma or 1.4 mg/100 μl protein of liver, as determined with Lowry assay) were assessed with

our in-house developed sandwich ELISA22. In short, the capture antibody α-pPB was diluted in

coating buffer (100 mM NaHCO3/33 mM Na2CO3 in water (pH 9.5) and incubated overnight

(100 μl, 6.5 mg/ml, 1:1000, custom prepared by Charles Rivers, Den Bosch, The Netherlands)

in a 96-well high protein binding plate (Corning, New York, NY, USA). The plate was washed

extensively with PBS containing 0.5‰ Tween-20 (PBS-T) and blocked with 200 μl 5 wt-%

nonfat dry milk in PBS-T for 1 h. After washing, 100 μl sample was incubated for 2 h. The plate

was washed again, and the detection antibody goat α-HSA was applied (100 μl, 1:8000, ICN

Biomedicals, Zoetermeer, The Netherlands) for 1 h. The appropriate HRP-conjugated

secondary antibody (100 μl, 1:2000, DAKO, Santa Clara, CA, USA) was applied for 1 h, and the

substrate tetramethyl benzidine (100 μl, R&D Systems, Minneapolis, MN, USA) was incubated

for 20 min after washing with PBS-T. The absorbance was measured at 450nm (THERMOmax

microplate reader, Molecular Devices, Sunnyvale, CA, USA) after addition of 50 μl 2N H2SO4.

ELISA for immunoglobulins. Plasma levels of immunoglobulins against pPB-HSA or pPB-MSA

were measured with an in-house developed ELISA. Either pPB-HSA or pPB-MSA (100 μl, 10 μg

per ml coating buffer) was incubated for 2 h at room temperature in a high protein binding

96-well plate (Corning). After extensive washing with PBS-T, non-specific binding sites were

blocked with 200 μl 5 wt-% nonfat dry milk in PBS-T for 1 h. Plasma samples (100 μl plasma,

diluted 1:100) were added for incubation of 1 h after washing with PBS-T. The plate was

washed again and HRP-conjugated anti-mouse immunoglobulins diluted in blocking buffer

(100 μl, 1:2000, DAKO) were incubated for 1 h. After washing with PBS-T, the substrate

tetramethyl benzidine (100 μl, R&D Systems) was incubated for 20 min. The absorbance was

measured at 450 nm after addition of 50 μl 2N H2SO4.

5

Page 113: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

112

Quantitative real-time PCR

Total RNA from was isolated from livers using a Maxwell® LEV simply RNA Cells/Tissue kit

(Promega, Madison, WI, USA) according to manufacturer’s instructions. RNA concentrations

were determined using NanoDrop ND-100 spectrophotometer (NanoDrop Technologies,

Wilmington, DE, USA). The primers used include procollagen α1(I) forward: 5’-

TGACTGGAAGAGCGGAGAGT-‘3; reverse: 3’-ATCCATCGGTCATGCTCTCT-‘5; PDGFβ-receptor

forward: 5’-AACCCCCTTACAGCTGTCCT-‘3; reverse: 3’-TTCCTCTATTGCCCATCTC-‘5; β-actin

forward: 5’-ATCGTGCGTGACATCAAAGA-‘3; reverse: 3’-ATGCCACAGGATTCCATACC-‘5 (all

Sigma-Aldrich). Quantitative real-time PCR analysis was performed with 10 ng cDNA per

sample according to manufacturer’s instructions (SensiMix™ SYBR kit, Bioline, Taunton, MA,

USA) and was analyzed by the ABI7900HT sequence detection system (Applied Biosystems,

Foster City, CA, USA). For each sample, the threshold cycles (Ct values) were calculated with

the SDS 2.3 software program (Applied Biosystems) and mRNA expression was normalized for

β-actin.

Immunohistochemistry

Cryosections of neck skin tissue were cut with a thickness of 4 μm (CryoStar NX70 cryostat,

Thermo Fisher Scientific), dried and fixed with acetone. Paraffin sections of livers were cut

with a Leica Reichert-Jung 2040 microtome (Leica Microsystems, Nussloch, Germany) with a

thickness of 4 μm. The sections were deparaffinized in xylene and ethanol. All sections were

rehydrated in PBS and were incubated for 1 h with the primary antibody (goat anti-collagen

I&III (both 1:200 + 5% normal mouse serum (Southern Biotech, Birmingham, AL, USA)) or

rabbit anti-HSA (1:1500 (ICN Biomedicals)) at room temperature or boiled in 10 mM Tris/1

mM EDTA (pH 9.0) for 15 minutes prior to overnight incubation with rabbit anti-PDGFβ-

receptor (1:50, Cell Signaling) at 4°C. Next, sections were incubated with the appropriate HRP-

conjugated secondary antibody (1:100, DAKO, Santa Clara, CA, USA) for 30 minutes at room

temperature, which were visualized with ImmPACT NovaRED (both Vector, Burlingame, CA,

USA). Hematoxylin counterstaining was performed. Digital photomicrographs were captured

at 400x magnification (Aperio, Burlingame, CA, USA).

Statistical analyses

At least 3 individual experiments were performed for the in vitro microsphere

characterization and these data are represented as mean ± SD. All other data are represented

Page 114: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

113

as mean ± SEM. The graphs and statistical analyses were performed with Graphpad Prism

version 6.0 (GraphPad Prism Software, Inc., La Jolla, CA, USA). The differences between the

groups were assessed by ordinary one-way ANOVA followed by Bonferroni’s multiple

comparison test unless stated otherwise. Basic pharmacokinetic modeling was performed

with the computer program Multifit for non-linear curve-fitting26.

Results

In vivo pharmacokinetics of pPB-HSA

To determine the in vivo kinetics and tissue distribution of pPB-HSA, all organs of mice with

CCl4-induced acute liver fibrogenesis were collected and examined after a single intravenous

injection with I125-labeled pPB-HSA. Ten minutes after injection, high amounts (65 ± 6% of the

dose) accumulated in the liver, expressing the PDGFβR15, whereas 15 ± 2% was still found in

the plasma. A minor amount of the remaining fraction was detected in the kidneys (2.6 ±

0.5%), spleen (2.1 ± 1.0%), lungs (1.4 ± 0.5%), heart (0.9 ± 0.3%) and brains (0.2 ± 0.1%). The

active targeting of pPB-HSA to myofibroblasts in the fibrotic liver has been shown in previous

studies18, and we herewith confirmed the steering of pPB-HSA to the organ with the highest

PDGFβR-expression (Fig. 1). Based on the plasma concentration per milliliter at 10, 30 and 60

minutes after injection, the in vivo plasma half-life was calculated to be approximately 40

minutes. Basic kinetic modeling assuming 1-compartment kinetics further yielded a rough

estimate for the clearance of 58 µl/min and for the volume of distribution of 3 ml (Fig. S1 and

Table S1).

Figure 1. In vivo distribution of I125

-labeled pPB-HSA in plasma, liver and off-target organs at 10, 30 and 60

minutes after intravenous injection in mice with CCl4-induced acute liver fibrosis.

5

Page 115: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

114

Microsphere characterization

Microspheres containing pPB-HSA that ensure gradual and prolonged release into plasma for

at least 7 days were developed. All microspheres were spherically shaped and had a smooth

surface with little to no pores, as shown with scanning electron microscopy (Fig. 2A, Fig. S2).

Laser diffraction analysis revealed that the two batches of protein-loaded microspheres had

similar particle size distributions with a median particle size of around 25 µm, while polymer-

only control microspheres were slightly smaller at all volume percentages (Table 1). The

polydispersity is expressed in the span (Eq. 1), which is comparable to values found in earlier

studies applying the same production process22. Both protein-loaded microsphere

formulations yielded high encapsulation efficiencies of protein (Table 1) and showed low

burst release and sustained release in vitro for at least 14 days (Fig. 2B), with a cumulative

release after 14 days of 52% for pPB-HSA and 55% for HSA. The slightly higher molecular

weight of pPB-HSA (~74 kDa) than HSA (67 kDa) did not affect the release rate, with average

values of 4.5 ± 0.65 %/day and 5.0 ± 0.29 %/day, respectively.

Table 1. Characteristics of microspheres with different content as used in vivo in the Mdr2-/- model.

Formulation Protein load

Particle size (µm ± SD)

Span

Encapsulation

efficiency (%) X10 X50 X90

pPB-HSA 3% pPB-HSA/

2% HSA

4.4 ± 0.3 23.3 ± 1.5 59.5 ± 5.0 2.4 99

HSA 5% HSA 4.1 ± 0.2 27.6 ± 0.7 59.1 ± 1.2 2.0 81

Control - 2.5 ± 0.1 16.7 ± 1.2 43.9 ± 0.7 2.5 -

Page 116: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

115

Figure 2. Morphology and in vitro release of pPB-HSA from microspheres used in the Mdr2-/- model for liver

fibrosis. (A) Representative scanning electron micrograph of pPB-HSA microspheres after freeze-drying (1,000x

magnification). The empty microspheres and microspheres containing HSA only exhibited similar morphology.

(B) Cumulative in vitro release of pPB-HSA and HSA from pPB-HSA microspheres and HSA microspheres,

respectively. Percentages are corrected for EE.

Pharmacokinetic profile of pPB-HSA released from microspheres in vivo

The pPB-HSA microspheres were subcutaneously injected in the neck of Mdr2-/- mice with

advanced biliary liver fibrosis. These mice exhibit a profound increase in procollagen α1(I) and

PDGFβR gene expression as compared to normal mice (Fig. 3A and D), which is a hallmark of

fibrosis25. The livers of these Mdr2-/- mice showed characteristic deposition of collagen types

I & III in the portal areas particularly around the bile ducts (Fig. 3B) as a consequence of the

toxicity of accumulated of phosphatidylcholine in the hepatocytes, due to the knock out of

the transporter gene. Collagen deposition extended into advanced portal to portal bridge

formation in the parenchyma (Fig. 3C). Accordingly, myofibroblasts lining the fibrotic bile

ducts and myofibroblasts in the parenchyma expressed the PDGFβR (Fig. 3E and F).

5

Page 117: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

116

Figure 3. (pro-) Collagen and PDGFβ-receptor expressions at mRNA (A and D, respectively) and protein level (B, C,

E, F) in livers of Mdr2-/- mice. Of note, the staining (in red) is observed in particular around the bile ducts (BD) (B

and E, respectively) and in the parenchyma (C and F, respectively). Differences between groups were assessed

by unpaired student t-test. Arrows indicate fibrous collagen bands.

The microsphere injection site was inspected for presence and appearance of microspheres

that resided for 7 days at the injection site. Clearly, during these 7 days the microspheres

remained subcutaneous (Fig. 4A) and the staining for HSA demonstrated that the

microspheres in vivo still contained protein after 7 days (HSA and pPB-HSA) (Fig. 4B, C), as was

expected from the in vitro release studies.

Figure 4. Immunohistochemical staining for HSA of skin tissue samples at 7 days after injection. These samples

were obtained at the subcutaneous injection site of the microspheres (MSP). MSP contained either (A) no

protein, (B) HSA or (C) pPB-HSA.

Page 118: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

117

The pharmacokinetic profile of the released pPB-HSA was determined at 1, 3, 5 and 7 days

after injection. We confirmed sustained release of pPB-HSA from the microspheres into the

plasma up to 7 days after injection reaching a steady state concentration of 44.9 ± 4.7 ng/ml,

which equals 2.4*10-3 ± 0.3*10-3 %/ml based on the total pPB-HSA content of the

microspheres, within 1 day after injection and remained constant for 5 days (Fig. 5A). The

infusion rate of pPB-HSA from the microspheres into the circulation was determined by

multiplying this value with the clearance as estimated in the acute CCl4-model, yielding an

infusion rate of 0.2% per day. Interestingly, the steady state concentration in plasma was 78.5

± 10.9% lower at 7 days after injection than at day 5 (p<0.0001, unpaired student t-test).

In line with the PDGFβ-receptor expression, pPB-HSA was present in the fibrotic livers at all

time points, reflecting a similar pattern as seen in plasma, reaching a steady state

concentration of 121 ± 28.3 ng/ liver within 1 day after injection (Fig. 5B). Similar to the

observations in plasma, a decline of 75.4 ± 5.3% in the pPB-HSA concentration was detected

in the livers after 7 days as compared to day 5 (p=0.002, unpaired student t-test). As

expected, for both plasma and livers, the control group with microspheres containing HSA

only did not show any presence of pPB-HSA.

Figure 5. Concentration of pPB-HSA in (A) plasma and (B) livers of Mdr2-/- mice that received microspheres

(MSP) subcutaneously for either 1, 3, 5 or 7 days containing pPB-HSA or HSA (only after 7 days) as determined

with ELISA.

The in vivo reduction in pPB-HSA levels 7 days after injection as compared to earlier time-

points was rather unexpected, as protein was still present in the subcutaneous microspheres

after 7 days (Fig. 4B and C) and the in vitro data showed minimal burst release of pPB-HSA

followed by pseudo-zero order sustained release kinetics for 10 to 14 days (Fig. 2B). To

5

Page 119: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

118

further investigate this phenomenon, plasma samples were analyzed for the presence of

antibodies against the albumin-based carrier as this is not a mouse based protein, which

might interfere with our results. Indeed, the decline in pPB-HSA concentration at day 7

coincided with the induction of an antibody response against pPB-HSA as compared to the

empty microspheres as control group. Mice that received microspheres containing HSA only

also showed an induction of the immune response to the same extent (Fig. 6A). This

immunological reaction in these mice to the drug carrier was completely absent when

microspheres contained pPB-MSA, in which human serum albumin was replaced with the

mouse equivalent (Fig. 6B), as the response was not exceeding background levels. The pPB-

MSA microspheres displayed similar size distribution (median 22 μm) and morphology, and

showed slightly faster in vitro release characteristics than the microspheres containing the

human albumin-based carrier (Fig. S3, Table S2).

Figure 6. Antibody (AB) response in plasma of Mdr2-/- mice that received microspheres loaded with either (A)

pPB-HSA, HSA or no protein at 1, 3, 5 or 7 days after injection, or (B) pPB-MSA or no protein at 7 days after

injection as measured by ELISA (statistics were performed using the unpaired student t-test). The control group

consisted of mice injected with empty MSP. Note that an antibody-response was only present when levels

exceeded the background levels in the control group.

Discussion

Protein-based therapeutics including fusion proteins, such as protein-based drugs modified

with targeting moieties, drastically increased the treatment perspective for a wide variety of

diseases13,14,27. In the past decades, we developed various compounds enriched with PDGFβ-

receptor recognizing peptides (pPB) including the myofibroblast-selective drug carrier

PDGFβR-targeted human serum albumin (pPB-HSA)18. We aim to apply this PDGFβR-targeted

Page 120: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

119

drug carrier, when coupled to an effective antifibrotic compound, for the treatment of

chronic fibrotic diseases. However, delivery of protein-based compounds involves parenteral

administration with consequent short-lasting peak-concentrations and the need for frequent

application, posing a high burden to the potential patient. For effective treatment of such

diseases the sustained release of therapeutics is essential. We therefore developed a patient-

friendly formulation providing sustained release of protein therapeutics, which can be applied

for protein constructs like pPB-HSA22. In the present study, we showed that pPB-HSA was

released from the subcutaneous polymeric microspheres into the plasma and reached easily

detectable levels in the fibrotic liver, where a steady state concentration was achieved within

1 day after microsphere injection, which lasted for 5 days. A sudden decrease of this

concentration at day 7 was associated with antibody formation, while antibody formation was

prevented with the mouse albumin-based carrier.

In an attempt to correlate the in vitro release of pPB-HSA from microspheres and the

estimated in vivo infusion-rate, based on the steady state concentration in plasma and the

plasma clearance, we calculated release rates of 4.5 %/day and 0.2 %/day, respectively. This

yields approximately a 20-fold lower value in the in vivo situation. According to Cardot and

Tomic a linear in vitro - in vivo correlation only holds true when either drug dissolution or

release from the formulation are the sole limiting factors for the systemic absorption28. In our

studies, multiple factors might have contributed to the limitation in the in vivo infusion-rate of

pPB-HSA, which will be discussed in more detail below.

