fig :fi ,. 5,. : m, . new“. . ya; .. A i , "a s. e. . .. :: $4... .6 ... 9w. 5. . _ but? “an! :2. Kai‘s: d 2mm... 5 .21. .z . :1 a). 2...: 0.9.3.6 \A 1! 3.19.3 .3 .2... i 1:? 2 1133.513: 24.1.. 3:: ~§3§ snail 1.1.3. :3- “gufinn ‘ \ "”101. o 5.. 4. whim a. ,2 a ,t 53 _ LIBRARY *' * Mmhtgan State , .2 University This is to certify that the dissertation entitled EVALUATING AND MODIFYING ADENOVIRUS VECTOR INTERACTIONS WITH MULTIPLE ARMS OF THE INNATE IMMUNE SYSTEM presented by Sergey Seregin has been accepted towards fulfillment of the requirements for the PhD degree in Microbiology and Molecular Genetics ' Major Pi'ofessof’s Signature O ‘l' O 41 ~63 o l 0 Date MSU is an Affirmative Action/Equal Opportunity Employer -._ - -.--a-.—.--u-t-.-p- 0'-l-'-C-O-O-I-I-I-U-l-‘-O-O-O-I-C-.-I-D-I-C-I-‘-O-O-0-O-I-I-'-I-l-l-n->-.-— a. PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5’08 K:IProleoc&Pres/ClRC/Dat90m.hdd EVALUATING AND MODIFYING ADENOVIRUS VECTOR INTERACTIONS WITH MULTIPLE ARMS OF THE INNATE IMMUNE SYSTEM By Sergey Seregin A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Microbiology and Molecular Genetics 2010 ABSTRACT EVALUATING AND MODIFYING ADENOVIRUS VECTOR INTERACTIONS WITH MULTIPLE ARMS OF THE INNATE IMMUNE SYSTEM By Sergey Seregin A single, universally adaptable gene transfer vector cannot be envisioned for use in all human clinical gene transfer applications. However, Adenovirus (Ad) based vectors offer several important benefits compelling their potential for use in a wide range of gene transfer applications. Ad vectors can be readily produced to cGMP guidelines, which allowed their safe usage in thousands of clinical trial subjects. Unfortunately, upon contact with the circulatory system and/or cellular membranes, Ads induce several, innate, complement dependent toxicities that limit more widespread utilization of this promising platform. In addition, Ad derived transgene expression is greatly diminished when pre-existing Ad immunity is present in the host. These limitations have driven initiation of several distinct approaches to improve the safety/efficacy profiles of Ad-based vectors, including: the generation of chemically modified Ad capsids and/or chimeric Ads; the complete replacement of common Ad-based serotypes with alternative (human and non-human origin) Ad serotypes; genome modification of common (Ad5) serotype in attempts to improve the efficacy of the platform as well minimize acute innate responses to the vector itself. In this dissertation, I describe the pivotal role that the complement system plays in regulating Ad safety and efficacy. l unveil the mechanism underlying the complement dependent induction of neutralizing antibodies to Ad capsids as a C3 and CR1I2-dependent phenomenon that correlates with B-oell activation. Moreover, I have constructed several novel Ad5-based vectors, “capsid- displaying” complement regulatory peptide (COMPinh), as fiber or plX fusion proteins. These novel Ads dramatically minimize Ad-dependent activation of the human and non-human primate complement systems. We have also demonstrated that a simple, pre-emptive and transient glucocorticoid pre- treatment is a viable approach to reduce Ad associated acute toxicities, without reducing efficacy of Ad mediated gene transfer, suggesting that glucocorticoid therapy can be combined with the use of novel capsid-displaying Ads to further improve the outcome. Previous studies have demonstrated that several signaling pathways are triggered by Ads, inclusive of TLR dependent pathways. Here, I have unveiled an important role for the G-protein coupled receptor adaptors B-arrestin1 (B-Arr1) and fl-arrestinz (B-Arr2) in Ad5 vector-induced inflammatory responses, thereby identifying them as new potential targets in attempts to improve Ad vector safety/efficacy profile. Future studies will expand upon these findings. I leave readers with the view that utilizing a combination of the approaches, summarized herein will likely improve the capabilities of this important vector platform for expanded use in a number of additional human and agricultural applications. DEDICATION I would like to dedicate this dissertation to my parents. My parents, who are both scientists, are happy that one of their four children followed their path. I'm extremely grateful for their paramount and unbelievable support, unconditional love and care, throughout all my educational steps. My parents live in Russia and I miss them very much — l have not seen my Father for over four years. I’m happy that upon Graduation I will finally have an opportunity to visit them in summer 2010. I would like to express my love to my beautiful sisters, Dina and Maria and my brother Alexander. I love my family so much, and I would not have made it this far without their support. iv ACKNOWLEDGEMENTS I would like to take the opportunity to thank everyone who contributed to my development as an independent investigator — the path I continue to follow to become one. First and foremost, I express my profound gratitude to my mentor Dr. Andrea Amalfitano, a leading researcher in a field of Adenovirus-based gene transfer. He kindly allowed me to join his newly formed lab, here at Michigan State University in early 2006, and since then constantly supported and encouraged me to develop scientific thinking, writing skills, critical data analysis and planning of experiments. His sincere guideless, patience and continuous support created, in my opinion, productive and positive atmosphere in our lab. Dr. Amalfitano has a unique ability to sense a “golden middle” in supervising graduate students, allowing them to reach an answer I accomplish a task by themselves and only when person is puzzled, he provides precious scientific advice. This “smart guideless” takes a lot of his time, patience and effort, and I’m extremely happy, Dr. Amalfitano utilized this approach and feel that l have greatly improved all my skills, associated with future scientific career. Dr. Amalfitano is my best role model for a scientist, scientific advisor, teacher and person. It is my privilege to have an opportunity to work with him. In my future career I will be trying to preserve and elaborate upon these important personal characteristics. I greatly appreciate the help provided by my guidance committee: Dr. Michele M. Fluck, Dr. Robert A. Roth and Dr. Ian York. Their insightful comments and constructive discussions have significantly improved my research progress as well as development of critical scientific thinking. My sincere thanks to all current and former members of Dr. Amalfitano lab: Dan Appledom, Yasser Aldhamen, Sarah Godbehere Roosa, Jeannine Scott, Nathaniel Schuldt, Aaron McBride, Tyler Voss, Joyce Liu, Dionisia Quiroga, William Nance, Mathew Bugold, William De Pas, Megan Hoban, Jenny Zehnder, Brandy Burke, Anthony Sisk, Ryan Stringer. All of them are genuinely nice and eager to help each other, and I’m glad, I have worked and interacted with them. Together we have created wonderful positive friendly atmosphere in a lab, an environment with a space for fruitful scientific and science unrelated discussions, productive team work and, of course, fun and jokes! I will forever be thankful to Jeannine Scott, who contributed greatly in my initial training here at MSU, she was the one who taught me how to design, construct, propagate and purify high titer Adenovirus vectors as well as other numerous techniques. She also provided advices many times during my graduate school career. I would like to give my particularly special thanks to the most extraordinary person (in all aspects), I have ever met in my life - Dan Appledom. He possesses amazing skills in countless science related issues, he is enthusiastic, and energetic and definitely one of the smartest people I know. He is my primary resource for getting my science questions answered and was instrumental in helping me to proofread this thesis. We had countless fruitful discussions, which helped me greatly to improve my thinking skills. I know that I could always ask him vi for advice and opinion on any issue. He is a wonderful person, and I admire his positive outlook and his ability to smile despite the situation. What makes Dan even more unique is together with all this great scientific thinking, he is the main source of fun and jokes, creating positive environment in our lab. It is very critical to get some good laughs in a tough, 12-hour day. I'm sure that Dan will soon become great and productive Pl. I also thank my friend and co-worker, Yasser Aldhamen, who is a wonderful person. It is a pleasure to work with him, in particular as a team on some projects. He is honest, smart and responsible; he has unique ability to dig out foundation for future experiments or projects, the skill, I tried to improve, as it is critical piece during my future independent career. My special thanks to Sarah Godbehere Roosa, who is a wonderful technician and exceptionally pleasant person to work with. If not for all Sarah’s help, my work would have taken me twice as long. I apologize that I can’t individually thank all members of Dr. Amalfitano lab, as this dissertation should contain at least a little bit of science. My sincere thanks go to our collaborators from Duke University Medical Center: Dr. Michael Frank, Zachary Hartman, Anne Kiang, Junping Wei, Delila Serra, Fang Xu, Xiao Yi Zhao, Hai Xiang Jiang. We had several papers published together and their contribution was significant, especially in complement assays. Anne and Zack are former Dr. Amalfitano Graduate students, who had started several projects we continued to develop. In particular, I have finished several projects, Zack has initiated back in 2003. Despite the fact that I never met him vii personally, we have exchanged numerous e-mails and he helped me greatly during my research and I know that he a great person. I would like to emphasize my paramount gratitude to Adenovirus - the best and the most pragmatic vector platform for numerous future gene transfer applications and all people who contributed in discovery of Ads and who started to design Ad-based vectors. Thank you! Special thanks to all wonderful MSU core facilities, including: ULAR and, in particular, all employees of BPS/biochemistry animal housing facility; MSU Histopathology labs (Amy S. Porter, Kathy A. Joseph, Rick A. Roseburry); MSU electron microscopy facility (Ralph Common); MSU Genomics Technology Support Facility: Flow Cytometry (Louis King), quantitative RT—PCR (Jeff Landgraf), sequencing (Christi Hemming), Proteomics (Douglas Whitten). Without their great performance and skilled assistance our research would not have been possible. I’m thankful to all faculty members and employees of the Department of Microbiology and Molecular Genetics, especially the Chair of the department (Dr. Walter J. Esselman), the Director of MMG Graduate program (Dr. Robert Hausinger) and Graduate Secretary (Suzanne Peacock) for their kindness and all their help and advises, provided during these years and their patience, when they had to deal with my numerous questions. I would like to give my special thanks to Administrative Assistant Coreena K. Spitzley for her countless assistance in issues associated with lntemational student status, health insurance, and Grants viii submission. She is the best and the smartest administrative employee, I have ever worked with. American Heart Association is one of the few agencies, which accept applications from international applicants. I’m extremely thankful to AHA for their policy and support, which they provided to me as 2-year pre-doctoral fellowship. My special warm thanks for my Aunt Julia Busik, who helped us a lot during all these years, especially during first year and upon arriving to MSU: new culture, new country, with 11-month old child. Our first couple of months were extremely tough and I don’t know how we would of got by if not her constant and unconditional help. I’m thankful to her husband Denis, who used to be semi- professional downhill skier, for teaching us how to ski and enjoy the slopes. I thank with all my heart to my parents for their encouragements, love and support which motivated me to accomplish all my educational aims. My special thanks go to my dear wife, Maria Tikhonenko and my wonderful daughter Anastasia, who have been a source of motivation and inspiration during all those tough moments. Maria has been a true and great supporter; she has unconditionally loved me during my good and bad times. I feel her support and the faith she has in me and I will continue to give her my absolute support and love to provide her with patience and power to complete her Graduate Studies. I feel that we became much wiser and calmer after these years; life has strengthened our commitment and determination to our family as a whole. ix TABLE OF CONTENTS LIST OF TABLES ............................................................................... xii LIST OF FIGURES ............................................................................ xiii List of Abbreviations .......................................................................... xvi Chapter 1 Introduction .................................................................... 1 1.1 Ad-based vectors: the most pragmatic vectors for gene transfer applications .......................................................... 2 1.2 Primary problem: Ad-triggered innate immune responses ......... 7 1.2.1 Toll-like receptors and Adenovirus ....................................... 8 1.2.2 The complement system: first line of defense against pathogens .................................................................................. 11 1.2.3 Complement and Adenovirus ............................................ 17 1.3 Approaches directed to minimize Ad-triggered immune responses ....................................................................... 19 1.3.1 Ad capsid modifications ................................................... 21 1.3.1.1 Chemical modifications of the Ad capsid ............................. 21 1.3.1.2 Insertion of novel peptides into Ad capsid proteins ............... 22 1.3.2 lmmunosuppressive glucocorticoid Dexamethasone ............. 25 Chapter 2 Ads-triggered signaling pathways: critical roles of BArr-1 and pArr-Z adaptor proteins ............................................ 29 2.1 Introduction .................................................................... 30 2.2 Results ........................................................................ 32 2.3 Discussion .................................................................... 57 Chapter 3. Ad interactions with the complement system are pivotal in understanding how to maximize the safety and potency of Ad mediated gene transfer for both gene therapy and vaccine applications ................................................................ 61 3.1 Introduction ................................................................... 62 3.2 Results ......................................................................... 65 3.3 Discussion .................................................................... 94 Chapter 4. Ads-based vectors “capsid-displaying” specific complement inhibitor: a novel approach to improve Ad vector safety profile ..................................................... 102 4.1 Introduction ................................................................. 103 4.2 Results and Discussion .................................................. 106 Chapter 5. Simple, pre-emptive and transient glucocorticoid pre-treatment reduces Ad5-associated acute toxicities ..... 123 5.1 Introduction ................................................................. 124 5.2 5.3 Chapter 6. 6.1. 6.1.1. 6.1.2. 6.2. 6.3. 6.4. 6.4.1. 6.4.2. 6.5. 6.6. 6.7. 6.8. 6.9. 6.10. 6.11. 6.12. 6.13. 6.14. 6.15. 6.16. 6.17. 6.18. 6.19. 6.20. 6.21. 6.22. 6.23. 6.24. Chapter 7. References. Results ....................................................................... 127 Discussion .................................................................. 162 Material and Methods ................................................... 170 Adenovirus vector construction ....................................... 171 Incorporation of COMPinh in HI loop of the fiber protein ........ 171 Incorporation of COMPinh in the C-terminus of protein IX ...... 171 Adenovirus vector production and characterization ............. 172 Electron Microscopy of purified Ad vectors ......................... 175 Validation of VP titers of Ads ........................................... 175 Silver Staining .............................................................. 176 Western Blotting ........................................................... 177 Capsid thermostability assay ............................................ 178 Animal procedures ........................................................ 180 CytokinelChemokine/Endothelial cells activation markers release measurement .................................................... 180 Cytokine quantification in human PBMCs ........................... 181 Complete blood count analysis, cell type differentiation ......... 181 Platelet enumeration ...................................................... 181 Ad genome copy number per liver or spleen cell ................. 182 B-Galactosidase enzyme activity, in situ X-gal staining.........182 qRT-PCR Analysis ........................................................ 183 Kupffer cell staining ...................................................... 184 Hematoxilin and Eosin staining ....................................... 185 IFNv secretion assay (Flow Cytometry) ............................. 189 B cell activation assay (Flow Cytometry) ........................... 190 Antibody Titering Assay .................................................. 191 Neutralizing Antibody Assay .......................................... 191 Western blotting ............................................................ 192 Peritoneal macrophages isolation and infection .................. 193 Complement activation AP50 serum-based assay ............... 193 C3a-desArg ELISA and CH50 assay ................................. 194 Statistical analysis ......................................................... 195 Summary and future perspectives .................................. 197 .................................................................................. 203 xi Table 1 Table 2 Table 3 Table 4 Table 5 Table 6 Table 7 Table 8 Table 9 LIST OF TABLES B-Arr1-KO mice had significantly reduced Ad5-triggered activation of a number of genes in livers, as compared to WT mice ........................................................................ 50 B-Arr1-KO mice had similar levels of Ad5-triggered activation of genes in spleens, as compared to WT mice ......................... 52 B-Arr2-KO mice had significantly higher levels of Ad5-triggered activation of a number of genes in livers, as compared to WT mice .......................................... 54 B—Arr2-KO mice had significantly higher levels of Ad5-triggered activation of a number of genes in spleens, as compared to WT mice ...................................... 56 mCR1I2 is an important suppressor of Ad5-LacZ induced Gene expression in livers of C57BU6 mice ............................. 75 Novel “capsid-displaying” Ads can be propagated to high titers, similar to conventional Ad vectors. Capsid modifications do not impair infectivity and transduction efficiency of novel Ads ......... 112 Dexamethasone pre—treatment abrogates Ad5-LacZ induced gene expression in livers of 057BL/6 mice ........................... 138 Complete list of primers and oligos, utilized to construct and validate Ad5-based vectors ............................. 174 List of primers, utilized in qRT-PCR experiments .................... 187 xii Figure 1 Figure 2 Figure 3 Figure 4 Figure 5 Figure 6 Figure 7 Figure 8 Figure 9 Figure 10 Figure 11 Figure 12 Figure 13 LIST OF FIGURES Images in this dissertation are presented in color Diagram of Adenovirus and its genome ..................................... 5 The complement system ...................................................... 16 Current approaches to design improved Adenovirus based gene transfer vectors ........................................................... 28 Functional B-arrestin-1 acts as positive regulator for several chemokines released in response to Ad infection in vitro ............ 34 Functional B—arrestin-Z acts as negative regulator for several chemokines released in response to Ad infection in vitro. ................................................................................................ 36 Functional B—arrestin-1 acts as positive regulator of Ad mediated systemic cytokine and chemokine release in 057BLI6 mice .................................................................... 39 Functional B-arrestin-Z acts as potent suppressor of Ad mediated systemic cytokine and chemokine release in 057BLI6 mice ..................................................................... 41 Functional B-arrestin-1 and B-arrestin-Z have modest impact upon liver and spleen transduction efficiency by Ad5 in 057BU6 mice .................................................................... 44 Functional B-arrestin-1 and B—arrestin-Z have modest impact upon Ad derived transgene expression in both liver and spleen tissues in CS7BL16 mice ............................................. 46 Plasma basal levels and Ad dependent activation of complement protein C3 were identical between C57BU6 WT and CR1l2-KO mice ............................................................. 67 Murine Complement Receptor 1/2 mitigates Ad mediated cytokine and chemokine release in C57BU6 mice ..................... 70 Murine Complement Receptor 1/2 reduces Ad dependent activation of endothelial cells in C57BU6 mice .......................... 73 mCR112-KO mice exhibit significantly reduced leukocyte infiltration into the liver of Ad treated mice at 28 dpi .................... 78 xiii Figure 14 Figure 15 Figure 16 Figure 17 Figure 18 Figure 19 Figure 20 Figure 21 Figure 22 Figure 23 Figure 24 Figure 25 Figure 26 Figure 27 The efficacy of Ad transduction of the liver of C57BU6 mice is not dependent upon murine Complement Receptor 112, but the levels of Ad derived transgene expression are complement and mCR112-dependent ...................................... 82 mCR1I2-KO mice exhibit significantly reduced Ad vector capsid specific humoral immune responses ................................. 85 mCR1I2-KO mice exhibit significantly reduced Ad capsid specific neutralizing antibodies titer .......................................... 88 mCR1I2 and C3-KO mice have impaired B cell activation in response to systemic Ad injection .......................................... 90 mCR1I2-KO mice have minimally reduced Ad vector derived transgene specific humoral immune responses as compared to WT mice ...................................................... 93 mCR112 protein plays a significant role in down-regulation of Ad mediated complement dependent pro-inflammatory cytokines production ............................................................ 96 Schematic diagram of all Ad vectors constructed and utilized in ourstudy.........‘ ............................................................. 108 Electron microscopy of purified Ad5 vectors ............................. 110 Novel COMPinh displaying Ads minimize Ad mediated complement activation in several serum-based assays .............. 115 Silver staining of purified Ad vectors revealed marginal differences in spectrophotometry determined vp titer .................. 118 Novel COMPinh displaying Ads reduce Ad-triggered activation of pro-inflammatory cytokines and chemokines in PBMCs... ......121 Dexamethasone (DEX) blocks Ad-mediated systemic cytokine and chemokine release in C57BU6 mice in a dose dependent manner ........................................................................... 129 Dexamethasone prevents Ad mediated thrombocytopenia in 057BU6 mice ................................................................... 133 Dexamethasone minimizes Ad dependent activation of endothelial cells in CS7BL/6 mice ......................................... 135 xiv Figure 28 Figure 29 Figure 30 Figure 31 Figure 32 Figure 33 Figure 34 Figure 35 Figure 36 Dexamethasone treatment causes an increase in lymphopenia and neutrophilia in a blood of C57BU6 mice, which is NOT related to Ad treatment ........................................................ 143 Dexamethasone treatment preserves the efficacy of Ad derived transgene expression and Ads genomes persistency in the livers of 057BL/6 mice ................................ 145 Dexamethasone treatment does not change Ad dependent Kupfer cells degradation in a liver of C57BU6 mice .................. 147 Dexamethasone treatment results in significantly reduced Ad mediated leukocytes infiltration to the liver of C57BU6 mice at 28 dpi ................................................................... 150 Dexamethasone treatment significantly reduced Ad vector capsid specific humoral immune responses, including capsid- neutralizing antibodies ....................................................... 153 Dexamethasone treatment significantly reduced Ad vector derived transgene specific humoral immune responses ............ 156 Dexamethasone treatment does not change natural levels of total non-specific lgG antibodies in a blood of 057BLI6 mice ...... 158 Dexamethasone treatment does not change Ad dependent 008 positive T cells activation ............................................. 161 Dexamethasone pre-treatment mediates blockage of Ad-induced innate immune responses: model of action ............. 