Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering...

143
Research Collection Doctoral Thesis Engineering delivery and release of protein therapeutics for mammalian systems Author(s): Fluri, David Andreas Publication Date: 2008 Permanent Link: https://doi.org/10.3929/ethz-a-005708602 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Transcript of Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering...

Page 1: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Research Collection

Doctoral Thesis

Engineering delivery and release of protein therapeutics formammalian systems

Author(s): Fluri, David Andreas

Publication Date: 2008

Permanent Link: https://doi.org/10.3929/ethz-a-005708602

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

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

ETH Library

Page 2: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

DISS. ETH NO. 17862

Engineering delivery and release of protein therapeutics for mammalian systems

A dissertation submitted to ETH ZURICH

for the degree of

Doctor of Natural Sciences

Presented by

David Andreas Fluri

Dipl. Biotechnologe ESBS Strasbourg

born April 12, 1977

citizen of Niederdorf (BL)

Accepted on the recommendation of

Prof. Dr. Martin Fussenegger, examiner Prof. Dr. Sabine Werner, co-examiner

2008

Page 3: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Table of Contents

2

Table of Contents Summary

Zusammenfassung

General Introduction

Chapter 1 Transduction Technologies

Chapter 2 Adeno-associated viral vectors engineered for

macrolide-adjustable transgene expression in

mammalian cells and mice

Chapter 3 InXy and SeXy, compact heterologous reporter

proteins for mammalian cells

Chapter 4 A novel system for trigger-controlled drug

release from polymer capsules

Conclusion

Acknowledgements

Curriculum vitae

Page 4: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Summary

3

Summary

Page 5: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Summary

4

Summary

In recent years conventional therapeutic strategies using small molecule drugs

have been complemented by new approaches in the field of modern biotechnology.

Innovative therapeutic strategies include the delivery of genetic information that is

capable of replacing inherited genetic defects and the delivery of protein therapeutics

such as monoclonal antibodies or recombinant cytokines or cell- and tissue-

engineering approaches.

A major hurdle still to overcome in gene-therapy protocols is the efficient

delivery of genetic information to a targeted cell and the stringent control of

expression of the delivered genes. Currently viral systems are the most promising

tools for gene therapy and the most comprehensive pre-clinical and clinical data is

available for these systems.

The first chapter of this work summarizes the protocols and compares the

production and use of different viral transduction systems for mammalian cells.

The second chapter describes the construction of recombinant adeno-

associated viral vectors (rAAV), which are attracting more and more attention for

gene therapy applications, for the efficient and safe delivery of transgenes controllable

by a small molecule antibiotic. Vectors for the delivery of binary ON- and OFF-type,

auto-regulated, self-regulated and bi-directional systems were constructed and

functionally tested in various mammalian cell lines. Delivery and regulation of these

systems in vivo was confirmed by injecting the engineered rAAV vectors into mice.

Model reporter proteins simplify the analysis of new delivery, release and

control scenarios in vitro and in vivo. The third chapter of this work describes the

design and evaluation of a novel enzyme-based reporter system for mammalian cells.

Engineered bacteria-derived reporter proteins specifically designed for localization

either intracellularly or extracellularly were generated. Functional expression of these

constructs was confirmed in different cell lines and extensive analysis of their

biological and chemical characteristics was performed. These new proteins provide

an interesting alternative to currently available reporter systems for mammalian cells

due to their simple and sensitive assay formats that require no sample pre-treatment as

well as their compactness and high stability.

Page 6: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Summary

5

A major limitation of the use of biologics in therapeutic interventions is that

their structural complexity and fragility causes difficulties in safe and efficient

administration. Treatments using these factors would need either repeated pulsed

administrations or a continued release of the active substance over a prolonged period

of time. In the fourth chapter of this work a versatile delivery system based on

biocompatible polymer microcapsules is described. This system can be implanted and

allows for the trigger-induced release of encapsulated biologics upon an external cue.

A release system was designed where sensor cells within the capsules express, upon

induction by a chemical cue, an enzyme that degrades the polymer structure and leads

to the collapse of the particle. Co-encapsulated biologics can be released “on demand”

and by changing the sensor cell line a variety of different cues can be used to trigger

the burst.

Overall, this work presents new strategies to improve the delivery, monitoring,

expression-control and release of biologics utlizing engineered mammalian cells, viral

vectors and polymer capsules.

Page 7: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Summary

6

Zusammenfassung In den letzten Jahren wurden traditionelle Behandlungsstrategien mittels

"small molecule drugs" durch neue Ansätze aus dem Gebiet der modernen

Biotechnologie ergänzt. Innovative Strategien beinhalten unter anderem:

(1) Die Administration genetischer Information, welche in der Lage ist vererbte

Defekte zu komplementieren.

(2) Die Anwendung von auf Proteinen basierenden Therapeutika wie z.B.

monoklonale Antikörper oder rekombinant hergestellte Zytokine.

(3) Neue Ansätze in den Gebieten der Zell- und Gewebetherapie.

Ein grundlegendes Problem, welches es für gentherapeutische Anwendungen

zu überwinden gilt, ist neben der effizienten Einschleusung fremder, genetischer

Information in Zielzellen deren präzise Expressionskontrolle. Gegenwärtig sind virale

Syteme die vielversprechendsten Werkzeuge für gentherapeutische Anwendungen

und dementsprechend ist auch die präklinische und klinische Datenlage für diese

Systeme vergleichsweise umfangreich. Das erste Kapitel dieser Arbeit beinhaltet

Protokolle und vergleicht die Produktion und Anwendung von unterschiedlichen

viralen Transduktionssystemen für Säugerzellen.

Im zweiten Kapitel wird die Konstruktion von rekombinanten Adeno-

Assozierten viralen Vektoren (rAAV), welche für gentherapeutische Anwendungen

immer mehr in den Mittelpunkt rücken, beschrieben. Vektoren für die Applikation

von binären, auto-regulierten, selbst-regulierten und bidirektionalen Systemen, welche

durch das Antibiotikum Erythromycin reguliert werden können, wurden konstruiert

und ihre Funktionalität in verschiedenen Säugerzellinien getestet. Zusätzlich wurde

die kontrollierte Expression eines Glykoproteins nach Administration eines selbst-

regulierten rAAV in vivo gezeigt.

Reporterproteine vereinfachen die Analyse von neuen Administrations-,

Freisetzungs- und Kontrollsystemen in vitro und in vivo. Das dritte Kapitel befasst

sich mit dem Design und der Evaluation eines neuen, enzymbasierenden

Reportersystems für Säugerzellen. Es wurden von einem bakteriellen Protein

abgeleitete Reporterkonstrukte, speziell konzipiert für die intra- und extrazelluläre

Lokalisation, entwickelt. Die funktionelle Expression dieser Enzyme wurde in

Page 8: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Summary

7

verschiedenen Säugerzellinien gezeigt und die biologischen und chemischen

Eigenschaften wurden detailliert analysiert. Diese neuen Reporterproteine stellen,

aufgrund ihrer Kompaktheit, der hohen Stabilität, sowie ihrer einfachen und

sensitiven Analyseformate, welche keine Vorbehandlung der Proben erfordern, eine

Alternative zu gegenwärtig erhältlichen Reportersystemen dar.

Eine grundlegende Einschränkung bei der Anwendung von biologischen

Wirkstoffen für therapeutische Eingriffe ist ihre strukturelle Komplexität und

Fragilität, welche zu Problemen bei der sicheren und effizienten Applikation führen

kann. Behandlungen mit solchen Stoffen müssen entweder durch wiederholte

Administrationen oder unter kontinuierlicher Freisetzung des Wirkstoffs im Körper

über längere Zeit erfolgen. Im vierten Kapitel dieser Arbeit wird ein vielseitiges

Applikationssystem, basierend auf biokompatiblen Polymermikrokapseln,

beschrieben. Das System erlaubt nach Implantation die stimulusabhängige

Freisetzung von verkapselten biologischen Präparaten. Sensor Zellen in den Kapseln

exprimieren triggerabhängig ein Enzym, welches die Polymer-Struktur destabilisiert

und die Kapsel kollabieren lässt. Co-verkapselte biologische Wirkstoffe können somit

bei Bedarf freigesetzt werden. Durch den Austausch der Sensor Zellen können

verschiedenste Stimuli zur Kapseldestabilisierung verwendet werden.

Diese Arbeit stellt neue Strategien zur verbesserten Applikation,

Überwachung, Expressionskontrolle und Freisetzung von biologischen Präparaten mit

Hilfe von Säugerzellen, viralen Vektoren und Polymerkapseln vor.

Page 9: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

Introduction

Page 10: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

9

General Introduction Over the last 25 years, since human insulin became the first medicine to be

produced using recombinant technologies, modern biotechnology has become a

promising field for the development of new drugs and therapies. Recombinant protein

drugs such as insulin, interferon, growth hormones, recombinant vaccines and

therapeutic antibodies have made it to the market over the years as new biotech and

pharmaceutical companies expand their research and development investments in the

field (Lahteenmaki and Lawrence, 2007). Besides the production of therapeutic

proteins, mammalian biotechnology also provides new tools to cure diseases. Gene

and cell based therapies provide new strategies to fight diseases that so far have been

challenging to treat by existing therapies (Edelstein et al., 2007; Lillicrap et al., 2006;

Melo et al., 2004; Mimeault et al., 2007; Sueblinvong et al., 2007).

Gene therapy

In the gene therapy field, more than 1300 clinical trials have been completed

since 1989 when Rosenberg et al. conducted the first immunotherapy for patients with

advanced melanoma (Rosenberg et al., 1990). Although gene therapy is still regarded

as the ultimate cure for genetic diseases by attacking the source of the disease,

optimally foregoing the need for a life-long therapy of symptoms, severe set-backs in

the field have slowed or even stalled the development of new therapies (Hacein-Bey-

Abina et al., 2003; Raper et al., 2003). Extensive risk/benefit examinations have been

conducted and research in the field has been directed towards the understanding of

potential risks associated with gene therapy protocols (Check, 2003; Gansbacher,

2002). In spite of the setbacks associated with first clinical trials, the effort in bringing

gene therapy to a safe and efficient state where it can be used to treat severely ill

patients is still prevailing and recent work shows promising results (Morgan et al.,

2006; Ott et al., 2006).

The precise delivery of genes and dose-dependant expression of the

corresponding protein products are the determining criteria for successful gene

therapy applications. Viral vectors are considered the most promising vehicles to

efficiently deliver foreign genetic information to a targeted cell-nucleus (Edelstein et

al., 2007). Today, the most commonly used viral vector systems are based on

oncoretrovirus (Landau and Littman, 1992; Pear et al., 1993), lentivirus (Naldini et

al., 1996; Zufferey et al., 1997), adenovirus (Bett et al., 1994; Chartier et al., 1996),

Page 11: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

10

herpes simplex virus (Krisky et al., 1997) and adeno-associated virus (Xiao et al.,

1998). Each of these vectors have unique properties that make them more or less

suitable for a particular application, and all of these systems, except the lentiviral

vectors, have been evaluated in clinical trials (Edelstein et al., 2007) .

Vectors were engineered to exploit the highly efficient viral machinery to

deliver genetic information to cells. These viral vectors are able to infect a broad

range of different cell types but are incapable of causing a productive infection. After

major setbacks using retroviral vectors and adenoviral vectors in clinical trials, much

of the hope in the gene therapy community relies on recombinant adeno-associated

viral vectors (rAAV). rAAV has shown promising pre-clinical and clinical results

(Jiang et al., 2006; Kaplitt et al., 2007; Li et al., 2005; Wang et al., 2000) and has

superior safety characteristics among all currently used viral vector systems. rAAV

vectors integrate into the host chromosome at a significantly lower frequency than

retroviral or lentiviral vectors and more than 95% of transgene expression stems from

stable circular episomes in the target cell. This characteristic reduces the risk of

insertional mutagenesis and thereby decreases proto-oncogene activation

significantly. These characteristics combined with a high transduction efficiency for a

broad variety of tissues as well as the stability of rAAV particles makes this system a

premium choice for gene therapy applications. A significant shortcoming of rAAV

vectors is their low packaging capacity. The delivery of an entire genomic locus

including spacious endogenous regulatory sequences is beyond the capacity of

currently used rAAV vectors. Even the delivery of smaller promoter-transgene

cassettes encounters size constraints in certain applications. As a result, vector design

has become crucial for successful rAAV-based gene therapy. Different strategies have

been employed to optimize and compress the delivered cassettes or engineer the

vector system in such a way that larger amounts of genetic information can be

transferred (Duan et al., 2000; Urabe et al., 1997; Yan et al., 2000).

Particularly in settings where the expression of delivered transgenes has to be

regulated, a compact architecture is a prerequisite to encoding of all the elements of

the regulation system. Many gene regulation systems for mammalian cells are

available. These regulation systems respond to an array of varying stimuli and allow

the precise control of expression levels of delivered transgenes (Weber and

Fussenegger, 2006). Vector designs encoding compact control systems, mainly the

tetracycline-responsive TET system (Gossen and Bujard, 1992), have been

Page 12: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

11

extensively studied in oncoretroviral, lentiviral systems (Kafri et al., 2000; Koponen

et al., 2003; Regulier et al., 2002; Reiser et al., 2000; Unsinger et al., 2001), and to a

lesser extent in rAAV (Chtarto et al., 2003; Gafni et al., 2004; Jiang et al., 2004).

Although the building blocks for safe delivery and the subsequent regulation of

transgene expression are established, a long road of improved safety and efficiency

lays ahead before these gene therapy techniques can be utilized as efficient weapons

for the treatment of diseases.

Reporter Proteins

Reporter proteins are valuable tools for quick, precise and sensitive analysis of

gene expression in biological systems. Fluorescent proteins such as GFP and its

derivatives (Hadjantonakis et al., 2003), luciferases (de Wet et al., 1985; Lorenz et al.,

1991), various enzymatic reporters (An et al., 1982; Gorman et al., 1982; Tjuvajev et

al., 1995) or secreted reporter genes (Berger et al., 1988; Schlatter et al., 2002)

changed the landscape of many domains of modern biology; allowing the detection

and quantification of effects that have not been analyzed before. Many aspects of the

development of biologics have to be closely monitored in order to optimize the

design, production, delivery and expression of the factors. Reporter proteins provide

an approach to monitor these crucial steps and aid in the simplification and

streamlining of the development process. Reporter proteins are commonly used and

provide important information on the optimization of production processes (Reischer

et al., 2004), the engineering of producer cell lines (Choe et al., 2005; Sleiman et al.,

2008), the tracing of implanted cells (Rome et al., 2007), the quantification of

expression levels upon delivery (Bartoli et al., 2006; Wang et al., 2001) and the fate

of certain cells following implantation (Schiedlmeier et al., 2003).

One group of reporter proteins is secreted from cells through conventional

pathways and then analyzed extracellularly. These reporter proteins represent the final

product of a complex cascade of events including transcription, transcript processing,

mRNA export from the nucleus, translation, posttranslational modification and

protein export. Utilizing secreted reporter proteins allows one to measure these

parameters and therefore they are a powerful tool for applications such as metabolic

engineering (Choi et al., 2006; Park et al., 2005), gene regulation (Weber et al., 2002),

synthetic biology (Kramer and Fussenegger, 2005; Weber et al., 2007), gene therapy

(Gronevik et al., 2005; Lamartina et al., 2003) or glycosylation monitoring (Lipscomb

et al., 2005).

Page 13: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

12

A limitation of certain secreted reporter proteins is the difficulty to distinguish

the activity of the engineered factor from signals arising from cognate endogenous

proteins. This often leads to lower signal to background ratios and thus reduces the

sensitivity of the assay. For some reporter systems, such as human placental secreted

alkaline phosphatase (SEAP), these ratios can be improved by inactivating

endogenous signals by heat treatment or specific inhibitors (Berger et al., 1988;

Schlatter et al., 2002). Other reporter protein, such as human growth hormone or

erythropoietin, have the intrinsic limitation that direct activity assays are nonexistent

and expression levels have to be quantified via labor intensive methods, such as an

ELISA (Muramatsu et al., 2001; Selden et al., 1986). For some applications gene size

restraints exist, such as for viral delivery or multi-cistronic expression systems, so the

length of the coding region of the reporter protein becomes crucial. To overcome

these limitations compact systems have been developed to free space for other

elements. The development of new secreted reporter proteins that are heterologous,

compact, sensitive, stable, high-throughput analysis capable and compatible to

existing reporter platforms represent a useful complement to existing systems.

Drug delivery

Apart from gene therapy, other fields of modern biotechnology have shown

promising results for the treatment of various severe diseases such as cancer, multiple

sclerosis, Alzheimer, Parkinson disease and autoimmune diseases. Protein

therapeutics employing monoclonal antibodies, hormones or cell-based platforms

have shown promising pre-clinical and clinical results (Dove, 2002; Leader et al.,

2008).

Due to their chemical and biological properties, protein and cell therapeutics

have to be administered by injection or released from special implanted devices.

Although repeated injection is currently, and will likely be in the future, the major

route of administration of biologics due to its convenience for many treatments other

delivery modes would be more advantageous in certain applications. The annual

growth in the field of drug delivery systems has been significant and an increasing

number of companies enter the market with innovative strategies for drug delivery

(Orive et al., 2003c). Systems for oral (Veronese and Harris, 2002), transdermal

(Langer, 2003) and transmucosal drug delivery (Illum et al., 2001) have been

developed over the last few years. In addition to these advancements in administration

and dosing of therapeutics, another promising approach is the immunoisolation and in

Page 14: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

13

situ production of biologics. Cells expressing biologics are isolated from the immune

system by polymeric membranes that enable the free transfer of nutrients and waste

products. These membranes also prevent the access of the different players of the

immune system to the contents of the capsule, be they allogeneic or xenogeneic cells

(Orive et al., 2004). Various promising research initiatives have been initiated

recently using encapsulation protocols (Orive et al., 2003a). Polymer capsules have

been used to encapsulate different types of biologics including protein therapeutics

(Blanco-Prieto et al., 2000; Han et al., 2001), vaccines (Rosas et al., 2002; Sturesson

et al., 1999), liposomes (Dhoot and Wheatley, 2003; Kibat et al., 1990; Lee et al.,

2003), DNA (Alexakis et al., 1995; Lengsfeld et al., 2002; Zhang et al., 2004), viral

vectors (Sailaja et al., 2002) and whole cells/tissues (Luca et al., 2007; Read et al.,

2001; Springer et al., 2000; Wang et al., 1997).

There are various modes of release for therapeutics depending on needs of the

system, these include: (i) continuous release over time (Hasse et al., 1997; Rinsch et

al., 2002), (ii) initial burst followed by decreasing release over time (Chretien and

Chaumeil, 2005; Pelegrin et al., 1998; Wang et al., 1997), (iii) pulsed release patterns

(Bussemer et al., 2003; De Geest et al., 2005) and (iv) inducible release triggered by

internal and external cues (Angelatos et al., 2005; Fischel-Ghodsian et al., 1988;

Skirtach et al., 2004). In particular the rupture of polymercapsules, and release of

their contents, “on-demand” represents an appealing approach to deliver therapeutic

molecules when they are needed in the body. Different approaches have been

investigated for the release of therapeutics on demand comprising inducible capsule

rupture and release of encapsulated cargo by triggers such as an electric field (Kiser et

al., 1998), IR light (Angelatos et al., 2005), a magnetic field (Lu et al., 2005), a pH

change (Dejugnat and Sukhorukov, 2004), swelling or osmotic bursting of the capsule

(De Geest et al., 2005; Iskakov et al., 2002), enzymatic degradation of the polymer

capsule (De Geest et al., 2006; Itoh et al., 2006) or destabilization of the capsule

induced by changes in salt and redox states (Haynie et al., 2005; Schuler and Caruso,

2001). Although significant progress in the field of delivery of biologic drugs has

been made there still remain numerous obstacles to overcome. These obstacles

include the mechanical and chemical stability of capsules, biosafety, long-term

survival of capsules, biocompatibility and long-term functionality (Orive et al.,

2003b).

Page 15: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

14

Due to the complexity of gene- and cell-therapies and protein therapeutics,

efficient administration, dosage and formulation is much more complex than for

traditional small molecule drugs. To harness the potential of these new classes of

therapeutics novel methods of biologic delivery have to be developed and existing

technologies have to be improved and adapted.

Contributions of this work Chapter 1: Transduction Technologies

This chapter presents basic methodologies for the production and use of the

most commonly utilized virus-based vectors for the delivery of genetic information to

mammalian cells. Optimized production protocols for viral delivery vectors namely,

oncoretrovirus, lentivirus, adenovirus and adeno-associated virus are provided. The

intention was to provide a thorough groundwork from where one can choose an

optimal transduction system for a given application. Each viral system has its inherent

advantages and disadvantages. The choice of a virus will depend on the required

characteristics such as the ability to integrate genetic material into the host genome,

the tropism, the packaging capacity, safety profiles, immunogenicity and available

production and concentration protocols.

The optimized protocols allow for the production of both crude and highly

pure vector preparations suitable for in vitro and in vivo use. The last section of the

chapter consists of a case study partly derived from chapter 2 of this work describing

the application of rAAV vectors in the transduction and regulated expression of

secreted and intracellular transgenes in mammalian cells.

The chapter is written in a protocol-based style as it has been published in a

methodology book for the establishment of cell lines for biotechnology applications.

Chapter 2: Adeno-associated viral vectors engineered for macrolide-adjustable

transgene expression in mammalian cells and mice

We have combined the efficiency and safety of recombinant adeno-associated

viral vectors of serotype 2 (rAAV2) with the effectiveness of a gene regulation system

(Weber et al., 2002) that responds to a clinically licensed antibiotic in order to deliver

and control transgenes in vitro as well as in vivo.

Page 16: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

15

rAAV vectors were constructed for binary ON-type and OFF-type systems

expressing either intracellular or secreted reporter protein under the control of

erythromycin responsive promoters. These engineered rAAV vectors were able to

regulate reporter expression in an array of different mammalian cell lines. Binary

systems have the advantage that relatively large transgenes can be delivered, which is

crucial for viral vector system having a small packaging capacity such as rAAV. A

considerable disadvantage of binary systems is the requirement that both parts of the

system i.e. both vectors have to infect the same cell in order to obtain functional

regulatable expression. To circumvent this we designed compact single vectors that

were auto-regulated, self-regulated and bi-directional which harbored all of the

necessary elements for functional regulation together with the gene of interest (GOI).

Although good induction factors were obtained for all three types of one-vector

systems, only the self-regulated rAAV2 vector was able to obtain high transduction

rates. Using the self-regulated one-vector rAAV2 system mice were transduced and

regulatable expression of a secreted reporter gene was observed in vivo.

Chapter 3: InXy and SeXy, compact heterologous reporter proteins for

mammalian cells

Intracellular (InXy) and secreted (SeXy) bacterial xylanase-derived reporter

proteins were created allowing for a quick and sensitive quantification of activity in

the supernatant and cell pellet of mammalian cells. InXy and SeXy are entirely

heterologuous proteins in mammalian cells and direct measurement of their activity

from cell culture supernatants or lysed cell pellets without pretreatment is possible.

Both reporters are robust and maintain activity after incubation at a broad range of

temperatures, pH conditions and after long term incubation in complex biological

mixtures such as human serum. InXy and SeXy reporters are compatible with other

commonly used reporter proteins thus permitting parallel analysis. SeXy is

extensively glycosylated when secreted from various mammalian cell lines and could

be utilized to monitor biopharmaceutical processes since most of the currently

produced biopharmaceuticals are glycoproteins. In vivo SeXy was shown to circulate

in the blood stream of mice after being infected with rAAV particles hosting a SeXy

expression cassette.

Page 17: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

16

Chapter 4: A novel versatile system for stimulus-controlled drug release from

polymer capsules

The tailored degradation of polymer capsules, upon the action of certain cues,

is a promising approach to design delivery scenarios for stimulus-dependent release of

therapeutic molecules.

A new, flexible system was created that was able to respond to selectable

triggers either applied from the outside, such as antibiotics, hormones, amino acids,

biotin, or directly linked to endogenous stimuli, such as oxygen levels or quorum-

sensing molecules at the site of infection.

Clinically tested cellulose sulfate (CS)/poly-dimethyl-diallylammonium

chloride (pDADMAC) encapsulation technology (Merten et al., 1991) was combined

with the controlled inducible expression of a bacteria-derived cellulase that

destabilizes the capsule resulting in the release of its therapeutic cargo. The

functionality of the modified secreted cellulase (SecCell) was tested in various

mammalian cell types and a stable cell line was created, using HEK293-T, which

expressed SecCell under the control of either a doxycycline- or an erythromycin-

responsive promoter. The molecular weight cut-off of the capsule system was

determined to be around 40 kDa which is in an attractive range for the delivery of

biologics. The trigger-induced release of the model glycoprotein SEAP was illustrated

by co-encapsulating a sensor cell population, which expresses SecCell that is

repressed by the presence of doxycycline, together with a cell line that constitutively

produces SEAP. In the presence of doxyclcline SEAP was observed to be exclusively

accumulating inside the capsule. In the absence of this molecule SecCell was

expressed by the sensor cells, disrupted the capsule and SEAP was released into the

supernatant. Micrographs and time-lapse images reveal a collapse of the capsule

structure upon SecCell expression. Release characteristics could be modulated by

varying the time of induction, the number of sensor cells encapsulated or by titrating

SecCell induction levels with different concentrations of inducer. It was also shown

that the capsules were exclusively ruptured when SecCell was expressed internally.

This characteristic of the capsules allows for the co-administration of varying capsule-

populations containing different sensor cells responsive to changing stimuli. One can

imagine applications where multiple capsule-populations containing specific biologics

co-encapsulated with different stimuli-responsive sensor cells could be administered

Page 18: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

17

at once. Then these different biologics could be released at desired points in time

according to the presence of different stimuli.

Moreover, it was shown by injecting capsules containing both a model

glycoprotein and doxycycline-responsive sensor cells into mice that an in vivo

inducible release of the glycoprotein into the bloodstream is possible.

Page 19: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

18

References

Alexakis, T., Boadi, D.K., Quong, D., Groboillot, A., O'Neill, I., Poncelet, D., and

Neufeld, R.J. (1995). Microencapsulation of DNA within alginate microspheres and

crosslinked chitosan membranes for in vivo application. Applied biochemistry and

biotechnology 50, 93-106.

An, G., Hidaka, K., and Siminovitch, L. (1982). Expression of bacterial beta-

galactosidase in animal cells. Molecular and cellular biology 2, 1628-1632.

Angelatos, A.S., Radt, B., and Caruso, F. (2005). Light-responsive

polyelectrolyte/gold nanoparticle microcapsules. The journal of physical chemistry

109, 3071-3076.

Bartoli, M., Poupiot, J., Goyenvalle, A., Perez, N., Garcia, L., Danos, O., and Richard,

I. (2006). Noninvasive monitoring of therapeutic gene transfer in animal models of

muscular dystrophies. Gene therapy 13, 20-28.

Berger, J., Hauber, J., Hauber, R., Geiger, R., and Cullen, B.R. (1988). Secreted

placental alkaline phosphatase: a powerful new quantitative indicator of gene

expression in eukaryotic cells. Gene 66, 1-10.

Bett, A.J., Haddara, W., Prevec, L., and Graham, F.L. (1994). An efficient and

flexible system for construction of adenovirus vectors with insertions or deletions in

early regions 1 and 3. Proceedings of the National Academy of Sciences of the United

States of America 91, 8802-8806.

Blanco-Prieto, M.J., Besseghir, K., Zerbe, O., Andris, D., Orsolini, P., Heimgartner,

F., Merkle, H.P., and Gander, B. (2000). In vitro and in vivo evaluation of a

somatostatin analogue released from PLGA microspheres. J Control Release 67, 19-

28.

Bussemer, T., Dashevsky, A., and Bodmeier, R. (2003). A pulsatile drug delivery

system based on rupturable coated hard gelatin capsules. J Control Release 93, 331-

339.

Chartier, C., Degryse, E., Gantzer, M., Dieterle, A., Pavirani, A., and Mehtali, M.

(1996). Efficient generation of recombinant adenovirus vectors by homologous

recombination in Escherichia coli. Journal of virology 70, 4805-4810.

Check, E. (2003). Harmful potential of viral vectors fuels doubts over gene therapy.

Nature 423, 573-574.

Page 20: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

19

Choe, J., Guo, H.H., and van den Engh, G. (2005). A dual-fluorescence reporter

system for high-throughput clone characterization and selection by cell sorting.

Nucleic acids research 33, e49.

Choi, S.S., Rhee, W.J., Kim, E.J., and Park, T.H. (2006). Enhancement of

recombinant protein production in Chinese hamster ovary cells through anti-apoptosis

engineering using 30Kc6 gene. Biotechnology and bioengineering 95, 459-467.

Chretien, C., and Chaumeil, J.C. (2005). Release of a macromolecular drug from

alginate-impregnated microspheres. International journal of pharmaceutics 304, 18-

28.

Chtarto, A., Bender, H.U., Hanemann, C.O., Kemp, T., Lehtonen, E., Levivier, M.,

Brotchi, J., Velu, T., and Tenenbaum, L. (2003). Tetracycline-inducible transgene

expression mediated by a single AAV vector. Gene therapy 10, 84-94.

De Geest, B.G., Dejugnat, C., Sukhorukov, G.B., Braeckmans, K., De Smedt, S.C.,

and Demeester, J. (2005). Self-rupturing microcapsules. Advanced Materials 17,

2357-+.

De Geest, B.G., Vandenbroucke, R.E., Guenther, A.M., Sukhorukov, G.B., Hennink,

W.E., Sanders, N.N., Demeester, J., and De Smedt, S.C. (2006). Intracellularly

degradable polyelectrolyte microcapsules. Advanced Materials 18, 1005-+.

de Wet, J.R., Wood, K.V., Helinski, D.R., and DeLuca, M. (1985). Cloning of firefly

luciferase cDNA and the expression of active luciferase in Escherichia coli.

Proceedings of the National Academy of Sciences of the United States of America 82,

7870-7873.

Dejugnat, C., and Sukhorukov, G.B. (2004). pH-responsive properties of hollow

polyelectrolyte microcapsules templated on various cores. Langmuir 20, 7265-7269.

Dhoot, N.O., and Wheatley, M.A. (2003). Microencapsulated liposomes in controlled

drug delivery: strategies to modulate drug release and eliminate the burst effect.

Journal of pharmaceutical sciences 92, 679-689.

Dove, A. (2002). Cell-based therapies go live. Nature biotechnology 20, 339-343.

Duan, D., Yue, Y., Yan, Z., and Engelhardt, J.F. (2000). A new dual-vector approach

to enhance recombinant adeno-associated virus-mediated gene expression through

intermolecular cis activation. Nature medicine 6, 595-598.

Edelstein, M.L., Abedi, M.R., and Wixon, J. (2007). Gene therapy clinical trials

worldwide to 2007--an update. The journal of gene medicine 9, 833-842.

Page 21: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

20

Fischel-Ghodsian, F., Brown, L., Mathiowitz, E., Brandenburg, D., and Langer, R.

(1988). Enzymatically controlled drug delivery. Proceedings of the National Academy

of Sciences of the United States of America 85, 2403-2406.

Gafni, Y., Pelled, G., Zilberman, Y., Turgeman, G., Apparailly, F., Yotvat, H., Galun,

E., Gazit, Z., Jorgensen, C., and Gazit, D. (2004). Gene therapy platform for bone

regeneration using an exogenously regulated, AAV-2-based gene expression system.

Mol Ther 9, 587-595.

Gansbacher, B. (2002). Policy statement on the social, ethical and public awareness

issues in gene therapy. The journal of gene medicine 4, 687-691.

Gorman, C.M., Moffat, L.F., and Howard, B.H. (1982). Recombinant genomes which

express chloramphenicol acetyltransferase in mammalian cells. Molecular and cellular

biology 2, 1044-1051.

Gossen, M., and Bujard, H. (1992). Tight control of gene expression in mammalian

cells by tetracycline-responsive promoters. Proceedings of the National Academy of

Sciences of the United States of America 89, 5547-5551.

Gronevik, E., von Steyern, F.V., Kalhovde, J.M., Tjelle, T.E., and Mathiesen, I.

(2005). Gene expression and immune response kinetics using electroporation-

mediated DNA delivery to muscle. The journal of gene medicine 7, 218-227.

Hacein-Bey-Abina, S., von Kalle, C., Schmidt, M., Le Deist, F., Wulffraat, N.,

McIntyre, E., Radford, I., Villeval, J.L., Fraser, C.C., Cavazzana-Calvo, M., et al.

(2003). A serious adverse event after successful gene therapy for X-linked severe

combined immunodeficiency. The New England journal of medicine 348, 255-256.

Hadjantonakis, A.K., Dickinson, M.E., Fraser, S.E., and Papaioannou, V.E. (2003).

Technicolour transgenics: imaging tools for functional genomics in the mouse. Nature

reviews 4, 613-625.

Han, K., Lee, K.D., Gao, Z.G., and Park, J.S. (2001). Preparation and evaluation of

poly(L-lactic acid) microspheres containing rhEGF for chronic gastric ulcer healing. J

Control Release 75, 259-269.

Hasse, C., Klock, G., Schlosser, A., Zimmermann, U., and Rothmund, M. (1997).

Parathyroid allotransplantation without immunosuppression. Lancet 350, 1296-1297.

Haynie, D.T., Palath, N., Liu, Y., Li, B., and Pargaonkar, N. (2005). Biomimetic

nanostructured materials: inherent reversible stabilization of polypeptide

microcapsules. Langmuir 21, 1136-1138.

Page 22: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

21

Illum, L., Jabbal-Gill, I., Hinchcliffe, M., Fisher, A.N., and Davis, S.S. (2001).

Chitosan as a novel nasal delivery system for vaccines. Advanced drug delivery

reviews 51, 81-96.

Iskakov, R.M., Kikuchi, A., and Okano, T. (2002). Time-programmed pulsatile

release of dextran from calcium-alginate gel beads coated with carboxy-n-

propylacrylamide copolymers. J Control Release 80, 57-68.

Itoh, Y., Matsusaki, M., Kida, T., and Akashi, M. (2006). Enzyme-responsive release

of encapsulated proteins from biodegradable hollow capsules. Biomacromolecules 7,

2715-2718.

Jiang, H., Pierce, G.F., Ozelo, M.C., de Paula, E.V., Vargas, J.A., Smith, P., Sommer,

J., Luk, A., Manno, C.S., High, K.A., et al. (2006). Evidence of multiyear factor IX

expression by AAV-mediated gene transfer to skeletal muscle in an individual with

severe hemophilia B. Mol Ther 14, 452-455.

