ENHANCEMENT OF RECOMBINANT ADENOVIRUS VECTOR VACCINE EFFICACY BY INNATE IMMUNE MODULATION By Dionisia Marie Quiroga A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of Cell and Molecular Biology – Doctor of Philosophy 2014 ABSTRACT ENHANCEMENT OF RECOMBINANT ADENOVIRUS VECTOR VACCINE EFFICACY BY INNATE IMMUNE MODULATION By Dionisia Marie Quiroga In the field of vaccine development, there is a clear need for novel, effective platforms and adjuvants that can elicit robust, multifaceted adaptive immune responses to antigenic targets. Unfortunately, many methods investigated thus far are either too toxic or too weak to induce a substantial response against difficult targets, such as tumor-associated antigens (TAAs) or antigens from highly-mutagenic pathogens. In this dissertation, we explore the use of recombinant adenovirus serotype 5 (rAd5) platforms as vaccine vectors and the ability of innate immunostimulants to act as concurrently-administered adjuvants. First, we investigated how different variations of rAd5 vectors may promote altered innate immune signaling, and downstream, improved transgene memory responses. rAd5 vectors are strong vaccine candidates due to their intrinsic immunogenicity and potent transgene expression, however, widespread pre-existing Ad5 immunity has been considered a developmental barrier to the use of traditional, first-generation (Ad5[E1-]) vectors. Recently published studies have revealed that additionally E2b-deleted rAd5 (Ad5[E1-,E2b-]) vectors circumvent anti-Ad5 immunity to induce stronger transgene-specific memory responses than Ad5[E1-] vectors, yet it is unknown why these vector differences exist. Therefore, we decided to compare Ad5[E1-] and Ad5[E1-,E2b-] vector-induced innate immune responses by using human peripheral mononuclear cell (hPBMC) samples from multiple donors. We found that Ad5[E1,E2b-] vectors trigger higher levels of pro-inflammatory cytokine secretion and Th1-dominant gene expression; yet lowered amounts of Ad viral-derived gene expression, independent of the donors' pre-existing anti-Ad5 adaptive responses. These results suggest that Ad5[E1-,E2b-], in contrast to Ad5[E1-], vaccines do not promote activities that suppress innate immune signaling, revealing a novel way they may produce superior efficacy and safety profiles, regardless of previous Ad5 immunity. Next, we investigated if and how a recombinant profilin-like immunostimulant derived from the Eimeria tenella protozoan (rEA) may promote innate immunity during rAd5-mediated vaccination. Upon finding that rEA could trigger activation of multiple types of hematopoietic immune cells in a MyD88-dependent, but TRIF-inhibitory, manner; we created a rAd5 vector expressing rEA to test its ability to enhance CMI responses against co-injected vaccine targets. rEA is an attractive vaccine candidate due to previous clinical investigations showing it to be a safe, immunogenic agent with the ability to trigger Th1-predominant responses in vivo. Subsequently, we investigated if an rEA-expressing rAd5 (rAd5-rEA) could promote and/or alter cytotoxic memory responses towards CEA, a colorectal cancer-related TAA. We found that the addition of rAd5-rEA to an Ad-based CEA vaccine (rAd5-CEA) induced a dose-dependent increase in the potency of anti-CEA T cell and B cell responses, elevated the number of CMIand IgG-recognized CEA-specific epitopes, and enhanced in vivo CEA-targeted cell killing, suggesting that co-injection of rAd5-rEA with a TAA-directed vaccine can substantially boost and broaden the TAA-specific memory response. Finally, we briefly discuss the use of an rAd5 expressing endoplasmic reticulum aminopeptidase 1 (ERAP1) and how different polymorphisms of this gene may promote innate immune and cellular stress signaling. Overall, these dissertation findings address how best the features of rAd5-based vectors can be utilized as vaccine models and how modulation of innate immunity can be used to direct and foster vaccine-promoting long-term memory responses. Copyright by DIONISIA MARIE QUIROGA 2014 I dedicate my dissertation to my friends and family, who have supported me whole-hearted throughout my graduate school process and will continue to support me afterwards. For every phone call, every meal, every hug, and every smile, I can’t thank you enough. v ACKNOWLEDGEMENTS To my mentor, Dr. Andy Amalfitano – It is clear that you serve as a model for what D.O./Ph.D students can hope one day become: a successful, intelligent physician scientist who has made tremendous strides in their area of research while still taking the time to provide compassionate treatment to his patients. Of all the hats you wear, you were still able to provide me with the mentorship and education I needed to excel in my studies. You taught me not only how to conduct medical research well, but also how to think outside of the box and be a more independent thinker. And while you have been a more than kind and generous mentor; you did not hesitate to gradually push me to do more than what I even thought I was capable of. Without this motivation, I doubt I would be able to come out of this program with as many skills and experiences as I have now; and I am eternally grateful for all of them you’ve equipped me with. Thank you for believing in me every step of the way; my hope is that one day, I will be able to pass on these same gifts to another young researcher like you did for me. To my guidance committee members, Dr. Walter Esselman, Dr. J. Justin McCormick, Dr. Richard Schwartz, and Dr. Kefei Yu – Thank you so much for your helpful comments and suggestions. You helped me to think constructively and analytically, while also providing input into how best to proceed with my experiments, funding opportunities, and future career goals. To my colleague, Dr. Yasser Aldhamen – Your insight, advice, and day-to-day assistance were invaluable tools to me during the entirety of my graduate experience. As you’ve progressed over the years from graduate student, to post-doctoral scientist, to professor; you’ve allowed me to advance in my skills as a researcher as well. I was so fortunate to have your guidance; it was truly like having a second mentor. vi To my labmates, past and current, including Dr. Fadel Alyaqoub, Dr. Daniel Appledorn, Dr. Charles Aylsworth, Dr. Joyce Liu, Aaron McBride, Yuliya Pepelyayeva, Cristiane Pereira, David Rastall, Sarah Godbehere Roosa, Dr. Nathan Schuldt, Dr. Sergey Seregin, and Dr. Tyler Voss – You have made coming to work every day a pleasure and were more than kind in giving me your assistance and advice throughout the years. I look forward to keeping in touch with everyone, both through your research work and personally. To our undergraduate assistants, past and current, including Marissa Bauters, Christopher Blum, Matthew Bujold, Brandi Burke, Christopher Busuito, John David, Dr. William DePas, Laura Harding, Megan Hoban, Kristen Kenny, William Nance, Doug Peters, Ashley Raedy, and Jennifer Zehnder – I thank you for your many hours of work that allow for the lab functions and experiments to run smoothly. I have enjoyed watching you progress from freshman to graduate/professional students yourself and I’m happy to have had the chance to be a part of that process. To the MSU ULAR and Research Technology Support Facility staff, especially Dr. Louis King in flow cytometry and Dr. Jeff Landgraf and Colleen Curry in genomics – I am so grateful for your aid in learning and working with the technologies and facilities necessary for me to complete my dissertation research. To my graduate program director Dr. Susan Conrad and program manager Becky Mansel – Thank you for giving me the opportunity to conduct my graduate studies in such a supportive and friendly environment. Additionally, your assistance with all the logistics that go into completing a Ph.D was priceless to me. To the MSU D.O./Ph.D program personnel, past and current, including Dr. Justin McCormick, Dr. Veronica Maher, and Bethany Heinlen – I cannot thank you enough for vii allowing me to join this exceptional program. You have helped me through every step and every stumble I made along the way, and always with compassion, resolve, and encouragement. Thank you for keeping me on track, both then and now. To my fellow D.O./Ph.D students, especially Steven Proper, Joseph Prinsen, Darin Quach, and Paul Beach – Thank you so much for all your great, honest advice as we went through this process together. I am so grateful for the friendship and good times I got to have with you during our years here. To Mrs. Lois Rood, and in memory of Suzan Jean Snyder – I thank you for your support and scholarship to assist me in my