166 XV Ad ADAR ALT ANOVA AP ATCC B-Arr-‘I , B-Arr-Z CAR C3 C3a-desArg CMV COMPinh CR CRAD CTL ‘ CXCL-9 DAF DEX DNA dpi EC LIST OF ABBREVATIONS Adena-associated virus Adenovirus Adenosine deaminase-RNA-specific (lFN-inducible) Alanine aminotransferase Analysis of variance Alternative complement pathway American type culture collection beta-Arrestins 1 and 2 Coxsackie and adenovirus receptor Complement component 3 stable protein, produced upon complement activation Cytomegalovirus peptide with ability to inhibit human complement Complement receptor Conditionally replicative adenoviruses Cytotoxic T lymphocyte Chemokine, induced by IFNy Decay accelerating factor Dexamethasone Deoxyribonucleic acid Days post injection Endothelial cells xvi EDTA EGTA EM fD, fI-l, fl FDAd GAPDH GFP G-CSF GM-CSF FDA HDAd HVR hpi lCAM-1 IFNa, IFNB I9 lL-6, IL-12p40 IRF3, IRF7, IRF8 JAK-1, JAK-3 KC (CXCL-1) KO LPS Ethylene-diamine-tetra-acetic acid Ethylene-glycol-tetra-acetic acid Electron microscopy Factor D, Factor H, Factor I (complement) Fully deleted adenovirus Glyceraldehyde 3-phosphate dehydrogenase Green fluorescent protein Granulocyte colony-stimulating factor Granulocyte-macrophage colony-stimulating factor Food and drug administration Helper-dependent adenovirus Hypervariable region of Ad hexon protein Hours post injection Inter-Cellular adhesion molecule 1 lnterferons a and B (type I IF Ns) Immunoglobulin Interleukins 6 and 12 (pro-inflammatory cytokines) Interferon Regulatory Factors 3, 7 and 8 Janus kinases 1 and 3 Keratinocyte derived chemokine, murine analog of human lL-8 Knockout Lipopolysaccharides xvii LacZ ml mM mg pl till #9 MAC MAPK MBL MOP-1 (CCL-2) MlP-1 p (COL-4) My088 NAb NFKB NHS NHPS NOD-1, NOD-2 OAS-1a OD qRT-PCR SPAMP PBMC Bacterial B-galactosidase Milliliter Milli-molar Milligram Micro-liter Micro-molar Microgram Membrane attack complex Mitogen-activated protein kinase Mannose Iectin binding pathway (complement) Monocyte chemotactic protein 1 Macrophage inflammatory protein 1 beta Myeloid differentiation factor 88 (T LR adaptor) Neutralizing antibody Nuclear factor kappa B Normal human serum Non-human primate serum Nucleotide-binding oligomerization domains 1 and 2 2'-5' Oligoadenylate synthetase (IF N-inducible) Optical density Reverse transcription and quantitative real-time PCR Pathogen-associated molecular pattern Peripheral blood mononuclear cells xviii PBS PCR PEG PTA PRR RANTES RNA SD 8003-1, 8005-3 TCID TBK-1 TLR TNFa TRAFpr TRAF6 TRIF VCAM-1 VP Phosphate buffer saline Polymerase chain reaction Polyethylene glycol Phosphotungstic acid (for EM staining) Pattern recognition receptor Normal T-cell Expressed, and Secreted Ribonucleic acid Standard deviation Suppressors of cytokine signaling 1 and 3 Tissue culture infectious dose TANK-binding kinase (T BK) Toll-Like Receptor Tumor necrosis factor alpha TNF receptor associated factor 2 binding protein TNF receptor associated factor 6 Toll/lnterleukin-1 receptor (TlR)-domain-containing adaptor-inducing interferon-[3 Vascular cell adhesion molecule 1 VIral particle WIld-type xix Chapter I Introduction 1.1. Adenovirus (Ad) based vectors: the most pragmatic vectors for gene transfer applications Ads are non-enveloped icosahedral viruses of 60-100 nm in diameter, that contain an ~36 kb double-stranded linear DNA genome. The human Adenovin'dae family is comprised of more than 50 Ad serotypes (based primarily upon the lack of cross reacting antibody neutralization between serotypes), which are categorized into six subgroups (A-F), primarily based upon different red blood cell agglutinating capabilities of the various subgroups. Classification of Ad serotypes have been historically based upon biological, biochemical as well as structural properties, including the lack of cross-neutralization between serotypes from different species. Subsequently, such parameters as oncogenicity, length of fiber protein and genomic sequence similarity were taken into consideration to further sub-classify Ads (1, 2). Ads have a worldwide distribution and account for ~5% of all respiratory infections in childhood (3). The viral capsid contains total of eleven structural proteins, of which hexon (protein ll), penton base (protein III) and fiber (protein IV) are termed major capsid proteins, and proteins llla, VI, VIII and IX minor capsid proteins, four additional proteins are packaged with the DNA inside the core (4-6). The capsid proteins hexon, and fiber are the major targets of Ad specific antibodies, many of which become neutralizing (7-12). Transcription and processing of Ad RNA occurs on both strands utilizing early (E) and late (L) regions, resulting, mainly by mechanism of alternative splicing, in production of over 30 non-structural proteins (13) (Figure 1). Early region (E1- E4), is transcribed and translated first, and resultant proteins assist is subsequent stages of Ad replication and packaging: E1a (immediate early gene), encodes transcription factor necessary for activation of early genes; E1b - encodes protein, blocking apoptosis, as well together with E1a, E3 and E4 products modulate cellular transcriptional machinery to transcribe predominantly viral genes and evade host immune responses; E2a - encodes DNA binding protein, E2b - encodes pretenninal protein and viral polymerase. Late region (L1-L5) predominantly encode structural proteins, necessary for virion assembly, for example, L2 region encodes core proteins V, VII and X as well as penton base (plll), L5 encodes fiber protein (2, 14). Several generations of recombinant Ad vector has been generated. First generation Ad vectors lack E1 region ([E1-] Ad) or E1 and portion of E3 ([E1-, E3-] Ad). These Ad vectors are specifically propagated on transcomplementing cell lines such as HEK293 or PER.C6, which supply the essential E1a and E1b proteins in trans (2, 14). Advanced generation vectors encompassing additional deletions in the E2b or E4 regions (as well as fully deleted “gutless” Ads) have also been developed. These vectors retain the capabilities of large-scale production noted with E1 deleted vectors. However, transcomplementing cell lines unique to each vector (or an additional helper virus) must also be provided to support their growth (15, 16). Recombinant, Ad-based gene transfer vector platforms are heavily utilized in both gene therapy and vaccine applications. This utility is based on a number of positive attributes inherent to the use of recombinant Ads. Recombinant Ads are capable of transducing foreign genes into a wide range of cell types inclusive Figure 1: Diagram of Adenovirus and its genome. (A) Adenoviruses are icosahedral, non-enveloped viruses that contain multiple features including a double stranded DNA genome, the fiber protein that terminates in the knob region (required for binding to the coxsackie adenovirus receptor (CAR)), and the penton base (required for facilitating viral entry). (B) Adenoviruses contain a ~36 kb genome generally organized into regions encoding. the Early (E) expressed genes and Late (L) genes. Various regions of the Ad genome can be removed and replaced with an expression cassette containing a “gene of choice" and its associated promoter and enhancer elements. Important regions of the Ad genome that have been removed to improve the persistence and/or safety of the vector are indicated with dotted arrows. These viruses can be grown to high viral titers using cell lines expressing the proteins encoded by these regions in trans. Knob Fiber ....r.s.1..., LJ,_L£,J£, estate, fees" *5? "'EZI'" I I I I l I l J 10,000 bp 20,000 bp 30,000 bp of antigen presenting cells; allow for efficient transgene expression, have large cloning capacities (8-36 Kb); and can be easily produced to extremely high titers in a current good manufacturing practices (cGMP) compliant fashion (up to 1x1013 vplml). The ability to easily “scale-up” traditional Ad vectors in a manner that is cGMP compliant, has resulted in thousands of patients safely receiving recombinant, Ad5-based gene transfer vectors. In fact, it is the author’s opinion that the proven ability to mass-produce Ad-based vectors makes them one of the most pragmatic of all gene transfer vectors currently proposed for direct in vivo human use. For example, the lack of simple, efficient, and large-scale cGMP production capabilities has limited the clinical utilization of a number of other proposed gene transfer platforms (i.e.: Adena-associated virus (AAV) or lentivirus based systems) to a handful of human clinical trials (see: http://www.wiley.co.uklwileychilgenmed/clinicall). There are, however, several limitations to the use of recombinant Ad vectors as a gene transfer platform, primarily: the decreased efficacy of these platforms in Ad-immune individuals, and vector-associated innate immunogenic toxicities. Many in the lay press and, in fact, the scientific theater wrongly hold the view that Ad-based clinical trials are no longer being conducted or pursued due to these important issues. Thus, it is very important to note the fact that Ad5- based vectors are currently the most widely used gene transfer vector in human clinical trials (http:/lwww.wiley.co.uk/wileychi/genmed/clinicall). More recently, a major advantage to the use of Ad-based vectors has also become apparent, with the confirmation that integrating viral vectors (i.e.: retrovirus, lentivirus, possibly AAVs) can cause insertional mutagenesis and cancer in animal models and in human clinical trial subjects, therefore such vectors may subject human recipients to similar and/or additional risks (17-21). The natural biology of the recombinant Ad genome to not integrate into the host chromosome as a nuclear episome largely mitigates insertional mutagenesis risks (22). 1.2. Primary problem: Ad-triggered innate immune responses The main problem with using Ad vectors in current applications is the frequent need for use of high doses of the vector to achieve successful evidence of gene transfer in several specific tissues, a problem that increases unwanted side effects, such as vector-associated innate immune responses (23-28). Importantly, these Ad-triggered responses are clinically relevant, unfortunately as primarily noted in the Ornithine transcarbamylase gene transfer tragedy (29). In that trial, the tragic death of a patient, who received 6x1011 vplkg of an Ad vector, coincided with induction of a “cytokine storm” — a massive release of pro- inflammatory cytokines (including IL-6), causing a systemic inflammatory response syndrome. Although not replicated in other patients receiving similar doses of the vector (both in this trial and in other subsequent studies (29-31), the trial highlighted the need for greater study of Ad vector induced innate immune responses. The lack of efficient vector platforms for systemic targeting of the liver has primarily driven the need more detailed investigation of Ad vector interactions with multiple arms of the innate immune system in attempts to find a way of improving the safety profile of this promising vector in these sorts of applications. It is now known that Ad vectors elicit multiple innate immune responses after systemic administration due to several processes: complement system activation, anaphylotoxins release, macrophage activation, cytokine/chemokine release, induction of granulocyte and mast cells infiltration, endothelial cell activation, generalized transcriptome dysregulation in multiple tissues, and thrombocytopenia (23-28). Several of the innate immune systems, known to interact with Ads are described in greater detail below. 1.2.1. Toll-like receptors and Adenovirus In the late eighteen hundreds, the famous Russian microbiologist llya llyich Mechnikov, Nobel prize laureate, described phagocytosis and suggested the term “innate immunity” to define an initial, nonspecific defensive mechanism that was present for defense against newly encountered pathogens. Almost a whole century passed before the modern scientific community discovered that pathogen recognition receptors (PRRs) function to recognize microbes and viruses at both the intra- and extracellular levels, triggering the induction of multiple innate immune responses. PRRs are able to recognize pathogen-associated molecular patterns (PAMPs), which are expressed on foreign agents. Bacterial Iipopolysaccharides (LPS), viral/bacterial nucleic acids, lipoproteins are all examples of PAMPs. Since these molecules are absent in host cells, they are recognized as non-self motifs by PRRs. The most studied PRRs include 82 integrins (CD11/CDIB), Nucleotide Oligomerization Domains (NODs), complement receptors (CR1ICD35, CR210021), RIG-I-like receptors and Toll- Like Receptors (T LRs). TLRs were discovered in late 1990s, giving rise to tremendous amount of studies, which shed light on TLRs signaling mechanisms associated with the innate (and downstream adaptive) immune responses (32, 33). TLRs are expressed in many cell types: non-hematopoietic epithelial and endothelial cells, macrophages, neutrophils and dendritic cells; 13 mammalian TLRs (10 in humans) have been identified to date. TLRs predominantly utilize conserved leucine rich repeat (LRR) at the N-terrnini for pathogen recognition, although amino acid insertions and non-consensus LRRs remarkably diversify TLR’s ability to recognize a wide range of PAMPs (34). Ad vectors have been confirmed to activate multiple PRRs both in vitro and in vivo, including RIG-I-like receptors, TLR2, TLR4 and TLR9 (for more detailed review please see (25)). These activations ultimately results in Ad-triggered innate toxicities. Specifically, we have shown that acute innate (and downstream adaptive) immune responses developed following systemic injection of Ad vectors, are significantly mediated by TLR adaptor proteins MyDBB and TRIF, as shRNA-mediated stable knockdown of these molecules resulted in diminished pro-inflammatory cytokine (IL-6) releases in vitro in response to Ad injection (35); moreover, cytokine/chemokine activation (G-CSF, IL-5, lL-6, lL-12p40, MOP-1, RANTES) and massive, pro-inflammatory genes induction was blunted in MyDBB knockout mice, as compared to identically Ad-treated WT mice (23). Ad-triggered activation of MAPK and NFkB signaling pathways, systemic cytokine release and generation of Ad-capsid specific neutralizing antibodies (NAb) are at least partially TLR2/TLR9/MyD88 dependent (36). Therefore, it is confirmed that TLRs play a pivotal role in mediating Ad-triggered innate immune responses both in vitro (35, 37, 38) and in vivo (38, 39), however, continued investigations of Ad- induwd innate immune responses and specific signaling pathways are required. B—Arrestins 1 and 2 (B—Arr—1 and -2) are ubiquitously expressed G-protein coupled receptor adaptors, known to have pivotal roles in regulating TLR signaling pathways (40). B-Arr-1 and -2 were originally discovered to play a major role in G protein coupled receptor desensitization, due to their ability to bind to phosphorylated G protein coupled receptors and sterically block their ability to signal downstream (40). Subsequent studies revealed more versatile roles of B- Arr1 (arrestin-2) and B—An2 (arrestin-3), including broader regulation of cell signaling in general, serving as scaffolds, as well as having roles in transcriptional regulation (40). Recent studies demonstrate B-arrestins as negative regulators of TLR-stimulated NFIcB and ERK signaling pathways in macrophages and fibroblasts (41, 42). Consistent with this role of B-arrestins, Wang et. al. showed that deficiency of B-Arr2 significantly enhances endotoxic mortality in mice (41). In contrast to these studies, Fan et. al. (43) demonstrated differential effects of B-Arr1 and -2 in lipopolysaccharide signaling in mouse embryo fibroblasts, suggesting that B-arrestin’s effect on TLR signaling might depend on the cellular context. Although a role for B-arrestins has been established using selective TLR ligands, their role in microbial infections (which activate multiple TLRs) is not known. Innate immune responses associated with systemic injection of Adenovirus (Ad) based vectors remain to be one of the most important obstacles limiting the usage of these vectors in numerous clinically important applications, 10 requiring high level vector administrations, for example for liver targeted therapy. Systemic (intravenous) administration of Ads results in acute thrombocytopenia (26, 44), activation of the complement, TLR and lFN systems, a robust cytokine release into the circulation sometimes referred to as a “cytokine storm" (24, 26, 27, 36), activation of endothelial cells (45) as well as massive inductions of pro- inflammatory gene expression in multiple tissues, including the liver, spleen and lung (23, 26, 46-50). Therefore, understanding the mechanisms mediating Ad induction of these processes may allow for focused efforts to interfere with these mechanisms to foster improved safety and/or efficacy of Ad mediated gene transfer applications. Because TLRs significantly modulate Ad vector responses, and since B-arrestins play a crucial role in TLR signaling, we analyzed the role of B-arr-1 and -2 in Ad5-induced innate immune responses both in vitm and in vivo. Our results confirm that multiple Ad5 induced innate immune responses are mediated by B—arrestin functionality. Moreover B—Arr1 and B-Arr2 differentially mediate Ad5 induced innate responses, having somewhat opposite functions: [3- Arr2 clearly down-regulates Ad5 induced gene induction and cytokine responses, whereas B-Arr1 enhances portions of these responses, as detailed in chapter II of the dissertation. 1.2.2. The complement system: first line of defense against pathogens The complement system, which is composed of over 30 membrane-bound as well as fluid phase proteins, represents the main non-cellular part of the innate immune system (Figure 2). Most complement components are synthesized by hepatic parenchymal cells in the liver, however, some may also be synthesized ll by macrophages (51). Complement proteins can bind and facilitate opsonization of different types of pathogens including those not previously encountered by the host. There are three major complement pathways: 1) Classical Complement Pathway is activated as a result of specific antibody interactions with a previously encountered pathogen. Pre-existing antibodies (lgG or lgM) bind to the microbe, which allows C1q complement protein to bind to the antibody/pathogen complex. This in turn triggers C1r and C1s binding, creating an active 01qu serine protease (Ca2+ dependent), which cleaves proteins C4 (releasing C4a and 04b) and 02 (releasing C2a and 02b). Reactive C2a and 04b associate with each other into a complex on the pathogen, giving rise to the classical pathway C3 convertase: C4b2a (Mg2+ dependent). C3 convertase cleaves large amounts of C3, generating C3a anaphylotoxin and C3b. C3b protein can activate endothelial cells (ECs) by binding to specific receptors on EC surfaces (49). 03b can also directly bind to pathogens, facilitating targeting of pathogens to complement receptors on phagocytic cells (macrophages, granulocytes) (49, 52). The C4b2a3b complex is a C5 convertase cleaving CS into 05a (a potent anaphylotoxin) and C5b. C5b protein binds to the surface of a pathogen (or infected cell), which triggers membrane attack complex (MAC) formation by recruitment of complement proteins 06-09. The MAC can form a pore in bacterial or enveloped viral pathogens, thereby initiating their .lysis. The potent anaphylotoxins C3a and C5a efficiently bind to their receptors (C5aR, C3aR) on mast cells, granulocytes and 12 macrophages facilitating their activation (49), (Figure 2), thereby activating the cellular part of the innate immune system. 2) The Iectin pathway for complement activation is homologous to the classical pathway but is initiated by the binding of the mannan-binding Iectin to mannose residues on the pathogen surface. This causes activation of the MBL- associated serine proteases, MASP-1, MASP-2, MASP-3, which cleave proteins C4 and C2, generating the C3 convertase C4b2a. 3) The alternative complement pathway has a role in combating pathogens not previously encountered by the host immune system (i.e. there are no pre-existing antibodies to the pathogen). Protein C3 possesses an internal thioester bond between a cysteine and the v-carboxyl group of a glutamine residue allowing low levels of reactive fluid phase C3b-Iike molecules to be spontaneously produced and rapidly inactivated in the blood via a “tick—over" mechanism. However, upon pathogen encounter, pre-existing C3-OH can bind stably to the surface of the pathogen, facilitating binding of factor B (Fb) to the C3blpathogen complex. The complex is recognized by factor D, and Fb is cleaved to generate the alternative pathway-derived C3 convertase C3bBb (Mg2+ dependent). Numerous C3 molecules are cleaved by this alternative pathway- derived C3 convertase. The alternative pathway can also serve as an amplification loop for classical pathway-initiated complement system activation (52-54). Excessive complement activation can cause a number of disorders including adult respiratory distress syndrome (ARDS), hypotensive shock, 13 anaphylactoid reactions, systemic inflammatory response syndrome, and thrombocytopenia. Many of these toxicities have been observed after high dose Ad administration into rodents, non-human primates, and humans (48-50). Our published data confirms that the intact Ad capsid interacts with human and murine complement immediately after delivery in vitro and in vivo thereby consuming complement components and inducing several toxicities, toxicities that can be avoided by complement inhibition (24, 26, 46, 55), as fully detailed in several chapters of this dissertation. l4 Figure 2: The complement system. Three major complement pathways are shown. Activation of either of the pathway leads to formation of C3- convertase, which cleaves main complement protein C3 and allows to generate CS-convertase, capable of cleaving C5 into 05a and 05b. Potent anaphylotoxins, released during complement activation, activate cellular arm of the innate immune system. C5b facilitates formation of membrane attack complex (MAC), capable of lysing infected cells and pathogens. 