Jiang, L., Rampalli, S., George, D., Press, C., Bremer, E.G., O'Gorman, M.R., and

Bohn, M.C. (2004). Tight regulation from a single tet-off rAAV vector as

demonstrated by flow cytometry and quantitative, real-time PCR. Gene therapy 11,

1057-1067.

Kafri, T., van Praag, H., Gage, F.H., and Verma, I.M. (2000). Lentiviral vectors:

regulated gene expression. Mol Ther 1, 516-521.

Kaplitt, M.G., Feigin, A., Tang, C., Fitzsimons, H.L., Mattis, P., Lawlor, P.A., Bland,

R.J., Young, D., Strybing, K., Eidelberg, D., et al. (2007). Safety and tolerability of

gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's

disease: an open label, phase I trial. Lancet 369, 2097-2105.

Kibat, P.G., Igari, Y., Wheatley, M.A., Eisen, H.N., and Langer, R. (1990).

Enzymatically activated microencapsulated liposomes can provide pulsatile drug

release. Faseb J 4, 2533-2539.

Kiser, P.F., Wilson, G., and Needham, D. (1998). A synthetic mimic of the secretory

granule for drug delivery. Nature 394, 459-462.

Koponen, J.K., Kankkonen, H., Kannasto, J., Wirth, T., Hillen, W., Bujard, H., and

Yla-Herttuala, S. (2003). Doxycycline-regulated lentiviral vector system with a novel

reverse transactivator rtTA2S-M2 shows a tight control of gene expression in vitro

and in vivo. Gene therapy 10, 459-466.

Page 23: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

22

Kramer, B.P., and Fussenegger, M. (2005). Hysteresis in a synthetic mammalian gene

network. Proceedings of the National Academy of Sciences of the United States of

America 102, 9517-9522.

Krisky, D.M., Marconi, P.C., Oligino, T., Rouse, R.J., Fink, D.J., and Glorioso, J.C.

(1997). Rapid method for construction of recombinant HSV gene transfer vectors.

Gene therapy 4, 1120-1125.

Lahteenmaki, R., and Lawrence, S. (2007). Public biotech 2006 - the numbers. Nature

biotechnology 25, 729-737.

Lamartina, S., Silvi, L., Roscilli, G., Casimiro, D., Simon, A.J., Davies, M.E., Shiver,

J.W., Rinaudo, C.D., Zampaglione, I., Fattori, E., et al. (2003). Construction of an

rtTA2(s)-m2/tts(kid)-based transcription regulatory switch that displays no basal

activity, good inducibility, and high responsiveness to doxycycline in mice and non-

human primates. Mol Ther 7, 271-280.

Landau, N.R., and Littman, D.R. (1992). Packaging system for rapid production of

murine leukemia virus vectors with variable tropism. Journal of virology 66, 5110-

5113.

Langer, R. (2003). Where a pill won't reach. Scientific American 288, 50-57.

Leader, B., Baca, Q.J., and Golan, D.E. (2008). Protein therapeutics: a summary and

pharmacological classification. Nat Rev Drug Discov 7, 21-39.

Lee, K.E., Cho, S.H., Lee, H.B., Jeong, S.Y., and Yuk, S.H. (2003).

Microencapsulation of lipid nanoparticles containing lipophilic drug. Journal of

microencapsulation 20, 489-496.

Lengsfeld, C.S., Manning, M.C., and Randolph, T.W. (2002). Encapsulating DNA

within biodegradable polymeric microparticles. Current pharmaceutical

biotechnology 3, 227-235.

Li, C., Bowles, D.E., van Dyke, T., and Samulski, R.J. (2005). Adeno-associated virus

vectors: potential applications for cancer gene therapy. Cancer gene therapy 12, 913-

925.

Lillicrap, D., VandenDriessche, T., and High, K. (2006). Cellular and genetic

therapies for haemophilia. Haemophilia 12 Suppl 3, 36-41.

Lipscomb, M.L., Palomares, L.A., Hernandez, V., Ramirez, O.T., and Kompala, D.S.

(2005). Effect of production method and gene amplification on the glycosylation

pattern of a secreted reporter protein in CHO cells. Biotechnology progress 21, 40-49.

Page 24: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

23

Lorenz, W.W., McCann, R.O., Longiaru, M., and Cormier, M.J. (1991). Isolation and

expression of a cDNA encoding Renilla reniformis luciferase. Proceedings of the

National Academy of Sciences of the United States of America 88, 4438-4442.

Lu, Z., Prouty, M.D., Guo, Z., Golub, V.O., Kumar, C.S., and Lvov, Y.M. (2005).

Magnetic switch of permeability for polyelectrolyte microcapsules embedded with

Co@Au nanoparticles. Langmuir 21, 2042-2050.

Luca, G., Calvitti, M., Nastruzzi, C., Bilancetti, L., Becchetti, E., Angeletti, G.,

Mancuso, F., and Calafiore, R. (2007). Encapsulation, in vitro characterization, and in

vivo biocompatibility of Sertoli cells in alginate-based microcapsules. Tissue

engineering 13, 641-648.

Melo, L.G., Pachori, A.S., Kong, D., Gnecchi, M., Wang, K., Pratt, R.E., and Dzau,

V.J. (2004). Gene and cell-based therapies for heart disease. Faseb J 18, 648-663.

Merten, O.W., Dautzenberg, H., and Palfi, G.E. (1991). A new method for the

encapsulation of mammalian cells. Cytotechnology 7, 121-130.

Mimeault, M., Hauke, R., and Batra, S.K. (2007). Stem cells: a revolution in

therapeutics-recent advances in stem cell biology and their therapeutic applications in

regenerative medicine and cancer therapies. Clinical pharmacology and therapeutics

82, 252-264.

Morgan, R.A., Dudley, M.E., Wunderlich, J.R., Hughes, M.S., Yang, J.C., Sherry,

R.M., Royal, R.E., Topalian, S.L., Kammula, U.S., Restifo, N.P., et al. (2006). Cancer

regression in patients after transfer of genetically engineered lymphocytes. Science

(New York, NY 314, 126-129.

Muramatsu, T., Arakawa, S., Fukazawa, K., Fujiwara, Y., Yoshida, T., Sasaki, R.,

Masuda, S., and Park, H.M. (2001). In vivo gene electroporation in skeletal muscle

with special reference to the duration of gene expression. International journal of

molecular medicine 7, 37-42.

Naldini, L., Blomer, U., Gallay, P., Ory, D., Mulligan, R., Gage, F.H., Verma, I.M.,

and Trono, D. (1996). In vivo gene delivery and stable transduction of nondividing

cells by a lentiviral vector. Science (New York, NY 272, 263-267.

Orive, G., Gascon, A.R., Hernandez, R.M., Igartua, M., and Luis Pedraz, J. (2003a).

Cell microencapsulation technology for biomedical purposes: novel insights and

challenges. Trends in pharmacological sciences 24, 207-210.

Page 25: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

24

Orive, G., Hernandez, R.M., Gascon, A.R., Calafiore, R., Chang, T.M., De Vos, P.,

Hortelano, G., Hunkeler, D., Lacik, I., Shapiro, A.M., et al. (2003b). Cell

encapsulation: promise and progress. Nature medicine 9, 104-107.

Orive, G., Hernandez, R.M., Rodriguez Gascon, A., Calafiore, R., Chang, T.M., de

Vos, P., Hortelano, G., Hunkeler, D., Lacik, I., and Pedraz, J.L. (2004). History,

challenges and perspectives of cell microencapsulation. Trends in biotechnology 22,

87-92.

Orive, G., Hernandez, R.M., Rodriguez Gascon, A., Dominguez-Gil, A., and Pedraz,

J.L. (2003c). Drug delivery in biotechnology: present and future. Current opinion in

biotechnology 14, 659-664.

Ott, M.G., Schmidt, M., Schwarzwaelder, K., Stein, S., Siler, U., Koehl, U., Glimm,

H., Kuhlcke, K., Schilz, A., Kunkel, H., et al. (2006). Correction of X-linked chronic

granulomatous disease by gene therapy, augmented by insertional activation of

MDS1-EVI1, PRDM16 or SETBP1. Nature medicine 12, 401-409.

Park, K.S., Seol, W., Yang, H.Y., Lee, S.I., Kim, S.K., Kwon, R.J., Kim, E.J., Roh,

Y.H., Seong, B.L., and Kim, J.S. (2005). Identification and use of zinc finger

transcription factors that increase production of recombinant proteins in yeast and

mammalian cells. Biotechnology progress 21, 664-670.

Pear, W.S., Nolan, G.P., Scott, M.L., and Baltimore, D. (1993). Production of high-

titer helper-free retroviruses by transient transfection. Proceedings of the National

Academy of Sciences of the United States of America 90, 8392-8396.

Pelegrin, M., Marin, M., Noel, D., Del Rio, M., Saller, R., Stange, J., Mitzner, S.,

Gunzburg, W.H., and Piechaczyk, M. (1998). Systemic long-term delivery of

antibodies in immunocompetent animals using cellulose sulphate capsules containing

antibody-producing cells. Gene therapy 5, 828-834.

Raper, S.E., Chirmule, N., Lee, F.S., Wivel, N.A., Bagg, A., Gao, G.P., Wilson, J.M.,

and Batshaw, M.L. (2003). Fatal systemic inflammatory response syndrome in a

ornithine transcarbamylase deficient patient following adenoviral gene transfer.

Molecular genetics and metabolism 80, 148-158.

Read, T.A., Sorensen, D.R., Mahesparan, R., Enger, P.O., Timpl, R., Olsen, B.R.,

Hjelstuen, M.H., Haraldseth, O., and Bjerkvig, R. (2001). Local endostatin treatment

of gliomas administered by microencapsulated producer cells. Nature biotechnology

19, 29-34.

Page 26: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

25

Regulier, E., Pereira de Almeida, L., Sommer, B., Aebischer, P., and Deglon, N.

(2002). Dose-dependent neuroprotective effect of ciliary neurotrophic factor delivered

via tetracycline-regulated lentiviral vectors in the quinolinic acid rat model of

Huntington's disease. Human gene therapy 13, 1981-1990.

Reischer, H., Schotola, I., Striedner, G., Potschacher, F., and Bayer, K. (2004).

Evaluation of the GFP signal and its aptitude for novel on-line monitoring strategies

of recombinant fermentation processes. Journal of biotechnology 108, 115-125.

Reiser, J., Lai, Z., Zhang, X.Y., and Brady, R.O. (2000). Development of multigene

and regulated lentivirus vectors. Journal of virology 74, 10589-10599.

Rinsch, C., Dupraz, P., Schneider, B.L., Deglon, N., Maxwell, P.H., Ratcliffe, P.J.,

and Aebischer, P. (2002). Delivery of erythropoietin by encapsulated myoblasts in a

genetic model of severe anemia. Kidney international 62, 1395-1401.

Rome, C., Couillaud, F., and Moonen, C.T. (2007). Gene expression and gene therapy

imaging. European radiology 17, 305-319.

Rosas, J.E., Pedraz, J.L., Hernandez, R.M., Gascon, A.R., Igartua, M., Guzman, F.,

Rodriguez, R., Cortes, J., and Patarroyo, M.E. (2002). Remarkably high antibody

levels and protection against P. falciparum malaria in Aotus monkeys after a single

immunisation of SPf66 encapsulated in PLGA microspheres. Vaccine 20, 1707-1710.

Rosenberg, S.A., Aebersold, P., Cornetta, K., Kasid, A., Morgan, R.A., Moen, R.,

Karson, E.M., Lotze, M.T., Yang, J.C., Topalian, S.L., et al. (1990). Gene transfer

into humans--immunotherapy of patients with advanced melanoma, using tumor-

infiltrating lymphocytes modified by retroviral gene transduction. The New England

journal of medicine 323, 570-578.

Sailaja, G., HogenEsch, H., North, A., Hays, J., and Mittal, S.K. (2002).

Encapsulation of recombinant adenovirus into alginate microspheres circumvents

vector-specific immune response. Gene therapy 9, 1722-1729.

Schiedlmeier, B., Klump, H., Will, E., Arman-Kalcek, G., Li, Z., Wang, Z., Rimek,

A., Friel, J., Baum, C., and Ostertag, W. (2003). High-level ectopic HOXB4

expression confers a profound in vivo competitive growth advantage on human cord

blood CD34+ cells, but impairs lymphomyeloid differentiation. Blood 101, 1759-

1768.

Schlatter, S., Rimann, M., Kelm, J., and Fussenegger, M. (2002). SAMY, a novel

mammalian reporter gene derived from Bacillus stearothermophilus alpha-amylase.

Gene 282, 19-31.

Page 27: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

26

Schuler, C., and Caruso, F. (2001). Decomposable hollow biopolymer-based capsules.

Biomacromolecules 2, 921-926.

Selden, R.F., Howie, K.B., Rowe, M.E., Goodman, H.M., and Moore, D.D. (1986).

Human growth hormone as a reporter gene in regulation studies employing transient

gene expression. Molecular and cellular biology 6, 3173-3179.

Skirtach, A.G., Antipov, A.A., Shchukin, D.G., and Sukhorukov, G.B. (2004).

Remote activation of capsules containing Ag nanoparticles and IR dye by laser light.

Langmuir 20, 6988-6992.

Sleiman, R.J., Gray, P.P., McCall, M.N., Codamo, J., and Sunstrom, N.A. (2008).

Accelerated cell line development using two-color fluorescence activated cell sorting

to select highly expressing antibody-producing clones. Biotechnology and

bioengineering 99, 578-587.

Springer, M.L., Hortelano, G., Bouley, D.M., Wong, J., Kraft, P.E., and Blau, H.M.

(2000). Induction of angiogenesis by implantation of encapsulated primary myoblasts

expressing vascular endothelial growth factor. The journal of gene medicine 2, 279-

288.

Sturesson, C., Artursson, P., Ghaderi, R., Johansen, K., Mirazimi, A., Uhnoo, I.,

Svensson, L., Albertsson, A.C., and Carlfors, J. (1999). Encapsulation of rotavirus

into poly(lactide-co-glycolide) microspheres. J Control Release 59, 377-389.

Sueblinvong, V., Suratt, B.T., and Weiss, D.J. (2007). Novel therapies for the

treatment of cystic fibrosis: new developments in gene and stem cell therapy. Clinics

in chest medicine 28, 361-379.

Tjuvajev, J.G., Stockhammer, G., Desai, R., Uehara, H., Watanabe, K., Gansbacher,

B., and Blasberg, R.G. (1995). Imaging the expression of transfected genes in vivo.

Cancer research 55, 6126-6132.

Unsinger, J., Kroger, A., Hauser, H., and Wirth, D. (2001). Retroviral vectors for the

transduction of autoregulated, bidirectional expression cassettes. Mol Ther 4, 484-

489.

Urabe, M., Hasumi, Y., Ogasawara, Y., Matsushita, T., Kamoshita, N., Nomoto, A.,

Colosi, P., Kurtzman, G.J., Tobita, K., and Ozawa, K. (1997). A novel dicistronic

AAV vector using a short IRES segment derived from hepatitis C virus genome. Gene

200, 157-162.

Veronese, F.M., and Harris, J.M. (2002). Introduction and overview of peptide and

protein pegylation. Advanced drug delivery reviews 54, 453-456.

Page 28: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Introduction

27

Wang, B., Li, J., and Xiao, X. (2000). Adeno-associated virus vector carrying human

minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse

model. Proceedings of the National Academy of Sciences of the United States of

America 97, 13714-13719.

Wang, M., Orsini, C., Casanova, D., Millan, J.L., Mahfoudi, A., and Thuillier, V.

(2001). MUSEAP, a novel reporter gene for the study of long-term gene expression in

immunocompetent mice. Gene 279, 99-108.

Wang, T., Lacik, I., Brissova, M., Anilkumar, A.V., Prokop, A., Hunkeler, D., Green,

R., Shahrokhi, K., and Powers, A.C. (1997). An encapsulation system for the

immunoisolation of pancreatic islets. Nature biotechnology 15, 358-362.

Weber, W., Daoud-El Baba, M., and Fussenegger, M. (2007). Synthetic ecosystems

based on airborne inter- and intrakingdom communication. Proceedings of the

National Academy of Sciences of the United States of America 104, 10435-10440.

Weber, W., and Fussenegger, M. (2006). Pharmacologic transgene control systems for

gene therapy. The journal of gene medicine 8, 535-556.

Weber, W., Fux, C., Daoud-el Baba, M., Keller, B., Weber, C.C., Kramer, B.P.,

Heinzen, C., Aubel, D., Bailey, J.E., and Fussenegger, M. (2002). Macrolide-based

transgene control in mammalian cells and mice. Nature biotechnology 20, 901-907.

Xiao, X., Li, J., and Samulski, R.J. (1998). Production of high-titer recombinant

adeno-associated virus vectors in the absence of helper adenovirus. Journal of

virology 72, 2224-2232.

Yan, Z., Zhang, Y., Duan, D., and Engelhardt, J.F. (2000). Trans-splicing vectors

expand the utility of adeno-associated virus for gene therapy. Proceedings of the

National Academy of Sciences of the United States of America 97, 6716-6721.

Zhang, J., Chua, L.S., and Lynn, D.M. (2004). Multilayered thin films that sustain the

release of functional DNA under physiological conditions. Langmuir 20, 8015-8021.

Zufferey, R., Nagy, D., Mandel, R.J., Naldini, L., and Trono, D. (1997). Multiply

attenuated lentiviral vector achieves efficient gene delivery in vivo. Nature

biotechnology 15, 871-875.

Page 29: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

28

CHAPTER 1

Transduction Technologies

David A. Fluri and Martin Fussenegger (2007). In Methods in

Biotechnology book series, Animal Cell Biotechnology: Methods and

Protocols 2nd Edition, Humana Press, 3-21

Page 30: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

29

Abstract Safe and reliable transfer of genetic information at high frequencies into desired

target cells remains one of the major challenges in therapeutic life sciences. Multiply

attenuated viral vector systems, which are able to transfer heterologous DNA or RNA

into almost any cell phenotype as well as in vivo, emerged as the most efficient

transgene delivery systems in modern biology. We provide a comprehensive technical

overview on the most commonly used viral transduction systems and compare their

specific characteristics in order for researchers to make the best choice in a particular

scientific setting.

1. Introduction

The successful transfer of genetic material to target cells still represents a major

experimental challenge in all aspects of molecular life sciences including

biotechnology and therapeutic initiatives. A promising delivery method involves the

use of engineered animal and human viruses. Such vectors are designed to transfer a

desired transgene by exploiting the efficient delivery machinery of the wild-type virus

while at the same time minimizing the risk of pathogenicity.

Different systems, based on animal as well as human viruses, have recently

been developed and, in some cases, advanced to clinical application. Worldwide more

than 400 clinical trials using viral vectors have been, or are currently being, conducted

(Edelstein et al., 2004). The majority of these trials have been performed using

retroviral vectors, followed by adenoviral and other viral vector systems, among them

adeno-associated viral (AAV) vectors. Due to the high transfer efficiency, and

reasonably straightforward production protocols, viral vectors have also become

increasingly popular for routine applications in various fields of life sciences.

The majority of viral vector systems have been designed to retain, and even

extend, the viruses’ cell tropism while ensuring high biosafety characteristics. For

some viruses the ability to transduce many cell types is intrinsically present (e.g.

adenovirus or AAV). Other viruses have a far narrower host range (e.g. lentiviruses

which are mainly restricted to CD4-expressing cells). Such viruses must be altered

through pseudotyping with envelope or capsid proteins from other compatible virus

Page 31: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

30

species thereby enabling the recombinant vector to transduce a broader variety of cell

types.

To ensure biosafety most vectors are rendered replication incompetent through

selective deletion of wild type viral genetic information, thereby preventing

productive viral life cycles within the target cell (Somia and Verma, 2000). In

addition to the removal of non-essential sequences from the wild-type genome, a

fundamental principle underlying the production of recombinant viral particles is the

redistribution of indispensable elements to either helper plasmids or producer cell

lines. The resulting free space flanked by the viral inverted/long terminal repeats

(ITRs/LTRs) is then employed to accommodate the heterologous DNA to be

delivered. For a number of vector types only one viral open reading frame (ORF) is

replaced and delivered in trans (e.g. first generation adenovirus) (Danthinne and

Werth, 2000), whereas for other types the entire viral coding sequence is replaced by

foreign DNA with only minimal viral elements retained (e.g. adeno-associated virus,

“gutless” adenovirus) (Morsy et al., 1998; Xiao et al., 1998).

The architecture of a viral vector has significant impact on the safety of the

transduction system (see Figure 1). Typically, the more the viral genome is

segregated and lacks overlapping sequences, the smaller is the probability for

recombination-based generation of replication-competent revertants during the

production process. Since wild-type virus is on the whole highly optimized and

adapted to its environment, altering its organization and expression patterns, as done

for the construction of viral vectors, is likely to decrease production efficiency. This

often manifests in lower titers, poor encapsidation of viral genomes or changes in

transduction efficiency of recombinant viral particles. It is therefore crucial to find an

optimal balance between high-safety profiles while retaining high efficiency and

quality of viral particle production.

Every viral transduction system has its unique characteristics which render it

more suitable for one or another application. Crucial parameters influencing selection

of appropriate transduction systems are outlined in Table 1. For every application one

should carefully consider the advantages and drawbacks of each system, and decide

according to the experimental requirements.

This chapter provides detailed protocols and guidelines for the production and

purification of the four most common viral transduction systems; namely,

oncoretroviral, lentiviral, adenoviral and adeno-associated viral vectors. At the end of

Page 32: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

31

the chapter we provide an example in which adeno-associated viral vectors have been

engineered to deliver adjustable reporter genes in vitro to exemplify the convenience

of viral delivery systems for various applications.

Figure 1. Schematic representation of the most popular viral transduction systems. Elements originating from the wild-type virus, which are retained in transfer vectors, are underlined. Only essential viral elements are shown and not drawn to scale. Abbreviations: cap, gene coding for structurual AAV proteins; cPPT, central polypurine tract; E1-E4, adenovirus early transcription units 1-4; env, gene encoding envelope protein; gag, gene encoding core proteins (capsid, matrix, nucleocapsid); ITR, inverted terminal repeat; pol, gene encoding viral reverse transcriptase and integrase proteins; L1-L5, adenovirus late transcription units 1-5; LTR, long terminal repeat; rep, gene coding for replication and multiple other AAV functions; RRE, rev responsive element; nef, rev, tat, vif, vpr, vpu, genes encoding for accessory lenitviral proteins; VA, gene encoding viral associated RNA; ψ, packaging signal

Page 33: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

32

Table 1. Selection criteria for viral transduction systems

2. Materials 2.1.1 General Materials

1. HEK293-T cells (Mitta et al., 2002)

2. DMEM complete medium: DMEM medium (DMEM, Invitrogen, Carlsbad, CA, USA) containing 10% heat-inactivated fetal calf serum (FCS; PAN Biotech GmbH, Aidenbach, Germany, cat. no. 3302-P231902) and 1% penicillin-streptomycin solution (Sigma, St. Louis, MO, cat. no. P4458-100)

Onc

ore t

r ovi

r al

ve c

tors

Lent

ivir a

l v

ecto

r s

Ade

nov i

ral

vec

tor s

Ade

no- a

ssoc

iate

d v

ir al v

e cto

rs

Transduction of non-dividing cel l s - + + +

Stability of viral particl e + / - + / - + / - +

Episomal maintenanc e - - + + / -

Integration into the chromosome + + - + / -

Extensive packaging capaci t y - - + -

Fast production protocol s + + - +

High safety profile + / - + / - + / - +

Low immunogenicit y + + + / - + / -

Clinical data available + - + +

Page 34: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

33

3. Trypsin solution: Trypsin-EDTA solution in PBS (PAN Biotech GmbH, Aidenbach, Germany, cat. no. P10-023100)

4. Sterile CaCl2 (1M) solution

5. Sterile 2X HeBS solution (HeBS; 0.28M NaCl, 0.05M HEPES, 1.5mM Na2HPO4, pH 7.08)

6. Phosphate-buffered saline solution without magnesium and calcium, pH 7.2 (PBS, Sigma, cat. no. D-5652)

7. Cell culture-certified disposable plastic ware: Petri dishes, 6-well plates, 24-well plates (TPP, Trasadingen, Switzerland), pipets, Falcon tubes 15ml and Eppendorf tubes 1.5ml (Greiner bio-one, Kremsmuenster, Austria),

8. Ultracentrifuge tubes (e.g. Beckman Quick-Seal tubes [Beckman cat. no. 342 413] or Sorvall,centrifuge tubes [Sorvall, cat. no. 03141 ])

9. 0.45µm filters (Schleicher & Schuell GmbH, Dassel, Germany)

10. Plasmid DNA purification kits (Jet-Star 2.0 Midiprep, Genomed AG, Bad Oyenhausen, Germany; Wizard mini-prep kit, Promega, Madison, USA)

11. Cell counting device (Casy1® counter, Schaerfe System, Reutlingen, Germany; alternatively, a hemacytometer can be used)

12. Incubator (e.g. Hereaus, HERAcell, Langenselbold, Germany) for cultivation of mammalian cells at 37°C in a humidified atmosphere containing 5% CO2

13. Ultracentrifuge equipped with swing-out rotor (e.g. Sorvall, Hanau, Germany)

2.1.2 Materials for Oncoretroviral/Lentiviral Vectors

1. GP-293 cells (Clontech, Palo Alto, CA, USA, cat. no. K1063-1)

2. pVSV-G (Clontech)

3. pLEGFP-N1 (Clontech)

4. pMF365 (Mitta et al., 2002)

5. pCD/NL-BH* (Mochizuki et al., 1998)

6. pLTR-G (Reiser et al., 1996)

7. Advanced DMEM (Gibco, cat. No. 12491-015)

8. Chemically defined lipid concentrate (Gibco, cat. no. 11905-031)

9. Egg lecithin (Serva Electrophoresis GmbH, cat. no. 27608)

10. Cholesterol (Sigma, cat. no. C-4951, diluted in PBS)

Page 35: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

34

2.1.3 Materials for Adenoviral Vectors

1. Buffer A (10mM Tris-HCl, 1mM MgCl2, 135mM NaCl, pH7.5)

2. Buffer B (buffer A +10% Glycerol)

3. Cesium chloride (Gibco, cat. no. 15507-023)

4. Agarose (Brunschwig, Basel, Switzerland, cat. no. 8008)

5. Phenol/chloroform/isoamylalcohol 25:24:1 saturated with 10mM Tris/1mM EDTA (Sigma, cat. no. P-3803)

6. Ethanol 96%

7. TE Buffer (10mM Tris, 1mM EDTA)

8. Glycerol (Fluka, cat. no. 49767)

9. 16G needles (Becton Dickinson, Franklin Lakes, NJ, USA)

10. Dialysis tubing (Sigma, cat. no. D9777-100FT)

11. Dialysis closures (Sigma, cat. no. Z370959-10EA)

12. HEK293 (Microbix, Toronto, Canada)

13. Adenoviral type 5 genomic vector pJM17 (Microbix)

14. Adenovector encoding desired transgene (e.g. pVN31 (Gonzalez-Nicolini and Fussenegger, 2005))

15. Proteinase K (Qiagen GmbH, Hilden, Germany, cat. no. 19131)

2.1.4 Materials for Adeno-Associated Viral Vectors

1. pDG (Grimm et al., 2003)

2. Spin columns MWCO 300’000 (Vivaspin, Vivascience, Germany)

3. Heparin affinity columns (HiTrap heparin HP, Amersham Biosciences, Sweden)

4. Pump, allowing flow rates of 2ml/min

5. Liquid nitrogen or dry ice/ethanol bath

6. Binding buffer (10mM Na2PO4, pH 7)

7. Elution buffer (10mM Na2PO4, 1M NaCl, pH 7)

8. Storage buffer (binding buffer containing 20% ethanol)

Page 36: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

35

9. PBS-MK buffer (PBS 1x containing 1mM MgCl2 and 2.5mM KCl)

10. Benzonase (Sigma, cat. no. E1014)

11. Optiprepiodixanol solution (Axis-Shield, Rodelokka, Norway)

12. Phenol Red solution 0.5% (Sigma, cat. no. P0290)

2.1.5 Materials for Trigger Inducible Transgene Expression

1. HT-1080 cells (ATCC, CCL-121)

2. AAV vector encoding desired transgene (e.g. pDF141, Figure 2)

3. Erythromycin (Fluka, Buchs, Switzerland, cat. no. 4573) stock solution of 1mg/ml in 96% ethanol

4. 2x SEAP buffer: (20mM homoarginine, 1mM MgCl2 and 21% (v/v) diethanolamine/HCl, pH 9.8.)

5. pNPP solution (120mM para-nitrophenylphosphate [pNPP hexahydrate, Chemie Brunschwig, Basel, Switzerland, cat. no. 12886-0100]) in 2x SEAP buffer

6. Microplate reader (GeniusPro, Tecan Group Ltd., Maennedorf, Switzerland)

7. Fluorescence microscope (DMI6000B, Leica Inc., Heerbrugg, Switzerland) equipped with CFP and YFP-specific filters

3. Methods

All of the viral vectors introduced in this manual are classified as biosafety level

2 according to the National Institutes of Health (NIH). Working with these systems

therefore requires appropriate safety equipment and governmental regulations may

apply. Additional precautions must be followed if working with vectors hosting toxic

or oncogenic inserts.

The production protocols for all of the following viral vectors are based on

comparable techniques. Nevertheless, the protocols for each viral vector system have

been separately provided (apart from general procedures which are discussed in the

first chapter) to ensure simplicity for the user and to provide comprehensive

instructions for each virus type.

Page 37: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

36

3.1. General Procedures

All plasmids were propagated in Escherichia coli DH5α. Standard cloning

techniques were performed to engineer plasmids (Sambrook and Russell, 2001). The

DNA used for virus production was purified using a silica-based anion-exchange

DNA purification kit (Genomed) or with “Wizard” mini-prep kits (Promega) (see

Note 1), according to the manufacturers’ protocols. DNA concentrations were

measured with an UV spectrometer (BioPhotometer, Eppendorf, Hamburg, Germany)

and only DNA with a 260/280 coefficient of greater than 1.75 was used for

transfections. Unless otherwise specified, HEK293 cells were cultivated in DMEM

supplemented with 10% FCS and 1% penicillin/streptomycin solution in a humidified

atmosphere at 37°C containing 5% CO2 (see Note 2).

3.2. Oncoretroviral/Lentiviral vectors

In this chapter oncoretroviral and lentiviral vectors are introduced together as

the two systems are closely related and share numerous features. However, separate

production protocols are provided for each vector system to simplify their utilization.

Both, oncoretroviral and lentiviral vectors are enveloped RNA viruses

packaging two copies of the viral RNA genome. Oncoretroviral vectors have been

derived from different animal viruses such as murine leukemia virus (MLV), rous

sarcoma virus (RSV) or avian leucosis virus (ALV) and were among the first viral

transduction systems used in gene therapy. The most commonly used lentiviral

vectors are derived from human immunodeficiency virus type 1 (HIV-1) (Mitta et al.,

2002; Mochizuki et al., 1998). However, members of the lentivirus family, such as

simian immunodeficiency viruses (SIV’s), feline immunodeficiency virus (FIV) or

equine infectious anaemia virus (EIAV), also serve as parental viruses for the

generation of vectors for gene therapy purposes (Curran and Nolan, 2002).

Oncoretroviruses and lentiviruses randomly integrate as proviruses into target

chromosomes. In contrast to oncoretroviruses, lentiviruses encode accessory genes,

which enable transduction of mitotically inert target cells.

Oncoretroviral vector design is based upon the replacement of the viral gag,

pol and env open reading frames (ORFs) by the desired transgene; either under the

control of a heterologous promoter, or the viral long terminal repeat (LTR). The only

Page 38: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

37

elements required in cis are the extended packaging signal (Ψ+) and the viral LTRs

together with adjacent regions essential for reverse transcription and integration

(Mann et al., 1983; Miller et al., 1993).

The design of lentivectors is very similar to oncoretroviral vectors but requires

additional viral elements provided in trans such as the Rev and Tat proteins. Besides

the LTRs and the packaging signal, lentiviral vectors also contain the Rev-responsive

element (RRE) and the central polypurine tract (cPPT) to enable efficient nuclear

transport and second DNA strand priming, respectively.

Both oncoretroviral and lentiviral vectors can be efficiently pseudotyped with

envelope proteins from other virus species enabling the relatively narrow host-range

of the wild-type virus to be broadened to a wider variety of cell types. The envelope

protein most frequently used for pseudotyping is derived from the vesicular stomatitis

virus glycoprotein (VSV-G), which shows fusogenic activity and uses ubiquitous cell-

surface receptors (Naldini et al., 1996; Reiser et al., 1996).

Retroviral vectors have become very attractive for gene therapy initiatives due

to their stable integration of genetic information combined with low immunogenicity,

improved safety profiles through use of self-inactivating third-generation vectors

(Bukovsky et al., 1999) and simple production protocols. Nonetheless, safety

concerns remain due to random transgene integration resulting in the risk of proto-

oncogene activation or insertional mutagenesis.

In the following chapter we cover the production of small-scale crude

oncoretroviral and lentiviral particle preparations by transient transfection methods

and provide simple purification and concentration protocols using ultracentrifugation.

