apresentação do powerpoint fernandes phd... · vectores cav-2, nomeadamente ... bioprocesso com...

180

Upload: dotu

Post on 13-Nov-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Bioprocess design of canine adenovirus vectors

for gene therapy applications

Paulo David Machado dos Santos Fernandes

Dissertation presented to obtain a Ph.D. degree in Engineering and Technology Sciences,

Biotechnology

Instituto de Tecnologia Química e Biológica António Xavier

Universidade Nova de Lisboa

Supervisor: Paula Marques Alves

Co-supervisor: Ana Sofia Coroadinha

With the financial support from FCT, under contract SFRH / BD / 70810 / 2010

ii

Bioprocess design of canine adenovirus vectors for gene therapy applications

By Paulo Fernandes

First edition:

Cover: MDCK cells in microcarriers infected with CAV-2 vectors.

ITQB-UNL/iBET Animal Cell Technology Unit

Instituto de Tecnologia Química e Biológica/

Instituto de Biologia Experimental e Tecnológica

Av. da República EAN, 2780-157 Oeiras, Portugal

http://tca.itqb.unl.pt

http://www.itqb.unl.pt

http://www.ibet.pt

Copyright ©2015 by Paulo Fernandes

All rights reserved

Printed in Portugal

iii

Supervisors:

Dr. Paula Marques Alves, CEO of iBET, Instituto de Biologia Experimental e Tecnológica

(iBET), Oeiras, Portugal; Invited associate professor at Faculdade de Ciências e

Tecnologia, NOVA university of Lisbon; Principal investigator and Director of Animal Cell

Tecnology Unit at Instituto de Tecnologia Química e Biológica (ITQB/UNL), Oeiras,

Portugal

Dr. Ana Sofia Coroadinha, Auxiliary investigator and Head of Cell line development and

molecular biotechnology laboratory, Animal Cell Technology Unit, iBET and ITQB/UNL,

Oeiras, Portugal

Jury:

Dr. Otto-Wilhelm Merten, Head of the Applied Vectorology and Innovation group of

Généthon, Généthon, Évry, France

Dr. Rubén Hernández-Alcoceba, Investigator of CIMA – Center for Applied Medical

Research, University of Navarra, Pamplona, Spain

Dr. Michael Parkhouse, Head of the Infections & Immunity group, IGC, Oeiras, Portugal

Prof. Manuel J. T. Carrondo, Professor of Chemical and Biochemical Engineering at

Faculdade de Ciências e Tecnologia, NOVA university of Lisbon; Director of iBET and

Head of the Engineering Cellular Applications Laboratory from Animal Cell Technology

Unit, iBET and ITQB/UNL, Oeiras, Portugal.

iv

Foreword

This thesis is the result of four years of research at the Animal Cell Technology Unit of

ITQB-UNL/iBET, Oeiras, Portugal, under the supervision of Paula Marques Alves and Ana

Sofia Coroadinha.

This dissertation aimed to contribute to the advance of gene therapy by developing and

optimizing the production of canine adenovirus type 2 vectors, and by understanding the

biological features modulating adenovirus vector propagation.

v

Acknowledgements

I would like to express my gratitude to all the people that contributed to this thesis and to

the hosting institutions, iBET and ITQB, for the excellent working conditions.

To my supervisor, Dr Paula M. Alves, for the opportunity to let me grow as a scientist in

animal cell technology unit (ACTU) and develop this work, for the example of dedication,

for the confidence, guidance, support, pragmatism and for granting me all the means to

work.

To my co-supervisor, Dr Ana Sofia Coroadinha, for the guidance, confidence and support,

for making me feel a peer and for pushing my enthusiasm to science.

To Dr Eric J. Kremer, from IGMM, Montpellier, France, for providing the materials

necessary to start the area of CAV-2 vectors at ACTU, for the time, enthusiasm, inspiration

and discussions during this journey.

To the financial support provided by Fundação para a Ciência e Tecnologia

(SFRH/BD/70810/2010) and by European Commission, through the FP7 project

BrainCAV.

To the key persons in any bioprocess developed at ACTU – Marcos Sousa (upstream

process) and Dr. Cristina Peixoto (downstream process) – for the guidance and support.

To former and current colleagues from ACTU for the technical help and great times I have

spent with you in these years. In particular, to Vanessa Bandeira, Carina Brilha, Rute

Castro, Sofia Rebelo, Filipa Rodrigues, Marta Silva, Daniel Simão and Hugo Soares.

Aos meus amigos de longa data, Francisca, André, Sofia e Mafalda, por todos os momentos

e aventuras que ajudam a recuperar energias e resolver problemas mesmo sem falar

deles. Em especial à Francisca pela companhia nesta caminhada de PhD students, por me

obrigar a apanhar ar, pelas conversas e gargalhadas importantes em não perder o foco e

continuar em frente. Ao grupo das sessões pós-laboratório no jardim do Cego – Francisca,

André e Raquel.

À Maria, pelo exemplo de genuidade, bondade e boa disposição, pelo carinho e pelo apoio

especial que faz com que tudo valha a pena.

vi

À minha maravilhosa Família, pelo exemplo de força, integridade, e pelo espírito positivo

mesmo nas situações mais difíceis. Em particular à minhas primas Armanda e Paulinha

pelo apoio especial neste último ano. À minha avó Pataca, à minha mãe e ao meu pai, pelo

altruísmo e sacrifício em dar a educação e a vida que sempre sonharam aos filhos. À

minha irmã Mónica, pelo exemplo de determinação e por mostrar o quão importante é o

pilar Família para ultrapassar qualquer obstáculo que nos apareça no caminho.

vii

Abstract

The potential of human adenovirus vectors as vehicles for gene transfer with clinical

applications in vaccination, cancer treatment and in many monogenic and acquired

diseases has been demonstrated in several studies and clinical trials. However, the

clinical use of these vectors can be limited by pre-existing humoral and cellular anti-

capsid immunity. One way to circumvent this bottleneck while keeping the advantages of

using adenovirus vectors is using non-human viruses such as Canine Adenovirus type 2

(CAV-2). Moreover, CAV-2 vectors present attractive features to develop potential

treatment of neurodegenerative and ocular disorders. While the interest in CAV-2 vectors

increases, scalable and robust production processes are required to meet the need for

preclinical and possibly clinical uses.

This PhD thesis aimed to develop and optimize the production process of CAV-2 vectors,

namely E1-deleted (ΔE1) and helper-dependent vectors (HDVs), by i) addressing

technical aspects to enable large-scale production compliant with the guidelines for

manufacturing clinical material and ii) understanding cell physiological state and virus

replication behind high production titers and product quality.

Being MDCK cells already approved by the regulatory agencies (FDA and EMA), the use of

MDCK-based cell lines can facilitate the acceptance of CAV-2 clinical grade production.

Accordingly, MDCK-E1 and MDCK-E1-Cre cell lines were established in Chapter II, with

titers comparable to those described for human adenovirus producing cells. In addition, a

clear correlation between E1 (mostly E1A) expression levels and CAV-2 production was

observed during clone characterization, corroborating the importance of evaluating E1

levels when screening and developing adenovirus producer cells.

The need of larger amounts of vectors requires a CAV-2 manufacturing protocol

compliant with process scales. To address this, in Chapter III bioprocess development for

ΔE1 CAV-2 vectors production in stirred tank bioreactors and serum-free medium was

done. To do so, two approaches were evaluated: i) using microcarriers technology (part

A) and ii) using cells adapted to grow in suspension (part B). While productivities in

microcarriers-based bioprocess were similar to those from monolayer static cultures,

serum was still necessary for cell attachment. The production process was completely

serum-free with cells adapted to suspension, although a slight decrease in cell specific

productivity was observed. Both processes developed constitute a progress in CAV-2

viii

vectors availability at larger scales, leveraging the manufacturing process of CAV-2

vectors to the level of human adenovirus vectors.

The HDV production system, that comprises the use of Cre recombinase-expressing cells

and infection schemes with two vectors (HDV and helper vector (HV)), is more complex

than ΔE1 vectors system. Therefore, special attention was paid to the propagation of

HDVs. HDV production was optimized by evaluating MDCK-E1-Cre cell line stability and

infection conditions in Chapter IV. MDCK-E1-Cre cells specific productivity decreased

with the increase of cell culture passage. Best infection conditions (defined MOI ratio

between HDV and HV) were sufficient to minimize HV contamination and surpass the

need of Cre-expressing cells. The amplifications performed under a defined MOI of HDV

and HV can be thus moved to MDCK-E1 cells, which presents a robust performance even

under high passage number (Chapter II and Chapter IV). Despite these advances, HDV

production yields were still lower than ΔE1 vectors. This was further explored in Chapter

V by monitoring HDV replication steps to understand what factors were still limiting

production yields. Replication progression was intensified during HDV production, as

genome replication occurred faster during HDV production, leading to higher and earlier

expression of structural proteins. Such high viral protein content was consistent with

increased cell death observed during production and contributed to the low volumetric

titers. The low infectivity of the HDVs generated suggested a defective maturation

process and/or increased levels of defective particles. Altogether, this study identifies

genome replication, viral protein synthesis and maturation as features to be modulated in

the pursuit of an optimal HDV production.

In conclusion, a bioprocess for CAV-2 vectors meeting the technical aspects for large-scale

and/or clinical material production was developed. Important bioprocess parameters

were defined and a better understanding of adenovirus replication and producer cell

physiology was achieved. This work contributes to the progress of gene therapy and

biotechnology by debottlenecking the availability of a new vector to clinical applications

and by improving the state-of-the-art on adenovirus manufacturing.

ix

Resumo

Os vectores adenovirais humanos têm demonstrado elevado potencial como veículos

para a transferência de genes. As suas aplicações clínicas são vastas, passando pelo

desenvolvimento de vacinas, tratamento de cancro e doenças monogénicas. No entanto, o

uso clínico destes vectores pode ser condicionado pela imunidade humoral e celular pré-

existente em pacientes, resultante da exposição a este tipo de vírus ao longo da vida. Este

problema pode ser ultrapassado com o uso de adenovírus não-humanos, tais como os

adenovírus caninos tipo 2 (CAV-2), que mantêm todas as vantagens associadas ao uso de

vectores adenovirais. Mais ainda, os vectores CAV-2 possuem características promissoras

para serem usados no desenvolvimento de tratamentos contra doenças

neurodenegerativas e oculares. Como tal, o interesse pelos vectores CAV-2 tem

aumentado, o que torna imperativo o desenvolvimento de processos de produção

robustos e escaláveis que possam ir de encontro à quantidade e qualidade necessárias de

material biológico para aplicação em ensaios pré-clínicos e clínicos.

Esta tese de doutoramento teve como objectivo desenvolver e optimizar a produção de

vectores CAV-2, nomeadamente vectores com delecção na região E1 (ΔE1) e vectores

helper-dependent (HDV), de modo a i) respeitar os aspectos técnicos que facilitam a

produção em larga-escala e/ou de material com grau clínico e ii) compreender a fisiologia

das células produtoras e a replicação viral para desenvolver um bioprocesso com

produtividade máxima.

As células MDCK, já usadas para produção de outros vírus e aprovadas pelas autoridades

reguladoras de produtos para uso humano (FDA e EMA), foram usadas no Capítulo II

para estabelecer linhas celulares produtoras de vectores CAV-2 (MDCK-E1 e MDCK-E1-

Cre). Os títulos de produção de CAV-2 com estas novas linhas celulares foram

semelhantes aos descritos para células produtoras de adenovírus humanos. Durante este

trabalho foi também observada uma correlação entre os níveis de E1 (principalmente

E1A) e produção de CAV-2, o que revela a importância da avaliação dos níveis de E1 no

rastreio e selecção de linhas celulares produtoras de vectores adenovirais.

A crescente procura de vectores CAV-2 torna necessário o desenvolvimento e

implementação de um protocolo de manufactura compatível com as escalas usadas em

bioprocesso. Nesse sentido, a produção foi transferida para biorreactores de tanque

agitado e para meio de cultura sem soro (Capítulo III) usando duas abordagens: i)

tecnologia de microcarriers, para permitir cultura de células aderentes em sistema

x

agitado (parte A) e ii) adaptação de células aderentes a crescimento em suspensão

(parte B). A produtividade de vectores CAV-2 no bioprocesso baseado em microcarriers

foi semelhante à obtida em culturas estáticas, no entanto foi necessário suplementar o

meio de cultura com soro para permitir a adesão das células aos microcarriers. O

bioprocesso com células adaptadas a suspensão ocorreu em condições livres de soro,

embora tenha resultado numa ligeira diminuição dos títulos volumétricos. Ambos os

bioprocessos permitem a produção de vectores CAV-2 a escalas maiores, tornando o

processo de manufactura equivalente ao usado para adenovírus humanos.

O sistema de produção de HDV, que inclui células produtoras a expressar Cre

recombinase e esquemas de co-infecção com dois vectores (HDV e vectores helper (HV)),

é mais complexo que o sistema de produção de vectores ΔE1. Por isso, a propagação de

HDV foi estudada em maior detalhe. Para definir os pontos-chave na produção de HDV, a

estabilidade da linha celular MDCK-E1-Cre e as condições de infecção foram avaliadas no

Capítulo IV. A produtividade específica das células MDCK-E1-Cre diminuiu com o

aumento do número de passagens em cultura. Por outro lado, a optimização da MOI de

HDV e de HV nas co-infecções foi suficiente para minimizar a contaminação de HV e, por

conseguinte, ultrapassar a necessidade de usar células que expressem Cre. Esta

observação indica que, usando uma MOI definida de HDV e de HV, a amplificação de HDV

pode ser transferida para células MDCK-E1, que apresentam maior robustez em termos

de performance, mesmo com passagens elevadas em cultura (Capítulo II e Capítulo IV).

Apesar destes avanços, a produção de HDV continua a ser inferior à de vectores ΔE1. Este

aspecto foi estudado no Capítulo V ao monitorizar a replicação de HDV para

compreender que factores estão a limitar a sua propagação. Curiosamente, o progresso da

replicação foi intensificado durante a produção de HDV. Mais especificamente, a

replicação de genomas foi mais rápida que aquela observada com vectores ΔE1, levando a

uma expressão precoce e mais elevada das proteínas virais. O conteúdo elevado de

proteínas virais foi consistente com um aumento da morte celular após infecção e

contribuiu para a diminuição dos títulos virais. Adicionalmente, a baixa infecciosidade

dos HDV produzidos sugere um processo defectivo de maturação. Em suma, este estudo

identificou a replicação do genoma viral, a síntese de proteínas virais e a produção de

partículas defectivas/maturação como aspectos a serem modulados para estabelecer uma

produção melhorada de HDV.

xi

Em conclusão, neste trabalho foi desenvolvido um bioprocesso para produção de vectores

CAV-2 compatível com os aspectos técnicos de produção em larga-escala e/ou de material

para uso clínico. Ao mesmo tempo, parâmetros relevantes para o sucesso do bioprocesso

foram definidos e combinados com um melhor conhecimento da replicação adenoviral e

da fisiologia da célula produtora. Como resultado, i) o estado da arte sobre a manufactura

de adenovírus foi melhorado e ii) a produção de vectores CAV-2 é agora compatível com

as escalas usadas em bioprocesso e com o desenvolvimento de material para uso humano,

o que constitui um avanço para o progresso da terapia génica e da biotecnologia.

xii

Thesis publications

Published:

1. Fernandes P, Almeida AI, Kremer EJ, Alves PM, Coroadinha AS. 2015. Canine helper-

dependent vectors production: implications of Cre activity and co-infection on adenovirus

propagation. Sci Rep. accepted (in press)

2. Fernandes P, Simão D, Guerreiro M, Kremer EJ, Coroadinha AS, Alves PM. 2015. Impact

of Adenovirus life cycle on the generation of canine helper-dependent vectors. Gene Ther.

Jan;22(11):40-9. http://dx.doi.org/10.1038/gt.2014.92

3. Fernandes P, Peixoto C, Santiago VM, Kremer EJ, Coroadinha AS, Alves PM, 2013.

Bioprocess development for canine adenovirus type 2 vectors. Gene Ther. Apr;20(4):353-

60. http://dx.doi.org/10.1038/gt.2012.52

4. Fernandes P, Santiago VM, Rodrigues AF, Tomás H, Kremer EJ, Alves PM, Coroadinha

AS. 2013. Impact of E1 and Cre on Adenovirus Vector Amplification: Developing MDCK CAV-

2-E1 and E1-Cre Transcomplementing Cell Lines. PLoS One. 2013;8(4):e60342.

http://dx.doi.org/10.1371/journal.pone.0060342

Submitted:

5. Fernandes P, Silva AC, Coroadinha AS, Alves PM. Propagation of Adenovirus vectors. In

D.T. Curriel (ed.) Adenoviral Vectors for Gene Therapy, 2nd edition, Academic Press

(submitted)

In preparation:

6. Fernandes P, Sousa MFQ, Kremer EJ, Coroadinha AS, Alves PM. Suspension MDCK-E1 cell

line for the manufacturing of CAV-2 vectors (in preparation)

Other publications in the scope of virus production:

7. Silva AC, Roldão A, Teixeira A, Fernandes P, Sousa MFQ, Alves PM 2015. Cell

immobilization for the production of viral vaccines. In: Al-Rubeai, Mohamed (Ed.), Animal

Cell Culture, Cell Engineering, Vol. 9, Springer. http://dx.doi.org/10.1007/978-3-319-

10320-4_17

8. Silva AC, Fernandes P, Sousa MFQ, and Alves PM. 2013. Scalable Production of

Adenovirus Vectors, p. 175-196. In M. Chillón and A. Bosch (ed.), Adenovirus, vol. 1089.

Humana Press. http://dx.doi.org/10.1007/978-1-62703-679-5_13

xiii

List of contents

Chapter I – Introduction ……………………………………………………………………………………………. 1

Chapter II – Developing MDCK CAV-2-E1 and E1-Cre transcomplementing cell lines …. 31

Chapter III – Upstream bioprocess development for E1-deleted CAV-2 vectors …………. 55

Part A – Production with adherent cells using microcarrier technology………... 57

Part B – Production with cells adapted to suspension …………………………………... 73

Chapter IV – Implications of Cre activity and co-infection on canine helper-dependent

vector production …………………………………………………………………………………………………….. 91

Chapter V –Impact of adenovirus life cycle on the generation of canine helper-dependent

vectors …………………………………………………………………………………………………………………... 113

Chapter VI – Discussion and perspectives ………………………………………………………………. 141

Appendix ………………………………………………………………………………………………………...……. 153

xiv

Chapter I

Introduction

This chapter is adapted from the book chapter:

Fernandes P, Silva AC, Coroadinha AS, Alves PM. Upstream bioprocess for adenovirus

vectors. David T Curiel (ed), Adenoviral vectors for gene therapy, Academic Press

(under review)

Chapter I

2

Contents

1. Gene therapy ................................................................................................................................................. 3

2. Adenovirus vectors for gene therapy ................................................................................................. 3

2.1. Structure and genome ............................................................................................................................ 4

2.2. Infection and replication cycle ........................................................................................................... 4

3. Manufacturing of adenovirus vectors ................................................................................................ 5

3.1. Adenovirus vectors and producer cells .......................................................................................... 5

3.1.1. 1st generation vectors ................................................................................................................... 6

3.1.2. Conditionally-replicating vectors ............................................................................................ 7

3.1.3 2nd generation vectors ................................................................................................................... 7

3.1.4. 3rd generation vectors ................................................................................................................... 8

3.1.5. Novel adenovirus vectors ............................................................................................................ 9

3.2. Upstream process for adenovirus vectors ................................................................................. 10

3.2.1. Cell culture and adenovirus production process ........................................................... 11

3.2.2. Adenovirus seed stocks ............................................................................................................. 12

3.2.3. The infection process and harvest strategy ..................................................................... 13

3.2.4. Productivity of Adenovirus vector manufacturing ....................................................... 14

3.2.5. Adenovirus production at high cell density ..................................................................... 14

3.2.6. Physical parameters and process monitoring................................................................. 15

3.3. Production process of helper-dependent vectors .................................................................. 16

3.4. Downstream process for adenovirus vectors .......................................................................... 17

4. Concerns in the manufacturing of adenovirus vectors for product release .................. 18

4.1. Extraneous agents ................................................................................................................................ 18

4.2. Replication competent adenovirus ............................................................................................... 19

4.3. Vector genetic stability ....................................................................................................................... 19

4.4. Quantity and potency .......................................................................................................................... 20

5. Scope of the thesis ................................................................................................................................... 20

6. Author contribution ................................................................................................................................ 21

7. References ................................................................................................................................................... 22

Introduction

3

1. Gene therapy

Gene therapy is defined as the transfer of genetic material into an individual’s cells or

tissues, treating or preventing a disease. After three decades of research, gene therapy is

currently considered solid (1), showing success in the clinic with an emphasis on rare,

inherited disorders including hemophilia (2), Leber Congenital Amaurosis (3-5) and X-

linked severe combined immunodeficiency (6). Moreover, a critical milestone was

achieved with the market authorization from European Medicines Agency of Glybera

(alipogene tiparvovec), an adeno-associated virus based product to treat deficiency in

lipoprotein lipase in patients with a severe form of hyperglyceridemia (7). Included in

gene therapy is also the sub-field of oncolytic virotherapy. In fact, most of the clinical

trials in gene therapy have been aimed at the treatment of cancer. Oncolytic virotherapy

utilises viruses capable of specifically targeting and replicating in tumor cells and causing

cell lysis, thereby killing the infected tumor cells (8). In some cases, these viruses are

modified to deliver suicide genes or immunostimulatory cytokines to enhance the

immune response against cancer.

Although nonviral approaches are becoming increasingly common to perform gene

delivery (9), viral vectors remain by far the most popular approach, having been used in

more than 70% of the trials performed to date (http://www.abedia.com/wiley/). Among

these, the top most used are based on: i) adenovirus, ii) retrovirus, iii) vaccinia virus, iv)

adeno-associated virus, v) poxvirus, vi) lentivirus and vii) herpes simplex virus. Worth

mentioning the emerging interest on vectors from adeno-associated viruses and

lentivirus, as depicted by the research dedicated to these vectors and growth rate of use

(9).

2. Adenovirus vectors for gene therapy

Viral vectors are currently the most efficient tools for in vivo gene transfer. Due to their in

vivo efficiency, adenovirus vectors (AdV) are used more often than any other vector in

clinical trials, in which the human adenovirus vector (HAdV) serotypes 5 and 2 are the

most characterized ones among all other serotypes of the same family (10, 11).

Adenovirus wide cell tropism in quiescent and non-quiescent cells, its inability to

integrate the host genome and its high production titre (12) make AdV very good

candidates for human gene therapy.

Chapter I

4

2.1. Structure and genome

Adenoviruses are non-enveloped, icosahedral viruses of 60 to 90 nm with a linear double

stranded DNA genome of ~36 kbp. The genome can be divided in two sets of

transcriptional units: early genes that are expressed before the onset of viral DNA

replication, and late genes which are preferentially expressed after viral DNA replication

(Figure 1). This classification also defines the early and late phase of the infectious cycle.

E1A is the first gene to be expressed following infection and is involved in the

transcription of the other viral early genes. The mRNAs of major late transcription unit

are grouped into five families (L1-L5) which are dependent on the activation of major late

promoter during viral DNA replication.

Figure 1. Schematic representation of wild-type adenovirus genome and the different generations of non-replicating vectors. E1 to E4: early region transcript units; L1 to L5: late region transcprits unit; ITR: inverted terminal repeats; MLP: major late promoter; Ψ: packaging signal; GOI: gene-of-interest.

2.2. Infection and replication cycle

The sequential uptake process relies on an initial contact with cellular receptors

responsible for attachment (13) and internalization of the virus particle (14).

Internalization occurs by receptor-mediated endocytosis (15). Once in the cytosol, the

virion is transported via microtubuli towards the nucleus. Meanwhile, the particle is

dismantled by an ordered elimination of structural proteins so that when it reaches the

nuclear membrane, only the core particle is left. Adenovirus uncoating culminates with

the release of the viral DNA into the nucleus via nuclear pore complexes (16). The early

genes are responsible for expressing mainly non-structural, regulatory proteins (17).

ITR

E1A/E1BMLP

E3L1 to L5

E2B E2A E4ITR

Ψ

ITR

∆E1 ∆E3Ψ

GOI ITR

ITR

∆E1 ∆E3Ψ

GOI ITR

∆E2B ∆E2A ∆E4

ITR

Ψ

GOI and stuffer DNA ITR

Wild-type

1st generation

2nd generation

3rd generation

Introduction

5

These proteins alter the expression of host proteins that are necessary for DNA synthesis,

activate other early genes (such as the virus-encoded DNA polymerase) and avoid

premature death of the infected cell by the host-immune defenses. During viral genome

replication, late phase transcription is activated (17). This infection phase is mainly

focused on producing capsid proteins and packaging the replicated viral genomes;

structural proteins are assembled into virions, viral DNA is packaged and viruses are

released from the cell as a result of virus induced cell lysis (Figure 2).

Figure 2. Adenovirus replication cycle. Briefly, after cell binding (1), the mature viral particle is internalized and transported towards the nucleus (2). During this transport, the particle is dismantled by an ordered elimination of some structural proteins and culminates with the delivery of the viral genome into the nucleus via nuclear pore complexes. Once in the nucleus, early genes are expressed (3, 4) and viral DNA replication starts (5). Late phase of infection is then activated and structural proteins expressed (6, 7). These proteins are likely assembled into empty virions (8), followed by the packaging of viral genome (9). Finally, viral particles are subjected to a maturation process becoming infectious (10) and cell lysis is accomplished (11).

3. Manufacturing of adenovirus vectors

3.1. Adenovirus vectors and producer cells

The development of adenovirus vectors is based on modifications of wild type genome

mainly by removing viral gene(s) and adding expression cassettes with the gene(s) of

Chapter I

6

interest (GOI). While these viral genes are essential for virus replication, to propagate

adenovirus vectors it is necessary the establishment of producer cell lines, i.e. cells

expressing the viral elements deleted from the vector genome. The adenovirus vectors

that are non-replicating in a clinical setting and strictly used for gene transfer can be

categorized as first, second or third generation vectors, depending on the extension of

viral genes removed (Figure 1). The conditionally-replicating vectors are another

category of adenovirus vectors and are designed to specifically target, propagate and

deliver gene(s) in cancer cells.

3.1.1. 1st generation vectors

The majority of adenovirus vectors used for gene therapy or many other therapeutic

purposes are non-replicating, in which the E1 region is deleted from the genome often in

combination with E3, providing space for the insertion of expression cassettes. Such

adenovirus vectors are denominated as first generation or E1-deleted (∆E1) vectors. E1

region can be divided in E1A and E1B subunits and codes for products involved in the

activation of other early and late genes expression and in making host cells more

amenable to initiate virus propagation (i.e. inhibition of antiviral response and apoptosis)

(18-24). Therefore, to produce ∆E1 vectors a producer cell line containing adenovirus E1

sequences is necessary to complement these functions. On the other hand E3 region,

which is involved in antagonizing host defense mechanisms, is not essential for viral

amplification in vitro (25). Together, the deletion of E1 and E3 permits a transgene insert

capacity up to 8.2 kb. HEK 293 represents the traditional cell line used to trans-

complement the lack of E1 and produce adenovirus vectors. Because of the significant

homology of first-generation adenovirus vectors with the HEK 293 DNA, the main

disadvantage in using HEK293 is its potential to generate replication-competent

adenoviruses (RCA), raising safety concerns in a therapeutic product (26-30). To avoid

this, several cell lines were established under the rational of reducing the homology of

viral DNA sequences incorporated in cell line with viral vectors genome (reviewed in

(31)). Following this effort, PER.C6 represents the first cell line in which the generation of

RCA is abolished and typical adenovirus yields are ensured (32).

HEK293 cell line was generated with sheared HAdV-5 DNA, however the current strategy

to generate producer cell lines for E1 complementation is through the incorporation of

the contiguous sequences of E1A and E1B sequences into the cell line genomes. Strategies

Introduction

7

to minimize any recombination between viral genome and cell DNA were further

developed and involve the integration of E1A and E1B into producer cell genome at

separate locations (33, 34). E1 complementing cell lines are the backbones for the

remaining producer cells used for other adenovirus vectors.

3.1.2. Conditionally-replicating vectors

Conditionally-replicating adenovirus vectors, being also denominated oncolytic

adenoviruses, have been employed for cancer treatment to specifically target and

replicate in cancer cells (8, 35, 36). The lytic nature of adenovirus replication directly kills

tumor-infected cells, releasing the associated antigens. On the other hand, progeny

viruses can be spread throughout a tumor, further infecting and destroying other cancer

cells.

The construction of oncolytic adenoviruses is based on the deletion or modification of

viral gene functions that are critical to viral replication in normal cells, but dispensable in

tumor cells. This includes insertion of mutations in E1A region (37, 38), or deletion of

E1B (39, 40) from wild-type genome, to target cancer cells with defects in the

retinoblastoma (Rb) and p53 pathways, respectively, as these pathways are defective in

most human tumors. Another strategy in targeting oncolytic adenoviruses replication in

cancer cells involves the control of E1 transcription by using tumor or tissue specific

promoters such as prostate-specific enhancer/promoter for prostate cancer (41), or E2F-

I for cancer cells with a defective Rb pathway (42). In addition to these modifications,

these vectors can further incorporate genes encoding immune stimulatory factors to

boost the anti-tumor immunity (reviewed in (8)), giving rise to the so-called armed-

vectors and to one of the most promising gene delivery systems for cancer therapy.

Production of oncolytic adenoviruses is similar to first generation vectors, in which E1

functions are provided by the producer cell line. Therefore, cell lines already established

for first generation vectors are used to the manufacture of these vectors and include HeLa

(43), A549 (44), HEK293 and PER.C6 (45).

3.1.3 2nd generation vectors

Although the removal of E1 region renders the virus replication-defective, the delivery of

high doses of first generation vectors and/or the presence of E1-like factors in many cells

can lead to the expression of other viral proteins in vivo (31, 46-48). This can induce a

Chapter I

8

strong immune response, reducing the efficacy of these vectors. To circumvent that,

further deletions in E2 and/or E4 regions were explored, leading to the establishment of

2nd generation vectors. The E2 region (E2A and E2B subunits) codes for three proteins

essential for viral replication: DNA-binding protein (DBP) transcribed from E2A subunit,

terminal protein and viral DNA polymerase from E2B subunit (49). E4 products modulate

transcription, the cell-cycle, cell signaling and DNA repair and are essential for productive

virus infection (50), but only one of the ORF3 or ORF6 is required for successful virus

production in cell culture (51, 52).

Given the toxicity associated with E2 and E4 viral products, most of the cell lines for 2nd

generation vectors rely on the use of an inducible system (reviewed in (31)). Similarly to

1st generation vectors, the majority of E1/E2 and E1/E4 complementary producer cells

lines use HEK 293 as parental cell line.

Despite the sophisticated systems available for 2nd generation vectors manufacturing,

including vector construct and incorporation of inducible systems in the producer cell

lines, the use of these vectors remains without advances. In general, reduced production

yields are obtained for vectors with multiple deletions. In fact, when compared to first

generation vectors, the yields of these vectors can be reduced, and no major

improvement in toxicity is observed in vivo (31, 53-58). Moreover, the transgene

expression of these vectors is also described as unstable, which probably made 3rd

generation vectors a more suitable choice to that end (reviewed in (59)).

3.1.4. 3rd generation vectors

3rd generation vectors, also known as gutted or gutless, high capacity or helper-

dependent vectors (HDVs), are the most advanced adenovirus vectors. They are devoid of

all viral coding genes, only harbouring on their genome the essential cis-acting elements:

inverted terminal repeats (ITRs) and packaging signal. This allows the insertion of

therapeutic gene or genes up to ~37 kb. Additional stuffer DNA is included to render

vector genome size similar to wild-type and maintain viral particle stability (60). Apart of

their increased safety, the use of these vectors result in long-term transgene expression

(reviewed in (59)). To produce these vectors, while E1 functions are provided by the

producer cell line, the remaining functions are provided by an helper vector (HV). In fact,

the need of a HV increases the complexity of production system and it is the main

disadvantage of these type of vectors. Adding HV to the production system implicates the

Introduction

9

production of both HDV and HV, raising the need to reomove HV contamination from the

final product. To date, the most elegant way to prevent HV propagation is the approach

described by Parks et al. in which the HV packaging signal is flanked by loxP sites, and

under the expression of Cre-recombinase the HV genome is cleaved (hampering its

encapsidation) and HV particles production is minimized (61). Thus, besides E1,

producer cell lines for HDV have to further express Cre recombinase. Following same

approach, FLPe recombinase system is also used to minimize HV contamination (62, 63).

Similarly to E1 transcomplementing cell lines, the most common producer cell lines using

recombinase system are derived from HEK 293 cells, although PER.C6 cells are also used

to that purpose (reviewed in (31)). Despite these efforts in HDV production system, two

major bottlenecks are still found when considering the use of HDV in patients: i) HV

contamination is still not fully eliminated and ii) HDV production yield still faces the need

of multiple amplification steps and inconsistency in infectious particles yields (discussed

in section 3.3). Despite these bottlenecks, the increased cassette incorporation, improved

expression and lower immunity is still pushing their further development (59).

3.1.5. Novel adenovirus vectors

While most research on vector development is based on the utilization of human

adenovirus serotypes 5 and 2, over 80% of the adult population has been naturally

exposed to these viruses (64). Therefore, pre-existing humoral and cellular immunity

may preclude efficient gene transfer when these adenovirus vectors are used (65-70).

Apart of limitations in therapeutic efficacy (71), immune responses against the vector

may result in a number of undesirable side effects, including liver toxicity (72) and

systemic inflammatory response syndrome due to repeated vector administration (73).

Eliminating liver tropism and the epitopes involved in viral proteins recognition by

neutralizing antibodies has been proposed to reduce the immune response. Alternatively,

vectors based on low seroprevalence AdVs can be also be used to circumvent the pre-

existing immunity. In addition, the diversity of AdV protein isoforms and their variety of

ligand-receptor interactions found in the different serotypes can also be the basis to

target different cell types (70). Adenovirus vectors derived from alternative human and

non-human serotypes, to which the human population has a lower or no prevalence of

neutralizing antibodies, are being currently investigated.

Chapter I

10

Human subgroup B adenoviruses, and in particular serotypes 11 and 35, are being used

to develop adenovirus vectors (reviewed in (31)). Typically, E1 (both E1A and E1B)-

deleted vectors from these serotypes cannot replicate in regular E1 producer cell lines

already established for serotype 5 vectors, although E1A-deleted vectors can be

propagated in PER.C6 cells (74). These vectors are therefore produced by modifying the

typical 1st generation cell lines already available (75-78) or replacing vectors genes by

HAdV5 sequences to permit viral propagation on unmodified cell lines, such as PER.C6

(79).

Several non-human adenoviruses derived from bovine, simian, porcine, ovine, murine

and canine sources have been used as backbone to develop E1-deleted vectors for gene

therapy or vaccine purposes (reviewed in (70)). For the majority of non-human vectors,

specialized E1 producer cells have been developed to propagate these vectors, such as

bovine kidney or fetal retinal cells expressing Bovine or human adenovirus E1 sequences

(80), or canine kidney cells expressing the canine adenovirus type 2 (CAV-2) E1 (81, 82).

Chimpanzee derived vectors of subgroup E can be produced on HEK293 cells already

developed (83, 84). However, similarly to HAdV11 and HAdV35, chimpanzee derived

vectors of subgroup B cannot be propagated on these cells, and a chimeric strategy is

used to allow propagation in HEK293 cells (85, 86). CAV-2 derived vectors are probably

the best described and advanced non-human vectors (87). The ability to preferentially

transduce neurons combined with a remarkable capacity of axonal transport, make CAV-

2 vectors candidates for the treatment of neurodegenerative diseases (88). Furthermore,

3rd generation CAV-2 vectors were already developed and tested in animal models,

confirming their potential for gene transfer based therapies (89, 90).

3.2. Upstream process for adenovirus vectors

An adenovirus production process starts by growing the cell line of choice to the desired

cell density for infection followed by the inoculation of an adenovirus stock to initiate the

infection and virus production cycle (Figure 3) (91). The need for significant amounts of

clinical grade adenovirus vectors, which in some cases may reach 1013 total

particles/patient (1011 infectious particles/patient), requires efficient and robust

processes for production and purification at large-scale compliant with good

manufacturing practices (GMP)(12). From a scalable point-of-view, the production of

adenovirus vectors encompasses several bioengineering aspects that must be carefully

Introduction

11

undertaken to maximize bioprocess yields and reduce production costs. In this section,

process considerations for the adenovirus production will be discussed mostly focusing

on first generation vectors, as the main bioprocess advances have been achieved with

these vectors.

Figure 3. Typical profile of an adenovirus production process, including cell growth, viability and infection kinetics. After inoculation, cells grow exponentially and are usually infected at the end of exponential growth phase (represented by an arrow). Viral particles are assembled intracellularly. Due to the lytic nature of adenovirus propagation, cell lysis is induced after the production of viral particles. As a result, cell concentration and viability decreases and viral particles released to the supernatant. ●Cell concentration at infection (CCI); ■ time of infection (TOI). Adapted from (91).

3.2.1. Cell culture and adenovirus production process

Adenovirus production process can be performed in static or stirred cultures. From a

scalable point-of-view, stirred cultures are preferred. The use of microcarriers represents

the simplest approach to transfer adherent cells to stirred culture systems. The initial

steps of microcarriers preparation and cell seed from adherent cultures represent the

main drawback when using this system (92), since cell manipulation is cumbersome

when working at larger scales. Most of the production processes for adenoviruses

reported in literature use cells growing in suspension (reviewed in (93)). Although

adaptation of cells to grow in suspension can be time-consuming at the expense of

lowering cell specific productivity, suspension cell lines are preferred as they greatly

facilitate large-scale productions. Furthermore, the adaptation of cells to suspension

involves transference to serum-free culture medium, which represents an additional

advantage for biopharmaceuticals production.

In general, the medium is first chosen and optimized for its ability to support cell growth.

During the infection and vector production phase the specific consumption rate of several

Viability

Viable cell concentration

Infe

cti

on

Ce

ll c

on

ce

ntr

atio

n (

)

Ce

ll v

iab

ility (

)

Extr

ace

lula

r V

ira

l pa

rtic

les (

)

Intr

ace

lula

r vir

al p

art

icle

s (

)

Time (h)

Chapter I

12

medium components is increased, and need to be taken into account when defining the

final medium formulation. Also, cells cultured with media containing animal serum

should be avoided. The undefined composition and high batch-to-batch variability of

serum, together with its potential source of contaminations, raises safety concerns and

hinders the standardization of cell culture processes for the production of

biopharmaceuticals (94). Almost all media manufacturers commercialize serum-free

formulations designed and optimized for specific cell lines and/or final product.

The majority of scalable adenovirus manufacturing processes are performed in stirred-

tank bioreactors both for microcarriers and suspension cultures (93). Technological

advances in disposable equipments, such as single-use Wave and stirred tank bioreactors,

have been shown by several companies (e.g. GE Healthcare, Sartorius, PBS). The use of

such systems poses several advantages for GMP production by avoiding the need of

cleaning and sterilization validation and alleviating facility requirements. Therefore, the

increasing use of disposable equipments for adenovirus manufacturing is anticipated.

3.2.2. Adenovirus seed stocks

AdV viral particles must be rescued from the initial vector genome construct. This is

usually performed by linearization of vector genome from the plasmid, and transfection

of producer cells. Viruses are then harvested once cytopathic effect is evident or, as

alternative, 2-3 days after transfection. This procedure is typically performed in static

culture conditions. When working with producer cells easily transfectable and first

generation vectors, the rescue of viral particles after this transfection step is relatively

high, and one or two more production rounds are performed to amplify the amount of

viral vectors and establish a purified viral seed stock. In such cases, scalable production

processes are usually established once a purified viral seed stock is obtained. When

developing viral vectors that typically present low productivities, such as HDV, process

development is defined from the transfection step to the final viral seed stock production

to maximize working-scale and volumetric productivity in all steps and minimize the

number of amplifications required to produce sufficient viral material (91) (discussed in

section 3.3.).

For GMP production, the purified viral seed stock must be certified and tested to confirm

the absence of adventitious agents (see section 4.1). On the other hand, the use of well

established purified viral seed stocks is advised even for research purposes as it ensures

Introduction

13

reproducibility between different production batches. Therefore, determination of viral

particles, physical to infectious particles ratio and selection of best storage conditions are

carefully undertaken to ensure that particles properties from viral seed stock are

maintained (95-99).

