© 2010 brittney l. gurda · 1 mapping dominant antigenic regions on the capsid surfaces of...

214
1 MAPPING DOMINANT ANTIGENIC REGIONS ON THE CAPSID SURFACES OF ADENO-ASSOCIATED VIRIONS By BRITTNEY LYNN GURDA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2010

Upload: others

Post on 02-Jan-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

1

MAPPING DOMINANT ANTIGENIC REGIONS ON THE CAPSID SURFACES OF ADENO-ASSOCIATED VIRIONS

By

BRITTNEY LYNN GURDA

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2010

Page 2: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

2

© 2010 Brittney L. Gurda

Page 3: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

3

To my mother, for reminding me that I could when I said “I couldn’t”, and my father, who said that he didn’t care what I grew up to be as long as it made me happy

Page 4: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

4

ACKNOWLEDGMENTS

I would like to begin with my mom, Christine Gurda. Not only has she been there

financially, but also spiritually and mentally. I can think of no other who has shown greater

support in all of my endeavors. My father, Thomas Gurda, my sister, CaryBeth, and my brother,

Dean were always there for support. I feel exceptionally lucky and honored to have had an

excellent mentor. Dr. Mavis Agbandje-McKenna’s patience and encouragement make her the

ideal mentor and I could have never found a better place to start. I would also like to thank Dr.

Robert McKenna for insightful conversations and general advice in not only science, but life as

well. I also need to thank Michael DiMattia and Edward Miller for help with computational

programs. The monoclonal work was done in collaboration at Cornell University by Wendy

Weichert under Dr Colin Parrish, and the neutralization data was mostly done by Dr. Michael

Schmidt and Beverly Handelman under Dr. Jay Chiorini at the NIH. As for data collection, all of

our cryoEM data was collected at UCSD under the direction of Dr. Tim Baker by Norm Olson.

My thanks also go to Dr. Baker’s staff especially Drs. Robert Sinkovits and Kristin Parent who

helped with the HBoV structure in Appendix A. The data in Appendix B is almost entirely the

work of Dr. Christina Raupp in Dr. Jurgen Kleinchmidt’s lab in Hieldelberg, Germany and I am

excited to have been able to offer data that helped shaped the efforts of the epitope mapping of

the mAb ADK8. I am honored to have had the expertise of my committee members at my

disposal; Dr. James B. Flanegan, Dr. Nicholas Muzyczka, Dr. Roland Herzog, and Dr. Arun

Srivastava. All of whom are not only experts in their field, but also in the shaping of a project

and the student involved. The IDP graduate student office was invaluable in making my first year

transition smooth, and I would also like to thank the staff in the biochemistry office. Last but not

least, I would like to thank the hordes of undergraduate students that I have taught over the years,

who have unknowingly taught me many valuable lessons in return.

Page 5: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

5

TABLE OF CONTENTS

page

ACKNOWLEDGMENTS.................................................................................................................... 4

LIST OF TABLES................................................................................................................................ 9

LIST OF FIGURES ............................................................................................................................ 10

LIST OF ABBREVIATIONS ............................................................................................................ 12

ABSTRACT ........................................................................................................................................ 14

CHAPTER

1 BACKGROUND AND INTRODUCTION .............................................................................. 15

Introduction to Genetic Disease and Gene Therapy ................................................................. 15 Gene Delivery Systems ............................................................................................................... 16

Non-Viral Delivery Methods .............................................................................................. 17 Physical approaches ..................................................................................................... 17 Chemical methods ........................................................................................................ 19

Viral Delivery Methods ....................................................................................................... 22 Retroviruses .................................................................................................................. 23 Adenoviruses ................................................................................................................ 25 Herpesvirus ................................................................................................................... 27 Alphaviruses ................................................................................................................. 29 Adeno-associated viruses............................................................................................. 31

Current Status and Issues in Gene Therapy ............................................................................... 33 Antibody Recognition of Viruses and Viral Neutralization ..................................................... 40 Parvovirus Capsid Structures, Receptor Binding, and Infection .............................................. 44 Parvovirus Antigenic Properties................................................................................................. 47 Parvovirus Antigenic Structures for Autonomous Viruses ...................................................... 48 The Dependoviruses and their Antigenic Properties................................................................. 51 AAV Capsid Structure and Serotypes........................................................................................ 54 Significance ................................................................................................................................. 56

2 MATERIALS AND METHODS ............................................................................................... 59

Production and Purification of Recombinant AAV VLPs for Structural Studies ................... 59 Fv Antibody Modeling Using the Web Antibody Modelling (WAM) Server ........................ 60 Preliminary Neutralization Assays ............................................................................................. 61 Preliminary In-House Green Cell Assays .................................................................................. 61 Dot Blot Analysis to check Cross-Reactivity among VLPs ..................................................... 62 Generation of Fragment Antibodies from Monoclonal Antibodies ......................................... 62 Preparation of Fab:VLP Complexes .......................................................................................... 63

Page 6: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

6

CryoEM Data Collection ............................................................................................................ 63 Three-Dimensional Reconstruction of AAV:Fab Complexes .................................................. 63 Fitting of Atomic and Pseudo-Atomic Models into CryoEM Density Maps .......................... 64 Mutagenesis of Proposed Antigenic Regions ............................................................................ 65 Production and Purification of Antigenic Mutants ................................................................... 67

Western Blot Analysis ......................................................................................................... 70 AAV1 Enzyme-Linked Immunosorbent Assay (ELISA) ................................................. 70 Dot Blot Analysis for Antigenic Reactivity ....................................................................... 71 Pixel Intensity Calculations from Native Dot Blots .......................................................... 71 Infectivity Assays ................................................................................................................ 71 DNA Extraction and Real-Time Polymerase Chain Reaction .......................................... 72

3 CHARACTERIZATION OF ANTIBODIES AGAINST AAV1 AND AAV5 CAPSIDS, CROSS-REACTIVITY AND NEUTRALIZATION CAPABILITIES .................................. 74

Introduction ................................................................................................................................. 74 Results .......................................................................................................................................... 76

Production of Anti-AAV Monoclonal Antibodies and Isotyping..................................... 76 Cross-Reactivity among Different AAV Serotypes .......................................................... 77 Neutralization Ability of the Monoclonal Antibody Panel ............................................... 77

Discussion .................................................................................................................................... 78

4 DETERMINATION OF DOMINANT ANTIGENIC SITES ON THE CAPSID SURFACE OF ADENO-ASOCIATED VIRUS SEROTYPES 1, 2, 5, 6, and 8 THROUGH CRYO-ELECTRON MICROSCOPY STUDIES ................................................ 90

Introduction ................................................................................................................................. 90 Results .......................................................................................................................................... 93

Analysis of the Fab-VLP CryoEM Complexes ................................................................. 93 Fab G7 and AAV1 ............................................................................................................... 94 Fab D11 with AAV1 and AAV6 Capsids .......................................................................... 94 Fab C37B and AAV2 .......................................................................................................... 95 Fab F4 and AAV5 Capsids ................................................................................................. 96 Fab ADK8 and AAV8 Capsids ........................................................................................... 97 Fab ADK1a and AAV1 Capsids ......................................................................................... 98 Common Features of Antibody Binding Sites on the AAV Capsid ................................. 99

Discussion .................................................................................................................................. 100 Viruses as Antigens - Features Revealed ......................................................................... 100 Structures of Bound Antibodies and Possible Mechanisms of Neutralization or

Effects on Capsid Functions .......................................................................................... 101

5 MUTATIONAL ANALYSIS OF PROPOSED DOMINANT ANTIGENIC SITES ON THE CAPSID SURFACE OF ADENO-ASSOCIATED VIRUS SEROTYPE 1 ................ 111

Introduction ............................................................................................................................... 111 Results ........................................................................................................................................ 112

Generation of AAV1 Capsid Mutants .............................................................................. 112

Page 7: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

7

Analysis of Mutant Protein Expression ............................................................................ 113 Characterization of Mutant Capsid Production................................................................ 113 Quantification of Mutant Particle Titers, Infectivity, and Packaging Ability................ 114 Ability of Antigenic Mutants to Evade Monoclonal Antibody Recognition ................. 115 Direct Effects of Mutations on Neutralization ................................................................. 116

Discussion .................................................................................................................................. 117 Preliminary Analysis on Mutant AAV1 Viruses Reveals a Retained Ability to

Assemble Infectious Capsids ........................................................................................ 117 Ability of Antigenic Escape is Limited to Specific Regions on the Capsid Surface .... 120

6 SUMMARY AND FUTURE DIRECTIONS ......................................................................... 128

Summary .................................................................................................................................... 128 Future Directions ....................................................................................................................... 131

APPENDIX

A HUMAN BOCAVIRUS CAPSID STRUTURE: INSIGHTS INTO THE STRUCTURAL REPERTOIRE OF THE PARVOVIRIDAE ................................................. 133

Introduction ............................................................................................................................... 133 Materials and Methods .............................................................................................................. 136

Expression and Purification .............................................................................................. 136 Negative Stain Transmission Electron Microscopy ........................................................ 137 CryoEM and Image Reconstruction ................................................................................. 138 Generation of Density Maps for Representative Members of the Parvoviridae

Genera ............................................................................................................................. 139 Sequence Analysis and Generation of Unrooted Phylogenic Tree ................................. 140 Generation and Docking of a Homology Model of HBoV VP2 into Cryo-

Reconstructed Density ................................................................................................... 140 Comparison of Parvovirus VP2 Structures ...................................................................... 141

Results and Discussion ............................................................................................................. 141 HBoV Capsid Structure ..................................................................................................... 141 Pseudo-Atomic Model of HBoV VP2 Built Based on the B19 VP2 Crystal

Structure.......................................................................................................................... 143 HBoV VP2 Shares Low Sequence Identity but High Structural Similarity to Other

Parvovirinae VP2s ......................................................................................................... 145 HBoV Capsid Surface Topology is Unique, but Shares Features Common to Other

Vertebrate Parvoviruses ................................................................................................. 146 Summary .................................................................................................................................... 149

B MONOCLONAL ANTIBODY AGAINST THE ADENO-ASSOCIATED VIRUS TYPE 8 CAPSID AND THE IDENTIFICATION OF CAPSID DOMAINS INVOLVED IN THE NEUTRALIZATION OF THE AAV8 INFECTION ........................ 159

Introduction ............................................................................................................................... 159 Materials and Methods .............................................................................................................. 162

Page 8: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

8

Animals, Cell Lines and Cell Culture............................................................................... 162 Plasmids and Site-Directed Mutagenesis ......................................................................... 163 Plasmids and Peptide Insertions ....................................................................................... 164 Transfection of 293T Cells and Vector Particle Purification .......................................... 164 Purification and Titration of Mutant AAV Stocks .......................................................... 165 Analysis of Viral Protein Expression ............................................................................... 165 Production and Purification of Monoclonal Antibody ADK8 ........................................ 165 Determination of Antibody Isotypes ................................................................................ 166 AAV Capture ELISA ........................................................................................................ 166 In Vitro and In Vivo Neutralization .................................................................................. 166 Binding Analysis by DNA Dot Blot ................................................................................. 167 Immune Fluorescence ........................................................................................................ 168 Native Dot Blot Assay ....................................................................................................... 168 Epitope Mapping................................................................................................................ 169

Results and Discussion ............................................................................................................. 169 Three-Dimensional Reconstruction and Model Docking of the AAV8:ADK8

Complex.......................................................................................................................... 169 Analysis of the 3D Models Identifies Proposed Monoclonal Antibody ADK8

Binding Sites on the AAV8 Capsid .............................................................................. 170 Characterization of rAAV8- and rAAV2- Capsid Mutants to Elucidate the ADK8

Binding Epitope ............................................................................................................. 170 In Vitro and In Vivo Neutralization of AAV8 by ADK8 ................................................ 172 Neutralization Analysis of ADK8 .................................................................................... 174 Impact on rAAV8 vector generation ................................................................................ 176

LIST OF REFERENCES ................................................................................................................. 184

BIOGRAPHICAL SKETCH ........................................................................................................... 214

Page 9: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

9

LIST OF TABLES

Table page 3-1 Mouse monoclonal antibodies selected for further studies.................................................. 85

3-2 Amino acid differences between AAV1-like viruses and AAV1* ..................................... 85

4-1 Summary of AAV-Fab complexes generated for cryoEM data studies ........................... 104

4-2 CryoEM data reconstruction statistics ................................................................................ 104

4-3 Scaling and fitting statistics of cryoEM 3D reconstructions with atomic models ........... 104

4-4 Proposed epitopes for each respective complex including residue numbers and sequence data ........................................................................................................................ 104

5-1 AAV1 mutational nomenclature and sequence differences .............................................. 122

5-2 AAV1 single-site mutant characterization.......................................................................... 122

A-1 Selected properties of representative members of Parvoviridae ...................................... 151

A-2 Sequence alignment statistics for representative members with structural data .............. 151

Page 10: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

10

LIST OF FIGURES

Figure page 1-1 The antigenic structure of AAV2. ......................................................................................... 57

1-2 Panel of available AAV atomic structures. .......................................................................... 58

2-1 Coomassiee stained SDS-PAGE and EM images of purified AAV VLPs for structural studies. .................................................................................................................... 73

2-2 Images of the Fab WAM models. ......................................................................................... 73

2-3 Production, purification, and characterization of mAb and Fabs. ....................................... 73

3-1 Basic immunoglobulin structure ........................................................................................... 85

3-2 Monoclonal antibody characterization and cross-reactivity.. .............................................. 86

3-3 Preliminary neutralization assay data for the anti-AAV5 monoclonals. ............................ 87

3-4 Preliminary neutralization assay data for the anti-AAV1 monoclonals. ............................ 87

3-5 The ability of the anti-AAV1 monoclonals to neutralize the AAV1-like viruses.............. 88

3-6 Differnces between the AAV1 and X1 sequences are mapped to the atomic coordinates of AAV1 and cluster at the three-fold axis of symmetry. ............................... 89

4-1 CryoEM 3D reconstructions shown with respective roadmap of epitope positions on the 3D capsid surface ........................................................................................................... 105

4-2 Variable regions of the AAV-VP3 monomer ..................................................................... 106

4-3 Docking of atomic models into cryoEM density ............................................................... 106

4-4 Difference map between the AAV5:F4 reconstruction and a similar scale AAV5-uncomplexed map ................................................................................................................ 107

4-5 Three-dimensional reconstruction and model fitting for the AAV8:ADK8 complex ..... 108

4-6 AAV1:ADK1a reconstruction and model fitting ............................................................... 109

4-7 Illustration of overlapping antigenic regions at the capsid surface level .......................... 109

4-8 Structure based sequence alignment of AAV1, 2, 5, and 6.. ............................................. 110

5-1 Western blot analysis on single-site mutant cell lysates .................................................... 123

5-2 Purification of single-site antigenic escape mutants .......................................................... 123

Page 11: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

11

5-3 Electron micrograph images of AAV1 mutant capsids.. ................................................... 124

5–4 ADK1a native dot blot of mutant cell lysates. ................................................................... 124

5-5 Analysis of a native dot blot of single and double-site mutants........................................ 125

5-6 Infectious green cell assay images of NAb treated single-site mutants. ........................... 126

5-7 Histograms for in vitro Nab treated mutant virus. ............................................................. 127

5-8 Heparin-binding analysis. .................................................................................................... 127

A-1 Characterization of recombinant HBoV VLPs................................................................... 152

A-2 HBoV cryo-reconstruction. ................................................................................................. 153

A-3 Comparison of six representative parvovirus capsid structures. ....................................... 154

A-4 Radial density projections of six representative parvoviruses. ......................................... 155

A-5 Pseudo-atomic model of HBoV VP2. ................................................................................. 156

A-6 Superposition of VP2 structures from six representative members of the Parvoviridae. ........................................................................................................................ 158

B-1 Three-dimensional reconstruction and model fitting for the AAV8:ADK8 complex.. ... 178

B-2 Homology alignment of a portion of the G-H loop between AAV2 and AAV8. ............ 179

B-3 Analysis of rAAV2 and rAAV8 mutant vector production. .............................................. 179

B-4 Binding ability of rAAV2 and 8 mutant capsids by the ADK8 monoclonal.................... 180

B-5 in vitro and in vivo neutralization data ................................................................................ 181

B-6 Neutralization analysis of the ADK8 mAb......................................................................... 182

B-7 ADK8 and its impact on the N terminus externalization in the post-entry process. ........ 183

Page 12: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

12

LIST OF ABBREVIATIONS

AAV adeno-associated virus

AAV1 adeno-associated virus serotype 1

AAVS1 adeno-associated virus integration site S1

Ab antibody

AC artificial chromosome

Ad adenovirus

Ag antigen

BCR B-cell receptor

CC correlation coefficient

cDNA copy deoxyribonucleic acid

CF cystic fibrosis

CFTR cystic fibrosis transmembrane conductance regulator

Ch19 human chromosome chromosome 19

CNS central nervous system

cryoEM cryo electon microscopy

DMEM Dubelco’s modified eagle media

DNA deoxyribonucleic acid

ELISA enzyme-linked immunosorbent assay

Fab fragment antibody-binding region

FBS fetal bovine serum

Fc fragment crystallizable region

FV foamy vector

HFV human foamy virus

HIV-1 human immunodeficiency virus type 1

Page 13: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

13

HSPG heparan-sulfate proteoglycan

HSV herpes simplex virus

LAT latency associated transcript

mAb monoclonal antibody

mRNA messanger ribonucleic acid

mtRNA mitochondrial ribonucleic acid

MHC major histocompatibility complex

PBS phosphate buffered saline

PDGFR platelet derived growth factor receptor

pDNA plasmid dexoyribonucleic acid

pEI polyethleneinimine

PLA2 Phospholipase A2

PLL poly-L-lysine

RNA ribonucleic acid

RT reverse transcriptase

RU resonance units

SFV simian foamy virus

SIN sindbis virus

SPR surface plasmon resonance

TCR T-cell recptor

TMB tetramethylbenzodine

VEE Venezuelan equine encephalitis

VH heavy chain

VL light chain

VLPs viral like-proteins

Page 14: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

14

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

MAPPING DOMINANT ANTIGENIC REGIONS ON THE CAPSID SURFACES OF

ADENO-ASSOCIATED VIRIONS

By

Brittney L. Gurda

May 2010 Chair: Mavis Agbandje-McKenna Major: Medical Sciences – Biochemistry and Molecular Biology

The Adeno-associated virus (AAV) is a small ssDNA virus that can package and deliver

non-genomic DNA. Its low toxicity and relative lack of pathology, along with broad tropisms,

has made it popular as a vector for gene delivery trials. However, despite the critical role of

antibodies in the success of gene therapy with AAV vectors, the nature of the antigenic structure

of the AAV capsids is not well understood. Here we defined the binding of monoclonal

antibodies against AAV serotypes 1, 2, 5, 6, and 8 using cryo-electron microscopy and three-

dimensional image reconstructions. Docking of capsid structures with fragment antibody models

showed that the footprints on these viruses all fell on or around the three-fold protrusions of the

capsid regardless of the serotype examined. Significant overlap between the binding sites led to

the conclusion that this is a common region of antigenicity among the AAV capsid surfaces. In

preliminary studies, mutagenesis of the proposed sites on the AAV1 capsid showed that certain

surface loop changes resulted in viruses that were able to evade previous recognition from

parental antibodies. These data support the hypothesis that the three-fold axes are antigenically

important and that mutations in this region may avert a pre-existing immune response. Further

characterization is necessary to better design escape mutants and fully understand if these

mutants are still viable in their original tissue tropisms and transduction efficiencies.

Page 15: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

15

CHAPTER 1 BACKGROUND AND INTRODUCTION

Introduction to Genetic Disease and Gene Therapy

In the central dogma of molecular biology the normal flow of biological information is

identified: DNA can be copied to DNA (DNA replication), DNA information can be copied into

mRNA (transcription), and proteins can be synthesized using the information in mRNA as a

template (translation) (Crick, 1970). There are numerous factors involved in each stage of this

flow that assist in the final outcome, which often leaves many opportunities for mistakes to

occur. Evolutionarily we have a distinct advantage in that there are also many checkpoints that

aid in the correct outcome of these processes. Even so, a mistake can occur, but it is often not

evident at what point it occurred, particularily for uncharacterized and multigenic diseases. When

the fault occurs at the level of DNA, it is ultimately our genes that become altered. These

abnormalities are generally referred to as genetic disorders and can range from a small mutation

in a single gene to the addition or subtraction of an entire chromosome or set of chromosomes. In

single gene disorders a mutation causes the protein product of a single gene to be altered or

missing, i.e., in cystic fibrosis. Multiple gene disorders result from mutations in multiple genes,

often coupled with environmental causes, i.e., Alzheimer’s disease. In chromosome

abnormalities, entire chromosomes, or large segments of them, are missing, duplicated, or

otherwise altered, i.e., Down syndrome.

With the completion of the human genome project in 2003 and recent technological and

medical advances, the number of genetic disorders discovered has escalated. There are currently

no cures for these diseases and most are treated symptomatically. In rare cases, replacement

protein therapies can alleviate the issues associated with the knock down or loss of a specific

protein. Another option is gene therapy. In this DNA-based therapeutic, a functional gene is

Page 16: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

16

inserted or delivered to the cell in order to replace a dysfunctional one. Although this concept is

still in its infancy, many advances have been made over the last few decades that have identified

this option as a possible mainstream therapy. Gene therapy can be seen in two schools of

thought. The first, germ line therapy, takes place in the germ line cells, i.e., sperm or eggs.

Modifications are created by the introduction of functional genes, which are ordinarily integrated

into their genomes. Therefore, the change due to therapy would be heritable and would be passed

on to later generations. The second is aptly termed somatic gene therapy and is a more accepted

option that involves the passage of genetic material into the somatic cells of a patient. This

allows for alterations to the patient’s genetic material only and does not transfer genetically to

immediate or distant offspring (http://www.dnapolicy.org/science.gm.php). There are a variety of

different methods to replace or repair the genes targeted in gene therapy: (I) An abnormal gene

could be swapped for a normal gene through homologous recombination, (II) The abnormal gene

could be repaired through selective reverse mutation, which returns the gene to its normal

function or, (III) The regulation (the degree to which a gene is turned on or off) of a particular

gene could be altered (http://www.ornl.gov/sci/techresources/Human_Genome/medicine/

genetherapy.shtml). In disorders where mitochondrial DNA (mtDNA) is defective, a new

technique referred to as ‘spindle transfer’ can be used to replace entire mitochondria (Tachibana

et al., 2009). The most common approach is the delivery of a functional gene to a cell in context

of an episome. This approach requires the use of a vector for delivery of the gene into the desired

tissue.

Gene Delivery Systems

The delivery of naked DNA, or DNA-based drugs, has had limited success due to its poor

cellular uptake profile and biological stability, as well as a short half-life resulting in

unpredictable pharmacokinetics (Patil et al., 2005). Furthermore, a broad application of naked

Page 17: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

17

DNA–mediated gene transfer to gene therapy may not be conceivable because DNA, being large

in size and highly hydrophilic, is efficiently kept out of the cells in a whole animal by several

physical barriers. These include the blood endothelium, the interstitial matrices, the mucus lining

and specialized ciliate/tight junction of epithelial cells, and the plasma membrane of all cells

(Gao et al., 2007). The use of DNA delivery systems has not only improved the

pharmacokinetics of DNA-based therapeutics but has also achieved efficient targeted

introduction of these molecules into desired tissues (Patil et al., 2005). An ideal vector should be

safe, stable, easy to produce in large quantities, and capable of achieving efficient and tissue

specific gene expression when directly administered in vivo (Li and Ma, 2001). These guidelines

have aided in the development of numerous delivery methods. There are currently two types of

vectors for gene delivery; non-viral and viral vectors. Each system has its own set of advantages

and disadvantages in both the production aspect and clinical counterpart.

Non-Viral Delivery Methods

Non-viral delivery vectors are particularly suitable with respect to simplicity, ease of large-

scale production, and lack of specific immune response. The delivery of therapeutic DNA in the

absence of viral counterparts employs both physical and chemical approaches. The physical

approaches include needle injection, hydrodynamic, electroporation, gene gun, and ultrasound

delivery, which employ a physical force that permeates the cell membrane and facilitates

intracellular gene transfer.

Physical approaches

Direct injection of naked plasmid DNA (pDNA) intramuscularly has been shown to

transfer genes to myofibers, although the levels of gene expression were shown to be low (Wolff

et al., 1990). Intratracheal administration of pDNA in the mouse airway has been shown to be

safe and effective (Meyer et al., 1995), which warrants further study for use in pulmonary

Page 18: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

18

diseases where gene transfer could be beneficial. Studies involving intravascular injection

generally show limited transfer in all major organs (Li and Ma, 2001), however Budker et al.

(1998) reported that intravascular injection of naked DNA under high hydrostatic pressure in rats

led to high levels of foreign gene expression throughout all of the muscles of the targeted limb.

These studies led to the observation that levels of gene expression could be increased by

improving the method of injection, such as using hydrostatic pressure and clamping veins near

the major injection vein to increase uptake of pDNA.

Electroporation works through the notion that exposing cells to an intense electrical field

induces a transmembrane potential that facilitates cell permeabilization (Neumann et al., 1982).

A number of tissues have been examined including liver, skin, subcutaneous tumors, and muscle

(Li and Ma, 2001). Most reports showed 10- to 1000-fold differences in gene expression between

naked DNA injection and DNA injection followed by electroporation, and in many cases, a

broad distribution of transfected cells was also achieved (Li and Ma, 2001). Many tissues,

including liver cells, testes, skin, arteries, and tumors responded with high gene expression after

electropartion (Parker et al., 2003), but it is noted that tissues may respond differently to

electroporation due to the variation in their local biological environment (Li and Ma, 2001).

Typical voltages used in studies can be 1300 V/cm with 100 μs pulses (Heller et al., 2000)

however; ultra-short pulses of nanosecond (ns) durations and 10–300 kV/cm have been used for

electroporation (Schoenbach et al., 2001). Since markers of apoptosis, including

phosphatidylserine translocation and caspase activation have been observed with high-intensity

nanopulse electroporation it is thought that these methods could be useful for cancer/gene

therapy (Sundararajan et al., 2009). Besides local tissue damage, other drawbacks include the

Page 19: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

19

amount of DNA needed to reach therapeutic levels of transgene expression as well as

translational methodologies to include internal organs (Parker et al., 2003).

The gene gun was originally designed to deliver genes to plant tissues. It has shown

promise in animal tissues as well, albeit limited to superficial tissues. In this method, particles of

varying sizes (1-3µm) are coated in pDNA and expelled out by a powerful burst of air. The

bombardment of the particles through the tissues allows for delivery of the genes. This approach

has been reported to be superior to other methods of plasmid delivery to the skin, but tends to

generate a Th2 (humoral) rather than the Th1 (cytotoxic) immune response that commonly

follows intramuscular administration (Wells, 2004). For this reason, biolistic approaches are

heavily studied in the delivery of DNA vaccines.

Ultrasound is considered a non-invasive technique and has been used in the clinical setting

for diagnostic and therapeutic applications. The application of ultrasound results in acoustic

cavitation that can disrupt tissues and produce transient membrane permeabilization, thus

enhancing the delivery of plasmid to the cytoplasm (Wells, 2004). Nucleation agents such as

ultrasound contrast agents can enhance cavitation, and several studies have examined a range of

such contrast agents, concluding that albumin-coated octa-fluoropropane gas microbubbles

(Optison) is preferable to several other commonly available agents (Wells, 2004). Researchers

have reported successful delivery of naked pDNA at low frequencies in lung (Xenariou et al.,

2010), heart (Bekenedjian et al., 2003), tendon (Delalende et al., 2010), and skeletal muscle

(Taniyama et al., 2002). However, current levels of cellular transfection have not yet reached

therapeutic levels (Parker et al., 2003) using this approach.

Chemical methods

The chemical approaches use synthetic or naturally occurring compounds as carriers to

deliver the transgene into cells (Gao et al., 2007). Efforts to develop deleivery systems that

Page 20: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

20

mimic the desired properties of a viral vector has incorporated the use of cationic lipid and

protein carriers, termed lipoplexes and polyplexes, respectively.

Lipoplexes consist of a positively charged or cationic lipid outer layer that encloses

negatively charged DNA molecules. The positively charged lipid layer can self assemble around

the negative DNA giving these lipoplexes more effective transfection efficiency of the negatively

charged host cell membrane. These particles can be ~100-300nm in diameter (Midoux et al.,

2009; Parker et al., 2003) and usually vary in shape depending on the DNA: lipid ratios.

Internalization of the lipoplex is thought to occur through clathrin-mediated endocytosis

(Midoux et al., 2009) and its escape still remains a mystery. Several thoughts about their escape

from the endosome have been proposed, but it seems that a majority of the DNA complexes

become trapped in the endosome and are eventually destroyed when fusion with a lysosome

occurs (Midoux et al., 2009). This has greatly limited the efficiency of lipoplexes in gene

delivery and has led to the use of fusogenic lipids and protein, as well as the incorporation of

histidines and imidazole, in the complexes for improved endosomal escape (Midoux et al., 2009)

to the nucleus. Another drawback is the tendency of the negatively charged particle to become

associated with positively charged proteins and glycans in the extracellular milieu (Parker et al.,

2003). Researchers are also investigating the interaction of lipoplexes with membrane-associated

proteoglycans into order to better characterize the effect of cell surface receptors and trafficking

pathways on this approach (Mislick and Baldeschweieler, 1996).

Polyplexes are cationic polymers that have been mixed with DNA to form stable

complexes. Several polymers have been studied for use in gene delivery including linear, i.e.

poly-lysine (PLL), and branched, i.e. polyethyleneimine (pEI), as well as block and grafted

copolymers (De Smedt et al., 2000). These particles are often nanoparticulate (<100nm) and

Page 21: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

21

homogenous (Parker et al., 2003). Many of these studies have also involved the addition of

ligands to aid in targeting specific tissues, i.e. galactose for hepatocyte delivery (Zanta et al.,

2007). The uptake of polyplexes appears to happen through both clathrin-mediated and

cholesterol-dependent pathways, which can also occur simultaneously (Midoux et al., 2008).

Peptides that mimic viral proteins have also been used in conjunction with nanoscopic material

to help target delivery to and aid in passage across the cell membrane (Roy et al., 2009). The

major barrier is, as for lipofection, endosomal escape (De Smedt et al., 2000).

Protocols that use PLL have incorporated the use of endosome-lytic functions, such as

inactivated adenovirus or fusogenic peptides (Li and Ma, 2001). The most promising studies

have used pEI as it appears to have an inherent mechanism for escape into the cytoplasm (Parker

et al., 2003). Although the mechanism of endosomal escape is still not well understood there are

several ideas that have been proposed. These include an endosomal buffering capacity,

quenching of lysosomal acidification through protonation of amine groups, and an increase in

osmolarity leading to eventual rupture of the endosomal membrane (Parker et al., 2003).

Other novel non-viral delivery methods include transposons, where the advantage of virus

and naked DNA is combined, and artificial chromosomes (ACs). Both are highly appealing for

gene therapy. Transposons are naturally occurring with a desirable safety/toxicity profile and

integrate into chromosomes, providing long-term expression of the gene of interest in cells

(Izsvak et al., 2009). Current studies include recombinant forms of a fish transposon, Sleeping

Beauty, and have been fairly successful, but require further optimization of cellular transduction

(Ivics and Izsvak, 2006). On the contrary, ACs are autonomous and do not integrate into the host

chromosome. Although they offer escape from other issues surrounding gene therapy including,

Page 22: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

22

insertional mutagenesis and immune responses, they are highly complex, difficult to deliver, and

technically challenging to construct (Macnab and Whitehouse, 2009).

Optimization of non-viral vector delivery methods is a growing field due to the safety of

the vectors, no or low immune response, and their relative ease of production. In summary,

major issues that surround these methods include specific tissue targeting, cellular uptake,

trafficking to the nucleus, and stable translation. Many of these issues are being addressed by

studying and applying viral methods, such as the addition of viral peptides, which will provide

safer forms of delivery and give an advantage over viral vectors.

Viral Delivery Methods

Viruses have spent thousands of years evolving to deliver their genomes into our tissues in

order to express the proteins needed to continue their lifecycle. The application of viruses as

vectors in gene transfer involves members who naturally infect mammalian tissues and can be

used in diseases which are genetic or environmentally acquired (Lundstrom, 2003). Although

progress has been slower than expected, many advances have been made over the past two

decades which have lead to various successes and unfortunately, some failures (Flotte, 2007).

The engineering of integrating (retroviruses) and non-integrating (herpes, adenovirus, and adeno-

associated viruses) has made it increasingly obvious that there are no universally applicable ideal

viral vector systems available (Lundstrom, 2003). Several factors should be reviewed before the

application of a vector including the various features of each vector as well as the type of disease

being treated (Lundstrom, 2003). Brief discussions of several popular viruses currently being

studied as vehicles in gene delivery are given below, as well as the major advantages and

disadvantages of each vector.

Page 23: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

23

Retroviruses

Retroviruses have a large 7-10kb single-stranded RNA (ssRNA) genome which must be

reverse-transcribed in order for viral replication to occur. Retroviruses follow a special case in

the central dogma of molecular biology by first transcribing its mRNA to DNA. In order to do

this, the virus encodes for its own reverse transcriptase as well as an integrase, to assist with

integration of the viral genome into the host genome for transcription. These proteins are

translated from the polymerase or pol gene which also encodes for a viral protease. Retroviruses

are enveloped viruses which obtain their membrane from the host during cell budding. This

envelope is decorated with surface antigens that are encoded in the envelope or env gene and

expressed on host cell membranes. The capsid proteins and membrane associated matrix proteins

are found inside the membrane and are encoded by the group-specific antigen or gag gene

(Coffin, 1992). Several of these viruses have been studied for use as gene therapy vectors,

including members of the Gamma-retroviruses (Buchschacher, 2001), Lentiviruses (Valori et al.,

2008) and Spumaviruses (Trobridge, 2009). Pioneering of in vivo gene transfer involved the

retroviruses, namely murine leukemia virus (Lundstrom, 2004). The lentiviruses were used to

show a vector’s potential to transduce cells which have no or very low mitotic activity, and have

further since been developed for clinical application in neurological disorders (Lundberg et al.,

2008). Vectors based on human immunodeficiency virus type 1 (HIV-1) have been developed for

enzyme replacement and/or delivery of neurotrophic factors for neurodegenerative diseases and

CNS manifestations of lysosomal storage diseases, as well as delivery of glial cell line-derived

neurotrophic factor and ciliary neurotrophic factor for Parkinson’s and Huntington’s diseases

(Lundberg et al., 2008). These vectors have also shown great promise in the delivery of small

hairpin RNA for gene therapy in neurological disorders and transgenic knockdown animals

(Singer and Verma, 2008).

Page 24: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

24

The retroviruses offer the advantage of large packaging capabilities, the ability to

efficiently transduce non-dividing cells, and to the ability to maintain stable gene expression

(Cockrell and Kafri, 2007). Their ability to integrate into the host genome is appealing for stable

gene expression, but is limited to cell division and has also been shown to randomly integrate (Li

et al., 2002). In comparison to conventional retroviruses, the lentiviruses can infect both dividing

and non-dividing cells (Lundstrom, 2004). One of the greatest successes in gene therapy trials to

date was the ex vivo retroviral mediated transfer of the γc gene into CD34+ cells for X-linked

severe combined immunodeficiency disease (X-SCID) (Hacein-Bey-Abina et al., 2002).

Unfortunately, aberrant viral integration into the LMO-2 locus was found to cause leukemia in a

few patients and the trial was halted (Hacein-Bey-Abina et al., 2003). With the increase in

knowledge of retrovirus biology, especially with the lentivirus, the engineering of non-

integrating vectors continues (Sarkis et al., 2008; Valori et al., 2008). As toxicity of replication

competent vectors have increased, efforts to improve the safety and titers of recombinant

retroviral vectors have included the creation of replication incompetent vectors by the

introduction of cis-acting elements in trans during vector production (Sinn et al., 2005). Tissue

targeting is also an issue with many of the retroviruses due to their strict receptor specificity. To

address this issue, researchers have developed ‘pseudotyping’approaches, which can greatly aid

in re-trageting (Sanders, 2002). In this method, the retrovirus’ native envelope is replaced with a

helper plasmid expressing heterologous envelope glycoproteins, including lyssaviruses,

arenaviruses, hepadnaviridae, flaviviridae, paramyxoviridae, baculovirus, filoviruses, and

alphaviruses (Sinn et al., 2005).

The Foamy virus (FV) is a spumavirus, another subfamily of the retroviruses, and is

unique in that they have evolved to exhibit no pathology in their host during transmission and

Page 25: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

25

infection (Trobridge, 2009). The human foamy virus (HFV) was found to have evolved from the

simian foamy virus (SFV) and it appears that humans are an ‘accidental’ host (Meiering and

Linial, 2001). There is no evidence of human-to-human transmission and it also appears to be

contained among individuals working amid non-human primates with active infections (Meiering

and Linial, 2001). Their other promising attributes for gene therapy consist of a desirable safety

profile, a broad tropism, a large transgene capacity, and the ability to persist in quiescent cells

(Trobridge, 2009). FV vectors have been shown to efficiently transduce embryonic stem cells

and neuronal progenitor cells (Trobridge, 2009), and their resistance to human sera provides

further advantage for in vivo usage (Russell and Miller, 1996).

Adenoviruses

The Adenoviruses (Ad) are large, non-enveloped, double-stranded DNA viruses that are

commonly associated with upper respiratory tract infections and have been linked to other

illnesses such as gastroenteritis, conjunctivitis, cystitis, and rash (Horwitz, 1995). Positive factors

making Ad an attractive vehicle for gene transfer include their ubiquitous nature; over 100

serotypes have been isolated from many different species, their ability to transfect a broad range

of human cells, including non-proliferating cells, their ability to yield a high rate of gene transfer,

their relative low pathogenicity; causing mild symptoms associated with the common cold, their

large 7.5 kb genome, and finally, their genome stability that allows for the maintenance of

recombinant genes (Vorburger and Hunt, 2002).

Ad vectors were originally studied for use as vehicles for gene transfer in patients suffering

from cystic fibrosis (CF), a monogenic disease where a mutation disables the cystic fibrosis

transmembrane conductance regulator (CFTR) gene, due to their specificity for pulmonary

epithelial cells (Wilson, 1996). Adenoviral DNA was also found in liver, skeletal muscle, heart,

brain, lung, pancreas, and tumor tissue (Bramson et al., 1995), expanding the range of cell lines

Page 26: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

26

for gene delivery. Interestingly, when Ad vectors are administered intravenously, most of the

virus accumulates in the liver (Vorburger and Hunt, 2002).

Preliminary trials were met with several problems, including transient expression and a

host immune response (Wilson, 1996). It was eventually realized that viral products needed to be

limited in order to minimize the immune response and from this realization came the second

generation vectors. The Ad genome contains early genes which encode for regulatory proteins,

and late genes which encode for structural proteins. In order to make the Ad virus less

immunogenic, additional early genes were removed to stop the up-regulation of structural

proteins that would continue the viral lifecycle (Wilson, 1996). Unfortunately, these vectors were

still toxic due to low transcription levels of the remaining viral genes (Yang e al., 1995). One

recombinant strategy to improve the vector included a Cre-loxP helper-dependent (HD-Ad)

vector in which all viral coding sequences were deleted and a helper virus supplied the necessary

functions in trans (Parks and Graham, 1997). Another novel design in Ad vectors incorporated a

transposon system in which the gene of interest could be inserted into the host genome (Yates et

al., 2002). Other strategies include the creation of hybrid vectors, which combine the infectivity

of Ad with the integration processes of retroviruses or adeno-associated viruses, retargeting of

Ad to non-CAR-expressing cells, and promoter regulation at the packaged genome level

(Vorburger and Hunt, 2002).

Although many of these systems have improved toxicity and prolonged gene expression

(Schiedner et al., 1998), re-administration of the vector is still problematic. In most cases, an

initial immune response is seen upon first administration of the vector, and furthermore, most

adults have been exposed to Ad2 and 5, the two serotypes most commonly used in gene therapy

(Vorburger and Hurt, 2002). Because the Ad gene does not integrate into the host genome, vector

Page 27: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

27

re-administration is necessary, especially in chronic problems, like CF. However, this transient

display of gene expression has been found to be useful in issues where a patient may benefit

from an induced immune response, especially against cancer cells (Vorburger and Hunt, 2002).

Ad vectors are commonly used in clinical trials for cancer, including head and neck,

prostate, breast cancer, melanomas (Vorburger and Hunt, 2002), and ovarian cancers (Matthews

et al., 2009). Several unique vectors have been designed to target cancer cells and are termed

conditionally replicating adenoviruses or CRAds (Matthews et al., 2009). One of the best

examples is ONYX-015 (Khuri et al., 2000). This system is modified in the early gene region,

specifically, E1b, which normally allows the virus to bind and inactivate the p53 gene (Barker

and Berk, 1987). Ironically, this mutated virus cannot replicate in normal cells with a functioning

p53 gene, but can replicate and kill cancer cells that have a defective p53 gene (Vorburger and

Hunt, 2002). One of the first commercial gene therapy products, Gendicine, which was approved

by the State Food and Drug of China (SFDA) in 2005, is based on an Ad5 vector that expresses

p53 (Wilson, 2009). More than 20 kinds of cancer indications have been treated with Gendicine,

such as head and neck squamous cell carcinoma, lung cancer, breast cancer, and liver cancers

(Peng, 2005). Another Ad-based therapy, Ocorine (H-101) was also released in China alongside

Gendicine in 2005 (Shi and Zheng, 2009).

Herpesvirus

The herpes simplex virus (HSV) is one of the largest viruses being studied for use in viral

mediated gene delivery. The two most commonly studied human species are HSV-1 and 2,

which are usually associated with watery blisters on the lips, mouth, and genitals. The HSV

genome is single stranded DNA and encodes for ~100-200 genes. This relatively large, complex

genome is ~152kb and is encapsulated in an icosahedral shell. The capsid is then surrounded by

the tegument, which contains about 20 different proteins that have structural and regulatory roles

Page 28: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

28

(Marconi et al., 2009). The tegument is encompassed by a membrane that is obtained when the

virus leaves the host cell. This membrane is decorated with glycoproteins that are translated from

viral genes at the late stage of infection. The HSV infection is characterized by immediate-early,

early, and late gene translation. The appealing features of the HSVare not only the large

packaging ability, but also its neurotropism and ability to generate latent infections (Lundstrom,

2004). A latent infection is established in the neural ganglia through the expression of a Latency

Associated Transcipt. This allows the virus to maintain the host cell, by-passing cell death

mechanisms, in order to preserve a copy of the virus (Wang et al., 2005).

Full-length HSV are neurotoxic and are generally manipulated to be either replication-

competent attenuated vectors, replication-incompetent recombinant vectors, or defective helper-

dependent vectors known as amplicons. In attenuated vectors, the removal of several non-

essential genes that are involved in the replication, immune evasion, and virulence of HSV

allows for an alteration of the HSV virulence (Marconi et al., 2009). These have been tested as

live viral vaccines, as oncolytic viruses, and as gene therapy vectors to deliver transgenes to the

nervous system (Marconi et al., 2009). The replication-deficient vectors have mutated or deleted

essential genes that render the virus incapable of replication (Marconi et al., 2009). These vectors

are generated in transformed cell lines that offer the missing replication genes in trans which

allows for the generation of ‘second-generation’ vectors that have reduced toxicity associated

with them (Marconi et al., 2009). The removal of these genes also allows for larger foreign gene

products, or multiple genes under separate promoters (Marconi et al., 2009). The HSV-amplicon

system has gained significance as a versatile gene transfer platform due to its extensive transgene

capacity, widespread cellular tropism, minimal immunogenicity, and its amenability to genetic

manipulation (de Silva and Bowers, 2009). Among other components, the amplicon has only two

Page 29: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

29

non-coding sequences from the wild-type HSV-1 genome, which are required for replication and

packaging of the amplicon into infectious particles (de Silva and Bowers, 2009). Upon delivery

to cells, the genome remains episomal and does not integrate into the host system (de Silva and

Bowers, 2009). Since this would be lost during cell replication, several factors have been added

to the amplicon to allow for replication-competency or to enhance its episomal retention within

the transduced cell nucleus (Lufino et al., 2008). Packaging systems for the amplicons have also

been developed and included either helper-free or helper-dependent systems based on HSV

cosmids or HSV replication-deficient vectors, respectively (reviewed in de Silva and Bowers,

2009). Re-targeting of the HSV vector’s natural tropism has also been attempted through the

genetic manipulation of HSV envelope glycoproteins, namely gC, gD, and gB (de Silva and

Bowers, 2009). Conversely, restriction of the vector to its natural cell targets has also been

attempted by replacing envelope glycoproteins with ligands (de Silva and Bowers, 2009). HSV-

amplicons have been incorporated into therapeutic studies for disease of the CNS, including

Parkinson’s, Ischemia, and hereditary ataxia, as well as anti-cancer therapies (de Silva and

Bowers, 2009). The limitations for implementation of HSV-amplicons in clinical trials continue

to be vector production, extension of transgene expression (mainly conventional amplicon

vectors), and regulation of transgene expression (de Silva and Bowers, 2009). Overall, the major

issue with HSV vectors is its innate and humoral immune response in the host system (Flotte,

2007).

Alphaviruses

The alphaviruses have been found to infect a wide variety of hosts, including humans,

rodents, birds, and horses. Their overall structure consists of a 9.7-11.8kb ssRNA genome that is

housed in a nucleocapsid, which is surrounded by a membrane (http://www.expasy.org/

viralzone/all_by_species/625.html). They have a relatively simple genome that encodes for both

Page 30: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

30

structural and non-structural genes. The structural genes give rise to the capsid protein, as well as

the envelope glycoproteins (E1-3), while the non-structural genes are important for transcription

and replication. Alphaviruses, such as Semliki Forest (SFV) (Liljestrom and Garoff, 1991),

Sindbis (SIN) (Xiong et al., 1989), and one of the encephalitis-producing members, Venezuelan

Equine Encephalitis (VEE) (Davis et al., 1989), have all been used to develop vehicles for gene

transfer. There are several general features that make the alphaviruses appealing for gene transfer

including; positive-sense RNA, which allows for direct translation of the message, mass

production of viral mRNA which shuts down host protein synthesis and eventually leads to

apoptosis (may be beneficial in cancer therapy), their efficient packaging ability, and finally,

their affinity for a broad range of host cells (Ehrengruber and Lundstrom, 2007).

The alphaviruses have been used extensively for recombinant protein expression in cell

lines, in expression studies in neurons, and in vaccine production (Lundstrom, 2003).

Furthermore, it appears that while SFV and SIN have been developed further on the gene therapy

front, VEE has had more promise as a vaccine vector (Pushko et al., 1997; Lundstrom, 2004),

specifically as a DNA vaccine for bioterrorism (Lee et al., 2005; Dupuy and Schmaljohn, 2009).

Animal studies using SFV vectors have shown efficient gene transfer of reporter and therapeutic

genes to quite a few tumor models (Lundstrom, 2003). In the case of SIN vectors, a

morphological mutant vector has been shown to allow for an increase in packaging size from

~12 kb to an 18 kb cDNA insert (Nanda et al., 2009). It has been speculated that this

heterologous vector could possibly package and transcribe an RNA insert as long as 32 kb

(Nanda et al., 2009). Unfortunately, alphavirus is cytotoxic to most cell lines causing cell death

between 12 and 24 hours post-infection (Schlesinger, 2001). On the other hand, studies in insect

cell lines have shown that a persistent, non-cytopathic infection occurs and that virus production

Page 31: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

31

can reach up to a 100-fold higher compared to mammalian cells (Schlesinger, 2001). Vector

design has included the use of replicons that eliminates most of the structural proteins, as well as

the use of two defective helpers, which offers the advantage of overcoming the ‘copy-choice’

mechanisms of the replicase in order to avoid infectious particle production (Schlesinger, 2001).