3.2.1 Retroviral Vector Production

Day 1: Seeding of cells for transfection:

1. Seed GP-293 cells at a concentration of 300’000 cells per 6 well and cultivate at 37°C in DMEM supplemented with 10% FCS, 1% penicillin/streptomycin solution

Page 39: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

38

Day 2: Calcium phosphate transfection of GP-293:

1. Prepare Eppendorf tube containing 80µl of HeBS solution

2. Prepare Eppendorf tube containing 2µg pVSV-G, and 2µg of plasmid encoding the retroviral expression construct (e.g. pLEGFP-N1) in 60µl ddH2O. Add 20µl of 1M CaCl2 solution

3. Add DNA mix drop-wise to HeBS solution while bubbling with a pipette, vortex 5s and let sit for 2min. Meanwhile, add chloroquine at a final concentration of 25µM to the cells. Add precipitates to the cells and let sit for 6h

4. Replace medium with fresh complete DMEM and incubate for another 48h

Day 4: Virus harvest:

1. Collect virus-containing supernatant from GP-293 culture and filter through a 0.45µm filter (Schleicher & Schuell)

2. Aliquot the viral particle preparation and freeze at -80°C

3. Concentrate viral stocks by ultracentrifugation for 3.5h at 4°C and 70’000g in an ultracentrifuge equipped with a swing-out rotor and optiseal tubes (Beckman Instruments inc., CA; cat. no. 361625)

4. Discard supernatant and resuspend the pellet in an appropriate volume of PBS and store aliquots at -80°C or in liquid nitrogen (see Note 5)

3.2.2 Lentiviral Vector Production

Day 1: Seeding of cells for transfection:

1. Seed 500’000 HEK293-T cells in one well of a 6-well plate and cultivate at 37°C in Advanced DMEM supplemented with 2% FCS, 0.01mM cholesterol, 0.01mM egg lecithin and 1x chemically defined lipid concentrate (see Note 3)

Page 40: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

39

Day 2: Calcium phosphate transfection of HEK293-T:

1. Prepare Eppendorf tube containing 80µl of HeBS solution

2. Prepare Eppendorf tube containing 1µg pCD-NL (helper construct), 1µg pLTR-G (construct encoding the VSV-G gene) and 1µg of the vector encoding the lentiviral expression construct (e.g. pMF365) in 60µl ddH2O. Add 20µl of 1M CaCl2 solution

3. Add DNA mix drop-wise to HeBS solution while bubbling with a pipette, vortex 5s and let sit for 2min. Meanwhile, add chloroquine at a final concentration of 25µM to the cells. Add precipitates and let sit for 5h

4. Replace medium with fresh Advanced DMEM containing 2% FCS, cholesterol, egg lecithin and chemically defined lipid concentrate. Let sit for a further 48h

Day 4: Virus harvest:

1. Collect virus-containing medium from HEK293-T cells and filter through a 0.45!m filter (Schleicher & Schuell) (see Note 4)

2. Aliquot the viral particle preparation and freeze at -80°C or in liquid nitrogen (see Note 5)

3. Concentration of viral stocks: lentiviral particles can be purified using the protocol for oncoretroviral vector ultracentrifugation

3.3. Adenovirus

Adenoviral vectors are among the oldest and most widely used vectors for gene

therapy applications. The advantages of this vector type are the extensive packaging

capacity, the ability to transduce a variety of different cell types (including non-

dividing cells), and its potential for high-titer preparations. Adenovirus is a non-

enveloped icosahedral structure, and for its genome contains a double-stranded DNA

molecule of 30-40kb. The genome can be classified according to the expression time

Page 41: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

40

point of two major overlapping regions; the early E and the late L open reading

frames. More than 50 distinct human adenovirus serotypes are known of which some

can cause respiratory, intestinal or eye infections.

Adenoviral vectors have been generated by deleting E1 and E3 (Danthinne

and Imperiale, 2000) (first generation), E1, E3 and E2/4 (Wang et al., 1995) (second

generation), and adenoviruses devoid of any viral genes containing only the

packaging signal and the viral ITRs (high capacity, “gutless” vectors) (Kochanek et

al., 2001). First-generation vectors are able to host transgenes of up to 8kb. Following

deletion of additional information in 2nd- and 3rd-generation vectors the packaging

capacity has been increased up to 35kb for gutless vectors which was associated with

a significant reduction in immunogenicity.

Several different production systems for first-generation adenoviral vectors are

available. Older systems use homologous recombination in HEK293 cells to generate

recombinant adenoviral vectors. Homologous recombination between sequences on

the left-end of the adenovirus genome, and sequences on the shuttle plasmid

containing the transgene as well as the right end of the adenoviral genome, lead to

recombinant virus (Bett et al., 1994). Newer systems take advantage of

recombination-triggering enzymes, such as the phage P1-derived enzyme Cre, to

enable efficient recombination of the viral components (i) in vitro (Wang et al., 1995)

(ii) in bacteria (Aoki et al., 1999; Chartier et al., 1996) or (iii) in a Cre-expressing

helper cell line (Ng et al., 1999).

The following protocol is a standard method for producing first-generation

adenoviral vectors by homologous recombination in HEK293 cells. This protocol can

be adapted to other production systems by changing helper plasmids and production

cell lines.

Page 42: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

41

3.3.1 Production of Adenoviral Particles

3.3.1.1 Production of First-Round Virus

Day 1: Seed 8x105 HEK293 (Microbix) cells in a 6cm culture dish

Day 2: CaCl2 transfection for virus production:

1. Prepare Eppendorf tube containing 240µl of HeBS solution

2. Prepare Eppendorf tube containing 3µg of pJM17 (genomic plasmid) and 3µg of shuttle plasmid (e.g. pVN31 (Gonzalez-Nicolini and Fussenegger, 2005)) encoding the transgene in 180µl ddH2O. Add 60µl of 1M CaCl2 solution

3. Add the DNA mix drop-wise to the HeBS solution while bubbling with a pipette, vortex 5s and let sit for 2min. Add the precipitates to the cells and let sit for 6h.

4. Remove medium and overlay cells with a mix of complete DMEM containing 0.5% agarose (see Note 6)

Day 5-10: Plaques should appear within 5-10 days

1. Pick plaque with sterile Pasteur pipette and transfer to 1ml of PBS containing 10% glycerol (optionally freeze at -80°C until use)

3.3.1.2 Analysis of Viral DNA by Test Digestion

It is recommended that restriction analysis of the backbone is performed prior to amplification of the virus:

Day 1: Seed 8x105 HEK293 (Microbix) cells in a 6cm dish

Day 2: Transduction:

1. Add 200µl of virus suspension from 3.3.1.1. to the cells (cells should be at 80% confluence) and adsorb virus for 30min by shaking plate from time to time to disperse solution over cells

2. Add 5ml of DMEM +10% FCS and incubate at 37°C

3. Cytopathic effect (CPE) should be visible after 3-4 days.

Page 43: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

42

Day 3-4: DNA isolation and restriction digests:

1. Remove medium gently by avoiding the transfer of cells, transfer to Falcon tube and freeze at -80°C

2. Remove remaining medium on the cells and add 360µl of TE buffer containing 0.5% SDS and 0.2mg/ml of Proteinase K and digest overnight

3. Transfer lysate to Eppendorf tube and extract 1x with saturated phenol. Transfer aqueous phase and transfer to new tube

4. Add 1ml of ethanol and vortex briefly

5. Wash twice with ethanol to remove phenol

6. Dry pellet and resuspend in 50µl TE buffer

7. Digest with appropriate restriction enzyme and run on agarose gel (see Note 7)

3.3.1.3 Amplification of Adenoviral Particle Stock

After vector verification, the initial stock can be used for further amplification steps:

Day 1: Seed desired numbers of HEK293 cells in 10cm dishes.

Day 2: When cells reach 80-90% confluence:

1. Remove medium and infect at a MOI of 1-10 (see Note 8) per cell.

2. After 30-60min remove virus solution and re-feed with DMEM +10% FCS. Incubate in humidified atmosphere and check daily for CPE

Day 4-6: When most of the cells are rounded up but not detached:

1. Scrape the cells from the plate with a cell-scraper and transfer cells with the medium to a falcon tube.

2. Centrifuge at 2000g for 15min, remove supernatant and resuspend pellet in desired volume of PBS and freeze at -80°

3. Freeze-thaw 3x by shuttling the sample between liquid nitrogen and 37°C water bath

4. Centrifuge the cell lysate for 15 minutes at 3000rpm to pellet cell debris

5. Transfer supernatant to new tube crude viral stock (see Note 5)

Page 44: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

43

3.3.1.4 Purification of Particles by Caesium Chloride (CsCl) Gradient

For certain applications (mostly in vivo experiments) subsequent purification of crude adenoviral preparations is required. Presented below is a standard purification method using CsCl centrifugation:

1. Add 1.5ml of a 1.45g/ml caesium chloride solution and 2.5ml of a 1.33g/ml of a cesium chloride solution to a 7ml ultraclear tube (place the least dense layer at the bottom of the tube, float this layer on top of the more dense layer again by placing the tip of the syringe at the bottom of the tube)

2. Thaw crude viral stock and layer the virus solution on top of the gradient

3. Add 2mm of mineral oil on top of the virus solution and equilibrate tubes

4. Centrifuge for 2h at 90’000g (at 4°C)

5. Remove the lowest of the three visible white bands by side-wall puncture with a 16G needle (Becton Dickinson)

6. Dilute the virus fraction with 0.5 volumes of TE buffer pH 7.8

7. Layer the solution on top of a second gradient prepared as above but this time with 1ml of 1.45g/ml and 1.5ml of 1.33g/ml cesium chloride and proceed as in step 1c, 2 and 3

8. Recover the virus particle-containing band, transfer to dialysis tubing, seal with clamps and dialyse for 1h against 1l of buffer A

9. Transfer tubing to another beaker containing 1l of buffer B (buffer A + 10% glycerol) and dialyse for another 2h

10. Aliquot virus and freeze at -80°C (see Note 5)

3.4. Adeno-Associated Virus (AAV)

In recent years adeno-associated viral vectors have become widely used in

clinical initiatives due to their unique safety properties. So far, no disease has been

attributed to the wild-type virus even though a majority of the human population is

sero-positive for one or more of the different subtypes. For productive infection AAV

is dependent on a helper virus, which is either adenovirus or herpes simplex virus. In

the absence of helper functions the AAV enters a non-productive life cycle and

integrates site specifically into a locus on human chromosome 19 (Kotin et al., 1990;

Samulski et al., 1991). Recombinant adeno-associated viruses lack this characteristic

Page 45: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

44

and remain predominantly episomal (ca. 90%), although a small proportion does still

randomly integrate into the host chromosome (ca. 10%) (Nakai et al., 2001).

The unique design of recombinant adeno-associated viral vectors (rAAVs),

where all viral open reading frames have been replaced and only the ITRs are

delivered in cis, renders this transduction system exceptionally safe. Furthermore, the

ability of adeno-associated viral vectors to transduce non-dividing cells and provide

sustained expression of the transgene in a wide variety of tissues makes AAVs one of

the most promising candidates for clinical applications.

Standard production protocols use transient transfection of plasmids encoding

the desired transgene(s) flanked by viral ITRs together with a plasmid providing the

AAV Rep and Cap proteins in trans. HEK293 production cells already provide the

crucial adenoviral functions performed by viral E1A and E1B. In addition, other

adenoviral proteins such as E2A, E4orf6 and VA are required for efficient production

of AAVs. In older protocols these functions are provided by helper adenovirus

infection of producer cells (Hermonat and Muzyczka, 1984; Samulski et al., 1989). A

major drawback of such protocols is the contamination of rAAV stocks with wild-

type helper adenovirus. State-of-the-art protocols replace helper virus infection

completely by providing the essential proteins on engineered helper plasmids.

In this section we provide a production protocol where all adenoviral helper

functions (except E1) are combined with Rep/Cap on a single plasmid (Grimm et al.,

2003). Even though titers obtained with such helper virus-free production protocols

are occasionally lower, the advantages in respect of purity and safety outbalance the

drawbacks.

Also provided in this section are detailed protocols for the production and

purification of crude adeno-associated viral stocks, as well as purification and

concentration strategies for use in animal studies.

Page 46: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

45

3.4.1 Production of Crude Adeno-Associated Viral Vector Preparations

Day 1: Seeding of cells for transfection:

1. Seed 300’000 HEK293-T cells per well of a 6-well plate (or for medium-scale production seed 2.5 Mio cells into a 10cm culture dish)

Day 2: Calcium phosphate transfection of HEK293-T:

1. Prepare tube containing 80µl (400µl) of HeBS solution

2. Prepare tube containing 1.2µg (8µg) plasmid containing transgene of interest flanked by the viral ITRs and 2.4µg (16µg) pDG28 (helper plasmid containing all required adenovirus functions plus AAV rep and cap ORFs) in 60µl (300µl) ddH2O. Add 20µl (100µl) of 1M CaCl2 solution

3. Add DNA drop-wise to HeBS solution while bubbling the solution using a pipette, vortex the mix for 5s and let sit for 2min. Add the precipitates drop-wise to the cells and let sit for another 6h

4. Replace medium with fresh complete medium and incubate for 60h

Day 5: Virus harvest:

1. In the morning, remove medium from cells, wash carefully with 400µl (2ml) PBS and add another 500µl (2ml) of PBS to the cells

2. Detach cells by pipetting with a 1000µl micropipet, transfer cell-suspension to an Eppendorf (15ml Falcon) tube and vortex vigorously

3. Freeze-thaw 3x by shuttling the sample between 37°C water bath and liquid nitrogen (vortex vigorously after each thawing step)

4. Centrifuge for 5min at 8000g

5. Transfer supernatant to new tube ( crude viral stock) and store at -80°C (see Note 5)

3.4.2 Virus Purification (for Medium-Scale High-Purity Virus Produced in 10cm Culture Dishes)

1. Add 50U/ml Benzonase (Sigma) to crude viral stock and incubate for 30min at 37°C

2. Dilute crude viral stock in PBS to a final volume of 12ml

3. In an ultracentrifuge tube prepare an iodixanol step gradient by pipetting the crude viral preparation onto a solution containing 15% iodixanol and 1M NaCl in PBS-MK buffer. Continue to sequentially pipet 5ml of a 25%, 4ml of a 40% and 4ml of a 60% iodixanol solution (all in PBS-MK) under the 15%

Page 47: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

46

iodixanol solution. For better distinction of the iodixanol layers, add 2.5µl/ml of a 0.5% Phenol Red stock solution to the 60% and 25% iodixanol layers.

4. Centrifuge for 3.5h at 150’000g (18°C)

5. Harvest the clear 40% iodixanol fraction after puncturing the tube on the side with a 16G needle equipped with a syringe.

6. Equilibrate heparin affinity column with 5-10 bed volumes of binding buffer

7. Filter gradient-purified virus preparation through a 0.45µm filter (Schleicher & Schuell), and run it over the column at a flow rate of approximately 1.5ml/min

8. Wash with 10 bed volumes of binding buffer

9. Elute bound virus with 5 bed volumes of elution buffer

10. Transfer eluate to a vivaspin column and spin for 20min at 3800g, resuspend in 5ml PBS and spin another 20min at 3800g

11. Resuspend in the desired volume of PBS, aliquot and freeze at -80°C

3.5. Trigger-Inducible Transgene Transduction Using Engineered AAV

Systems enabling regulated transgene expression are invaluable tools in

different aspects of molecular life sciences. Delivery of a desired transgene coupled

with the capability to tightly regulate its expression is essential for various

applications (Clackson, 1997; Fussenegger, 2001; Fussenegger et al., 1998; Weber

and Fussenegger, 2002). Presented above are different methods for efficient delivery

of transgenes to target cells capitalizing on viral transduction systems. Here we

provide an application example using adeno-associated viral vectors to transfer and

regulate intracellular and secreted reporter transgenes. This protocol may be amenable

to other combinations of viral vectors, transgenes and transgene control systems.

Page 48: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

47

Figure 2. Plasmid maps of adeno-associated viral vectors engineered for macrolide-responsive expression of transduced transgenes. Vectors encoding (i) the macrolide-dependent transactivator ET1 driven by a constitutive promoter PhCMV (pDF51), (ii) SEAP driven by the erythromycin-responsive promoter PETR (pDF77) and (iii) a macrolide-responsive self-regulated one-vector configuration combining PSV40- driven ET1 and PETR-driven EYFP expression (pDF141). Abbreviations: bla, beta lactamase; ET1, erythromycin transactivator; EYFP, enhanced yellow fluorescent protein; ITR, inverted terminal repeat; ORI, origin of replication; pAhgh, human growth hormone polyadenylation signal; pASV40, simian virus 40 polyadenylation signal; PhCMV, human immediate early cytomegalovirus promoter; PETR, erythromycin-responsive promoter; PSV40, simian virus 40 promoter; SEAP, secreted alkaline phosphatase

NotI 142 MluI 150

HindIII 435

EcoRI 637

PmlI 2454

NotI 2468

NotI 2210BstBIAscIBclISphISpeIPacISwaIMluIPmeIBsaBI 2295

ITR

PETR

SEAP

pASV40

ITR

bla

Ori

pDF77

5213 bp

NotI 142

MluI 150

ClaI 1319

EcoRI 1326

NotI 2889

XbaI 2324HindIIIBglII 2373

ITR

PhCMV

!-globin intron

ET1

pAhghITR

bla

Ori

pDF51

5634 bp

ClaI 390

HpaI 776

EcoRI 821

PacI 1833

SwaI 1840

PmeI 1852

PmlI 3234

EcoRI 2224AflIINruINdeIAgeI 2280

BstBI 3021SpeIPacISwaIPmeI 3066

ITR

PSV40

ET1

pASV40

PETR

EYFPpASV40

ITR

bla

Ori

pDF141

5993 bp

Page 49: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

48

3.5.1 Viral Transduction using adeno-associated virus

Day 1: Seeding of cells:

1. Seed 40’000 HT-1080 cells per well of a 24-well plate (9 wells) and 200’000 HT-1080 cells per well of a 6-well plate (2 wells)

Day 2: Transduction:

1. Thaw vector stocks in a 37°C water bath and vortex tubes briefly

2. Add pDF141-derived AAVs at a multiplicity of infection (MOI) of 5 (see Note 8) to 2 wells of 6-well plate and pDF77- and pDF51-derived AAVs at identical MOIs to 6 wells of a 24-well plate (see Figure 2 for plasmid maps)

3. Add Erythromycin (EM) at 50ng/ml to one of the wells transduced with pDF141 and to 3 of the wells containing pDF143

4. Incubate overnight at 37°C in a humidified atmosphere containing 5% CO2

Day 3: Medium change:

1. Change medium the following day and add new DMEM complete, supplemented with 50ng/ml EM where transgene expression should be repressed

2. Incubate for another 48h in a humidified atmosphere containing 5% CO2

Day 5: Analysis:

Fluorescence microscopy:

Analyze samples using a fluorescence microscope (Leica) equipped with appropriate filters (CFP/YFP filter cube, Leica) (see Figure 3)

SEAP quantification:

1. Harvest 120µl supernatants per well from pDF77/51-transduced cells and 120µl from non-transduced cells as a negative control

2. Heat inactivate supernatants for 20min at 65°C

3. Incubate for 5min on ice and then spin down at top speed for 5min

4. Prepare 9 wells of a 96-well assay plate with 100µl of 2x SEAP assay buffer, pre-warm at 37°C

5. Transfer 80µl culture supernatant to wells containing 2x SEAP buffer

6. Add 20µl substrate solution to each well

Page 50: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

49

7. Quantify absorbance at 405nm for up to 1h in a microplate reader (Tecan) (see Figure 3).

1. Subtract values of the blank from sample values

2. Calculate enzymatic activity

EA [U/L] = ΔAbs/min*ν/(ε*d)*106

Where:

Dilution factor n: volume measurement/volume sample = 200/80

Absorption factor of ε=18600 M-1cm-1

Lightpath d=0.5cm

Figure 3. Macrolide-responsive transgene transduction using engineered adeno-associated viral particles. (A) SEAP expression levels of HT-1080 cells co-transduced with pDF51- and pDF77-derived AAV particles (see Figure 2). (B) Fluorescence micrographs of HT-1080 transduced with pDF141-derived AAV particles delivering a self-regulated macrolide-responsive EYFP expression unit and grown in the presence (+EM) and absence of erythromycin (-EM). Abbreviations: EM, erythromycin; SEAP, secreted alkaline phosphatase

Page 51: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

50

4. Notes

1. To perform transfections we recommend using ethanol or iso-propanol-precipitated DNA for sterility reasons. For constructs used in larger amounts (e.g. helper constructs) we recommend performing midi- or maxi- DNA preparations in order to minimize batch-to-batch variations of virus preparations. For small-scale virus preparations, DNA purification methods based on rapid precipitation-free protocols (e.g. Wizard mini DNA purification system) are sufficient.

2. We observed decreasing efficiency of viral particle production upon transient transfection when using HEK293 cells at higher passage number and recommend therefore to work with low-passage cell populations.

3. Production of lentiviral particle stock is feasible using standard DMEM containing 10% FCS without providing cholesterol, egg lecithin and chemically defined lipid concentrate but infectious titers are significantly lower (Mitta et al., 2005).

4. It is crucial to use low protein binding filters.

5. Storage at -80° (up to 6 months) is sufficient. For long-term storage we recommend liquid nitrogen. Oncoretroviral and lentiviral vectors should be aliquoted in small volumes since preparations loose approximately half of their infectivity with every freeze-thaw step.

6. Some protocols forgo this step by directly cultivating the cells in liquid medium and then harvesting the entire supernatant for further procedures. We recommend initial plaque purification and analysis of the first produced viral stocks to ensure consistent starting material for further steps.

7. Upon complete CPE the DNA preparation should be sufficiently pure for restriction analysis. Nevertheless, genomic DNA will be visible as background smear and some enzymes (e.g. HindIII) repeatedly cut genomic DNA yielding bands which are not to be confused with viral DNA.

8. Vectors encoding for fluorescent proteins are titrated by applying serial dilutions

of vector preparations to target cells in 96-well plates (at low density) and subsequent counting of transgene-expressing cells by fluorescence microscopy.

Page 52: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

51

5. References

Aoki, K., Barker, C., Danthinne, X., Imperiale, M.J., and Nabel, G.J. (1999). Efficient

generation of recombinant adenoviral vectors by Cre-lox recombination in vitro. Mol

Med 5, 224-231.

Bett, A.J., Haddara, W., Prevec, L., and Graham, F.L. (1994). An efficient and

flexible system for construction of adenovirus vectors with insertions or deletions in

early regions 1 and 3. Proc Natl Acad Sci U S A 91, 8802-8806.

Bukovsky, A.A., Song, J.P., and Naldini, L. (1999). Interaction of human

immunodeficiency virus-derived vectors with wild-type virus in transduced cells. J

Virol 73, 7087-7092.

Chartier, C., Degryse, E., Gantzer, M., Dieterle, A., Pavirani, A., and Mehtali, M.

(1996). Efficient generation of recombinant adenovirus vectors by homologous

recombination in Escherichia coli. J Virol 70, 4805-4810.

Clackson, T. (1997). Controlling mammalian gene expression with small molecules.

Curr Opin Chem Biol 1, 210-218.

Curran, M.A., and Nolan, G.P. (2002). Nonprimate lentiviral vectors. Curr Top

Microbiol Immunol 261, 75-105.

Danthinne, X., and Imperiale, M.J. (2000). Production of first generation adenovirus

vectors: a review. Gene Ther 7, 1707-1714.

Danthinne, X., and Werth, E. (2000). New tools for the generation of E1- and/or E3-

substituted adenoviral vectors. Gene Ther 7, 80-87.

Edelstein, M.L., Abedi, M.R., Wixon, J., and Edelstein, R.M. (2004). Gene therapy

clinical trials worldwide 1989-2004-an overview. J Gene Med 6, 597-602.

Fussenegger, M. (2001). The impact of mammalian gene regulation concepts on

functional genomic research, metabolic engineering, and advanced gene therapies.

Biotechnol Prog 17, 1-51.

Fussenegger, M., Schlatter, S., Datwyler, D., Mazur, X., and Bailey, J.E. (1998).

Controlled proliferation by multigene metabolic engineering enhances the

productivity of Chinese hamster ovary cells. Nat Biotechnol 16, 468-472.

Gonzalez-Nicolini, V., and Fussenegger, M. (2005). A novel binary adenovirus-based

dual-regulated expression system for independent transcription control of two

different transgenes. J Gene Med.

Page 53: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

52

Grimm, D., Kay, M.A., and Kleinschmidt, J.A. (2003). Helper virus-free, optically

controllable, and two-plasmid-based production of adeno-associated virus vectors of

serotypes 1 to 6. Mol Ther 7, 839-850.

Hermonat, P.L., and Muzyczka, N. (1984). Use of adeno-associated virus as a

mammalian DNA cloning vector: transduction of neomycin resistance into

mammalian tissue culture cells. Proc Natl Acad Sci U S A 81, 6466-6470.

Kochanek, S., Schiedner, G., and Volpers, C. (2001). High-capacity 'gutless'

adenoviral vectors. Curr Opin Mol Ther 3, 454-463.

Kotin, R.M., Siniscalco, M., Samulski, R.J., Zhu, X.D., Hunter, L., Laughlin, C.A.,

McLaughlin, S., Muzyczka, N., Rocchi, M., and Berns, K.I. (1990). Site-specific

integration by adeno-associated virus. Proc Natl Acad Sci U S A 87, 2211-2215.

Mann, R., Mulligan, R.C., and Baltimore, D. (1983). Construction of a retrovirus

packaging mutant and its use to produce helper-free defective retrovirus. Cell 33, 153-

159.

Miller, A.D., Miller, D.G., Garcia, J.V., and Lynch, C.M. (1993). Use of retroviral

vectors for gene transfer and expression. Methods Enzymol 217, 581-599.

Mitta, B., Rimann, M., Ehrengruber, M.U., Ehrbar, M., Djonov, V., Kelm, J., and

Fussenegger, M. (2002). Advanced modular self-inactivating lentiviral expression

vectors for multigene interventions in mammalian cells and in vivo transduction.

Nucleic Acids Res 30, e113.

Mitta, B., Rimann, M., and Fussenegger, M. (2005). Detailed design and comparative

analysis of protocols for optimized production of high-performance HIV-1-derived

lentiviral particles. Metab Eng.

Mochizuki, H., Schwartz, J.P., Tanaka, K., Brady, R.O., and Reiser, J. (1998). High-

titer human immunodeficiency virus type 1-based vector systems for gene delivery

into nondividing cells. J Virol 72, 8873-8883.

Morsy, M.A., Gu, M., Motzel, S., Zhao, J., Lin, J., Su, Q., Allen, H., Franlin, L.,

Parks, R.J., Graham, F.L., et al. (1998). An adenoviral vector deleted for all viral

coding sequences results in enhanced safety and extended expression of a leptin

transgene. Proc Natl Acad Sci U S A 95, 7866-7871.

Nakai, H., Yant, S.R., Storm, T.A., Fuess, S., Meuse, L., and Kay, M.A. (2001).

Extrachromosomal recombinant adeno-associated virus vector genomes are primarily

responsible for stable liver transduction in vivo. J Virol 75, 6969-6976.

Page 54: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 1

53

Naldini, L., Blomer, U., Gallay, P., Ory, D., Mulligan, R., Gage, F.H., Verma, I.M.,

and Trono, D. (1996). In vivo gene delivery and stable transduction of nondividing

cells by a lentiviral vector. Science 272, 263-267.

Ng, P., Parks, R.J., Cummings, D.T., Evelegh, C.M., Sankar, U., and Graham, F.L.

(1999). A high-efficiency Cre/loxP-based system for construction of adenoviral

vectors. Hum Gene Ther 10, 2667-2672.

Reiser, J., Harmison, G., Kluepfel-Stahl, S., Brady, R.O., Karlsson, S., and Schubert,

M. (1996). Transduction of nondividing cells using pseudotyped defective high-titer

HIV type 1 particles. Proc Natl Acad Sci U S A 93, 15266-15271.

Sambrook, J., and Russell, D.W. (2001). Molecular cloning a laboratory manual, 3rd

edn (Cold Spring Harbor, New York, Cold Spring Harbor Laboratory Press).

Samulski, R.J., Chang, L.S., and Shenk, T. (1989). Helper-free stocks of recombinant

adeno-associated viruses: normal integration does not require viral gene expression. J

Virol 63, 3822-3828.

Samulski, R.J., Zhu, X., Xiao, X., Brook, J.D., Housman, D.E., Epstein, N., and

Hunter, L.A. (1991). Targeted integration of adeno-associated virus (AAV) into

human chromosome 19. Embo J 10, 3941-3950.

Somia, N., and Verma, I.M. (2000). Gene therapy: trials and tribulations. Nat Rev

Genet 1, 91-99.

Wang, Q., Jia, X.C., and Finer, M.H. (1995). A packaging cell line for propagation of

recombinant adenovirus vectors containing two lethal gene-region deletions. Gene

Ther 2, 775-783.

Weber, W., and Fussenegger, M. (2002). Artificial mammalian gene regulation

networks-novel approaches for gene therapy and bioengineering. J Biotechnol 98,

161-187.

Xiao, X., Li, J., and Samulski, R.J. (1998). Production of high-titer recombinant

adeno-associated virus vectors in the absence of helper adenovirus. J Virol 72, 2224-

2232.

Page 55: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

54

CHAPTER 2

Adeno-Associated Viral Vectors Engineered For

Macrolide-Adjustable Transgene Expression In

Mammalian Cells and Mice

David A. Fluri , Marie Daoud-El Baba and Martin Fussenegger (2007).

BMC Biotechnology, Nov 6;7:75

Page 56: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

55

Abstract Background

Adjustable gene expression is crucial in a number of applications such as de- or

transdifferentiation of cell phenotypes, tissue engineering, various production processes as

well as gene-therapy initiatives. Viral vectors, based on the Adeno-Associated Virus (AAV)

type 2, have emerged as one of the most promising types of vectors for therapeutic

applications due to excellent transduction efficiencies of a broad variety of dividing and

mitotically inert cell types and due to their unique safety features.

Results

We designed recombinant adeno-associated virus (rAAV) vectors for the regulated

expression of transgenes in different configurations. We integrated the macrolide-responsive

E.REX systems (EON and EOFF) into rAAV backbones and investigated the delivery and

expression of intracellular as well as secreted transgenes for binary set-ups and for self- and

auto-regulated one-vector configurations. Extensive quantitative analysis of an array of

vectors revealed a high level of adjustability as well as tight transgene regulation with low

levels of leaky expression, both crucial for therapeutical applications. We tested the

performance of the different vectors in selected biotechnologically and therapeutically

relevant cell types (CHO-K1, HT-1080, NHDF, MCF-7). Moreover, we investigated key

characteristics of the systems, such as reversibility and adjustability to the regulating agent, to

determine promising candidates for in vivo studies. To validate the functionality of delivery

and regulation we performed in vivo studies by injecting particles, coding for compact self-

regulated expression units, into mice and adjusting transgene expression.

Conclusion

Capitalizing on established safety features and a track record of high transduction

efficiencies of mammalian cells, adeno- associated virus type 2 were successfully engineered

to provide new powerful tools for macrolide-adjustable transgene expression in mammalian

cells as well as in mice.

Background An array of different viral transduction systems are being used currently in pre-clinical

and clinical trials (Edelstein et al., 2004; Tomanin and Scarpa, 2004; Vandendriessche, 2002).

Among these, vectors based on the replication-defective adeno-associated virus type 2 have

attracted special attention as tools for clinical gene transfer. Different characteristics, such as

Page 57: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

56

(i) the ability to transduce dividing as well as non-dividing cells, (ii) high transduction rates in

a wide range of tissues, and notably, (iii) the unique safety properties, make AAVs a

promising vector in gene therapy initiatives (Flotte et al., 1992; Grimm and Kay, 2003;

Kaplitt et al., 1994; Li et al., 2005; Nakai et al., 2001; Wang et al., 2000; Xu et al., 2000).

Over the past few years, extensive studies have been carried out on different systems

to regulate transgenes with small-molecule stimuli, preferentially clinically licensed agents.

Started by the tetraycline-responsive TET system (Gossen and Bujard, 1992; Gossen et al.,

1995), numerous other control modalities have followed including those responsive to

streptogramin (Fussenegger et al., 2000), macrolide (Weber et al., 2002a) and

aminocoumarines (Zhao et al., 2003), immunosuppressive agents (rapamycin) (Rivera et al.,

1996), hormones (Beerli et al., 2000; Braselmann et al., 1993; No et al., 1996), or susceptible

to temperature (Boorsma et al., 2000; Weber et al., 2003), quorum sensing molecules

(Neddermann et al., 2003; Weber et al., 2003) and gaseous acetaldehyde (Weber et al., 2004).

To date, most of the experimental work with AAV vectors in the gene-therapy field has

focused on (i) the expression of therapeutic transgenes driven by strong constitutive

promoters (Acland et al., 2005; Flotte et al., 1996; Haberman et al., 2003), (ii) the regulated

expression of transgenes based on the tetracycline-responsive TETON and TETOFF systems

(Bohl et al., 1998; Chtarto et al., 2003; Gafni et al., 2004; Jiang et al., 2004) and, to lesser

extent, (iii) the regulated expression of transgenes by rapamycin-controlled transgene

expression (Rivera et al., 2005; Wang et al., 2005).

Until recently, in-vivo studies using recombinant AAV particles have been limited by

the production of high-titer and helper virus-free preparations. However, with the

development of helper-free production methods (Grimm et al., 1998; Xiao et al., 1998) and

improved purification and concentration protocols (Clark et al., 1999; Zolotukhin et al.,

1999), high-titer production of AAV particles in the absence of helper-virus contamination

was achieved, thereby opening the way to in vivo and clinical studies with AAV-derived

particles.

We report the design and validation of different AAV type 2-based expression vectors,

which enable macrolide-controlled transgene expression capitalizing on the recently

developed erythromycin-responsive expression technology (E.REX) (Weber et al., 2002a).

E.REX systems exist in two different configurations: (i) the EOFF arrangement consisting of a

macrolide-dependent transactivator (ET, a fusion of the Escherichia coli MphR(A) repressor

protein [E] and the Herpes simplex virus VP16 transactivation domain) which binds and

activates chimeric promoters (PETR) assembled by placing ET-specific operator modules 5’ of

Page 58: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

57

a minimal eukaryotic promoter in a macrolide-responsive manner. Since the presence of

erythromycin turns transgene expression off by abolishing the ET-PETR interaction these

control modalities are known as OFF-type or EOFF systems (Weber et al., 2002a). (ii) The EON

technology consists of a macrolide-dependent transrepressor (E-KRAB (ET4, (Weber et al.,

2002a)), a fusion of the E. coli MphR(A) repressor protein [E] and the KRAB (Krueppel-

associated box) transsilencing domain of the human kox-1 gene), which binds and represses

chimeric promoters (PETRON) assembled by placing E-KRAB-specific operator modules 3’ of

a constitutive eukaryotic promoter in a macrolide-inducible manner. Since the presence of

erythromycin turns transgene expression ON by releasing E-KRAB from PETRON, these

control arrangements are known as ON-type or EON systems (Weber et al., 2002a).

We have designed a set of recombinant AAV vectors harboring EOFF or EON-

controlled expression units (i) on two independent vectors (binary system), (ii) on a single

vector expressing the transgene and the transactivator in a dicistronic or bidirectional

configuration and (iii) on a single vector containing a constitutive promoter, driving

transactivator expression, and the regulatable PETR promoter, driving the gene of interest. We

have optimized performance, delivery and regulation, analyzed key characteristics such as

reversibility and adjustability of the systems and validated the results with quantitative in-

vitro as well as mouse studies.