3.2.3. The infection process and harvest strategy

Production process starts once producer cells are growing, being infected afterwards and

the newly produced adenoviruses harvested and the end of process (Figures 2 and 3).

Defining best process-related parameters, such as multiplicity of infection (MOI), cell

concentration at infection (CCI), time of infection (TOI) and time of harvest (TOH) is

determinant to ensure an optimal production process. Because adenovirus infections are

relatively fast and lytic, processes are established as single-round infections with MOI > 1

to ensure that all cells are infected. Although MOI > 1 needs to be established, infecting

the cells with more virus than those optimally required may have a negative effect on

cells viability, compromising virus productivity. TOI or CCI is usually selected when cells

are at exponential growth phase. For adherent, static cultures, cells are infected when 60-

80% confluence is achieved, while for stirred cultures the typical cell densities range used

for adenovirus production is 0.5-1 x 106 cells/mL in batch mode with medium exchange

at infection. To produce adenovirus above this cell density and maintain cell specific-

productivity, fed-batch or perfusion modes must be added to the bioprocess (discussed in

section 3.2.5.).

After infection, the cell growth is arrested within 24 hpi. Viral DNA replication and virus

assembly occurs between 10-24 hpi and 20-48 hpi, respectively. As virus production

progresses, cell viability decreases after 24 hpi. Typically, at 48 hpi, when the volumetric

virus productivity reaches its plateau, the cell viability is around 40-80%. At this point, a

percentage of the virus (between 10-50%) has already been released from lysed cells, but

the rest of the virus remains intracellular. The cultivation process can proceed further

with no significant increase in virus production, but with a significant increase in virus

found in the culture medium. Harvest timing and method can be tailored to the entire

bioreaction bulk, or only to the intra- or extracellular fractions. The remaining cells are

lysed either by freeze-thaw cycles (100, 101) or using a detergent (102, 103).

Chapter I

14

3.2.4. Productivity of Adenovirus vector manufacturing

One of the most attractive features for adenovirus vectors, namely ∆E1-deleted vectors,

is the high production yields obtained when compared to other viral vectors. Typically,

titers of ∆E1-deleted vectors range from 103 to 104 IP per cell, whereas genome-

containing particles are one log higher (reviewed in (93)). Considering the usual CCI used

for adenovirus production, this corresponds to a volumetric productivity of 109 – 1010

IP/mL or 1010 – 1011 PP/mL. Worth mentioning that such yields are dependent on

adenovirus vector construct and culture conditions. Some production processes for HDV

are described as holding relatively low specific (102 IP/cell) and volumetric (108 IP/mL)

titers (104, 105) or inconsistent PP:IP ratios (106) (see section 3.3). In addition, cell

specific productivity under batch mode is limited to CCI below 1x106 cells/mL; producing

adenoviruses above this cell concentration results in lower specific yields (see section

3.2.5). Finally, cell physiological state can also impact the infectivity of the newly

produced adenoviruses. Indeed, when producing adenoviruses with MOI higher than the

optimal, the resulting PP:IP ratio tends to increase.

3.2.5. Adenovirus production at high cell density

In a batch operation mode, the cell density at infection is a very important parameter as it

impacts virus production. In fact, the narrow range for an optimal infection cell density

between 0.5-1.0x106 cells/mL is well documented (reviewed elsewhere (91)): a drop in

specific productivity at higher cell densities occurs even though medium formulations

allow cell growth up to 5 x 106 cells/mL. This is often referred to as the “cell density

effect”. While this phenomenon is not totally understood, the decrease in productivity is

thought to be due to limitations in a nutrient critical for virus propagation, accumulation

of inhibitory byproducts, or a combination of both (107). Following this hypothesis, the

typical approach to maintain cell-specific productivity at increased cell densities consists

of exchanging medium at infection time. This operation mode, widely used for viral vector

production, enables maximum cell-specific productivity up to 2x106 cells/mL. The main

limitation of such approach is the incorporation of a medium exchange/cell separation

step. While at scales of few liters, this is relatively easy to perform by centrifugation,

when dealing with hundreds or thousands of liters such manipulation is highly

cumbersome. At these scales, fed-batch or perfusion are more feasible approaches.

Introduction

15

Several fed-batch approaches were devoted to adenovirus production based on the

control or addition of glutamine, glucose and aminoacids (108-110). Despite the

simplicity to implement, most of the fed-batch strategies towards maintaining cell-

specific productivity have failed.

Perfusion processes are capable of achieving high cell densities, through a continuous

renewal of fresh medium, providing new nutrients and diluting/removing inhibiting-

byproducts. Therefore, perfursion must be such that nutrient supply and byproducts

removal rates are sufficient to ensure robust cell performance at increasing cell

concentration. The perfusion rates described for adenovirus production can range from

0.5 to 3 culture volumes per day (V/day), being 2 V/day the rate mostly used (43, 111-

113). When compared to low cell density batch productions, specific productivity of 293

cells infected at 3 – 6x106 cells/mL was maintained, which constituted a 5-fold increase in

the final production yield (111, 113). The highest cell density tested to successfully

produce adenoviruses under perfusion mode is described in (43) using Hela cells at 107

cells/mL.

Other efforts were made to better understand the cell density effect. The specific

metabolic demands of producer cells during growth and virus production have been

analyzed through a metabolic flux analysis approach, showing that a favorable metabolic

state for adenovirus production should have an increase in glycolytic and TCA fluxes

(113, 114), and in ATP production rates upon infection. This state is also extended to

perfusion modes (113). Furthermore, it was demonstrated that a decrease in the

proportion of cells in S phase was related to a decrease in specific productivity at high cell

densities (115). Following this observation, the synchronization of HEK293 cells

chemically or by lowering temperature led to an enrichment of cells in S phase and up to

a 7.3 fold increase in AdV cell specific titer (116). Despite these insights, perfusion modes

still remain the best strategy to successfully produce AdV at high cell densities.

3.2.6. Physical parameters and process monitoring

Physical parameters such as temperature, pH and dissolved oxygen are usually well

established when manufacturing AdV at larger scales. The impact of such parameters on

AdV production has been previously reviewed (91, 93), therefore only optimal values

used to produce AdV are discussed.

Chapter I

16

Production of adenoviruses is typically set to cell growth temperature of 37°C, although

previous works showed that by decreasing the temperature to 35°C an improvement in

virus production can be obtained. The common pH values for adenoviruses have been

established as 7.2 – 7.3. On the other hand, production of canine adenoviruses in stirred

tank bioreactor at pH 7.4 showed production yields similar to human adenoviruses.

Despite the lack of reports addressing the effect of dissolved oxygen (DO) on adenovirus

production, most of the controlled processes set DO to values higher than 30% (92, 93).

Small-scale productions are typically monitored with offline sampling. While such offline

methods can be used to monitor some parameters, such as cell concentration and pH,

online methods are very useful for stirred-tank bioreactor productions at large-scale,

specially to select TOI and TOH. The cell/infection status can be monitored through a

variety of noninvasive online measurements. In particular, cell density can be estimated

by the oxygen uptake rate and the capacitance levels (113, 117, 118), as they can also be

used to monitor infection kinetics and confirm the maximum productivity point and

harvest timing.

3.3. Production process of helper-dependent vectors

Despite the potential of HDV, extensive use of such vectors for gene transfer experiments

has been restrained by two main bottlenecks: production yields and HV contamination.

Similarly to first generation vectors, HDV particles need to be rescued from linearized

plasmid after transfecting producer cells. To provide all the viral elements required for

HDV replication, cells must be also infected with HV (see section 3.1.4.). Typically, low

yields from this initial step are obtained and/or multiple rounds of HDV production are

needed until the desired vectors titer is achieved. Reducing the number of amplifications

is a primary condition to establish a robust production process. This also minimizes the

possibility of recombination between HV/HDV and producer cell line, avoiding the

occurrence of RCA, packaging-competent or recombinant HV, which have a propagation

advantage when compared to HDV (119, 120). Therefore, the most significant advances

for scalable production of HDV are designed early in the transfection step to maximize

volumetric productivity and set amplification rounds to its minimum (105).

The importance of recombinase system, such as Cre/loxP, with high levels of Cre to avoid

HV propagation is unquestionable (121). In fact, considering that the remaining HV

contaminant was due to limiting Cre levels that permitted HV to escape packaging signal

Introduction

17

excision (121), major advances were made to increase the levels of recombinase during

HDV production (106). Moreover, the definition of optimal MOI ratio showed a critical

impact either in maximizing HDV propagation, but also in reducing HV contamination.

The use of high HV MOI, besides being unnecessary and even negative for HDV

production (104), implicates the development of alternative designs to attain higher

levels of Cre than those supported by the cell line to reduce HV contamination (106).

Further, this also leads to the accumulation of higher levels of excised helper DNA

molecules (after Cre recombinase excision), increasing the chance of recombination

between viral vectors. In fact, some authors showed that these helper DNA molecules,

being prone to rearrangements, contributed to the generation of recombination between

viral vectors, in which helper vectors with rearranged genomes had a growth advantage

(120). In addition, some studies show a relatively high and/or great inconsistency in

maintaining PP:IP ratios among different preparations of same HDV (106, 122). Special

attention must be paid to this, as the PP:IP ratio of clinical grade adenoviruses is limited

by Food and Drug Administration (see section 4.4.).

3.4. Downstream process for adenovirus vectors

Since downstream processing is not in the scope of this work, only a brief overview is

presented. Traditional purifications of adenovirus vectors are performed by cesium

chloride (CsCl) density gradient ultracentrifugation. While useful for preparations at

laboratory scales, scalable purification of adenovirus vectors relies on membrane and

chromatographic processes (91, 123, 124). The main aim of downstream processing is to

eliminate contaminants, either process related (e.g., bovine serum albumin, Benzonase,

extractables, and leachables) or product related (e.g., host cell proteins, DNA,

proteoglycans, and glycosaminoglycans); other product-related impurities include free

proteins, aggregates, and empty capsids (125). The ultimate goal is to obtain a product

with high purity, potency, and quality, which can meet the stringent guidelines of the

regulatory authorities, such as the FDA and EMA. After harvesting and prior purification

two main steps are employed to production bulk: cell lysis, to release intracellular

adenoviruses and increase the yield, and genomic DNA breakdown, to facilitate DNA

removal. Purification can then be divided in three major steps: clarification,

concentration/purification and polishing. A suitable clarification step should remove cell

debris and large aggregates and can be performed by centrifugation, widely used at

Chapter I

18

laboratory scales, or continuous flow centrifugation and microfiltration at industrially

relevant scales (91, 123, 126, 127). The concentration/purification step aims to reduce

the stream volume and facilitate the upfront equipment and materials. In this step, low

molecular weight proteins, fragmented DNA and other impurities are further removed

and ultrafiltration or chromatography columns are usually employed (91, 123, 128). The

step of polishing is usually performed using chromatographic processes and applied to

remove remaining impurities closely related to the product of interest (91, 128). The final

step for the production of GMP-grade adenovirus product is a filtration using a 0.2 μm

sterile membrane. In the final product, host cell DNA and protein levels should be bellow

the specifications set by the European Pharmacopoeia (Ph. Eur 5.2.3) and the World

Health Organization (WHO expert Committee on Biological Standardization, WHO

Technical Report Series 878, 47th report, 1998) (124).

4. Concerns in the manufacturing of adenovirus vectors for product release

Commonly to vaccines for human use, release of adenovirus vectored products includes

tests for potency, general safety, sterility, purity, identity, and constituent materials. In

addition, screening for bacterial endotoxins and pyrogens are added when the product is

intended for use by injection (U.S. Pharmacopeial Convention and USP-NF bulletin,

http://www.usp.org). Every biopharmaceutical has its own characteristics that are

considered when developing and qualifying these tests. Therefore this section is focused

in the tests connected to the upstream manufacturing process of adenovirus vectors:

extraneous agents, RCAs, vector genetic stability and quantity/potency.

4.1. Extraneous agents

Viral inactivation and clearance steps are generally performed in the production of

inactivated vaccines which contribute in removing potential extraneous agents from final

product. However, these procedures cannot be applied in adenovirus vectors for gene

therapy, as their bioactivity is critical to gene transfer. Safety strategies need to be thus

implemented to lower the risk of introduction and carryover of contaminants, such as

extraneous agents, adventitious viruses, and transmissible spongiforme encephalopathy

causing agents. According to good manufacturing practices, a track record of all the

materials used in construction and manufacturing of adenovirus vectors must be kept.

Master cell banks and master viral seeds must be extensively tested to confirm the

Introduction

19

absence of adventitious viruses according to the ICH Q5A and Q5D guidelines. The use of

animal-derived components should be avoided as much as possible, as it represents a

potential source of contamination (94). In situations where the use of animal components

is unavoidable, excellent traceability and testing are performed to discard any risk of

enter and transfer of extraneous agents to the manufacturing process. The screening for

adventitious has relied on the use of in vitro infectivity assays, in vivo studies and specific

polymerase chain reaction (PCR) tests. More recently, massive parallel sequencing has

been also applied to this end (129).

4.2. Replication competent adenovirus

Contamination of clinical batches with RCA is an important safety concern. Possible

consequences range from increased local inflammatory responses and tissue damage due

to uncontrolled systemic replication in immune-compromised individuals. Although the

issue of RCA formation by homologous recombination was solved by the introduction of

new cell lines like PER.C6 (see section 3.1), regulatory agencies still require testing to

confirm their absence in clinical batches (124). The maximum contamination level is set

to 1 RCA per 3x1010 VP and is based on the current FDA guidelines for HAdV5 vectors

(FDA Gene Therapy Letter, 2000). The detection of replicating virus is based on the

screening for cytopatic effect (CPE) after inoculating non-complementing cell lines. CPE

positive results are then confirmed by a more specific assay such as PCR or

immunofluorescence (130).

4.3. Vector genetic stability

To confirm product stability throughout the manufacturing process, genetic stability of

the viral vector is tested. To demonstrate this, viral vector is propagated for a number of

passages beyond the level used in production, generally 5 passages (more information at

U.S. Pharmacopeial Convention and USP-NF bulletin, http://www.usp.org). Mutation

frequency in replication-deficient adenovirus is considered rare, however such analysis is

important to evaluate that transgene region and corresponding expression is maintained,

and critical when considering the use of oncolytic adenoviruses, which, unlike gene

transfer-strict vectors, are intended to further propagate in a clinical setting (45).

Extended propagation permits the detection of any recombinant or mutant vector that

Chapter I

20

have a replication advantage over the target vector. For this purpose, PCR detection

combined with sequence analysis of PCR fragments can be used for screening any

variations in transgene sequence that might occur (124).

4.4. Quantity and potency

Quantification of viral particles is important for monitoring yields during process

development, but also in final product to control of the amount of viral protein injected

within an acceptable safety window. Typically, absorvance measurements are used to

quantify physical particles based in the correlations between adenovirus preparations

and absorvance at 260 nm described by Maizel et al. (1968). While these measurements

require purified and concentrated preparations, alternative methods have been

developed to allow adenovirus quantification during process development and,

simultaneously, improve the limit of detection (see bellow) (91).

Adenovirus vectors exert their clinical effect by transducing target cells. Therefore,

quantification of infectious particles is required during the overall production process, for

product release, and stability assessments of viral preparations. In fact, a parameter

considered to represent the quality of preparations is the ratio between the amount of

physical and infectious particles. For gene therapy programs using adenoviruses, PP:IP

ratio must be <30:1 (FDA Gene Therapy Letter, 2000).

Standard plaque assays or tissue culture infectious dose (TCID50) assays using

complementing cell lines can be employed to quantify infectious titers. To improve

accuracy and precision of these assays, alternative methods can be applied which are

based in quantitative PCR techniques or in the detection of the transgene expression.

5. Scope of the thesis

The central objective of this PhD thesis was to develop a scalable production process for

∆E1 and HD CAV-2 vectors using MDCK derived cell lines as producer cells, assuring

optimal cell performance and maximum productivity. Critical cell culture and virus

infection parameters were studied and combined with a better understanding of cell

physiological state and viral cycle steps behind a successful production process.

To accomplish this, the work was organized according to the following:

(I) Evaluate MDCK derived cell lines as cell substrates for the production of CAV-2 vectors.

Introduction

21

Since MDCK cells are accepted in Food and Drug Administration (FDA) and European

Medicine Agency (EMA) and used for the production of many viral vaccines, the use of an

MDCK-based cell lines would facilitate the regulatory approval for the clinical grade

production of CAV-2 vectors. Accordingly, MDCK-E1 and MDCK-E1-Cre cell lines were

established (Chapter II).

(II) Develop a scalable production process for CAV-2 vectors

The need for significant amounts of clinical grade adenovirus vectors, which in some

cases may reach 1013 total particles/patient or 1011 infectious particles/patient, requires

efficient and robust processes for production and purification at large-scale compliant

with good manufacturing practices (GMP) (12). Therefore, current lab-scale protocols

must be adapted and transferred to a scalable stirred culture system (Chapter III).

(III) Establish best infection conditions for HD CAV-2 production

Process optimization for ΔE1 vectors is relatively straightforward. However, given the

production system complexity, special attention must be paid to HDV propagation. Using

the Cre/loxP system, producer cells must express Cre-recombinase, which can be toxic to

cells (131), and production is performed under co-infection. Therefore, cell line stability

and co-infection conditions must be well defined to ensure production process

reproducibility (Chapter IV).

(IV) Identify the limitations in HD CAV-2 vectors production from an adenovirus replication

cycle perspective.

Despite the potentialities of HDV for gene therapy, one important bottleneck is the yields,

which compromises process scale-up, viral particles quality and its availability to pre and

clinical trials. Still, the low productive viral cycle of HDV has been somehow neglected

(Chapter V).

6. Author contribution

Paulo Fernandes wrote this chapter based on the referred bibliography.

Chapter I

22

7. References

1. Naldini, L. 2009. Medicine. A comeback for gene therapy. Science 326:805-806. 2. Nathwani, A. C., E. G. Tuddenham, S. Rangarajan, C. Rosales, J. McIntosh, D. C. Linch,

P. Chowdary, A. Riddell, A. J. Pie, C. Harrington, J. O'Beirne, K. Smith, J. Pasi, B. Glader, P. Rustagi, C. Y. Ng, M. A. Kay, J. Zhou, Y. Spence, C. L. Morton, J. Allay, J. Coleman, S. Sleep, J. M. Cunningham, D. Srivastava, E. Basner-Tschakarjan, F. Mingozzi, K. A. High, J. T. Gray, U. M. Reiss, A. W. Nienhuis, and A. M. Davidoff. 2011. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med 365:2357-2365.

3. Bainbridge, J. W., A. J. Smith, S. S. Barker, S. Robbie, R. Henderson, K. Balaggan, A. Viswanathan, G. E. Holder, A. Stockman, N. Tyler, S. Petersen-Jones, S. S. Bhattacharya, A. J. Thrasher, F. W. Fitzke, B. J. Carter, G. S. Rubin, A. T. Moore, and R. R. Ali. 2008. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med 358:2231-2239.

4. Maguire, A. M., F. Simonelli, E. A. Pierce, E. N. Pugh, Jr., F. Mingozzi, J. Bennicelli, S. Banfi, K. A. Marshall, F. Testa, E. M. Surace, S. Rossi, A. Lyubarsky, V. R. Arruda, B. Konkle, E. Stone, J. Sun, J. Jacobs, L. Dell'Osso, R. Hertle, J. X. Ma, T. M. Redmond, X. Zhu, B. Hauck, O. Zelenaia, K. S. Shindler, M. G. Maguire, J. F. Wright, N. J. Volpe, J. W. McDonnell, A. Auricchio, K. A. High, and J. Bennett. 2008. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med 358:2240-2248.

5. Testa, F., A. M. Maguire, S. Rossi, E. A. Pierce, P. Melillo, K. Marshall, S. Banfi, E. M. Surace, J. Sun, C. Acerra, J. F. Wright, J. Wellman, K. A. High, A. Auricchio, J. Bennett, and F. Simonelli. 2013. Three-year follow-up after unilateral subretinal delivery of adeno-associated virus in patients with Leber congenital Amaurosis type 2. Ophthalmology 120:1283-1291.

6. Hacein-Bey-Abina, S., J. Hauer, A. Lim, C. Picard, G. P. Wang, C. C. Berry, C. Martinache, F. Rieux-Laucat, S. Latour, B. H. Belohradsky, L. Leiva, R. Sorensen, M. Debre, J. L. Casanova, S. Blanche, A. Durandy, F. D. Bushman, A. Fischer, and M. Cavazzana-Calvo. 2010. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 363:355-364.

7. Bryant, L. M., D. M. Christopher, A. R. Giles, C. Hinderer, J. L. Rodriguez, J. B. Smith, E. A. Traxler, J. Tycko, A. P. Wojno, and J. M. Wilson. 2013. Lessons learned from the clinical development and market authorization of Glybera. Human gene therapy. Clinical development 24:55-64.

8. Choi, I. K., and C. O. Yun. 2013. Recent developments in oncolytic adenovirus-based immunotherapeutic agents for use against metastatic cancers. Cancer gene therapy 20:70-76.

9. Ledley, F. D., L. M. McNamee, V. Uzdil, and I. W. Morgan. 2014. Why commercialization of gene therapy stalled; examining the life cycles of gene therapy technologies. Gene Ther 21:188-194.

10. McConnell, M. J., and M. J. Imperiale. 2004. Biology of adenovirus and its use as a vector for gene therapy. Hum Gene Ther 15:1022-1033.

11. Tatsis, N., and H. C. Ertl. 2004. Adenoviruses as vaccine vectors. Mol Ther 10:616-629. 12. Dormond, E., M. Perrier, and A. Kamen. 2009. From the first to the third generation

adenoviral vector: what parameters are governing the production yield? Biotechnol Adv 27:133-144.

13. Soudais, C., S. Boutin, S. S. Hong, M. Chillon, O. Danos, J. M. Bergelson, P. Boulanger, and E. J. Kremer. 2000. Canine adenovirus type 2 attachment and internalization: coxsackievirus-adenovirus receptor, alternative receptors, and an RGD-independent pathway. J Virol 74:10639-10649.

14. Wickham, T. J., P. Mathias, D. A. Cheresh, and G. R. Nemerow. 1993. Integrins alpha v beta 3 and alpha v beta 5 promote adenovirus internalization but not virus attachment. Cell 73:309-319.

Introduction

23

15. Nemerow, G. R., and P. L. Stewart. 1999. Role of alpha(v) integrins in adenovirus cell entry and gene delivery. Microbiology and molecular biology reviews : MMBR 63:725-734.

16. Greber, U. F., M. Suomalainen, R. P. Stidwill, K. Boucke, M. W. Ebersold, and A. Helenius. 1997. The role of the nuclear pore complex in adenovirus DNA entry. EMBO J 16:5998-6007.

17. Russell, W. C. 2000. Update on adenovirus and its vectors. J Gen Virol 81:2573-2604. 18. Berk, A. J. 1986. Functions of adenovirus E1A. Cancer surveys 5:367-387. 19. Rao, L., M. Debbas, P. Sabbatini, D. Hockenbery, S. Korsmeyer, and E. White. 1992.

The adenovirus E1A proteins induce apoptosis, which is inhibited by the E1B 19-kDa and Bcl-2 proteins. Proceedings of the National Academy of Sciences of the United States of America 89:7742-7746.

20. Gallimore, P. H., and A. S. Turnell. 2001. Adenovirus E1A: remodelling the host cell, a life or death experience. Oncogene 20:7824-7835.

21. Moran, E. 1993. DNA tumor virus transforming proteins and the cell cycle. Current opinion in genetics & development 3:63-70.

22. Berk, A. J. 1986. Adenovirus promoters and E1A transactivation. Annual review of genetics 20:45-79.

23. Debbas, M., and E. White. 1993. Wild-type p53 mediates apoptosis by E1A, which is inhibited by E1B. Genes & development 7:546-554.

24. Schmitz, M. L., A. Indorf, F. P. Limbourg, H. Stadtler, E. B. Traenckner, and P. A. Baeuerle. 1996. The dual effect of adenovirus type 5 E1A 13S protein on NF-kappaB activation is antagonized by E1B 19K. Molecular and cellular biology 16:4052-4063.

25. Doerfler, W., and P. Böhm. 1995. The Molecular repertoire of adenoviruses. Springer. 26. Lochmuller, H., A. Jani, J. Huard, S. Prescott, M. Simoneau, B. Massie, G. Karpati, and

G. Acsadi. 1994. Emergence of early region 1-containing replication-competent adenovirus in stocks of replication-defective adenovirus recombinants (delta E1 + delta E3) during multiple passages in 293 cells. Hum Gene Ther 5:1485-1491.

27. Hehir, K. M., D. Armentano, L. M. Cardoza, T. L. Choquette, P. B. Berthelette, G. A. White, L. A. Couture, M. B. Everton, J. Keegan, J. M. Martin, D. A. Pratt, M. P. Smith, A. E. Smith, and S. C. Wadsworth. 1996. Molecular characterization of replication-competent variants of adenovirus vectors and genome modifications to prevent their occurrence. J Virol 70:8459-8467.

28. Smith, J. G., and S. L. Eck. 1999. Molecular characterization of an adenoviral vector resulting from both homologous and nonhomologous recombination. Cancer gene therapy 6:475-481.

29. Zhu, J., M. Grace, J. Casale, A. T. Chang, M. L. Musco, R. Bordens, R. Greenberg, E. Schaefer, and S. R. Indelicato. 1999. Characterization of replication-competent adenovirus isolates from large-scale production of a recombinant adenoviral vector. Hum Gene Ther 10:113-121.

30. Murakami, P., E. Pungor, J. Files, L. Do, R. van Rijnsoever, R. Vogels, A. Bout, and M. McCaman. 2002. A single short stretch of homology between adenoviral vector and packaging cell line can give rise to cytopathic effect-inducing, helper-dependent E1-positive particles. Hum Gene Ther 13:909-920.

31. Kovesdi, I., and S. J. Hedley. 2010. Adenoviral producer cells. Viruses 2:1681-1703. 32. Fallaux, F. J., A. Bout, I. van der Velde, D. J. van den Wollenberg, K. M. Hehir, J.

Keegan, C. Auger, S. J. Cramer, H. van Ormondt, A. J. van der Eb, D. Valerio, and R. C. Hoeben. 1998. New helper cells and matched early region 1-deleted adenovirus vectors prevent generation of replication-competent adenoviruses. Hum Gene Ther 9:1909-1917.

33. Farson, D., L. Tao, D. Ko, Q. Li, D. Brignetti, K. Segawa, D. Mittelstaedt, T. Harding, D. C. Yu, and Y. Li. 2006. Development of novel E1-complementary cells for adenoviral production free of replication-competent adenovirus. Mol Ther 14:305-311.

34. Howe, J. A., P. Pelka, D. Antelman, C. Wilson, D. Cornell, W. Hancock, M. Ramachandra, J. Avanzini, M. Horn, K. Wills, S. Sutjipto, and R. Ralston. 2006.

Chapter I

24

Matching complementing functions of transformed cells with stable expression of selected viral genes for production of E1-deleted adenovirus vectors. Virology 345:220-230.

35. Liu, T. C., E. Galanis, and D. Kirn. 2007. Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress. Nature clinical practice. Oncology 4:101-117.

36. Choi, J. W., J. S. Lee, S. W. Kim, and C. O. Yun. 2012. Evolution of oncolytic adenovirus for cancer treatment. Advanced drug delivery reviews 64:720-729.

37. Heise, C., T. Hermiston, L. Johnson, G. Brooks, A. Sampson-Johannes, A. Williams, L. Hawkins, and D. Kirn. 2000. An adenovirus E1A mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nat Med 6:1134-1139.

38. Lamfers, M. L., J. Grill, C. M. Dirven, V. W. Van Beusechem, B. Geoerger, J. Van Den Berg, R. Alemany, J. Fueyo, D. T. Curiel, G. Vassal, H. M. Pinedo, W. P. Vandertop, and W. R. Gerritsen. 2002. Potential of the conditionally replicative adenovirus Ad5-Delta24RGD in the treatment of malignant gliomas and its enhanced effect with radiotherapy. Cancer Res 62:5736-5742.

39. Bischoff, J. R., D. H. Kirn, A. Williams, C. Heise, S. Horn, M. Muna, L. Ng, J. A. Nye, A. Sampson-Johannes, A. Fattaey, and F. McCormick. 1996. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 274:373-376.

40. Freytag, S. O., K. R. Rogulski, D. L. Paielli, J. D. Gilbert, and J. H. Kim. 1998. A novel three-pronged approach to kill cancer cells selectively: concomitant viral, double suicide gene, and radiotherapy. Hum Gene Ther 9:1323-1333.

41. Yu, D. C., Y. Chen, J. Dilley, Y. Li, M. Embry, H. Zhang, N. Nguyen, P. Amin, J. Oh, and D. R. Henderson. 2001. Antitumor synergy of CV787, a prostate cancer-specific adenovirus, and paclitaxel and docetaxel. Cancer Res 61:517-525.

42. Bristol, J. A., M. Zhu, H. Ji, M. Mina, Y. Xie, L. Clarke, S. Forry-Schaudies, and D. L. Ennist. 2003. In vitro and in vivo activities of an oncolytic adenoviral vector designed to express GM-CSF. Molecular Therapy 7:755-764.

43. Yuk, I. H., M. M. Olsen, S. Geyer, and S. P. Forestell. 2004. Perfusion cultures of human tumor cells: a scalable production platform for oncolytic adenoviral vectors. Biotechnol Bioeng 86:637-642.

44. Longley, R., Jr., L. Radzniak, M. Santoro, Y.-S. Tsao, R. G. Condon, P. Lio, M. Voloch, and Z. Liu. 2005. Development of a Serum-free Suspension Process for the Production of a Conditionally Replicating Adenovirus using A549 Cells. Cytotechnology 49:161-171.

45. Working, P. K., A. Lin, and F. Borellini. 2005. Meeting product development challenges in manufacturing clinical grade oncolytic adenoviruses. Oncogene 24:7792-7801.

46. Nevins, J. R. 1981. Mechanism of activation of early viral transcription by the adenovirus E1A gene product. Cell 26:213-220.

47. Gaynor, R. B., and A. J. Berk. 1983. Cis-acting induction of adenovirus transcription. Cell 33:683-693.

48. Imperiale, M. J., H. T. Kao, L. T. Feldman, J. R. Nevins, and S. Strickland. 1984. Common control of the heat shock gene and early adenovirus genes: evidence for a cellular E1A-like activity. Molecular and cellular biology 4:867-874.

49. Swaminathan, S., and B. Thimmapaya. 1995. Regulation of Adenovirus E2 Transcription Unit, p. 177-194. In W. Doerfler and P. Böhm (ed.), The Molecular Repertoire of Adenoviruses III, vol. 199/3. Springer Berlin Heidelberg.

50. Weitzman, M. D. 2005. Functions of the adenovirus E4 proteins and their impact on viral vectors. Frontiers in bioscience : a journal and virtual library 10:1106-1117.

51. Bridge, E., and G. Ketner. 1989. Redundant control of adenovirus late gene expression by early region 4. J Virol 63:631-638.

52. Huang, M. M., and P. Hearing. 1989. Adenovirus early region 4 encodes two gene products with redundant effects in lytic infection. J Virol 63:2605-2615.

53. Gao, G. P., Y. Yang, and J. M. Wilson. 1996. Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy. J Virol 70:8934-8943.

54. Lusky, M., M. Christ, K. Rittner, A. Dieterle, D. Dreyer, B. Mourot, H. Schultz, F. Stoeckel, A. Pavirani, and M. Mehtali. 1998. In vitro and in vivo biology of recombinant adenovirus vectors with E1, E1/E2A, or E1/E4 deleted. J Virol 72:2022-2032.

Introduction

25

55. O'Neal, W. K., H. Zhou, N. Morral, E. Aguilar-Cordova, J. Pestaner, C. Langston, B. Mull, Y. Wang, A. L. Beaudet, and B. Lee. 1998. Toxicological comparison of E2a-deleted and first-generation adenoviral vectors expressing alpha1-antitrypsin after systemic delivery. Hum Gene Ther 9:1587-1598.

56. Christ, M., B. Louis, F. Stoeckel, A. Dieterle, L. Grave, D. Dreyer, J. Kintz, D. Ali Hadji, M. Lusky, and M. Mehtali. 2000. Modulation of the inflammatory properties and hepatotoxicity of recombinant adenovirus vectors by the viral E4 gene products. Hum Gene Ther 11:415-427.

57. Brough, D. E., C. Hsu, V. A. Kulesa, G. M. Lee, L. J. Cantolupo, A. Lizonova, and I. Kovesdi. 1997. Activation of transgene expression by early region 4 is responsible for a high level of persistent transgene expression from adenovirus vectors in vivo. J Virol 71:9206-9213.

58. Wang, Q., G. Greenburg, D. Bunch, D. Farson, and M. H. Finer. 1997. Persistent transgene expression in mouse liver following in vivo gene transfer with a delta E1/delta E4 adenovirus vector. Gene Ther 4:393-400.

59. Segura, M. M., R. Alba, A. Bosch, and M. Chillon. 2008. Advances in helper-dependent adenoviral vector research. Curr Gene Ther 8:222-235.

60. Parks, R. J., and F. L. Graham. 1997. A helper-dependent system for adenovirus vector production helps define a lower limit for efficient DNA packaging. J Virol 71:3293-3298.

61. Parks, R. J., L. Chen, M. Anton, U. Sankar, M. A. Rudnicki, and F. L. Graham. 1996. A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proceedings of the National Academy of Sciences of the United States of America 93:13565-13570.

62. Ng, P., C. Beauchamp, C. Evelegh, R. Parks, and F. L. Graham. 2001. Development of a FLP/frt system for generating helper-dependent adenoviral vectors. Mol Ther 3:809-815.

63. Umana, P., C. A. Gerdes, D. Stone, J. R. Davis, D. Ward, M. G. Castro, and P. R. Lowenstein. 2001. Efficient FLPe recombinase enables scalable production of helper-dependent adenoviral vectors with negligible helper-virus contamination. Nat Biotechnol 19:582-585.

64. Garnett, C. T., D. Erdman, W. Xu, and L. R. Gooding. 2002. Prevalence and quantitation of species C adenovirus DNA in human mucosal lymphocytes. J Virol 76:10608-10616.

65. Bangari, D. S., and S. K. Mittal. 2006. Current strategies and future directions for eluding adenoviral vector immunity. Current gene therapy 6:215-226.

66. Paillard, F. 1997. Advantages of non-human adenoviruses versus human adenoviruses. Human gene therapy 8:2007-2009.

67. Thomas, C. E., D. Birkett, I. Anozie, M. G. Castro, and P. R. Lowenstein. 2001. Acute direct adenoviral vector cytotoxicity and chronic, but not acute, inflammatory responses correlate with decreased vector-mediated transgene expression in the brain. Mol Ther 3:36-46.

68. Yang, Y., Q. Su, and J. M. Wilson. 1996. Role of viral antigens in destructive cellular immune responses to adenovirus vector-transduced cells in mouse lungs. Journal of virology 70:7209-7212.

69. Sumida, S. M., D. M. Truitt, A. A. Lemckert, R. Vogels, J. H. Custers, M. M. Addo, S. Lockman, T. Peter, F. W. Peyerl, M. G. Kishko, S. S. Jackson, D. A. Gorgone, M. A. Lifton, M. Essex, B. D. Walker, J. Goudsmit, M. J. Havenga, and D. H. Barouch. 2005. Neutralizing antibodies to adenovirus serotype 5 vaccine vectors are directed primarily against the adenovirus hexon protein. J Immunol 174:7179-7185.

70. Lopez-Gordo, E., Podgorski, II, N. Downes, and R. Alemany. 2014. Circumventing antivector immunity: potential use of nonhuman adenoviral vectors. Hum Gene Ther 25:285-300.

71. Kuriyama, S., K. Tominaga, M. Kikukawa, T. Nakatani, H. Tsujinoue, M. Yamazaki, S. Nagao, Y. Toyokawa, A. Mitoro, and H. Fukui. 1998. Inhibitory effects of human sera on adenovirus-mediated gene transfer into rat liver. Anticancer research 18:2345-2351.

72. Vlachaki, M. T., A. Hernandez-Garcia, M. Ittmann, M. Chhikara, L. K. Aguilar, X. Zhu, B. S. Teh, E. B. Butler, S. Woo, T. C. Thompson, H. Barrera-Saldana, and E. Aguilar-

Chapter I

26

Cordova. 2002. Impact of preimmunization on adenoviral vector expression and toxicity in a subcutaneous mouse cancer model. Mol Ther 6:342-348.

73. Raper, S. E., N. Chirmule, F. S. Lee, N. A. Wivel, A. Bagg, G. P. Gao, J. M. Wilson, and M. L. Batshaw. 2003. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 80:148-158.

74. Seshidhar Reddy, P., S. Ganesh, M. P. Limbach, T. Brann, A. Pinkstaff, M. Kaloss, M. Kaleko, and S. Connelly. 2003. Development of adenovirus serotype 35 as a gene transfer vector. Virology 311:384-393.

75. Vogels, R., D. Zuijdgeest, R. van Rijnsoever, E. Hartkoorn, I. Damen, M. P. de Bethune, S. Kostense, G. Penders, N. Helmus, W. Koudstaal, M. Cecchini, A. Wetterwald, M. Sprangers, A. Lemckert, O. Ophorst, B. Koel, M. van Meerendonk, P. Quax, L. Panitti, J. Grimbergen, A. Bout, J. Goudsmit, and M. Havenga. 2003. Replication-deficient human adenovirus type 35 vectors for gene transfer and vaccination: efficient human cell infection and bypass of preexisting adenovirus immunity. J Virol 77:8263-8271.

76. Gao, W., P. D. Robbins, and A. Gambotto. 2003. Human adenovirus type 35: nucleotide sequence and vector development. Gene Ther 10:1941-1949.

77. Sirena, D., Z. Ruzsics, W. Schaffner, U. F. Greber, and S. Hemmi. 2005. The nucleotide sequence and a first generation gene transfer vector of species B human adenovirus serotype 3. Virology 343:283-298.

78. Stone, D., S. Ni, Z. Y. Li, A. Gaggar, N. DiPaolo, Q. Feng, V. Sandig, and A. Lieber. 2005. Development and assessment of human adenovirus type 11 as a gene transfer vector. J Virol 79:5090-5104.

79. Havenga, M., R. Vogels, D. Zuijdgeest, K. Radosevic, S. Mueller, M. Sieuwerts, F. Weichold, I. Damen, J. Kaspers, A. Lemckert, M. van Meerendonk, R. van der Vlugt, L. Holterman, D. Hone, Y. Skeiky, R. Mintardjo, G. Gillissen, D. Barouch, J. Sadoff, and J. Goudsmit. 2006. Novel replication-incompetent adenoviral B-group vectors: high vector stability and yield in PER.C6 cells. J Gen Virol 87:2135-2143.

80. van Olphen, A. L., S. K. Tikoo, and S. K. Mittal. 2002. Characterization of bovine adenovirus type 3 E1 proteins and isolation of E1-expressing cell lines. Virology 295:108-118.

81. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

82. Fernandes, P., V. M. Santiago, A. F. Rodrigues, H. Tomas, E. J. Kremer, P. M. Alves, and A. S. Coroadinha. 2013. Impact of E1 and Cre on adenovirus vector amplification: developing MDCK CAV-2-E1 and E1-Cre transcomplementing cell lines. PLoS One 8:e60342.

83. Farina, S. F., G. P. Gao, Z. Q. Xiang, J. J. Rux, R. M. Burnett, M. R. Alvira, J. Marsh, H. C. Ertl, and J. M. Wilson. 2001. Replication-defective vector based on a chimpanzee adenovirus. J Virol 75:11603-11613.

84. Roy, S., G. Gao, Y. Lu, X. Zhou, M. Lock, R. Calcedo, and J. M. Wilson. 2004. Characterization of a family of chimpanzee adenoviruses and development of molecular clones for gene transfer vectors. Hum Gene Ther 15:519-530.

85. Roy, S., Y. Zhi, G. P. Kobinger, J. Figueredo, R. Calcedo, J. R. Miller, H. Feldmann, and J. M. Wilson. 2006. Generation of an adenoviral vaccine vector based on simian adenovirus 21. J Gen Virol 87:2477-2485.

86. Roy, S., D. S. Clawson, O. Lavrukhin, A. Sandhu, J. Miller, and J. M. Wilson. 2007. Rescue of chimeric adenoviral vectors to expand the serotype repertoire. J Virol Methods 141:14-21.

87. Bru, T., S. Salinas, and E. J. Kremer. 2010. An update on canine adenovirus type 2 and its vectors. Viruses 2:2134-2153.

88. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

Introduction

27

89. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

90. Lau, A. A., T. Rozaklis, S. Ibanes, A. J. Luck, H. Beard, S. Hassiotis, K. Mazouni, J. J. Hopwood, E. J. Kremer, and K. M. Hemsley. 2012. Helper-dependent canine adenovirus vector-mediated transgene expression in a neurodegenerative lysosomal storage disorder. Gene 491:53-57.

91. Altaras, N. E., J. G. Aunins, R. K. Evans, A. Kamen, J. O. Konz, and J. J. Wolf. 2005. Production and formulation of adenovirus vectors. Adv Biochem Eng Biotechnol 99:193-260.

92. Silva, A., P. Fernandes, M. Q. Sousa, and P. Alves. 2014. Scalable Production of Adenovirus Vectors, p. 175-196. In M. Chillón and A. Bosch (ed.), Adenovirus, vol. 1089. Humana Press.

93. Silva, A. C., C. Peixoto, T. Lucas, C. Kuppers, P. E. Cruz, P. M. Alves, and S. Kochanek. 2010. Adenovirus vector production and purification. Curr Gene Ther 10:437-455.

94. Falkner, E., H. Appl, C. Eder, U. M. Losert, H. Schoffl, and W. Pfaller. 2006. Serum free cell culture: the free access online database. Toxicol In Vitro 20:395-400.

95. Croyle, M. A., B. J. Roessler, B. L. Davidson, J. M. Hilfinger, and G. L. Amidon. 1998. Factors that influence stability of recombinant adenoviral preparations for human gene therapy. Pharm Dev Technol 3:373-383.

96. Croyle, M. A., X. Cheng, and J. M. Wilson. 2001. Development of formulations that enhance physical stability of viral vectors for gene therapy. Gene Ther 8:1281-1290.

97. Obenauer-Kutner, L. J., P. M. Ihnat, T. Y. Yang, B. J. Dovey-Hartman, A. Balu, C. Cullen, R. W. Bordens, and M. J. Grace. 2002. The use of field emission scanning electron microscopy to assess recombinant adenovirus stability. Hum Gene Ther 13:1687-1696.

98. Evans, R. K., D. K. Nawrocki, L. A. Isopi, D. M. Williams, D. R. Casimiro, S. Chin, M. Chen, D. M. Zhu, J. W. Shiver, and D. B. Volkin. 2004. Development of stable liquid formulations for adenovirus-based vaccines. J Pharm Sci 93:2458-2475.

99. Cruz, P. E., A. C. Silva, A. Roldao, M. Carmo, M. J. Carrondo, and P. M. Alves. 2006. Screening of novel excipients for improving the stability of retroviral and adenoviral vectors. Biotechnol Prog 22:568-576.

100. Huyghe, B. G., X. Liu, S. Sutjipto, B. J. Sugarman, M. T. Horn, H. M. Shepard, C. J. Scandella, and P. Shabram. 1995. Purification of a type 5 recombinant adenovirus encoding human p53 by column chromatography. Hum Gene Ther 6:1403-1416.

101. Blanche, F., B. Cameron, A. Barbot, L. Ferrero, T. Guillemin, S. Guyot, S. Somarriba, and D. Bisch. 2000. An improved anion-exchange HPLC method for the detection and purification of adenoviral particles. Gene Ther 7:1055-1062.

102. Zhang, S., C. Thwin, Z. Wu, and T. Cho. 2001. US Patent 6,194,191 B1. 103. Goerke, A. R., B. C. To, A. L. Lee, S. L. Sagar, and J. O. Konz. 2005. Development of a

novel adenovirus purification process utilizing selective precipitation of cellular DNA. Biotechnol Bioeng 91:12-21.

104. Dormond, E., M. Perrier, and A. Kamen. 2009. Identification of critical infection parameters to control helper-dependent adenoviral vector production. J Biotechnol 142:142-150.

105. Dormond, E., A. Meneses-Acosta, D. Jacob, Y. Durocher, R. Gilbert, M. Perrier, and A. Kamen. 2009. An efficient and scalable process for helper-dependent adenoviral vector production using polyethylenimine-adenofection. Biotechnol Bioeng 102:800-810.

106. Gonzalez-Aparicio, M., I. Mauleon, P. Alzuguren, M. Bunuales, G. Gonzalez-Aseguinolaza, C. San Martin, J. Prieto, and R. Hernandez-Alcoceba. 2011. Self-inactivating helper virus for the production of high-capacity adenoviral vectors. Gene Ther 18:1025-1033.

107. Nadeau, I., and A. Kamen. 2003. Production of adenovirus vector for gene therapy. Biotechnol Adv 20:475-489.

Chapter I

28

108. Nadeau, I., A. Garnier, J. Cote, B. Massie, C. Chavarie, and A. Kamen. 1996. Improvement of recombinant protein production with the human adenovirus/293S expression system using fed-batch strategies. Biotechnol Bioeng 51:613-623.

109. Nadeau, I., P. A. Gilbert, D. Jacob, M. Perrier, and A. Kamen. 2002. Low-protein medium affects the 293SF central metabolism during growth and infection with adenovirus. Biotechnol Bioeng 77:91-104.

110. Ferreira, T. B., A. L. Ferreira, M. J. Carrondo, and P. M. Alves. 2005. Effect of re-feed strategies and non-ammoniagenic medium on adenovirus production at high cell densities. J Biotechnol 119:272-280.

111. Henry, O., E. Dormond, M. Perrier, and A. Kamen. 2004. Insights into adenoviral vector production kinetics in acoustic filter-based perfusion cultures. Biotechnol Bioeng 86:765-774.

112. Cortin, V., J. Thibault, D. Jacob, and A. Garnier. 2004. High-titer adenovirus vector production in 293S cell perfusion culture. Biotechnol Prog 20:858-863.

113. Henry, O., M. Perrier, and A. Kamen. 2005. Metabolic flux analysis of HEK-293 cells in perfusion cultures for the production of adenoviral vectors. Metab Eng 7:467-476.

114. Nadeau, I., D. Jacob, M. Perrier, and A. Kamen. 2000. 293SF metabolic flux analysis during cell growth and infection with an adenoviral vector. Biotechnol Prog 16:872-884.

115. Zhang, C. H., T. B. Ferreira, P. E. Cruz, P. M. Alves, M. Haury, and M. J. T. Carrondo. 2006. The importance of 293 cell cycle phase on adenovirus vector production. Enzyme and Microbial Technology 39:1328-1332.

116. Ferreira, T. B., R. Perdigao, A. C. Silva, C. Zhang, J. G. Aunins, M. J. Carrondo, and P. M. Alves. 2009. 293 cell cycle synchronisation adenovirus vector production. Biotechnol Prog 25:235-243.

117. Garnier, A., J. Cote, I. Nadeau, A. Kamen, and B. Massie. 1994. Scale-up of the adenovirus expression system for the production of recombinant protein in human 293S cells. Cytotechnology 15:145-155.

118. Monica, T. J., T. Montgomery, J. L. Ayala, G. M. Schoofs, E. M. Whiteley, G. Roth, J. J. Garbutt, S. Harvey, and F. J. Castillo. 2000. Monitoring adenovirus infections with on-line and off-line methods. Biotechnol Prog 16:866-871.

119. Sandig, V., R. Youil, A. J. Bett, L. L. Franlin, M. Oshima, D. Maione, F. Wang, M. L. Metzker, R. Savino, and C. T. Caskey. 2000. Optimization of the helper-dependent adenovirus system for production and potency in vivo. Proc Natl Acad Sci U S A 97:1002-1007.

120. Ahn, M., A. Gamble, S. R. Witting, J. Magrisso, S. Surendran, S. Obici, and N. Morral. 2013. Vector and helper genome rearrangements occur during production of helper-dependent adenoviral vectors. Human gene therapy methods 24:1-10.

121. Ng, P., C. Evelegh, D. Cummings, and F. L. Graham. 2002. Cre levels limit packaging signal excision efficiency in the Cre/loxP helper-dependent adenoviral vector system. J Virol 76:4181-4189.

122. Kreppel, F., V. Biermann, S. Kochanek, and G. Schiedner. 2002. A DNA-based method to assay total and infectious particle contents and helper virus contamination in high-capacity adenoviral vector preparations. Hum Gene Ther 13:1151-1156.

123. Vicente, T., J. P. Mota, C. Peixoto, P. M. Alves, and M. J. Carrondo. 2011. Rational design and optimization of downstream processes of virus particles for biopharmaceutical applications: current advances. Biotechnol Adv 29:869-878.

124. Vellinga, J., J. P. Smith, A. Lipiec, D. Majhen, A. Lemckert, M. van Ooij, P. Ives, C. Yallop, J. Custers, and M. Havenga. 2014. Challenges in manufacturing adenoviral vectors for global vaccine product deployment. Hum Gene Ther 25:318-327.

125. Lee, D. S., B. M. Kim, and D. W. Seol. 2009. Improved purification of recombinant adenoviral vector by metal affinity membrane chromatography. Biochem Biophys Res Commun 378:640 -644.

126. Puig, M., J. Piedra, S. Miravet, and M. M. Segura. 2014. Canine adenovirus downstream processing protocol. Methods Mol Biol 1089:197-210.

Introduction

29

127. Jungbauer, A. 2013. Continuous downstream processing of biopharmaceuticals. Trends Biotechnol 31:479-492.

128. Segura, M. M., A. A. Kamen, and A. Garnier. 2011. Overview of current scalable methods for purification of viral vectors. Methods Mol Biol 737:89-116.

129. Onions, D., and J. Kolman. 2010. Massively parallel sequencing, a new method for detecting adventitious agents. Biologicals 38:377-380.

130. Marzio, G., E. Kerkvliet, J. A. Bogaards, S. Koelewijn, A. De Groot, L. Gijsbers, G. J. Weverling, R. Vogels, M. Havenga, J. Custers, M. G. Pau, J. Y. Guichoux, J. Lewis, and J. Goudsmit. 2007. A replication-competent adenovirus assay for E1-deleted Ad35 vectors produced in PER.C6 cells. Vaccine 25:2228-2237.

131. Loonstra, A., M. Vooijs, H. B. Beverloo, B. A. Allak, E. van Drunen, R. Kanaar, A. Berns, and J. Jonkers. 2001. Growth inhibition and DNA damage induced by Cre recombinase in mammalian cells. Proc Natl Acad Sci U S A 98:9209-9214.

Chapter I

30

Chapter II

Developing MDCK CAV-2-E1 and E1-Cre

Transcomplementing Cell Lines

This chapter is adapted from:

Fernandes P, Santiago VM, Rodrigues AF, Tomás H, Kremer EJ, Alves PM, Coroadinha AS.

(2013) Impact of E1 and Cre on Adenovirus Vector Amplification: Developing MDCK CAV-2-

E1 and E1-Cre Transcomplementing Cell Lines. PLoS ONE 8(4): e60342.

Chapter II

32

Abstract

Adenovirus vectors have been extensively studied through the manipulation of viral

genome. However, little attention is being paid to their producer cell-lines; cells are

selected according to virus yields, neglecting the expression profile of

transcomplementing gene products underlying cell performance. This work evaluates the

impact of E1 (E1A and E1B) and Cre recombinase levels in the production of E1-deleted

and helper-dependent canine adenovirus type 2 (CAV-2) vectors using MDCK cells. E1A

and E1B gene expression and Cre activity were evaluated in different cell clones and

compared with the corresponding cell productivity and susceptibility to oxidative stress

injury. CAV-2 production was proportional to E1A expression (the highest levels of E1A

corresponding to productivities of 3000-5000 I.P./cell), while E1B prolonged host cell

viability after infection, conferring protection against apoptosis. Cre recombinase

counteracted E1B anti-apoptotic properties, however viral production was maintained

under high levels of Cre. Yet, Cre recombinase side effects can be reduced using cell lines

with lower Cre-activities, without compromising the excision efficiency of helper vector

packaging signal. These results highlight the influence of transcomplementing gene

products on CAV-2 producer cell line performance, and the ability to express high levels

of E1A and E1B as an important feature for cell line establishment and high adenovirus

titers.

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

33

Contents

1. Introduction ............................................................................................................................................... 34

2. Materials and Methods .......................................................................................................................... 35

2.1. Plasmids .................................................................................................................................................... 35

2.2. Cell lines and culture media. ............................................................................................................ 36

2.3. Vectors ....................................................................................................................................................... 36

2.4. Establishment of MDCK-E1 and MDCK-E1-Cre clones. ........................................................ 37

2.5. E1 Gene expression analysis ............................................................................................................ 37

2.6. Cre activity ............................................................................................................................................... 38

2.7. Oxidative stress induction and cell viability assessment .................................................... 39

2.8. CAVGFP, JB∆5 and HD CAVGFP amplification by MDCK cells ........................................... 39

2.9. Infectious vectors titration ............................................................................................................... 40

2.10. Statistical analysis .............................................................................................................................. 40

3. Results ........................................................................................................................................................... 41

3.1. E1 gene expression and ΔE1 CAV-2 amplification ................................................................. 41

3.2. Cre expression in MDCK-E1 and HD CAV-2 amplification .................................................. 43

3.3. Physiological implications of E1 and Cre on ΔE1 and HD CAV-2 vectors producer

cell lines ............................................................................................................................................................. 46

4. Discussion.................................................................................................................................................... 48

5. Acknowledgments and author contribution ................................................................................ 51

6. References ................................................................................................................................................... 51

Chapter II

34

1. Introduction

Adenovirus vectors (AdV) are efficient gene transfer vectors due to the ability to

efficiently infect a wide variety of quiescent and proliferating cell types leading to high-

level gene expression (1). These vectors have been extensively studied for gene therapy

applications, where the AdV genome has been progressively modified from the wild-type

to improve its safety and efficacy in therapeutic applications. However, little attention has

been paid to the producer cell lines. So far, the main efforts during cell line development

are focused in avoiding the generation of replicative-competent adenovirus (RCA) by

sophisticated designs of the transforming plasmid; afterwards, cell lines that allow high-

titers of AdV are selected (reviewed in (2)). Thus, the expression profile of

transcomplementing genes behind cell lines performance has been neglected.

Most replication-defective adenovirus vectors require, for manufacturing and replication,

a cell line that expresses the adenoviral E1 functions in trans. E1 region codes for two

subunits – E1A and E1B – important to direct cellular and viral gene expression to enable

a productive virus cycle (3). E1A function is crucial for viral DNA replication; it

transactivates other early units and deregulates various cell cycle controls. E1A gene is

composed by two exons and several E1A polypeptides are produced following alternative

splicing of a primary RNA transcript. The most abundant E1A proteins are derived from

differentially spliced 12S and 13S mRNA that give rise to two proteins of variable size,

depending on the adenovirus serotype and species, and act as major regulators of early

viral transcription activating other early promoter regions (3, 4). Adenovirus E1A gene

expression products stimulate infected cells to enter S phase of the cell cycle and provide

an intracellular environment for viral replication. The E1A also contributes to the

transforming ability attributed to the virus, but requires the expression of E1B (5, 6). E1B

gene encodes two major proteins generally designated according to adenovirus

terminology by E1B 19 kDa and E1B 55 kDa, whose main functions are inhibition of

apoptosis, protection of viral and cellular DNA from degradation during infection and

further intracellular environment modification, to make the cell more hospitable to viral

protein production and viral DNA replication (7-10).

From vectors with the deletion of the E1 region to helper-dependent AdV with the

deletion of viral genes, an enhanced capacity for a gene therapeutic insertion from ~7 kb

to ~36 kb has been achieved (11). Helper-dependent vectors (HDV) harbors the inverted

terminal repeats (ITRs), where replication starts, the packaging signal (Ψ), and an

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

35

expression cassette. For that reason, their production requires trans-acting elements

provided by an helper vector (usually a ∆E1 vector) after co-infecting cells. To prevent

helper vector (HV) packaging, during HDV production, the Ψ sequence can be removed by

flanking the Ψ with recombinase recognition sequences that are recognized by the

recombinase stably expressed in the transcomplementing cell line. Therefore, the

transcomplementing cell line must also express a recombinase, such as Cre recombinase

(12-14).

In this work, we studied the impact of E1 expression in the development of MDCK-E1 and

MDCK-E1-Cre cell lines for E1-deleted and HD canine adenovirus type 2 (CAV-2) vectors.

Although human AdV is generally the preferred prototype vector backbone, memory

immunity (humoral and cellular) may limit the efficiency in humans (15-21). One

alternative to circumvent this drawback, while keeping other AdV advantages, is the use

of non-human adenovirus. CAV-2 vectors, showing longer transgene expression due to

the absence of neutralizing CAV-2 antibodies in the serum of healthy human individuals

(20-23), preferentially transduce neurons and have a remarkable capacity of axonal

transport, making them promising tools for the treatment of neurodegenerative diseases

(22, 24, 25). To investigate the production of CAV-2 vectors under different levels of E1

(E1A and E1B) different cell-clones were analyzed. This work confirms similar function of

adenovirus E1 proteins, predicted by conserved sequences (26). We show that viral

replication was influenced by the levels of E1A and E1B gene products. Higher levels of

Cre recombinase, although counteracting E1B anti-apoptotic properties, had no effect on

viral yields. Mechanisms underlying virus production and its relationship with E1A, E1B

and Cre are discussed in the context of cell line development.

2. Materials and Methods

2.1. Plasmids

For the transfection of MDCK with E1 genes, pCI-neo plasmid backbone containing CAV-2

E1A gene under the CMV promoter control and CAV-2 E1B gene under the control of its

own promoter was used and named pCI-NeoK9 (16, 20). E1 region corresponds to the

sequence 354- 3609 bp available in GenBank accession no. J04368 (20). Additionally, pCI-

NeoK9 confers neomycin resistance allowing the selection of cells after transfection. Cre

Chapter II

36

recombinase expression was obtained with pZeoCre plasmid (23) conferring Zeocin

resistance and in which Cre was cloned under the control of SV40 promoter.

2.2. Cell lines and culture media.

MDCK cells (ECCAC, Nr 84121903), MDCK-E1 clones and MDCK-E1-Cre subclones were

grown in Dulbecco’s Modified Eagle’s Medium (DMEM) (Gibco, Paisley, UK)

supplemented with 10% (v/v) FBS (Gibco). Dog Kidney cells expressing E1 from CAV-2

(DKZeo) and expressing E1 and Cre (DKCre) (20, 23) were used as control cells and

maintained in serum-supplemented DMEM (Gibco). All cells were maintained at 37⁰ C in

a humidified atmosphere air containing 5% CO2.

Cell concentration and viability was determined by the trypan blue exclusion method

using a 0.1% (v/v) solution prepared in PBS and counting cells in a Fuchs-Rosenthal

haemacytometer (Brand, Wertheim, Germany).

2.3. Vectors

CAVGFP, JB∆5 and HD CAVGFP are vectors derived from CAV-2 strain Toronto A 26/61,

GenBank U77082. CAVGFP (20) and HD CAVGFP (25) are E1-deleted (ΔE1) and HD

vectors, respectively, and contain an eGFP expression cassette. JB∆5 is a helper vector

containing loxP flanking the packaging domains and a RSV-lacZ expression cassette (25).

For CAVGFP and JB∆5 viral stocks preparation, 150 cm2 T-flasks with DKZeo cells at a

confluency of 80 - 90% were infected with a MOI of 5 infectious particles (I.P.) per cell

with medium exchange at the time of infection. 40 hours post infection (hpi) cells were

collected and lysed with 0.1% (v/v) Triton 100 (Sigma-Aldrich, Steinhein, Germany) in

Tris-HCl 10 mM, pH 8. The lysate was clarified by centrifugation at 3000 g during 10 min

at 4ºC and purified by CsCl gradients as described previously (20). The purified vectors

were stored in phosphate buffered saline (PBS) with 10% (v/v) glycerol in aliquots at -

85ºC. HD CAVGFP viral stock used in this work was prepared from a purified HD CAVGFP

obtained as described previously (25). Then, two amplification rounds were performed

by infecting DKCre cells, using an MOI ratio between HD CAVGFP and JB∆5 of 5 to 1. Viral

vectors from the second amplification round were purified as mentioned above.

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

37

2.4. Establishment of MDCK-E1 and MDCK-E1-Cre clones.

MDCK cells were transfected in suspension using 10 µg of polyethylenimine (PEI)

(Polysciences, Eppelheim, Germany) per 1×106 cells, with the plasmid pCI-NeoK9 (4µg).

Selection was performed in medium containing 500 µg/mL of Geneticin (G418)

(Invivogen, Toulouse, France). This cell population was cloned by limiting dilution with

50% (v/v) conditioned medium, 20% FBS (v/v) and 250 µg/mL of G418 (Invivogen). 121

clones were isolated. MDCK-E1-Cre subclones were generated using a similar protocol;

after transfecting MDCK-E1#106 cell clone with pZeoCre, the cells were selected using

500 µg/mL of Zeocin (Invivogen) followed by cloning by limiting dilution.

2.5. E1 Gene expression analysis

RNA samples were extracted from 1 × 106 cells of MDCK, DKZeo and MDCK-E1 clones

using RNeasy mini kit (QIAGEN, Germantown, MD, USA). cDNA synthesis from extracted

RNA was performed using “Transcriptor first strand cDNA synthesis kit” (Roche

Diagnostics, Mannheim, Germany). cDNA synthesis mix was prepared to the

manufacturer’s indicated end-concentrations. Amplification of cDNA was performed

using Fast Start PCR Master kit (Roche Diagnostics). Detection of E1A and E1B (for E1B

19 kDa and E1B 55 kDa) genes was performed using the following primers: for E1A gene

sense primer sequence 5’-CCGCGCAATCTCCATGATTA-3’ and antisense primer sequence

5’-AGTGCTCGCACTCGAATCAG-3’; for E1B 19 kDa sense primer sequence 5’-

TACTTTGTCGCCTGGATTTT-3’ and antisense primer sequence 5’-

CTGTCTACCTCTATTTTCCAGC-3’ and for E1B 55 kDa sense primer sequence 5’-

CACACTTTAGAAATGCCCAG-3’ and antisense primer sequence 5’-

CCGTAACCCTAATCTTAGAA-3’. The primers used for E1A gene were designed to consider

both mRNA transcripts originating E1A 12S and E1A 13S RNAs, since they amplify in

exon 1 containing CR1 and CR2 regions present in both transcripts (Figure 1). E1B codes

for two unrelated proteins, the 19 kDa and 55 kDa proteins, with partially overlapping

reading frames. Thus two different pairs of primers against non-overlapping regions

accounting for the two different mRNA transcripts were designed (Figure 1). GAPDH

(Glyceraldehyde 3-phosphate dehydrogenase) was selected as the endogenous control

gene and was amplified using sense primer sequence 5’-AACATCATCCCTGCTTCCAC-3’

and antisense primer sequence 5’-GACCACCTGTTCCTCAGTGT-3’.

Chapter II

38

For LightCycler real time PCR a master mix with the following reaction components was

prepared to the indicated end-concentrations: 2 µL LightCycler master (Fast start DNA

master SYBR Green I; Roche Diagnostics), 2.4 µL MgCl2 (3 mM), 2 µL forward and reverse

primer (0.3 µM), and 9.6 µL PCR grade water. LightCycler master mix was distributed into

the LightCycler capillaries (18 µL each) and either 2 µL of pCI-neoE1K9, pGAPDH

(standard curves for E1 and GAPDH gene) or cDNA was added. The following LightCycler

run protocol was used: denaturation program (95ºC, 10 minutes); amplification and

quantification program repeated 40 times (95ºC for 10 seconds; 60ºC (E1A gene) and

55ºC (E1B 19 kDa, E1B 55 kDa) for 10 seconds; 72ºC for 9 seconds); melting curve

program (95ºC 0 seconds; 65ºC for 15 seconds; 95ºC 0 seconds); and finally a cooling

step to 40ºC. The results obtained were analyzed with LightCycler software 4.1 (Roche

Diagnostics).

Figure 1 – Schematic representation of the DNA E1 region used to express in MDCK-E1 transcomplementing cells and the predicted major mRNA and protein derived from E1A and E1B genes. Predicted mRNAs and proteins were inferred by sequence homology (26) with human adenovirus. E1A 32 kDa and 28 kDa are translated from 13S and 12S mRNAs from E1A respectively. E1B 55 kDa is translated from 22S mRNA and E1B 19 kDa protein can be translated from the 22S or 13S mRNAs from E1B. Grey dashes underlying the mRNA products represent the generated PCR amplicon in the expression analysis of E1A and E1B genes. One set of primers was designed to assess E1A expression since it is expected that most generated splicing transcripts will contain the initial 5’ region of exon 1. Two sets of primers were design for E1B to confirm the formation of mRNA transcripts containing the 55 KDa open reading frames.

2.6. Cre activity

Relative Cre activity was assessed by the luciferase activity generated by the adenovirus

vector AdMA19 after infecting cells (27). AdMA19 contains the luciferase cDNA under

CMV exon 1 exon 2intron

E1A

19 kDa

55 kDa

E1B

13S 22S

13S

pIX

32 kDa

28 kDa

55 kDa

19 kDa

Predicted proteins:

DNA:

Predicted mRNA:

12S

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

39

control of the human cytomegalovirus promoter but separated from it by an extraneous

spacer sequence composed by a series of initiation and stop codons in several reading

frames. This translational “start/stop sign”, flanked by loxP, disrupts translation. If the

cell clone expresses Cre, the stop sign is floxed out and translation proceeds leading to

luciferase expression. Briefly, cells were seeded in 24 well-plates in quadruplicates and

infected the day after with 20 I.P./cell of AdM19 with medium exchange. Twenty-four hpi,

cells were counted and lysed to release luciferase, both in duplicates. The resulting

supernatant was collected and the light units were quantified with a Modulus

Luminometer from Turner Biosystems (Sunnyvale, CA, USA) after adding luciferin and

normalized to the cell concentration.

2.7. Oxidative stress induction and cell viability assessment

The susceptibility to oxidative stress injury was assessed by analyzing tert-butyl

hydroperoxide induced death rates. Briefly, cells were inoculated at 0.8 x 104 cells/cm2 in

24 well plates. At the 2nd day of culture, when cells reached approximately 90% of

confluence, tert-butyl hydroperoxide (Sigma) was added in increasing concentrations

ranging from 1.3 to 608.4 μM. Cell viability was quantified 18 h later by negative

propidium iodide (Sigma) staining and IC50 values calculated by non-linear fitting with a

95% confidence interval using Graphpad Prism (Graphpad Software, Inc., La Jolla, CA,

USA) and same approach as the one previously described (28).

2.8. CAVGFP, JB∆5 and HD CAVGFP amplification by MDCK cells

Cell clones were screened for CAVGFP vector amplification in tissue-culture Petri dishes

with 10 cm diameter (BD Biosciences, USA). MDCK-E1 clones were seeded at

1.5×104cells/cm2, infected the day after with a MOI of 5 I.P. and incubated for further 40

hours. CAVGFP vectors were collected using triton (Sigma-Aldrich) 0.1% (v/v) in Tris-

HCl, clarified at 3000 g for 10 min at 4ºC and stored at -85ºC until further analysis. To

evaluate the production decrease of helper JB∆5 under the presence of Cre, MDCK-E1-Cre

subclones, DKCre cells and corresponding parental cells were infected with 5 I.P./cell of

JB∆5 using the same protocol described above. The production assays for HD CAVGFP

was performed accordingly, by infecting cells with 5 I.P./cell of HD CAVGFP and 1 I.P./cell

of JB∆5.

Chapter II

40

2.9. Infectious vectors titration

Quantification of infectious CAVGFP and HD CAVGFP was performed by monitoring the

expression of GFP. MDCK cells were seeded in 24-well plates at 2 x 104 cells/cm2 and

infected the day after using serial dilutions of viral suspensions in fresh DMEM (Gibco).

Infected cells were harvested 24 hpi and the percentage of GFP-positive cells determined

by flow cytometry (CyFlow Space, Partec, Germany). Two replicates were performed for

each dilution. CAVGFP titer was determined by multiplying the number of cells at

infection with the percentage of GFP expressing cells and the respective dilution factor.

Titration of infectious JB∆5 vectors was based in lacZ gene expression and β-

galactosidase activity by adapting a previously described protocol (29). Briefly, DKZeo

cells were seeded at 1 x 105 cells/cm2 in 96-well plates. After 24 h, cells were infected

with serial dilutions of viral suspension. The day after, infected cells were fixed and

stained using a solution with X-gal (5-bromo-4-chloro-3-indolyl-b-D-galactopyranoside)

(Stratagene, La Jolla, CA, USA). Four replicates were performed for each dilution. JB∆5

titer was determined by counting the stained blue cells using an inverted phase contrast

microscope, multiplied by the dilution factor.

Productivities were shown as amplification ratio, corresponding to the ratio between I.P.

harvested and those used at infection (I.P. Out/I.P. In), or as cell specific viral titers,

corresponding to total I.P. normalized to cell concentration at virus harvesting time.

2.10. Statistical analysis

Statistics were performed using Microsoft Office Excel and Graphpad Prism software.

Data are presented as mean ± standard-deviation. Statistical significance was determined

by single factor Anova analysis with P value set at < 0.05.

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

41

3. Results

3.1. E1 gene expression and ΔE1 CAV-2 amplification

MDCK cells were stably transfected with the plasmid encoding E1 genes from CAV-2

(Figure 1) and clones with different expression levels were obtained. To evaluate the

ability of the clones to produce ΔE1 CAV-2 vectors, a production assay using CAVGFP was

performed. Nine of the cell clones efficiently propagated CAVGFP with productivities

above 300 I.P./cell.

To assess the effect of E1 expression on virus replication, five MDCK-E1 clones showing

different levels of virus productivities were analyzed (Table 1): MDCK-E1#106 and

MDCK-E1#121 had high amplification yields of 469 and 497 (I.P. Out/I.P. In) respectively,

MDCK-E1#139 had an intermediate amplification yield of 349, and finally MDCK-E1#21

and MDCK-E1#131 had amplification yields below 100. Although MDCK-E1#121

presented higher amplification yields than clones #106 and #139, attained lower cell

specific virus productivity (I.P./Cell) (Figure 2a). This can be justified by the significant

higher cell density at the virus harvesting time of MDCK-E1#121 (Figure 2d).

E1 region expression was investigated by determining the mRNA levels of E1A and E1B

transcripts. No relation between E1A and E1B expression was observed. Lower

expression of E1B versus E1A was obtained because the former is driven by its own

weaker promoter. The highest E1A gene expression was obtained for clone MDCK-

E1#106, followed by clone #139 and #121 (Figure 2b). MDCK-E1#21 and #31 had the

lowest levels of E1A expression. Concerning E1B expression, MDCK-E1#121 had levels on

the average 3- and 7-fold more than the other clones for E1B 19 kDa and E1B 55 kDa,

respectively (Figure 2c).

Table 1. Virus amplification in MDCK-E1 clones and DKZeo cells.

Cells Amplification ratio

MDCK-E1#21 94 ± 23

MDCK-E1#106 469 ± 117

MDCK-E1#121 497 ± 124

MDCK-E1#131 92 ± 23

MDCK-E1#139 349 ± 87

DKZeo 984 ± 246

The error corresponds to 25% inter-assays variability.

Chapter II

42

A correlation between E1A mRNA expression (Figure 2b) and cell specific viral

production (Figure 2a) can be observed, suggesting that higher E1A expression favors

vector production. On the other hand MDCK-E1#121 presented a 2-fold increase in viable

cells concentration at virus harvesting time (Figure 2d), which may indicate cell survival

advantages. E1B contribution for vector production may thus be indirect by preventing

loss of cell viability.

Figure 2 – Cell specific productivity and corresponding E1 gene expression of MDCK-E1 cell clones. a. Cell specific infectious viral titer produced in MDCK-E1 clones using DMEM 10% (v/v) FBS and 1% (v/v) NEAA. Error bars represent a 25% inter-assay variability error. b and c. Levels of mRNA E1A (b) and E1B expression (c) obtained for the different MDCK-E1 clones. The gene expression was determined by Real Time reverse transcriptase PCR and normalized to the housekeeping gene (GAPDH). Error bars represent a 10% variability error associated with the method. d. Cell concentration obtained for the different MDCK-E1 clones at virus harvest time. Cells were infected at the same cell concentration. Error bars represent the standard deviation of three independent experiments (n=3).

The long-term stability of E1 expression in MDCK-E1 clones was also assessed by

analyzing cells without antibiotic selection pressure with increasing subcultures

passages: low (passage 20), intermediate (passages 30 - 40) and high (passages 45 - 55)

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

43

passages. Similar CAVGFP amplifications were obtained in the different cell passages of

most MDCK-E1 clones tested. However, for MDCK-E1#139 clone, a decrease in the viral

production was observed at high passages (Figure 3). Also, a 2-fold reduction in specific

productivity of our reference cell line DKZeo (20) at high cell passage was observed. The

highest cell culture passages tested for MDCK-E1#106 and DKZeo were 52 and 56,

respectively.

E1 gene expression was determined in the three different ranges of passages for MDCK-

E1#106 and MDCK-E1#139 (Figure 4). No significant variation in E1A expression with

the increasing cell passages of MDCK-E1#106 was observed (Figure 4a). However, a

gradual increase in E1B expression with increasing cell passages was observed for the

same cell clone (Figure 4a). A clear reduction on E1A expression during subculture was

observed for MDCK-E1#139, particularly at high passage, possibly explaining the drop in

viral production for this clone (Figure 4b). The expression of E1B was maintained in

MDCK-E1#139. A small decrease was observed in E1A expression of DKZeo cells at high

passage (data not shown).

Figure 4 – Levels of mRNA of E1A and E1B over increasing cell culture passages for MDCK-E1 clone #106 (a) and #139 (b). The gene expression was determined by Real Time reverse transcriptase PCR normalized to the housekeeping gene (GAPDH). Error bars represent a 10% variability error associated with the method.

3.2. Cre expression in MDCK-E1 and HD CAV-2 amplification

To amplify helper-dependent (HD) CAV-2 vectors, and avoid helper-vector (HV)

contamination, MDCK-E1 cells have to express Cre-recombinase. MDCK-E1#106, being

Chapter II

44

the clone with the highest CAV-2 productivity (Figures 2a and 3), was stably transfected

with Cre recombinase gene and several clones were obtained. Cre activity was

determined indirectly by infecting the subclones with AdMA19 (27), which when cleaved

in the loxP site expresses luciferase (see materials and methods section), and compared

with DKCre cells (Figure 5). In comparison to DKCre cells, 25% presented either equal or

higher Cre activity. The subclones with higher Cre activity presented lower cell growth

(data not shown).

Figure 5 – Cell specific Cre activity of MDCK-E1#106-Cre subclones and DKCre cells measured indirectly through a luciferase assay (see materials and methods). Error assumes 20% inter-assay variability.

To determine if high productivities are maintained after Cre expression, CAVGFP

production was evaluated in five subclones with different Cre activities: Cre#2, Cre#10,

Cre#19, Cre#16 and Cre#30. The results showed same range of productivities for all the

subclones tested (Table 2).

In the current HDV production protocol it is fundamental to avoid HV contamination (13,

14), via Cre excision of the packaging signal of HV genome. However, a relative level of

Cre activity is required to attain the effective excision levels. Thus, considering that

subclones and the corresponding parental cells present same capability to produce viral

vectors, we evaluated the excision efficiency of the packaging signal of JB∆5 (CAV-2 HV),

flanked by loxP, by analyzing its amplification in MDCK-E1-Cre subclones (Table 2).

MDCK-E1#106-Cre subclones presented a reduced amplification capacity of ~26-fold,

which is in the range of that obtained with DKCre cells. MDCK-E1#106-Cre#10 was the

exception, showing the highest fold-decrease among all MDCK-E1#106-Cre subclones. No

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

45

evident relation was observed between the level of Cre activity and the amplification

reduction occurring in cell subclones. Furthermore, all MDCK-E1-Cre subclones

presented an estimated excision efficiency ≥ 96% (Table 2).

In order to establish the best conditions to produce HD CAV-2 vectors (maximum

productivity and lowest HV contamination) before testing MDCK-E1-Cre subclones,

production assays under different MOI ratios between HD CAVGFP and JBΔ5 (5 to 5, 5 to

1 and 5 to 0.5 I.P.) were performed using control DKCre cells. While under high MOI of

JB∆5 the specific productivity of HD CAVGFP was decreased to 3-fold with 60% of JB∆5

I.P. in the final product, maximum production of HD CAVGFP and lowest contamination of

JB∆5 (ranging from 2-4%) were both obtained using the ratios 5 to 1 and 5 to 0.5.

To test the amplification of HD CAV-2 vectors in MDCK-E1-Cre cells, three subclones were

co-infected with HD CAVGFP and JB∆5 and evaluated regarding the production of HD

CAVGFP and contamination level of JB∆5. The selected MOI ratio was 5 to 1 to ensure at

least one I.P. of JB∆5 per cell. The results showed same productivity between the MDCK-

E1-Cre subclones and DKCre cells, as well as a similar low contamination of HV (Table 3).

Table 2. Amplification of CAVGFP and JB∆5 in Cre-expressing clones and the corresponding parental cells.

Cells

Viral vectors

CAVGFP JB∆5

Amplification ratio

Productivity ratio

Amplification ratio

Fold decrease

Excision efficiency

(%) MDCK-E1#106 688 ± 172 - 605 ± 151 - -

MDCK-E1#106-Cre#2 537 ± 134 0.8 22 ± 6 28 96 MDCK-E1#106-Cre#10 928 ± 232 1.3 3 ± 1 201 99 MDCK-E1#106-Cre#19 590 ± 148 0.9 22 ± 6 28 96 MDCK-E1#106-Cre#26 798 ± 200 1.2 22 ± 6 28 96 MDCK-E1#106-Cre#30 882 ± 221 1.3 25 ± 6 24 96

DKZeo 475 ± 106 - 299 ± 75 - - DKCre 604 ± 151 1.3 10 ± 3 30 97

The error in amplification ratio corresponds to 25% inter-assays variability. The productivity ratio corresponds to the ratio between CAVGFP amplification using Cre expressing and corresponding parental cells. Fold decrease in JB∆5 production was calculated using the ratio between amplification value of parental cells (MDCK-E1#106 and DKZeo) and the corresponding Cre-expressing cells. Excision efficiency was calculated assuming that the difference in the amplification ratio of parental and corresponding Cre-expressing cells corresponds to unpackaged viral genomes.

Chapter II

46

Table 3. Cell specific productivity of HD CAVGFP and corresponding contamination with helper JB∆5.

Cells HD CAVGFP JB∆5

I.P./cell Amplification

ratio I.P./cell

Amplification ratio

Contamination (%)

MDCK-E1#106-Cre#10

454 ± 114 91 ± 23 6 ± 2 6 ± 2 1.3

MDCK-E1#106-Cre#26

469 ± 117 94 ± 24 8 ± 2 8 ± 2 1.6

MDCK-E1#106-Cre#30

281 ± 70 61 ± 15 5 ± 1 5 ± 1 1.9

DKCre 531 ± 133 106 ± 27 17 ± 4 17 ± 4 2.5

Errors correspond to 25 % inter-assay variability. The contamination levels of JB∆5 correspond to the ratio between the I.P titers of JB∆5 and HD CAVGFP.

3.3. Physiological implications of E1 and Cre on ΔE1 and HD CAV-2 vectors

producer cell lines

Adenoviral E1B gene products are involved in the inhibition of apoptosis (8-10), thereby

prolonging cell viability. On the other hand, Cre activity can induce growth inhibition and

apoptosis (30). To investigate the cellular response to the presence of E1 and Cre, death

susceptibility under oxidative stress conditions was analyzed. MDCK-E1#106 and MDCK-

E1#121 are shown as representatives of low and high E1B expression (Figure 2c), while

MDCK-E1#106-Cre#10 and MDCK-E1#106-Cre#19 as representatives of high and low

Cre activity (Figure 5), respectively.

Higher half maximal inhibitory concentration values (IC50) were obtained for MDCK-

E1#121 clone (Figure 6). On the other hand, MDCK-E1#106-Cre#10, being the subclone

with highest Cre activity, presented the highest susceptibility to oxidative stress. IC50

values of MDCK-E1#106-Cre#19, although higher than its counterpart (Cre#10) with

higher Cre-activity, were not significantly different from either MDCK or MDCK-E1#106

cell clone. High levels of Cre in MDCK-E1#106 subclones resulted in an higher cell

susceptibility to cell death that upon infection was translated in a decrease in cell density

by approximately 10% after 48 hpi (data not shown). However, no significant impact in

virus production was observed, as shown by the CAVGFP amplification yields that were

maintained in MDCK-E1#106-Cre subclones (Table 2).