15 Classical Lectin Alternative . Pathwa Pathway Pathway -\ ® C3 convertases P "D Anaphylatoxins 1.2.3. Complement and Adenovirus Ad-based vectors are capable of activating human complement via both classical and alternative complement pathways (24, 26, 46, 47, 55). While it was well established that Ad5 can activate the classical complement pathway in the presence of Ad-specific antibodies, our group first confirmed that immobilized Ad5 capsids can mediate the activation of the alternative complement pathway in Ad-na‘lve serum (47, 55). Ad5 is also capable of activating murine complement, justifying the use of a mouse model to investigate the role of complement in Ad- triggered responses (26). In this regard, intravenous administration of high doses of Ad5-based vectors in mice deficient in various complement factors including C1q and C4 (classical pathway), FB (alternative pathway), or C3 (shared pathway) has revealed that Ad-triggered induction of thrombocytopenia is dependent upon a functional complement system and is alternative pathway dependent (24, 26, 46). It was also confirmed that the induction of several important pro-inflammatory cytokines/chemokines by Ads is also complement dependent. The induction by Ads of some cytokines is dependent upon alternative pathway proteins, while the induction of other cytokines by Ads is depended upon classical pathway activation. However, the induction of most cytokines and chemokines by Ads was found to be primarily dependent upon the presence C3 - the complement pathway component that is a key converging point of both the classical and alternative complement pathways (24, 26, 46, 56). It is also known that complement system activation acts to optimize induction of pathogen specific antibody responses (57-59). Subsequent to 17 opsonization by activated C3, pathogens are bound to B cells and dendritic cells via binding of the pathogen bound C3 to the complement receptors (CRs). The human CR1 and CR2 receptors have well known roles in modulating both innate and adaptive immune responses. Human CR1 (hCR1) is a potent inhibitor of complement activation, having both decay-accelerating and cofactor activities. Furthermore, hCR1 has a critical role in the clearance of immune complexes, and B cell maturation, as thoroughly reviewed elsewhere (60, 61). Human CR2 (hCR2) expression is restricted to the surface of B cells, follicular dendritic cells and thymocytes. When hCR2 binds C3d opsonised pathogens and becomes associated with CD19, it lowers the threshold for B cell activation by up to 1000 fold (62). HCR1 and hCR2 also play a role in T cell biology, for example, crosslinking of hCR1 inhibits T cell proliferation and lL-12 production (63). In contrast to their human counterparts, the murine complement receptors (mCRs) 1 and 2 (CD35ICDZ1) are products of alternative splicing from the same gene. mCR1 contains 21 complement control protein repeats (CCPRs), whereas the smaller mCR2 contains only 15 C-terrninal CCPRs of mCR1 (64). mCR1/2 is known to be expressed on B cells and dendritic cells (64). Therefore, the expression pattern of mCR1/2 resembles that of hCR2, but not hCR1. Similar to their human homologues, the functions of mCR1/2 related to generation of maximal humoral responses have also been well described (65—69). Interestingly, mCR1/2 functionality prevents excessive myocardial tissue damage subsequent to coxsackievirus B3 infection (70), as well prevents lethal Streptococcus 18 pneumoniae infection, a role potentially indirectly reflective of the complement inhibitory activities of the CRs (71). While the role of murine CR1/2 protein has been extensively studied in regards to adaptive immune responses, its function in inhibiting/regulating murine complement dependent responses has not been intensely investigated, possibly since in most mouse models, the Crry protein was suggested to play the predominant role in controlling complement activation. Our studies, however, now confirm that the role of murine CR1/2 protein in innate immune responses (including the ones which are known to be complement dependent) may be more important than previously considered, as suggested by our present studies of Adenovirus mediated gene transfer into mCR1I2-KO mice. Our results in murine models revealed dual roles for mCR1/2; roles that include down-regulation of multiple aspects of the Ad induced innate immune responses, while also playing the major role in the complement dependent induction of neutralizing antibody responses to Ads. 1.3. Approaches directed to minimize Ad-triggered immune responses Innate toxicities, rapidly developed in response to systemic Ad injections, significantly limit more widespread utilization of this gene transfer platform. Additionally, Ad-based gene transfer approaches can be limited due to adaptive immune responses to the virus or the transgene it encodes. These adaptive immune responses can limit the duration of transgene expression and/or limit the ability to re-administer the vector, although this highly depends on the immunogenicity of the transgene delivered (14). Though it is often noted that Ad 19 mediated gene delivery is transient due to these responses, there are multiple examples that even first generation Ads, and certainly advanced generation Ads, can allow for long-terrn gene expression in vivo (14). For example, first generation Ad mediated delivery of non-immunogenic transgenes can persist for long periods of time in both murine and non-human primate models. Similar levels of improved efficacy are more likely to occur with the use of the advanced generation Ad-based vectors (16, 72, 73). An important problem in regards to potential use of Ads in human clinical applications is that the majority of the human population has been infected by wild type Ads in childhood, and often these individuals develop pre-existing immunity to a number of common Ad serotypes (i.e.: serotypes 2, 4, 5, and 7). The percentage of the human population with medium to high Ad specific antibody titers largely depends upon geographic location, with 30-50% of the United States population, and up to 90% of the sub-Saharan African populations having significant Ad5 antibodies with an overall trend to having higher titers in developing countries (74-76). Pre-existing immunity to Ad can include both neutralizing antibodies (NAb), as well as Ad protein specific T cell responses. Additionally, recent studies highlight a critical role for CDB“ T cells in pre-existing Ad immunity (9, 77). This latter point is very important, as these pre-existing responses can in fact still be harnessed when a host is exposed to a novel (previously unencountered) Ad serotype (78-81). Therefore, several strategies have been proposed to improve the efficacy or utility of Ad-based gene transfer vectors (78, 82, 83). Below we discuss three 20 approaches, developed to minimize Ad vector induced innate toxicities. Importantly, these approaches have shown improved efficacy in Ad-immune hosts and/or allowed for efficient re-administration of Ad vector, generating significantly blunted Ad-specific adaptive immune responses, including NAb. 1.3.1. Ad capsid modifications 1.3.1.1. Chemical modifications of the Ad capsid Several groups have chemically modified the Ad5 capsid (i.e.: in a non- covalent fashion) in an attempt to shield or hide highly antigenic epitopes on the viral capsid (mainly hexon protein) from the immune system of the Ad-immune host. This strategy primarily involves the addition of synthetic polymers onto the Ad capsid. These polymers can include polyethylene glycol (PEG) (84), polyactic glycolic acid (PLGA) (85) or other lipids (86). PEGylation appears to be the most promising approach for non-covalent modification of Ad capsids. Ad5 PEGylation may not only reduce Ad-triggered toxicities, but also allow for re-administration of such modified Ads in mice. For example, animals were intravenously injected with an Ad5 vector expressing an irrelevant antigen, and then re-injected with conventional or PEGylated Ad5 vectors each expressing the bacterial B-galactosidase gene (B-Gal) (87). This simple study revealed high levels of B-Gal expression only in Ad5-immune mice re-injected with the PEGylated Ad5 vector (87). Moreover, it was shown that PEGylated Ad5 vectors also reduce the induction of Ad5-triggered innate toxicities, such as thrombocytopenia, plasma ALT elevations, and release of pro- 21 inflammatory cytokines such as lL-6 (87, 88). PEGylation with a small PEG (5 kDa) moiety increases transduction efficiency of the Ad5 capsid in Ad-immune mice (89). Use of a 5 kDa PEGylation of Ad5 can also reduce Ad5 specific neutralizing antibody (NAb) titers generated after intranasal injection into Ad- naive BALBIc mice (90). How PEGylation may affect the efficiency or tropism of Ad5 mediated gene transduction in vivo has not been fully delineated. For example, a dramatic reduction of liver transduction was reported after Ad PEGylation (91), while other studies reported equal and/or increased liver transduction by PEGylated Ads (87, 88). In summary, characterization of how PEGylation affects the ability of so modified Ad vectors relative to tropism changes, ability to reduce Ad-triggered innate and adaptive immune responses, is still required to determine if these strategies afford improved utility of PEGylated Ad-based platforms. 1.3.1.2. Insertion of novel peptides into Ad capsid proteins Several Ad capsid proteins, including the fiber, penton, protein IX .and hexon proteins have been exploited for genetic insertion of foreign peptides into these specific capsid proteins, either as “in-frame” insertions within the proteins, or as “in-frame” C-terrninal fusions. If the insertion does not cause a deleterious change in the biology of the so-modified protein, (i.e.: causes misfolding, lack of stability, lack of trimerization or lack of incorporation into the tertiary structure of the Ad capsid) novel, recombinant Ad vectors “capsid-displaying” the respective foreign peptide can be isolated. The penton and hexon proteins tolerate relatively small peptide insertions: up to 18 amino acids (aa) for penton base (92, 93), and 22 up to 36 aa within the hypervariable region 5 (HVR5) of hexon (94, 95), whereas the HI loop of the fiber knob and C-tenninus of protein IX allow for incorporation of peptides of substantial size: up to 83 aa for fiber (96) and up to 1018 aa for plX (97). Issues regarding capability to generate these recombinant viruses consistently, to very high titers, or in a cGMP compliant fashion have many times not been addressed in studies published to date, but will need to be if clinical deployment of these vectors is to be achieved. The insertion of foreign peptides into capsid proteins in attempts to change the natural tropism of the Ad vector have been well-described (98, 99). Ad5-based vectors depend on coxsackievirus-adenovirus receptor (CAR) receptor for cell transduction: fiber protein, consisting of three domains (tail, shaft, and trimeric knob), is responsible for initial cell attachment. Fiber knob I CAR interaction is followed by the binding of the penton proteins to (NBS or dv85 integrins through a conserved Arg-Gly-Asp (RGD) consensus motif. In addition, factor X Gla domain interactions with hypervariable region of Ad hexon were recently identified as critical factor mediating liver Ad transduction via Heparan- sulfate proteo-glycans (100). Based on this simple Ad biology, fiber domains (and knob in particular) as well as RGD motif from penton and hypervariable region of hexon protein are the best targets in attempts to modify Ad5 vector tropism. Therefore, several studies described the incorporation of polylysine or RGD motifs into different locations of Ad5 capsid, resulting in modified tropism. Specifically, incorporation of a polylysine motif into the plX was shown to augment Ad fiber knob independent 23 infection of CAR-deficient cell types in vitro (101). RGD and polylysine motifs have been incorporated into the fiber knob, providing an ability to infect CAR- deficient cells in vitro (102-105). Additional studies, utilizing in vitro comparative analysis of Ads, capsid-displaying RGD peptide at HI loop of fiber knob, C- terminus of fiber knob, C-ten'ninus of plX or HVR5 of hexon, revealed that the most efficient transduction was achieved when HI loop of fiber was used for RGD incorporation (99). It has been subsequently confirmed that RGD motifs incorporated into C-tenninus of protein IX allowed for transduction of CAR- deficient cell types in vitro (106). The use of 30-75 Angstrom a—helical spacers for incorporation of foreign peptides in pIX was suggested (106), but it is still unclear, however, if these spacers provide any detectable benefit, since numerous studies showed promising results incorporating peptides in pIX without spacers (99, 101). More detailed direct comparison between several alternative plX- displaying Ads (with and without linkers) is required to answer this question. A major limitation of these targeting approaches is that numerous promising in vitro results fail to provide any significant benefit in vivo, primarily due to inability to overcome natural liver tropism of Ad-based vectors. However, several studies provide optimism by showing reduced metastasis formation and increase survival in mice with usage of E1a CRAD (107) or 2-fold increased tumor uptake of targeting Ad-displaying avB6 integrin-selective peptide as compared to intact capsid-Ad, which was in parallel with reduced liver transduction by targeting Ad (108). Future development of advanced targeting Ads, possibly by combining approaches undertaken up to date, should be 24 continued in order to more fully determine the applicability of targeting Ad for clinical applications (109). Note that in this dissertation we describe a novel class of Ad5-based vectors, capsid-displaying specific complement inhibitory peptide (COMPinh). These novel Ads minimize complement activation (with a potential to minimize Ad-induced complement dependent toxicities) and represent a promising tool for numerous future gene transfer applications. 1.3.2. lmmunosuppressive glucocorticoid dexamethasone The synthetic anti-inflammatory glucocorticoid dexamethasone (DEX) is an FDA approved drug widely used to treat a number of transient and/or chronic inflammatory conditions (110-115). Importantly, mechanisms of action of DEX include preventing the activation of NFkB and AP1 transcription factors, as well as MAP kinases, all of which have been shown by us and others to also be important mediators of the Ad induced inflammatory response complex (116- 118). lL-8 and lFNB production have also been shown to be inhibited by DEX (117, 119). As a result, the maturation of mast cell progenitors, as well GM-CSF and TNFa secretion by mast cells are also altered by DEX treatment (120). Very limited data is available regarding the effect of DEX treatment on animals treated with gene transfer agents. Use of glucocorticoids has been shown to increase the efficacy of gene transfer in vitro, while in vivo studies showed that dexamethasone could improve non-viral gene and virus derived gene expression in experimental animal models (121-125). Relative to Ad 25 mediated gene transfer, DEX can improve Ad vector derived gene expression when these vectors are directly administered into the spinal cord, nasal mucosa, inner ear or lungs (124-128). Pulmonary delivery of Ad vectors into mice pre- treated with Dexamethasonelspermine resulted in a reduced activation of limited portions of the lung specific innate and adaptive immune response to the Ad vector utilized (128). These results prompted us to evaluate the efficacy of DEX to prevent the numerous innate toxicities induced by systemic administration of Ad vectors, as detailed in chapter V. In summary, all of the approaches, summarized on figure 3 have shown promising results in terms of improving some aspects of Ad vector safety and/or efficacy. Further development of genome modified Ad vectors, “capsid- displaying” Ads, chemically modified Ads, chimeric Ads, as well as Ad vectors, derived from alternative Ad serotypes is now justified. Possibly, a combination of some (or all) of these approaches may result in construction of safer and a more efficacious Ad vector platform. In this dissertation we describe a novel class of Ad vectors, “capsid-displaying” specific peptides, inhibiting complement activation, and outline critical role of complement in mediating Ad-triggered innate and adaptive immune responses. We also propose to use an FDA approved potent immunosuppressive glucocorticoid Dexamethasone to block Ad- induced immune responses, potentially in combination with other approaches to further improve the outcome of gene transfer applications. 26 Figure 3: Current approaches to design improved Adenovirus based gene transfer vectors. Group 1. Starting with a wild-type Ad5 virus the E1 region is deleted to generate [E1-] Ad5 vectors; Group 2. Adenovirus vectors of any group can be PEGylated; Group 3. Capsid “display” of antigens at various locations on the Ad capsid where indicated; Group 4. Chimeric Adenovirus vectors, that contain capsid proteins derived from 2 different Ad serotypes; Group 5. Adenovirus vectors derived completely from alternative serotypes; Group 6. Genome Modified Adenovirus vectors, that retain the parental serotype capsid, but are deleted for portions of the parental genome. All of Group 1-6 vectors can be utilized in combination with transient pre-treatment 'with synthetic immunosuppressive glucocorticoid Dexamethasone to further improve the outcome of an Ad-based gene transfer. Note, that we describe Group 3 approach in this dissertation. 27 Ad5 WT M P “2:2?" " 0' Alternative human r \ displaying .7 . _ Ad serotype / Ad5 . ,. 5 7i" ”Protein IX” Alternative non- displaying Ad5 [EH Ad human Ad serotype upentonu l 4 ‘ l 5‘ Ad5/3 Fiber - - E1-, E2b- Ad displaying i l - a v Chimera 6 "Fully . y x, “Fiber” ,i deleted” Ad5/25 HVR5/7 displaying Ad5 HDAd-J ChImera j "PEGylated”_. Ad vectors 28 Chapter II Ad5-triggered signaling pathways: critical roles of BArr-1 and BArr-2 adaptor proteins This chapter is the edited version of a research article that was published in the Journal of Virus Research, Volume 147, Issue 1 (123-134), November 10, 2009. Authors: Seregin, S. S., Appledom, D. M., Patial, S., Bujold, M., Nance, W., Godbehere, S., Parameswaran, N., and A. Amalfitano. 29 2.1. Introduction B—Arrestin-1 and -2 (B-Arr1 and 2) were originally discovered to play a major role in G protein coupled receptor desensitization, due to their ability to bind to phosphorylated G protein coupled receptors and sterically block their ability to signal downstream (40). Subsequent studies revealed more versatile roles of B-Arr1 (arrestin-2) and B—Arr2 (arrestin-3), including broader regulation of cell signaling in general, serving as scaffolds, as well as having roles in transcriptional regulation (40). Recent studies demonstrate B-arrestins as negative regulators of TLR-stimulated NFIcB and ERK signaling pathways in macrophages and fibroblasts (41, 42). Consistent with this role of B-arrestins, Wang et. al. showed that deficiency of B-An2 significantly enhanced endotoxic mortality in mice (41). In contrast to these studies, Fan et. al. (43) demonstrated ' differential effects of B-Arr1 and -2 in lipopolysaccharide signaling in mouse embryo fibroblasts, suggesting that B-arrestin’s effect on TLR signaling might depend on the cellular context. Although a role for B-arrestins has been established using selective TLR ligands, their role in microbial infections (which activate multiple TLRs) is not known. Innate immune responses associated with systemic injection of Adenovirus (Ad) based vectors remain to be one of the most important obstacles limiting the usage of these vectors in numerous clinically important applications. For example, systemic administration of Ads results in acute thrombocytopenia (26, 44), activation of the complement, TLR and IFN systems, a robust cytokine 30 release into the circulation sometimes referred to as a “cytokine storm” (24, 26, 27, 36), activation of endothelial cells (45) as well as massive inductions of pro- inflammatory gene expression in multiple tissues, including the liver, spleen and lung (23, 26, 46-50). Therefore, understanding the mechanisms mediating Ad induction of these processes may allow for focused efforts to interfere with these mechanisms to foster improved safety and/or efficacy of Ad mediated gene transfer applications. Because TLRs significantly modulate Ad vector responses, and since B-arrestins play a crucial role in TLR signaling, in this study we analyzed the role of B-arr-1 and -2 in Ad5-induced innate immune responses both in vitro and in vivo. Our results confirm that multiple Ad5 induced innate immune responses are mediated by B-arrestin functionality. Moreover B-Arr1 and B—Arr2 differentially mediate Ad5 induced innate responses, having somewhat opposite functions: B-An2 clearly down-regulates Ad5 induced gene induction and cytokine responses, whereas B—Arr1 enhances portions of these responses. 31 2.2. Results B-Arrestins differentially mediate Ad5 induced cytokine and chemokine release from peritoneal macrophages in vitro. Macrophages participate in the first line of defense against invading pathogens by functioning as phagocytes, antigen presenting cells, and activators of both innate and adaptive immune responses via secretion of cytokines and chemokines that serve as co-activating factors. Numerous studies have indicated that the induction of cytokines and chemokines by Ad5 injection is dependent upon the presence of macrophages, specifically liver resident macrophages known as Kupffer cells. We have shown that the inductions of KC, MOP-1, lL-12(p40) and RANTES are Kupffer cell dependent in vivo (36). To determine if B-Arrestins also play a role in the secretion of cytokines and chemokines from macrophages, we isolated peritoneal macrophages from wild type, B-Arr1, and B—Arr2 knockout mice, exposed them to Ad5-LacZ, and measured the concentration of pro- inflammatory cytokines in the growth media at various time points post Ad exposure (Figures 4-5). Interestingly we observed that levels of MIP-1B, MCP-1 and RANTES were significantly lower (p<0.05) in the B-Arr-I-KO macrophages compared to the wild types. We also observed that the decrease was time- dependent, i.e. MIP1B was lower at 6 h.p.i., whereas MCP-1 and RANTES were decreased at 24 h.p.i., suggesting that these are potentially regulated by distinct mechanisms (Figure 4). In contrast to these results, levels of lL-12(p40) and MCP-1 (at 24 and 48 h.p.i.) were significantly enhanced (p<0.05) in 32 Figure 4: Functional 8-arrestin-1 acts as positive regulator for several chemokines released in response to Ad infection in vitro. Peritoneal macrophages derived from WT or B-Arr1-KO C57BU6 mice were isolated, plated and infected with Ad5-LacZ as described in Materials and Methods. Media samples were collected at 6, 24 and 48 hours post virus infection (hpi) and assayed using a multiplexed bead array based system. The bars represent Mean 1 SD. Statistical analysis was completed using Two Way ANOVA with a Bonferroni post-hoc test. The N=4 for all groups of peritoneal macrophages, including mock-infected groups. *, ** - indicate media cytokine values that are statistically different from those in Mock-infected groups of the same genotype at the same time point, p<0.05, p<0.001 respectively. ft, #11! - indicate statistically different values in the same treatment group at the same time point, p<0.05, p<0.001 respectively. 33 lL-6 IL-12p40 fl, n n .0- 1” it t, E 60' n \ n U n. . 40 5°. 20d 0- 6 Mil 24 1101 48 1191 S hDI 24 hal 48 hpi I ! 0- U l I 6 "DI 2‘ ml OB hpi 6 Elm 24 "BI 4B 119' MCP-1 = WT_Mock E B-Arrl-KO_Mock - WT_Ad5-LacZ E: B-ArrI—KO_Ad5—Lacz C 1191 24 hol 4. 1191 34 Figure 5: Functional B-arrestin-2 acts as negative regulator for several chemokines released in response to Ad infection in vitro. Peritoneal macrophages derived from WT or B—Arr2-KO CS7BL/6 mice were isolated, plated and infected with Ad5-LacZ as described in Materials and Methods. Media samples were collected at 6, 24 and 48 hours post virus infection (hpi) and assayed using a multiplexed bead array based system. The bars represent Mean 1 SD. Statistical analysis was completed using Two Way ANOVA with a Bonferroni post-hoc test. The N=4 for WT_Mock and WT_Ad5-LacZ groups, N=6 for B-Arr2-KO_Mock and B-An2-KO_Ad5-LacZ groups. *, ** - indicate media cytokine values that are statistically different from those in Mock-infected groups of the same genotype at the same time point, p<0.05, p<0.001 respectively. #, #If - indicate statistically different values in the same treatment group at the same time point, p<0.05, p<0.001 respectively. 35 ## IL-B IL12p40 F1 1* ** 200 “I 100 ** fl 0 6 hot 24 hpi 49 hpi 6 Mil 24 hpl 48 hpi GCSF KC it it fit *‘l' I1- I‘* 9* * 200 fl. *i * 100 6 her 24 hpi 48 her a hpi 24 run 4: MI MOP-1 Ill MIP-1fl r3. ..m .. 20000 10000 0 c not 24 MI 4. trial 6 hot 24 hot 48 MI RANTES _ E: WT_Mock . an B—Arr2—KO_Mock ** -WT_Ad5—LacZ - B-Arr2—KO_Ad5-Lacz 3 MI 24 hol 48 nor macrophages, derived from B-Arr-Z-KO mice compared to WT macrophages (Figure 5). These results, taken together, suggest that B-Arr1 may act as a positive regulator, and B-Arr2 as a negative regulator of Ad5-induced inflammatory responses. Positive and negative roles for B-Arrestins in Ad5 induced cytokine and chemokine release. Our results using primary macrophages from B-Arr-1 and -2 KO mice clearly demonstrate important but differential roles for B-arrestins in Ad-induced inflammatory responses. Therefore, we evaluated the potential that B-arrestin-1 and -2 also play a pivotal role in the release of inflammatory cytokines and chemokines following systemic Ad5 administrations in vivo. To accomplish this, the plasma levels of seven cytokines/chemokines, commonly induced following Ad5 injection were evaluated, following systemic delivery of 19:10” vps Ad5-LacZ. As expected, all seven cytokines/chemokines tested were significantly induced within 1 h.p.i. (IL-6, MCP-1, MlP-1B, and KC, p<0.001, p<0.05), and/or 6 h.p.i (IL-12(p40), G-CSF, MCP-1, MlP-1B, and RANTES; p<0.001, p<0.05) in wild-type CS7BU6 mice (Figures 6-7). Identically injected B- Arr-1-KO mice also exhibited significant inductions of MCP-1 and MlP-1B at 1 h.p.i. (p<0.05 and p<0.001, respectively). However, at this time point, levels of IL- 6 and KC were significantly lower than levels found in the wild types (p<0.05) and were not statistically induced over mock levels. Furthermore, the induction of IL- 12(p40) was significantly lower at 6 h.p.i. in Ad5-injected B-Arr-1-KO as compared to wild-type animals. Although levels of G-CSF and MCP-1 in B—Arr1- 37 Figure 6: Functional B-arrestin-1 acts as positive regulator of Ad mediated systemic cytokine and chemokine release in C57BU6 mice. WT or B-Arr1-KO 057BLI6 mice were intravenously injected with 0.75x1011 vplmouse of Ad5-LacZ vector. Plasma samples were collected at 1 and 6 hours post virus injection (hpi). Plasma samples were analyzed using a multiplexed bead array based system. The bars represent Mean :t SD. Statistical analysis was completed using Two Way ANOVA with a Bonferroni post-hoc test. The N=3 for Mock (PBS) injected animals, N=4 for virus injected mice. *, ** - indicate plasma cytokine values that are statistically different from those in Mock-injected animals of the same genotype at the same time point, p<0.05, p<0.001 respectively. it, #If - indicate statistically different values in the same treatment group at the same time point, p<0.05, p<0.001 respectively. 38 Iii IL-6 rL-12p40 # 1 hpl e hpl c-csr MOP-1 1 hpi a hpi RANTES = WT_Mock E 6—Arr1—KO_Mocl< -WT_Ad5-LacZ :3 B—ArrI-KO_Ad5—LaCZ 39 Figure 7: Functional B-arrestin-Z acts as potent suppressor of Ad mediated systemic cytokine and chemokine release in C573lJ6 mice. WT or B-Arr2-KO C57BU6 mice were intravenously injected with 0.75x1011 vplmouse of Ad5-LacZ vector. Plasma samples were collected at 1 and 6 hours post virus injection (hpi). Plasma samples were analyzed using a multiplexed bead array based system. The bars represent Mean :I: SD. Statistical analysis was completed using Two Way ANOVA with a Bonferroni post-hoc test. The N=3 for Mock (PBS) injected animals, N=4 for virus injected mice. *, ** - indicate plasma cytokine values that are statistically different from those in Mock-injected animals of the same genotype at the same time point, p<0.05, p<0.001 respectively. #, #If - indicate statistically different values in the same treatment group at the same time point, p<0.05, p<0.001 respectively. 40 ** IL-6 r31 IL-12p4o IE' 300 SM ** 'g' ...| i I 2000 B *1: Q 1% 1M 0 0 1 hpl 6 hpl 1 hpl 6 hpI c-csr MOP-1 .... r32. .. tit. _ mo- * m, «E 1500- 21M- ”00‘ 500- zoom 0- o. 1 hpl 6 hpl 1 hpi 6 hpl MIP-Ip RANTES .... fig .0. a; —- m. E «oo- DD 100- a. 1000~ * m- 0. 