Results Design of recombinant adeno-associated viral particles for transduction of EOFF-

controlled transgene expression

We have engineered serotype 2-based adeno-associated viral particles for transduction

of macrolide-responsive expression of the enhanced yellow fluorescent protein (EYFP). The

generic design consisted of a set of two vectors: pDF51 (ITR-PhCMV-intronβ-globin-ET1-pAHGH-

ITR) harboring an ITR (inverted terminal repeats)-flanked PhCMV- (human cytomegalovirus

immediate early promoter) driven and pAhgh- (human growth hormone-derived

polyadenylation site) terminated ET1 (macrolide-dependent transactivator) expression unit

containing a β-globin intron (intronβ-globin) reported to increase transcript processing and

overall ET1 production levels and pDF54 (ITR-PETR-EYFP-pASV40-ITR) containing an ITR-

flanked expression unit encoding PETR- (macrolide-responsive promoter) driven and pASV40-

(simian virus 40-derived polyadenylation site) terminated EYFP (enhanced yellow

fluorescent protein-encoding gene) expression cassette (Figure 1A). Transgenic AAV-derived

Page 59: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

58

particles produced by transient co-transfection of either pDF60, pDF51 or pDF54 with the

helper construct pDG providing constitutive processing and assembly functions (adenovirus

E2A, E4 and VA as well as AAV rep and cap genes; (Grimm et al., 2003)) in trans into

HEK293-T were validated for regulated EYFP expression by co-transduction of pDF51- and

pDF54-derived AAV particles into representative cell types such as Chinese hamster ovary

cells (CHO-K1), human fibrosarcoma cells (HT-1080), primary normal human dermal

fibroblasts (NHDF) and human breast cancer cells (MCF-7) cultivated in the presence (+EM)

or absence (-EM) of the macrolide antibiotic erythromycin (EM). Fluorescence micrographs

of transduced populations maintained for 48h in erythromycin-free medium revealed bright

green fluorescence in all cell types. Transduced populations cultivated for two days in the

presence of 1µg/ml erythromycin showed no important EYFP expression. As a positive

control all cell types were transduced with AAV particles harboring EYFP driven by a

constitutive human cytomegolovirus immediate early promoter (PhCMV) (Figure 1B).

One vector-based macrolide-responsive AAV expression vectors

In order to enable delivery of macrolide-responsive transgene expression in a most

compact format and a single transduction event we engineered expression of the macrolide-

dependent transactivator ET1 and the desired target gene into a single AAV vector

configuration. The pioneering one-vector design concept consisted of tandem constitutive ET1 (PSV40-ET1-pASV40) and macrolide-responsive EYFP expression units (PETR-EYFP-

pASV40) placed between two ITRs (Figure 2A). pDF141- (ITR-PSV40-ET1-pASV40-PETR-EYFP-

pASV40-ITR) derived AAV particles efficiently transduced HT-1080 and MCF-7 and mediated

high-level EYFP expression when transgenic populations were grown for 48h in the absence

of erythromycin (Figure 2B). To prevent any deregulation of PETR by promoters or enhancers

encoded in cis and to minimize the overall size of the transgene unit, we assembled (i)

pDF124 (ITR-PETR-EYFP-IRESEMCV-ET1-pASV40-ITR) a dicistronic autoregulated PETR-

driven AAV expression configuration harboring an ITR-flanked PETR-driven dicistronic

expression cassette with EYFP in the first and ET1 in the second cistron separated by a

internal ribosome entry site of encephalomyocarditisviral origin (IRESEMCV) and (ii) pDF89

(ITR-pAI-EYFPPhCMVmin-ETR-PHSP70minET1-pASV40-ITR) containing a bidirectional

expression unit consisting of a central ET1-specific operator ETR flanked by PETR driving

EYFP expression in one direction and a PHSP70min (minimal version of the Drosophila

melanogaster heat-shock protein 70 promoter) driving ET1 in the opposite direction.

Page 60: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

59

Figure 1. AAV type 2-based macrolide-responsive EYFP expression. (A) Schematic representation of AAV

type 2-based vectors encoding ET1 under control of the constitutive PhCMV promoter (pDF51) and EYFP driven

by PETR (pDF54) or a constitutive hCMV promoter (pDF60). (B) Fluorescence micrographs of CHO-K1, HT-

1080, NHDF and MCF-7 either co-transduced with pDF51/pDF54 and cultivated in the presence (+) and

absence (-) of erythromycin (EM) or transduced with pDF60 (1000 genomic particles/cell for each vector).

Abbreviations: EM, erythromycin; ET1, erythromycin-dependent transactivator; EYFP, enhanced yellow

fluorescent protein; ITR, inverted terminal repeat; pAhgh, human growth hormone polyadenylation signal;

pASV40, simian virus 40 polyadenylation signal; PETR, erythromycin-responsive promoter; PhCMV, human

immediate early cytomegalovirus promoter; Selected restriction sites: A, AccI; As, AscI; Bg, BglII; Bs, BstBI;

Bsa, BsaBI; C, ClaI; E, EcoRI; H, HindIII; Hi, HincII; M, MluI; N, NdeI; Nh, NheI; Nr, NruI; P, PacI; Pm,

PmeI; Pml, PmlI; S, SalI; Sa, SacII Sp, SpeI; Sph, SphI; Sw, SwaI; X, XbaI.

Page 61: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

60

Table 1. Plasmids used and designed in this study

Plasmid Description and Cloning Strategy Reference or source

pAAV-MCS

AAV transfer vector Stratagene

pAAV-lacZ

AAV vector expressing lacZ Stratagene

pDG Helper construct encoding AAV Rep/Cap as well as Adeno virus E2A, E4 and VA.

(Grimm et al., 2003)

pCF18 Plasmid containing ECFP driven by a tetracycline responsive promoter and EYFP driven by a pristinamycin responsive promoter

(Fux and Fussenegger, 2003)

pCF19 Plasmid containing SEAP cassette (Fux et al., 2004)

pCF125 Plasmid expressing ECFP and ET1 from a bidirectional promoter (Fux et al., 2003)

pSS134 Plasmid containing SEAP cassette unpublished pWW43 Plasmid expressing E-KRAB (Weber et al.,

2002a) pWW76 Plasmid containing tricistronic expression configuration driven by PETRON8 (Weber et al.,

2002a) pWW78 pTRIDENT1-based tricistronic expression vector for macrolide-responsive

auto-regulated expression of up to two desired transgenes. (Weber et al., 2002b)

pBP141 Vector expressing SEAP and ET1 under tetracycline-responsive promoter: PhCMV*-1-SEAP-IRESPV-ET-pA

unpublished

pMF123 Plasmid encoding tricistronic expression cassette driven by a constitutive SV40 promoter.

(Fussenegger et al., 1998)

pMF351 Lentiviral vector encoding EYFP driven by a constitutive hCMV promoter (Mitta et al., 2002)

pDF37 AAV2 vector containing a tricistronic PETR driven expression unit. The entire expression unit from pWW73 was excised using SspI/XbaI, polished by Klenow and cloned into pAAV-lacZ which was NotI digested and Klenow polished before, thus resulting in pDF37 (ITR-PETR-IRESPV-IRESEMCV-pASV40-ITR).

this work

pDF51 AAV2 vector containing a constitutive hCMV driven ET1 cassette. ET1 was excised from pWW078 using EcoRI/HindIII and cloned into the corresponding sites of pAAV-MCS, thus resulting in pDF51 (ITR-PETR-Intronβ-globin-ET1-pAhgh-ITR).

this work

pDF54 AAV2 vector encoding EYFP driven by the erythromycin responsive PETR promoter. PETR was excised from pDF55 using AccI/NheI and cloned into the corresponding sites of pDF60, thus resulting in pDF54 (ITR-PETR-EYFP-pASV40-ITR).

this work

pDF55 AAV2 vector encoding divergent expression units for ECFP driven by PETR and ET1 driven by a HSP70 minimal promoter. The entire expression cassette was excised from pCF125 using EcoRV/XbaI and cloned into the HincII/SpeI sites of pDF60, thus resulting in pDF55 (ITR-pAI-ECFP←PETR-ETR-PHSP70min→ET1-pASV40-ITR).

pDF56 AAV2 vector encoding ET1 driven by a constitutive SV40 promoter. ET1 was this work

Page 62: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

61

excised from pWW78 using EcoRI/XbaI and cloned into the EcoRI/SpeI sites of pDF63, thus resulting in pDF56 (ITR-PSV40-ET1-pASV40-ITR).

pDF60 AAV2 vector encoding EYFP driven by a constitutive hCMV promoter. PhCMV-EYFP was excised from pMF351 using XbaI/PacI and cloned into NheI/PacI sites of pDF63, thus resulting in pDF60 (ITR-PhCMV-EYFP-pASV40-ITR).

this work

pDF61 AAV2 vector encoding SEAP driven by an erythromycin responsive PETR promoter. SEAP was excised from pCF019 using NheI/ClaI and cloned into the NheI/BstBI sites of pDF54, thus resulting in pDF61 (ITR-PETR-SEAP-pASV40-ITR).

this work

pDF63 AAV2 vector containing an SV40 promoter followed by an IRESPV and a IRESEMCV element. PSV40-IRESPV-IRESEMCV was excised from pMF123 using SspI/BglII, polished with Klenow and cloned into the polished NcoI/SpeI sites of pAAV-lacZ, thus resulting in pDF63 (ITR- PSV40-IRESPV-IRESEMCV-pASV40-ITR).

this work

pDF74 AAV2 vector containing tricistronic expression cassette driven by a PETRON8 promoter. The PETRON8 promoter was excised from pWW76 using NheI/EcoRI and cloned into the corresponding sites of pDF63, thus resulting in pDF74 (ITR-PETRON8-IRESPV-IRESEMCV-pASV40-ITR).

this work

pDF75 AAV2 vector encoding dicistronic expression unit consisting of SEAP followed by an IRESPV element followed by ET1 driven by PETR. Dicistronic expression cassette was excised from pBP141 using XbaI/PacI and cloned into the NheI/PacI sites of pDF54, thus resulting in pDF75 (ITR-PETR-SEAP-IRESPV-ET1-pASV40-ITR).

this work

pDF76 AAV2 vector encoding SEAP driven by a PETRON8 promoter. SEAP was excised from pSS134 using EcoRI/HindIII and cloned into pDF37. This vector was digested using EcoRI/AscI and the SEAP containing insert was cloned into the corresponding sites of pDF74, thus resulting in pDF76 (ITR-PETRON8-SEAP-IRESEMCV-pASV40-ITR).

this work

pDF77 AAV2 vector encoding SEAP under the control of an erythromycin responsive PETR promoter (additional upstream ATG deleted). PETR was excised from pDF54 using AccI/EcoRI and cloned into the ClaI/EcoRI sites of pDF61, thus resulting in pDF77 (ITR-PETR-SEAP-pASV40-ITR)

this work

pDF89 AAV2 vector encoding divergent expression units for EYFP driven by PETR and ET1 driven by a HSP70 minimal promoter. The EYFP cassette was excised from pCF18 using AccI/EcoRV and cloned into the NruI/ClaI sites of pDF55, thus resulting in pDF89 (ITR-pAI-EYFP←PETR-ETR-PHSP70min→ET1-pASV40-ITR)

this work

pDF98 Plasmid containing hEF1α promoter flanked by multiple cloning sites unpublished pDF109 AAV2 vector encoding SEAP driven by a constitutive hCMV promoter. SEAP

was excised from pDF61 using EcoRI/SpeI and cloned into the EcoRI/XbaI sites of pAAV-MCS, thus resulting in pDF109 (ITR-PhCMV-Intronβ-globin-SEAP-pAhgh-ITR)

pDF124 AAV2 vector encoding dicistronic expression unit consisting of EYFP followed by an IRESEMCV element followed by ET1. The IRES-ET1 containing insert was excised from pDF75 using HindIII, polished with Pfu polymerase, digested using BstXI and cloned into the SwaI/BstXI sites of pDF54, thus resulting in pDF124 (ITR-PETR-EYFP-IRESEMCV-ET1-pASV40-ITR).

this work

pDF126 AAV2 vector encoding E-KRAB under the control of a constitutive hCMV promoter. The E-KRAB containing insert was excised from pWW043 using

this work

Page 63: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

62

EcoRI/HpaI and cloned into the EcoRI/HincII sites of pAAV-MCS, thus resulting in pDF126 (ITR-PhCMV-Intronβ-globin-E-KRAB-pAhgh-ITR).

pDF141 AAV2 vector encoding self-regulated expression cassette consisting of ET1 driven by a constitutive SV40 promoter and EYFP driven by PETR. The entire ET1 expression cassette of pDF56 was excised using ClaI/PmlI and cloned into pDF54 which was digested by HindIII and polished by Pfu before, thus resulting in pDF141 (ITR-PSV40-ET1-pASV40-PETR-EYFP-pASV40-ITR).

this work

pDF143 AAV2 vector encoding self-regulated expression cassette consisting of ET1 driven by a constitutive SV40 promoter and SEAP driven by PETR. The SEAP containing insert was excised from pDF77 using KpnI/SpeI and cloned into the corresponding sites of pDF141, thus resulting in pDF143 (ITR-PSV40-ET1-pASV40-PETR-SEAP-pASV40-ITR).

this work

pDF199

AAV2 vector encoding SEAP under the control of a constitutive SV40 promoter followed by 2 binding sites for the transrepressor E-KRAB. The 4*ETR binding site containing fragment was excised from pWW55 using BstBI/NdeI and cloned into the corresponding sites of pDF76. Two of the binding sites were deleted by recombination during the cloning procedure, thus resulting in pDF199 (ITR-PETRON2-SEAP-IRESEMCV-pASV40-ITR).

this work

pDF200 AAV2 vector encoding SEAP under the control of a constitutive SV40 promoter followed by 4 binding sites for the transrepressor E-KRAB. The 4*ETR binding site-containing fragment was excised from pWW55 using BstBI/NdeI and cloned into the corresponding sites of pDF76, thus resulting in pDF200 (ITR-PETRON4-SEAP-IRESEMCV-pASV40-ITR).

this work

pDF207 AAV2 vector encoding EYFP under the control of a constitutive SV40 promoter followed by 8 binding sites for the transrepressor E-KRAB. EYFP was excised from pDF34 using EcoRI/PacI and cloned into the corresponding sites of pDF76, thus resulting in pDF207 (ITR-PETRON8-EYFP-pASV40-ITR).

this work

pDF208 AAV2 vector encoding EYFP under the control of a constitutive SV40 promoter followed by 4 binding sites for the transrepressor E-KRAB. EYFP was excised from pDF34 using EcoRI/PacI and cloned into the corresponding sites of pDF200, thus resulting in pDF208 (ITR-PETRON4-EYFP-pASV40-ITR).

this work

pDF209 AAV2 vector encoding EYFP under the control of a constitutive SV40 promoter followed by 2 binding sites for the transrepressor E-KRAB. EYFP was excised from pDF34 using EcoRI/PacI and cloned into the corresponding sites of pDF199, thus resulting in pDF209 (ITR-PETRON2-EYFP-pASV40-ITR).

this work

Abbreviations: AAV2, adeno-associated virus type 2; ECFP, enhanced cyan fluorescent protein (720bp); EM,

erythromycin; ET1, erythromycin transactivator (972bp); EYFP, enhanced yellow fluorescent protein (720bp);

IRESEMCV, internal ribosome entry site of encephalomyocarditisviral origin (502bp); IRESPV, internal ribosome

entry site of polioviral origin (635bp); ITR, inverted terminal repeat (141bp); KRAB, kruppel-associated box

(450bp); pAI, artificial polyadenylation signal (91bp); pAhgh, human growth hormone polyadenylation signal

(478bp); pASV40, simian virus 40 polyadenylation signal (145bp); PhEF1α, human elongation factor 1α promoter

(1185bp); PETR, erythromycin responsive promoter (200bp); PETRON, macrolide inducible promoter (530bp);

PhCMV, human immediate early cytomegalovirus promoter (663bp); PHSP70min, heat shock protein 70 minimal

promoter (350bp); PSV40, simian virus 40 promoter (308bp); SEAP, human placental secreted alkaline

phosphatase (1560bp)

Page 64: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

63

Following transduction of pDF124-derived AAV particles, active PETR produces a

single transcript from which EYFP is translated in a classic cap-dependent manner whereas

ET1 production depends on cap-independent IRESEMCV-mediated translation initiation.

Undetectable PETR-mediated transcripts result in few initial ET1 proteins, which, in the

absence of erythromycin trigger auto-regulated high-level expression of this transactivator

along with the cocistronically encoded EYFP. In the presence of erythromycin, ET1

originating from basal PETR activity is inactivated, which interrupts the auto-regulated feed-

forward transcription and results in repression of EYFP production. After transduction of

pDF89-derived AAV particles, leaky ET1 transcript initiate an auto-regulated expression

circuit resulting in simultaneous expression of ET1 and EYFP until ET1-ETR binding is

abolished by erythromycin. All three configurations yielded AAV particles, which displayed

good regulation performance upon trandsduction of HT-1080 and MCF-7 and cultivation in

the absence or in the presence of erythromycin (Figure 2B).While using equal genomic

particle numbers the transduction efficiency varied between pDF124, pDF89 and pDF141.

Trandsuction rates of the bi-directional (pDF89) and the dicistronic expression units

(pDF124) were significantly lower compared to the two-promoter set-up (pDF141) (Fig. 2B)

which made FACS-based quantitative analysis was rather difficult (Figure 2C). However

qualitative fluorescence microscopy suggested excellent regulation performance in individual

transduced cells (Fig. 2B).

Engineered AAV-derived particles transducing tightly regulated production of secreted

proteins

Tight regulation of therapeutic transgenes remains a major challenge for

current gene therapy initiatives. In order to assess whether AAV-based transduction systems

enable delivery of tightly regulated expression of a human model glycoprotein, we

constructed pDF77 harboring a PETR-driven SEAP (human placental secreted alkaline

phosphatase) (ITR-PETR-SEAP-pASV40-ITR) (Figure 3A). Co-transduction of HT-1080 and

MCF-7 with pDF77- and pDF51- (ITR-PhCMV-ET1-pASV40-ITR) derived AAV particles

provided excellent regulation performance characterized by high-level SEAP production in

the absence of erythromycin and repressed SEAP levels in the presence of erythromycin

(Figure 3B).

Page 65: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

64

Figure 2. Self- and auto-regulated AAV type 2-based expression of fluorescent proteins. (A) Schematic

representation of an auto-regulated (pDF124), a bidirectional (pDF89) and a self-regulated (pDF141) AAV type

2-based expression unit. (B) Fluorescence micrographs of human fibrosarcoma cells (HT-1080) and a human

breast cancer cell line (MCF-7) transduced with pDF124-, pDF89- and pDF141-derived AAV particles (2000

genomic particles/cell) cultivated in the presence (+) and absence (-) of EM. (C) FACS-mediated quantification

of EYFP in HT-1080 transduced with equal amounts of viral particles (2000 genomic particles/cell) and

cultivated in the presence (+EM) and absence (-EM) of erythromycin. Abbreviations: EM, erythromycin; ET1,

erythromycin-dependent transactivator; ETR, ET1-specific operator; EYFP, enhanced yellow fluorescent

protein; IRES, internal ribosome entry site; ITR, inverted terminal repeat; pAI, synthetic polyadenylation signal;

pASV40, simian virus 40 polyadenylation signal; PETR, erythromycin-responsive promoter; PHSP70min, minimal

version of the Drosophila melanogaster heat-shock protein 70 promoter; PSV40, simian virus 40 promoter.

Page 66: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

65

Furthermore, we have designed and evaluated pDF143 (ITR-PSV40-ET1-pASV40-PETR-SEAP-

pASV40-ITR), a one-vector expression configuration which is isogenic to pDF141 but

encoding SEAP instead of EYFP. pDF143 exhibited similar SEAP regulation profiles in HT-

1080 and MCF-7 compared to co-transduction of pDF77-/pDF51-derived AAV particles

suggesting that there is no interference between ET1-driving PSV40 and ET1-specific PETR

(Figure 3A and 3B). Overall, maximum expression levels for the one-vector configuration

were even higher than for the binary setting likely because for the binary system two different

particles have to transduce a single cell which is a more rare event. Transduction of HT-1080

and MCF-7 with a constitutive control vector, pDF109 (ITR-PhCMV-intronβ-globin-SEAP-pAhgh-

ITR) showed increased SEAP production compared to fully induced pDF143-derived

particles, which might be associated with either higher promoter strenght or the β-globin

intron. AAVs engineered for EON-controlled transgene expression

Various therapeutic initiatives require transgene control systems, which trigger

heterologous target genes after administration of the regulating agent and remain repressed in

the absence of the inducer (Weber and Fussenegger, 2006). We have constructed binary EON-

controlled AAV-based expression systems consisting of pDF126 (ITR-PhCMV-Intron-β-globin-

E-KRAB-pAhgh-ITR), and pDF207 (ITR-PETRON8-EYFP-pASV40-ITR), pDF208 (ITR-

PETRON4-EYFP-pASV40-ITR) or pDF209 (ITR-PETRON2-EYFP-pASV40-ITR), which harbor

EYFP under control of different E-KRAB-specific macrolide-inducible promoters

characterized by different ETR tandem repeats placed 3’ of the constitutive PSV40 promoter (Weber et al., 2002a). Functionality of the ON-type system was validated by co-transduction

of pDF126/pDF209-derived into HT-1080 and MCF-7 and profiling of EYFP expression after

48h cultivation in the presence or absence of erythromycin (Figure 4B). FACS-based

comparison of HT-1080 co-transduced with pDF126-/pDF207-, pDF126-/pDF208- and

pDF126-/pDF209-derived AAV particles and cultivated for 48h in the presence and absence

of erythromycin revealed that although overall induction factors were almost identical among

different particle combinations, basal and maximum expression levels were a function of the

number of ETR repeats associated with PSV40 (Figures 4A and 4C).

Page 67: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

66

Figure 3. AAV type 2-based regulated expression of secreted proteins. (A) Schematic representation of

pDF77, harboring a SEAP expression cassette under the control of PETR, pDF143, a self-regulated expression

unit expressing ET1 from PSV40 and driving SEAP from PETR and pDF109, harboring a constitutive SEAP

expression unit driven by PhCMV (B) SEAP expression levels profiled 48h after (co-)transduction of transgenic

AAV particles (1000 genomic particles/cell for pDF51/77, 1000 genomic particles/cell for pDF143 and 1000

genomic particles/cell for pDF109) derived from indicated vectors and cultivated in the presence (+) or absence

(-) of EM. SEAP expression is shown in units/liter (U/l) as defined by Schlatter et al.(Schlatter et al., 2002). Abbreviations: EM, erythromycin; ET1, erythromycin transactivator; ITR, inverted terminal repeat; pASV40,

simian virus 40 polyadenylation signal; PhEF1α, human elongation factor 1α promoter; PETR, erythromycin-

responsive promoter; SEAP, human placental secreted alkaline phosphatase; PSV40, simian virus 40 promoter.

Page 68: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

67

Figure 4. AAV type 2-based erythromycin-inducible EYFP expression. (A) Schematic representation

of pDF126 harboring a PhCMV-driven E-KRAB expression unit and pDF206/207/208 encoding EYFP transcribed

by PETRON promoter derivatives containing 8 (pDF207), 4 (pDF208) and 2 (pDF209) E-KRAB-specific ETR

operator modules. (B) Fluorescence micrographs of HT-1080 and MCF-7 co-transduced with pDF209/pDF126-

derived AAV particles (1000 genomic particles/cell for pDF207/208/209 and 4000 genomic particles/cell for

pDF126) and cultivated in the presence (+EM) and absence (-EM) of erythromycin (C) FACS-mediated

quantification of EYFP expression in HT-1080. EYFP-specific FACS diagrams of HT-1080 co-transduced (1000

genomic particles/cell) by pDF126/pDF207, pDF126/pDF208 and pDF126/pDF209 were cultivated for 48h in

the presence (+) and absence (-) of EM. Abbreviations: EM, erythromycin; EYFP, enhanced yellow fluorescent

protein; ITR, inverted terminal repeat; KRAB, kruppel associated box; PETRON, erythromycin-inducible

promoter.

Page 69: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

68

Reversibility and adjustability of AAV-transduced macrolide-controlled transgene

expression

Reversibility and adjustability are key characteristics for future clinical

implementation of human-compatible transgene control modalities. In order to assess the

reversibility of macrolide-responsive transgene expression in an AAV expression

configuration we transduced HT-1080 and MCF-7 with pDF143-derived AAV particles and

cultivated (i) in the presence (+++) or absence (---) of EM over 9 days, (ii) in the presence

(++-) or absence (--+) of EM over 6 days and then incubated in reversed EM conditions for

the remaining three days and (iii) in medium whose EM status was alternated every three days

from +EM to –EM to +EM (+-+) or from –EM to +EM to –EM (-+-).SEAP accumulation was

always measured prior to any EM status switch.

HT-1080 and MCF-7 showed excellent switching characteristics upon application of

removal of erythromycin (Figure 5A and 5B)

In order to assess the dose-response characteristics of binary and one-vector-based

macrolide-responsive AAV transduction systems HT-1080 and MCF-7 were transduced with

pDF143- or co-transduced with pDF77-/pDF51-derived AAV particles and cultivated for 48h

in the presence of different EM concentrations before SEAP production was quantified.

Whereas SEAP expression was completely repressed between 20 and 100ng/ml for both

configurations gradual decrease in antibiotic treatment resulted in dose-dependent increase of

transgene expression until maximum expression levels were reached in the absence of EM

(Figure 5C and 5D). The differences in the dose-response characteristics of one-vector and

binary configurations may result from different ET1-PETR stoichiometries associated with

those technologies.

Transgenic AAV-derived particles mediate EOFF-controlled transgene expression in mice

For in-vivo validation of AAV-based macrolide-responsive transgene transduction we

injected 5*1010 pDF143-derived genomic AAV particles intramuscularly into mice and

treated them optionally with intraperitoneal erythromycin injections (2mg/mouse every 24h).

Profiling of serum SEAP levels of treated mice showed significant production of this

glycoprotein in mice, which had not received EM doses whereas SEAP production was

repressed whenever EM was administered (Figure 6).

Page 70: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

69

Figure 5. Reversibility and adjustability of macrolide-responsive SEAP expression in HT-1080 and

MCF-7 transduced by transgenic AAV type 2-derived particles. (A) HT-1080 and (B) MCF-7 were transduced

with pDF143-derived AAV particles (2000 genomic particles/cell) and six equal populations (1-6) were

cultivated in media containing different antibiotic concentrations. Cells were (i) cultivated in the presence (+++)

or absence (---) of EM over 9 days, (ii) cultivated in the presence (++-) or absence (--+) of EM over 6 days and

then incubated in reversed EM conditions for the remaining three days or (iii) cells were cultivated in medium

whose EM status was alternated every three days +EM to –EM to +EM (+-+) or from –EM to +EM to –EM (-+-)

and SEAP expression was quantified. Adjustability of SEAP expression of pDF143 (1000 genomic

particles/cell) (C) and pDF51/77 (500 genomic particles/cell) (D) in HT-1080 and MCF-7 cultivated for 48h in

the presence of increasing EM concentrations. SEAP expression is shown in units/liter (U/l) as defined by

Schlatter et al.(Schlatter et al., 2002) . Abbreviations: EM, erythromycin; SEAP, human placental secreted

alkaline phosphatase.

Page 71: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

70

Figure 6. Macrolide-triggered SEAP expression in mice injected with pDF143 (ITR-PSV40-ET1-pASV40-

PETR-SEAP-pASV40-ITR)-derived AAV type 2 particles. pDF143-derived AAV particles were administered

intramuscularly and SEAP levels in the serum were measured at two different time points for one group in the

absence of erythromycin, and for the other group with EM injections every 24h. SEAP expression is shown in

milliunits/liter (mU/l) as defined by Schlatter et al. (Schlatter et al., 2002). Abbreviations: EM, erythromycin;

SEAP, human placental secreted alkaline phosphatase.

Discussion In recent years, gene delivery systems based on adeno-associated viral vectors have

become one of the most promising tools for delivering transgenes in vivo. After major

setbacks in trials with retroviral as well as adenoviral vectors (Check, 2002; Lehrman, 1999), AAVs present a strong alternative for the safe and efficient delivery of transgenes. We have

pioneered the delivery and regulation of transgenes by integrating the recently developed

erythromycin-controllable E.REX system into AAV type 2-derived backbones. Considering

that erythromycin is a clinically licensed drug and that AAV vectors have an excellent safety

profile record, such a system is suitable for in vivo applications and is promising for clinical

initiatives. Maximum erythromycin doses of up to 4g/day in adults are in accordance with

FDA and AHFS (American Society of Health-System Pharmacists) guidlines.

(WWW.FDA.gov; WWW.AHFSdruginformation.com). These doses are within the range of

Page 72: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

71

the amounts we have tested in mice. We are therefore confident that the AAV-encoded and

erythromycin-controlled transgenes will be compatible with future clinical applications.

Research, in which AAV vectors deliver adjustable transgenes, has been conducted

mainly with tetracycline responsive systems, which trigger or repress expression upon the

addition of tetracycline (Chtarto et al., 2003; Gafni et al., 2004; Haberman et al., 1998; Jiang

et al., 2004; Stieger et al., 2006).

We have designed a range of AAV type 2-based vectors encoding different

configurations of the EOFF expression system to evaluate optimal arrangements for further in

vivo studies. A binary set-up, where the transactivator is delivered on one vector and the gene

of interest is driven by PETR on another vector, worked well in vitro. The expression levels,

vector titers and regulation performance of the tested configurations were excellent, indicating

an efficient co-transduction of the two types of transgenic AAV particles. Regardless of

whether intracellular or secreted transgenes were expressed, the regulation factors of all the

tested cell lines were between 10 and 400. Although transgene expression mediated by AAVs

engineered for macrolide-responsive transgenes expression were typically lower after

induction compared to isogenic AAV derivatives containing constitutive promoters the

overall production levels were in the same order of magnitude in vitro. Isogenic EON systems

also enabled excellent regulation performance but overall induction factors were typically

lower, since cells exclusively transduced with the transgene-encoding AAV particle exhibit

constitutive expression. Therefore, to achieve optimized EON-controlled transgene expression

from transgenic AAVs arranged in a binary vector set, the transrepressor-encoding AAV has

to be administered in excess compared to the transgene-encoding counterpart or cell lines

have to be used, which are particular susceptible to AAV-based transduction.

Generally, even though binary systems have the disadvantage that two vectors must

enter the same cell to obtain regulated expression, this design is advantageous for some

applications, namely when delivering large transgenes. Although binary systems are powerful

tools, in a therapeutic setting it may be desirable to combine the regulation modules on a

single vector. The advantages of such a setting are: (i) only one virus must enter the target cell

to obtain regulated expression of the transgene, (ii) exact virus titration is easier and (iii)

lower virus doses are required for comparable transgene expression.

We have pioneered both self-regulated and auto-regulated macrolide-responsive

recombinant adeno-associated viral vectors for tightly controlled transgene expression.

Although auto-regulated expression units are more compact than self-regulated configurations

we had difficulty achieving reasonably high transduction efficiencies and expression levels.

Page 73: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

72

Since qRT-PCR-based analysis of viral particles revealed identical number of encapsidated

genomes among our AAV portfolio, the lower transduction rates of auto-regulated AAVs may

result from their genetic configuration rather than virus assembly. In contrast, self-regulated

expression units, consisting of a strong constitutive promoter driving the expression of the

transactivator as well as a macrolide-responsive promoter driving the desired transgene on the

same vector, showed excellent performance in a variety of cell types. The vector design using

compact promoter and polyadenylation elements in a self-regulated configuration allows the

integration of a transgene with a size of up to 2500bp which is sufficient for various clinically

important transgenes (for example, erythropoietin or vascular endothelial growth factor). We

did not observe promoter interference in the tested cell types, although the strong constitutive

PSV40 promoter driving ET1 and the erythromycin-responsive promoter were on the same

construct. We were successful in expressing intracellular as well as secreted transgenes in a

highly controlled manner and showed precise titration of transgene expression upon addition

of varying erythromycin concentrations as well as reversibility upon the addition or removal

of the regulating agent. After switching from a fully repressed to a fully induced state or vice

versa a short lag phase was observed before the new expression state was reached. This

behaviour might be due to trace amounts of antibiotic remaining in the culture (by switching

from +EM to –EM) or due to the degradation kinetics of transcripts and transactivators (from

–EM to +EM)

One-vector configurations would be suitable for in vivo studies and would enable

efficient targeting of a wide variety of cells by systemic administration or by local injection of

the viral particles encoding the therapeutic transgene. We exemplified the functionality of the

system in vivo by administrating pDF143-derived AAV particles intramuscularly to mice and

analyzing transgene expression in the presence and absence of antibiotic for several days.

Most current studies on regulated transgene expression are carried out using retroviral

vectors (Markusic et al., 2005; Mitta et al., 2005; Mitta et al., 2004; Pluta et al., 2005; Vigna

et al., 2005). Although these systems are powerful and allow the efficient long-term

expression of transgenes, they have several drawbacks, such as (i) the necessity of retroviral

vectors for integration into transcriptionally active regions, which may interfere with desired

transgene control, (ii) random integration, possibly triggering oncogene activation and (iii)

silencing of the integrated transgene upon integration into the host chromosome. Adeno-

associated viral vectors on the contrary, remain mainly episomal after entering the target cell

and, therefore, do not have aforementioned drawbacks, thus providing a powerful alternative

for upcoming gene-therapy studies.

Page 74: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

73

Conclusions We have designed an array of novel AAV type 2-based expression vectors, which enable safe

and efficient transduction of mammalian cells for macrolide-adjustable transgene expression.

We have pioneered binary ON- and OFF-type systems as well as compact bidirectional, auto-

regulated and self-regulated one vector expression configurations for regulation of

intracellular as well as secreted proteins in mammalian cells. We have also engineered a

compact self-regulated AAV which demonstrated efficient transduction, expression and

regulation of a reporter gene in mice.

Methods Plasmid construction. Table 1 lists all the plasmids used in this study as well as

detailed information on their construction and the length of the genetic elements.