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

47

Figure 6 – Half maximal inhibitory concentrations (IC50) of MDCK-E1 and MDCK-E1-Cre cell clones in response to increasing concentrations of tert-butyl hydroperoxide (t-BHP) (causing oxidative stress injury) in the culture medium. MDCK-E1#121 and MDCK-E1#106 represent high and low E1B expression, while MDCK-E1#106-Cre#10 and MDCK-E1#106-Cre#19 represent high and low Cre-activity, respectively. Error bars correspond to standard-deviation of quadruplicate assays. * p < 4x10-5, ** p < 0.05, *** p < 0.04, indicating the significance of a single factor Anova analysis

Chapter II

48

4. Discussion

Most replication-defective AdV require E1 transcomplementing cell lines that provide

E1A and E1B gene products essential for viral replication. The establishment of these

producer cell lines is mainly focused in developing cells that allow high-yield production,

while avoiding RCA generation (2). To our knowledge, the impact of E1A and E1B

expression levels on the production of AdV has never been investigated. To address this

issue, and given the attractive features of these vectors to fundamental neurobiological

questions and to develop potential treatment of neurodegenerative disorders (24, 25, 31,

32), we selected CAV-2 vectors production in MDCK-derived cell lines. Moreover, MDCK

cells are already approved by the regulatory authorities for the manufacture of vaccines

and thus representing a suitable cell substrate that might facilitate the regulatory

approval for the production of clinical grade CAV-2 vectors (33). Thus, the MDCK-derived

cell lines developed herein, holding production yields similar to the ones obtained with

the previously established DK cell lines, constitute a step forward for achieving that

purpose.

This work confirms the similar functions of E1A and E1B gene products from canine and

human adenoviruses, as extrapolated by sequence analogy. E1A encoded proteins from

adenovirus play an essential role in viral production by acting as transcriptional

activators of early viral genes (4), inducing cells to enter S-phase (34, 35) and allowing

the use of cellular DNA replication machinery to support viral DNA replication (3, 36).

Therefore, it is not surprising that high production yields were attained under high levels

of E1A transcripts (Figures 2a,b). When plotting E1A transcripts as a function of viral

production from the different MDCK-E1 clones, a linear tendency was obtained (r2= 0.99).

MDCK-E1#121 was an exception: the higher expression of E1B seemed to compensate

the intermediate level of E1A expression. MDCK-E1#121, showing the highest expression

of E1B among all clones tested, also presented less susceptibility to oxidative stress injury

(Figure 6) and higher cell concentration at virus harvesting time (Figure 2d). This

indicates the involvement of E1B in cell ability to hold stressful conditions such as

CAVGFP infection, which is supported by the functions dedicated to E1B gene products.

E1B 19 kDa and E1B 55 kDa proteins from human adenovirus can independently inhibit

the activation of apoptosis as a response to infection and foreign DNA replication (8, 37,

38). E1B 55 kDa also inhibits cellular mRNA export and promotes viral mRNA export and

translation during late infection (39). Therefore, the high levels E1B expression had also

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

49

an effect on viral production by extending cell viability. More interestingly, MDCK-

E1#106 cell clone presented a concomitant increase on E1B expression with cell passages

(Figures 3 and 4a). This gradual increase of expression can be due to an epigenetic

mechanism started in a few cells, which conferred proliferating/anti-apoptotic selection

advantage. Considering the main principle to choose cell lines (high production of AdV)

and the direct relation between viral yields and E1A expression levels, the cell lines

usually selected are likely to attain high levels of E1A. On the other hand, it is not

guaranteed that these same cells hold appreciable E1B expression levels, due to its

indirect role on viral yields. This has not a major impact on the production of E1-deleted

AdV, however high expression of E1B should be pursued in scenarios where cell line has

to further express toxic products, such as Cre recombinase during helper-dependent

vectors (HDV) production.

Cre expression can result in a markedly reduced proliferation and genotoxic effects in

cultured cells (30). The aberrant activity on multiple pseudo loxP sites presented in

mammalian genomes (40) induces DNA double-strand breaks (DSBs) or nicks that are

converted to DSBs during DNA replication, leading to arrest of cells in G2/M phase

because of intolerable DNA damage (30). Indeed, MDCK-E1-Cre subclones with very high

Cre activity presented reduced growth rates and we were unable to maintain some of

them at long term in culture (data not shown). In addition to disturbed growth patterns,

the subclones with high Cre activity become more susceptible to cell death (Figure 6),

showing an average of 10% lower cell densities at virus harvesting time, in comparison

with MDCK-E1 parental cells. The anti-apoptotic improvement conferred by E1B was

decreased, and in this context, the impact of Cre should be further investigated under

higher levels of E1B. Despite these observations, the MDCK-E1-Cre subclones used in this

work attained reproducible growth rates and maximum cell concentrations similar to

MDCK-E1 parental cells (data not shown). More important, similar CAV-2 vector

production was observed between MDCK-E1-Cre and corresponding MDCK-E1 cells

(Table 2). Although we hypothesized that the inhibitory effects of Cre could be extended

to vector production, because adenoviruses inhibit key components of cellular DNA

damage response (41, 42), compromising the repair of the Cre-induced DSBs, these

findings showed no effect of Cre on infected cells productivity, probably due to the

inhibition of DNA damage response taking place later in the lytic cycle (42).

Chapter II

50

The ideal situation to avoid cytotoxicity would be the minimum possible level of Cre that

would ensure an efficient excision of the Ψ in the helper vector (HV) genome. Herein, the

excision efficiency of the HV was assessed in subclones presenting different Cre activities

(Table 2). Overall, all MDCK-E1#106-Cre cell subclones presented similar excision

efficiency (above 96%), suggesting that above a minimal level of Cre, the excision of HV

packaging signal is equal. However, in a situation where HV production is maximized,

either by different viral construct or improved E1 expression of producer cell lines, more

viral genomes would be replicated and the threshold of Cre for the same excision

efficiency would probably increase. Nevertheless, under the conditions assayed herein,

MDCK-E1-Cre cells are suitable producer cell lines, with similar excision efficiencies to

the ones obtained for human HV (43). Moreover, MDCK-E1-Cre clones attained

appreciable HD CAV-2 titers; the HD CAVGFP amplification corresponded to a volumetric

productivity of 1.5 – 4.0 x 108 I.P./mL, which is similar to the one described for human

HDV (44, 45). The production of HD CAVGFP using 5 I.P./cell of this vector was

compromised when co-infection was performed with high MOI of JB∆5. In fact, the

negative effect of higher MOI of HV on HDV yield, together with the generation of higher

HV concentration, was already reported for human HDV by Dormond et al. (44). Thus,

lower MOI of JB∆5 was used to test MDCK-E1-Cre clones. Under these conditions, lower

amplification of HV was obtained, when comparing to what was observed in single

infections with high MOI (Tables 2 and 3). This may be due to the use of a non-optimal

MOI for HV replication or the presence of non-lethal mutations in HV packaging region

(24), further reducing DNA packaging in co-infection scenarios (23), such as the

production of HDV.

In this work, we show that the expression of E1 had an essential role in adenoviral vector

production, using CAV-2 as a case study. While E1A levels had a direct effect on viral

production, the expression of E1B conferred protection against cell death. The further

activity of Cre, although impairing cell growth and physiology, had no effect on viral

production and can be substantially reduced without compromising the excision of

packaging signal from HV. These findings highlight the need to understand the influence

of trans-complementing genes on the biology of host-cell and virus, providing important

knowledge to choose and design superior cell lines and enhance adenovirus yields.

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

51

5. Acknowledgments and author contribution

The authors are grateful to Núria Viana and Telma Lança for their support in the initial

MDCK cell line transfections and screening. The authors acknowledge the financial

support received from the Fundação para a Ciência e a Tecnologia (FCT) – Portugal

(PTDC/BIO/69452/2006 and PTDC/EBB-BIO/118615/2010) and European Commission

through FP7 project BrainCAV (222992). P. Fernandes, A.F. Rodrigues and H. Tomás also

acknowledge FCT for their Ph.D. grants (SFRH/BD/70810/2010, SFRH/BD/48393/2008

and SFRH/BD/79022/2011, respectively).

Paulo Fernandes participated in the experimental setup and design, performed part of the

experiments, analyzed the data and wrote the chapter.

6. References

1. Bramson, J. L., F. L. Graham, and J. Gauldie. 1995. The use of adenoviral vectors for gene therapy and gene transfer in vivo. Current opinion in biotechnology 6:590-595.

2. Kovesdi, I., and S. J. Hedley. 2010. Adenoviral producer cells. Viruses 2:1681-1703. 3. Berk, A. J. 1986. Functions of adenovirus E1A. Cancer surveys 5:367-387. 4. Liao, Y., D. Yu, and M. C. Hung. 2007. Novel approaches for chemosensitization of breast

cancer cells: the E1A story. Advances in experimental medicine and biology 608:144-169. 5. Bernards, R., M. G. de Leeuw, A. Houweling, and A. J. van der Eb. 1986. Role of the

adenovirus early region 1B tumor antigens in transformation and lytic infection. Virology 150:126-139.

6. Houweling, A., P. J. van den Elsen, and A. J. van der Eb. 1980. Partial transformation of primary rat cells by the leftmost 4.5% fragment of adenovirus 5 DNA. Virology 105:537-550.

7. Curriel, D. T., and J. T. Douglas (ed.). 2002. Adenoviral Vectors for Gene Therapy. Academic Press, California, U.S.A.

8. Rao, L., M. Debbas, P. Sabbatini, D. Hockenbery, S. Korsmeyer, and E. White. 1992. The adenovirus E1A proteins induce apoptosis, which is inhibited by the E1B 19-kDa and Bcl-2 proteins. Proceedings of the National Academy of Sciences of the United States of America 89:7742-7746.

9. White, E. 1998. Regulation of Apoptosis by Adenovirus E1A and E1B Oncogenes. Seminars in Virology 8:505-513.

10. White, E., P. Sabbatini, M. Debbas, W. S. Wold, D. I. Kusher, and L. R. Gooding. 1992. The 19-kilodalton adenovirus E1B transforming protein inhibits programmed cell death and prevents cytolysis by tumor necrosis factor alpha. Molecular and cellular biology 12:2570-2580.

11. Dormond, E., M. Perrier, and A. Kamen. 2009. From the first to the third generation adenoviral vector: what parameters are governing the production yield? Biotechnol Adv 27:133-144.

12. Lieber, A., C. Y. He, I. Kirillova, and M. A. Kay. 1996. Recombinant adenoviruses with large deletions generated by Cre-mediated excision exhibit different biological properties compared with first-generation vectors in vitro and in vivo. J Virol 70:8944-8960.

13. Hardy, S., M. Kitamura, T. Harris-Stansil, Y. Dai, and M. L. Phipps. 1997. Construction of adenovirus vectors through Cre-lox recombination. J Virol 71:1842-1849.

Chapter II

52

14. Parks, R. J., L. Chen, M. Anton, U. Sankar, M. A. Rudnicki, and F. L. Graham. 1996. A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci U S A 93:13565-13570.

15. Bangari, D. S., and S. K. Mittal. 2006. Current strategies and future directions for eluding adenoviral vector immunity. Current gene therapy 6:215-226.

16. Klonjkowski, B., P. Gilardi-Hebenstreit, J. Hadchouel, V. Randrianarison, S. Boutin, P. Yeh, M. Perricaudet, and E. J. Kremer. 1997. A recombinant E1-deleted canine adenoviral vector capable of transduction and expression of a transgene in human-derived cells and in vivo. Human gene therapy 8:2103-2115.

17. Paillard, F. 1997. Advantages of non-human adenoviruses versus human adenoviruses. Human gene therapy 8:2007-2009.

18. Thomas, C. E., D. Birkett, I. Anozie, M. G. Castro, and P. R. Lowenstein. 2001. Acute direct adenoviral vector cytotoxicity and chronic, but not acute, inflammatory responses correlate with decreased vector-mediated transgene expression in the brain. Mol Ther 3:36-46.

19. Yang, Y., Q. Su, and J. M. Wilson. 1996. Role of viral antigens in destructive cellular immune responses to adenovirus vector-transduced cells in mouse lungs. Journal of virology 70:7209-7212.

20. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

21. Perreau, M., and E. J. Kremer. 2005. Frequency, proliferation, and activation of human memory T cells induced by a nonhuman adenovirus. J Virol 79:14595-14605.

22. Keriel, A., C. Rene, C. Galer, J. Zabner, and E. J. Kremer. 2006. Canine adenovirus vectors for lung-directed gene transfer: efficacy, immune response, and duration of transgene expression using helper-dependent vectors. J Virol 80:1487-1496.

23. Soudais, C., S. Boutin, and E. J. Kremer. 2001. Characterization of cis-acting sequences involved in canine adenovirus packaging. Mol Ther 3:631-640.

24. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

25. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

26. Shibata, R., M. Shinagawa, Y. Iida, and T. Tsukiyama. 1989. Nucleotide sequence of E1 region of canine adenovirus type 2. Virology 172:460-467.

27. Anton, M., and F. L. Graham. 1995. Site-specific recombination mediated by an adenovirus vector expressing the Cre recombinase protein: a molecular switch for control of gene expression. J Virol 69:4600-4606.

28. Rodrigues, A. F., M. R. Guerreiro, V. M. Santiago, C. Dalba, D. Klatzmann, P. M. Alves, M. J. Carrondo, and A. S. Coroadinha. 2011. Down-regulation of CD81 tetraspanin in human cells producing retroviral-based particles: tailoring vector composition. Biotechnology and bioengineering 108:2623-2633.

29. Coroadinha, A. S., R. Schucht, L. Gama-Norton, D. Wirth, H. Hauser, and M. J. Carrondo. 2006. The use of recombinase mediated cassette exchange in retroviral vector producer cell lines: predictability and efficiency by transgene exchange. J Biotechnol 124:457-468.

30. Loonstra, A., M. Vooijs, H. B. Beverloo, B. A. Allak, E. van Drunen, R. Kanaar, A. Berns, and J. Jonkers. 2001. Growth inhibition and DNA damage induced by Cre recombinase in mammalian cells. Proc Natl Acad Sci U S A 98:9209-9214.

31. Salinas, S., L. G. Bilsland, D. Henaff, A. E. Weston, A. Keriel, G. Schiavo, and E. J. Kremer. 2009. CAR-associated vesicular transport of an adenovirus in motor neuron axons. PLoS pathogens 5:e1000442.

32. Henaff, D., and S. Salinas. 2010. An endocytic CARriage tale: Adenoviruses internalization and trafficking in neurons. Virulence 1:188-191.

Developing MDCK CAV-2-E1 and E1-Cre Transcomplementing Cell Lines

53

33. Fernandes, P., C. Peixoto, V. M. Santiago, E. J. Kremer, A. S. Coroadinha, and P. M. Alves. 2013. Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20:353-360.

34. Gallimore, P. H., and A. S. Turnell. 2001. Adenovirus E1A: remodelling the host cell, a life or death experience. Oncogene 20:7824-7835.

35. Moran, E. 1993. DNA tumor virus transforming proteins and the cell cycle. Current opinion in genetics & development 3:63-70.

36. Berk, A. J. 1986. Adenovirus promoters and E1A transactivation. Annual review of genetics 20:45-79.

37. Debbas, M., and E. White. 1993. Wild-type p53 mediates apoptosis by E1A, which is inhibited by E1B. Genes & development 7:546-554.

38. Schmitz, M. L., A. Indorf, F. P. Limbourg, H. Stadtler, E. B. Traenckner, and P. A. Baeuerle. 1996. The dual effect of adenovirus type 5 E1A 13S protein on NF-kappaB activation is antagonized by E1B 19K. Molecular and cellular biology 16:4052-4063.

39. Blackford, A. N., and R. J. Grand. 2009. Adenovirus E1B 55-kilodalton protein: multiple roles in viral infection and cell transformation. Journal of virology 83:4000-4012.

40. Thyagarajan, B., M. J. Guimaraes, A. C. Groth, and M. P. Calos. 2000. Mammalian genomes contain active recombinase recognition sites. Gene 244:47-54.

41. Araujo, F. D., T. H. Stracker, C. T. Carson, D. V. Lee, and M. D. Weitzman. 2005. Adenovirus type 5 E4orf3 protein targets the Mre11 complex to cytoplasmic aggresomes. J Virol 79:11382-11391.

42. Baker, A., K. J. Rohleder, L. A. Hanakahi, and G. Ketner. 2007. Adenovirus E4 34k and E1b 55k oncoproteins target host DNA ligase IV for proteasomal degradation. J Virol 81:7034-7040.

43. Ng, P., C. Evelegh, D. Cummings, and F. L. Graham. 2002. Cre levels limit packaging signal excision efficiency in the Cre/loxP helper-dependent adenoviral vector system. J Virol 76:4181-4189.

44. Dormond, E., M. Perrier, and A. Kamen. 2009. Identification of critical infection parameters to control helper-dependent adenoviral vector production. J Biotechnol 142:142-150.

45. Dormond, E., A. Meneses-Acosta, D. Jacob, Y. Durocher, R. Gilbert, M. Perrier, and A. Kamen. 2009. An efficient and scalable process for helper-dependent adenoviral vector production using polyethylenimine-adenofection. Biotechnol Bioeng 102:800-810.

Chapter II

54

Chapter III

Upstream bioprocess development for

E1-deleted CAV-2 vectors

This chapter is adapted from:

Fernandes P, Peixoto C, Santiago VM, Kremer EJ, Coroadinha AS, Alves PM (2013)

Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20(4): 353-60.

Fernandes P, Sousa M, Kremer EJ, Coroadinha AS, Alves PM. Suspension MDCK-E1 cell line

for the manufacturing of CAV-2 vectors. (under preparation)

Part A

Production with adherent cells using

microcarrier technology

This chapter is adapted from:

Fernandes P, Peixoto C, Santiago VM, Kremer EJ, Coroadinha AS, Alves PM (2013)

Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20(4): 353-60.

Chapter III – Part A

58

Abstract

Canine adenovirus type 2 (CAV-2) vectors overcome many of the clinical immunogenic

concerns related to vectors derived from human adenoviruses (AdVs). In addition, CAV-2

vectors preferentially transduce neurons and efficient traffic via axons to afferent regions

when injected in brain. To meet the need for preclinical and possibly clinical uses,

scalable and robust production processes are required. CAV-2 vectors are currently

produced in E1-transcomplementing DK (dog kidney) cells, which might raise obstacles

in regulatory approval for clinical grade material production. Here a GMP-compliant

bioprocess was developed. An MDCK-E1 cell line, developed by our group, was grown in

scalable stirred tank bioreactors, using serum-free medium, and used to produce CAV-2

vectors. Vectors produced in MDCK-E1 cells were identical as those produced in DK cells

as assessed by SDS-page and DLS measurements (diameter and Zeta potential).

Productivities of ~109 infectious particles (IP)/mL and 2 x 103 IP/cell were possible.

These results constitute a step towards a scalable process for CAV-2 vectors production

compliant with clinical material specifications.

Production with adherent cells using microcarrier technology

59

Contents

1. Introduction ............................................................................................................................................... 60

2. Materials and methods .......................................................................................................................... 61

2.1. Cell lines and Culture Media ............................................................................................................ 61

2.2. Cell Growth Assays ............................................................................................................................... 61

2.3. Cell concentration determination.................................................................................................. 62

2.4. ΔE1 CAV-2 vector stock preparation ........................................................................................... 62

2.5. CAVGFP production assays ............................................................................................................... 62

2.6. Titration of infectious CAVGFP particles .................................................................................... 63

2.7. Measurement of total CAVGFP particles by Real Time-PCR (RT-PCR) ......................... 63

2.8. Protein profile of CAVGFP by SDS-PAGE and western blot ................................................ 64

2.9. Size and Electrodynamics measurements of CAVGFP .......................................................... 64

2.10. Bioreactor culture and CAVGFP production .......................................................................... 64

3. Results ........................................................................................................................................................... 65

3.1. MDCK-E1 transcomplementing cells for the production of a ∆E1 CAV-2 vector ..... 65

3.2. Effect of culture medium on cell growth and vector production under static

conditions ......................................................................................................................................................... 66

3.3. Production optimization under stirring conditions .............................................................. 67

3.3.1. Effect of cell inoculum and microcarrier type on cell growth .................................. 67

3.3.2. Effect of time of harvest on virus yield .............................................................................. 68

3.3.3. Stirred tank bioreactor for ∆E1 CAV-2 production ...................................................... 68

4. Discussion.................................................................................................................................................... 69

5. Acknowledgements and author contribution ............................................................................. 71

6. References ................................................................................................................................................... 71

Chapter III – Part A

60

1. Introduction

Viral vectors are currently the most efficient tools for in vivo gene transfer. Due in part to

their in vivo efficiency, adenovirus vectors (AdV) are used more often than any other

vector in clinical trials (http://www.wiley.com/legacy/wileychi/genmed/clinical/).

Several characteristics make this diverse family of vectors attractive, namely wide cell

tropism in quiescent and non-quiescent cells, the poor ability to integrate the host

genome and the high production titers obtained in culture (1). However, gene transfer

efficacy and the clinical use of human AdV can be hampered by the pre-existing humoral

and cellular immunity in most humans (2, 3).

Canine adenovirus type 2 (CAdV-2, or more common referred to as CAV-2) is probably

the best characterized nonhuman adenovirus vector (2-5). The paucity of neutralizing

antibodies and memory T cells in humans and efficient gene delivery obtained for E1-

deleted (∆E1) CAV-2 vectors in the central nervous system, make these viruses promising

tools for human gene therapy (2, 5). The extraordinary ability to preferentially transduce

neurons combined with a remarkable capacity of axonal transport, make CAV-2 vectors

candidates for the treatment of neurodegenerative diseases (6).

The need for significant amounts of clinical grade adenovirus vectors, which in some

cases may reach 1013 total particles/patient or 1011 infectious particles/patient, requires

efficient and robust processes for production and purification at large-scale compliant

with good manufacturing practices (GMP) (1). Therefore, current lab-scale protocols

must be adapted and transferred to a scalable stirred culture system

Moreover, cells cultured with media containing animal serum should be avoided. The

undefined composition and high batch-to-batch variability of serum, together with its

potential source of contaminations, raises safety concerns and hinders the

standardization of cell culture processes for the production of biopharmaceuticals (7).

To date, CAV-2 vectors have been produced in a cell-line generated from dog kidney (DK)

(2). However these cells are not regulated for biopharmaceuticals production, which

precludes the potential clinical use of CAV-2 vectors. Recently, we developed a CAV-2 E1-

trascomplementing cell-line based in MDCK. MDCK cells are accepted in Food and Drug

Administration (FDA) and European Medicine Agency (EMA) and used for the production

of many viral vaccines. In fact, recently, a cell based flu vaccine produced in MDCK was

launched in the market by Novartis (Optaflu®). Thus, the use of an MDCK-based cell-line

would facilitate the regulatory approval for the production of CAV-2 vectors.

Production with adherent cells using microcarrier technology

61

Here, MDCK-E1 cells grown in microcarriers was used as host cells for viral production in

stirred tank bioreactors. The impact of upstream parameters, such as cell inoculum

concentration, microcarrier type on cell growth and time of harvesting on total viral

productivity were evaluated. Our results constitute a significant step in a scalable CAV-2

vector production

2. Materials and methods

2.1. Cell lines and Culture Media

DKZeo cells, generated from DK cell line as described by Kremer et al. (6), were

maintained in DMEM (Gibco, Paisley, UK) with 10% (v/v) fetal bovine serum (FBS)

(Gibco) and 1% (v/v) non-essential amino acids (Sigma-Aldrich, St. Louis, MO, USA).

MDCK-E1 cells were obtained by transfecting MDCK (ECCAC 841211903) with pCI-neo

plasmid (Promega, Madison, WI, USA) harboring E1 gene from CAV-2, using similar

approach to the one described for DKZeo cells.(2, 8). MDCK-E1 cells were cultivated in

MEM (Sigma) with 10% (v/v) FBS, 2 mM glutamine (Gibco) and 1% (v/v) non-essential

amino acids (Sigma). These cells were sequentially adapted to Optipro serum-free

medium (Gibco) with 4 mM glutamine (Gibco) by increasing the percentage of Optipro

medium in subculture passages. Cells were subcultured twice a week in 150 cm2 t-flasks

and maintained in an incubator with humidified atmosphere of 5% CO2 in air at 37ºC. For

splitting or collecting cells, the monolayer was washed with PBS, incubated with 0.25%

trypsin-EDTA (Gibco) at 37ºC until detachment started to become evident (up to 15 min)

and cells suspended in culture medium. Cells were seeded directly using culture media

containing FBS (MEM and DMEM), or pelleted at 300 g during 10 min and re-suspended

in fresh medium when serum-free medium was used.

2.2. Cell Growth Assays

MDCK-E1 growth assays in static cultures were performed using an inoculum of 1 x 104

cells/cm2 in 25 cm2 t-flasks with a working volume of 10 mL.

The stirred cultures were performed in 125 mL spinner flasks (Wheaton, Millville, NJ,

USA) previously siliconized with a solution of dimethyldichlorosilane in toluene (Merck,

Darmstadt, Germany) to prevent sticking of cells and microcarriers to the flasks walls.

Chapter III – Part A

62

Two non-porous microcarriers, Cytodex 3 and Cytodex 1 (GE Healthcare, Uppsala,

Sweden), were prepared according to the manufacturers’ instructions. The growth curves

were performed using an inoculum of 1 x 105 and 2 x 105 cells/mL, a microcarrier

concentration of 2 and 3 mg/mL and a stirring of 50 rpm.

2.3. Cell concentration determination

Cells were counted using a Fuchs–Rosenthal haemocytometer chamber with the trypan

blue exclusion method. In static cultures, cells were counted directly from cell suspension

after trypsin detachment. In stirred/microcarriers cultures, a similar procedure was

adapted. From 1 mL sample, microcarriers were let to settle-down and supernatant

removed (~800 μL). PBS was added in same volume to wash cells and removed after new

microcarriers settle-down. Equal volume of trypsin was added, cells were incubated at

37ºC until evident detachment, suspended by pipetting up and down and finally counted.

2.4. ΔE1 CAV-2 vector stock preparation

CAVGFP, an E1-deleted CAV-2 vector, was prepared as described previously (2). For

stock preparation, 150 cm2 t-flasks with DKZeo cells at a confluency of 80 - 90% were

infected with a MOI of 5 (ip) and medium exchange at time of infection. 40 hours post

infection (hpi) cells were collected and lysed with 0.1% (v/v) Triton 100x (Sigma-

Aldrich). The lysate was clarified by centrifugation at 3 000 g during 10 min at 4ºC and

purified by CsCl gradients as described previously (2). The purified vectors were stored

in phosphate buffered saline (PBS) with 10% (v/v) glycerol in aliquots at -85ºC. The

ip/physical particles (pp) ratio was 1:21 based on FACS analysis and Real-Time PCR.

2.5. CAVGFP production assays

The infection assays were performed in tissue-culture (TC) Petri dishes with 10 cm

diameter (BD Biosciences, Franklin Lakes, NJ, USA) and 125 ml spinner vessels

(Wheaton). A MOI of 5 was used and the medium was exchanged at the time of infection.

When using TC Petri dishes 8 x 104 cells/cm2 were seeded and infected 24h after with a

cell confluence of 80 - 90% and a working volume of 10 mL. In spinner flasks cultures 1 x

105 cells/mL and 2 x 105 cells/mL were inoculated using MEM and Optipro media,

respectively. Cells were infected at the end of the exponential growth phase (approx. 48

h). Extracellular viruses were collected from the cell supernatant. Intracellular viruses

Production with adherent cells using microcarrier technology

63

were collected by disrupting cells in a known volume of lysing buffer (Tris/HCl 10 mM,

pH 8.0 and 0.1% (v/v) Triton-X 100). The resulting intracellular and extracellular

samples were clarified at 3000 g for 10 min at 4ºC and stored at -85ºC.

2.6. Titration of infectious CAVGFP particles

Quantification of infectious particles was performed by monitoring the expression of GFP

reporter gene using slightly modifications to the method described by Ferreira et al. (9).

Briefly, 8 x 104 MDCK-E1 cells were seeded in 24-well plates and infected with serial

dilutions of viral suspensions. Cells were trypsin-detached 24 hpi and the percentage of

GFP-positive cells determined by flow cytometry (CyFlow Space, Partec, Münster,

Germany).

2.7. Measurement of total CAVGFP particles by Real Time-PCR (RT-PCR)

CAVGFP DNA was extracted and purified by High Pure Viral Nucleic Acid Kit (Roche

Diagnostics, Penzberg, Germany). Previously purified and quantified plasmid coding for

the E1-deleted CAV-2 vector genome (pJB19) (10) was used as standard.

Amplifications were performed by the Light Cycler system with ‘‘Fast Start Master SYBR

Green I kit’’ (Roche Diagnostics) based on the method developed previously (11). For a 20

µL PCR reaction, 10 µL DNA templates and 10 µL of mastermix were added to each

capillary. The mastermix content was 3 mM MgCl2 and 0.5 µM of each primer. The

forward primer used was 5’AAATATACAGGACAAAGAGGTGTGG3’ and the reverse

5’GAACTCGCCCTGTCGTAAAA3’. The standard program was performed according in Light

Cycler RT-PCR (Roche Diagnostics) and analyzed using the Light Cycler software. A

standard curve was obtained by linear regression analysis of the threshold cycle (Ct)

value (y axis) versus the log of the initial copy number present in each sample dilution (x

axis). PCR efficiency (E) was calculated as E= 10(-1/slope) – 1 and corresponded to 2.021.

The error associated was 0.0849. The PCR limit of detection was ~1 x 102

copies/reaction, detected using 40 cycles. The viral DNA loss during DNA

extraction/purification showed to be constant and was further considered to calculate

physical particles concentration (11).

Chapter III – Part A

64

2.8. Protein profile of CAVGFP by SDS-PAGE and western blot

Viral particles were precipitated in absolute ethanol at -20ºC overnight, centrifuged at

10.000 g for 10 min and concentrated in the SDS-PAGE loading-buffer at 30 μg of protein

per total volume to apply on gel. SDS-PAGE was performed according to NuPAGE®

Novex® Bis-Tris Mini Gels protocol using the Xcell SureLock™ equipment (Invitrogen).

Western blot analysis was carried out using a polyclonal rabbit anti-CAV-2 primary

antibody (12). Proteins were revealed via BCIP/NBT substrate (Pierce) after incubation

with an alkaline phosphatase conjugated anti-rabbit secondary antibody (Sigma-Aldrich).

2.9. Size and Electrodynamics measurements of CAVGFP

Size and zeta potential of viral particles were determined by dynamic light scatering

(DLS) using a Zetasizer Nano-ZS Series ZEN3600 equipped with a 633nm He-Ne laser

(Malvern, Worcestershire, UK). Zeta potential was measured using a 40 μl sample in 660

μl 10 mM phosphate buffer (Sigma-Aldrich) for pH ranging from 3 to 10 in 10x10 mm

plastic cuvette (Sarstedt, USA) using a dip-cell (Malvern). Particles size was determined

using the same sample and buffer volumes at pH 8.

2.10. Bioreactor culture and CAVGFP production

MDCK-E1 cells were cultivated in siliconized 2 L stirred tank bioreactors (Biostat Q-Plus,

Sartorius Stedim Biotech, Goettingen, Germany) equipped with 3-blade impellers, under

defined conditions (working volume: 1.5 L; pH: 7.4; temperature: 37ºC; surface aeration

at 40% pO2; agitation rate: 50 - 60 rpm). Cells (2 x 105 cells/mL) were seeded on 3

mg/mL of Cytodex 1 microcarriers placed inside glass bottles and transferred to

bioreactor vessels. Culture medium Optipro was supplemented with 5% (v/v) FBS during

inoculation (12 h). Then, a complete medium replacement was done by stopping agitation

and letting microcarriers settle-down. After 2 days cultivation (1 x 106 cells/mL), the

medium was exchanged to regular Optipro (without FBS) and cells were infected with

MOI 5.

Production with adherent cells using microcarrier technology

65

3. Results

The ability of MDCK-E1 cells to produce CAV-2 particles of correct size, charge and SDS

profile was evaluated by comparison with viral vector production in DKZeo cells. The

impact of serum-free (SF) Optipro medium on cellular growth and viral production was

also assessed by comparing the results obtained with serum-supplemented (SS) MEM.

Moreover, to develop a scalable process, the best inoculation strategy (cell concentration

inoculum and microcarrier type) for improved cell growth and the best time of harvest

for maximum virus productions yield in stirring conditions were evaluated and

confirmed in 2 L bioreactor.

3.1. MDCK-E1 transcomplementing cells for the production of a ∆E1 CAV-2 vector

To evaluate MDCK-E1 cell line potential for CAV-2 vectors production, specific

productivity/amplification of CAVGFP and its physical characteristics were investigated.

DKZeo cells were used as control because they are currently used for the production of

these vectors; vectors were produced in both cell lines using DMEM culture medium.

Similar cell specific productivities and infectious particles amplification were obtained for

both cell lines (Table 1). No significant difference in particles size, charge or protein SDS

profile was detected for viral particles produced with DKZeo and MDCK-E1 (Figure 1).

These results suggest that MDCK-E1 cells are suitable for CAV-2 vectors production.

Table 1. Effect of producer cell line on production and properties of viral particles

Criteria MDCK-E1 DKZeo MDCK-E1

Serum-free conditions

Cell specific productivity (ip/cell) 2 345 ±

5861

2 605 ± 6511

2 223 ± 5561

Amplification (ip out/ip in) 469 ± 1171 521 ± 1301 445 ± 1381

Total physical particles per ip in purified preparations

21 ± 61 17 ± 51 26 ± 81

Viral particles diameter (nm) 116 ± 172 115 ± 172 n.d.

1 Standard deviation of triplicate measurements; 2 Assumes 15% standard instrument error associated with the measurement. n.d.: non-determined.

Chapter III – Part A

66

Figure 1. A) SDS-PAGE of purified viral particles produced in DKZeo (lane 2) and MDCK-E1 (lane 3). Gel was loaded with 10 μl of concentrated sample per well. Lane 1: molecular weight markers. B) Zeta (ζ) potential of purified viral samples produced in DK E1 (○ circle) and MDCK E1 (♦ diamond).

3.2. Effect of culture medium on cell growth and vector production under static

conditions

The impact of SF medium (Optipro) in cell growth and viral productivity was evaluated

by comparison with SS medium (MEM).

Although the specific growth rate of MDCK-E1 cells adapted to SF medium in static

conditions was almost half of the value obtained when using SS medium, the maximum

cell concentration attained was 3.5-fold higher (1.9 x 106 cells/mL) (Table 2).

Cell-specific production of 2 223 infectious particles (ip)/cell and 391 ip/cell were

obtained for SF and SS medium, respectively, corresponding to amplification ratios of 445

and 78 infectious particles.

Table 2. Impact of culture medium and culture strategy on specific growth rate (µ) and maximum cell concentration (Xvmax) of MDCK-E1 cells.

Culture SS medium (MEM) SF medium (Optipro)

Static µ (h-1) 0.039 ± 0.0011 0.023 ± 0.0011 Xvmax

(106 cell/mL) 0.54 ± 0.052 1.9 ± 0.22

Stirred µ (h-1) 0.039 ± 0.0011 0.037 ± 0.0011

Xvmax (106 cell/mL)

0.53 ± 0.052 1.1 ± 0.12

1 Standard error associated with the exponential data regression; 2 Assumes 10% standard instrument error associated with the measurement.

Production with adherent cells using microcarrier technology

67

3.3. Production optimization under stirring conditions

3.3.1. Effect of cell inoculum and microcarrier type on cell growth

The effect of inoculum concentration in stirred cultures was evaluated in SS and SF

medium testing two concentrations: 1 x 105 cells/mL and 2 x 105 cells/mL. The ability of

Cytodex 1 and Cytodex 3 to support cell growth in serum-free medium was also

evaluated.

MDCK-E1 cells did not attach to microcarriers when SF medium was used. To overcome

this and promote cell adhesion, 5% (v/v) FBS was added. After 12 h this culture medium

was replaced by regular SF medium (without FBS).

Maximum cell concentration obtained for both culture media and cell inoculums tested is

shown in Figure 2. The highest cell concentration was obtained for SF medium at

approximately 50 h of culture using an inoculum of 2 x 105 cells/mL. No differences were

observed between the two microcarriers evaluated as assessed by total cell number and

viability. Moreover, a similar range of total cell concentration was obtained for static or

stirred cultures using both culture media (Table 2).

Figure 2. Maximum cell concentration obtained in spinner flasks using different cell inoculums in serum-supplemented (SS) and serum-free (SF) media. All experiences were performed with MDCK-E1 and Cytodex 1 except in ♦ (diamond) where Cytodex 3 was used. Medium exchange was performed after cell attachment (12 h) and considered to exclude any effect of serum on cell growth under stirring conditions. Error bars represent error associated with the measurement (~10%).

Chapter III – Part A

68

3.3.2. Effect of time of harvest on virus yield

After infecting cells, the maximum number of infectious particles was achieved between

24 and 36 hpi (Figure 3A), the majority being in the intracellular fraction. At this time,

infectious particles amplification ratio of 21 and 28, corresponding to 132 ip/cell and 141

ip/cell, were obtained for MDCK-E1 cells in SF medium and SS medium, respectively.

3.3.3. Stirred tank bioreactor for ∆E1 CAV-2 production

Taking into account the results obtained in spinner vessels (2.3.1 and 2.3.2) MDCK-E1

cells were cultivated in a 2 L stirred tank bioreactor using an inoculum of 2 x 105 cells/mL

and 3 mg/mL of Cytodex 1 microcarriers. Maximum cell concentration of 1.1 x 106

cells/mL was obtained being the maximum infectious particles productivity obtained at

24 – 36 hpi. However, in bioreactor an amplification ratio of 256 was attained

corresponding to a volumetric productivity of 1.8 x 109 ip/mL (Figure 3B).

Figure 3. A) Amplification ratio of infectious particles along time after infecting MDCK-E1 cells in spinner flasks with serum supplemented medium and serum-free medium. Between 24 and 36 hpi most of viable cells attained attached to microcarriers. Error bars represent standard deviation of triplicate measurements. B) Volumetric productivity (ip/mL) obtained with MDCK-E1 cultured in static cultures (T-Flasks) and bioreactor (in Cytodex 1) using serum supplemented medium and serum-free medium. Viral vectors were collected at 40 hpi and 36 hpi in static and bioreactor cultures, respectively. Error bars represent standard deviation of triplicate measurements.

Production with adherent cells using microcarrier technology

69

4. Discussion

The potential of CAV-2 vectors for fundamental and clinical gene transfer and the

amounts needed require a robust and scalable bioprocess. MDCK cells are accepted by the

FDA and EMEA as producer cells for clinical grade vaccines and recombinant protein. In

this study, an MDCK derived cell-line was used to produce ∆E1 CAV-2 vectors using

serum-free medium.

Productivities and the impact on viral particles were consistent with the robustness of

MDCK-E1 cells to produce ∆E1 CAV-2 vectors when compared with DKZeo cells.

The main differences in MDCK-E1 cells performance observed between SF Optipro and SS

MEM is likely due to the more complex composition of SF medium. Cells cultured in SF

medium, although showing slower growth rates, can achieve higher cell concentration

since more nutrients and/or nutrients concentration are available. Moreover, a similar

range of cell specific productivity was obtained when using cells cultivated in a richer

serum supplemented medium, namely DMEM.

Adaptation of cells to SF medium affected the cell specific grown as adherent monolayer

in static cultures. However, for stirring conditions similar growth rates were observed,

when compared with MDCK-E1 cells growing in SS medium. This observation suggested a

relation between agitation and promotion of cell growth rate when SF medium was used.

Serum provides many factors that promote cell adhesion. Although several reports in the

literature describe effective cell attachment to microcarriers, for different cell types

including MDCK (13), in commercially available serum free SF formulations, we were not

able to accomplish this for the MDCK-E1 cells used herein. Attachment was only

significant in the presence of serum, being possible to remove it afterwards without

compromising virus productivity. From a biopharmaceutical production process

perspective, the use of animal compounds, namely serum, should be avoided. However, as

the serum was only required for the cell attachment phase (inoculation) and was washed

out by two complete medium replacement steps, the content of serum during bioprocess

cell growth, virus infection and virus production phases was low and the risks associated

with its use significantly reduced.