0- 1 hpi e hpl 1 hpi 6 hpl m r31 “C = WT_Mock _ .. ** m B-Arr2—KO_MOCI< E - WT_AdSLacZ 901000 ‘1 - B-Arr2—KO_Ad5—LacZ 1 I'Ipl 0 hp! 41 KO did not reach statistically significant differences as compared to identically injected 057BLI6 mice, the levels of these factors were not statistically induced over background levels in B-Arr1-KO mice in contrast to WT mice. No significant differences in either MIP-1B or RANTES were observed (Figure 6). Overall, the results demonstrate that B-Arr1 regulates induction of an important subset of Ad5-induced cytokines and chemokines following systemic injection of Ad5 vectors in vivo. In contrast to observations in B—Arr1-KO mice, levels of all cytokines and chemokines tested in the plasma of Ad5-injected B-Arr2-KO mice were significantly enhanced when compared to identically injected wild type mic». Specifically, at the peak of the KC response, significantly higher levels of this chemokine were detected in Ad-injected B-Arr2-KO animals as compared to wild- type mice. Additionally, significantly higher levels of lL-6 (2-fold), lL-12(p40) (2- fold), G-CSF (1.4-fold), and MCP-1 (6-fold) were detected in the plasma of 8- Arr2-KO compared to wild type mice at 6 h.p.i. (Figure 7). Importantly, the absolute numbers of Ad5 genomes present in the liver in either B-Arr1-KO or B- Arr2-KO mice were not different from WT mice as measured by Ad5-genome levels (Figure 8A-B). While there were differences in Ad genome content of the spleens, these differences did not result in differential transduction of these tissues as measured by LacZ protein expression, as determined using a qualitative analysis of X-gal staining and quantitative B-Gal activity 42 Figure 8: Functional B-arrestin-1 and B-arrestin-Z have modest impact upon liver and spleen transduction efficiency by Ad5 in C573U6 mice. qPCR based quantification of Ad5 genomes in liver and spleen tissues harvested from (A) WT and B-Arr1-KO C57BU6 mice or (8) WT and B-Arr2-KO C57BU6 mice at 6 hpi was performed. The bars represent Mean :I: SD. Statistical analysis was completed using tvvo-tailed Student t-test to compare each B-Arr-KO with WT group of virus injected animals. # - indicate statistically different values in B-Arr1-KO_Ad5-LacZ group or B-Arr2-KO_Ad5-LacZ group as compared to WT_Ad5-LacZ group, p<0.05. Experiments were performed in duplicate; representative experiment is shown. N=4 for all groups of mice, including Mock-injected animals. 43 .1 il w m... m c I» In m .m. .. =3 .52. \ meEocou v< :3 cue...» \ «0.553 3 B TI— . # m. w m. h c e.» u 9.. .w A =3 .52. \ 380cc» 3 =3 new...» \ 3505» 3 _ _ pl wr Ad5-LacZ 6h ' WI'Mock6h' D - _ _ PI WTAdS-LacZ WTMOdtSh D - pl 6111* cZ_6hpi -KO Ads-Ca mMer-Ko Moo—k -I3A"Q _6hpi O_Ad5-LacZ_6hpi EW-KO_Mcck_6hpi 1:3 W -K 44 Figure 9: Functional B-arrestin-1 and B-arrestin-Z have modest impact upon Ad derived transgene expression in both liver and spleen tissues in C57BU6 mice. Bacterial B-galactosidase activity levels were analyzed in liver and spleen protein homogenates prepared at 6 hpi from (A) WT and B-Arr1-KO C57BU6 mice or (8) WI' and B-ArrZ-KO CS7BU6 mice. Activity levels were presented as Units per mg of total protein (see Materials and Methods). The bars represent Mean :I: SD. Statistical analysis was completed using two-tailed Student t-test to compare each B-Arr-KO with WT group of virus injected animals. No significant differences were detected. 45 > U / mg liver U / mg spleen E:WT_Mock_6hpi -WT_Ad5-LacZ_6hpi EflArr1-KO_Mock_6hpi DpArrI-K0_Ad5-Lacz_6hpi U / mg liver U / mg spleen 46 DWT_Mock_6hpi -WT__Ad5-LacZ_6hpi EBArfl-KO_Mock_6hpi -8Arr2-KO_Ad5-LacZ_6hpi measurements in both liver and spleen tissues (Figure 9A-B). Therefore the results presented here were most likely not attributable to differential transduction of murine tissues per 39. Together these data indicate that B-Arr1 positively regulates the induction of a subset of Ad5-induced cytokines and chemokines, while B—Arr2 functions more globally as a negative regulator of the induction of these innate immune factors. B-Arrestins differentially mediate the induction of pro-inflammatory genes in livers and spleens following systemic Ad5 injection. We have previously characterized tissue specific transcriptome changes rapidly induced after transduction by Ad5 vectors both in vitm and in vivo (23, 26, 28, 35). Therefore we have selected a panel of genes and analyzed their expression in the liver and spleen of Ad5-injected WI', B—Arr1-KO and B—Arr2-KO mice at 6 h.p.i. (Tables 1-4). Selected genes include those involved in innate immune responses, such as pattern recognition receptors (T LRs, NODs), TLR signaling pathways (MyDBB, TRIF, TRAF6, TRAF2bp, TBK1), markers of endothelial cells activation (e-Selectin, ICAM-1, VCAM-1), interferon responsive genes (OAS1a, IRF7, IRF8), negative regulators of cytokine signaling (8008-1, 8008-3), and dsRNA editing enzymes (ADAR) (23, 28, 35). Our results demonstrate that Ad5- induced activation of a number of genes in the liver of Ad5-injected B-Arr1-KO mice were significantly reduced compared to corresponding Ad treated WT mice (Table 1). Specifically, B-Arr1-KO mice completely lacked Ad5-induced ADAR, CD14, TLR6 and VCAM-1 transcripts. However, baseline levels of TLR6 were significantly lower in the tissues of B-Arr1-KO mock-injected animals, which may 47 have partially obscured these results. In contrast to Ad5-injected WT mice, NOD- 2 and TRIM-30 were not induced in Ad5 treated B-Arr1-KO mice. We also observed a significant reduction in TLR3 transcripts in the liver of Ad5-injected B- Arr1-KO mice as compared to identically injected WI’ mice. A transcriptome profile observed in spleens isolated from these same animals, revealed significantly reduced levels of TLR3 transcript in B-Arr1-KO mice (Table 2). Opposite results were obtained in Ad5-injected B-ArrZ-KO mice, results that positively correlated with the increased cytokines and chemokines levels observed in these same mice. Over half of the genes tested were induced to significantly higher levels in livers of Ad-injected B-Arr2-KO as compared to identically injected WT mice (Table 3). Specifically, we detected significantly higher amounts of CXCL-9, lCAM-1, lRF-7, MyDBB, SOCS-1, SOCS-3, TBK-1, TLR-2, and TRAF-2bp transcripts in livers of Ad5-injected B-Arr2-KO mice, as compared to wild type mice. Moreover, several genes, including ADAR, IRF8, Oas1a, which were not induced in Ad5-injected WT mice, were significantly induced in Ad5-injected B-Arr2-KO mice. The levels of these same transcripts in the spleen followed a similar trend, as IRF-7, lRF-8 and MyD88 were present in significantly higher levels in spleens of Ad5-injected B-An’Z-KO compared to wild type mice. In addition, CXCL9, lFNa, SOCS-1 and TLR9, which were not induced in spleens of wild type C57BU6 mice, were significantly induced in [3-Arr2-KO mice following systemic Ad5 injection (Table 4). These results further support the notion that B-Arr2 functions as a negative regulator of Ad5-induced innate 48 Table 1: B-Arr1-KO mice had significantly reduced Ad5-triggered activation of a number of genes in livers, as compared to WT mice. Values represent Mean :I: SD. Statistical analysis was completed using One Way ANOVA with a Student-Newman-Keuls post-hoc test, p<0.05 was deemed statistically significant. N=3 for Mock-injected groups, N=4 for Ad5-LacZ injected groups. Significant differences compared to WT_Mock are highlighted in light grey color. Significant differences in transcriptional activation in B-Arr1-KO (Ad5-LacZ) group compared to WT (Ad5-LacZ) group are indicated in table by black frame and boldface font. * Indicate significant differences between mock-injected animals. For full gene names please refer to abbreviation list. 49 Ad5-LacZ Induced gene expression In the liver (fold over CS7BL/6 Mock, 6 h.p.I.) C57BL/6 B-Arrl-KO C57BL/6 (AdS- B-Arrl-KO (Ad5- mgck) (Mock) Laczl LacZ] ADAR 1.0 1 0.2 0.7 1 0.1 2.2 1 0.5 1.6 1 0.2 c014 1.0 1 0.2 0.7 1 0.1 2.31 0.5 1.3 1 0.7 was 1.0 1 0.1 1.0 1 0.1 ' ‘ 10.3 1 2.4 ._ 9.91547“ ‘-, DAF 1.0 1 0.2 0.9 1 0.1 1.2 1 0.1 1.4 1 0.2 e—Selectin 1.0 t 0.1 0.6 1 0.2 - 2.1 t 0.3 2.0 1:10.7‘fi 3;]: ' ,' ICAM 1.0 1 0.2 0.7 1 0.1 2.2 1 0.2 2.2 1 0.4 ' IFNd 1.0 1 0.2 0.6 1 0.1 1.2 1 0.3 0.7 1 0.2 IFNB 1.0 1 0.4 0.8 1 0.2 1.6 1 0.4 1.0 1 0.1 IRF-3 1.0 1 0.2 0.9 1 0.1 1210.2 1.0 1 0.1 IRF-7 1.0 1 0.1 0.9 1 0.2 15.3 1 1.5 , ' 16.0115; ) [RF-8 1.0 1 0.1 0.8 1 0.1* . 2.11.0.3; g,- 7, 234101427621" jak-l 1.0 1 0.2 0.8 1 0.1 1.4 1 0.2 1.1 1 0.1 jak-3 1.0 1 0.1 0.7 1 0.2 1.3 1 0.1 1.3 10.1 MyD88 1.0 1 0.1 0.8 1 0.2 4.110.5'_ .- 4.65: 0.1). -. , » NFkB" 1.0 1 0.1 1.0 1 0.1 1.7 10.2 1.6 10.2 RelA . . . . NOD-1 1.0 1 0.1 0.7 1 01* 1.7102 1.4 1 0.4 NOD-2 1.0 1 0.1 0.6 1 02* 1.3 1 0.3 _ 1.0 1 0.2 OAS-la 1.0 1 0.3 0.7 1 0.1 . 3.1103 2.61 0.5' . : SOCS-l 1.0 1 0.2 0.7 1 0.1 4.6 1‘ 0.6 4.6 11.4 ‘ socs-3 1.0 1 0.2 0.3 1 0.1 2.3 1 0.2 2.5 1 1.4 TBK-l 1.0 1 0.1 0.9 1 0.1 3.91 0.2 3.8110 TLR-2 1.0 1 0.1 0.7 1 0.1 ' 21.7 17.0 25.91109 57 ' TLR-3 1.0 1 0.2 0.7 1 0.1 _ 11.6 1 1.4 ‘ 7.9 1 3.8- TLR-6 1.0 1 0.1 0.6 1 0.1* 2.0 1 0.5 1.5 1 0.2 TLR-9 1.0 1 0.2 0.6 1 0.1 2.0 10.3 1.81 0.2 -1 - TRAFpr 1.0 1 0.2 0.6 1 0.2 6.1 1 1.1 5.9 1 3.3 ' TRAF6 1.0 1 0.2 1.2 1 0.1 1.3 1 0.1 1.3 1 0.1 TRIF 1.0 1 0.1 1.2 1 0.1 1.4 1 0.3' 1.7 1 0.1 TRIM30 1.0 1 0.2 0.6 1 0.1 11.4 1 1.5 7.0 1 5.1 VCAM 1.0 1 0.2 0.7 1 0.1 1.5 1 0.2 1.0 1 0.1 50 Table 2: B-Arr1-KO mice had similar levels of Ad5-triggered activation of genes in spleens, as compared to WT mice. Values represent Mean 1: SD. Statistical analysis was completed using One Way ANOVA with a Student-Newman-Keuls post-hoc test, p<0.05 was deemed statistically significant. N=3 for Mock-injected groups, N=4 for Ad5-LacZ injected groups. Significant differences compared to WT_Mock are highlighted in light grey color. Significant differences in transcriptional activation in B-Arr1-KO (Ad5-LacZ) group compared to WT (Ad5-LacZ) group are indicated in table by black frame and boldface font. * Indicate significant differences between mock-injected animals. For full gene names please refer to abbreviation list. 51 Ad5-LacZ Induced gene expression in the spleen (fold over C57BL/ 6 Mock, 6 h.p.i.) CS7BL/6 B-Arrl—KO CS7BL/6 [AdS- B-Arrl-KO (Ad5- (Mock) (Mock) LacZ) LacZ) ADAR 1.0 1 0.6 1.5 1 0.4 2.7 10.2. :4 ' f ‘ 3.0.1 0.2;.“ 1‘. c014 1.0 1 0.5 1.9 1 0.7 2.0 1 0.1 1.5 1 0.5 CXCL-9 1.1 1 1.0 3.3 1 1.0 12.4129. 4 31.9 13.11:. 2 . _e-Selectin 1.0 1 0.2 1.5 1 0.4 1.2 1 0.1 1.5 t 0.3 ICAM 1.0 1 0.4 1.3 1 0.1 , 4.1-10.2 -~ '3 -' «43111164?- " “I IFNd 1.0 1 0.6 2.0 1 0.8 1.8 1 0.6 1.7 1 0.3 [RF-7 1.0 1 0.3 1.4 1 0.3 34.3 14.7 ”38.4 11.1“} , IRF-8 1.0 1 0.3 1.2 1 0.2 2.6 1 0.4 2.41.0.3 ‘ , j jak-l 1.0 1 0.5 1.9 1 0.3 0.7 1 0.1 0.7 1 0.1 MyD88 1.0 1 0.1 1.5 1 0.4 3.3 1 0.3 3.4 10.4:- "1 .1 NFkB-ReIA 1.0 1 0.4 0.8 1 0.1 0.9 1 0.1 1.0 1 0.4 OAS-la 1.0 1 0.5 2.6 1 04* 13.4 12.6 7 11.441113 , , , . _..; socs-1 1.1 1 0.8 1.3 1 0.2 13:91 02 a " {33.471110 ' is, socs-3 1.1 1 0.9 2.2 1 0.1 . 30.413.6‘ . . 33.3 1'1Q.a:,-._ ; TBK-l 1.0 1 0.1 1.0 1 0.1 ' x: 91.61 0.1 I ' , . 2.110135} I TLR-2 1.0 1 0.4 1.6 1 0.1 3.8 10.1 ' 46311.0 r ' ,1: TLR-3 1.0 1 0.3 1.9 1 0.2* 10.0 1 0.6 . ., 1.6)1f‘oféi91‘f f TLR-6 1.0 1 0.5 1.6 1 0.5 0.9 1 0.1 1.0 1 0.1 , TLR-9 1.1 1 0.8 1.1 1 0.1 ' 14.3 10.7 5.2 11.1" 35.1 TRAFpr 1.0 1 0.1 1.3 1 0.1 1.3 1 0.1 1.7 1 0.5 52 Table 3: B-ArrZ-KO mice had significantly higher levels of Ad5- triggered activation of a number of genes in livers, as compared to WT mice. Values represent Mean 1 SD. Statistical analysis was completed using One Way ANOVA with a Student-Newman-Keuls post- hoc test, p<0.05 was deemed statistically significant. N=3 for Mock- injected groups, N=4 for Ad5-LacZ injected groups. Significant differences compared to WT_Mock are highlighted in light grey color. Significant differences in transcriptional activation in B-Arr2-KO (Ad5-LacZ) group compared to WT (Ad5-LacZ) group are indicated in table by black frame and boldface font. * Indicate significant differences between mock-injected animals. For full gene names please refer to abbreviation list. 53 Ad5-LacZ induced gene expression in the llver (fold over C57BL/6 Mock. 6 h.p.l.) C57BL/6 B-ArrZ-KO CS7BL/6 (AdS- B-ArrZ-KO (Ad5- (Mock) (Mock) LacZ) LacZ) . ADAR 1.0 1 0.1 0.7 1 0.1 1.3 1 0.3 * 21.6 10.11 ‘ c014 1.0 1 0.2 0.9 1 0.1 2.1 1 1.2 1.9 1 1.2 CXCL-9 1.0 1 0.3 0.6 1 0.1 5.2 1 0.7 ‘ "9".9;1E‘f2i‘l. _. DAF 1.0 1 0.1 0.7 1 0.1 1.1 1 0.2 0.9 1 0.1 e-Selectin 1.0 1 0.2 0.7 1 0.2 2.0 1 0.0.7 ‘ “33.0 111 ‘ » lCAM 1.0 1 0.2 0.7 1 0.2 1.9 1 0.2 ' ‘ I 7 3.231046; lFNa 1.0 1 0.2 0.7 1 0.1 0.6 1 0.2 0.6 1 0.1 IFNB 1.0 1 0.2 0.6 1 0.1 0.6 1 0.2 0.6 1 0.1 lRF-3 1.0 1 0.1 0.6 1 0.1* 0.7 1 0.1 0.6 1 0.1 [RF-7 1.0 1 0.1 0.7 1 0.1 ' . 9112.4 . ‘ . 1 ~ ‘ [RF-8 1.0 1 0.1 0.7 1 01* 1.6 1 0.4 2.01 f . , lak-l 1.0 1 0.1 0.7 1 0.1* 0.7 1 0.2 0.7 1 0.1 Jak-3 1.0 1 0.1 0.7 1 0.1 1.0 1 0.3 1.11 0.1 MyD88 1.0 1 0.1 0.6 1 0.1* i -' 2.7.11.0 1411; {1.0 "i ‘ NFkB-RelA 1.0 1 0.1 0.7 1 0.1 1.0 1 0.3 1.4 1 0.2 NOD-1 1.0 1 0.3 0.8 1 0.1 1.1 1 0.1 1.4 1 0.1 NOD-2 1.0 1 0.2 0.7 1 0.1 0.7 1 0.1 0.9 1 0.1 OAS-1a 1.0 1 0.2 0.6 1 0.1 1.5 1 0.4 2,210.3 ‘ socs-1 1.0 1 0.1 0.8 1 0.1 3.9 1 1.4 75.113, .‘ socs-3 1.0 1 0.4 0.9 1 0.6 2.2 10.7 ' 415' 10.5 . j j .1 _ TBK—l 1.0 1 0.1 0.8 1 0.1 ' 17 1 0.4 . 10.5 "f TLR-2 1.0 1 0.1 0.9 1 0.3 18.6 1 8.5 75312” ,v ,' TLR-3 1.0 1 0.1 0.6 1 0.1* 5.9 1 1.3 , 9.0 1 3.0 TLR-6 1.0 1 0.1 0.9 1 0.1 1.2 1 0.2 1.6 1 0.4 TLR-9 1.0 1 0.1 0.6 1 01* 1.2 1 0.3 1.2 1 0.1 TRAFpr 1.0 1 0.3 0.6 1 0.1 3.7 1 1.5 11512.7 ’ ' TRAF6 1.0 1 0.1 0.8 1 01* 0.9 1 0.1 0.8 1 0.1 TRlF 1.0 1 0.1 0.9 1 0.1* 0.9 1 0.2 1.1 1 0.2 TRlM30 1.0 1 0.1 0.6 1 0.1* . 6.7 1 2.3 8.7 1 1.0 VCAM 1.0 1 0.1 0.8 1 0.1 0.7 1 0.2 0.9 1 0.1 54 Table 4: B-ArrZ-KO mice had significantly higher levels of Ad5- triggered activation of a number of genes in spleens, as compared to WT mice. Values represent Mean 1 SD. Statistical analysis was completed using One Way ANOVA with a Student-Newman-Keuls post- hoc test, p<0.05 was deemed statistically significant. N=3 for Mock- injected groups, N=4 for Ad5-LacZ injected groups. Significant differences compared to WT_Mock are highlighted in light grey color. Significant differences in transcriptional activation in B—ArrZ-KO (Ad5-LacZ) group compared to WT (Ad5-LacZ) group are indicated in table by black frame and boldface font. * Indicate significant differences between mock—injected animals. For full gene names please refer to abbreviation list. 55 Ad5-LacZ Induced gene expression in the spleen (fold over C57BU6 Mock, 6 h.p.i.) CS7BL/6 B-ArrZ-KO C57BL/6 [AdS- B-ArrZ-KO (Ad5- [MockL (Mock) LacZ) LacZ) ADAR 1.1 1 0.8 0.9 1 0.3 1.3 1 0.5 1.6 1 0.1 CXCL-9 1.1 1 0.9 1.2 1 0.9 3.6 1 3.3 4.61 1.9 e-Selecu'n 1.0 1 0.2 1.0 1 0.3 0.7 1 0.1 1.0 1 0.2 lCAM 1.0 1 0.5 0.5 1 0.2 1.3 1 0.4 lFNa 1.0 1 0.1 1.6 1 1.1 1.5 1 1.0 lRF-7 1.0 1 0.1 0.7 1 0.2 21.41 53 [RF-8 1.0 1 0.1 0.7 1 0.4 0.9 1 0.2 MyD88 1.0 1 0.4 1.0 1 0.1 25 1 0.1 NFkB-RelA 1.0 1 0.3 0.7 1 0.6 0.7 1 0.4 OAS-1a 1.0 1 0.1 3.4 1 3.1 12.9 1 33 ' socs-1 1.0 1 0.4 0.11 1 004* 1.9 1 1.0 socs-3 1.0 1 0.4 2.9 1 0.4* ' 12.5 1 4.4 TBK-l 1.0 1 0.2 0.7 1 0.4 0.9 1 0.5 TLR-2 1.0 1 0.3 0.9 1 0.3 2.0 1 0.2 TLR-3 1.0 1 0.3 2.2 1 1.4 ‘ 6.11 0§ ' " ' TLR-6 1.0 1 0.3 2.0 1 1.4 0.9 1 0.1 TLR-9 1.0 1 0.2 0.6 1 0.3 1.5 1 1.3 TRAFpr 1.0 1 0.7 1.1 1 0.5 1.0 1 0.2 56 immune responses, whereas similar responses are positively mediated by B- Arr1 . 2.3. Discussion Ad-based vectors possess an enormous potential for numerous gene transfer applications based upon their broad tropism, highly efficient transductional capability, and their ease for scalable production. Based upon these facts, Ad vectors have been the most utilized gene transfer vector in humans to date (http://www.wiley.co.uk/wileychi/genmedlclinicall). However, the robust innate immune response elicited shortly after intravascular Ad delivery poses a significant limitation to the use of this vector for numerous applications that could benefit from such administrations, such as gene transduction into the liver. lntravascular delivery of Ads into animal models facilitates detection of Ad induced innate immune responses, which may not be detectable when other routes of administration (such as intramuscular) are utilized. These innate responses include acute thrombocytopenia (26, 44), robust cytokine and chemokine releases (24, 26, 28, 36), activation of endothelial cells (45, 129) and inductions of inflammatory gene networks in multiple tissues, including both the liver and the spleen (23, 26, 46). It is well established that Ad5-induced innate immune responses are mediated by the TLR adaptor proteins MyDBB and TRIF. As a receptor that utilizes both MyD88 and TRIF, TLR4 is activated by LPS (41-43). Because both B—Arr1 and B-Arr2 proteins have been shown to modulate LPS induced TLR4 57 mediated signaling, we sought to determine if these proteins also played a role in Ad5-induced innate immune responses. Utilizing B—Arr1-KO and B—An’Z-KO mouse models, we show that whereas B-Arr1 served as positive regulator of a portion of Ad5 induced innate immune responses, B—Arr2 appears to function as a negative regulator of Ad5 induced innate immune responses. For example, following intravenous Ad5 injection into B-Arr1-KO mice, we observed significantly reduced levels of pro-inflammatory mediators including IL- 6, lL-12(p40) and KC, relative to levels measured in Ad5-injected WT mice. Furthermore, we observed significantly reduced transcription of innate immune response genes in transdcued tissues of Ad5-injected B—Arr1-KO mice. We additionally detected reduced expression levels of MCP-1, MlP-1B, and RANTES in peritoneal macrophages from B-Arr1-KO mice following exposure to Ad5 vectors in vitro. The results suggest that B-Arr1 may serve as a mediator of Ad- induced immune responses downstream of TLR signaling, specifically those that require MyDBB as an adaptor. Consistent with this notion, we have found significantly reduced activation of IRF7, TLR3 and TLR6 genes in livers of [SAM- KO mice, which further suggest the role of B—Arr1 in mediating TLR/MyD88/IRF7 pathways. The findings also highlight a relatively underappreciated role for B—Arr1 as a positive regulator of pro-inflammatory responses, a role that contrasts the previously reported role for B-Arr1 as playing a negative role in the induction of cytokine responses following LPS challenge (41). Our present results however, are consistent with our recent study where we found that B-Arr1 is necessary for 58 the sustained production of some cytokines/chemokines after LPS challenge in vivo (Porter et al, 2009. Manuscript submitted for publication). The role of B-Arr2 in the induction of innate immune responses by Ad vectors was diametrically opposite to that of B-Arr1. This was evidenced by significantly higher inductions by Ads of multiple cytokines and chemokines, and inflammatory gene expression responses after injection into B-ArIQ-KO mice. These responses were corroborated in vitro as detected in Ad5 infected peritoneal macrophages, suggesting that this cell type may play an important role in mediating this response in vivo. Interestingly, the profile of innate immune responses observed in B-Arr2-KO mice paralleled the role that TLR4 plays in innate immune responses noted after systemic Ad5 injection(130). Our previous work indicated that TLR4 plays a negative role in the induction of IL-12(p40) and G-CSF following intravenous Ad5 injection. It is possible that the negative role that TLR4 plays in Ad5 induced innate immune responses is mediated by B-Arr2, such that loss of either protein results in a more pronounced innate immune response triggered by Ad5. Our results also suggest that B-Arr2 may act as a negative regulator of TLR2/MyD88/IRF7 pathways. However, it is also clear that other factors must also play a role in these responses, since the phenotype in Ad5-injected B—ArrZ-KO animals did not completely mimic that found in Ad5- injected TLR4-KO mice (130). Interestingly, in a recent study, we found that B- Arr-2 also mediates LPS/TLR4-induced cytokine responses in vivo (Porter et al, 2009, Manuscript submitted for publication). 59 In summary, our data demonstrate important roles for both B-Arr1 and B- Arr2 as positive and negative regulators in modulating Ad5 induced innate immune responses. B-Arr-1 and -2 interaction partners that mediate B-arrestin’s actions in response to Ad5 administration may include proteins of the TLR pathways or other unique proteins of the Ad5 responsive signaling pathways. Acknowledgements We wish to thank Michigan State University Laboratory Animal support facility for their assistance in the humane care and maintenance of the animals utilized in this work. 8.8.8. was supported by American Heart Association Midwest Affiliate Fellowship 08156606. A.A. was supported by the National Institutes of Health grants RO1DK-069884, P01 CA078673, the MSU Foundation and the Osteopathic Heritage Foundation. N.P. is supported by the National Institutes of Health grants R01AR056680, R01 HL095637, and R21AR055726. 60 Chapter III Ad interactions with the complement system are pivotal in understanding how to maximize the safety and potency of Ad mediated gene transfer for both gene therapy and vaccine applications This chapter is the edited version of a research article that was published in the Gene Therapy Journal, Volume 16, Issue 10 (1245-1259), June 25, 2009. Authors: Seregin, S. S., Y. A. Aldhamen, D. M. Appledom, N. J. Schuldt, A. J. McBride, M. Bujold, S. Godbehere, and A. Amalfitano. 61 3.1. Introduction Adenovirus (Ad) based vectors offer tremendous capabilities as gene transfer platforms. However, Ad-triggered innate immune responses and Ad- specific neutralizing antibodies hinder the ability to repeatedly administer the vector and preserve transgene expression for long periods of time, prompting multiple efforts to develop alternative Ad vectors to overcome this problem. It is with these realities in mind that studies investigating the mechanisms underlying the induction of neutralizing antibodies to the well-characterized Ad5 vector platform are required. Importantly, the Ad5 serotype is the only Ad serotype utilized in all human gene transfer clinical trials (>367 as per September 2008, comprising 24.9% of all gene therapy trials to date), please see http://www.wiley.co.uklwileychi/genmed/clinicall). Our previous studies have confirmed that the induction of neutralizing antibodies to Ads is dependent upon the presence of a functional complement system in the host (24). Although we have discovered that the protein C3 is essential in this response, the mechanisms underlying CS-dependent induction of neutralizing antibody by Ad vectors is currently unknown. Our previous results confirm that lack of Ad interactions with the C3 protein results in a diminished induction of pro-inflammatory cytokines and acute phase responses early after Ad administration, suggesting that a lack of this initial response may contribute to diminished induction of neutralizing antibody responses. This hypothesis is supported by recent studies in lFN receptor KO mice that also showed 62 diminished induction of neutralizing antibody titers after Ad treatments (131). We have utilized Ad treated CR1I2-KO mice in this study to test this hypothesis. It is known that complement system activation acts to optimize induction of pathogen specific antibody responses (57-59). Subsequent to opsonization by activated C3, pathogens are bound to B cells and dendritic cells via binding of pathogen bound C3 to the complement receptors (CRs). The human CR1 and CR2 receptors have well known roles in modulating both innate and adaptive immune responses. Human CR1 (hCR1) is a potent inhibitor of complement activation, having both decay-accelerating and cofactor activities. Furthermore, hCR1 has a critical role in the clearance of immune complexes and B cell maturation, as thoroughly reviewed elsewhere (60, 61). Human CR2 (hCR2) expression is restricted to the surface of B cells, follicular dendritic cells and thymocytes. When hCR2 binds C3d opsonised pathogens and becomes associated with 0019, it lowers the threshold for B cell activation by up to 1000 fold (62). HCR1 and hCR2 also play a role in T cell biology, for example, crosslinking of hCR1 inhibits T cell proliferation and lL-12 production (63). Murine complement receptors (mCRs) 1 and 2 (CD35ICD21) are products of alternative splicing from the same gene. mCR1 contains 21 complement control protein repeats (CCPRs), whereas the smaller mCR2 contains only 15 C- terrninal CCPRsof mCR1 (64). mCR1/2 is known to be expressed on B cells and dendritic cells (64). Therefore, the expression pattern of mCR1/2 resembles that of hCR2, but not hCR1. Similar to their human homologues, the functions of mCR1/2 related to generation of maximal humoral responses have been well 63 described (65-69). Interestingly, mCR1/2 functionality prevents excessive myocardial tissue damage subsequent to coxsackievirus B3 infection (70), as well prevents lethal Streptococcus pneumoniae infection, a role potentially indirectly reflective of the complement inhibitory activities of the CRs (71). While the role of murine CR1/2 protein has been extensively studied in regards to adaptive immune responses, its function in inhibiting/regulating murine complement has not been demonstrated, possibly since in most mouse models, the Crry protein was suggested to play the predominant role in controlling complement activation. We think that the role of murine CR1/2 protein in innate immune responses (including the ones which are known to be complement dependent) may be more important than previously considered, as suggested by our present studies of Adenovirus mediated gene transfer into mCR1/2-KO mice. Our results in murine models revealed dual roles for mCR1/2; roles that include down—regulation of multiple aspects of the Ad induced innate immune responses, while also playing the major role in the complement dependent induction of neutralizing antibody responses to Ads. 3.2. Results Murine Complement Receptor 1I2 regulates Ad mediated cytokine and chemokine release in CSTBUB mice. To study the role of mCR1/2 protein in Ad induced innate and adaptive immune responses, we utilized mCR1/2-KO mice. These mice have been previously demonstrated to completely lack expression of CR1/2 on B cells (71). It is also known that CR1/2 activities also impact upon levels of activated C3, by virtue of CR1/2’s decay accelerating properties. Utilizing western blotting with C3-specific antibodies, we confirmed that mock- injected CR1I2-KO mice have normal overall levels of C3 no different than wild- type mice, and equivalent amounts of C3 cleavage products were present in the plasma of virus injected WT and CR1l2-KO mice, as investigated both at 10 minutes post injection and 6 hours post injection (Figure 10). These results suggest that CR1I2-KO mice do not have significant alterations in the ability of C3 to initially interact with Ads, an interaction that we have previously confirmed mediates many Ad induced innate and adaptive immune responses (23-26, 46). Inflammatory cytokines and chemokines are rapidly released after systemic Ad injection. We have identified 7 cytokines and chemokines (KC or CXCL1, MCP-1, MIP-1B, G-CSF, RANTES, lL-6, IL-12p40) that become significantly elevated within hours of systemic administration of Ad vectors, some of which are elevated in a C3—dependent fashion (24, 26). We investigated the role that mCR1/2 has in the induction of these cytokines by administering Ads into wild type and mCR1/2 knockout mice. Plasma samples, collected at 1 and 6 65 Figure 10: Plasma basal levels and Ad dependent activation of complement protein C3 were identical between CSTBIIG WT and CR1I2-KO mice. Plasma samples from WT C57BU6 and CR1/2-KO mice were collected and Western blotting was performed as described in Materials and Methods utilizing quantitative Licor’s Odyssey system. Intact and activated a-chains of C3 protein are shown. There were no significant differences in levels of basal C3 protein or C3 protein cleavage detected. 