Cell culture and transfection. Human embryonic kidney cells, transgenic for the

adenovirus type 5-derived E1 region as well as for the simian virus 40 (SV40) large T-antigen

(HEK293-T; (Mitta et al., 2002)), human fibrosarcoma cells (HT-1080; ATCC CCL-121),

human breast cancer cells (MCF-7, ATCC HTB-22) and normal human dermal fibroblasts

(NHDF; PromoCell, Heidelberg, Germany; cat. no. C-12300, lot no. 1070402) were

cultivated in Dulbecco’s modified Eagle’s medium (DMEM, Invitrogen, Carlsbad, CA, USA)

supplemented with 10% fetal calf serum (FCS; PAN Biotech GmbH, Aidenbach, Germany;

cat. no. 3302-P231902, lot no. P231902) and 1% penicillin/streptomycin solution (Sigma

Chemicals, St. Louis, MO, USA). Chinese hamster ovary cells (CHO-K1; ATCC CCL-61)

were cultivated in ChoMaster® HTS medium (Cell Culture Technologies GmbH, Gravesano,

Switzerland) supplemented with 5% FCS (PAN Biotech GmbH) and 1%

penicillin/streptomycin solution. All cell lines were cultivated at 37°C in a 5% CO2-

containing humid atmosphere.

Production of adeno-associated viral particles. AAV particles were produced by co-

transfection of the helper plasmid pDG (Grimm et al., 2003) and the transgene-encoding

AAV vector into HEK293-T using optimized calcium phosphate transfection protocols. In

brief, 2x106 HEK293-T were seeded per culture dish (diameter 10cm) and cultivated

overnight. For each plate 15!g of helper plasmid were mixed with 5µg AAV vector and

CaCl2 was added to a final concentration of 0.25M. The mixture was added slowly to an equal

Page 75: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

74

volume of HEPES-buffered saline solution (HeBS; 50mM HEPES, 280mM NaCl, 1.5mM

Na2HPO4, pH 7.1), vortexed briefly, and incubated for 2min. before adding the DNA-calcium

phosphate precipitate solution to the monolayer culture. Precipitates were removed after 6h,

cells were supplemented with 1% FCS-supplemented DMEM and incubated for 60h. The

supernatant was discarded, the cells were detached using a cell scraper and resuspended in

2ml PBS (138mM NaCl, 8.1mM Na2HPO4, 1.47mM KH2PO4, 2.67mM KCl; Invitrogen, cat.

no. 21600-069, lot. no. 1255481) per plate. Each cell suspension was transferred to a Falcon

tube and pelleted by centrifugation for 3min. at 280xg. Following another washing step using

5ml PBS, the cells were resuspended in 2ml PBS and the intracellular viral particles were

released after cell lysis induced by three consecutive freeze-thaw cycles consisting of

shuttling the tubes between liquid nitrogen and a 37°C water bath (tubes were vortexed

vigorously after each thawing step). The cell debris was eliminated by centrifugation for

5min. at 10’000xg and the supernatant containing the crude virus stock was collected,

supplemented with 50U/ml of Benzonase (Sigma, cat. no. E1014) and incubated for 30min at

37°C. Iodixanol density-gradient purification of viral particles was performed using a

protocol adapted from Zolotukhin et al. (Zolotukhin et al., 1999). In brief, crude viral stocks

were diluted in PBS to a final volume of 12ml. Iodixanol step gradients were prepared in an

ultracentrifuge tube by sequential underlaying of the crude viral preparation with 5ml of a

15% (plus 1M NaCl in the first layer), 5ml of a 25%, 4ml of a 40% and 4ml of a 60%

iodixanol-containing PBS. For better distinction of the gradient layers 2.5µl/ml of a 0.5%

Phenol Red (Sigma, cat. no. P0290) stock solution was added to the 60% and the 25% layers.

Step gradients were centrifuged for 3.5h at 150’000xg at 18°C. The clear 40% fraction was

harvested after puncturing the ultracentrifuge tube on the side with a syringe equipped with a

16G needle. A heparin affinity column (HiTrap Heparin HP, Amersham Biosciences,

Sweden; cat. no. 17-0406-01) was equilibrated with 10 bed volumes of binding buffer (10mM

Na2PO4, pH 7) and run at a flow rate of 1ml/min. The AAV particle-containing 40% fraction

harvested from the step gradient was loaded onto the heparin affinity column. After washing

the column with 10 bed volumes of binding buffer the viral particles were eluted with 5 bed

volumes of elution buffer (10mM Na2PO4, 1M NaCl, pH 7) and then concentrated using a

spin column with a molecular weight cut-off (MWCO) of 30 kDa (Vivaspin, Vivascience,

Germany; cat. no. VS1521).

Page 76: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

75

Virus titration by quantitative real-time PCR. Crude viral preparations were treated

as described in (Clark et al., 1999). Primers and the Taqman probe were designed to anneal to

(i) the SV40 polyadenylation signal (pA) (forward primer, 5’-

AGCAATAGCATCACAAATTTCACAA-3’, reverse primer, 5’-

GACATGATAAGATACATTGATGAGTTTGG-3’, Taqman FAM/TAMRA probe, 5’-

AGCATTTTTTTCACTGCATTCTAGTTGTGGTTTG-3’), (ii) the SEAP open reading

frame (forward primer, 5’-AGGCCCGGGACAGGAA-3’, reverse primer, 5"-

GCCGTCCTTGAGCACATAGC-3") or (iii) the erythromycin-dependent transactivator open

reading frame (forward primer, 5’-CCAACTCCTCCAGGCACA-3’, reverse primer, 5’-

AGCAGGCCCTCGATGGTA-3’). Absolute quantification was performed using Taqman

Universal PCR Master Mix (Applied Biosystems, Warrington, UK, cat. no. 4324018) or

Power SYBR Green PCR Master Mix (Applied Biosystems, Warrington, UK, cat. no.

4367659) on AB Prism 7500 RT-PCR quantitative PCR hardware according to the

manufacturer’s instructions (Applied Biosystems, Weiterstadt, Germany). The reference

standard consisted of a four log-spanning dilution of pDF60 harboring a single SV40 pA

sequence, pDF51 harboring the ET1 transactivator or pDF109 harboring the SEAP open

reading frame.

Quantification of reporter gene expression. Enhanced yellow fluorescent protein

(EYFP) was visualized using a Leica DM-RB fluorescence microscope (Leica Inc.,

Heerbrugg, Switzerland) equipped with an XF114 filter (Omega Optical Inc., Brattleboro,

VT, USA). Fluorescence was quantified 48h after transduction using a fluorescence-activated

cell sorter (Coulter FC500, Beckman Coulter Inc., FL, USA) with CXP software (Beckman

Coulter) installed. SEAP production was quantified in cell-culture supernatants 48h after

transduction and in mouse serum 3 days to 7 days after injection as described in (Schlatter et

al., 2002).

Chemicals used for transgene regulation. For all in-vitro experiments, erythromycin

(Fluka, Buchs, Switzerland cat. no. E-5389) was prepared as a stock solution of 1mg/ml in

ethanol and used at a final concentration of 1!g/ml. For in-vivo studies, 200!l of a 10mg/ml

erythromycin solution (10% ethanol, 90% PBS) were daily intraperitoneally injected into

each animal.

In vivo studies. Female OF1 (oncins france souche 1) mice were obtained from

Charles River Laboratories (Lyon, France). Mice were treated with intramuscular injections

5x1010 vector genomes/mouse. Erythromycin was administered intraperitoneally 1h after

injection of the transgenic AAV particles and repeated every 24h. Blood samples were

Page 77: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

76

collected retroorbitally and serum was produced using microtainer SST tubes (Beckton

Dickinson, Plymouth, UK). All animal experiments were approved by the French Ministry of

Agriculture and Fishery and performed by M.D.-E. at the Institut Universitaire de

Technologie, IUTA, F-69622 Villeurbanne Cedex, France.

Authors’ contributions DAF designed and performed all experiments except the animal studies which were

conducted by MDE. MF participated in the conception and design of the study and helped to

draft the manuscript.

Acknowledgements We thank Marcia Schoenberg for critical comments on the manuscript. This work was

supported by the Swiss National Science Foundation (grant no. 3100A0-112549) and

the Swiss State Secretariat for Education and Research within EC Framework 6.

References

Acland, G.M., Aguirre, G.D., Bennett, J., Aleman, T.S., Cideciyan, A.V., Bennicelli, J.,

Dejneka, N.S., Pearce-Kelling, S.E., Maguire, A.M., Palczewski, K., et al. (2005). Long-

Term Restoration of Rod and Cone Vision by Single Dose rAAV-Mediated Gene Transfer to

the Retina in a Canine Model of Childhood Blindness. Mol Ther.

Beerli, R.R., Schopfer, U., Dreier, B., and Barbas, C.F., 3rd (2000). Chemically regulated

zinc finger transcription factors. J Biol Chem 275, 32617-32627.

Bohl, D., Salvetti, A., Moullier, P., and Heard, J.M. (1998). Control of erythropoietin delivery

by doxycycline in mice after intramuscular injection of adeno-associated vector. Blood 92,

1512-1517.

Boorsma, M., Nieba, L., Koller, D., Bachmann, M.F., Bailey, J.E., and Renner, W.A. (2000).

A temperature-regulated replicon-based DNA expression system. Nat Biotechnol 18, 429-

432.

Braselmann, S., Graninger, P., and Busslinger, M. (1993). A selective transcriptional

induction system for mammalian cells based on Gal4-estrogen receptor fusion proteins. Proc

Natl Acad Sci U S A 90, 1657-1661.

Page 78: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

77

Check, E. (2002). Regulators split on gene therapy as patient shows signs of cancer. Nature

419, 545-546.

Chtarto, A., Bender, H.U., Hanemann, C.O., Kemp, T., Lehtonen, E., Levivier, M., Brotchi,

J., Velu, T., and Tenenbaum, L. (2003). Tetracycline-inducible transgene expression mediated

by a single AAV vector. Gene Ther 10, 84-94.

Clark, K.R., Liu, X., McGrath, J.P., and Johnson, P.R. (1999). Highly purified recombinant

adeno-associated virus vectors are biologically active and free of detectable helper and wild-

type viruses. Hum Gene Ther 10, 1031-1039.

Edelstein, M.L., Abedi, M.R., Wixon, J., and Edelstein, R.M. (2004). Gene therapy clinical

trials worldwide 1989-2004-an overview. J Gene Med 6, 597-602.

Flotte, T., Carter, B., Conrad, C., Guggino, W., Reynolds, T., Rosenstein, B., Taylor, G.,

Walden, S., and Wetzel, R. (1996). A phase I study of an adeno-associated virus-CFTR gene

vector in adult CF patients with mild lung disease. Hum Gene Ther 7, 1145-1159.

Flotte, T.R., Solow, R., Owens, R.A., Afione, S., Zeitlin, P.L., and Carter, B.J. (1992). Gene

expression from adeno-associated virus vectors in airway epithelial cells. Am J Respir Cell

Mol Biol 7, 349-356.

Fussenegger, M., Mazur, X., and Bailey, J.E. (1998). pTRIDENT, a novel vector family for

tricistronic gene expression in mammalian cells. Biotechnol Bioeng 57, 1-10.

Fussenegger, M., Morris, R.P., Fux, C., Rimann, M., von Stockar, B., Thompson, C.J., and

Bailey, J.E. (2000). Streptogramin-based gene regulation systems for mammalian cells. Nat

Biotechnol 18, 1203-1208.

Fux, C., and Fussenegger, M. (2003). Toward higher order control modalities in mammalian

cells-independent adjustment of two different gene activities. Biotechnol Prog 19, 109-120.

Fux, C., Langer, D., and Fussenegger, M. (2004). Dual-regulated myoD- and msx1-based

interventions in C2C12-derived cells enable precise myogenic/osteogenic/adipogenic lineage

control. J Gene Med 6, 1159-1169.

Fux, C., Weber, W., Daoud-El Baba, M., Heinzen, C., Aubel, D., and Fussenegger, M.

(2003). Novel macrolide-adjustable bidirectional expression modules for coordinated

expression of two different transgenes in mice. J Gene Med 5, 1067-1079.

Gafni, Y., Pelled, G., Zilberman, Y., Turgeman, G., Apparailly, F., Yotvat, H., Galun, E.,

Gazit, Z., Jorgensen, C., and Gazit, D. (2004). Gene therapy platform for bone regeneration

using an exogenously regulated, AAV-2-based gene expression system. Mol Ther 9, 587-595.

Page 79: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

78

Gossen, M., and Bujard, H. (1992). Tight control of gene expression in mammalian cells by

tetracycline-responsive promoters. Proc Natl Acad Sci U S A 89, 5547-5551.

Gossen, M., Freundlieb, S., Bender, G., Muller, G., Hillen, W., and Bujard, H. (1995).

Transcriptional activation by tetracyclines in mammalian cells. Science 268, 1766-1769.

Grimm, D., and Kay, M.A. (2003). From virus evolution to vector revolution: use of naturally

occurring serotypes of adeno-associated virus (AAV) as novel vectors for human gene

therapy. Curr Gene Ther 3, 281-304.

Grimm, D., Kay, M.A., and Kleinschmidt, J.A. (2003). Helper virus-free, optically

controllable, and two-plasmid-based production of adeno-associated virus vectors of

serotypes 1 to 6. Mol Ther 7, 839-850.

Grimm, D., Kern, A., Rittner, K., and Kleinschmidt, J.A. (1998). Novel tools for production

and purification of recombinant adenoassociated virus vectors. Hum Gene Ther 9, 2745-2760.

Haberman, R.P., McCown, T.J., and Samulski, R.J. (1998). Inducible long-term gene

expression in brain with adeno-associated virus gene transfer. Gene Ther 5, 1604-1611.

Haberman, R.P., Samulski, R.J., and McCown, T.J. (2003). Attenuation of seizures and

neuronal death by adeno-associated virus vector galanin expression and secretion. Nat Med 9,

1076-1080.

Jiang, L., Rampalli, S., George, D., Press, C., Bremer, E.G., O'Gorman, M.R., and Bohn,

M.C. (2004). Tight regulation from a single tet-off rAAV vector as demonstrated by flow

cytometry and quantitative, real-time PCR. Gene Ther 11, 1057-1067.

Kaplitt, M.G., Leone, P., Samulski, R.J., Xiao, X., Pfaff, D.W., O'Malley, K.L., and During,

M.J. (1994). Long-term gene expression and phenotypic correction using adeno-associated

virus vectors in the mammalian brain. Nat Genet 8, 148-154.

Lehrman, S. (1999). Virus treatment questioned after gene therapy death. Nature 401, 517-

518.

Li, C., Bowles, D.E., van Dyke, T., and Samulski, R.J. (2005). Adeno-associated virus

vectors: potential applications for cancer gene therapy. Cancer Gene Ther.

Markusic, D., Oude-Elferink, R., Das, A.T., Berkhout, B., and Seppen, J. (2005). Comparison

of single regulated lentiviral vectors with rtTA expression driven by an autoregulatory loop or

a constitutive promoter. Nucleic Acids Res 33, e63.

Mitta, B., Rimann, M., Ehrengruber, M.U., Ehrbar, M., Djonov, V., Kelm, J., and

Fussenegger, M. (2002). Advanced modular self-inactivating lentiviral expression vectors for

Page 80: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

79

multigene interventions in mammalian cells and in vivo transduction. Nucleic Acids Res 30,

e113.

Mitta, B., Weber, C.C., and Fussenegger, M. (2005). In vivo transduction of HIV-1-derived

lentiviral particles engineered for macrolide-adjustable transgene expression. J Gene Med.

Mitta, B., Weber, C.C., Rimann, M., and Fussenegger, M. (2004). Design and in vivo

characterization of self-inactivating human and non-human lentiviral expression vectors

engineered for streptogramin-adjustable transgene expression. Nucleic Acids Res 32, e106.

Nakai, H., Yant, S.R., Storm, T.A., Fuess, S., Meuse, L., and Kay, M.A. (2001).

Extrachromosomal recombinant adeno-associated virus vector genomes are primarily

responsible for stable liver transduction in vivo. J Virol 75, 6969-6976.

Neddermann, P., Gargioli, C., Muraglia, E., Sambucini, S., Bonelli, F., De Francesco, R., and

Cortese, R. (2003). A novel, inducible, eukaryotic gene expression system based on the

quorum-sensing transcription factor TraR. EMBO Rep 4, 159-165.

No, D., Yao, T.P., and Evans, R.M. (1996). Ecdysone-inducible gene expression in

mammalian cells and transgenic mice. Proc Natl Acad Sci U S A 93, 3346-3351.

Pluta, K., Luce, M.J., Bao, L., Agha-Mohammadi, S., and Reiser, J. (2005). Tight control of

transgene expression by lentivirus vectors containing second-generation tetracycline-

responsive promoters. J Gene Med 7, 803-817.

Rivera, V.M., Clackson, T., Natesan, S., Pollock, R., Amara, J.F., Keenan, T., Magari, S.R.,

Phillips, T., Courage, N.L., Cerasoli, F., Jr., et al. (1996). A humanized system for

pharmacologic control of gene expression. Nat Med 2, 1028-1032.

Rivera, V.M., Gao, G.P., Grant, R.L., Schnell, M.A., Zoltick, P.W., Rozamus, L.W.,

Clackson, T., and Wilson, J.M. (2005). Long-term pharmacologically regulated expression of

erythropoietin in primates following AAV-mediated gene transfer. Blood 105, 1424-1430.

Schlatter, S., Rimann, M., Kelm, J., and Fussenegger, M. (2002). SAMY, a novel mammalian

reporter gene derived from Bacillus stearothermophilus alpha-amylase. Gene 282, 19-31.

Stieger, K., Le Meur, G., Lasne, F., Weber, M., Deschamps, J.Y., Nivard, D., Mendes-

Madeira, A., Provost, N., Martin, L., Moullier, P., et al. (2006). Long-term doxycycline-

regulated transgene expression in the retina of nonhuman primates following subretinal

injection of recombinant AAV vectors. Mol Ther.

Tomanin, R., and Scarpa, M. (2004). Why do we need new gene therapy viral vectors?

Characteristics, limitations and future perspectives of viral vector transduction. Curr Gene

Ther 4, 357-372.

Page 81: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

80

Vandendriessche, T.C. (2002). Clinical gene therapy - first international conference. 24-26

January 2002, Groningen, the Netherlands. IDrugs 5, 209-212.

Vigna, E., Amendola, M., Benedicenti, F., Simmons, A.D., Follenzi, A., and Naldini, L.

(2005). Efficient Tet-dependent expression of human factor IX in vivo by a new self-

regulating lentiviral vector. Mol Ther 11, 763-775.

Wang, B., Li, J., and Xiao, X. (2000). Adeno-associated virus vector carrying human

minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proc

Natl Acad Sci U S A 97, 13714-13719.

Wang, J., Voutetakis, A., Papa, M., Rivera, V.M., Clackson, T., Lodde, B.M., Mineshiba, F.,

and Baum, B.J. (2005). Rapamycin control of transgene expression from a single AAV vector

in mouse salivary glands. Gene Ther.

Weber, W., and Fussenegger, M. (2006). Pharmacologic transgene control systems for gene

therapy. J Gene Med.

Weber, W., Fux, C., Daoud-el Baba, M., Keller, B., Weber, C.C., Kramer, B.P., Heinzen, C.,

Aubel, D., Bailey, J.E., and Fussenegger, M. (2002a). Macrolide-based transgene control in

mammalian cells and mice. Nat Biotechnol 20, 901-907.

Weber, W., Marty, R.R., Keller, B., Rimann, M., Kramer, B.P., and Fussenegger, M. (2002b).

Versatile macrolide-responsive mammalian expression vectors for multiregulated multigene

metabolic engineering. Biotechnol Bioeng 80, 691-705.

Weber, W., Marty, R.R., Link, N., Ehrbar, M., Keller, B., Weber, C.C., Zisch, A.H., Heinzen,

C., Djonov, V., and Fussenegger, M. (2003). Conditional human VEGF-mediated

vascularization in chicken embryos using a novel temperature-inducible gene regulation

(TIGR) system. Nucleic Acids Res 31, e69.

Weber, W., Rimann, M., Spielmann, M., Keller, B., Daoud-El Baba, M., Aubel, D., Weber,

C.C., and Fussenegger, M. (2004). Gas-inducible transgene expression in mammalian cells

and mice. Nat Biotechnol 22, 1440-1444.

Xiao, X., Li, J., and Samulski, R.J. (1998). Production of high-titer recombinant adeno-

associated virus vectors in the absence of helper adenovirus. J Virol 72, 2224-2232.

Xu, A., Song, L., Wang, C., Wang, A., Xu, Q., Xiao, Z., Wang, S., Li, M., Hao, S., and Li, Z.

(2000). [An observation of the immuno-persistence after inoculating with the domestic BRD

II strain rubella vaccine among infants and young children]. Zhonghua Liu Xing Bing Xue Za

Zhi 21, 117-120.

Page 82: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 2

81

Zhao, H.F., Boyd, J., Jolicoeur, N., and Shen, S.H. (2003). A coumermycin/novobiocin-

regulated gene expression system. Hum Gene Ther 14, 1619-1629.

Zolotukhin, S., Byrne, B.J., Mason, E., Zolotukhin, I., Potter, M., Chesnut, K., Summerford,

C., Samulski, R.J., and Muzyczka, N. (1999). Recombinant adeno-associated virus

purification using novel methods improves infectious titer and yield. Gene Ther 6, 973-985.

Page 83: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

82

CHAPTER 3

InXy and SeXy, Compact Heterologous Reporter Proteins

For Mammalian Cells

David A. Fluri, Jens M. Kelm, Guillaume Lesage, Marie Daoud-El Baba

and Martin Fussenegger (2007).

Biotechnology and Bioengineering, 98(3): 655-667

Page 84: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

83

Abstract Mammalian reporter proteins are essential for gene-function analysis, drugscreening

initiatives and as model product proteins for biopharmaceutical manufacturing. Bacillus

subtilis can maintain its metabolism by secreting Xylanase A (XynA), which converts xylan

into shorter xylose oligosaccharides. XynA is a family 11 xylanase monospecific for D-xylose

containing substrates. Mammalian cells transgenic for constitutive expression of wild-type

xynA showed substantial secretion of this prokaryotic enzyme. Deletion analysis confirmed

that a prokaryotic signal sequence encoded within the first 82 nucleotides was compatible

with the secretory pathway of mammalian cells. Codon optimization combined with

elimination of the prokaryotic signal sequence resulted in an exclusively intracellular

mammalian Xylanase A variant (InXy) while replacement by an immunoglobulin-derived

secretion signal created an optimal secreted Xylanase A derivative (SeXy). A variety of

chromogenic and fluorescence-based assays adapted for use with mammalian cells detected

InXy and SeXy with high sensitivity and showed that both reporter proteins resisted repeated

freeze/thaw cycles, remained active over wide temperature and pH ranges, were extremely

stable in human serum stored at room temperature and could independently be quantified in

samples also containing other prominent reporter proteins such as the human placental

alkaline phosphatase (SEAP) and the Bacillus stearothermophilus-derived secreted α-amylase

(SAMY). Glycoprofiling revealed that SeXy produced in mammalian cells was N-

glycosylated at four different sites, mutation of which resulted in impaired secretion. SeXy

was successfully expressed in a variety of mammalian cell lines and primary cells following

transient transfection and transduction with adeno-associated virus particles (AAV)

engineered for constitutive SeXy expression. Intramuscular injection of transgenic AAVs into

mice showed significant SeXy levels in the bloodstream. InXy and SeXy are highly sensitive,

compact and robust reporter proteins, fully compatible with pre-existing marker genes and

can be assayed in high-throughput formats using very small sample volumes.

Abbreviations: AAV, adeno-associated virus; BHK, baby hamster kidney cells; CHO-K1, chinese

hamster ovary cells; Cy3, cyanine 3; Cy5, cyanine 5; HEK293-T, human embryonic kidney

cells transgenic for simian virus 40 large T antigen; His-Tag, hexa-histidine tag; HT-1080,

Page 85: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

84

human sarcoma cell line; InXy, intracellular xylanase A; NHDF, normal human dermal

fibroblasts; SAMY, Bacillus stearothermophilus-derived secreted α-amylase; SEAP, human

placental secreted alkaline phosphatase; SeXy, secreted xylanase A; SSIgk, signal sequence

derived from the murine Igκ-chain V-12-C region; SSIL-2, signal sequence derived from the

human interleukin 2 gene; XynA, Bacillus subtilis xylanase A

Introduction Secreted mammalian reporter proteins have been used extensively for functional

genomic research (Kramer and Fussenegger, 2005), as marker genes in vivo (Mitta et al.,

2005), for prototype gene therapy scenarios (Liu et al., 2004; Weber et al., 2004) as well as

for high-throughput screening initiatives (Durocher et al., 2000; Lee et al., 2004). A variety of

intracellular reporter proteins are available for use in mammalian cells such as (i) specific

enzymes (β-galactosidase, thymidine kinase or chloramphenicol acetyltransferase) (An et al.,

1982; Gorman et al., 1982; Mannhaupt et al., 1988; Searle et al., 1985), (ii) fluorescent

reporter proteins represented by the pioneering green fluorescent protein (e.g. GFP, YFP or

RFP) (Hadjantonakis et al., 2003; Yang et al., 2000), or (iii) bioluminescent proteins of the

luciferase type (de Wet et al., 1985; Lorenz et al., 1991). While secreted reporter proteins are

(i) easily accessible in the cell culture supernatant, (ii) do not require complex cell lysis and

sample preparations, (iii) enable simple profiling of expression kinetics using a single culture

and (iv) could serve as model product proteins for generic biopharmaceutical manufacturing

scenarios, only a few secreted reporter genes are available including the human placental

secreted alkaline phosphatase (SEAP) (Berger et al., 1988), secreted luciferase (Markova et

al., 2004) and the Bacillus stearothermophilus-derived secreted α-amylase (Schlatter et al.,

2002). SEAP and SAMY reporter proteins have functional endogenous homologs for which

phosphatase and α-amylase activities must be inactivated before the heterologous marker

protein can be reliably quantified (Berger et al., 1988).

Bacillus subtilis is able to metabolize the carbohydrate xylan, which is found

associated with cellulose in plant cell walls (Roncero, 1983). Xylan is a polymer consisting of

a β-1,4-linked xylose backbone with branches formed by xylose, other pentoses, hexoses, and

uronic acid. Xylanases have been expressed and used as reporter proteins in bacteria and

plants (Kamionka et al., 2004; Vickers et al., 2003). Bacterial family 10 xylanases (displaying

activity on both, cellulose and hemi- cellulose) as well as true cellulases have been previously

expressed in mammalian cells in order to improve nutritional performance of transgenic

Page 86: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

85

animals (Fontes et al., 1999; Soole et al., 1993; Zhang et al., 1997; Zhang et al., 1999).We

have engineered B. subtilis xylanase A, a family 11 xylanase, which, in contrast to family 10

xylanases, exclusively catabolizes D-xylose-containing substrates for optimal intracellular

production as well as for high-level secretion in mammalian cells. The modified xylanases

produced by different mammalian cell lines and primary cells were extremely stable and

could be quantified using a variety of chromogenic and fluorescent assays. Even after

intramuscular transduction using adeno-associated virus particles, secreted xylanase could be

detected in the bloodstream of mice.

Plasmid Description and Cloning Strategy Reference or source

pEF4-MycHis

Mammalian expression vector containing PhEF1α Invitrogen

pSEAP2-control

Expression plasmid encoding SEAP Clontech

pSS158 Expression plasmid encoding SAMY Schlatter et al. 2002

pDF38 AAV expression vector harboring PhCMV unpublished pDF149 Expression plasmid encoding full length B. subtilis xylanase A

(XynA) driven by PhEF1α. XynA was amplified from genomic B. subtilis DNA (strain 168) using ODF42/ODF43 and digested with BamHI/XbaI before cloning the fragment into the corresponding sites of pEF4-MycHis in-frame with C-terminal Myc- and His-tags resulting in pDF149 (PhEF1α-xynA-pABGH).

This work

pDF163 Expression plasmid encoding a XynA variant (XynAM), harboring several codon modifications at its N-terminus for optimized translation in mammalian cells, driven by PhEF1α. XynAM was amplified from pDF149 using ODF53/ODF43 digested with EcoRI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF163 (PhEF1α-xynAM-pABGH).

This work

pDF164 Expression plasmid encoding XynAM fused to Igκ secretion signal (METDTLLLWVLLLWVPGSTGD, Coloma et al., 1992), derived from the murine Igκ-chain V-J2-C region, driven by PhEF1α. SSIgκ-xynAM was amplified from pDF163 using ODF41/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF164 (PhEF1α-SSIgk-xynAM-pABGH).

This work

Page 87: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

86

Plasmid Description and Cloning Strategy Reference or source

pDF184 Expression plasmid encoding N-terminally truncated form of XynAM (xynAM!1-27) driven by PhEF1α. XynAM∆1-27 was amplified from pDF163 using ODF56/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF184 (PhEF1α-xynAM∆1-27-pABGH)

This work

pDF186 Expression plasmid encoding N-terminally truncated form of XynAM (xynAM!1-87) driven by PhEF1α. XynAM∆1-87 was amplified from pDF163 using ODF58/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF186 (PhEF1α-xynAM∆1-87-pABGH)

This work

pDF187 Expression plasmid encoding N-terminally truncated form of XynAM (xynAM!1-115) driven by PhEF1α. XynAM∆1-115 was amplified from pDF163 using ODF59/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF187 (PhEF1α-xynAM∆1-115-pABGH)

This work

pDF188 Expression plasmid encoding N-terminally truncated form of XynAM (xynAM!1-139) driven by PhEF1α. XynAM∆1-139 was amplified from pDF163 using ODF60/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF188 (PhEF1α-xynAM∆1-139-pABGH)

This work

pDF189 AAV2 vector encoding intracellular xylanase (InXy) fused to human IL-2 secretion signal (MYRMQLLSCIALSLALVTNS, Sasada et al. 1998), driven by PhCMV. SSIL-2-InXy was excised from pDF190 with BamHI/PmeI and cloned without Myc- and His tags into the BamHI/EcoRV sites of pDF38 resulting in pDF189 (ITR-PhCMV-Intronβ-globin-SSIL-2-InXy-pAHGH-ITR)

This work

pDF190 Expression plasmid encoding InXy fused to a human interleukin 2 (IL-2) secretion signal, driven by PhEF1α. SSIL-2-InXy was amplified from pDF186 using ODF71/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF190 (PhEF1α-SSIL-2-InXy-pABGH)

This work

pDF191 Expression plasmid encoding SeXy, a minimal intracellular version of XynA (InXy) fused to Igκ secretion signal, driven by PhEF1α. SSIgk-InXy (SeXy) was amplified from pDF186 using ODF72/ODF43 digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF191 (PhEF1α-SeXy-pABGH)

This work

pDF193 AAV2 vector encoding SeXy driven by PhCMV. SeXy was excised from pDF191 with BamHI/PmeI and cloned into the BamHI/EcoRV sites of pDF38 resulting in pDF193 (ITR-PhCMV-Intronβ-globin-SeXy-pAHGH-ITR)

This work

Page 88: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

87

Plasmid Description and Cloning Strategy Reference or source

pDF213 Expression vector encoding mutated SeXy (N30A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF74/ODF43 and ODF75/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF213 (PhEF1α-SeXyN30A-pABGH).

This work

pDF214 Expression vector encoding mutated SeXy (N42A, N47A, N51A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF76/ODF43 and ODF77/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF214 (PhEF1α-SeXyN42A,N47A,N51A-pABGH).

This work

pDF215 Expression vector encoding mutated SeXy (N163A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF78/ODF43 and ODF79/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF215 (PhEF1α-SeXyN163A-pABGH).

This work

pDF216 Expression vector encoding mutated SeXy (N203A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF80/ODC10 and ODF81/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODC10. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF216 (PhEF1α-SeXyN203A-pABGH).

This work

pDF218 Expression vector encoding mutated SeXy (N42A,N47A,N51A,N203A). Mutated SeXy was amplified from pDF214 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF80/ODC10 and ODF81/ODF72 on pDF214. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODC10. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF218 (PhEF1α-SeXyN42A,N47A,N51A,N203A-pABGH).

This work

Page 89: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

88

Plasmid Description and Cloning Strategy Reference or source

pDF220 Expression vector encoding mutated SeXy (N42A,N47A,N51A,N163A,N203A). Mutated SeXy was amplified from pDF218 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF78/ODC10 and ODF79/ODF72 on pDF218. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODC10. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF220 (PhEF1α-SeXyN42A,N47A,N51A,N163A,N203A-pABGH).

This work

pDF234 Expression vector encoding mutated SeXy (N42A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF97/ODF43 and ODF98/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF234 (PhEF1α-SeXyN42A-pABGH).

This work

pDF235 Expression vector encoding mutated SeXy (N47A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF99/ODF43 and ODF100/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF235 (PhEF1α-SeXyN47A-pABGH).

This work

pDF236 Expression vector encoding mutated SeXy (N51A). Mutated SeXy was amplified from pDF191 in a two-step fusion PCR reaction. (i) Two overlapping fragments were generated by PCR amplification using ODF101/ODF43 and ODF102/ODF72 on pDF191. (ii) The two fragments were purified and used as a template for the amplification of mutated SeXy, using ODF72/ODF43. The final product was digested with BamHI/XbaI and cloned into the corresponding sites of pEF4-MycHis resulting in pDF236 (PhEF1α-SeXyN51A-pABGH).

This work

Table 1. Plasmids used and designed in this study. Abbreviations: AAV2, adeno-associated virus type 2;

InXy, intracellular xylanase; His-tag, hexa-histidine tag; ITR, inverted terminal repeat; Myc-tag, myc

proto-oncogene epitope tag; pABGH, bovine growth hormone polyadenylation signal; pASV40, simian virus

40 polyadenylation signal; pAHGH, human growth hormone polyadenylation signal; PhEF1α, human

elongation factor 1α promoter; PhCMV, human immediate early cytomegalovirus promoter; SAMY, Bacillus

stearothermophilus-derived secreted α-amylase; SeXy, secreted xylanase; SEAP, human placental secreted

alkaline phosphatase; SSIgκ, signal sequence derived from the murine Igk-chain V-12-C region; SSIL-2,

signal sequence derived from the human interleukin 2 gene

Page 90: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

89

Table 2. Oligonucleotides used in this study.