graduate and medical studies into the field of oncology. I hope that I can contribute back to the field of cancer research and patient care in a way that would honor the memory of your daughter who lost her battle with cancer. To the McNair/SROP program staff and scholars, especially Dr. Nettavia Curry – I have you to thank for first introducing me to the research world as an undergraduate student and helping me to begin development of the skills I would need to pursue a Ph.D and career as a scientist. To my lovely friends, especially Lola Alvarez, Ruby Carrillo, Allie Holschbach, and Jessica Jean – Whatever day or time it was, I could always count on you to be a lending hand, a study buddy, or a dance partner with me. You are overly generous and patient with me and I’m so proud and happy to have you all in my life. To my aunts, uncles, cousins, half-siblings, and other extended family members – Since day one, you’ve always encouraged me to follow my passions. Also, thank you for teaching me about the importance of learning about one’s culture, telling a good joke, reading great books, and drinking excellent wine. viii To my little brother, Michael – Thank you so much for assisting me in the editing and design of some of the dissertation figures. More importantly, thank you for being the steadythinking and collected sibling in our family. You’re an intelligent, caring, and witty person that I am so lucky to call my friend and my brother. To my mom, Lenore – You have always supported me through thick and thin; always putting the needs of your family and others before yourself . Once I got into college, you may not have been able to help me with my homework anymore, but you took the time to help me with every other difficulty I approached, whether it was fixing up things around the house, giving me a place to stay when I had none, making big financial decisions, or planning my next move. You are were and are a shining example to me of what a strong, smart, and caring woman should be like. To my dad, Pablo – Since I was a little girl, you’ve always been my hero; my first and best inspiration to pursue medicine and graduate school. You never tried to push or persuade me into the medical profession; all you needed to do is show me by example what a great physician could do. I can only hope that after getting both my medical degree and my Ph.D, I could possibly be even half the doctor you are. Thank you all for your generosity; I couldn’t have done this without you! ix TABLE OF CONTENTS LIST OF TABLES ....................................................................................................................... xiii LIST OF FIGURES ..................................................................................................................... xiv KEY TO ABBREVIATIONS ...................................................................................................... xvi Chapter 1: Introduction ............................................................................................................... 1 1.1 Innate immunity .................................................................................................................. 2 1.1.1 Definition and overview ......................................................................................... 2 1.1.2 Toll-like receptors and Toll-like receptor agonists ................................................. 5 1.1.3 Anti-viral innate immunity ..................................................................................... 6 1.2 Adaptive immunity ............................................................................................................. 8 1.2.1 T cell-based adaptive immunity .............................................................................. 8 1.2.2 B cell-based adaptive immunity.............................................................................. 9 1.2.3 Effects of TLR signaling on adaptive memory response ...................................... 10 1.3 Vaccine adjuvants ............................................................................................................. 12 1.3.1 Definition and overview ....................................................................................... 12 1.3.2 Currently utilized adjuvants in human vaccines ................................................... 12 1.3.3 TLR agonist adjuvants .......................................................................................... 13 1.4 Recombinant adenoviruses ............................................................................................... 15 1.4.1 Adenovirus biology and structure ......................................................................... 15 1.4.2 Synthesis and utilization of rAd for vaccine vectors ............................................ 17 1.4.3 Innate immune recognition of rAd ........................................................................ 19 1.4.4 Pre-existing immunity against rAd vaccine vectors ............................................. 21 Chapter 2: Decreased vector gene expression from second-generation, E2b-deleted Ad5 vaccines paradoxically intensifies pro-inflammatory immune responses.............................. 23 2.1 Introduction ....................................................................................................................... 24 2.2 Methods............................................................................................................................. 27 2.2.1 Microarray analysis ............................................................................................... 27 2.2.2 Recombinant adenovirus vector construction ....................................................... 27 2.2.3 Cell isolation, culture, and adenovirus infection .................................................. 28 2.2.4 ELISA analysis ..................................................................................................... 28 2.2.5 Cell staining and flow cytometry .......................................................................... 29 2.2.6 RNA extraction, cDNA synthesis, and quantitative RT-PCR .............................. 29 2.2.7 Statistical analysis ................................................................................................. 32 2.3 Results ............................................................................................................................... 33 2.3.1 Hepatocyte microarray analysis following first- or second-generation rAd infection ................................................................................................................ 33 2.3.2 Ad5[E1-,E2b-] vectors induce an increased pro-inflammatory cytokine response upon hPBMC infection ......................................................................................... 48 x 2.3.3 Ad5-induced pro-inflammatory cytokine responses are not correlated to preexisting Ad5 immunity in human blood samples ............................................... 556 2.3.4 Ad5[E1-,E2b-] infection produces dampened Ad gene expression ...................... 61 2.3.5 Ad5[E1-] and Ad5[E1-,E2b-] vectors induce distinct innate immune gene expression profiles ................................................................................................ 68 2.4 Discussion ......................................................................................................................... 70 Acknowledgements ....................................................................................................................... 74 Chapter 3: Utilization of a recombinant adenovirus vector expressing an Eimeria antigenderived TLR agonist as a vaccine adjuvant. ............................................................................. 75 3.1 Introduction ....................................................................................................................... 76 3.2 Methods............................................................................................................................. 78 3.2.1 Animal care and procedures.................................................................................. 78 3.2.2 Recombinant adenovirus vector construction ....................................................... 78 3.2.3 Splenocyte processing ........................................................................................... 79 3.2.4 Cell staining and flow cytometry .......................................................................... 79 3.2.5 Cytokine and chemokine analysis ......................................................................... 