The release of any drug from a subcutaneous depot occurs, when dissolved, within the

subcutaneous tissue (or hypodermis) predominantly by diffusion or convection. The anatomy

and microenvironment of the hypodermis can largely affect the release and transport of the

drug29. Firstly, components of the subcutaneous extracellular matrix including collagens,

noncollagenous glycoproteins, hyaluronic acid and chondroitin, heparan or dermatan sulfates

can interfere with locally dissolved drugs and impede their passage to the circulation30. Such

interference can happen in several ways, for example by electrostatic or (non-) specific

interactions with the released protein drug. This ultimately leads to the formation of charged

and/or large complexes that are unable to diffuse through the tissue and consequently

accumulate subcutaneously instead31,32. Also, steric exclusion of the drug by the ECM can play

a role, causing not only limited diffusion, but might also influence the solubility of the protein

drug33. Secondly, other microenvironmental parameters including the ionic composition and

the pH of the injection site could limit protein absorption and stability30,34. Lastly, the uptake

of drug in the systemic circulation can be reduced due to degradation of the protein at the

injection site by the presence of proteases in the interstitial fluid35.

5

Page 121: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

120

After diffusion through the hypodermis, the released drug can reach the systemic circulation

via local semipermeable capillaries or by lymphatic vessels, depending on the size of the

protein. When the sustained release formulation is subcutaneously injected, the pressure in

the interstitial fluid is increased. As compensatory mechanism, the local lymphatic flow is

stimulated in order to transport excess of fluid into the lymphatic system. Consequently,

more macromolecules with a diameter of up to 100 nm can be taken up, including pPB-HSA

(which we assume to have a diameter of <25 nm36), and eventually these molecules will be

delivered to the bloodstream37,38. However, in mice Wu et al. demonstrated a limited uptake

of a similar protein, i.e. bovine serum albumin, into the lymphatics following subcutaneous

injection, thereby causing lower drug plasma concentrations39. Another reason could be pre-

systemic metabolism of the drug which may occur in the draining lymphatics before reaching

the circulation35. Such parameters cannot be easily mimicked in vitro. In summary, the in vitro

studies are oversimplified as compared to the in vivo circumstances, which is clearly

illustrated by our data. Recently, Kinnunen et al. made the first step towards an in vitro model

that provides a better representation of subcutaneous conditions40 and this model potentially

yields better correlations with in vivo data in future studies.

Despite the expected discrepancy between the in vitro and in vivo release, we were able to

show protein release and measured pPB-HSA at a plasma steady state concentration of

approximately 45 ng/ml during 5 days after injection (Fig. 5A), a sound result in view of the

extremely low in vivo half-life of 40 minutes. It is noteworthy that the calculated half-life

might be an overestimation, as the PDGFβR is most likely expressed at a lower level in the

acute CCl4-model than in the more clinical relevant Mdr2-/- model, mimicking many disease

features seen in human cirrhotic patients as well. Furthermore, a comparable concentration

profile was demonstrated in the fibrotic liver as our target organ, in which the PDGFβ-

receptor was highly expressed, with a steady state concentration of 121 ± 28.3 ng/liver within

1 day after injection. Whether this concentration is sufficient to reach therapeutic levels

would depend on the potency of the attached antifibrotic compound.

Several studies showed the receptor selectivity of PDGFβR-targeted constructs in different

animal models18,41,42. With the use of I125-labeled pPB-HSA we confirmed the specific

accumulation of this particular construct in mice suffering from acute liver fibrogenesis (Fig.

1). Because of the confirmation of the specific accumulation and the limited accumulation

found in other organs (between 0.2 and 2.6%), we did not consider any accumulation in off-

target organs in the pharmacokinetic study in the Mdr2-/- model.

The rapid decline in plasma concentration after 7 days was not anticipated, based on the

pseudo zero-order in vitro release kinetics (Fig. 2B) and the presence of protein in the

Page 122: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

121

microspheres in vivo 7 days after injection (Fig. 4). We therefore hypothesized that this

decline might be induced by antibody formation in mice against the human albumin-based

carrier. Indeed, we demonstrated low antibody levels during the first 5 days, drastically

increasing at day 7, which paralleled the decline in plasma and liver levels of pPB-HSA (Fig. 5

and 6A). As part of the adaptive immune defense, B cells produce immunoglobulin antibodies

against an antigen, i.e. a foreign protein like pPB-HSA or HSA in mice, upon prolonged

exposure. The induced antibodies form complexes with pPB-HSA or HSA that are ultimately

eliminated by the complement system43.

Eventually, the induction of the immune response was successfully circumvented by replacing

human albumin with mouse albumin (Fig. 6B), thereby creating a non-immunogenic drug

carrier pPB-MSA. This alternative approach proved that the antibody formation was directed

against the albumin part of pPB-HSA, and not against the pPB targeting moieties or the

crosslinking domain. We now constructed a drug carrier that is compatible with our polymeric

microsphere formulation and applicable for chronic administration of proteins.

In conclusion, we demonstrated that PDGFβR-targeted albumin was released in vivo from the

subcutaneously injected polymeric microspheres using hydrophilic multi-block copolymers

composed of poly(L-lactide) and poly ethylene glycol/poly(ϵ-caprolactone) blocks. pPB-HSA

was detectable in the plasma and in the fibrotic liver up to 7 days after injection. This implies

that sustained release microsphere formulations composed of these polymers are suitable for

the parenteral delivery of therapeutic proteins and might constitute a realistic option for

future application in chronic diseases such as liver fibrosis, bearing in mind that the origin of

the potential therapeutic protein should be complementary to the species used as model.

Future research should focus on pharmacodynamic aspects of the drug formulation, by

incorporating the PDGFβR-targeted albumin-based drug carrier coupled to an antifibrotic

drug, thereby directing towards the clinical application of this combined delivery system

ensuring sustained release of targeted therapeutic proteins.

Acknowledgements

The authors thank Herman Steen (BiOrion Technologies BV, Groningen) for providing pPB-

HSA, Marlies Schippers (Department of Pharmacokinetics, Toxicology and Targeting,

University of Groningen) and Marjolein van der Putten (student Pharmacy, University of

Groningen) for their practical contributions, and Imco Sibum (Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen) for his assistance with the scanning

electron micrographs. This research was performed in the framework of the Transition II and

5

Page 123: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

122

Peaks 2011 (Transitie II en Pieken 2011) subsidy program of The Northern Netherlands

Provinces alliance (Samenwerkingsverband Noord-Nederand), and financially supported by

the Province of Groningen, the municipality of Groningen and The Netherlands Institute for

Regenerative Medicine, and the ERC Advanced Grant FIBROIMAGING to Detlef Schuppan.

Page 124: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

123

References

1. Anselmo AC, Mitragotri S. An overview of clinical and commercial impact of drug delivery

systems. J Control Release. 2014;190:15-28.

2. Tibbitt MW, Dahlman JE, Langer R. Emerging frontiers in drug delivery. J Am Chem Soc.

2016;138(3):704-717.

3. Steinijans VW. Pharmacokinetic characterization of controlled-release formulations. Eur

J Drug Metab Pharmacokinet. 1990;15(2):173-181.

4. Freitas S, Merkle HP, Gander B. Microencapsulation by solvent extraction/evaporation:

Reviewing the state of the art of microsphere preparation process technology. J Control

Release. 2005;102(2):313-332.

5. Vaishya R, Khurana V, Patel S, Mitra AK. Long-term delivery of protein therapeutics.

Expert Opin Drug Deliv. 2015;12(3):415-440.

6. Cohen-Naftaly M, Friedman SL. Current status of novel antifibrotic therapies in patients

with chronic liver disease. Therap Adv Gastroenterol. 2011;4(6):391-417.

7. Friedman SL. Liver fibrosis -- from bench to bedside. J Hepatol. 2003;38 Suppl 1:S38-53.

8. Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. 2008;371(9615):838-851.

9. Mehal WZ, Schuppan D. Antifibrotic therapies in the liver. Semin Liver Dis.

2015;35(2):184-198.

10. Said A, Lucey MR. Liver transplantation: An update 2008. Curr Opin Gastroenterol.

2008;24(3):339-345.

11. Popov Y, Schuppan D. Targeting liver fibrosis: Strategies for development and validation

of antifibrotic therapies. Hepatology. 2009;50(4):1294-1306.

12. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest. 2013;123(5):1887-

1901.

13. Strohl WR. Fusion proteins for half-life extension of biologics as a strategy to make

biobetters. BioDrugs. 2015;29(4):215-239.

14. Walsh G. Biopharmaceutical benchmarks 2014. Nat Biotechnol. 2014;32(10):992-1000.

15. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted

interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology. 2011;54(2):586-596.

16. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

17. Wong L, Yamasaki G, Johnson RJ, Friedman SL. Induction of beta-platelet-derived growth

factor receptor in rat hepatic lipocytes during cellular activation in vivo and in culture. J

Clin Invest. 1994;94(4):1563-1569.

5

Page 125: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

124

18. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

19. Mao S, Guo C, Shi Y, Li LC. Recent advances in polymeric microspheres for parenteral

drug delivery--part 1. Expert Opin Drug Deliv. 2012;9(9):1161-1176.

20. Stankovic M, Tomar J, Hiemstra C, Steendam R, Frijlink HW, Hinrichs WL. Tailored

protein release from biodegradable poly(epsilon-caprolactone-PEG)-b-poly(epsilon-

caprolactone) multiblock-copolymer implants. Eur J Pharm Biopharm. 2014;87(2):329-

337.

21. Stankovic M, Hiemstra C, de Waard H, et al. Protein release from water-swellable

poly(D,L-lactide-PEG)-b-poly(-caprolactone) implants. Int J Pharm. 2015;480(1-2):73-83.

22. Teekamp N, Van Dijk F, Broesder A, et al. Polymeric microspheres for the sustained

release of a protein-based drug carrier targeting the PDGFbeta-receptor in the fibrotic

kidney. Int J Pharm. 2017;534(1-2):229-236.

23. Stankovic M, de Waard H, Steendam R, et al. Low temperature extruded implants based

on novel hydrophilic multiblock copolymer for long-term protein delivery. Eur J Pharm

Sci. 2013;49(4):578-587.

24. Beljaars L, Molema G, Schuppan D, et al. Successful targeting to rat hepatic stellate cells

using albumin modified with cyclic peptides that recognize the collagen type VI receptor.

J Biol Chem. 2000;275(17):12743-12751.

25. Popov Y, Patsenker E, Fickert P, Trauner M, Schuppan D. Mdr2 (Abcb4)-/- mice

spontaneously develop severe biliary fibrosis via massive dysregulation of pro- and

antifibrogenic genes. J Hepatol. 2005;43(6):1045-1054.

26. Proost J. MULTIFIT: An interactive program for nin-linear curve-fitting of several

pharmacokinetic models and mathematical functions. 1992.

27. PhRMA. Medicines in development: Biologics. Washington, DC. 2013.

28. Cardot JM, Tomic I. In vitro in vivo correlation basis and application to slow release

injectable formulation. Farmacia. 2015;63:781-791.

29. Weiser JR, Saltzman WM. Controlled release for local delivery of drugs: Barriers and

models. J Control Release. 2014;190:664-673.

30. Kinnunen HM, Mrsny RJ. Improving the outcomes of biopharmaceutical delivery via the

subcutaneous route by understanding the chemical, physical and physiological

properties of the subcutaneous injection site. J Control Release. 2014;182:22-32.

31. Mach H, Gregory SM, Mackiewicz A, et al. Electrostatic interactions of monoclonal

antibodies with subcutaneous tissue. Ther Deliv. 2011;2(6):727-736.

32. Yaoi Y, Hashimoto K, Takahara K, Kato I. Insulin binds to type V collagen with retention of

mitogenic activity. Exp Cell Res. 1991;194(2):180-185.

Page 126: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

125

33. Laurent TC, Persson H. On the interaction between polysaccharides and other

macromolecules. iii. the use of hyaluronic acid for the separation of macromolecules in

the ultracentrifuge. Biochim Biophys Acta. 1963;78:360-366.

34. Cugia C, Monduzzi M, Ninham BW, Salis A. Interplay of ion specificity, pH and buffers:

Insights from electrophoretic mobility and pH measurements of lysozyme solutions. RSC

Adv. 2013;3:5882-5888.

35. Richter WF, Bhansali SG, Morris ME. Mechanistic determinants of biotherapeutics

absorption following SC administration. AAPS J. 2012;14(3):559-570.

36. Ferrer ML, Duchowicz R, Carrasco B, de la Torre JG, Acuna AU. The conformation of

serum albumin in solution: A combined phosphorescence depolarization-hydrodynamic

modeling study. Biophys J. 2001;80(5):2422-2430.

37. Hommel E, Mathiesen ER, Aukland K, Parving HH. Pathophysiological aspects of edema

formation in diabetic nephropathy. Kidney Int. 1990;38(6):1187-1192.

38. Porter CJ, Edwards GA, Charman SA. Lymphatic transport of proteins after s.c. injection:

Implications of animal model selection. Adv Drug Deliv Rev. 2001;50(1-2):157-171.

39. Wu F, Bhansali SG, Tamhane M, et al. Noninvasive real-time fluorescence imaging of the

lymphatic uptake of BSA-IRDye 680 conjugate administered subcutaneously in mice. J

Pharm Sci. 2012;101(5):1744-1754.

40. Kinnunen HM, Sharma V, Contreras-Rojas LR, et al. A novel in vitro method to model the

fate of subcutaneously administered biopharmaceuticals and associated formulation

components. J Control Release. 2015;214:94-102.

41. Bansal R, Tomar T, Ostman A, Poelstra K, Prakash J. Selective targeting of interferon

gamma to stromal fibroblasts and pericytes as a novel therapeutic approach to inhibit

angiogenesis and tumor growth. Mol Cancer Ther. 2012;11(11):2419-2428.

42. Prakash J, de Jong E, Post E, Gouw AS, Beljaars L, Poelstra K. A novel approach to deliver

anticancer drugs to key cell types in tumors using a PDGF receptor-binding cyclic peptide

containing carrier. J Control Release. 2010;145(2):91-101.

43. Warrington R, Watson W, Kim HL, Antonetti FR. An introduction to immunology and

immunopathology. Allergy Asthma Clin Immunol. 2011;7 Suppl 1:S1-1492-7-S1-S1.

5

Page 127: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 5

126

Supplementary data

Figure S1. Basic pharmacokinetic modeling using the plasma concentration of I125

-labeled pPB-HSA per ml plasma

at 10, 30 and 60 minutes after intravenous injection in mice with CCl4-induced acute liver fibrosis, assuming 1-

compartment kinetics. Fitting curve is shown in red, individual data points in yellow.

Table S1. Estimated pharmacokinetic parameters of I125

-labeled pPB-HSA, based on plasma levels following a

single intravenous injection in the acute CCl4-mouse model for liver fibrosis and calculated assuming 1-

compartment kinetics.

Pharmacokinetic parameter Estimated value

Plasma half-life 39 ± 5 min

Clearance 58 ± 4 µl/min

Volume of distribution 3 ± 0.3 ml

Page 128: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Pharmacokinetics of a sustained release formulation of pPB-HSA

127

Figure S2. Morphology of (A) 5% HSA microspheres and (B) empty microspheres depicted by representative

scanning electron micrographs (1,000x magnification).

Figure S3. Characteristics of pPB-MSA microspheres. (A) Cumulative in vitro release of protein as determined

with the BCA assay from pPB-MSA microspheres. Percentages are corrected for EE. (B) Representative scanning

electron micrograph of microspheres containing pPB-MSA (1,000x magnification).

Table S2. Characteristics of pPB-MSA microspheres as used in the in vivo study.

Formulation Protein load

Particle size (µm ± SD)

Span

Encapsulation

efficiency (%) X10 X50 X90

pPB-MSA 3% pPB-MSA/

2% MSA 3.4 ± 0.2 21.6 ± 1.0 66.7 ± 2.2 2.9 74

5

Page 129: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

6

Page 130: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

F. van Dijka,b, N. Teekampa, E. Postb, D. Schuppanc,d, Y.O. Kimd, J. Zuidemae, R.

Steendame, A. Casinif, P.L. Horvatovichg, N.J. Sijbrandih, H.W. Frijlinka, W.L.J.

Hinrichsa, K. Poelstrab, L. Beljaarsb and P. Olingaa

aGroningen Research Institute of Pharmacy, Department of Pharmaceutical

Technology and Biopharmacy, University of Groningen, Groningen, The

Netherlands, bGroningen Research Institute of Pharmacy, Department of

Pharmacokinetics, Toxicology and Targeting, University of Groningen,

Groningen, The Netherlands, cInstitute of Translational Immunology and

Research Center for Immune Therapy, University Medical Center, Johannes

Gutenberg University, Mainz, Germany, dBeth Israel Deaconess Medical Center,

Harvard Medical School, Boston, MA, USA, eInnoCore Pharmaceuticals,

Groningen, The Netherlands, fSchool of Chemistry, Cardiff University, Cardiff,

United Kingdom, gGroningen Research Institute of Pharmacy, Department of

Analytical Biochemistry, University of Groningen, Groningen, The Netherlands, hLinXis BV, Amsterdam, The Netherlands.