Replicons with reduced cytopathogenicity have been identified that allow replication of the virus

in certain cell lines without causing cell death (Schlesinger, 2001). Further limitations of the

vector include transient transgene expression, a broad host tropism, and a neuron-specific

transgene expression (Lundstrom, 2003). In order to overcome these issues, advances in

targeting have included genetic alteration of the E2 glycoproteins (Schlesinger, 2001), which

determines binding to cell receptors (Dubuisson and Rice, 1993), in addition to encapsulation of

the vector into liposomes (Lundstrom, 2003). A phase I trial using encapsulated SFV-particles

produced no toxicity to the liposome or SFV-particle and allowed for repeat treatment with the

vector (Lundstrom, 2003).

Adeno-associated viruses

The adeno-associated viruses (AAVs) are the smallest of the gene transfer vectors, but

have had a large impact in the field of gene therapy. Although over 100 different genotypes of

AAV have been isolated from human and non-human primate tissues alone, only a handful of

them have been serotyped and studied through structural analysis (Gao et al., 2005; Xie et al.,

2002; Govindasamy et al., 2006; Nam et al., 2007, Agbandje-McKenna unpublished data). The

serotyped members include AAV1-12, with AAV2 being the best characterized and most studied

among them (Mori et al., 2004). The AAVs are members of the genus dependovirus and are

dependent on a larger DNA virus, like adenovirus, for replication. Ironically, they were

originally found as contaminants of adenoviral stocks (Atchison et al., 1965) and have been

continually isolated from a very broad range of hosts, including goats, birds, sheep, snakes, and

Page 32: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

32

cows (Olson et al., 2004; Luchsinger et al., 1970; Farkas et al., 2004; Clarke et al., 1979; Yates et

al., 1973). The AAVs are non-enveloped and have a very simple genome, encoding just two

open readings frames, rep and cap. Because of its simplicity, as well as relative lack of

pathogenicity, low toxicity, and ease of manipulation, the AAVs have been extensively studied

as vectors in gene transfer. These vectors are also able to infect dividing and non-dividing cells,

and can offer integration into a unique site, AAVS1, on human chromosome 19 (Ch19) under

certain constraints, i.e. inclusion of rep proteins and specific integration site sequences (Surosky

et al., 1997). The different serotypes have also been identified in animal studies as having

different specificities for different tissues. For example, AAV1 has shown to have the best gene

expression in the muscle, while AAV8 has repeatedly been superior in the liver (Thomas et al,

2004; Davidoff et al., 2005; Jiang et al., 2006; Wang et al., 2005). Although AAV9 has been

found to be best in cardiac tissue (Pacak et al., 2006), a recent study with AAV9 has identified an

ability to cross the blood-brain barrier resulting in widespread transduction in neurons

throughout the brain including the neocortex, hippocampus and cerebellum (Foust et al., 2009).

AAV vectors have already been tested in clinical trials for CF, hemophilia B, Parkinson’s

and Canavan disease, late infantile neuronal ceroid lipofuscinosis, arthritis, and an ex vivo study

to treat prostate cancer (Carter, 2005). The greatest success story involves a recombinant AAV2

(rAAV2) vector packaging the RPE65 gene for treatment in patients with Leber’s Congenital

Amaurosis (LCA). These studies have not only improved eyesight in both animal models and

human patients, but have also produced stable gene expression for over a year for continued

improvement of the disease (Bainbridge et al., 2008; Maguire et al., 2008; Hauswirth et al.,

2008; Cideciyan et al., 2009). Although these vectors have shown relative success in the field of

gene therapy they also have a few disadvantages. First, their small size allows for minimal

Page 33: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

33

packaging, and attempts to package more than 5 kb have been quite unsuccessful (Lai et al.,

2010). This limits the size of the therapeutic gene that can be packaged as well as the disease

targets, as many monogenic diseases can also involve large gene defects. Second, many of the

recombinant vectors are difficult to produce in the laboratory setting and efficient yields are

difficult to produce for large scale studies (Flotte, 2007). Third, wild-type AAV is known to have

an integration site on the human chromosome during latent infection (Kotin et al., 1990;

Samulski et al., 1991) and efforts to characterize this phenomenon will aid in the exploitation of

AAV integration (Daya et al., 2009). The development of integrating AAV vectors may also help

deal with the transient expression of AAV delivered therapies in dividing cells. Lastly and one of

the largest concerns, is the immune response against AAV vectors. Fortunately the immune

response is not lethal, but does raise concerns for re-administration of vectors and cell-mediated

death of transduced cells. Several trials in animal models have been able to express therapeutic

genes stably in target tissues often resulting in impressive levels of protein expression, but

eventually are overcome by an immune response in human trials (Wang et al., 2007; Zaiss and

Muruve, 2008). The types and extent of these responses will be addressed further in the

following sections, since the focus of this study is on the antigenic nature of the AAV capsid,

more specifically, the humoral response.

Current Status and Issues in Gene Therapy

To date, the Food and Drug Administration (FDA) has not approved any human gene

therapy products. Although many animal trials have been relatively successful (hemophilia B,

RPE, color-blindness), past human clinical trials have had moderate to little success. However,

success that has occurred throughout the last decade has gained gene therapy a runners-up title in

Science’s 2009 “breakthrough of the year” issue (The News Staff, 2009). Gene therapy is

considered experimental and still has many hurdles to overcome. The field has definitely grown

Page 34: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

34

in knowledge since its conception and there are many different approaches to the current issues.

These issues, which are briefly discussed in the section following, include short-lived gene

transfer, insertional mutagenesis, immune responses to the vector and the transgene, vector

toxicity, ability of viral vectors to revert to wild-type virus, horizontal gene transfer, and the

correction of multi-gene disorders.

Retroviral and lentiviral vectors appear to stand at the fore-front of integrating vectors due

to their inherent ability to integrate into the host genome. Although this allows for long-term

gene expression, there are concerns involving the efficiency and effects of integration.

Integration of the vector into host genes or regulatory elements can knock out the gene or cause

truncation products, or even lead to changes in expression patterns (Voigt et al., 2008). These

results can have devastating effects on the host, including causing cancer (Voigt et al., 2008).

The AAVs also have the ability to integrate into the host genome at Ch19 where a persistent

latency is established. Studies have shown that vectors containing rep gene were more efficient

at integration compared to vectors without AAV Rep (McCarty et al., 2004). The mechanisms

and efficiency for AAV integration have been studied by several groups (McCarty et al., 2004;

Daya et al., 2009; Linden et al., 1996) and have had difficulty in defining either the mechanism

or its actual efficiency, which is apparently very low (McCarty et al., 2004). However, these

studies have also identified important regions on Ch19 for integration as well as cellular

components and DNA sequences (McCarty et al., 2004; Daya et al., 2009; Linden et al., 1996).

One such study examines the rate of integration in mouse neonate livers. This study found that at

least 0.05% of hepatocytes contained rAAV integration, while 68% of integrations occurred in

genes, but none of them were near the mir-341 locus, the common rAAV integration site found

in mouse hepatocellular carcinoma (Inagaki et al., 2008). Of course, integration is not limited to

Page 35: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

35

viral vectors. Non-viral methods rely on the synthetic fish and frog transposons, Sleeping Beauty

and Frog Prince, respectively, for integration (Voigt et al., 2008). These allow for a DNA ‘cut-

and-paste’ mechanism that has been shown to be relatively safe in animal models as well as

human tissues (Voigt et al., 2008). Non-viral vectors still lack targeting efficiency, but may be

overcome by the combination of viral delivery with non-viral gene transfer. Most vector systems

to date lack well characterized integration and generally exist as episomes in host cells, which

are eventually lost in dividing cells or silenced (Voight et al., 2008). Efforts to stabilize episomes

include the removal of bacterial sequences and the inclusion of elements such as nuclear antigen

I, from the Epstein - Barr virus, and human scaffold or matrix attachment regions (S/MARs) can

prolong persistence of the episome (Wu, Xiao, et al., 2000; Piechaczek et al., 1999).

The use of viral vectors in gene delivery has many advantages, especially in targeting and

gene transfer efficiency, but their disadvantages are rather serious. The most obvious is that of a

host immune response and the toxicity that can ensue. Of course, the types and severity of these

factors will depend on the target tissue, mode and route of delivery, vector being used and doses,

as well as the immunocompetence of the host (Waters and Lillicrap, 2009). For one, delivery of

vector to the eye has shown only an attenuated response to new immunogens while systemic

delivery through the circulatory system results in activation of the innate and adaptive system at

various sites (Waters and Lillicrap, 2009). Although non-viral transfer has shown better tolerance

by the immune system, methods using hydrodynamic protocols have been shown to cause tissue

damage which activates inflammatory responses and subsequent reaction can include the

delivered transgene (Miao et al., 2006). Viral vectors are naturally immunogenic and most have

developed methods to evade detection by the host immune system. At large doses, like those

given in gene therapy settings, these reponses can be detrimental to the host or the therapeutic

Page 36: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

36

outcome. The adenovirus is a good example. This vector has great potential as it transduces

many tissues, including antigenic presentic cells (APCs), and although large doses up to 1 x 1013

are generally well tolerated in mice, studies in baboons highlight lethal toxicity (Di Paolo and

Shayakhmetov, 2009). Compared to adenoviral-vectors, the AAVs do not transduce APCs well

and therefore, do not tend to create a robust innate response (Waters and Lillicrap, 2009).

Although studies have shown that a cellular response can be elicited depending upon delivery

methods, transgene, and species (Vandenberghe and Wilson, 2007; Mingozzi et al., 2007; Lu and

Song, 2009). The AAVs have also been shown to generate neutralizing antibodies against the

vector capsid, which can cause issues upon re-administration using the same AAV vector (Zaiss

and Muruve, 2008; Huang and Yang, 2009). Interstingly, responses to lentiviral vectors injected

into animal tissues, such as non-human primate brain, cat eye, and mouse thymus have shown

no, or limited, immune response (Chang, 2009). However, the ability of lenti-vectors to

transduce APCs, such as dendritic cells, can result in immunological responses to the transgene

as it will most likely be displayed in either major histocompatability complexes I (MHCI) or

MHCII (Waters and Lillicrap, 2009).

The next issue would be the possibility of a recombinant virus reverting to its wild-type

disease causing form. An extensive amount of time and research has gone into protecting

ourselves from the very viruses that we have placed such high hopes in for gene delivery. Almost

all vectors studied today use minimal wild-type genome from the infectious viral parent and,

when needed can usually incorporate a helper replication system to provide replication proteins

in trans (Walther and Stein, 2000; de Silva and Bowers, 2009; Valori et al., 2008). Specifically

in the lenti-vectors, removal of as much viral sequence as possible is important because there are

several genes that can cause detrimental effects on cells including, oncogenesis (Tat), apoptosis

Page 37: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

37

(Vpr), MHC down regulation (Nef), and differentiation (Vpu) (Azzouz and Mazarakis, 2004).

This ensures that replication of the vector cannot occur in the patient and minimizes the cellular

toxicity seen in response to parental virus (de Silva and Bowers, 2009; Azzouz and Mazarakis,

2004).

Another concern is that of vertical gene transfer, or the transfer of these therapeutic

elements to the next generation. This line of thought breaches the Weismann barrier, proposed by

August Weismann in 1885. Weismann’s theory basically states that the germ cells are

responsible for heredity and that the somatic cells, or other cells of the body, do not influence

this heredity. Apparently there is enough evidence to suggest that there is a selective

permeability to this theory. One such study using retroviral vectors delivered angiotensin II type

I receptor antisense (AT1R-AS) cDNA to spontaneously hypertensive rats (SHR) and found that

subsequent generations had cardiovascular protection against hypertension as a result of a

genomic integration and germ line transmission (Reaves et al., 1999). Another study for

hemophilia using a viral delivery system was halted after viral DNA was identified in a patient’s

semen (Boyce, 2001). However, since semen contains other material, further testing was ordered

and isolated sperm cells were eventually found to be negative for the viral DNA (Boyce, 2001).

Katherine A. High, an M.D. and Howard Hughs Medical Institute investigator suggests that:

Even if foreign DNA gains access to a germ cell, in order for harm to result, it must lodge in a site where it can have an effect. Based on reasonable assumptions about the human genome and on data from a recent AAV trial, the risk of detecting a birth defect caused by gene transfer appears to be less than one in one million—a negligible increase over the baseline risk of birth defects of about one in one hundred (high-risk of gene transfer).

Among concern for vertical transmission is also horizontal gene transfer. In this case, an

organism receives DNA from another organism, without being the offspring of that organism.

Almost all vectors are typically gutted or have minimal elements retained in the vector construct,

Page 38: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

38

but this does not rule out the chance of viral integration, or homologous recombination events

leading to residual viral DNA elements. Interestingly enough, researchers have recently

identified the presence of bornavirus, a negative-sense RNA virus incorporated into the genomes

of multiple mammalian lineages and at different times, ranging from more than 40 million years

ago in anthropoid primates to less than 10 million years ago in squirrels (Horie et al., 2010;

Feschotte, 2010). This novel find, along with the data identified in gene transfer trials indicate

that viruses have indeed optimized themselves for future revival and could have profound

implications in viral vector mediated gene therapy.

An additional consideration is one of eco-pharmcovigilance. The excretion of active

pharmaceutical material in the environment has been going on since the introduction of these

compounds, but we have just recently begun to quantify their levels (Boxall, 2004). The best

known example is the effects of birth-control hormones in freshwater fish populations (Kidd et

al, 2007; Boxall, 2004). A workshop, held in October 2007 by the International Conference on

Harmonisation Gene Therapy Discussion Group, aimed to provide a better understanding of the

contribution of viral vector-shedding studies to the benefit-risk assessment of gene therapy

products (Kühler et al., 2009). Topics discussed included advantages and disadvantages of assays

used for detection of viral vector shed into excreta (Kühler et al., 2009). It is expected that more

sensitive assays should be developed to test for viral nucleic acids and among that, the detection

of transmission rates of shed vector to third-party persons or animals should be included in all

trials (Kühler et al., 2009).

Current strategies in gene therapy have focused on monogenic disorders or those resulting

from a single gene disorder, such as CF. These disorders have been well characterized and the

therapeutic target is generally one form of a gene. Unfortunately, most genetic diseases are the

Page 39: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

39

result of multigene disorders, such as heart disease, high blood pressure, and diabetes. These

diseases pose multiple issues for gene therapy due to targeting and coordination of multiple gene

delivery. More complicated diseases will most likely need to be well characterized so that the

molecular factors involved can be fully corrected. Regrettably, it seems that many of these

disorders, monogenic and multifactorial alike have environmental factors involved that may not

be reversible with gene therapy. The stage of disease will also make this more difficult, as many

effects of disease, such as tissue damage, are limited in reversibility depending on the extent of

damage. However, for the more characterized diseases, prevention may be the best cure here and

may at some point involve gene therapy of a single disorder at a time.

It is obvious, from even the small amount of information presented here, that the issues in

gene therapy are quite numerous and still require more detailed knowledge before a solution may

be found. Depending on the nature of the disease in question, therapies through gene transfer will

most likely not be able to rely on one method alone. An interesting combination is that of RNAi

and Ad vectors, in which poor in vivo gene transfer of RNAi alone was overcome with the use of

an Ad delivery system (Lundstrom, 2004). Immune responses may be overcome with the

encapsulation of viral vectors in liposomes (Lundstrom, 2003), while pre-existing responses may

be overcome by the use of vectors that have not been seen by the human patient, i.e. Goat-AAV

(Arbetman et al., 2005). It may be that stable inducible, gene expression is achieved through the

combination of well characterized integrating vectors combined with drug-regulating

transcription (Auricchio et al., 2002). There seem to be endless possibilities and multiple

combinations that have yet to be tried or optimized. Unfortunately, beyond the specific concerns

associated with each method still exists the translation of observed therapies in animal models to

the human patient. This major obstacle is currently being tackled as researchers advance to larger

Page 40: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

40

animal models and aim to further characterize the toxicity of introducing foreign genes and

proteins into our system. As one last point, beyond the biological concerns, are also the ethical

concerns of human modification and enhancement. These issues are beyond the strictly

biological aspect of this study, but are definitely another barrier in the advancement of gene

therapy and many of the techniques surrounding this modern marvel.

As the focus of this project is on the antigenic nature of viruses as a vehicle for gene

therapy, specifically the AAVs, it is necessary to look further at the events surrounding an

immune response. As these vectors are foreign to our systems, we have adapted to respond to

them for our own good. On the first insult our bodies will naturally work to clear the infection,

generally beginning with a cellular response. The cellular response will then alert the humoral

responses which will aid in continued protection through antibody production and memory

responses. This memory response will be a factor if treatment is needed again. As mentioned

earlier, many of the current therapies are transient and will require a secondary, or even, more

consistent delivery. This type of treatment will require superior engineering to evade the immune

responses and it is our hope that the data generated here will eventually add to the understanding

of the antigenic nature of viral vectors and their further exploitation and manipulation for

successful gene delivery therapies.

Antibody Recognition of Viruses and Viral Neutralization

Antibodies are critical for the control of many viruses and generally target proteins on the

viral surface or on the surface of infected cells. Neutralization of a virus by these antibodies is

defined as “the loss of infectivity which ensues when antibody molecule(s) bind to a virus

particle, and usually occurs without the involvement of any other agency” (Burton, 2002). The

protective effects of neutralizing antibodies can be achieved not only by neutralization of free

particles, but also by several activities directed against infected cells. Although there is much

Page 41: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

41

controversy surrounding the production and mechanisms of neutralizing antibodies (Burton,

2002; Dimmock, 1984; Burton et al., 2000; Klasse and Sattentau, 2002), they remain imperative

to most vaccine-mediated protective strategies.

Efficient B-cell induction and generation of B-cell memory requires two signals. The first

is mediated by the B-cell receptor (BCR) while the second signal is usually provided by T-cells.

It has been postulated that extensive BCR cross-linking can activate B-cells to produce IgMs

without the aid of T-cells. Antigens that conatin highly repetitive epitope have been correlated to

these specific B-cell responses (Bachmann and Zinkernagel, 1996). In fact, it has been suggested

that for an antigen to be T-cell independent it must contain a threshold number of appropriately

spaced epitopes in order to stimulate the B-cell receptor (Dintzis et al., 1983). This includes all

non-enveloped viruses, especially those that have a highly organized icosahedral structure, like

AAV. Interestingly, in hepatitis B (which is an enveloped structure) studies identified the

nucleocapsid antigen to generate both T-cell dependent and independent response, while the

surface antigen is T-cell dependent (Milich and McLachlan, 1986). Any antigen may have

several epitopes, or specific sites that are recognized by a lymphocyte or an antibody. Epitopes

for antibodies on proteins can be linear or continuous, like those recognized by T-cell receptors

(TCRs) or they can be made up of different regions of a polypeptide that come together in a

three-dimensional structure. The latter is often referred to as a discontinuous or a conformational

epitope. However, not all conformational epitopes are discontinuous (Harris et al., 2006). Indeed,

linear stretches of residues have been identified in conformational epitopes where the unique

structure of the paratope, like a loop-helix motif, plays an important role in recognition (Harris et

al., 2006). These antibodies are generally raised against native folded proteins, but may

recognize fragments of the protein if the proper binding elements of the structure are retained.

Page 42: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

42

When the heavy-chain (Hc) and light-chain (Lc) regions of the antibody’s variable region

are paired their hypervariable loops form the complementarity-detemining regions or CDRs.

These regions determine antigen specificity by forming a surface complementary to the antigen.

The formation of these shape complementary regions is an important determinant of the

antibody-antigen interaction along with solvent accessibility and contact energies (Rapberger et

al., 2007). This interaction can be disrupted by extreme salt concentrations, large pH shifts,

detergents, and even competition with high concentrations of pure epitope. The binding is

therefore a reversible, non-covalent interaction that involves electrostatic forces, hydrogen

bonds, Van der Waals forces, and hydrophobic interactions. A striking difference seen in

antibody-protein interactions is the involvement of aromatic residues (Janeway et al., 2005).

Several groups have addressed if an unusual amino acid composition exists in antigen-antibody

interactions. Studies involving the physical properties of these interfaces have revealed that high

percentages (34%) of the antibody-contacting residues are aromatic, with tyrosines and

tryptophans in the CDRs being more solvent exposed (Davies and Cohen, 1996). In contrast, the

contacting residues on the antigens contain relatively few aromatic amino acids (Davies and

Cohen, 1996). The pattern of binding that emerges is similar to that found in other protein-

protein interactions, with good shape complementarity between the interacting surfaces (Davies

and Cohen, 1996).

Viruses are eliminated by the host through many different mechanisms, including

cytokines, cytotoxic T-cells (CTLs), complement, and antibodies. It follows that viruses that are

efficiently controlled by antibodies are under a direct evolutionary pressure to form serotypes

(Bachmann and Zinkernagel, 1996). These serotypes exist so that neutralizing antibodies against

serotype A do not work against serotype B, thus an individual can be infected by two (or more)

Page 43: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

43

serotypes despite the pre-existence of neutralizing antibodies to the first. The existence of many

different antigenic forms of viruses (e.g. Rhinoviruses) makes them harder to control as they

often have the ability to mutate rapidly to generate many different variants in a single host (e.g.

HIV-1 in humans). The ability of viruses to produce structural variation while maintaining

function and the effectiveness of the antibody to neutralize are other factors involved in their

response. The success of a vaccine will be determined by an antibody’s ability to neutralize and

the subsequent protection after a natural infection. However, in the case of viral-vectored gene

therapy the ability of engineered viruses to escape neutralization may be beneficial to patients

with pre-existing immunity or those in need of repeated treatment with the same vector.

Antibodies directed at viruses may be able to completely block infectivity after direct

binding to the virion, while others are less efficient. Mechanisms of neutralization can be

classified according to the event in virus replication that they block. Studies have noted antibody

interference at cellular attachment sites, post-attachment interactions of the virus with receptor

and co-receptor, internalization, and fusion at the cell surface or in endosomal compartments of

enveloped virions (Klasse and Sattentau, 2002). Additional mechanisms can occur in vivo

including Fc-mediated phagocytosis, complement binding and activation, opsonization, and

antibody-dependent cellular cytotoxicity of infected cells (Burton, 2002). It has also been noted

that some antibodies neutralize through mechanisms that have yet to be explored. Apparently

some antibodies to Sindbis virus and poliovirus can eliminate infection or reduce progression in

vitro even when added to cells several hours post-infection (Smith, 2003). There is also evidence

that some antibodies show non-neutralizing activity in vitro, but offer protective immunity in

vivo (Schmaljohn et al., 1982; McCullough et al., 1992). Although neutralizing antibody is one

of the main arms of protective immunity and the type that is often desired in vaccination, there is

Page 44: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

44

surprisingly little known about these interactions. Some of the best studied examples of

neutralization include many common human pathogens, such as HIV-1, rhinovirus, influenza

virus, and poliovirus (Reading and Dimmock, 2007; Huber and Trkola, 2007; Smith, 2001). The

list has recently grown to include emerging pathogens like West Nile, Hendra, and Nipah virus

(Reading and Dimmock, 2007; Oliphant and Diamond, 2007).

Parvovirus Capsid Structures, Receptor Binding, and Infection

It appears that the immune responses seen among patients and animals alike are ultimately

dependent upon the structural nature and infectivity profile of the insulting virus, or vector. Since

this work is dedicated to characterizing the immune responses seen in our vector of choice,

AAV, it is best to have a detailed look at the infectious pathway and structural nature of this

virus in comparison with other family members and in context of the host.

Members of the family Parvoviridae infect a variety of hosts, ranging from insects to

primates, predominantly causing disease in young vertebrates. These viruses are non-enveloped

with a capsid shell of ~260 Å in diameter that exhibits a T=1 icosahedral symmetry. They are

made up of 60 copies of between two and four overlapping capsid proteins designated viral

protein (VP) 1-VP4 (Table A-1). The autonomous parvoviruses (including minute virus of mice

(MVM) and canine parvovirus (CPV)) are composed of ~50 copies of VP2, while the adeno-

associated viruses (AAVs), a member of the genus Dependovirus, contain mostly VP3. The VP1

protein makes up about 5% of the capsid and has been reported to contain phospholipase A2

(PLA2) activity in the VP1 unique region, as well as basic sequences which appear to act as

nuclear localization sequences (Girod et al., 2002; Zadori et al., 2001; Grieger et al., 2007). The

AAVs are also composed of ~5% of an alternatively spliced protein, also called VP2, which is

reported to not be required for infectivity. Parvovirus capsids are similar in that there is one

capsid protein that forms the majority of the structure, as well as minor proteins that form the

Page 45: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

45

same core structure, but have additional sequences on their amino-termini. Despite differences in

sequence there are certain structural elements that have been identified as common among the

parvoviruses. These include raised regions at the five-fold axes of symmetry (Figure A-3),

depressed regions or canyons surrounding the five-fold axes, one or three protrusions at or

surrounding the three-fold axes of symmetry (three-fold spikes or peaks; Figure A-3), and

depressed or dimpled regions at the two-fold axes of symmetry (Chapman and Agbandje-

McKenna, 2006). All the viruses have a superimposable core eight-stranded β-barrel domain that

forms the interior of the capsid (Figure A-6A).

The parvoviruses use a variety of proteins and carbohydrates as receptors or co-receptors

for infectivity and entry. Sialic acid has been shown to be important for infectivity of MVM and

several of the AAV serotypes. However, studies have identified that an AAV serotypes’ ability

to bind sialic acid depends on the linkages involved in the carbohydrate chemistry; AAV4 binds

2,3-O-linked sialic acids, and AAV1, 5, and 6 bind both 2,3- and 2,6-N-linked sialic acids

(Kaludov et al., 2001; Seiler et al., 2006; Wu et al, 2006). Receptors and co-receptors for AAV2

include heparin sulfate proteoglycan (HSPG), human fibroblast growth factor receptor-1, αvβ5

and α5β1 integrins, and hepatocyte growth factor receptor (c-met) (Qing et al., 1999; Qiu and

Brown, 1999; Summerford et al., 1999; Kashiwakura et al., 2005). Platelet-derived growth factor

receptor (PDGFR) is the protein that mediates infection for AAV5 (Di Pasquale et al., 2003)

while the transferrin receptor has been reported as a receptor for CPV and feline panleukopenia

(FPV) (Parker et al., 2001). The sites for interaction of some of these receptors with the

parvovirus capsid have been mapped either by mutagenesis or through direct interactions at the

structural level. In AAV2 HSPG binds to the three-fold axis of symmetry, whereas for CPV the

transferrin receptor attachment site is near the top of a raised region around this three-fold axis of

Page 46: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

46

symmetry (Opie et al., 2003; Kern et al., 2003; Hafenstein et al., 2007; O’Donnell et al., 2009;

Levy et al., 2009). Mutations in AAV2 have been used to alter binding specificity and infectivity

(Rabinowitz et al., 1999; Wu, Xiao, et al., 2000; Xu et al., 2005; Lochrie et al., 2006; Shi et al.,

2006). The receptor attachment sites are still unknown for the other AAV serotypes.

Although the attachment sites vary among different parvoviruses, it is thought that they all

enter by receptor-mediated endocytosis (Weitzman, 2006). Entry of AAV2 and CPV into the cell

occurs by endocytosis through clathrin-coated pits and is inhibited by overexpression of a

dominant interfering mutant of dynamin (Duan et al., 1999; Parker and Parrish, 2000; Bartlett et

al., 2000). After uptake into the cell the pathway is less well characterized. Acidification has

been suggested as being necessary for infectivity by using drugs that disrupt endosomal pH

(Bartlett et al., 2000; Ros et al., 2002; Basak and Turner, 1992). Escape from the endosome

seems less efficient, but studies identify a release of the VP1 unique region and its associated

PLA2 activity, allowing the virion to enter the cytoplasm (Girod et al., 2002; Suikkanen et al.,

2003; Zadori et al., 2001). The capsid is then proposed to move through the cytoplasm by

microtubule-mediated processes to the perinuclear region, the capsid then enters the nucleus

possibly via a NLS that is found in the already exposed VP1 unique region (Ros and Kempf,

2004; Vihinen-Ranta et al., 2002; Girod et al., 2002; Greiger et al., 2007). Modifications to the

capsid during transport may also result in the exposure of sequences that aid in transport through

the nuclear membrane (Cotmore, 1999). Different entry and transport patterns by the different

parvoviruses may also exist. For example, transduction of cells by AAV capsids can be enhanced

by inhibitors of proteosomal degradation proteases, while the MVM and CPV capsids are not

affected by those inhibitors (Denby et al., 2005; Yan et al., 2002, Ros et al., 2002, Yan et al.,

2004).

Page 47: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

47

Parvovirus Antigenic Properties

There have been many studies involving antibody binding and neutralization of the

parvoviruses, most notably on MVM, CPV, FPV, AAV2, and human parvovirus B19 (B19).

These viruses differ in their host range, pathogenic nature, and capsid protein composition, but

still have structural similarities that determine the antigenic response due to infection (Agbandje-

McKenna and Chapman, 2006). The mechanisms of antibody induction, binding, and

neutralization are not well understood, but the data presented in this work adds to the knowledge

in this field and may eventually aid in their definition. It appears that the elaborate loop regions

between the strands of the viral β-barrel, which make up the protrusions at or surrounding the

icosahedral threefold axes and the walls between the depressions at the two- and five-fold axes,

are the most immunogenic sites in the parvovirus capsid (Agbandje et al., 1995; Govindasamy et

al., 2003; Bloom et al., 2001; Xie et al., 2002; Simpson et al., 2002; Strassheim et al., 1994).

Studies of CPV and MVM have used escape mutations from neutralizing mAbs to map epitopes

on the CPV, FPV and MVM capsids (Lopez-Bueno et al., 2003; Strassheim et al., 1994), while

others have used peptide scans and ELISA based assays to map immunodominant regions on the

parvovirus capsids, including AAV2 (Wobus et al., 2000; Brown et al., 1992). Structural studies

based on CPV and FPV have also used cryoelectron microscopy (cryoEM) to confirm previously

identified antigenic regions on these capsid surfaces (Hafenstein, 2009), much like this study has

done with AAV. The capsid surface of the insect infecting densoviruses appear quite smooth

compared to those of the viruses infecting vertebrates (Figure A-3) (Simpson et al., 1998;

Bruemmer et al., 2005), suggesting that the raised structures in the vertebrate viruses provide a

benefit when the virus is under antibody selection. These data also confirm the idea that only

viruses that are efficiently controlled by antibodies are under evolutionary pressure to form

serotypes (Bachmann and Zinkernagel, 1996) as described below for the parvoviruses.

Page 48: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

48

Parvovirus Antigenic Structures for Autonomous Viruses

FPV isolates infect cats, mink, and other carnivores, while CPV infects dogs and close

relatives, such as wolves, coyotes, and Asiatic raccoon dogs. CPV emerged in 1978 and the

original strain (CPV type-2) was replaced between 1979 and 1981 by a genetically and

antigenically distinct virus (type-2a), which has recently been replaced by further variants

(Parrish, 2006). FPV and CPV differ antigenically in at least 1 epitope, and after emerging in

dogs CPV later gained 3 or 4 additional mutations in the VP2 gene which changed 2 different

epitopes (Parrish et al., 1991; Parrish et al., 1985). There are close connections between the host

ranges of the viruses and their antigenic structures and variation. The effects of antibody binding

on the capsid surface of CPV and FPV have been previously examined (Parrish and Carmichael,

1983; Strassheim et al., 1994; Wikoff et al., 1994). Efforts to map epitope regions on the capsid

have included the use of natural variants or mAb-selected escape mutant analysis (Strassheim et

al., 1994; Wikoff et al., 1994), peptide mapping, and cryoEM analysis with image reconstruction

of Fab:capsid complexes. Most of these studies have identified two major regions of antigenicity

on the virus, while a recent structural study using cryoEM has confirmed these regions

(Hafenstein, 2009). Site A is close to the top of the three-fold spike of the capsid, while site B is

located on the shoulder of the three-fold spike (Chang et al., 1992; Langeveld et al., 1993; Lopez

de Turiso et al., 1991; Strassheim et al., 1994). The capsid regions around epitopes A and B also

show the greatest structural variation between CPV and FPV, and their host range and antigenic

mutants (Agbandje et al., 1993; Govindasamy et al., 2003; Llamas-Saiz et al., 1996; Simpson et

al., 2002). These methods have determined that VP2 residue 93 in epitope A and 299 and 300 in

site B determine the canine host range of the viruses, by controlling the interaction of the capsid

with canine and feline TfRs (Govindasamy et al., 2003; Hueffer et al., 2003). Antibodies to these

regions preclude receptor attachment ultimately causing neutralization.

Page 49: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

49

Studies of MVM during long term immune therapy with a single neutralizing mAb in

immunodeficient mice resulted in the isolation of mutant viruses escaping neutralization through

single amino acid changes within the VP2 protein, residues 433-439, which are located on a

surface-exposed loop at the three-fold axes of the capsid (Lopez-Bueno et al., 2003). Recent

studies using cryoEM have led to the image reconstruction, at 7Å resolution, of the structure of

the immunosuppressive strain of MVM (MVMi) bound to the neutralizing antibody, B7

(Kaufmann et al., 2007). The VP2 residues involved in the contact are 225, 228, 229, 425, 426,

and the loop 432-440, which correlate with the escape mutant findings. The structural results,

together with the analysis of B7 binding and neutralization activity, suggest that this antibody

may hinder conformational transitions in the viral capsid required for the MVMi infection

process (Kaufmann et al., 2007).

Antigenic studies on Aleutian mink disease virus (AMDV) have revealed some rather

interesting results. Adult mink infected with AMDV develop a persistent infection associated

with high levels of antiviral antibodies and hypergammaglobulinemia (Aasted et al., 1984;

Bloom et al., 1994). In spite of this robust immune response, virus is not eliminated in vivo and

complications usually arise. Furthermore, antiviral antibody enables AMDV to infect cells such

as macrophages via an Fc-receptor-dependent mechanism termed antibody-dependent

enhancement (ADE) (Bloom et al., 2001). It is also interesting to note that antiviral antibody

does not protect adult mink from infection, but rather, leads to an accelerated form of disease

upon challenge (Aasted et al., 1998). Sites on the AMDV capsid have been mapped through

peptide studies (Bloom et al., 1997; Bloom et al., 2001; McKenna et al., 1999) to regions similar

for AAV (see below) and CPV/FPV, located on the wall between the two-, three-, and five-fold

axes, and on the inner wall of its three-fold mounds. Immunoreactive spans of peptides were

Page 50: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

50

located in VP2 residues 429-524, which correlates with a residue span of 428-446 that is

recognized by infected minks (Bloom et al., 1997). This region is located in the wall of the two-

fold depression, and antibodies directed at this peptide, although elicit ADE, are able to

neutralize infectivity in CrFK (feline kidney origin) cells. However, another well characterized

epitope spanning from residues 487-501 at the inner wall of the mounds of the icosahedral three-

fold axes, is capable of inducing ADE, but is not neutralizing. The observation that the same

amino acid sequence can both neutralize virus and induce ADE may explain the inability of

capsid-based vaccines to protect against AMDV infection (Bloom et al., 2001).

B19, a member of the erythrovirus genus, is a human pathogen that predominantly infects

young children with mild flu-like symptoms and a characteristic red rash that gives the

appearance of a “slapped cheek”. The disease usually runs its course with no complications, but

has been known to have adverse effects in pregnant women and those with immune disorders.

The B19 capsid epitopes have been mapped to the VP1 unique region and to the VP1/VP2

overlapping sequence, and most of the VP1/VP2 neutralizing epitopes are located on the loops

that make up the protrusions on the B19 capsid surface (Kaufmann et al., 2004; Rosenfeld et al.,

1992; Sato et al., 1991; Yoshimoto et al., 1991). Antibodies that recognize VP2 residues 57-77,

345-365, and 446-466 inhibit hemagglutination by B19 (Brown et al., 1992), while those

generated for peptides with the residues 253-515 are neutralizing. Mapping these residues onto

the crystal structure of B19 identifies residues on the wall between the two-and five-fold

depressions and at the base of the three-fold protrusions. The loop containing residues 309-319 is

disordered in the B19 structure (Kaufmann et al., 2004), but residues 314-330 structurally

superimpose onto a portion of the C37B epitope for the dependovirus AAV2 (discussed below)

(Wobus et al., 2000).

Page 51: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

51

The Dependoviruses and their Antigenic Properties

The genus dependovirus houses the AAVs, which share a common, but not absolute

requirement for an unrelated DNA helper virus (e.g. adenovirus, herpesvirus, papillomavirus,

vaccinia virus) to aid in the completion of their otherwise replication deficient life cycle

(Muzyczka and Berns, 2001; Bowles et al., 2006). Phylogenic analysis now places the

autonomously replicating goose (GPV) and duck parvoviruses into this genus where they cluster

with the avian adeno-associated virus (AAAV) (Tattersal, 2006). Probing studies have lead to the

identification and cloning of 53 new AAV capsid DNA sequences from non-human primates

(Gao et al., 2002; Gao et al., 2003) and 64 from human tissues (Gao et al., 2004). The

identification of so many isolates has brought about the need for a classification system in order

to move the field forward. The isolates have therefore been divided by three different

classification systems based on differences in the capsid: serology, transcapsidation, and clades

based on genetic relatedness. Much of the virus variation is concentrated into the regions of the

sequence that make up the surface exposed loops of the capsids, and they therefore are mostly

antigenically distinct when tested with polyclonal antibodies, being classified as AAV1 through

AAV11. There are currently six clades; A,B,D,E, and F, which house AAV1, AAV2, AAV7,

AAV8, and AAV9 respectively, while the AAV2/3 hybrids belong to Clade C. AAV4, AAV5,

do not fit any of the clades and are regarded as clonal isolates (Gao et al., 2004). Serology is no

longer used to group the AAVs, but the isolates still fall into serologically distinct ‘groups’.

In addition to their genetic and antigenic differences, the AAV serotypes exhibit a wide

range of distinct biological properties including receptor binding and tissue tropism (Chiorini et

al., 1997, 1999; Xiao et al., 1999; Davidson et al., 2000; Rabinowitz et al., 2002, Choi et al.,

2005). Their dependency upon helper viruses has generated a promiscuity among the AAVs that

enables them to be in many different cell lines awaiting helper functions. In the absence of this

Page 52: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

52

helper, the virus has the ability to integrate into the host chromosome to set up a latent infection.

This relatively simple strategy is not harmful to the host and has not been associated with any

known disease or pathogenicity. These qualities have enabled the development of recombinant

AAVs for use as vectors in gene therapy. Recombinant gene transfer vectors based on AAV2

have proven effective in animal models for the correction of genetic diseases of the eye, brain,

muscle, liver, and lung (Warrington and Herzog, 2006). However, numerous studies have shown

that other serotypes possess unique tissue tropisms and have improved gene transfer activities

compared to that of AAV2 for some cell types and are also being studied to avoid the pre-

existing antibodies that are widespread in the human population, or which develop after primary

AAV treatments and which reduce the efficacy of subsequent treatments (Jooss and Chirmule

2003; Zaiss and Muruve 2005, Calcedo 2009). AAV1, for example, can transduce rodent skeletal

muscle more efficiently than AAV2 (Xiao et al., 1999). AAV4 has strong tropism for ependymal

cells in the central nervous systems of mice (Davidson et al., 2000; Liu et al., 2005) and for

retinal pigmented epithelium in rats, dogs, and nonhuman primates (Weber et al., 2003). Among

the more recently discovered serotypes, AAV7 has also been shown to have superior muscle

transduction compared to AAV2, while AAV8 and AAV9 are the most efficient serotypes

discovered so far for transducing the liver (Gao et al., 2004; Gao et al., 2002). Studies on AAV2

capsids and their mutants suggests that the polyclonal antibody response is focused on a

relatively small number of sites on the capsid, and that reactivity can be reduced by mutations of

residues within those sites (Lochrie et al., 2006). However, until the developments of new mAbs

to AAV1 and AAV5, discussed in this study, there have been only a few mAbs reported that

react with intact capsids (Kuck et al., 2007; Wobus et al., 2000). There is currently little

Page 53: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

53

information available about how the various anti-AAV IgGs recognize capsids, their mechanisms

of neutralization, or the distinction between cross-reactive and specific epitopes.

The antigenic structure of AAV2 is the best characterized among the dependoviruses, since

peptide mapping has been used to identify both linear and conformation-dependent epitopes on

the capsid that are recognized by mAb (Wobus et al., 2000). Linear epitopes (recognized by

antibodies A1, A69, and B1) and conformational epitopes (recognized by antibodies A20, C24-

B, C37B, and D3) have been reported, although the C24-B site has not been mapped. The linear

epitopes are not relevant to the infectious virus as A1 recognizes VP1 only, while A69

recognizes a peptide in the VP1/VP2 common region, and B1 recognizes all three capsid proteins

via an epitope at their C-termini (Wobus et al., 2000). Antibodies to the conformation-dependent

epitopes show multiple mechanisms of neutralization, as the C24-B and C37B antibodies inhibit

receptor attachment, while the A20 antibody apparently neutralizes at a post-attachment step, and

D3 antibodies are non-neutralizing (Wobus et al., 2000; Figure 1-1). The C37B epitope (residues

493-502, 602-610, VP1 numbering) is close to the HSPG binding site on the capsid, which is

consistent with its ability to block receptor binding. Antibodies to the C37B epitope specifically

recognize AAV2, but not AAV1, 3, 4, or 5, likely related to the fact that surface loops near the

heparin binding region are the most variable in the AAV capsid structures (Govindasamy et al.,

2006; Padron et al., 2005). The D3 epitope is fairly conserved in the sequences of most AAV

serotypes (Padron et al., 2005). D3 antibodies recognize serotypes AAV1, 3 and 5, but not 4, and

residues in that epitope are close to structural differences between AAV2 and AAV4 within the

mounds surrounding the icosahedral three-fold axes (Govindasamy et al., 2006; Padron et al.,

2005). The A20 antibody reacts only with AAV2 and AAV3, and this epitope contains three

different regions in the primary sequence (residues 272-281, 369-378, and 566-575) (Wobus et

Page 54: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

54

al., 2000) that come together in the capsid structure (Xie et al., 2002). Comparisons of the

AAV4, AAV5, AAV8, and AAV2 coat protein structures identified the A20 binding regions as

some of the most variable between the three viruses (Govindasamy et al., 2006; Padron et al.,

2005; Walters et al., 2004; Nam et al., 2007). The AAV4 capsid protein is not recognized by the

B1 antibody, which recognizes a linear epitope (residues 726-733) of denatured capsids near the

C-terminus of the VP3 protein (Wobus et al., 2000). A comparison of the AAV2 and AAV4 VP3

crystal structures showed that there is no major conformational difference in the B1 epitope

region, suggesting that amino acid sequence differences may play a role in the capsid-antibody

recognition (Govindasamy et al., 2006).

A panel of mutations of surface residues of AAV2 to either conservative substitutions (to

Ala) or to radical changes were prepared and examined for their ability to bind HSPG and to

bind or be neutralized by the A20 mAb, by polyclonal antibodies in 3 human sera, or by a pool

of human IgGs (Huttner et al., 2003; Lochrie et al., 2006). Mutations that reduce binding and

neutralization by polyclonal human and monoclonal murine antibodies were identified as well as

mutations that reduce, have no effect on, or increase transduction. There was not always a direct

relationship between the change in IgG binding and neutralization, indicating that not all IgGs

neutralize equally efficiently as has been reported for other virus families such as rhinovirus

(Smith et al., 2001).

AAV Capsid Structure and Serotypes

The AAV2 capsid structure has been determined by cryoEM and by X-ray crystallography

to ~3 Å (Kronenberg et al., 2001; Xie et al., 2002). Even more recently the structure of AAV2 at

65˚C was solved by cryoEM (Kronenberg et al., 2005). This structure gave some insight into the

elusive location of the VP1 unique region in that it is known to be required for infectivity, but

has not been identified in previous reconstructions. This structural region was described as

Page 55: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

55

“globules” located under the icosahedral two-fold axes of symmetry. Other serotype structures

that have been solved by cryoEM and X-ray crystallography include AAV1, AAV4, and AAV5 -

9 by the Agbandje-McKenna lab (Figure 1-2A) (Govindasamy et al., 2006; Nam et al., 2007;

Walters et al., 2004; and Agbandje-McKenna, unpublished data. Sequence analysis has identified

AAV4 and AAV5 as being the most diverse serotypes at the amino acid level and also

antigenically (Gao et al., 2004).

The AAV serotype structures can be placed into two categories based on their surface

topology. AAV2 (Kronenberg et al., 2001; Xie et al., 2002) and AAV1, and AAV6-8 (Nam et

al., 2007 and unpublished data) exhibit pointed “finger-like” protrusions around the threefold

axes, while AAV4, AAV5, and AAV9 (Govindasamy et al., 2006; Padron et al., 2005; Walters et

al., 2004; and unpublished data) have “rounder” mounds surrounding the icosahedral three-fold

axis of symmetry (Figure 1-2B). Sequence alignments and 3D-modeling of AAV3, 6 and 10

shows that they will be similar to the AAV1, 2, and 8 class, while AAV11 will be more like

AAV4, 5, and 9. Comparison of all the available AAV structures or models revealed a conserved

β-barrel motif, while the more variable regions were mapped to the exposed surface loops

(Govindasamy et al., 2006; Figure 1-2C). This has also held true for other parvovirus capsids

(reviewed in Kontou et al., 2005). The surface loop regions become clustered as VP monomers

come together to form the capsids icosahedral symmetry. This assembly creates variations on the

capsid surface at the five-fold axes, the “shoulder” regions and the tips of the three-fold

protrusions, which surround the two-fold axes. These variable regions localize with previously

identified surface areas that affect receptor recognition, capsid recognition by conformational

antibodies, specifically A20 (Wobus et al., 2000), and transduction efficiency for AAV2

(Lochrie et al., 2006). This study has mapped antigenic sites on five serotypes, including the

Page 56: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

56

already well characterized AAV2, the structurally similar AAV1 and AAV6, and the more

distinctly related AAV5, as well as hepatrotrophic AAV8, which provides a strong insight on the

recognition of antibodies by the AAV capsids and their resultant binding abilities.

Significance

The generation of neutralizing antibodies during viral infections is often crucial to recovery

and is usually the target response in vaccination strategies. Despite the important role that

immunoglobulins have in animal virology there are still many aspects that are not completely

understood. There are many other viruses, including such important pathogens as HIV,

rhinovirus, hepatitis C, and influenza that induce only partially protective antibodies that do not

aid in clearance of the virus. The rules that govern neutralization mechanisms are even less well

known, as it has not yet been determined how the step in the viral lifecycle to be blocked is

chosen. The evolution of viruses is partially driven by the host’s mechanisms of defense in that

they must choose mutations that still allow for receptor attachment, replication, and immune

evasion in order to survive. Through the data presented here it will be possible to define

dominant antigenic regions that are directly involved in binding and neutralization by mAbs

against the capsid of a simple and well characterized virus, the AAVs.

The parvoviruses are important pathogens in the veterinary sciences, as well as among

humans including the erythrovirus B19, which had been recognized for many years as one of the

classical rash illnesses of childhood. There are many unknown autonomous parvoviruses

identified by PCR (Candotti et al., 2004; Terossi et al., 2007), as well as different AAVs (Chen et

al., 2005; Gao et al., 2004). Because the AAVs are associated with no known pathology, have a

wide range of infectivity, and can establish long-term transgene expression, they have great

potential as a viral vector in gene therapy (Muzyczka, 1994; Choi et al., 2005; Muzyczka and

Warrington, 2005). The AAVs under examination here have distinct tissue tropisms likely due to

Page 57: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

57

differences in cellular receptor utilization which is thought to arise from structural differences

between AAV serotypes at the capsid level (Wu, Asokan, et al., 2006). Similar to wild-type

viruses, recombinant vectors are detected by the immune system and generate a response that

becomes effective usually before the virus infects its target cell. This immunity not only acts

against the vector and the infected cells, but also results in a memory response that affects further

efforts to use the same vector or even transgene again (Bessis et al., 2004). Efforts to enhance the

safety and delivery of vectors are ongoing as well as the determination of strategies to sidestep

the antiviral response. This study supports the idea that it would be useful to have well

characterized AAV vectors with predictable tropisms and altered antigenic sites that can be used

in the treatment of individuals with a pre-existing immune response as well as for subsequent

therapies.