66 N8..-mu N03¢n 3.99% i I... . . ~ “ % . % _ % m 5 W 6 _ m 9 w J . . . . _ 1 . . ..c\ 4 . J.v 4 r... . L .. U. W . a 2 la . m, . WT DEX Ad5 Iono 6:80:23 9:08 moo lNFy positive 161 5.3. Discussion Toxicities that rapidly develop after intravenous injection of Ad vectors is a major problem, limiting the usage of Ads in gene therapy applications requiring systemic administration; i.e.: for high-level liver transduction (46, 78, 183, 184). To improve the risk/benefit ratio of systemic administration of Ad vectors requires simultaneous blockade of several Ad induced innate immune responses, such as acute thrombocytopenia (44), cytokines/chemokine release (26, 185, 186), induction of pro-inflammatory gene expression (23, 25, 26, 46) and activation of endothelial cells (45, 129, 132). In this study, we attempted to block all of these responses with a simple pretreatment with a clinically convenient and the widely utilized glucocorticoid, in this instance, Dexamethasone. Our results confirmed that transient pretreatment of mice with several doses of Dexamethasone can abrogate most innate toxicities attributable to systemic delivery of Ad-based vectors, improving the risk/benefit ratio of this vector class for numerous applications such as liver gene therapy. The drug dosages used in our studies parallel doses routinely utilized in humans, doses that range from those attempting to capitalize upon the anti-inflammatory effects of DEX to treat mild conditions, to doses used to treat complications of sepsis and clinical shock (187- 189). The mechanism of action of glucocorticoids is relatively well known. Glucocorticoid hormones bind to their respective glucocorticoid receptors (GR), which dimerize and translocate to the nucleus, where they bind to specific DNA sequences, the Glucocorticoid response elements (GRE) and either activate 162 transcription of anti-inflammatory genes (IL-10, lL-1_RA) or indirectly downregulate transcription of a number of pro-inflammatory genes (cytokines, adhesion molecules) through a variety of mechanisms (113-115). Glucocorticoids are known to change chromatin structure allowing for increased (or decreased) accessibility for transcriptional machinery (173). A number of reports confirm the role of high dose glucocorticoids in reducing transcription of pro-inflammatory genes mediated by GRs (113-115), in particular in LPS-induced models (190- 192). Systemic activation of the innate immune system can also be minimized by high dose DEX pre-treatment (or other glucocorticoids) at least in part by GR- mediated blocking of pro-inflammatory genes transcription (127, 187, 190, 192, 193). Based upon this analysis, we thought that many of the same innate immune response pathways that are induced by Ad vectors may also be impacted upon by glucocorticoids. Clearly, our results confirm this notion, as DEX pretreatment can minimize or prevent numerous Ad vector induced innate immune responses. In Figure 36, we have attempted to summarize some of the known innate immune response pathways induced by Ad that appear to be responsive to DEX pretreatment (Figure 36). We found that DEX pre-treatment significantly diminished the Ad dependent activation of most innate immune responses noted after systemic delivery of Ads (44-46, 129, 132, 185, 186). Specifically, the systemic release of pro- inflammatory cytokines/chemokines within 6 hours of Ad injection was partially or completely blocked by pretreatment of mice with escalating doses of DEX. At 163 maximal DEX dosages, Ad induction of thrombocytopenia was prevented, the transcriptional activation of multiple, pro-inflammatory liver genes at 6 hpi was completely blocked, and endothelial cell activation were all mitigated by transient DEX pre-treatment of Ad-injected mice. DEX pre-treatment also blocked leukocyte infiltration into the Ad transduced liver at 28 dpi. Our previous studies and those of others suggests that this effect was. likely mediated by significantly altering Ad interactions with the TLR and complement systems, since these systems are known to mediate Ad interactions with macrophages, dendritic cells, mast cells, endothelial cells, and/or granulocytes (23-26, 35, 36, 46, 163, 194, 195). Cellular infiltration of the liver by elements of the mammalian innate immune response is dependent upon activation of endothelial cells. Elevated expression of adhesion molecules on their surface, such as the P and E selectins, mediate tethering and rolling of leukocytes, whereas increased expression of ICAM-1, lCAM-2 and VCAM-l mediate firm adhesion of leukocytes. It is known that high dose Ad vectors cause rapid activation of endothelial cells both in vitro (45) and in vivo (45, 129, 132). We have shown that plasma levels of endothelial cell derived activation markers were induced at least 3 fold in Ad-injected mice as compared to mock-injected animals (e-Selectin, lCAM-1). This induction was completely blocked with DEX pre-treatment of Ad-injected mice, a result verified by qRT-PCR analysis of lCAM-1 and VCAM-1 gene transcriptions. Since we confirmed that systemic Ad injection into DEX pretreated mice still resulted in 164 Figure 36: Dexamethasone pre-treatment mediates blockage of Ad induced innate immune responses: model of action. Ad interactions with the complement system, as and/or Ad capsid triggered TLR signaling results in nuclear translocation of inflammatory Transcription Factors, such as NFkB, IRF7 and others. The expression of a number of genes involved 0 in innate immune responses becomes upregulated in response to systemic injection of Ads. This leads to cytokine and chemokine release and signal amplification by autocrine and paracrine cytokine signaling. DEX, when injected prior to Ad treatment, causes Glucocorticoid Receptor dimerization and nuclear translocation, where GR interacts with genes containing the target DNA sequence (GRE). Thus, GR activation by DEX pretreatment interferes with induction of transcription of pro-inflammatory genes, to block Ad induced transcription of adhesion molecules (lCAM-1, VCAM-1) minimize systemic cytokine release and therefore in lack of induction of IFN responsive genes, such as Oas1a or ADAR. See (23-26, 35, 36, 44-46, 129, 132, 163, 185, 186, 194, 195) for further details. 165 3&2 sagas» 2...; I \ 166 Kupffer cell necrosis, we have now confirmed that Kupffer cell dependent innate immune responses induced by Ads are likely consequent to immediate interactions with Kupffer cells, rather than due to Ad induced Kupffer cell necrosis per se (36, 129, 196). Interestingly, DEX pre-treatment did not have any effect on Ad-dependent Kupffer cell necrosis, which suggests that DEX does not mediate damage associated molecular pattern molecules inflammatory responses. We further confirmed that pre-treatment with DEX did not prevent Ad vectors from transducing the murine liver with transgenes. Interestingly, DEX pre- treatment increased the detectable number of Ad genomes in the murine liver early after injection, but this did not elevate Ad derived B-Gal expression levels in murine liver at 24 hpi and 28 dpi. This paradox may be due to the fact that in the vectors utilized in this study the LacZ gene was expressed under control of the CMV promoter/enhancer element. This enhancer is known to contain Nuclear factor KB (NF-KB) response elements (197). NF-KB is a key transcription factor activating pro-inflammatory gene expression, and is induced over 2 fold in the livers of Ad5-LacZ treated C57BU6 mice at 6 hpi compared to Mock-injected animals. In contrast, DEX pre-treated virus injected animals had NF-KB transcript levels no different from mock-injected animals; in fact they had 1.6 fold less NF- KB transcripts compared to WT_Mock group. This over 3.5 fold difference between DEX treated and non-treated Ad5-LacZ injected animals may explain why CMV promoter/enhancer derived LacZ gene transcription did not correlate with Ad vector genome copy numbers (198). 167 DEX pre-treatment of mice minimized Ad induced innate immune responses, an effect that also resulted in blunted adaptive immune responses. Neutralizing antibody titers, anti-Ad and anti-LacZ antibody titers were significantly reduced in DEX pre-treated Ad-injected mice relative to non-DEX treated, Ad vector treated mice. The fact that DEX pre-treatment diminished the generation of anti-Ad NAb may have significant applicability in Ad re- administration scenarios (199). In summary, results obtained in this study confirm that the simple pre- emptive and transient use of a potent glucocorticoid prior to systemic delivery of an Ad vector can allow for safer systemic Ad mediated gene transfer. This approach avoids the complications associated with long-terrn glucocorticoid usage, while simultaneously significantly improving the safety profile of systemic Ad administration for use in gene therapy strategies requiring widespread liver transduction. Furthermore, this method can be simply applied in conjunction with use of either advanced generation Ad vectors, and/or other pre-emptive strategies previously shown to impact on Ad induced innate immune responses to further improve the safety profile of this important gene transfer platform. These benefits, coupled with DEX dependent diminishment of generation of Ad specific neutralizing antibodies further highlights the importance of this study. 168 Acknowledgement: We wish to thank Michigan State University Laboratory Animal support facility for their assistance in the humane care and maintenance of the animals utilized in this work. 8.8.8. was supported by American Heart Association Midwest Affiliate Fellowship 0815660G. A.A. was supported by the National Institutes of Health grants RO1DK-069884, P01 CA078673, the MSU Foundation as well the Osteopathic Heritage Foundation. 169 Chapter VI Materials and Methods 170 6.1. Adenovirus vector construction 6.1.1. Incorporation of COMPinh in HI loop of the fiber protein All novel Ad vectors were constructed utilizing pAdEasy based system (200) with modifications. pAdEasy plasmid was digested with Spa! and Feel restriction enzymes and the 6.20 kb fragment containing the fiber gene was gel purified and subcloned into the pBSX plasmid, giving rise to pBSX-FiberHI. The Fiber HI loop was flanked with in frame, Natl and Xbal restriction sites, using an approach similar to that described in Fontana et.a|. (201) generating pBSX- FiberHI-I-Not/Xba. Next, 45-mer complementary oliogonucleotides, encoding the 13-mer COMPinh nucleotide sequence were synthesized. When hybridized together, these oligomers yielded Net! and Xbal compatible overhangs, allowing in-frame subcloning into the HI fiber loop of pBSX-FiberHl+Not/Xba. The plasmid obtained, pBSX-FiberHI+Not/Xba-COMPinh, was digested with Spa! and Feel and the 6.25 kb fragment was cloned back into the pAdEasy backbone, giving rise to pAd-Fiber-COMPinh. Bacterial homologous recombination of pAd-Fiber- COMPinh with Pmel linearised pShuttle-CMV-LacZ yielded the pAd-CMV-LacZ- Fiber-COMPinh plasmid. Further manipulations with this plasmid are identical to the ones described below. 6.1.2. Incorporation of COMPinh in the C-terminus of protein IX Oligonucleotides encoding COMPinh with Nhel compatible ends were subcloned in-frame into the C-terminus of viral protein IX into pShuttle-lX/Nhel, the latter contains an Nhel site just upstream of the pix stop codon (101). A LacZ 171 expression cassette was inserted into the MOS of the pShuttle-lX-COMPinh as previously described (15, 26). pShuttle-LacZ-lX-COMPinh, was linearised with Pmel restriction enzyme and homologously recombined with the plasmid pAdEasyl (200), yielding pAd-LacZ-lX-COMPinh plasmid. HEK293 cells were transfected with Pacl linearised Ad5-LacZ-lX-COMPinh or Ad5-LacZ-Fiber- COMPinh plasmids. Recombinant viable viruses were isolated, amplified, and purified in CSCIz gradients as previously described (185, 202). Note that a complete list of primer sequences utilized for construction and validation studies is in table 8. All viruses were designed to be [E1-,E3-] and found to be RCA free (23). 6.2. Adenovirus vector production and characterization A first-generation, human Adenovirus type 5 derived replication deficient vector (deleted for the E1 and E3 genes) encoding B-galactosidase (LacZ) as a transgene (Ad5-LacZ) was used in these studies. Virus construction, propagation and purification was performed as previously described (27, 28, 185, 202). Briefly, a number of serial passages on HEK293 cells allowed high titer purification of Ad5-LacZ by sequential, cesium chloride density gradient centrifugations. Purified virus was dialyzed against 10 mM Tris (pH 8.0) and stored in 1% sucrose, 1 X PBS at -80° C until use. VIral particle (vp) and transducing unit titers (bfu/ml) were determined as previously described, and were 2.6 x 1012 vplml and 1.8 x 1011 bfulml respectively (15, 26). The vp to bfu ratio was ~14:1. Infectious titers of all Ads were determined by standard Tissue 172 Table 8: Complete list of primers and oligos, utilized to construct and validate Ad5-based vectors. Oligos, utilized to subclone COMPinh into plX and fiber are shown. Important sequencing primers used to confirm integrity of plasmids constructed are also presented. Note, validation primers were used to sequence plasmids at all stages and purified virus derived DNA. 173 Primer name Primer sequence (5’-3’) Used for Comp-Nhe-F CTAGCatctgcgtgtggcaggattggggcgcccacaggtgcaccG compinh in plX Cloning Comp-Nhe-R CTAGngtgcacctgtgggcgccccaatcctgccacacgcagatg compinh in pIX Cloning Comp-Xba-Fc CTAGAatctgcgtgtggcaggattggggcgcccacaggtgcaccCG Cloning compinh in Fiber Comp-Not-Rc GGCCGngtgcacctgtgggcgccccaatcctgccacacgcagatT Cloning compinh in Fiber Knob-F1 AGGCAG'ITI'GGCTCCAATATCTG Natl/Xbal insertion in Fiber knob-XbalNot—R1 GCGGCCGCACCTCTAGATGTGTCTCCTGTITCCTGTGTA Natl/Xbal insertion in Fiber knob-XbalNot-FZ TCTAGAGGTGCGGCCGCTCCAAGTGCATACTCTATGTCATI’ T Natl/Xbal insertion in Fiber knob-R2 GCTATGTGGTGGTGGGGCTATACTA Natl/Xbal insertion in Fiber CMV-F TGGGAGTTTGTITI'GGCACC Sequencing transgene SV40polyA-R TTCATTTTATG'ITTCAGG'ITCAGGG Sequencing transgene plX-S EQ—F 1 GCAAGCAGTGCAGCTI’CCCG Sequencing plX display plX-SEQ-F2 GATCTGCGCCAGCAGG'ITI'C Sequencing plX display plX-SEQ-R CAGGACCCTCAACGACCGAG Sequencing plX display Ad5-Comp-R CCGCCCTATCCTGATGCACG Sequencing Fiber display Cominh-SEQ-F ATCTGCGTGTGGCAGGATI’G Sequencing compinh plX-upstream-R CCACGCCCACACATTTCAGTACC Sequencing of Ads to test for RCA 174 Culture Infectious Dose 50 (T CIDSO) method (AdEasy Adenoviral vector system manual, Qbiogene, Carlsbad, CA). Infections titer was calculated by using KARBER statistical method: TCID50/ml titer=10 x101 * “(Sf-5’, where d is the log(10) of the dilution and S is the sum of ratios from the first dilution. VP/T U/T CID results are summarized in table 6. Note, that VP/T UIT CID ratios of capsid-displaying Ads were not dramatically different from the same ratios for control Ads (table 6). These experiments validated the viability of novel capsid- displaying Ads. All viruses were found to be RCA free both by RCA PCR (E1 region amplification) and direct sequencing, methods as previously described (23). Ad vectors have also been tested for the presence of bacterial endotoxin as previously described (136) and were found to contain <0.01 EU per injection dose. 6.3. Electron Microscopy of purified Ad vectors Negative staining of CSCIz purified Ad vectors was performed as follows. Ads diluted to 1012 vplml in 10 mM Tris were adsorbed to FormvarlCarbon film 300 mesh Copper grids (Electron Microscopy Sciences, Hatfield, PA) and stained with a freshly prepared, 1% solution of phosphotungstic acid (1 g PTA, 50 pl of FBS, 50 ml miliQ water, pH 6.0, adjusted by KOH) for 30 seconds and examined by using transmission electron microscope (Philips EM410). Photographs were taken from representative areas from each sample (Figure 21). 6.4. Validation of VP titers of Ads 175 6.4.1. Silver Staining To verify that particle number quantification was accurate across all Ads constructed, 101o vps of lysed purified virions of each Ad were separated by 10% SDS-PAGE and subsequently stained with silver nitrate utilizing a Silver stain kit for proteins (Sigma, St. Louis, MO). The amount of hexon protein was quantified for each Ad vector by scanning densitometry using lmageJ software, ver. 1.29 (developed at the US. National Institutes of Health and available on the lntemet at http://rsb.info.nih.govlnih-image/). Results from this analysis indicated that the VP titers of all viruses determined by spectrophotometry fall within ~1.13 fold of each other, thus capsid-displaying Ad5 vector preparations did not contain less virions as compared to first generation Ad5 vectors, based on this assay (Figure 23 and data not shown). 6.4.2. Western Blotting To further verify that particle number quantification was accurate across all Ads constructed, 1010 of lysed purified virions of each Ad were separated by 10% SDS-PAGE and Western blotting was performed utilizing hexon specific antibodies (Abcam, Cambridge, MA). Electrophoretically separated capsid protein samples were transferred onto nitrocellulose membranes and probed with rabbit polyclonal Ad5 hexon specific antibody, followed by probing with a fluorescent secondary antibody as previously described (36). Membranes were scanned and hexon concentrations quantified using Licor's Odyssey scanner (36). Results from this analysis indicated that the VP titers determined by 176 spectrophotometry fall within ~1.3 fold of each other based on this assay, thus capsid-displaying Ad5 vector preparations did not contain less virions as compared to conventional first generation Ad5 vectors based on this assay (data not shown). 6.5. Capsid thermostability assay It has been shown that Ad capsids containing functional ple are more resistant to temperature inactivation as compared to Ad capsids lacking plX or plX functionality (203, 204). Therefore, we pre-incubated control Ads, and plX- COMPinh displaying Ads at 45° C or 56° C, infected HEK293 cells and determined the percentage of LacZ positive HEK293 cells as previously described (15, 26). We did not detect significant differences between the ability of Ad-LacZ-lX-dCOMPinh to transduce 293 cells compared to control vectors Ad- LacZ. This confirms that COMPinh displaying vectors contain a functional protein IX. Capsid thermostability assay was performed according to previously described protocol (101, 203, 204) with modifications. 400,000 HEK293 cells were plated in each well of a 24 well tissue culture plate. Ads were diluted from CsClz purified stocks in complete media (1.55x107 vp in 500 pl of media) and were either not heat treated, or heated to 45° C or 56° C for 1 hour. Following incubation viruses were added to 293 cells. Following a 12-hour incubation, the cells were stained with X—gal (15, 26) and the percentage of LacZ positive cells was determined for every sample. The results indicated that all Ads retained 177 about 10-20% of LacZ positive cells after incubation at 45° C as compared to the non-heat treated Ads. 6.6. Animal procedures Adult C57BU6 WT and B612984-C3tmlCrr (C3-KO) mice were purchased from Jackson Laboratory (Bar Harbor, ME). mCR1l2-KO mice in CS7BLI6 background were a kind gift from Dr. Tedder, Duke University Medical Center (71, 205). B-arrestin-1 and -2 KO mice have been backcrossed to CS7BL6 background for more than 10 generations and have been described previously (206). These mice were kindly provided by Dr. Robert Lefltowitz (Duke University). The Ad vector was injected intravenously (via the retro-orbital sinus) into 8- 10 week old male C57BU6 mice after performing proper anesthesia with isofluorane. A total of 0.75 x 1011 vp in 200 pl of PBS was injected per mouse. Dexamethasone (America Pharmaceutical Partners, INC, Schauburg IL, USA) was administered by intraperitoneal injection (10 mglkg, 1 mglkg, 0.1 mglkg), at 15 hours and 2 hours prior to Ad vector administration. Note, that 0.1 mglkg and 1 mglkg DEX was used only in measuring Ad induced cytokines/chemokines release. The highest dose of DEX was selected based upon current dose regimens utilized to treat bacterial sepsis and shock human clinical applications (187-189), Ad vector induced inflammatory responses closely resemble these conditions. These dosages have been widely used in a number of studies on animal models (127, 193). 178 Four groups of mice were analyzed in DEX study: Wild-type (WT) mice mock-injected with PBS, WT mice pre-treated with DEX and then mock-injected with PBS (to identify DEX mediated responses not directly attributed to Ad), WT mice injected only with Ad5-LacZ, and WT mice pre-treated with DEX and subsequently injected with Ad5-LacZ. Control and experimental mice were sacrificed at different times after mock or virus treatment: 6 hours post injection (hpi), (N=6 for virus injected groups, N=4 for Mock-injected groups), 24 hpi (N=4 for all groups), 28 dpi (N=5 for all groups). Four groups of mice were analyzed in CR1/2 study: C57BU6 Wild-type (WT) and CR1/2-KO mice, Mock-injected with PBS and C57BU6_WI' or CR1I2- KO mice injected with Ad5-LacZ. For some of the control experiments C3-KO mice were utilized (N=4 for all C3-KO groups). Mice were sacrificed at different times after mock or virus treatment: 6 hours post injection (hpi), (N=6 for virus injected groups, N=4 for Mock-injected groups), 24 hpi (N=4 for all groups), 28 dpi (N=5 for all groups). For both B-Arr and B-Arr2 four groups of mice were analyzed: VVIld-type (WT) mice mock-injected with PBS, B-Arr or B-ArrZ-KO mice mock-injected with PBS, WT mice injected with Ad5-LacZ, and B—Arr or B-Arr2-KO mice injected with Ad5-LacZ. Control and experimental mice were sacrificed at 6 hours after mock or virus treatment: N=4 for virus injected groups, N=3 for Mock-injected groups). Plasma and tissue samples were collected and processed at the indicated time points in accordance with Michigan State University Institutional Animal 179 Care and Use Committee. All procedures with recombinant Ads were performed under BSL-2, and all vector treated animals were maintained in ABSL-2 conditions. All animal procedures were reviewed and approved by the Michigan State University ORCBS and IACUC. Care for mice was provided in accordance with PHS and AAALAC standards. 6.7. CytokineIChemokineIEndothelial cells activation markers release measurement Ad induced systemic release of pro-inflammatory cytokines/chemokines in murine plasma was measured in all groups of mice utilizing a multiplex bead array system. Plasma samples were collected using heparinized capillary tubes and EDTA coated microvettes (Sarstedt, Nlimbrecht, Germany) and centrifuged at 3400 rpm for 10 min to retrieve plasma samples. Samples were assayed for 7 independent cytokines/chemokines, which we have previously shown to be rapidly induced by systemically injected Ad vectors (MCP-1, KC, MlP-1B, lL-6, lL- 12p40, G-SCF, RANTES) (23, 25, 26). All procedures were performed exactly as previously described according to manufacturer’s instructions (Bio-Rad, Hercules, CA) via Luminex 100 technology (Luminex, Austin, TX) (23). For in vitro experiments utilizing peritoneal macrophages, media collected at specified time points was assayed for the same 7 analytes as per manufacturer’s instructions. The measurement of soluble lCAM-1 and e-Selectin molecules (endothelial cells activation markers) in murine plasma (collected at 6 hpi) was performed utilizing mouse cardiovascular disease panel LlNCOplex kit (Millipore, Billerica, MA) as per manufacturer's instructions. 180 6.8. Cytokine quantification in human PBMCs Human PBMCs were plated as previously described (207). Briefly, PBMCs were resuspended in RPMI-1640 with 10% FBS, 1% PSF and plated into 24-well tissue culture plate at a concentration of 106 PBMClmI. Upon 24 hour incubation, cells were washed two times with HBSS and exposed to the following Ad5 vectors at a multiplicity of infection 5000 (5x109 vplwell): Ad5-LacZ, Ad5-LacZ-IX- dCOMPinh, Ad5-LacZ-Fiber—dCOMPinh. Media was collected at 6, 24 and 48 hpi, stored at -20° C until use, and levels of human lL-6,_ IL-8, RANTES, GCSF, lL-10, MOP-1 and MlP-1B measured utilizing a multiplex bead array system exactly as previously described (27, 28). Only levels of lL-6, "-8 and RANTES are reported, since levels of other analytes were not induced over Mock (IL-10, GCSF) or were not significantly different between Ad-injected groups (MOP-1, MlP-1 B). 