Manuscript in preparation

The antifibrotic potential of a sustained

release formulation of a PDGFβ-receptor

targeted rho kinase inhibitor

Page 131: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

130

Abstract

Rho kinase is associated with the development of portal hypertension in liver cirrhosis.

Inhibition of rho kinase activity can successfully reduce the portal pressure, but this also leads

to severe side effects related to a systemic drop in blood pressure. This can be circumvented

by the liver-specific delivery of a rho kinase inhibitor such as Y27632, which reduces portal

pressure and additionally possesses antifibrotic activity. For this, we targeted Y27632 with the

drug carrier pPB-MSA to the key pathogenic cells in liver cirrhosis, i.e. the myofibroblasts,

which highly express the PDGFβ-receptor. pPB-MSA consists of mouse serum albumin (MSA)

covalently coupled to several PDGFβR-recognizing moieties (pPB). In this study, we aimed to

create a combined drug delivery system providing the prolonged release of such targeted

antifibrotic protein constructs by encapsulating pPB-MSA-Y27632 in biodegradable polymeric

microspheres, thereby reducing short-lasting peak concentrations and the need for frequent

administrations. Firstly, we confirmed the relaxing potency of pPB-MSA-Y27632 in vitro in a

contraction assay using hepatic stellate cells seeded on collagen gels. Our results showed that

PDGFβ-receptor targeted Y27632 was able to exert its effect in the designated target cell. We

subsequently established the in vivo antifibrotic effect of pPB-MSA-Y27632 at 7 days after a

single subcutaneous microsphere administration in the Mdr2-/- mouse model for advanced

liver fibrosis. pPB-MSA-Y27632 loaded microspheres significantly reduced the gene

expression level of multiple extracellular matrix proteins and additionally reduced the

collagen I&III protein expression. This effect was associated with a reduction in profibrogenic

cytokines and matrix metalloproteinases, and not accompanied by an effect on

proteoglycans, proinflammatory cytokines, proteases or protease inhibitors on the gene level.

In conclusion, this study shows that polymeric microspheres are suitable as drug delivery

system for the sustained systemic delivery of targeted protein constructs with antifibrotic

potential such as pPB-MSA-Y27632. These formulations could be applied for the long-term

treatment of chronic diseases such as liver fibrosis.

Page 132: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

131

Introduction

In liver cirrhosis, fibrotic scarring and contraction of fibrogenic cells leads to portal

hypertension, representing one of the major complications of this disease1,2. Cirrhotic

patients and animals commonly have a reduced mean arterial pressure due to peripheral

vasodilation, causing an increase in splanchnic flow. In contrast to this peripheral vasodilation,

portal hypertension occurs in these patients, due to increased hepatic resistance to portal

inflow. This complicates options for treatment2,3.

Upregulation of intrahepatic rho-associated protein kinase activity, or in short rho kinase, may

contribute to the development of portal hypertension in liver cirrhosis4,5. Rho kinase is a

major downstream effector protein of Rho GTPase, which is a pivotal player in the regulation

of cell morphology and shape, chemotaxis, actin-cytoskeletal reorganization, and contraction

in different cell types6,7. The effector protein rho kinase is particularly involved in cell

migration and contractility8,9. Inhibition of this protein using rho kinase inhibitors, such as

Y27632, was found to reduce portal vascular resistance and thus portal pressure5,10. In

addition to the hemodynamic effects, Y27632 was shown to possess antifibrotic activity as

well in several animal models of liver fibrosis11-13. However, systemic administration of a rho

kinase inhibitor will affect many cell types, as kinase-controlled signaling occurs in virtually

every cell type. This causes the induction of severe adverse effects, including a reduction in

vascular smooth muscle contractility and therefore an even further decline in systemic blood

pressure10,14.

This can be circumvented by the cell-specific delivery of such a rho kinase inhibitor to the key

pathogenic cells, without affecting other cells such as vascular smooth muscle cells. The

pathogenic cells involved in the excessive production of extracellular matrix (ECM) proteins in

liver fibrosis are myofibroblasts including hepatic stellate cells (HSCs) with a contractile

phenotype15. These cells can be reached with an albumin-based drug carrier (pPB-MSA), that

targets to the myofibroblasts by binding to the highly and specifically expressed PDGFβ-

receptor, via several PDGFβ-receptor recognizing peptides (pPB) attached to an albumin core

(mouse serum albumin, MSA)16,17. By inhibiting rho kinase activity specifically in the crucial

fibrogenic cells, the migration, contraction and transdifferentiation of hepatic stellate cells

into myofibroblasts can be impeded. The antifibrotic effect of the rho kinase inhibitor Y27632

when targeted to the myofibroblasts using a similar albumin carrier binding to the M6P/IGFII-

receptor was previously demonstrated9. In that study, Y27632 was coupled to the carrier via a

platinum-based linker, allowing slow intracellular release of Y27632 from the conjugate

during several days after endocytic uptake9,18.

6

Page 133: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

132

An essential challenge in the application of therapeutic proteins is the route of

administration. For such drugs the most common route is parenteral administration, generally

causing undesirable fluctuations in plasma levels and moreover high burden to the patient19.

Therefore, next to the formation of an intracellular slow release depot of Y27632 by taking

advantage of the properties of the chemical linker, a patient-friendly formulation providing

gradual and prolonged release of such therapeutic proteins for application in chronic diseases

such as fibrosis may ensure further sustained release20. We previously established the

sustained controlled release of a similar albumin-based carrier for at least 7 days in vivo by

encapsulating that protein in biodegradable polymeric microspheres21 and explored the

pharmacokinetic release profile17. We were able to demonstrate long lasting serum levels of

the carrier and additionally localization in the fibrotic organ.

In the present study, we explored the pharmacodynamic properties of biodegradable

polymeric microspheres loaded with the albumin-based carrier (pPB-MSA) coupled to an

antifibrotic compound (Y27632) via the platinum-based Lx-linker. We first confirmed the

relaxing potential of pPB-MSA-Y27632 in vitro. Subsequently, we prepared polymeric

microspheres containing pPB-MSA-Y27632 that allow release of proteins for at least 7 days

and determined the antifibrotic activity at 7 days after a single microsphere injection in Mdr2-

/- mice with established biliary liver fibrosis.

Materials and methods

Synthesis and characterization of proteins

pPB-MSA was synthesized as described before16,17. The Lx-linker (LinXis, Amsterdam, the

Netherlands) was used as platinum-based linker to couple Y27632 to pPB-MSA. The Lx-linker

was conjugated to trans-4-[(1R)-1-Aminoethyl]-N-4-pyridinylcyclohexanecarboxamide

dihydrochloride, i.e. Y27632 (Tocris Bioscience, Bristol, UK), and characterized with NMR,

mass spectrometry and HPLC as described in the supplementary materials and methods, and

was subsequently coupled to pPB-MSA. In short, 0.214 μmol Y27632-Lx reacted with 14 nmol

pPB-MSA in 20 mM tricine/NaNO3 buffer pH 8.5 for 30 minutes at room temperature while

stirring, and subsequently incubated overnight at 37°C. The mixture was dialyzed against PBS

for 48 hours, and freeze dried.

The product was characterized by MALDI-TOF mass spectrometry (Voyager DE-Pro MALDI-

TOF, Applied Biosystems, Foster City, CA, USA) in a sinapinic acid matrix according to standard

protocols and silver staining. For the silver staining, samples (10 μg) were applied on a 10%

SDS polyacrylamide gel according to standard procedures. In short, the gel was fixed in

Page 134: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

133

H2O/MeOH = 1/1 containing acetic acid. After washing in 25% EtOH in H2O, the gel was

incubated in H2O containing Na2S2O3. Next, the gel was extensively washed in H2O, incubated

in H2O supplemented with AgNO3 and formaldehyde, washed again and developed in H2O

containing Na2S2O3, formaldehyde and Na2CO3. The reaction was stopped by washing and

subsequent incubation in H2O/MeOH = 4/5 containing acetic acid, after which the gel was

photographed.

Contraction assay

Human LX2 hepatic stellate cells were kindly provided by prof. Scott Friedman (Mount Sinai

Hospital, New York) and cultured in Dulbecco’s modified Eagle’s medium (DMEM, Invitrogen,

Carlsbad, CA) supplemented with 10% FCS and antibiotics (100 U/ml penicillin and 100 µg/ml

streptomycin). Cells (50.000/well) were seeded in complete medium containing 2% FCS on

collagen gels, composed of 1.2 mg/ml collagen I (rat tail, Corning, New York), 6 mM NaOH,

0.4x PBS (55 mM NaCl, 1 mM KCl, 4 mM PO43-, pH 7.4) and 20 mM HEPES diluted in medium,

which were allowed to solidify for 1 hour at 37°C and 5% CO2. After 3 hours, the medium was

replaced by complete medium and cells incubated with 10 µM Y27632, pPB-MSA-Y27632 or

pPB-MSA for 48 hours. Gels were photographed and contraction was determined as the ratio

between the surface of the gel and the surface of the well as measured with ImageJ (National

Institutes of Health, USA).

Production and characterization of microspheres

Microspheres were produced using a similar double emulsification evaporation method as

described previously21. The phase-separated multi-block copolymers [PCL-PEG1000-PCL]-[PLLA]

(50/50 weight ratio) and [PCL-PEG3000-PEG]-[PLLA] (30/70 weight ratio) (obtained from

InnoCore Pharmaceuticals, Groningen, The Netherlands) were used at a 1:1 weight ratio. PBS

(control), or pPB-MSA-Y27632 and MSA in a 1:4 weight ratio, or pPB-MSA and MSA in a 3:2

weight ratio were encapsulated at a 5 wt-% theoretical protein load. Microspheres were

characterized for morphology by scanning electron microscopy, particle size distribution by

laser diffraction, protein content and in vitro protein release as described before by Teekamp

et al.21.

6

Page 135: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

134

Animal experiments

Experiments with the Mdr2-/- mouse model were approved by the Animal Ethical Committee

of the State of Rhineland Palatinate. Female FVB mice (n=8) were purchased from Jackson

Laboratory (Jackson Laboratory, Bar Harbor, ME, USA) and FVB Mdr2-/- mice (n=26) (20-26

grams) were bred in homozygosity at the Institute of Translational Immunology at Mainz

University Medical Center. All animals were housed with a 12 h light/dark cycle with ad

libitum chow and water. At age 8-9 weeks Mdr2-/- mice were injected subcutaneously with

500 μl of 12.6 wt-% microspheres dispersed in 0.4% carboxymethyl cellulose (CMC, Aqualon

high Mw, Ashland, pH 7.0-7.4) when microspheres contained 3 wt-% pPB-MSA + 2 wt-% MSA

(n=8), or with 500 μl of 20 wt-% microspheres dispersed in CMC when microspheres

contained 1 wt-% pPB-MSA-Y27632 + 4 wt-% MSA (n=4) or no protein (polymer only) (n=8).

Mice were injected subcutaneously once daily for 7 days with 250 μg/ml Y27632 (Tocris

Bioscience, Bristol, UK) in PBS for a final dose of 1 mg/kg (n=6). All mice were sacrificed 7 days

after microsphere administration or after 7 injections with plain Y27632. Livers were collected

for further analysis.

Inductively coupled plasma mass spectrometry

The platinum content in the livers was quantitated with inductively coupled plasma mass

spectrometry (ICP-MS), using an ICP-MS Agilent 7500ce (Agilent Technologies, Waldbronn,

Germany) instrument, equipped with a CETAC ASC-520 autosampler (CETAC Technologies,

Omaha, Nebraska, USA) and a MicroMist nebulizer at a sample uptake rate of 0.25 ml/min.

The instrument was calibrated on a daily basis. ICP-MS parameters were: RF power 1560 W,

cone material nickel, carrier gas 0.9-1.0 L/min, make up gas 0.2-0.3 L/min, plasma gas 15

L/min, dwell time 0.3 s, replicates 10, monitored isotopes 194Pt and 195Pt. The Agilent

MassHunter software package (Workstation Software, version B.01.01, Build 123.11, Patch 4,

2012) was used for data processing. Samples were prepared by digestion of 15-30 mg tissue

in 2 ml 20% nitric acid using a microwave system (Discover SP-D, CEM Microwave Technology,

Germany) (200°C, ramp time 5 min, hold time 6 min, maximal power 300 W). Digested

samples were diluted with Milli-Q water resulting in nitric acid concentrations <4% and

platinum concentrations <15 µg/kg.

Page 136: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

135

Low density array

Total RNA from was isolated from livers using a Maxwell® LEV simply RNA Cells/Tissue kit

(Promega, Madison, WI, USA) according to manufacturer’s instructions. RNA concentrations

were determined using NanoDrop One spectrophotometer (Thermo Fisher Scientific,

Waltham, MA, USA). The expression of 24 fibrosis-related genes (Table S1) was studied with a

custom-designed low density array. For this, a reaction mixture containing 50 μl of 6 ng/μl

cDNA and 50 μl 2x TaqMan PCR Master Mix was loaded per sample. PCR amplification was

performed on a ViiA7 Real-Time PCR system (all Applied Biosystems). For each sample, mRNA

expression was normalized for GAPDH, β-actin and YWHAZ. Fold induction values were

calculated using the 2^-ΔΔCt method and subsequently Z-normalized22. Heat maps for

different sets of genes were generated using R Studio (version 1.1.383).

Immunohistochemistry

Paraffin sections of livers were cut with a Leica Reichert-Jung 2040 microtome (Leica

Microsystems, Nussloch, Germany) with a thickness of 4 μm. The sections were

deparaffinized in xylene and ethanol. Sections were rehydrated in PBS and were incubated for

1 h with the primary antibodies (goat anti-collagen I&III (both 1:200 + 5% normal mouse

serum (Southern Biotech, Birmingham, AL, USA)) at room temperature. Next, sections were

incubated with the appropriate HRP-conjugated secondary antibody (1:100, DAKO, Santa

Clara, CA, USA) for 30 minutes at room temperature and were visualized with ImmPACT

NovaRED (both Vector, Burlingame, CA, USA). Hematoxylin counterstaining was performed.

Digital photomicrographs were captured at 400x magnification (Aperio, Burlingame, CA, USA).

Statistical analyses

At least 3 individual experiments were performed for the in vitro microsphere

characterization and these data are represented as mean ± SD. All other data are represented

as mean ± SEM. The graphs were made with Graphpad Prism version 6.0 (GraphPad Prism

Software, Inc., La Jolla, CA, USA). The statistics were performed with R (version 3.4.0, 2017-

04-21, 64 bit). Statistical differences were assessed by Kruskal Wallis test, and if applicable

pairwise comparison was done by Mann-Whitney test corrected with Benjamini-Hochberg

test, unless stated otherwise in the figure caption.

6

Page 137: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

136

Results

Characterization of pPB-MSA-Y27632

The rho kinase inhibitor Y27632 was coupled to pPB-MSA as HSC-selective drug carrier using

the platinum-based Lx-linker. Characterization of the constructs using silver staining and

MALDI-TOF mass spectrometry showed monomeric protein products with average molecular

weights of 66.0 kDa, 71.0 kDa and 73.6 kDa for MSA, pPB-MSA and pPB-MSA-Y27632,

respectively (Fig. 1, Table 1, Fig. S1). Based on these data, we calculated that per mouse

albumin molecule approximately 5 pPB-moieties (Mw = 1 kDa) and 5 Y27632 molecules (Mw

= 538 Da) were coupled (Table 1).

Figure 1. Analysis of MSA-based protein constructs using silver staining.

Table 1. Characterization of MSA-based protein constructs using MALDI-TOF mass spectrometry.