Figure 1-1. The antigenic structure of AAV2. Antigenic epitopes of AAV2 (Wobus et al., 2000) displayed on the 3D capsid surface of PDB 1LP3 (Xie et al., 2002). Amino acids that form the A20 (green spheres), C37B (magenta), D3 (orange), and B1 (blue) epitopes are displayed in context of the entire capsid surface depicted in grey cartoon. The icsahedral symmetrical axes are indicated by red arrows and the 5-fold is also highlighted in dark grey for orientation purposes.

Page 58: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

58

Figure 1-2. Panel of available AAV atomic structures A) VP3 is shown for each serotype that has been solved to date by X-ray crystallography. B) The two major surface topologies of the existing serotype structures can be grouped into two classes. AAV5 represents those serotypes with “rounder” protrusion. The other class depicts “finger-like” protrusions and is represented by AAV2. C) Superimposed VP3 monomers reveal a common motif with variable regions at the exposed surface loops (I-IX). The symmetrical axes are represented by black shapes (same in A and C); the pentamer represents the five-fold axis, while the triangle and the oval represent the three-fold and the two-fold, repectively. An asymmetric unit is depicted as a white-outlined triangle in B.

Page 59: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

59

CHAPTER 2 MATERIALS AND METHODS

Production and Purification of Recombinant AAV VLPs for Structural Studies

The AAV capsid proteins VP1, VP2, and VP3, for serotypes 1, 4, and 5-9, were expressed

in sf9 insect cells under the baculovirus polyhedron promoter as previously reported (Miller et

al., 2006). These proteins self-assemble into virus-like particles (VLPs). After 72 hours

incubation at 27̊ C the cells were spun down for 15 minutes at 1 500 rev min-1 (4ºC), and the

pellet was resuspended in lysis buffer (50mM.Tris pH8.0, 100mM NaCl, 0.2% Triton X-100).

The pellets were then subjected to three freeze/thaws with the addition of benzonase (Novagen)

after the second cycle and then centrifuged for 15-30 minutes at 10 000 rev min-1 (4ºC). The

supernatant was then diluted with TNET buffer (50mM Tris pH 8.0, 100mM NaCl, 1mM EDTA,

0.2% Triton X-100) and pelleted through a 20% sucrose cushion for 3 hours at 45 000 rev min-1

(4ºC). This pellet was resuspended overnight at 4ºC and further purified by ultracentrifugation on

a step gradient (5-40% sucrose) at 35 000 rev min-1 (4 ºC). A visible blue fraction containing

empty (no DNA) viral capsids, sedimenting at ~20-25% sucrose, is expected and will be

extracted and dialyzed.

AAV2 capsids were purified using iodixanol gradients and a heparin agarose affinity

column. Discontinuous iodixanol step gradients were formed in quick-seal tubes (25×89 mm,

Beckman) by underlaying and displacing the less dense cell lysate (15 ml) with iodixanol (5,5′-

[(2-hydroxy-1,3 propanediyl)bis(acetylamino)] bis[N,N′-bis(2,3dihydroxypropyl-2,4,6-triiodo-

1,3-benzenecarboxamide] prepared using a 60% (w/v) sterile solution of OptiPrep (Nycomed)

and PBS-MK buffer (1× PBS containing 1 mM MgCl2 and 2.5 mM KCl). Therefore, each

gradient consists of (from the bottom) 5 ml 60%, 5 ml 40%, 6 ml 25%, and 9 ml of 15%

iodixanol; the 15% density step also contains 1 M NaCl. Tubes were sealed and centrifuged in a

Page 60: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

60

Type 70 Ti rotor at 69 000 rev min-1 for 1 h at 18°C. Approximately 5 ml of the 40–25% step

interface was aspirated after side-puncturing each tube with a syringe equipped with an 18-gauge

needle (modified from Zolotukhin et al., 2002). A heparin-agarose (H6508, Sigma) column with

a 2.5ml bed volume was then prepared and equilibrated with 10 ml of PBS-MK buffer (1× PBS

containing 1 mM MgCl2 and 2.5 mM KCl) then 10 ml of PBS-MK/1 M NaCl, followed by 20 ml

of PBS-MK buffer. The AAV2 capsid containing iodixanol fraction(s) were then diluted with

twice the original volume and applied to the column. The column was washed with 3-10 column

volumes of PBS-MK buffer and sample was eluted with PBS-MK/1M NaCl. The high sodium

chloride content was reduced by buffer exchange or dialysis and the eluent was checked for

protein by optical density. Samples were then concentrated on centrifugal filter units (Millipore)

and checked for the VP1, VP2, and VP3 proteins by SDS-PAGE analysis (Figure 2-1). Intact

capsids were verified by negative stain electron microscopy (EM) (Figure 2-1).

Fv Antibody Modeling Using the Web Antibody Modelling (WAM) Server

Sequences (obtained for the Fab variable domain of AA4E4.G7, AA5H7.D11, and

BB3C5.F4 by the Parrish group at Cornell) were were submitted to the WAM server

(http://antibody.bath.ac.uk/index.html). The modeling is done piecewise for the heavy and light

chains and the canonical loops (Figure 2-2), which is based on the WAM algorithm

(http://antibody.bath.ac.uk/algorithm.html). Briefly, homology modeling is used to build the

framework of the antibody structure, i.e., heavy and light chains. The canonical loops are also

built through homology modeling with 5 rounds of minimization to smooth out joint regions.

The more diverse, non-canonical regions are built based on a different algorithm, the Combined

Antibody Modelling Algorithm (CAMAL) (Martin et al., 1989).

Page 61: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

61

Preliminary Neutralization Assays

Neutralization assays were conducted at NIH (Bethesda, MD) in collaboration with Dr.

John Chiorini. Briefly, AAV1 and AAV5 capsids that were packaging the green fluorescing

protein (GFP) gene were pre-incubated with serial dilutions of hybridoma supernatant for 30

minutes at room temperature prior to addition to cos7 cells and incubated for 1 hour at 37°C.

Fresh media was then added to the cells and the cultures incubated for 48 hours at 37°C. The

percentage of infected cells was evaluated by measuring the GFP-expressing cells using flow

cytometry.

Preliminary In-House Green Cell Assays

Purified monoclonal antibodies of AA4E4.G7, AA9A8.B12, and BB3C5.F4 were used in

green cell assays to check their neutralization properties (for future studies aimed at deciphering

theirmechanisms of neutralization). Briefly, cos7 cells grown to ~100% confluency in complete

Dulbecco’s Modified Eagle Media (with 10% FBS and 1% P/S; DMEM) were plated at a

dilution of 1:4 in 96-well dishes. Assuming 60 binding sites per capsid, purified virus and

purified mAb were complexed at different ratios. As an example, one capsid to 240mAbs is a

ratio of 1:4 (VP:mAb). Ratios of 1:1 and 1:0.5 VP:mAb were also examined. (To note, a mAb

was considered to have one binding site although the presence of bivalent binding was not ruled

out.) These capsid:mAb mixtures were incubated at 22°C for ~30 minutes. Media was removed

from the cells and the mixture was applied to the wells. The mixture was left on cells for 1 hour

and incubated at 37°C with 5% CO2. The mixtures were then removed and complete DMEM was

added. At ~48 hours post-infection green cells were visualized by UV excitation on an Olympus

scope. These experiments were done in triplicate for each VP:mAb ratio.

Page 62: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

62

Dot Blot Analysis to check Cross-Reactivity among VLPs

To examine the reactivity of the mAbs against different AAV serotypes, dot blots of

previously purified VLPs were performed. PVDF membrane (Millipore) was activated in 100%

methanol for 30 seconds and then soaked in PBS for ~15 minutes. Approximately 100-300ngs of

each AAV serotype was applied to the prepared membrane through a vacuum-manifold dot blot

system. PVDF membrane containing samples were then blocked in 10% non-fat milk (Bio-Rad)

in PBS for ~1 hour at 22°C. Membranes were then incubated with primary antibody for 1 hour

and then secondary antibody conjugated to horse radish peroxidase (HRP) for another hour. The

bound antibody-HRP was detected with Supersignal luminescent substrate and exposed to X-ray

film.

Generation of Fragment Antibodies from Monoclonal Antibodies

Intact IgGs were purified using 1 ml Hi TrapTM Protein G HP columns (GE Healthcare).

Briefly, hybridoma sera were run through a 0.45μm syringe filter while protein G columns and

were equilibrated with 20mM sodium phosphate pH7.0. The clarified supernatant was then

diluted and applied to the Protein G column at 1ml/min as specified in the manufactures protocol

(GE Healthcare). Whole IgGs were eluted from the column with Glycine pH 2.5 into prepared

collection tubes containing 200μls 1M Tris pH9.0, to ensure a final pH of 7.5-8.0. The IgGs were

then concentrated and buffer exchanged into 20mM sodium phoshate pH7.0, 10mM EDTA for

papin cleavage. Samples were then incubated with activated immobilized papain (Pierce) at a

suggested enzyme: substrate ratio of 1:160 w/w at 37°C for ~12 hours. The Fc fragment was then

captured on a Hi Trap™ Protein A column (GE Healthcare) and the fragment antibody-binding

portions (Fabs) were collected in the flow-through, which was then subjected to gel filtration on

a Superdex 75 column (GE Healthcare) in PBS. Samples were checked through-out the

Page 63: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

63

purification and production process to ensure purity and further characterize products (Figure 2-

3).

Preparation of Fab:VLP Complexes

Purified capsids were mixed together with Fabs at a ratio of approximately 2 Fabs per

potential binding site on the capsid, giving a final complexed ratio of ~1:120 capsid:Fab.

Complexes were left at 4°C for 1 hour and checked by negative stain electron microscopy for the

presence of decorated particles before cryoEM data collection.

CryoEM Data Collection

Sample aliquots of 3.5 µl were vitrified with a manual plunge freezing device either on

grids with Quantifoil films or on grids with continuous carbon films. The samples were

examined at -193°C on a FEI Tecnai G2 Polara electron microscope at an accelerating voltage of

200 keV and at a nominal magnification of 59,000 X (in collaboration with Timothy Baker,

UCSD). Images were recorded with a Gatan Ultrascan 4000 CCD camera at a step size of 1.883

Å / pixel. The CCD images were saved in the Gatan DM3 format and later converted to the tiff

format. All images were recorded with an objective lens underfocus value of 1.25 to 3.0 µm. The

total electron dose on each image was ~24 to 28 e- / Å2.

Three-Dimensional Reconstruction of AAV:Fab Complexes

The software package RobEM (http://cryoEM.ucsd.edu/programs.shtm) was used to

extract individual particle images from collected micrographs or film. Preprocessing of the

selected images occurred as previously described, as well as estimation of the defocus levels of

each micrograph was as previously described (Baker et al., 1999). A random-model computation

procedure (Yan, Dryden, et al., 2007) was used to generate a starting model at ~30Å resolution

from 150 particle images. This map was then used to initiate full orientation and origin

determinations and refinement of the entire set of images using the current version of

Page 64: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

64

AUTO3DEM (Yan, Sinkovits, et al, 2007). Corrections to compensate for the effects of phase

reversals in the contrast-transfer functions of the images were performed as previously described

(Bowman et al., 2002; Zhang et al., 2003), but amplitude corrections were not applied. Final 3D

maps, reconstructed from the selected particle images were estimated to be reliable to ~10.8-

22.8Å resolution (Table 4-2), using the 0.5 threshold of the Fourier Shell Correlation (FSC0.5)

criterion (data not shown). The handedness of the maps was determined by comparison of the

surface features with those of AAV structures determined by X-ray crystallography. Graphical

representations of the constructed maps were generated with the RobEM and Chimera

(http://www.cgl.ucsf.edu/CHIMERA; Peterson et al., 2004) visualization software packages.

Fitting of Atomic and Pseudo-Atomic Models into CryoEM Density Maps

The atomic coordinates for the capsid structures of AAV2 (PDB accession number 1LP3;

Xie et al., 2002) and AAV8 (Nam et al., 2007), AAV1 and 5 (unpublished data), and a model

built for AAV6 based upon AAV1, were used for capsid docking and scaling of the cryoEM

reconstruction density maps. When necessary, the capsid coordinates were rigid-body docked

into the cryo-reconstructed AAV-complexed maps using the COLORES subroutine in the SITUS

program, version 2.3 (Chacon and Wriggers, 2002). A visual inspection of the docked model in

the program COOT (Emsley and Cowtan, 2004) identified either several small surface loop

regions extending above the density envelope or into the capsid interior density depending on the

specific map, possibly indicating an incorrect initial map scaling factor. The absolute scales for

the maps were thus determined by creating electron density maps of the cryo-reconstructed

AAV:Fab structures at various Å/pixel values and comparing these maps to an electron density

map generated for structure factors calculated for the docked homology model at cell-dimensions

and grid sizes matching each of the scaled maps. The various cryo-reconstructed and model

maps were then normalized in the program MAPMAN (Kleywegt, 1996) and compared using the

Page 65: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

65

program’s similarity function (http://xray.bmc.uu.se/usf/mapman_man.html). The highest

correlation coefficient was observed between the maps generated at 1.80 and 1.883Å/pixel (the

original scale), depending on the respective structure (Table 4-3) and was considered the

absolute map scale. Difference maps between the cryoEM reconstructed maps and the atomic

data (or AAV6 3D model) maps (as used above in scaling) were then generated in MAPMAN

using the operation function (OP). The excess density that was displayed in the difference maps

were identified as the bound Fab. Atomic coordinates for an unrelated (generic) Fab (PDB 2FBJ;

Suh et al., 1986) structure was manually docked into the Fab density, as web-antibody models

(WAM) models of the respective Fabs were only generated for the Fv arm of the antibody. Once

the unrelated Fab was docked into the respective density, indicated by the least amount of amino

acids seen jutting from the density, a 60-mer of the capsid and Fab model was generated in order

to mimic the fully bound structure of the AAV:Fab complex. Electron density maps generated

from structure factors calculated for these fully bound models (at cell-dimensions and grid sizes

matching each of the scaled maps) were then compared to their respective complex density map

using the similarity function in MAPMAN. Models with the highest correlation (Table 4-3), or

similarity index were then chosen as the properly docked representative Fab model. WAM

models generated from the sequenced Fabs were then superimposed onto the docked unrelated

Fab (2FBJ) in COOT (SSM Superpose; Krissinel and Henrick, 2004) in order to visualize the

variable loops on the AAV capsid surface that appear to interact with the canonical and non-

canonical loop regions on the Fab models. Visualization and analysis of these models in context

of their respective maps were completed in Chimera (Petteresen et al., 2004).

Mutagenesis of Proposed Antigenic Regions

Since a majority of the structures solved in the cryoEM studies were of AAV1,

mutagenesis studies were focused on this serotype. Specific regions of the capsid surface that

Page 66: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

66

appeared to be important in capsid recognition were identified by the previous docking

procedures (Table 4-4) and primers were designed to generate antigenic escape mutants in these

regions by muatatons of the AAV1 cap gene. The recombinant AAV1 plasmid (pAAV1cap,

~8kb) was a gift from Dr. Jay Chiorini (NIH, Bethesda, MD) and contained the AAV1 cap gene

and the AAV2 rep gene. Forward (Fwd) and reverse (Rvs) primers were designed to mutate

multiple regions in the AAV1 cap gene to structurally equivalent regions in AAV2 (Table 4-4).

The mutations from AAV1 to AAV2 were based on the observation that AAV2 was not

recognized by anti-AAV1 antibodies (See Figure 3-1A and C). The method involved mutating 1-

2 amino acids at a time so several primers were designed for subsequent rounds of mutagenesis.

For example, mutant 1AB (Mut1AB) was created by mutating AQNK (residues (aa) 456-459) of

the AAV1 cap gene to TTQS (of AAV2) using, Fwd1a: 5’

GTCCGGAAGTACCCAACAGAAGGACTTGCTGTTTAGCCGTGG 3’, Rev1a: 5’

CCACGGCTAAACAGCAAGTCCTTCTGTTGGGTACTTCCGGAC 3’ and Fwd1b: 5’

GTCCGGAAGTACCACACAGAGCGACTTGCTGTTTAGCCGTGG 3’ , Rev1b: 5’

CCACGGCTAAACATGAAGTCAGTCTGTTGGGTACTTCCGGAC 3’, where 1a mutated

AQNK TQQK and 1b mutated TQQKTTQS. This method was repeated for all other

mutants. Mut2AB was mutated from TK (aa492-493) to SA using, Fwd2a: 5’

GCAGCGCGTTTCTAAAACAAGCACAGACAACAACAACAGC 3’, Rev2a: 5’

GCTGTTGTTGTTGTCTGTGCTTGTTTTAGAAACGCGCTGC 3’ and Fwd2b: 5’

GCAGCGCGTTTCTAAAACAAGCGCAGACAACAACAACAGC 3’, and Rev2b: 5’

GCTGTTGTTGTTGTCTGCGCTTGTTTTAGAAACGCGCTGC 3’. Finally, Mut3AB was

mutated from SSST (aa586-591) to RGNR using, Fwd3a: 5’

GCAGTCAATTTCCAGAGAGCAACACAGACCCTGCGACCGG 3’, Rev3b:5’

Page 67: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

67

CCGGTCGCAGGGTCTGTGTTGCTTCTCTGGAAATTGACTGC 3’ and Fwd3b: 5’

GCAGTCAATTTCCAGAGAGGCAACCGAGACCCTGCGACCGG 3’ and Rev3b:5’

CCGGTCGCAGGGTCTCGGTTGCCTCTCTGGAAATTGACTGC 3’. These mutants were

then used in combination with the designed primers to generate AAV1 capsid with multiple

AAV2 region suvstitutions. For example, Mut1AB served as the template to generate

Mut1AB2AB, which served as the template to create a mutant with all three regions mutated,

Mut1AB2AB3AB. Mutations were made with the QuickChange LightningTM kit (Stratagene)

following recommended parameters, and primer and reagent volumes. Final products were run

out on a 0.8% agarose gel with ethidium bromide to check for purity and sent to the UF ICBR

core for Sanger sequencing.

Production and Purification of Antigenic Mutants

Plasmids with positive mutations, as well as plasmids containing Ad helper functions

(pXX6, ~18kb; Xiao et al., 1998) and green fluorescing protein (GFP) gene (UF11, ~7.5kb;

Klein et al., 1998), were amplified by inoculating 1L cultures of TB with glycerol stocks.

Cultures were shaken at ~225 rev min-1 with incubation at 37°C for ~24 hours and were

harvested by low-speed centrifugation (15 minutes, 5000 rev min-1, 4°C). 1L Cell pellets were

resuspended in 20 mls buffer ( 25mM Tris pH8.0, 10mM EDTA, 15% sucrose (w/v), 0.1mg/ml

RNAse) and incubated with 50mgs lysozyme for 5-10 minutes on ice. Cells were lysed with the

addition of 48 mls lysis buffer (1% SDS, 0.2 N NaOH) and gentle shaking. The solution was

neutralized with the addition of 36 mls 3M sodium acetate (C2H3NaO2) pH5.2, and gently

inverted several times. Chloroform (0.2 mls) was added and the solution was mixed by gentle

inversion. The lysed cells were then spun at 9000 rev min-1 for 20 minutes and the supernatant

was poured into a new centrifuge bottle through a layer of gauze to remove any debris. The

plasmid DNA was then precipitated by the addition of an equal volume of isopropanol and left

Page 68: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

68

overnight at 4°C. The DNA was then pelleted at 9000 rev min-1 for 10 minutes at 4°C and

resuspended in 20 mls double distilled filtered water (ddH2O). After allowing the DNA to

dissolve, 20 mls 5.5 M lithium chloride (LiCl) was added to precipitate any proteinaceous

material. The solution was incubated for 30 minutes on ice and then spun at 9000 rev min-1 for

10 minutes at 4°C. To avoid disrupting the protein pellet, the supernatant was carefully decanted

into two 50 ml conical tubes and equal volumes of isopropanol were added to re-precipitate the

plasmid DNA. This was spun at 9000 rev min-1 for 10 minutes at 4°C and the supernatant was

discarded and the pellet was air-dried for several minutes. The pellet was allowed to gently

dissolve in 7 mls ddH2O and cesium chloride (CsCl) was added at 1 g/ml. The CsCl was allowed

to completely dissolve on ice and then 125uls of 10mg/ml ethidium bromide (EtBr) was added.

This solution was then split into two 4.9ml Optiseal tubes (Beckman) and sealed. The EtBr

gradient was transferred to an NVT90 rotor and allowd to run at 78000 rev min-1 for 4 hours at

15°C, or 55000 rev min-1 overnight (15°C). Bands were collected with an 18 gauge needle and

transferred to a 15 ml centrifuge tube. EtBr was removed through several iso-amyl alcohol

extractions and the DNA was re-precipitated by the addition of 2.5 volumes ddH2O and two-

times the combined volume of 95% ethanol. The DNA was then pelleted at 9000 rev min-1 for 15

minutes at 4°C. The supernatant was discarded and the DNA pellet was allowed to air-dry, and

then allowed to resuspended in 500 uls ddH2O. The sample was subjected to two rounds of

phenol/chloroform/iso-amyl extractions and one chloroform/iso-amyl extraction before a final

round of re-precipitation. Finally, 0.1 volumes of 3 M C2H3NaO2 pH5.2, followed by 2.5

volumes 95% ethanol were added to precipitate the DNA and the mixture was spun at 14000 rev

min-1 at room temp. The pellet was then washed with 1 ml 75% ethanol and allowed to resuspend

in 1 ml ddH2O. Purified DNA concentrations were obtained through optical density

Page 69: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

69

measurements at OD260 and DNA isoforms were verified on agarose gels. The plasmid DNA was

also checked for purity and identified by restriction digests.

Human endothelial kidney cells (HEK 293) were grown to ~70% confluency in 15cm

plates with DMEM supplemented with 1% antibiotics and 10% serum (complete DMEM) at

37°C and 5% carbon dioxide (CO2). Cell media was aspirated and replaced with 12 mls of fresh

complete DMEM and were then triple transfected using polyethyleneimine (pEI) inoculums.

Transfection inoculums were made using 45ugs/plate pXX6, 15ugs/plate UF11, 15ugs/plate

mutant plasmid, 160uls/plate pEI, and 1000uls/plate of DMEM (no FBS). Transfected cells were

incubated for 24 hours and assayed for quality of transfection on a florescence scope by green

cell visualization. Virus infected cells were then harvested at ~60 hours post-infection by

scraping and low-speed centrifugation (1200 rev min-1, 15 minutes, 4°C). Cell pellets were

resuspended in 1 ml TD (1x PBS, 5mM MgCl2, 2.5mM KCl) per plate supplemented with

protease inhibitor cocktail (Roche) and cells were lysed by three rapid freeze/thaws with the

addition of benzonase (Novagen) at the second thaw, and subjected to a low-speed spin to pellet

cell debris (10000 rev min-1, 15 minutes, 4°C). Clarified cell lysates were then used in dot blot,

infectivity, and packaging assays.

For further purification, clarified cell lysates were loaded onto an iodixanol gradient and

fractions were collected (as previously explained). Ion-exchange chromatography was used to

further purify capsids and remove iodixanol as previously described in Zolotukhin et al., 2002.

Briefly, a 1 ml HiTrap Q-column (GE Healthcare) was equilibrated with 5 mls buffer A (20mM

Tris pH8.5, 15mM NaCl) followed by 5 mls buffer B (20mM Tris pH8.05, 500mM NaCl) and

another round of 5 mls buffer A on a Gilson peristaltic pump at 1 ml/minute. The iodixanol

fractions were then diluted 1:1 with buffer A and loaded onto the equilibrated column at 1

Page 70: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

70

ml/minute and washed with 50 mls of buffer A. Bound virus was eluted with a gradient reaching

100% buffer B over a 5 minute time span at 1 ml/minute on a Pharmacia ATKA FPLC system.

One ml fractions were collected and assayed for protein concentrations by OD280 optical density

readings.

Western Blot Analysis

Fractions were assayed for VPs by western bolt analysis. Fractions were denatured by 10%

SDS-PAGE and transferred to PVDF membranes (Millipore) overnight at 27V. The membrane

was then blocked in 10% non-fat milk (BioRad) for 1 hour and probed with B1 (Wistuba et al.,

1997), a mouse monoclonal that recognizes denatured AAV capsids (except AAV serotype 4).

The blot was then treated with secondary antibody conjugated to horse radish peroxidase (HRP)

for another hour. The bound antibody-HRP was detected with Supersignal luminescent substrate

and exposed to X-ray film.

AAV1 Enzyme-Linked Immunosorbent Assay (ELISA)

Mutant AAV1 capsids were tittered using an AAV1 titration ELISA kit (PROGEN).

Briefly, standards were prepared by diluting the provided kit standard from 1:2 to 1:64 and cell

lysates were initially diluted from 1:250 to 1:100000. Prepared dilutions and a blank were added

to a microtiter strip and incubated at 37°C for 1 hour. The strips were washed in 1x wash buffer

provided by the kit and anti-AAV1 biotin conjugate was added to the wells for 1 hour at 37°C.

After washing, a streptavidin peroxidase conjugate was then added and the strips were incubated

again at 37°C for 1 hour. Strips were washed and then reacted with tetramethylbenzidine (TMB)

or substrate for 10 minutes. A blue color indicated bound capsid and the color reaction was

stopped with Stop Buffer, provided by the kit. The plate was then read on an ELx800 plate

reader (BioTek) at a wavelength of 450nm.

Page 71: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

71

Dot Blot Analysis for Antigenic Reactivity

Recombinant AAV1 mutant capsids (rAAV1muts) were generated, expressed, and

purified as described above and further characterized for antibody recognition and cross-

reactivity against mAbs originally generated against the rAAV1 parental virus. Native dot blots

using cell lysates of the rAAV1muts were performed as described above.

Pixel Intensity Calculations from Native Dot Blots

Native dot blots were scanned at about 600 pixels per inch (or higher) and analyzed using

the freeware, imageJ (http://rsbweb.nih.gov/ij). Briefly, the stacked RGB image was divided into

the different gamma channels; red, green, and blue, and stacks containing the most information

were combined. Background and noise was removed from the images using a rolling ball

function at a threshold just below the actual data points so as not to smooth the actual data.

Finally, data points were chosen and analyzed using the “plot lanes” function. Pixels intensities

were given as integrated optical densities. Negative cell lysates values were subtracted from the

raw data and the final values were plotted in Excel against the respective mutant. Percent (%)

calculations were performed by setting the rAAV1 pixel intensities for each mAB to 100% and

subsequent data was calculated from this normalized value.

Infectivity Assays

Infectivity of the rAAV1muts was checked by green cell assays in HEK293 cells as

previously described (Wu et al., 2000). Briefly, HEK293 cells were plated at a 1:4 dilution in 96-

well dishes and were allowed to attach for ~24 hours. In a separate 96-well dish 20uls of cell

lysates were added to the first column and serial dilutions were made from 10-1 to 10-8 in DMEM

(no FBS) spiked with Ad 5 virus at a MOI of 10. To the cells, 100 uls of the dilutions were added

to corresponding rows and plates were incubated for 1 hour at 37°C with 5% CO2. The dilutions

were removed and wells were refilled with 100 uls complete DMEM. Cells were allowed to

Page 72: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

72

grow for 48-60 hours and infectivity was checked by counting green cells (GFP production)

under a fluorescent microscope. The total number of green cells counted was multiplied by the

original dilution factor and the plate dilution to get a final infectious particle count. Negative

controls included wells with Ad 5 virus only and no virus or cell lysate.

DNA Extraction and Real-Time Polymerase Chain Reaction

Packaging efficiency was checked by determining the amount of vector genomes (vgs) in

the cell lysate virus. Briefly, 2 uls of benzonase and benzonase buffer were added to 10 uls of

cell lystate and incubated at 37°C for ~2 hours. To the sample, 2 uls proteinase K and 22 uls

proteinase K buffer was added for a final volume of 224 uls. This reaction was allowed to

incubate for 1 hour at 37°C. Samples were then subjected to two rounds of

phenol/chloroform/iso-amyl extractions and one round of plain chloroform extraction. After the

final extraction, 22.4 uls of 3 M C2H3NaO2 pH5.2, 2 uls glycogen (Novagen), and 670 uls of

100% ethanol were added to precipitate the DNA. Samples were left overnight at -20°C and then

spun at 14000 rev min-1 for 20 minutes. The pellet was then washed in 75% ethanol and spun

again at 14000 rev min-1 for 5 minutes. The DNA pellet was allowed to air-dry and was then

resuspended in 20 uls ddH2O. Concentrations were checked by OD260 readings and were then

diluted to be between 10 and 1 ng/ul for real-time polymerase chain reaction (qPCR) analysis.

A master mix containing iQ SYBR Green Supermix (Bio-Rad) and forward and reverse

primers for each sample, was made and then delivered to 96-well PCR plates (BioRad). To this

mix, extracted DNA samples or standards (ranging from 10 ngs to 0.0001 ngs) were added to

respective wells for a final reaction volume of 25 uls and qPCR capture was performed. PCR

parameters were set as suggested in the iQ SYBR Green Supermix handbook and analysis of the

data was performed on the My iQ 2.0 software (Bio-Rad).

Page 73: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

73

Figure 2-1. Coomassiee stained SDS-PAGE and EM images of purified AAV VLPs for structural studies. Images for AAV serotypes illustrate stained bands for VP1, 2, and 3 at ~92, 78, and 67kDa, repectively and a corresponding EM image for each VLP depicting white, negatively stained intact VLPs against a black carbon background.

Figure 2-2. Images of the Fab WAM models. Complimentary determining regions are colored differently an the heavy and light chain portions are indicated in gray. Heavy and light chain hyper-variable regions are indicated as H1-3 and L1-3, respectively. Models were generated for A) AA5H7.D11 B) AA4E4.G7 C) BB3C5.F4

Figure 2-3. Production, purification, and characterization of mAb and Fabs. A) Superdex-75 column data showing the milli-absorbance units (mAU) at OD280 on the y-axis and peak fractions (vertical red dotted lines) and column volume on the x-axis. The Fab peak at fraction 11-12 corresponds to a molecular weight calculation of ~41.7kDa. B) An SDS-PAGE analysis of the Superdex-75 fraction 11 and the whole IgG before papain digestion indicating the loss of the Fc portion. Both images indicate by size and purity that Fabs have been obtained for the example IgG.

Page 74: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

74

CHAPTER 3 CHARACTERIZATION OF ANTIBODIES AGAINST AAV1 AND AAV5 CAPSIDS,

CROSS-REACTIVITY AND NEUTRALIZATION CAPABILITIES

Introduction

Antibodies are unique structures that have been found in various tissues of the body and

are responsible for recognition and neutralization of foreign objects. There are five main classes,

some of which have several subtypes. These classes represent the effector function and heavy-

chain structure of each isotype, which are: Immunoglobulin G (IgG), IgM, IgD, IgE, and IgA.

The IgG and IgM molecules are the first of the secreted Igs to be present in early infection, while

the IgD molecule functions at the B-cell surface as a receptor. The IgE molecules are generally

associated with allergies and parasites, whereas the IgA has roles in preventing colonization at

the mucosal surface. Of these isotypes, IgG is the most abundant isotype in plasma and

represents the typical antibody structure. This structure is composed of four polypeptide chains;

two heavy chain molecules (Hc) and two light chain molecules (Lc) (Figure 3-1A). These chains

are broken down further into constant (C) and variable (V) regions. The variable regions VH and

VL make up the Fab and contain the complimentary determining region (CDR) (Figure 3-1A).

This region is important for interaction with antigens and contains the paratope specific for

antigen recognition. Both the heavy and light chains have three CDRs, generally referred to as

hypervariable regions 1-3 (Figure 2-2). Structures among the different isotypes are generally

similar for IgG, IgD, and IgE, where differences occur mainly in the hinge regions and

glycosylation. The IgM molecule exists as a pentameric structure (Figure 3-1C) with five IgE-

like molecules attached by a small peptide referred to as the J-chain. The IgA is also held

together by the J-chain, but exists in a dimeric form (Figure 3-1B). These structures can be

abundantly produced by activated B-cells, which has led to the creation of hybridomas. In this

methodology, hybridomas were made by fusing B-cells with myeloma cells (a B-cell cancer cell

Page 75: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

75

line) (Alkan, 2004). Hybridomas specific for a certain antigen could then be separated and

propagated to generate mAbs. This technology won Cesar Milstein, Georges J. F. Köhler and

Niels Kaj Jerne the Nobel Peace Prize in Physiology or Medicine in 1984 (Alkan, 2004). Since a

virus can be neutralized in several different ways that are determined primarily by the specificity

of the reacting antibody, analysis of specific antibody-virus interactions is only possible using

these mAbs (Reading and Dimmock, 2007). Thus mAbs are used in various clinical and research

approaches, including diagnosis of disease and production of vaccines (Nabel, 2004).

The antibody response is a key immunity that develops against pathogens and foreign

objects alike, making viral vectors no different than their wild-type parents. Many people have

already been infected with some parvoviruses (Schneider et al., 2008; Lindner and Modrow,

2008; Candotti et al., 2004) and AAV serotypes (Gao et al., 2004; Calcedo et al., 2009) and have

developed antibodies which can interfere with the application of AAV vectors (Manno et al.,

2006). In addition, it is likely that therapeutic application of AAV vectors will induce immune

responses, which may interfere with re-administration. The primate and human AAV serotypes

are mostly antigenically distinct when tested with polyclonal antibodies, although much of the

sequences are similar, especially in structurally conserved domains (Gao et al., 2004; Agbandje-

Mckenna and Chapman, 2006). However, despite the important role of antibodies in the

recognition of AAV, the mechanisms of binding and neutralization or other effects are not well

understood. Information regarding antigenicity has been obtained for AAV2, as well as the

autonomous viruses; CPV, human B19 parvovirus (B19), and MVM (Wobus et al., 2000;

Hafenstein et al., 2009; Tolfvenstam et al., 2000). These studies suggest that although most

antibodies appear to bind on or near the protrusions at or surrounding the icosahedral three-fold

axes, there is a complex interaction between the capsid surface and the host antibody response,

Page 76: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

76

and that the sites involved in antibody binding may be at least partially constrained by the capsid

surface. Studies of AAV2 examining a small number of mAbs, or polyclonal anti-capsid

antibodies, show that the antibody response is affected by changes in only a small number of

sites on the capsids, and that reactivity can be reduced by mutations of residues within those sites

(Lochrie et al., 2006; Huttner et al., 2003; Wu et al., 2000b). For AAV2 some epitopes have been

described by mutational analysis and peptide mapping. These show neutralization of infectivity

at either attachment or post-attachment steps (Wobus et al., 2000).

Here, a number of mAbs (both IgGs and IgMs) directed against the capsids of AAV1 and

AAV5 have been generated. By comparing their reactivities with the parental capsids and other

AAV serotypes we seek to define the general mechanisms of antibody attachment, the processes

of neutralization, and to compare the structures that allow these different serotypes to be

successful in nature. This study also aims to characterize the antigenic nature, mainly the B-cell

response, of the AAV serotypes and provide optimized tools for serotype recognition.

Results

Production of Anti-AAV Monoclonal Antibodies and Isotyping

A panel of mAbs against AAV1 and AAV5 capsids (Table 3-1) were prepared in the

Parrish laboratory at Cornell University. Two different immunization strategies were utilized.

The first involved immunization 2-3 months prior to the fusion of spleen cells, giving 4 IgG- and

4 IgM-secreting hybridomas against AAV1. Seeking to derive cross-reactive antibodies between

AAV1 and AAV5 capsids, mice were immunized with AAV1 capsids, then given an intravenous

immunization with AAV5 capsids 4 days prior to the fusion. This protocol resulted in the

isolation of several hybridomas producing 9 IgMs and 1 IgG against AAV5. Three antibodies

were chosen from each panel to continue on with the structural studies: 4 IgGs and 2 IgMs. The

anti-AAV1 monoclonals were AA4E4.G7, AA5H7.D11, both IgG2a subtypes, and the IgG1

Page 77: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

77

subtype, AA9A8.B12 (Table 3-1). For the anti-AAV5 final panel; the two IgMs were

BB9F7.F12 and BB8F1.E8, with the last being BB3C5.F4, an IgG3 (Table 3-1).

Cross-Reactivity among Different AAV Serotypes

Cross-reactivity among the available serotypes was tested by dot blot analysis using the

anti-AAV panel antibodies previously chosen. At low dilutions of ~1:4 cross- reactivity was

observed with the anti-AAV5 mAbs against AAV1 (Figure 3-2A) however, at higher dilutions of

hybridoma sera, this phenomenon was completely abolished (Figure 3-2B). The slight

recognition of AAV1 by A20 (Figure 3-2A) has been previously reported at high concentrations

of the A20 monoclonal (Kuck et al., 2007). All antibodies showed specific reactions with their

homologous antigen, but no cross-reaction with the other capsids among AAV1, 2, 4, and 5-9

other than those previously mentioned (Figure 3-2C). All of the antibodies tested recognize

conformational epitopes on assembled capsids rather than linear epitopes in denatured capsid

proteins (Figure 1-1), as is seen for antibodies against other parvovirus capsids (Wobus et al.,

2000; Kuck et al., 2007, Hafenstein et al., 2009; Wistuba et al., 1997; Parrish and Carmichael,

1983).

Neutralization Ability of the Monoclonal Antibody Panel

Each of the antibodies was tested for its ability to neutralize virus infectivity by

determining the concentration of immunoglobulin required to neutralize AAV1 and AAV5

vectors carrying the GFP-gene. Most of the antibodies tested in this study neutralized AAV,

where some of the antibodies were non-neutralizing. In the anti-AAV5 study, both IgMs were

able to neutralize approximately 50% of cells at hybridoma supernatant dilutions of 1:200 and

1:400, BB9F7.F12 and BB8F1.E8, respectively (Figure 3-3). The only IgG in the anti-AAV5

panel, BB3C5.F4, was non-neutralizing, with the undiluted stock leading to only a 20%

reduction in gene expression (Figure 3-3). This observation may have been due to an aggregation

Page 78: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

78

of viral particles due to excess of antibody and interstingly, the ability of non-neutralizing

antibodies to reduce gene transfer has been noted (Li and Ertl, 2009). This occurrence may be

due to the aggregation of viral particles and further characterization of these events may

eventually allow for useful generalizations in the future. Similar results were observed for the

anti-AAV1 panel. Both IgG2a antibodies, AA5H7.D11 and AA4E4.G7, neutralized cells at

approximately 50% at dilutions of ~1:1800 and ~1:1200, respectively (Figure 3-3), while the

IgG1, AA9A8.B12, was not able to neutralize AAV1-GFP vector (Figure 3-4). Studies were also

completed using available AAV1-like vectors, X1, X25, and AAV6 (Figure 3-5). Neutralization

of 50% was achieved for AAV6 with AA4E4.G7 at ~1:2000 (Figure 3-5A) and ~1:3200 for

AA5H7.D11 (Figure 3-4C), while AA9A8.B12 was not neutralizing (Figure 3-5B). The X25

vector was neutralized at ~50% at dilutions of ~1:3200 for both AA4E4.G7 and AA5H7.D11

(Figures 3-5A and 3-5C). AA9A8.B12 was also non-neutralizing for X25 (Figure 3-5B).

Interestingly, the X1 capsid was not neutralized with AA5H7.D11, whereas this antibody

neutralized the X25, AAV1, and AAV6 vectors (Figures 3-4 and 3-5C). It showed similar

profiles as seen before with AA4E4.G7, neutralizing 50% of vector at a dilution of 1:1200, while

AA9A8.B12 was again non-neutralizing (Figures 3-5A and 3-5B).

Discussion

The antigenic nature of viral capsids recognized by antibodies is crucial to the

development of protective immune responses. The AAVs provide an important model system to

examine the antigenic structure of theses simple capsids as there are several different genetically

distinct viruses, which have appeared to be antigenically distinct when analyzed in limited

serological studies. Antibodies are an important protective immunity against viral infection,

including the AAVs. Immunoglobulins can develop shortly after initial infection, and can

continue to provide protection through the duration of infection and by generation of a memory

Page 79: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

79

response (Dörner and Radbruch, 2007). These responses are also effective against infection by

antigenically related viruses. The use of monoclonals in viral neutralization assays is beneficial

due to the existence of several epitopes on the capsid surface, which can lead to various

mechanisms of neutralization through different antibodies (Reading and Dimmock, 2007). Here

we developed a variety of new IgG and IgM mAbs against the capsids of two AAV serotypes to

examine the antigenic structures of the viruses, the cross-reactivity with the representative AAV

clade members, and also the effects of IgG and IgM antibodies. The AAV capsid was a potent

antigen; generating a panel of mAbs against AAV5 four days post immunization (Dr. Parrish,

Cornell University). This is consistent with the fact that multivalent viral capsids and particles

appear to be potent B-cell antigens and to give some immediate T-independent responses

(Reading and Dimmock, 2007; Janeway et al., 2005). For AAV5, although the mice had been

previously immunized with AAV1, most of the antibodies produced were IgMs directed against

AAV5, with only 1 IgG. The IgM response is one of the first responses to be seen in early

infection due to circulating low-affinity molecules (Janeway et al., 2005). It is possible that T-

helper cell epitopes shared between the AAV1 used to pre-immunize and the AAV5 capsids

given in the boost enhanced the responses to the second virus.

Characterization of antibodies produced after vector delivery has become increasingly

more important. In the data presented here Balb/c mice were used to generate a diverse response

of antibodies to AAV1 and 5 capsids. Most animal studies have found patterns in antibody

generation depending upon the animal strain and route of administration. Both IgG2a and IgG3

responses were seen in intramuscularly (i.m.) injected Balb/c mice, while C57B1/6 mice

exhibited IgG2a (Chirmule et al., 1999). The subcutaneous (s.q.) injections used by the Parrish

group produced IgG2a/b and IgG1 responses against AAV1, but AAV5 immunizations produced

Page 80: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

80

an IgG3 with the majority of the response being IgMs (Table 3-1). Intravenous (i.v.)

administration in C57B1/6 mice also produced IgG2a and IgG2b responses (Zhang et al., 2005).

This issue becomes important due to the different responses seen against both vector and

transgene (Lu and Song, 2009; Lu et al., 2006; Vandenberghe et al., 2006; Lin and Ertl, 2009;

Hernandez et al., 1999). Few studies have fully characterized the antibody response in humans,

although it has been repeatedly reported that the human population has titerable antibodies to the

better characterized AAV serotypes 1-9 (Erles et al., 1999, Chirmule et al., 1999; Moskalenko et

al., 2000; Calcedo et al., 2009; Xiao et al., 1999; Mingozzi and High, 2007). One study sought to

examine IgG subsets in serum samples of normal human subjects exposed to wild-type AAV,

injected i.m. with AAV vectors, or subjects injected i.v. with AAV vectors (Murphy et al., 2009).

Although the ultimate findings were that a complex, non-uniform pattern of responses existed, it

was interesting to note that IgG1 and IgG3 responses appeared more prominent in those exposed

to AAV vector, while IgG2 and IgG4 appeared more robust in normal donors (Murphy et al.,

2009). Thus, the inclusion of different isotypes into antigenic studies, such as this study, will

further characterize the antigenic nature of the AAV vector and may help to isolate immunogenic

differences among vector delivery routes, concentrations, and target tissues. Another such study

identified that IgG1 responses were the highest among the naturally infected population and

suggests that this may be indicative of a cured subject or a chronic carrier of this virus (Sylvie et

al., 2010). Most importantly, it appears that the antibody response is subjective to many factors,

including immunocompetence of the patient and their pre-existing immunity. Further

characterization of these responses may one day enable us to predict the immune outcome based

on the patient, the disease, and the vector type and administration.

Page 81: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

81

The new monoclonal antibodies described here react with the AAV1 and AAV5 capsids,

providing a variety of useful reagents for the study of AAVs in particular. Cross-reactivity was

checked by dot blot to determine if conserved regions on the AAV capsid serotypes were

recognized among the different antibodies. Of note, the anti-AAV1 antibodies recognized the

AAV6 capsid structure. This serotype was not used in the immunizations, but is 99% identical to

AAV1 (Xiao et al., 1999; Schmidt et al, 2006). This cross-recognition implies that the

differences between AAV1 and 6 are probably not involved in the antibody epitope, but different

affinities may be seen due to these differences. However, it is interesting to find that in a recent

seroprevalence study AAV6 IgG levels in human subjects was lower (46%) than those found for

AAV1, suggesting that the differences at the capsid surface level play an important role in

immunogenicity among humans (Sylvie et al., 2010). Although no cross-reactivity was seen

among other serotypes not used in the mouse immunizations, a weak cross-reactivity of the anti-

AAV5 antibodies with the AAV1 capsid was observed. This is likely a result of pre-

immunization with AAV1 before the AAV5 boost. However, the recognition was very weak, as

serial dilutions in hybridoma supernatant drastically lowered the recognition seen (Figure 3-2B).

Similarily, in an unrelated study looking at pre-existing immune response models, mice

immunized with Ad-AAV8cap vectors and then subsequently challenged with AAV2-transgene

vectors produced low-levels of non-neutralizing antibodies that bound AAV2 capsids (Li and

Ertl, 2009). Also of interest, these low-levels of non-neutralizing antibodies against AAV8,

found to be of IgG2a isotype, were able to disrupt gene transfer in AAV2 studies (Li and Ertl,

2009).

Preliminary neutralization studies on the anti-AAV1 and AAV5 mAbs (conducted by Dr.

J. Chiorini, NIH) identified neutralizing ability for most of the mAbs generated. Among the anti-

Page 82: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

82

AAV5 mAbs, both of the IgMs selected for further study were neutralizing. Here the expression

of GFP was reduced by ~50% at hybridoma supernatant (sup) dilutions of ~1/300-1/150 for

BB9F7.F12 and ~1/400 for BB8F1.E8 (Figure 3-3). These data were only repeated once at this

point in the study and the jump seen for BB9F7.F12 between dilutions of 1/300 and 1/150 may

have to do with pipeting errors or the avidity of the IgM itself. Repetitions will be needed to

confirm the values for this anti-AAV5 IgM. The only IgG selected from the anti-AAV5 mAbs

was non-neutralizing. Neutralization assays did not identify a diltion of BB3C5.F4 sup that was

able to knock down GFP expression by 50%. In fact, at a 1/25 dilution, green cells were still

sustained at about 80% (Figure 3-3). However, neutralization assays for AAV5:BB3C5.F4 at a

sup dilution of 1/25 show almost no change in GFP expression patterns (Figure 3-3). Since

particles that escape from this aggregate become infectious once again, the on and off rates, or

kinetics of antibody binding may allow for enough viral particles to escape and cause infectious

events. Of note, these infections were done in the absence of helper virus and since few cells are

infected to begin with, a significant change may not been seen in the materials used here. A

similar trend was seen in anti-AAV1 neutralization assays. Two of the three mAbs chosen for

further study were also neutralizing while one was non-neutralizing. The two IgG2a mAbs,

AA4E4.G7 and AA5H7.D11, both neutralized ~50% of AAV1-GFP vectors at sup dilutions of

~1/1200 and >1/1600, respectively (Figure 3-4). Both of these mAbs are strongly neutralizing as

almost complete knockdown of GFP transduction was seen at dilutions of ~1/100. In comparison

to the non-neutralizing anti-AAV5 mAb, BB3C5.F4, gene transfer of the AAV1 capsids was

slightly more susceptible to the non-neutralizing anti-AAV1 mAb, AA9A8.B12, as ~50%

knockdown was seen at sup dilutions of ~1/50 (Figures 3-3 and 3-4). Again, in-house green cell

Page 83: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

83

assays mimic this observation as green cells are reduced by almost half in capsid:mAb ratios of

1:4 (data not shown).