6.9. Complete blood count analysis and cell type differentiation Total blood (0.3-0.4 ml) was collected into EDTA coated 1.0 ml Lavender tubes (BD Microtainer, Franklin Lakes, NJ) at 24 hpi. For complete blood counts (CBCs), blood was analyzed on an Advia 120 Hematology System (Bayer, New York) by the Clinical Pathology Laboratory of the Diagnostic Center for Population and Animal Health at Michigan State University (East Lansing, MI). In addition, all blood samples were examined microscopically and underwent manual differential count. 6.1 0. Platelet enumeration 181 To access Ad vector induced thrombocytopenia in mice, platelets were measured 24 hpi after systemic Ad injection by using Unopette (Fisher Scientific) system as previously described (23, 26) as per manufacturer's recommendations. Platelets were subsequently manually counted using Neubauer hemocytometer. 6.11. Ad genome copy number per liver or spleen cell To determine the number of Ad genome copies per spleen and/or liver cell at different time points post-transduction, tissues (<01 9) were snap frozen in liquid nitrogen, crushed to a fine powder using a mortar and pestle and total DNA was extracted from as previously described (208). Ad genome copy numbers were assessed using Real-Time PCR based quantification. PCR reactions were performed on an ABI 7900HT Fast Real-Time PCR System using the SYBR Green PCR Mastennix as described for qRT-PCR technique. Primers generated against the Ad5 Hexon gene have been previously described (46). As an intemal control for ensuring adequate DNA amplification, DNA was quantified using primers spanning the GAPDH gene. Standard curves were run in duplicate and consisted of 6 half-log dilutions using total genomic DNA, or DNA extracted from the purified Ad5-LacZ virus. These standard curves were used to determine the number of viral genomes present per liver/spleen cell. Melting curve analysis confirmed the quality and specificity of the PCR (data not shown). 6.12. B-Galactosidase enzyme activity and in situ X-gal staining 182 Ad mediated expression of the transgene LacZ was measured both qualitatively and quantitatively. Liver and spleen sections from animals sacrificed at 6 hpi, 24 hpi and (in case of DEX and CR1/2 experiments) 28 dpi were embedded in Optimal Cutting Temperature (OCT) compound, frozen and stored at -80° C until use. Frozen samples were sectioned (7 pm sections) on a Leica cryostat and were fixed and in situ stained for LacZ expression using 5-bromo-4- chloro-3-indolyl-b-D-galactopyranoside (X-Gal, 1 mg/ml) as previously described (138). For quantitative assay, enzyme B-Galactosidase (B-gal) activity was measured in snap frozen liver samples. Liver samples (<01 9) were homogenized and total protein concentration was determined by bicinchoninic acid (BCA) assay, Pierce (Rockford, IL). B-gal activity was quantified by use of a B—gal activity detection kit (Stratagene, La Jolla, CA) according to manufacturers instructions and as previously described (137). Data were reported as Units of [3- gal activity per pg of total protein. 6.13. qRT-PCR Analysis To determine relative levels of a specific, liver or spleen derived RNA transcript, corresponding tissues were snap frozen in liquid nitrogen and RNA was harvested from =100mg of frozen tissue using TRIzol reagent (Invitrogen, Carlsbad, CA) per the manufacturer's protocol. Following RNA isolation, reverse transcription was performed on 180ng of total RNA using SuperScript ll (Invitrogen, Carlsbad, CA) reverse transcriptase and random hexamers (Applied Biosystems, Foster City, CA) per manufacturer's protocol. RT reactions were diluted to a total volume of 60pl and 2pl was used as the template in the 183 subsequent PCR reactions. Primers were designed using Primer Bank web based software (http:llpga.mgh.harvard.edulprimerbankl). Some primers used for amplification have been previously described (26, 28, 46). Complete list of primers utilized in this study is available in table 9. Q-PCR was carried out on an ABI 7900HT Fast Real-Time PCR System using SYBR Green PCR Masterrnix (Applied Biosystems, Foster City, CA) in a 15pl reaction. All PCRs were subjected to the following procedure: 95.0° C for 10 minutes followed by 40 cycles of 950° C for 15 seconds followed by 60.0° C for 1 minute. The comparative Ct method was used to determine relative gene expression using GAPDH to standardize expression levels across all samples. Relative expression changes were calculated based on comparing experimental levels of a respective liver/spleen transcript to those quantified in liver/spleen samples, derived from Mock-injected animals. 6.14. Kupffer cell staining Liver blocks, preserved in OCT compound at -80°C, were sliced into 7pM sections using a cryostat, placed on glass slides and frozen at -80°C for future use. Slides were thawed, fixed in 100% ethanol for 15min, and washed in KPBS containing 0.2% gelatin and 0.05% tween-20. Sections were permeabilized in 0.1% triton-X100 and blocked with KPBS containing 0.1% gelatin, 1% tween-20, and 5% BSA for 60 min at RT. To prevent non-specific binding of the secondary antibody, 5% rabbit serum in wash buffer was added and incubated at RT for 30 min. Rat anti-mouse F4I80 antibody (Invitrogen, Carlsbad, CA) diluted to 5.2 pg/mL in wash buffer containing 2% rabbit serum was added to the sections and 184 incubated overnight at 4° C. Sections were washed and incubated at RT for 45 min with rabbit anti-rat Alexafluor—488 (Invitrogen, Carlsbad, CA) secondary antibody diluted 1:25.000 in wash buffer containing 2% rabbit serum. Sections were then washed, stained with DAPI (Invitrogen, Carlsbad, CA) for 5 min at RT and mounted using VECTASHIELD (Vector Laboratories, Burlingame, CA). Images were obtained using a Leica DMLB microscope, and captured using SPOT software v.3.5.9. Both DAPI and F4I80 stained images were transferred to Adobe Photoshop and converted to grayscale. The background threshold was set based on mock-injected livers, and pixels were quantified. F4I80 fluorescence values were normalized to DAPI fluorescence values to control for cell number differences. Values are reported as percent relative to number of Kupffer cells identically enumerated in liver tissues derived from mock-injected control mice. 6.15. Hematoxilin and Eosin staining To study Ad vector mediated hepatic inflammation (at 28 dpi), Hematoxilin and Eosin (H&E) staining of mouse liver samples was performed as previously described (137). Briefly, tissues were fixed in 10% neutral formalin for 12 hours, washed in 70% ethanol, embedded in paraffin and 6-pm sectioned were stained with H&E. We have adapted a previously developed semi-quantitative scoring system, which allows the level of hepatic pathology between different liver sections to be quantified and statistically compared (137). For every mouse, 10 liver sections obtained at different portions of the liver (0-1000 pm from liver surface) were analyzed and given a numerical score (0-3) for three different 185 Table 9: List of primers, utilized in qRT-PCR experiment. A pair of Fonivard (For) and Reverse (Rev) primers is provided for every transcript tested by qRT-PCR based methods. The primers were designed as described in Materials and Methods section; the length of resulted PCR products was 100-160 nucleotides. 186 ADAR CXCL-9» DAF GAPDH GATAS ICAM-l IFNa Imp IRE-'7 IRF- 8 Jak-l Jak-3 :My088 NFkB-RelA NOD - 1 NOD-2 OAS—1a SOCS-l SOCS-3 TEX-1 TLR-2 TLR-3 TLR-6 TRAFZbP TRAFG TRIF VCAM-l ADAR-Mm-Rev 5' CXCLQ-Mm-Por S' CXCL9-Mm-Rev 5' DAF~Mm For 5’ DAF-Mm-Rev 5' — GAPDH—Mm—For 5’ GAPDH-Mm-Rev 5’ GATA3—Mm—For 5’ GATAB—Mm-Rev 5’ ICAMl—Mm-For 5' ICAMl-Mm-Rev 5' IFNal—Mm—For 5’ IFNal-Mm—Rev 5’ IFNB-Mm-For 5’ IFNB-Mm-Rev 5’ IRF7—Mm—For 5' IRFI—Mm—Rev 5' TRF8-Mm—For S' IRF8—Mm—Rev 5' Jakl-Mm—For 5' Jakl-Mm—Rev 5’ Jak3—Mm—For 5' JakB—Mm-Rev 5’ MyD88—Mm-For 5' MyD88-Mm-Rev 5' NFkB-RelA-Mn-For 5' - NFkB-RelA—Mn-Rev 5’ NODl-Mm-For 5' NODI-Mm-Rev 5’ NODZ-Mm-For 5’ NODZ—Mm—Rev 5' OASla-Mm—For 5' CASla-Mm—Rev 5' SOCSi-Mm-Pnr 5' SOCSI-Mm-Rev 5' SOCSB—Mm~For 5' SOCS3-Mm-Rev 5’ TBKl—Mm—For 5' TBKl-Mm-Rev 5’ TLRZ-Mm—For 5’ TLRZ-Mm-Rev 5’ TLR3-Mm-For 5’ TLR3-Mm-Rev 5' TLR6—Mm-For 5' TLR6-Mm-Rev 5' TRAF2bp-Mm-For TRAFpr Mm-Rev IRAF6-Mm-F01 5’ TRAF6—Mm-Rev 5’ TRTP-Mm-For 5' TRIF—Mm-Rev 5' VCAMl-Mm-For 5' VCAMl—Mm—Rev 5' ADAR-Mm-For LI - AGGATTGGTSAGCTCGTCAG - CCCCTCTTTCTTCCTCTCTC - GCCCCAATTCCAACAAAACTC - CCTCTTTTGCTTTTTCTTTTGGCTG GGCCAATGGTCGAGCCACG CGAAATCCTGGCCGACACTC - AGAACATCATCCCTGCATCC - CACATTGGGGGTAGGAACAC - CCTACTACGGAAACTCCGTCAGG - GCCSCCATCCAGCCAGG - GGCATTGTTCTCTAATGTCTCCG - GCTCCAGGTATATCCGAGCTTC - GCCTTGACACTCCTGGTACAAATGAG - CAGCACATTGGCAGACGAASACAG - TGGGTGGAATGAGACTATTGTTG CTCCCACGTCAATCTTTCCTC - TGAACGAGGCTCGCACAGTC - GGCAGGTTAACTCCACTAGGTG - GTTTACCGAATTGTCCCCGAS - CTCCTCTGGSTCATACCCATGTA GCTGTGCATCAGGGCCGCC - GCGGTAGTGGAGCCGGAGAG - GTGTGCGAGCTGCCAAGG - GCCTGTAGACCAAGACTTGAGTGTCC - AGAGCTGCTGGCCTTGTTAG - TTCTCGGACTCCTGGTTCTG GGCGCTCAGCGGGCAGTATTC - CCACAAGTTCATGTSGATGAGGC - CCCCTTCCCAGCTCATTCG - TGTGTCCATATAGGTCTCCTCCA - CAGGTCTCCGAGAGGGTACTG GCTACGGATGAGCCAAATGAAG - CAGGAGGTGGAGTTTGATGTG - CCGTGAAGCAGGTAGAGAACTC - CTGCGGCTTCTATTGGGGAC — AAAAGGCAGTCGAAGGTCTCG CAAGAACCTACGCATCCAGTG - CCAGCTTGAGTACACAGTCGAA - AGGACCATCAGAAGAAGTACGG - CCCCTCGAASGGTCTAAACG - CCAGACACTGGGGGTAACATC - CGGATCGACTTTAGACTTTGGG - GGGGTCCAACTGGAGAACCT - CCGGCGAGAACTCTTTAAGTGG - AGGAACCTTACTCATGTCCCC - TGTTGTGGGACAGTCTCAGAA 5' - CAGGTTCGGAGAGTATCAGTTCC 5' - CTCTGGTATGGGATTCTSTGTTG - GCGAGAGATTCTTTCCCTGACG - CGTTGGCACTGGGGACAATTC - AACCTCCACATCCCCTCTHTT — CGGGCACCTSAAATTCCTCA - AGTTGGGGATTCGGTTGTTCT - CCCCTCATTCCTTACCACCC 187 categories of liver pathology: 1. Portal inflammation: 0 — no portal inflammation 1 — low-moderate number of inflammatory cells (macrophages, lymphocytes) evident in <1I3 of portal tracts 2 — moderate number of inflammatory cells in 1/3-2/3 of portal tracts 3 — high number of inflammatory cells in over 2/3 of portal tracts 2. Periportal inflammation 0 - no inflammation 1 - low-moderate number of inflammatory cells infiltration evident around <1I3 of portal tracts 2 — moderate number of inflammatory cells infiltrated through 1l3-2l3 of portal tracts, in majority of which they take less that 50% of circumference. Minimum hepato-cellular necrosis observed. 3 - moderate-high number of inflammatory cells infiltrated through over 2/3 of portal tracts or infiltrated through over 1l3 of tracts but occupy over 50% of circumference in at least 50% of them. Significant hepato-cellular necrosis observed. 3. Lobular inflammation / damage 0 — no inflammation 1 - minimum-moderate necrosis observed in <1I3 of lobules 2 - moderate hepatocellular necrosis observed in 1l3-2I3 of lobules 188 3 - moderate-severe hepato-cellular necrosis observed in over 2/3 of lobules Two independent researchers have scored all the slides in a blind manner and the averages of their scores were taken. The sum of scores (10 slides) for each mouse was taken and individual category scores were averaged for each group. Total inflammation index was computed by averaging the sum of all three individual category scores for each mouse. 6.16. lFNy secretion assay (Flow Cytometry) lFNy-secreting cells were detected using the APC-IFNy secretion assay (Miltenyi Biotec, Auburn, CA) according to the manufacturer’s instructions. Briefly, after harvesting splenocytes from individual mice at 28 days post injection, RBCs were lysed by using ACK lysis buffer (Invitrogen, Carlsbad, CA). Splenocytes were subsequently washed two times with RPMI medium 1640 (Invitrogen, Carlsbad, CA) supplemented with 10% FBS, 2 mM L-glutamine, 1% PSF (penicillin, streptomycin, fungizone), counted, and stimulated for 6 hours with PMA (5 nglml), ionomycin (25 nglml) or H2b restricted LacZ peptide (DAPIYTNV, 3 pg/ml) constructed by Genscript (Piscataway, NJ) at 37° C under 5% 002 (or left unstimulated). After stimulation splenocytes (1x106) were transferred to 5 ml tubes and washed with 3 ml cold FACS-buffer (1600 rpm, 5 min, 4° C). The cell pellet was resuspended in 90 pl of cold complete RPMI and 15 pl lFNy catch reagent. After 5 min of incubation (labeling) at 4° C, 2 ml of warm (37° C) medium was added to each tube. The cells were then incubated at 37° C and briefly shaken every 10 min to allow cytokine secretion for 55 min. 189 Following incubation, cells were placed on ice and washed with 4 ml cold FACS- buffer (1600 rpm, 5 min, 4° C), resuspended in 90 pl ‘cold FACS-buffer. The secreted lFNy was stained with 15 pl APO-conjugated lFNy-specific antibody (lFNy detection reagent). After 10 min incubation at 4° C, the cells were washed with cold FACS- buffer, pelleted (1600 rpm, 5 min, 4° C), and resuspended in FACS-buffer. For surface molecules staining, the antibodies were prepared in the supernatant of the 2.462 hybridoma cell line, cells were stained with PE-CD8 (6 jig/ml), PeGCCy5.5-CD19 (8 pglml) (all BD Biosciences, San Diego, CA), and . incubated on ice for 30 minutes. Samples were analyzed on BD LSR Il instrument and analyzed using FlowJo software. 6.17. B cell activation assay (Flow Cytometry) Early activation of B cells was studied by Flow Cytometry based methods. 0.75x1011 vplmouse of Ad5-lacZ was injected intravenously into WT, CR1I2-KO or C3-KO mice. Forty-eight hours after Ad injection splenocytes were harvested and CD19+ cells examined for expression of CD69 activation marker. Briefly, after harvesting splenocytes at 48 hpi from individual mice, RBCs were lysed by using ACK lysis buffer (Invitrogen, Carlsbad, CA). Splenocytes were subsequently washed two times with RPMI medium 1640 (Invitrogen, Carlsbad, CA) supplemented with 10% FBS, 2 mM L-glutamine, 1% PSF (penicillin, streptomycin, fungizone), resuspended and counted. Two million cells were first washed two times with cold FACS buffer and then incubated for 15 minutes with purified rat anti-mouse CD16/CD32 Fc block (BD Biosciences, San Diego, CA). For surface staining, cells were washed one time with FACS buffer and 190 incubated on ice for 30 minutes with the following antibodies, APC-CD3 (8 pglml), PeGCCy5.5-CD19 (8 pg/ml) and FlTC-CDB9 (10 pg/ml), (all BD Biosciences, San Diego, CA). Samples were analyzed on BD LSR ll instrument and analyzed using FlowJo software (Tree Star, San Carlos, CA, USA). 6.18. Antibody Titering Assay ELISA based titering experiments were essentially completed as previously described (209). Briefly, 5 x 108 vplwell or 0.2 pg recombinant LacZ protein/well (each diluted in PBS) was used to coat wells of a 96 well plate overnight at 4°C. Plates were washed with PBS-Tween (0.05%) solution, and blocking buffer (3% BSA in PBS) was added to each well and incubated for 1-3 hours at room temperature. For titering of total lgG antibodies, plasma was diluted in 1:800 in blocking buffer, added to the wells, and incubated at RT for 1 h. Wells were washed using PBS-Tween (0.05%) and HRP conjugated rabbit anti-mouse antibody (BioRad, Hercules, CA) was added at a 1:4000 dilution in PBS-Tween. TMB (Sigma-Aldrich, St. Louis, MO) substrate was added to each well, and the reaction was stopped with 1 N phosphoric acid. Plates were read at 450nm in a microplate spectrophotometer. Subisotype titering was completed using a hybridoma subisotyping kit (Calbiochem, La Jolla, CA) using plasma at a dilution of 1:200 per manufacturer’s recommendations. 6.19. Neutralizing Antibody Assay 2 x 103 HEK293 cells were seeded in microwells in 125pl of complete media (DMEM, 10% FBS, 1% PSF). Cells were cultured overnight in a 37° C, 5% 191 CO; incubator. To inactivate complement, plasma was heat inactivated for 60 min at 56° C and brought to room temperature. Dilutions were made as indicated in complete media in a total volume of 100pl for each well. 1.3 x 106 viral particles of Ad5-LacZ (~650 vplcell) was next added to each dilution, mixed well and incubated at room temperature (RT) for 1 hour. 100 pl of the medium/plasmalvirus mixture was applied to cells and incubated for 2-3 days. Control samples were incubated with either virus alone or complete media alone. CELLTITER 96 A0...mus One solution (Promega, Madison, WI) was added to each well and incubated for 2 hours in a 37° C, 5% 002 incubator. 150 pl of media from each well was removed into a new microtiter plate and read at 492 nm in a microplate spectrophotometer. Blank subtracted OD492 values are reported. 6.20. Western blotting To determine if mCR1/2 protein has any effect on levels of major complement protein C3 (or its cleavage products) in murine plasma, we have performed Western blotting utilizing murine C3 specific polyclonal antibody, which also recognizes C3 cleavage products (Abcam, Cambridge, MA). Assay was performed as previously described (144). Briefly, 5 pl of murine plasma, collected at 10 minutes post injection or 6 hpi from all 4 groups of mice, utilized in this study was loaded on 7.5% SDS-PAGE, transferred onto PVDF membranes and probed with C3 antibody. Subsequently, the blot was probed with secondary fluorescent antibodies, scanned and quantified utilizing Licor’s Odyssey scanner (210). 192 6.21. Peritoneal macrophages isolation and infection Murine peritoneal macrophages were elicited by injection of 1 ml of sterile Brewer's thioglycollate medium (4.05 gl100 ml; Sigma) into the peritoneal cavity of CS7BU6 WT, B—Arr1-KO or [3-Arr2-KO mice. After 4 days, mice were euthanized. PBS (10 ml) was injected into the peritoneum, and lavage fluid was removed as previously described (211, 212). Peritoneal cells were washed twice by centrifugation, resuspended in 10% fetal bovine serum (US origin, endotoxin tested (less than 0.3 EU/ml), heat inactivated, mycoplasma, virus and bacteriophage tested from Gibco/Invitrogen) supplemented RPMI-1640 medium (Gibcollnvitrogen) and then plated in 12-well plates and allowed to adhere to the wells for 8 h in 5% 002 at 37 °C. Nonadherent cells were removed by two washes with fresh culture medium (RPMI supplemented with 0.5% FBS), and the remaining adherent peritoneal macrophages were used for the subsequent infection with 20,000 vplcell of Ad5-LacZ. Perotineal macrophages were extracted from four WT mice, four B-Arr1-KO mice and six 8-Arr2-KO mice and plated onto multiple wells at the concentration of 1.5x106 cells/well. For every time point independent wells were utilized: N=4 for WT and B-Arr1-KO, N=6 for [3- Arr2-KO. Total media was collected at corresponding time point (6, 24 or 48 hpi) and stored at -80° C until use. 6.22. Complement activation AP50 serum-based assay Normal adult human serum (pooled from 30 individuals) was purchased from Complement Technology Inc. (Tyler, TX), Rhesus monkey serum was 193 purchased from Innovative Research (Novi, MI), human PBMCs were obtained from Astarte Biologics (Redmond, WA). Alternative pathway 50 complement activation assay was performed as previously described (24) with modifications. NHS was diluted 1l5 in EGTA-GVBS++ (8 mM EGTA) and mixed with the respective Ads (2x1011 vp). NHS/Ad mixtures were incubated at 37° c for 30 min., and rabbit erythrocytes were added to each tube (0.75x108 in EGTA- GVBS”) After incubation at 37° C for 1 hour, 2 ml of EDTA-GVBS (10 mM EDTA) was added to each tube and tubes were centrifuged at 3500 rpm for 5 min. Following centrifugation absorbance of all tubes was read at 414 nm. All reagents including NHS (pooled from 30 healthy individuals) were purchased from Complement Technology Inc. (Tyler, TX). AP50 assay experiment was repeated 4 times yielding similar results. Data from one representative experiment are reported. In every experiment we have utilized N=3 technical replicates. 6.23. C3a-desArg ELISA and CH50 assay 200 pl of NHS or NHPS was mixed with ma” vp of each or: the respective Ad vectors (final concentration equal to 5x1010 vplml of serum) and incubated at 37° C for 90 min. The reaction was then stopped by adding EDTA to a final concentration of 10 mM. C3a-desArg was then quantified using ELISA as per the manufacturer's instructions (Fitzgerald Industries lntl, former Research Diagnostic lnc., Concord, MA) or (Quidel Corporation, San Diego, CA). Activation 194 of Classical complement pathway was measured by CH50 assay using the MicroVue CH50 Eq E1A Kit (Quidel Corporation, San Diego, CA). For this assay, after exposure of the serum to the control or respective Ad5 vectors aliquots were taken, without exposure to EDTA, mixed with complement activator and an ELISA that specifically detects the complement terminal complexes was performed according to the manufacturer’s instructions (Quidel Corporation, San Diego, CA). Note, that the residual amounts of terminal complement complexes present in the control or Ad vector exposed serum samples were quantified as CH50 unit equivalents per ml. 6.24. Statistical analysis For every experiment, pilot trials were performed with 3 mice per group (or N=3 for in vitro experiments). This allowed us to determine effect size and sample variance so that Power Analysis could be performed to correctly determine the number of subjects per group required to achieve a statistical Power > 0.8 at the 95% confidence level. Statistically significant differences in toxicities associated with innate immune response (i.e. platelet counts, gene induction, etc.) were determined using One Way ANOVA with a Student- Newman-Keuls post-hoc test (p value < 0.05). Furthermore, a Two Way ANOVA with a Bonferroni post-hoc test was used to analyze the levels of cytokines at 1 and 6 hpi (or other specified time points) to determine significant differences (p value < 0.05) between groups. For antibody titering assays, liver H&E stains, 8- Gal activity and Ad genomes in mouse liver, a two-tailed Student t-test was used to compare 2 groups of virus-injected animals (p < 0.05). All graphs are 195 presented as Mean of the average :I: SD. GraphPad Prism software was utilized for statistical analysis. 196 Chapter VII Summary and Future Perspectives 197 The use of Ad5 vectors as a platform for gene transfer applications has been gaining steady momentum. The large number of patients safely treated with the Ad5 platform confirms its high likelihood for acceptance by regulatory bodies, relative to less well tested platforms, an important point when considering the considerable risks and costs involved in developing new therapeutics. Unfortunately, despite this tremendous track record for safe and efficacious delivery of transgenes in numerous applications, conventional, E1 deleted Ad5- based vectors have several confirmed limitations. These include Ad vector triggered innate toxicities and a dramatically reduced efficacy for gene transfer in Ad-immune hosts. These facts indicate a need for development of more efficacious Ad-based vectors. We have summarized how many groups have attempted to address the need to improve the efficacy of Ad-based vectors in general, and/or avoid the problem of pre-existing Ad5 immunity specifically, as summarized in Figure 3. Some groups have proposed the utilization of alternative human Ad serotype based vectors (some derived from non-human primates) as a relatively simple method to avoid pre-existing Ad5 immunity (209, 213-215). However, use of alternative serotype based Ad vectors also poses several new serious problems including: (1) lack of previous usage in humans, (2) significantly altered biodistribution profiles and (3) induction of more deleterious inflammatory immune responses relative to Ad5, in some cases causing excessive morbidity and mortality in animal models (46, 75, 163, 209). All of these caveats may prevent/hinder regulatory approval of alternative serotype based Ad vectors for widespread deployment. 