Mw MALDI-TOF (Da) # pPB coupled # Y27632 coupled

MSA 65.950 - -

pPB-MSA 70.950 5 -

pPB-MSA-Y27632 73.570 5 5

pPB-MSA-Y27632 reduced fibrotic parameters in vitro

As we aimed to determine the antifibrotic effect of pPB-MSA-Y27632 encapsulated in

polymeric microspheres administered to mice suffering from liver fibrosis, we firstly verified

the in vitro effects of pPB-MSA-Y27632. Since rho kinase is known to be involved particularly

in cell contractility, we assessed the relaxing potency of the targeted rho kinase inhibitor in an

in vitro contraction assay using LX2 hepatic stellate cells seeded on collagen gels (Fig. 2). Both

Page 138: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

137

pPB-MSA-Y27632 and free Y27632 (in equimolar amounts) significantly reduced the

contractility of LX2 cells after 48 hours by 38.7 ± 8.4% and 41.0 ± 5.5%, respectively. The

carrier pPB-MSA alone did not affect cell contraction. These results indicate that PDGFβ-

receptor directed Y27632 is able to exert its effect in the designated target cells.

neg c

trl

pPB-M

SA-Y

27

pPB-M

SA

Y27

0

20

40

60

80

100

p=0.008

p=0.04

Rela

tive g

el

co

ntr

acti

on

(%

)

Figure 2. The in vitro effect of pPB-MSA-Y27632 as determined by a contraction assay in cultures of LX2 cells

seeded on collagen gels. The graph shows gel contraction following treatment of cells for 48 hours with pPB-

MSA-Y27632, pPB-MSA or Y27632 (n=4), relative to vehicle treatment. Differences between groups were

assessed in Graphpad Prism version 6.0 by Friedman test followed by Dunn’s multiple comparisons test.

Microsphere characterization

With the confirmed in vitro activity of pPB-MSA-Y27632 on the contractility of HSCs, we

continued our studies with the development of a sustained release formulation aiming for at

least 7 days of gradual release of pPB-MSA-Y27632. We prepared polymeric

microspheres17,21, and encapsulated our construct. The microspheres displayed a

polydisperse size distribution and a median particle size of 28 µm (Fig. 3A, Table 2). Control

microspheres containing either pPB-MSA or no protein, were slightly smaller in size, with

median sizes of 21.6 and 22 μm, respectively. Scanning electron microscopy showed that all

particles were spherical with a smooth surface (Fig. 3A) and analysis of their protein content

of the microspheres revealed a high encapsulation efficiency of 81% for pPB-MSA-Y27632

loaded microspheres and 103% for pPB-MSA encapsulated particles (Table 2). The in vitro

release profile of proteins from the pPB-MSA-Y27632 microspheres revealed sustained and

gradual release for several days. There was only a minimal initial burst release. The

cumulative protein release from pPB-MSA-Y27632 loaded microspheres in release buffer was

6

Page 139: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

138

78 ± 7.2% after 14 days (Fig. 3B). Morphology and in vitro release characteristics of pPB-MSA

loaded microspheres were published before17.

Figure 3. Morphology and in vitro release of proteins from polymeric microspheres containing pPB-MSA-Y27632.

(A) Representative scanning electron micrograph of pPB-MSA-Y27632 microspheres after freeze-drying (1,000x

magnification) showing smooth spherical particles. (B) Cumulative in vitro release of proteins from pPB-MSA-

Y27632 loaded microspheres as measured with BCA assay. Percentages are corrected for the encapsulation

efficiency.

Table 2. Characteristics of microspheres with different contents used in vivo in the Mdr2-/- model.

Formulation Protein load

Particle size (µm ± SD)

Span

Encapsulation

efficiency (%) X10 X50 X90

pPB-MSA-

Y27632

1% pPB-MSA-

Y27632/

4% MSA

5.7 ± 0.2 27.9 ± 0.5 67.1 ± 1.6 2.2 81

pPB-MSA 3% pPB-MSA/

2% MSA 3.4 ± 0.2 21.6 ± 1.0 66.7 ± 2.2 2.9 103

Control - 3.1 ± 0.1 22.0 ± 0.8 63.9 ± 1.5 2.8 -

pPB-MSA-Y27632 reduced fibrotic parameters in vivo

Having confirmed the pharmacological activity of our construct and the sustained release of

these constructs from microspheres in vitro, Mdr2-/- mice aged 8-9 weeks, suffering from

advanced biliary liver fibrosis, were injected once subcutaneously with pPB-MSA-Y27632

loaded microspheres. 7 days after injection, we were able to detect levels of 721 ± 23 ng

platinum/liver, reflecting the Lx-linker as part of pPB-MSA-Y27632. We performed a gene

array on liver samples. In these animals, the gene expression levels for the extracellular matrix

Page 140: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

139

proteins (collagens 1a1, 1a2, 3a1, 4a1, 5a1 and 6a1, elastin1 and fibronectin1) were

significantly reduced (p=0.001) in the group of mice treated with microspheres containing

pPB-MSA-Y27632 as compared to mice that received empty microspheres (Fig. 4A). In

addition, gene expression levels for the profibrogenic cytokines (TGFβ and PDGF-BB) and the

matrix metalloproteinases (MMP2 and 14) were markedly reduced (p=0.026 and 0.021,

respectively) following treatment with pPB-MSA-Y27632 loaded microspheres as compared to

diseased control animals that received empty microspheres (Fig. 4B and C, respectively). This

is demonstrated by the reduced z-normalized values of these genes as compared to the

diseased control mice (empty MSP) and the accessory heat maps (expressions of the separate

genes in Fig. S2). The treatment did not affect the expression levels for proteoglycans

(decorin, biglycan and fibromodulin), proinflammatory cytokines (interleukin 1β, tumor

necrosis factor and chemokine ligand 2), proteases (cathepsin K, bone morphogenetic protein

1 and ADAM metallopeptidase) or protease inhibitors (TIMP2, plasminogen activator inhibitor

1) (data not shown). The antifibrotic effect on ECM proteins was confirmed at the protein

level by immunohistochemical staining for collagen I&III, that clearly demonstrated regression

of bridging fibrosis in the liver parenchyma in mice treated with pPB-MSA-Y27632 containing

microspheres as compared to the control groups (empty MSP or pPB-MSA MSP) or plain

Y27632 (Fig. 4D).

Free Y27632 injected subcutaneously once daily did not have any antifibrotic effect in the

livers of these mice. Previous studies did show an effect of Y276329, albeit to a lesser extent

than the targeted equivalent, but in these studies free Y27632 was administered

intravenously.

6

Page 141: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

140

Figure 4. In vivo effect of pPB-MSA-Y27632 released from subcutaneously residing polymeric microspheres. Z-

normalized values of fold inductions (mRNA levels relative to healthy controls) and heat maps of (A) extracellular

matrix proteins (collagens 1a1, 1a2, 3a1, 4a1, 5a1 and 6a1, elastin1 and fibronectin1), (B) profibrogenic

cytokines (TGFβ and PDGF-BB) and (C) matrix metalloproteinases (MMP2 and 14) at mRNA level in the different

treatment groups. Two missing z-normalized values for fibronectin1 are depicted in grey. (D)

Immunohistochemical staining for collagens I&III of Mdr2-/- livers of mice at 7 days after microsphere injection

containing pPB-MSA-Y27632 or controls.

Page 142: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

141

Discussion

Kinase inhibitors are upcoming therapeutics in the cancer field. However, they also seem

promising for a future treatment of fibrotic diseases, as they inhibit the proliferation and

contractility of hepatic stellate cells (HSCs) being key pathogenic cells23. In particular, the rho

kinase inhibitor Y27632 was shown both in vitro and in vivo to effectively reduce fibrotic

parameters11-13. Despite its antifibrotic potential, this kinase inhibitor induced several serious

adverse effects10,14. Therefore, we aimed to deliver Y27632 to the key hepatic cell type in the

liver involved in the excessive extracellular matrix (ECM) protein deposition and blood flow

regulation in the cirrhotic liver, i.e. the activated HSC. We delivered this compound to the HSC

via the highly and specifically expressed PDGFβ-receptor24. For this, we attached multiple

Y27632 moieties to the HSC-selective drug carrier pPB-MSA16,17. For effective treatment of

chronic diseases such as fibrosis we developed a patient-friendly formulation that could

facilitate the gradual and prolonged release of this pPB-MSA-Y27632, thereby circumventing

high plasma concentrations following intravenous injections and avoiding multiple

injections19. In the present study, we achieved the sustained release of HSC-targeted Y27632

from polymeric microspheres in vitro and demonstrated its antifibrotic activity in vivo.

We produced a HSC-selective protein endowed with pharmacological activity and determined

its activity as compared to its untargeted equivalent. The in vitro experiments, assessing HSCs

contractility, demonstrated that pPB-MSA-Y27632 was equally active as free Y27632 in

equimolar concentrations. Our results are in line with those of previous in vitro studies with

Y27632-constructs targeted to the M6P/IGFII-receptor or the PDGFβ-receptor9,25. These

results confirm that our protein construct is pharmacologically active and also show that in

vitro there is no added value to the targeted construct as compared to the free Y27632.

The rho kinase inhibitor was coupled to the HSC-selective drug carrier pPB-MSA via the

platinum (II)-based Lx-linker, creating a coordinative bond between the two constituents. The

use of this linker offers advantages in terms of synthesis and stability of the construct18. It was

previously shown that this linker, when attached to another kinase inhibitor and carrier

protein, after endocytosis could provide a local drug reservoir for several days following a

single injection assuring slow intracellular release of the drug18. This can be explained by the

slow ligand-exchange kinetics of platinum allowing easy replacement of attached drugs by

other (intracellular) ligands such as glutathione26, or by the degradation of the construct by

cellular enzymes e.g. in the lysosomes, enabling the release of the active compound18.

Although platinum-based compounds like cisplatin are notorious nephrotoxic agents, this

particular platinum (II)-based linker did neither induce any toxicity in vitro in renal tubular

cells nor in vivo in the kidneys18. This is due to the relatively low concentrations of the linker

6

Page 143: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

142

(far below toxicity levels) and to the fact that platinum is not present in its unconjugated

(free) form; it is either coupled to albumin or to ligands like glutathione.

In addition to the slow intracellular release of Y2763218, continuous inhibition of rho kinase

activity by sustained release is important for the treatment of chronic diseases such as liver

fibrosis, because high plasma levels possibly leading to toxicity are prevented. Moreover, a

discontinuous effect of drugs on for instance portal pressure may still lead to a perpetuation

of disease activity. An additional benefit of a sustained release formulation is that multiple

injections are not necessary, and thus improves patient compliance and comfort20. Previous

studies demonstrated the antifibrotic potential of Y27632 when targeted to the M6P/IGFII-

receptor expressed on activated HSCs following multiple intravenous injections9. We now

were able to show this effectivity on regression of fibrosis in the fibrotic liver resulting from

one single subcutaneous injection with polymeric microspheres containing pPB-MSA-Y27632,

thereby providing prolonged release of proteins for several weeks.

The previously developed polymeric microsphere formulation is suitable for the sustained

release of large therapeutic proteins, and the in vivo release kinetics of a similar carrier

protein and subsequent localization in the UUO-model for kidney fibrosis and the Mdr2-/-

model for liver fibrosis were demonstrated before17,21. We proceeded with the same

formulation and now encapsulated the rho kinase inhibitor Y27632 coupled to the drug

carrier pPB-MSA. In vitro, the formulation showed diffusion-controlled sustained release for

at least 14 days. At 7 days after injection the concentration reached in the liver was 721 ± 23

ng/liver, based on the platinum content present in the Lx-linker. This gives an indication for

the amount of pPB-MSA-Y27632 present in the liver, keeping in mind that 5 Y27632

molecules are attached via the Lx-linker per 1 pPB-MSA molecule. The steady state

concentration in the fibrotic livers of mice that received microspheres containing pPB-HSA

was 121 ± 28.3 ng/liver, as determined with ELISA in a previous study17, which is in the same

range. It could be that the actual level of pPB-MSA-Y27632 in the liver is lower, as the total

amount of platinum present is measured and some accumulation of platinum during the 7

days cannot be excluded.

However, the essential question is whether these levels are high enough to obtain

therapeutic concentrations to exert an antifibrotic effect. Clearly, the gene array showed a

reduction in gene expression levels of several extracellular matrix proteins, profibrotic

cytokines and matrix metalloproteinases (MMPs), and additionally a decrease in collagen I&III

protein expression in Mdr2-/- mice that received microspheres containing pPB-MSA-Y27632.

According to all our gene array data, this could either be due to a direct effect related to the

reduced expression of profibrotic cytokines, or indirectly as the consequence of reduced

Page 144: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

143

expression of matrix metalloproteinases (MMPs), but evidently mechanistic studies at protein

level are necessary in order to gain more insight. MMPs are involved in the maintenance of

the ECM and processes of tissue repair. These proteinases not only resolve the excess matrix,

but certain MMPs can have profibrotic functions as well27. For example, both MMP-2 and

MMP-14 were reported to be antifibrotic via degradation of ECM proteins27,28, while other

studies demonstrated contribution to fibrosis via the proliferation of stellate cells29 or by

processing profibrotic signaling molecules like transforming growth factor beta (TGFβ),

thereby stimulating collagen synthesis and accumulation30.

In this present study we only considered the antifibrotic effects of pPB-MSA-Y27632

containing microspheres, and did not determine the hemodynamic effects. Our group and

others previously demonstrated that the inhibition of rho kinase with Y27632 in activated

HSCs using several selective carriers, including M6P-HSA and pPB-HSA, significantly decreased

portal pressure and hepatic vascular resistance in different animal models of liver cirrhosis

with portal hypertension4,9,25. This confirms the importance of rho kinase, next to other well-

known mechanisms that regulate the hepatic vascular resistance and blood pressure, for

example nitric oxide4,31. This selective inhibition of rho kinase did not cause any off-target

hemodynamic or toxic effects4,25. The effects on portal pressure and fibrosis were explained

by a reduction in phosphorylation of myosin light chain (MLC), as one of the downstream

proteins in the rho kinase pathway4,9,25. Also, the reduction of hepatic ROCK2 mRNA and

reduced phosphorylation of moesin in cirrhotic livers probably contributed to the therapeutic

effects4,25.

In conclusion, we demonstrated the antifibrotic effectivity of polymeric microspheres

containing the rho kinase inhibitor Y27632 targeted to the PDGFβ-receptor by using the

carrier pPB-MSA. This study showed that sustained release formulations composed of these

polymers are suitable for the patient-friendly delivery of therapeutic proteins including pPB-

MSA-Y27632, representing an important step towards the clinical application of this

combined delivery system.

Acknowledgements

The authors thank HendrikJan Houthoff (LinXis BV, Amsterdam) for providing Y27632-Lx,

Miriam Boersema (Department of Pharmaceutical Technology and Biopharmacy) for her help

with the low density array, and Imco Sibum for his assistance at the scanning electron

microscope. This study was performed using a grant of the Netherlands Institute of

Regenerative Medicine (NIRM) and a grant from NanoNext.NL (program 03.10).

6

Page 145: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

144

References

1. Bosch J, Garcia-Pagan JC. Complications of cirrhosis. I. portal hypertension. J Hepatol.

2000;32(1 Suppl):141-156.

2. Hennenberg M, Trebicka J, Sauerbruch T, Heller J. Mechanisms of extrahepatic

vasodilation in portal hypertension. Gut. 2008;57(9):1300-1314.

3. Ekataksin W, Kaneda K. Liver microvascular architecture: An insight into the

pathophysiology of portal hypertension. Semin Liver Dis. 1999;19(4):359-382.

4. Klein S, Van Beuge MM, Granzow M, et al. HSC-specific inhibition of rho-kinase reduces

portal pressure in cirrhotic rats without major systemic effects. J Hepatol.

2012;57(6):1220-1227.

5. Zhou Q, Hennenberg M, Trebicka J, et al. Intrahepatic upregulation of RhoA and rho-

kinase signalling contributes to increased hepatic vascular resistance in rats with

secondary biliary cirrhosis. Gut. 2006;55(9):1296-1305.

6. Charest PG, Firtel RA. Big roles for small GTPases in the control of directed cell

movement. Biochem J. 2007;401(2):377-390.

7. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420(6916):629-

635.

8. Amano M, Nakayama M, Kaibuchi K. Rho-kinase/ROCK: A key regulator of the

cytoskeleton and cell polarity. Cytoskeleton (Hoboken). 2010;67(9):545-554.

9. van Beuge MM, Prakash J, Lacombe M, et al. Reduction of fibrogenesis by selective

delivery of a rho kinase inhibitor to hepatic stellate cells in mice. J Pharmacol Exp Ther.

2011;337(3):628-635.

10. Hennenberg M, Biecker E, Trebicka J, et al. Defective RhoA/rho-kinase signaling

contributes to vascular hypocontractility and vasodilation in cirrhotic rats.

Gastroenterology. 2006;130(3):838-854.

11. Tada S, Iwamoto H, Nakamuta M, et al. A selective ROCK inhibitor, Y27632, prevents

dimethylnitrosamine-induced hepatic fibrosis in rats. J Hepatol. 2001;34(4):529-536.