Neutralization assays were also done for the AAV1 mAbs against AAV1-like viruses, X1,

X25, and AAV6. These vectors differ from AAV1 by only a few amino acids at the capsid

surface. AAV6 differs from AAV1 at several sites on the VP1 protein, but only three of these are

exposed at the capsid surface and reside in the common C-terminus, VP3. These residues are

E418D, E531K, and E584L (Table 3-2). The X25 capsid differs from AAV1 at T162S, V198L,

which are buried in the capsid on VP2, and Q386K, which is surface exposed (Table 3-2). The

X1 capsid differs from AAV1 at N327S, D495G, R514H, and D590H (Table 3-2). All 4 of these

residues can be mapped to the capsid surface and fall in or around the three-fold axis (Figure 3-

6). These viruses gave results similar to wtAAV1 in that AA9A8.B12 was still non-neutralizing

while AA4E4.G7 was still able to neutralize (Figure 3-5). However, a change was noted in the

ability of AA5H7.D11 to neutralize against the X1 virus. A reduction of ~50% in green cells was

now seen at a sup dilution of ~1/200 (Figure 3-5C). The decline in sensitivity to this previously

neutralizing virus is most likely due to the differences seen at the aa level. Interestingly, single

mutations on the AAV1 capsid surface to mimic the X1 surface (made in the Chiorini lab, NIH),

resulted in no significant changes in neutralizing activity against the AA4E4.G7 and

AA5H7.D11 monoclonals (data not shown). This supports that idea that several amino acids are

involved in the epitope and that it may be conformational in nature. This data corresponds to the

mapping of the AAV2 mAb epitopes (Figure 1-1), supporting the idea that these symmetrical

axes are important in the antigenicity of the AAV capsids and also implies that some of these

residues are involved in the epitope itself, or at least plays a role in stabilization.

Page 84: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

84

The mAbs presented here will aid in the further characterization of antigenic regions on the

AAV capsid surface in hopes of engineering AAV capsids for efficient immune response

evasion, whether it be pre-existing or an initial response. Conversely, this data will also aid in the

manipulation of these sites to generate a desired response, as in vaccine development. The next

step in these studies is to determine the epitopes for these mAbs using a structural approach as

detailed in the following chapter. These data may then define the consequence of manipulation in

these capsid regions and better define the antigenic nature of the capsid surface while adding to

the general knowledge of antigen-antibody interactions.

Page 85: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

85

Table 3-1. Mouse monoclonal antibodies selected for further studies Monoclonal antibody (mAb) Parental AAV antigen Isotype AA4E4.G7 AAV1 IgG2a AA5H7.D11 AAV1 IgG2a AA9A8.B12 AAV1 IgG1 BB3C5.F4 AAV5 IgG3 BB9F7.F12 AAV5 IgM BB8F1.E8 AAV5 IgM Table 3-2. Amino acid differences between AAV1-like viruses and AAV1* 162 198 327 386 418 495 514 531 584 AAV6 T V N Q D D R K L X1 T V S Q E G H E F X25 S L N K E D R E F AAV1 T V N Q E D R E F *Red residues signify different amino acids

Figure 3-1. Basic immunoglobulin structure. A) General structure of a monomeric antibody representing the IgG, IgE, and IgD classes. The heavy chains (Hc) are colored in dark shades which are connected to the light chains (Lc, lighter shade) by disulfide bridges. The Hc and Lc portion of the Fab structure is further divided into a variable (V) and constant (C) region, VH and VL, and CH and CL. The variable region contains the CDRs housing the hypervariable domains responsible for the antibody paratope. B) The IgA class is represented by a dimer which is held together by a J-chain (small structure colored gray). C) The pentameric structure depicts an IgM molecule. These structures are also held together by a J-chain (gray). Different colored structures represent a separate functional Ig unit.

Page 86: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

86

Figure 3-2. Monoclonal antibody characterization and cross-reactivity. A) Native blot using a mAb hybridoma supernatant dilution of 1:4. Recognition of native AAV1 by the anti-AAV5 monoclonals is evident at this dilution. B) Serial dilutions of the anti-AAV5 mAb hybridoma supernatant tested against native AAV1 and 5 capsids. A lack of recognition is already seen at a dilution of 1:25. C) Native dot blot analysis of AAV1, 2, and 4-9 against the anti-AAV1 and -AAV5 monoclonal antibodies. No cross-reactivity is seen, except for anti-AAV1 recognition of AAV6 capsids. The B1 antibody recognizes denatured capsids at a linear epitope near the two-fold axis and is used here to emphasize lack of native virion binding. The ADK4 monoclonal only recognizes native AAV4 capsids, as the B1 epitope does not exist in this serotype.

Page 87: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

87

Figure 3-3. Preliminary neutralization assay data for the anti-AAV5 monoclonals. The non-

neutralizing effects of the IgG BB3C5.F4 (blue) can be seen from this assay, as GFP expression has shown only ~20% knockdown at dilutions of even 1:25. Both IgMs, BB8F1.E8 (magenta) and BB9F7.F12 (yellow) appear to be completely abolishing the ability of the vector to deliver GFP cDNA at dilutions of ~1:100 and ~1:50, respectively.

Figure 3-4. Preliminary neutralization assay data for the anti-AAV1 monoclonals. The non-

neutralizing monoclonal, AA9A8.B12 (A8.B12; green) among the AAV1 antibodies appears to have an effect on the vectors ability to transduce cells at dilutions below ~1:100. While the other monoclonal, AA4E4.G7 (E4.G7; red) and AA5H7.D11 (H7.D11; blue), appear to be able to completely abrogate transduction at dilutions of ~1:100 and ~1:200, respectively.

Page 88: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

88

Figure 3-5. The ability of the anti-AAV1 monoclonals to neutralize the AAV1-like viruses: X1, X25, and AAV6. A) AA4E4.G7 data. The G7 monoclonal antibody appears to retain neutralizing ability, although compared to the AAV1 data, a slight increase in transduction capability is seen for all vectors, especially among the X1 virus. B) AA9A8.B12 data. B12 monoclonal appears to retain its overall non-neutralizing effect even against the AAV1-like viruses. C) AA5H7.D11 data. Neutralization data for D11 shows a significant reduction in neutralizing activity against the X1 virus, while X25 and AAV6 are still neutralized at levels similar to AAV1. AAV6 is shown in blue, X1 is red, and X25 is yellow.

Page 89: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

89

Figure 3-6. Differnces between the AAV1 and X1 sequences are mapped to the atomic coordinates of AAV1 and cluster at the three-fold axis of symmetry. A 3D surface representation of the AAV1 capsid is shown in radial coloring from blue to red, where red represents the furthest distance from the capsid center. A three-fold axis is circled to illustrate the region of interest shown in more detail on the right. Here, three symmetry realted monomers, shown in green, red, and blue, make up the interactions of the there-fold spike. Residue differences netween AAV1 and X1 are highlighted in corresponding colors to depict the highly interactive nature of this region. It is also evident from this image how easily a conformational epitope could be generated from the capsid surface.

Page 90: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

90

CHAPTER 4 DETERMINATION OF DOMINANT ANTIGENIC SITES ON THE CAPSID SURFACE OF

ADENO-ASOCIATED VIRUS SEROTYPES 1, 2, 5, 6, AND 8 THROUGH CRYO-ELECTRON MICROSCOPY STUDIES

Introduction

The use of structure approaches in biology has shed light on the mechanisms of many

interactions, especially macromolecular assemblies and their function in general (Johnson and

Chiu, 2000). It has also been equally important in virology, as it has given insight into the

structural basis of assembly, nucleic acid packaging, particle dynamics, and interactions with

cellular molecules (Johnson and Chiu, 2000). Structural characterizations of viruses are crucial to

the design of therapeutics and vaccines against them and has allowed for the elucidation of

mechanistic pathways. There are many well developed methods for structural determination

approaches available, and the one undertaken for a particular study is often dependent on the

resolution and type of information desired. For instance, both nuclear magnetic resonance

(NMR) and crystallography can give atomic resolution detail on bond interaction and backbone

placement, but NMR also provides dynamic (ensemble) information while crystallography

provides a “snapshot” and is often considered static. In cryoEM, macromolecules are frozen in

their native state allowing for discrete selection of dynamic states to be visualized, albielt at

lower resolution. The largest issue separating cryoEM from crystallography, besides size and the

limitations of crystal formation, is resolution. CryoEM has generally been considered a low-

resolution technique, giving reconstructions around 15-30Å, but with advances in sample

handling, instrumentation, image processing, and model building, near-atomic resolution can

now be achieved (Zhou, 2008). In reality, hybrid approaches, combining NMR, X-ray

crystallography and cryoEM, is often adopted, which provides a powerful means of filling gaps

that can arise in structural characterization of macromolecues. For example, in studies where

Page 91: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

91

large complexes or macromolecular assemblages cannot be crystallized, subcomponents can be

crystallized to obtain high resolution information which can then be used to interprete the

complex structure at lower resolution obtained by cryoEM (Rossmann, 2000). An excellent

example of this approach is highlighted in a recent review on the influenza virus where several

crystallographic studies involving smaller portions of the viral polymerase were combined with a

large cryoEM complex to enable functional interpretation (Ruigrok et al., 2010). Insights into the

function of the replication machinery, like those of the viral polymerase, were deemed from

these studies. This information has become increasingly important due to rising concerns of

influenza pandemics, much like that reported for the 2009 H1N1 strain, and forms a basis for

virtual screening of drug compounds for clinical trials (Ruigrok et al., 2010).

A number of reported cryoEM virus complex structures offer insights into receptor or

antibody interactions, as well as conformational changes associated with replication and virion

assembly (Baker, 1999). CryoEM of immune complexes provides a method for visualizing

antigen–antibody interactions for viral capsids (Smith, 2003). These structures are often

interpreted by docking the known structures of the capsid and FAb or homologous 3D models

(from the RCSB protein data bank, (http://www.rcsb.org/pdb/home/home.do) into a cryoEM

density map of the Fab-decorated capsid (Harris et al., 2006). The resulting pseudo-atomic model

may then be used to define the FAb binding sites in an attempt to identify the peptide(s) that

make up the epitope (Harris et al., 2006). These studies better define the often dynamic surface

of viruses and also further characterize antibody-antigen interactions while offering insight into

their mechanisms of neutralization and potential therapeutic targets.

Numerous virus structures have been determined to date, big or small, pathogenic or non-

pathogenic. While these viruses have unique host characters and function, many similarities can

Page 92: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

92

be found among them. Most of the structural studies on virus-Ab complexes began with the

influenza virus, which laid the foundations for thoughts on how viruses avoid immune

surveillance (Smith, 2003). However, the first study to visualize an intact virion complexed with

a neutralizing antibody was done with rotavirus in 1990 (Prasad et al., 1990). This study had

remarkable impact on the field at two levels. First, it established that protein-ligand interactions

could be visualized by cryoEM and second, it correlated structural and functional information for

the rotavirus capsid proteins (Smith, 2003). Later on, studies with Cowpea Mosaic Virus

(CPMV) would show that pseudo-atomic models could be generated from atomic models fitted

in the density maps, which were then used to identify important regions on the capsid surface

(Wang et al., 1992; Porta et al., 1994). Studies with the Human Rhinovirus 14 (HRV14) took it

one step further and demonstrated that pseudo-atomic models could be generated using all of the

structures in the complex and subsequently used to determine binding regions (Smith, 2003).

There are several structures of HRV14 bound to an antibody molecule (Smith, 2003), which all

seem to be localized in one region. Studies with HRV2 brought more insight into antibody

flexibility, with the observation that Fabs binding to the same site can have distinctly different

orientations and mode of binding, such as bivalency, as well as the identification of a second

antigenic site on the rhinovirus capsid (Smith, 2003). Peptide mapping and analysis of escape

mutants were approaches originally used to identify dominant neutralizing sites to CPV and FPV

capsids (Smith, 2003). CryoEM structures of virus-Ab complexes confirmed these sites (for

CPV) and further demonstrated the recognition of a common antigenic epiope by several

different mAbs generated against AAV the capsid as was observed for the HRVs (Hafenstein et

al., 2009; Wikoff et al., 1994). Many of these studies highlight the fact that the location of these

antigenic loops is a critical factor in determining how to engineer vectors and vaccines and the

Page 93: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

93

notion that dominant antigenic regions exist on the capsid surfaces. All of these data further

support the techniques used here and the work presented in this chapter.

Here cryoEM and image reconstruction of capsid-Fab complexes was used to define the

specific structural contacts between 6 different mAbs (5 neutralizaing and one not) and the

capsids of AAV1, AAV2, AAV5, AAV6, and AAV8, to provide a comprehensive view of the

antigenic structure of the AAVs. These viruses are representative members of the AAV antigenic

clade and clonal isolate groups (Gao etal., 2004) and are ~55 to 99% identical in VP amino acid

sequence, with AAV1 and AAV6 being the most similar and belonging to the same clade group

A. Docking of atomic structures and models into the reconstructed density identified the AAV

surface loops which interact with the respective Fab.

Results

Analysis of the Fab-VLP CryoEM Complexes

In order to identify the binding sites of antibodies recognizing different AAV serotypes,

six different mAbs were chosen for analysis in complex with their respective capsid VLP (Table

4-1). These studies report seven different complexes with resolutions between ~10.8 -22.8Å

(Table 4-2). Two complexes involved the same Fab AA9A8.D11 in complex with AAV1 and

AAV6 serotypes, while the other three mAbs do not cross-react with other serotypes. One

complex, AAV5:F4 appeared to be non-neutralizing in preliminary studies, while the other mAbs

neutralize the virus that they recognize (Figures 3-2 and 3-3). For each complex the final map

was scaled by comparison with known atomic virus capsid coordinates, which were then docked

into the cryoEM reconstruction with fairly high confidence (ranging from a cc of 0.6-0.75).

Excess Fab density most likely accounted for correlations less than 100% (Table 4-3). Models of

the Fab Fv portions were derived from the sequencings of the heavy and light variable domains

of the antibodies which were submitted to the WAM antibody modeler server (Figure 2-2; see

Page 94: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

94

chapter 2). These Fab models were then manually fit into the Fab density that extended from the

capsid surface. Once the atomic capsid coordinates and the Fab homology models were fit into

the cryoEM maps the exposed capsid surface loops that extended into the open Fab density could

be analyzed for possible contact areas. The docked Fab models served as guide points to

determine which loops were most likely involved in protein-protein contacts.

Fab G7 and AAV1

A final map of ~13Å resolution was derived from 2,870 particle images. The most notable

extensions of density occurred at the capsid three-fold axis of symmetry, and extended across the

two-fold region to overlap with the density extension from a symmetry related two-fold region

(Figure 4-1A). This extension of density was recognized as the binding site of the Fab on the

viral capsid surface. The Fab density (~1.0σ) at this region is not as strong as the capsid density

(2.30σ), indicating that steric hindrance may prevent two Fabs binding at the same time. Capsid

surface loops that play a role in the footprint are clustered in VRIV, V, and VIII (Figure 4-1D

and 4-2). These are loops from symmetry-related capsid VPs that must be forming a

conformational epitope. Residues that contact the fitted Fab in the model of a VP1 monomer

include residues 456-459 (AAV1 numbering), giving the short peptide AQNK in VRIV, and

residues 585-590 highlighting the QSSSTD peptide in VRVIII; while residues in a three-fold

related monomer include residues 492-498 - TKTDNNN in VRV (Table 4-4).

Fab D11 with AAV1 and AAV6 Capsids

Reconstructions were generated from 262 particles for AAV1:D11 and 2,527 particles for

AAV6:D11, at resolutions of 22.8Å and 15.3Å, respectively (Table 4-2). Docking of the atomic

coordinates and Fab models identified the binding region as the large protrusion from the center

of the three-fold symmetry axis of both complexes (Figure 4-1B and 4-1C). Density closer to the

three-fold protrusions was stronger (2.20σ) than that seen for the region above the capsid (0.60σ)

Page 95: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

95

correlating to the constant portions of the D11 Fab. Indeed, fitting of a model in both density

portions, i.e. capsid and Fab density, revealed that the constant region of the Fab arm was angled

over the three-fold canyon, just above the center of the two remaining three-fold peaks. This

suggests that only one Fab at a time was bound, possibly due to steric hindrance. Capsid surface

loops in the Fab contact region were identified as VRIV, V, and VIII (Figure 4-1B and 4-1C,

Figure 4-2), which create the three-fold protrusion. AAV1 and AAV6 differ by only six amino

acids, with three of them, E418D, E531K, and F584L located on the capsid surface at the three-

fold axis (Table 3-2). As the Fab bound both viruses in the same position, those residues are

most likely not directly involved directly in the Fab binding, but they may play a role in stability

of the protein interactions. Residues mapped to one VP3 monomer include 456-459 (AAV1

numbering) with the sequence AQNK in VRIV; and 585-590 of QSSSTD in VRVIII (Table 4-4).

A three-fold related interaction places VRV between the VRIV and VIII fingers of the reference

monomer; notably, D495, N496, N497, and N498. The same sequences were identified in the

AAV6 complex due to the high degree of conservation among these two serotypes (Table 3-2).

Fab C37B and AAV2

A total of 4,392 particles were extracted from 149 CCD images to generate a final 3D map

with a resolution of ~10.8Å (Table 4-2). Density was present at each three-fold protrusion,

indicating that there is no steric hindrance for the binding of all three protrusions simultaneously

(Figure 4-1D). The latter region has been identified as being important for the binding of the

AAV2 primary receptor, HSPG (Opie et al., 2003; Kern et al., 2003; Levy et al., 2009;

O’Donnell et al., 2009), and incubation of mAb C37B with AAV2 capsids has shown a strong

reduction of infectivity, possibly due to blocking of receptor mediated entry (Wobus et al.,

2000). The C37B mAb is not cross-reactive with any of the serotypes studied here (Wobus et al.,

2000). Peptide scans identified two binding regions on the AAV2 capsid surface, including

Page 96: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

96

residues 493 to 502 (SADNNNSEYSWT) which showed the highest binding, while residues 601

to 610 (LPGMVWQDRD) was involved to a lesser extent (Wobus et al., 2000). Those peptides

are found near the three-fold axis of symmetry with 493-502 located on the surface of the three-

fold protrusions in VRV, which is cradled by the fingers (VRIV and VIII) of a three-fold related

symmetry monomer. Residues 601-610 map to an interior region just below the central

depression of the three-fold protrusions which is not accessible from the surface and is therefore

most likely important for the structure of the three-fold axes. Currently there is no sequence data

available for this mAb, thus an unrelated Fab structure (PDB 2FBJ) was used for docking into

the Fab density (Figure 4-3). Close inspection of the docked models revealed Fab coverage at the

tips of the three-fold protrusions highlighting the variable regions (VR) within this region;

VRIV, V, and VIII (Figure 4-2). The most obvious residues involved include a portion of the

sequence identified as the major epitope by Wobus et al (2000), residues 496-500 (Table 4-4).

The peptide identified here, NNNSE, includes the two asparagines (underlined residues, N497

and N498) that when mutated in peptide competition experiments caused a partial reversal of

competition (Wobus et al., 2000). The other two proposed regions, in VRIV and VIII (peptides

RGNRQ and TTT, respectively) flank VRV as symmetry-related monomers that come together

to form the three-fold protrusions, and possibly only contribute stability to the main interaction

found at VRV. A surface exposed sequence identified to be important in receptor binding of

HSPG (residues 585-590), located in VRVIII was also identified here (Opie et al., 2003; Kern et

al., 2003), consistent with the reduction in infectivity of AAV2 capsids when incubated with

C37B mAbs due to receptor blocking (Wobus et al., 2000).

Fab F4 and AAV5 Capsids

The fnal density map of AAV5 with Fab BB3C5.F4 included 2,694 particles submitted for

3D rendering, producing a density map to 15.7Å (Table 4-2). These Fabs obscure the capsid

Page 97: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

97

topology, and the 3D map shows a stronger portion of the Fab arm extending from the shoulder

of the three-fold protrusion towards the five-fold axis of symmetry (Figure 4-1E). Additional

analysis also reveals a weaker extension between the shoulders of the three-fold protrusions at

the base of the two-fold axis of symmetry (Figure 4-4). It may be possible that these two regions

are the binding sites for the heavy and light chain of the Fv portion, where residues at the end of

the chain may be stabilized in a non-specific manner. Docking of the AAV5 atomic coordinates

and analysis of the bound Fab structure were done at a high sigma level (~3.5-4.0σ) in order to

identify the capsid regions below the strongest density - the likely contact sites. These regions

were visually mapped to the same symmetry regions that make up the three-fold spike on the

capsid surface at VRIV, V, and VIII. Residues involved in this region were mapped to a

reference monomer at the three-fold as 442-448 (AAV5 VP1 numbering) giving the peptide

NNTGVQ at VRIV; and 572-578 of NNQSSTT at VRVIII. The three-fold related monomer

contributes peptide SGVNR (residues 479-483) at VRV (Table 4-4).

Fab ADK8 and AAV8 Capsids

A total of 1,123 particles were extracted from 78 CCD images to generate a final 3D map

with a resolution of ~18.4Å (Figure 4-5A). Density was present at each three-fold protrusion,

indicating that there was no steric hindrance for the binding of all three protrusions

simultaneously, as was seen for a previous reconstruction, AAV2:C37B (Figure 4-1D). An entire

capsid molecule, or 60-mer, was generated from the AAV8 atomic coordinates (PDB 2QA0;

Nam et al., 2007) and was docked into the respective capsid density with a correlation coefficient

(cc) of ~0.42. The cc of the fit was most likely low due to the presence of the uninterpretted

density belonging to the bound Fabs as well as the use of a C-α model (Figure 4-5B). Since the

ADK8 antibody has yet to be sequenced, a model has not been generated. The atomic

coordinates for a generic Fab model (PDB 2FBJ) were then docked into the identified Fab

Page 98: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

98

density and a density map was generated from this new model (Figure 4-5C). A new cc was

calculated (see chapter 2) and was found to be ~0.68. The correlation was improved when the

excess density was filled, but this low correlation is most likely due to the use of C-α model, as

previously mentioned, in addition to the use of rigid-body and manual fitting techniques. In this

case, these approaches do not account for conformational differences in the docked structures.

This is evident from a comparison of the maps used in the determination of the cc between the

cryoEM reconstruction and the fitted models (Figure 4-5D).

Docking atomic models into the cryoEM map allowed for further characterization of the

ADK8 binding site on the AAV8 capsid (data presented in Appendix B). Visual inspection

identified the Fab binding to the spikes at the three-fold axis of symmetry, leaving the two- and

five-fold axes un-occluded. A more detailed inspection of the docked VP3 capsid monomers

identified VRIV, V, and VIII (Figure 4-2) of the three-fold axes to be important in the antibody

interaction. A lesser, but possible interaction was also noticed at VRVI corresponding to a

KDDEE peptide at aa530-534. These regions were also noted in the previously determined

reconstructions (Table 4-4). Sequence identification of this region proposes that the peptides

GTANTQ (VRIV; aa455-460), TTTGQNNNS (VRV; aa493-501), and LQQQNT (VRVIII;

aa586-591) could be involved in the epitope. It is interesting to note that portions of the peptides

from VRIV and V in AAV8 have been identified in studies as being immunogenic in cytotoxic

T-cell assays, while the peptide identified in VRVIII was found as an epitope in MHC II

molecules (Chen et al., 2006). Another interesting note includes the observation the sequences at

VRV and VIII both have triplicate repeats of hydrophilic, polar residues, i.e. QQQ (Table 4-4).

Fab ADK1a and AAV1 Capsids

The data collected for the ADK1 Fab and AAV1 yielded a reconstruction to ~10.8Å from

1,271 particles (Figure 4-6A). Data for this particular complex was taken from 42 film images at

Page 99: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

99

a magnification of 50,000. The data was collected on the 200KV FEI Sphera, instead of the

300KV FEI Polara used for the other complexes, from 1.5-2.75µm underfocus. Scanning of the

films was done on a Nikon super coolscan 8000ED at a scan step size of 6.35μm/pixel, giving a

final image size of 1.27Å/pixel. Film data was most likely the deciding factor in the higher

resolution obtained for this structure.

Fab density can be seen at each individual three-fold protrusion and appears to bind at an

estimated angle ~45° to the capsid surface. The binding indicates that all axes are open for

interactions, and if the antibody is neutralizing, it most likely does so by steric interference. The

ADK1a monoclonal antibody has been shown to recognize assembled capsids (Kuck et al., 2009)

most likely implying a conformational epitope at the tip of the three-fold protrusion. Fitting of

atomic models into the cryoEM map (Figure 4-6B) has illustrated that the most probable region

of interaction is at VRIV and corresponds to the peptide 453SGSAQ457. These residues flank

the region previously found at VRIV in the other structures. It appears that the region also

previously identified at VRV, 496NNNSNF501 may help to stabilize the interaction. Because

this region is also found in AAV2 (495 NNNSN498) it is unlikely that it is involved in the

epitope itself. However, it is possible that the last two residues at AAV1, 500NF501 are

involved, since these differ from AAV2 at 499SE500. Mutagenesis will be able to confirm this

interaction. This antibody has also been found to recognize AAV6 capsids, which further

supports the region identified here due to the identity between AAV1 and AAV6 at these

regions.

Common Features of Antibody Binding Sites on the AAV Capsid

Mapping the antigenic regions of the different virus-antibody complexes shows that there

is significant overlap among these epitopes at the three-fold axis of symmetry (Figure 4-7). A

structure-based sequence alignment of AAV1, 2, 5, 6, and 8 (Figure 4-8) shows that the epitope

Page 100: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

100

residues have stretchess of high variability, adopt different surface loop conformations, but form

similar structural features, such as the protrusions in the proposed regions of antibody binding.

Discussion

Here the antigenic structures of the AAV capsids that are recognized by a set of antibodies

have been characterized. This study also shows that there are common antibody recognition

properties that are shared by the capsids of these four distinct AAV serotypes. The two mAbs

tested that recognize the intact AAV1 capsid, AA4E4.G7 and AA5H7.D11, were produced after

extended immunization of mice and likely represent affinity mature IgGs, including the C37B,

ADK1a, and ADK8 IgGs. The BB3C5.F4 anti-AAV5 mAb was generated only 4 days after

intact AAV5 capsid immunization of a mouse which had been previously immunized with

AAV1and may represent an immature antibody.

Viruses as Antigens - Features Revealed

The antibodies tested here all bound on or near the three-fold protrusions on the capsids of

the four distinct serotypes. While the specific sites of interaction vary, with some binding nearer

the top and others on the side of these structures, there was a common overlapping area within

the footprints of all the antibodies on all capsids (Figure 4-7). All epitopes appeared

conformational in nature, as each antibody made multiple contacts with sequences from

symmetry-related monomers, which were displayed as variable loops in the assembled structure

of the capsid. The capsid surface contacted by the Fabs did not show any specific features, other

than being a raised structure with three-fold symmetry, and the distributions of amino acids in

the interacting regions were not significantly different from those seen in the remainder of the

capsid surfaces. However, it is interesting to note that some residues identified in these proposed

epitopes were highly repetitive. Indeed, further examination of the AAV1-9 sequences show that

there are several instances of identical triplet aa stretches, i.e. TTT, almost all occurring at the

Page 101: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

101

capsid surface regions. However, the significance of this find is yet to be determined. The

regions identified as possible epitopes were also found to exhibit an overall hydrophilic

environment with predominantly polar side chains.

The most obvious feature of the structures bound by the antibodies was that they were

raised above the capsid surface. This feature is a structural property that has been revealed for

antigenic sites on the canine and feline parvoviruses, as well as other viral capsids (Kaufmann et

al., 2007; Hafenstein et al., 2009; Moskalenko et al., 2000; Smith, 2003). However, the other

raised region of the AAV capsid, the cylinder that surrounds the pore structure at the five-fold

axis of symmetry, did not bind the antibodies in this study or among the 8 antibodies examined

for the canine and feline parvovirus capsid (Hafenstein et al., 2009), and it may therefore require

more than just structural prominence to induce the formation and affinity maturation of

antibodies. The loops in that structure are flexible which may influence the attachment of

antibodies and the selection of high affinity IgGs (Agbandje-McKenna and Chapman, 2006).

Structures of Bound Antibodies and Possible Mechanisms of Neutralization or Effects on Capsid Functions

Studies of other viruses and antibodies have shown that there are several possible

mechanisms of neutralization by different antibody-virus combinations (Smith, 2003). The

neutralization mechanisms of many anti-AAV antibodies have not been investigated in detail,

but some examples have been reported. Although we have not yet examined the capsid-cell

infection processes of all the antibodies and Fabs tested here, the structural analysis provides a

number of predictions about the likely effects of antibody binding. At least for the AAV2:C37B

structure there is more conclusive evidence that the mechanism of neutralization is indeed

blocking by obscuring cellular receptor attachment as biochemical data (Wistuba et al., 1997)

and now structural data identify the HSPG receptor site as being involved.

Page 102: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

102

Since the structures were determined in context with Fabs instead of the full mAb it is

difficult to conclude that bivalent binding had occurred for any of the antibodies studied here.

However, there are some generalizations that can be made that suggest bivalent binding for the

AA4E4.G7 mAb. In the case of the AA4E4.G7 Fab, there is strong density seen at the base of the

three-fold protrusion and hovereing above the two-fold axes in a cross-linking manner (Figure 4-

2A). However, it has been shown that bivalent binding does not have to occur over the two-fold

axis (Thouvenin et al., 1997). Bivalency on such a repetitive antigenic surface as a virl capsid

generally hints at an increased avidity causing both Fab arms to bind the capsid surface, resulting

in a marked decrease in capsid aggregation. There is no structure of the AA9A8.B12 Fab bound

to an AAV1 capsid, but preliminary complexing studies using negative staining techniques

identified a lack of aggregagtion at several dilutions of mAb (Figure 3-6). It was previously

noted in studies with a bivalent mAb against RHDV that small amounts of aggregation may be

indicative of bivalency (Thouvenin et al., 1997). There are two thoughts here, one being the

direct overlap of the antibody attachment site and the receptor binding site that would likely lead

to competition for the binding site, and the other would be the occlusion of these regions on the

capsid surface by the Fab structures, particularly where the Fabs bind at oblique angles so that

they would block an even larger region of the capsid than the direct footprint. The effectiveness

of competition may be lower for the AA5H7.D11 Fab, as that only occupies one of 3 possible

binding sites near the three-fold axis, meaning that forty sites would remain open even when the

capsid was saturated for antibody binding. However, other studies have noted that not all sites

need to be covered to induce neutralization (Thouvenin et al., 1997). Some antibodies can

stabilize the capsid against structural changes required for uncoating within the cell, which

would still allow cell binding and internalization (Smith, 2003). It appears that AA4E4.G7, if

Page 103: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

103

binding across the two-fold as proposed, could lock the capsid in a position where uncoating may

not be possible, if the two-fold is involved in uncoating. Indeed, further studies are required to

isolate the step in the life cycle that is neutralized by these antibodies, but studies in reovirus

have identified large structural changes at the binding site (Nason et al., 2001). Conversly, it has

been proposed that antibodies do not neutralize by gross conformational changes (Smith, 2003).

Most likely due to the thought that for antibody-induced conformational changes to be important

for neutralization, clonal expansion of B-cells would need a mechanism to choose for B-cells

that would generate antibodies that induce conformational changes (Smith, 2003). Although

small structural changes were noted in a few virus-antibody reconstructions (Li et al., 1994;

Bothner et al., 1998, Lewis et al., 1998), it is more likely that capsid dynamics play a role here

instead of antibody-induced changes (Smith, 2003).

Collaborative studies with Drs. Jurgen Kleinschmidt and Christina Raupp included the

ADK8 data to further characterize this mAb and its interaction with the AAV capsid. In these

studies mutations were made in the proposed antigenic regions on the AAV8 capsid (Table 4-4)

generating similar regions corresponding to AAV2 and ultimately found that when the AAV8

peptide 586LQQQNT591 at VRVIII was changed to the AAV2 peptide 586LQNRGNR591

ADK8 recognition was abolished (Appendix B). This region was further confirmed when AAV2

capsids were mutated to the corresponding AAV8 antigenic peptide (Appendix B). Further

analysis included investigating neutralization efficiency, using in vivo and in vitro techniques,

and mechanisms of neutralization. Both in vivo and in vitro data confirmed neutralization activity

by the ADK8 monoclonal and preliminary mechanistic studies further identified that binding and

internalization of AAV8 in HepG2 cells was not perturbed (Appendix B).

Page 104: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

104

Table 4-1. Summary of AAV-Fab complexes generated for cryoEM data studies Monoclonal antibody (mAb)

Parental Antigen

Antigen used in complex

Complex formed

AAV4E4.G7 AAV1 AAV1 AAV1:G7 AA5H7.D11 AAV1 AAV1 AAV1:D11 C37B AAV2 AAV2 AAV2:C37B BB3C5.F4 AAV5 AAV5 AAV5:F4 AA5H7.D11 AAV1 AAV6 AAV6:D11 ADK1a AAV1 AAV1 AAV1:ADK1a ADK8 AAV8 AAV8 AAV8:ADK8

Table 4-2. CryoEM data reconstruction statistics Antigen: antibody complex

Number of micrographs

Range of defocus

Includes focal pair data

Number of particles boxed

Number of particles used

Final resolution (~Å)

AAV1:G7 95 1.50-3.0 No 2,376 1,914 12.3 AAV1:D11 31 1.25-3.0 No 313 262 22.8 AAV2:C37B 149 1.50-3.0 Yes 4,731 4,392 10.8 AAV5:F4 122 1.25-3.0 No 2,806 2,552 15.7 AAV6:D11 49 1.00-3.0 Yes 5,265 2,527 15.3 AAV1:ADK1a 49/film 1.00-3.0 No 1,387 1,271 10.8 AAV8:ADK8 78 1.00-3.0 No 1,923 1,123 18.4

Table 4-3. Scaling and fitting statistics of cryoEM 3D reconstructions with atomic models 3D Reconstruction Scaling CC Final Å/pixel Fitted AAV + Fab

model CC AAV1:G7 0.80 1.863 0.82 AAV1:D11 0.81 1.850 0.65 AAV2:C37B 0.65 1.883 0.73 AAV5:F4 0.67 1.800 n/a AAV6:D11 0.77 1.850 0.63 AAV8:ADK8 0.60 1.900 0.68

Table 4-4. Proposed epitopes for each respective complex including residue numbers and

sequence data Complex VRIV* peptide: amino

acid sequence VRV* peptide: amino acid sequence

VRVIII* peptide: amino acid sequence

AAV1:G7 456-459:AQNK 492-498:TKTDNNN 585-590:QSSSTD AAV1:D11 456-459:AQNK 492-498:TKTDNNN 585-590:QSSSTD AAV1:ADK1a 453-456:SGSA -- -- AAV6:D11 456-459:AQNK 492-498:TKTDNNN 585-590:QSSSTD AAV2:C37B 454-456:TTT 496-500:NNNSE 585-590:RGNRQ AAV5:F4 442-448:NNTGVQ 479-483:SGVNR 572-578:NNQSSTT AAV8:ADK8 455-460:GTANTQ 493-501:TTTGQNNNS 586-591:LQQQNT

*residue numbering is respective to the serotype indicated in each complex

Page 105: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

105

Figure 4-1. CryoEM 3D reconstructions shown with respective roadmap of epitope positions on the 3D capsid surface A) AAV1:G7 complex to ~12.3Å B) AAV1:D11 complex to ~22.8 Å C) AAV6:D11 complex to ~15.3 Å D) AAV2:C37B complex to ~10.8 Å E) AAV5:F4 complex to ~15.3 Å. All viral surfaces are colored respectively, AAV1-purple, AAV2-blue, AAV5-grey, AAV6-pink, while the Fab density is shown in light grey. Maps are viewed down the two-fold axis as indicated in the roadmap image.

Page 106: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

106

Figure 4-2. Variable regions of the AAV-VP3 monomer A) Colored loops on the monomer represent the variable regions (VR) of the monomeric unit and are indicated with Roman numerals. The conserved core structure is shown in grey. Icosahedral symmetrical axes are depicted with a triangle and the number 3 for the three-fold axis, while the two-fold axis is depicted by an oval and the number 2 and the five-fold is shown as the number 5 set into a pentagon shape. B) The same monomer repeated 60-times generates an entire capsid structure and is viewed here in a surface representation. The VRs can be seen interacting with one another in context of the surface and it is evident that the capsid surface is highly variable. The VRs are colored the same as in part A and a triangle represents the asymmetric unit of the AAV capsid.

Figure 4-3. Docking of atomic models into cryoEM density A) The atomic coordinates of AAV2 (PDB 1LP3) were docked into the capsid density for the AAV2:C37B complex while atomic coordinates of a generic Fab (PDB 2FBJ) was used for the Fab density. Symmetry related capsid monomers at a three-fold axis are depicted in green, blue, and magenta and the Fab is shown in yellow. B) Zoomed in image of the broken line-boxed region shown in part A. Surface loops from the Fab model can be seen in close proximity to the capsid surface monomers. These regions were chosen as sights of possible interaction.

Page 107: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

107

Figure 4-4. Difference map between the AAV5:F4 reconstruction and a similar scale AAV5-uncomplexed map. Difference map density can be seen as black mesh on the light grey capsid surface. The red-dash oval depicts the region covered by an entire Fab molecule. Within this region there appeared to be two sites contacting the capsid surface. The first can be seen at the regions between the five and three-fold axes, in a black-dash box and the second at the shoulders of the three-fold axis, in a black-solid box, spilling into the two-fold. Model docking appears to correlate better with Fab fitting to the region identified with the black-solid box. The 3D map is viewed down the two-fold axis at a 90 degree rotation from the maps in Figure 4-1.

Page 108: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

108

Figure 4-5. Three-dimensional reconstruction and model fitting for the AAV8:ADK8 complex. A) Final density map for the AAV8:ADK8 structure to ~18.4Å. The AAV8 viral surface is colored green while the excess density in white was identified as the Fab density. B) Fitting of the AAV8 (PDB 2QA0) structure into the respective capsid density. Density map is shown in grey and the atomic coordinates are in green. The excess white space remaining for the Fabs could easily account for the low fitting statistics. C) Fitting of both Fab and AAV8 capsid models into the reconstructed density. One Fab portion, modeled in as PDB 2FBJ, is shown in red and is hovering above three symmetry related monomers, colored in green, blue, and magenta, at the three-fold axis of symmetry. D) Visual comparison of the maps used to generate the final correlation of the docked models (green) and the original cryoEM reconstruction (grey). An excess of green density from the fit model map can be seen extending beyond the cryoEM map, indicating regions that would decrease the final corrolation. Black arrows mark example regions of density extension.

Page 109: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

109

Figure 4-6. AAV1:ADK1a reconstruction and model fitting. A) Three-dimensional rendering of the ~10.8Å density map. The virus is shown in purple with the Fabs depicted in white and the three-fold axis is marked by a black triangle. B) Atomic models were fit into the virus:Fab cryoEM map to isolate regions on the capsid that are interacting with capsid surface. The capsid monomers making up the highest point on one three-fold protrusion are colored in green and blue. It appears that one major loop, identified as VRIV, is most involved in the interaction. It is highly possible that other VRs close by are helping to stabilize or interact directly with the antibody.

Figure 4-7. Illustration of overlapping antigenic regions at the capsid surface level A) An AAV capsid surface is depicted in cartoon diagram illustrating the overlapping regions identified through structural epitope mapping. The DE and HI loops that make up the five-fold pore and surrounding floor, respectively, are offset in dark grey for orientation, while the overlapping antigenic residues are highlighted in black spheres. These regions sit at the shoulders of the three-fold protrusions. B) A roadmap of the 3D capsid surface maps the regions of interest in black to the three-fold axes. An assymetrical unit is shown on both surfaces, red triangle in A and a black triangle in B. A pentamer shape is representing the five-fold axes, while an oval depicts the two-fold, and filled triangles map the three-fold axes.

Page 110: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

110

Figure 4-8. Structure based sequence alignment of AAV1, 2, 5, and 6. Variable regions (VR) are marked by roman numerals and specific sequences of interest are underlined in red. The numbering to the above the sequences follows AAV2 VP3 numbering.

Page 111: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

111

CHAPTER 5 MUTATIONAL ANALYSIS OF PROPOSED DOMINANT ANTIGENIC SITES ON THE

CAPSID SURFACE OF ADENO-ASSOCIATED VIRUS SEROTYPE 1

Introduction

The AAV capsid surface is responsible for a multitude of interactions and functions within

the viral life-cycle including host cell receptor recognition, pathogenicity determination, viral

genome encapsidation, capsid assembly, nuclear import and export, endosomal escape,

maturation to infectious virions, and recognition by and evasion from the host immune response

(Agbandje-Mckenna and Chapman, 2006). Mutational analysis, including the use of insertions

and cross-packaging, of the AAV capsids have answered many questions about the role of the

VPs in assembly, tissue tropisms, antigenicity, and even gene transfer (Lochrie et al., 2006;

Hauck et al., 2006; Opie et al., 2003; DiPrimo et al., 2009; Wu et al., 2000; Xu et al., 2005; Shi

et al., 2006 Rabinowitz et al., 1999, 2002; Grifman; et al., 2001; Huttner et al., 2003). However,

these studies have also created more questions. For example, AAV2 mutants that lack HSPG

binding ability can no longer transduce the liver at comparable levels, but still retain a robust

tropism for heart tissue (Kern et al., 2003). Since the atomic structures of almost all of the known

AAV serotypes have been solved, examination of the capsid surface in a three-dimensional

context has aided in the identification of functional regions important in the viral lifecycle (Xie

et al., 2002; Nam et al., 2007; Govinda et al., 2006; unpublished data).

Almost all of the structural studies conferring functionality to domains on the AAV capsid

have involved AAV2. These studies include the identification of the primary cell-surface

receptor, HSPG (Summerford and Samulski, 1998) and the capsid residues that confer binding

(Opie et al., 2003; Kern et al., 2003) as well as the identification of potential co-receptors

necessary for internalization of AAV2 (Qing et al., 1999; Kashiwakura et al., 2005; Summerford

et a., 1999; Akache et al., 2006). Previous studies aimed at identifying antigenic regions have

Page 112: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

112

also identified both T-cell and B-cell epitopes which generally map to the core structural

components and the capsid surface, respectively (Figure 1-1; Wobus et al., 2000; Mays et al.,

2009). Further studies in cellular uptake and trafficking have hinted that rearrangements at the

capsid surface level occur during receptor interaction that may allow for the exposure of residues

necessary for subsequent steps in the trafficking pathway (Levy et al., 2009; Lochrie et al.,

2006). This work has given new insight into the dynamics of these regions and the role that they

play in the capsid framework. In further support, there are several structures available which

incorporate receptors and antibodies against AAV, including the work presented here, to further

identify the multitude of interactions at the capsid surface level and beyond (Levy et al., 2009;

O’Donnell et al., 2009; unpublished data).

Increased interest in alternate serotypes has broadened many of these types of

investigations to include other serotypes. Several studies have already identified cell-surface

receptors for AAV1, 3, 4, 5, and 6 (Walters et al., 2001; Kaludov et al., 2001; Wu et al., 2006;

Di Pasquale et al., 2003). In this study we aim to further characterize the antigenic properties of

AAV1 through mutational analysis at variable regions previously identified to be involved in

antibody recognition (Chapter 4).

Results

Generation of AAV1 Capsid Mutants

Recombinant AAV1 mutants were generated in a plasmid encoding the AAV1 cap gene

and an AAV2 rep gene. Both the packaged genome, GFP, and the Ad virus helper functions

were supplied in trans on another plasmid, using UF11 with the pXX6, respectively. Mutational

primers were designed against the VRs IV, V, and VIII and targeted the previously determined

sequences (Table 4-4). The idea was to change the AAV1 sequence to that of the structurally

similar region in AAV2 because the AAV2 capsids were not recognized in native dot blot

Page 113: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

113

analyses (Figures 3-2A and 3-2C). This would generate 7 mutants where the three regions could

be tested separately and in context with one another. The first ‘single-site’ mutant at VRIV was

changed from 456AQNK459 on AAV1 to 455TTQS458 on AAV2 and named mut1AB as all the

mutations were done in a two-step process (see methods chapter 2). The second mutant had

mutations at VRV, from 492KT493 on AAV1 to 493SA494 on AAV2 and was referred to as

mut2AB. The final single-site was at VRVIII and was changed from 586SSST589 on AAV1 to

585RGNR588 on AAV2, which has been associated with HSPG binding, and was referred to as

mut3AB. The four remaining mutants included all the possible combinations of the single-sites

to create ‘double-sites’, 1AB2AB, 1AB3AB, and 2AB3AB, and finally a mutant which harbors

AAV2 sequences at VRIV, V, and VIII, 1AB2AB3AB, which has yet to be generated (Table 5-

1).

Analysis of Mutant Protein Expression

The ability of the capsid mutants to be expressed in the 293 cell system was checked by

standard western blotting techniques using the B1 antibody that recognizes a common, linear

epitope at the C-terminus (see chapter 2). The monoclonal, 1F, was also used to detect that Rep

proteins were being expressed. Western blots on cell lysates of single-site mutants revealed that a

similar amount of the VPs were being expressed during transfections (Figure 5-1). This blot also

identified the expression of Rep proteins 52 and 42 (Figure 5-1).

Characterization of Mutant Capsid Production

The observation of VPs in a western blot does not indicate that intact capsids are being

generated during the transfection process. To check for the ability of the antigenic mutants to

form particles a native dot blot could not be done as the nature of the mutants was to evade

detection by capsid antibodies, and the lack of recognition could therefore not confirm antibody

escape, but possibly just be due to the lack of intact particles. Thus, in order to identify capsid

Page 114: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

114

populations, cell lysates were further purified by iodixanol-gradients and ion-exchange

chromatography (see chapter 2 methods). Fractions from the iodixanol gradient were similar to

the rAAV1 vector having B1 positive fractions at the 40/60% and 40%, except for mut3AB

which showed VPs in the 40% and 40/25% (Figure 5-2A). Anion exchange chromatography also

showed identical elution fractions for all samples (Figure 5-2B). These purified samples were

then visually inspected by negative stain EM for the presence of intact particles. The single-site

mutants were all capable of forming intact virions as indicated by the presence of particles in the

EMs (Figure 5-3).

Quantification of Mutant Particle Titers, Infectivity, and Packaging Ability

In order to determine mutant capsid titers an ELISA was performed using the ADK1a

mAb. The binding region for this antibody has been mapped to regions just flanking the mutants

at VRIV (Table 4-4) and it was determined by native dot blot (Figure 5-4) that this antibody will

still bind the antigenic mutants and that any changes in titer seen can be directly correlated with a

defect in the mutant itself. Capsid titers were determined from separate analyses using cell

lysates from independent transfections performed at different times. Titers show that mut1AB

and mut2AB produced similar intact capsid amounts when compared to each other at 1.06 x 1014

and 1.72 x 1014 respectively (Table 5-2). However, there was at least a two log decrease in the

titers seen for mut3AB at 1.22 x 1012 (Table 5-2). rAAV1 controls produced higher amounts of

intact capsid than all three single-site mutants at an average titer (n=3) of 2.70 x 1014 (Table 5-2).

Preliminary particle assembly values for the double-site mutants confer a similar observation

where mutants involving aa swapps from AAV1 to AAV2 at both VRIV and VRV

(mut1AB2AB) have near control values (~2.34 x 1014 particles/ml), while those harboring

swapps from VRVIII show a slight loss when combined with VRV (2AB3AB; ~2.91 x 1013) and

a drastic drop when VRIV is included (mut1AB3AB; 1.55 x 1011) (Table 5-2).

Page 115: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

115

Infectivity of the mutants was checked by infectious assays using the direct visualization of

green cells (see methods chapter 2). The mutants were all determined to be infectious at similar

values with mut1AB being lowest by less than one log (Table 5-2). Double-site mutants have yet

to be checked for infectivity.

Quantitative PCR (qPCR) was used to determine the packaging ability of each mutant in

context of rAAV1 as a control. However, results indicated that more genome was packaged than

capsids were made (Table 5-2). The error could easily be due to several instances, including

background interference of non-specific primer recognition, pipetting errors, technique, or is a

reflection of the sensitivity differences between ELISA and qPCR. Data for the double-site

mutations is currently under evaluation and is not available for inclusion in this report. Assays

will need to be repeated to ensure accurate data collection, possibly including optimization of

DNA extraction protocols. Acquisition of the data for the double-site mutations is currently in

progress.