80 3.2.6 Enzyme-linked immunosorbent spot (ELISpot) assay analysis............................ 80 3.2.7 Statistical analysis ................................................................................................. 81 3.3 Results ............................................................................................................................... 82 3.3.1 Regulation of rEA-induced dendritic cell responses by TLR adaptor proteins ... …………………………………………………………………………………82 3.3.2 Regulation of rEA-induced natural killer cell responses by TLR adaptor proteins………………………………………………………………………..….88 3.3.3 Murine cytokine response to intramuscular or intraperitoneal rEA protein injections ............................................................................................................... 93 3.3.4 An rEA-expressing recombinant Ad5 vector enhances co-expressed transgenespecific immunity.................................................................................................. 97 3.3.5 Addition of rAd5-rEA improves CMI against a co-injected rAd-expressed transgene ............................................................................................................. 102 3.4 Discussion ....................................................................................................................... 104 Acknowledgements ..................................................................................................................... 106 Chapter 4: Strengthened tumor antigen immune recognition by inclusion of a recombinant Eimeria antigen in therapeutic cancer vaccination ................................................................ 108 4.1 Introduction ..................................................................................................................... 109 4.2 Methods........................................................................................................................... 111 4.2.1 Recombinant adenovirus vector construction ..................................................... 111 4.2.2 Animal care and procedures................................................................................ 111 4.2.3 ELISpot assay analysis ....................................................................................... 111 4.2.4 Cell staining and flow cytometry ........................................................................ 112 4.2.5 In vivo cytotoxic T lymphocyte (CTL) assay ..................................................... 112 4.2.6 Anti-CEA and anti-Ad ELISA analysis .............................................................. 113 4.2.7 Statistical analysis ............................................................................................... 114 4.3 Results ............................................................................................................................. 115 xi 4.3.1 Increased rAd5-CEA transgene-specific CMI responses induced in the presence of rAd5-rEA ........................................................................................................ 115 4.3.2 Improved cytotoxic T cell responses against CEA following high-dose rAd5-CEA + rAd5-rEA co-administration ............................................................................ 117 4.3.3 Dose-dependent increase in anti-CEA IgG production following rAd5-CEA + rAd5-rEA co-administration ............................................................................... 120 4.3.4 rAd5-CEA + rAd5-rEA co-injection enhances breadth of T cell-recognized CEA peptide epitopes .................................................................................................. 122 4.3.5 rAd5-CEA + rAd5-rEA co-injection enhances diversity of anti-CEA IgG antibody-identified CEA peptide epitopes .......................................................... 130 4.3.6 Enhanced in vivo CEA-targeted CTL killing following rAd5-CEA + rAd5-rEA co-administration ................................................................................................ 134 4.4 Discussion ....................................................................................................................... 137 Acknowledgments....................................................................................................................... 142 Chapter 5: Recombinant adenoviruses expressing antigen-trimming protein ERAP1 trigger a robust innate immune response. ........................................................................................... 143 5.1 Introduction ..................................................................................................................... 144 5.2 Methods........................................................................................................................... 146 5.2.1 Cell transfection and culture ............................................................................... 146 5.2.2 Recombinant adenovirus construction and cell infection ................................... 147 5.2.3 RNA extraction, cDNA synthesis, and quantitative RT-PCR ............................ 147 5.2.4 Protein isolation and western blotting................................................................. 149 5.2.5 In vitro phagocytosis assay ................................................................................. 149 5.2.6 Statistical analysis ............................................................................................... 150 5.3 Results ............................................................................................................................. 151 5.3.1 ERAP-1 expression following transfection of HeLa-Kb-B27/47 cells............... 151 5.3.2 Ad-ERAPlow and Ad-ERAPhigh activation of AS-related gene immune responses ............................................................................................................. 156 5.3.3 Phagocytosis activity of RAW macrophages following Ad-ERAP stimulation..159 5.4 Discussion ....................................................................................................................... 163 Acknowledgments....................................................................................................................... 166 Chapter 6: Summary and future directions ........................................................................... 167 BIBLIOGRAPHY ....................................................................................................................... 176 xii LIST OF TABLES Table 1 Quantitative RT-PCR primers for rAd5[E1-,E2b-] studies……………………...31 Table 2 Ontological categorization of genes differentially and significantly expressed following in vivo Ad5[E1-] or Ad5[E1-,E2b-] infection………………………...40 Table 3 Individual hPBMC sample cytokine inductions…………………………………52 Table 4 CEA peptide epitope sequences and assigned numbering………………...……123 Table 5 Quantitative RT-PCR primers for ERAP1 studies……………………………...148 xiii LIST OF FIGURES Figure 1 rAd5 genome and recombinant rAd5 platforms…………………………………16 Figure 2 Functional analysis of murine hepatocytes following in vivo Ad5[E1-] and Ad5[E1-,E2b-] infection ………………………………………………………...35 Figure 3 Ad5[E1-,E2b-] infection induces increased hPBMC pro-inflammatory cytokine secretion……………………………………………………………………….…50 Figure 4 Quantity of Ad5[E1-,E2b-]-induced hPBMC pro-inflammatory cytokine secretion is not correlated to pre-existing anti-Ad5 immunity……………………………..57 Figure 5 Ratio of Ad5[E1-,E2b-] to Ad5[E1-] induced cytokine production is not correlated to pre-existing anti-Ad5 immunity……………………………………60 Figure 6 Ad5[E1-,E2b-] infection produces dampened Ad gene expression……………...63 Figure 7 Ad5[E1-] and Ad5[E1-,E2b-] viral genome quantification following human PBMC infection………………………………………………………………….67 Figure 8 Ad5 vectors promote distinct innate immune gene expression patterns dependent on vector generation……………………………...………………………………69 Figure 9 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF……………………………………………………………………..……….83 Figure 10 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF (histogram plots)…………………………………………………....……..85 Figure 11 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF (MFI)…………………………………………………………..……..…....87 Figure 12 rEA-mediated NK cell activation is differentially regulated by MyD88 and TRIF.......................................................................................................................90 Figure 13 rEA-mediated NK cell activation is differentially regulated by MyD88 and TRIF (histogram plots)………………………………………………………..……..…92 Figure 14 rEA protein promotes cytokine and chemokine induction mainly independent of injection route…………………………………………………………..