12. Murata T, Arii S, Mori A, Imamura M. Therapeutic significance of Y-27632, a rho-kinase

inhibitor, on the established liver fibrosis. J Surg Res. 2003;114(1):64-71.

13. Murata T, Arii S, Nakamura T, et al. Inhibitory effect of Y-27632, a ROCK inhibitor, on

progression of rat liver fibrosis in association with inactivation of hepatic stellate cells. J

Hepatol. 2001;35(4):474-481.

14. Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity of tyrosine

kinase inhibition. Nat Rev Cancer. 2007;7(5):332-344.

15. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology. 2008;134(6):1655-

1669.

Page 146: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

145

16. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

17. van Dijk F, Teekamp N, Beljaars L, et al. Pharmacokinetics of a sustained release

formulation of PDGFbeta-receptor directed carrier proteins to target the fibrotic liver. J

Control Release. 2017;269:258-265.

18. Prakash J, Sandovici M, Saluja V, et al. Intracellular delivery of the p38 mitogen-activated

protein kinase inhibitor SB202190 [4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-

pyridyl)1H-imidazole] in renal tubular cells: A novel strategy to treat renal fibrosis. J

Pharmacol Exp Ther. 2006;319(1):8-19.

19. Steinijans VW. Pharmacokinetic characterization of controlled-release formulations. Eur

J Drug Metab Pharmacokinet. 1990;15(2):173-181.

20. Vaishya R, Khurana V, Patel S, Mitra AK. Long-term delivery of protein therapeutics.

Expert Opin Drug Deliv. 2015;12(3):415-440.

21. Teekamp N, Van Dijk F, Broesder A, et al. Polymeric microspheres for the sustained

release of a protein-based drug carrier targeting the PDGFbeta-receptor in the fibrotic

kidney. Int J Pharm. 2017;534(1-2):229-236.

22. Cheadle C, Vawter MP, Freed WJ, Becker KG. Analysis of microarray data using Z score

transformation. J Mol Diagn. 2003;5(2):73-81.

23. Koyama Y, Xu J, Liu X, Brenner DA. New developments on the treatment of liver fibrosis.

Dig Dis. 2016;34(5):589-596.

24. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

25. Magdaleno F, Klein S, Frohn F, Reker-Smit C, Uschner FE, Schierwagen R, Poelstra K,

Beljaars L, Trebicka J. Rho-kinase inhibitor coupled with peptide-modified albumin

carrier reduces fibrogenesis and portal pressure in cirrhotic rats without systemic

effects. Hepatology. 2017;66(142A).

26. Ikeda K, Miura K, Himeno S, Imura N, Naganuma A. Glutathione content is correlated

with the sensitivity of lines of PC12 cells to cisplatin without a corresponding change in

the accumulation of platinum. Mol Cell Biochem. 2001;219(1-2):51-56.

27. Giannandrea M, Parks WC. Diverse functions of matrix metalloproteinases during

fibrosis. Dis Model Mech. 2014;7(2):193-203.

28. Spinale FG, Janicki JS, Zile MR. Membrane-associated matrix proteolysis and heart

failure. Circ Res. 2013;112(1):195-208.

6

Page 147: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

146

29. Benyon RC, Hovell CJ, Da Gaca M, Jones EH, Iredale JP, Arthur MJ. Progelatinase A is

produced and activated by rat hepatic stellate cells and promotes their proliferation.

Hepatology. 1999;30(4):977-986.

30. Mu D, Cambier S, Fjellbirkeland L, et al. The integrin alpha(v)beta8 mediates epithelial

homeostasis through MT1-MMP-dependent activation of TGF-beta1. J Cell Biol.

2002;157(3):493-507.

31. Trebicka J, Hennenberg M, Laleman W, et al. Atorvastatin lowers portal pressure in

cirrhotic rats by inhibition of RhoA/rho-kinase and activation of endothelial nitric oxide

synthase. Hepatology. 2007;46(1):242-253.

Page 148: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

147

Supplementary materials and methods

Lx-linker conjugation to Y27632 and purification

AgNO3 (85 mg, 500 µmol) was added to a suspension of [PtCl2(en)] (163 mg, 500 µmol) in

DMF (24.8 ml) and stirred overnight at room temperature in the dark under argon. The

mixture was filtered through a 0.2 µm filter. To a solution of Y27632.2HCl (40 mg, 125 µmol)

in MilliQ water (14 ml, pH adjusted to 6.95 using 1 M NaOH) was added 12.4 ml (250 µmol) of

the former solution. The mixture was stirred for 4.5 hours at room temperature in the dark

under argon. Subsequently, the reaction mixture was filtered through a 0.2 µm filter. To the

solution was added 0.9% NaCl (1 ml), followed by the removal of the solvent under reduced

pressure (40 °C, 4.5 h). The product was dissolved in 15% MeOH/MilliQ water (6 ml), filtered

through a 0.2 µm filter, and purified in two batches (2 x 3 ml) by preparative reverse-phase

HPLC (Grace Alltima C18 5 µm column, 22 x 250 mm; gradient: 15 to 35% MeOH/0.1% TFA in

water/0.1% TFA in 36 min). Product fractions were lyophilized and obtained as colorless solids

(total yield: 41.8 mg, 43.8% total yield).

Characterization of Y27632-Lx

HRMS (ESI+) C16H2935ClN5O195Pt+ [M]+ calc 538.1776, found 538.1716

1H NMR (400 MHz, CD3OD) δ 8.57 – 8.47 (m, 2H), 7.74 – 7.68 (m, 2H), 6.09 – 5.78 (m, 2H),

5.72 – 5.40 (m, 2H), 3.21 – 3.11 (m, 1H), 2.80 – 2.51 (m, 4H), 2.48 – 2.37 (m, 1H), 2.10 – 2.01

(m, 2H), 1.98 – 1.84 (m, 2H), 1.67 – 1.51 (m, 3H), 1.32 – 1.15 (m, 5H)

195Pt NMR (86 MHz, CD3OD) δ -2512 ppm

HPLC (Grace Alltima C18 5 µm column, 25 x 4.6 mm) indicated that the product was 95.6%

pure (retention time 15.8 min; gradient: 5 to 25% MeCN/0.1% TFA in water/0.1% TFA in 18

min measured at a wavelength of 273 nm).

6

Page 149: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

148

Table S1. Taqman expression ID’s used for the custom-designed low density array.

Gene Full gene name Taqman expression ID

Extracellular matrix proteins

Col1a1 Collagen, type I, alpha 1 Col1a1-Mm00801666_g1

Col1a2 Collagen, type I, alpha 2 Col1a2-Mm00483888_m1

Col3a1 Collagen, type III, alpha 1 Col3a1-Mm00802300_m1

Col4a1 Collagen, type IV, alpha 1 Col4a1-Mm01210125_m1

Col5a1 Collagen, type V, alpha 1 Col5a1-Mm00489299_m1

Col6a1 Collagen, type VI, alpha 1 Col6a1-Mm00487160_m1

Eln Elastin Eln-Mm00514670_m1

Fn1 Fibronectin 1 Fn1-Mm01256725_m1

Profibrotic cytokines

Tgfb1 Transforming growth factor, beta 1 Tgfb1-Mm01178820_m1

Pdgfb Platelet derived growth factor, B polypeptide Pdgfb-Mm00440677_m1

Matrix Metalloproteinases

Mmp2 Matrix metallopeptidase 2 Mmp2-Mm00439498_m1

Mmp9* Matrix metallopeptidase 9 Mmp9-Mm00442991_m1

Mmp13* Matrix metallopeptidase 13 Mmp13-Mm00439491_m1

Mmp14 Matrix metallopeptidase 14 (membrane-inserted) Mmp14-Mm00485054_m1

Proteoglycans

Dcn Decorin Dcn-Mm00514535_m1

Bgn Biglycan Bgn-Mm01191753_m1

Fmod Fibromodulin Fmod-Mm00491215_m1

Proinflammatory cytokines

Il1b Interleukin 1 beta Il1b-Mm00434228_m1

Tnf Tumor necrosis factor Tnf-Mm00443258_m1

Ccl2 Chemokine (C-C motif) ligand 2 Ccl2-Mm00441242_m1

Ifng* Interferon gamma Ifng-Mm01168134_m1

Proteases

Ctsk Cathepsin K Ctsk-Mm00484039_m1

Bmp1 Bone morphogenetic protein 1 Bmp1-Mm00802220_m1

Adamts2 A disintegrin-like and metallopeptidase (reprolysin type) with thrombospondin type 1 motif, 2

Adamts2-Mm00805170_m1

Protease inhibitors

Timp1* Tissue inhibitor of metalloproteinase 1 Timp1-Mm01341361_m1

Timp2 Tissue inhibitor of metalloproteinase 2 Timp2-Mm00441825_m1

Serpine1 Serine (or cysteine) peptidase inhibitor, clade E, member 1

Serpine1-Mm00435858_m1

Housekeeping genes

Gapdh Glyceraldehyde-3-phosphate dehydrogenase Gapdh-Mm99999915_g1

Actb Actin, beta Actb-Mm00607939_s1

Ywhaz Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide

Ywhaz-Mm03950126_s1

NB. Genes with an asterisk (*) were excluded from the array as the expression was below

detection limits.

Page 150: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

149

Supplementary data

A

B

C

Figure S1. MALDI-TOF mass spectrometry spectra of (A) MSA, (B) pPB-MSA and (C) pPB-MSA-Y27632.

6

Page 151: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

150

Extracellular matrix proteins

Page 152: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

The antifibrotic potential of a sustained release formulation of pPB-MSA-Y27632

151

Profibrotic cytokines

6

Page 153: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 6

152

MMPs

Figure S2. Gene expression levels of the extracellular matrix proteins (collagens 1a1, 1a2, 3a1, 4a1, 5a1 and 6a1,

elastin1 and fibronectin1), profibrotic cytokines (TGFβ and PDGF-BB) and matrix metalloproteinases (MMP2 and

14) as expressed by 2^-ΔCt. The fold inductions of these individual genes were used for the Z-normalization and

combined as depicted in figure 4.

Page 154: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 155: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

7

Page 156: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

Page 157: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

156

Therapeutic proteins in fibrosis

Biologicals, such as antibodies, siRNA, cytokines and enzymes, are becoming increasingly

popular in the treatment of a wide range of disorders, as reflected by the rapidly increasing

number amongst new therapeutic products1,2. In fact, at present well over 100 different

protein therapeutics are FDA-approved and it is predicted, based on the large number of

therapeutic proteins currently in clinical trials and in clinical use, that these numbers will

greatly expand in the coming years1. Such proteins are promising therapeutic agents, as they

display high affinity and specificity for their ligands and additional high tolerability as

compared to small-molecule drugs1.

The most common route of administration for (therapeutic) proteins is an (intravenous)

injection. Frequent injections are necessary when the tissue permeability is low and the in

vivo half-life is short causing large fluctuations in plasma levels3. Therefore, this

administration route is associated with a high burden to the patient, in particular with chronic

indications when long-term treatment is required4. These disadvantages can be easily avoided

by applying a sustained release formulation such as polymeric microspheres, providing the

gradual and prolonged release of proteins following a single injection5.

A prevalent chronic condition of which the progression cannot be fully reversed by

therapeutic intervention yet, is fibrosis. This is a condition hallmarked by the dynamic

deposition of extracellular matrix (ECM) proteins by myofibroblasts in response to acute or

chronic injury, like viral infections or metabolic syndrome6-8. In the liver, fibrosis can progress

to cirrhosis; a process that may take decades, during which the overall liver architecture is

irreversibly distorted, causing high morbidity and mortality in patients worldwide9. In 2015,

liver cirrhosis was ranked in the top 20 of leading causes of death worldwide, accounting for

more than 1 million of deaths per year10. With the increasing prevalence of obesity, as

important cause of cirrhosis, it is expected that the number of patients suffering from liver

cirrhosis will continue to rise the coming years11. However, the convincing evidence that

fibrosis is a dynamic and, depending on the disease stage, an even reversible process has

boosted research to gain insights in the molecular mechanisms involved in fibrosis that could

advance to new and effective therapeutic strategies in the near future9.

Currently, the only available curative treatment option for liver cirrhosis is transplantation

surgery. Since viral hepatitis is one of the major causes of cirrhosis, some patients benefit

from treatment with a new generation antiviral agent against hepatitis B and C viral

infections7. However, new therapeutic strategies are urgently needed to stop fibrosis

progression and even induce fibrosis reversal in all patients suffering from this severe

disease7,9,12. Many experimental studies focus on myofibroblasts as the primary target for

Page 158: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

157

antifibrotic therapeutics, as they are the key fibrogenic cells in fibrosis. New treatment

strategies should not aim at eradication of these cells, but should reduce their fibrogenic

activity and induce ECM-degrading enzymes instead9. These cells as primary producers of scar

tissue should not only be regarded as the main contributors to fibrosis, but they have a crucial

role in maintaining organ integrity in general and can even stimulate resolution of fibrosis by

induction of ECM-degrading proteases13. There are multiple strategies aiming at reducing the

pathogenic function of myofibroblasts, as reviewed by Schuppan et al.9. This includes

manipulation of fibrosis-specific pathways upregulated in these cells, such as procollagen type

I or profibrogenic signaling14, and using advanced siRNA delivery techniques, like liposomes

that passively accumulate in the liver15. Another approach is to use ligands that can recognize

receptors induced on myofibroblasts in order to target drugs16,17 or siRNA18.

As summarized in chapter 1, the focus of this thesis is on several therapeutic proteins

targeted to the disease-induced platelet-derived growth factor receptor beta (PDGFβR) via

this latter approach. We aimed to apply a sustained release formulation for such proteins, in

order to develop a patient-friendly formulation providing the gradual and prolonged release

of targeted protein therapeutics, thereby assuring a constant plasma drug concentration as

desired for chronic treatment.

Targeting of antifibrotic cytokines

During fibrosis, cells interact with each other via the release of numerous growth factors and

cytokines, which can be either disease-inducing (e.g. the profibrogenic platelet-derived

growth factor BB (PDGF-BB) and transforming growth factor beta (TGFβ)) or disease-inhibiting

(e.g. the antifibrogenic interferon gamma (IFNγ))19. Such disease inhibiting cytokines are very

interesting as therapeutic proteins, but as they are notorious for their pleiotropic effects on

different cell types, they rarely lead to the desired effects. Targeting could improve their

future clinical use20. In fact, the clinical use of many biological-based therapeutics in general

could be drastically improved using cell-selective delivery. In this way, the therapeutic

effectivity is enhanced due to the improved pharmacokinetic and pharmacodynamic profile,

while circumventing adverse effects, as reviewed recently17.

It is widely known that the PDGFβ-receptor is a disease-induced receptor in the context of

fibrosis, highly and specifically expressed on myofibroblasts21, which makes it a perfect target

for site-specific delivery of antifibrotic compounds. Another extensively studied target highly

expressed on activated fibroblasts includes for example mannose 6-phosphate/insulin-like

growth factor II receptor22, but the sugar-moieties used in this approach are commonly

7

Page 159: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

158

associated with immunogenicity. Therefore, the PDGFβR was further explored as drug target

and a series of compounds all based on the same PDGFβR-targeting moiety were developed

in the past decades 17: the cyclic PDGFβR-recognizing peptide pPB, that is able to bind to the

receptor without activating the downstream signaling cascade23. Chapter 2 describes the

rationale behind the PDGFβ-receptor targeting and summarizes the different targeted

constructs that were developed in time.

One of the most promising compounds developed in our lab appeared to be the minimalized

construct Fibroferon. This construct contains the signaling moiety of the potent antifibrotic

cytokine IFNγ, while its extracellular receptor binding part is removed. This so called mimetic

IFNγ was targeted to the PDGFβ-receptor by coupling of two pPB-moieties via a PEG linker. In

this way, the construct can only interact with and bind to the extracellular PDGFβR, thereby

preventing interaction with the widely expressed IFNγR and thus reducing side effects. The

improved antifibrotic potential and the reduction of adverse effects of Fibroferon as

compared to full length IFNγ were previously shown in mouse models of both acute and

chronic liver fibrosis as induced with CCl424 and in the UUO model for kidney fibrosis25,

respectively. Although antifibrotic effects of Fibroferon were noted and this compound might

reach the clinical phase, it remained elusive how this compound worked at the cellular level.

For that reason, in chapter 3, we evaluated the mechanism of action of this chimeric

construct by studying the cellular binding, uptake and intracellular signaling and trafficking.