Ability of Antigenic Mutants to Evade Monoclonal Antibody Recognition

Cell lysates were used in a native dot blot analysis to check for the ability of the parental

mAbs to recognize the mutants. Of the single-site mutants, it appears that the mutations in VRIV

and VRV reduce the ability of the AA4E4.G7 antibody to recognize the AAV1 capsid, seen as a

~27% and ~75% drop in pixel intensity, respectively, (Figure 5-5). Prelimianry analysis thus

indicates VRIV and VRV as important in AA4E4.G7 mAb recognition, with mut2AB (VRV)

having a greater role, but further analysis with combinatorial VR mutations in VRV and VRVIII

(mut2AB3AB) shows a drastic reduction in recognition (Figure 5-5B). Intersetingly, there is an

increase (~32%; Figure 5-5B) in AA4E4.G7 recognition of mut3AB, possibly indicating a

stabilization or increased affinity for these capsids. The AA5H7.D11 antibody was still able to

recognize all of the single-site mutants (Figure 5-5A), but a reduced pixel intensity was observed

Page 116: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

116

for mut3AB (~56% reduction) as well as the double-site mutants mut1AB3AB and 2AB3AB (at

~61% and 62% respectively). The AA9A8.B12 antibody still recognizes mut2AB and mut1AB,

however there is definitely a loss in the AA9A8.B12 ability to recognize mut3AB (~86%

reduction; Figure 5-5B). This shows that the epitope for B12 exists in the AAV1 VRVIII. A

negative control of un-transfected cells was also used to isolate any unspecific recognition from

the cell lysates, as this background was subtracted in pixel intensity calculations.

Direct Effects of Mutations on Neutralization

An infectious assay for the singe-site mutants was repeated (as indicated for the

neutralization assays done by the Chiorini lab, NIH) and the AA4E4.G7 and AA5H7.D11

neutralizing mAbs (Nabs) were included at a ratio of one mAb per one capsid binding site (or

monomer). As seen in the native dot blot, both neutralizing antibodies knocked down rAAV1

infectivity to below visible detectable levels (Figure 5-6A-C), while infectious units were

drastically knocked down to ~10% of the original recombinant infectivity units (Figure 5-7A). In

conjunction with mut1AB un-treated levels (Figure 5-6D) the mutant’s ability to escape

recognition by the AA4E4.G7 Nabs (Figure 5-6F) was evident as comparable levels of infectious

units were accounted for in the green cell assays (Figure 5-7B). This data also confirms that

mut1AB has retained recognition by AA5H7.D11 as was previously seen in the native dot blot

(Figure 5-6E and Figure 5-5). Although, mut2AB has shown comparable results to rAAV1 titers,

infectivity, and packaged genomes (Figure 5-6A and Figure 5-6G; Table 2), as well as

recognition by the AA5H7.D11 NAb (Figure 5-6H), it has now acquired the ability to escape

recognition by Nab AA4E4.G7 (Figure 5-6I). The levels of infectious mutant virus treated with

AA4E4.G7 are near un-treated levels of mut2AB (Figure 5-7C). In accordance with the native

dot blot (Figure 5-5), mut3AB was still recognized by AA4E4.G7 and AA5H7.D11 (Figure 5-6J-

L). However, a slight drop in recognition by the AA5H7.D11 mAb for mut3AB was observed as

Page 117: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

117

seen by the appearance of green cells in the infectious assay, which is also in accordance with the

pixel intensity calculations for this mutant (Figure 5-5B).

Discussion

Previous structural studies involving complexes of AAV capsids and anti-AAV Fabs

(Chapter 4) led to the observation that antigenic regions on the capsid surface cluster at and

around the three-fold axis. A majority of these structures involved AAV1 and a different

monoclonal antibody, including ADK1a, AA4E4.G7, and AA5H7.D11, hence the use of AAV1

as the backbone for mutational analyses. Further analysis of these structures allowed for the

selection of VRs within the capsid monomers (VP3) that appeared to have contact with the

antigen binding regions. The capsid sites were identified as VRIV, VRV, and VRVIII (Table 4-

4) and although there are several VRs that give rise to the topology seen at the three-fold, these

VRs specifically make up the three-fold protrusions. In order to confirm that these VRs were

involved in the antigenic epitope, the AAV1 cap gene was mutated in VRs IV, V, and VIII to

corresponding VRs in AAV2 and subsequently tested against the anti-AAV1 antibody panel.

Preliminary Analysis on Mutant AAV1 Viruses Reveals a Retained Ability to Assemble Infectious Capsids

Assembly studies and mutational analysis on the AAV2 VPs have identified various

regions that confer properties important for assembly, packaging, and infectivity which have

variable levels of tolerance for mutagenesis (Wu et al., 2000, DiPrimio et al., 2008; unpublished

data). In addition, combinatorial studies among several different serotypes also eluded that

certain VRs can be switched between the different AAVs in order to produce chimeric vectors

with different functional profiles (Rabinowitz et al., 2002). The mutations made here included

swapping amino acids in variable surface loop regions among two different serotypes, AAV1

and AAV2, which have almost 83% identity. For the singe-site mutations, particle assembly

Page 118: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

118

appeared to be near recombinant AAV1 control values (Table 5-2) indicating that these regions

in the AAV1 capsid tolerate the corresponding functional domains from the AAV2 capsid.

However, inclusion of the AAV2 receptor binding domain (VRVIII) onto AAV1 (mut3AB)

appears to have decreased particle production by at least two logs (Table 5-2). This region is

known to carry a highly basic charge and may offer a challenge for the structural integrity of

these residues in context of the AAV1 background. Similar observations have been identified for

the double-site mutants where mut1AB3AB shows a drastic reduction (~3 logs) in particle

assembly compared to recombinant controls (Table 5-2). Although the addition of VRV from

AAV2 (mut2AB3AB) appears to rescue this defect as reasonable (near control values) capsid

tites were identified (Table 5-2). This phenomenon is likely due to the structural arrangement of

these VRs, as VRV from a symmetry related monomer sits between VRIV and VIII of another

three-fold related monomer. The addition of residues from AAV2 in VRV most likely act as a

‘buffer’ by helping to further stabilize the normal interactions of the AAV2 type residues in the

AAV1 background. Interestingly, VRV is fairly conserved between AAV1 and 2 (Figure 4-9) yet

the inclusion of the two residues that differ here (AAV1:KT AAV2:SA) possibly offer more

stability for capsid assembly at this region. This data may also support the importance of these

residues for AAV2 stability in this region.

Packaging ability was determined by qPCR analysis of extracted DNA and indicated that

all capsids created retained the ability to package. Since the AAV1 single-site mutant capsids

now harbor amino acids in the outermost extensions of the three-fold VRs IV, V, and VIII,

which were functional in the parental AAV2 capsid, it appears to be possible that no packaging

defect exists for these mutants. Previous studies in AAV2 indicated packaging defects for 4-5aa

linker insertion at residues 595 and 597(VP1 numbering) (Rabinowitz et al., 2000), however this

Page 119: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

119

phenotype is not observed here as substitutions, and not insertions, were created. Also, previous

studies have not exploited these regions as being important for interactions with the Rep proteins

or packaging (Wu et al., 2002). However, since the values for PCR were higher than those values

obtained for capsid production (Table 5-2), it is hard to conclude that the packaging data is

accurate. In further analysis, this may be an artifact of the sensitivity difference between the two

techniques and will require more thorough confirmation, possibly with optimization of the DNA

extraction protocol.

Green cell assays revealed that two of the three single-site mutants retained near control

levels of infectivity, with the exception of mut1AB, which harbors VRIV residues from AAV2.

Since total capsid levels of this mutant were lower compared to the others, it is possible that this

drop is due to the difference in capsid production and also implies that infectivity assays should

be repeated holding capsid titers comparable during the assay. With the data available, it appears

that infectivity levels are within the same range suggesting that infectivity has not been altered

and that these regions are not interfering with any primary cell surface receptor recognition in the

cell type (HEK 293) tested. However, infectivity data will most likely be different depending on

the cell type used and studies will benefit greatly from taking these mutants into in vivo

infectivity assays. AAV1 capsids have been shown to very weakly bind heparin agarose beads

(Opie et al., 2003), and AAV1 mutants with VRVIII (mut3AB, mut1AB3AB, and 2AB3AB)

from AAV2 now contain the heparin-binding motif RGNR (Opie et al., 2003; Kern et al., 2003).

Subsequently, these mutants were tested on heparin-agarose columns and results concluded that

the more intense heparin-binding ability of AAV2 was also transferred to the AAV1 capsids

(Figure 5-8). Overall, these data show that the single-site mutant capsids were assembly

competent (Figure 5-3) and retained the ability to infect cells at levels comparable to the rAAV1

Page 120: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

120

control (Table 5-2). The double-site mutants were only assumed to create capsids based on

recognition by the ADK1a ELISA and further studies are currently underway (Table 5-2).

Ability of Antigenic Escape is Limited to Specific Regions on the Capsid Surface

Single-site AAV1 mutants were tested for recognition by the anti-AAV1 panel of

antibodies that were used in the structure determination of this project (Chapter 4). Although

these studies identified the three VRs IV, V, and VIII as being involved in the epitope,

mutational analysis has made it increasingly more evident that antigen recognition is not a broad

activity. The site for AA4E4.G7 binding has been disrupted with the mutation of either VRIV

(mut1AB) or VRV (mut2AB), although the reduction appears greater for mut2AB (Figure 5-5B).

It is quite possible that either both sites are involved in a conformational epitope, or that steric

hindrance occurs when one site or the other is altered. Intriguingly, the double-site mutants with

either one or both of these regions mutated appears to have a reduction in recognition of the

capsid antigen, with ~65% and 56% reductions in VRIV+VRV (mut1AB2AB) and

VRIV+VRVIII (mut1AB3AB), with VRV+VRVIII (mut2AB3AB) having nearly abolished

recognition completely (Figure 5-5B). This drastic reduction indicates that the combination of

VR substitutions in mut2AB3AB (both VRV and VRVIII) contribute to the AA4E4.G7 epitope.

In order to confirm this, it is necessary to place these regions of AAV1 onto another serotype and

re-probe for recognition. The lack of recognition for mut3AB mutants by AA9A8.B12 indicates

that this region is involved in the antibody epitope (Figure 5-5). Double-site mutants

incorporating AAV2 VRVIII residues also confirm this observation as mut1AB3AB and

mut2AB3AB appear to have escaped recognition by the AA9A8.B12 antibody with an ~87% and

91% reduction respectively (Figure5-5B). Since there is no structure yet of the AAV1 capsid and

the AA9A8.B12 Fab, this find illustrates the strength of this technique in the elucidation of

common antigenic regions. Again, transferring these residues into the background of another

Page 121: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

121

serotype will confer the binding region for this antibody. As an aside, it is intriguing to note that

this specific monoclonal is non-neutralizing, and following that its binding site is structurally

superimposable with the HSPG binding site of AAV2, the primary cell surface receptor-binding

region is most likely not within this region. On the other hand, it has been noted before that

parvoviruses have differing sites of receptor attachment (Agbandje-McKenna and Chapman,

2006). AA5H7.D11 still appears to recognize all double-site mutant capsids through native dot

blot analysis, although recognition reductions were seen in the pixel intensity calculations for

mut3AB, mut1AB3AB and 2AB3AB (Figure 5-5B). Infectivity assays conducted in the presence

of the AA5H7.D11 Nab indicate that mutations in VRVIII (mut3AB) may play a role in escape

from this mAB (Figure 5-6), as double-site mutants with VRVIII mutations also indicate a

reduced binding ability (Figure 5-5B). Overall, it appears that mutations in VRV and VRVIII

(mut2AB3AB) have the greatest affect on antibody recognition for the specific panel of

antibodies studied here as native dot blots revealed reduced recognition for AA4E4.G7 (near

100%), AA5H7.D11 (~62%), and AA9A8.B12 (~91%). In support, the AAV1-like virus X1 has

mutations D495G and D590H, which are also mutated in mut2AB3AB, and correlate to the drop

in AA5H7.D11 mAb recognition (Figure 5-5B). Infectivity assays for this double-site mutant

should confirm this observation.

Page 122: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

122

Table 5-1. AAV1 mutational nomenclature and sequence differences AAV1 mutant AAV2 VRIV:

455TTQS458 AAV2 VRV: 493SA494

AAV2 VRVIII: 585RGNR588

mut1AB X - - mut2AB - X - mut3AB - - X mut1AB2AB X X - mut1AB3AB X - X mut2AB3AB - X X

X indicates by that column is contained within the AAV1 capsid sequence. A dash (-) indicates that it does not exist. Table 5-2. AAV1 single-site mutant characterization

Vector Total capsid/ml Infectious capsid/ml* Vector genomes/ml* rAAV1 2.70 x 1014 6.90 x 107 1.07 x 1015

mut1AB 1.06 x 1014 4.88 x 106 3.50 x 1014

mut2AB 1.72 x 1014 2.75 x 107 1.53 x 1014

mut3AB 1.22 x 1012 1.38 x 107 1.38 x 1014

mut1AB2AB 2.34 x 1014 Nd Nd mut1AB3AB 1.50 x 1011 Nd Nd mut2AB3AB 2.91 x 1013 Nd Nd

*(nd) not determined.

Page 123: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

123

Figure 5-1. Western blot analysis on single-site mutant cell lysates. Cell lysates were probed with B1 antibody to detect VP1, 2, and 3 ratios and 1F to detect Rep proteins in the cell lysates. Both were used to ensure that all proteins from the rep/cap plasmid were being expressed. Mutant VP levels appear comparable to the rAAV1 control. Viral proteins are indicated with text (VP1, 2, and3) where VP3 is also indicated by a red arrow. Rep 52 and 42 are also indicated by text where rep52 is indicated below VP3 with a green arrow.

Figure 5-2. Purification of single-site antigenic escape mutants A) B1 western blot analysis on iodixanol fractions for each mutant and the rAAV1 control. Bands for VP1, 2, and 3 can be seen in the 60/40% and 40% fractions for the rAAV1 control as well as mut1AB and 2AB, while the VPs are seen in the 40% and 40/25% for mut3AB. The lower bands in the 40% and 40/25% fractions appear to be degredation products, possibly an indication of unstable capsids. B) An example elution (mut1AB) from the ion-exchange column indicating the peak that capsids were seen (Figure 5-3). Inset shows B1 positive western blot analysis for the ion-exchange elutin fractionfor each single-site mutant. VP3 is indicated by a red arrow.

Page 124: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

124

Figure 5-3. Electron micrograph images of AAV1 mutant capsids. Ion-exchange fractions buffer

exchanged out of high salt and were spotted onto carbon-coated electron microscopy grids which were viewed at 47, 000 times magnification. White spherical images indicate negatively stained AAV capsids against a black carbon background. A) mut1AB capsids B) mut2AB capsids C) mut3AB capsids.

Figure 5–4. ADK1a native dot blot of mutant cell lysates. Mutant cell lystates were blotted against ADK1a to determine if capsids could be detected by the ADK1a monoclonal for use by an ADK1a ELISA. The AA5H7.D11 is used as a positive control as it has already shown to recognize the single-site mutants.

Page 125: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

125

Figure 5-5. Analysis of a native dot blot of single and double-site mutants. A) Mutants were checked for recognition with anti-AAV1 mAbs AA4E4.G7, AA5H7.D11, and AA9A8.B12. The rAAV1 was used as a positive control and non-transduced cell lysates as a negative control. B) ImageJ analysis results indicating pixel intensities for the blots. Preliminary double-site mutant analysis revealed a reduced recognition by AA4E4.G7 against all double-site mutants; 1AB2AB, 1AB3AB, and 2AB3AB, indicating that VRIV and V are involved in the AA4E4.G7 epitope. A slightly reduced recognition for 3AB, 1AB3AB, and 2AB3AB by AA5H7.D11 is seen from the pixel intensity calculations indicating VRVIII in the D11 epitope. A similar observation is indicated with the AA9A8.B12 mAb.

Page 126: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

126

Figure 5-6. Infectious green cell assay images of NAb treated single-site mutants. Mutant cell lysates and rAAV1 control were left un-treated or were saturated (1 mAb:1 VP) with the AA4E4.G7 and AA5H7.D11 Nabs and applied to 293 cells. Cells were imaged under UV light for the production of GFP. A) rAAV1 control untreated virus. B) rAAV1:D11 NAb. C) rAAV1:G7 NAb. Both AA5H7.D11 and AA4E4.G7 neutralize rAAV1 infections as indicated by the lack of green cells in (B) and (C). D) Mut1AB untreated virus. E) Mut1AB:D11. F) Mut1AB:G7. G) Mut2AB untreated virus. H) Mut2AB:D11. I) Mut2AB:G7. AA5H7.D11 treated virus shows retained recognition and neutralization of mut1AB and mut2AB, while AA4E4.G7 appears to have lost the ability to recognize and subsequently neutralize both of these capsids. J) Mut3AB untreated virus. K) Mut3AB:D11. L) Mut3AB:G7. rAAV1 capsids with VRVIII mutations (mut3AB) appears to have retained recognition by both the neutralizing Nabs, although quantification (Figure 5-7D) and visualization indicate a slight loss in recognition and neutralization of this particular mutant.

Page 127: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

127

Figure 5-7. Histograms for in vitro Nab treated mutant virus. Group ANOVA statistics and

Tukey’s post hoc results are indicated separately for each mutant as their comparisons are internal. A) rAAV1 control virus with D11(AA5H7.D11) and G7 (AA4E4.G7) where F[2,3]=496.6, P= 0.0002 (***). B) mut1AB with D11 and G7 where F[2,3]=2217, P=0.0001 (***). C) mut2AB with D11 and G7 where F[2,3]=266.6, P=0.0004 (***). Here, a slightly significant value is noted as; *=P<0.05 D) mut3AB with D11 and G7, where F[2,3]=230.2, P=0.0005 (***).

Figure 5-8. Heparin-binding analysis. Single and double-site mutants with VRVIII (mutations including 3AB) from AAV2 were analyzed on a heparin-agarose column. Mutants 3AB, 1AB3AB, and 2AB3AB were loaded onto the column and eluted off with 1 M sodium chloride. Empty baculovirus expressed AAV2 capsids (bAAV2) were loaded as a positive control and non-transfected cell lysates were loaded as a negative control. The flow-through (FT) was collected during loading of the samples. bAAV2 blot samples were checked using mAB C37B, while AAV1 mutants were tested with mAb ADK1a.

Page 128: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

128

CHAPTER 6 SUMMARY AND FUTURE DIRECTIONS

Summary

The AAVs have shown great promise as vectors for gene delivery in therapeutic medicine.

However, since the AAV capsid is a foreign antigen, patient immune responses are activated

causing significant decrease in the therapeutic potential of gene therapy. The future aims of the

field include generating vectors that escape immune surveilence, specifically pre-existing

humoral responses. Biochemical engineering of the capsid surface requires a working knowledge

of not only capsid regions that confer antigenicity, but also receptor binding and tissue tropism.

This study used a structural approach to determine antigenic regions on the capsid surface of

several different AAV serotypes using monoclonal antibodies directed against intact capsid

antigen. Although different serotypes (AAV1, 2, 5, 6, and 8) and monoclonal antibodies were

used, data identified that the monoclonals studied were all recognizing overlapping regions at the

three-fold axes of the capsid surface, regardless of the serotype examined (Figure 4-7). This led

to the conclusion that AAV capsids harbor common antigenic regions at the three-fold axes

including VRs IV, V, and VIII. A majority of the structures included AAV1 as the anti-AAV1

panel was largest at the time the study was initiated and thus this data was used for further

verification of structurally mapped antigenic sites. Based on the structural analyses, two to four

amino acid stretches in the AAV1 capsid VRs IV, V, and VIII were chosen for mutation (Table

4-4). These stretches were subsequently mutated to amino acid residues present in the AAV2 VP

at structurally equivalent positions since this serotype was observed to have no cross-reactivity

with the anti-AAV1 mAbs and is also ~83% similar to AAV1, possibly allowing for tolerable

mutations. This led to the creation of six AAV1 mutant capsids, three single-site mutants termed

mut1AB, mut2AB, and mut3AB, and three double-sites mutants termed mut1AB2AB,

Page 129: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

129

mut1AB3AB, and mut2AB3AB. All mutants appeared to have comparable amounts of capsid

production, although mutants harboring mut3AB (AAV2 VRVIII) had a decrease in capsid

production (Table 5-2). It was proposed that these mutants will most likely not have packaging

defects as the regions mutated have not been previously identified in packaging or assembly

deficiencies of the recombinant virion. Preliminary vector genome evaluation could not be

trusted due to the error seen between the ELISA and qPCR values. Undeniably, PCR is a much

more sensitive technique and will most likely require optimization of the DNA extraction

protocol in order to reduce contaminating cellular DNA.

Infectivity assays with single-site mutant cell lysates revealed comparable levels of

infectivity, although mut1AB had a slightly lower infectivity among them (Table 5-2). However,

the lower value for mut1AB may be due to lower capsid production and/or packaged genome.

Interstingly, double-site mutant capsid production for mut1AB3AB and mut2AB3AB had

markedly less particles/ml (Table 5-2), possibly indicating a challenge in maintaining such a

basic patch (AAV2 VRVIII residues 585RGNR588) within the background of AAV1 VP3.

Interstingly, but not unexpected, mutants which contained the AAV2 heparin-binding motif in

VRVIII, were able to bind heparin (Figure 5-8). It is appealing to propose that receptor binding

regions are heavily supported in the context of the capsid structure through neighboring residues

and that these regions may offer dual functionality in governing receptor-mediated entry as well

as capsid stability. This observation will most likely cause issues in retaining desired

functionality during capsid engineering, but may also reveal more simplistic engineering as

changing one site to generate a desirable phenotype may remove the previous functionality that

was conferring therapeutic challenge.

Page 130: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

130

Following preliminary mutant characterization, in vitro neutralization assays using a GFP

reporter mirrored recognition levels seen on a native dot blot. Here, capsids with mutations in

VRIV and V showed the ability to escape recognition by neutralizing antibody AA4E4.G7

(Figure 5-6A-C). Interstingly, green cell assays involving mut3AB illustrate a slight reduction in

infectivity when incubated with the neutralizing mAb AA4E4.G7 which contradicts the dot blot

analysis (Figure 5-5B). Here, an increase in recognition was observed by AA4E4.G7 with

mut3AB capsids. As the epitope for the non-neutralizing mAb AA9A8.B12 was found to include

VRVIII (mut3AB), it is likely that the increased recognition of the capsid has hindered its ability

to either bind a receptor or traffic properly. This observation supports the notion that the close

proximity of these variable regions confers a mixed functionality through indirect structural

support. Also of note, a native dot blot indicated that all capsids were still recognized by the Nab

AA5H7.D11, but pixel intensity calculations and green cell assays identified a slight drop in

recognition by mutants with VRVIII muations (Figure 5-5B) and an enhanced ability to infect in

the presence of the Nab (Figures 5-6D and 5-7K), respectively. It is likely that engineering of

unique vectors will also include the replacement of adjacent residues to support the desired

mutations/deletions in order to retain viability.

Taken as a whole, all three VRs identified through the structural analysis of complexed

AAV capsid revealed antigenic nature as mutations in these regions altered recognition to some

level. From the data presented for this antibody panel, it appears that VRVIII plays an important

role in the antigenic nature of the AAV1 capsid, including the surrounding regions of VRIV and

VRV. This study has at least developed a foundation for mutational analysis of antigenic regions

on the AAV capsid and offers a starting point for engineering antigenic escape vectors. Ideally,

the whole AAV capsid is antigenic in nature and current data shows that the majority of the

Page 131: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

131

raised regions on the AAV capsid surface, other than the top of the five-fold cylindrical channel,

are succestible to antibody recognition. Thus it is anticipated that new antigenic regions will

arise following the use of escape mutant vectors and that new antibodies against the engineered

vectors will arise. Further analysis of these responses will lead to strategies for better vector

development because it will provide information on capsid regionswhere minimal mutations

create novel antigenic profiles. This information can then inform the engineering of new esacape

vectors which encompase all potential epitopes.

Future Directions

The studies initiated here offer a pathway for further elucidation and definition of the

common antigenic regions on the AAV capsid surface. Including more diverse serotypes, like

AAV4, and more similar serotypes, like AAV9, and expanding the number of antibodies tested,

will most likely offer more support for these common antigenic regions, but may also identify

other areas of the capsid that should be included in engineering antibody escape vectors.

Broadening the antibody panel to include different isotypes, i.e. IgA, may also offer a different

perspective among antibody binding and may also indicate preferential sites among isotypes.

These studies could also identify mechanistical differences among the isotypes and the types of

response mounted, i.e. IgM vs. IgG. Also, the use of serum from patients involved in AAV trials,

and those with natural exposure, may also better identify which antigenic sites are more

common, if such a thing exists. Structural studies including receptor binding regions will also

support the functionality of the variable surface loops and will help to better guide capsid

engineering.

Future studies involving characterization of the mechanisms of neutralization for the

antibodies proposed here may also link specific isotypes or binding regions with certain

mechanisms, allowing for the prediction of antigen-antibody interactions. More immediate goals

Page 132: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

132

will be to swap the identified epitope regions from AAV1 onto other serotypes to confer their

specificity for the antibodies studied here. Also, testing of the intermediate mutants, i.e. 1A,

where only half of the site has been altered may confer more specific residue interactions at the

capsid surface level. Furthermore, a more thorough mutational analysis through this region will

likely better indicate residues involved in antigenicity and may further define those residues

important for the structural integrity of this region.

Page 133: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

133

APPENDIX A HUMAN BOCAVIRUS CAPSID STRUTURE: INSIGHTS INTO THE STRUCTURAL

REPERTOIRE OF THE PARVOVIRIDAE

Introduction

Human Bocavirus (HBoV), a newly discovered member of the family Parvoviridae, was

originally isolated in randomly selected nasopharyngeal aspirates (Allander et al., 2005). Since

this initial discovery, HBoV has also been detected worldwide, predominantly in children under

the age of two with respiratory infections, in serum, urine, and fecal samples (Lindner and

Modrow, 2008). Symptomatic children commonly exhibit acute diseases of the upper and lower

respiratory tracts (Arnold et al., 2006; Kesebir et al., 2006; Monteny et al., 2007; Vicente et al.,

2007), and possibly, gastroenteritis (Kapoor et al., 2009; Vicente et al., 2007), though the link to

gastroenteritis outbreaks has been questioned (Campe et al., 2008). It is still unclear if HBoV is

the sole etiologic agent of respiratory disease as higher rates of co-infections with other

respiratory pathogens such as human rhinovirus and Streptococcus spp. are often observed

(Allander et al., 2007). However, Allander et al. recently reported (2007) that HBoV was found

in 19% of children with acute wheezing, thereby making it the fourth most common virus, after

rhinoviruses, enteroviruses, and Rous Sarcoma Virus, detected in children exhibiting this

symptom. These findings suggest that, at high viral load, HBoV could be an etiologic agent of

respiratory tract disease (Allander et al., 2007). HBoV infection is common in the first few years

of life and clinical research suggests it may follow the primary period for acquisition of Human

Parvovirus B19 (B19), though there is no antigenic cross-reactivity between B19 and HBoV

(Kahn et al., 2008; Kantola et al., 2008). By age five most people have circulating antibodies

against HBoV as is also true for other respiratory viruses such as rous sarcoma virus,

rhinoviruses, and Human Metapneumovirus (Chow and Esper, 2008). HBoV has also been

Page 134: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

134

identified in adults, with ~63% of samples tested being seropositive, showing a positive

correlation with age and a slight positive bias towards women (Cecchini et al., 2009).

The Parvoviridae is a family of small, non-enveloped viruses that package a single-

stranded DNA (ssDNA) genome of ~5000 bases. These viruses are subdivided into two

subfamilies: Parvovirinae and Densovirinae (Table A-1). The Parvovirinae are further

subdivided into five genera, all of whose members infect vertebrates. The Densovirinae (four

genera) only infect invertebrates. Phylogenetic analysis places HBoV in the recently classified

Bocavirus genus (Table A-1). In addition to HBoV, numerous parvoviruses circulate among the

human population. Among these are several dependoviruses, Adeno-associated virus (AAV)

serotypes AAV1-3, AAV5, and AAV9, the Erythrovirus B19, and the newly discovered human

parvovirus genotypes 4 (Parv4) and 5 (Parv5) (Jones et al., 2006; Fryer et al., 2006; Schneider et

al., 2008). Of these, only B19 had been implicated in disease until the discovery of HBoV and

Parv4, which has been isolated from patients who present symptoms of acute HIV infection

(Schneider et al., 2008).

The HBoV genome, like that of all members of the Bocavirus genus, contains three open

reading frames (ORFs). The first ORF (ns), at the 5’ end, encodes for NS1, a nonstructural

protein. The next ORF, unique to the Bocaviruses, encodes for NP1, a second nonstructural

protein. The third ORF (cap), at the 3’ end, encodes the two structural capsid viral proteins

(VPs), VP1 and VP2. The HBoV VPs share 42% and 43% amino acid sequence identity with the

corresponding VPs of bovine parvovirus and canine minute virus, respectively (Allander et al.,

2005). More recently, two additional HBoV-like viruses, HBoV-2 and HBoV-3, were identified

in stool samples from children (Arthur et al., 2009; Kapoor et al., 2009). The genome

organization of these viruses is identical to that of HBoV, with the NS1, NP1, and VP proteins of

Page 135: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

135

HBoV2/3 being ~80/90%, ~70/80%, and ~80/80% identical to the respective proteins in HBoV

(Arthur et al., 2009; Kapoor et al., 2009).

Parvovirus genomes are packaged into a T=1 icosahedral capsid that is assembled from 60

copies of a combination of up to six types of capsid VPs (VP1-VP6), all of which share a

common C-terminus. VP1 is always a minor component, typically comprising about five copies

per capsid, whereas the smallest VP is always the major component. The unique N-terminal

region of VP1 (VP1u) contains a conserved phospholipase A2 (PLA2) motif within the first 131

amino acids that is essential for infection (Qu et al., 2008; Zadori et al., 2001). Interestingly,

AMDV, the only member of the Amdovirus genus, is the only exception in that this motif is

absent, which suggests this virus employs a different mechanism to escape the endosome during

infection (Tijssen et al., 2006).

The X-ray crystal structures of several parvoviruses show that all VPs contain a conserved,

eight-stranded β–barrel motif (βB-I) that forms the core of the capsid (Chapman and Agbandje-

McKenna, 2006). There is also a conserved α-helix (α-A) observed in all parvovirus structures

determined to date. The bulk of the VP consists of elaborate loops between the strands that form

the surface of the capsid. For example, the GH loop between the βG and βH strands is ~230

residues. The composition and topology of these loops encode several important functions,

including tissue tropism, pathogenicity, and the antigenic response directed against each

parvovirus during infection (Agbandje-McKenna and Chapman, 2006).

A number of parvoviruses have been studied by cryo-electron microscopy (cryoEM) and

three-dimensional (3D) image reconstruction in concert with and complementary to X-ray

crystallographic studies (reviewed in Agbandje-McKenna and Chapman, 2006). Report here is

the 3D structure of a recombinant HBoV capsid solved to 7.9-Å resolution using cryoEM. The

Page 136: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

136

capsid of HBoV was compared to representative members of Parvoviridae (Table A-1) with

known atomic structures (AAV2, MVM, B19, GmDNV) or pseudo-atomic models built into

cryoEM reconstructed density (AMDV) to identify similarities and differences. The capsid

topology of the newly emerging HBoV incorporates a combination of structural elements at the

surface seen in other members of the Parvovirinae. However, homology modeling, based on a

comparison of sequences and available parvovirus structures, places the HBoV VP2 topology

closest to B19, the only other structurally characterized parvovirus that is pathogenic to humans.

Materials and Methods

Expression and Purification

The putative HBoV VP2 gene was constructed by de novo synthesis using the published

HBoV ST1 sequence (NCBI Accession No. DQ000495; start codon, nt 3373), and cloned into a

pUC vector (Epoch Biolabs, Inc.). The vector was cut with EcoRV and HindIII to release the

insert, and recloned into a Fastbac1 plasmid (Invitrogen). The sequence and orientation of the

resulting plasmid were confirmed, and the plasmid was used to produce a recombinant

baculovirus using the standard Bac-to-Bac technology (Invitrogen).

Sf9 insect cells (ATCC) were maintained in Grace’s medium (Invitrogen) with 10% fetal

calf serum (FCS) and antibiotics, infected with recombinant baculovirus, harvested 4 or 7 days

post infection into the medium, and collected by centrifugation. Production of baculovirus was

confirmed by a combination of immunofluorescence of infected cells with mouse antibody

against baculovirus (eBioscience), and the typical cytopathic effect of baculovirus-infected cells.

For the immunofluorescence testing, slides were prepared by cytocentrifugation (200 x g for 8

min in a Shandon Cytospin 4) or by spotting a concentrated cell suspension onto each of 8 spots

of a fluorescent antibody slide (Bellco Glass, Inc). The slides were allowed to air dry and were

fixed in 1:1 acetone:methanol at -20°C for 10 min. The monoclonal antibody was diluted 1:50 in

Page 137: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

137

PBS plus 10% FCS, and 25 µl of the solution was incubated with each cell spot (~200 cells) for

1 h at 37 °C. After washing with PBS, the cells were incubated with 1:100 anti-mouse-FITC

conjugated antibody in PBS plus 10% FCS and 1:200 Evans Blue (Sigma) for 1 h at 37°C. After

further washing, the cells were examined by UV microscopy.

Sf9 cells were grown in suspension at 27 °C in Sf-900 II SFM media (Gibco/Invitrogen

Corporation) and infected at a multiplicity of infection of five plaque-forming units per cell.

Resultant HBoV virus-like particles (VLPs) were released from infected cells by three cycles of

freeze–thaws in lysis buffer (50 mM Tris–HCl pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.2%

Triton X-100), with the addition of benzonase (Merck KGaA) after the second cycle. The sample

was clarified by centrifugation at 12,100 x g for 15 min at 4 °C. The cell lysate was pelleted

through a 20% (w/v) sucrose cushion (in 25 mM Tris–HCl pH 8.0, 100 mM NaCl, 0.2% Triton

X-100, 1 mM EDTA) by ultracentrifugation at 149,000 x g for 3 h at 4 °C. The pellet from the

cushion was resuspended in the same buffer overnight at 4 ˚C. The sample was subjected to

multiple low-speed spins at 10,000 x g in order to remove insoluble material. The clarified

sample was purified by separation within a 10-40 % (w/v) sucrose-step gradient spun at 151,000

x g for 3 h at 4 °C. Equilibrium dialysis was performed against 20 mM Tris–HCl pH 7.5, 150

mM NaCl, 2 mM MgCl2 at 4 °C. The final concentration of the sample was 10 mg mL-1

calculated from optical density measurements at 280 nm with an extinction coefficient of 1.7 M-1

cm-1. The purity and integrity of the viral capsids were monitored by SDS-PAGE and negative-

stain electron microscopy, respectively.

Negative Stain Transmission Electron Microscopy

Small (3.5 µL) aliquots of purified VLPs (~5 mg mL-1) were applied to a continuous

carbon support film that had been glow-discharged for 15 s in an Emitech K350 glow discharge

unit and subsequently stained with 1% aqueous uranyl acetate. Micrographs were recorded on a

Page 138: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

138

2k x 2k Gatan CCD camera in an FEI Sphera microscope at a nominal magnification of 40,000 x

at an accelerating voltage of 200 keV.

CryoEM and Image Reconstruction

Small (3.5 µL) aliquots of purified VLPs (~10 mg mL-1) were vitrified via standard rapid

freeze-plunging procedures (Adrian et al., 1984; Dubochet et al., 1988). Samples were applied to

Quantifoil holey grids that had been glow-discharged for ~15 s in an Emitech K350 glow

discharge unit. Grids were then blotted for ~5 s, plunged into liquid ethane, and transferred into a

pre-cooled FEI Polara multi-specimen holder, which maintained the specimen at liquid nitrogen

temperature. Micrographs were recorded on Kodak SO-163 electron-image film at 200 keV in a

FEI Polara microscope under minimal-dose conditions (~9 e/Å2) at a nominal magnification of

39,000 x.

Sixteen micrographs exhibiting minimal astigmatism and specimen drift, recorded at

underfocus settings ranging between 0.95 and 3.25 µm, were digitized at 7 µm intervals

(representing 1.795 Å pixels) on a Zeiss SCAI scanner. The software package RobEM

(http://cryoEM.ucsd.edu/programs.shtm) was used to extract 3,754 individual particle images

(each 1,872 pixels), pre-process the images, and estimate the defocus level of each micrograph

(Baker et al., 1999). The random-model procedure (Yan, Dryden, et al., 2007) was used to

generate an initial reconstruction at ~ 25 Å resolutions from 150 particle images. This was then

used as a starting model to initiate full orientation and origin determinations and refinement of

the entire set of images using AUTO3DEM (Yan, Sinkovits, et al., 2007). Corrections to

compensate for the effects of phase reversals in the contrast-transfer functions of the images

were performed as previously described (Zhang et al., 2003; Bowman et al., 2002), but

amplitude corrections were not applied. A final 3D map at an estimated resolution limit of 7.9 Å,

based on the 0.5 threshold of the Fourier Shell Correlation (FSC0.5) criterion (van Heel and

Page 139: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

139

Schatz, 2005) (data not shown), was reconstructed from all particle images. To verify the new

structure, we computed two, completely independent, ab initio reconstructions from separate sets

of micrographs consisting of 1770 and 1839 particle images. The resultant reconstructions

showed excellent correspondence in all coarse and fine features as evidenced by good agreement

(FSC0.5) at all spatial frequencies out to 1/9.8 Å-1. An inverse temperature factor of 1/100 Å2

was applied to the final 3D reconstruction (Havelka et al., 1995) to enhance fine details and to

aid in the analysis and interpretation of the capsid structure. A Gaussian function was applied to

attenuate the Fourier data smoothly to zero between 7.24 and 6.75 Å. The absolute handedness of

the reconstruction was set to be consistent with surface features seen in parvovirus crystal

structures (see below).

Generation of Density Maps for Representative Members of the Parvoviridae Genera

Density maps were generated from the crystal structure Cα coordinates of AAV2 (PDB

accession No. 1LP3 (Xie et al., 2002)), B19 (PDB accession No. 1S58 (Kaufmann et al., 2004)),

GmDNV (PDB accession No. 1DNV (Simpson et al., 1998)), and MVM (PDB accession No.

1Z1C (Kontou et al., 2005)), and from the Cα pseudo atomic model of AMDV (McKenna et al.,

1999). The coordinates for each VP2 subunit were used to generate a complete icosahedral

capsid using the Oligomer generator in VIPERdb (Carrillo-Tripp et al., 2009). The CCP4

software suite was used to calculate structure factors (7.9 Å resolution cutoff, B-factor =100 Å2)

and electron density maps, using the SFALL and FFT routines, respectively (The CCP4 suite:

programs for protein crystallography, 1994). The calculated maps were rendered with RobEM

(http://cryoEM.ucsd.edu/programs.shtm) and Chimera (Pettersen et al., 2004).

Page 140: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

140

Sequence Analysis and Generation of Unrooted Phylogenic Tree

VP2 sequences of representative members of the six Parvoviridae genera (taken from the

PDB accession files listed above), AMDV (NCBI accession No. ABG20974), and HBoV (NCBI

accession number ABN46897) were uploaded to the Biology Workbench (Subramaniam, 1998).

A multiple sequence alignment was conducted using ClustalW (Thompson et al., 1994) with

default values and with B19 as the reference sequence. An unrooted phylogenetic tree was then

created using the PHYLIP algorithm (Felsenstein, 1989).

Generation and Docking of a Homology Model of HBoV VP2 into Cryo-Reconstructed Density

A homology model was built for the HBoV VP2 (amino acids 37-542) using the Swiss-

PDBviewer-Deep View 3.7 software program (Guex and Peitsch, 1997), using the VP2 sequence

(NCBI accession No. ABN46897) and the B19 VP2 coordinates (PDB accession No. 1S58)

supplied as a template. B19 VP2 was chosen as a template for building the HBoV VP2 pseudo-

atomic model because the comparison of density maps generated for the members of the

different Parvoviridae genera (see above) identified B19 as being the most structurally similar to

HBoV even though the AAV2 VP2 sequence had a slightly higher sequence identity.

The HBoV VP2 homology model was used to generate the coordinates for a complete, 60-

subunit capsid through icosahedral matrix multiplication in VIPERdb (Carrillo-Tripp et al.,

2009). The 60-mer was then visualized within the HBoV cryo-EM density map in COOT

(Emsley and Cowtan, 2004). The core β-barrel (β-strands B-I) and helix αA regions fitted into

density contoured at a 3.3-σ threshold without further adjustment. The loops between β-strands

D and E (DE loop) and between β-strands H and I (HI loop) of a reference VP2 monomer were

manually adjusted to fit into density contoured at a threshold of 1.0 σ guided by their distinct

structural features. The remaining surface loops were adjusted to be within the reconstructed

Page 141: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

141

envelope at a density threshold of 0.5 σ. Further model building included addition of amino acids

at the N-terminus of the VP2 monomer (residues 28-36) to satisfy density (0.5-σ threshold)

inside the capsid under the icosahedral five-fold axes. Following these adjustments, the geometry

of the model was regularized under idealized geometric constraints in COOT. The NCS function

in COOT was used to generate the remaining 59 icosahedral symmetry related VP2 monomers to

verify their fit within the cryo-EM density map. The pseudo-atomic capsid model coordinates

were used to generate a density map for comparison with the HBoV cryo-EM density in

MAPMAN ( Kleywegt and Jones, 1996) using the program’s similarity function

(http://xray.bmc.uu.se/usf/mapman_man.html).

Comparison of Parvovirus VP2 Structures

The structures of VP2 from representative members of six Parvoviridae genera (Table A-

1) were aligned by superposition of the Cα positions of their atomic coordinates (AAV2, B19,

GmDNV, and MVM) or pseudo-atomic coordinates built into cryo-reconstructed densities

(AMDV and HBoV) using the MatcherMaker Tool (Meng et al., 2006) in Chimera.

Results and Discussion

HBoV Capsid Structure

Self-assembled VP2 VLPs expressed and purified from insect cells were checked for

composition and purity by SDS-PAGE (Figure A-1A), and for integrity by negative-stain

electron microscopy (Figure A-1B) prior to sample vitrification and cryoEM (Figure A-1C). A

3D reconstruction of the HBoV capsid, at an estimated resolution limit of 7.9 Å, was computed

from 3,754 individual particle images (Figure A-2) and compared to the capsid structures of

representative members of the Parvovirinae (B19, AAV2, AMDV, MVM) and Densovirinae

(GmDNV) subfamilies (Figure A-3).

Page 142: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

142

The HBoV capsid exhibits three characteristic features common to other vertebrate

parvoviruses. The first of these is a dimple-like depression at each icosahedral two-fold axis

(“dimple”) (Figures A-2A and A-3). The second is a large, trimeric protrusion that surrounds

each three-fold axis or is located at the three-fold axis (“protrusion”) (Figures A-2A and A-3).

The third is a channel at each five-fold axis (“channel”), whose outermost opening is formed by

a small, pentameric structure encircled by a wide, canyon-like region (“canyon”) (Figures A-2A

and A-3). While the dimple is also observed among the invertebrate parvoviruses, these members

lack the three-fold protrusions and canyon around the five-fold channel (e.g. GmDNV in Figure

A-3).

The external diameter of the HBoV capsid ranges from ~215 Å at the lowest points of the

dimple and canyon to ~280 Å at the top of the protrusion. For comparison, the maximum

diameters of AMDV and GmDNV (the largest and smallest capsids of the representative

parvoviruses being compared) are 310 and 270 Å respectively, and the minimum diameter

calculated from the capsid density maps generated from the atomic or pseudo-atomic coordinates

of all the viruses being compared is ~220 Å (Figure A-3). The spherical, relatively smooth

topology of the invertebrate GmDNV clearly distinguishes it from the others, especially those

with more prominent surface features such as AMDV, MVM, and AAV2. HBoV appears to be

morphologically most similar to B19 since both display features intermediate to the above

extremes.

Radial density projections of the six representative Parvoviridae capsids reveal that

structural similarity among them is greatest at low radii and most divergent at the capsid surface

(Figure A-4). Of note, all the vertebrate parvoviruses have strikingly similar structures at radii <

Page 143: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

143

98 Å, suggesting they share a common core. Again, GmDNV stands out among the parvoviruses

as having a structure that is most distinct from the others at all radii.

Pseudo-Atomic Model of HBoV VP2 Built Based on the B19 VP2 Crystal Structure

The HBoV capsid surface topology was observed to be most similar to B19 (Figure A-3),

even though the AAV2 VP2 protein sequence is closer to that of HBoV in percentage identity

(Table A-2). Preliminary docking of the crystal structure of the VP2 subunit for both AAV2 and

B19 into the HBoV cryo-reconstruction showed that the β-barrel and α-A helix in both viruses

fitted into the HBoV density (with a MAPMAN correlation coefficient of 0.6, but for AAV2, the

loop regions that form each protrusion extended outside the HBoV envelope (not shown)

consistent with HBOV’s smoother surface features (Figure A-3). Thus, the B19 crystal structure

coordinates (VP2 residues 19-554) were used to construct the initial homology model of HBoV

VP2 (residues 37-542) for docking into the cryo-EM density map (Figure A-5A and A-5B).

A map of HBoV at sub-nanometer resolution enabled precise, rigid-body fitting of the

conserved secondary structural elements (βA, β-barrel core, and α-A helix) as well as the DE

and HI loops, which required only small manual adjustments followed by bond geometry

refinement (Figure A-5B-D). The remaining inter-strand loops and the C-terminus of the VP2,

including variable regions (VRs as defined in (Govindasamy et al., 2006)), were adjusted to

conform to the HBoV density envelope (Fig. 5A and B). The initial homology model of HBoV

started at residue 37 which corresponds to B19 residue 19, the first N-terminal residue reported

in the crystal structure (Kaufmann et al., 2004). However, a clearly defined region of density at

the base of the channel adjacent to residue 37 of the initial HBoV model remained un-interpreted

following the fitting procedure. This density (denoted by an asterisk in Figure A-2C and A-2D)

was modeled as residues 28-36 contributed by five VP2 monomers (Figure A-5E). These nine

Page 144: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

144

residues were modeled as a small loop that projects into the additional density and extends

towards the channel as seen in B19 and MVM (Agbandje-McKenna et al., 1998; Kaufmann et

al., 2008) (dark blue loop in Figure A-5A and A-5E). This interpretation positions residue 28 of

the HBoV model towards the remaining un-interpreted density that lies close to the entrance of

the channel and appears to form a “plug-like” structure inside the capsid (black arrow in Figure

A-2C and A-5E). The correlation coefficient between the map of the 60-subunit capsid

(generated from the final HBoV VP2 pseudo-atomic model, residues 28-542) and the cryo-EM

density map is 0.70.

A similar plug at the base of the five-fold channel was also observed in the cryo-

reconstruction of B19 VLPs comprised solely of VP2 (Kaufmann et al., 2008). This plug

appeared as negative difference density when the B19 VLP cryo-reconstruction was subtracted

from reconstructions of wild-type full (VP1, VP2, and DNA) and empty (VP1 and VP2) B19

capsid structures (Kaufmann et al., 2008). The location of the first ordered N-terminal residue in

the B19 VP2 VLP crystal structure (starting with residue 18) overlapped this negative density,

suggesting that the remaining, un-ordered residues lie inside the VLP capsid (Kaufmann et al.,

2008). In the B19 report no effort was made to model the missing residues into the negative

difference density. However, positive difference density was observed, in the B19 full and empty

capsid minus VLP density map subtraction, projecting into the channel towards the capsid

surface, emerging between the DE loop β-ribbons onto the canyon at the capsid surface. This

density was modeled as B19 VP2 residues 1-17 (Kaufmann et al., 2008). The differences in

locations of the N-terminal residues in B19 VLPs versus capsids with or without DNA and VP1

are likely dictated by the presence of the DNA and/or VP1. The VP1u motif was not observed in

B19 capsids (Kaufmann t al., 2008) most likely because only a few copies are present per

Page 145: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

145

particle, and their contributions to the density map would be averaged out by the icosahedral

symmetry imposed during structure determination. The B19 channel appears to be closed or

tightly constricted at the capsid surface (Figure A-3) (Kaufmann et al., 2008; Kaufmann et al.,

2004). Thus, disposition of the N-terminal residues of the B19 capsid proteins on the outer

surface is consistent with previous reports that placed VP1u on the B19 surface (Anderson et al.,

1995; Kajigaya et al., 1991; Kawase et al., 1995), which differs from other parvoviruses in which

externalization is proposed to occur via the channel. In HBoV the channel is constricted at its

base but open at the top (Figure A-2C and A-D, and Figure A-3). Thus, it is difficult to assess

whether the HBoV VP2 and/or VP1 N-termini are also already located on the capsid surface. Our

current model of HBoV suggests the N-terminal residues (Figure A-5E) may protrude out

through the channel in virions. Structural studies of capsids that contain VP1 or both VP1 and

DNA are needed to verify this prediction.