In contrast to the reported cultivation of MDCK cells in microcarrier under stirring

conditions, we observed similar maximum cell concentration using Cytodex 1 or Cytodex

3 without significant cell detachment during growth (13, 14). In studies using MDCK cells

for influenza production, growth rates of about 0.02 to 0.035 h-1 and maximum cell

Chapter III – Part A

70

concentrations of 1.1 x 106 – 1.3 x 106 cells/mL obtained 4 and 5 - 6 days after inoculation

using SS and SF medium, respectively, were reported (13, 15, 16). In our hands, the same

range of maximum cell concentration was obtained. However, the higher specific growth

and shorter lag phase may have been responsible for a maximum cell concentration

attained only 2 days after inoculating same cell number. Host cell-line effect was

excluded, because a similar performance was observed between MDCK-E1 cells and

MDCK progenitor cell line under same conditions (data not shown). These discrepancies

may be due to the different culture media used in each study. Moreover, these variations

in specific growth rate and maximum cell concentrations should be viewed in the context

of cell metabolic state. From a bioprocess perspective, the need for higher cell

concentration to increase volumetric productivity is expected; thus the next step towards

CAV-2 vectors production process is to design a feeding strategy to obtain high MDCK-E1

cells concentration, while maintaining the corresponding cell-specific virus yield.

Although this is a nonhuman adenovirus, the mechanisms underlying viral replication

cycle and its production are similar (2, 6, 17). Therefore, a high MOI was used for vector

production assays. In most reports, typical titers range from 104 to 105 adenovirus total

particles per cell, while infectious particles are 1 log lower (2, 18, 19). Although high viral

productivity is obtained using SF medium with adherent monolayer cells in static

cultures, the fast decrease of pH after infecting cells in spinner cultures may be

responsible for the 20-fold lower productivity. By controlling pH in 2 L stirred tank

bioreactor during process validation, the productivity can be restored to yields

comparable with in static cultures. The optimal harvest times described for adenoviruses

are in the range of 40-48 hpi (20, 21).However, under stirring conditions the maximum

yield of ∆E1 CAV-2 was attained earlier. Considering that similar cell-specific productivity

was obtained in 2 L stirred tank bioreactor (24 - 36 hpi) and static cultures (40 hpi), we

speculate that stirring promotes MDCK cell metabolic activity, which ultimately leads to

faster virus production.

In conclusion, MDCK-E1 cells were an efficient producer cell line for ∆E1 CAV-2 vectors.

Cultivation of MDCK-E1 cells in stirred tank bioreactor readily reached a viable cell

concentration, and a high ∆E1 CAV-2 specific productivity at 36 hpi. For manufacturing

purposes, faster cell growth and earlier virus production time point improve process

economics.

Production with adherent cells using microcarrier technology

71

The work described herein presents the first results concerning the stirring cell culture of

MDCK-E1 cells for the production of CAV-2 vectors using SF medium, providing valuable

information for bioprocess implementation.

5. Acknowledgements and author contribution

This work was supported by Fundação para a Ciência e Tecnologia – Portugal (through

the projects PTDC/BIO/69452/2006, PTDC/EBB-BIO/119501/2010 and PTDC/EBB-

BIO/118615/2010) and European Commission (BrainCAV HEALTH – HS_2008_222992).

Paulo Fernandes acknowledges Fundação para a Ciência e a Tecnologia (FCT) for his

Ph.D. grant (SFRH/BD/70810/2010). The authors acknowledge Eng. Marcos Sousa for

the technical support in bioreaction.

Paulo Fernandes conceived the experimental setup and design, performed the

experiments, analyzed the data and wrote the chapter.

6. References

1. Dormond, E., M. Perrier, and A. Kamen. 2009. From the first to the third generation adenoviral vector: what parameters are governing the production yield? Biotechnol Adv 27:133-144.

2. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

3. Perreau, M., and E. J. Kremer. 2005. Frequency, proliferation, and activation of human memory T cells induced by a nonhuman adenovirus. J Virol 79:14595-14605.

4. Soudais, C., S. Boutin, and E. J. Kremer. 2001. Characterization of cis-acting sequences involved in canine adenovirus packaging. Mol Ther 3:631-640.

5. Bru, T., S. Salinas, and E. J. Kremer. 2010. An update on canine adenovirus type 2 and its vectors. Viruses 2:2134-2153.

6. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

7. Falkner, E., H. Appl, C. Eder, U. M. Losert, H. Schoffl, and W. Pfaller. 2006. Serum free cell culture: the free access online database. Toxicol In Vitro 20:395-400.

8. Klonjkowski, B., P. Gilardi-Hebenstreit, J. Hadchouel, V. Randrianarison, S. Boutin, P. Yeh, M. Perricaudet, and E. J. Kremer. 1997. A recombinant E1-deleted canine adenoviral vector capable of transduction and expression of a transgene in human-derived cells and in vivo. Human gene therapy 8:2103-2115.

9. Ferreira, T. B., R. Perdigao, A. C. Silva, C. Zhang, J. G. Aunins, M. J. Carrondo, and P. M. Alves. 2009. 293 cell cycle synchronisation adenovirus vector production. Biotechnol Prog 25:235-243.

10. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

11. Segura, M. M., M. Monfar, M. Puig, F. Mennechet, S. Ibanes, and M. Chillon. 2009. A real-time PCR assay for quantification of canine adenoviral vectors. J Virol Methods.

Chapter III – Part A

72

12. Salinas, S., L. G. Bilsland, D. Henaff, A. E. Weston, A. Keriel, G. Schiavo, and E. J. Kremer. 2009. CAR-associated vesicular transport of an adenovirus in motor neuron axons. PLoS pathogens 5:e1000442.

13. Genzel, Y., M. Fischer, and U. Reichl. 2006. Serum-free influenza virus production avoiding washing steps and medium exchange in large-scale microcarrier culture. Vaccine 24:3261-3272.

14. Tree, J. A., C. Richardson, A. R. Fooks, J. C. Clegg, and D. Looby. 2001. Comparison of large-scale mammalian cell culture systems with egg culture for the production of influenza virus A vaccine strains. Vaccine 19:3444-3450.

15. Genzel, Y., I. Behrendt, S. Konig, H. Sann, and U. Reichl. 2004. Metabolism of MDCK cells during cell growth and influenza virus production in large-scale microcarrier culture. Vaccine 22:2202-2208.

16. Genzel, Y., J. B. Ritter, S. Konig, R. Alt, and U. Reichl. 2005. Substitution of glutamine by pyruvate to reduce ammonia formation and growth inhibition of mammalian cells. Biotechnol Prog 21:58-69.

17. Chillon, M., and E. J. Kremer. 2001. Trafficking and propagation of canine adenovirus vectors lacking a known integrin-interacting motif. Hum Gene Ther 12:1815-1823.

18. Nadeau, I., and A. Kamen. 2003. Production of adenovirus vector for gene therapy. Biotechnol Adv 20:475-489.

19. Ferreira, T. B., A. L. Ferreira, M. J. Carrondo, and P. M. Alves. 2005. Effect of re-feed strategies and non-ammoniagenic medium on adenovirus production at high cell densities. J Biotechnol 119:272-280.

20. Altaras, N. E., J. G. Aunins, R. K. Evans, A. Kamen, J. O. Konz, and J. J. Wolf. 2005. Production and formulation of adenovirus vectors. Adv Biochem Eng Biotechnol 99:193-260.

21. Kamen, A., and O. Henry. 2004. Development and optimization of an adenovirus production process. J Gene Med 6:184-192.

Part B

Production with cells adapted to suspension

This chapter is based in the data to be published as:

Fernandes P, Sousa MFQ, Kremer EJ, Coroadinha AS, Alves PM. Suspension MDCK-E1 cell

line for the manufacturing of CAV-2 vectors. (under preparation)

Chapter III – Part B

74

Abstract

MDCK-derived cell lines can be used for canine adenovirus type 2 (CAV-2) vectors

manufacturing. However, these cells are anchorage-dependent, which complicates

process scale-up. This work aimed at streamline CAV-2 production process by adapting

our MDCK-E1 cell line to grow in suspension using serum-free medium. After directly

transferring MDCK-E1 cells to SFM4BHK medium and shaker flasks, cells were adapted to

suspension growth in 5 weeks. The newly adapted cells presented reproducible cell

growth profiles and maximum cell density up to 2.6×106 cells/mL. Adapted cells

maintained their capacity to be infected by CAV-2 and titers up to 5×108 IP/mL were

obtained. Here, we show that MDCK-E1 cells adapted to suspension can have a good

performance in what concerns virus production, providing important infection conditions

to be undertaken when streamlining CAV-2 bioprocess. This constitutes a step forward in

developing a scalable production process, contributing to make the process of CAV-2

vectors manufacturing equivalent to human adenovirus vectors.

Production with cells adapted to suspension

75

Contents

1. Introduction ............................................................................................................................................... 76

2. Materials and methods .......................................................................................................................... 76

2.1. Cell lines and culture media ............................................................................................................. 76

2.2. Viral vectors ............................................................................................................................................ 76

2.3. Adaptation to suspension growth in serum-free medium ................................................. 77

2.4. Cell growth assays ................................................................................................................................ 77

2.5. Evaluation of CAV-2 production in suspension adapted cells .......................................... 77

2.6. Stirred tank bioreactor for CAV-2 production ......................................................................... 78

2.7. Virus quantification ............................................................................................................................. 78

3. Results ........................................................................................................................................................... 79

3.1. Adaptation of MDCK-E1 cells to suspension ............................................................................. 79

3.2. Cell growth of suspension-adapted cells .................................................................................... 79

3.3. CAV-2 production in cells adapted to suspension growth under different MOIs .... 80

3.4. Reducing process steps: avoiding medium exchange at infection .................................. 81

3.5. Effect of infection strategy on the infectivity of viral particles ........................................ 83

4. Discussion.................................................................................................................................................... 85

5. Acknowledgements and author contribution ............................................................................. 88

6. References ................................................................................................................................................... 88

Chapter III – Part B

76

1. Introduction

The need for large amounts of vectors for pre-clinical and clinical assays is anticipated

due to the increasing interest in canine adenovirus type 2 (CAV-2) vectors for gene

therapy applications (1, 2), When scaling-up processes, stirred culture systems are

preferred over the typical static cultures. Previously, we established a CAV-2 E1-

trascomplementing cell line derived from MDCK and developed a scalable production

process based in the use of microcarriers (Chapters II and III, part A). Although

microcarriers represent the traditional approach to transfer anchorage-dependent cells

to stirred culture systems, their use poses additional costs and processing steps

associated with microcarriers preparation, cell expansion and monitoring that can be

cumbersome at industrial scales (3). Therefore, adaptation of cell lines to grow in

suspension, being performed for many cell lines including HEK293 for human adenovirus

production (4), has been recently explored with parental MDCK cells by gene expression

(5, 6) or culture medium manipulation (7-9).

In this work, aiming to streamline the production process of CAV-2 vectors, we

established a suspension cell line in completely serum-free conditions. MDCK-E1 cell line

was adapted to suspension growth and serum-free medium and its potential as CAV-2

producer evaluated in stirred cultures.

2. Materials and methods

2.1. Cell lines and culture media

MDCK-E1 cells were obtained by transfecting MDCK (ECCAC 841211903) with pCI-neo

plasmid (Promega, Madison, WI, USA) harboring E1 gene from CAV-2, as described

previously (10). Cells were cultivated in DMEM (Gibco, Paisley, UK) with 10% (v/v) FBS,

subcultured twice a week in 150 cm2 t-flasks and maintained in an incubator with

humidified atmosphere of 5% CO2 in air at 37°C.

2.2. Viral vectors

CAVGFP is an E1-deleted (ΔE1) vector containing an eGFP expression cassette derived

from CAV-2 strain Toronto A 26/61, GenBank J04368. Viral vectors stocks were prepared

and purified by CsCl gradients as described previously (10, 11).

Production with cells adapted to suspension

77

2.3. Adaptation to suspension growth in serum-free medium

Cells were adapted to grow in suspension with serum-free medium using a direct

approach, by transferring cells from adherent cultures directly in SFM4BHK21 serum-

free medium (HyClone GE Healthcare, Aalst, Belgium; currently manufactured upon

request) supplemented with 4mM of glutamax (Gibco) in 125 mL shaker flasks. Cells from

adherent cultures were pelleted by centrifugation (300 g, 10 min, 4°C) and re-suspended

in SFM4BHK21 medium at 1×106 cells/mL in a final working volume of 20 mL.

Suspension cultures were placed at 130-150 rpm stirrer speed on an orbital stirrer plate

(IKA-Werke, Germany) at 37°C in a humidified atmosphere containing 8% CO2. Culture

medium was then replaced every 2-3 days until cell growth became evident. Once cell

growth was observed (~3 weeks) and concentration reached 2×106 cells/mL, cells were

seeded at 0.5×106 cells/mL in fresh medium and cultured twice a week. Cells were

considered adapted after observing consistent cell growth in 3 subsequent culture

passages, and frozen thereafter.

2.4. Cell growth assays

Grothw profiles of suspension adapted cells were obtained using an inoculum of

0.5 × 106 cells/mL in 125 mL shaker flasks with a working volume of 20-25 mL. Growth

was monitored by determining cell concentration and viability at least every 24 h.

2.5. Evaluation of CAV-2 production in suspension adapted cells

Production of CAV-2 in suspension adapted MDCK-E1 cells were performed in 125 mL

shaker flasks, seeding 0.5×106 cells/mL in 20-25 mL working volume. Cells were infected

at 1×106 cells/mL or 2×106 cells/mL, with medium exchange or culture dilution with

fresh medium at infection, using CAVGFP at MOI 0.5, 1, 2.5 or 5. To determine the

percentage of infected cells in the different MOIs, GFP-positive cells were determined 24

hpi by flow cytometry (CyFlow Space, Partec, Münster, Germany). Sampling and cell

monitoring were performed at 24 h intervals. Viruses were collected by disrupting cells

with lysis buffer (Tris/HCl 10 mM, pH 8.0 and 0.1% (v/v) Triton-X 100). The resulting

sample was clarified at 3000 g for 10 min at 4°C and stored at -85°C until further analysis.

Chapter III – Part B

78

2.6. Stirred tank bioreactor for CAV-2 production

Suspension adapted MDCK-E1 cells were cultivated in 0.5 L stirred tank bioreactor

(Biostat Q-Plus, Sartorius Stedim Biotech, Goettingen, Germany) equipped with Rushton

6-blade disk impeller and a microsparger, under defined conditions (working volume: 0.3

L; pH: 7.4; temperature: 37°C; gas flow rate: 0.01 vvm; agitation rate: 50-110 rpm).

Dissolved oxygen concentration was kept constant at 40% of air saturation by controlling

gas inlet mixture with nitrogen, air and/or oxygen. pH was controlled using CO2

percentage in gas inlet. Cells were inoculated at 0.5×106 cells/mL and infected at 1×106

cells/mL with MOI 0.5 and medium exchange. For medium exchange, cells were collected

from bioreactor, centrifuged at 300 g for 15 min at 4°C and re-suspended in fresh

medium. Bioreactor was re-inoculated afterwards. Sampling and cell monitoring were

performed at 24 h intervals as described above.

2.7. Virus quantification

Quantification of infectious particles was performed by monitoring the expression of GFP

through flow cytometry from DK cells subjected to serial dilutions of viral samples, as

described previously (10). Physical (genome-containing) particles were titrated by

quantifying viral genomes through qPCR. Viral genomes were extracted and purified by

High Pure Viral Nucleic Acid Kit (Roche Diagnostics, Penzberg, Germany). SYBR Green I

dye chemistry was used to detect PCR products using Lightcycler system. Previously

purified and quantified plasmids coding for E1-deleted CAV-2 genome or GFP. To ensure

the removal of free viral genomes from samples and accurately quantify viral physical

particles, a benzonase treatment was performed prior to DNA extraction by adding 2 μl of

benzonase (250 U/μl) (Merck Millipore, Darmstadt, Germany) to the same sample

volume and incubating for 1 h at 37 °C. Benzonase was inhibited during the proteinase K

step of DNA extraction protocol. To estimate the number of viral genomes, primers

against the expression cassette of each viral vector were used. Primers for GFP gene:

forward 5’-CAGAAGAACGGCATCAAGGT-3’ and reverse 5’-CTGGGTGCTCAGGTAGTGG-3’.

Production with cells adapted to suspension

79

3. Results

3.1. Adaptation of MDCK-E1 cells to suspension

After confirming that parental MDCK cells can be successfully adapted to suspension

growth using SFM4BHK21 serum-free medium with a direct adaptation strategy (9),

MDCK-E1 cells, initially growing in DMEM with 10 % (v/v) FBS, were subjected to this

adaptation process. As a control, the parental MDCK cells were adapted in parallel

(Figure 1). After initial inoculation in SFM4BHK21 medium in shaker flasks, MDCK-E1

cells showed a 5-fold reduction in the number of viable cells (Figure 1A), and under

these conditions they took more to start growing than parental MDCK cells (Figure 1B).

MDCK-E1 cell growth in suspension was observed 3 weeks after initial inoculation and

considered adapted after 3 culture passages.

Figure 1. Cell concentration of MDCK-E1 (A) and parental MDCK cells (B) during adaptation process. Cells were considered adapted after showing consistent growth in 3 consecutive passages. P: passage number.

3.2. Cell growth of suspension-adapted cells

The growth of MDCK-E1 cells was evaluated at culture passage 4 and compared to

parental MDCK cells subjected to same adaptation procedure. Similarly to parental MDCK

cells, MDCK-E1 cells showed a specific growth rate of 0.3 day-1, although with a lower

maximum cell concentration (1.6×106 cells/mL for MDCK-E1 cells versus 2.3×106

cells/mL for parental MDCK cells) (Figure 2). MDCK-E1 cells were maintained for more

Chapter III – Part B

80

20 culture passages. An increase of 1.5 fold in specific cell growth and maximum cell

density of MDCK-E1 cells was obtained after 20 passages: cells were able to grow up to

2.6×106 cells/mL (Figure 2). Parental MDCK cells after 24 passages kept the same growth

profile.

Figure 2 - Cell growth of MDCK and MDCK-E1 in suspension cultures after adaptation. Two passage numbers were evaluated for each cell line. P: passage number.

3.3. CAV-2 production in cells adapted to suspension growth under different MOIs

Due to stirring, infection is usually more efficient in suspension cultures than in adherent

cultures. MOI should be thus tuned to ensure that infection is not compromising cell

viability that can impact virus production yields. MDCK-E1 cells were infected at 1×106

cells/mL with medium exchange at infection and MOI 5, 2.5, 1 and 0.5. Then, cell

concentration, percentage of infected cells and virus production were evaluated (Figure

3). The results show that the majority of cells were infected with the different MOIs

assayed (Figure 3A). In fact, using a MOI as low as 0.5, 75% of cells were shown to be

infected. Cell death was in proportion to the MOI used, in which high virus input led to

earlier decrease in cell concentration (Figure 3B). While amplification ratios where

higher when low MOIs were used (Figure 3C), the total volumetric productivity was

higher when using high MOIs, with maximum productivity of 4.9 × 108 infectious particles

(IP)/mL obtained at MOI 5 (Figure 3D). On the other hand, MOI 0.5 led to a maximum

volumetric productivity of 1.4×108 IP/mL. CAV-2 production was then assayed in stirred

tank bioreactor cultures as a proof of concept. Cell culture and infection was prepared as

described above: infection at 1×106 cells/mL with medium exchange and MOI of 0.5.

Production with cells adapted to suspension

81

Similarly to shaker flasks results, a volumetric productivity of 1.3×108 IP/mL was

obtained. The best time of harvest was at 48 hpi. At this time point, CAV-2 vectors

presented 43 physical particles (PP) per each IP quantified.

Figure 3. Effect of MOI on CAV-2 production in suspension cultures of MDCK-E1 cells. (A) percentage of infected cells at 24 hpi. (B) Cell concentration after infection. (C) Amplification ratio of CAV-2 under different MOIs. (D) Total volumetric productivity (IP/mL). Cells were infected at 1×106 cells/mL with medium exchange. Values are shown as average ± standard-deviation (n=2).

3.4. Reducing process steps: avoiding medium exchange at infection

Production of adenovirus in a lab-scale is typically performed with medium exchange at

infection. Although easy to perform in static cultures, medium exchange with suspension

cultures implies additional steps including centrifugation and re-inoculation of cells in

fresh medium. To overcome this, we diluted the cell culture at infection time with fresh

Chapter III – Part B

82

medium (Figure 4). Dilutions were tested at two cell concentrations: 1×106 cells/mL and

2×106 cells/mL. Cultures at 1×106 cells/mL were diluted to 0.5×106 cells/mL, while

cultures at 2×106 cells/mL were diluted to 1 × 106 cells/mL and 0.5×106 cells/mL. To

compare with previous work (Chapter III, part A), cells were infected at MOI 5. Cultures

infected at 1×106 cells/mL and 2×106 cells/mL with medium exchange were used as

controls.

Figure 4. Scheme with the experimental design used to test the effect of culture dilution with fresh medium on CAV-2 production. Cells were grown up to 1×106 cells/mL and 2×106 cells/mL. Cultures at 1×106 cells/mL were diluted to a cell concentration of 0.5×106 cells/mL (1:2) and infected afterwards; Cultures at 2×106 cells/mL were diluted to 1×106 cells/mL (1:2) and 0.5×106 cells/mL (1:4) at infection. In addition, cultures with total medium exchange at infection by centrifugation were used as controls. Cells were infected at MOI 5.

The assays performed without medium exchange showed a ~8-fold decay in virus

productivities, when compared to assays with medium exchange (data not shown).

The results show a slight decay, although without statistical mean, in total infectious

titers (volumetric titers normalized to dilution) in all diluted cultures, when compared to

control cultures (performed with medium exchange at infection) (Figure 5). The

normalized volumetric productivity of cultures diluted from 2×106 cells/mL to 1×106

cells/mL or to 0.5×106 cells/mL was higher than that obtained in cultures obtained from

cultures at 1×106 cells/mL. Together, these results show the possibility to maintain CAV-

2 titers by diluting the culture to be infected with fresh medium (Figure 5). To ensure the

Production with cells adapted to suspension

83

success of this strategy, MDCK-E1 cells culture must be at 2×106 cells/mL at

dilution/infection time.

Figure 5. Effect of cell concentration and dilution with fresh medium on volumetric productivity (IP/mL) measured at 96 hpi. Cell cultures were prepared as described in Figure 4. w/ ME: with medium exchange. Values are shown as average ± standard-deviation (n=3).

3.5. Effect of infection strategy on the infectivity of viral particles

To evaluate the infectivity of CAV-2 vectors produced in suspension cultures under the

different infection strategies (Figure 4), physical (genome containing) particles were

quantified and compared to IP. Results are shown as PP per cell and IP per cell,

respectively.

Similarly to volumetric titers (Figure 5), diluted cultures showed a slight decay in cell

specific infectious titer (Figure 6). In addition, a tendency to obtain less PP per cell in

cultures resulting from 2×106 cells/mL cells was observed.

PP to IP ratios were calculated to better evaluate the quality of CAV-2 vectors generated

under the different conditions (Table 1). PP:IP ratio of CAV-2 vectors generated

according to the bioprocess previously described (12) was used as a control. Consistent

to Figure 6 data, cultures diluted from 2×106 cells/mL delivered viruses with better

quality (infectivity) than cultures from 1×106 cells/mL (Table 1). Still, a slight increase

(1.8 to 2.3-fold) in non-infectious particles was observed in these cultures, when

compared to the control culture (bioreactor culture of MDCK-E1 grown in microcarriers

and using Optipro SFM) (Chapter III, part A). Worth mentioning that in bioreactor

Chapter III – Part B

84

cultures PP:IP ratio of CAV-2 vectors produced in suspension adapted MDCK-E1 cells was

similar to that obtained in the control culture (43:1 and 32:1, respectively).

Figure 6. Cell specific productivity of PP (black bars, left axis) and IP (white bars, right axis) in the different cultures at 96 hpi. Cell cultures were prepared as described in Figure 4. Values are shown as average ± standard-deviation (n=3).

Table 1. Physical to infectious particles ratio of CAV-2 vectors obtained in different cultures.

Cultures PP per each IP

Prior dilution After dilution/at infection

1x106 cells/mL 1x106 cells/mL (w/ ME) 93 ± 10 *

0.5x106 cells/mL 114 ± 7 **

2x106 cells/mL

2x106 cells/mL (w/ ME) 59 ± 13

1x106 cells/mL 72 ± 11 ***

0.5x106 cells/mL 74 ± 20

MDCK-E1 in Optipro SFM (control) 32 ± 16

Values are shown as average ± standard-deviation (n=3); * p = 0.006, ** p = 0.001 and *** p = 0.03, given by a single factor Anova analysis against the values obtained with control. Worth mentioning that suspension-adapted MDCK-E1 cells in bioreactor cultures delivered CAV-2 vectors with 43 PP per each IP (data in the text, n=2). w/ ME: with medium exchange.

Production with cells adapted to suspension

85

4. Discussion

The increasing demands of CAV-2 vectors for gene transfer (1, 2, 13), require larger

amounts of high quality vectors to conduct preclinical and possibly clinical trials. To

develop a virus manufacturing GMP-compliant and scalable process, producer cell line

should be well characterized, preferably growing in suspension in serum-free medium,

and permit virus replication to high titers. To fully meet these requirements, the

previously established MDCK-E1 cell line (10) (Chapter II) was adapted to grow in

suspension with serum-free medium. The ΔE1 CAV-2 vectors production was analyzed.

Several methods to achieve parental MDCK cells stably growing in suspension by either

medium (7-9) or genetic manipulations (5, 6) have been described. The work of van

Wielink et al. (9) probably represents the fastest strategy to adapt MDCK cells to

suspension growth and was adapted for this work. Based on that, we were able to adapt

MDCK cells by directly transferring cells to shaker flasks with SFM4BHK21 medium,

skipping the adaptation phase in adherent cultures previously described and therefore

reducing the adaptation process in 4-6 weeks. Accordingly, single MDCK-E1 cells growing

in suspension were obtained. Two additional weeks were required for adaptation of

MDCK-E1 when comparing with the adaptation of parental MDCK cells (Figure 1). This

suggests that the adaptive potential of MDCK was reduced during the establishment of

MDCK-E1 cell line. As shown previously, parental MDCK cells depict a heterogeneous

population (14, 15). The original variability of MDCK cell population, which possibly

favored its adaptation to suspension by selecting a clone or a subset of clones more prone

to survive and/or grow in suspension, may not be well represented in MDCK-E1 cell line;

MDCK-E1 cell line represents a clone selected through limiting dilution in static cultures,

which might explain why the adaptation of this cell line was more difficult.

In accordance to the work of van Wielink et al. (9), parental MDCK cells adapted herein

showed similar maximum cell density (Figure 2). However, in our hands growth rate of

MDCK and was 2-fold lower than that described. MDCK-E1 cells showed a slight decrease

in the maximum cell density at culture passage 4, which can be correlated with the

adaptive profile of this cell line (Figure 1). We could overcome this by maintaining

MDCK-E1 cells for 20 more culture passages (Figure 2).

To counteract the absence of serum and/or adhesion surface with the survival and

growth in a suspension culture environment, the levels of membrane proteins and cell

surface receptors of suspension adapted cells can change from the original cell (9, 16).

Chapter III – Part B

86

However, the receptors involved in virus entry, such as CAR (17), must be still present in

the newly adapted MDCK-E1 cells, since the permissiveness to CAV-2 infection was

maintained (Figure 3A).

Ideally, the MOI to be used in suspension cultures should be optimized, as the high MOIs

used in adherent cultures might compromise the cell viability and final virus titers in

stirred cultures. In addition, agitation maximizes the number of infected cells and low

MOIs are suitable for stirred cultures. In fact, earlier cell death was observed in cells

infected at high MOI (Figure 3B). Volumetric titers ranged from 1.4 - 4.9×108 IP/mL in

proportion to the MOI used (Figure 3D). When comparing MOI 5 with MOI 0.5, 3.5-fold

improvement in volumetric titer was observed although 10-times more viral vectors

were spent to infect same cell number. Accordingly, the amplification ratio under MOI 0.5

was higher than that obtained with MOI 5 (Figure 3C). This indicates that infection of

MDCK-E1 cells in suspension with MOI 0.5 is preferable: the majority of cells are infected,

productivity (per virus particle) is maximized and virus seed stock is saved. Still, given

the process previously established in stirred tank bioreactors (12) and the typical MOI

used for adenovirus production (18), we decided to perform the following assays with

MOI 5.

To avoid any limitation in nutrient(s) that could affect for virus propagation and/or

accumulation of inhibitory byproducts, medium exchange at infection time is typically

applied for small-scale productions of adenoviruses. However, from a bioprocess point of

view this is difficult to manage with suspension cells, as it implies transfer of bioreactor

culture, centrifugation and re-inoculation. To address this possible bottleneck, we

evaluated the effect of diluting culture prior infection with new culture medium (Figure

4). This would allow the addition of fresh nutrients, dilute any toxic byproduct and

circumvent centrifugation step. Indeed, when using diluted cultures from cells at 2×106

cells/mL, total CAV-2 titers obtained were similar to those assays with medium exchange

(Figure 5 and Figure 6). More specifically, similar CAV-2 titers can be obtained either by

diluting the culture 2 or 4 times (Figure 5). From a bioprocess point of view, CAV-2

vectors should be thus produced by diluting the culture to half: this would save culture

medium and alleviate downstream processing, as less working volume is being managed.

Assuming the medium nutrient limitation/byproducts accumulation hypothesis, one

should expect that cultures with less growing time (i.e. at cell concentration of 1×106

cells/mL) would have less medium-related limitations than those in which cells were

Production with cells adapted to suspension

87

grown for longer time periods (i.e. at cell concentration of 2×106 cells/mL). However, the

infectious titers of cultures generated from cells at 2×106 cells/mL were higher than that

obtained with cultures generated from cells at 1×106 cells/mL (Figure 5). This indicates

that cell status and/or physiology gathered at concentration of 2×106 cells/mL is better

suited for virus production in terms of infectious titers (Figures 5 and 6) and quality of

viral particles (Table 1). The relation between metabolites, cell cycle phase and CAV-2

production should be thus further explored to take fully profit of this feature (19).

When transferring to stirred tank bioreactor, no significant improvements in productivity

were observed, although the resulting vectors had higher infectivity (PP:IP ratio of 43:1)

than those obtained in shaker flasks and similar to our control culture (bioreactor culture

with MDCK-E1 in microcarriers) (Table 1). In the bioreactors established previously with

MOI 5, a volumetric productivity of ~2.8×109 IP/mL (n=4) is typically obtained (Chapter

III, part A), which is 5-fold higher than that obtained with the new suspension cells

(Figure 3D). Nevertheless, worth mentioning that the use of suspension cells greatly

facilitates culture manipulation in what regards microcarriers preparation and cell seed

from adherent cultures. In addition, unlike the bioprocess previously established

(Chapter III, part A), serum is now totally abolished from production process.

In summary, we show the adaptation process of MDCK-E1 cell line to suspension, by

transferring them directly to stirred cultures in SFM4BHK21 culture medium. Using

adapted cells, CAV-2 vectors titers up to 5.1×108 IP/mL were possible, and a PP value of

43 per each IP was obtained in bioreactor cultures. Feeding cells at infection time with

fresh medium enabled same specific titers, overcoming the need of medium

exchange/cell centrifugation at infection time. Together, this work constitutes a progress

in CAV-2 vectors availability at larger scales, contributing to make the manufacturing

process of CAV-2 vectors equivalent to human adenovirus vectors.

Chapter III – Part B

88

5. Acknowledgements and author contribution

This work was supported by Fundação para a Ciência e Tecnologia – Portugal (through

the project PTDC/EBB-BIO/118615/2010) and European Commission (BrainCAV

HEALTH – HS_2008_222992). Paulo Fernandes acknowledges Fundação para a Ciência e

a Tecnologia (FCT) for his Ph.D. grant (SFRH/BD/70810/2010). The authors

acknowledge Rute Castro and Tanja Laske for the fruitful discussions about the

adaptation of MDCK cells to suspension growth.

Paulo Fernandes conceived the experimental setup and design, performed the

experiments, analyzed the data and wrote the chapter.

6. References

1. Cubizolle, A., N. Serratrice, N. Skander, M. A. Colle, S. Ibanes, A. Gennetier, N. Bayo-Puxan, K. Mazouni, F. Mennechet, B. Joussemet, Y. Cherel, Y. Lajat, C. Vite, F. Bernex, V. Kalatzis, M. E. Haskins, and E. J. Kremer. 2014. Corrective GUSB Transfer to the Canine Mucopolysaccharidosis VII Brain. Mol Ther 22:762-773.

2. Ariza, L., L. Gimenez-Llort, A. Cubizolle, G. Pages, B. Garcia-Lareu, N. Serratrice, D. Cots, R. Thwaite, M. Chillon, E. J. Kremer, and A. Bosch. 2014. Central Nervous System Delivery of Helper-Dependent Canine Adenovirus Corrects Neuropathology and Behavior in Mucopolysaccharidosis Type VII Mice. Hum Gene Ther 25:199-211.

3. Silva, A., P. Fernandes, M. Q. Sousa, and P. Alves. 2014. Scalable Production of Adenovirus Vectors, p. 175-196. In M. Chillón and A. Bosch (ed.), Adenovirus, vol. 1089. Humana Press.

4. Silva, A. C., C. Peixoto, T. Lucas, C. Kuppers, P. E. Cruz, P. M. Alves, and S. Kochanek. 2010. Adenovirus vector production and purification. Curr Gene Ther 10:437-455.

5. Chu, C., V. Lugovtsev, H. Golding, M. Betenbaugh, and J. Shiloach. 2009. Conversion of MDCK cell line to suspension culture by transfecting with human siat7e gene and its application for influenza virus production. Proc Natl Acad Sci U S A 106:14802-14807.

6. Chu, C., V. Lugovtsev, A. Lewis, M. Betenbaugh, and J. Shiloach. 2010. Production and antigenic properties of influenza virus from suspension MDCK-siat7e cells in a bench-scale bioreactor. Vaccine 28:7193-7201.

7. Tsutsumi, R., S. Fujisaki, M. Shozushima, K. Saito, and S. Sato. 2006. Anoikis-resistant MDCK cells carrying susceptibilities to TNF-alpha and verotoxin that are suitable for influenza virus cultivation. Cytotechnology 52:71-85.

8. Lohr, V., Y. Genzel, I. Behrendt, K. Scharfenberg, and U. Reichl. 2010. A new MDCK suspension line cultivated in a fully defined medium in stirred-tank and wave bioreactor. Vaccine 28:6256-6264.

9. van Wielink, R., H. C. Kant-Eenbergen, M. M. Harmsen, D. E. Martens, R. H. Wijffels, and J. M. Coco-Martin. 2011. Adaptation of a Madin-Darby canine kidney cell line to suspension growth in serum-free media and comparison of its ability to produce avian influenza virus to Vero and BHK21 cell lines. J Virol Methods 171:53-60.

10. Fernandes, P., V. M. Santiago, A. F. Rodrigues, H. Tomas, E. J. Kremer, P. M. Alves, and A. S. Coroadinha. 2013. Impact of E1 and Cre on adenovirus vector amplification: developing MDCK CAV-2-E1 and E1-Cre transcomplementing cell lines. PLoS One 8:e60342.

11. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

Production with cells adapted to suspension

89

12. Fernandes, P., C. Peixoto, V. M. Santiago, E. J. Kremer, A. S. Coroadinha, and P. M. Alves. 2013. Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20:353-360.

13. Serratrice, N., A. Cubizolle, S. Ibanes, N. Mestre-Frances, N. Bayo-Puxan, S. Creyssels, A. Gennetier, F. Bernex, J. M. Verdier, M. E. Haskins, G. Couderc, F. Malecaze, V. Kalatzis, and E. J. Kremer. 2014. Corrective GUSB transfer to the canine mucopolysaccharidosis VII cornea using a helper-dependent canine adenovirus vector. Journal of controlled release : official journal of the Controlled Release Society 181:22-31.

14. Dukes, J. D., P. Whitley, and A. D. Chalmers. 2011. The MDCK variety pack: choosing the right strain. BMC cell biology 12:43.

15. Lugovtsev, V. Y., D. Melnyk, and J. P. Weir. 2013. Heterogeneity of the MDCK cell line and its applicability for influenza virus research. PLoS One 8:e75014.

16. Jaluria, P., M. Betenbaugh, K. Konstantopoulos, B. Frank, and J. Shiloach. 2007. Application of microarrays to identify and characterize genes involved in attachment dependence in HeLa cells. Metab Eng 9:241-251.

17. Soudais, C., S. Boutin, S. S. Hong, M. Chillon, O. Danos, J. M. Bergelson, P. Boulanger, and E. J. Kremer. 2000. Canine adenovirus type 2 attachment and internalization: coxsackievirus-adenovirus receptor, alternative receptors, and an RGD-independent pathway. J Virol 74:10639-10649.

18. Altaras, N. E., J. G. Aunins, R. K. Evans, A. Kamen, J. O. Konz, and J. J. Wolf. 2005. Production and formulation of adenovirus vectors. Adv Biochem Eng Biotechnol 99:193-260.

19. Ferreira, T. B., R. Perdigao, A. C. Silva, C. Zhang, J. G. Aunins, M. J. Carrondo, and P. M. Alves. 2009. 293 cell cycle synchronisation adenovirus vector production. Biotechnol Prog 25:235-243.

Chapter III – Part B

90

Chapter IV

Implications of Cre activity and

co-infection on canine

helper-dependent vector production

This chapter is adapted from:

Fernandes P, Almeida AI, Kremer EJ, Alves PM, Coroadinha AS. (2015). Production of

canine helper-dependent vectors: implications of Cre activity and co-infection on adenovirus

propagation. Sci Rep. (accepted).

Chapter IV

92

Abstract

The importance of Cre recombinase to minimize helper vector (HV) contamination during

helper-dependent adenovirus vectors (HDVs) production is well documented. However,

Cre recombinase, by inducing DNA double-strand breaks (DSBs), can cause a reduced

proliferation and genotoxic effects in cultured cells. In this work, Cre-expressing cell

stability, co-infection and their relation to adenovirus amplification/HV contamination

were evaluated to develop a production protocol for HD canine adenovirus type 2 (CAV-

2) vectors. Long-term Cre expression reduced the capacity of MDCK-E1-Cre cells to

produce CAV-2 by 7-fold, although cell growth was maintained. High HDV/HV MOI ratio

(5:0.1) led to low HV contamination without compromising HDV yields. Indeed, such MOI

ratio was sufficient to reduce HV levels, as these were similar either in MDCK-E1 or

MDCK-E1-Cre cells. This raises the possibility of producing HDVs without Cre-expressing

cells, which would circumvent the negative effects that this recombinase holds to the

production system. Here, we show how Cre and MOI ratio impact adenovirus vectors

yields and infectivity, providing key-information to design an improved manufacturing of

HDV. Potential mechanisms to explain how Cre is specifically impacting cell productivity

without critically compromising its growth are presented.

Implications of Cre activity and co-infection on canine helper-dependent vector production

93

Contents

1. Introduction ............................................................................................................................................... 94

2. Materials and Methods .......................................................................................................................... 95

2.1. Cell lines and culture media ............................................................................................................. 95

2.2. Viral vectors ............................................................................................................................................ 96

2.3. Cell growth assays ................................................................................................................................ 96

2.4. Cre activity ............................................................................................................................................... 96

2.5. Viral vectors production assays ..................................................................................................... 97

2.6. Infectious vectors titration ............................................................................................................... 97

2.7. Quantification of physical particles .............................................................................................. 97

3. Results ........................................................................................................................................................... 99

3.1. Cell line evaluation at different culture passages ................................................................... 99

3.1.1. Growth characteristics of MDCK-E1-Cre ........................................................................... 99

3.1.2. Cre expression and excision efficiency of MDCK-E1-Cre .........................................100

3.1.3. CAV-2 production of MDCK-E1-Cre ...................................................................................100

3.2. Establishing the best conditions for HD CAV-2 production .............................................102

3.2.1. HDV /HV MOI ratio ..................................................................................................................102

3.2.2. The impact of MOI ratio in co-infections .........................................................................102

3.2.3. The role of Cre recombinase and MOI on the propagation of HV .........................104

3.2.4. The relation between viral particles infectivity, Cre and packaging ..................104

4. Discussion..................................................................................................................................................106

5. Acknowledgments and author contribution ..............................................................................109

6. References .................................................................................................................................................109

Chapter IV

94

1. Introduction

Adenoviruses have received considerable attention as vectors for gene therapy and their

genome has been progressively modified to improve safety and efficacy in therapeutic

applications. From vectors with the deletion of the E1 region to helper-dependent vectors

(HDVs) with the deletion of viral genes, an enhanced capacity for a gene therapeutic

insertion from ~7 kb to ~36 kb has been achieved (1). Most replication-defective

adenovirus vectors require for manufacturing and replication a cell line that expresses

the adenoviral E1 functions in trans. However, since HDVs retain only the inverted

terminal repeats and the packaging signal, early and late viral functions need to be

provided in a synchronized fashion. Standard productions of HDVs are based on co-

infection of HDV and a helper vector (HV), which provides all viral proteins in trans.