………..95 Figure 15 rAd5-rEA construction……………………………………………………..…….99 xiv Figure 16 Improved cell-mediated immunity towards co-expressed transgene following rAd5-rEA vaccination…………………………………………..…………........101 Figure 17 rAd5-rEA induces heightened HIV Gag-specific CMI responses………..….....103 Figure 18 Strong CEA-specific cell-mediated immunity produced by rAd5-CEA and rAd5rEA co-vaccination……………………………………………………………..116 Figure 19 High-dose rAd5-CEA and rAd5-rEA administration induces a robust, CEAspecific CD8+ cytotoxic T cell response…………………………………….…119 Figure 20 Anti-CEA IgG production is potently induced by rAd5-CEA and rAd5-rEA coinjection…………………………………………………………………………121 Figure 21 rAd5-CEA and rAd5-rEA co-vaccination induces CMI against multiple unique CEA peptide epitopes………………………………………………………..…126 Figure 22 Addition of rAd5-rEA to an rAd5-CEA vaccine regimen induces CMI against multiple CEA peptide regions…………………………………………………..129 Figure 23 rAd5-CEA and rAd5-rEA co-injection induces creation of IgG antibodies against a diverse area of CEA peptide regions………………………………………….131 Figure 24 High anti-CEA IgG binding affinity following rAd5-CEA and rAd5-rEA coinjection…………………………………………………………………………133 Figure 25 rAd5-CEA and rAd5-rEA co-injection induces significant CEA-specific in vivo cytolytic T cell killing activity……………………………………………….…135 Figure 26 ERAP1 expression following HeLa-Kb-B27/47 transfection…………………..152 Figure 27 Expression of AS-related genes in ERAP1-transfected HeLa-Kb-B27/47 cells………………………………………………………………..……………155 Figure 28 Ad-ERAPlow and Ad-ERAPhigh induce AS-related gene expression………...158 Figure 29 Ad-ERAP stimulates RAW macrophage phagocytosis activation……………..162 xv KEY TO ABBREVIATIONS μL microliter(s) μM micromolar Ad adenovirus Ad5[E1-] first-generation recombinant adenovirus Ad5[E1-,E2b-] E2b-deleted, second-generation recombinant adenovirus Ad5[E1-,E4-] E4-deleted, second-generation recombinant adenovirus ADCC antibody-dependent cell-mediated cytotoxicity ANOVA analysis of variance APC antigen presenting cell AS ankylosing spondylitis AS03 adjuvant system 03 AS04 adjuvant system 04 ATP adenosine triphosphate BCG Bacillus Calmette–Guérin BCR B cell receptor BH FDR MTC Benjamini-Hochberg false discover rate multiple test correction BiP binding immunoglobulin protein bp base pair(s) CA-125 cancer antigen 125 CAR coxsackievirus and adenovirus receptor CD cluster of differentiation xvi CD40L cluster of differentiation 40 ligand cDNA complementary deoxyribonucleic acid CEA carcinoembryonic antigen CFSE carboxyfluorescein succinimidyl ester CLR C-type lectin receptor CMI cell-mediated immunity CMV cytomegalovirus CNS central nervous system CpG cytosine – phosphate – guanine CTL cytotoxic T lymphocyte CXCL10 C-X-C motif chemokine 10 DC dendritic cell DKO double knockout DNA deoxyribonucleic acid dpi day(s) post-injection dsDNA double-stranded deoxynucleic acid dsRNA double-stranded deoxynucleic acid EASE Expression Analysis Systematic Explorer ELISA enzyme-linked immunosorbent assay ELISpot enzyme-linked immunospot EMCV encephalomyocarditis virus ER endoplasmic reticulum ERAP1 endoplasmic reticulum aminopeptidase 1 xvii FACS fluorescence-activated cell sorting FBS fetal bovine serum Fc fragment crystallizable FDA Food and Drug Administration GAPDH glyceraldehyde 3-phosphate dehydrogenase GBP2 interferon-induced guanylate-binding protein 2 G-CSF granulocyte colony-stimulating factor GFP green fluorescent protein GI gastrointestinal GM-CSF granulocyte-macrophage colony-stimulating factor GO gene ontology HBV hepatitis B virus HCV hepatitis C virus HD-Ad5 helper-dependent adenovirus serotype 5 HEK293 human embryonic kidney 293 HIV human immunodeficiency virus HLA human leukocyte antigen hPBMC human peripheral blood mononuclear cell hpi hour(s) post-injection hpt hour(s) post-transfection HPV human papillomavirus HRP horseradish peroxidase HSV herpes simplex virus xviii ICS intracellular staining IFIT interferon-induced protein with tetratricopeptide repeats 1 IFN interferon Ig immunoglobulin IκB nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor IKK-β inhibitor of nuclear factor kappa-B kinase subunit beta IL interleukin IM intramuscular(ly) IP intraperitoneal(ly) IPA Ingenuity Pathway Analysis IRAK interleukin-1 receptor-associated kinase IRF interferon regulatory factor ITR inverted terminal repeat IV intravenous(ly) kb kilobase(s) kDa kilodalton(s) KO knockout LPS lipopolysaccharide LRR leucine-rich repeat LTA lipoteichoic acid MALP-2 macrophage-activating lipopeptide 2 MAP3K7 mitogen-activated protein kinase kinase kinase 7 xix MAPK mitogen-activated protein kinase MAVS mitochondrial antiviral-signaling protein MDa megadalton(s) MDA5 melanoma differentiation-associated protein 5 MFI mean fluroescence intensity mg milligram(s) MHC major histocompatibility complex miRNA micro ribonucleic acid mL milliliter(s) MLP major late promoter MPL monophosphoryl lipid A MyD88 myeloid differentiation primary response gene 88 NALP3 nucleotide binding oligomerization domain-like receptor-like receptor family, pyrin domain containing 3 NCBI National Center for Biotechnology Information NFκB nuclear factor kappa B ng nanogram(s) NK natural killer NKT natural killer T NLR nucleotide binding oligomerization domain-like receptor OD optical density ODN oligodeoxynucleotide(s) OTC ornithine transcarbamylase PAMP pattern-associated molecular pattern xx PBS phosphate buffer saline PCR polymerase chain reaction PKR protein kinase ribonucleic acid-activated PMA phorbol myristate acetate Pol DNA polymerase poly(I:C) polyinosinic:polycytidylic acid PRR pattern recognition receptor PSF penicillin-streptomycin-fungizone PTP precursor terminal protein rAd recombinant adenovirus rAd5 recombinant adenovirus, serotype 5 RANTES regulated on activation, normal T cell expressed and secreted RBC red blood cell rEA recombinant Eimeria antigen RFP red fluorescent protein RIG-I retinoic acid-inducible gene I RIPK1 receptor-interacting serine/threonine-protein kinase 1 RLR retinoic acid-inducible gene-I-like receptor RNA ribonucleic acid RNAi ribonucleic acid interference RT-PCR reverse transcription polymerase chain reaction SFC spot-forming cells ssRNA single-stranded ribonucleic acid xxi TBK1 tumor necrosis factor receptor-associated factor family memberassociated nuclear factor kappa B activator-binding kinase 1 TCR T cell receptor Th T helper TIR Toll/interleukin-1 receptor TIRAP Toll-interleukin 1 receptor domain containing adaptor protein TLR Toll-like receptor TMB tetramethylbenzidine TNF tumor necrosis factor alpha TRAF tumor necrosis factor receptor associated factor TRAM Toll/interleukin-1 receptor -domain-containing adapter-inducing interferon-β-related adaptor molecule Treg Regulatory T cell TRIF Toll/interleukin-1 receptor -domain-containing adapter-inducing interferon-β UPR unfolded protein response U.S. United States UV ultraviolet VARNA viral-associated ribonucleic acid viperin virus inhibitory protein, endoplasmic reticulum-associated, interferon-inducible vp viral particle(s) VSV vesicular stomatitis virus wpi week(s) post-infection XBP1s X-box binding protein 1s xxii Chapter 1: Introduction 1 1.1 Innate immunity 1.1.1 Definition and overview In all mammalian organisms, innate immunity serves as the first-line defense against invading pathogens. The initially encountered components of this system include basic physical barriers by epithelial cells, constitutive expression of peptide antibiotics, and phagocytosismediated killing by neutrophils (1). This response progresses to cell-mediated promotion of inflammatory signals and processing of microbial antigens so as to recruit downstream adaptive responses (2,3). As opposed to adaptive immunity, elements of the innate immune system are intrinsic to the organism’s germline DNA and are ubiquitous across all cells within that lineage (2). While this limits the diversity of molecules that the innate immune response can detect, its molecules mainly recognize microbially-conserved components called pathogen-associated molecular patterns (PAMPs) (3,4). Importantly, PAMPs remain recognizable even across species or when minor genetic variations are present. For example, retinoic acid-inducible gene-I-like receptors (RLRs) detect viral dsRNA from many different viruses and in a manner that is dependent on the dsRNA’s length, but not sequence (3). Human cells contain a considerable amount of PAMP-recognizing surface, endosomal, and cytoplasmic pattern recognition receptors (PRRs), including C-type lectin receptors (CLRs), nucleotide binding oligomerization domainlike