HBoV VP2 Shares Low Sequence Identity but High Structural Similarity to Other Parvovirinae VP2s

Sequence alignments and an un-rooted phylogenetic tree were generated to quantitatively

compare the HBoV VP2 sequence with the corresponding VP sequences in five other parvovirus

genera (Table A-1) and to help guide homology modeling based on the HBoV cryo-EM density

map. The pairwise sequence identities ranged between 12 and 23%, with GmDNV, the sole

invertebrate member in the comparison, being the most divergent from HBoV (Table A-2).

Phylogenetic analysis based on a multiple sequence alignment of the six viruses being compared

placed them almost equidistant from each other in an un-rooted tree (data not shown) suggesting

high divergence.

Structural superimposition of the Cα positions of the atomic (B19, AAV2, MVM) and

pseudo-atomic (AMDV) coordinates of the Parvovirinae VP2s with the pseudo-atomic model

Page 146: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

146

generated for HBoV (residues 28-542) showed that, despite the high (~50%) sequence

variability, the eight-stranded β-barrel (BIDG-CHEF) core, βA, and αA helix are conserved in all

the viruses (Figure A-6A). The most VRs in the VP2 structures of the Parvovirinae being

compared to HBoV are located within the loops between the β-strands and most occur at and

around the three-fold axis (Chapman and Agbandje-McKenna, 2006). In the HBoV pseudo-

atomic model, the VRs in the loops are also predicted to be located around this axis (Figure A-

5A). However, at the current resolution of the HBoV density map, the exact positions of the loop

structures are unknown and thus preclude a comparison with those of the other viruses. As with

the Parvovirinae members, the core β-strands are structurally very similar between HBoV and

GmDNV, though the exact position of αA differs slightly (Figure A-6A).

HBoV Capsid Surface Topology is Unique, but Shares Features Common to Other Vertebrate Parvoviruses

Based on the overall surface morphologies of their capsids, parvoviruses have been

structurally assigned to three groups (Padron et al., 2005). Groups I and III comprise members of

the Parvovirinae subfamily, all of which share three features as described earlier: dimples at

two-fold axes, protrusions at or surrounding three-fold axes, and channels surrounded by

canyons at the five-fold axes (Figure A-3). The primary difference between these two groups

occurs in the vicinity of each three-fold axis. Group I capsids have a single, relatively flat,

pinwheel-shaped protrusion, whereas group III capsids have three distinct protrusions. Current

structural data place members of the Parvovirus genus in group I and members of the

Dependovirus, Amdovirus, and Erythrovirus genera in group III (Table A-1, (Padron et al.,

2005)). A third topology (group II), seen in GmDNV and other members of the Densovirinae

subfamily (Bruemmer et al., 2005; Simpson et al., 1998), is characterized by capsids which are

relatively spherical and featureless compared to the vertebrate parvoviruses (Table 1, (Padron et

Page 147: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

147

al., 2005)). This morphology, exemplified by GmDNV, results from the reduced length of the

surface loops compared to members of the other parvovirus genera (Simpson et al., 1998).

HBoV possesses features common to groups I and III and also others that are unique. One

distinguishing feature in HBoV arises from a difference in surface topology at the two-fold axis,

where the HBoV dimple is narrower and shallower than that in the other parvoviruses (Figure A-

3). However, like B19, the HBoV dimple is flanked by wider “walls” between the two- and five-

fold depressions compared to the other viruses which may be due a conformational loop

difference in modeled VR-IX located close to the two-fold axis (Figure A-5A). Residues that

form the walls of the parvovirus dimple have been implicated in receptor attachment for MVM

(Lόpez-Bueno et al., 2006) and as determinants of tissue tropism and pathogenicity for other

members of the parvovirus genus (reviewed in (Kontou et al., 2005)). There is no evidence

however that this region plays an analogous role in B19 or HBoV.

Other characteristic parvovirus features at the two-fold region are conserved in HBoV.

Helix αA, observed in all parvovirus structures determined to date (reviewed in (Chapman and

Agbandje-McKenna, 2006)), is also present in HBoV (Figure A-5C and A-6A). This helix forms

stabilizing interactions at the icosahedral two-fold interface (Govindasamy et al., 2006). Even

though there are fewer interactions at the two-fold interface compared to those at the three-fold

interface, they are proposed to play a crucial role in parvovirus capsid assembly (Govindasamy

et al., 2006; Wu et al., 2000). Conservation of this structural feature in the HBoV VP2 thus

suggests that it plays a similar, central role in capsid assembly.

HBoV topology at the three-fold axis is intermediate between that of groups I and III.

AAV2 and AMDV, group III viruses, have large protrusions that arise from the βEF and βGH

loops. The protrusions in HBoV are less pronounced than those of the group III parvoviruses,

Page 148: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

148

which may be due to shorter sequences and/or conformational differences in its βEF and βGH

loops. This results in a less pronounced depression in this region (Figure A-3). MVM and

AMDV, group I viruses, also lack depressions at their three-fold axes (Figure A-3). Residues

near the three-fold axes in AAV2 and B19 have been implicated as receptor attachment sites

(Chipman et al., 1996; Kern et al., 2003; Levy et al., 2009; O'Donnell et al., 2009, Opie et al.,

2003). The protrusions surrounding this axis are responsible for the antigenic reactivity of

members of the Parvovirinae subfamily (reviewed in (Agbandje-McKenna and Chapman,

2006)). Given this antigenic role of the group I and III surface protrusions, the lack of similar

structures in group II viruses is consistent with the fact that invertebrate viruses are not subject to

adaptive pressures of the host immune response (Simpson et al., 1998).

As mentioned above, the capsid structures of members of the Parvovirinae have a

conserved channel at the icosahedral five-fold axis, surrounded by a canyon (Figure A-3). The

channel is a β-cylinder formed by five symmetry-related β-ribbons inserted between β-strands D

and E with a loop at the top (referred to as the DE loop). In HBoV the DE loops form finger-like

protrusions that surround the channel (Figures A-5A, A-5D, and A-6B) and project away from

the five-fold axis in a manner similar to the conformation observed in AAV2, resulting in an

open channel (Figure A-3). In contrast, for the other Parvovirinae structures, the DE loops

cluster nearer the five-fold axis and constrict the channel (Figure A-3 and A-6B). This channel in

parvoviruses is postulated to serve several functions such as the site of (a) VP2 externalization in

members of the parvovirus genus (e.g. MVM, in which VP3 is generated by a maturation

cleavage event), (b) VP1u externalization (except in B19 and GmDNV) for its PLA2 function,

and (c) genomic DNA packaging. However, because the diameter of the channel varies between

~5 and 25 Å at its narrowest and widest points, respectively, this is likely too small to

Page 149: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

149

accommodate extrusion of the ~130aa in the PLA2 domain. It has thus been proposed that

flexibility and structural re-arrangement of the DE loop (and possibly the HI loop, see below) are

required to facilitate these functions. Conservation of this channel in HBoV suggests that it

performs analogous functions. Notably, HBoV appears to have the largest surface opening of the

parvovirus structures determined to date (Figure A-3).

The canyon depression in HBoV is less pronounced compared to that seen in all other

parvovirus structures except for GmDNV, which lacks that feature (Figure A-3). In HBoV the

HI loop lies in the canyon and is structurally most similar to the HI loop of AAV2, and although

shorter in HBoV, generates a similar surface topology for the two viruses (Figure A-3 (red loop

features on the canyon floor), Figure A-5D, and A-6B). The HI loop conformation differs

significantly in B19 because it projects radially from the capsid surface and lies very close to the

DE loop to form a continuous, raised surface in the canyon (Figure A-3). This B19 HI loop

conformation is similar to that proposed to occur in AAV2 following receptor attachment (Levy

et al., 2009). The HI loops in MVM and AMDV, which also lie on the canyon floor, are

structurally similar to each other but differ from HBoV (Figure A-6B). In GmDNV the HI loop

lies very close to the DE loop, and together they “fill in” the canyon floor (Fig. 3 and 6B). The

HI loop which has been proposed to undergo structural re-arrangements in AAV2 to facilitate

VP1 externalization upon receptor binding has also been reported to play a role in capsid

assembly (DiPrimio et al., 2008; Levy et al., 2009).

Summary

The structure of the HBoV capsid presented here represents the first for a member of the

newly established Bocavirus genus of the Parvoviridae. The sub-nanometer resolution obtained

in the cryo-reconstruction clearly reveals that the newly identified HBoV assembles a capsid that

retains the highly conserved structural core adopted by all other parvoviruses, and does so

Page 150: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

150

despite low sequence similarity between its major capsid protein and those of the other viruses

which have existed for many years. Conservation of core structural features in the major coat

protein, including the eight-stranded β-barrel, the αA helix, the DE loop, and the HI loop, is

consistent with their contributing essential functions in capsid assembly and stability.

Conservation of the β-cylinder channel formed by the DE loop strongly suggests that this

channel is essential for productive HBoV infection as it likely serves as a portal for VP1u

externalization and DNA packaging as reported for other parvoviruses.

Overall, HBoV shares most of its capsid surface features with the two other human

parvoviruses (B19 and AAV2) in this comparative study, and these result in a unique capsid

morphology whose functional domains are yet to be verified. The resemblance of B19 and

AAV2 capsids at the three-fold axes suggests that this region confers similar, receptor

attachment capability. On the other hand, the structural difference of the HBoV five-fold axis

from B19 (reported to have its VP1/VP2 localized on the capsid surface) and its similarity to

AAV2 (proposed to externalize its VP1u for its PLA2 function via the channel) suggests that the

HBoV capsid is required to undergo similar conformational transitions as AAV2 during cellular

trafficking. Variable HBoV VP2 surface loops, as built in the pseudo-atomic model based on

known parvovirus structures (Chapman and Agbandje-McKenna, 2006) and constrained by the

reconstructed density envelope, likely reflect adaptations related to host cell recognition and

evasion of the adaptive immune response. Significantly, the structure of HBoV provides an

additional framework for the molecular annotation of parvovirus VP structures, which have

evolved to facilitate infectivity through the utilization of structural elements and capsid surface

features that fulfill myriad functions.

Page 151: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

151

Table A-1. Selected properties of representative members of Parvoviridae Genus Members Number of VPs Group Major VP Subfamily Parvovirinae – vertebrate hosts Parvovirus

MVM*, CPV*, FPV*

3

I

VP2

Erythrovirus

B19*†, SPV 2 III VP2

Dependovirus AAV2*, AAV4*, GPV

3 III VP3

Amdovirus

AMDV† 2 III VP2

Bocavirus HBoV†, BPV, CnMV

2 III VP2

Subfamily Densovirinae - invertebrate hosts Densovirus

GmDNV*, JcDNV†

4

II

VP4

Iteravirus BmDNV† 4-6 II VP1-4

Brevidensovirus AaeDNV, AalDNV

2 or 3 n/a VP1 or 2/3

Pefudensovirus PfDNV 5 n/a VP1 *Denotes structures determined by X-ray crystallography, †Denotes structures determined by cryo-EM, (n/a) denotes structural group not assigned

Table A-2. Sequence alignment statistics for representative members with structural data Sequence % Identity* % Strong

similarity* % Weak similarity*

% Different*

AAV2 23 17 14 46 B19 19 17 14 50 MVM 16 17 16 51 AMDV 14 17 13 56 GmDNV 12 13 11 64

*Percentage identity, similarity, and difference are as defined in Thompson et al., 1994.

Page 152: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

152

Figure A-1. Characterization of recombinant HBoV VLPs. A) 10% SDS-PAGE of HBoV VLPs

(right lane) and molecular mass marker (left lane), showing that VLPs consist solely of VP2 (~61 kDa). B) Micrograph of negatively stained VLPs shows that most capsids exhibit a circular profile and fill with stain, consistent with them being empty, spherical shells. C) Micrograph of unstained, vitrified VLP sample highlights that these HBoV particles are intact, empty, and spherical.

Page 153: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

153

Figure A-2. HBoV cryo-reconstruction. A) Stereo, shaded-surface representation of the entire capsid viewed along a two-fold direction. The black equilateral triangle bounded by two three-fold (3) axes (separated by a two-fold (2) axis) and a five-fold (5) axis identifies one asymmetric unit of the icosahedral capsid. B) Stereo, half particle image of (A) sectioned at the center and generated as a perspective view. C) Central cross-section of the reconstructed map, with highest and lowest density features depicted in black and white, respectively. Arrows highlight the two- (2), three- (3), and five-fold (5) axes that lie in the plane of the cross-section in one quadrant. Density “plugging” the five-fold channel at low radii is indicated by the short, black arrow for one channel. D) Enlarged view of upper left quadrant from (C) with circles drawn at radii corresponding to the radial density projections shown in Figure A-4.

Page 154: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

154

Figure A-3. Comparison of six representative parvovirus capsid structures. Stereo, radially color-cued, surface representation of HBoV cryo-reconstruction shown with corresponding two-fold views of B19, AAV2, AMDV, MVM, and GmDNV highlight similarities and differences in the outer surfaces of the capsids. Density maps of the latter five virus capsids were computed from atomic (B19, AAV2, MVM, and GmDNV) or pseudo-atomic (AMDV) coordinates and all rendered at the same resolution (7.9 Å)

Page 155: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

155

and magnification. Vertical bar depicts color cueing as a function of particle radius in Å.

Figure A-4. Radial density projections of six representative parvoviruses. The virus labels are as defined in Table 1. Density distributions at different radii (see Figure A-2D) illustrate similarities (inside) and differences (towards the outside) among the virus capsids. Contrast same as that depicted in Figure A-2C.

Page 156: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

156

Figure A-5. Pseudo-atomic model of HBoV VP2. A) A Cα-trace of the VP2 pseudo-atomic model. Labels highlight the β-strands regions (BIDG-CHEF) that form the core β-barrel, the α- helix A (α-A), the DE and HI loops, and the nine variable surface regions (VR-1 to VR-IX). The approximate two-, three- and five-fold axis are shown by the numbers 2, 3, and 5, respectively. The dark blue, N-terminal portion of the model represents nine residues that were not included in the initial HBoV homology model, but that can be fit into the HBoV cryo-reconstruction. B) An equatorial slab of pseudo-atomic HBoV VP2 monomers docked into the reconstructed density map. Several icosahedral symmetry axes are labeled as in Figure A-2B. Regions outlined in red are enlarged in subsequent panels to further emphasize the fit of the model. C) Stereo view of a small portion of the VP2 model (in red) showing the structurally conserved, β–barrel core (β-strands A, B, I, D and G) and α-A helix modeled into the reconstructed density (grey mesh, contoured at a threshold of 3.3σ). D) Same as (C) for a view of the region of the capsid near the five-fold axis, showing the fit of five, symmetry-related, DE loop β-ribbon models that form the cylindrical channel and the fit of five HI loops that form the petal-like structures on the canyon floor. The five VP2 monomers are distinguished by color. E, left) Same as (D) but for a view from inside the capsid that highlights the channel “plug”. Residues 28-36 in the HBoV VP2 model are colored blue. E, right) Same as (E, left) but viewed from the side to show the model of one monomer and the DE-loops of four symmetry-related monomers at the pore of the five-fold channel. N-terminal residues 28-36 (in blue) were modeled as extending up into the five-fold channel. Color scheme same as in (D).

Page 157: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

157

Figure A-5. Continued

Page 158: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

158

Figure A-6. Superposition of VP2 structures from six representative members of the Parvoviridae. The orientation of the VP monomer is relative to the exterior and interior of the capsid is indicated by labels. A) Enlarged view of the conserved β-barrel motif (BIDG and CHEF sheets), strand βA, and helix α-A. Structural elements are labeled as in Figure A-5A. B) Enlarged view of the HI loops

Page 159: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

159

APPENDIX B MONOCLONAL ANTIBODY AGAINST THE ADENO-ASSOCIATED VIRUS TYPE 8 CAPSID AND THE IDENTIFICATION OF CAPSID DOMAINS INVOLVED IN THE

NEUTRALIZATION OF THE AAV8 INFECTION

Introduction

As previously mentioned, the AAVs have become a useful tool in gene transfer. Several

serotypes have been tested for use as vectors in gene therapy, with AAV2 being the best

characterized and most tested (Rabinowitz et al., 1999; Nicklin et al., 2001; Wu et al., 2000;

Bartlett et al., 1998; Fischer et al., 2003; Simonelli et al., 2009; Wobus et al., 2000; Opie et al.,

2003; Moskalenko et al., 2000; Flotte, 2005). AAV vectors based on other serotypes show

transduction efficiency in various tissues: AAV1 in muscle, pancreatic islets, heart, vascular

endothelium, brain and central nervous system (CNS), and liver; AAV3 in Cochlear inner hair

cells; AAV4 in brain; AAV5 in brain and CNS, lung, eye, arthritic joints, and liver; AAV6 in

muscle, heart, and airway epithelium; AAV7 in muscle; and AAV8 in muscle, pancreas, heart,

and liver (Jiang et al., 2006). Most vectors are broad in their tropism, but many have specific

points of localized infectivity that make them more efficient for one tissue over the other.

Interstingly, i.v. and intraperitoneal (i.p.) injection of AAV8-based vectors have been found to

cross the blood vessel barrier and deliver transgene efficiently to striated muscles where as

AAVs 1 and 6 have been shown to better deliver localized responses (Wang et al., 2003). Also,

in studies to identify the best vector for transgene expression in specific tissues the AAV8

serotype repeatedly out-transduces other serotype-based vectors in the mouse liver (Gao et al.,

2002; Jiang et al., 2006; Moscioni et al., 2006; Nakai et al., 2005; Davidoff et al., 2005; Sarker et

al., 2004; Conlon et al., 2005; Sun et al., 2005). Concordantly, the first study to successfully

correct hemophilia A in mice used an AAV8 vector and demonstrated the same phenomenon

independent of vector cassette or injection route (Sarkar et al., 2004). Studies comparing AAV2,

Page 160: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

160

6, and 8 vectors in the liver have identified that AAV8 can efficiently transduce liver cells due to

rapid uncoating of the viral capsid, thus allowing for a constant presence of ssDNA available for

annealing and dsDNA production (Thomas et al., 2004). This phenomenon allows for more than

10% transduction of mouse liver cells compared to AAV2 vectors (Thomas et al., 2004).

Although AAV2/8 vectors have been successful in the correction or reduction of pathology in

many murine disease models, there are many differences noted in the efficiency of treatment and

gene expression. One study successfully transduced adult mouse livers resulting in a sustained

correction of a metabolic disease, but noted that juvenile livers, still growing, had markedly

decreased gene expression over time (Cunningham et al., 2008, 2009). A follow up study

identified that re-administration was necessary, but difficult due to an anti-capsid humoral

condition and further concludes that efficient vector design and re-administration techniques will

be necessary (Cunningham et al., 2009). Although, the fold increase of transformed-hepatic cells

with AAV8-based vectors seen in the mouse is yet to be repeated in larger animal models

(Murphy et al., 2009), recent studies in hemophilia dog models have demonstrated AAV-

mediated continuous expression of canine factor VIIa (cFVIIa) (Margaritis et al., 2009). These

results provide the first evidence for efficacy and safety of the gene-based FVIII/FIX bypassing

agent FVIIa for the treatment of hemophilia in a large animal model using rAAV8 vectors

(Margaritis et al., 2009). However, antibody responses to the capsid have been shown to

efficiently block gene transfer in large animal models (Wang et al., 2010; Davidoff et al., 2005).

The same has held true in human patients (Manno et al., 2006; Li et al., 2009). Indeed, lower

transgene expression was primarily linked to AAV-binding antibodies rather than to AAV

capsid–specific CD8+ T cells (Manno et al., 2006).

Page 161: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

161

Few animal models exist where pre-existing antibody responses can be studied that will

relate to the human conditions. Many studies use passive immunity models where animals are

injected with human-derived immunoglobulin, often termed intravenous immunoglobulin (IVIG)

(Scallan et al., 2006; Wang et al., 2010). Pre-existing immune response studies in mice using

AAV2 and 8 vectors indeed support that even non-neutralizing antibodies can have a negative

effect on gene transfer stability, as well as an interesting find that mice immunized with

AAV8cap vector and subsequently challenged with an AAV2-gene vector induced cross-reactive

antibodies to AAV2, but the converse was not found in the opposite studies (Li et al., 2009).

However, when compared to rh32.33 vectors, AAV8 generated a lower immune response to the

viral vector allowing for longer sustainment of the transduced gene (Mays and Wilson, 2009).

The use of alternate serotypes to bypass pre-existing immune responses (Halberd et al., 2006; De

et al., 2006; Wang et al., 2005) has drawn much attention as the list of naturally and engineered

capsids continue to expand (Gao et al., 2004; Koerber et al., 2008; Excoffon., 2009; Choi et al.,

2005). These new vectors utilize variability among the surface loops as a general means of

antibody escape and retargeting. It is a well-known fact that the human population already has

high neutralizing antibody titers to the more common AAV, serotype 2 (Blacklow et al., 1968,

1971; Chirmule et al., 1999; Erles et al., 1999; Calcedo et al., 2009; Boutin et al., 2010). A more

comprehensive study looking at serum samples from around the world, implicated that titers to

the non-human primate isolates AAV7 and 8 were relatively low compared to AAV2 and 1,

while rh32.33, an engineered vector from primate tissue, was rarely detected (Calcedo et al.,

2009). Of note, a more recent study identified that AAV8 seroprevelence was the lowest, less

than 40%, when compared to AAV1,2,5,6, and 9 (Boutin et al., 2010). These vectors may

therefore be useful in clinical trials with patients who have already been exposed to other

Page 162: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

162

serotypes (Calcedo et al., 2009; Boutin et al., 2010). However, given that AAV-reactive

antibodies are present in a large portion of the population (Moskalenko et al., 2000), combined

with the fact that a high degree of homology exists among the serotypes, it may become

increasing difficult to overcome pre-existing immunity (Lin and Ertl, 2009). Indeed, it has been

noted that patients who have titerable antibodies to AAV1 will generally have antibodies to the

rest of the serotypes currently being studied (Dr. Luk Vandenberghe, personal communication).

This underlines the increasing importance of understanding the antigenic nature of the AAV

vector in context with the patient response.

A better understanding of these regions will aid in engineering or choosing vectors that

have specific properties for use with a certain disease or special circumstances that arise among

the different patients. This study specifically focuses on identifying regions on the AAV8 capsid

surface that are recognized by a monoclonal antibody, ADK8. In order to identify regions

involved in the antigen-antibody interaction, 3D reconstructions were generated from cryoEM

data of the AAV8:ADK8 complex (see chapter 2). Possible sites of antibody recognition were

identified on the capsid surface and mutational analysis was then performed. These vectors were

tested in vivo and in vitro for their inherent ability to evade pre-existing responses while

retaining the transduction efficiency previously noted for the first generation AAV8 vector.

Materials and Methods

Animals, Cell Lines and Cell Culture

Six-to-nine weeks old, female NMRI mice were used for all experiments. Mice were

purchased from Charles River Wiga (Sulzfeld, Germany). Mice were kept according to the

guidelines of the German Cancer Research Center. 293T (Pear et al., 1993), HeLa cells

(laboratory stock) and HepG2 (Knowles et al., 1980) cells were maintained in Dulbecco’s

Modified Eagles Medium (DMEM) supplemented with 10% heat inactivated fetal-calf serum

Page 163: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

163

and 100 U/ml penicillin and 100 µg streptomycin at 37°C in 5% CO2. For transfection, a

confluency of 70% of 293T cells is needed. For binding assays, a 90 % confluency of Hep G2

cells should be ensured. X63/Ag8 lymphoma cells (kindly provided by P.Krammer, DKFZ) were

cultured in RPMI 1640 supplemented with 10% heat-inactivated fetal-calf serum, 100 U/ml

penicillin, 100 µg/ml streptomycin, 2 mM L-Glutamin, 20 mM Hepes pH 7.2, at 37°C, 5% CO2.

Plasmids and Site-Directed Mutagenesis

Vector plasmid pTRUF2CMV-Luc is a recombinant AAV2 plasmid expressing a

luciferase reporter gene from a cytomegalovirus (CMV) promoter and is flanked by inverted

terminal repeats (Zolotukhin et al., 1996). Plasmid pDGΔVP, expresses all essential Ad helper

proteins. It is based on plasmid pDG but prevents expression of VPs (Grimm et al., 1998). The

p5E18-VD2/8 helper construct (Gao et al., 2002) supports the synthesis of AAV2 Rep proteins

and AAV8 VPs, pBSΔTR18 provides AAV2 Rep and Vp proteins (Weger et al., 1997). Both

plasmids served as templates for site-directed mutagenesis reactions. Mutagenesis was

performed using the QuickChange site-directed mutagenesis kit (Stratagene, Amsterdam, The

Netherlands) according to the manufacturer’s manual. For each mutant two complementary PCR

primers were designed containing the substitution, flanked by 10 to 15 homologous base pairs on

each site of the mutation. Three sets of primer were produced and inserted into the cap gene of

P5E18-VD2/8: forward primer 5´GGAGGCACGACAACTCAGTC GACCCTCTGGGC 3´,

reverse primer 5´GCCCAGAGTCGACTGAGTTGTCGTGCCTCC 3´

(455GTANTQ→GTTTQS460); forward primer 5´GCTATTGTTGTTGTCCGCGCTTGT

CTTTGAGACGCG 3´, reverse primer 5´CGCGTCTCAAAGACAAGCGCGGACAACAAC

AA TAGC 3´ (493TTTGQ→KTSAD497); forward primer 5´ GCAGATAACTTGCAGC

GGGGAAACAGGGCTCCTC 3´, reverse primer 5´GAGGAGCCCTGTTTCCCCGCTGCA

AGTTATCTGC 3´ (586LQQQNT→LQRGNR591). Two sets of primer were produced and

Page 164: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

164

inserted into the cap gene of pBSΔTR18: forward primer 5́CCAAGTGG

AACCACCAATACGCAAACTCTTGGGTTTTCTCAGGC 3´, reverse primer 5`GCCTG

AGAAAACCCAAGAGTTTGCGTATTGGTGGTTCCACTTGG 3´ (457QSRLQ→TQ TLG

461); forward primer 5´CCACCTCCAGCAACAAAACACAGCGGCAGCTACCGC 3´, reverse

primer 5´GCGGTAGCTGCCGCTGTGTTTTGTTGCTGGAGGTTGG3´ (585RGNR

Q→QQNTA589). The complete fragments were sequenced to exclude additional mutations.

Plasmids and Peptide Insertions

To insert a peptide into the AAV8 VPs, a SFII binding site had to be constructed (Muller,

Kaul et al. 2003) and a fragment of 743 bp was synthesized (GeneArt, Regensburg, Germany). It

was cloned with XcmI 1277 - EcoRV 2020 into the plasmid P5E18-VD2/8. Four peptides were

chosen for insertion into the SFII binding site: PSVPRPP, VNSTRLP, GQHPRPG (Ying Y. et

al., paper under revision) and ASSLNIA (Yang et al., 2009). The fragments containing the

insertion sites were sequenced for verification.

Transfection of 293T Cells and Vector Particle Purification

A triple transfection was carried out by calcium phosphate precipitation. For each mutant,

twenty plates with 5E+6 cells each, were seeded out and transfected 24 h later. Per plate, 50 – 60

µg DNA was resuspended in 1.125ml sterile Braun H20, mixed with 125 µl CaCl2 (Sigma, St.

Louis, USA) and added slowly to 1.25 ml of 2 x HBSS (280 mM NaCl, 50 mM Hepes, 1.5mM

Na2HPO4, 10 mM KCL, 12 mM glucose, pH 7.05), shaking constantly. After 1 min of

incubation, the mix was added to 7.5 ml medium and append to the cells. After 48 h at 37°C, 5%

CO2, cells were harvested, washed with PBS, centrifuged at 200x g for 15min and stored at -

80°C until further purification steps took place.

Page 165: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

165

Purification and Titration of Mutant AAV Stocks

15 ml of a lysis buffer (150 mM NaCl, 50 mM Tris-HCl, pH8.5) was added to the cell

pellets, followed by 5 rounds of freeze-thawing (N2 and 37°C), 30 min of Benzonase (50 U per

ml) treatment at 37°C and 15 min at 3000 x g . Via an iodixanol-step-gradient ( 40 ml Beckman

Quick Seal Tube), AAV particles were purified from the crude cell lysate (Zolotukhin et al.,

1999). Sealed gradients were centrifuged at 50,000 x g at 4°C for 2 h. Viral particles were

aspirated from the 40 % iodixanol phase and deep-frozen at 20°C. Quantitation of AAV genomic

particles was achieved by quantitative real-time PCR adapted from previously described methods

(Veldwijk et al., 2002). After the alkaline lysis of the AAV particles, genomes were subjected to

Taqman Universal Master mix including primer (for-5´TGCCCAGTACATGAC CTTATGG 3´,

rev- 5´GAAATCCCCGTGAGTCAAACC 3´), probe (6-fam-AGTCATCGC TATTACCATGG-

MGB) and analysed under standard QR-PCR conditions (Applied Biosystems Inc, Foster City,

USA).

Analysis of Viral Protein Expression

Western blot analysis was performed with equal amounts of genome containing particles

according to standard methods (Harlow and Lane, 1988). Monoclonal antibodies were applied as

previously described (Wistuba et al., 1995).

Production and Purification of Monoclonal Antibody ADK8

After fusion (Wobus et al., 2000) and screening of hybridoma supernatants (Kuck et al.,

2007), a monoclonal antibody was identified and its hybridoma cells were cultivated with

increasing medium amounts (maximum 1.5 ml) in expanded surface roller bottles (Sigma-

Aldrich, St. Louis, USA) for 12 days. Cells were centrifuged at 4000 x g, 10min, and supernatant

was collected. For preservation, 0.01% thimerosal was added to the supernatant. A monoclonal

antibody was purified from the hybridoma supernatant by affinity chromatography using a

Page 166: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

166

protein G sepharose column (GE Healthcare Europe GmbH, Munich, Germany). Hybridoma

culture was filtered through a 0.45µm filter and applied onto the column at room temperature

overnight. Wash column with PBS and elute the antibody with 10 x 1ml Na azide (0.1 M)/NaCl

(0.15 M), pH 4.4. The eluted antibody was neutralized with 5% Tris pH 9.5. The total antibody

amount was determined (Nanodrop ND-1000, Peqlab Biotechnology, Erlangen, Germany) at an

OD of 280nm.

Determination of Antibody Isotypes

Subclasses of antibodies were determined by using the Amersham mouse antibody

isotyping kit (RPN29, Braunschweig, Germany) according to manufactuere’s guidelines.

AAV Capture ELISA

ELISA was carried out using ADK8 (50ng per well) coated flexible microtiter plates

(Becton Dickinson, Heidelberg, Germany). Particles of rAAV8 for the standard were determined

by particle calculation of negative staining from 10 randomly taken pictures on an electron

microscope. 1E+10 genome containing particles were applied and serial diluted. HRP-labelled

(ApD Serotech, Oxford, UK) ADK8 (1µg/ml) antibodies were applied and analysed by an

ELISA reader (Ascent FL, Thermo Labsystems, Egelsbach, Germany).

In Vitro and In Vivo Neutralization

HepG2 cells were seeded out on a 96-well plate (NuncTM, Rochester, USA) 24 h in

advance. Monoclonal antibodies were preincubated with vector particles at 37°C for 30 min.

Antibody particle mix was added to FCS-free medium of HepG2 cells. After 4 h, medium was

aspirated, cells were washed with PBS and DMEM+FCS was added. After 72 h medium was

aspirated, cells were washed with PBS once again and 1x RLB (Reporter lysis buffer, Promega

GmbH, Mannheim, Germany) was applied. Cell extracts were stored at -80°C until expression

analysis was performed.

Page 167: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

167

Mice were injected intraperitoneal with 50 or 500 µg monoclonal antibodies 4 h prior to

intravenous injection of the viral particles. 1E+11 viral genome containing particles were

injected into each mouse. 14 days after injection animals were analysed by the IVIS Imaging

software ( Xenogen, Alameda, USA) for 5 min, 10 min after D-Luciferin (30 mg/ml) (Synchem

OHG, Altenburg, Germany) injection. Mice were sacrificed, liver and heart were extracted and a

luciferase expression analysis was performed. The amount of protein was determined with a

NanoOrange protein quantitation kit (Invitrogen, Karlsruhe, Germany).

Binding Analysis by DNA Dot Blot

For the DNA binding analysis, HeLa cells were seeded out on 6-well plates 24 h before

infection. Viral particles were pre-incubated with 50 µg or 250 µg of antibody at 37°C for 30

min. After incubation, virus antibody mix was shifted to 4°C before being applied onto the cells

for 30 min. Cells were washed with medium and either harvested directly or transferred to 37°C

for 2 h and harvested. Without a temperature shift, cells were centrifuged at 200 x g for 10 min

and medium aspirated. The latter were centrifuged, medium aspirated, 100 µl of pre-warmed

trypsin (0.05%) added for 5 min, centrifuged again and medium carefully removed. Pellets were

stored at -20°C. After Nuclease (1mg/ml) and Proteinase K (10 mg/ml) treatment (Roche,

Pensberg, Germany), phenol-chloroform extraction was performed, DNA was spotted onto a

nylon membrane (Gene ScreenTM, DuPont , Boston, USA)in a dot blot chamber, 10 min

denaturated (1.5 M NaCl, 0.5 M NaOH), 10 min neutralized (0.5 M Tris-HCL pH 7.0, 0.3 M

Tris-Sodium-Citrate, 3M NaCl) and by UV-cross-linking at 1200 Joule. A radioactive probe,

directed against CMV was produced with standard protocol of the DNA Labeling kit (Roche,

Pensberg, Germany) and 5µl [α-32P]dCTP (50 μCi). On the nylon membrane immobilized

template DNA was pre-hybridized in 15 ml hybridization buffer (125 mM Na2HPO4, 250 mM

NaCL, 1 mMEDTA, 45% formamide, 7% SDS), probe was added and DNA was labelled in a

Page 168: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

168

hybridization oven with rotation at 42°C overnight. Membrane was washed 6 x 5 min with wash

buffer I (2 x SSC, 0.1% SDS) at 42°C and 3 x 20 min with wash buffer II (0.1 x SSC, 0.1% SDS)

at 68 °C. Membrane was exposed to an X-ray film (Kodak BioMax MS, Sigma, St. Louise,

USA).

Immune Fluorescence

HeLa cells were seeded out on cover slides in 24-well plates. Viral particles were pre-

incubated with 0.1 mg antibody ADK8 or IVA7 (own stock) at 37°C for 30 min then on ice for

10 min. Virus-antibody mix was added to pre-cooled cell slides and kept at 4°C for 30 min. Cells

were washed with PBS and directly fixed with ice-cold methanol for 10 min. After fixation,

ADK8 hybridoma culture was applied overnight, then washed 3 x 10 min with PBS, kept 1 h at

room temperature in the dark after secondary antibody (Dianova GmbH, Hamburg, Germany)

goat-anti-mouse Alexa-Fluor 488, diluted 1:700 in PBS 1% BSA, was applied. Finally cell slides

were washed twice with PBS and embedded in Permafluor mounting medium. Examination was

carried out with a fluorescence microscope (Leica DMRD).

Native Dot Blot Assay

Viral genome containing particles per mutant vector were incubated at 37°C for 30 min

either without or with the monoclonal antibody ADK8 (2 mg/ml), then 10 min on ice. Next,

samples were subjected to a temperature treatment, 37°C, 65°C, 75°C or 99°C for 5 min.

Samples were spotted on a nitrocellulose membrane (Schleicher&Schuell, Dassel, Germany) in

a dot blot chamber. Membrane was blocked in PBS/10% skimmed milk powder and incubated

with A1 antibody, which had been linked to HRP (Zenon labelling Kit, Invitrogen). Particle

visualization was performed using an enhanced chemiluminescence detection kit (Perkin-Elmer,

Boston, USA)

Page 169: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

169

Epitope Mapping

See above section in Chapter 2.

Results and Discussion

Three-Dimensional Reconstruction and Model Docking of the AAV8:ADK8 Complex

A total of 982 particles were extracted from 78 CCD images to generate a final 3D map

with a resolution of ~18.4Å (Figure B-1A). Independent density was present at each three-fold

protrusion, indicating that there was no steric hindrance for the binding of all three protrusions

simultaneously, as was seen for a previous reconstruction, AAV2:C37B (Figure 4-2). An entire

capsid molecule, or 60-mer, was generated from the AAV8 atomic coordinates (PDB 2QA0;

Nam et al., 2007) and was docked into the respective capsid density with a correlation coefficient

(cc) of ~0.42. The cc of the fit was most likely low due to the presence of the excess density

belonging to the bound Fabs as well as the use of a C-α model (Figure B-1B). Since the ADK8

antibody has yet to be sequenced, a model has not been generated. The atomic coordinates for a

generic Fab model (PDB 2FBJ; Suh et al., 1986) were then docked into the identified Fab density

and a density map was generated from this new model (Figure B-1C). A new cc was calculated

(see chapter 2) and was found to be ~0.68. The correlation was improved when the excess

density was filled, but this low correlation is most likely due to the use of C-α model, as

previously mentioned, in addition to the use of rigid-body and manual fitting techniques. In this

case, these functions do not refine or take possible low-energy conformational movements into

account, causing portions of the atomic structures to be outside of the density in order to better fit

more important regions. This is evident from a comparison of the maps used in the determination

of the cc between the cryoEM reconstruction and the fitted models (Figure B-1D).

Page 170: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

170

Analysis of the 3D Models Identifies Proposed Monoclonal Antibody ADK8 Binding Sites on the AAV8 Capsid

Docking atomic models into the cryoEM map allowed for further characterization of the

ADK8 binding site on the AAV8 capsid. First glance identifies the Fab binding to the protrusions

at the three-fold axis of symmetry, leaving the two- and five-fold axes un-occluded. A more

detailed inspection of the docked VP3 capsid monomers identifies VRIV, V, and VIII (Figure 4-

3) of the three-fold axes to be prominent in the antibody interaction. A lesser, but possible

interaction was also noticed at VRVI corresponding to a KDDEE peptide at aa530-534. The

symmetry related manner of this region brings VRV into a position to be ‘cradled’ by VRs IV

and VIII from another symmetry related monomer. These regions were also noted in the

previously determined reconstructions (Table 4-4). Sequence identification of this region

proposes that the peptides GTANTQ (VRIV; aa455-460), TTTGQNNNS (VRV; aa493-501),

and LQQQNT (VRVIII; aa586-591) could be involved in the epitope. It is also interesting to

note that portions of the peptides from VRIV and V have been identified in studies as being

immunogenic in cytotoxic T-cell assays, while the peptide identified in VRVIII was found as an

epitope in MHC II molecules (Chen et al., 2006). Indeed, these regions also overlap with the

sequence based structural alignment from previously determined reconstructions and show

similar patterns. Mainly, the sequences at VRV and VIII both have triplicate repeats of

hydrophilic, polar residues, i.e. QQQ (Table 4-4).

Characterization of rAAV8- and rAAV2- Capsid Mutants to Elucidate the ADK8 Binding Epitope

Many monoclonal antibodies, such as B1, A1, A69 and A20, have been well studied and

characterized in the past (Wistuba et al., 1995; Wobus et al., 2000). The collection of generated

monoclonal antibodies has been rapidly increasing and has greatly contributed to a detailed

analysis of AAV2 (Grimm et al., 1999; Kern et al., 2003), the best studied serotype. The

Page 171: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

171

monoclonal antibody ADK8 is of importance as it could help to further analyze the rhesus

monkey derived AAV8, which is of particular interest to gene therapy (Gao et al., 2002; Wang et

al., 2005).

To investigate possible binding epitopes of ADK8, we chose to construct mutants with

amino acid exchanges of AAV8 towards AAV2 and vice versa. Mutations in the cap gene were

chosen according to the aa identified in the previously described antibody mapping. However, a

homology alignment confirmed that only three of the four optional binding sites are localized in

non-homologous region between AAV 2 and AAV8 (Figure B-2). Therefore, binding site

KDDEE (aa sequence underlined in green) was excluded in the next set of experiments. A

reduced VP expression combined with an impaired capsid assembly has been observed for

several mutants in previous studies (Bleker et al., 2005). On that account, quantitative real-time

PCR and western blot analysis was assayed. In two independent productions, rAAV2 and

rAAV8 as well as the five mutants had been produced and purified. The results of the

quantitative real-time PCR indicate that all mutants, except mutant rAAV8→rAAV2

586LQRGNR591 showed no decrease in capsid assembly (Figure B-3A). The latter did show a

20-fold decrease, but VP expression was not impaired for any mutant (Figure B-3B). It is

interesting to note an extra band for the rAAV2rAAV8 493KTSAD497 mutant and a change

in the banding position for all three rAAV2rAAV8 vectors. It is quite possible that these small

changes at the capsid surface cause differences in glycosylation events or a change in the break-

down pattern of the capsid during denaturing. These results permitted an ADK8-ELISA to parse

mutant vectors for their monoclonal antibody binding ability. We decided to use 1E+10 viral

genome containing particles to check if the assay could be used to detect the amount of mutant

capsids. In case of the first set of produced mutants (aa exchanged from rAAV8→ rAAV2), the

Page 172: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

172

capsids of two mutant vectors, rAAV8→ rAAV2 455GTTTQS460 and rAAV8→ rAAV2

493KTSAD497, could be detected in range of rAAV8. Ten-fold more capsids were detected than

viral genome containing particles had been applied (Figure B-4A). This finding is conform with

the literature, a 10- to 100- fold higher amount of capsids is normally found after the purification

with a single iodixanol step-gradient (Grimm et al., 1999). In contrast to the previously described

mutants, the third mutant vector, rAAV8→ rAAV2 586LQNRGNR591, could not be detected

with the ADK8-ELISA. It indicates that the binding site 586-591 LQQQNT is of utmost

importance in ADK8 binding to rAAV8. This finding was further substantiated by a second set

of produced mutants (aa exchange from rAAV2→ rAAV8). Whereas the mutant vector

rAAV2→ rAAV8 457TQTLG461 could not be detected by the ADK-ELISA, just like rAAV2,

the mutant vector rAAV2→ rAAV8 585QQNTA589, was detected (Figure B-4B). If a seven aa

peptide was inserted at position 590 of rAAV8, ADK8 ELISA was not able to detect any capsids

(Figure B-4C). The motifs had been chosen either from an AAV2 in vitro and in vivo heart tissue

selection (Ying et al. paper under revision) and a muscle-targeting peptide (Yu et al., 2009).

Taken together, the experiments could demonstrate that of all possible binding sites, the position

586-591 LQQQNT is directly involved in ADK8 binding to AAV8. Due to a mutation from

AAV8 to AAV2 at this position, ADK8 was not able to recognize the AAV8 mutant anymore.

Additionally, AAV2 mutated to AAV8 at the given locus, allowed mutant vector capsid

detection in the ADK8-ELISA. Insertion of a 7 aa peptide motif in the binding site at aa 590

caused a complete loss of capsid recognition in the ADK8-ELISA.

In Vitro and In Vivo Neutralization of AAV8 by ADK8

It has been revealed that the seroprevalence of AAV8 is lower than AAV1 and AAV2 in

ten different countries on four different continents (Calcedo et al., 2009). Due to the fact that

AAV2 directed antibodies dominate the global seroprevalence, the AAV8 serotype is more

Page 173: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

173

feasible for the avoidance of antibody-mediated AAV vector neutralization in clinical

application.

To gain insight into the basis of neutralization, it is of importance to characterize

neutralization competency of the derived monoclonal antibody ADK8. We decided to investigate

if ADK8 impacts transduction efficiency in vitro and in vivo. At first, three different vectors,

rAAV2, rAAV8 and mutant rAAV2→ rAAV8 585QQNTA589 were pre-incubated either

without, with 250 ng or 500 ng of monoclonal antibodies (ADK8 or A20). ADK8 was able to

decrease transduction efficiency of AAV8 as well as of mutant rAAV2→ rAAV8

585QQNTA589 by more than 50-fold compared to vector transduction without antibody addition

(Figure B-5A). Furthermore, neutralization was already found if less antibody (250 ng) ADK8

had been applied. However, ADK8 was not able to neutralize AAV2 as this sequence is not

identical in AAV2. As a second control, vectors were pre-incubated with A20, a monoclonal

antibody for AAV2. AAV2 transduction was almost completely neutralized after A20 pre-

incubation. Conversely, A20 did not have any influence on AAV8 transduction, indicating that

the sequence responsible for A20 binding to AAV2 is not found in AAV8. This further implies

that the A20 epitope is most likely in a variable region where AAV2 and 8 differ. Transduction

efficiency for mutant rAAV2→ rAAV8 585QQNTA589 was slightly reduced. Moreover, ADK8

neutralization was also analyzed in vivo. Four groups of 4 (6-9 week old, female NMRI) mice

were chosen for this experiment. As a control, mice were injected with rAAV8 without any

antibodies. The other three groups were i.p. injected with ADK8 (50 µg and 250 µg) or with

ADK4 (250 µg) and 4 h later with rAAV8. Two weeks later, luciferase expression was measured

with an IVIS imaging system. The imaging ascertained that already the amount of 50 µg ADK8

was enough to achieve a complete neutralization of rAAV8 vector transduction (Figure B-5B).

Page 174: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

174

Without i.p. antibody injection, transduction efficiency had a typical imaging pattern of a

hepatotropic AAV vector (Sarkar et al., 2004). The results of the fourth group of mice which had

been injected with ADK4, a monoclonal antibody against AAV4 only, clearly demonstrated that

a monoclonal antibody has to bind rAAV8 for neutralization. Antibodies present in the mouse

system which are not able to bind AAV8, are rapidly cleared from the system and do not impact

reporter expression. Results were verified by reporter expression analysis of liver and muscle

after dissection of the animals (Figure B-5C). As already observed in the imaging, luciferase

reporter expression was reduced more than 100-fold in liver and heart after the 50 µg ADK8 i.p.

injections. After a 250 µg ADK8 i.p. injection, RLU were even below the detection limit.

Injection of ADK4 did not have any impact on luciferase reporter expression. Similar

transduction efficiency was confirmed as for rAAV8 transduction without antibody injection.

The obtained results are in agreement with the fact that ADK8 is a monoclonal antibody

which neutralizes rAAV8 vector and mutant rAAV2 (with inserted binding epitope of ADK8,

585QQNTA589) transduction in vitro and in vivo. Therefore, this monoclonal antibody can be

used to elucidate the basis of neutralization of rAAV8.

Neutralization Analysis of ADK8

Clear attempts have been made to understand the molecular mechanisms of novel

hepatotropic AAVs (Thomas et al., 2004). However, the transduction pathway itself has not

been analyzed as a whole yet. ADK8 is known to neutralize AAV8 in vitro and in vivo, it is of

interest to ask if neutralization occurs at cell-surface binding, endosomal release, or uncoating.

To identify the point of ADK 8 neutralization, a series of experiments was designed.

Binding of AAV8 in presence of ADK8 was analysed by immune fluorescence and DNA dot

blot. In the past it has been demonstrated that monoclonal antibodies such as C37B or C24-B

(Wobus et al., 2000) prevent AAV2 binding. This does not appear to be the case for ADK8.