Oligo Sequence Description ODC10 TAGAAGGCACAGTCGAGG Annealing sequence capital

letters ODF41 cgggatccaccatggagacagacacactcctgctatgggtactgctgctctg

ggttccaggttccactggtgacATGTTTAAGTTTAAAAAGAA BamHI [underlined], Kozak sequence [italics], secretion signal [bold], annealing sequence [capital letters]

ODF42 cgggatccaccATGTTTAAGTTTAAAAAGAA BamHI [underlined], Kozak sequence [italics], annealing sequence [capital letters]

ODF43 cttggcgcgcttatctagACCACACTGTTACGTTAG BssHII and XbaI [underlined], annealing sequence [capital letters]

ODF53 ggaattcaccatgtttaagtttaaaaagaatttcctggtgggactgtccgccgccctgatgagcattagcctgttttccGCAACCGCCTCTGCAGCTAGCACA

EcoRI site [underlined], optimized codons [bold], annealing sequence [capital letters]

ODF56 ODF58 ODF59 ODF60

cgggatccaccatgGTGGGACTGTCCGCCGCCCTG cgggatccaccatgGCAGCTAGCACAGACTACTGG cgggatccaccatgGATGGGGGCGGTATAGTAAAC cgggatccaccatgGTCAATGGGTCTGGCGGGAAT

BamHI site [underlined], Kozak sequence [italics], annealing sequence [capital letters]

ODF71 cgggatccaccatgtacaggatgcaactcctgtcttgcattgcactaagtctt gcacttgtcacaaacagtGCAGCTAGCACAGACTACTG

BamHI [underlined], Kozak sequence [italics], SSIL-2 [bold], annealing sequence [capital letters]

ODF72 cgggatccaccatggagacagacacactcctgctatgggtactgctgctctg ggttccaggttccactggtgacGCAGCTAGCACAGACTACTG

BamHI [underlined], Kozak sequence [italics], SSIgk [bold], annealing sequence [capital letters]

ODF74 ODF75 ODF76 ODF77 ODF78 ODF79 ODF80 ODF81 ODF97 ODF98 ODF99 ODF100 ODF101 ODF102

agactactggcaagcttggactgatgg ccatcagtccaagcttgccagtagtct atagtaaacgctgtcgctgggtctggcggggcttacagtgttgcttggtctaataccgga tccggtattagaccaagcaacactgtaagccccgccagacccagcgacagcgtttactat ccaaccggaagcgccgctacaatcact agtgattgtagcggcgcttccggttgg agtggaagttctgccgtaacagtgtgg ccacactgttacggcagaacttccact gtaaacgctgtcgctgggtctggcggg cccgccagacccagcgacagcgtttac gggtctggcggggcttacagtgttaat attaacactgtaagccccgccagaccc aattacagtgttgcttggtctaatacc ggtattagaccaagcaacactgtaatt

Oligonucleotide pairs used to introduce mutations [bold]

Page 91: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

90

Materials and Methods Vector construction. A detailed description of plasmid construction and

oligonucleotides used is provided in Tables 1 and 2.

Cell culture, transfection and transduction. Human embryonic kidney cells,

transgenic for the adenovirus type 5-derived E1 region as well as for the simian virus 40

(SV40) large T-antigen (HEK293-T; (Mitta et al., 2002)), human fibrosarcoma cells (HT-

1080; ATCC CCL-121) and normal human dermal fibroblasts (NHDF; PromoCell,

Heidelberg, Germany; cat. no. C-12300, lot 1070402) were cultivated in Dulbecco’s modified

Eagle’s medium (DMEM, Invitrogen, Carlsbad, CA) supplemented with 10% fetal calf serum

(FCS; PAN Biotech GmbH, Aidenbach, Germany; cat. no. 3302-P231902, lot no. P231902)

and 1% penicillin/streptomycin solution (Sigma Chemicals, St. Louis, MO). Chinese hamster

ovary cells (CHO-K1; ATCC CCL-61) were cultivated in ChoMaster® HTS medium (Cell

Culture Technologies GmbH, Gravesano, Switzerland) supplemented with 5% FCS and 1%

penicillin/streptomycin solution. CHO-K1 cell lines stably expressing secreted xylanase were

constructed by transfection of pDF164 into CHO-K1 cells and selection in ChoMaster® HTS

medium supplemented with 5% FCS, 1% penicillin/streptomycin and 100 mg/ml zeocin

(Invitrogen). All cell lines were cultivated at 37°C in a 5% CO2-containing humid

atmosphere. Transient transfections were performed using standard CaPO4-based protocols or

FuGENE® (Roche Diagnostics AG, Basel, Switzerland) transfection reagent according to the

manufacturers protocol.

For in-vitro experiments an MOI of 100 genomic particles/cell was used.

Production of adeno-associated virus (AAV) particles. AAV particles were

produced by co-transfection of the helper plasmid pDG (Grimm et al., 2003) and the

transgene-encoding AAV vector into HEK293-T using an optimized calcium phosphate-based

transfection protocol. In brief, 30x 2x106 HEK293-T cells were seeded into 30 culture dishes

(diameter 10cm) and cultivated overnight. Per 10cm dish 15µg of pDG were mixed with 5µg

AAV vector and 0.25M CaCl2 was added to reach a final volume of 400µl. The mixture was

added slowly to an equal volume (400µl) of HEPES-buffered saline solution (HeBS; 50mM

HEPES, 280mM NaCl, 1.5mM Na2HPO4, pH 7.1), vortexed briefly and incubated for 2min

before adding the DNA-CaPO4 precipitate solution to the monolayer culture. The precipitate

was removed after 6h, and the cells were incubated in 10% FCS-supplemented DMEM for

60h. For AAV particle recovery, the culture supernatant was discarded, the cells were

detached using a cell scraper and resuspended in 1ml PBS (137.9mM NaCl, 8.1mM

Page 92: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

91

Na2HPO4, 1.47mM KH2PO4, 2.67mM KCl; Invitrogen, Carlsbad, CA) per 10cm dish. The

combined cell suspensions were then transferred to a 50ml Falcon tube and pelleted by

centrifugation for 3min at 1200xg. After another washing step using 30ml PBS, the cells were

resuspended in 20ml PBS and the intracellular viral particles were released following cell

lysis, then induced by three consecutive freeze-thaw cycles consisting of shuttling the tube

between liquid nitrogen and a 37°C water bath (tube was vortexed vigorously after each

thawing step). The cell debris was eliminated by centrifugation for 5min at 10,000xg and the

supernatant containing the crude viral particle stock was collected, supplemented with 50U/ml

of Benzonase (Sigma, cat. no. E1014) and incubated for 30min at 37°C. Iodixanol density

gradient purification of viral particles was performed according to Zolotukhin et al.

(Zolotukhin et al., 1999). Step gradients were centrifuged for 3.5h at 150,000xg (18°C). The

clear 40% iodixanol layer was harvested by puncturing the side of the tube with a 16G needle

attached to a syringe. A heparin affinity column (HiTrap Heparin HP, Amersham

Biosciences, Sweden; cat. no. 17-0407-01) was equilibrated with 10 bed volumes of binding

buffer (10mM Na2PO4, pH 7) at a flow rate of 4ml/min. The 40% iodixanol layer of the

gradient containing the viral particles was loaded onto the column. After washing with 10 bed

volumes of binding buffer the viral particles were eluted with 5 bed volumes of elution buffer

(10mM Na2PO4, 1M NaCl, pH 7) and were then concentrated using a spin column with a

molecular weight cut-off (MWCO) of 30 kDa (Vivaspin, Vivascience, Germany; cat. no.

VS1521).

Xylanase assays. DNS (3,5-dinitrosalicylic acid) assay: 50µl of xylanase A-

containing cell culture supernatant were centrifuged at 16,000xg for 1min to remove cell

debris and 30µl were then transferred to 90µl of substrate solution (100mM sodium acetate,

pH5.5, 1% birchwood xylan [Sigma, cat. no. X0502, lot no. 044K0169]) and incubated for

30min at 37°C. Xylanase A-mediated xylan degradation was stopped by addition of 120µl

DNS solution (dissolve 1g 3,5-dinitrosalicylic acid [Sigma, cat. no. D0550, lot no. 80K3490]

and 30g sodium potassium tartrate in 20ml 2N NaOH and adjust to 100ml using dH2O).

Boiling of the reaction mixture for 5min results in DNS-mediated covalent chromogenic

modification of glucose liberated by xylanase A-driven xylan breakdown, which can be

quantified after addition of 760µl H2O by scoring absorbance at 540nm and comparing the

value to a calibration curve produced with pure glucose (Fluka, cat. no. 49139, lot R00354).

Xylanase units were calculated according to the following equation:

Page 93: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

92

Enzymatic activity (U*l-1): EA= µmol*min-1l-1 glucose produced

PAHBAH (p-hydroxybenzoic acid hydrazide) assay: 50µl of xylanase A-containing

cell culture supernatant were centrifuged at 16,000xg for 1min to remove cell debris and 30µl

were then transferred to 90µl substrate solution (100mM sodium acetate, pH5.5, 1%

birchwood xylan [Sigma, cat. no. X0502, lot no. 044K0169]) and incubated for 30min at

37°C. Xylanase A-mediated xylan degradation was stopped by addition of 1ml PAHBAH

reagent (1% p-hydroxybenzoic acid hydrazide [ABCR, cat. no. AV12702] in 0.5M NaOH).

Boiling of the reaction mixture for 5 min results in PAHBAH-mediated covalent chromogenic

modification of glucose liberated by xylanase A-driven xylan breakdown, which can be

quantified by scoring absorbance at 405nm and comparison to a calibration curve generated

using pure glucose (Fluka). Xylanase units were calculated using the equation indicated

above.

Remazol brilliant blue xylan (RBB-xylan) assay: 50µl of xylanase A-containing cell

culture supernatant were centrifuged at 16,000xg for 1min to remove cell debris. 30µl were

then transferred to 90µl RBB-xylan solution and incubated for 60 min at 37°C. Xylanase-

mediated RBB release from RBB-xylan was stopped by addition of two volumes of 100%

ethanol and the reaction mixture was centrifuged for 2min at 16,000g. 200µl of the

supernatant were transferred to a 96-well plate, absorbance was measured at 600nm and

compared to readings taken from standardized solutions containing defined xylanase (Sigma,

cat. no. X3876) concentrations.

Fluorescence-based xylanase assay: The EnzCheck® xylanase assay (Molecular

Probes, Eugene, OR, USA; cat. no. E33650, lot no. 35416A) was performed according to the

manufacturer’s protocol. In brief, 20µl of cell culture supernatant or cleared cell lysate were

centrifuged at 16,000g for 1min to remove debris. 15µl were transferred to a 384-well plate

and incubated for 30min at 37°C after addition of a substrate mix (fluorescence substrate

[3.3% v/v] in 100mM sodium acetate). Fluorescence intensity was measured at an excitation

wavelength of 340nm and an emission wavelength of 450nm on a Genios Pro multiwell plate

reader (Tecan AG, Männedorf, Switzerland). Xylanase from Trichoderma viride (Sigma, cat.

no. X3876, lot no. 036K4147) was used as an internal standard.

Glycosylation profiling and immunofluorescence. Cell extracts were prepared by

detaching HEK293-T cells from 6-well plates using a cell scraper, collecting them by

centrifugation at 280xg and washing the cells once with 2ml PBS. The cells were then

Page 94: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

93

resuspended in 400µl PBS and lysed by three freeze/thaw cycles consisting of shuttling the

sample twice between a 37°C water bath and liquid nitrogen before clearing the lysate by

centrifugation at 16,000xg and 4°C for 5min. Preparation of secreted proteins was performed

by harvesting cell culture supernatants, followed by centrifugation for 2min at 16,000g to

remove cells and cell debris and subsequent transfer of cleared supernatants to fresh tubes.

For glycosylation analysis, 90µl of cell culture supernatants were treated with PNGase

F (New England Biolabs, Beverly, MA; cat. no. P0704S, lot no. 30) as indicated in the

manufacturer’s protocol. 40µl of the de-glycosylated samples were mixed with 10ml 5x SDS

loading buffer (50% glycerol, 10%SDS, 250mM Tris, pH6.8 containing 10% (v/v) β-

mercaptoethanol) and boiled for 5min before loading on a 9% SDS-polyacrylamide gel.

Proteins were electroblotted onto polyvinylidene difluoride membranes (Millipore, Bedford,

MA; cat. no. IPVH20200, lot no. K3AN0841E). After blocking in TBS (50mM Tris, 150mM

NaCl, pH7.5) containing 3% non-fat dry milk, the membrane was probed with a monoclonal

antibody specific for the hexa-histidine tag (HIS, Novagen, Madison, WI, USA; cat. no.

70796-3 lot N56036) or mouse serum (diluted 1/50) and visualized using an anti-mouse IgG

coupled to a horseradish peroxidase (ECLTM Anti Mouse IgGxHRP, Amersham,

Buckinghamshire, UK) and a chemiluminescence-based assay (ECLTM plus, Amersham)

according to the manufacturer’s protocol.

Immunofluorescence-based analysis. An 8-chamber culture slide (NUNCTM,

Roskilde, Denmark) was inoculated with 5000 HT-1080 cells per well. After 2 days in culture

the cells were fixed with 4% paraformaldehyde (Sigma, cat. no. 76240) in phosphate-buffered

saline (PBS; Invitrogen) for 15 min, and subsequently washed twice for 5min in PBS. Cell

monolayers were then permeabilized for 15min using 0.5% Triton X-100 in PBS containing

1%BSA and then washed twice for 5min with PBS. Cells were then blocked for 2h in PBS

containing 1% BSA and 5% goat serum (Sigma, cat. no. G9023). Primary antibodies specific

for the endoplasmic reticulum marker protein GRP78/BiP (rat polyclonal; Sigma, cat. no.

G8918, lot 073K4875) and hexa-histidine tag (mouse monoclonal; Novagen, Madison, WI,

USA; cat. no. 70796-3 lot N56036) were diluted in 1% BSA and 5% goat serum-containing

PBS. Primary antibodies were incubated for 4h at 4°C and subsequently washed three times in

PBS. Cy3-coupled anti-mouse (Jackson Immunochemicals, West Grove, PA; cat. no. 115-

165-146) and Cy5-coupled anti-rabbit (Jackson Immunochemicals, West Grove, PA; cat. no.

111-175-144) secondary antibodies were incubated for 4 hours at room temperature. Finally,

the cells were washed three times with PBS and embedded using Tris-buffered glycerol (a 3:7

Page 95: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

94

mixture of 0.1 M Tris-HCl [pH 9.5] and glycerol supplemented with 50 mg/ml n-propyl-

gallat).

The imaging system consisted of an inverted fluorescence microscope (Leica DMIRB/E,

Glattbrugg, Switzerland) equipped with a Leica 63x oil immersion objective, a confocal

scanner (Leica TCS SP1) featuring an argon and helium-neon laser and a Silicon Graphics

Workstation (SGI, Schlieren, Switzerland) with Imaris 3D multi-channel image processing

software installed (Bitplane, AG, Zurich, Switzerland) (Messerli et al., 1993).

Animal studies. Female OF1 (oncins france souche 1) mice were obtained from Iffa-

Credo (Lyon, France). Mice were treated with intramuscular injections of 200µl PBS

containing pDF193-derived AAV particles adjusted to 1x1010 particles/ml. Blood samples

were collected retroorbitally at day 6, 19 and 34 and serum was prepared using microtainer

SST tubes (Beckton Dickinson, Plymouth, UK). All experiments involving animals were

conducted according to European Community legislation (86/609/EEC), and have been

approved by the French Republic (No. 69266310) and performed by M.D-E. at the Institut

Universitaire de Technologie, IUTA, F-69622 Villeurbanne Cedex, France.

Prediction of N-glycosylation sites. All predictions of possible glycosylation sites

were performed by using NetNGlyc software (www.cbs.dtu.dk/services/NetNGlyc).

Results Cloning and expression of Bacillus subtilis xylanase A in mammalian cells

PCR-mediated amplification of full-length genomic Bacillus subtilis xylanase A

(xynA) and constitutive production in a transfected human embryonic kidney cell line

(HEK293-T) of an entirely eukaryotic expression platform (pDF149; PhEF1α-xynA-pABGH)

showed that Xylanase A is efficiently secreted (Figure 1A). Computational analysis of the

bacterial coding sequence revealed several codons in the N-terminus, which were predicted to

be suboptimal for efficient translation in mammalian cells. We have therefore designed a

modified mammalian cell-compatible xynA derivative (xynAM) harboring a codon-optimized

5’-terminus (pDF163; PhEF1α-xynAM-pABGH). Comparative expression profiling using the

isogenic vectors pDF149 (PhEF1α-xynA-pABGH) and pDF163 (PhEF1α-xynAM-pABGH) showed an

almost two-fold higher production level of XynAM compared to the native prokaryotic XynA

(Figure 1A).

Page 96: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

95

In order to characterize the sequence motif mediating Xylanase A secretion in

mammalian cells, we have designed a variety of N-terminal xynA deletion mutants and

expressed them in HEK293-T using an expression configuration isogenic to pDF163. The

observation (i) that pDF184 (PhEF1α-xynAM!1-pABGH) mediated Xylanase A secretion levels

identical to pDF163 (PhEF1α-xynAM-pABGH), (ii) that pDF187 (PhEF1α-xynAM!1-115-pABGH) as

well as pDF188 (PhEF1α-xynAM!1-139-pABGH) showed no Xylanase A production at all and (iii)

that Xylanase A mutants expressed from pDF186 (PhEF1α-xynAM!1-82-pABGH) were produced

but no longer secreted, suggests that the mammalian cell-compatible prokaryotic secretion

signal is encoded within the first 27 amino acids of XynA’s N-terminus (Figure 1A). pDF186-

encoded XynAM!1-82 is therefore an ideal intracellular mammalian reporter protein referred to

as InXy (intracellular xylanase A).

In order to modify XynA for high-level secretion in mammalian cells we have fused

InXy N-terminally to two strong mammalian secretion signals (SS) derived from human

interleukin 2 (IL-2; SSIL-2) (pDF190; PhEF1α-SSIL-2-InXy-pABGH) or from the murine

immunoglobulin Igk-chain V-J2-C region (SSIgκ) (pDF164; PhEF1a-SSIgk-xynAM-pABGH;

pDF191; PhEF1α-SSIgκ-InXy-pABGH). Comparative production profiling in transiently

transfected HEK293-T cells showed that SSIgκ-containing XynA variants were more

efficiently secreted than the SSIL-2-InXy fusion protein (pDF190; PhEF1α-SSIL-2-InXy-pABGH)

and the codon-optimized full-length prokaryotic variant (pDF163; PhEF1α-xynAM-pABGH)

(Figure 1B). Although the secretion performance of pDF164 (PhEF1α-SSIgκ-xynAM-pABGH) and

pDF191 (PhEF1α-SSIgκ-InXy-pABGH) was equivalent, SSIgκ-InXy was smaller and therefore

considered the ideal secreted xylanase (SeXy), which was used for further studies.

Transient transfection of SeXy-encoding expression vectors into different cell types

showed comparable expression and secretion levels (i) in the human cell lines HEK293-T and

HT-1080, (ii) in the biotechnologically relevant rodent cell lines CHO-K1 and BHK-21 as

well as to a lower extent (iii) in human primary fibroblasts (NHDF) (Figure 1C).

Immunofluorescence analysis of HEK293-T transiently transfected for InXy (pDF186; PhEF1α-

InXy-pABGH) and SeXy (pDF191; PhEF1α-SeXy-pABGH) production showed that SeXy co-

localized with the ER marker protein GRP78/BiP indicating that it is secreted via the classical

mammalian secretory pathway, whereas InXy was found exclusively in the cytosol (Figure 2).

Page 97: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

96

Enzymatic assays

We have refined different assays originally established for quantification of fungal and

bacterial xylanases and evaluated their sensitivity for detection of SeXy production in the

culture supernatants of mammalian cells. Two assays (DNS, 3,5-dinitrosalicylic acid;

PAHBAH, p-hydroxybenzoic acid hydrazide) capitalize on chromogenic modification of a

substrate by reducing sugar liberated by xylanase-mediated degradation of purified birchwood

xylan (Lever, 1972) (Bailey and Poutanen, 1989), the remazol brilliant blue (RBB) assay is

based on xylanase-driven release of RBB from modified RBB-xylan (Biely et al., 1985) and

the fluorescence assay which takes advantage of xylanase-based conversion of a synthetic

substrate into a fluorescent product (EnzChek ultra xylanase assay, Molecular Probes). Key

performance parameters including detection limit and background activity of all xylanase

assays were determined by analysis of serially diluted HEK293-T culture supernatants 48h

after transient production of pDF191-encoded SeXy.

The DNS method was the least sensitive of the four xylanase assays (detection limit at

20mU/ml) and also displayed elevated levels of background signal originating from the

residual glucose in the culture medium or endogenous glycoproteins produced by the cells.

The PAHBAH assay showed increased sensitivity (detection limit at 10mU/ml) but featured

the same elevated background levels as the DNS method. RBB-Xylan is a convenient

substrate with very low background activity but only average sensitivity (detection limit at

5mU/ml). The most powerful assay in terms of detection limits, ease of handling and low

background activity is the EnzCheck® Ultra Xylanase Assay Kit (Molecular Probes) allowing

fast detection of xylanase activity as low as 0.5 mU/ml at negligible background levels (Table

3).

Page 98: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

97

Figure 1. Quantification of xylanase A activity in HEK293-T cells. (A) Culture supernatants (black) as well as

cell lysates (grey) of HEK293-T cells transiently transfected with pDF149 (PhEF1α-xynA-pABGH), pDF163 (PhEF1α-

xynAM-pABGH), pDF184 (PhEF1α-xynAMD1-27-pABGH), pDF186 (PhEF1α-xynAMD1-82-pABGH), pDF187 (PhEF1α-

xynAMD1-115-pABGH) and pDF188 (PhEF1α-xynAMD1-139-pABGH) were assayed for xylanase production. (B)

XylanaseA production was determined in cell culture supernatants transiently transfected with pDF164 (PhEF1α-

SSIgκ-xynAM-pABGH), pDF186 (PhEF1α-InXy-pABGH), pDF190 (PhEF1α-SSIL-2-InXy-pABGH), pDF191 (PhEF1α-SeXy-

pABGH) and pDF163 (PhEF1α-xynAM-pABGH). (C) Xylanase A production was determined in different cell types

transiently transfected with pDF191 (PhEF1α-SeXy-pABGH).

Page 99: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

98

Figure 2. Subcellular localization of xylanase A in HEK293-T cells. HEK293-T cells were transfected either

with pDF191 (PhEF1α-SeXy-pABGH) and pDF186 (PhEF1α-InXy-pABGH) or no vector (control). Intracellular

localization was visualized by immunofluorescence microscopy using SeXy- and 78kDa glucose regulated

protein (GRP78/BiP)-specific primary antibodies as well as secondary Cy3-coupled anti-mouse and Cy5-

coupled anti-rabbit antibodies. The third and fourth column show an overlay of the Cy3 and the Cy5 channels to

visualize xylanase localization relative to the endoplasmic reticulum (ER) and a differential interference contrast

(DIC) image showing all cells in range.

Page 100: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

99

Detection limit [mU/ml]

Assay time [min]

Background

DNS 20 60 High (>50mU/ml) PAHBAH 10 60 High (>50mU/ml) RBB-Xylan 5 80 Medium (10mU/ml) EnzCheck® 0.5 40 Low (1mU/ml) Table 3. Comparison of different assays used to quantify xylanase A activity from cell culture supernatants. Stability of SeXy and InXy

Stability is a key characteristic of reporter proteins as it enables precise detection of

actual protein levels without considering protein degradation or a considerable loss of activity

during the assay procedure. We have assayed the stability of SeXy in human serum by mixing

cell culture supernatants of pDF191-transfected HEK293-T cells with human AB serum and

incubated the samples at 37°C for 15 days. Samples were removed every three days and

frozen at -20°C. The stability of SeXy in serum was excellent with only a minor loss of

activity over the investigated time period (Figure 3A). SeXy’s half-life (t1/2) of 1500h

compared favorably with the best-in-class reporter protein the human placental secreted

alkaline phosphatase (SEAP, t1/2 502h; (Schlatter et al., 2002)) as well as the low molecular

weight urokinase-type plasminogen activator (u-PALMW, t1/2 61h; (Schlatter et al., 2002)) and

was of the same order of magnitude as the Bacillus stearothermophilus-derived secreted α-

amylase (SAMY, t1/2 1505h; (Schlatter et al., 2002)) (Figure 3A).

Besides stability at room temperature in complex solutions such as human serum,

stable activity over a broad pH range, the robustness in multiple freeze/thaw cycles and

resistance to increased temperatures qualify perfect reporter proteins. SeXy and InXy,

incubated for 48h in buffers adjusted to different pH, retained its standard activity within a pH

range from 3.5 to 9 (Figure 3B). Although not derived from an extremophilic organism as

SAMY, SeXy and InXy retained their activity over a broad temperature range with a peak

activity around 45°C (Figure 3C). Multiple freeze/thaw (-20°C/+20°C) cycles were tolerated

by SeXy and InXy without showing any decrease in activity (Figure 3D). The observation

that InXy resists freeze/thaw cycles significantly simplifies assay protocols as InXy-

producing cells could simply be frozen for storage and thawed for immediate analysis as InXy

is completely liberated from disrupted cells (Figure 3D).

Page 101: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

100

Figure 3. Quantification of SeXy and InXy activity in culture supernatants of HEK293-T transiently transfected

with pDF191 (PhEF1α-SeXy-pABGH) and pDF186 (PhEF1α-InXy-pABGH) treated under different conditions. (A)

Stability of native () and peptide, N-glycosidase F (PNGase F)-treated () SeXy incubated in human serum.

(B) Activity of SeXy (black) and InXy (grey) after incubation at different pH values for 48h at 37°C. (C) SeXy

(black) and InXy (grey) activity at different incubation temperatures. (D) A change in enzyme activity was

determined after up to six consecutive freeze/thaw cycles with an assay of SeXy (black) and InXy (grey) activity

after each round.

Page 102: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

101

Compatibility of SeXy with other secreted reporter proteins and stable expression of

SeXy in transgenic CHO cells

Combinatorial expression analysis and elucidation of complex regulatory cascades

require the simultaneous use of different compatible reporter proteins whose activity can be

independently assayed in the same cell culture supernatant (Fux and Fussenegger, 2003). In

order to assess the enzymatic compatibility of SeXy with other established reporter proteins

such as SEAP and SAMY we cotransfected HEK293-T cells with pSEAP2-control (PSV40-

SEAP-pASV40-ESV40), pSS158 (PhCMV-SAMY-pABGH), and pDF191 (PSV40-SeXy-pABGH) and

followed expression of the three secreted reporter proteins for 48h after transfection. All three

reporter proteins showed a comparable increase in reporter activity in the supernatant over

time, suggesting that they would be compatible in simultaneous analysis (Figure 4A).

SeXy was stably expressed in a clonal CHO-K1 cell line and proliferation rate as well as

specific reporter productivity were monitored over three days. Transgenic CHO-K1 cells

showed wild type-like growth characteristics and sustained SeXy production (Figure 4B).

Glycosylation analysis of SeXy

Most of today’s blockbuster protein pharmaceuticals are glycoproteins. In order for a

reporter protein to capture product parameters relevant for biopharmaceutical manufacturing

it needs to be glycosylated. In order to characterize SeXy’s glycoprofile after production in

mammalian cells, we incubated pDF191 (PSV40-SeXy-pABGH)-transfected HEK293-T cells

with tunicamycin (inhibiting N- as well as O-glycosylation), or treated SeXy-containing

culture supernatants with PNGase F (exclusive elimination of N-glycans).

Western blot analysis of SeXy isoforms revealed that untreated cell culture

supernatants showed distinct SeXy-specific bands suggesting that SeXy is produced in several

N-glycosylated isoforms. Cultivation of SeXy-producing cells in tunicamycin-containing cell

culture medium or treating the supernatants with PNGase F produced a single band of 22kDa

which is expected to represent the unglycosylated form of SeXy (Figure 5). Software-based

analysis of SeXy suggested seven N-glycosylation sites which we have eliminated by

designing the following mutants: pDF213 (PhEF1α-SeXyN30A-pABGH), pDF214 pDF234 (PhEF1α-

SeXyN42A-pABGH), pDF235 (PhEF1α-SeXyN47A-pABGH), pDF236 (PhEF1α-SeXyN51A-pABGH),

pDF215 (PhEF1α-SeXyN163A-pABGH) and pDF216 (PhEF1α-SeXyN203A-pABGH). Whereas

pDF213- and pDF236 -encoded SeXy yielded the same glycosylation patterns as wild-type

SeXy (Figure 6), all other SeXy mutants showed a distinct but different glycoprofile,

Page 103: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

102

suggesting that Asn 42, 47, 163 and 203 are indeed glycosylated (Figure 6). We were able to

detect all glycoforms in the supernatant except for SeXyN42A,N47A,N51A,N163A,N203A (pDF220)

which has all of its glyocosylation sites eliminated and was found exclusively in the cytosol.

This observation indicated that glycosylation is essential for secretion of SeXy.

Figure 4. (A) Simultaneous expression and quantification of three different secreted mammalian reporter

proteins in the same culture supernatant. pDF191 (PhEF1α-SeXy-pABGH), pSS158 (PhCMV-SAMY-pABGH) and

pSEAP2-control (PSV40-SEAP-pASV40-ESV40) were co-transfected at equimolar ratios and reporter proteins were

quantified in the same supernatants. (B) Analysis of proliferation and specific production rates of a CHO-K1 cell

clone stably expression SeXy.

Page 104: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

103

SeXy expression by different cell lines and in mice after transduction with transgenic

adeno-associated virus particles

In order to validate SeXy as a universal reporter protein for in-vitro as well as in-vivo

use, we have engineered adeno-associated virus (AAV) particles for transduction of a variety

of mammalian cell lines (HEK293-T; HT-1080; CHO-K1), primary cells (normal human

dermal fibroblasts [NHDF]) as well as mice with constitutive SeXy expression. Transduction

of HEK293-T, HT-1080, CHO-K1 and NHDF with pDF193- (ITR-PhCMV-Intronβ-globin-SeXy-

pAHGH-ITR)-and pDF189- (ITR-PhCMV-Intronβ-globin-SSIL-2-InXy-pAHGH-ITR) derived AAV

particles induced SeXy production in all cell types after 48h (Figure 7).

Figure 5. Western blot analysis of SeXy derived from pDF191 (PhEF1a-SeXy-pABGH)-transfected HEK293-T cells

cultivated in the presence and absence of 2µg/ml tunicamycin and optionally treated with Peptide N-Glycosidase

F (PNGase F).

Intramuscular injection of mice with pDF193-derived AAV, showed significant SeXy

levels in their serum after 6 days (2U±0.3U/l). Mouse sera analyzed 19 and 34 days after

injection of transgenic AAVs showed no further SeXy production but contained SeXy-

specific antibodies when used to probe Western blot membranes with bound SeXy from

Page 105: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

104

supernatants of pDF191-transfected HEK293-T (Figure 8). The Western blot signals were

identical to those shown in Figures 5 and 6 using HIS tag-specific antibodies, which confirms

the specificity of the mouse antibodies for a highly glycosylated SeXy protein. These SeXy-

specific mouse antibodies, which suggest that SeXy is immunogenic, may clear the reporter

protein from the blood stream.

Figure 6. Western blot analysis of SeXy mutants harboring specific mutations at predicted N-glycosylation sites.

pDF191 (PhEF1a-SeXy-pABGH), pDF213 (PhEF1α-SeXyN30A-pABGH), pDF234 (PhEF1α-SeXyN42A-pABGH), pDF235

(PhEF1α-SeXyN47A-pABGH), pDF236 (PhEF1α-SeXyN51A-pABGH), pDF215 (PhEF1α-SeXyN163A-pABGH), pDF216

(PhEF1α-SeXyN203A-pABGH) and pDF220 (PhEF1α-SeXyN42A,N47A,N51A,N163A,N203A-pABGH) were transiently transfected

into HEK293-T and supernatants as well as cell pellets (after three times freeze/thaw) were used for Western

Blot-based analysis of SeXy glycoprofiles.

Page 106: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

105

Figure 7. Quantification of SeXy in cell culture supernatants of different mammalian cell lines and primary cells

transduced with transgenic AAV particles. pDF193 (ITR-PhCMV-Intron-β-globin-SeXy-pAHGH-ITR) as well as

pDF189 (ITR-PhCMV-Intron-β-globin-SSIL2-xynAM-pAHGH-ITR)-derived AAV particles were used to transduce

HEK293-T, HT-1080, CHO-K1 and NHDF monolayer cultures. Expression was quantified in each supernatant

48h after transduction.

Figure 8. Western blot analysis of SeXy produced by pDF193-transduced HEK293-T using serum of mice

intramuscularly injected with SeXy-producing AAV particles. Mouse serum was probed for the presence of

circulating antibodies against SeXy after 19 and after 34 days. A membrane containing bound SeXy was probed

with diluted mouse serum and bands were visualized using an horseradish peroxidase (HRP)-coupled anti-mouse

antibody.

Page 107: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

106

Discussion

Bacillus subtilis-derived xylanase A versions engineered for intracellular (InXy) and

secreted (SeXy) production in mammalian cells share several characteristics of an ideal

mammalian reporter protein and have considerable advantages over currently used secreted

reporter systems: (i) with a size of only 642bp (InXy) and 702bp (SeXy) the xylanase A

variants are amongst the most compact (secreted) reporter genes which favors their use in

space-limited expression scenarios such as viral transduction systems. (ii) The lack of

structural and functional homologs in mammalian cells enables interference-free

quantification of InXy and SeXy in the absence of endogenous background signals and

without the need for complex sample preparations. (iii) Since InXy and SeXy process an

unphysiologic substrate their expression is unlikely to elicit any pleiotropic effects. (iv) InXy

and SeXy are an isogenic pair of reporter proteins enabling precise assessment of intracellular

production and secretion capacities of mammalian cells. (v) Both reporter proteins are

compatible with a variety of different assay systems including the highly sensitive

fluorescence-based detection technology. (vi) Xylanase A-based reporter proteins are

extremely stable in complex solutions and at room temperature over extended periods of time,

which enables consistent quantifications any time after sampling without considering complex

degradation profiles. (vii) InXy and SeXy activities are highly robust allowing precise

quantification over a wide range of temperature and pH changes. (viii) InXy and SeXy are

resistant to repeated freeze/thaw cycles facilitating handling and reproducible analysis. Also,

a single freeze/thaw cycle releases ready-to-assay InXy from mammalian cells. (ix) Since

SeXy is N-glycosylated like most marketed protein pharmaceuticals it could be used for

evaluation of generic biopharmaceutical manufacturing scenarios. (x) SeXy is compatible

with viral transduction and can be expressed in a variety of mammalian cell types. (xi) InXy

and SeXy are functionally compatible with other prominent reporter proteins such as SEAP

and SAMY and can be independently quantified in a sample containing both of these proteins.