receptors (NLRs), RLRs, and Toll-like receptors (TLRs) (2,3); the latter of which will be detailed in the next section. While activation of PRRs trigger a general pro-inflammatory signaling cascade, they can also lead to unique effects depending upon the specific PRR, the cell type that is expressing it, and the immune environment that surrounds it (5). The human cells that express PRRs are mainly derived from a hematopoietic lineage, including monocytes, macrophages, dendritic cells 2 (DCs), natural killer (NK) cells, B cells, and some T cells; however, PRRs can also be found on non-immune cells, such as epithelial and endothelial cells (2,6). Macrophages, DCs, and a small subset of B cells are considered professional antigen-presenting cells (APCs), meaning they are particularly effective at internalizing microbial pathogens (typically through receptor-mediated endocytosis or phagocytosis), processing them into small antigenic peptides, and displaying them extracellularly on class II MHC molecules (7). Tissue macrophages are blood monocyte-derived cells that both act as professional APCs as well as phagocytes. While neutrophils may be the most predominant phagocytic cells in the human body and are typically considered “first-responder” immune cells, macrophages are able to differentiate into specialized phagocyte forms (e.g. Kupffer cells in the liver, osteoclasts in the bone, alveolar macrophages in the lungs, microglial cells in the central nervous system (CNS)), become activated to kill vacuolated pathogens, and present antigens to T cells (3,8). Macrophages can also be activated by the binding of cytokines, chemokines, or PAMPs to the appropriate PRRs (e.g. mannose receptor binding triggers phagocytosis; TLR or cytokine receptor triggering activates inflammation, antigen presentation, and co-stimulatory molecule expression) (8). These cells can also release their own cytokines and chemokines, which help to alert other macrophages and innate immune cells to become activated and migrate towards the site of infection. The other major professional APC that humans possess are DCs. These cells possess many of the same traits as macrophages, but their function is more specific to the effective display of antigenic epitopes; often being seen as the “bridge” between the initial innate response and the developing adaptive immune response (9). Immature/migratory DCs are present in many parts of the human body, for example, in local lymph nodes, as Langerhans cells in the skin, and 3 as Peyer’s patch colonic DCs in the gastrointestinal (GI) system (9–11). When activated by PRR stimulation, DCs also express cytokine molecules like macrophages, but do not destroy engulfed microbes. Instead, DCs quickly migrate to lymph nodes or other areas of high T cell concentrations to initiate the required signals to form an eventual adaptive immune response (9,12). NK cells serve as another subset of immune cells that contribute to the innate immune response. While recent studies have found them to have properties of adaptive immune lymphocytes as well (13,14), the main function of NK cells are to specifically browse their environment for cells that either lack surface class I MHC molecules or display other danger/immune activation signals that would suggest the cell was infected or possibly tumorigenic (15,16). When NK cells identify dysfunctional cells, they come into close contact with them and release α-defensins, perforin, and other granzymes, thereby triggering the targeted cell to undergo cytolytic granule-mediated apoptosis (15,16). While other lymphocytes are slower to act and can only identify a single antigen epitope per cell, NK cells are able to quickly kill infected or abnormal cells that may have otherwise been able to evade T and B cell recognition due to lack of antigen presentation (17). Phenotypically similar to NK cells and T cells, natural killer T (NKT) cells are also contributors towards the overall innate immune schema. As the existence of these as an independent lymphocyte group is still moderately new (18), their exact mechanisms and purpose are not completely clear. However, it has been found that NKT cells are able to secrete large amounts of pro-inflammatory cytokines (e.g. GM-CSF, IFNγ, IL-2, TNFα) upon activation (19). These cells have also been found to assist in maturation of DCs when working in concert with certain TLR agonists (20). 4 1.1.2 Toll-like receptors and Toll-like receptor agonists TLRs were named as such due to their homology with the Drosophila fly species’ Toll protein, which was found to be an anti-microbial agent (21,22). All TLR molecules share sequence homology in that they are composed of a cytosolic Toll/IL-1R (TIR) domain and several lumenal or extracellular leucine-rich repeat (LRR) domains. TLRs are unique receptors in that they can recognize both nucleic acids and proteins. For example, TLR1, TLR2, and TLR6 are activated by binding to lipopeptides, LTA, peptidoglycans, porins, mannans, and zymosan. TLR4 is also able to recognize mannans, but is best characterized as responding to Gramnegative bacteria LPS and viral envelope proteins (6,21,23). TLR5 is more restricted, and is mainly cited as recognizing bacterial flagellins (24). While these aforementioned TLRs are positioned within the plasma membrane and mostly recognize protein structures; TLR3, TLR7, TLR8, and TLR9 are embedded in endosomes and are mainly activated by foreign forms of nucleic acids (e.g. unmethylated CpG dinucleotides, ssRNA, dsRNA) (25,26). To transmit signals to the nucleus following ligand binding, TLRs form homodimers or, in some cases, heterodimers to other TLR molecules (e.g. TLR2 with TLR1 or TLR6 (27,28)). Conformational protein changes upon dimerization then allow for recruitment of one or more cytosolic adaptor proteins. All human TLRs, with the exception of TLR3, utilize the adaptor protein MyD88 for signal transduction (5). MyD88 works to recruit IRAK kinases, which in turn phosphorylates TRAF6; subsequently leading to the polyubiquitination of itself and MAP3K7. This action allows for IKK-β, then IκB, to be phosphorylated; freeing NFκB to migrate to the nucleus and activate pro-inflammatory gene signaling (29,30). TLR3 and TLR4 use a TRIF-dependent pathway to initiate NFκB signaling; this involves TRIF activating RIPK1, thereby inducing polyubiquitination of itself and MAP3K7, and from 5 there on, proceeding in the same manner as the MyD88-dependent signaling cascade. TRIF also can form a complex with TBK1 which can phosphorylate IRF3 and allow it to travel to the nucleus and induce IRF3-mediated type I IFN production and signaling (6,23). TIRAP and TRAM are also used in conjunction with TLRs and other adaptor proteins to signal NFκB and IRF3, as well as multiple MAPK systems (23). 1.1.3 Anti-viral innate immunity During virus infection, innate mechanisms are triggered by many unique components of these microbes. For example, TLR2 is able to recognize extracellular glycoproteins produced by CMV and HSV (31,32). TLR4, another plasma membrane-bound PRR originally believed to only recognize bacterial motifs, has also been found to recognize viral proteins from such microbes as VSV and Ebola virus (33,34). When viruses are taken into cells via endosomal transport, they are exposed to several endosomal TLRs (35). Endosome-bound TLR3 is able to detect dsRNA, therefore, is able to be triggered by dsRNA from viral genomes or secondary dsRNA-like structures (25,30). TLR7/TLR8 also contribute to endosomal virus recognition in that they are directly activated by viral-derived ssRNA (36). Moreover, TLR9 binds to and is activated by unmethylated CpG sequences, which are rare in typical human cell systems, but much more common in viruses and bacterial organisms (37,38). Cytosolic PRRs, such as RLRs, also contribute greatly to the initial immune response to viral infection (39). RIG-I binds and reacts to viral short dsRNA and 5’ triphosphate ssRNA, which are produced by such microbes as the influenza virus, hepatitis C virus (HCV), and Newcastle disease virus (40,41). Alternatively, MDA5 uses long dsRNA as a ligand; this structure is often derived from such pathogens as the West Nile virus and encephalomyocarditis virus (EMCV) (42,43). 