Page 175: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

175

Neither in presence of IVA7 nor in presence of ADK8, could a reduction in rAAV8 vector

binding to HeLa cells be demonstrated (Figure B-6A), which was also consistent with the DNA

dot blot analysis (Figure B-6B). The same amounts of vector genomes of bound particles could

be identified with and without ADK8 pre-incubation. Interestingly, the addition of ADK4

improved rAAV8 vector binding to the HepG2 cells, but not for rAAV2. To investigate if all

bound particles can enter, infected cells were incubated in 0.05% trypsin to deplete defective or

un-internalized particles from cell surfaces. For rAAV8 and mutant rAAV2 (with inserted

binding epitope of ADK8, 585QQNTA589), less particles could be detected after trypsin

treatment, but not rAAV2. Additionally, pre-incubation of ADK8 showed a reduction in detected

vector genomes after trypsin treatment. However, spots are very low in contrast and needed

confirmation by the analysis of the spots with the plot lane analysis software ImageJ (Figure B-

6C and D). According to the pixel intensity, AAV2 binding and internalization is not hampered

but is even improved by the addition of ADK8 (about 15% increase in pixel intensity). In case of

rAAV8 and mutant rAAV2, ADK8 does not impact vector binding, but viral entry. About 15%

less viral genomes for AAV8 and the AAV2 mutant could be detected after trypsin treatment.

ADK8 pre-incubation caused an additional decrease of detectable viral genomes, the pixel

intensity was 20% reduced in these spots compared to vector genomes without previous antibody

treatment.

It has been observed previously that the A20 antibody has a direct impact on N-terminus

externalization (data not shown). The exposure of the VP1 N termini was conducted by using a

monoclonal antibody A1. A1 is known to bind a defined epitope close to the PLA2 domain on

the N terminus of VP1 (Wobus et al., 2000). Figure B-7 shows that after the incubation of virions

for 5 min at 37°C, no reaction upon A1 supernatant application to the nitrocellulose membrane

Page 176: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

176

could be detected. Detection was found after 5 min treatment at 65°C for rAAV2 and mutant

rAAV2, but not for rAAV8. N termini became accessible at this point. At 71°C and 99°C, signal

detection was shown for all analysed vectors after A1 supernatant was applied. Due to the fact

that the secondary antibody GαM-HRP recognizes A1 supernatant and ADK8 which is used in

the preincubation step, the experiment had to be repeated with purified monoclonal A1 directly

coupled to HR-peroxidase. The results were similar, although the N-terminus was already

partially accessible for rAAV8 at 65°C. If rAAV8 particles were pre-incubated with ADK8, the

N-terminus became even better accessible at 65°C. Reduction of detection signal due to ADK8

pre-treatment was not observed.

Taken together, these results indicate, that ADK8 does not influence AAV8 binding to the

cell but has a slight impact on endocytosis. Additionally, there is no direct or indirect influence

on the N-terminus externalization. This may lead to the hypothesis that ADK8 prevents

uncoating and neutralizes reporter expression of vectors below detection limit if antibody

binding site is present on the capsids.

Impact on rAAV8 vector generation

Pre-existing immune reponses have proven to be a challenge in the field of gene therapy.

Current studies involved in circumventing these responses include shuffling of the cap gene to

produce chimeric vectors and generating wild-type capsids under immunoselective strain, as well

as identifying natural AAVs from other animal species. These methods create highly diversified

AAVs with capsids that incorporate different VRs that often mirror those seen in already

identified genotypes, yet also yield radically different phenotypes. Preliminary charcterization of

many of these vectors have identified poor capsid production, as well as reduced packaging and

altered infectivity profiles, which generally leads to a limited number of viable vectors. Studies

aimed at determining functional regions on the capsid surface have identified VRs that are

Page 177: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

177

necessary for structural integrity, receptor binding, infectivity, and antigenicity. All of these

types of studies add to the basic biology and further characterization of the capsid surface as

human engineering of the capsid component is only fruitful when applied appropriately in a

manner that allows for the generation of vectors that mimic or exceed wt virus production and

infectivity values. The knowledge provided in the study presented here has identified VRs on the

AAV8 capsid surface that are specifically important in antigenicity and thus add to the basic

biology of the capsid surface. The VRs identified here will provide a basis where vector

development using mutational analysis in these regions may help generate antigenic escape

vectors.

Page 178: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

178

Figure B-1. Three-dimensional reconstruction and model fitting for the AAV8:ADK8 complex. A) Final density map for the AAV8:ADK8 structure to ~21Å. The AAV8 viral surface is colored green while the excess density in white was identified as the Fab density. B) Fitting of the AAV8 (PDB 2QA0) structure into the respective capsid density. Density map is shown in grey and the atomic coordinates are in green. The excess white space remaining for the Fabs could easily account for the low fitting statistics. C) Fitting of both Fab and AAV8 capsid models into the reconstructed density. One Fab portion, modeled in as PDB 2FBJ, is shown in red and is hovering above three symmetry related monomers, colored in green, blue, and magenta, at the three-fold axis of symmetry. D) Visual comparison of the maps used to generate the final correlation of the docked models (green) and the original cryoEM reconstruction (grey). An excess of green density from the fit model map can be seen extending beyond the cryoEM map, indicating regions that would decrease the final corrolation. Black arrows mark example regions of density extension.

Page 179: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

179

Figure B-2. Homology alignment of a portion of the G-H loop between AAV2 and AAV8. Amino acid differences are highlighted in blue and sequences of interest are underlined in black. The sequence underlined in green was found to have a possible interaction in the structure analysis, but is obviously conserved among the two viruses, indicating little or no involvement in the epitope.

Figure B-3. Analysis of rAAV2 and rAAV8 mutant vector production. A) A quantitative real-time analysis demonstrated viral genome containing particles for all produced mutants above the detection level (detection limit 5E+7Vg/ml). Compared to AAV2 and AAV8, none of the produced mutants showed a completely diminished packaging capacity. Production of viral genome containing particles was performed twice for each mutant and an average is shown. B) In a western blot analysis, VP proteins are analyzed with a monoclonal antibody (B1). VP1, 2 and 3 are marked with arrows, the stoichiometry of ~1:1:10 is evident.

Page 180: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

180

Figure B-4. Binding ability of rAAV2 and 8 mutant capsids by the ADK8 monoclonal. A) Shows the reaction of the ADK8 antibody with capsids containing 1E+10 viral genomes in an AAV8 capsid ELISA. The effects on the mAb ADK8 detection due to the exchanges of aa 455 - 460 (GTANTQ → GTTTQS), aa 493 - 497 (TTTGQ → KTSAD), aa 586 - 591 (LQQQNT → LQRGNR) from rAAV8 to rAAV2 are demonstrated. B) Detection of AAV2 capsid mutants derived from the exchange of aa 457- 461 (QSRLG → TQTLG) and aa 585 - 589 (RGNRQ → QQNTA) in the AAV 2 capsid. C) Seven aa peptide insertion at position 590 into the rAAV8 capsid and disrupt sequence QNTA influence the mAb ADK8 detection. Mean standard deviations from four independent experiments are shown, an asterik indicates mutants for which capsids could not be detected in the ADK8 ELISA (detection limit, 1E+8 capsids/ml).

Page 181: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

181

Figure B-5. in vitro and in vivo neutralization data A) Prior toHepG2 cell infection (MOI 5E+4), viral vectors were pre-incubated with different amounts (250 ng and 500 ng) of antibody for 30 min at 37°C. After 6 h, viral vector-antibody mix was washed off and cells were harvested two days after infection for the determination of reporter gene expression. B) Four hours after i.p. injection of ADK8 (50 µg and 250 µg) or ADK4, respectively, NMRI mice (female, 9 weeks old, n= 4) were i.v. injected with 1E+11 viral genome containing rAAV8 vectors. Two weeks after injection, mice were injected with D-luciferin, imaged for 5 min (10 min after D-luciferin injection) and sacrificed. C) Transgene expression per mg protein was determined for heart and liver of sacrificed mice with a luciferase reporter assay and NanoOrange. Dashed line indicates the detection limit of the luciferase reporter assay

Page 182: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

182

Figure B-6. Neutralization analysis of the ADK8 mAb. A) Hela cells which had been seeded out on glas slides 24 h earlier, were infected with rAAV8 (MOI 1E+6). After viral vector preincubation with or without ADK8 (0.1 mg) for 30 min at 37°C,. infected Hela cells were kept at 4°C for 30 min. Next, glas slides were washed with PBS, then cells were fixed with ice cold methanol for 10 min and washed with PBS once again. As a negative control, ADK8 was added to the cells or no virus without antibody preincubation at all was applied. As a positive control, rAAV8 infected Hela cells without ADK8 preincubation or with IVA7 preincubation. After fixation, ADK8 hybridoma supernatant was applied for 24 h. Three washes with PBS were applied before GαM-A488 was added (1:700). After a last washing step, glas slides were embedded in Permafluor. B).Binding analysis was performed by DNA dot blot analysis after infection of HepG2 cells with rAAV8, rAAV2 and rAAV2 with aa exchanges at position (585RGNRQ → QQNTA589). At first, genomes containing particles (MOI5E+4) were preincubation with ADK8 or ADK4 for 30 min. Then, cells were infected for 30 min and washed with PBS. Cells were either directly harvested or kept at 37°C for 2 h before harvest and trypsin treatment. For all samples, DNA was extracted and transfered to a membrane for radioactive labelling. Positive control is the viral load without cell infection. The dot blot was performed three times. C) Analysis of spots by a plot lane analysis was performed using ImageJ software. Pixel intensity was determined for all spots containing viral genomes of bound vector particles to the cells. D) shows pixel intensities of viral genome containing particles which entered the cells at 37°C.

Page 183: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

183

Figure B-6. Continued.

Figure B-7. ADK8 and its impact on the N terminus externalization in the post-entry process. AAV8 genome containing particles (1E+9) were incubated with or without ADK8 (1 μg /spot) for 30 min at 37°C. Thereafter, they were kept at either 37°C, 65°C, 71°C or 95°C for 5 min. Particles or particle-antibody mixes were spotted onto a nitrocellulose membrane and reacted with A1 hybridoma supernatant or HR peroxidase coupled A1 antibodies.

Page 184: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

184

LIST OF REFERENCES

Aasted, B., Alexandersen, S., and Christensen, J. (1998). Vaccination with Aleutian mink disease parvovirus (AMDV) capsid proteins enhances disease, while vaccination with the major non-structural AMDV protein causes partial protection from disease. Vaccine 16, 1158-1165.

Aasted, B., Tierney, G. S., and Bloom, M. E. (1984). Analysis of the quantity of antiviral

antibodies from mink infected with different Aleutian disease virus strains. Scand. J. Immunol 19, 395-402.

Adrian, M., Dubochet, J., Lepault, J., and McDowall, A. W. (1984). Cryo-electron microscopy of

viruses. Nature 308, 32-36. Agbandje, M., McKenna, R., Rossmann, M. G., Strassheim, M. L., and Parrish, C. R. (1993).

Structure determination of feline panleukopenia virus empty particles. Proteins 16, 155-171.

Agbandje, M., Parrish, C. R., and Rossmann, M. G. (1995). The structure of parvoviruses. Semin

Virol 6, 299-309. Agbandje-McKenna, M., Llamas-Saiz, A. L., Wang, F., Tattersall, P., and Rossmann, M. G.

(1998). Functional implications of the structure of the murine parvovirus, minute virus of mice. Structure 6, 1369-1381.

Akache, B., Grimm, D., Pandey, K., Yant, S. R., Xu, H., and Kay, M. A. (2006). The 37/67-

kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9. J. Virol 80, 9831-9836.

Alkan, S. S. (2004). Monoclonal antibodies: the story of a discovery that revolutionized science

and medicine. Nat Rev Immunol 4, 153-156. Allander, T., Jartti, T., Gupta, S., Niesters, H. G. M., Lehtinen, P., Osterback, R., Vuorinen, T.,

Waris, M., Bjerkner, A., Tiveljung-Lindell, A., et al. (2007). Human bocavirus and acute wheezing in children. Clin. Infect. Dis 44, 904-910.

Allander, T., Tammi, M. T., Eriksson, M., Bjerkner, A., Tiveljung-Lindell, A., and Andersson,

B. (2005). Cloning of a human parvovirus by molecular screening of respiratory tract samples. Proc. Natl. Acad. Sci. U.S.A 102, 12891-12896.

Anderson, S., Momoeda, M., Kawase, M., Kajigaya, S., and Young, N. S. (1995). Peptides

derived from the unique region of B19 parvovirus minor capsid protein elicit neutralizing antibodies in rabbits. Virology 206, 626-632.

Page 185: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

185

Arbetman, A. E., Lochrie, M., Zhou, S., Wellman, J., Scallan, C., Doroudchi, M. M., Randlev, B., Patarroyo-White, S., Liu, T., Smith, P., et al. (2005). Novel caprine adeno-associated virus (AAV) capsid (AAV-Go.1) is closely related to the primate AAV-5 and has unique tropism and neutralization properties. J. Virol 79, 15238-15245.

Arnold, J. C., Singh, K. K., Spector, S. A., and Sawyer, M. H. (2006). Human bocavirus:

prevalence and clinical spectrum at a children's hospital. Clin. Infect. Dis 43, 283-288. Arthur, J. L., Higgins, G. D., Davidson, G. P., Givney, R. C., and Ratcliff, R. M. (2009). A novel

bocavirus associated with acute gastroenteritis in Australian children. PLoS Pathog 5, e1000391.

Atchison, R. W., Casto, B. C., and Hammon, W. M. (1965). Adenovirus-Associated Defective

Virus Particles. Science 149, 754-755. Auricchio, A., Rivera, V. M., Clackson, T., O'Connor, E. E., Maguire, A. M., Tolentino, M. J.,

Bennett, J., and Wilson, J. M. (2002). Pharmacological regulation of protein expression from adeno-associated viral vectors in the eye. Mol. Ther 6, 238-242.

Azzouz, M., and Mazarakis, N. (2004). Non-primate EIAV-based lentiviral vectors as gene

delivery system for motor neuron diseases. Curr Gene Ther 4, 277-286. Baac, H., Hajós, J. P., Lee, J., Kim, D., Kim, S. J., and Shuler, M. L. (2006). Antibody-based

surface plasmon resonance detection of intact viral pathogen. Biotechnol. Bioeng 94, 815-819.

Bachmann, M. F., and Zinkernagel, R. M. (1996). The influence of virus structure on antibody

responses and virus serotype formation. Immunol. Today 17, 553-558. Bainbridge, J. W. B., Smith, A. J., Barker, S. S., Robbie, S., Henderson, R., Balaggan, K.,

Viswanathan, A., Holder, G. E., Stockman, A., Tyler, N., et al. (2008). Effect of gene therapy on visual function in Leber's congenital amaurosis. N. Engl. J. Med 358, 2231-2239.

Baker, T. S., Olson, N. H., and Fuller, S. D. (1999). Adding the third dimension to virus life

cycles: three-dimensional reconstruction of icosahedral viruses from cryo-electron micrographs. Microbiol. Mol. Biol. Rev 63, 862-922, table of contents.

Barker, D. D., and Berk, A. J. (1987). Adenovirus proteins from both E1B reading frames are

required for transformation of rodent cells by viral infection and DNA transfection. Virology 156, 107-121.

Bartlett, J. S., Samulski, R. J., and McCown, T. J. (1998). Selective and rapid uptake of adeno-

associated virus type 2 in brain. Hum. Gene Ther 9, 1181-1186.

Page 186: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

186

Bartlett, J. S., Wilcher, R., and Samulski, R. J. (2000). Infectious entry pathway of adeno-associated virus and adeno-associated virus vectors. J. Virol 74, 2777-2785.

Basak, S., and Turner, H. (1992). Infectious entry pathway for canine parvovirus. Virology 186,

368-376. Bekeredjian, R., Chen, S., Frenkel, P. A., Grayburn, P. A., and Shohet, R. V. (2003). Ultrasound-

Targeted Microbubble Destruction Can Repeatedly Direct Highly Specific Plasmid Expression to the Heart. Circulation 108, 1022-1026.

Bessis, N., GarciaCozar, F. J., and Boissier, M. (2004). Immune responses to gene therapy

vectors: influence on vector function and effector mechanisms. Gene Ther 11 Suppl 1, S10-17.

Blacklow, N. R., Hoggan, M. D., and Rowe, W. P. (1968). Serologic evidence for human

infection with adenovirus-associated viruses. J. Natl. Cancer Inst 40, 319-327. Blacklow, N. R., Hoggan, M. D., Sereno, M. S., Brandt, C. D., Kim, H. W., Parrott, R. H., and

Chanock, R. M. (1971). A seroepidemiologic study of adenovirus-associated virus infection in infants and children. Am. J. Epidemiol 94, 359-366.

Bleker, S., Sonntag, F., and Kleinschmidt, J. A. (2005). Mutational analysis of narrow pores at

the fivefold symmetry axes of adeno-associated virus type 2 capsids reveals a dual role in genome packaging and activation of phospholipase A2 activity. J. Virol 79, 2528-2540.

Bloom, M. E., Best, S. M., Hayes, S. F., Wells, R. D., Wolfinbarger, J. B., McKenna, R., and

Agbandje-McKenna, M. (2001). Identification of aleutian mink disease parvovirus capsid sequences mediating antibody-dependent enhancement of infection, virus neutralization, and immune complex formation. J. Virol 75, 11116-11127.

Bloom, M. E., Kanno, H., Mori, S., and Wolfinbarger, J. B. (1994). Aleutian mink disease:

puzzles and paradigms. Infect Agents Dis 3, 279-301. Bloom, M. E., Martin, D. A., Oie, K. L., Huhtanen, M. E., Costello, F., Wolfinbarger, J. B.,

Hayes, S. F., and Agbandje-McKenna, M. (1997). Expression of Aleutian mink disease parvovirus capsid proteins in defined segments: localization of immunoreactive sites and neutralizing epitopes to specific regions. J. Virol 71, 705-714.

Bothner, B., Dong, X. F., Bibbs, L., Johnson, J. E., and Siuzdak, G. (1998). Evidence of viral

capsid dynamics using limited proteolysis and mass spectrometry. J. Biol. Chem 273, 673-676.

Boutin, S., Monteilhet, V., Veron, P., Leborgne, C., Benveniste, O., Montus, M., and Masurier,

C. (2010). Prevalence of serum IgG and neutralizing factors against adeno-associated virus types 1, 2, 5, 6, 8 and 9 in the healthy population: implications for gene therapy using AAV vectors. Human Gene Therapy, 100122110120098.

Page 187: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

187

Bowles, D., J.E.R, and Samulski, R. J. (2006). The genus Dependovirus. In Parvoviruses, J. R.

Kerr, M. E. Bloom, R. Linden, and C. Parrish, eds. (New York: Oxford University Press Inc.), pp. 15-23.

Boxall, A. B. (2004). The environmental side effects of medication. EMBO Rep 5, 1110-1116. Boyce, N. (2001). Trial halted after gene shows up in semen. Nature 414, 677. Bramson, J. L., Graham, F. L., and Gauldie, J. (1995). The use of adenoviral vectors for gene

therapy and gene transfer in vivo. Current Opinion in Biotechnology 6, 590-595. Brown, C. S., Jensen, T., Meloen, R. H., Puijk, W., Sugamura, K., Sato, H., and Spaan, W. J.

(1992). Localization of an immunodominant domain on baculovirus-produced parvovirus B19 capsids: correlation to a major surface region on the native virus particle. J. Virol 66, 6989-6996.

Bruemmer, A., Scholari, F., Lopez-Ferber, M., Conway, J. F., and Hewat, E. A. (2005). Structure

of an insect parvovirus (Junonia coenia Densovirus) determined by cryo-electron microscopy. J. Mol. Biol 347, 791-801.

Buchschacher, G. L. (2001). Introduction to retroviruses and retroviral vectors. Somat. Cell Mol.

Genet 26, 1-11. Budker, V., Zhang, G., Danko, I., Williams, P., and Wolff, J. (1998). The efficient expression of

intravascularly delivered DNA in rat muscle. Gene Ther 5, 272-276. Burton, D. R., Saphire, E. O., and Parren, P. W. (2001). A model for neutralization of viruses

based on antibody coating of the virion surface. Curr. Top. Microbiol. Immunol 260, 109-143.

Burton, D. R. (2002). Antibodies, viruses and vaccines. Nat Rev Immunol 2, 706-713. Calcedo, R., Vandenberghe, L. H., Gao, G., Lin, J., and Wilson, J. M. (2009). Worldwide

epidemiology of neutralizing antibodies to adeno-associated viruses. J. Infect. Dis 199, 381-390.

Campe, H., Hartberger, C., and Sing, A. (2008). Role of Human Bocavirus infections in

outbreaks of gastroenteritis. J. Clin. Virol 43, 340-342. Candotti, D., Etiz, N., Parsyan, A., and Allain, J. (2004). Identification and characterization of

persistent human erythrovirus infection in blood donor samples. J. Virol 78, 12169-12178.

Page 188: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

188

Carrillo-Tripp, M., Shepherd, C. M., Borelli, I. A., Venkataraman, S., Lander, G., Natarajan, P., Johnson, J. E., Brooks, C. L., and Reddy, V. S. (2009). VIPERdb2: an enhanced and web API enabled relational database for structural virology. Nucleic Acids Res 37, D436-442.

Carter, B. J. (2005). Adeno-associated virus vectors in clinical trials. Hum. Gene Ther 16, 541-

550. Cecchini, S., Negrete, A., Virag, T., Graham, B. S., Cohen, J. I., and Kotin, R. M. (2009).

Evidence of prior exposure to human bocavirus as determined by a retrospective serological study of 404 serum samples from adults in the United States. Clin. Vaccine Immunol 16, 597-604.

Chacón, P., and Wriggers, W. (2002). Multi-resolution contour-based fitting of macromolecular

structures. J. Mol. Biol 317, 375-384. Chang, L. (2009). Immune responses to lentiviral vectors. In Gene Therapy Immunology (New

Jersey: Wiley-Blackwell), pp. 85-96. Chang, S. F., Sgro, J. Y., and Parrish, C. R. (1992). Multiple amino acids in the capsid structure

of canine parvovirus coordinately determine the canine host range and specific antigenic and hemagglutination properties. J. Virol 66, 6858-6867.

Chao, H., Liu, Y., Rabinowitz, J., Li, C., Samulski, R. J., and Walsh, C. E. (2000). Several log

increase in therapeutic transgene delivery by distinct adeno-associated viral serotype vectors. Mol. Ther 2, 619-623.

Chapman, M. S., and Agbandje-McKenna, M. (2006). Atomic structures of viral particles. In

Parvoviruses, J. R. Kerr, S. F. Cotmore, M. E. Bloom, Linden, R L, and Parrish, C R, eds. (New York, NY: Edward Arnold,Ltd.), pp. 107-123.

Chen, C., Jensen, R. L., Schnepp, B. C., Connell, M. J., Shell, R., Sferra, T. J., Bartlett, J. S.,

Clark, K. R., and Johnson, P. R. (2005). Molecular characterization of adeno-associated viruses infecting children. J. Virol 79, 14781-14792.

Chen, J., Wu, Q., Yang, P., Hsu, H., and Mountz, J. D. (2006). Determination of specific CD4

and CD8 T cell epitopes after AAV2- and AAV8-hF.IX gene therapy. Mol. Ther 13, 260-269.

Chiorini, J. A., Kim, F., Yang, L., and Kotin, R. M. (1999). Cloning and characterization of

adeno-associated virus type 5. J. Virol 73, 1309-1319. Chiorini, J. A., Yang, L., Liu, Y., Safer, B., and Kotin, R. M. (1997). Cloning of adeno-

associated virus type 4 (AAV4) and generation of recombinant AAV4 particles. J. Virol 71, 6823-6833.

Page 189: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

189

Chipman, P. R., Agbandje-McKenna, M., Kajigaya, S., Brown, K. E., Young, N. S., Baker, T. S., and Rossmann, M. G. (1996). Cryo-electron microscopy studies of empty capsids of human parvovirus B19 complexed with its cellular receptor. Proc. Natl. Acad. Sci. U.S.A 93, 7502-7506.

Chirmule, N., Propert, K., Magosin, S., Qian, Y., Qian, R., and Wilson, J. (1999). Immune

responses to adenovirus and adeno-associated virus in humans. Gene Ther 6, 1574-1583. Choi, V. W., McCarty, D. M., and Samulski, R. J. (2005). AAV hybrid serotypes: improved

vectors for gene delivery. Curr Gene Ther 5, 299-310. Chow, B. D. W., and Esper, F. P. (2009). The human bocaviruses: a review and discussion of

their role in infection. Clin. Lab. Med 29, 695-713. Cideciyan, A. V., Hauswirth, W. W., Aleman, T. S., Kaushal, S., Schwartz, S. B., Boye, S. L.,

Windsor, E. A. M., Conlon, T. J., Sumaroka, A., Roman, A. J., et al. (2009). Vision 1 year after gene therapy for Leber's congenital amaurosis. N. Engl. J. Med 361, 725-727.

Clarke, J. K., McFerran, J. B., McKillop, E. R., and Curran, W. L. (1979). Isolation of an adeno

associated virus from sheep. Archives of Virology 60, 171-176. Cockrell, A., and Kafri, T. (2007). Gene delivery by lentivirus vectors. Molecular Biotechnology

36, 184-204. Conlon, T. J., Cossette, T., Erger, K., Choi, Y., Clarke, T., Scott-Jorgensen, M., Song, S.,

Campbell-Thompson, M., Crawford, J., and Flotte, T. R. (2005). Efficient hepatic delivery and expression from a recombinant adeno-associated virus 8 pseudotyped alpha1-antitrypsin vector. Mol. Ther 12, 867-875.

Cotmore, S. F., D'abramo, A. M., Ticknor, C. M., and Tattersall, P. (1999). Controlled

conformational transitions in the MVM virion expose the VP1 N-terminus and viral genome without particle disassembly. Virology 254, 169-181.

Crick, F. (1970). Central dogma of molecular biology. Nature 227, 561-563. Cunningham, S. C., Dane, A. P., Spinoulas, A., Logan, G. J., and Alexander, I. E. (2008). Gene

delivery to the juvenile mouse liver using AAV2/8 vectors. Mol. Ther 16, 1081-1088. Cunningham, S. C., Spinoulas, A., Carpenter, K. H., Wilcken, B., Kuchel, P. W., and Alexander,

I. E. (2009). AAV2/8-mediated correction of OTC deficiency is robust in adult but not neonatal Spf(ash) mice. Mol. Ther 17, 1340-1346.

Davidoff, A. M., Gray, J. T., Ng, C. Y. C., Zhang, Y., Zhou, J., Spence, Y., Bakar, Y., and

Nathwani, A. C. (2005). Comparison of the ability of adeno-associated viral vectors pseudotyped with serotype 2, 5, and 8 capsid proteins to mediate efficient transduction of the liver in murine and nonhuman primate models. Mol. Ther 11, 875-888.

Page 190: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

190

Davidson, B. L., Stein, C. S., Heth, J. A., Martins, I., Kotin, R. M., Derksen, T. A., Zabner, J., Ghodsi, A., and Chiorini, J. A. (2000). Recombinant adeno-associated virus type 2, 4, and 5 vectors: transduction of variant cell types and regions in the mammalian central nervous system. Proc. Natl. Acad. Sci. U.S.A 97, 3428-3432.

Davies, D. R., and Cohen, G. H. (1996). Interactions of protein antigens with antibodies. Proc.

Natl. Acad. Sci. U.S.A 93, 7-12. Davis, N. L., Willis, L. V., Smith, J. F., and Johnston, R. E. (1989). In vitro synthesis of

infectious venezuelan equine encephalitis virus RNA from a cDNA clone: analysis of a viable deletion mutant. Virology 171, 189-204.

Daya, S., Cortez, N., and Berns, K. I. (2009). Adeno-associated virus site-specific integration is

mediated by proteins of the nonhomologous end-joining pathway. J. Virol 83, 11655-11664.

De Smedt, S. C., Demeester, J., and Hennink, W. E. (2000). Cationic polymer based gene

delivery systems. Pharm. Res 17, 113-126. De, B. P., Heguy, A., Hackett, N. R., Ferris, B., Leopold, P. L., Lee, J., Pierre, L., Gao, G.,

Wilson, J. M., and Crystal, R. G. (2006). High levels of persistent expression of alpha1-antitrypsin mediated by the nonhuman primate serotype rh.10 adeno-associated virus despite preexisting immunity to common human adeno-associated viruses. Mol. Ther 13, 67-76.

Delalande, A., Bureau, M., Midoux, P., Bouakaz, A., and Pichon, C. (2010). Ultrasound-assisted

microbubbles gene transfer in tendons for gene therapy. Ultrasonics 50, 269-272. Denby, L., Nicklin, S. A., and Baker, A. H. (2005). Adeno-associated virus (AAV)-7 and -8

poorly transduce vascular endothelial cells and are sensitive to proteasomal degradation. Gene Ther 12, 1534-1538.

Di Paolo, N., and Shayakhmetov, D. (2009). Immune responses to adenoviral vectors. In Gene

Therapy Immunology, R. W. Herzog, ed. (New Jersey: Wiley-Blackwell), pp. 57-84. Di Pasquale, G., Davidson, B. L., Stein, C. S., Martins, I., Scudiero, D., Monks, A., and Chiorini,

J. A. (2003). Identification of PDGFR as a receptor for AAV-5 transduction. Nat. Med 9, 1306-1312.

Dimmock, N. J. (1984). Mechanisms of neutralization of animal viruses. J. Gen. Virol 65 ( Pt 6),

1015-1022. Dimmock, N. J., and Hardy, S. A. (2004). Valency of antibody binding to virions and its

determination by surface plasmon resonance. Rev. Med. Virol 14, 123-135.

Page 191: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

191

Dintzis, R. Z., Middleton, M. H., and Dintzis, H. M. (1983). Studies on the immunogenicity and tolerogenicity of T-independent antigens. J. Immunol 131, 2196-2203.

DiPrimio, N., Asokan, A., Govindasamy, L., Agbandje-McKenna, M., and Samulski, R. J.

(2008). Surface loop dynamics in adeno-associated virus capsid assembly. J. Virol 82, 5178-5189.

Dörner, T., and Radbruch, A. (2007). Antibodies and B cell memory in viral immunity.

Immunity 27, 384-392. Duan, D., Li, Q., Kao, A. W., Yue, Y., Pessin, J. E., and Engelhardt, J. F. (1999). Dynamin is

required for recombinant adeno-associated virus type 2 infection. J. Virol 73, 10371-10376.

Dubochet, J., Adrian, M., Chang, J. J., Homo, J. C., Lepault, J., McDowall, A. W., and Schultz,

P. (1988). Cryo-electron microscopy of vitrified specimens. Q. Rev. Biophys 21, 129-228.

Dubuisson, J., and Rice, C. M. (1993). Sindbis virus attachment: isolation and characterization of

mutants with impaired binding to vertebrate cells. J. Virol 67, 3363-3374. Dupuy, L. C., and Schmaljohn, C. S. (2009). DNA vaccines for biodefense. Expert Rev Vaccines

8, 1739-1754. Ehrengruber, M. U., and Lundstrom, K. (2007). Alphaviruses: Semliki Forest virus and Sindbis

virus vectors for gene transfer into neurons. Curr Protoc Neurosci Chapter 4, Unit 4.22. Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molecular graphics. Acta

Crystallogr. D Biol. Crystallogr 60, 2126-2132. Erles, K., Sebökovà, P., and Schlehofer, J. R. (1999). Update on the prevalence of serum

antibodies (IgG and IgM) to adeno-associated virus (AAV). J. Med. Virol 59, 406-411. Excoffon, K. J. D. A., Koerber, J. T., Dickey, D. D., Murtha, M., Keshavjee, S., Kaspar, B. K.,

Zabner, J., and Schaffer, D. V. (2009). Directed evolution of adeno-associated virus to an infectious respiratory virus. Proc. Natl. Acad. Sci. U.S.A 106, 3865-3870.

Farkas, S. L., Zadori, Z., Benko, M., Essbauer, S., Harrach, B., and Tijssen, P. (2004). A

parvovirus isolated from royal python (Python regius) is a member of the genus Dependovirus. J Gen Virol 85, 555-561.

Felsenstein, J. (1989). PHYLIP—Phylogeny inference package (version 3.2). Cladistics 5, 164-

166. Feschotte, C. (2010). Virology: Bornavirus enters the genome. Nature 463, 39-40.

Page 192: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

192

Fischer, A. C., Beck, S. E., Smith, C. I., Laube, B. L., Askin, F. B., Guggino, S. E., Adams, R. J., Flotte, T. R., and Guggino, W. B. (2003). Successful transgene expression with serial doses of aerosolized rAAV2 vectors in rhesus macaques. Mol. Ther 8, 918-926.

Flotte, T. R. (2005). Adeno-associated virus-based gene therapy for inherited disorders. Pediatr.

Res 58, 1143-1147. Flotte, T. R. (2007). Gene therapy: The first two decades and the current state-of-the-art. J. Cell.

Physiol. 213, 301-305. Foust, K. D., Nurre, E., Montgomery, C. L., Hernandez, A., Chan, C. M., and Kaspar, B. K.

(2009). Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat. Biotechnol 27, 59-65.

Fryer, J. F., Kapoor, A., Minor, P. D., Delwart, E., and Baylis, S. A. (2006). Novel parvovirus

and related variant in human plasma. Emerging Infect. Dis 12, 151-154. Gao, G., Alvira, M. R., Somanathan, S., Lu, Y., Vandenberghe, L. H., Rux, J. J., Calcedo, R.,

Sanmiguel, J., Abbas, Z., and Wilson, J. M. (2003). Adeno-associated viruses undergo substantial evolution in primates during natural infections. Proc. Natl. Acad. Sci. U.S.A 100, 6081-6086.

Gao, G., Vandenberghe, L. H., Alvira, M. R., Lu, Y., Calcedo, R., Zhou, X., and Wilson, J. M.

(2004). Clades of Adeno-associated viruses are widely disseminated in human tissues. J. Virol 78, 6381-6388.

Gao, G., Vandenberghe, L. H., and Wilson, J. M. (2005). New recombinant serotypes of AAV

vectors. Curr Gene Ther 5, 285-297. Gao, G., Alvira, M. R., Wang, L., Calcedo, R., Johnston, J., and Wilson, J. M. (2002). Novel

adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc. Natl. Acad. Sci. U.S.A 99, 11854-11859.

Gao, X., Kim, K., and Liu, D. (2007). Nonviral gene delivery: What we know and what is next.

AAPS J 9, E92-E104. Girod, A., Wobus, C. E., Zádori, Z., Ried, M., Leike, K., Tijssen, P., Kleinschmidt, J. A., and

Hallek, M. (2002). The VP1 capsid protein of adeno-associated virus type 2 is carrying a phospholipase A2 domain required for virus infectivity. J. Gen. Virol 83, 973-978.

Govindasamy, L., Hueffer, K., Parrish, C. R., and Agbandje-McKenna, M. (2003). Structures of

host range-controlling regions of the capsids of canine and feline parvoviruses and mutants. J. Virol 77, 12211-12221.

Page 193: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

193

Govindasamy, L., Padron, E., McKenna, R., Muzyczka, N., Kaludov, N., Chiorini, J. A., and Agbandje-McKenna, M. (2006). Structurally mapping the diverse phenotype of adeno-associated virus serotype 4. J. Virol 80, 11556-11570.

Grieger, J. C., Johnson, J. S., Gurda-Whitaker, B., Agbandje-McKenna, M., and Samulski, R. J.

(2007). Surface-exposed adeno-associated virus Vp1-NLS capsid fusion protein rescues infectivity of noninfectious wild-type Vp2/Vp3 and Vp3-only capsids but not that of fivefold pore mutant virions. J. Virol 81, 7833-7843.

Grifman, M., Trepel, M., Speece, P., Gilbert, L. B., Arap, W., Pasqualini, R., and Weitzman, M.

D. (2001). Incorporation of tumor-targeting peptides into recombinant adeno-associated virus capsids. Mol. Ther 3, 964-975.

Grimm, D., Kern, A., Pawlita, M., Ferrari, F., Samulski, R., and Kleinschmidt, J. (1999).

Titration of AAV-2 particles via a novel capsid ELISA: packaging of genomes can limit production of recombinant AAV-2. Gene Ther 6, 1322-1330.

Grimm, D., Kern, A., Rittner, K., and Kleinschmidt, J. A. (1998). Novel tools for production and

purification of recombinant adenoassociated virus vectors. Hum. Gene Ther 9, 2745-2760.

Guex, N., and Peitsch, M. C. (1997). SWISS-MODEL and the Swiss-PdbViewer: an

environment for comparative protein modeling. Electrophoresis 18, 2714-2723. Hacein-Bey-Abina, S., von Kalle, C., Schmidt, M., Le Deist, F., Wulffraat, N., McIntyre, E.,

Radford, I., Villeval, J., Fraser, C. C., Cavazzana-Calvo, M., et al. (2003). A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N. Engl. J. Med 348, 255-256.

Hacein-Bey-Abina, S., Le Deist, F., Carlier, F., Bouneaud, C., Hue, C., De Villartay, J.,

Thrasher, A. J., Wulffraat, N., Sorensen, R., Dupuis-Girod, S., et al. (2002). Sustained Correction of X-Linked Severe Combined Immunodeficiency by ex Vivo Gene Therapy. N Engl J Med 346, 1185-1193.

Hafenstein, S., Bowman, V. D., Sun, T., Nelson, C. D. S., Palermo, L. M., Chipman, P. R.,

Battisti, A. J., Parrish, C. R., and Rossmann, M. G. (2009). Structural comparison of different antibodies interacting with parvovirus capsids. J. Virol 83, 5556-5566.

Hafenstein, S., Palermo, L. M., Kostyuchenko, V. A., Xiao, C., Morais, M. C., Nelson, C. D. S.,

Bowman, V. D., Battisti, A. J., Chipman, P. R., Parrish, C. R., et al. (2007). Asymmetric binding of transferrin receptor to parvovirus capsids. Proc. Natl. Acad. Sci. U.S.A 104, 6585-6589.

Page 194: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

194

Halbert, C. L., Miller, A. D., McNamara, S., Emerson, J., Gibson, R. L., Ramsey, B., and Aitken, M. L. (2006). Prevalence of neutralizing antibodies against adeno-associated virus (AAV) types 2, 5, and 6 in cystic fibrosis and normal populations: Implications for gene therapy using AAV vectors. Hum. Gene Ther 17, 440-447.

Hardy, S. A., and Dimmock, N. J. (2003). Valency of antibody binding to enveloped virus

particles as determined by surface plasmon resonance. J. Virol 77, 1649-1652. Harris, A., Belnap, D., Watts, N., Conway, J., Cheng, N., Stahl, S., Vethanayagam, J., Wingfield,

P., and Steven, A. (2006). Epitope Diversity of Hepatitis B Virus Capsids: Quasi-equivalent Variations in Spike Epitopes and Binding of Different Antibodies to the same Epitope. Journal of Molecular Biology 355, 562-576.

Hauck, B., Xu, R. R., Xie, J., Wu, W., Ding, Q., Sipler, M., Wang, H., Chen, L., Wright, J. F.,

and Xiao, W. (2006). Efficient AAV1-AAV2 hybrid vector for gene therapy of hemophilia. Hum. Gene Ther 17, 46-54.

Hauswirth, W. W., Aleman, T. S., Kaushal, S., Cideciyan, A. V., Schwartz, S. B., Wang, L.,

Conlon, T. J., Boye, S. L., Flotte, T. R., Byrne, B. J., et al. (2008). Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum. Gene Ther 19, 979-990.

Havelka, W. A., Henderson, R., and Oesterhelt, D. (1995). Three-dimensional structure of

halorhodopsin at 7 A resolution. J. Mol. Biol 247, 726-738. van Heel, M., and Schatz, M. (2005). Fourier shell correlation threshold criteria. J. Struct. Biol

151, 250-262. Heller, R., Gilbert, R., and Jaroszeski, M. J. (2000). Clinical Trials for Solid Tumors Using

Electrochemotherapy Available at: http://dx.doi.org.lp.hscl.ufl.edu/10.1385/1-59259-080-2:137 [Accessed January 7, 2010].

Horie, M., Honda, T., Suzuki, Y., Kobayashi, Y., Daito, T., Oshida, T., Ikuta, K., Jern, P.,

Gojobori, T., Coffin, J. M., et al. (2010). Endogenous non-retroviral RNA virus elements in mammalian genomes. Nature 463, 84-87.

Huang, X., and Yang, Y. (2009). Innate immune recognition of viruses and viral vectors. Hum.

Gene Ther 20, 293-301. Huber, M., and Trkola, A. (2007). Humoral immunity to HIV-1: neutralization and beyond. J.

Intern. Med 262, 5-25. Hueffer, K., Govindasamy, L., Agbandje-McKenna, M., and Parrish, C. R. (2003). Combinations

of two capsid regions controlling canine host range determine canine transferrin receptor binding by canine and feline parvoviruses. J. Virol 77, 10099-10105.

Page 195: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

195

Huttner, N. A., Girod, A., Perabo, L., Edbauer, D., Kleinschmidt, J. A., Büning, H., and Hallek,

M. (2003). Genetic modifications of the adeno-associated virus type 2 capsid reduce the affinity and the neutralizing effects of human serum antibodies. Gene Ther 10, 2139-2147.

Inagaki, K., Piao, C., Kotchey, N. M., Wu, X., and Nakai, H. (2008). Frequency and spectrum of

genomic integration of recombinant adeno-associated virus serotype 8 vector in neonatal mouse liver. J. Virol 82, 9513-9524.

Ivics, Z., and Izsvák, Z. (2006). Transposons for gene therapy! Curr Gene Ther 6, 593-607. Izsvák, Z., Chuah, M. K. L., Vandendriessche, T., and Ivics, Z. (2009). Efficient stable gene

transfer into human cells by the Sleeping Beauty transposon vectors. Methods 49, 287-297.

Janeway, C., Travers, P., Walport, M., and Shlomchik, M. (2005). Immunobiology: the immune

system in health and disease (New York: Garland Science Publishing). Jiang, H., Lillicrap, D., Patarroyo-White, S., Liu, T., Qian, X., Scallan, C. D., Powell, S., Keller,

T., McMurray, M., Labelle, A., et al. (2006). Multiyear therapeutic benefit of AAV serotypes 2, 6, and 8 delivering factor VIII to hemophilia A mice and dogs. Blood 108, 107-115.

Johnson, J. E., and Chiu, W. (2000). Structures of virus and virus-like particles. Curr. Opin.

Struct. Biol 10, 229-235. Jooss, K., and Chirmule, N. (2003). Immunity to adenovirus and adeno-associated viral vectors:

implications for gene therapy. Gene Ther 10, 955-963. Kahn, J. S., Kesebir, D., Cotmore, S. F., D'Abramo, A., Cosby, C., Weibel, C., and Tattersall, P.

(2008). Seroepidemiology of human bocavirus defined using recombinant virus-like particles. J. Infect. Dis 198, 41-50.

Kajigaya, S., Fujii, H., Field, A., Anderson, S., Rosenfeld, S., Anderson, L. J., Shimada, T., and

Young, N. S. (1991). Self-assembled B19 parvovirus capsids, produced in a baculovirus system, are antigenically and immunogenically similar to native virions. Proc. Natl. Acad. Sci. U.S.A 88, 4646-4650.

Kaludov, N., Brown, K. E., Walters, R. W., Zabner, J., and Chiorini, J. A. (2001). Adeno-

associated virus serotype 4 (AAV4) and AAV5 both require sialic acid binding for hemagglutination and efficient transduction but differ in sialic acid linkage specificity. J. Virol 75, 6884-6893.

Page 196: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

196

Kantola, K., Hedman, L., Allander, T., Jartti, T., Lehtinen, P., Ruuskanen, O., Hedman, K., and Söderlund-Venermo, M. (2008). Serodiagnosis of human bocavirus infection. Clin. Infect. Dis 46, 540-546.

Kapoor, A., Slikas, E., Simmonds, P., Chieochansin, T., Naeem, A., Shaukat, S., Alam, M. M.,

Sharif, S., Angez, M., Zaidi, S., et al. (2009). A newly identified bocavirus species in human stool. J. Infect. Dis 199, 196-200.

Kashiwakura, Y., Tamayose, K., Iwabuchi, K., Hirai, Y., Shimada, T., Matsumoto, K.,

Nakamura, T., Watanabe, M., Oshimi, K., and Daida, H. (2005). Hepatocyte growth factor receptor is a coreceptor for adeno-associated virus type 2 infection. J. Virol 79, 609-614.

Kaufmann, B., Chipman, P. R., Kostyuchenko, V. A., Modrow, S., and Rossmann, M. G. (2008).

Visualization of the externalized VP2 N termini of infectious human parvovirus B19. J. Virol 82, 7306-7312.

Kaufmann, B., López-Bueno, A., Mateu, M. G., Chipman, P. R., Nelson, C. D. S., Parrish, C. R.,

Almendral, J. M., and Rossmann, M. G. (2007). Minute virus of mice, a parvovirus, in complex with the Fab fragment of a neutralizing monoclonal antibody. J. Virol 81, 9851-9858.

Kaufmann, B., Simpson, A. A., and Rossmann, M. G. (2004). The structure of human parvovirus

B19. Proc. Natl. Acad. Sci. U.S.A 101, 11628-11633. Kawase, M., Momoeda, M., Young, N. S., and Kajigaya, S. (1995). Most of the VP1 unique

region of B19 parvovirus is on the capsid surface. Virology 211, 359-366. Kern, A., Schmidt, K., Leder, C., Müller, O. J., Wobus, C. E., Bettinger, K., Von der Lieth, C.

W., King, J. A., and Kleinschmidt, J. A. (2003). Identification of a heparin-binding motif on adeno-associated virus type 2 capsids. J. Virol 77, 11072-11081.

Kesebir, D., Vazquez, M., Weibel, C., Shapiro, E. D., Ferguson, D., Landry, M. L., and Kahn, J.

S. (2006). Human bocavirus infection in young children in the United States: molecular epidemiological profile and clinical characteristics of a newly emerging respiratory virus. J. Infect. Dis 194, 1276-1282.

Khuri, F. R., Nemunaitis, J., Ganly, I., Arseneau, J., Tannock, I. F., Romel, L., Gore, M.,

Ironside, J., MacDougall, R. H., Heise, C., et al. (2000). a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat. Med 6, 879-885.

Kidd, K. A., Blanchfield, P. J., Mills, K. H., Palace, V. P., Evans, R. E., Lazorchak, J. M., and

Flick, R. W. (2007). Collapse of a fish population after exposure to a synthetic estrogen. Proc. Natl. Acad. Sci. U.S.A 104, 8897-8901.

Page 197: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

197

Klasse, P. J., and Sattentau, Q. J. (2002). Occupancy and mechanism in antibody-mediated neutralization of animal viruses. J. Gen. Virol 83, 2091-2108.

Kleywegt, G. J., and Jones, T. A. (1996). xdlMAPMAN and xdlDATAMAN - programs for

reformatting, analysis and manipulation of biomacromolecular electron-density maps and reflection data sets. Acta Crystallogr. D Biol. Crystallogr 52, 826-828.

Knowles, B. B., Howe, C. C., and Aden, D. P. (1980). Human hepatocellular carcinoma cell

lines secrete the major plasma proteins and hepatitis B surface antigen. Science 209, 497-499.

Koerber, J. T., Jang, J., and Schaffer, D. V. (2008). DNA shuffling of adeno-associated virus

yields functionally diverse viral progeny. Mol. Ther 16, 1703-1709. Kontou, M., Govindasamy, L., Nam, H., Bryant, N., Llamas-Saiz, A. L., Foces-Foces, C.,

Hernando, E., Rubio, M., McKenna, R., Almendral, J. M., et al. (2005). Structural determinants of tissue tropism and in vivo pathogenicity for the parvovirus minute virus of mice. J. Virol 79, 10931-10943.

Kotin, R. M., Siniscalco, M., Samulski, R. J., Zhu, X. D., Hunter, L., Laughlin, C. A.,

McLaughlin, S., Muzyczka, N., Rocchi, M., and Berns, K. I. (1990). Site-specific integration by adeno-associated virus. Proceedings of the National Academy of Sciences of the United States of America 87, 2211-2215.

Krissinel, E., and Henrick, K. (2004). Secondary-structure matching (SSM), a new tool for fast

protein structure alignment in three dimensions. Acta Crystallogr. D Biol. Crystallogr 60, 2256-2268.