During production, both HDV and HV genomes replicate inside the cells, but the

encapsidation of the HV genome is minimized, by flanking its packaging signal (Ψ) with

recognition sequences for a recombinase constitutively expressed by the cells, such as

Cre (2). Therefore, the E1 transcomplementing cell line for HDV production must also

express the recombinase (2-4). Cre/loxP system was the first system described to

efficiently reduce HV contaminant, and is still the most common system used for HDV

propagation.

The importance of high levels of Cre to avoid HV propagation is unquestionable (5). In

fact, considering that the remaining HV contaminant was due to limited Cre activity levels

that permitted HV to escape packaging signal excision (5), major advances were made to

increase the levels of recombinase during HDV production (6). On the other hand, the

effect of Cre expression on producer cell homeostasis has been undervalued. Cre

expression can result in a markedly reduced proliferation and genotoxic effects in

cultured cells (7). The aberrant activity on multiple pseudo loxP sites presented in the

mammalian genome (8) induces DNA double-strand breaks (DSBs) or nicks that are

converted to DSBs during DNA replication, leading to arrest of cells in G2/M phase

because of intolerable DNA damage (7). Furthermore, although sustained low-levels

might be supported by cells without significant toxicity, they cause a slow, cumulative

increase in recombination and chromosome abnormalities. Therefore, the stability of

cells constitutively expressing Cre recombinase is likely to be impaired. While these

effects are not substantiated in literature in the scope of adenovirus production, they are

considerably described for several cell lines (7, 9-11), including HEK 293 cells (9),

Implications of Cre activity and co-infection on canine helper-dependent vector production

95

traditionally used to propagate adenoviruses. From a bioprocess point-of-view, failure of

producer cell line robustness to maintain performance compromises the viral

preparations quality, process yields, reproducibility, economics and the regulatory

approval.

Previously, we established MDCK-derived cell lines for the production of canine

adenovirus type 2 (CAV-2) vectors (12). In this work, we studied the impact of Cre

recombinase in the scope of an optimal production process for HD CAV-2 production with

MDCK-derived cells in serum-free medium, emphasizing two aspects: cell line stability

and infection conditions. Canine adenovirus type 2 (CAV-2) vectors were selected

because of their attractive features to bypass the clinical disadvantages of using human

adenoviruses while keeping other advantages (13, 14), to address fundamental

neurobiological questions (15-19) and to develop potential treatment of

neurodegenerative and ocular disorders (20-22). Moreover, MDCK cells are already

approved by the regulatory authorities for the manufacture of vaccines and thus

represent a suitable cell substrate that might facilitate the approval of clinical grade CAV-

2 vectors production (23). MDCK-E1-Cre cell line stability was addressed by evaluating

growth, Cre activity levels and virus production in cells with increasing culture passages.

Different HDV/HV MOI ratios were assayed to determine the best infection conditions to

amplify HD CAV-2 and to understand the relation between Cre expression, infection

conditions and HV propagation. Here, we show how Cre expression impacts cell

performance and adenovirus propagation and how infection conditions significantly

contribute to reduce HV contamination during HDV production even in the absence of

Cre-recombinase.

2. Materials and Methods

2.1. Cell lines and culture media

MDCK-E1 cells (12) are MDCK (ECCAC 841211903) derived and stably express the E1

region from CAV-2 and the neomycin resistance gene. MDCK-E1-Cre cells (12), in addition

to E1, also express Cre recombinase and zeocin resistance gene. MDCK-E1 and MDCK-E1-

Cre were sequentially adapted to Optipro serum-free medium (Gibco, Paisley, UK) with 4

mM glutamax (Gibco) by increasing the percentage of Optipro medium in subculture

passages. Cells were subcultured twice a week in 150 cm2 t-flasks and maintained in an

Chapter IV

96

incubator with humidified atmosphere of 5% CO2 in air at 37 °C. For splitting or collecting

cells, the monolayer was washed with PBS, incubated with 0.25% (w/v) trypsin-EDTA

(Gibco) at 37 °C until detachment started to become evident (up to 15 min) and cells

suspended in culture medium. Cells were then pelleted at 300 g during 10 min and re-

suspended in fresh medium. To evaluate the effect of culture passages, cells were

maintained in duplicates, either with or without antibiotic pressure. For antibiotic

pressure, 500 µg/mL of Geneticin (G418) (Invivogen, Toulouse, France) was added to

culture medium for MDCK-E1 cells maintenance, while for MDCK-E1-Cre cells G418 plus

500 µg/mL of Zeocin (Invivogen) was used. Cells were frozen after 10 and 20 cell culture

passages in CryoStor CS10 (Sigma-Aldrich, St. Louis, MO, USA) using a Mr. Frosty Freezing

Container (Thermo-scientific, Rockford, IL, USA).

2.2. Viral vectors

CAVGFP, JB∆5 and HD CAVGFP are vectors derived from CAV-2 strain Toronto A 26/61,

GenBank J04368. CAVGFP (13) and HD CAVGFP (16) are E1-deleted (ΔE1) and HD

vectors, respectively, and contain an eGFP expression cassette. JB∆5 is a helper vector

(HV) containing loxP sites flanking the packaging domain and a RSV-lacZ expression

cassette (16). CAVGFP, JB∆5 and HD CAVGFP viral stocks were prepared and purified by

CsCl gradients as described previously (12, 13).

2.3. Cell growth assays

MDCK-E1-Cre growth assays in static cultures were performed using an inoculum of 1 x

104 cells/cm2 in 25 cm2 t-flasks with a working volume of 10 mL. Cell concentration and

viability was determined by the trypan blue exclusion method using a 0.1% (v/v) solution

prepared in PBS and counting cells in a Fuchs-Rosenthal haemacytometer (Brand,

Wertheim, Germany).

2.4. Cre activity

Relative Cre activity was assayed by the level of luciferase activity generated by the

adenovirus vector AdMA19 (a gift from F. Graham) after infecting cells (24). AdMA19

contains the luciferase cDNA under control of the human cytomegalovirus promoter but

separated from it by an extraneous spacer sequence composed by a series of initiation

Implications of Cre activity and co-infection on canine helper-dependent vector production

97

and stop codons in several reading frames. This translational “start/stop sequence”,

flanked by loxP, disrupts translation. If the cell clone expresses Cre, the stop sequence is

floxed out and translation proceeds leading to luciferase expression. Briefly, cells were

seeded in 24 well-plates in quadruplicates and infected the day after with 20 IP/cell of

AdM19 with medium exchange. Twenty-four hpi, cells were counted and lysed to release

luciferase, both in duplicates. The resulting supernatant was collected and the light units

were quantified with a Modulus Luminometer from Turner Biosystems (Sunnyvale, CA,

USA) after adding luciferin and normalized to cell concentration.

2.5. Viral vectors production assays

Cells were seeded at 3 × 104 cells/cm2, infected the day after with medium exchange. To

evaluate cells productivity at different passage number, infections of CAVGFP at MOI 5

were performed. Excision efficiency was estimated by determining the fold-reduction in

helper JB∆5 production with MDCK-E1-Cre cells infected at MOI 5. The assays for HD

CAVGFP production optimization were performed by co-infecting cells with HD CAVGFP

at MOI 5 and JB∆5 at MOI 1, 0.5 and 0.1. To further understand how Cre recombinase and

co-infection determine optimal HD CAVGFP production, co-infections of CAVGFP (MOI 5)

with JB∆5 (MOI 5, 1 and 0.5), and single infections of JB∆5 (MOI 5, 1 and 0.5) were also

performed. Moreover, all the infection assays were performed in MDCK-E1-Cre and

MDCK-E1 cells. Once infected, cells were incubated for further 48 h and viral samples

collected using triton (Sigma-Aldrich) 0.1% (v/v) in Tris-HCl, clarified at 3000 g for 10

min at 4 °C and stored at -85 °C until further analysis.

2.6. Infectious vectors titration

Quantification of infectious CAVGFP and HD CAVGFP was performed by monitoring the

expression of GFP, while titration of infectious JB∆5 vectors was based in lacZ gene

expression and β-galactosidase activity as described previously (12).

2.7. Quantification of physical particles

Viral genomes were extracted and purified by High Pure Viral Nucleic Acid Kit (Roche

Diagnostics, Penzberg, Germany). SYBR Green I dye chemistry was used to detect PCR

products using Lightcycler system. Previously purified and quantified plasmids coding for

Chapter IV

98

E1-deleted CAV-2 genome, GFP or lacZ were used as standards. To ensure the removal of

free viral genomes from samples and accurately quantify viral physical particles, a

benzonase treatment was performed prior to DNA extraction by adding 2 μl of benzonase

(250 U/μl) (Merck Millipore, Darmstadt, Germany) to the same sample volume and

incubating for 1 h at 37 °C. Benzonase was inhibited during the proteinase K step of DNA

extraction protocol. To estimate the number of viral genomes, primers against the

expression cassette of each viral vector were used. Primers for GFP gene: forward 5’-

CAGAAGAACGGCATCAAGGT-3’ and reverse 5’-CTGGGTGCTCAGGTAGTGG-3’; primers for

lacZ gene: forward 5’-ACTATCCCGACCGCCTTACT-3’ and reverse 5’-

TAGCGGCTGATGTTGAACTG-3’.

Implications of Cre activity and co-infection on canine helper-dependent vector production

99

3. Results

MDCK-E1-Cre cells, once adapted to grow in Optipro SFM, were maintained in culture for

20 more passages (P) in duplicates, with and without antibiotic pressure, and frozen at

P0, P10 and P20 (see material and methods section). MDCK-E1 cells, subjected to same

procedure, were used as a control for CAV-2 vector production. The experiments to

determine the best infection conditions for HD CAVGFP (HDV) production were

performed with cells at P0.

3.1. Cell line evaluation at different culture passages

3.1.1. Growth characteristics of MDCK-E1-Cre

Growth curves of MDCK-E1-Cre cells obtained in the different cell culture passages were

performed in 25 cm2 t-flasks. Concentration and viability were determined every day

(Figure 1).

The results show that cell growth was similar in all culture passages, regardless of

antibiotic pressure. On average, MDCK-E1-Cre cells showed a specific growth rate of 1.00

± 0.07 day-1 (Figure 1A) and a minimum cell viability of 63 ± 3 % obtained during

stationary phase (Figure 1B). These results indicate that increasing passages and

selective pressure had no impact on MDCK-E1-Cre cell growth.

Figure 1. Effect of cell culture passage and antibiotic pressure on MDCK-E1-Cre cell growth (A) and viability (B). Dashed line represents the average of data points and error bars the corresponding standard-deviation. w/: with antibiotic pressure; w/o: without antibiotic pressure.

Chapter IV

100

3.1.2. Cre expression and excision efficiency of MDCK-E1-Cre

To evaluate the impact of increasing passages in Cre expression, two approaches were

used. The first estimates Cre activity through the level of luciferase activated by the

excision of a stop sequence flanked by loxP sites (view materials and methods section)

(Figure 2). In the second approach, Cre activity was estimated by evaluating the

reduction in HV JB∆5 propagation (due to excision of packaging genome) in MDCK-E1-Cre

cells (Table 1). The results obtained from both methods were similar. A decrease in Cre

activity up to 1.5 ± 0.3 fold, was obtained in cells at P20 when maintaining the antibiotic

pressure (Figure 2), nevertheless this reduction did not significantly alter the HV

excision efficiency (Table 1). Without the selective pressure, a further 2.5 ± 0.5 fold

decrease in Cre activity was observed from P10 to P20 (Figure 2), which contributed to

approximately 3-fold reduction in the capacity for HV genomes excision (Table 1).

Figure 2. Effect of cell culture passage and antibiotic pressure on Cre-activity of MDCK-E1-Cre cells. Light units were normalized to cell concentration and background obtained with MDCK-E1 cells. Values are shown as average ± standard-deviation (n=3). w/ AB: with antibiotic pressure; w/o AB: without antibiotic pressure. * p = 0.01, given by a single factor Anova analysis against the corresponding P0 values.

3.1.3. CAV-2 production of MDCK-E1-Cre

To evaluate the impact of increasing subculture passages on the capacity to produce viral

vectors, production assays with CAVGFP were performed. MDCK-E1 cells maintained in

parallel under same passages number were added to the experiments and used as

controls for ∆E1 production.

Implications of Cre activity and co-infection on canine helper-dependent vector production

101

Table 1. Effect of cell culture passage and antibiotic pressure on excision efficiency of HV genomes in MDCK-E1- Cre cells.

Passages Excised/unpackaged HV genomes (%)

w/ AB w/o AB

P0 98.9 ± 0.6

P10 98.4 ± 1.5 97.7 ± 2.3

P20 92.4 ± 7.2 28.5 ± 2.3

Values are shown as average ± standard-deviation (n=2). Since MDCK-E1 and MDCK-E1-Cre cells have similar productivities at P0 (Figure 3), the HV amplification reduction obtained in MDCK-E1-Cre cells corresponds to Cre-mediated excision of HV genomes avoiding their packaging. The percentage of excised HV genomes was estimated assuming that the differences of HV amplification titers between (a) MDCK-E1 and (b) MDCK-E1-Cre cells corresponds to excised/unpackaged HV genomes as follows: ([a-b]/a). Value (b) was also normalized to the fold-reduction of MDCK-E1-Cre cells productivity observed during passages (Figure 3). w/ AB: with antibiotic pressure; w/o AB: without antibiotic pressure.

In MDCK-E1 cells, an increase of 2.1 ± 0.3 fold in productivity was observed with

increasing cell passages and regardless of selective pressure (Figure 3). Such feature has

been reported for these cells (12). On the other hand, MDCK-E1-Cre cells showed a

decrease in the production of ∆E1. A slight reduction of 1.6 ± 0.5 fold was observed in

cells without selective pressure at P10, in which productivity was then maintained up to

P20. On the other hand, MDCK-E1-Cre under selective pressure showed a consecutive

reduction in ∆E1 production capacity with increasing passages, achieving a 7.1 ± 1.7 fold-

decrease at P20. These observations indicate that production capacity of cells was

hampered by maintaining Cre activity through selective pressure.

Figure 3. Effect of cell culture passage and antibiotic pressure on MDCK-E1-Cre cells specific productivity. As a control, MDCK-E1 cells under same conditions were added to the experiments. Values are shown as average ± standard-deviation (n=3). w/: with antibiotic pressure; w/o: without antibiotic pressure. * p < 0.05, ** p < 0.005, given by a single factor Anova analysis against the corresponding P0 values.

Chapter IV

102

3.2. Establishing the best conditions for HD CAV-2 production

To exclude any cell passage effect, the following experiments were performed is MDCK-

E1-Cre and MDCK-E1 cells at P0.

3.2.1. HDV /HV MOI ratio

To establish optimal conditions to produce HD CAVGFP (HDV) (maximum productivity

and lowest HV contamination), production assays under different MOI ratios (5 to 1, 5 to

0.5, and 5 to 0.1 IP) were performed. MOI ratio of 5 to 5 was initially tested, but critically

comprised cell viability and production of either HDV or HV (data not shown).

Similar HDV specific productivities of approximately 300 IP/cell were obtained using the

different MOI ratios tested (Figure 4A). Even with a MOI as low as 0.1, HDV productivity

was still maintained. When using MDCK-E1-Cre cells, HV contamination levels were

similar in the three co-infections (2-3%) (Figure 4B).

To further understand how Cre recombinase impacts HV propagation in the different MOI

ratios, the assays were also performed in MDCK-E1 cells (Figure 4A, B). The results show

a similar HDV production in both cells (Figure 4A), however a clear proportion between

HV MOI and its contaminant levels was observed (Figure 4B). While at MOI 1 of HV a

high contamination was observed (16.3%), a decrease in HV contaminant to 2.0% was

possible when decreasing MOI of this vector. Moreover, contaminant levels of HV with

MDCK-E1 cells in 5HDV : 0.1HV co-infections were similar to those obtained with MDCK-

E1-Cre cells. These results indicate that co-infection with the lowest HV MOI tested

herein, i.e. 0.1 of HV was sufficient to minimize contamination during HDV production.

3.2.2. The impact of MOI ratio in co-infections

Given that HDV production requires co-infection, the impact of co-infection conditions on

adenovirus propagation and HV contaminant levels were further analyzed. Since that

HDV production yields are low, in which cell homeostasis is highly impaired (25), we

reasoned that the effect of co-infection and vectors competition for amplification would

be much more evident if using a CAV-2 vector that typically holds high propagation levels.

Therefore, infections assays under different MOI ratios were reproduced by replacing

HDV for ∆E1 CAV-2 vectors. Similarly to HDV assays, cells were co-infected with ∆E1

(MOI 5) and HV at MOI 5, MOI 1 or MOI 0.5 using MDCK-E1 and MDCK-E1-Cre cell-lines.

Implications of Cre activity and co-infection on canine helper-dependent vector production

103

The production of ∆E1 vector increased when HV MOI was decreased for both cell lines

(Figure 4C). However, MDCK-E1-Cre cells showed lower capacity to produce CAVGFP

than MDCK-E1 cells, especially under high virus input. Similarly to the assays with HDV,

the proportion of HV was minimal in MDCK-E1-Cre cells, and highly dependent on the

MOI when MDCK-E1 cells were used (Figure 4D). However, in each infection condition

assayed, a constant fold-reduction (5 to 6 fold) in HV contamination levels was observed

when comparing MDCK-E1 to MDCK-E1-Cre cells. These results corroborate that the MOI

ratio used to infect cells determines the proportion of each vector in the final production

pool, i.e which vector and its propagation is favored.

Figure 4. Effect of MOI ratio on virus production and HV contaminant using MDCK-E1-Cre and MDCK-E1 cells. (A, B) Cell specific productivity of HDV (A) and resulting HV contaminant (B). (C, D) Cell specific productivity of ∆E1 (C) and resulting HV contaminant (D). Values are shown as average ± standard-deviation (n=3). * p < 0.05, ** p < 0.005, given by a single factor Anova analysis.

Chapter IV

104

3.2.3. The role of Cre recombinase and MOI on the propagation of HV

To gather a better understanding of the factor behind a low HV contamination in HDV

production, the role of Cre recombinase and MOI were evaluated from an HV propagation

point-of-view. To understand the relation between Cre recombinase and HV propagation

under different MOIs, infections with HV using MOI 5, 1 and 0.5 were assayed in MDCK-

E1-Cre and compared to those obtained with MDCK-E1 cells (Figure 5). A decrease in HV

production was observed in MDCK-E1-Cre cells in comparison to MDCK-E1 cells due to

excision of the packaging single by Cre activity. Cell specific productivity was

proportional to the MOI used in both cell lines. Furthermore, the differences in IP/cell of

MDCK-E1-Cre versus MDCK-E1 cells in the different MOIs tested were shown to be

similar, following on average a fold-reduction of 145 ± 23. These observations indicate

that HVs escaping from excision were proportional to the MOI used.

Figure 5. Effect of MOI on HV production in MDCK-E1-Cre and MDCK-E1 cells. Values are shown as

average ± standard-deviation (n=3).

3.2.4. The relation between viral particles infectivity, Cre and packaging

Physical (genome-containing) particles of HDV, ∆E1 and HV were quantified and

normalized to the infectious titer to analyze the infectivity in the different conditions

assayed.

The results show a higher PP:IP ratio with HDVs than ∆E1 (Figure 6), as previously

described (25). When using MDCK-E1 as producer cells, the PP:IP ratios of HDV and ∆E1

were similar along the different co-infections (Figure 6A,B). However, PP:IP ratios

obtained with MDCK-E1-Cre cells were consistently higher than those obtained with

Implications of Cre activity and co-infection on canine helper-dependent vector production

105

MDCK-E1 cells. In addition, the highest PP:IP ratio for HDV and ∆E1 was obtained with

MDCK-E1-Cre cells under high MOI. Together, these observations show that MDCK-E1-

Cre cells generate more non-infectious particles, which is further aggravated by the use of

a high MOI of virus.

When evaluating HV, no significant differences in PP:IP ratios were observed between the

different MOIs or cell lines used, although a slight tendency to obtain more non-infectious

particles of HV in co-infections with ∆E1 was also obtained with MDCK-E1-Cre cells

(Table 2).

Figure 6. Comparison of physical to infectious particles ratio for HDV (A) and ∆E1 (B) obtained in MDCK-E1 and MDCK-E1-Cre cells in the different co-infections. Values are shown as average ± standard-deviation (n=3). * p < 0.05, given by a single factor Anova analysis.

Table 2. Physical to infectious particles ratio of HV obtained with MDCK-E1-Cre and MDCK-E1 cells under the different co-infections

Co-infection with

Cells MOI of HV

5 HV 1HV 0.5 HV 0.1HV

HDV MDCK-E1-Cre n.d. 86 ± 12 93 ± 17 85 ± 16

MDCK-E1 n.d. 75 ± 18 102 ± 33 93 ± 16

∆E1 MDCK-E1-Cre 58 ± 8 50 ± 9 42 ± 3 n.d.

MDCK-E1 37 ± 19 38 ± 15 37 ± 7 n.d.

Values are shown as average ± standard-deviation (n=3); No statistical mean (p value < 0.05) was obtained when comparing values of MDCK-E1-Cre with MDCK-E1 cells under same co-infection conditions. n.d.: not determined.

Chapter IV

106

4. Discussion

While helper-dependent vectors (HDVs) continue to demonstrate a high therapeutic

potential, the production standardization of these vectors is still limiting their wide

availability in gene delivery assays and trials. In this study, we evaluated Cre-expressing

cells stability, infection conditions and their relation to adenovirus amplification and

helper vector (HV) contamination in serum-free medium conditions to establish a

reproducible production process for HD canine adenovirus type 2 (CAV-2) vectors. We

found that constitutive Cre expression affects the long-term capacity of cells to produce

adenoviruses, and co-infection conditions using low HV MOI can effectively reduce HV

contamination even when using cells without Cre recombinase.

HDV producer cells need to be in culture for longer periods of time as cells are needed for

serial amplifications until sufficient vectors are generated (2, 26-28). On the other hand,

the expression of Cre recombinase, due to its toxicity (7), is likely to impair cell line

performance. Therefore, the evaluation of producer cell line stability in the scope of HDV

production becomes critical. The consistent MDCK-E1-Cre cells growth profile under

increasing subculture passages (Figure 1) indicates that Cre expression was supported

by the cell line, probably because of non-toxic levels and/or the cells capacity to correct

Cre-induced DNA double-stranded breaks (DSBs) (7). However, when evaluating virus

production, a consecutive reduction in ΔE1 vectors (CAVGFP) productivity along culture

passages was observed (Figure 3), namely in the cell lines forced to express Cre

recombinase through selective pressure from P10 to P20 (Figure 2). MDCK-E1-Cre cells

in Optipro SFM medium can be thus used for virus production during 10 culture passages

after thawing. The factor(s) and/or mechanism(s) behind these observations must be

such that despite impacting virus replication, cell homeostasis is balanced enough to

sustain growth under the presence of Cre. Furthermore, any effect of E1 expression levels

on CAV-2 production should be ruled out, since E1 levels between MDCK-E1-Cre and

MDCK-E1 cells in same conditions (passage number and antibiotic pressure) were found

to be similar (data not shown). The 116 cell line, one of the several Cre-expressing cell

lines derived from HEK 293 (29) and established by Palmer and Ng (27), was able to keep

adenovirus productivities up to 86 passages when maintained under selective pressure.

While these results differ from ours, the evaluation of other Cre-expressing cells

established for HDV production(29) should be pursued to understand how host cell

Implications of Cre activity and co-infection on canine helper-dependent vector production

107

and/or cell clone can also contribute to the impact that Cre has on the resulting cell

performance.

The best infection conditions for HDV production must be well established as the MOI

ratio of HDV/HV impacts the contamination levels of HV in co-infections. Similarly to

what was shown with human HDV production (30), the best strategy to produce HD CAV-

2 was under co-infections of 5 HDV : 0.1 HV, where maximum HDV productivity were

ensured and HV contamination the lowest (Figure 4). Indeed, a low HV MOI can benefit

HDV production by reducing the levels of excised helper DNA molecules after Cre

recombinase activity, that are known to contribute to recombination between viral

vectors (31), and circumventing the need to develop alternative strategies to attain

higher levels of Cre than those supported by the cell line (6). The relation between MOI

ratio and HV contamination levels, namely that obtained in co-infections using MDCK-E1

cells (Figure 4B,D), indicates that MOI ratio is an effective strategy to unbalance the

competition for amplification of HV in relation to HDV or ∆E1. In fact, this led to

contamination levels as low as those obtained with MDCK-E1-Cre cells when producing

HDV (Figure 4B), raising the possibility of producing HDV without the need of a

recombinase when optimal MOI ratios are used. Therefore, as soon as HDV titers permit

the establishment of such MOI ratio (usually after rescue step and 1st amplification), the

subsequent amplifications can be moved to cell lines without Cre. This would lead to

better vector yields, surpassing the negative effects that Cre has on cell performance (7),

recombination between vectors genomes (31) and virus amplification (Figure 3).

The analysis of PP:IP ratios also indicated that more non-infectious particles were

generated when using MDCK-E1-Cre cells as producer cells, especially when co-infections

were performed with high MOI of HV (Figure 6). Although the differences in PP:IP ratio

between MDCK-E1 and MDCK-E1-Cre cells were rather low (from 1.2 to 1.6-fold

differences), they were statistically significant in some cases and reproducible in the

different experiments. More importantly, given that the PP:IP ratio of clinical grade

adenovirus vector is limited by the Food and Drug Administration, special attention must

be paid to this. These findings substantiate the need to minimize the use of Cre

recombinase (expressing cells) and lower HV MOI to maximize HDV quality. Following

these observations, cell physiological state imposed by the expression of Cre

recombinase, probably aggravated under high HV MOI, may be compromising the success

of virus replication cycle and virus particle maturation process. The relation between Cre

Chapter IV

108

activity, infection conditions and virus maturation should be thus further clarified to

understand the mechanism behind the generation of less infectious particles.

Apart of generating more non-infectious particles (Figure 6, Table 2) and reducing virus

production at high passage number (Figure 3), MDCK-E1-Cre cells also impacted CAV-2

production when co-infected with HV. In fact, when comparing the specific productivity

of MDCK-E1 and MDCK-E1-Cre cells in ∆E1 + HV co-infections, a reduction in ∆E1

amplification was observed with MDCK-E1-Cre cells, specially under high HV MOI

(Figure 4C). This feature is specific to co-infections with HV in MDCK-E1-Cre cells as this

was not observed after infecting MDCK-E1-Cre and MDCK-E1 with increasing MOIs (from

5 to 40) of ∆E1 alone (Figure 3 and data not shown). Since the difference between co-

infections with MDCK-E1-Cre and MDCK-E1 cells is the occurrence of HV genomes

excision, these observations substantiate a relation between high number of excised HV

genomes and inhibition of ∆E1 amplification.

To support Cre recombinase activity, cells must present a consistent DSBs repair system.

Such mechanism is involved in the inhibition of adenovirus replication: viral genomes are

sensed as DSBs activating DNA damage response and the repair mechanism concatenate

viral genomes making them unavailable for the subsequent replication process (32).

Adenoviral E4 11k (E4orf3) or the complex of E4 34k (E4orf6) and E1B proteins

counteract and are sufficient to inhibit DSBs repair system, thereby preventing genome

concatenation (33-36). Given the link of Cre – DSBs repair – virus amplification, we

hypothesize that the reduction in CAV-2 production could be due to an up regulation of

DSBs repair mechanism. Assuming that Cre-expressing cells should have an up regulated

DSBs repair system, MDCK-E1-Cre cells are likely to be more prone in sensing viral DNA

as DSBs than MDCK-E1 cells. If MDCK-E1-Cre cells are challenged with high MOIs of HV,

more DNA breaks are exposed as more viral genomes are being excised, which might

result in a higher activation of DSBs repair system, explaining the inhibition of ∆E1

propagation under high HV MOIs (Figure 4C). Accordingly, MDCK-E1-Cre cells with

increased DSBs repair system, being more prone to repair Cre-induced damages, might

dominate cell population during culture passages due to proliferative advantage.

Therefore, the DSBs repair system of cells maintained in culture for long periods would

be consistently more active and sensitive to viral genomes, which might explain the

subsequent reduction of ∆E1 production in MDCK-E1-Cre cells along passages (Figure 3).

A model where DSB repair system interfere with the typical adenovirus propagation must

Implications of Cre activity and co-infection on canine helper-dependent vector production

109

fit with a scenario which E4 levels attained during infection are no longer enough to

inhibit this mechanism (33-35). In addition, the occurrence of genome concatenation

should be observed to confirm these assumptions (32).

In conclusion, despite similar growth profile of MDCK-E1-Cre cells with increasing

passages, the long-term Cre recombinase expression reduced their capacity to produce

adenoviruses. Not only that, but more non-infectious particles were generated with these

cells (up to 1.6 fold-increase) in HV co-infections, especially under high MOIs. The best

conditions to produce HDV followed a high HDV/HV MOI ratio with a HV MOI as low as

0.1. This favored HDV propagation and infectivity and limited the generation of

contaminant HV. In fact, such MOI ratio was capable of attaining low HV contamination

levels, with or without the activity of Cre, raising the possibility of producing HDVs

without Cre-expressing cells when optimal infection conditions are used. This work

unveils the impact of the Cre recombinase system and MOI ratio on adenovirus

propagation, highlighting important bioprocess parameters that must be considered to

design an improved manufacturing of HDVs.

5. Acknowledgments and author contribution

We thank Sandy Ibanes and Aurelie Gennetier for the initial preparations of CAV-2

vectors. This work was supported by Fundação para a Ciência e a Tecnologia (FCT) –

Portugal, through the project PTDC/EBB-BIO/118615/2010, and European Commission

through FP7 project BrainCAV (222992). Paulo Fernandes acknowledges FCT for his

Ph.D. grant (SFRH/BD/70810/2010). Paulo Fernandes conceived the experimental setup

and design, performed the experiments, analyzed the data and wrote the chapter.

6. References

1. Dormond, E., M. Perrier, and A. Kamen. 2009. From the first to the third generation adenoviral vector: what parameters are governing the production yield? Biotechnol Adv 27:133-144.

2. Parks, R. J., L. Chen, M. Anton, U. Sankar, M. A. Rudnicki, and F. L. Graham. 1996. A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci U S A 93:13565-13570.

3. Lieber, A., C. Y. He, I. Kirillova, and M. A. Kay. 1996. Recombinant adenoviruses with large deletions generated by Cre-mediated excision exhibit different biological properties compared with first-generation vectors in vitro and in vivo. J Virol 70:8944-8960.

4. Hardy, S., M. Kitamura, T. Harris-Stansil, Y. Dai, and M. L. Phipps. 1997. Construction of adenovirus vectors through Cre-lox recombination. J Virol 71:1842-1849.

Chapter IV

110

5. Ng, P., C. Evelegh, D. Cummings, and F. L. Graham. 2002. Cre levels limit packaging signal excision efficiency in the Cre/loxP helper-dependent adenoviral vector system. J Virol 76:4181-4189.

6. Gonzalez-Aparicio, M., I. Mauleon, P. Alzuguren, M. Bunuales, G. Gonzalez-Aseguinolaza, C. San Martin, J. Prieto, and R. Hernandez-Alcoceba. 2011. Self-inactivating helper virus for the production of high-capacity adenoviral vectors. Gene Ther 18:1025-1033.

7. Loonstra, A., M. Vooijs, H. B. Beverloo, B. A. Allak, E. van Drunen, R. Kanaar, A. Berns, and J. Jonkers. 2001. Growth inhibition and DNA damage induced by Cre recombinase in mammalian cells. Proc Natl Acad Sci U S A 98:9209-9214.

8. Thyagarajan, B., M. J. Guimaraes, A. C. Groth, and M. P. Calos. 2000. Mammalian genomes contain active recombinase recognition sites. Gene 244:47-54.

9. Silver, D. P., and D. M. Livingston. 2001. Self-excising retroviral vectors encoding the Cre recombinase overcome Cre-mediated cellular toxicity. Molecular cell 8:233-243.

10. Pfeifer, A., E. P. Brandon, N. Kootstra, F. H. Gage, and I. M. Verma. 2001. Delivery of the Cre recombinase by a self-deleting lentiviral vector: efficient gene targeting in vivo. Proc Natl Acad Sci U S A 98:11450-11455.

11. Baba, Y., M. Nakano, Y. Yamada, I. Saito, and Y. Kanegae. 2005. Practical range of effective dose for Cre recombinase-expressing recombinant adenovirus without cell toxicity in mammalian cells. Microbiol Immunol 49:559-570.

12. Fernandes, P., V. M. Santiago, A. F. Rodrigues, H. Tomas, E. J. Kremer, P. M. Alves, and A. S. Coroadinha. 2013. Impact of E1 and Cre on adenovirus vector amplification: developing MDCK CAV-2-E1 and E1-Cre transcomplementing cell lines. PLoS One 8:e60342.

13. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

14. Perreau, M., F. Mennechet, N. Serratrice, J. N. Glasgow, D. T. Curiel, H. Wodrich, and E. J. Kremer. 2007. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: implications for clinical efficacy. J Virol 81:3272-3284.

15. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

16. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

17. Hnasko, T. S., F. A. Perez, A. D. Scouras, E. A. Stoll, S. D. Gale, S. Luquet, P. E. Phillips, E. J. Kremer, and R. D. Palmiter. 2006. Cre recombinase-mediated restoration of nigrostriatal dopamine in dopamine-deficient mice reverses hypophagia and bradykinesia. Proc Natl Acad Sci U S A 103:8858-8863.

18. Pivetta, C., M. S. Esposito, M. Sigrist, and S. Arber. 2014. Motor-circuit communication matrix from spinal cord to brainstem neurons revealed by developmental origin. Cell 156:537-548.

19. Ekstrand, M. I., A. R. Nectow, Z. A. Knight, K. N. Latcha, L. E. Pomeranz, and J. M. Friedman. 2014. Molecular profiling of neurons based on connectivity. Cell 157:1230-1242.

20. Ariza, L., L. Gimenez-Llort, A. Cubizolle, G. Pages, B. Garcia-Lareu, N. Serratrice, D. Cots, R. Thwaite, M. Chillon, E. J. Kremer, and A. Bosch. 2014. Central Nervous System Delivery of Helper-Dependent Canine Adenovirus Corrects Neuropathology and Behavior in Mucopolysaccharidosis Type VII Mice. Hum Gene Ther 25:199-211.

21. Cubizolle, A., N. Serratrice, N. Skander, M. A. Colle, S. Ibanes, A. Gennetier, N. Bayo-Puxan, K. Mazouni, F. Mennechet, B. Joussemet, Y. Cherel, Y. Lajat, C. Vite, F. Bernex, V. Kalatzis, M. E. Haskins, and E. J. Kremer. 2014. Corrective GUSB Transfer to the Canine Mucopolysaccharidosis VII Brain. Mol Ther 22:762-773.

Implications of Cre activity and co-infection on canine helper-dependent vector production

111

22. Serratrice, N., A. Cubizolle, S. Ibanes, N. Mestre-Frances, N. Bayo-Puxan, S. Creyssels, A. Gennetier, F. Bernex, J. M. Verdier, M. E. Haskins, G. Couderc, F. Malecaze, V. Kalatzis, and E. J. Kremer. 2014. Corrective GUSB transfer to the canine mucopolysaccharidosis VII cornea using a helper-dependent canine adenovirus vector. Journal of controlled release : official journal of the Controlled Release Society 181:22-31.

23. Fernandes, P., C. Peixoto, V. M. Santiago, E. J. Kremer, A. S. Coroadinha, and P. M. Alves. 2013. Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20:353-360.

24. Soudais, C., S. Boutin, and E. J. Kremer. 2001. Characterization of cis-acting sequences involved in canine adenovirus packaging. Mol Ther 3:631-640.

25. Fernandes, P., D. Simão, M. R. Guerreiro, E. J. Kremer, A. S. Coroadinha, and P. M. Alves. 2014. The impact of Adenovirus life cycle on the generation of canine helper-dependent vectors. Gene Ther in press.

26. Dormond, E., A. Meneses-Acosta, D. Jacob, Y. Durocher, R. Gilbert, M. Perrier, and A. Kamen. 2009. An efficient and scalable process for helper-dependent adenoviral vector production using polyethylenimine-adenofection. Biotechnol Bioeng 102:800-810.

27. Palmer, D., and P. Ng. 2003. Improved system for helper-dependent adenoviral vector production. Mol Ther 8:846-852.

28. Suzuki, M., R. Cela, C. Clarke, T. K. Bertin, S. Mourino, and B. Lee. 2010. Large-scale production of high-quality helper-dependent adenoviral vectors using adherent cells in cell factories. Hum Gene Ther 21:120-126.

29. Kovesdi, I., and S. J. Hedley. 2010. Adenoviral producer cells. Viruses 2:1681-1703. 30. Dormond, E., M. Perrier, and A. Kamen. 2009. Identification of critical infection

parameters to control helper-dependent adenoviral vector production. J Biotechnol 142:142-150.

31. Ahn, M., A. Gamble, S. R. Witting, J. Magrisso, S. Surendran, S. Obici, and N. Morral. 2013. Vector and helper genome rearrangements occur during production of helper-dependent adenoviral vectors. Human gene therapy methods 24:1-10.

32. Weiden, M. D., and H. S. Ginsberg. 1994. Deletion of the E4 region of the genome produces adenovirus DNA concatemers. Proc Natl Acad Sci U S A 91:153-157.

33. Boyer, J., K. Rohleder, and G. Ketner. 1999. Adenovirus E4 34k and E4 11k inhibit double strand break repair and are physically associated with the cellular DNA-dependent protein kinase. Virology 263:307-312.

34. Stracker, T. H., C. T. Carson, and M. D. Weitzman. 2002. Adenovirus oncoproteins inactivate the Mre11-Rad50-NBS1 DNA repair complex. Nature 418:348-352.

35. Araujo, F. D., T. H. Stracker, C. T. Carson, D. V. Lee, and M. D. Weitzman. 2005. Adenovirus type 5 E4orf3 protein targets the Mre11 complex to cytoplasmic aggresomes. J Virol 79:11382-11391.

36. Baker, A., K. J. Rohleder, L. A. Hanakahi, and G. Ketner. 2007. Adenovirus E4 34k and E1b 55k oncoproteins target host DNA ligase IV for proteasomal degradation. J Virol 81:7034-7040.

Chapter IV

112

Chapter V

The impact of adenovirus life cycle on the

generation of canine helper-dependent

vectors

This chapter is adapted from:

Fernandes P, Simão D, Guerreiro MR, Kremer EJ, Coroadinha AS, Alves PM (2015). Impact

of adenovirus life cycle on the generation of canine helper-dependent vectors. Gen Ther.

Jan;22(11):40-9

Chapter V

114

Abstract

Helper-dependent adenovirus vectors (HDVs) are safe and efficient tools for gene

transfer with high cloning capacity. However, the multiple amplification steps needed to

produce HDV hamper a robust production process and in turn the availability of high

quality vectors. To understand the factors behind the low productivity, we analyzed the

progression of HDVs life cycle. Canine adenovirus type 2 (CAV-2) vectors, holding

attractive features to overcome immunogenic concerns and treat neurobiological

disorders, were the focus of this work.