6 Despite the plethora of anti-viral mechanisms in place, it is important to recognize that they are not fool-proof, as many viruses have evolved to evade innate immune detection and response (44). Human CMV protein products are able to keep MHC I from being expressed on the cell surface so as to avoid T cell recognition (45). Similar MHC I inhibitory viral defense phenomena have also been described with infection by HIV and adenoviruses (46,47). While NK cells typically would identify these class I MHC-lacking cells as damaged and cause them to undergo cytolysis, the some viruses are also able to block this action by producing class I MHC molecule homologs to trick the NK cells into seeing the virus-infected cell as normal (48). Viruses are also able to create cytokine receptor homologs or directly bind to and inhibit cytokine and cytokine receptors which are important for the anti-viral response, including such targets as TNF receptors (49,50), IFNs (51,52), and chemokine receptors (53,54). Anti-viral innate immune responses also work sequentially to affect adaptive memory responses in positive and/or negative ways. For example, in a HIV vaccine study, TLR3 stimulation was found to decrease cytotoxic lymphocyte (CTL) responses against the HIV protein Gag, while application of a TLR4 agonist boosted CD8+ T cell function (55). These innate immune responses are important to consider during the utilization of viral-based therapeutic delivery methods (56) and will be further addressed in the subsequent sections. 7 1.2 Adaptive immunity 1.2.1 T cell-based adaptive immunity T cells represent one of the two major cells involved in the adaptive memory response. Naïve versions of these cells are residents of the spleen and lymph nodes, where they can frequently interact with antigenic peptides being presented on MHC molecules by APCs (57). When combined with the expression of co-stimulatory factors, T cell receptors (TCRs) can react to non-self antigens in a multitude of ways. Each T cell contains only one specific type of TCR; therefore, during T cell activation, it replicates by clonal expansion to respond with greater numbers to the specific antigen recognized (58). For CD4 receptor-expressing T helper (Th) cells, recognition of foreign antigens presented by class II MHC molecules leads to an extensive production of cytokines that recruit other T cells, B cells, and APCs (58). These CD4+ T cells can also be stimulated to differentiate into different Th subtypes, allowing them to respond in a manner that is specific to the type of immune response required to act against the pathogenic threat (59). Alternatively, CD8-expressing CTLs identify antigens presented by class I MHC molecules, and upon recognition of foreign antigen epitopes alongside the appropriate costimulatory signals, are activated to kill the MHC-antigen complex-bearing cell by release of cytotoxins or Fas-Fas ligand binding-mediated apoptosis, a process known as cell-mediated immunity (CMI) (60). The final prominent group of T cells present in the human body are called regulatory T cells (Tregs). These cells use similar antigen recognition techniques as Th cells and CTLs, but function so to suppress antigenic reactivity and promote antigen immune tolerance instead (61). Treg dysfunctions can lead to the development of autoimmune diseases as they act as a major protectant against self-antigen immunogenicity (62). On the other hand, they have also been 8 implicated in perpetuating immunological tolerance towards tumor cells due to their phenotypic similarities with normal, healthy cells (63,64). 1.2.2 B cell-based adaptive immunity B cells are another adaptive immune cell that possesses unique cellular receptors for specific antigenic epitopes and can induce robust responses against identified foreign antigens (65,66). As opposed to T cells, B cell receptors (BCRs) have the ability to recognize protein and non-protein antigens, like those that are membrane-bound or repetitive (66,67). Following exposure to an antigen, naïve B cells undergo clonal expansion and differentiation into effector plasma cells, which secrete antibodies containing the identical epitope binding receptor pattern as the cell’s original BCRs (68). Initially, B cells can perpetuate a “primary response”, during which it produces low strength, variable IgM antibodies (67). During the “secondary response”, a later and larger wave of antibodies is produced, including higher-affinity IgG antibodies and sometimes specialized antibody classes (e.g. IgA, IgE) (69). Besides microbial antibody binding, B cells can also interact with CD4+ Th cells in lymphoid follicles and present antigens to them via class II MHC complex (70). This union allows for Th cell activation, leading to the secretion of cytokines and surface expression of the co-stimulatory molecule CD40L. In turn, CD40 molecules and cytokine receptors on the B cell surface can engage CD40L and cytokine proteins, respectively, allowing them to be subsequently activated themselves (71). Moreover, antibodies secreted from B cells can assist in NK cell killing. Following IgG antibody binding to a surface antigen, NK cells are able to recognize their exposed Fc regions and attach to them with their own Fcγ receptors. This antibody-dependent cell-mediated cytotoxicity (ADCC) interaction allows for NK cells to be activated and kill antibody-bound cells that may have evaded cytolytic killing otherwise (16,72). 9 1.2.3 Effects of TLR signaling on adaptive memory response For there to be a strong and useful adaptive memory response mounted against an antigenic target, it is vital that a robust innate immune response be created upon initial antigen encounter. Cellular TLR activation has been linked in multiple cell and pathogen models as being an important promoter of pro-inflammatory cytokine expression, thereby activating other immune cells, steering CD4+ Th cells away from an anergic state, and even reprogramming Tregs into Th cells (73). Moreover, animals deficient in MyD88 TLR adaptor protein have been found to have blunted Th cell differentiation abilities and absent Th cell-dependent IgG antibody responses (74). The functions of CD8+ T cells are also dependent upon innate immune function. In TLR2 knockout (KO) mice, memory CD8+ T cells were found to be completely absent while other types of CD8+ T cells were also dysfunctional in that they were unable to resolve a Listeria monocytogenes infection (75). Alternatively, TLR2 stimulation in wild-type animals was found to promote the expression of granzymes from activated CTLs (76). Signaling through TLRs is also required in situations where the reversal of Treg-induced CD8+ T cell tolerance towards a specific antigen is desired (77). NK cells have been found to be important mediators of the adaptive memory response as well. When these cells induce cell-mediated killing of infected targets, NK cells are able to induce CD4+ T cell activation in a MyD88- and TRIF-dependent fashion (78). NK cells also use this pathway and IFN cytokine signaling to stimulate humoral and CD8+ T cell memory responses, making them an important component in several facets of adaptive immunity (78,79). Additionally, NKT-dependent activation of DCs has been shown to specifically prime these cells for a more robust anti-antigen CD4+ and CD8+ T cell response (80,81). 10 Finally, TLRs present on DCs have a vital role in determining how T cells they interact with will differentiate and activate (82,83). TLR9 stimulation within certain DC subsets increase their expression of pro-migratory cytokines that propel them to the lymph nodes and allow for increased rates of T cell maturation (84). Both TLR9-deficient and MyD88-deficient mice have been found to have virtually non-existent levels of DC proliferation and maturation, meaning they are unable to enact any meaningful antigen presentation to T cells (36). 11 1.3 Vaccine adjuvants 1.3.1 Definition and overview Vaccines provide prophylactic and/or therapeutic protection against specific pathogens or cell types by inducing a memory response against one or more pathogen-derived antigens that the vaccine provides or expresses. It is common that introduction of a target antigen/pathogen alone is not sufficient to induce protective/therapeutic immunity, therefore, vaccine adjuvants must be added so as to boost the strength, speed, and breadth of immunologic memory response that the host will eventually mount against the desired antigen(s) (85,86). 