198 As a result of these considerations, it is clear that each of the modified Ad vectors may be better utilized for some clinical applications, but not for others. Despite this context specific utility, one should also be impressed that the Ad platform appears to be highly “plastic” and capable of tolerating a number of elegant molecular manipulations. In many instances, these modifications not only preserve important benefits inherent to the use of the traditional Ad5-based platform, but also provide for improved efficacy, even in the context of pre- existing Ad5 immunity. Future studies will expand upon these findings. Incorporation of some of the modifications summarized herein will likely improve the capabilities of this important platform for expanded use in a number of additional human and agricultural applications, not targeted to date. One can also envision combining some of the approaches described in this dissertation. However, theoretical contemplation of these multiply modified vectors is not a guarantee for reduction to practice, as nuances regarding compatibility of multiple manipulations to allow for viability and large scale production of the resultant vector must be respected. In regards to Ad-triggered innate toxicities, various strategies were designed to minimize these acute immune responses, including non-covalent modification of Ad capsids (PEGylation) (87), usage of immunosuppressive M, such as Dexamethasone (28), Makines ([NFa) blockers (167) or fl (lLR9) inhibitors (38). They have all shown initial promising results, but may have limited applicability due to either non-specific off target effects, a significant alteration of Ad transductional efficiency, lack of scalability, and/or alteration of 199 Ad vector biodistribution profiles. As a result, continued investigations of Ad- induced innate immune responses are required in order to more fully understand the mechanisms by which immune responses against Ad vectors are generated within the host. In attempts to investigate the role of PRRs in mediating Ad-triggered responses, we have studied the roles of B-Arr-1 and B-Arr—2 in modulating these inflammatory responses. We were the first to identify p-Arr-2 as potent inhibitor of Ad-induced immune responses in mice. Although the biochemical mechanisms by which B-arrestins mediate or inhibit the innate immune responses of Ad5 are not known, previously reported interactions of B-arrestins with TRAF6, lea and p105 might play a role in this process (41, 42, 216). It is also possible that scaffolding functions of p-arrestins unique to Ad5 signaling pathways might be important. It has recently been shown that Ad5 vectors trigger TLR-independent pathways, such as NALP3/ASC inflammasomes (cytoplasmic NOD-like receptors) (58), resulting in activation of lL-1B and lL-18, lL-1odlL-1Rl mediated, nucleic acid independent signaling, and Ad interactions with mucosal defensins have also been identified as mediating Ad5-induced innate immune responses (217, 218). The potential roles of B-arrestins in mediating these responses remain to be determined and represent a target for future studies. In this dissertation, we have described the pivotal role the complement system plays in generating robust innate and adaptive immune responses subsequent to Ad treatment, (including systemic pro-inflammatory 200 cytokines/chemokine release, EC activation, acute thrombocytopenia, and liver transcriptome dysregulation (24, 26)), while identifying that Ad-triggered toxicities are modulated by C3 and CR1/2. Importantly, in our studies we have demonstrated the critical role of these complement components (C3 and CR1/2) in mediating generation of Ad-capsid specific humoral responses, including Ad capsid-specific neutralizing antibodies. Future studies will shed light if the use of C3-blockers or CR1/2 receptor antagonists may be a viable approach to improve the efficacy of Ad-mediated gene transfer, possibly by reducing the generation of Ad capsid specific neutralizing antibodies. The role of cytokines/chemokines in mediating Ad-triggered cellular immune responses has not been fully investigated. Our studies provide a body of evidence confirming that the systemic release of pro-inflammatory cytokines/chemokines is Cit-dependent, and regulated by CR1/2 protein (Figure 19). Specifically, since both MCP-1 and G-CSF are known to activate macrophages (219), and both RANTES and G-CSF activate neutrophils (219), it might be hypothesized that lack of mCR1/2 might result in an enhanced infiltration of these inflammatory cells to the sites normally transduced by Ad vectors (i.e. liver). However, our results were not able to detect substantial increases in acute, Ad induced cellular responses in the livers of Ad-injected mCR1l2-KO mice, suggesting that recruitment of these cells requires additional factors. In one of the most exciting chapters of this dissertation we have described the construction and analysis of a novel class of Ad5-based vectors, “capsid- 201 displaying" specific complement inhibitors. We have began to test these novel COMPinh-displaying Ads in several models and reported improved properties of such-modified vectors. Furthermore, the use of COMPinh displaying Ads could be combined with other methods (i.e.: prophylactic glucocorticoid therapy, or surgical bypass techniques) (28, 170) to further reduce aspects of the Ad vector triggered innate toxicities and improve the outcomes of gene transfer applications. Based upon these data, future, more advanced studies are now justified. We leave our readers with the view that despite all pragmatic limitations, fully described in this dissertation, the continued improvement of Ad-based vectors is likely to yield several important vector platforms that will have high utility in a number of clinical and agricultural applications. 202 References 203 10. 11. 12. Bailey, 11., and V. Mautner. 1994. Phylogenetic relationships among adenovirus serotypes. Virology 205:438-452. Russell, W.C. 2000. Update on adenovirus and its vectors. 1 Gen Virol 81:2573-2604. Volpers, C., and S. Kochanek. 2004. Adenoviral vectors for gene transfer and therapy. ] Gene Med 6 Suppl 1:8164-171. Parks, RJ. 2005. Adenovirus protein 1X: a new look at an old protein. Moi Ther 11:19-25. Rux, 1.1., and RM. Burnett. 2004. Adenovirus structure. Hum Gene Ther 15:1167-1176. Vellinga, 1., S. Van der Heijdt, and RC. Hoeben. 2005. The adenovirus capsid: major progress in minor proteins. ] Gen Virol 86:1581-1588. Cali, 1., A. Kass-Eisler, L. Leinwand, and E. Falck-Pedersen. 1996. Adenovirus type 5 and 7 capsid chimera: fiber replacement alters receptor tropism without affecting primary immune neutralization epitopes. 1 Viral 70:2116-2123. Gall, 1.6., KG. Crystal, and E. Falck-Pedersen. 1998. Construction and characterization of hexon-chimeric adenoviruses: specification of adenovirus serotype. 1 Viral 7 2:10260-10264. Molinier-Frenkel, V., R. Lengagne, F. Gaden, 5.8. Hong, I. Choppin, H. Gahery-Segard, P. Boulanger, and LG. Guillet. 2002. Adenovirus hexon protein is a potent adjuvant for activation of a cellular immune response.) Viral 76:127-135. Roy, 5., PS. Shirley, A. McClelland, and M. Kaleko. 1998. Circumvention of immunity to the adenovirus major coat protein hexon. 1 Viral 72:6875-6879. Wu, 11., 1. Dmitriev, E. Kashentseva, 'l'. Seki, M. Wang. and D.’l‘. Curiel. 2002. Construction and characterization of adenovirus serotype 5 packaged by serotype 3 hexon. ] Viral 76:12775-12782. Myhre, 8., P. Henning, 0. Granio, A.S. Tylo, P.A. Nygren, S. Olofsson, P. Boulanger, L. Lindholm, and 8.8. Hong. 2007. Decreased immune reactivity towards a knobless, affibody-targeted adenovirus type 5 vector. Gene Ther 14:376-381. 204 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Appledom, D.M., S. Seregin, and A. Amalfitano. 2008. Adenovirus vectors for renal-targeted gene delivery. Contrib Nephrol 159:47-62. Amalfitano, A., and RJ. Parks. 2002. Separating fact from fiction: assessing the potential of modified adenovirus vectors for use in human gene therapy. Current gene therapy 2:111-133. Amalfitano, A., M.A. Hauser, H. Hu, D. Serra, C.R. Begy, and LS. Chamberlain. 1998. Production and characterization of improved adenovirus vectors with the E1, E2b, and E3 genes deleted. 1 Viral 72:926-933. Palmer, D.]., and P. Ng. 2005. Helper-dependent adenoviral vectors for gene therapy. Hum Gene Ther 16:1-16. Hacein-Bey-Abina, S., C. Von Kalle, M. Schmidt, M.P. McCormack, N. Wulffraat, P. Leboulch, A. Lim, C.S. Osborne, R. Pawliuk, E. Morillon, R. Sorensen, A. Forster, P. Fraser, [.1. Cohen, G. de Saint Basile, l. Alexander, U. Wintergerst, T. Frebourg, A. Aurias, D. Stoppa-Lyonnet, S. Romana, l. Radford-Weiss, F. Gross, F. Valensi, E. Delabesse, E. Macintyre, F. Sigaux, I. Soulier, L.E. Leiva, M. Wissler, C. Prinz, T.H. Rabbitts, F. Le Deist, A. Fischer, and M. Cavazzana-Calvo. 2003. LMOZ-associated clonal T cell proliferation in two patients after gene therapy for SClD-XI. Science 302:415-419. Seggewiss, R., and C.E. Dunbar. 2005. A new direction for gene therapy: intrathymic T cell-specific lentiviral gene transfer. 1 (.‘lin Invest 115:2064-2067. Puck, ].M., and BL. Malech. 2006. Gene therapy for immune disorders: good news tempered by bad news. The journal of allergy and clinical immunology 117:865-869. Donsante, A., D.G. Miller, Y. Li, C. Vogler, E.M. Brunt, D.W. Russell, and MS. Sands. 2007. AAV vector integration sites in mouse hepatocellular carcinoma. Science 317:477. Dave, U.P., K. Akagi, R. Tripathi, S.M. Cleveland, M.A. Thompson, M. Yi, R. Stephens, ].R. Downing, NA. lenkins, and N.G. Copeland. 2009. Murine leukemias with retroviral insertions at Lmoz are predictive of the leukemias induced in SClD-Xl patients following retroviral gene therapy. PLoS Genet 5:e1000491. Harui, A., S. Suzuki, 8. Kochanek, and K. Mitani. 1999. Frequency. and stability of chromosomal integration of adenovirus vectors. ] Viral 73:6141-6146. 205 23. 24. 25. 26. 27. 28. 29. 30. 31. Hartman, Z.C., A. Kiang, R.S. Everett, D. Serra, XX. Yang, T.M. Clay, and A. Amalfitano. 2007. Adenovirus infection triggers a rapid, MyD88- regulated transcriptome response critical to acute-phase and adaptive immune responses in vivo. ] Vir0181:1796-1812. Appledom, D.M., A. McBride, S. Seregin, ].M. Scott, N. Schuldt, A. Kiang, S. Godbehere, and A. Amalfitano. 2008. Complex interactions with several arms of the complement system dictate innate and humoral immunity to adenoviral vectors. Gene Ther 15:1606-1617. Hartman, Z.C., D.M. Appledom, and A. Amalfitano. 2008. Adenovirus vector induced innate immune responses: impact upon efficacy and toxicity in gene therapy and vaccine applications. Virus research 132:1- 14. Kiang, A., Z.C. Hartman, R.S. Everett, D. Serra, H. Jiang, M.M. Frank, and A. Amalfitano. 2006. Multiple innate inflammatory responses induced after systemic adenovirus vector delivery depend on a functional complement system. Mol Ther 14:588-598. Seregin, S.S., Y.A. Aldhamen, D.M. Appledorn, NJ. Schuldt, AJ. McBride, M. Bujold, S.S. Godbehere, and A. Amalfitano. 2009. CR1/2 is an important suppressor of Adenovirus-induced innate immune responses and is required for induction of neutralizing antibodies. Gene Ther 16:1245-1259. Seregin, S.S., D.M. Appledom, A.l. McBride, NJ. Schuldt, Y.A. Aldhamen, T. Voss, I. Wei, M. Bujold, W. Nance, S. Godbehere, and A. Amalfitano. 2009. Transient pretreatment with glucocorticoid ablates innate toxicity of systemically delivered adenoviral vectors without reducing efficacy. Mal Ther 17:685-696. Raper, S.E., N. Chirmule, F.S. Lee, N.A. Wivel, A. Bagg. G.P. Gao, I.M. Wilson, and M.L. Batshaw. 2003. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 80:148-158. Makower, D., A. Rozenblit, H. Kaufman, M. Edelman, M.E. Lane, I. Zwiebel, H. Haynes, and S. Wadler. 2003. Phase ll clinical trial of intralesional administration of the oncolyfic adenovirus ONYX-015 in patients with hepatobiliary tumors with correlative p53 studies. Clin Cancer Res 9:693-702. Grines, C.L., M.W. Watkins, 1.]. Mahmarian, A.E. lskandrian, 1.]. Rade, P. Marrott, C. Pratt, and N. Kleiman. 2003. A randomized, double-blind, placebo-controlled trial of Ad5FGF-4 gene therapy and its effect on 206 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. myocardial perfusion in patients with stable angina. journal of the American College of Cardiology 42:1339-1347. lwasaki, A., and R. Medzhitov. 2004. Toll-like receptor control of the adaptive immune responses. Nature immunology 5:987-995. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat Rev Immunol 4:499-511. Lemmers, B., L. Salmena, N. Bidere, H. Su, E. Matysiak-Zablocki, K. Murakami, P.S. Ohashi, A. lurisicova, M. Lenardo, R. Hakem, and A. Hakem. 2007. Essential role for caspase-8 in Toll-like receptors and NFkappaB signaling] Biol Chem 282:7416-742 3. Hartman, Z.C., E.P. Black, and A. Amalfitano. 2007. Adenoviral infection induces a multi-faceted innate cellular immune response that is mediated by the toll-like receptor pathway in A549 cells. Virology 358:357-372. Appledom, D.M., S. Patial, A. McBride, S. Godbehere, N. Van Rooiien, N. Parameswaran, and A. Amalfitano. 2008. Adenovirus vector-induced innate inflammatory mediators, MAPK signaling, as well as adaptive immune responses are dependent upon both TLR2 and TLR9 in vivo. J Immunol 181:2134-2144. Nociari, M., 0. Ocheretina, ].W. Schoggins, and E. Falck-Pedersen. 2007. Sensing infection by adenovirus: Toll-like receptor-independent viral DNA recognition signals activation of the interferon regulatory factor 3 master regulator. 1 Viral 81:4145-4157. Cerullo, V., M.P. Seiler, V. Mane, N. Brunetti-Pierri, C. Clarke, T.K. Bertin, ].R. Rodgers, and B. Lee. 2007. Toll-like receptor 9 triggers an innate immune response to helper-dependent adenoviral vectors. Mal Ther 15:378-385. Shayakhmetov, D.M., A. Gaggar, 8. Ni, Z.Y. Li, and A. Lieber. 2005. Adenovirus binding to blood factors results in liver cell infection and hepatotoxicity. 1 Viral 79:7478-7491. DeWire, S.M., S. Ahn, RJ. Leflmwitz, and S.K. Shenoy. 2007. Beta- arrestins and cell signaling. Annual review of physiology 69:483-510. Wang, Y., Y. Tang, L. Teng, Y. Wu, K. Zhao, and G. Pei. 2006. Association of beta-arrestin and TRAF6 negatively regulates Toll-like receptor- interleukin 1 receptor signaling. Nature immunology 7:139-147. 207 42. 43. 45. 46. 47. 48. 49. 50. 51. 52. Parameswaran, N., C.S. Pao, K.S. Leonhard, D.S. Kang, M. Kratz, S.C. Ley, and 1.L. Benovic. 2006. Arrestin-Z and G protein-coupled receptor kinase 5 interact with NFkappaBl p105 and negatively regulate lipopolysaccharide-stimulated ERK1/2 activation in macrophages. 1 Biol Chem 281:34159-34170. Fan, H., L.M. Luttrell, G.E. Tempel, ].1. Senn, P.V. Halushka, and ].A. Cook. 2007. Beta-arrestins 1 and 2 differentially regulate LPS-induced signaling and pro-inflammatory gene expression. Molecular immunology 44:3092-3099. Wolins, N., ]. Lozier, 'I‘.L. Eggerman, E. ]ones, E. Aguilar-Cordova, and 1.6. Vostal. 2003. Intravenous administration of replication-incompetent adenovirus to rhesus monkeys induces thrombocytopenia by increasing in vivo platelet clearance. British journal of haematology 123:903-905. Liu, Q., A.K. Zaiss, P. Colarusso, K. Patel, G. Halian, T.]. Wickham, and D.A. Muruve. 2003. The role of capsid-endothelial interactions in the innate immune response to adenovirus vectors. Hum Gene Ther 14:62 7-643. Appledom, D.M., A. Kiang, A. McBride, H. ]iang, S. Seregin, ].M. Scott, R. Stringer, Y. Kousa, M. Hoban, M.M. Frank, and A. Amalfitano. 2008. Wild- type adenoviruses from groups A-F evoke unique innate immune responses, of which HAd3 and SAd23 are partially complement dependent. Gene Ther 15:885-901. Cichon, G., S. Boeckh-Herwig, H.H. Schmidt, E. Wehnes, T. Muller, P. Pring-Akerblom, and R. Burger. 2001. Complement activation by recombinant adenoviruses. Gene Ther 8:1794-1800. Kirschfink, M. 1997. Controlling the complement system in inflammation. Immunopharmacology 38:51-62. Makrides, S.C. 1998. Therapeutic inhibition of the complement system. Pharmacological reviews 50:59-87. Barrington, R., M. Zhang, M. Fischer, and MC. Carroll. 2001. The role of complement in inflammation and adaptive immunity. Immunol Rev 180:5-15. Wagner, E., and MM. Frank. Therapeutic potential of complement modulation. Nature reviews 9:43-56. Mollnes, T.E., W.C. Song, and ].D. Lambris. 2002. Complement in inflammatory tissue damage and disease. Trends in immunology 23:61- 64. 208 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. Atkinson, 1.P. 2006. C5a and chamma receptors: a mutual admiration society. 1 Clin Invest 116:512-52 0. Atkinson, 1.P., and MM. Frank. 2006. Bypassing complement: evolutionary lessons and future implications. 1 Clin Invest 116:1215- 1218. 1iang, H., Z. Wang, D. Serra, M.M. Frank, and A. Amalfitano. 2004. Recombinant adenovirus vectors activate the alternative complement pathway, leading to the binding of human complement protein C3 independent of anti-ad antibodies. Mal Ther 10:1140-1142. Zaiss, A.K., M.]. Cotter, L.R. White, S.A. Clark, N.C. Wong, V.M. Holers, ].S. Bartlett, and D.A. Muruve. 2008. Complement is an essential component of the immune response to adeno-associated virus vectors. ] Virol 82:2727-2740. Carlisle, R.C., Y. Di, A.M. Cerny, A.F. Sonnen, R.B. Sim, N.K. Green, V. Subr, K. Ulbrich, R.]. Gilbert, K.D. Fisher, R.W. Finberg, and L.W. Seymour. 2009. Human erythrocytes bind and inactivate type 5 adenovirus by presenting Coxsackie virus-adenovirus receptor and complement receptor 1. Blood 113:1909-1918. Muruve, D.A., V. Petrilli, A.K. Zaiss, L.R. White, S.A. Clark, P.1. Ross, R.]. Parks, and 1. Tschopp. 2008. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature 452:103-107. Xu, 2., ]. Tian, ].S. Smith, and A.P. Byrnes. 2008. Clearance of adenovirus by Kupffer cells is mediated by scavenger receptors, natural antibodies, and complement] Viral 82:11705-11713. Gasque, P. 2004. Complement: a unique innate immune sensor for danger signals. Molecular immunology 41:1089-1098. Kawano, M. 2000. Complement regulatory proteins and autoimmunity. Archivum immunologiae et therapiae experimentalis 48:367-372. Morgan, B.P., K.]. Marchbank, M.P. Longhi, C.L. Harris, and AM. Gallimore. 2005. Complement: central to innate immunity and bridging to adaptive responses. Immunol Lett 97:171-179. Kemper, C., and 1.P. Atkinson. 2007. T-cell regulation: with complements from innate immunity. Nat Rev Immunol 7:9-18. Prechl, 1., and A. Erdei. 2000. lmmunomodulatory functions of murine CR1/2. Immunopharmacology 49:117-124. 209 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. Tedder, T.F., M. lnaoki, and S. Sato. 1997. The CD19-CD21 complex regulates signal transduction thresholds governing humoral immunity and autoimmunity. Immunity 6:107-118. Tedder, T.F., K.M. Haas, and ].C. Poe. 2002. CD19-CD21 complex regulates an intrinsic Src family kinase amplification loop that links innate immunity with B-lymphocyte intracellular calcium responses. Biochemical Society transactions 30:807-811. Carroll, M.C. 2004. The complement system in B cell regulation. Molecular immunology 41:141-146. Boackle, S.A., V.M. Holers, and DR. Karp. 1997. CD21 augments antigen presentation in immune individuals. Eur] Immunol 27:122-129. Holers, V.M., and L. Kulik. 2007. Complement receptor 2, natural antibodies and innate immunity: Inter-relationships in B cell selection and activation. Molecular immunology 44:64-72. Fairweather, D., S. Frisancho-Kiss, D.B. Nioku, ].F. Nyland, Z. Kaya, S.A. Yusung, S.E. Davis, ].A. Frisancho, M.A. Barrett, and N.R. Rose. 2006. Complement receptor 1 and 2 deficiency increases coxsackievirus B3- induced myocarditis, dilated cardiomyopathy, and heart failure by increasing macrophages, lL-1beta, and immune complex deposition in the heart.) Immunol 176:3516-3524. Haas, K.M., M. Hasegawa, D.A. Steeber, ].C. Poe, M.D. Zabel, C.B. Bock, D.R. Karp. D.E. Briles, ].H. Weis, and T.F. Tedder. 2002. Complement receptors CD21/35 link innate and protective immunity during Streptococcus pneumoniae infection by regulating lgG3 antibody responses. Immunity 17:713-723. Ding, E.Y., B.L. Hodges, H. Hu, A.1. McVie-Wylie, D. Serra, F.K. Migone, D. Pressley, Y.T. Chen, and A. Amalfitano. 2001. Long-term efficacy after [E1-, polymerase-l adenovirus-mediated transfer of human acid-alpha- glucosidase gene into glycogen storage disease type ll knockout mice. Hum Gene Ther 12:955-965. Svensson, E.C., H.B. Black, D.L. Dugger, S.K. Tripathy, E. Goldwasser, z. Hao, L. Chu, and ].M. Leiden. 1997. Long-term erythropoietin expression in rodents and non-human primates following intramuscular injection of a replication-defective adenoviral vector. Hum Gene Ther 8:1797- 1806. Lasaro, M.C., and H.C. Ertl. 2009. New Insights on Adenovirus as Vaccine Vectors. Mol Ther 210 75. 76. 77. 78. 79. 81. 82. 83. Abbink, P., A.A. Lemckert, B.A. Ewald, D.M. Lynch, M. Denholtz, S. Smits, L. Holterman, l. Damen, R. Vogels, A.R. Thomer, K.L. O'Brien, A. Carville, K.G. Mansfield, 1. Goudsmit, M.1. Havenga, and D.H. Barouch. 2007. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. 1 Viral 81:4654-4663. Cohen, P. 2006. lmmunity's yin and yang. A successful vaccine must first avoid being eliminated by pre-existing immunity before it can promote a protective immune response. IAVIRep 10:1-5. Molinier-Frenkel, V., H. Gahery-Segard, M. Mehtali, C. Le Boulaire, S. Ribault, P. Boulanger, T. Tursz, 1.G. Guillet, and F. Farace. 2000. Immune response to recombinant adenovirus in humans: capsid components from viral input are targets for vector-specific cytotoxic T lymphocytes. ] Viral 74:7678-7682. Sumida, S.M., D.M. Truitt, M.G. Kishko, ].C. Arthur, S.S. 1ackson, D.A. Gorgone, M.A. Lifton, W. Koudstaal, M.G. Pau, S. Kostense, M.1. Havenga, 1. Goudsmit, N.L. Letvin, and D.H. Barouch. 2004. Neutralizing antibodies and CD8+ T lymphocytes both contribute to immunity to adenovirus serotype 5 vaccine vectors. 1 Viral 78:2666-2673. Flomenberg, P., V. Piaskowski, R.I.. Truitt, and 1.T. Casper. 1995. Characterization of human proliferative T cell responses to adenovirus. [Infect Dis 171:1090-1096. Tang. 1., M. Olive, K. Champagne, N. Flomenberg, L. Eisenlohr, S. Hsu, and P. Flomenberg. 2004. Adenovirus hexon T-cell epitope is recognized by most adults and is restricted by HLA DP4, the most common class II allele. Gene Ther 11:1408-1415. Tang, 1., M. Olive, R. Pulmanausahakul, M. Schnell, N. Flomenberg, L. Eisenlohr, and P. Flomenberg. .2006. Human CD8+ cytotoxic T cell responses to adenovirus capsid proteins. Virology 350:312-322. Hashimoto, M., 1.L. Boyer, N.R. Hackett, ].M. Wilson, and R.G. Crystal. 2005. Induction of protective immunity to anthrax lethal toxin with a nonhuman primate adenovirus-based vaccine in the presence of preexisting anti-human adenovirus immunity. Infect Immun 73:6885- 6891. Hackett, N.R., S.M. Kaminsky, D. Sondhi, and R.G. Crystal. 2000. Antivector and antitransgene host responses in gene therapy. Curr Opin Mol Ther 2:376-382. 211 84. 85. 86. 87. 89. 90. 91. 92. 93. O'Riordan, C.R., A. Lachapelle, C. Delgado, V. Parkes, S.C. Wadsworth, A.E. Smith, and G.E. Francis. 1999. PEGylation of adenovirus with retention of infectivity and protection from neutralizing antibody in vitro and in vivo. Hum Gene Ther 10:1349-1358. Matthews, C., G. 1enkins, 1. Hilfinger, and B. Davidson. 1999. Poly-L- lysine improves gene transfer with adenovirus formulated in PLGA microspheres. Gene Ther 6:1558-1564. Lee, S.G., 5.]. Yoon, C.D. Kim, K. Kim, D.S. Lim, Y.I. Yeom, M.W. Sung, D.S. Heo, and N.K. Kim. 2000. Enhancement of adenoviral transduction with polycationic liposomes in vivo. Cancer Gene Ther 7:1329-1335. Croyle, M.A., H.T. Le, K.D. Linse, V. Cerullo, G. Toietta, A. Beaudet, and L. Pastore. 2005. PEGylated helper-dependent adenoviral vectors: highly efficient vectors with an enhanced safety profile. Gene Ther 12:579-587. Croyle, M.A., N. Chirmule, Y. Zhang, and ].M. Wilson. 2002. PEGylation of E1-deleted adenovirus vectors allows significant gene expression on readministration to liver. Hum Gene Ther 13:1887-1900. Wortrnann, A., S. Vohringer, T. Engler, S. Corjon, R. Schirmbeck, 1. Reimann, S. Kochanek, and F. Kreppel. 2008. Fully detargeted polyethylene glycol-coated adenovirus vectors are potent genetic vaccines and escape from pre-existing anti-adenovirus antibodies. Mal Ther 16:154-162. Weaver, E.A., and M.A. Barry. 2008. Effects of Shielding Adenoviral Vectors with Polyethylene Glycol (PEG) on Vector-specific and Vaccine- mediated Immune Responses. Hum Gene Ther Ogawara, K., M.G. Rots, R.1. Kok, H.E. Moorlag, A.M. Van Loenen, D.K. Meiier, H.]. Haisma, and G. Molema. 2004. A novel strategy to modify adenovirus tropism and enhance transgene delivery to activated vascular endothelial cells in vitro and in vivo. Hum Gene Ther 15:433- 443. Einfeld, D.A., D.E. Brough, P.W. Roelvink, l. Kovesdi, and T.1. Wickham. 1999. Construction of a pseudoreceptor that mediates transduction by adenoviruses expressing a ligand in fiber or penton base. 1 Viral 73:9130-9136. Wickham, T.1., D.M. Sega], P.W. Roelvink, M.E. Carrion, A. Lizonova, G.M. Lee, and I. Kovesdi. 1996. Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. 1 Viral 70:6831-6838. 212 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. McConnell, M.1., X. Danthinne, and M.1. lmperiale. 2006. Characterization of a permissive epitope insertion site in adenovirus hexonJ Viral 80:5361-5370. McConnell, M.1., P.C. Hanna, and M.1. lmperiale. 2006. Cytokine response and survival of mice immunized with an adenovirus expressing Bacillus anthracis protective antigen domain 4. Infect Immun 74:1009-1015. Belousova, N., V. Krendelchtchikova, D.T. Curiel. and V. Krasnykh. 2002. Modulation of adenovirus vector tropism via incorporation of polypeptide ligands into the fiber protein. ] Viral 76:8621-8631. Matthews, Q.L., D.A. Sibley, H. Wu, 1. Li, M.A. Stoff-Khalili, R. Waehler, ].M. Mathis, and D.T. Curiel. 2006. Genetic incorporation of a herpes simplex virus type 1 thymidine kinase and firefly luciferase fusion into the adenovirus protein [X for functional display on the virion. Mal Imaging 5:510-519. Worgall, S., A. Busch, M. Rivara, D. Bonnyay, P.L. Leopold, R. Merritt, N.R. Hackett, P.W. Rovelink, 1.T. Bruder, T.1. Wickham, I. Kovesdi, and R.G. Crystal. 2004. Modification to the capsid of the adenovirus vector that enhances dendritic cell infection and transgene-specific cellular immune responses. 1 Viral 7 8:2572-2 580. Kurachi, S., N. Koizumi, F. Sakurai, K. Kawabata, H. Sakurai, S. Nakagawa, T. Hayakawa, and H. Mizuguchi. 2007. Characterization of capsid-modified adenovirus vectors containing heterologous peptides in the fiber knob, protein IX, or hexon. Gene Ther 14:266-274. Waddington, S.N., ].H. McVey, D. Bhella, A.L. Parker, K. Barker, H. Atoda, R. Pink, S.M. Buckley, ].A. Greig, L. Denby, 1. Custers, T. Morita, l.M. Francischetti, R.Q. Monteiro, D.H. Barouch, N. van Rooiien, C. Napoli, M.1. Havenga, S.A. Nicklin, and A.H. Baker. 2008. Adenovirus serotype 5 hexon mediates liver gene transfer. Cell 132:397-409. Dmitriev, l.P., E.A. Kashentseva, and D.T. Curiel. 2002. Engineering of adenovirus vectors containing heterologous peptide sequences in the C terminus of capsid protein IX. 1 Viral 76:6893-6899. Wickham, T.1., P.W. Roelvink, D.E. Brough, and I. Kovesdi. 1996. Adenovirus targeted to heparan-containing receptors increases its gene delivery efficiency to multiple cell types. Nat Biatechnal 14:1570-1573. Koizumi, N., H. Mizuguchi, N. Utoguchi, Y. Watanabe, and T. Hayakawa. 2003. Generation of fiber-modified adenovirus vectors containing heterologous peptides in both the HI loop and C terminus of the fiber knob. ] Gene Med 5:267-276. 213 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. Dmitriev, I., V. Krasnykh, C.R. Miller, M. Wang, E. Kashentseva, G. Mikheeva, N. Belousova, and D.T. Curiel. 