We confirmed the binding of Fibroferon to the PDGFβR expressed by myofibroblasts, and

interestingly were able to show the absence of activation of the PDGFβ-signaling route.

Alternatively, we observed the induction of the signaling cascade of the IFNγ-receptor

following clathrin-dependent internalization, and subsequent nuclear translocation and

uptake. We showed that after delivery of the signaling moiety of IFNγ to the PDGFβ-receptor

we do get IFNγ-associated signaling effects. Therefore, in this study, we demonstrated the

possibility of redirecting cytokines to a cellular target of interest in general. Intriguing

questions that remained unanswered in this study, concern the switch of Fibroferon from the

targeting receptor (PDGFβR) to the effector receptor (IFNγR), escape from the endosomal

degradation pathways after uptake via the target receptor, and how the nuclear uptake of

such a construct occurs. Moreover, a fascinating question is whether this trafficking is unique

for the PDGFβR pathway, or if it is also possible with similar constructs targeting other

receptors using other intracellular routes.

Apart from IFNγ, in the past years various studies reported many other cytokines that possess

antifibrotic activity as well19. This raised the question whether IFNγ was the absolute best

choice for targeting to the main pathogenic cells in fibrosis, or whether there are other even

Page 160: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

159

more promising candidates available. For example, hepatocyte growth factor (HGF) and

interferon alpha (IFNα) were reported as antifibrogenic cytokines involved in liver disease19.

Another example is the adipocytokine adiponectin, which was mainly recognized for its anti-

inflammatory properties by inhibiting the synthesis of the proinflammatory tumor necrosis

factor alpha (TNFα) and by inducing the two important anti-inflammatory cytokines IL-10 and

IL-1 receptor antagonist26. However, it can also influence fibrosis in a direct manner by

suppressing the proliferation and migration of activated hepatic stellate cells (HSCs) as main

myofibroblast-like cells in liver fibrosis. In fact, it was shown by Kamada et al. that adiponectin

attenuated CCl4-induced liver fibrosis in mice27.

The aforementioned cytokine, TNFα, and interleukin-1 beta (IL-1β) are commonly known as

proinflammatory mediators. Actually, TNFα was shown to contribute to hepatic fibrosis by

activating local macrophages to secrete the profibrotic cytokine transforming growth factor

beta (TGFβ)19. However, a more recent study revealed that both TNFα and IL-1β can also

reverse in vitro the profibrogenic phenotype of HSCs, by upregulation of several matrix

metalloproteinases and downregulation of certain profibrogenic genes (including α-SMA and

PDGF-BB)28. The targeting of antifibrotic cytokines such as adiponectin and TNFα could

possibly be a novel therapeutic strategy in chronic (liver) disease. The essential challenge

there is to bring the signaling moiety of the cytokine of interest to the right location inside the

cell, i.e. cytoplasm or nucleus, following delivery of this moiety to a disease-induced receptor.

It remains to be seen whether this exciting strategy is applicable for other cytokines apart

from IFNγ. Future research should thus focus on developing other constructs targeting potent

cytokines to various disease-induced receptors, in this way creating several new treatment

options with fewer side effects for chronic conditions.

In the past, such targeted protein constructs were synthesized chemically in our group,

allowing the production of a well-defined product29. However, the synthetic upscaling and

purification of such constructs sometimes appeared to be problematic in terms of

reproducibility, yield and activity of the synthetized protein. Therefore, to explore the

applicability of multiple proteins in large in vivo studies and eventually clinical trials, an

alternative method providing more flexibility in protein design is necessary, such as the

biological production of complex molecules or fragments via recombinant techniques30. A

commonly used host is Escherichia coli, which is approved by drug regulatory authorities for

recombinant protein production31. Inside the host, proper folding into the native

conformation is guaranteed by a so-called protein quality control network32; a feature that is

evidently absent during chemical synthesis of proteins. However, release of the recombinant

protein from the cytoplasm of the E. coli is accompanied by binding to cellular components

including endotoxins (lipopolysaccharides) from which the protein has to be purified before

7

Page 161: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

160

administration in vivo30. This can be done with affinity chromatography, but this technique is

generally associated with low yields of purified protein. An alternative is switching to a

different bacterium strain, for example the Gram-positive and thus endotoxin free

Lactococcus lactis. Although production in L. lactis allows easy scale-up for large protein

amounts, many proteins are degraded by the internal proteolytic system. However, past and

current developments made many advances in improving the protein production and

secretion in this alternative host32.

Polymeric microspheres for sustained release of therapeutic proteins

As mentioned earlier in this chapter, polymeric microspheres are a suitable sustained release

formulation for therapeutic proteins in the lifelong treatment of chronic diseases like fibrosis,

by providing constant plasma concentrations and thus avoiding peak levels. This formulation

can be a patient-friendly alternative to repetitive parenteral administrations that are

accompanied by high patient discomfort5. Therefore, this formulation can bring the clinical

application of such proteins one step closer. Obviously, to date various sustained release

formulations are available, like injectable implants33 or nanoparticles34. Polymeric

microspheres are particularly interesting as they provide stability of the encapsulated

therapeutic proteins by physical protection from cells and enzymes in adjacent tissues5.

Proteins encapsulated in and released from polymeric microspheres do not have

denaturation issues following exposure to high temperatures which is common during the

production of implants, generally produced via hot melt extrusion35. Moreover, such

polymeric microspheres can be produced relatively easy and their composition can be simply

adjusted depending on the preferred release profile and dosage of the protein of interest.

Another advantage is that the formulation can be easily administrated via a single

subcutaneous injection5.

The most commonly used polymer in this microsphere application is the biodegradable

polymer poly (lactic-co-glycolic) acid (PLGA), from which the encapsulated protein is released

via degradation of the polymer by bulk erosion36. Currently, there are several protein-delivery

systems approved for marketing based on PLGA microspheres. Two examples include

Nutropin Depot®, containing recombinant human growth hormone that is indicated for the

long-term treatment of growth failure, and Lupron Depot® which contains leuprolide, a

synthetic analog of gonadotropin-releasing hormone, indicated for amongst others

endometriosis. Such depot products can be administered at 1, 3 or even 6 month intervals37.

However, the acidic degradation products and the hydrophobic nature of PLGA can cause

(chemical) denaturation or aggregation of encapsulated proteins and this could contribute to

Page 162: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

161

incomplete release and an undesirable release profile38. Alternative biodegradable synthetic

polymers for microsphere production include polycaprolactones, or natural polymers such as

starch and hyaluronic acid, but unlike PLGA-based microparticles, these polymers never

reached clinical approval by the FDA because they displayed lower biocompatibility and

biodegradability, or did not possess similar mechanical properties5.

In our studies we used a combination of two biodegradable and biocompatible multi-block co-

polymers as matrix of the microspheres. The two different polymers were based on

amorphous blocks, composed of poly (ε-caprolactone) (PCL) and poly (ethylene glycol) (PEG),

and semi-crystalline blocks, created with poly (L-lactic acid) (PLLA)39. Different to PLGA, the

release of proteins from microspheres produced with these polymers is diffusion-mediated

instead of degradation-mediated. This diffusion is regulated by the swelling of the PEG

fragments in the amorphous blocks when in an aqueous environment following in vivo

injection40.

We developed a microsphere formulation for PDGFβ-receptor directed human serum albumin

(pPB-HSA), which is a cell-specific drug carrier23. We extensively studied the in vitro release of

pPB-HSA from microsphere compositions with different blends of the copolymers in chapter

4, thereby aiming at a 14-day release. Moreover, we conducted an in vivo proof of concept

study in the UUO mouse model for kidney fibrosis, as the chronic nature of liver fibrosis

demands sustained release for a prolonged period of time. We were able to demonstrate

gradual and complete in vitro release of pPB-HSA within 14 days in the optimized microsphere

formulation, with both co-polymers in a 50:50 ratio. Moreover, we showed the in vivo release

of pPB-HSA in plasma 7 days after a single subcutaneous injection of these microspheres and

the enhanced accumulation of the targeted protein in the fibrotic kidney expressing the

PDGFβ-receptor. In this study, we showed that this microsphere technology is suitable as

sustained release formulation for large complex proteins, and can thus be successfully

combined with a second technology, i.e. drug targeting39.

In chapter 5, we took this concept one step further, by studying the in vivo pharmacokinetics

of pPB-HSA released from the optimized microsphere formulation in the Mdr2-/- model for

liver fibrosis. This study showed that pPB-HSA was released from the subcutaneously residing

microspheres. Both the plasma concentration and liver concentrations already reached

steady state levels within 1 day after injection and lasted for 5 days. A rather unexpected

decline in plasma and liver pPB-HSA levels 7 days after injection was associated with the

induction of an antibody response in these mice against the human albumin. This was avoided

by substituting human serum albumin with the mouse equivalent, creating the drug carrier

pPB-MSA that may be suitable for long-term studies in mice41. Obviously, for clinical studies

7

Page 163: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

162

and ultimate application in humans the carrier protein should be complementary for use in

humans, i.e. pPB-HSA. Hence, our results demonstrate a relevant pitfall that can be

encountered with exploring the therapeutic potential of biologicals; species differences

should always be taken into account, particularly when extrapolating fundamental,

experimental studies of protein therapeutics to clinical studies.

We also correlated the in vitro release data to our in vivo pharmacokinetic data, and

estimated a 20-fold lower release rate of pPB-HSA from the microspheres in vivo, relative to

in vitro studies. This lower release rate in vivo does not necessarily mean that it will be

challenging to obtain therapeutic effects when an active drug is coupled; the antifibrotic

effect of the construct greatly depends on the potency of the attached pharmacon. Evidently,

the in vitro studies are oversimplified as compared to the more complicated situation in vivo,

in which the anatomy and microenvironment of the subcutaneous space play a key role42, in

part explaining the discrepancy between these studies. The in vivo plasma levels are the net

result of release from the subcutaneous depot, uptake by the target cells and metabolism41.

The essential question that was subsequently raised, was whether the steady state

concentrations reached in the target organ (i.e. liver) were sufficient to obtain therapeutic

levels. In order to find an answer to this question, we attached the potential antifibrotic rho-

kinase inhibitor Y27632 to the drug carrier pPB-MSA, encapsulated this in the microsphere

formulation and determined the pharmacodynamics in vivo in Mdr2-/- mice chapter 6. We

confirmed that pPB-MSA-Y27632 displayed in vitro a relaxing effect on HSCs. After confirming

that PDGFβ-receptor directed Y27632 was able to exert its effect in the stellate cells as target

cells, we determined its effect in vivo, and demonstrated antifibrotic effects as reflected by a

reduction in expression of several fibrosis-related genes. Clearly, the concentration of Y27632

in the target cell is high enough to exert its effects, while the untargeted, unencapsulated

equivalent does not show any effect following daily subcutaneous injections. However, it

would still be interesting to determine the plasma levels of pPB-MSA in the treated mice, to

be able to compare these values to the ones found in the pPB-HSA treated animals.

Although the application of the polymeric microspheres used in this thesis was promising,

there is still room for improvement. These microspheres were produced by water-in-oil-in-

water emulsification by homogenization, resulting in a broad particle size distribution as

visible from scanning electron microscopy images and as reflected by high span values.

Particle size is an essential property in governing the release profile of the encapsulated

therapeutic protein and plays an important role in the fate of particles as well when they

might be recognized by (local) macrophages, which can opsonize such non-self objects43,44. By

production of the microspheres with a similar double emulsification method via membrane

Page 164: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

163

emulsification, in which the primary emulsion is forced through uniform pores of a glass

membrane into the external water phase, a narrow particle size distribution can be

obtained45. In this way, the release behavior of the encapsulated drug can be controlled more

precisely and additionally better biocompatibility can be obtained with cells and tissues in

vivo45.

Concluding remarks and new directions

Liver fibrosis can be caused by viral infections, alcohol and genetic disorders, but obesity

leading to non-alcoholic fatty liver disease (NAFLD) including non-alcoholic steatohepatitis

(NASH) is nowadays forming one of the major threats to global health46,47. Currently, the only

available therapeutics for patients with liver fibrosis or cirrhosis are the new generation

antiviral agents, but they are only effective in case the disease is caused by hepatitis B and C

viral infections7,9. Various recent studies suggest that the fibrotic process is halted or even

reversed after successful eradication of the causative agent9. Several therapeutics are in

clinical trials, some of them protein-based, including antibodies for example against the

profibrotic cytokine TGFβ1 or the regulator of the ECM-crosslinking enzyme LOXL29. However,

most therapeutic proteins to treat liver diseases are not ready yet to enter the market48.

Protein-based drugs represent a group of fastest growing new medicines for the treatment of

nearly 150 indications, including cancer, diabetes, hepatitis and rheumatoid arthritis.

However, clinical application is still limited or associated with severe side effects, frequently

related to the administration route, immunogenicity or hepatotoxicity20. Sustained release

formulations such as the microsphere technology might therefore open patient-friendly

therapeutic treatment options for patients suffering from many chronic indications, and such

formulations may also enhance the therapeutic efficacy by providing a sustained release3,49.

In this thesis we were able to show the successful application of the microsphere technology

for the delivery of (targeted) therapeutic compounds in the context of (liver) fibrosis.

Clearly, many advances are ongoing in (pre-) clinical drug development for liver diseases,

however, there are still many scientific hurdles to take. The focus of future research should be

both on the search for the most potent targeted antifibrotic proteins that show the least

adverse effects and on a suitable patient-friendly (microsphere) formulation with the right

polymer composition. When using the in this thesis described versatile polymer platform,

suitable formulations can be designed and developed for any potent proteinaceous

antifibrotic drug, possibly derived from endogenous mediators, with any desired release

profile. In this way, we will gradually move towards the clinical application of a combined

7

Page 165: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

164

delivery system that provides the sustained release of effective (targeted) antifibrotic

proteins. It is important to unite the forces from both the pharmaceutical technology and

drug targeting field, because if you want to go fast, go alone, but if you want to go far, go

together (African proverb).

Page 166: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

165

References

1. Leader B, Baca QJ, Golan DE. Protein therapeutics: A summary and pharmacological

classification. Nat Rev Drug Discov. 2008;7(1):21-39.

2. Kinch MS. An overview of FDA-approved biologics medicines. Drug Discov Today.

2015;20(4):393-398.

3. Wu F, Jin T. Polymer-based sustained-release dosage forms for protein drugs,

challenges, and recent advances. AAPS PharmSciTech. 2008;9(4):1218-1229.

4. Prajapati VD, Jani GK, Kapadia JR. Current knowledge on biodegradable microspheres in

drug delivery. Expert Opin Drug Deliv. 2015;12(8):1283-1299.

5. Vaishya R, Khurana V, Patel S, Mitra AK. Long-term delivery of protein therapeutics.

Expert Opin Drug Deliv. 2015;12(3):415-440.

6. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology. 2008;134(6):1655-

1669.

7. Popov Y, Schuppan D. Targeting liver fibrosis: Strategies for development and validation

of antifibrotic therapies. Hepatology. 2009;50(4):1294-1306.

8. Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. 2008;371(9615):838-851.

9. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest. 2013;123(5):1887-

1901.

10. World Health Organisation. Health statistics and information systems; estimates for

2000-2015.

http://www.who.int/healthinfo/global_burden_disease/estimates/en/index1.html.

Accessed 12/13, 2017.

11. Farrell GC, Larter CZ. Nonalcoholic fatty liver disease: From steatosis to cirrhosis.

Hepatology. 2006;43(2 Suppl 1):S99-S112.

12. Said A, Lucey MR. Liver transplantation: An update 2008. Curr Opin Gastroenterol.

2008;24(3):339-345.

13. Eckes B, Zweers MC, Zhang ZG, et al. Mechanical tension and integrin alpha 2 beta 1

regulate fibroblast functions. J Investig Dermatol Symp Proc. 2006;11(1):66-72.

14. Neef M, Ledermann M, Saegesser H, et al. Oral imatinib treatment reduces early

fibrogenesis but does not prevent progression in the long term. J Hepatol.

2006;44(1):167-175.

15. Love KT, Mahon KP, Levins CG, et al. Lipid-like materials for low-dose, in vivo gene

silencing. Proc Natl Acad Sci U S A. 2010;107(5):1864-1869.

16. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted

interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology. 2011;54(2):586-596.

7

Page 167: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

166

17. van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted therapies in liver fibrosis: Combining

the best parts of platelet-derived growth factor BB and interferon gamma. Front Med

(Lausanne). 2015;2:72.