1.3.2 Currently utilized adjuvants in human vaccines Vaccine adjuvants can be composed of a wide variety of substances, including salts, proteins, oils, virosomes, and nucleic acids (87). Despite the considerably great amount of time that has been spent studying a wide spectrum of these compounds, there are only two specific adjuvants approved by the U.S. FDA for use in the general human population (86). The first, and by far most popular, adjuvant licensed for use is alum, which is composed of aluminum salts (88). Many vaccines contain some form of alum, including those for the prevention of diphtheria, tetanus, pertussis, and hepatitis A (89). The mechanism by which alum functions is believed to be TLR-independent, involving the NALP3 inflammasome instead to potentiate IL-1β production, DC activation, and upregulated pathogen internalization by DC and B cells (90,91). However, using alum as an adjuvant is not a perfect fit for all vaccine regimens as it mainly promotes humoral and Th2-dominant responses, both adaptive pathways that favor B cell over T cell activation (89). Alternatively, a strong T cell-mediated Th1 response would be needed to boost therapeutic cancer vaccines, as these typically require robust CTL killing activities over development of neutralizing antibodies to be effective (92). 12 AS04 is another approved adjuvant and is made up of both monophosphoryl lipid A (MPL) and either aluminum hydroxide or aluminum phosphate (87). It can be found both in vaccines against human papillomavirus (HPV) and in newer versions of the hepatitis B virus (HBV) vaccines, which have actually been shown to be more immunogenic than alum-adjuvant HBV vaccine preparations (93–95). In comparison to alum alone, AS04 has been found to increase both humoral and Th1 responses against co-administered antigens; this is most likely due to MPL being a TLR4 agonist, thereby, it can substantially activate NFκB and MAPK pathway signaling (95,96). Technically, the influenza vaccine adjuvant AS03 (a combination of squalene, DL-αtocopherol, and polysorbate 80) could be counted as a third U.S. FDA-approved adjuvant (97). However, due to some safety concerns, including the possibility that it may induce severe anaphylactic reactions and acute narcoleptic symptoms upon administration, this adjuvant and vaccine formulation are not commercially available and are only stockpiled by the U.S. government for cases in which public health crises may validate it’s use (98,99). 1.3.3 TLR agonist adjuvants As the TLR4 agonist-containing AS04 and other similar adjuvants have shown great promise in the past couple decades, there has been much interest in developing other adjuvants that trigger TLR signaling, perhaps in an even more robust manner. As established earlier, TLRs help to bridge innate and adaptive responses in a multitude of ways, including assisting in the maturation/activation/migration of APCs, elevating pro-inflammatory cytokine secretion, inducing lymphocyte differentiation into more specialized memory cells, and combating antigen tolerance signals (20,73–84,100). Besides MPL, other examples of TLR agonist adjuvants that have been previously studied include MALP-2 (TLR2 and TLR6 agonist), poly(I:C) (TLR3 13 agonist), flagellin (TLR5 agonist), imidazoquinolins (TLR7/8 agonists), and CgG ODN (TLR9 agonist) (101–106), resulting in different levels of efficacy. It should be stated that while many of these components have the potential to activate strong anti-antigen immune responses, some produce such an excessive amount of TLR stimulation that it may cause significant side effects. One example of such a reaction was seen in human trials utilizing the tuberculosis vaccine and TLR agonist, BCG, which triggered acute pneumonitis and hepatitis in some participants (107– 109). Consequently, it is important that the actions of an adjuvant be balanced precisely to allow for beneficial immune stimulation without toxicity. 14 1.4 Recombinant adenoviruses 1.4.1 Adenovirus biology and structure Human adenoviruses (Ads) are a non-enveloped dsDNA viral species which belong to the Mastadenovirus genus and Adenoviridae family (110). Currently, there are over 50 identified human Ad serotypes, each belonging to one of six subgroups (A, B, C, D, E) (110). Each possess an outer icosahedral capsid structure that measures about 90 nm in length with a weight of approximately 150 MDa (111,112). The capsid structure includes seven proteins (II, III, IIIa, IV, VI, VIII, and IX) and is predominantly made up of hexon and penton formations (113). Some penton complexes are also attached to fiber homotrimers that project externally from the virus; these serve as viral binding knobs which can attach to host cell coxsackievirus and adenovirus receptors (CARs), thereby allowing Ad to be internalized (112,114). Internally, the Ad core is made up of four proteins (V, VII, X, terminal 5’ DNA-linked protein) and the viral genome (113). The Ad genome is approximately 36 kb long, which includes the early (E1a, E1b, E2a, E2b, E3, and E4), late (L1, L2, L3, L4, and L5), and other smaller transcriptional units (IVa2, IX, VAI, and VAII) (Figure 1) (110,115,116). Additionally, Ads also contain a major late promoter (MLP) sequence that assists in enhancing late (as well as early) Ad gene transcription and 100-140 bp inverted terminal repeat (ITR) elements that flank the genomic ends (110,115– 118). 15 Figure 1 rAd5 genome and recombinant rAd5 platforms The top figure represents the wild-type adenovirus serotype 5 genome. The genome extends for approximately 36 kb and is divided into the early (E1A, E1B, E2A, E2B, E3, and E4) and late (L1, L2, L3, L4, and L5) transcription units. pIX, IVa2, VAI, and VAII are transcribed during this process as well. ITRs and MLP are also pictured. Below this genome schematic are bars highlighting the areas in which different generations of rAd5 vectors are modified from the original Ad5 genome, as indicated by Δ marked transcription units. Areas of transgene insertion are also indicated. 16 Viral replication occurs at a rapid rate; with a typical 24 hour life cycle yielding approximately 10,000 viral particles (vp) per infected cell (116). While some human Ad species are symptomatically benign, many are able to induce mild respiratory disease, conjunctivitis, pharyngitis, and/or gastroenteritis (119). Occasional cases of severe Ad-induced acute respiratory disease have also been described, although outbreaks seem to be confined in areas where living conditions are crowded, such as in military barracks (119,120). 1.4.2 Synthesis and utilization of rAd for vaccine vectors Recombinant adenoviruses (rAds), particularly serotype 5, are currently one of the most studied therapeutic viral vectors and possess many attractive attributes for clinical use. For example, as wild-type adenoviruses typically include mild cold-like symptoms and are then processed to rAds to make them replication incompetent, these resulting vectors typically produce only minor side effects when used as vaccine platforms (e.g. erythema at injection site) (121). rAd5 vectors have particularly well-characterized genomes, allowing for researchers to design transgene-expressing Ad-based vectors simply and easily. Once the rAd5 construct is synthesized, growing the virus to a substantial amount and concentration is quick and effective, allowing for the accumulation of a potent dosage that can easily be scaled up or down, dependent on the desired utilization. rAds also do not integrate their own genome into that of the host cell; effectively eliminating the risk of vector-induced mutagenesis and oncogenesis that is present with other viral vectors (e.g. retroviruses) (122,123). rAd5 vectors can be utilized to infect both dividing and non-dividing cells; allowing them to have a large infection tropism as they are able to enter cells via the largely ubiquitous CAR and α-integrin binding, as well as other host cell receptors (124–126). Moreover, rAds5 vectors possess intrinsic immunogenicity that improve 17 adaptive memory responses when they are used as vaccine platforms; this characteristic will be further discussed in the following section (127). To date, there have been many recombinant constructs created from the Ad5 genome backbone (Figure 1). The most prolifically used rAd5 platform is surely the first-generation rAd5 vector (Ad5[E1-]), which contains E1 and E3 deletions. The E1 transcription region encodes for a trans-acting transcriptional regulatory factor needed for efficient transcriptional activation and viral replication. In addition, E1 encodes for multiple proteins that can work to suppress apoptosis, inhibit IFN signaling, and block cellular p53 signaling; allowing for virus to persist and defer recognition (121,128). The E3 region also contains several genes which translate into inhibitors that work against class I MHC molecule mobilization and other host anti-viral responses, however, E3 is not required for viral replication and is mainly removed from rAds to allow for a larger transgene insertion area (116,121). Deletion of both of these regions allows for a total transgene insertion size of about 8 kb and can be easily grown when introduced to HEK293 cells (115,116,129). Unfortunately, Ad5[E1-] vectors do not persist for