When compared to E1-deleted (∆E1) vectors, we found a faster helper genome

replication during HDV production. This was consistent with an upregulation of the

adenovirus polymerase (Adpol) and pre-terminal protein (pTP) and led to higher and

earlier expression of structural proteins. While genome packaging occurred similarly to

∆E1 vectors, more immature capsids were obtained during HDV production, which led to

a ~4-fold increase in physical to infectious particles ratio. The higher viral protein

content in HDV-producing cells was also consistent with an increased activation of

autophagy and cell death, in which earlier cell death compromised volumetric

productivity. The increased empty capsids and earlier cell death found in HDV production

may partially contribute to the lower vector infectivity. However, an HDV specific factor

responsible for a defective maturation process should be also involved to fully explain the

low infectious titers. This study showed how a deregulated adenovirus cycle progression

affected cell line homeostasis and HDV propagation, highlighting the impact of vector

genome design on virus-cell interaction.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

115

Contents

1. Introduction .............................................................................................................................................116

2. Materials and Methods ........................................................................................................................118

2.1. Cell lines and culture medium .......................................................................................................118

2.2. Vector .......................................................................................................................................................118

2.3. Gene expression analysis ................................................................................................................118

2.4. Viral vectors amplification studies .............................................................................................119

2.5. Infectious vectors titration .............................................................................................................119

2.6. Viral genomes and physical particles by real-time PCR ....................................................119

2.7. Quantification of assembled capsids ..........................................................................................119

2.8. Cell death analysis ..............................................................................................................................120

2.9. Western blot analysis ........................................................................................................................121

2.10. Nuclei extracts ...................................................................................................................................121

3. Results .........................................................................................................................................................122

3.1. Volumetric productivity and viral particles infectivity are impaired in HDV

production ......................................................................................................................................................123

3.2. Cell binding and trafficking into the nucleus does not affect HDV production .......124

3.3. Replication of vector genome increases in HDV-producing cells ..................................125

3.4. Expression of genes implicated in viral DNA replication and late phase is observed

earlier in HDV-producing cells ..............................................................................................................126

3.5. Despite more assembled particles during HDV production, packaging occurs

similarly to ∆E1 control ............................................................................................................................128

3.6. Low cell viability during HDV production is linked to activation of autophagy and

high levels of viral proteins affecting virus infectivity ...............................................................129

4. Discussion..................................................................................................................................................132

5. Acknowledgements and author contribution ...........................................................................135

6. References .................................................................................................................................................136

Chapter V

116

1. Introduction

Adenoviruses (Ads) are non enveloped, icosahedral particles containing a linear, double-

stranded DNA genome. Ad vectors have received considerable attention as gene transfer

tools in which human adenovirus serotype 5 (HAd5) is the typical prototype vector

backbone. However, the hurdles facing the clinical use of human adenoviral vectors,

including innate and pre-existing immunity, may limit the efficiency and time of

transgene expression (1). One of the alternatives to circumvent these drawbacks is using

non human Ads, such as canine adenovirus type 2 (CAV-2) vectors (2). In addition to the

paucity of anti-CAV-2 neutralizing antibodies in humans (2, 3), their vectors induce a low

level of innate response and no activation of human complement pathways (4, 5). Their

ability to preferentially transduce neurons, combined with a remarkable capacity of

axonal transport (6, 7), make CAV-2 vectors promising candidates for the treatment of

neurodegenerative diseases (8, 9).

Adenovirus genome has been progressively modified from the wild-type to improve

vector safety and efficacy in therapeutic applications (reviewed elsewhere (10)). Helper-

dependent vectors (HDVs), devoid of viral coding regions, are powerful tools for gene

transfer in vivo. HDVs combine high transduction efficacies, long-term gene expression,

and a cloning capacity of >30 kb. These vectors retain only on their genomes the inverted

terminal repeats, the packaging region (Ψ) and an expression cassette(s). To produce

HDVs, both early and late viral functions need to be complemented in a synchronized

fashion. Standard production methods are based on the co-infection of E1-

transcomplementing cells with HDV and an E1-deleted helper vector (HV), which

provides the viral proteins in trans. During production, both HDV and HV genomes

replicate inside the cells, but the encapsidation of the HV genome is minimized, by

flanking its Ψ with recognition sequences for a recombinase constitutively expressed by

the cells, such as FLP (11, 12) or Cre (13).

Despite their enormous potential, the use of HDVs in preclinical and clinical settings is

limited because of two bottlenecks: contamination with HV and production yields.

Several efforts to reduce HV contamination have been described. Based on alternative

vector designs, deletion of the Ψ in the HV genome was improved through the

development of an inducible self-inactivating vector (vector expressing a recombinase

that can block its own genome packaging) (14). Moreover, tweaking the HV packaging

domain can delay its packaging (15), or reduce its packaging efficiency in co-infection

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

117

contexts (16, 17). Reduced HDV production obtained after the initial transfection step,

limited by transfection efficiency and initiation of viral DNA replication from plasmid

substrates, has also prompted others to improve the strategies. With a focus on this step,

production of HDV was improved by creating cell lines expressing pre-terminal protein

(pTP) and adenoviral DNA polymerase (Adpol) (18), optimizing transfection methods

compliant with scalable processes (19), and developing an intracellular system to release

HDV genome from transfected circular plasmids, which increased transfection efficiencies

of ~10-fold (20). However, even using improved conditions, multiple amplification steps

are needed to rescue HDV amplification to its maximum and prepare sufficient quantities

of vector (14, 19, 21, 22). Moreover, most reports show a relatively low infectivity of HDV

in comparison to physical particles (optical density based) titers (14, 23) and, equally

importantly, inconsistent infectious yields obtained in different preparations of the same

HDV (14). Although most studies were conducted with human HDVs, consistently low

infectious titers are also extended to canine HDVs (4, 6, 9).

Ads have a regulated infection cycle where multiple viral and cellular proteins interact to

enable a productive virus replication and generate infectious virus particles (reviewed in

(24)). Therefore, understanding the adenovirus replication steps during the generation of

HDV particles could help break the bottlenecks behind the productivity of these vectors.

After cell binding, the mature viral particle is internalized and transported towards the

nucleus. During this transport, the particle is dismantled by an ordered elimination of

some structural proteins and culminates with the delivery of the viral genome into the

nucleus via nuclear pore complexes (25). Once in the nucleus, early genes are expressed

and viral DNA replication starts. Late phase of infection is then activated and structural

proteins expressed. These proteins are likely assembled into empty virions, followed by

the packaging of viral genome. Finally, viral particles undergo a protease-mediated

maturation process to becomes infectious (26-28).

In the present study, we investigated the steps of Ad cycle that could be affected during

HD CAV-2 production. As CAV-2 vectors use keeps increasing(8, 9), larger amounts of

high quality vectors to conduct preclinical and possibly clinical assays are required.

Therefore, understanding HDV productivity is critical to identify and hopefully overcome

the manufacturing limitations. Here, we provide evidences of a distinct replication cycle

progression during HDV production, which resulted in changes in the cell physiology and

vector productivity.

Chapter V

118

2. Materials and Methods

2.1. Cell lines and culture medium

MDCK (ECCAC, Nr 84121903) and MDCK-E1-Cre cells (29) were grown in Dulbecco’s

Modified Eagle’s Medium (DMEM) (Gibco, Paisley, UK) supplemented with 10% (v/v) FBS

(Gibco). Cells were maintained at 37ºC in a humidified atmosphere air containing 5% CO2.

Cell concentration and viability were determined by the trypan blue exclusion method

using a 0.1% (v/v) solution prepared in PBS and counting cells in a Fuchs-Rosenthal

haemacytometer (Brand, Wertheim, Germany).

2.2. Vector

CAVGFP, JB∆5 and HD CAVGFP are vectors derived from CAV-2 strain Toronto A 26/61,

GenBank J04368. CAVGFP (2) and HD CAVGFP (7) are E1-deleted (ΔE1) and HD vectors,

respectively, and contain an CMV-eGFP expression cassette. JB∆5 is a helper vector

containing loxP flanking the packaging domains and a RSV-lacZ expression cassette (7).

CAVGFP, JB∆5 and HD CAVGFP viral stocks were prepared and purified by CsCl gradients

as described previously (2, 29).

2.3. Gene expression analysis

To assess gene expression, total RNA was extracted from 1 × 106 cells using RNeasy mini

kit (QIAGEN, Germantown, MD, USA). In infected cells, two rounds of RNA cleaning were

performed. For real-time PCR, RPL-22 was chosen as a control gene. Reverse

transcription of total RNA was performed according to Transcriptor high fidelity cDNA

synthesis kit (Roche applied science, Mannheim, Germany) protocol for cDNA synthesis

using 1 μg of total RNA and oligo dT primer for total mRNA reverse transcription (RT).

The reverse transcribed cDNA was aliquoted and stored at -20ºC until further processing.

SYBR Green I dye chemistry was used to detect the PCR products using LightCycler 480

SYBR Green I Master (Roche applied science) according to the manufacturer’s

instructions using LightCycler 480 Real Time PCR System (Roche applied science). cDNA

samples were run with a RT-reaction free control and a sample without cDNA template.

The primers used for each target are shown in Table S1 (Appendix).

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

119

2.4. Viral vectors amplification studies

MDCK-E1-Cre cells were seeded at 1.5 × 104 cells/cm2, infected the day after with

medium exchange. Unless stated otherwise, HD CAVGFP production assays were

performed using MOI 5 and MOI 1 of HD CAVGFP and JB∆5, respectively. As control,

CAVGFP production assays were performed by co-infecting JB∆5 under same conditions.

Production assays using single infection of JB∆5 with MOI 1 were also performed to

isolate the role of this vector on production kinetics. Intracellular samples were collected

using triton (Sigma-Aldrich) 0.1% (v/v) in Tris-HCl, clarified at 3000 g for 10 min at 4ºC

and stored at -85ºC until further analysis.

2.5. Infectious vectors titration

Quantification of infectious HD CAVGFP and CAVGFP was performed by monitoring the

expression of GFP, while titration of infectious JB∆5 vectors was based in lacZ gene

expression and β-galactosidase activity as described previously (29).

2.6. Viral genomes and physical particles by real-time PCR

Viral genomes were extracted and purified by High Pure Viral Nucleic Acid Kit (Roche

Diagnostics, Penzberg, Germany). SYBR Green I dye chemistry was used to detect PCR

products using Light Cycler system. Quantification was based on the detection of GFP (for

HD CAVGFP and CAVGFP) or lacZ (for JB∆5). Previously purified and quantified plasmids

coding for E1-deleted CAV-2 genome, GFP or lacZ were used as standards. To ensure the

removal of free viral genomes from samples and accurately quantify viral physical

particles, a benzonase treatment was performed prior to DNA extraction by adding 2 μl of

benzonase (250 U/μl) (Merck Millipore, Darmstadt, Germany) to the same sample

volume and incubating for 1 h at 37ºC. Benzonase was inhibited using the proteinase K

step of DNA extraction protocol. GFP and lacZ primers are shown in Table S1 (Appendix).

2.7. Quantification of assembled capsids

After confirming that virions and empty capsids were quantifiable through Nanoparticle

Tracking Analysis (NTA), assembled capsids were titrated using LM10 instrument

(NanoSight, Amesbury, UK). Following the manufacturer’s instructions, samples were

diluted with Dulbecco’s phosphate buffered saline (PBS) to reach a particle concentration

Chapter V

120

suitable for analysis with NTA (1.0 × 108 to 2.5 × 109 particles/mL). At least two different

sample dilutions were prepared for each sample and analyzed twice. The NanoSight

LM10 recorded 60 second sample videos which were then analyzed with the NTA 2.0

Analytical software release version build 0125 (NanoSight, Amesbury, UK). The NTA

software is then able to identify and track individual nanoparticles moving under

Brownian motion and the results allow particle size and concentration to be recovered

(30).

2.8. Cell death analysis

Cells were seeded in 24 well plates, infected under the conditions above mentioned and

analysed for necrosis and autophagy at 20 hpi. Necrosis was assessed by staining cells

with propidium iodide (Sigma-Aldrich, St. Louis, MO, USA). After 20 hpi, cells were

collected from wells in duplicates, stained with 2 μl of propidium iodide solution (Sigma-

Aldrich) in 2 mL final volume, incubated in the dark during 10 min and analyzed by flow

cytometry using a CyFlow Space (Partec, Münster, Germany) equipped with a 561 nm

yellow laser and 630 ± 22 nm filter. Non-stained and non-infected controls were also

analyzed. Autophagy was evaluated through the detection of autophagosomes by

immunostaining. Cells grown in coverslips inside wells were fixed in 4% (w/v)

paraformaldehyde + 4% sucrose (w/v) in PBS, permeabilized with 0.1% (v/v) Triton X-

100 solution (Sigma) and processed as described previously (31). Briefly, after blocking

with 0.2% (w/v) fish skin gelatin (FSG), samples were incubated with anti-LC3B antibody

(Sigma-Aldrich), diluted to 1:100 in 0.125% (w/v) FSG + 0.1% (v/v) Triton X-100 in PBS.

Alexa Fluor® 647 Goat Anti-Rabbit IgG (H+L) antibody (Invitrogen, Carlsbad, CA, USA)

was used as secondary antibody diluted to 1:500. Cell nuclei were counter stained with

ProLong Gold antifade reagent with DAPI (Invitrogen). Samples were visualized using

fluorescence (DMI6000, Leica) microscopy. Images were processed using the open source

ImageJ software (Rasband, W.S., ImageJ, U. S. National Institutes of Health, Bethesda,

Maryland, USA, http://imagej.nih.gov/ij/, 1997-2012.). Autophagy-positive cells were

quantified by counting the number of cells presenting autophagosomes in relation to the

number of stained nuclei. Quantifications were performed in two biological replicates,

using at least three different sections per replicate.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

121

2.9. Western blot analysis

Cell protein extracts were obtained from 1 × 106 cells using 500 μl of mammalian protein

extraction reagent M-PER (Thermoscientific, Rockford, IL, USA) according to

manufacturer’s instructions. Equal volumes of sample were subjected to SDS-PAGE gel

according to NuPAGE® SDS-PAGE gel system (Invitrogen), and transfered to Hybond-C

extra membranes (GE Healthcare, Uppsala, Sweden) using Trans-blot turbo transfer

system (Bio-rad, Hercules, CA, USA). Membranes were blocked with 5% (w/v) dry milk in

tris-buffered saline-Tween 20, and probed with polyclonal rabbit anti-CAV-2 primary

antibody (32) and anti-β actin abcam8226 (housekeeping control). Both antibodies were

diluted to 1:1000. Western blot membrane detection was performed using HPR-linked

antibodies and Amersham ECL reagents and protocols.

2.10. Nuclei extracts

Cells at 80% confluence in four 150 cm2 T-flasks were infected and harvested 8 hpi in

cold PBS using a cell scraper. The extract from nuclei were prepared by adapting the

protocol provided in CelLytic NuCLEAR extraction kit (Sigma-Aldrich). Briefly, cells were

centrifuged and suspended in Lysis buffer (5 mM Tris HCl, pH 7.5 with 1 mM MgCl2, 1.5

mM CaCl2 and 1 mM DTT), then cells were let to swell on ice for 15 min and disrupted

using a syringe with a narrow-gauge needle (No. 26). After, the suspension was

centrifuged during 20 min at 11 000 g, and the crude nuclei pellet suspended in

extraction buffer (20 mM HEPES pH 7.9, with 1.5 mM MgCl2 and 0.42 M NaCl, 0.2 mM

EDTA, 1 mM DTT and 25% (v/v) Glycerol). Finally, 0.1 % (v/v) of Triton (Sigma-Aldrich)

in Tris HCl was added, the suspended nuclei was agitated for 30 min and centrifuged for

3000 g during 10 min at 4 ºC. Extracts were stored at -85ºC. Viral genomes were isolated

from nuclei extracts using the High Pure Viral Nucleic Acid Kit (Roche) and quantified as

stated above.

Chapter V

122

3. Results

We initially asked whether the limitations in helper-dependent vectors (HDVs)

production were related to differences in adenovirus propagation kinetics. To address

this possibility, HD CAVGFP production was characterized and the main steps of

adenovirus life cycle were evaluated by comparing it to CAVGFP (an E1-deleted vector)

and JB∆5 (a helper vector containing a loxP flanked packaging domain). Figure 1 depicts

the experimental design used in this study (Figure 1A) and the relevant characteristics of

each vector (Figure 1B). CAVGFP (herein designated as ∆E1), harbouring the same eGFP

expression cassette as HD CAVGFP (herein designated as HDV), was used as the control.

Moreover, considering that to produce HDV, cells are co-infected with helper vector (HV),

co-infection with HV JB∆5 was also performed in the ∆E1 production control assays.

Finally, the propagation of HV alone was also performed. Thus, three

infections/production scenarios in Cre-expressing cells were analyzed: i) HDV co-infected

with HV (HDV + HV), ii) ∆E1 co-infected with HV (∆E1 + HV) and iii) HV.

Figure 1. (A) Experimental design of the infections performed in this work. (B) Viral vector genome construct that should be considered in each infection scheme. The amplification of HDV was analyzed by comparison to ∆E1 and HV, and three infection/production assays were performed: HDV, ∆E1 (which share same expression cassette than HDV), and HV. The production of HDV requires co-infection of HDV and HV. As a control of co-infection, ∆E1 assays were performed by co-infecting producer cells with HV. To isolate the role of HV, the third group of production assays was performed by infecting producer cells with only HV. HDV or ∆E1 production assays mentioned throughout the text correspond to HDV + HV or ∆E1 + HV co-infections, respectively. MDCK-E1-Cre cells were used as producer cells and infected using MOI 5 of HDV and ∆E1 and MOI 1 of HV. Ψ – Packaging sequence.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

123

3.1. Volumetric productivity and viral particles infectivity are impaired in HDV

production

To determine the productivities of HDV, production assays with HDV+HV, ∆E1+HV and

HV alone were performed (Figure 1A). Vectors were harvested 48 hpi and quantified for

physical (genome-containing capsids) particles (PP) and infectious particles (IP) (Figure

2). We found a similar cell-specific production of PP for both HDV and ∆E1 vectors

(Figure 2A). However, fewer infectious HDV particles were detected. Concomitantly,

similar PP/cell of HV was obtained in the three infection schemes, but at a lower number

of IP in HDV+HV-producing profile (Figures 1A-i and 2B). From 24 hpi onwards the

viability of HDV+HV-producing cells was half of that obtained for both control-infections

(Figure 2C), which likely affected the volumetric productivity (Figure 2D).

Figure 2. Characterization of HDV production. (A) Cell specific productivity of physical (genome-containing) and infectious HDV and ∆E1 vectors and the physical-to-infectious particles ratio (PP:IP). * p = 0.007, given by a single factor Anova analysis against the corresponding IP values of ∆E1 vectors (B) Cell specific productivity of HV and the physical-to-infectious particles ratio (PP:IP) in the three infection schemes. * p = 0.03, ** p = 0.01, given by a single factor Anova analysis against the corresponding IP values of HV in ∆E1+HV and HV infections, respectively (C) Changes in viable cell concentration of HDV-, ∆E1- and HV-producing cells over time. Viable cell concentration was compared by calculating the fold-change in viable cell number between HDV+HV and ∆E1+HV or HV infections. * p < 0.05, given by a single factor Anova analysis against the corresponding cell concentration values of ∆E1+HV and HV infections (D) Volumetric productivity (Physical particles (PP)/mL) obtained 48 hpi for each viral vector in the three infections. Values are shown as average ± standard-deviation (n=3).

Chapter V

124

3.2. Cell binding and trafficking into the nucleus does not affect HDV production

As mentioned above, there are several steps in vector amplification. To address the

capacity of binding, trafficking and genome delivery of HDV, the progression of infection

was monitored in extracellular and intracellular fractions. The decrease of extracellular

vector concentration (i.e. culture medium) typically occurs during the first 24 hpi (33)

and can be used to quantify binding and/or internalization. In parallel, intracellular

genome accumulation can be accurately measured until the vector replication starts at

approximately 8 hpi.

To evaluate the extracellular fraction, infectious particles during HDV+HV production

were quantified from culture medium at 0, 12 and 24 hpi and the profiles were compared

to ∆E1+HV production. We found that the profiles of HDV and ∆E1 vectors were similar

(Figure 3A), which suggests that the ability to bind cells was maintained in HDV.

The progression from binding to internalization and trafficking was evaluated by

quantifying viral genomes from intracellular fractions collected at 3, 6 and 9 hpi in the

three infection scenarios (Figure 1A). To analyze vector genomes reaching nucleus,

nuclear extracts were collected 8 hpi. We found that the intracellular accumulation of

vector genomes was similar for HDV and ∆E1 at the three time points (Figure 3B). Also,

we found similar levels of HDV and ∆E1 viral genomes in nuclear extracts (Figure 3C),

indicating that these steps were not affected during HDV+HV production. To complement

these assays, the intracellular accumulation of HV genomes was also evaluated. We found

similar intracellular HV genomes were obtained at 3 and 6 hpi in all production schemes.

However, a slight increase in HV genomes in HDV+HV-producing cells compared to

∆E1+HV and HV productions was observed at 9 hpi (Figure 3D). Because vector genome

replication starts at ~8 hpi, this observation might be connected to differences in HV

genome replication in HDV+HV production scenario, rather than to the capacity to infect

cells.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

125

3.3. Replication of vector genome increases in HDV-producing cells

After the delivery of viral genomes into the nucleus, transcription of early region gene

products and viral DNA replication are the subsequent steps in adenovirus propagation.

To analyze whether viral DNA replication was altered in HDV+HV-producing cells,

intracellular viral genomes were quantified from 12 to 48 hpi (Figure 4). Similar

quantities of vector genomes were obtained in the three vector production scenarios at

48 hpi (Figure 4A,B). However, the quantity of HV genomes in HDV+HV-producing cells

(scenario (i) from Figure 1A) was higher from 12 hpi to 24 hpi, especially at 15 hpi:

increases of a 10 and 71-fold were observed when compared to ∆E1+HV- and HV-

producing cells, respectively (Figure 4B). These data indicated that HV genome

replication occurred faster in HDV+HV-producing cells.

Figure 3. Characterization of HDV infection and viral genomes reaching cell nucleus. (A) Infectious particles in cell supernatant after cell infection. (B) Intracellular genomes of HDV and ∆E1 vectors during infection time. Values were normalized to intracellular viral genomes obtained at 3hpi in control ∆E1 infection. (C) Viral genomes in cell nucleus at 8 hpi. (D) Intracelullar genomes of HV in HDV+HV, ∆E1+HV and HV infections during infection time. Values were normalized to intracellular viral genomes obtained at 3hpi for control HV infection. The legends HDV+HV, ∆E1+HV and HV correspond to infections schemes depicted in Figure 1. Values are shown as average ± standard-deviation (n=2).

Chapter V

126

The transgene expression in terms of mRNA, was also evaluated as an indicator of

transcription capacity since to some extent may represent the immediate-early

transcription. These showed lower levels of GFP and slightly higher levels of β-

galactosidase in HDV+HV-infected cells when compared to ∆E1+HV and HV infections

(Figure S1, Appendix).

3.4. Expression of genes implicated in viral DNA replication and late phase is

observed earlier in HDV-producing cells

While early phase genes are primarily transcribed before genome replication and the

corresponding mRNA is transcribed from the input genomes, late region transcription

becomes robust once DNA replication starts. To analyze whether the expression of viral

genes was altered during HDV production, transcription and translation were analyzed.

Transcription analysis was performed by determining mRNA levels from several regions

to have representatives of the different infection phases. Table S2 (Appendix) describes

the viral genes analyzed and the corresponding infection phase and biological role. Viral

genes to be analyzed were selected according to two main criteria: choose at least two

viral genes from early and late phases, and viral genes involved in processes that

occurred differently during HDV production, namely those involved in DNA replication

(E2 region (34)), and those involved in capsid maturation (protease and pVI (28, 35)). In

addition, IVa2 was analyzed because of its role during mid-late phase in activating late

region expression (36) and later as key player in viral genome packaging (37).

Regardless of the phase, the expression of all viral genes increased from 12 hpi to 48 hpi

(Table S3, Appendix). Notably, we found earlier and higher levels of viral transcripts in

Figure 4. Analysis of viral genomes replication of viral vectors in each infection scheme. (A) HDV and ∆E1 genomes (B) HV genomes in each infection. Values were normalized to intracellular viral genomes obtained at 3hpi for control ∆E1+HV and HV infections. Values are shown as average ± standard-deviation (n=2).

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

127

HDV+HV-producing cells. To better understand the gene expression pattern and the

corresponding percentage of each viral product in the different infections, gene

expression values were further normalized to the overall expression at each time. After

this normalization, the results show the switch of expression pattern from early phase

(12 hpi) to late phase transcription (24 and 48 hpi) (Figure S2, Appendix). Also, at 24

and 48 hpi the expression pattern was similar and thus proportional in the different

infections scenarios. However, an over-representation of E2 (E2B), IVa2 and L3 gene

expression was observed at 12 hpi (Figure 5) in HDV+HV-producing cells, when

compared to ∆E1+HV- and HV-producing cells. This indicates that, in addition to the viral

genes involved in genome replication (Adpol and pTP) (34), late phase activator (IVa2)

(36) and late genes (L3) were also differentially expressed during HDV production,

earlier in infection.

To evaluate translation, western blot analysis was performed using rabbit sera that

detects capsid proteins against cell extracts obtained at 12, 24 and 48 hpi. In accordance

to transcription analysis, the results show that higher production of viral proteins was

obtained earlier during HDV+HV production (Figure 6).

Figure 5. Fold-change in gene expression pattern between HDV+HV and ∆E1+HV (A) and HDV+HV and HV (B) of the different viral genes. Gene expression pattern values, representing the contribution of each viral target to the overall expression, were calculated as the percentage of mRNA level of each viral gene relative to total mRNAs levels obtained at each infection point. Only fold-changes >2 were considered relevant. Values are shown as average ± standard-deviation (n=2).

Chapter V

128

3.5. Despite more assembled particles during HDV production, packaging occurs

similarly to ∆E1 control

Once the translation of late region transcripts is accomplished, the structural proteins are

assembled into capsids and genomes are packaged to generate full virions. However,

when producing adenovirus vectors, two types of viral particles are routinely produced:

assembled genome-containing or empty capsids, which are assembled particles with little

or no viral genome (38). Therefore, to understand the type of particles generated during

HDV production, assembly and packaging were further analyzed.

Assembly was analyzed by quantifying the assembled viral particles obtained 48 hpi

using the NanoSight technology, which measured assembled particles, the sum of empty

capsids and full virions. In addition to a positive correlation between the number of

assembled viral particles and viral proteins expression in each infection (Figure 6), we

found more assembled viral particles in HDV+HV production (Figure 7A).

Genome packaging was evaluated by comparing free viral genomes with total viral

genomes (free genomes plus packaged genomes) at 48 hpi. To do so, viral DNA was

extracted from intracellular samples in duplicate, in which one replicate was subjected to

benzonase treatment prior DNA extraction; the decrease of quantified viral genomes in

the samples treated with benzonase before DNA extraction, in comparison with non-

treated sample, indicates the presence of free viral genomes. We found no significant

changes in the free viral genomes obtained in HDV+HV- and ∆E1+HV-producing cells

(Figure 7B). By comparison, the quantity of HV free viral genomes was similar in the

three infection schemes. More free viral genomes should be obtained for HV than for HDV

Figure 6. Western blot of cell extracts obtained in each infection scheme. Cell extracts were normalized to cell concentration and each lane was loaded with same sample volume. Image corresponds to a single membrane. β-actin blot (loading control) was obtained by cutting a fragment which included the 42 kDa size region from the original membrane, using the bands of SeeBlue® Plus2 Pre-stained Protein Standard (Invitrogen) as reference. After detection, Magic MagicMark™ XP Western Protein Standard (Invitrogen) was used as molecular weight marker. For a matter of clarity, blot is shown cropped.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

129

or ΔE1 vectors, as Cre recombinase specifically impacts HV genome packaging. This

observation was not clear in Figure 7B, probably because of the considerable inter-assay

variability (depicted by the error bars) that disguised the expected differences between

free genomes percentage of HV and HDV or ΔE1 vector.

Together, these results show that despite more viral capsids generated, the percentage of

packaged HDV genomes did not increased. Notably, the total DNA of HDV and ∆E1 were

similar at 48 hpi (Figure 4A).

3.6. Low cell viability during HDV production is linked to activation of autophagy

and high levels of viral proteins affecting virus infectivity

When compared to non-infected cells, a 3.32 ± 0.23 fold-increase in necrotic cells was

obtained at 24 hpi during HDV production, versus 1.74 ± 0.15 and 1.42 ± 0.01 for ∆E1 and

HV, respectively, which is consistent with the cell viability analysis (Figure 2D). To

further understand the cell death during HDV production, we evaluated the effect of HDV

infection on cell viability, the activation of autophagy, an adenovirus-related programmed

cell death (39, 40), and the role of virus cycle progression on producer cells viability and

viral productive outcome.

To determine the effect of infection on cell death, HDV infections were performed in

MDCK-E1-Cre cells and MDCK cells at same MOI (Figure 1A-i), which do not express the

CAV-2 E1 region and are therefore unable to allow productive infectious cycle of ∆E1

CAV-2 vectors. At 48 hpi, MDCK cell viability was maintained, indicating that HDV

Figure 7. Assembled viral particles (A) and free viral genomes (B) obtained in each infection at 48 hpi. Values are shown as average ± standard-deviation (n=2).

Chapter V

130

transduction (binding, trafficking and delivery of vector genome to the nucleus) per se did

not induce cell death (data not shown).

The decay in cell viability (Figure 2C) should be connected to an adenovirus-specific

programmed cell death. Autophagy, a form of programmed cell death induced by

adenoviruses (39, 40), was therefore evaluated via the detection of LC3-incorporated

autophagosomes. One of the major hallmarks of activation of autophagic process is the

formation of cellular autophagosome puncta containing LC3-II. We found an increase in

LC3-II autophagosomes in HDV+HV-producing cells (Figure 8). While HDV+HV

producing cells presented 16 ± 4 % autophagy-positive cells, ∆E1+HV-producing cells

presented 2 ± 1 %, corresponding to an 8-fold difference. HV-producing cells or mock-

infected cells did not present detectable autophagosome-like structures in the six

analyzed sections. Moreover, the number of autophagosomes per autophagy-positive

cells was higher in cells producing HDV+HV (Figure 8). This indicates that autophagy

was more strongly induced during the production of these vectors.

To determine if cell death was due to the viral cycle progression, the intracellular

environment observed during HDV+HV production (high levels of HV genomes and viral

proteins earlier in time) was induced during ∆E1 production by increasing the MOI of HV

(Figure 9). Forty-eight hpi, cell concentration and viral proteins levels were determined.

Consistent with an increase in viral proteins (Figure 9A), we found a decrease in cell

viability when the HDV+HV production scenario was pursued (Figure 9B). Furthermore,

to analyze the effect of this scenario on production of viral vectors, PP and IP were

Figure 8. Detection of autophagy activation through LC3-integrated autophagosomes (shown as light blue spots) in infected cells 24 hpi. White arrowheads point to LC3B-positive cells with punctuate pattern indicating the integration of LC3 into the autophagosomes. High magnification inset represents the region indicated by the white square. Scale bars, 50 μm and 10 μm for high-magnification inset.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

131

quantified in the above mentioned assay (Figure 9B). The results showed a relation

between cell death and low viral particles infectivity (increase in PP:IP ratio). However,

although a 1.7 ± 0.3-fold increase in PP:IP ratio of ∆E1 vectors was obtained when co-

infecting cells with a HV MOI 40, fewer viable cells compared to those observed during

HDV+HV production were observed. Moreover, even under these circumstances, the

decrease in viral particles infectivity did not reach the 4-fold change when comparing

HDV with ∆E1 vectors (Figure 2A).

Figure 9. Production of ∆E1 vectors by inducing the intracellular environment obtained in HDV-producing cells at 48 hpi. The intracellular environment of HDV+HV-producing cells was induced by co-infecting cells with MOI 5 of ∆E1 and increasing MOIs of HV. (A) Western blot of cell extracts to compare production of viral proteins. Cell extracts were normalized to cell concentration and each lane was loaded with same sample volume. Cell extracts of HDV+HV-producing cells were added as a control to show the level of proteins produced under these conditions. Image corresponds to a single membrane and was processed as described previously (B) Fold-change on viable cell concentration and physical to infectious particles (PP:IP) ratio relative to co-infection with MOI 1 of HV. Values are shown as average ± standard-deviation (n=2).

Chapter V

132

4. Discussion

The production of helper-dependent vectors (HDVs) presents two important bottlenecks:

helper vector (HV) contamination and production yields. Several efforts toward the

reduction of HV contamination (11-15) and the improvement of transfection step (18-20)

have been described, however less attention has been paid to the overall production of

HDV. More specifically, while the interest in canine adenovirus type 2 (CAV-2) vectors for

gene therapy increases (8, 9), understanding canine HDV productivities is important to

accomplish a consistent production system, streamline the bioprocess and debottleneck

high quality preparations.

Despite the difficulty in comparing the titers described in the literature, due to the use

different protocols, vectors and samples purity, cell specific productivity of HD CAV-2

vectors (~105 physical particles/cell (Figure 2A)) was similar to the production yields

reported for human HDV (14, 21, 22). On the other hand, a ~4-fold decrease in infectious

particles of HDV was observed when compared to ∆E1 control vectors (Figure 2A).

Canine HDV preparations with higher physical to infectious particle (PP:IP) ratios than

those obtained with ∆E1 vectors, being already reported (4, 9), were consistently

obtained in our hands (data not shown). Some studies show a relatively high and/or

inconsistent PP:IP ratios among different preparations of same human HDV (14, 23).

Special attention must be paid to this, as the PP:IP ratio of clinical grade adenoviruses is

limited by Food and Drug Administration. In addition, we found a reduction in HDV

volumetric productivity related with an higher producer cell death (Figure 2C,D).

To further understand the limitations in HDV amplification we analyzed virus cycle

during vectors production. We found that HDV has a distinct viral cycle progression,

which impaired producer cell homeostasis and vector production and quality.

Despite similar internalization and nuclear delivery (Figure 3), a faster vector genome

replication of HV from 9 hpi onwards was obtained during the co-infection of HDV+HV

(Figures 3D and 4B). Any HV-specific effect was ruled out because this was not observed

in HV-producing cells (Figure 1A-iii). Although an increase in HV genome replication was

observed during co-infection with ∆E1 vector production (Figure 1A-ii), HV genome

replication was even faster in HDV+HV-producing cells (Figure 1A-i). The viral proteins

involved in adenovirus genome replication, DNA-binding protein (DBP), adenoviral DNA

polymerase (Adpol) and pre-terminal protein (pTP) belong to E2 transcription unit (34).

Indeed, an upregulation of subunit E2B products was observed in HDV-producing cells

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

133

(Figure 5), which was consistent with the replication profile of HV genome in these

conditions. Why this increase occurred during HDV+HV production, and the mechanism

behind this up-regulation, is still not clear. First, the activation of early phase

transcription is amplified by E1 (E1A) expression (41). However, no differences were

detected in E1 levels in the different infections assayed in this work (data not shown).

The mechanism behind the upregulation of E2B gene products appears to be specific,

because this was not observed in E2A subunit, which share same promoter, or E4. Unlike

HV, the replication profile of HDV genome was more modest and in accordance with that

obtained in ∆E1 control vector infection (Figure 4A). Assuming that genome replication

was upregulated in HDV+HV-producing cells, this might indicate that the stuffer

sequence, besides affecting transgene expression (Figure S1, Appendix) as previously

described (42, 43), may impact HDV genome replication.

Faster HV genome replication contributed to an increase in expression of viral products,

namely IVa2 and late-phase proteins (Figures 5 and 6). IVa2, expressed during DNA

replication, is implicated in the activation of major late promoter during late-phase

infection (36). Thus, upregulation of IVa2 can be correlated with the overexpression of

late-phase transcripts and proteins. Such high levels of viral proteins were likely

responsible for the increase in cell death during HDV+HV production, which

compromised the volumetric productivity (Figures 2C,D). Concomitantly, activation of

cell death was more pronounced in HDV+HV-producing cells, in which autophagy was

evident in these cells (Figure 8). Autophagy, in addition to its role in degradation and

recycling of cellular constituents (44, 45), can act as a form of programmed cell death (46,

47). Recent studies link autophagy to the main mechanisms for adenovirus-mediated cell

lysis (39, 40). This type of programmed cell death was therefore more activated during

HDV+HV production, which might be implicated in a critical physiological state. Still,

autophagy may be necessary for a successful adenovirus replication (40). Together, our

results are in line with the importance of a balanced autophagy activation to pursue both

a sustainable host cell homeostasis and an adequate environment for virus amplification

and maturation.

Consistent with the overall increase in viral protein production in HDV+HV-producing

cells, more viral capsids (empty plus genome-containing) were obtained when compared

to ∆E1+HV-infected cells (Figure 7A). This demonstrates that assembly occurred in

proportion to the viral protein content (Figure 6). Given the similar number of physical

Chapter V

134

(genome-containing) particles between HDV and ∆E1 infection (Figure 2A), this also

indicates that more empty and/or defective particles were obtained during HDV+HV

production. Assembly of adenovirus particles proceeds packaging: the adenovirus

genome, along with packaging proteins, interacts with structural and non-structural

proteins resulting in the formation of a “procapsid” structure with the viral DNA (48).

Then, the genome is encapsidated. The quantification of DNA-containing particles showed

that HDV packaging occurred similarly to control infections (Figure 2A). Empty capsids

and free-viral genomes were obtained in all of the assayed productions schemes (Figure

7). This indicates that a threshold for packaging was obtained, even under higher levels of

viral transcripts involved in packaging, namely IVa2 (Figure 5) (37), L1 52/55K (49), L4

22K (50) and IIIa (51), as observed during HDV+HV production (data not shown).

Together, these observations raise the hypothesis that post-transcriptional events or the

levels of host cell factors, such as P-complex (52), might influence genome packaging in a

rate limiting manner.

Following assembly and packaging, adenovirus undergoes a final maturation step driven

by the virus encoded protease. This process, involving the cleavage of several viral

proteins and increased by the presence of two co-factors (C-terminal of pVI and viral

dsDNA) renders virus particles infectious (28, 53). High levels of pVI and protease

transcripts were obtained during HDV+HV production (Figure 5), however the low

infectious titer of these vectors suggests a defect in capsid maturation. On the other hand,

late phase viral proteins produced earlier might compromise cell homeostasis and the

success of virus replication progress. For instance, protease is also responsible for the

cleavage of cell cytokeratin system, weakening mechanical integrity of the cell and

promoting host cell lysis late in infection (54). Thus, such high levels of protease earlier

(or any late-phase product) might contribute to impaired cell integrity in a way that

maturation step is not fully accomplished. Taking this into account, we hypothesized a

link between cell death/physiological state imposed by HDV infection and impaired virus

maturation. To test this possibility, an intracellular environment similar to HDV+HV

producing cells was induced in ∆E1 producing cells. Similar protein levels and cell death

profiles to those obtained during HDV production were detected using this strategy

(Figure 9), consistent with a correlation of cell death with viral protein content.

However, the impact of earlier cell death on virus maturation is only partial, because the

differences in ∆E1 infectivity (PP:IP ratio) (Figure 9B) did not reach the 4-fold decrease

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

135

observed in HDV (Figure 2A). This suggests that the physiological state of HDV+HV-

producing cells was not fully simulated using the increasing HV MOI strategy. In fact,

higher levels of autophagosomes were always obtained in HDV+HV-producing cells, even

in cells co-infected with ΔE1 and HV at MOI 40 (data not shown), which supports the

hypothesis of a HDV-specific interaction with producer cell. In addition, other intrinsic

feature of HDV might also be implicated in maturation and viral particles infectivity. The

maturation process is thought to be independent of DNA sequence (27), thus the effect of

HDV genome sequence should be ruled out. However, it is not known how viral genome

structurally interacts with core proteins, and if that affects viral capsid assembly and

subsequent maturation. In the light of these results, the role of stuffer sequence and the

clarification of HDV structure would be important to understand any connection with

viral particle maturation state (26).

In summary, in HDV+HV-producing cells viral genome replication occurred faster, leading

to higher levels of late region proteins. Such high viral protein content was consistent

with increased cell death observed during production. Despite similar cell-specific

productivities of total HDV and ∆E1 virions, infectious to physical particles ratios

obtained in HDV were 4-times lower, indicating lower infectivity. Increased levels of

empty capsids and defective particles interfere with the infectious titers. Therefore,

factors affecting the maturation process should be involved. For instance, it is likely that

the cell physiological state contributed to reduced vectors infectivity. Nevertheless, a

particular feature of HDV must be further affecting its maturation, which would fully

explain the differences in infectious titers.

This work identifies faster genome replication, viral protein synthesis, higher production

of defective particles and lower maturation as bottlenecks in helper-dependent CAV-2

production, highlighting the effect of vector design on the virus replication cycle and

virus-cell interaction as an important feature to understand the viral vector productive

outcome.

5. Acknowledgements and author contribution

We thank Sandy Ibanes and Aurelie Gennetier for the initial preparations of CAV-2

vectors, Francisca Monteiro, Vanessa Bandeira and Rute Castro for technical support.

This work was supported by Fundação para a Ciência e a Tecnologia (FCT) – Portugal,

through the project PTDC/EBB-BIO/118615/2010, and European Commission through

Chapter V

136

FP7 project BrainCAV (222992). Paulo Fernandes acknowledges FCT for his Ph.D. grant

(SFRH/BD/70810/2010).

Paulo Fernandes conceived the experimental setup and design, performed the

experiments, analyzed the data and wrote the chapter.

6. References

1. Lowenstein, P. R., R. J. Mandel, W. D. Xiong, K. Kroeger, and M. G. Castro. 2007. Immune responses to adenovirus and adeno-associated vectors used for gene therapy of brain diseases: the role of immunological synapses in understanding the cell biology of neuroimmune interactions. Curr Gene Ther 7:347-360.

2. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

3. Perreau, M., and E. J. Kremer. 2005. Frequency, proliferation, and activation of human memory T cells induced by a nonhuman adenovirus. J Virol 79:14595-14605.

4. Keriel, A., C. Rene, C. Galer, J. Zabner, and E. J. Kremer. 2006. Canine adenovirus vectors for lung-directed gene transfer: efficacy, immune response, and duration of transgene expression using helper-dependent vectors. J Virol 80:1487-1496.

5. Perreau, M., F. Mennechet, N. Serratrice, J. N. Glasgow, D. T. Curiel, H. Wodrich, and E. J. Kremer. 2007. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: implications for clinical efficacy. J Virol 81:3272-3284.

6. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

7. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

8. Cubizolle, A., N. Serratrice, N. Skander, M. A. Colle, S. Ibanes, A. Gennetier, N. Bayo-Puxan, K. Mazouni, F. Mennechet, B. Joussemet, Y. Cherel, Y. Lajat, C. Vite, F. Bernex, V. Kalatzis, M. E. Haskins, and E. J. Kremer. 2014. Corrective GUSB Transfer to the Canine Mucopolysaccharidosis VII Brain. Mol Ther 22:762-773.

9. Ariza, L., L. Gimenez-Llort, A. Cubizolle, G. Pages, B. Garcia-Lareu, N. Serratrice, D. Cots, R. Thwaite, M. Chillon, E. J. Kremer, and A. Bosch. 2014. Central Nervous System Delivery of Helper-Dependent Canine Adenovirus Corrects Neuropathology and Behavior in Mucopolysaccharidosis Type VII Mice. Hum Gene Ther 25:199-211.

10. Dormond, E., M. Perrier, and A. Kamen. 2009. From the first to the third generation adenoviral vector: what parameters are governing the production yield? Biotechnol Adv 27:133-144.

11. Ng, P., C. Beauchamp, C. Evelegh, R. Parks, and F. L. Graham. 2001. Development of a FLP/frt system for generating helper-dependent adenoviral vectors. Mol Ther 3:809-815.

12. Umana, P., C. A. Gerdes, D. Stone, J. R. Davis, D. Ward, M. G. Castro, and P. R. Lowenstein. 2001. Efficient FLPe recombinase enables scalable production of helper-dependent adenoviral vectors with negligible helper-virus contamination. Nat Biotechnol 19:582-585.

13. Parks, R. J., L. Chen, M. Anton, U. Sankar, M. A. Rudnicki, and F. L. Graham. 1996. A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci U S A 93:13565-13570.

14. Gonzalez-Aparicio, M., I. Mauleon, P. Alzuguren, M. Bunuales, G. Gonzalez-Aseguinolaza, C. San Martin, J. Prieto, and R. Hernandez-Alcoceba. 2011. Self-

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

137

inactivating helper virus for the production of high-capacity adenoviral vectors. Gene Ther 18:1025-1033.

15. Alba, R., P. Hearing, A. Bosch, and M. Chillon. 2007. Differential amplification of adenovirus vectors by flanking the packaging signal with attB/attP-PhiC31 sequences: implications for helper-dependent adenovirus production. Virology 367:51-58.

16. Soudais, C., S. Boutin, and E. J. Kremer. 2001. Characterization of cis-acting sequences involved in canine adenovirus packaging. Mol Ther 3:631-640.

17. Kanegae, Y., M. Ishimura, S. Kondo, and I. Saito. 2012. Influence of loxP insertion upstream of the cis-acting packaging domain on adenovirus packaging efficiency. Microbiol Immunol 56:447-455.

18. Hartigan-O'Connor, D., A. Amalfitano, and J. S. Chamberlain. 1999. Improved production of gutted adenovirus in cells expressing adenovirus preterminal protein and DNA polymerase. J Virol 73:7835-7841.

19. Dormond, E., A. Meneses-Acosta, D. Jacob, Y. Durocher, R. Gilbert, M. Perrier, and A. Kamen. 2009. An efficient and scalable process for helper-dependent adenoviral vector production using polyethylenimine-adenofection. Biotechnol Bioeng 102:800-810.

20. Ibanes, S., and E. J. Kremer. 2013. Canine Adenovirus Type 2 Vector Generation via I-Sce1-Mediated Intracellular Genome Release. PLoS One 8:e71032.

21. Palmer, D., and P. Ng. 2003. Improved system for helper-dependent adenoviral vector production. Mol Ther 8:846-852.

22. Suzuki, M., R. Cela, C. Clarke, T. K. Bertin, S. Mourino, and B. Lee. 2010. Large-scale production of high-quality helper-dependent adenoviral vectors using adherent cells in cell factories. Hum Gene Ther 21:120-126.

23. Kreppel, F., V. Biermann, S. Kochanek, and G. Schiedner. 2002. A DNA-based method to assay total and infectious particle contents and helper virus contamination in high-capacity adenoviral vector preparations. Hum Gene Ther 13:1151-1156.

24. Flint, S. J., L. W. Enquist, V. R. Racaniello, and A. M. Skalka. 2004. Principles of virology: molecular biology, pathogenesis, and control of animal viruses, 2nd ed. ASM Press, Washington, DC.

25. Greber, U. F., M. Suomalainen, R. P. Stidwill, K. Boucke, M. W. Ebersold, and A. Helenius. 1997. The role of the nuclear pore complex in adenovirus DNA entry. EMBO J 16:5998-6007.

26. Perez-Berna, A. J., A. Ortega-Esteban, R. Menendez-Conejero, D. C. Winkler, M. Menendez, A. C. Steven, S. J. Flint, P. J. de Pablo, and C. San Martin. 2012. The role of capsid maturation on adenovirus priming for sequential uncoating. J Biol Chem 287:31582-31595.

27. Graziano, V., W. J. McGrath, M. Suomalainen, U. F. Greber, P. Freimuth, P. C. Blainey, G. Luo, X. S. Xie, and W. F. Mangel. 2013. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: I. binding to DNA AND to hexon of the precursor to protein VI, pVI, of human adenovirus. J Biol Chem 288:2059-2067.

28. Blainey, P. C., V. Graziano, A. J. Perez-Berna, W. J. McGrath, S. J. Flint, C. San Martin, X. S. Xie, and W. F. Mangel. 2013. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: IV. viral proteinase slides along DNA to locate and process its substrates. J Biol Chem 288:2092-2102.

29. Fernandes, P., V. M. Santiago, A. F. Rodrigues, H. Tomas, E. J. Kremer, P. M. Alves, and A. S. Coroadinha. 2013. Impact of E1 and Cre on adenovirus vector amplification: developing MDCK CAV-2-E1 and E1-Cre transcomplementing cell lines. PLoS One 8:e60342.

30. Kramberger, P., M. Ciringer, A. Strancar, and M. Peterka. 2012. Evaluation of nanoparticle tracking analysis for total virus particle determination. Virology journal 9:265.

31. Serra, M., S. B. Leite, C. Brito, J. Costa, M. J. Carrondo, and P. M. Alves. 2007. Novel culture strategy for human stem cell proliferation and neuronal differentiation. Journal of neuroscience research 85:3557-3566.

Chapter V

138

32. Salinas, S., L. G. Bilsland, D. Henaff, A. E. Weston, A. Keriel, G. Schiavo, and E. J. Kremer. 2009. CAR-associated vesicular transport of an adenovirus in motor neuron axons. PLoS pathogens 5:e1000442.

33. Kamen, A., and O. Henry. 2004. Development and optimization of an adenovirus production process. J Gene Med 6:184-192.

34. Swaminathan, S., and B. Thimmapaya. 1995. Regulation of Adenovirus E2 Transcription Unit, p. 177-194. In W. Doerfler and P. Böhm (ed.), The Molecular Repertoire of Adenoviruses III, vol. 199/3. Springer Berlin Heidelberg.

35. Weber, J. 1976. Genetic analysis of adenovirus type 2 III. Temperature sensitivity of processing viral proteins. J Virol 17:462-471.

36. Tribouley, C., P. Lutz, A. Staub, and C. Kedinger. 1994. The product of the adenovirus intermediate gene IVa2 is a transcriptional activator of the major late promoter. J Virol 68:4450-4457.

37. Zhang, W., J. A. Low, J. B. Christensen, and M. J. Imperiale. 2001. Role for the adenovirus IVa2 protein in packaging of viral DNA. J Virol 75:10446-10454.

38. Altaras, N. E., J. G. Aunins, R. K. Evans, A. Kamen, J. O. Konz, and J. J. Wolf. 2005. Production and formulation of adenovirus vectors. Adv Biochem Eng Biotechnol 99:193-260.

39. Jiang, H., E. J. White, C. I. Rios-Vicil, J. Xu, C. Gomez-Manzano, and J. Fueyo. 2011. Human adenovirus type 5 induces cell lysis through autophagy and autophagy-triggered caspase activity. J Virol 85:4720-4729.

40. Rodriguez-Rocha, H., J. G. Gomez-Gutierrez, A. Garcia-Garcia, X. M. Rao, L. Chen, K. M. McMasters, and H. S. Zhou. 2011. Adenoviruses induce autophagy to promote virus replication and oncolysis. Virology 416:9-15.

41. Nevins, J. R. 1987. Regulation of early adenovirus gene expression. Microbiological reviews 51:419-430.

42. Ross, P. J., M. A. Kennedy, and R. J. Parks. 2009. Host cell detection of noncoding stuffer DNA contained in helper-dependent adenovirus vectors leads to epigenetic repression of transgene expression. J Virol 83:8409-8417.

43. Parks, R. J., J. L. Bramson, Y. Wan, C. L. Addison, and F. L. Graham. 1999. Effects of stuffer DNA on transgene expression from helper-dependent adenovirus vectors. J Virol 73:8027-8034.

44. Ohsumi, Y. 2001. Molecular dissection of autophagy: two ubiquitin-like systems. Nature reviews. Molecular cell biology 2:211-216.

45. Levine, B., and D. J. Klionsky. 2004. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Developmental cell 6:463-477.

46. Baehrecke, E. H. 2005. Autophagy: dual roles in life and death? Nature reviews. Molecular cell biology 6:505-510.

47. Gozuacik, D., and A. Kimchi. 2007. Autophagy and cell death. Current topics in developmental biology 78:217-245.

48. Ostapchuk, P., and P. Hearing. 2005. Control of adenovirus packaging. Journal of cellular biochemistry 96:25-35.

49. Perez-Romero, P., K. E. Gustin, and M. J. Imperiale. 2006. Dependence of the encapsidation function of the adenovirus L1 52/55-kilodalton protein on its ability to bind the packaging sequence. J Virol 80:1965-1971.

50. Ostapchuk, P., M. E. Anderson, S. Chandrasekhar, and P. Hearing. 2006. The L4 22-kilodalton protein plays a role in packaging of the adenovirus genome. J Virol 80:6973-6981.

51. Ma, H. C., and P. Hearing. 2011. Adenovirus structural protein IIIa is involved in the serotype specificity of viral DNA packaging. J Virol 85:7849-7855.

52. Erturk, E., P. Ostapchuk, S. I. Wells, J. Yang, K. Gregg, A. Nepveu, J. P. Dudley, and P. Hearing. 2003. Binding of CCAAT displacement protein CDP to adenovirus packaging sequences. J Virol 77:6255-6264.

The impact of adenovirus life cycle on the generation of canine helper-dependent vectors

139

53. Mangel, W. F., W. J. McGrath, D. L. Toledo, and C. W. Anderson. 1993. Viral DNA and a viral peptide can act as cofactors of adenovirus virion proteinase activity. Nature 361:274-275.

54. Chen, P. H., D. A. Ornelles, and T. Shenk. 1993. The adenovirus L3 23-kilodalton proteinase cleaves the amino-terminal head domain from cytokeratin 18 and disrupts the cytokeratin network of HeLa cells. J Virol 67:3507-3514.

Chapter V

140

Chapter VI

Discussion and perspectives

Chapter VI

142

Contents

1. Discussion ................................................................................................................................................. 143

1.1. Upstream bioprocess for CAV-2 vectors: from producer cell lines to stirred tank

bioreactors..................................................................................................................................................... 145

1.2. Players and challenges behind canine helper-dependent vectors propagation .... 146

2. Future directions and perspectives .............................................................................................. 148

2.1. Considerations to develop superior adenovirus producer cells ................................... 148

2.2. Improve the performance of cells adapted to suspension............................................... 148

2.3. Bioprocess development for HDV............................................................................................... 149

2.4. Understand the relation between HDV construct, replication profile and

productivity ................................................................................................................................................... 149

2.5. Enhance genome packaging to maximize the generation of CAV-2 vectors ............ 150

3. Final remarks .......................................................................................................................................... 150

4. Author contribution ............................................................................................................................. 151

5. References ................................................................................................................................................ 151

Discussion and perspectives

143

1. Discussion

Decades of intense research and several clinical trials have demonstrated that gene

therapy is feasible and has the potential to provide an etiological treatment to a wide

array of human pathologies. Currently, a considerable attention is being paid to adeno-

associated and lentivirus vectors. Still, adenovirus vectors represent the majority of

clinical trials (1), are the blockbuster for oncolytic therapies (2) and comprise the best

tool to gene editing through CRISPR/Cas9 (3), an hot topic in gene therapy and

biotechnology fields. In addition, high-capacity/helper-dependent adenovirus vectors

overcome the reduced cargo capacity of adeno-associated vectors, if delivery of large

genes or complex expression systems is required.

Despite the potentialities of human adenovirus vectors, the hurdles facing their clinical

utilization, including innate and pre-existing immunity, may limit the efficiency and time

of transgene expression (4). One of the alternatives to circumvent these drawbacks is the

use of non human adenovirus, such as canine adenovirus type 2 (CAV-2) vectors (5). In

addition to the paucity of anti-CAV-2 neutralizing antibodies in humans (5, 6), CAV-2

vectors induce a low level of innate response and no activation of human complement

pathways (7, 8). Their ability to preferentially transduce neurons, combined with a

remarkable capacity of axonal transport (9, 10), make CAV-2 vectors promising

candidates for the treatment of neurodegenerative diseases (11, 12). As the interest in

CAV-2 vectors keeps increasing (11, 12), larger amounts of high quality vectors to

conduct preclinical and possibly clinical assays are required. Still, prior to this work,

production process for CAV-2 was lagging behind those already established for human

adenovirus vectors.

This PhD thesis aimed at leveraging CAV-2 vectors manufacturing to process scales.

Technical aspects that would facilitate regulatory approval of clinical grade material

production were addressed and adapted to E1-deleted (ΔE1) and helper-dependent

vectors (HDV) production. The detailed characterization of cell line and bioprocess

parameters increased the knowledge on cell physiology and virus replication behind an

improved production, expanding the state-of-the-art of adenovirus manufacturing. The

specific aims and outcomes of this thesis are summarized in Figure 1.

Chapter VI

144

Figure 1. Schematic view of the work developed in this thesis, including initial bottlenecks, specific aims, strategies adopted and main achievements/findings. The central objective of this PhD thesis was to develop a scalable production for two CAV-2 vectors – ΔE1 and HDVs – compliant with clinical grade material production. To that end, new MDCK-based cell lines were developed (A) and production process transferred to stirred tank bioreactors (B). Given the low production yields and the complexity of HDV production system, a deeper characterization of this vector production was necessary before considering the upscale, by defining the infection parameters for optimal productivities (C) and identifying bottlenecks during HDV replication (D). ΔE1: E1-deleted; HDV: helper-dependent vector; HV: helper vector; IP: infectious particles; PP: physical particles; SFM: serum-free medium.

Discussion and perspectives

145

1.1. Upstream bioprocess for CAV-2 vectors: from producer cell lines to stirred tank

bioreactors

CAV-2 vectors were initially developed and produced using dog kidney (DK) cells (11-

13). From the point-of-view of a regulatory approval for clinical material production, the

use of a cell substrate already acknowledged by Food and Drug Administration and

European Medical Agency should be considered. Unlike DK cells, Madin Darby canine

kidney (MDCK) cells represent a suitable cell substrate to facilitate the regulatory

approval for the production of clinical grade CAV-2 vectors, as they are already approved

by the regulatory authorities for vaccines manufacturing (14), Accordingly, we

successfully develop new MDCK-derived cell lines – MDCK-E1 and MDCK-E1-Cre

(Chapter II). These cell lines showed productivity titers as high as those described for

DKZeo and DKCre cells and for human adenovirus producing cell lines (15). In addition,

our MDCK-E1 cell line showed a remarkable stability, maintaining the titers with

increasing cultures passages (Chapter II and Chapter IV).

To meet the need of vectors at larger quantities compliant with good manufacturing

practices, ΔE1 CAV-2 production in MDCK-E1 cells was adapted to stirred tank

bioreactors and serum-free medium. Microcarrier technology, representing the most

common approach to transfer anchorage-dependent cells to stirred culture systems, was

used to develop the first bioprocess for CAV-2 vectors (Chapter III, part A). The CAV-2

productivities of 2×103 IP/cell obtained in 2 L scale vessels were similar to monolayer

static cultures. Despite these advances, a competitive CAV-2 bioprocess should meet the

features already accomplished for human adenoviruses, i.e. producing vectors with cells

adapted to suspension (15). By changing culture medium to a specific serum-free

formulation (16), MDCK-E1 cell line was successfully adapted to grow in suspension as

single cells (Chapter III, part B). However, productivities were ~5-fold (between 3- and

8-fold) lower than those from bioreactor microcarrier cultures. Despite the lower titers,

single cells suspension cultures contributes to streamline CAV-2 production process. For

instance, the additional costs and processing steps associated with microcarriers

preparation, adherent cell expansion and monitoring are now abolished. Therefore, it will

be worth to explore the performance of the new suspension-adapted MDCK-E1 cells to

meet the titers normally accomplished. This would greatly improve the cost-effectiveness

of the final CAV-2 bioprocess.

Chapter VI

146

1.2. Players and challenges behind canine helper-dependent vectors propagation

Process optimization of ΔE1 vectors is relatively straightforward. However, due to the

complexity of production system, helper-dependent vector (HDV) production is

considered more challenging than ΔE1 vectors. More specifically, producer cells must

express Cre recombinase, which can be toxic to cells (17), and cells need to be infected by

two vectors (HDV and helper vector (HV)). A deeper evaluation of HDV production was

therefore pursued before considering the transfer to stirred cultures and the upscale.

MDCK-E1-Cre cells stability was analyzed, best infection conditions determined and

bottlenecks behind HDV replication identified.

Given Cre genotoxicity (17), it becomes crucial to understand how Cre expression can

affect MDCK-E1-Cre cell line phenotype in the long term. Indeed, and unlike MDCK-E1

cells, MDCK-E1-Cre cells were less stable with increasing culture passages in what

regards virus production, despite showing reproducible growth profiles (Chapter IV).

This indicates that passage number of our Cre-expressing MDCK must be kept to a

minimum to maintain the specific yields. To our knowledge, these findings were never

reported for other HDV production systems. In fact, Cre-expressing HEK293 cells

established by Palmer & Ng (18) were able to keep adenovirus productivities up to 86

passages. However, Cre-expressing HEK293 cells developed by others ceased to

proliferate (19). In the light of these contradictory observations, the evaluation of the

different Cre-expressing cells established for HDV production (20) should be pursued to

understand how host cell and/or cell clone is implicated in the impact that Cre has on the

resulting cell performance.

Multiple amplification steps are needed to rescue HDV amplification to its maximum and

prepare sufficient quantities of vector. This increases the chance of recombination

between HDV and HV. In fact, the excised DNA from HV after Cre activity greatly

contributes to recombination between vectors (21). To minimize this, the levels of

excised HV genomes and/or HV genome excision should be reduced in the sequential

amplifications of HDV. We found a simple way to do this by adjusting the HDV/HV MOI

ratio used to infect cells. High HDV/HV MOI ratio was sufficient to maintain HDV yields

and minimize HV levels. In fact, under such MOI ratio, HV contamination was similar in

MDCK-E1 or MDCK-E1-Cre cells (Chapter IV). This indicates the possibility to produce

HDV with low HV levels without the need of Cre-expressing cells and, therefore, avoiding

HV genome excision. Accordingly, as soon as HDV titers permit the establishment of such

Discussion and perspectives

147

MOI ratio (usually after rescue step and 1st amplification), the subsequent amplifications

can be moved to MDCK-E1 cells.

Even under optimized conditions, the production yields of canine HDV were 4-times

lower than those obtained for ΔE1 vectors. To identify the cause of this observation, we

evaluated HDV replication from an adenovirus life cycle perspective, i.e. by characterizing

the steps comprising the generation of adenovirus particles (Chapter V). Several steps

were intensified during HDV propagation, including genome replication, expression of

viral products and assembly of viral particles. This not only affected cell viability after

infection but also compromised HDV infectivity and volumetric titers. Such observations

were not found with other CAV-2 vectors, indicating a specific vector/cell interaction

during HDV propagation. The relation between vector construct and replication profile

should be thus elucidated to understand these observations and debottleneck higher

titers of HDV.

Similarly to what was done to ΔE1 CAV-2 (Chapter III), HDV production was tested in

stirred tank bioreactors with microcarriers technology and MDCK-E1-Cre cells were

subjected to suspension adaptation. In the first approach, high cell death after infection

and very low volumetric titers (106 – 107 IP/mL) were obtained (data not shown), which

corroborates the findings of Chapter V. Secondly, we were unable to adapt MDCK-E1-Cre

cells to suspension using the approach described on Chapter III, Part B: cell viability

consistently decreased during adaptation until no more viable cells were observed. This

indicates that Cre expression indeed makes cells more susceptible to manipulation and

death. Higher volumetric titers and MDCK-E1-Cre cells adapted to suspension should be

pursued to develop a robust production process for canine HDV in stirred tank

bioreactors.

Despite not succeeding in establishing a production process for HDV with high yields, this

thesis contributes to the i) definition of critical infection parameters for an improved HDV

production and ii) identification of the biological factors that should be modulated in the

future to debottleneck HDV at higher titers and with improved quality.

Chapter VI

148

2. Future directions and perspectives

The accomplishments and findings of this work (Figure 1) raised further aspects and

questions to be explored in the pursuit of an improved production process for CAV-2

vectors but also for other adenovirus vectors.

2.1. Considerations to develop superior adenovirus producer cells

While adenovirus producing cell clone selection is typically based on virus titers

screening, additional aspects behind cell performance should be undertaken to tune the

selection of more promising clones. In fact, a clear correlation between E1 (mostly E1A)

expression levels and CAV-2 production was observed during cell line development and

clone characterization (Chapter II), which corroborates the importance of evaluating E1

levels when screening and developing adenovirus producer cells. Accordingly, it is worth

to explore if attaining expression levels of E1 higher than those achieved up to now can

further increase CAV-2 titers.

2.2. Improve the performance of cells adapted to suspension

The accomplishment of adapting MDCK-E1 cells to suspension (Chapter III, part B)

contribute to the development of a cost-effective CAV-2 bioprocess. Still, the adapted

MDCK-E1 cells decreased their specific productivity in ~5-fold, when compared to those

previously obtained (Chapter II and Chapter III, part A). From the manufacturing

perspective, higher volumetric titers should be thus pursued in these cultures. One

possibility is testing other culture media commercially available that might allow higher

virus replication. Despite easy to evaluate, the success of new culture media delivering

higher titers is limited. In addition, any information on what is actually critical for

adenovirus replication in this scenario would remain unknown. One should regard the

changes in membrane receptors and signaling pathways that a cell is challenged to evolve

from an adherent phenotype to a suspension environment (22), while avoiding anoikis

(23). Therefore, understanding what changes MDCK-E1 went through and how they affect

virus production is of upmost importance, namely by studying the transcriptome of

MDCK-E1 producing cell in adherent versus suspension cultures. Besides the scientific

relevance of such findings, this information would be extremely helpful to improve cell

performance in a more targeted orientation.

Discussion and perspectives

149

2.3. Bioprocess development for HDV

MDCK-E1-Cre cells growing in suspension would be important to transfer production

process to stirred tank bioreactors. An adaptation procedure more gradual than that

performed (Chapter III, part B) can be explored for MDCK-E1-Cre cells. However, if Cre

recombinase is actually impairing cell survival during adaptation process as

hypothesized, an inducible expression system to control Cre expression in cells adapted

to suspension must be considered. Nevertheless, using what we accomplished so far, we

can propose a production protocol with rescue step (vector genome transfection) and

initial amplifications with MDCK-E1-Cre cells in adherent cultures. After obtaining

sufficient HDV, and since high HDV/HV ratio is sufficient to minimize HV contamination

(Chapter IV), HDV production can be moved to MDCK-E1 cells in stirred tank

bioreactors.

Aiming to develop a complete bioprocess for HDV vectors, the downstream processing

should be also envisaged. From a scalable perspective, chromatographic columns are the

typical choice for purification processes. However, such technology is incompatible with

the need of removing HV from HDV. One possibility to circumvent this is developing and

using self-inactivating HVs, as described by Gonzalez-Aparicio et al (24). Following this

approach, the HV contamination would be already avoided during HDV production, and

downstream process could be similar to any ΔE1 vector (25, 26).

2.4. Understand the relation between HDV construct, replication profile and

productivity

The HDV titers and quality are constraining a high yield, cost-effective production

process. Besides low infectivity, HDV showed a distinct replication profile resulting in

high cell death and low volumetric titers (Chapter V). These observations suggest a

specific HDV-producer cell interaction, as this was not observed for other CAV-2 vectors.

In the light of these results, it would be important to understand how vector construct

impacts its replication and producer cell homeostasis. Unlike ΔE1 vectors, HDV genome

harbors stuffer DNA sequences to bring the genome size to its normal and be properly

packaged. Therefore, it is important to evaluate the effect of different HDV constructs, i.e.

different stuffer DNA sequences, on replication profile and infectivity. If the replication

profile would indeed be extended to the different HDV constructs, HDV replication and

cell death must be delayed to improve volumetric titers and hopefully viral particles

Chapter VI

150

quality. For instance, cell survival after infection can be improved by expressing higher

levels of E1B (Chapter II) or other anti-apoptotic genes.

2.5. Enhance genome packaging to maximize the generation of CAV-2 vectors

Besides assembled genome-containing capsids, empty capsids are typically obtained after

adenovirus production (27). At the same time, 60-80% of free viral genomes were

observed at the end of production for both HDV and ΔE1 vectors (Chapter V). Together,

this shows that genome packaging constitutes a bottleneck during CAV-2 vectors

production. These observations also indicate enhancing packaging efficiency as a

potential approach to generate higher number of CAV-2 vectors. This can be explored by

manipulating cell and viral factors involved in packaging (28-32) and/or provide more

time for this process to occur by delaying cell lysis. Worth mentioning that adenovirus

particle generation is an orchestrated process, and the adenovirus life cycle steps

following packaging should occur accordingly. For instance, it will be crucial to ensure

that maturation step, which brings infectivity to adenoviruses, is not restricted to a

limited number of CAV-2 particles, i.e. is capable to process an increasing number of viral

particles.

3. Final remarks

This PhD thesis contributes to the progress of gene therapy by enlarging the repertoire of

vectors ready for clinical applications, merging biotechnology with virology and cell

biology areas. Upstream bioprocesses for CAV-2 vectors that meet the technical aspects

for process transfer and clinical grade production were accomplished. At the same time,

important features behind optimal cell performance and virus replication were identified.

Together, this work improved the state-of-the-art on adenovirus vectors manufacturing

and raised future perspectives on how adenovirus productivity can be further enhanced.

Discussion and perspectives

151

4. Author contribution

Paulo Fernandes wrote the chapter.

5. References

1. Ginn, S. L., I. E. Alexander, M. L. Edelstein, M. R. Abedi, and J. Wixon. 2013. Gene therapy clinical trials worldwide to 2012 - an update. J Gene Med 15:65-77.

2. Choi, I. K., and C. O. Yun. 2013. Recent developments in oncolytic adenovirus-based immunotherapeutic agents for use against metastatic cancers. Cancer gene therapy 20:70-76.

3. Holkers, M., I. Maggio, S. F. Henriques, J. M. Janssen, T. Cathomen, and M. A. Goncalves. 2014. Adenoviral vector DNA for accurate genome editing with engineered nucleases. Nat Methods 11:1051-1057.

4. Lowenstein, P. R., R. J. Mandel, W. D. Xiong, K. Kroeger, and M. G. Castro. 2007. Immune responses to adenovirus and adeno-associated vectors used for gene therapy of brain diseases: the role of immunological synapses in understanding the cell biology of neuroimmune interactions. Curr Gene Ther 7:347-360.

5. Kremer, E. J., S. Boutin, M. Chillon, and O. Danos. 2000. Canine adenovirus vectors: an alternative for adenovirus-mediated gene transfer. J Virol 74:505-512.

6. Perreau, M., and E. J. Kremer. 2005. Frequency, proliferation, and activation of human memory T cells induced by a nonhuman adenovirus. J Virol 79:14595-14605.

7. Keriel, A., C. Rene, C. Galer, J. Zabner, and E. J. Kremer. 2006. Canine adenovirus vectors for lung-directed gene transfer: efficacy, immune response, and duration of transgene expression using helper-dependent vectors. J Virol 80:1487-1496.

8. Perreau, M., F. Mennechet, N. Serratrice, J. N. Glasgow, D. T. Curiel, H. Wodrich, and E. J. Kremer. 2007. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: implications for clinical efficacy. J Virol 81:3272-3284.

9. Soudais, C., C. Laplace-Builhe, K. Kissa, and E. J. Kremer. 2001. Preferential transduction of neurons by canine adenovirus vectors and their efficient retrograde transport in vivo. Faseb J 15:2283-2285.

10. Soudais, C., N. Skander, and E. J. Kremer. 2004. Long-term in vivo transduction of neurons throughout the rat CNS using novel helper-dependent CAV-2 vectors. FASEB J 18:391-393.

11. Cubizolle, A., N. Serratrice, N. Skander, M. A. Colle, S. Ibanes, A. Gennetier, N. Bayo-Puxan, K. Mazouni, F. Mennechet, B. Joussemet, Y. Cherel, Y. Lajat, C. Vite, F. Bernex, V. Kalatzis, M. E. Haskins, and E. J. Kremer. 2014. Corrective GUSB Transfer to the Canine Mucopolysaccharidosis VII Brain. Mol Ther 22:762-773.

12. Ariza, L., L. Gimenez-Llort, A. Cubizolle, G. Pages, B. Garcia-Lareu, N. Serratrice, D. Cots, R. Thwaite, M. Chillon, E. J. Kremer, and A. Bosch. 2014. Central Nervous System Delivery of Helper-Dependent Canine Adenovirus Corrects Neuropathology and Behavior in Mucopolysaccharidosis Type VII Mice. Hum Gene Ther 25:199-211.

13. Bru, T., S. Salinas, and E. J. Kremer. 2010. An update on canine adenovirus type 2 and its vectors. Viruses 2:2134-2153.

14. Doroshenko, A., and S. A. Halperin. 2009. Trivalent MDCK cell culture-derived influenza vaccine Optaflu (Novartis Vaccines). Expert Rev Vaccines 8:679-688.

15. Silva, A. C., C. Peixoto, T. Lucas, C. Kuppers, P. E. Cruz, P. M. Alves, and S. Kochanek. 2010. Adenovirus vector production and purification. Curr Gene Ther 10:437-455.

16. van Wielink, R., H. C. Kant-Eenbergen, M. M. Harmsen, D. E. Martens, R. H. Wijffels, and J. M. Coco-Martin. 2011. Adaptation of a Madin-Darby canine kidney cell line to suspension growth in serum-free media and comparison of its ability to produce avian influenza virus to Vero and BHK21 cell lines. J Virol Methods 171:53-60.

Chapter VI

152

17. Loonstra, A., M. Vooijs, H. B. Beverloo, B. A. Allak, E. van Drunen, R. Kanaar, A. Berns, and J. Jonkers. 2001. Growth inhibition and DNA damage induced by Cre recombinase in mammalian cells. Proc Natl Acad Sci U S A 98:9209-9214.

18. Palmer, D., and P. Ng. 2003. Improved system for helper-dependent adenoviral vector production. Mol Ther 8:846-852.

19. Silver, D. P., and D. M. Livingston. 2001. Self-excising retroviral vectors encoding the Cre recombinase overcome Cre-mediated cellular toxicity. Molecular cell 8:233-243.

20. Kovesdi, I., and S. J. Hedley. 2010. Adenoviral producer cells. Viruses 2:1681-1703. 21. Ahn, M., A. Gamble, S. R. Witting, J. Magrisso, S. Surendran, S. Obici, and N. Morral.

2013. Vector and helper genome rearrangements occur during production of helper-dependent adenoviral vectors. Human gene therapy methods 24:1-10.

22. Jaluria, P., M. Betenbaugh, K. Konstantopoulos, B. Frank, and J. Shiloach. 2007. Application of microarrays to identify and characterize genes involved in attachment dependence in HeLa cells. Metab Eng 9:241-251.

23. Taddei, M. L., E. Giannoni, T. Fiaschi, and P. Chiarugi. 2012. Anoikis: an emerging hallmark in health and diseases. J Pathol 226:380-393.

24. Gonzalez-Aparicio, M., I. Mauleon, P. Alzuguren, M. Bunuales, G. Gonzalez-Aseguinolaza, C. San Martin, J. Prieto, and R. Hernandez-Alcoceba. 2011. Self-inactivating helper virus for the production of high-capacity adenoviral vectors. Gene Ther 18:1025-1033.

25. Fernandes, P., C. Peixoto, V. M. Santiago, E. J. Kremer, A. S. Coroadinha, and P. M. Alves. 2013. Bioprocess development for canine adenovirus type 2 vectors. Gene Ther 20:353-360.

26. Segura, M. M., M. Puig, M. Monfar, and M. Chillon. 2012. Chromatography purification of canine adenoviral vectors. Human gene therapy methods 23:182-197.

27. Altaras, N. E., J. G. Aunins, R. K. Evans, A. Kamen, J. O. Konz, and J. J. Wolf. 2005. Production and formulation of adenovirus vectors. Adv Biochem Eng Biotechnol 99:193-260.

28. Zhang, W., J. A. Low, J. B. Christensen, and M. J. Imperiale. 2001. Role for the adenovirus IVa2 protein in packaging of viral DNA. J Virol 75:10446-10454.

29. Perez-Romero, P., K. E. Gustin, and M. J. Imperiale. 2006. Dependence of the encapsidation function of the adenovirus L1 52/55-kilodalton protein on its ability to bind the packaging sequence. J Virol 80:1965-1971.

30. Ostapchuk, P., M. E. Anderson, S. Chandrasekhar, and P. Hearing. 2006. The L4 22-kilodalton protein plays a role in packaging of the adenovirus genome. J Virol 80:6973-6981.

31. Ma, H. C., and P. Hearing. 2011. Adenovirus structural protein IIIa is involved in the serotype specificity of viral DNA packaging. J Virol 85:7849-7855.

32. Erturk, E., P. Ostapchuk, S. I. Wells, J. Yang, K. Gregg, A. Nepveu, J. P. Dudley, and P. Hearing. 2003. Binding of CCAAT displacement protein CDP to adenovirus packaging sequences. J Virol 77:6255-6264.

Appendix

Appendix

154

Contents

1. Table S1 ..................................................................................................................................................... 155

2. Table S2 ..................................................................................................................................................... 156

3. Table S3 ..................................................................................................................................................... 157

4. Figure S1 ................................................................................................................................................... 158

5. Figure S2 ................................................................................................................................................... 159

6. References ................................................................................................................................................ 160

Appendix

155

1. Table S1

Primer sequences used to analyze each DNA target.

Target

Primer sequence

Forward Reverse

GFP CAGAAGAACGGCATCAAGGT CTGGGTGCTCAGGTAGTGG

lacZ ACTATCCCGACCGCCTTACT TAGCGGCTGATGTTGAACTG

Adpol ATGTGGCATGTATGCAAGTG CAGCACGTCCAAAAACTCTT

DBP AATGAGGATGCTTTCTGCTG TGTTCCAATTGCACTCACAC

pTP TTAATGAGCTCACCATCCGC CATTCCACACCCGACTGTAG

E4 Orf3 TGAAATTTTGCTCTCCTTGG TGGTCATCCCCTCAGTTAAA

E4 Orf6 TGCTGTGGTGAGAGTTCCTA GAGCAGTGACAGTGTACTCG

IVa2 AACTGTCCATAAAGCCTGCA TCCAGCGTTGTCTGTCTGTA

protease AAGCAATGAAACCATTCAGG CATACTGCACTGTGGGATCA

pVI TATCGCCAACCAGGAGTTAG GTCCGCAGTTATGAGCAAGT

II AGACAGGTCCCGCTACTTTT TTTCTGCCTGAAAGTTTTGG

RPL22 CTGCCAATTTTGAGCAGTTT CTTTGCTGTTAGCAACTACGC

Appendix

156

2. Table S2

Viral transcripts analyzed and the corresponding infection phase and biological function

on adenovirus amplification.

Target Phase Main Role

Adpol

Early (E2) genome replication (1) DBP

pTP

E4 Orf3

Early (E4) mRNA transport (2, 3)

E4 Orf6

IVa2 Middle and Late Activation of late gene expression (4) and genome

packaging (5)

Protease

Late (L3)

Maturation (6, 7)

pVI

II (hexon)

Capsid protein (2)

Appendix

157

3. Table S3

RPL22-normalized mRNA levels of selected viral genes during production time in

HDV+HV-, ∆E1+HV- and HV-producing cells.

HDV ∆E1 HV

12 hpi 24 hpi 48 hpi 12 hpi 24 hpi 48 hpi 12 hpi 24 hpi 48 hpi

Adpol 2.186 45.631 96.395 0.170 5.876 32.429 0.009 4.531 15.008

DBP 25.109 87.607 121.144 6.692 16.202 50.576 1.020 26.540 28.121

orf3 4.219 4.957 8.724 1.068 1.656 6.472 0.209 2.952 4.638

orf6 10.353 11.121 15.751 2.691 3.812 12.777 0.594 7.454 10.008

pTP 3.495 13.713 11.155 0.258 3.439 6.768 0.010 3.865 3.951

protease 8.964 315.629 490.248 0.258 55.742 241.287 0.005 15.180 63.504

pVI 5.801 150.904 245.405 0.236 26.326 129.790 0.003 10.453 36.367

IVa2 1.531 13.487 26.340 0.110 1.949 9.009 0.008 1.381 3.615

II 3.593 133.725 265.863 0.129 21.431 126.768 0.002 7.777 27.074

Values correspond to the average of two independent assays.

Appendix

158

4. Figure S1

Figure S1. mRNA levels of GFP and lacZ in each infection normalized to housekeeping

RPL-22. (A) Relative expression of GFP from HDV and ∆E1 genomes. (B) Relative

expression of lacZ from HV genome in HDV, ∆E1 and HV infection. Values are shown as

average ± standard-deviation (n=2).

Appendix

159

5. Figure S2

Figure S2. Gene expression pattern of

HDV (A), ∆E1 (B) and HV (C)

producing cells at 12, 24 and 48 hpi.

Gene expression pattern values,

representing the contribution of each

viral target to the overall expression,

were calculated as the percentage of

mRNA level of each viral gene relative

to total mRNAs levels obtained at each

infection point. Values are shown as

average ± standard-deviation (n=2).

Appendix

160

6. References

1. Swaminathan, S., and B. Thimmapaya. 1995. Regulation of Adenovirus E2 Transcription Unit, p. 177-194. In W. Doerfler and P. Böhm (ed.), The Molecular Repertoire of Adenoviruses III, vol. 199/3. Springer Berlin Heidelberg.

2. Russell, W. C. 2000. Update on adenovirus and its vectors. J Gen Virol 81:2573-2604.

3. Weitzman, M. D., and D. A. Ornelles. 2005. Inactivating intracellular antiviral responses during adenovirus infection. Oncogene 24:7686-7696.

4. Tribouley, C., P. Lutz, A. Staub, and C. Kedinger. 1994. The product of the adenovirus intermediate gene IVa2 is a transcriptional activator of the major late promoter. J Virol 68:4450-4457.

5. Zhang, W., J. A. Low, J. B. Christensen, and M. J. Imperiale. 2001. Role for the adenovirus IVa2 protein in packaging of viral DNA. J Virol 75:10446-10454.

6. Weber, J. 1976. Genetic analysis of adenovirus type 2 III. Temperature sensitivity of processing viral proteins. J Virol 17:462-471.

7. Blainey, P. C., V. Graziano, A. J. Perez-Berna, W. J. McGrath, S. J. Flint, C. San Martin, X. S. Xie, and W. F. Mangel. 2013. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: IV. viral proteinase slides along DNA to locate and process its substrates. J Biol Chem 288:2092-2102.