18. Sato Y, Murase K, Kato J, et al. Resolution of liver cirrhosis using vitamin A-coupled

liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol.

2008;26(4):431-442.

19. Tilg H, Kaser A, Moschen AR. How to modulate inflammatory cytokines in liver diseases.

Liver Int. 2006;26(9):1029-1039.

20. Pisal DS, Kosloski MP, Balu-Iyer SV. Delivery of therapeutic proteins. J Pharm Sci.

2010;99(6):2557-2575.

21. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, et al. Platelet-derived growth factor

isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest.

2008;88(10):1090-1100.

22. Beljaars L, Molema G, Weert B, et al. Albumin modified with mannose 6-phosphate: A

potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic

stellate cells. Hepatology. 1999;29(5):1486-1493.

23. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet

derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in

fibrotic tissue. Biochem Pharmacol. 2003;66(7):1307-1317.

24. Bansal R, Prakash J, De Ruiter M, Poelstra K. Interferon gamma peptidomimetic targeted

to hepatic stellate cells ameliorates acute and chronic liver fibrosis in vivo. J Control

Release. 2014;179:18-24.

25. Poosti F, Bansal R, Yazdani S, et al. Interferon gamma peptidomimetic targeted to

interstitial myofibroblasts attenuates renal fibrosis after unilateral ureteral obstruction

in mice. Oncotarget. 2016;7(34):54240-54252.

26. Wolf AM, Wolf D, Rumpold H, Enrich B, Tilg H. Adiponectin induces the anti-

inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res

Commun. 2004;323(2):630-635.

27. Kamada Y, Tamura S, Kiso S, et al. Enhanced carbon tetrachloride-induced liver fibrosis in

mice lacking adiponectin. Gastroenterology. 2003;125(6):1796-1807.

28. Robert S, Gicquel T, Bodin A, Lagente V, Boichot E. Characterization of the MMP/TIMP

imbalance and collagen production induced by IL-1beta or TNF-alpha release from

human hepatic stellate cells. PLoS One. 2016;11(4):e0153118.

29. Nilsson BL, Soellner MB, Raines RT. Chemical synthesis of proteins. Annu Rev Biophys

Biomol Struct. 2005;34:91-118.

30. Overton TW. Recombinant protein production in bacterial hosts. Drug Discov Today.

2014;19(5):590-601.

Page 168: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Summary and general discussion

167

31. Huang CJ, Lin H, Yang X. Industrial production of recombinant therapeutics in escherichia

coli and its recent advancements. J Ind Microbiol Biotechnol. 2012;39(3):383-399.

32. Morello E, Bermudez-Humaran LG, Llull D, et al. Lactococcus lactis, an efficient cell

factory for recombinant protein production and secretion. J Mol Microbiol Biotechnol.

2008;14(1-3):48-58.

33. Agarwal P, Rupenthal ID. Injectable implants for the sustained release of protein and

peptide drugs. Drug Discov Today. 2013;18(7-8):337-349.

34. Lamprecht A, Ubrich N, Yamamoto H, et al. Biodegradable nanoparticles for targeted

drug delivery in treatment of inflammatory bowel disease. J Pharmacol Exp Ther.

2001;299(2):775-781.

35. Stankovic M, Frijlink HW, Hinrichs WL. Polymeric formulations for drug release prepared

by hot melt extrusion: Application and characterization. Drug Discov Today.

2015;20(7):812-823.

36. van de Weert M, Hennink WE, Jiskoot W. Protein instability in poly(lactic-co-glycolic acid)

microparticles. Pharm Res. 2000;17(10):1159-1167.

37. Jain A, Kunduru KR, Basu A, Mizrahi B, Domb AJ, Khan W. Injectable formulations of

poly(lactic acid) and its copolymers in clinical use. Adv Drug Deliv Rev. 2016;107:213-

227.

38. Houchin ML, Topp EM. Chemical degradation of peptides and proteins in PLGA: A review

of reactions and mechanisms. J Pharm Sci. 2008;97(7):2395-2404.

39. Teekamp N, Van Dijk F, Broesder A, et al. Polymeric microspheres for the sustained

release of a protein-based drug carrier targeting the PDGFbeta-receptor in the fibrotic

kidney. Int J Pharm. 2017;534(1-2):229-236.

40. Stankovic M, de Waard H, Steendam R, et al. Low temperature extruded implants based

on novel hydrophilic multiblock copolymer for long-term protein delivery. Eur J Pharm

Sci. 2013;49(4):578-587.

41. van Dijk F, Teekamp N, Beljaars L, et al. Pharmacokinetics of a sustained release

formulation of PDGFbeta-receptor directed carrier proteins to target the fibrotic liver. J

Control Release. 2017;269:258-265.

42. Weiser JR, Saltzman WM. Controlled release for local delivery of drugs: Barriers and

models. J Control Release. 2014;190:664-673.

43. Shanbhag AS, Jacobs JJ, Black J, Galante JO, Glant TT. Macrophage/particle interactions:

Effect of size, composition and surface area. J Biomed Mater Res. 1994;28(1):81-90.

44. Yue H, Wei W, Yue Z, et al. Particle size affects the cellular response in macrophages. Eur

J Pharm Sci. 2010;41(5):650-657.

7

Page 169: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 7

168

45. Liu W, Yang XL, Ho WS. Preparation of uniform-sized multiple emulsions and micro/nano

particulates for drug delivery by membrane emulsification. J Pharm Sci. 2011;100(1):75-

93.

46. Mehal WZ, Iredale J, Friedman SL. Scraping fibrosis: Expressway to the core of fibrosis.

Nat Med. 2011;17(5):552-553.

47. Stal P. Liver fibrosis in non-alcoholic fatty liver disease - diagnostic challenge with

prognostic significance. World J Gastroenterol. 2015;21(39):11077-11087.

48. ClinicalTrials. www.clinicaltrials.gov. Accessed 12/13, 2017.

49. Frokjaer S, Otzen DE. Protein drug stability: A formulation challenge. Nat Rev Drug

Discov. 2005;4(4):298-306.

Page 170: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department
Page 171: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

8

Page 172: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

[Type text]

Nederlandse samenvatting

Dankwoord

Curriculum vitae

List of publications

Page 173: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

172

Nederlandse samenvatting

Fibrose is een chronische ziekte die steeds meer voorkomt en wereldwijd veel dodelijke

slachtoffers eist. De aandoening wordt gekenmerkt door de vorming van littekenweefsel in

een orgaan, als gevolg van een ontspoorde wondgenezing, waardoor een juiste werking van

het orgaan wordt aangetast. Dit proces kan optreden in verschillende organen, waaronder de

lever, nieren en longen, maar kan op een soortgelijke manier ontstaan in tumoren. Mogelijke

oorzaken van fibrose kunnen het metabool syndroom of genetische aandoeningen zijn.

In een gezond persoon komt een wondgenezingsreactie op gang wanneer er schade wordt

aangericht aan epitheelcellen in een bepaald orgaan. Deze wondgenezing gaat gepaard met

het aantrekken en activeren van immuuncellen, die vervolgens het orgaan infiltreren. Aldaar

scheiden de immuuncellen verschillende mediatoren uit (cytokines en groeifactoren), die een

activerende werking hebben op de centrale cel betrokken bij het fibrotische proces, te weten

de fibroblast. In geactiveerde toestand wordt deze cel myofibroblast genoemd, en produceert

dan extracellulaire matrix (ECM) eiwitten, met name fibrillaire collagenen, om de wond te

dichten. Als de schadelijke prikkel van tijdelijke aard was en dus na verloop van tijd verdwijnt,

schakelen de myofibroblasten zichzelf uit middels een proces genaamd apoptose

(geprogrammeerde celdood), of ze keren terug naar hun inactieve, rustende toestand, en het

littekenweefsel wordt netjes opgeruimd.

In het geval van een aanhoudende prikkel leidend tot chronische ontsteking, versterkt de

wondgenezingsreactie zichzelf en is er overmatige productie en afzetting van ECM eiwitten in

het orgaan. In dat geval spreekt men van fibrose (Fig. 1). Als de ziekte zich op deze manier

verder ontwikkelt, worden in een gevorderd stadium functionele cellen vervangen door ECM

eiwitten. Het orgaan kan daardoor steeds moeilijker zijn normale functies uitvoeren. Ondanks

het groeiende aantal patiënten zijn er momenteel zeer beperkte mogelijkheden voor

behandeling en in veel gevallen is de enige effectieve behandeling orgaantransplantatie.

Page 174: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Nederlandse samenvatting

173

Figuur 1. Schematische weergave van fibrose, onder andere voorkomend in lever, nier, hart, longen, darmen,

huid en tumoren. Wanneer epitheelcellen beschadigd raken, leidt dit tot een ontstekingsreactie door de

aantrekking van immuuncellen. Dit zorgt voor activatie van fibroblasten tot myofibroblasten, die de PDGFβ-

receptor verhoogd tot expressie brengen. De activatie gaat gepaard met de productie van extracellulaire matrix

eiwitten die bij langdurige schade de functionele cellen vervangen. Afbeelding aangepast van Yazdani et al.

(2017), Advanced Drug Delivery Reviews.

Het is intussen een algemeen geaccepteerde opvatting dat het fibrotische proces tot op

zekere hoogte omkeerbaar is. Dit inzicht heeft een nieuwe impuls gegeven aan het fibrose

onderzoek om de moleculaire mechanismen die ten grondslag liggen aan het fibrotische

proces op te helderen, en om op basis hiervan nieuwe en effectieve therapieën te

ontwikkelen. In de huidige zoektocht naar een effectieve therapie voor fibrose richten veel

studies zich op de myofibroblast als doelwitcel, omdat zij de belangrijkste matrix

producerende cellen zijn in het ziekteproces. Een veelbelovende strategie is de zogenaamde

drug targeting aanpak, waarbij de activiteit van deze myofibroblasten in het bijzonder wordt

verminderd door potente geneesmiddelen specifiek in deze cellen af te leveren.

Celspecifieke afgifte is mogelijk door gebruik te maken van bepaalde eiwitten die de fibroblast

uitsluitend in zijn geactiveerde toestand op zijn celmembraan tot expressie brengt,

waaronder een eiwit genaamd PDGFβ-receptor (PDGFβR). Door het ontwerpen van kleine

peptides die deze receptor kunnen herkennen en hier specifiek aan kunnen binden, en een

aantal van deze eenheden te koppelen aan groter eiwit, wordt een PDGFβR gestuurde

geneesmiddeldrager (carrier) gecreëerd. Wanneer een geneesmiddel wordt gebonden aan

deze carrier, kan het gekoppelde geneesmiddel op deze manier worden afgeleverd bij of in de

8

Page 175: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

174

myofibroblast als doelwitcel (Fig. 2). Het eerste doel van dit proefschrift is om de

verschillende ontwikkelde PDGFβR gestuurde eiwit constructen te evalueren, en meer inzicht

te verkrijgen in het werkingsmechanisme achter de meest veelbelovende kandidaat met

potentie om in de toekomst als therapie voor fibrose te dienen (Fibroferon). Dit wordt kort

uiteengezet in hoofdstuk 1, waarna in hoofdstuk 2 de ontwikkeling van deze zogeheten

PDGFβR targeting wordt beschreven en een overzicht geeft van alle ontworpen constructen.

Vervolgens licht hoofdstuk 3 het veelbelovende therapeutisch eiwit Fibroferon uit, en laat zien

hoe dit op celniveau bindt, wordt opgenomen en verwerkt.

Figuur 2. Schematische voorstelling van celspecifieke geneesmiddel afgifte. Als de fibroblast geactiveerd wordt,

zoals gebeurt tijdens de ontwikkeling van orgaanfibrose, transformeert deze in een myofibroblast. In deze staat

brengt de cel verhoogd en specifiek de PDGFβ-receptor (PDGFβR) tot expressie op zijn celmembraan.

Geneesmiddelen kunnen selectief worden afgeleverd bij de myofibroblast door deze te koppelen aan meerdere

PDGFβR herkennende peptides. Als het geneesmiddel is opgenomen in de cel, kan het daar zijn werking

uitoefenen en de myofibroblast terugbrengen naar zijn oorspronkelijke rustende toestand.

Een grote uitdaging bij de uiteindelijke toepassing van dergelijke therapeutische eiwitten is de

toedieningsvorm. Over het algemeen worden zulke therapeutica toegediend via injecties

omdat ze worden afgebroken in het maagdarm kanaal. Directe injectie in de bloedbaan levert

de grootste biologische beschikbaarheid van het middel op, maar wanneer frequente

toediening noodzakelijk is, zorgt dit voor een grote belasting voor de patiënt, zeker in het

Page 176: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Nederlandse samenvatting

175

geval van een chronische aandoening waarbij langdurige behandeling vereist is. Dit probleem

kan worden omzeild door gebruik te maken van preparaten die gereguleerde afgifte

verschaffen, waardoor er langdurige en geleidelijke afgifte van het therapeutische eiwit

gegarandeerd wordt na een eenmalige injectie. Hiervoor zijn meerdere alternatieven

beschikbaar, waaronder zogenaamde polymere microsferen. Dit zijn bolletjes gemaakt van

polymeren met diameters in de micrometer orde, waarin het therapeutische eiwit zorgvuldig

wordt verpakt.

Het tweede doel van dit onderzoek is om de geneesmiddel targeting naar de PDGFβR te

combineren met de gereguleerde afgifte technologie en dit te testen in proefdieren. De

combinatie van beide technologieën kan de toekomstige behandeling van fibrose een stap

dichterbij brengen (Fig. 3). Dit principe is onderzocht in hoofdstuk 4, waarin een PDGFβ-

receptor gestuurde geneesmiddeldrager, dus zonder gekoppeld geneesmiddel, in microsferen

met verschillende polymeercomposities is verpakt op zoek naar het gewenste afgifte profiel.

Het hoofdstuk laat zien hoe het therapeutische eiwit in de tijd vrijkomt uit de microsferen, en

bovendien dat het eiwit na 7 dagen nog terug te vinden is in het bloed en voornamelijk in de

fibrotische nier van muizen, waar de PDGFβR verhoogd tot expressie wordt gebracht op de

myofibroblasten. Dit is best indrukwekkend aangezien de geneesmiddeldrager in het bloed

snel wordt afgebroken (de halfwaardetijd in bloed is ongeveer 40 minuten).

Figuur 3. Grafische weergave van de gereguleerde afgifte van een celspecifiek geneesmiddel. Zieke muizen

lijdend aan fibrose worden eenmalig geïnjecteerd met een gereguleerde afgifte preparaat (polymere

microsferen) voor een PDGFβR gestuurd geneesmiddel. Dit preparaat garandeert langdurige en geleidelijke

afgifte van het geneesmiddel uit het depot in de bloedcirculatie van de muis, terwijl de microsferen op de

injectieplek blijven zitten. Op deze manier bereikt het celspecifieke geneesmiddel de myofibroblasten in het

8

Page 177: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

176

fibrotische orgaan, alwaar zij hun werking kunnen uitoefenen en door remming van het fibrotische proces de

muis kunnen genezen.

Hoofdstuk 5 gaat nog een stapje verder, door de afgifte van de geneesmiddeldrager in de tijd

uitgebreider in proefdieren te bestuderen. Studies in een muismodel voor leverfibrose laten

zien dat al binnen 1 dag na injectie een constante bloed- en leverconcentratie van het

afgegeven eiwit te meten is, die minstens een week aanhoudt. Dit betekent dat met slechts 1

injectie langdurig een constante bloedwaarde kan worden bereikt, en dus dat schommelingen

in de eiwit bloedwaarden als gevolg van frequente injecties, vaak geassocieerd met

bijwerkingen, kunnen worden voorkomen. Verder komt in dit hoofdstuk aan bod dat er een

immunologische reactie in proefdieren kan optreden wanneer een lichaamsvreemd eiwit

wordt geïnjecteerd. Gelukkig kan deze respons vermeden worden door het proefdier te

behandelen met een voor zijn soort bekend, al dan niet gemodificeerd, eiwit.

Omdat een nieuwe behandeling voor fibrose noodzakelijk is, laat hoofdstuk 6 zien dat de

toepassing van deze microsferen ook bruikbaar is voor een potentieel geneesmiddel

gekoppeld aan de drager. Zieke muizen werden eenmalig geïnjecteerd met microsferen die

de PDGFβR gestuurde geneesmiddeldrager bevatten, met daaraan gekoppeld een actief

geneesmiddel. In deze muizen werd een duidelijke verlaging van de fibrose in de lever

gemeten. Omdat door het gebruik van microsferen frequente injecties niet meer nodig zijn,

heeft dit grote implicaties voor de toekomstige behandeling van niet alleen fibrose, maar van

chronische ziekten in het algemeen.