Kronenberg, S., Böttcher, B., von der Lieth, C. W., Bleker, S., and Kleinschmidt, J. A. (2005). A

conformational change in the adeno-associated virus type 2 capsid leads to the exposure of hidden VP1 N termini. J. Virol 79, 5296-5303.

Kuck, D., Kern, A., and Kleinschmidt, J. A. (2007). Development of AAV serotype-specific

ELISAs using novel monoclonal antibodies. J. Virol. Methods 140, 17-24. Kühler, T. C., Andersson, M., Carlin, G., Johnsson, A., and Akerblom, L. (2009). Do biological

medicinal products pose a risk to the environment?: a current view on ecopharmacovigilance. Drug Saf 32, 995-1000.

Lai, Y., Yue, Y., and Duan, D. (2010). Evidence for the failure of adeno-associated virus

serotype 5 to package a viral genome > or = 8.2 kb. Mol. Ther 18, 75-79. Langeveld, J. P., Casal, J. I., Vela, C., Dalsgaard, K., Smale, S. H., Puijk, W. C., and Meloen, R.

H. (1993). B-cell epitopes of canine parvovirus: distribution on the primary structure and exposure on the viral surface. J. Virol 67, 765-772.

Page 198: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

198

Lee, J. S., Hadjipanayis, A. G., and Parker, M. D. (2005). Viral vectors for use in the development of biodefense vaccines. Adv. Drug Deliv. Rev 57, 1293-1314.

Levy, H. C., Bowman, V. D., Govindasamy, L., McKenna, R., Nash, K., Warrington, K., Chen,

W., Muzyczka, N., Yan, X., Baker, T. S., et al. (2009). Heparin binding induces conformational changes in Adeno-associated virus serotype 2. J. Struct. Biol 165, 146-156.

Lewis, J. K., Bothner, B., Smith, T. J., and Siuzdak, G. (1998). Antiviral agent blocks breathing

of the common cold virus. Proc. Natl. Acad. Sci. U.S.A 95, 6774-6778. Li, H., Lin, S., Giles-Davis, W., Li, Y., Zhou, D., Xiang, Z. Q., High, K. A., and Ertl, H. C. J.

(2009). A preclinical animal model to assess the effect of pre-existing immunity on AAV-mediated gene transfer. Mol. Ther 17, 1215-1224.

Li, Q., Yafal, A. G., Lee, Y. M., Hogle, J., and Chow, M. (1994). Poliovirus neutralization by

antibodies to internal epitopes of VP4 and VP1 results from reversible exposure of these sequences at physiological temperature. J. Virol 68, 3965-3970.

Li, S., and Ma, Z. (2001). Nonviral gene therapy. Curr Gene Ther 1, 201-226. Li, Z., Dullmann, J., Schiedlmeier, B., Schmidt, M., von Kalle, C., Meyer, J., Forster, M.,

Stocking, C., Wahlers, A., Frank, O., et al. (2002). Murine Leukemia Induced by Retroviral Gene Marking. Science 296, 497.

Liljeström, P., and Garoff, H. (1991). A new generation of animal cell expression vectors based

on the Semliki Forest virus replicon. Biotechnology (N.Y.) 9, 1356-1361. Lin, S., and Ertl, H. C. (2009). Immune responses to Adeno-associated virus vectors. In Gene

Therapy Immunology, Roland Herzog, ed. (Hoboken, New Jersey: John Wiley & Sons, Inc.), pp. 97-124.

Linden, R. M., Ward, P., Giraud, C., Winocour, E., and Berns, K. I. (1996). Site-specific

integration by adeno-associated virus. Proceedings of the National Academy of Sciences of the United States of America 93, 11288-11294.

Lindner, J., and Modrow, S. (2008). Human bocavirus--a novel parvovirus to infect humans.

Intervirology 51, 116-122. Liu, G., Martins, I. H., Chiorini, J. A., and Davidson, B. L. (2005). Adeno-associated virus type

4 (AAV4) targets ependyma and astrocytes in the subventricular zone and RMS. Gene Ther 12, 1503-1508.

Llamas-Saiz, A. L., Agbandje-McKenna, M., Parker, J. S., Wahid, A. T., Parrish, C. R., and

Rossmann, M. G. (1996). Structural analysis of a mutation in canine parvovirus which controls antigenicity and host range. Virology 225, 65-71.

Page 199: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

199

Lochrie, M. A., Tatsuno, G. P., Christie, B., McDonnell, J. W., Zhou, S., Surosky, R., Pierce, G.

F., and Colosi, P. (2006). Mutations on the external surfaces of adeno-associated virus type 2 capsids that affect transduction and neutralization. J. Virol 80, 821-834.

López de Turiso, J. A., Cortés, E., Ranz, A., García, J., Sanz, A., Vela, C., and Casal, J. I. (1991).

Fine mapping of canine parvovirus B cell epitopes. J. Gen. Virol 72 ( Pt 10), 2445-2456. López-Bueno, A., Mateu, M. G., and Almendral, J. M. (2003). High mutant frequency in

populations of a DNA virus allows evasion from antibody therapy in an immunodeficient host. J. Virol 77, 2701-2708.

Lu, Y., and Song, S. (2009). Distinct immune responses to transgene products from rAAV1 and

rAAV8 vectors. Proc. Natl. Acad. Sci. U.S.A 106, 17158-17162. Luchsinger, E., Strobbe, R., Wellemans, G., Dekegel, D., and Sprececher-Goldbeeger, S. (1970).

Haemagglutinating adeno-associated virus (AAV) in association with bovine adenovirus type 1. Archives of Virology 31, 390-392.

Lufino, M. M. P., Edser, P. A. H., and Wade-Martins, R. (2008). Advances in high-capacity

extrachromosomal vector technology: episomal maintenance, vector delivery, and transgene expression. Mol. Ther 16, 1525-1538.

Lundberg, C., Björklund, T., Carlsson, T., Jakobsson, J., Hantraye, P., Déglon, N., and Kirik, D.

(2008). Applications of lentiviral vectors for biology and gene therapy of neurological disorders. Curr Gene Ther 8, 461-473.

Lundstrom, K. (2004). Gene therapy applications of viral vectors. Technol. Cancer Res. Treat 3,

467-477. Lundstrom, K. (2003). Latest development in viral vectors for gene therapy. Trends Biotechnol

21, 117-122. M, A., and Chapman, M S (2006). Corrleating structure with function in the viral capsid. In

Parvoviruses, Kerr, J R, Cotmore, S F, Bloom, M E, Linden, R M, and Parrish, C R, eds. (New York, NY: Edward Arnold,Ltd.), pp. 125-139.

Macnab, S., and Whitehouse, A. (2009). Progress and prospects: human artificial chromosomes.

Gene Ther 16, 1180-1188. Maguire, A. M., Simonelli, F., Pierce, E. A., Pugh, E. N., Mingozzi, F., Bennicelli, J., Banfi, S.,

Marshall, K. A., Testa, F., Surace, E. M., et al. (2008). Safety and efficacy of gene transfer for Leber's congenital amaurosis. N. Engl. J. Med 358, 2240-2248.

Page 200: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

200

Manno, C. S., Pierce, G. F., Arruda, V. R., Glader, B., Ragni, M., Rasko, J. J., Rasko, J., Ozelo, M. C., Hoots, K., Blatt, P., et al. (2006). Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat. Med 12, 342-347.

Marconi, P., Argnani, R., Epstein, A. L., and Manservigi, R. (2009). HSV as a Vector in Vaccine

Development and Gene Therapy. Adv. Exp. Med. Biol 655, 118-144. Margaritis, P., Roy, E., Aljamali, M. N., Downey, H. D., Giger, U., Zhou, S., Merricks, E.,

Dillow, A., Ezban, M., Nichols, T. C., et al. (2009). Successful treatment of canine hemophilia by continuous expression of canine FVIIa. Blood 113, 3682-3689.

Martin, A. C., Cheetham, J. C., and Rees, A. R. (1989). Modeling antibody hypervariable loops:

a combined algorithm. Proc. Natl. Acad. Sci. U.S.A 86, 9268-9272. Matthews, K. S., Alvarez, R. D., and Curiel, D. T. (2009). Advancements in adenoviral based

virotherapy for ovarian cancer. Advanced Drug Delivery Reviews 61, 836-841. Mays, L. E., and Wilson, J. M. (2009). Identification of the murine AAVrh32.33 capsid-specific

CD8+ T cell epitopes. J Gene Med 11, 1095-1102. McCarty, D. M., Young, S. M., and Samulski, R. J. (2004). INTEGRATION OF ADENO-

ASSOCIATED VIRUS (AAV) AND RECOMBINANT AAV VECTORS. Annu. Rev. Genet. 38, 819-845.

McCullough, K. C., De Simone, F., Brocchi, E., Capucci, L., Crowther, J. R., and Kihm, U.

(1992). Protective immune response against foot-and-mouth disease. J. Virol 66, 1835-1840.

McKenna, R., Olson, N. H., Chipman, P. R., Baker, T. S., Booth, T. F., Christensen, J., Aasted,

B., Fox, J. M., Bloom, M. E., Wolfinbarger, J. B., et al. (1999). Three-dimensional structure of Aleutian mink disease parvovirus: implications for disease pathogenicity. J. Virol 73, 6882-6891.

Meng, E. C., Pettersen, E. F., Couch, G. S., Huang, C. C., and Ferrin, T. E. (2006). Tools for

integrated sequence-structure analysis with UCSF Chimera. BMC Bioinformatics 7, 339. Meyer, K. B., Thompson, M. M., Levy, M. Y., Barron, L. G., and Szoka, F. C. (1995).

Intratracheal gene delivery to the mouse airway: characterization of plasmid DNA expression and pharmacokinetics. Gene Ther 2, 450-460.

Miao, C. H., Ye, P., Thompson, A. R., Rawlings, D. J., and Ochs, H. D. (2006).

Immunomodulation of transgene responses following naked DNA transfer of human factor VIII into hemophilia A mice. Blood 108, 19-27.

Page 201: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

201

Midoux, P., Breuzard, G., Gomez, J. P., and Pichon, C. (2008). Polymer-based gene delivery: a current review on the uptake and intracellular trafficking of polyplexes. Curr Gene Ther 8, 335-352.

Midoux, P., Pichon, C., Yaouanc, J., and Jaffrès, P. (2009). Chemical vectors for gene delivery: a

current review on polymers, peptides and lipids containing histidine or imidazole as nucleic acids carriers. British Journal of Pharmacology 157, 166-178.

Miller, E. B., Gurda-Whitaker, B., Govindasamy, L., McKenna, R., Zolotukhin, S., Muzyczka,

N., and Agbandje-McKenna, M. (2006). Production, purification and preliminary X-ray crystallographic studies of adeno-associated virus serotype 1. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun 62, 1271-1274.

Milich, D. R., and McLachlan, A. (1986). The nucleocapsid of hepatitis B virus is both a T-cell-

independent and a T-cell-dependent antigen. Science 234, 1398-1401. Mingozzi, F., Maus, M. V., Hui, D. J., Sabatino, D. E., Murphy, S. L., Rasko, J. E. J., Ragni, M.

V., Manno, C. S., Sommer, J., Jiang, H., et al. (2007). CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat. Med 13, 419-422.

Mislick, K. A., and Baldeschwieler, J. D. (1996). Evidence for the role of proteoglycans in

cation-mediated gene transfer. Proceedings of the National Academy of Sciences of the United States of America 93, 12349-12354.

Monteny, M., Niesters, H. G. M., Moll, H. A., and Berger, M. Y. (2007). Human bocavirus in

febrile children, The Netherlands. Emerging Infect. Dis 13, 180-182. Mori, S., Wang, L., Takeuchi, T., and Kanda, T. (2004). Two novel adeno-associated viruses

from cynomolgus monkey: pseudotyping characterization of capsid protein. Virology 330, 375-383.

Moscioni, D., Morizono, H., McCarter, R. J., Stern, A., Cabrera-Luque, J., Hoang, A.,

Sanmiguel, J., Wu, D., Bell, P., Gao, G., et al. (2006). Long-term correction of ammonia metabolism and prolonged survival in ornithine transcarbamylase-deficient mice following liver-directed treatment with adeno-associated viral vectors. Mol. Ther 14, 25-33.

Moskalenko, M., Chen, L., van Roey, M., Donahue, B. A., Snyder, R. O., McArthur, J. G., and

Patel, S. D. (2000). Epitope mapping of human anti-adeno-associated virus type 2 neutralizing antibodies: implications for gene therapy and virus structure. J. Virol 74, 1761-1766.

Muzyczka, N. (1994). Adeno-associated virus (AAV) vectors: will they work? J. Clin. Invest 94,

1351.

Page 202: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

202

Muzyczka, N., and Berns, K. I. (2001). In Fields Virology, D. Knipe and P. Howley, eds. (New York: Lippincott, Williams and Wilkins), pp. 2327-60.

Muzyczka, N., and Warrington, K. H. (2005). Custom adeno-associated virus capsids: the next

generation of recombinant vectors with novel tropism. Hum. Gene Ther 16, 408-416. Nabel, G. J. (2004). Genetic, cellular and immune approaches to disease therapy: past and future.

Nat Med 10, 135-141. Nakai, H., Fuess, S., Storm, T. A., Muramatsu, S., Nara, Y., and Kay, M. A. (2005). Unrestricted

hepatocyte transduction with adeno-associated virus serotype 8 vectors in mice. J. Virol 79, 214-224.

Nam, H., Lane, M. D., Padron, E., Gurda, B., McKenna, R., Kohlbrenner, E., Aslanidi, G.,

Byrne, B., Muzyczka, N., Zolotukhin, S., et al. (2007). Structure of adeno-associated virus serotype 8, a gene therapy vector. J. Virol 81, 12260-12271.

Nanda, K., Vancini, R., Ribeiro, M., Brown, D. T., and Hernandez, R. (2009). A high capacity

Alphavirus heterologous gene delivery system. Virology 390, 368-373. Nason, E. L., Wetzel, J. D., Mukherjee, S. K., Barton, E. S., Prasad, B. V., and Dermody, T. S.

(2001). A monoclonal antibody specific for reovirus outer-capsid protein sigma3 inhibits sigma1-mediated hemagglutination by steric hindrance. J. Virol 75, 6625-6634.

Neumann, E., Schaefer-Ridder, M., Wang, Y., and Hofschneider, P. H. (1982). Gene transfer

into mouse lyoma cells by electroporation in high electric fields. EMBO J 1, 841-845. Nicklin, S. A., Buening, H., Dishart, K. L., de Alwis, M., Girod, A., Hacker, U., Thrasher, A. J.,

Ali, R. R., Hallek, M., and Baker, A. H. (2001). Efficient and selective AAV2-mediated gene transfer directed to human vascular endothelial cells. Mol. Ther 4, 174-181.

O'Donnell, J., Taylor, K. A., and Chapman, M. S. (2009). Adeno-associated virus-2 and its

primary cellular receptor--Cryo-EM structure of a heparin complex. Virology 385, 434-443.

Oliphant, T., and Diamond, M. S. (2007). The molecular basis of antibody-mediated

neutralization of West Nile virus. Expert Opin Biol Ther 7, 885-892. Olson, E. J., Haskell, S. R. R., Frank, R. K., Lehmkuhl, H. D., Hobbs, L. A., Warg, J. V.,

Landgraf, J. G., and Wünschmann, A. (2004). Isolation of an adenovirus and an adeno-associated virus from goat kids with enteritis. J. Vet. Diagn. Invest 16, 461-464.

Opie, S. R., Warrington, K. H., Agbandje-McKenna, M., Zolotukhin, S., and Muzyczka, N.

(2003). Identification of amino acid residues in the capsid proteins of adeno-associated virus type 2 that contribute to heparan sulfate proteoglycan binding. J. Virol 77, 6995-7006.

Page 203: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

203

Pacak, C. A., Mah, C. S., Thattaliyath, B. D., Conlon, T. J., Lewis, M. A., Cloutier, D. E.,

Zolotukhin, I., Tarantal, A. F., and Byrne, B. J. (2006). Recombinant adeno-associated virus serotype 9 leads to preferential cardiac transduction in vivo. Circ. Res 99, e3-9.

Padron, E., Bowman, V., Kaludov, N., Govindasamy, L., Levy, H., Nick, P., McKenna, R.,

Muzyczka, N., Chiorini, J. A., Baker, T. S., et al. (2005). Structure of adeno-associated virus type 4. J. Virol 79, 5047-5058.

Parker, A. L., Newman, C., Briggs, S., Seymour, L., and Sheridan, P. J. (2003). Nonviral gene

delivery: techniques and implications for molecular medicine. Expert Rev Mol Med 5, 1-15.

Parker, J. S., Murphy, W. J., Wang, D., O'Brien, S. J., and Parrish, C. R. (2001). Canine and

feline parvoviruses can use human or feline transferrin receptors to bind, enter, and infect cells. J. Virol 75, 3896-3902.

Parker, J. S., and Parrish, C. R. (2000). Cellular uptake and infection by canine parvovirus

involves rapid dynamin-regulated clathrin-mediated endocytosis, followed by slower intracellular trafficking. J. Virol 74, 1919-1930.

Parks, R., and Graham, F. (1997). A helper-dependent system for adenovirus vector production

helps define a lower limit for efficient DNA packaging. J. Virol. 71, 3293-3298. Parrish, C. R., Aquadro, C. F., Strassheim, M. L., Evermann, J. F., Sgro, J. Y., and Mohammed,

H. O. (1991). Rapid antigenic-type replacement and DNA sequence evolution of canine parvovirus. J. Virol 65, 6544-6552.

Parrish, C. R., O'Connell, P. H., Evermann, J. F., and Carmichael, L. E. (1985). Natural variation

of canine parvovirus. Science 230, 1046-1048. Parrish, C. R. (2006). Pathogenesis of feline panleukopenia virus and canine parvovirus. In

Parvoviruses, J. R. Kerr, M. E. Bloom, R. Linden, and C. Parrish, eds. (New York: Oxford University Press Inc.), pp. 429-434.

Parrish, C., and Carmichael, L. (1983). Antigenic structure and variation of canine parvovirus

type-2, feline panleukopenia virus, and mink enteritis virus. Virology 129, 401-414. Patil, S. D., Rhodes, D. G., and Burgess, D. J. (2005). DNA-based therapeutics and DNA

delivery systems: A comprehensive review. AAPS J 7, E61-E77. Pear, W. S., Nolan, G. P., Scott, M. L., and Baltimore, D. (1993). Production of high-titer helper-

free retroviruses by transient transfection. Proc. Natl. Acad. Sci. U.S.A 90, 8392-8396. Peng, Z. (2005). Current status of gendicine in China: recombinant human Ad-p53 agent for

treatment of cancers. Hum. Gene Ther 16, 1016-1027.

Page 204: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

204

Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., and

Ferrin, T. E. (2004). UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem 25, 1605-1612.

Piechaczek, C., Fetzer, C., Baiker, A., Bode, J., and Lipps, H. J. (1999). A vector based on the

SV40 origin of replication and chromosomal S/MARs replicates episomally in CHO cells. Nucleic Acids Res 27, 426-428.

Porta, C., Wang, G., Cheng, H., Chen, Z., Baker, T. S., and Johnson, J. E. (1994). Direct Imaging

of Interactions between an Icosahedral Virus and Conjugate Fab Fragments by Cryoelectron Microscopy and X-Ray Crystallography. Virology 204, 777-788.

Prasad, B. V., Burns, J. W., Marietta, E., Estes, M. K., and Chiu, W. (1990). Localization of VP4

neutralization sites in rotavirus by three-dimensional cryo-electron microscopy. Nature 343, 476-479.

Pushko, P., Parker, M., Ludwig, G. V., Davis, N. L., Johnston, R. E., and Smith, J. F. (1997).

Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 239, 389-401.

Qing, K., Mah, C., Hansen, J., Zhou, S., Dwarki, V., and Srivastava, A. (1999). Human

fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated virus 2. Nat. Med 5, 71-77.

Qiu, J., and Brown, K. E. (1999). Integrin alphaVbeta5 is not involved in adeno-associated virus

type 2 (AAV2) infection. Virology 264, 436-440. Qu, X., Liu, W., Qi, Z., Duan, Z., Zheng, L., Kuang, Z., Zhang, W., and Hou, Y. (2008).

Phospholipase A2-like activity of human bocavirus VP1 unique region. Biochem. Biophys. Res. Commun 365, 158-163.

Rabinowitz, J. E., Xiao, W., and Samulski, R. J. (1999). Insertional mutagenesis of AAV2 capsid

and the production of recombinant virus. Virology 265, 274-285. Rabinowitz, J. E., Rolling, F., Li, C., Conrath, H., Xiao, W., Xiao, X., and Samulski, R. J.

(2002). Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity. J. Virol 76, 791-801.

Rapberger, R., Lukas, A., and Mayer, B. (2007). Identification of discontinuous antigenic

determinants on proteins based on shape complementarities. J. Mol. Recognit 20, 113-121.

Page 205: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

205

Reading, S. A., and Dimmock, N. J. (2007). Neutralization of animal virus infectivity by antibody. Arch. Virol 152, 1047-1059.

Reaves, P. Y., Gelband, C. H., Wang, H., Yang, H., Lu, D., Berecek, K. H., Katovich, M. J., and

Raizada, M. K. (1999). Permanent Cardiovascular Protection From Hypertension by the AT1 Receptor Antisense Gene Therapy in Hypertensive Rat Offspring. Circ Res 85, e44-50.

Rini, J. M., Schulze-Gahmen, U., and Wilson, I. A. (1992). Structural evidence for induced fit as

a mechanism for antibody-antigen recognition. Science 255, 959-965. Ros, C., Burckhardt, C. J., and Kempf, C. (2002). Cytoplasmic trafficking of minute virus of

mice: low-pH requirement, routing to late endosomes, and proteasome interaction. J. Virol 76, 12634-12645.

Ros, C., and Kempf, C. (2004). The ubiquitin-proteasome machinery is essential for nuclear

translocation of incoming minute virus of mice. Virology 324, 350-360. Rosenfeld, S. J., Yoshimoto, K., Kajigaya, S., Anderson, S., Young, N. S., Field, A., Warrener,

P., Bansal, G., and Collett, M. S. (1992). Unique region of the minor capsid protein of human parvovirus B19 is exposed on the virion surface. J. Clin. Invest 89, 2023-2029.

Rossmann, M. G. (2000). Fitting atomic models into electron-microscopy maps. Acta

Crystallogr. D Biol. Crystallogr 56, 1341-1349. Roy, R., Jerry, D. J., and Thayumanavan, S. (2009). Virus-Inspired Approach to Nonviral Gene

Delivery Vehicles. Biomacromolecules 10, 2189-2193. Ruigrok, R. W., Crépin, T., Hart, D. J., and Cusack, S. (2010). Towards an atomic resolution

understanding of the influenza virus replication machinery. Curr Opin Struct Biol. Available at: http://www.ncbi.nlm.nih.gov.lp.hscl.ufl.edu/pubmed/20061134 [Accessed January 28, 2010].

Russell, D. W., and Miller, A. D. (1996). Foamy virus vectors. J. Virol 70, 217-222. Samulski, R. J., Zhu, X., Xiao, X., Brook, J. D., Housman, D. E., Epstein, N., and Hunter, L. A.

(1991). Targeted integration of adeno-associated virus (AAV) into human chromosome 19. EMBO J 10, 3941-3950.

Sanders, D. A. (2002). No false start for novel pseudotyped vectors. Curr. Opin. Biotechnol 13,

437-442. Sarkar, R., Tetreault, R., Gao, G., Wang, L., Bell, P., Chandler, R., Wilson, J. M., and Kazazian,

H. H. (2004). Total correction of hemophilia A mice with canine FVIII using an AAV 8 serotype. Blood 103, 1253-1260.

Page 206: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

206

Sarkis, C., Philippe, S., Mallet, J., and Serguera, C. (2008). Non-integrating lentiviral vectors. Curr Gene Ther 8, 430-437.

Sato, H., Hirata, J., Kuroda, N., Shiraki, H., Maeda, Y., and Okochi, K. (1991). Identification

and mapping of neutralizing epitopes of human parvovirus B19 by using human antibodies. J. Virol 65, 5485-5490.

Scallan, C. D., Jiang, H., Liu, T., Patarroyo-White, S., Sommer, J. M., Zhou, S., Couto, L. B.,

and Pierce, G. F. (2006). Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice. Blood 107, 1810-1817.

Schiedner, G., Morral, N., Parks, R. J., Wu, Y., Koopmans, S. C., Langston, C., Graham, F. L.,

Beaudet, A. L., and Kochanek, S. (1998). Genomic DNA transfer with a high-capacity adenovirus vector results in improved in vivo gene expression and decreased toxicity. Nat. Genet 18, 180-183.

Schlesinger, S. (2001). Alphavirus vectors: development and potential therapeutic applications.

Expert Opin Biol Ther 1, 177-191. Schmaljohn, A. L., Johnson, E. D., Dalrymple, J. M., and Cole, G. A. (1982). Non-neutralizing

monoclonal antibodies can prevent lethal alphavirus encephalitis. Nature 297, 70-72. Schmidt, M., Grot, E., Cervenka, P., Wainer, S., Buck, C., and Chiorini, J. A. (2006).

Identification and characterization of novel adeno-associated virus isolates in ATCC virus stocks. J. Virol 80, 5082-5085.

Schmidt, M., Voutetakis, A., Afione, S., Zheng, C., Mandikian, D., and Chiorini, J. A. (2008).

Adeno-associated virus type 12 (AAV12): a novel AAV serotype with sialic acid- and heparan sulfate proteoglycan-independent transduction activity. J. Virol 82, 1399-1406.

Schneider, B., Fryer, J. F., Oldenburg, J., Brackmann, H., Baylis, S. A., and Eis-Hübinger, A. M.

(2008). Frequency of contamination of coagulation factor concentrates with novel human parvovirus PARV4. Haemophilia 14, 978-986.

Schoenbach, K. H., Beebe, S. J., and Buescher, E. S. (2001). Intracellular effect of ultrashort

electrical pulses. Bioelectromagnetics 22, 440-448. Seiler, M. P., Miller, A. D., Zabner, J., and Halbert, C. L. (2006). Adeno-associated virus types 5

and 6 use distinct receptors for cell entry. Hum. Gene Ther 17, 10-19. Shi, J., and Zheng, D. (2009). An update on gene therapy in China. Curr. Opin. Mol. Ther 11,

547-553.

Page 207: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

207

Shi, X., Fang, G., Shi, W., and Bartlett, J. S. (2006). Insertional mutagenesis at positions 520 and 584 of adeno-associated virus type 2 (AAV2) capsid gene and generation of AAV2 vectors with eliminated heparin- binding ability and introduced novel tropism. Hum. Gene Ther 17, 353-361.

de Silva, S., and Bowers, W. J. (2009). Herpes Virus Amplicon Vectors. Viruses 1, 594-629. Simonelli, F., Maguire, A. M., Testa, F., Pierce, E. A., Mingozzi, F., Bennicelli, J. L., Rossi, S.,

Marshall, K., Banfi, S., Surace, E. M., et al. (2009). Gene Therapy for Leber's Congenital Amaurosis is Safe and Effective Through 1.5 Years After Vector Administration. Mol. Ther. Available at: http://www.ncbi.nlm.nih.gov.lp.hscl.ufl.edu/pubmed/19953081 [Accessed January 19, 2010].

Simpson, A. A., Chipman, P. R., Baker, T. S., Tijssen, P., and Rossmann, M. G. (1998). The

structure of an insect parvovirus (Galleria mellonella densovirus) at 3.7 A resolution. Structure 6, 1355-1367.

Simpson, A. A., Hébert, B., Sullivan, G. M., Parrish, C. R., Zádori, Z., Tijssen, P., and

Rossmann, M. G. (2002). The structure of porcine parvovirus: comparison with related viruses. J. Mol. Biol 315, 1189-1198.

Singer, O., and Verma, I. M. (2008). Applications of lentiviral vectors for shRNA delivery and

transgenesis. Curr Gene Ther 8, 483-488. Sinn, P. L., Sauter, S. L., and McCray, P. B. (2005). Gene therapy progress and prospects:

development of improved lentiviral and retroviral vectors--design, biosafety, and production. Gene Ther 12, 1089-1098.

Smith, T. J. (2001). Antibody interactions with rhinovirus: lessons for mechanisms of

neutralization and the role of immunity in viral evolution. Curr. Top. Microbiol. Immunol 260, 1-28.

Smith, T. J. (2003). Structural studies on antibody-virus complexes. Adv. Protein Chem 64, 409-

453. Strassheim, M. L., Gruenberg, A., Veijalainen, P., Sgro, J. Y., and Parrish, C. R. (1994). Two

dominant neutralizing antigenic determinants of canine parvovirus are found on the threefold spike of the virus capsid. Virology 198, 175-184.

Subramaniam, S. (1998). The Biology Workbench--a seamless database and analysis

environment for the biologist. Proteins 32, 1-2. Suh, S. W., Bhat, T. N., Navia, M. A., Cohen, G. H., Rao, D. N., Rudikoff, S., and Davies, D. R.

(1986). The galactan-binding immunoglobulin Fab J539: an X-ray diffraction study at 2.6-A resolution. Proteins 1, 74-80.

Page 208: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

208

Suikkanen, S., Antila, M., Jaatinen, A., Vihinen-Ranta, M., and Vuento, M. (2003). Release of canine parvovirus from endocytic vesicles. Virology 316, 267-280.

Summerford, C., Bartlett, J. S., and Samulski, R. J. (1999). AlphaVbeta5 integrin: a co-receptor

for adeno-associated virus type 2 infection. Nat. Med 5, 78-82. Sun, B., Zhang, H., Franco, L. M., Young, S. P., Schneider, A., Bird, A., Amalfitano, A., Chen,

Y., and Koeberl, D. D. (2005). Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II. Mol. Ther 11, 57-65.

Sundararajan, R. (2009). Nanosecond electroporation: another look. Mol. Biotechnol 41, 69-82. Surosky, R. T., Urabe, M., Godwin, S. G., McQuiston, S. A., Kurtzman, G. J., Ozawa, K., and

Natsoulis, G. (1997). Adeno-associated virus Rep proteins target DNA sequences to a unique locus in the human genome. J Virol 71, 7951-7959.

Tachibana, M., Sparman, M., Sritanaudomchai, H., Ma, H., Clepper, L., Woodward, J., Li, Y.,

Ramsey, C., Kolotushkina, O., and Mitalipov, S. (2009). Mitochondrial gene replacement in primate offspring and embryonic stem cells. Nature 461, 367-372.

Taniyama, Y., Tachibana, K., Hiraoka, K., Aoki, M., Yamamoto, S., Matsumoto, K., Nakamura,

T., Ogihara, T., Kaneda, Y., and Morishita, R. (2002). Development of safe and efficient novel nonviral gene transfer using ultrasound: enhancement of transfection efficiency of naked plasmid DNA in skeletal muscle. Gene Ther 9, 372-380.

Tattersall, P. (2006). The evolution of parvovirus taxonomy. In Parvoviruses, J. R. Kerr, S.

Cotmore, M. E. Bloom, R. Linden, and C. R. Parrish, eds. (Hodder Arnold), pp. 5-14. Terrosi, C., Fabbiani, M., Cellesi, C., and Cusi, M. G. (2007). Human bocavirus detection in an

atopic child affected by pneumonia associated with wheezing. J. Clin. Virol 40, 43-45. The CCP4 suite: programs for protein crystallography (1994). Acta Crystallogr. D Biol.

Crystallogr 50, 760-763. The News Staff (2009). The Runners-Up. Science 326, 1600-1607. Thomas, C. E., Storm, T. A., Huang, Z., and Kay, M. A. (2004). Rapid uncoating of vector

genomes is the key to efficient liver transduction with pseudotyped adeno-associated virus vectors. J. Virol 78, 3110-3122.

Thompson, J. D., Higgins, D. G., and Gibson, T. J. (1994). CLUSTAL W: improving the

sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res 22, 4673-4680.

Page 209: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

209

Tijssen, P., Szelei, J., and Zádori, Z. (2006). Phospholipase A2 domains in structural proteins of parvoviruses. In Parvoviruses, J. Kerr, S. Cotmore, M. E. Bloom, R. Linden, and C. R. Parrish, eds. (New York, NY: Edward Arnold,Ltd.), pp. 95-105.

Tolfvenstam, T., Lundqvist, A., Levi, M., Wahren, B., and Broliden, K. (2000). Mapping of B-

cell epitopes on human Parvovirus B19 non-structural and structural proteins. Vaccine 19, 758-763.

Trobridge, G. D. (2009). Foamy virus vectors for gene transfer. Expert Opin Biol Ther 9, 1427-

1436. Valori, C. F., Ning, K., Wyles, M., and Azzouz, M. (2008). Development and applications of

non-HIV-based lentiviral vectors in neurological disorders. Curr Gene Ther 8, 406-418. Vandenberghe, L. H., Wilson, J. M., and Gao, G. (2009). Tailoring the AAV vector capsid for

gene therapy. Gene Ther 16, 311-319. Vandenberghe, L. H., and Wilson, J. M. (2007). AAV as an immunogen. Curr Gene Ther 7, 325-

333. Veldwijk, M. R., Topaly, J., Laufs, S., Hengge, U. R., Wenz, F., Zeller, W. J., and Fruehauf, S.

(2002). Development and optimization of a real-time quantitative PCR-based method for the titration of AAV-2 vector stocks. Mol. Ther 6, 272-278.

Vicente, D., Cilla, G., Montes, M., Pérez-Yarza, E. G., and Pérez-Trallero, E. (2007). Human

bocavirus, a respiratory and enteric virus. Emerging Infect. Dis 13, 636-637. Vihinen-Ranta, M., Wang, D., Weichert, W. S., and Parrish, C. R. (2002). The VP1 N-terminal

sequence of canine parvovirus affects nuclear transport of capsids and efficient cell infection. J. Virol 76, 1884-1891.

Voigt, K., Izsvák, Z., and Ivics, Z. (2008). Targeted gene insertion for molecular medicine. J.

Mol. Med 86, 1205-1219. Vorburger, S. A., and Hunt, K. K. (2002). Adenoviral Gene Therapy. Oncologist 7, 46-59. Walters, R. W., Yi, S. M., Keshavjee, S., Brown, K. E., Welsh, M. J., Chiorini, J. A., and Zabner,

J. (2001). Binding of adeno-associated virus type 5 to 2,3-linked sialic acid is required for gene transfer. J. Biol. Chem 276, 20610-20616.

Walters, R. W., Agbandje-McKenna, M., Bowman, V. D., Moninger, T. O., Olson, N. H., Seiler,

M., Chiorini, J. A., Baker, T. S., and Zabner, J. (2004). Structure of adeno-associated virus serotype 5. J. Virol 78, 3361-3371.

Walther, W., and Stein, U. (2000). Viral vectors for gene transfer: a review of their use in the

treatment of human diseases. Drugs 60, 249-271.

Page 210: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

210

Wang, G., Porta, C., Chen, Z., Baker, T. S., and Johnson, J. E. (1992). Identification of a Fab

interaction footprint site on an icosahedral virus by cryoelectron microscopy and X-ray crystallography. Nature 355, 275-278. Wang, L., Calcedo, R., Nichols, T. C., Bellinger, D. A., Dillow, A., Verma, I. M., and Wilson, J.

M. (2005). Sustained correction of disease in naive and AAV2-pretreated hemophilia B dogs: AAV2/8-mediated, liver-directed gene therapy. Blood 105, 3079-3086.

Wang, L., Calcedo, R., Wang, H., Bell, P., Grant, R., Vandenberghe, L. H., Sanmiguel, J.,

Morizono, H., Batshaw, M. L., and Wilson, J. M. (2010). The pleiotropic effects of natural AAV infections on liver-directed gene transfer in macaques. Mol. Ther 18, 126-134.

Wang, L., Figueredo, J., Calcedo, R., Lin, J., and Wilson, J. M. (2007). Cross-presentation of

adeno-associated virus serotype 2 capsids activates cytotoxic T cells but does not render hepatocytes effective cytolytic targets. Hum. Gene Ther 18, 185-194.

Wang, Z., Zhu, T., Qiao, C., Zhou, L., Wang, B., Zhang, J., Chen, C., Li, J., and Xiao, X. (2005).

Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat. Biotechnol 23, 321-328.

Warrington, K. H., and Herzog, R. W. (2006). Treatment of human disease by adeno-associated

viral gene transfer. Hum Genet 119, 571-603. Waters, B., and Lillicrap, D. (2009). The immunology of gene transfer: an overview. In Gene

Therapy Immunology, R. W. Herzog, ed. (New Jersey: Wiley-Blackwell), pp. 1-18. Weber, M., Rabinowitz, J., Provost, N., Conrath, H., Folliot, S., Briot, D., Chérel, Y., Chenuaud,

P., Samulski, J., Moullier, P., et al. (2003). Recombinant adeno-associated virus serotype 4 mediates unique and exclusive long-term transduction of retinal pigmented epithelium in rat, dog, and nonhuman primate after subretinal delivery. Mol. Ther 7, 774-781.

Weger, S., Wistuba, A., Grimm, D., and Kleinschmidt, J. A. (1997). Control of adeno-associated

virus type 2 cap gene expression: relative influence of helper virus, terminal repeats, and Rep proteins. J. Virol 71, 8437-8447.

Weitzman, M. (2006). The parvovirus life cycle: an introduction to molecular interactions

important for infection. In Parvoviruses, J. R. Kerr, M. E. Bloom, R. Linden, and C. R. Parrish, eds. (New York: Oxford University Press Inc.), pp. 143-56.

Wells, D. J. (2004). Gene Therapy Progress and Prospects: Electroporation and other physical

methods. Gene Ther 11, 1363-1369.

Page 211: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

211

Wikoff, W. R., Wang, G., Parrish, C. R., Cheng, R. H., Strassheim, M. L., Baker, T. S., and Rossmann, M. G. (1994b). The structure of a neutralized virus: canine parvovirus complexed with neutralizing antibody fragment. Structure 2, 595-607.

Wilson, J. M. (1996). Adenoviruses as gene-delivery vehicles. N. Engl. J. Med 334, 1185-1187. Wilson, J. M. (2009). Progress in the Commercial-Scale Production of Adeno-Associated Viral

Vectors. Human Gene Therapy 20, 695-695. Wistuba, A., Kern, A., Weger, S., Grimm, D., and Kleinschmidt, J. A. (1997). Subcellular

compartmentalization of adeno-associated virus type 2 assembly. J. Virol 71, 1341-1352. Wistuba, A., Weger, S., Kern, A., and Kleinschmidt, J. A. (1995). Intermediates of adeno-

associated virus type 2 assembly: identification of soluble complexes containing Rep and Cap proteins. J. Virol 69, 5311-5319.

Wolff, J. A., Malone, R. W., Williams, P., Chong, W., Acsadi, G., Jani, A., and Felgner, P. L.

(1990). Direct gene transfer into mouse muscle in vivo. Science 247, 1465-1468. Wu, H., Ceccarelli, D. F., and Frappier, L. (2000). The DNA segregation mechanism of Epstein-

Barr virus nuclear antigen 1. EMBO Rep 1, 140-144. Wu, P., Xiao, W., Conlon, T., Hughes, J., Agbandje-McKenna, M., Ferkol, T., Flotte, T., and

Muzyczka, N. (2000b). Mutational Analysis of the Adeno-Associated Virus Type 2 (AAV2) Capsid Gene and Construction of AAV2 Vectors with Altered Tropism. J Virol 74, 8635-8647.

Wu, Z., Asokan, A., and Samulski, R. J. (2006). Adeno-associated virus serotypes: vector toolkit

for human gene therapy. Mol. Ther 14, 316-327. Wu, Z., Miller, E., Agbandje-McKenna, M., and Samulski, R. J. (2006). Alpha2,3 and alpha2,6

N-linked sialic acids facilitate efficient binding and transduction by adeno-associated virus types 1 and 6. J. Virol 80, 9093-9103.

Xenariou, S., Liang, H., Griesenbach, U., Zhu, J., Farley, R., Somerton, L., Singh, C., Jeffery, P.

K., Scheule, R. K., Cheng, S. H., et al. (2010). Low-frequency ultrasound increases non-viral gene transfer to the mouse lung. Acta Biochim. Biophys. Sin. (Shanghai) 42, 45-51.

Xiao, W., Chirmule, N., Berta, S. C., McCullough, B., Gao, G., and Wilson, J. M. (1999). Gene

therapy vectors based on adeno-associated virus type 1. J. Virol 73, 3994-4003. Xie, Q., Bu, W., Bhatia, S., Hare, J., Somasundaram, T., Azzi, A., and Chapman, M. S. (2002).

The atomic structure of adeno-associated virus (AAV-2), a vector for human gene therapy. Proc. Natl. Acad. Sci. U.S.A 99, 10405-10410.

Page 212: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

212

Xiong, C., Levis, R., Shen, P., Schlesinger, S., Rice, C. M., and Huang, H. V. (1989). Sindbis virus: an efficient, broad host range vector for gene expression in animal cells. Science 243, 1188-1191.

Xu, J., Ma, C., Bass, C., and Terwilliger, E. F. (2005). A combination of mutations enhances the

neurotropism of AAV-2. Virology 341, 203-214. Yan, X., Dryden, K. A., Tang, J., and Baker, T. S. (2007). Ab initio random model method

facilitates 3D reconstruction of icosahedral particles. J. Struct. Biol 157, 211-225. Yan, X., Sinkovits, R. S., and Baker, T. S. (2007). AUTO3DEM--an automated and high

throughput program for image reconstruction of icosahedral particles. J. Struct. Biol 157, 73-82.

Yan, Z., Zak, R., Luxton, G. W. G., Ritchie, T. C., Bantel-Schaal, U., and Engelhardt, J. F.

(2002). Ubiquitination of both adeno-associated virus type 2 and 5 capsid proteins affects the transduction efficiency of recombinant vectors. J. Virol 76, 2043-2053.

Yan, Z., Zak, R., Zhang, Y., Ding, W., Godwin, S., Munson, K., Peluso, R., and Engelhardt, J. F.

(2004). Distinct classes of proteasome-modulating agents cooperatively augment recombinant adeno-associated virus type 2 and type 5-mediated transduction from the apical surfaces of human airway epithelia. J. Virol 78, 2863-2874.

Yang, L., Jiang, J., Drouin, L. M., Agbandje-McKenna, M., Chen, C., Qiao, C., Pu, D., Hu, X.,

Wang, D., Li, J., et al. (2009). A myocardium tropic adeno-associated virus (AAV) evolved by DNA shuffling and in vivo selection. Proc. Natl. Acad. Sci. U.S.A 106, 3946-3951.

Yang, Y., Li, Q., Ertl, H. C., and Wilson, J. M. (1995). Cellular and humoral immune responses

to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J. Virol 69, 2004-2015.

Yates, F., Malassis-Séris, M., Stockholm, D., Bouneaud, C., Larousserie, F., Noguiez-Hellin, P.,

Danos, O., Kohn, D. B., Fischer, A., de Villartay, J., et al. (2002). Gene therapy of RAG-2-/- mice: sustained correction of the immunodeficiency. Blood 100, 3942-3949.

Yates, V. J., el-Mishad, A. M., McCormick, K. J., and Trentin, J. J. (1973). Isolation and

characterization of an Avian adenovirus-associated virus. Infect. Immun 7, 973-980. Yoshimoto, K., Rosenfeld, S., Frickhofen, N., Kennedy, D., Hills, R., Kajigaya, S., and Young,

N. S. (1991). A second neutralizing epitope of B19 parvovirus implicates the spike region in the immune response. J. Virol 65, 7056-7060.

Yu, C., Yuan, Z., Cao, Z., Wang, B., Qiao, C., Li, J., and Xiao, X. (2009). A muscle-targeting

peptide displayed on AAV2 improves muscle tropism on systemic delivery. Gene Ther 16, 953-962.

Page 213: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

213

Zádori, Z., Szelei, J., Lacoste, M. C., Li, Y., Gariépy, S., Raymond, P., Allaire, M., Nabi, I. R.,

and Tijssen, P. (2001). A viral phospholipase A2 is required for parvovirus infectivity. Dev. Cell 1, 291-302.

Zaiss, A. K., and Muruve, D. A. (2008). Immunity to adeno-associated virus vectors in animals

and humans: a continued challenge. Gene Ther 15, 808-816. Zaiss, A. K., and Muruve, D. A. (2005). Immune responses to adeno-associated virus vectors.

Curr Gene Ther 5, 323-331. Zanta, M. A., Boussif, O., Adib, A., and Behr, J. P. (1997). In vitro gene delivery to hepatocytes

with galactosylated polyethylenimine. Bioconjug. Chem 8, 839-844. Zhang, H., High, K. A., Wu, Q., Yang, P., Schlachterman, A., Yu, S., Yi, N., Hsu, H., and

Mountz, J. D. (2005). Genetic analysis of the antibody response to AAV2 and factor IX. Mol. Ther 11, 866-874.

Zhang, X., Walker, S. B., Chipman, P. R., Nibert, M. L., and Baker, T. S. (2003). Reovirus

polymerase lambda 3 localized by cryo-electron microscopy of virions at a resolution of 7.6 A. Nat. Struct. Biol 10, 1011-1018.

Zhou, Z. H. (2008). Towards atomic resolution structural determination by single-particle cryo-

electron microscopy. Current Opinion in Structural Biology 18, 218-228. Zolotukhin, S., Byrne, B. J., Mason, E., Zolotukhin, I., Potter, M., Chesnut, K., Summerford, C.,

Samulski, R. J., and Muzyczka, N. (1999). Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield. Gene Ther 6, 973-985.

Zolotukhin, S., Potter, M., Hauswirth, W. W., Guy, J., and Muzyczka, N. (1996). A "humanized"

green fluorescent protein cDNA adapted for high-level expression in mammalian cells. J. Virol 70, 4646-4654.

Zolotukhin, S., Potter, M., Zolotukhin, I., Sakai, Y., Loiler, S., Fraites, T. J., Chiodo, V. A.,

Phillipsberg, T., Muzyczka, N., Hauswirth, W. W., et al. (2002). Production and purification of serotype 1, 2, and 5 recombinant adeno-associated viral vectors. Methods 28, 158-167.

Page 214: © 2010 Brittney L. Gurda · 1 mapping dominant antigenic regions on the capsid surfaces of adeno-associated virions by brittney lynn gurda a dissertation presented to the graduate

214

BIOGRAPHICAL SKETCH

Brittney Lynn Gurda was born in Hinsdale, Illinois to Thomas and Christine Gurda. Her

family moved from Chicago to Orlando, Florida in the spring of 1985. She has two siblings.

Senior to her is CaryBeth Bevan, who is married to Nathan Bevan. She has one nephew, Corbin

Bailey Stevens, and two nieces, Eden and Rowan Bevan. Her brother, Dean Thomas, is younger

only by 25 months. Like most kids with a love for her pets, Brittney’s original goal was to

become a veterinarian. She graduated from Dr. Philips High School in May 1996 and

immediately moved to New Smyrna Beach to begin work at Beachwood Animal Hospital under

Dr. Donald Needham, D.V.M. While acquiring experience in the veterinary setting she went to

school full time at Daytona Beach Community College. She returned to Orlando, Fl in the

summer of 1997 and continued her undergraduate education at Valencia Community College

while working part-time to fund her schooling. She eventually obtained her Associate in Arts

from VCC in 1999 and began the necessary requirements for a Bachelor in Science in

microbiology and molecular biology at the University of Central Florida. Wanting to eventually

transfer to the University of Florida for vet school, Brittney moved to Gainesville, Florida in

2003. While attending UF, Brittney began undergraduate research under the guidance of Dr.

Mavis Agbandje-McKenna, Ph.D. in the department of Biochemistry and Molecular Biology.

This experience opened her up to a whole new realm of medicine and after graduating in the

spring of 2004 from UF with a BS in microbiology and cell science she acquired a full-time

position with Dr. Agbandje-McKenna as a laboratory technician. This role eventually led to her

acceptance into the Interdisciplinary Program in Biomedical Sciences in the College of Medicine

in the fall of 2005 where she continued her research as a graduate assistant in the Agbandje-

McKenna laboratory.