1998. An adenovirus vector with genetically modified fibers demonstrates expanded tropism via utilization of a coxsackievirus and adenovirus receptor-independent cell entry mechanism. I Viral 72:9706-9713. Wu, H., T. Han, N. Belousova, V. Krasnykh, E. Kashentseva, l. Dmitriev, M. Kataram, P.1. Mahasreshti, and D.T. Curiel. 2005. Identification of sites in adenovirus hexon for foreign peptide incorporation. ] Viral 79:3382- 3390. Vellinga, 1., M.1. Rabelink, S.1. Cramer, D.]. van den Wollenberg. H. Van der Meulen, K.N. Leppard, F.1. Fallaux, and RC. Hoeben. 2004. Spacers increase the accessibility of peptide ligands linked to the carboxyl terminus of adenovirus minor capsid protein lX. ] Viral 78:3470-3479. Heise, C., T. Hermiston, L. ]ohnson, G. Brooks, A. Sampson-1ohannes, A. Williams, L. Hawkins, and D. Kim. 2000. An adenovirus ElA mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nat Med 6:1134—1139. Coughlan, L., S. Vallath, A. Saha, M. Flak, LA. McNeish, G. Vassaux, ].F. Marshall, LR. Hart, and G1. Thomas. 2009. In vivo retargeting of adenovirus type 5 to alphavbeta6 integrin results in reduced hepatotoxicity and improved tumor uptake following systemic delivery. 1 Viro183:6416-6428. Rein, D.T., M. Breidenbach, and D.T. Curiel. 2006. Current developments in adenovirus-based cancer gene therapy. Future oncology (London, England) 2:137-143. Kroot, E.1., A.M. Huisman, 1. Van Zeben, ].M. Wouters, and H.C. Van Paassen. 2006. Oral pulsed dexamethasone therapy in early rheumatoid arthritis: a pilot study. Ann N YAcad Sci 1069:300-3 06. Kardash, K.1., F. Sarrazin, M.1. Tessler, and A.M. Velly. 2008. Single-dose dexamethasone reduces dynamic pain after total hip arthroplasty. Anesthesia and analgesia 106:12 53-12 57, table of contents. Ebrahimi, 8., and B. Sarkari. 2007. Comparative efficacy of dexamethasone versus hydrocortisone in severe acute pediatric asthma. Iranian journal of allergy, asthma, and immunology 6:159-160. Pace, T.W., F. Hu, and A.H. Miller. 2007. Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain, behavior, and immunity 21:9-19. 214 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. Chinenov, Y., and I. Rogatsky. 2007. Glucocorticoids and the innate immune system: crosstalk with the toll-like receptor signaling network. Molecular and cellular endocrinology 275:30-42. Liberman, A.C., 1. Druker, M.1. Perone, and E. Arzt. 2007. Glucocorticoids in the regulation of transcription factors that control cytokine synthesis. cytokine & growth factor reviews 18:45-56. Ma, W., K. Gee, W. Lim, K. Chambers, ].B. Angel, M. Kozlowski, and A. Kumar. 2004. Dexamethasone inhibits IL-12p40 production in lipopolysaccharide-sfimulated human monocytic cells by down- regulating the activity of c-1un N-terminal kinase, the activation protein-1, and NF-kappa B transcription factors. I Immunol 172:318- 330. Mogensen, T.H., R.S. Berg, S.R. Paludan, and L. Ostergaard. 2008. Mechanisms of dexamethasone-mediated inhibition of Toll-like receptor signaling induced by Neisseria meningitidis and Streptococcus pneumoniae. Infect Immun 76:189-197. Vital, A.L., M. Goncalo, M.T. Cruz, A. Figueiredo, C.B. Duarte, and M.C. Lopes. 2003. Dexamethasone prevents granulocyte-macrophage colony-stimulating factor-induced nuclear factor-kappaB activation, inducible nitric oxide synthase expression and nitric oxide production in a skin dendritic cell line. Mediators of inflammation 12:71-78. Gessani, S., S. McCandless, and C. Baglioni. 1988. The glucocorticoid dexamethasone inhibits synthesis of interferon by decreasing the level of its mRNA. 1 Biol Chem 263:7454-7457. Smith, S.1., A.M. Piliponsky, F. Rosenhead, U. Elchalal, A. Nagler, and F. Levi-Schaffer. 2002. Dexamethasone inhibits maturation, cytokine production and Fe epsilon RI expression of human cord blood-derived mast cells. Clin Exp Allergy 32:906-913. Braun, S., C. 1enny, C. Thioudellet, F. Perraud, M.C. Claudepierre, F. Langle-Rouault, D. Ali-Hadii, K. Schughart, and A. Pavirani. 1999. In vitro and in vivo effects of glucocorticoids on gene transfer to skeletal muscle. FEBS letters 454:277-282. Liu, Y., H.D. Liggitt, 8. Dow, C. Handumrongkul, T.D. Heath, and R1. Debs. 2002. Strain-based genetic differences regulate the efficiency of systemic gene delivery as well as expression. 1 Biol Chem 277:4966- 4972. Malone, R.W., M.A. Hickman, K. Lehmann-Bruinsma, T.R. Sih, R. Walzem, D.M. Carlson, and IS. Powell. 1994. Dexamethasone enhancement of 215 124. 125. 126. 127. 128. 129. 130. 131. 132. gene expression after direct hepatic DNA injection. 1 Biol Chem 269:29903-29907. Boulis, N.M., D.E. Turner, ].A. Dice, V. Bhatia, and EL. Feldman. 1999. Characterization of adenoviral gene expression in spinal card after remote vector delivery. Neurosurgery 45:131-137; discussion 13 7-138. lshimoto, S., K. Kawamoto, T. Stover, S. Kanzaki, T. Yamasoba, and Y. Raphael. 2003. A glucocorticoid reduces adverse effects of adenovirus vectors in the cochlea. Audiology & neuro-atalagy 8:70-79. Otake, K., D.L. Ennist, K. Harrod, and BC. Trapnell. 1998. Nonspecific inflammation inhibits adenovirus-mediated pulmonary gene transfer and expression independent of specific acquired immune responses. Hum Gene Ther 9:2207-2222. Kumahara, K., H. Nagata, K. Watanabe, N. Shimizu, Y. Arimoto, K. lsoyama, Y. Okamoto, and H. Shirasawa. 2005. Suppression of inflammation by dexamethasone prolongs adenoviral vector-mediated transgene expression in murine nasal mucosa. Acta ata-laryngalagica 125:1301-1306. Price, A.R., M.P. Limberis, ].M. Wilson, and S.L. Diamond. 2007. Pulmonary delivery of adenovirus vector formulated with dexamethasone-spermine facilitates homologous vector re- administration. Gene Ther 14:1594—1604. Schiedner, G., W. Bloch, S. Hertel, M. 1ohnston, A. Molojavyi, V. Dries, G. Varga, N. Van Rooijen, and S. Kochanek. 2003. A hemodynamic response to intravenous adenovirus vector particles is caused by systemic Kupffer cell-mediated activation of endothelial cells. Hum Gene Ther 14:1631-1641. Appledom, D.M., S. Patial, S. Godbehere, N. Parameswaran, and A. Amalfitano. 2009. TRIF, and TRIF-lnteracting TLRs Differentially Modulate Several Adenovirus Vector-Induced Immune Responses. ] Inn Imm 1:376-388. Zhu,1., X. Huang, and Y. Yang. 2007. Type I lFN signaling on both B and CD4 T cells is required for protective antibody response to adenovirus. ] Immunol 178:3505-3 510. Li, Y., D.A. Muruve, R.G. Collins, S.S. Lee, and P. Kubes. 2002. The role of selectins and integrins in adenovirus vector-induced neutrophil recruitment to the liver. Eur] Immunol 32:3443-3452. 216 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. Schiedner, G., S. Hertel, and S. Kochanek. 2000. Efficient transformation of primary human amniocytes by E1 functions of Ad5: generation of new cell lines for adenoviral vector production. Hum Gene I'her 11:2105-2116. Miwa, K., A. Igawa, and H. Inoue. 1997. Soluble E-selectin, ICAM-1 and VCAM-l levels in systemic and coronary circulation in patients with variant angina. Cardiovasc Res 36:37-44. Manickan, E., 1.8. Smith, 1. Tian, T.L. Eggerman, 1.N. Lozier, 1. Muller, and A.P. Byrnes. 2006. Rapid Kupffer cell death after intravenous injection of adenovirus vectors. Mal Ther 13:108-117. Cotter, M.1., A.K. Zaiss, and D.A. Muruve. 2005. Neutrophils interact with adenovirus vectors via Fc receptors and complement receptor 1. 1 Viral 79:14622-14631. Hu, H., D. Serra, and A. Amalfitano. 1999. Persistence of an [E1-, polymerase-1 adenovirus vector despite transduction of a neoantigen into immune-competent mice. Hum Gene Ther 10:355-364. Everett, R.S., B.L. Hodges, E.Y. Ding, F. Xu, D. Serra, and A. Amalfitano. 2003. Liver toxicities typically induced by first-generation adenoviral vectors can be reduced by use of E1, E2b-deleted adenoviral vectors. Hum Gene Ther 14:1715-1726. Zinn, K.R., A.]. Szalai, A. Stargel, V. Krasnykh, and TR Chaudhuri. 2004. Bioluminescence imaging reveals a significant role for complement in liver transduction following intravenous delivery of adenovirus. Gene Ther 11:1482-1486. Hawlisch, H., and 1. Kohl. 2006. Complement and Toll-like receptors: key regulators of adaptive immune responses. Molecular immunology 43:13-21. Carroll, M. 1999. Role of complement receptors CD21/CD35 in B lymphocyte activation and survival. Current topics in microbiology and immunology 246:63-68; discussion 69. Sato, S., A.S. Miller, M.C. Howard, and T.F. Tedder. 1997. Regulation of B lymphocyte development and activation by the CD19/CD21/CD81/Leu 13 complex requires the cytoplasmic domain of CD19. ] Immunol 159:3278-3287. Miwa, T., and W.C. Song. 2001. Membrane complement regulatory proteins: insight from animal studies and relevance to human diseases. lntemational immunapharmacalagy 1:445-459. 217 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. Miwa, T., L. Zhou, R. Tudoran, ].D. Lambris. M.P. Madaio, M. Nangaku, H. Molina, and W.C. Song. 2007. DAF/Crry double deficiency in mice exacerbates nephrotoxic serum-induced proteinuria despite markedly reduced systemic complement activity. Molecular immunology 44:139- 146. Turnberg, D., and M. Botto. 2003. The regulation of the complement system: insights from genetically-engineered mice. Molecular immunology 40:145-153. Tian,]., Z. Xu, 1.S. Smith, S.E. Hoflrerr, M.A. Barry, and A.P. Byrnes. 2009. Adenovirus activates complement by distinctly different mechanisms in vitro and in vivo: indirect complement activation by virions in vivo. ] Viral . Croyle, M.A., N. Chirmule, Y. Zhang, and ].M. Wilson. 2001. ”Stealth" adenoviruses blunt cell-mediated and humoral immune responses against the virus and allow for significant gene expression upon readministration in the lung. 1 Viral 75:4792-4801. Ricklin, D., and ].D. Lambris. 2008. Compstatin: a complement inhibitor on its way to clinical application. Advances in experimental medicine and biology 632:273-292. Green, T.D., B.R. Newton, P.A. Rota, Y. Xu, H.L. Robinson, and TM. Ross. 2001. C3d enhancement of neutralizing antibodies to measles hemagglutinin. Vaccine 20:242-248. Ross, T.M., Y. Xu, R.A. Bright, and H.L. Robinson. 2000. C3d enhancement of antibodies to hemagglutinin accelerates protection against influenza virus challenge. Nature immunology 1:12 7-13 1. Draper, 5.1., A.C. Moore, A.L. Goodman, C.A. Long, A.A. Holder, S.C. Gilbert, F. Hill, and AN. Hill. 2008. Effective induction of high-titer antibodies by viral vector vaccines. Nat Med 14:819-821. Ogun, S.A., L. Dumon-Seignovert1.B. Marchand, A.A. Holder, and F. Hill. 2008. The oligomerization domain of C4-binding protein (C4bp) acts as an adjuvant, and the fusion protein comprised of the 19-kilodalton merozoite surface protein 1 fused with the murine C4bp domain protects mice against malaria. Infect Immun 76:3817-3823. Ricklin, D., and ].D. Lambris. 2007. Complement-targeted therapeutics. Nat Biotechnol 25:1265-12 7 5. Muruve, D.A. 2004. The innate immune response to adenovirus vectors. Hum Gene Ther 15:1157-1166. 218 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. Sahu, A., B.K. Kay, and ].D. Lambris. 1996. Inhibition of human complement by a C3-binding peptide isolated from a phage-displayed random peptide library. ] Im‘munal 157:884-891. Mallik, B., M. Katragadda, L.A. Spruce, C. Carafides, C.G. Tsokos, D. Morikis, and ].D. Lambris. 2005. Design and NMR characterization of active analogues of compstatin containing non-natural amino acids. journal of medicinal chemistry 48:2 74-2 86. Sahu, A., D. Morikis, and ].D. Lambris. 2003. Compstatin, a peptide inhibitor of complement, exhibits species-specific binding to complement component C3. Molecular immunology 39:557-566. Yeatman, M., C.W. Daggett, C.L. Lau, G.W. Byrne, 1.8. Logan, 1.L. Platt, and RD. Davis. 1999. Human complement regulatory proteins protect swine lungs from xenogeneic injury. Ann Thorac Surg 67:769-775. Maelandsmo, G.M., P.1. Ross, M. Pavliv, R.A. Meulenbroek, C. Evelegh, D.A. Muruve, F.L. Graham, and R1. Parks. 2005. Use of a murine secreted alkaline phosphatase as a non-immunogenic reporter gene in mice. 1 Gene Med 7:307-315. Van Linthout, S., M. Lusky, D. Collen, and B. De Geest. 2002. Persistent hepatic expression of human apo A-l after transfer with a helper-virus independent adenoviral vector. Gene Ther 9:152 0-1528. Mathis, ].M., M.A. Stoff-lflralili, and D.T. Curiel. 2005. Oncolytic adenoviruses - selective retargeting to tumor cells. Oncogene 24:7775- 7791. Stone, D., Y. Liu, Z.Y. Li, S. Tuve, R. Strauss, and A. Lieber. 2007. Comparison of adenoviruses from species B, C, e, and f after intravenous delivery. Mal Ther 15:2146-2153. Hartman, Z.C., D.M. Appledom, D. Serra, 0. Glass, T.B. Mendelson, T.M. Clay, and A. Amalfitano. 2008. Replication-attenuated Human Adenoviral Type 4 vectors elicit capsid dependent enhanced innate immune responses that are partially dependent upon interactions with the complement system. Virology 374:453-467. Kuzmin, A.l., M.1. Finegold, and RC. Eisensmith. 1997. Macrophage depletion increases the safety, efficacy and persistence of adenovirus- mediated gene transfer in vivo. Gene Ther 4:309-316. Kay, M.A., L. Meuse, A.M. Gown, P. Linsley, D. Hollenbaugh, A. Aruffo, H.D. Ochs, and CB. Wilson. 1997. Transient immunomodulation with anti- CD40 ligand antibody and CTLA4lg enhances persistence and secondary 219 166. 167. 168. 169. 170. 171. 172. 173. adenovirus-mediated gene transfer into mouse liver. Proc Natl Acad Sci U SA 94:4686-4691. Kuriyama, S., K. Tominaga, A. Mitoro, H. Tsujinoue, T. Nakatani, M. Yamazaki, S. Nagao, Y. Toyokawa, S. Okamoto, and H. Fukui. 2000. lmmunomodulation with FK506 around the time of intravenous re- administration of an adenoviral vector facilitates gene transfer into primed rat liver. lntemational journal of cancer 85:83 9-844. Song, M.Y., S.K. Park, C.S. Kim, T.H. Yoo, B. Kim, M.S. Kim, Y.S. Kim, W.]. Kwag, B.K. Lee, and K. Baek. 2008. Characterization of a novel anti- human TNF-alpha murine monoclonal antibody with high binding affinity and neutralizing activity. Experimental & molecular medicine 40:35-42. Ho, P.P., P. Fontoura, M. Platten, R.A. Sobel, 1.1. DeVoss, L.Y. Lee, B.A. Kidd, B.H. Tomooka, 1. Capers, A. Agrawal, R. Gupta, 1. Zernik, M.K. Yee, B.1. Lee, H. Garren, W.H. Robinson, and L. Steinman. 2005. A suppressive oligodeoxynucleotide enhances the efficacy of myelin cocktail/lL-4- tolerizing DNA vaccination and treats autoimmune disease. 1 Immunol 175:6226-6234. Ren, L., ].B. Blanchette, L.R. White, S.A. Clark, D.]. Heffner, LA. Tibbles, and D.A. Muruve. 2005. Soluble fibronectin induces chemokine gene expression in renal tubular epithelial cells. Kidney international 68:2111-2120. Brunetti-Pierri, N., G.E. Stapleton, D.1. Palmer, Y. Zuo, V.P. Mane, M.1. Finegold, A.L. Beaudet, M.M. Leland, C.E. Mullins, and P. Ng. 2007. Pseudo-hydrodynamic delivery of helper-dependent adenoviral vectors into non-human primates for liver-directed gene therapy. Mal Ther 15:732-740. Shayakhmetov, D.M., Z.Y. Li, S. Ni, and A. Lieber. 2004. Analysis of adenovirus sequestration in the liver, transduction of hepatic cells, and innate toxicity after injection of fiber-modified vectors. 1 Viral 78:5368- 5381. Shayakhmetov, D.M., A.M. Eberly, Z.Y. Li, and A. Lieber. 2005. Deletion of penton RGD motifs affects the efficiency of both the internalization and the endosome escape of viral particles containing adenovirus serotype 5 or 35 fiber knobs. ] Viral 79:1053-1061. Barnes, P.1. 1998. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci (Land) 94:557-572. 220 174. 175. 176. 177. 178. 179. 180. 181. 182. 183. Huang, H.W., W. Bi, G.N. 1enkins, and 1.L. Alcorn. 2008. Glucocorticoid regulation of human pulmonary surfactant protein-B mRNA stability involves the 3'-untranslated region. American journal of respiratory cell and molecular biology 38:47 3-482. Schnell, M.A., Y. Zhang, 1. Tazelaar, G.P. Gao, Q.C. Yu, R. Qian, 8.]. Chen, A.N. Varnavski, C. LeClair, S.E. Raper, and ].M. Wilson. 2001. Activation of innate immunity in nonhuman primates following intraportal administration of adenoviral vectors. Mal Ther 3:708-722. Miller, T.A., and F.W. Schaefer, 3rd. 2007. Changes in mouse circulating leukocyte numbers in C57BL/6 mice immunosuppressed with dexamethasone for Cryptosporidium parvum oocyst production. Veterinary parasitalagy 149:147-157. 1eklova, E., L. Leva, Z. ]aglic, and M. Faldyna. 2008. Dexamethasone- induced immunosuppression: a rabbit model. Veterinary immunology and immunapathalagy 122:231-240. Anderson, B.H., D.L. Watson, and LG. Colditz. 1999. The effect of dexamethasone on some immunological parameters in cattle. Veterinary research communications 23:399-413. Lieber, A., C.Y. He, L. Meuse, D. Schowalter, l. Kirillova, B. Winther, and M.A. Kay. 1997. The role of Kupffer cell activation and viral gene expression in early liver toxicity after infusion of recombinant adenovirus vectors. 1 Viral 71:8798-8807. Stone, D., Y. Liu, D. Shayakhmetov, Z.Y. Li, S. Ni, and A. Lieber. 2007 . Adenovirus-platelet interaction in blood causes virus sequestration to the reticuloendothelial system of the liver. 1 Viral 81:4866-4871. Franchimont, D., 1. Galon, M. Gadina, R. Visconti, Y. Zhou, M. Aringer, D.M. Frucht, G.P. Chrousos, and 1.1. O'Shea. 2000. Inhibition of Th1 immune response by glucocorticoids: dexamethasone selectively inhibits IL-12-induced Stat4 phosphorylation in T lymphocytes. 1 Immunol 164:1768-1774. Ramirez, F., D.]. Fowell, M. Puklavec, S. Simmonds, and D. Mason. 1996. Glucocorticoids promote a TH2 cytokine response by CD4+ T cells in vitro. ] Immunol 156:2406-2412. Olive, M., L. Eisenlohr, N. Flomenberg, S. Hsu, and P. Flomenberg. 2002. The adenovirus capsid protein hexon contains a highly conserved human CD4+ T-cell epitope. Hum Gene Ther 13:1167-1178. 221 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. Sumida, S.M., D.M. Truitt, A.A. Lemckert, R. Vogels, ].H. Custers, M.M. Adda, S. Lockman, T. Peter, F.W. Peyerl, M.G. Kishko, S.S. 1ackson, D.A. Gorgone, M.A. Lifton, M. Essex, B.D. Walker, 1. Goudsmit, M.1. Havenga, and D.H. Barouch. 2005. Neutralizing antibodies to adenovirus serotype 5 vaccine vectors are directed primarily against the adenovirus hexon protein. j Immunol 174:7179-7185. Hodges, B.L., H.K. Evans, R.S. Everett, E.Y. Ding, D. Serra, and A. Amalfitano. 2001. Adenovirus vectors with the 100K gene deleted and their potential for multiple gene therapy applications. 1 Viral 75:5913- 5920. Hodges, B.L., D. Serra, H. Hu, C.A. Begy, 1.5. Chamberlain, and A. Amalfitano. 2000. Multiply deleted [E1, polymerase-, and pTP-] adenovirus vector persists despite deletion of the preterminal protein. j Gene Med 2:2 50-2 59. Dietzman, R.H., R.A. Ersek, ].M. Bloch, and RC. Lillehei. 1969. High- output, low-resistance gram-negative septic shock in man. Angialagy 20:691-700. Lefering, R., and EA. Neugebauer. 1995. Steroid controversy in sepsis and septic shock: a meta-analysis. Critical care medicine 23:12 94-1303. Oaks, W.W., and H.B. Cohen. 1967. Endotoxin shock in the geriatric patient. Geriatrics 22:120-130. Wang, X.Q., X. Zhou, Y. Zhou, L. Rang, L. Gao, and W. Xu. 2008. Low-dose dexamethasone alleviates lipopolysaccharide-induced acute lung injury in rats and upregulates pulmonary glucocorticoid receptors. Respirolagy (Carlton, Vic Zhou, Y., EA. Ling, and S.T. Dheen. 2007. Dexamethasone suppresses monocyte chemoattractant protein-1 production via mitogen activated protein kinase phosphatase-1 dependent inhibition of 1un N-terminal kinase and p38 mitogen-activated protein kinase in activated rat microglia. journal of neurachemistry 102:667-678. Lee, C.W., ].H. Chuang, P.W. Wang, N.K. Chang, H.C. Wang, CC. Huang, M.M. Tiao, and S.K. Lo. 2006. Effect of glucocorticoid pretreatment on oxidative liver injury and survival in jaundiced rats with endotoxin cholangitis. Warldjaumal of surgery 30:2217-2226. Hafner, D., and P.G. Germann. 1999. Dexamethasone enhances the activity of rSP-C surfactant but not of exosurf in a rat model of the acute lung injury. journal of pharmacological and toxicological methods 42:39- 48. 222 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. Schiedner, G., S. Hertel, M. ]ohnston, V. Dries, N. van Rooijen, and S. Kochanek. 2003. Selective depletion or blockade of Kupffer cells leads to enhanced and prolonged hepatic transgene expression using high- capacity adenoviral vectors. Mal Ther 7:35-43. Zsengeller, 2., K. Otake, S.A. Hossain, P.Y. Berclaz, and B.C. Trapnell. 2000. lntemalization of adenovirus by alveolar macrophages initiates early proinflammatory signaling during acute respiratory tract infection. j Viral 74:9655-9667. Tao, N., G.P. Gao, M. Parr, 1. ]ohnston, T. Baradet, ].M. Wilson, 1. Barsoum, and S.E. Fawell. 2001. Sequestration of adenoviral vector by Kupffer cells leads to a nonlinear dose response of transduction in liver. Mal Ther 3:28-35. Everett, R.S., H.K. Evans, B.L. Hodges, E.Y. Ding, D.M. Serra, and A. Amalfitano. 2004. Strain-specific rate of shutdown of CMV enhancer activity in murine liver confirmed by use of persistent [E1(-), E2b(-)] adenoviral vectors. Virology 32 5:96-105. Cross, S.L., . IN.F. Halden, M.1. Lenardo, and w.]. Leonard. 1989. Functionally distinct NF-kappa B binding sites in the immunoglobulin kappa and lL-2 receptor alpha chain genes. Science 244:466-469. Yang, T.C., ].B. Millar, N. Grinshtein, 1. Bassett, 1. Finn, and 1.L. Bramson. 2007. T-cell immunity generated by recombinant adenovirus vaccines. Expert review of vaccines 6:347 -3 56. He, T.C., S. Zhou, L.T. da Costa, 1. Yu, K.W. Kinzler, and B. Vogelstein. 1998. A simplified system for generating recombinant adenoviruses. PracNatlAcad Sci U S A 95:2509-2514. Fontana, L., M. Nuzzo, L. Urbanelli, and P. Monaci. 2003. General strategy for broadening adenovirus tropism. j Viral 77:11094—11104. Ng. P., and F.L. Graham. 2002. Construction of first-generation adenoviral vectors. Methods in molecular medicine 69:389-414. Sargent, K.L., P. Ng. C. Evelegh, F.L. Graham, and R1. Parks. 2004. Development of a size-restricted pix-deleted helper virus for amplification of helper-dependent adenovirus vectors. Gene 'l'her 11:504-511. Colby, W.W., and T. Shenk. 1981. Adenovirus type 5 virions can be assembled in vivo in the absence of detectable polypeptide IX. j Viral 39:977-980. 223 205. 206. 207. 208. 209. 210. 211. 212. 213. 214. Gommerman, 1.L., D.Y. Oh, X. Zhou, T.F. Tedder, M. Maurer, S.1. Galli, and M.C. Carroll. 2000. A role for CDZ 1/CD35 and CD19 in responses to acute septic peritonitis: a potential mechanism for mast cell activation. 1 Immunol 165:6915-6921. Rajagopal, K., E.1. Whalen, ].D. Violin, ].A. Stiber, P.B. Rosenberg, R.T. Premont, T.M. Coffrnan, H.A. Rockman, and RJ. Lefltowitz. 2006. Beta- arrestinZ-mediated inotropic effects of the angiotensin II type 1A receptor in isolated cardiac myocytes. Proc Natl Acad Sci U S A 103:16284-16289. Higginbotham, 1.N., P. Seth, R.M. Blaese, and W1. Ramsey. 2002. The release of inflammatory cytokines from human peripheral blood mononuclear cells in vitro following exposure to adenovirus variants and capsid. Hum Gene Ther 13:129-141. Hofstetter, 1.R., A. Zhang, A.R. Mayeda, T. Guscar,1.l. Nurnberger, ]r., and D.K. Lahiri. 1997. Genomic DNA from mice: a comparison of recovery methods and tissue sources. Biochemical and molecular medicine 62:197-202. Hensley, S.E., A.S. Cun, W. Giles-Davis, Y. Li, Z. Xiang, M.O. Lasaro, B.R. Williams, R.H. Silverman, and H.C. Ertl. 2007. Type I interferon inhibits antibody responses induced by a chimpanzee adenovirus vector. Mal Ther 15:393-403. Loniewski, K.1., S. Patial, and N. Parameswaran. 2007. Sensitivity of TLR4- and -7-induced NF kappa Bl p105-TPL2-ERK pathway to TNF- receptor-associated-factor-6 revealed by RNAi in mouse macrophages. Molecular immunology 44:37 15-3 723. 1ia, Q., H.R. Zhou, Y. Shi, and 1.1. Pestka. 2006. Docosahexaenoic acid consumption inhibits deoxynivalenol-induced CREB/ATFI activation and lL-6 gene transcription in mouse macrophages. The journal of nutrition 136:366-372. Loniewski, K., Y. Shi, 1. Pestka, and N. Parameswaran. 2008. Toll-like receptors differentially regulate GPCR kinases and arrestins in primary macrophages. Molecular immunology 45:23 12-2322. Farina, S.E., G.P. Gao, Z.Q. Xiang, 1.]. Rux, R.M. Burnett, M.R. Alvira, 1. Marsh, H.C. Ertl, and ].M. Wilson. 2001. Replication-defective vector based on a chimpanzee adenovirus. 1 Viral 75:11603-11613. Lemckert, A.A., S.M. Sumida, L. Holterman, R. Vogels, D.M. Truitt, D.M. Lynch, A. Nanda, B.A. Ewald, D.A. Gorgone, M.A. Lifton, 1. Goudsmit, M.1. Havenga, and D.H. Barouch. 2005. Immunogenicity of heterologous 224 215. 216. 217. 218. 219. prime-boost regimens involving recombinant adenovirus serotype 11 (Adll) and Ad35 vaccine vectors in the presence of anti-ads immunity. 1 Viral 79:9694-9701. Liu, 1., K.L. O'Brien, D.M. Lynch, N.L. Simmons, A. La Porte, A.M. Riggs, P. Abbink, R.T. Coffey, L.E. Grandpre, M.S. Seaman, G. Landucci, D.N. Forthal, D.C. Montefiori, A. Carville, K.G. Mansfield, M.1. Havenga, M.G. Pau, 1. Goudsmit, and D.H. Barouch. 2009. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457:87- 91. Gao, H., Y. Sun, Y. Wu, B. Luan, Y. Wang, B. Qu, and G. Pei. 2004. Identification of beta-arrestin2 as a G protein-coupled receptor- stimulated regulator of NF-kappaB pathways. Molecular cell 14:303- 317. Di Paolo, N.C., E.A. Miao, Y. Iwakura, K. MuraIi-Krishna, A. Aderem, R.A. Flavell, T. Papayannopoulou, and D.M. Shayakhmetov. 2009. Virus binding to a plasma membrane receptor triggers interleukin-1 alpha- mediated proinflammatory macrophage response in viva. Immunity 31:110-121. Smith, 1.G., and G.R. Nemerow. 2008. Mechanism of adenovirus neutralization by Human alpha-defensins. Cell Host Microbe 3:11-19. Maser, B., and K. Willimann. 2004. Chemokines: role in inflammation and immune surveillance. Annals of the rheumatic diseases 63 Suppl 2:ii84-ii89. 225