Samenvattend heeft het onderzoek zoals beschreven in dit proefschrift aangetoond dat

verschillende soorten therapeutische eiwitten in een doelwitcel (de myofibroblast) kunnen

worden afgeleverd via de PDGFβR. Door deze PDGFβR gestuurde eiwitten middels een

gereguleerde afgifte preparaat toe te dienen, kunnen schommelingen in bloedwaarden van

het geneesmiddel worden voorkomen, zoals blijkt uit de langdurig constante bloedspiegel na

1 microsfeerinjectie met het eiwit met een zeer korte halfwaardetijd, en dus bijwerkingen

worden verminderd. Hierdoor kan de injectiefrequentie en daarmee de belasting voor de

patiënt drastisch worden verlaagd; dit alles terwijl het gemodificeerde geneesmiddel zijn

effect behoudt. Met deze resultaten is de klinische toepassing van een dergelijk

geneesmiddelpreparaat, die de langdurige afgifte van een celspecifiek geneesmiddel

waarborgt, een stap dichterbij gekomen.

Page 178: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Dankwoord

177

Dankwoord

Vier jaren zijn voorbij gevlogen en ineens ligt daar dit boekje als tastbaar bewijs. Het is gelukt!

De steun van een aantal mensen was hierbij voor mij van onschatbare waarde, en deze

mensen wil ik in dit laatste hoofdstuk in het bijzonder bedanken. Promoveren kun je namelijk

onmogelijk in je eentje.

Allereerst wil ik graag mijn begeleiders bedanken. Leonie, omdat jouw betrokkenheid voor mij

heel waardevol was, ben je vanzelfsprekend de eerste in dit rijtje. Je positieve, kundige

inbreng en je open houding hebben grote invloed gehad op de succesvolle afronding van mijn

project. Met je persoonlijke en vrolijke benadering wist je me middels een paar kleine

suggesties altijd weer in de goede richting te duwen of over een ogenschijnlijk

onoverkomelijke hobbel te helpen. Ik heb heel veel van je geleerd en kijk uit naar het vervolg

van onze samenwerking. Ook gaat mijn dank uit naar mijn promotores Klaas en Peter, voor

jullie enthousiasme, kundigheid, aangename begeleiding en de wetenschappelijke roze bril

die jullie beiden dragen. Klaas, bedankt voor de kans die je me hebt gegeven om in jouw

groep onderzoek te kunnen doen. Je hebt me alle vrijheid gegeven om mijn eigen weg te

zoeken en daarbij te leren van mijn eigen fouten; iets waar ik de rest van mijn leven wat aan

zal hebben. Ik heb met heel veel plezier in jouw groep gewerkt, en ben erg verheugd dat ik

nog even kan blijven om me verder te ontwikkelen. Peter, niemand kan beter improviseren

dan jij. Elke keer wanneer het lood mij in de schoenen was gezakt en ik het project echt niet

meer zag zitten bedacht jij weer een nieuwe richting om verder te kunnen. Jij laat je door

niets of niemand kisten, en daar heb ik veel bewondering voor. Ik vind het lovenswaardig hoe

je zoveel verschillende PhD studenten met evenveel aandacht begeleidt en samen brengt.

Bedankt voor de leuke tijd in je groep. Verder wil ik mijn tweede copromotor Wouter

bedanken, voor je belangrijke bijdrage aan met name het tweede, meer technische deel van

mijn project. Ik heb inhoudelijk veel van je geleerd, en wil je ook zeker bedanken voor de

opbouwende kritieken op manuscripten, welke je altijd vliegensvlug aanleverde.

I would like to thank the assessment committee, prof. Klaas Nico Faber, prof. Reinoud Gosens

and prof. Jonel Trebicka, for reading and evaluating this thesis.

Naomi, wat hebben we een leuke samenwerking gehad de afgelopen jaren. Ik vond het een

verrijking om jou als maatje te hebben om inhoudelijk mee te sparren, maar ook om

persoonlijke dingen mee te delen. Als kroon op ons werk hebben we twee publicaties

gescoord om trots op te zijn, en hopelijk is de derde onderweg. Ik bewaar goede

herinneringen aan ons Duitse avontuur en hoop dat we in de toekomst contact zullen

houden.

8

Page 179: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

178

Mijn steun en toeverlaat Eduard, bedankt voor alles. Je weet: zonder jou was het nooit wat

geworden. Niet voor niets ben je coauteur op alle hoofdstukken van dit boekje. Je hebt me

heel veel geleerd op het lab, maar door je openhartigheid en omdat je eigenlijk best wijs bent

(dat komt vast door je leeftijd) ook zeker over het leven. Ik zal onze road trip naar Mainz nooit

vergeten. Wie anders dan jij kan mijn paranimf zijn.

Als jij er niet was geweest Barbro, was ik überhaupt nooit aan een PhD binnen de farmacie

begonnen. Jij hebt mij tijdens mijn eerste onderzoeksproject wegwijs gemaakt binnen de

(farmaceutische) wetenschappen en aangestoken met je enthousiasme. Jij bent een rolmodel

voor jonge wetenschappers, en voor vrouwen in het bijzonder. Bedankt voor je

vriendelijkheid, betrokkenheid en je waardevolle inhoudelijke suggesties.

Graag zou ik Erik Frijlink willen bedanken, omdat zonder jouw bijdrage dit project niet van

start had kunnen gaan. Ik vind het bewonderenswaardig hoe je jonge onderzoekers stimuleert

en helpt om hun weg te vinden in de wetenschap.

As part of my research I had the chance to visit the lab of prof. Schuppan in Mainz twice.

Thank you for having us in your lab. It was very nice to participate in your group for this short

period, during which I learned a lot. I’m really pleased that our collaboration resulted in one

publication and a second paper in preparation. I would like to sincerely thank Yong Ook,

Kyoung-Sook and Rosario for your hospitality and all the effort you made during our stays.

De voortgang van mijn onderzoek werd gedurende de afgelopen jaren enorm bevorderd door

samenwerkingen met verschillende bedrijven. Bedankt Rob Steendam en Johan Zuidema, voor

de leerzame maandelijkse werkbesprekingen, jullie nuttige input en de prettige

samenwerking. Herman Steen en Guus van Scharrenburg, bedankt voor jullie tijd en inbreng

tijdens inhoudelijke discussies, met name gedurende het eerste deel van mijn project.

Daarnaast wil ik HendrikJan Houthoff en Niels Sijbrandi bedanken voor jullie snelle leveringen

en aangename communicatie.

I would like to thank Jan-Luuk Hillebrands and Peter Horvatovich for the pleasant

collaboration.

Tijdens de periodes dat mijn onderzoek niet van een leien dakje ging, heeft dit me nooit mijn

werkplezier ontnomen. De analisten vormen de vaste basis van de groep en hebben daar een

groot aandeel in gehad. Marina, jouw positieve houding is goud waard en mag een voorbeeld

zijn voor velen. Bedankt voor je geduldige hulp met het slicen! Catharina, je bent altijd bereid

om anderen te helpen. Zonder jouw hulp was ik nu waarschijnlijk nog steeds allerlei

kleuringen en andere proeven aan het optimaliseren, waarvoor veel dank! Ook wil ik Dorenda

bedanken voor je hulp met alles, maar vooral voor de gezelligheid en levendigheid die je met

Page 180: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Dankwoord

179

je meebrengt. Ik hoop dat we in de toekomst nog regelmatig een kopje thee zullen drinken.

Jan V., bedankt voor je vrolijkheid en alle gezellige praatjes. Het is een stuk stiller geworden

op de afdeling sinds je met pensioen bent, dus des te gezelliger dat je af en toe nog eens

stiekem langskomt.

Niet alleen de analisten vormen een vaste basis, ook de staff en het secretariaat vervullen

hierin een belangrijke rol. The one and only Gillian, je bent de zon van de afdeling. Je helpt

iedereen waar mogelijk, je bent altijd in voor een grap, en je biedt een luisterend oor als

iemand het nodig heeft. Anna, Inge, Geny, Hans and Daan, thank you for the good working

atmosphere in the department and moreover for your questions and valuable suggestions

during work discussions. Angela, it was nice to collaborate with your lab in Cardiff.

Ik heb tijdens mijn PhD het geluk gehad het kantoor te delen met de allerleukste

kamergenoten van heel farmacie. Lieve Carian en Christina, wat hebben we het onwijs gezellig

gehad samen. Cariantje, met jou is het altijd lachen, maar ook heel fijn praten. Je

onverwoestbare energie, je oneindige enthousiasme en je eerlijkheid zijn een voorbeeld voor

mij. Om nog maar te zwijgen over je talent om mensen aan te sturen en taken te delegeren.

Dat die opgedane ervaring nog eens goed van pas zou komen in je persoonlijke leven had

niemand kunnen voorspellen. Ik ben onwijs trots op hoe jij en Sniegy het samen regelen en ik

weet zeker dat er niemand is in de wereld die het beter zou kunnen. Chrisje, jij bent altijd

vrolijk, opgewekt, nieuwsgierig en hebt een niet te stillen eetlust. Ik heb onze traditie van de

tweede lunch om vier uur er nog altijd ingehouden hoor! Wat was het leuk om elkaar

afgelopen herfst weer te zien in Ann Arbor, en om daar de draad gewoon weer op te pakken

waar we in Groningen waren gebleven. Geweldig om te zien hoe jij en Frank in zo’n korte tijd

een heel nieuw bestaan hebben opgebouwd. Als jullie maar wel terugkomen! Dear Adhy, we

both started our projects four years ago and grew up together as roommates. I enjoyed

learning from each other’s cultures from the conversations we had. I think many people can

learn from your kindness and your positivity. All the best in wrapping up your project and I

hope that you will be reunited soon with your beautiful family. Lieve Anienke, we delen al

bijna twee jaren elke dag lief en leed van het lab en vooral van daarbuiten. We lachen wat af,

maar daarnaast weet je mij regelmatig net een andere, frisse kijk op allerhande zaken te

geven. Je hebt zo veel in huis dat ik zeker weet dat je alles kunt bereiken wat je voor ogen

hebt. Ik waardeer je grenzeloze solidariteit enorm en hoop dat we nog heel wat jaren aan

onze vriendschap vast kunnen plakken. Habibie, our time together in room 318 was only short

but fun. Lots of success with your project! I hope that your family will soon live with you here

in Groningen.

8

Page 181: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

180

Thanks to all the nice current and former colleagues from both the PTT department (Amirah,

Aldy, Andreia, Roberta, Benoit, Henk, Christa, Natalia, Laura, Valentina, Hector, Marcel, Ming,

Marjolijn, Daphne, Xiaoyu, Sylvia, Shanshan, Sarah, Viktoriia, Bert, Amit, Gwenda, Suresh,

Natasha, Keni, Harita) and the PTB department (Isabel, BT, Emilia, Emma, Inge, Jasper, Rick, Su,

Tobias, Gerian, Mitchel, Nia, Radit, Jo, Imco, Max, Sonja, Jan), for the fun in and outside the

lab. I wish you all the best and good luck with your projects. Kaisa, kiitos for your good

company in the lab. It was a pleasure to have you as a colleague and a shame that you had to

leave. Miriam, dank je wel voor je geduldige en kundige hulp met de arrays. Ik hoop dat je

succes hebt met je beursaanvraag!

The enthusiastic work of a number of bachelor- and master students generated interesting

results, of which the majority can be found in this thesis. Thanks a lot for your contributions

Xu, Marjolein Bouwhuis, Niek, Marjolein van der Putten, Merel, Annemarie, Jitske and Valmira!

Met dit dankwoord dat op zijn einde loopt, kom ik steeds dichter bij mijn inner circle:

vrienden en familie. Aan mijn vrienden, bedankt voor jullie vriendschap door dik en dun.

Dankzij jullie weet ik wat er echt toe doet en jullie maken het leven zoveel leuker.

Lieve Hessel, vanaf het einde van de middelbare school hebben we elkaar gevonden en niet

meer losgelaten. Met een half woord begrijpen we elkaar. Het is heerlijk een vriendin te

hebben die van dezelfde vieze grapjes houdt, maar die tegelijk als geen ander begrijpt en

geïnteresseerd is in waar ik me mee bezig hou wanneer ik doe alsof ik volwassen ben. Ik vind

het een eer dat je mijn paranimf bent.

Dan blijft mijn familie over: lieve Pieter, Reinette, Huib, Hanne en Pieter, al bijna 10 jaar mijn

tweede familie. Bedankt voor de welkome afleiding tijdens alle gezellige weekenden met

heerlijke etentjes. Ik waardeer jullie gastvrijheid en open kijk op de wereld zeer.

Lieve papa, mama en Simon, zonder jullie onvoorwaardelijke steun was ik nergens gekomen.

Pap en mam, ik bewonder hoeveel rust en liefde jullie samen hebben en geven aan anderen.

Jullie hebben voor mij alle voorwaarden gecreëerd om met vertrouwen datgene te gaan doen

waar ik me goed bij voel en hebben me daarbij geleidelijk op eigen benen laten staan. Siem,

jouw onbevangen, relaxte houding is voor mij een gezond tegenwicht wanneer ik iets weer

eens te serieus neem. Ik kan me geen lievere ouders en broer wensen dan jullie.

Mijn liefde Jakko, mijn betere helft. Jij bent alles wat ik nodig heb en meer dan ik me kan

wensen. Je laat me elke dag opnieuw zien wat het allerbelangrijkste is in ons leven, en je laat

bijzaken naar de achtergrond verdwijnen. Jouw onvoorwaardelijke liefde en steun maken mij

het gelukkigste mens op aarde.

Page 182: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Curriculum vitae

181

Curriculum vitae

21 October 1989 Born in Groningen, The Netherlands

2001-2007 Preuniversity education at Augustinus College,

Groningen, The Netherlands

2007-2011 Bachelor Chemistry (track Biochemistry) with minor Pharmacy,

University of Groningen, The Netherlands

2011-2013 Master Medical Pharmaceutical Sciences, University of Groningen,

The Netherlands

2014-2018 Ph.D. project at the Groningen Research Institute for Pharmacy,

University of Groningen, The Netherlands, under the supervision of

prof. dr. K. Poelstra and prof. dr. P. Olinga. Subject: delivery of

biologicals.

8

Page 183: University of Groningen Delivery of biologicals van Dijk ...Fransien van Dijk . The work described in this thesis was performed at the Groningen Research Institute of Pharmacy, department

Chapter 8

182

List of publications

Published articles

F. van Dijk, P. Olinga, K. Poelstra and L. Beljaars (2015): Targeted Therapies in Liver Fibrosis:

Combining the Best Parts of Platelet-Derived Growth Factor BB and Interferon Gamma;

Frontiers in Medicine 2 (72).

N. Teekamp*, F. van Dijk*, A. Broesder, M. Evers, J. Zuidema, R. Steendam, E. Post, J.L.

Hillebrands, H.W. Frijlink, K. Poelstra, L. Beljaars, P. Olinga and W.L.J. Hinrichs: Polymeric

microspheres for the sustained release of a protein-based drug carrier targeting the PDGFβ-

receptor in the fibrotic kidney; International Journal of Pharmaceutics 534 (2017).

F. van Dijk*, N. Teekamp*, L. Beljaars, E. Post, J. Zuidema, R. Steendam, Y.O. Kim, H.W. Frijlink,

D. Schuppan, K. Poelstra, W.L.J. Hinrichs and P. Olinga: Pharmacokinetics of a sustained

release formulation of PDGFβ-receptor directed carrier proteins to target the fibrotic liver;

Journal of Controlled Release 269 (2018).

* authors contributed equally

Manuscripts in preparation

F. van Dijk, L. Beljaars, E. Post, C. Draijer, X. Hu, K. Luoto, P. Olinga and K. Poelstra: Steering

therapeutic proteins: the mechanism of action of mimetic IFNγ delivered to the disease-

induced PDGFβ-receptor.

F. van Dijk, N. Teekamp, E. Post, D. Schuppan, Y.O. Kim, J. Zuidema, R. Steendam, A. Casini,

P.L. Horvatovich, N.J. Sijbrandi, H.W. Frijlink, W.L.J. Hinrichs, K. Poelstra, L. Beljaars and P.

Olinga: The antifibrotic potential of a sustained release formulation of a PDGFβ-receptor

targeted rho kinase inhibitor.