(xii) SeXy circulates in the bloodstream of mice after intramuscular transduction.

Earlier findings exemplified the targeting of proteins harboring prokaryotic secretion

signals to the mammalian secretion apparatus (Fontes et al., 1999; Schlatter et al., 2002; Soole

et al., 1993) and it was therefore not unexpected that wild type Xylanase A transiently

expressed in mammalian cells was secreted to the supernatant. Although secretion of B.

subtilis Xylanase A was substantial, it could be further increased by optimizing the codons for

Page 108: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

107

optimal translation in mammalian cells and by replacing the microbial signal sequence by

mammalian homologues. An immunoglobulin-derived signal sequence promoted the highest

secretion level of different Xylanase A variants and was therefore used as the standard

secreted xylanase A (SeXy) for use in mammalian cells. Preliminary glycoprofiling suggested

SeXy to be N-glycosylated at four different sites. Since most protein pharmaceuticals are N-

glycosylated SeXy could be an attractive reporter protein to study prototype

biopharmaceutical scenarios and their impact on overall product quality. Glycosylation

analysis using SeXy is particularly straightforward as the protein (i) is very stable in cell

culture supernatants, (ii) yields characteristic banding patterns with distinct signals for all five

glycosylation states and (iii) is C-terminally tagged with a hexa-histidine tag for convenient

direct analysis by western blot or purification on nickel resins. InXy and SeXy are easily

traceable using different assays, which vary in cost, speed and sensitivity. Although and

PAHBAH tests capitalizing on chromogenic detection of reducing sugars released by the

reporter proteins from standardized xylan are rapid and low cost, sugars in the cell culture

medium or released by cells increase background and compromise assay sensitivity. While

the RBB assay proved to be reasonably robust, cheap and sensitive for every-day use, the

fluorescence-based InXy and SeXy detection assay is the most sensitive and should be

applied for reference studies.

For most assay and protein characteristics InXy and SeXy compare favorably with

other secreted reporter genes such as SEAP and SAMY. However, unlike SEAP and SAMY

for which functional homologs (phosphatases and amylases, exist in mammalian cells and

which may contribute to significant background, InXy and SeXy are functionally

heterologous and if xylanase activity is detected in mammalian samples it is exclusively

originating from these reporter proteins. Sensitive and robust secreted reporter proteins will

become increasingly important as model proteins for biopharmaceutical manufacturing, for

gene-function analysis and promoter characterization as well as for drug screening initiatives.

InXy and SeXy are expected to meet with those applications and challenges at a high standard

and represent a unique addition to the available portfolio of secreted reporter proteins.

Page 109: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

108

Acknowledgements We thank Simon Taennler for providing genomic B. subtilis DNA and Marcia

Schoenberg as well as Nilou Arden and Wilfried Weber for critical comments on the

manuscript. This work was supported by the Swiss National Science Foundation (grant no.

3100A0-112549) and by the EC Framework 6 program (COBIOS).

References An, G., Hidaka, K., and Siminovitch, L. (1982). Expression of bacterial beta-galactosidase in

animal cells. Mol Cell Biol 2, 1628-1632.

Bailey, M.J., and Poutanen, K. (1989). Production of xylanases by strains of Aspergillus.

Applied Microbiology and Biotechnology 30, 5-10.

Berger, J., Hauber, J., Hauber, R., Geiger, R., and Cullen, B.R. (1988). Secreted placental

alkaline phosphatase: a powerful new quantitative indicator of gene expression in eukaryotic

cells. Gene 66, 1-10.

Biely, P., Mislovicova, D., and Toman, R. (1985). Soluble chromogenic substrates for the

assay of endo-1,4-beta-xylanases and endo-1,4-beta-glucanases. Anal Biochem 144, 142-146.

de Wet, J.R., Wood, K.V., Helinski, D.R., and DeLuca, M. (1985). Cloning of firefly

luciferase cDNA and the expression of active luciferase in Escherichia coli. Proc Natl Acad

Sci U S A 82, 7870-7873.

Durocher, Y., Perret, S., Thibaudeau, E., Gaumond, M.H., Kamen, A., Stocco, R., and

Abramovitz, M. (2000). A reporter gene assay for high-throughput screening of G-protein-

coupled receptors stably or transiently expressed in HEK293 EBNA cells grown in

suspension culture. Anal Biochem 284, 316-326.

Fontes, C.M., Ali, S., Gilbert, H.J., Hazlewood, G.P., Hirst, B.H., and Hall, J. (1999).

Bacterial xylanase expression in mammalian cells and transgenic mice. J Biotechnol 72, 95-

101.

Fux, C., and Fussenegger, M. (2003). Toward higher order control modalities in mammalian

cells-independent adjustment of two different gene activities. Biotechnol Prog 19, 109-120.

Gorman, C.M., Moffat, L.F., and Howard, B.H. (1982). Recombinant genomes which express

chloramphenicol acetyltransferase in mammalian cells. Mol Cell Biol 2, 1044-1051.

Page 110: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

109

Grimm, D., Kay, M.A., and Kleinschmidt, J.A. (2003). Helper virus-free, optically

controllable, and two-plasmid-based production of adeno-associated virus vectors of

serotypes 1 to 6. Mol Ther 7, 839-850.

Hadjantonakis, A.K., Dickinson, M.E., Fraser, S.E., and Papaioannou, V.E. (2003).

Technicolour transgenics: imaging tools for functional genomics in the mouse. Nat Rev Genet

4, 613-625.

Kamionka, A., Sehnal, M., Scholz, O., and Hillen, W. (2004). Independent regulation of two

genes in Escherichia coli by tetracyclines and Tet repressor variants. J Bacteriol 186, 4399-

4401.

Kramer, B.P., and Fussenegger, M. (2005). Hysteresis in a synthetic mammalian gene

network. Proc Natl Acad Sci U S A 102, 9517-9522.

Lee, J.C., Chang, C.F., Chi, Y.H., Hwang, D.R., and Hsu, J.T. (2004). A reporter-based assay

for identifying hepatitis C virus inhibitors based on subgenomic replicon cells. J Virol

Methods 116, 27-33.

Lever, M. (1972). A new reaction for colorimetric determination of carbohydrates. Anal

Biochem 47, 273-279.

Liu, L., Sanz, S., Heggestad, A.D., Antharam, V., Notterpek, L., and Fletcher, B.S. (2004).

Endothelial targeting of the Sleeping Beauty transposon within lung. Mol Ther 10, 97-105.

Lorenz, W.W., McCann, R.O., Longiaru, M., and Cormier, M.J. (1991). Isolation and

expression of a cDNA encoding Renilla reniformis luciferase. Proc Natl Acad Sci U S A 88,

4438-4442.

Mannhaupt, G., Pilz, U., and Feldmann, H. (1988). A series of shuttle vectors using

chloramphenicol acetyltransferase as a reporter enzyme in yeast. Gene 67, 287-294.

Markova, S.V., Golz, S., Frank, L.A., Kalthof, B., and Vysotski, E.S. (2004). Cloning and

expression of cDNA for a luciferase from the marine copepod Metridia longa. A novel

secreted bioluminescent reporter enzyme. J Biol Chem 279, 3212-3217.

Messerli, J.M., van der Voort, H.T., Rungger-Brandle, E., and Perriard, J.C. (1993). Three-

dimensional visualization of multi-channel volume data: the amSFP algorithm. Cytometry 14,

725-735.

Mitta, B., Rimann, M., Ehrengruber, M.U., Ehrbar, M., Djonov, V., Kelm, J., and

Fussenegger, M. (2002). Advanced modular self-inactivating lentiviral expression vectors for

multigene interventions in mammalian cells and in vivo transduction. Nucleic Acids Res 30,

e113.

Page 111: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 3

110

Mitta, B., Weber, C.C., and Fussenegger, M. (2005). In vivo transduction of HIV-1-derived

lentiviral particles engineered for macrolide-adjustable transgene expression. J Gene Med 7,

1400-1408.

Roncero, M.I. (1983). Genes controlling xylan utilization by Bacillus subtilis. J Bacteriol 156,

257-263.

Schlatter, S., Rimann, M., Kelm, J., and Fussenegger, M. (2002). SAMY, a novel mammalian

reporter gene derived from Bacillus stearothermophilus alpha-amylase. Gene 282, 19-31.

Searle, P.F., Stuart, G.W., and Palmiter, R.D. (1985). Building a metal-responsive promoter

with synthetic regulatory elements. Mol Cell Biol 5, 1480-1489.

Soole, K.L., Hirst, B.H., Hazlewood, G.P., Gilbert, H.J., Laurie, J.L., and Hall, J. (1993).

Secretion of a prokaryotic cellulase in bacterial and mammalian cells. Gene 125, 85-89.

Vickers, C.E., Xue, G.P., and Gresshoff, P.M. (2003). A synthetic xylanase as a novel

reporter in plants. Plant Cell Rep 22, 135-140.

Weber, W., Rimann, M., Spielmann, M., Keller, B., Daoud-El Baba, M., Aubel, D., Weber,

C.C., and Fussenegger, M. (2004). Gas-inducible transgene expression in mammalian cells

and mice. Nat Biotechnol 22, 1440-1444.

Yang, M., Baranov, E., Jiang, P., Sun, F.X., Li, X.M., Li, L., Hasegawa, S., Bouvet, M., Al-

Tuwaijri, M., Chishima, T., et al. (2000). Whole-body optical imaging of green fluorescent

protein-expressing tumors and metastases. Proc Natl Acad Sci U S A 97, 1206-1211.

Zhang, J.X., Krell, P.J., Phillips, J.P., and Forsberg, C.W. (1997). Expression of a bacterial

endo (1-4)-beta-glucanase gene in mammalian cells and post translational modification of the

gene product. Biochim Biophys Acta 1357, 215-224.

Zhang, J.X., Meidinger, R., Forsberg, C.W., Krell, P.J., and Phillips, J.P. (1999). Expression

and processing of a bacterial endoglucanase in transgenic mice. Arch Biochem Biophys 367,

317-321.

Zolotukhin, S., Byrne, B.J., Mason, E., Zolotukhin, I., Potter, M., Chesnut, K., Summerford,

C., Samulski, R.J., and Muzyczka, N. (1999). Recombinant adeno-associated virus

purification using novel methods improves infectious titer and yield. Gene Ther 6, 973-985.

Page 112: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

111

CHAPTER 4

A Novel System For Trigger-Controlled Drug Release

From Polymer Capsules

David A. Fluri, Christian Kemmer, Marie Daoud-El Baba and

Martin Fussenegger (2008)

submitted

Page 113: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

112

Abstract Technologies currently available for the controlled release of protein therapeutics

involve either continuous or tissue-specific discharge from implants or engineered

extracellular matrix mimetics. For some therapeutic applications the trigger-controlled release

of protein cargo from a synthetic implant could be highly desirable. We have designed the

CellEase technology, a two-component system consisting of cellulose sulfate (CS) poly-

dimethyl-diallylammonium chloride (pDADMAC) capsules harboring mammalian sensor

cells transgenic for trigger-inducible expression of an engineered secreted mammalian

cellulase (SecCell). SecCell is a Bacillus subtilis-derived (1-4)-β-glucanase, which was

modified by replacing the N-terminal part of the bacterial enzyme with a murine Igκ-chain V-

12-C region-derived secretion signal. SecCell was engineered for doxycycline- or

erythromycin-inducible expression to enable trigger-controlled secretion by mammalian cells.

Detailed characterization of SecCell showed that it was glycosylated and efficiently secreted

by a variety of mammalian sensor cells such that it could internally rupture CS-pDADMAC

capsules within which the cells had been encapsulated. When SecCell was inducibly

expressed in sender cells, that were co-encapsulated with producer cell lines expressing

therapeutic proteins, the removal of relevant inducer molecules enabled the time-dependent

discharge of these therapeutic proteins, the kinetics of which could be modified by varying

the concentration of inducer molecules or the amount of encapsulated sender cells. SecCell’s

capacity to rupture CS-pDADMAC capsules exclusively internally also enabled the

independent trigger-induced release of different proteins from two capsule populations

harboring different inducible SecCell sensor cells. CellEase-based protein release was

demonstrated in vivo using capsules implanted intraperitoneally into mice that enabled the

doxycycline-controlled release of a model glycoprotein and accumulation in the bloodstream

of treated animals. Trigger-induced breakdown of tissue-compatible implants which provide a

timely controlled release of biologics may foster novel opportunities in human therapy.

Introduction The controlled release of bioactive molecules from polymer matrices or polymer

capsules has been proposed as a promising approach in various therapeutic interventions in

order to avoid multiple dosing and to sustain continuous or pulsed release over time. The

Page 114: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

113

entrapment and immuno-isolation of small-molecule drugs, hormones, protein therapeutics or

cell lines engineered for production of biologics in the patient’s body have been designed for

the treatment of various diseases such as infections (Sanchez et al., 1996; Suzuki et al., 1998),

cancer (Lohr et al., 2002; Pelegrin et al., 1998; Read et al., 2001; Sakai et al., 2005), diabetes

(Chang, 1997; Stadlbauer et al., 2006; Wang et al., 1997) and different genetic disorders

(Hortelano et al., 1996; Ross et al., 2000).

Most controlled release systems currently available have either been chemically

designed for sustained auto-catalytic or tissue-specific discharge of the therapeutic cargo, or

engineered to release the therapeutic load in response to physical cues such as pH

(Balabushevich et al., 2003; Dejugnat and Sukhorukov, 2004), light (Skirtach et al., 2004),

ionic strength (Wang et al., 2005) magnetic resonance, (Edelman et al., 1985; Langer et al.,

1985) or an electric field (Kiser et al., 1998). Unfortunately, polymers designed for controlled

release are often limited in their chemical flexibility, while most physical stimuli are

impractical for in vivo applications. Also, the timing of release and overall release kinetics are

often difficult to control.

We have designed a versatile two-component controlled release technology (CellEase)

which consists of a biocompatible polymer capsule and a transgenic sensor cell line

engineered for trigger-inducible expression of a chimeric secreted cellulase which catalyses

capsule breakdown thereby enabling the release of biopharmaceuticals produced by co-

encapsulated producer cells.

Capitalizing on established cellulose sulfate (CS)-poly-diallyldimethyl ammonium

chloride (pDADMAC)-based polymerization chemistry, we have established a high-

throughput protocol for the production of cell- or biologics-containing microcapsules (Weber

et al., 2006). CS-pDADMAC polymers are known for their high biocompatibility (Pelegrin et

al., 1998; Wang et al., 1997), lack of cytotoxicity (Dautzenberg et al., 1999) and their cheap

and straightforward production protocols (Merten et al., 1991; Stiegler et al., 2006). Owing to

their covalent polymer structure and their chemistry, implanted CS-pDADMAC capsules are

inert to metabolic breakdown and survive for several months in vivo (Pelegrin et al., 1998;

Wang et al., 1997).

Cellulases, which can cleave the polymer backbone of CS-pDADMAC capsules, are

typically absent from mammalian tissues. We have engineered a Bacillus subtilis (1-4)-β-

glucanase (cellulase) by the N-terminal fusion to an IgG-type secretion signal sequence with

expression placed under control of the TET (Gossen and Bujard, 1992) or E.REX (Weber et

al., 2002) systems for trigger-inducible expression and secretion by mammalian cells.

Page 115: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

114

TET/E.REX are prototypic transgene control system which are responsive to clinically

licensed antibiotics (tetracycline/doxycycline, erythromycin) and consist of chimeric

transactivators (tTA/ET1), designed by fusing bacterial response regulators (TetR/E) to a

eukaryotic transactivation domain (VP16), which binds and activates promoters (PhCMV*-

1/PETR) containing transactivator-specific operator sites (tetO7/OETR) 5’ of minimal eukaryotic

promoters. In the presence of regulating antibiotics the transactivators are released from their

cognate promoters and transgene expression is silenced in a dose-dependent manner (Weber

and Fussenegger, 2006, 2007). TET and E.REX systems have been shown to be compatible

and could be used for independent control of different transgenes in a single mammalian cell

or mixed cell populations (Weber et al., 2002).

By co-encapsulating sensor cells, engineered for TET-/E.REX-controlled expression

of the secreted mammalian cellulase, with biologics-secreting production cell lines into CS-

pDADMAC capsule we could trigger-control capsule rupture and fine-tune release kinetics of

protein therapeutics in vitro as well as in mice. The CellEase technology may foster advances

in timely delivery of specific therapeutic doses in future gene therapy and tissue engineering

protocols.

Material and Methods Vector design. pAAV-MCS (PhCMV-Intronb-globin-pAhGH)[Stratagene, La Jolla, CA,

USA], pEF4-MycHisA (PhEF1α-MCS-TMYC-THIS-pAbGH) [Invitrogen, Carlsbad, CA, USA],

pIRESbleo (PhCMV-MCS-Intronsynthetic-IRESEMCV-Bleo-pAbGH) [Clontech, Palo Alto, CA,

USA], pPur (PSV40-Puro-pASV40) [Clontech], pSEAP2-control (PSV40-MCS-SEAP-pASV40-

ESV40; ESV40, enhancer of SV40 virus) [Clontech], pDF51 (PhCMV-Intronβ-globin-ET1-pAhGH)

(Fluri et al., 2007a), pDF75 (PETR-SEAP-IRESPV-ET1-pASV40) (Fluri et al., 2007a), pDF109

(PhCMV-Intronβ-globin-SEAP-pAhGH) (Fluri et al., 2007a) pDF191 (PhEF1α-SeXy-pAbGH) (Fluri et

al., 2007b), pMF111 (PhCMV*-1-SEAP-pASV40) (Fussenegger et al., 1997), pMF172 (PPIR-

SEAP-pASV40) (Fussenegger et al., 2000), pSAM200 (PSV40-tTA-pASV40) (Fussenegger et al.,

1997) and pWW35 (PSV40-ET1-pASV40) (Weber et al., 2002) have been described previously.

The cellulase of Bacillus subtilis 168 (GenBank accession no.: AY044252) was PCR-

amplified from genomic DNA using oligonucleotides ODF67: (5’-

CGGGATCCACCATGGAGACAGACACACTCCTGCTATGGGTACTGCTGCTCTGGGT

TCCAGGTTCCACTGGTGACGCAGGGACAAAAACGCCAGTAGCC; BamHI,

underlined; start codon in italics) and ODF68: (5’-

Page 116: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

115

GGAATTCTCATCTAGAATTTGGTTCTGTTCCCCAAAT; EcoRI and XbaI, underlined),

restricted with BamHI/XbaI and cloned into the corresponding sites (BamHI/XbaI) of pEF4-

MycHisA (pDF195 [PhEF1α-SecCell-pAbGH]). This fused the cellulase 5’ to the signal

sequence derived from the murine Igk-chain V-12-C region (SSIgκ) replacing the first 115

nucleotides of the open reading frame and 3’ to the Myc tag (TMYC) which resulted in a

secreted cellulase (SecCell; SSIgκ-CELLULASE-TMYC). SecCell was excised from pDF195 by

BamHI/PmeI and cloned into the compatible sites (BamHI/HincII) of pAAV-MCS resulting

in pDF196 (PhCMV-Intronβ-globin-SecCell-pAhGH). pDF260 (PETR-SecCell-pAhGH) was

assembled by restricting pDF196 with ClaI/BglII and ligating SecCell into the corresponding

sites (ClaI/BglII) of pDF85 (PETR-Intronβ-globin-SEAP-pAhGH). pDF85 had been constructed by

excising PETR from pDF75 with BssHII/NruI and cloning the insert into the MluI/BsaBI sites

of pDF109. pDF301 (PhCMV*-1-SecCell-pASV40) was constructed by a multistep cloning

procedure including (i) excision of SecCell from pDF196 using EcoRI/XhoI, (ii) subcloning

(EcoRI/XhoI) into pMF172 resulting in pDF300 (PPIR-SecCell-pASV40) and (iii) excising

SecCell from pDF300 with SpeI/HindIII and cloning the fragment into the compatible sites

(XbaI/HindIII) of pMF111. pDF323 (PhCMV-ET1-Intronsynthetic-IRESEMCV-Bleo-pAbGH was

assembled by excising ET1 from pDF51 (EcoRI/BglII) and inserting it (EcoRI/BamHI) into

pIRESbleo.

Cell culture, transfection and construction of stable cell lines. Human embryonic

kidney cells, transgenic for the adenovirus type 5-derived E1 region and the simian virus 40

(SV40) large T-antigen (HEK293-T; (Mitta et al., 2002)), human fibrosarcoma cells (HT-

1080; ATCC CCL-121), human cervical carcinoma cells (HeLa; ATCC CCL-2), baby

hamster kidney cells (BHK-21, ATCC CCL-10) and all HEK293-T derivatives (HEK-ET15,

HEK-tTA2, HEK-DF26011, HEK-DF3019) were cultivated in Dulbecco’s modified Eagle’s

medium (DMEM, Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal calf serum

(FCS; PAN Biotech GmbH, Aidenbach, Germany; cat. no. 3302-P251110, lot no. P251110)

and 1% penicillin/streptomycin solution (Sigma Chemicals, St. Louis, MO, USA). Chinese

hamster ovary cells (CHO-K1; ATCC CCL-61; CHO-B13-24; ATCC CRL-11397) and the

CHO-K1 derivative CHO-SEAP18 (see below) were cultivated in ChoMaster® HTS medium

(Cell Culture Technologies GmbH, Gravesano, Switzerland) supplemented with 5% FCS and

1% penicillin/streptomycin solution. HEK-ET15 was created by transfecting pDF323 into

HEK293-T and selecting for two weeks using 100µg/ml zeocin (Invitrogen) before clonal

selection in 96-well plates. HEK-tTA2 was engineered by co-transfecting pSAM200 and

Page 117: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

116

pIRES-bleo into HEK293-T and selection using 100!g/ml zeocin for two weeks before clonal

selection in 96-well plates. HEK-26011 was created by co-transfecting pDF260 and pPur into

HEK-ET15 and subsequent selection for two weeks using 1µg/ml puromycin. HEK-3019 was

engineered by co-transfecting pDF301 and pPur into HEK-tTA2 before selection for two

weeks using puromycin (Calbiochem, San Diego, CA, USA). Both HEK-26011 and HEK-3019

were screened clonally in 96-well plates for optimal regulation performance. CHO-SEAP18

was obtained by co-transfecting pPur and pSEAP2-control (Clontech) into CHO-K1 followed

by selection for two weeks in medium containing 10µg/ml puromycin and screening for

maximum human placental secreted alkaline phosphatase (SEAP) expression.

Capsules containing cells were cultivated in mixed medium consisting of 50% DMEM

(Invitrogen) and 50% HTS (Cell Culture Technologies GmbH) supplemented with 7.5% FCS

(PAN biotech GmbH).

Chemicals used for transgene regulation. For all in vitro experiments, erythromycin

(Sigma, E-5289) was dissolved in ethanol and used at a final concentration of 1µg/ml.

Doxycycline (Sigma, D-9891) was dissolved in PBS and used at a final concentration of

1µg/ml for in vitro experiments and at a final concentration of 25mg/kg for in vivo

experiments.

Quantification of cellulase, SEAP and antibodies. Cellulase was quantified using

EnzCheck® fluorescent substrate (Molecular Probes, Eugene, OR, USA) according to the

manufacturer’s protocol. In brief, 20µl of cell culture supernatant was centrifuged at 16,000xg

for one minute to remove cell debris and 15µl of appropriate dilutions in PBS (Invitrogen, cat.

no. 21600-069) were transferred to a 384-well plate containing 15µl of sodium acetate buffer

(200mM sodium acetate, pH 5.5) per well supplemented with 3µl of substrate solution. Plates

were incubated at 37°C for 30 minutes and fluorescence intensity was measured subsequently

at 340nm (excitation wavelength) and 450nm (emission wavelength) in a Genios Pro

multiwell plate reader (Tecan AG, Maennedorf, Switzerland). Absolute cellulase levels were

determined by comparing the fluorescence intensities to readings of standardized solutions

containing known concentrations of Aspergillus niger cellulase (Sigma, cat. no. 22178).

Quantification of human placental secreted alkaline phosphatase (SEAP) in the supernatant of

microencapsulated cells or in mouse serum was performed as described previously (Berger et

al., 1988). Anti CD18 IgG was quantified using a standard sandwich ELISA. In brief, high

protein-binding 96-well plates (Corning, NY, USA) were coated overnight with Fc-specific

anti-human IgG (Sigma, cat. no. I2136, lot no. 105K4774) before blocking for 2h with 1%

Page 118: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

117

BSA in PBS (Invitrogen). Diluted culture supernatants were added to individual wells,

incubated for 2h at 4°C before washing three times with 200µl PBS containing 0.02% Tween

and adding a secondary peroxidase-coupled anti-human IgG (Sigma, cat. no. A0170, lot no.

026K4784). After another three washing steps, chromogenic tetramethyl benzidine (TMB)

substrate (Interchim, Montluçon, France) was added and the reaction was stopped after 10

minutes by adding 50µl/well of 1M H2SO4 before measuring absorbance at 450nm in a multi-

well plate reader (TECAN). Absolute antibody levels were determined by comparing

absorbance readings to those for purified IgG antibody standard (gammanorm®, Octapharma

GmbH, Langenfeld, Germany).

Encapsulation. Cells were encapsulated using the Inotech Encapsulator IE-50R

(Inotech AG, Basel, Switzerland) according to the manufacturer’s protocol at the following

settings: 0.2mm nozzle, 20ml syringe at a flow rate of 410 units, a nozzle vibration frequency

of 1250s-1 and 1150V for bead dispersion. Cellulose sulfate (CS) (Euroferm GmbH, Erlangen,

Germany, batch no. FCY-71) was dissolved to 2% (w/v) in PBS and stirred for 16h. The

polymerization solution consisted of 1% Poly-diallyl-dimethyl-ammoniumchloride

(pDADMAC) (Euroferm) in PBS containing 0.0002% Tween-20. For encapsulation, the cells,

proteins or fluorescently labeled molecules were re-suspended at desired concentrations in CS

and transferred to a syringe before injecting the solution into the encapsulator which

generated CS droplets and sprayed them into the agitated polymerization solution where

product-containing CS-pDADMAC capsules formed. After hardening of the CS-pDADMAC

capsules, the polymerization solution was exchanged with PBS and the capsules were

incubated in tissue culture plates at 37°C and 5% CO2 in a humidified atmosphere.

Immunoblotting. Culture supernatants from transiently transfected HEK293-T were

mixed with 5x SDS loading buffer (50% glycerol, 10% SDS, 250mM Tris, pH 6.8 containing

10% (v/v) β-mercaptoethanol) and boiled for five minutes before loading onto a 10%

denaturing SDS-PAGE gel. Proteins were electroblotted onto polyvinylidene difluoride

(PVDF) membranes (Millipore, Bedford, MA, USA). After blocking in Tris buffered saline

(TBS, 50mM Tris, 150mM NaCl, pH7.5) containing 3% skimmed dry milk (Rapilait, Migros,

Switzerland), the membranes were incubated with a primary antibody specific for the C-myc

tag (Santa Cruz Biotechnology, Santa Cruz, CA, USA; cat. no. SC-40, lot no. E2207).

Specific bands were visualized using an anti-mouse IgG coupled to horseradish peroxidase

(ECLTM anti-mouse IgGxHRP, Amersham, Buckinghamshire, UK; cat. no. NA931V, lot no.

Page 119: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

118

357597) and a chemiluminescence-based assay (ECLTM plus, Amersham) according to the

manufacturer’s protocol.

Microscopy. Microscopic analysis and time-lapse movies were generated on a LEICA

DMI-6000 microscope equipped with appropriate filters for EYFP, FITC and Cy3 detection,

and a heated, humidified incubation chamber containing 5% CO2.

Animal studies. Female OF1 (oncins france souche 1) mice were obtained from

Charles River Laboratories (Lyon, France). 700µl of PBS solution containing 50% capsules

was administered intraperitoneally to mice. A PBS solution containing DOX was injected

every 24 hours. Blood samples were collected retroorbitally 3, 6 and 9 days after capsule

injection.

All experiments involving mice were performed according to the European

Community Council directive (86/609/EEC), approved by the French Ministry of Agriculture

and Fishery (Paris, France) and performed by M.D-E.-B. at the Institut Universitaire de

Technologie, IUTA, F-69622 Villeurbanne Cedex, France.

Results Design and characterization of a secreted mammalian cellulase

The Bacillus subtilis endo (1-4)-β-glucanase, was PCR-amplified from genomic DNA

and fused to a strong mammalian secretion signal sequence derived from the murine Igκ-

chain V-12-C region. The secreted cellulase (SecCell) was cloned downstream of a

constitutive promoter (PhCMV) as well as two different inducible promoters which enabled

doxycycline- (PhCMV*-1) and erythromycin- (PETR) adjustable transgene expression (Fig. 1A).

Transient transfection of SecCell into different human (HEK293-T, HeLa, HT-1080) and

hamster (CHO-K1 and BHK-21) cell lines resulted in high-level cellulase activity in the

culture supernatants unless SecCell production was repressed by doxycycline (DOX) or

erythromycin (EM) (Fig. 1B).

The apparent size of the modified cellulase as well as possible glycosylation was

assayed by producing SecCell in HEK293-T in the presence and absence of the glycosylation

inhibitor tunicamycin. Western blot analysis of culture supernatants showed that the SecCell

produced from tunicamycin-free cultures had a molecular weight of 55kDa which was lower

when the cells were treated with tunicamycin. This suggested that the cellulase was

glycosylated in mammalian cells (Fig. 1C).

Page 120: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

119

Cellulase properties were further explored by assaying enzymatic activity after

incubation at different temperatures. Activity was relatively constant between 30 and 55°C

but dropped sharply upon incubation above 60°C (Fig. 1D). The stability of the modified

cellulase in human serum was also assayed by mixing HEK293-T-produced enzyme with

human AB serum before incubating the mixture for a total period of 12 days during which

samples were taken every 96 hours (Fig. 1E). The enzymatic half-life was calculated by linear

regression of decreasing enzymatic activities to be around 42 days, which is comparable to

other secreted proteins of bacterial origin (Fluri et al., 2007b; Schlatter et al., 2002).

Page 121: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

120

Figure 1. Transient expression of modified B. Subtilis cellulase in mammalian cells. (A) Schematic

representation of vectors used for the CellEase system. (B) Transient expression of cellulase in HEK293-T,

HeLa, HT-1080, BHK-21 and CHO-K1 cells. Cellulase was either driven by a constitutive (PhCMV) or antibiotic-

responsive promoters (PhCMV*-1 and PETR). (C) Western blot analysis of cell culture supernatants derived from

HEK293-T populations transiently transfected with (i) pDF196 and cultivated in the presence or absence of the

glycosylation inhibitor tunicamycin (2!g/ml), (ii) pDF301/pSAM200 (+/- DOX [2!g/ml]) or (iii) mock-

transfected to provide a negative control.

Abbreviations: DOX, doxycycline; ET1, erythromycin-dependent transactivator; pAhgh, polyadenylation signal

of the human growth hormone; pASV40, polyadylation signal of the simian virus 40; PhCMV*-1, tetracycline-

responsive promoter; PETR, erythromycin-responsive promoter; PhCMV, human cytomegalovirus immediate early

promoter; PSV40, simian virus 40 promoter; SSIgκ, signal sequence derived from the murine Igk-chain V-12-C

region; tTA, tetracycline-dependent transactivator; Tmyc protein tag encoding a c-myc epitope

Page 122: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

121

Capsule properties and rupture characteristics

Molecular weight cut-off (MWCO): Cellulose sulfate capsules were incubated for 16

hours in the presence of FITC-dextran solutions of different molecular weight. After

incubation, capsules were washed thoroughly and then analyzed by fluorescence microscopy.

Fluorescence micrographs show strong signals for the two FITC-dextrans of lower molecular

weight (10kDA and 20kDA) and no signal for FITC-dextrans of higher molecular weight

(40kDa, 70kDa), indicating a molecular weight cut-off between 20 and 40kDa for the 2%

capsules (Fig. 2A).

Upon incubation of 2% CS-containing capsules for 30 minutes at 37°C with cellulases

of the same target specificity, either Aspergillus niger cellulase (MW 25kDa, below molecular

cut-off) or SecCell (50kDa, above molecular cut-off) only the A. niger cellulase was able to

trigger capsule rupture at concentrations as low as 1U/ml. The larger-sized SecCell which is

above the MWCO and unable to penetrate the capsules failed to induce capsule degradation

even at 5U/ml (Fig. 2B). This observation suggests that capsule breakdown can only be

initiated from the inside and not from the outside which would require SecCell to be co-

encapsulated or produced inside the capsules in order to control capsule breakdown and

release of microencapsulated drugs in a robust and reliable manner. Although the underlying

mechanistic details of SecCell’s inside-specific capsule breakdown characteristics remain

elusive, this quality may enable sequential or independent release of various therapeutic

cargos using different stimuli (see below).

CellEase technology – trigger-inducible capsule rupture and protein release

In order to establish and characterize CellEase technology for trigger-inducible release of

protein drugs we have co-encapsulated HEK-3019 (5x105 cells/ml of CS), transgenic for

tetracycline-responsive SecCell expression, and CHO-SEAP18 (2.5x105 cells/ml of CS),

engineered for constitutive SEAP (58 kDa, above MWCO) production into CS-pDADMAC

capsules and cultivated them for 6 days in the presence or absence of doxycycline (DOX).

Control capsules contained the same number of the parental cell line HEK293-T instead of the

SecCell producing HEK-3019 and CHO-SEAP18. Every 24 hours, capsule micrographs were

taken and accumulated SEAP activity was assayed in the supernatant.

Page 123: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

122

Figure 2. Membrane properties of CS-pDADMAC capsules. (A) CS capsules were incubated overnight in the

presence of FITC-dextrans with different molecular weight and then washed and analyzed by fluorescence

microscopy. (B) CS-pDADMAC capsules were incubated for 30 min in the presence of increasing

concentrations of either a fungal cellulase (MW 25kDa) or SecCell derived from mammalian culture

supernatants (MW 55kDa). The percentage of intact capsules was normalized to capsules which had not been

exposed to any cellulase.

Page 124: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

123

Capsules cultivated in the absence of DOX (maximum induction of SecCell in HEK-

3019) started to collapse after an initial lag phase of around 36 hours after encapsulation,

leading to a strong increase in SEAP activity in the supernatant compared to intact capsules

cultivated in the presence of DOX (repression of SecCell in HEK-3019) (Fig. 3A and 3B).

The control capsules containing CHO-SEAP18 cells together with parental HEK293-T

behaved in exactly the same manner as the CellEase capsules containing HEK-3019 in which

SecCell was repressed by doxycycline (Fig. 3A and 3B).

The induction of cellulase expression lead to destabilization and rupture of the

capsules followed by discharge of their contents. To illustrate the capsule breakdown, two

time-lapse microscopy experiments were performed; they revealed (i) the immediate rupture

of a cellulose sulfate capsule after addition of cellulase solution (A. Niger cellulase MW.

25kDA) http://www.fussenegger.ethz.ch/people/flurida/movie/movie1 and (ii) that the rupture

process was triggered after 36h by encapsulated HEK-3019. Capsules with SecCell production

fully induced were traced by co-encapsulating 150kDa FITC-dextran whereas negative-

control capsules containing HEK293-T were not fluorescently labeled

http://www.fussenegger.ethz.ch/people/flurida/movie/movie2.

Adjusting protein release by fine-tuning capsule rupture kinetics

We also sought to control capsule rupture by applying an external stimulus at a

defined point in time, which would lead to the induction of SecCell production and liberation

of the therapeutic cargo at a later required time. Four groups of identical capsules, containing

HEK-3019 (5x105 cells/ml of CS) and CHO-SEAP18 (2.5x105 cells/ml of CS), were cultivated

with SecCell expression de-repressed sequentially by DOX removal either (i) immediately

after (group 1), (ii) 24h after (group 2) or (iii) 48 h after encapsulation and seeding (group 3).

Group 4 was cultivated in the continued presence of doxycycline and served as a baseline to

illustrate the tightness of the CellEase system. The cumulative SEAP activity of the

supernatants of the four groups was monitored for six days (Fig. 4A). An increase in

supernatant SEAP activity reflected the induction time frame of the respective capsule

population and demonstrated timely controlled induction of capsule rupture.

Page 125: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

124

Figure 3. Capsule rupture induced by DOX-controlled SecCell expression. (A) CHO-SEAP18 were co-

encapsulated with HEK-3019, and capsules were cultivated in the presence (2µg/ml) or absence of DOX for 6

days. Light micrographs of capsule populations were taken at 24h intervals to profile capsule integrity. (B)

Cumulative SEAP activity in the supernatant was measured every 24h. Capsule populations containing CHO-

SEAP18 together with parental HEK293-T served as a negative control. Time-lapse movies showing capsule

rupture following addition of 5U/ml A. niger cellulase

(http://www.fussenegger.ethz.ch/people/flurida/movie/movie1) or DOX-responsive SecCell induction

(http://www.fussenegger.ethz.ch/people/flurida/movie/movie2) can be viewed online.

Another option to control the time of release is the titration of SecCell expression by

using different DOX concentrations. Capsules containing HEK-3019 (5x105 cells/ml of CS)

and CHO-SEAP18 (2.5x105 cells/ml of CS) were seeded into a 24-well plate and incubated

with 0, 0.1, 0.5, 2, and 20ng/ml DOX. Again, the cumulative SEAP activity of the

supernatants was scored for 6 days. Capsule rupture and SEAP accumulation in the

supernatant took progressively longer as DOX concentrations increased (from 0 ng/ml to 2

ng/ml). For the two highest DOX concentrations (2ng/ml and 20ng/ml), SEAP readings in the

supernatant dropped to background levels indicating full repression of SecCell expression

(Fig. 4B).

Page 126: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

125

As a third option to control SecCell-mediated protein release from cellulose

sulfate/pDADMAC capsules, we have varied the numbers of HEK-3019 responder cells

(5x104/ml of CS, 5x105/ml of CS and 1x106/ml of CS) co-encapsulated with a constant

amount of CHO-SEAP18 (2.5x105 cells/ml of CS). SEAP accumulation profiles were again

compared in repressed (+ DOX) and induced (- DOX) SecCell expression states for six days

(Fig. 4C). Encapsulation of higher numbers of the responder cell line HEK-3019 lead to a

quicker response to the stimulus, but also slightly increased the overall leakiness of the

CellEase system possibly due to capsule rupture as a result of leaky cellulase expression from

the tetracycline-responsive promoter.

Co-cultivation of different capsule populations

For various applications such as a single-shot vaccine or temporally spaced, treatments

we sought to test whether it would be possible to deliver a mixed capsule population, and to

release the cargo at different points in time by inducing release with different stimuli. The

capacity of the CellEase system to control the release of different molecules after inducing

capsule rupture by two different antibiotics was investigated. We encapsulated (i) HEK-3019

(5x105 cells/ml of CS) together with CHO-SEAP18 (2.5x105 cells/ml of CS) for one capsule

population, and (ii) HEK-26011 (0.75x105 cells/ml of CS) engineered for macrolide-

responsive SecCell expression, together with IgG-producing CHO-B13-24 cells (5x105

cells/ml of CS) for the other. The two populations were mixed in a 1:1 ratio and incubated

under various antibiotic conditions. To distinguish the two capsule populations in the mixture

the HEK-3019/CHO-SEAP18-containing capsules were stained with FITC-dextran (150kDa)

whereas the HEK-26011/CHO-B13-24-containing capsules were stained with

tetramethylrhodamine isothiocyanate (TRITC)(150kDa). The mixed capsule populations were

seeded into four different groups which were incubated in the absence or presence of one or

both antibiotics (DOX and EM) for five days. Accumulated SEAP and anti CD-18 antibody

were measured in the culture supernatants after 5 days (Fig 5A). SEAP and IgG levels

indicated that the respective release from the capsules was entirely controlled by the presence

or absence of either antibiotic. Exclusive rupture of one capsule population did not appear to

influence the other one since SecCell can only degrade capsules internally. Capsule integrity

was also analyzed by microscopy 5 days after incubation. Bright-field and fluorescence

micrographs showed exclusive rupture of capsule populations harboring cells with induced

SecCell and confirmed the integrity of capsules containing HEK-3019 or HEK-26011with

repressed SecCell (Fig 5B).

Page 127: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

126

Figure 4. Modulation of release patterns using the CellEase system. (A) CHO-SEAP18 were co-encapsulated

with HEK-3019 and capsule rupture was induced at different points in time (0, 24 and 48 hours) after

encapsulation. Cumulative SEAP activity in the supernatant was then analyzed every 24h for the next 6 days and

compared to readings for capsules cultivated in the presence of doxycycline (DOX) (SecCell production

repressed) for the entire period of time. (B) CHO-SEAP18, co-encapsulated with HEK-3019 and incubated at

different DOX concentrations ( 20ng/ml, 2ng/ml, 0.5ng/ml, 0.1ng/ml and 0ng/ml). SEAP

accumulation in the supernatant was measured every 24 hours for 6 days. (C) CHO-SEAP18 was co-encapsulated

with different numbers of cells (5x104 cells/ml, 5x105 cells/ml and 1x106 cells/ml) of HEK-3019 and incubated in

the presence and absence of DOX for 6 days. Cumulative SEAP activity was measured in the supernatant every

24h.

Page 128: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

127

Figure 5. Co-cultivation of two different capsule populations. SEAP-producing CHO-SEAP18 were co-

encapsulated with HEK3019 and a high molecular weight FITC-Dextran (150 kDa), whereas antibody-producing

CHO-B13-24 were co-encapsulated with HEK-26011 and a high molecular weight TRITC-Dextran (150kDa).

The two capsule populations were subsequently mixed at a ratio of 1:1 and cultivated under different antibiotic

conditions for 5 days. (A) SEAP and IgG levels in the culture supernatant were analyzed for the different

conditions 120 hours after seeding the capsules. (B) Integrity of the two capsule populations was shown by

bright field (upper row) and fluorescence micrographs (lower two rows) taken 120 hours after capsule seeding.

Abbreviations: BF, bright field; FITC, fluorescein isohiocyanate; TRITC, tetramethylrhodamine isothiocyanate.

Page 129: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

128

In vivo release of a model glycoprotein upon withdrawal of doxycycline

Capsules containing HEK-3019 (5x105 cells/ml of CS), transgenic for doxycycline-

responsive SecCell expression, and 100µl SEAP (2U/ml) concentrated from serum free CHO-

SEAP18 supernatants were injected intraperitoneally into two groups of mice. One group was

given daily doxycycline injections (+DOX) to maintain CellEase in a repressed state, whereas

the second group was left untreated to induce capsule rupture (Fig. 6A). Parallel to the in vivo

experiment, the same capsule populations were cultivated in the presence and absence of

DOX in petri dishes and release profiles were compared to the in vivo situation (Fig. 6B).

Figure 6. Antibiotic-controlled release of capsule cargo in mice. (A) SEAP activity in mouse serum 3, 6 and 9

days after intraperitoneal injection of capsules containing concentrated SEAP and HEK-3019 and intraperitoneal

administration of doxycycline (+DOX) or PBS (-DOX). (B) At the same time, capsule populations were cultured

in vitro and SEAP expression levels in the supernatant were analyzed every 24 hours. Cell culture medium was

exchanged and capsules were washed daily after sample removal.

Page 130: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

129

Discussion Therapeutic interventions using novel drug carriers are being continuously designed to

overcome limitations in traditional drug delivery. To avoid multiple dosing and to sustain a

therapeutic level of the active drug over a prolonged period of time, different approaches have

been established: (i) drug-containing implants (Langer, 1990; Richards Grayson et al., 2003;

Saito et al., 2001), (ii) micro-pumps (Ryu et al., 2007; Webb et al., 2002), (iii) the

encapsulation of cell lines or primary cells continuously producing biologics (Pelegrin et al.,

1998; Regulier et al., 1998; Sailaja et al., 2002; Wang et al., 1997), (iv) biodegradable

polymers containing embedded therapeutic substances (Jain, 2000; Mathiowitz et al., 1997) or

(v) implants of drug-containing capsules designed to rupture in response to biological,

chemical or physical stimuli (Angelatos et al., 2005; De Geest et al., 2005; Itoh et al., 2006;

Kiser et al., 1998).

Active bacterial cellulases can be efficiently expressed in mammalian cells without

showing significant cytotoxicity (Soole et al., 1993; Zhang et al., 1997; Zhang et al., 1999).

Enzyme-induced breakdown of physiologically inert polymer capsules triggering the release

of the encapsulated material represents a powerful tool to precisely adjust time and delivery

kinetics of biologics to therapeutic requirements. We have successfully combined

encapsulation technology adapted for cellulose sulfate/pDADMAC polymers, which have a

track record in production scalability (Dautzenberg et al., 1999; Stadlbauer et al., 2006)

biocompatibility (Dautzenberg et al., 1999) and metabolic stability (Lohr et al., 2001; Wang

et al., 1997), with tight expression control of a mammalian designer cellulase using two

prototype transcription control systems (TET and E.REX (Gossen and Bujard, 1992; Weber et

al., 2002). This CellEase technology enabled precise and controllable rupture of the cellulose-

containing polymer shell and timely release of therapeutic content.

By varying the relative cellulose-sulfate/pDADMAC concentrations and production

parameters capsule porosity could in principle be tuned for selective retention of specific

biologics while sustaining cell growth and viability by a free flow of nutrients and waste

product across the capsule membrane. We have chosen to use a molecular weight cut-off

between 25 and 40kDa which enables retention of most protein therapeutics including IgGs.

CellEase capsules could either accommodate biologics or cells producing desired protein

therapeutics. Microencapsulation of therapeutic proteins requires separate production and

downstream processing efforts making drug delivery as expensive as classic injection-based

therapies. In-situ production of biologics by microencapsulated cells alleviates classical

Page 131: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

130

biopharmaceutical manufacturing which could make therapy more efficient and affordable.

Although the fate of the sensor and producer cells released into the body might be considered

problematic, our studies using xenotypic cell lines in mice did not reveal any immediate

adverse effects in the animal. Alternatively, the use of autologous cells may completely

eliminate concerns about side effects of heterologous cell material. The recently developed

protein-transducing nanoparticles also enable simultaneous production and encapsulation by

packaging of therapeutic proteins into lentivirus-derived nucleic acid-free nanoparticles (Link

et al., 2006) However, in comparison to CellEase technology, protein-transducing

nanoparticles release their therapeutic cargo in an uncontrolled manner directly upon contact

into any cells, which, unlike CellEase, lacks release control and limits therapeutic impact to

intracellular targets (Link et al., 2006).

CellEase technology is relatively straightforward, robust and advantageous for the

following reasons: (i) CS-pDADMAC capsules can be produced at low cost and large scale

using multi-nozzle devices for production of clinical-grade capsules (Lohr et al., 1999;

Salmons et al., 2003) (ii) Capsule parameters such as molecular weight cut-off and cellulose

content could easily be varied to modify growth and production characteristics inside capsules

and adjust release kinetics. (iii) The availability of an engineered ready-to-use sensor cell line

transgenic for trigger-controlled SecCell production increases flexibility as this cell line can

be co-encapsulated with any established constitutive producer cell line or primary cell via co-

encapsulation. We have shown that the relative number of encapsulated sensor and producer

cells can be used to adjust drug-release kinetics. For one-cell line solutions producer cells

could also be engineered for regulated SecCell expression. (iv) Having used the TET and

E.REX systems to trigger SecCell expression, capsule rupture and biologics release, we

assume that CellEase is compatible with any transcription control system (Weber and

Fussenegger, 2007). (v) Release kinetics can be modified by timing and dosing of the SecCell

trigger molecules. (vi) The unique characteristic of SecCell to exclusively breakdown CS-

pDADMAC capsules internally and leave the outer surface of the capsule intact makes

CellEase technology suitable for the administration of mixed capsule populations, which

differ, for instance, in the type and number of sensor cells, thereby allowing the controlled

release of different molecules at different points in time and with different release kinetics

without having to repeat capsule administration. (vii) Although we have used antibiotics as a

heterologous inducer in vivo it may also become possible to wire CellEase to endogenous

signals, which are increased during a disease or infection state (e.g., hormone or interleukins),

Page 132: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

131

to trigger release of therapeutic biologics and provide a rapid and self-sufficient first-line

protection.

By providing in-situ production of biologics and being robust, precise, versatile and

functional in vivo, CellEase technology has the potential to make future drug delivery more

efficient and affordable.

Conclusion In this work we present a new approach for the trigger-induced release of biologics

from biocompatible CS-pDADMAC capsules. Capsule rupture and the accompanying cargo

release was controlled by the inducible expression of a modified bacterial cellulase (SecCell)

from mammalian sensor cells. We have proved the functionality of the system by engineering

sensor cells that express SecCell under the control of either doxycycline or erythromycin

responsive promoters. After induction, with the corresponding antibiotic, SecCell was

expressed and secreted from the sensor cells and caused the destabilization of the capsule

from the inside, rupturing the capsule and releasing its contents. Model glycoproteins, either

expressed by co-encapsulated cells or co-encapsulated purified factors, were able to be

released from capsules upon induction in vitro as well as in vivo. This technology may open

new possibilities for trigger-induced release of therapeutic cargos. And by combining

CellEase with available gene-regulation systems one can create the tools to utilize a broad

variety of cues to control the release of biologics in therapeutic settings.

Acknowledgements We thank Martine Gilet for skillful assistance in animal experimentation and

Domenick Grasso as well as David Greber for critical comments on the manuscript. This

work was in part supported by the Swiss National Science Foundation (3200-64488).

Page 133: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

132

References Angelatos, A.S., Radt, B., and Caruso, F. (2005). Light-responsive polyelectrolyte/gold

nanoparticle microcapsules. The journal of physical chemistry 109, 3071-3076.

Balabushevich, N.G., Tiourina, O.P., Volodkin, D.V., Larionova, N.I., and Sukhorukov, G.B.

(2003). Loading the multilayer dextran sulfate/protamine microsized capsules with

peroxidase. Biomacromolecules 4, 1191-1197.

Berger, J., Hauber, J., Hauber, R., Geiger, R., and Cullen, B.R. (1988). Secreted placental

alkaline phosphatase: a powerful new quantitative indicator of gene expression in eukaryotic

cells. Gene 66, 1-10.

Chang, P.L. (1997). Microcapsules as bio-organs for somatic gene therapy. Annals of the

New York Academy of Sciences 831, 461-473.

Dautzenberg, H., Schuldt, U., Grasnick, G., Karle, P., Muller, P., Lohr, M., Pelegrin, M.,

Piechaczyk, M., Rombs, K.V., Gunzburg, W.H., et al. (1999). Development of cellulose

sulfate-based polyelectrolyte complex microcapsules for medical applications. Annals of the

New York Academy of Sciences 875, 46-63.

De Geest, B.G., Dejugnat, C., Sukhorukov, G.B., Braeckmans, K., De Smedt, S.C., and

Demeester, J. (2005). Self-rupturing microcapsules. Advanced Materials 17, 2357-+.

Dejugnat, C., and Sukhorukov, G.B. (2004). pH-responsive properties of hollow

polyelectrolyte microcapsules templated on various cores. Langmuir 20, 7265-7269.

Edelman, E.R., Kost, J., Bobeck, H., and Langer, R. (1985). Regulation of drug release from

polymer matrices by oscillating magnetic fields. Journal of biomedical materials research 19,

67-83.

Fluri, D.A., Baba, M.D., and Fussenegger, M. (2007a). Adeno-associated viral vectors

engineered for macrolide-adjustable transgene expression in mammalian cells and mice. BMC

biotechnology 7, 75.

Fluri, D.A., Kelm, J.M., Lesage, G., Baba, M.D., and Fussenegger, M. (2007b). InXy and

SeXy, compact heterologous reporter proteins for mammalian cells. Biotechnology and

bioengineering 98, 655-667.

Fussenegger, M., Morris, R.P., Fux, C., Rimann, M., von Stockar, B., Thompson, C.J., and

Bailey, J.E. (2000). Streptogramin-based gene regulation systems for mammalian cells.

Nature biotechnology 18, 1203-1208.

Page 134: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

133

Fussenegger, M., Moser, S., Mazur, X., and Bailey, J.E. (1997). Autoregulated multicistronic

expression vectors provide one-step cloning of regulated product gene expression in

mammalian cells. Biotechnology progress 13, 733-740.

Gossen, M., and Bujard, H. (1992). Tight control of gene expression in mammalian cells by

tetracycline-responsive promoters. Proceedings of the National Academy of Sciences of the

United States of America 89, 5547-5551.

Hortelano, G., Al-Hendy, A., Ofosu, F.A., and Chang, P.L. (1996). Delivery of human factor

IX in mice by encapsulated recombinant myoblasts: a novel approach towards allogeneic gene

therapy of hemophilia B. Blood 87, 5095-5103.

Itoh, Y., Matsusaki, M., Kida, T., and Akashi, M. (2006). Enzyme-responsive release of

encapsulated proteins from biodegradable hollow capsules. Biomacromolecules 7, 2715-2718.

Jain, R.A. (2000). The manufacturing techniques of various drug loaded biodegradable

poly(lactide-co-glycolide) (PLGA) devices. Biomaterials 21, 2475-2490.

Kiser, P.F., Wilson, G., and Needham, D. (1998). A synthetic mimic of the secretory granule

for drug delivery. Nature 394, 459-462.

Langer, R. (1990). New methods of drug delivery. Science (New York, NY 249, 1527-1533.

Langer, R., Siegel, R., Brown, L., Leong, K., Kost, J., and Edelman, E. (1985). Controlled

release and magnetically modulated systems for macromolecular drugs. Annals of the New

York Academy of Sciences 446, 1-13.

Link, N., Aubel, C., Kelm, J.M., Marty, R.R., Greber, D., Djonov, V., Bourhis, J., Weber, W.,

and Fussenegger, M. (2006). Therapeutic protein transduction of mammalian cells and mice

by nucleic acid-free lentiviral nanoparticles. Nucleic acids research 34, e16.

Lohr, M., Bago, Z.T., Bergmeister, H., Ceijna, M., Freund, M., Gelbmann, W., Gunzburg,

W.H., Jesnowski, R., Hain, J., Hauenstein, K., et al. (1999). Cell therapy using

microencapsulated 293 cells transfected with a gene construct expressing CYP2B1, an

ifosfamide converting enzyme, instilled intra-arterially in patients with advanced-stage

pancreatic carcinoma: a phase I/II study. Journal of molecular medicine (Berlin, Germany)

77, 393-398.

Lohr, M., Hoffmeyer, A., Kroger, J., Freund, M., Hain, J., Holle, A., Karle, P., Knofel, W.T.,

Liebe, S., Muller, P., et al. (2001). Microencapsulated cell-mediated treatment of inoperable

pancreatic carcinoma. Lancet 357, 1591-1592.

Lohr, M., Hummel, F., Faulmann, G., Ringel, J., Saller, R., Hain, J., Gunzburg, W.H., and

Salmons, B. (2002). Microencapsulated, CYP2B1-transfected cells activating ifosfamide at

Page 135: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

134

the site of the tumor: the magic bullets of the 21st century. Cancer chemotherapy and

pharmacology 49 Suppl 1, S21-24.

Mathiowitz, E., Jacob, J.S., Jong, Y.S., Carino, G.P., Chickering, D.E., Chaturvedi, P.,

Santos, C.A., Vijayaraghavan, K., Montgomery, S., Bassett, M., et al. (1997). Biologically

erodable microspheres as potential oral drug delivery systems. Nature 386, 410-414.

Merten, O.W., Dautzenberg, H., and Palfi, G.E. (1991). A new method for the encapsulation

of mammalian cells. Cytotechnology 7, 121-130.

Mitta, B., Rimann, M., Ehrengruber, M.U., Ehrbar, M., Djonov, V., Kelm, J., and

Fussenegger, M. (2002). Advanced modular self-inactivating lentiviral expression vectors for

multigene interventions in mammalian cells and in vivo transduction. Nucleic Acids Res 30,

e113.

Pelegrin, M., Marin, M., Noel, D., Del Rio, M., Saller, R., Stange, J., Mitzner, S., Gunzburg,

W.H., and Piechaczyk, M. (1998). Systemic long-term delivery of antibodies in

immunocompetent animals using cellulose sulphate capsules containing antibody-producing

cells. Gene therapy 5, 828-834.

Read, T.A., Sorensen, D.R., Mahesparan, R., Enger, P.O., Timpl, R., Olsen, B.R., Hjelstuen,

M.H., Haraldseth, O., and Bjerkvig, R. (2001). Local endostatin treatment of gliomas

administered by microencapsulated producer cells. Nature biotechnology 19, 29-34.

Regulier, E., Schneider, B.L., Deglon, N., Beuzard, Y., and Aebischer, P. (1998). Continuous

delivery of human and mouse erythropoietin in mice by genetically engineered polymer

encapsulated myoblasts. Gene therapy 5, 1014-1022.

Richards Grayson, A.C., Choi, I.S., Tyler, B.M., Wang, P.P., Brem, H., Cima, M.J., and

Langer, R. (2003). Multi-pulse drug delivery from a resorbable polymeric microchip device.

Nature materials 2, 767-772.

Ross, C.J., Ralph, M., and Chang, P.L. (2000). Somatic gene therapy for a neurodegenerative

disease using microencapsulated recombinant cells. Experimental neurology 166, 276-286.

Ryu, W., Huang, Z., Prinz, F.B., Goodman, S.B., and Fasching, R. (2007). Biodegradable

micro-osmotic pump for long-term and controlled release of basic fibroblast growth factor. J

Control Release 124, 98-105.

Sailaja, G., HogenEsch, H., North, A., Hays, J., and Mittal, S.K. (2002). Encapsulation of

recombinant adenovirus into alginate microspheres circumvents vector-specific immune

response. Gene therapy 9, 1722-1729.

Page 136: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

135

Saito, N., Okada, T., Horiuchi, H., Murakami, N., Takahashi, J., Nawata, M., Ota, H., Nozaki,

K., and Takaoka, K. (2001). A biodegradable polymer as a cytokine delivery system for

inducing bone formation. Nature biotechnology 19, 332-335.

Sakai, S., Kawabata, K., Tanaka, S., Harimoto, N., Hashimoto, I., Mu, C., Salmons, B., Ijima,

H., and Kawakami, K. (2005). Subsieve-size agarose capsules enclosing ifosfamide-activating

cells: a strategy toward chemotherapeutic targeting to tumors. Molecular cancer therapeutics

4, 1786-1790.

Salmons, B., Lohr, M., and Gunzburg, W.H. (2003). Treatment of inoperable pancreatic

carcinoma using a cell-based local chemotherapy: results of a phase I/II clinical trial. Journal

of gastroenterology 38 Suppl 15, 78-84.

Sanchez, A., Gupta, R.K., Alonso, M.J., Siber, G.R., and Langer, R. (1996). Pulsed

controlled-released system for potential use in vaccine delivery. Journal of pharmaceutical

sciences 85, 547-552.

Schlatter, S., Rimann, M., Kelm, J., and Fussenegger, M. (2002). SAMY, a novel mammalian

reporter gene derived from Bacillus stearothermophilus alpha-amylase. Gene 282, 19-31.

Skirtach, A.G., Antipov, A.A., Shchukin, D.G., and Sukhorukov, G.B. (2004). Remote

activation of capsules containing Ag nanoparticles and IR dye by laser light. Langmuir 20,

6988-6992.

Soole, K.L., Hirst, B.H., Hazlewood, G.P., Gilbert, H.J., Laurie, J.L., and Hall, J. (1993).

Secretion of a prokaryotic cellulase in bacterial and mammalian cells. Gene 125, 85-89.

Stadlbauer, V., Stiegler, P.B., Schaffellner, S., Hauser, O., Halwachs, G., Iberer, F.,

Tscheliessnigg, K.H., and Lackner, C. (2006). Morphological and functional characterization

of a pancreatic beta-cell line microencapsulated in sodium cellulose

sulfate/poly(diallyldimethylammonium chloride). Xenotransplantation 13, 337-344.

Stiegler, P.B., Stadlbauer, V., Schaffellner, S., Halwachs, G., Lackner, C., Hauser, O., Iberer,

F., and Tscheliessnigg, K. (2006). Cryopreservation of insulin-producing cells

microencapsulated in sodium cellulose sulfate. Transplantation proceedings 38, 3026-3030.

Suzuki, Y., Tanihara, M., Nishimura, Y., Suzuki, K., Kakimaru, Y., and Shimizu, Y. (1998).

A new drug delivery system with controlled release of antibiotic only in the presence of

infection. Journal of biomedical materials research 42, 112-116.

Wang, T., Lacik, I., Brissova, M., Anilkumar, A.V., Prokop, A., Hunkeler, D., Green, R.,

Shahrokhi, K., and Powers, A.C. (1997). An encapsulation system for the immunoisolation of

pancreatic islets. Nature biotechnology 15, 358-362.

Page 137: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Chapter 4

136

Wang, Y., Yu, A., and Caruso, F. (2005). Nanoporous Polyelectrolyte Spheres Prepared by

Sequentially Coating Sacrificial Mesoporous Silica Spheres. Angewandte Chemie

(International ed 44, 2888-2892.

Webb, G.C., Akbar, M.S., Zhao, C., Swift, H.H., and Steiner, D.F. (2002). Glucagon

replacement via micro-osmotic pump corrects hypoglycemia and alpha-cell hyperplasia in

prohormone convertase 2 knockout mice. Diabetes 51, 398-405.

Weber, W., and Fussenegger, M. (2006). Pharmacologic transgene control systems for gene

therapy. The journal of gene medicine 8, 535-556.

Weber, W., and Fussenegger, M. (2007). Inducible product gene expression technology

tailored to bioprocess engineering. Current opinion in biotechnology 18, 399-410.

Weber, W., Fux, C., Daoud-el Baba, M., Keller, B., Weber, C.C., Kramer, B.P., Heinzen, C.,

Aubel, D., Bailey, J.E., and Fussenegger, M. (2002). Macrolide-based transgene control in

mammalian cells and mice. Nature biotechnology 20, 901-907.

Weber, W., Rimann, M., Schafroth, T., Witschi, U., and Fussenegger, M. (2006). Design of

high-throughput-compatible protocols for microencapsulation, cryopreservation and release of

bovine spermatozoa. Journal of biotechnology 123, 155-163.

Zhang, J.X., Krell, P.J., Phillips, J.P., and Forsberg, C.W. (1997). Expression of a bacterial

endo (1-4)-beta-glucanase gene in mammalian cells and post translational modification of the

gene product. Biochimica et biophysica acta 1357, 215-224.

Zhang, J.X., Meidinger, R., Forsberg, C.W., Krell, P.J., and Phillips, J.P. (1999). Expression

and processing of a bacterial endoglucanase in transgenic mice. Archives of biochemistry and

biophysics 367, 317-321.

Page 138: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Conclusion

137

Conclusion

Page 139: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Conclusion

138

Conclusion Modern Biotechnology has made enormous progress in recent years and become an

important field of investment for many big pharmaceutical companies. Established disciplines

of this new field includes the recombinant production of growth hormones, cytokines and

monoclonal antibodies. Research into these areas has produced novel and innovative

technologies such as gene- and cell-therapies some of which have even entered clinical trials

over the few last years. This work details the development of new tools that may help to

overcome certain problems accompanying the production, delivery, expression, and

controlled delivery of biologics.

New recombinant adeno-associated virus type 2 (rAAV2) based gene therapy vectors

were designed and allow for tight regulation of transgene expression by a clinically licensed

antibiotic after infection of target cells. Many different configurations of vector systems were

characterized such as a binary ON-and OFF-type, an auto-regulated, a self-regulated and a bi-

directional setup. Due to the safety and efficiency of rAAV2, these new vectors are valuable

tools for transgene delivery in vitro as well as in vivo and further down the road may be

promising tools for the delivery and regulation of therapeutic transgenes in clinical settings.

Reporter proteins are valuable tools to monitor many essential parameters in

biotechnological applications. Complex questions in metabolic engineering, process

optimization, gene- and cell-therapies and basic research can be answered by an array of

available reporter proteins specialized for such tasks. In this work a pair of novel heterologous

reporter proteins were designed, for both intracellular and secreted applications, for use in

mammalian cells. These new reporters are sensitive, compact, stable, easy to assay and

compatible with existing reporter proteins. Future applications may benefit from these new

reporter proteins where size and stability of delivered reporter-transgenes are critical or direct

assays need to be made without pretreatment of the samples.

The delivery of therapeutics has become key in the advent of new, complex therapies

such as protein drugs, gene-therapy and cell-therapies. Owing to the complexity, low stability

and immunogeneicity of these new drugs, alternatives to traditional routes of delivery have to

be developed. A versatile system for the delivery of biologics is presented based on the

rupture of bio-compatible polymer capsules upon the application of an external cue. This

system potentially allows for the release of varying cargos including protein therapeutics,

viral particles, liposomes or entire cells. The sensor cell line, which captures the applied

signal and induces capsule rupture, can be engineered to be responsive to different stimuli

Page 140: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Conclusion

139

such as various small molecules, hormones, gaseous molecules or even physical cues such as

temperature fluctuations. The combination of bio-compatible encapsulation protocols together

with the drug release “on-demand” may foster applications in future initiatives for controlled

drug delivery.

The optimization of the development and deployment of efficient new biologics is an

active field of academic and industrial research and we hope to have contributed with this

work in the development of more efficient future therapies.

Page 141: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Acknowledgements

140

Acknowledgements I would like to thank everyone who helped me over the last years to accomplish this work. Martin Fussenegger for the opportunity to conduct my PhD thesis in his lab. Sabine Werner for her work as a co-referee. All the members of the Fussenegger group over the years for the nice work atmosphere, the scientific support and for all the good moments we spent together. Marie Daoud-El Baba for her help with in vivo experiments. Jens Kelm for introducing me into lab work back when I started, many good discussions and for his support during my PhD thesis. David Greber and Domenick Grasso for their language support and many entertaining coffee breaks. My parents for their continuous support. Nadja Stohler for her support, patience and her love.

Page 142: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Curriculum Vitae

141

Curriculum Vitae Personal Data Name: David Fluri Date of Birth: April 12, 1977 Nationality: Swiss Private Address: Langstrasse 241, 8005 Zürich Education 2002-current Doctoral Studies at the ETH Zurich, Switzerland, Institutes of

Biotechnology/Chemical- and Bioengineering, supervised by Prof. Dr. Martin Fussenegger. Title: “Engineering delivery and release of protein therapeutics for mammalian systems”.

2002 Diploma Thesis at the Benaroya Research Institute, Seattle,

USA, supervised by Dr. Tongwen Wang. Title: ”The regulation of a transcriptional co-repressor SNIP1 via a multimeric protein complex in BMP signaling”.

1999-2002 Preparation of diploma degree in Biotechnology from the

European School of Biotechnology Strasbourg (ESBS), France 1997-1999 Preparation of “Vordiplom” in molecular biology at the

Biozentrum of the University of Basel, Switzerland 1993-1996 Matura Typ E at the Gymnasium Liestal, Switzerland Publications Fluri, D.A., Kemmer C, Baba, M.D., and Fussenegger, M (2008). A novel system for trigger-controlled drug release from polymer capsules. (submitted) Fluri, D.A., Baba, M.D., and Fussenegger, M. (2007). Adeno-associated viral vectors engineered for macrolide-adjustable transgene expression in mammalian cells and mice. BMC Biotechnology 7, 75.

Page 143: Rights / License: Research Collection In Copyright - Non ...31014/eth-31014-02.pdfEngineering delivery and release of protein therapeutics for mammalian systems A dissertation submitted

Curriculum Vitae

142

Fluri, D.A., Kelm, J.M., Lesage, G., Baba, M.D., and Fussenegger, M. (2007). InXy and SeXy, compact heterologous reporter proteins for mammalian cells. Biotechnology and Bioengineering 98, 655-667. Fluri, D.A. and Fussenegger, M (2007). Transduction technologies. In Methods in Biotechnology book series: Animal Cell Biotechnology, Methods and Protocols 2nd Edition, 3-21, Humana Press, New Jersey, USA Kelm, J.M., Djonov, V., Ittner, L.M., Fluri, D., Born, W., Hoerstrup, S.P., and Fussenegger, M. (2006). Design of custom-shaped vascularized tissues using microtissue spheroids as minimal building units. Tissue Engineering 12, 2151-2160. Presentations David A. Fluri and Martin Fussenegger (2004). Regulated Gene Expression Using Adeno-Associated Viral Vectors. Poster presentation at the 5th symposium of the department of biology ETH Zurich, Davos, Switzerland David A. Fluri, Guillaume Lesage and Martin Fussenegger (2006) Secreted Xylanase - A Novel Mammalian Reporter Gene. Poster presentation at the 4th European biotech workshop, Ittingen, Switzerland