long periods of time in vivo and of all of the rAd5 vector options, produces the highest amount of inflammatory side effects (130–132). Second-generation rAd5 vectors contain E1 and E3 transcriptional unit deletions too, but also have additional genomic areas removed, such as E2b or E4. Vectors with the additional E4 deletion (Ad5[E1-,E4-]) lack genes that promote viral gene expression and replication, as well as lytic function signals (133). These vectors have been found to induce slightly lower levels of inflammation, but have profound issues in maintaining transgene expression (134,135). In contrast, second-generation, E2b-deleted vectors (Ad5[E1-,E2b-]) lack the genes that encode for viral DNA polymerase (Pol) as well as a precursor terminal protein (PTP) region which acts as a 18 primer for viral DNA replication (136,137). While no more difficult to reproduce than Ad5[E1-] vectors (requiring instead a modified HEK-293-based cell line that also expresses the Ad5 E2b genes in trans), Ad5[E1-,E2b-] vectors have been found to induce less toxicity and persist for long periods of time within various tissues of immune-competent animals (116,131,138). A “third” generation of rAd5 vector also exists which are referred to as gutted or helperdependent rAd5, as they lack any Ad-specific genes and require trans-complementation from a first-generation Ad5 vector to propagate (139,140). As can be expected, these vectors have the largest transgene cloning capacity possible for a rAd-based system (about 37 kb). The functional benefits and disadvantages of using each rAd generation will be further compared in the last section below. 1.4.3 Innate immune recognition of rAd Using rAd5 vectors as a vaccine platform carries the added benefit of introducing intrinsically immunogenic virus-derived components that can act as an innate immune boosting mechanism, further enhancing adaptive transgene memory responses. rAd5 is able to trigger activation of multiple TLRs by MyD88-dependent signaling (141); these include positively regulating TLR2, TLR6, and TLR9, and negatively regulating TLR4 (142,143). As described above, TLR innate triggering is very much linked to the development of strong vaccine responses, therefore, rAd exposure stimulates these PRRs to promote increased DC maturation (144). In addition to TLRs, our laboratory has provided evidence to suggest that rAds also signal through RLR pathways, as the knockdown of MAVS adaptor protein results in loss of NFκB signaling and downstream pro-inflammatory cytokine expression (145). Furthermore, rAd5 vectors are activators of the complement system (146), which, in turn, is intimately linked to the development of antibodies against Ad5 (147). 19 Most innate immunity directed towards rAd5 vectors is known to be against its outer, exposed protein components, such as the hexon, penton, and fiber proteins (148). rAd5 nucleic acid products have been historically considered less important in the early, inflammatory response as UV-inactivated rAds are still able to promote production of cytokines, including IL6, IL-10, and RANTES (149,150). That being said, adenoviral DNA has been implicated in triggering innate immune responses as well, only more predominantly through the NALP3 inflammasome pathway (151). Additionally, rAd viral-associated RNAs (VARNAs) have been found to stimulate a later innate immune response upon intracellular expression, specifically, by binding to and activating RIG-I, resulting in the upregulation of type I IFN production and signaling (152,153). Moreover, VARNAs can suppress cellular RNA interference (RNAi) machinery and act as competitive inhibitors of the dsRNA-activated protein kinase PKR, thereby causing NFκB pathway inhibition (154–157). While there have been a multitude of studies that have shown rAd5-induced innate immune responses can help to promote the development of adaptive memory responses (148), it must also be recognized that excessive early immune stimulation can lead to substantial toxicity and clinical side effects. In one infamous case, an 18-year old man enrolled in a gene therapy trial was intravenously (IV) administered high doses of an Ad5[E1-,E4-] vector for the treatment of ornithine transcarbamylase (OTC) deficiency. Within 18 hours after administration, the man developed thrombocytopenia and multi-organ failure, leading to his death four days later (132). This response was found to be induced by a cytokine storm reaction during which his plasma IL6 and IL-10 levels were profoundly elevated (132). Since its occurrence, this misfortune has served as a precaution to those who study the use of any viral vector and how the immune response to them may provoke beneficial or harmful reactions (158). 20 1.4.4 Pre-existing immunity against rAd vaccine vectors Due to their multitude of benefits, rAd5 vectors are consistently ranked among the most utilized forms of viral delivery. These vectors are also intrinsically strong immunostimulants, as was discussed in the previous section, and therefore provide a substantial boost to the immune response towards their transgenes. On the other hand, the immunogenicity of these vectors can swing both ways. Epidemiologically, the majority of the human population has had exposure to naturally-existent adenoviruses within their adolescent years of life, thus carry measurable titers of anti-adenovirus antibodies (159–161). As with innate immunity, strong neutralizing antibodies and anti-Ad specific T cells directed against wild-type and recombinant Ads are more exclusive to external capsid proteins (162–164), while lower strength adaptive responses can be developed against other adenovirus components as well (165,166). These responses are thought to be the main reason for the Merck STEP HIV trial failure, during which the testing of this HIV vaccine was prematurely halted due to the observation that individuals with high anti-Ad5 titers prior to vaccine administration were more likely to contract HIV than those administered a placebo vaccine (167,168). Consequently, this ineffectual attempt has given researchers much hesitation towards future utilization of unmodified, first-generation rAd vectors in populations at high-risk for HIV infection (169). A number of methods to evade pre-existing immunity against Ad vectors have been developed and attempted, including lower viral particle dose regimens (170,171), modification of Ad capsid proteins (163,172), and use of alterative Ad serotypes that do not typically infect humans (e.g. chimpanzee-endogenous Ads) (173,174). While some of these methods may help in avoiding anti-Ad5 immune neutralization, unfortunately, they also appear to blunt the adaptive memory response towards the encoded transgene, which is adversative to the properties of a 21 good vaccine (175–178). Another investigated technique to combat this issue has been the inclusion of additional early Ad gene deletions within the typical Ad5[E1-] vectors. As was mentioned above, these “later-generation” rAds have been developed as a means to reduce responsivity to overly excessive anti-viral immune responses generated by first-generation vectors. We and other research groups have found that the additionally-deleted rAd5 vectors prevent “leaky” expression of Ad5-specific proteins and promote longer durations of transgene expression compared to that which first-generation, Ad5[E1-] vectors induce (179–182). Most importantly, later-generation rAd5 vectors, such as Ad5[E1-,E2b-], are able to produce superior transgene-specific adaptive immunity following vaccination of Ad-immune mice, non-human primates, and humans (181,183–186). These platforms present novel and interesting ways to induce potent adaptive immunity against evasive pathogen-derived and TAA targets. 22 Chapter 2: Decreased vector gene expression from second-generation, E2b-deleted Ad5 vaccines paradoxically intensifies pro-inflammatory immune responses This chapter is the edited version of a research article under review for publication; submitted August 20th, 2014. Authors: Quiroga D, Aldhamen YA, Godbehere S, Harding L, Amalfitano A 23 2.1 Introduction Recombinant vaccine platforms derived from adenoviruses, particularly Ad5, are some of the best studied and characterized of the viral vector-based vaccines. Ad5-based vaccines have several benefits for use in humans, including broad human cell tropism, relative ease of development, and low incidence of adverse side effects (116). Multiple studies have demonstrated the tremendous ability of Ad5 vaccines to create robust, transgene-specific CMI against antigens derived from pathogens such as HIV (185,187–190), malaria (191–193), tuberculosis (194,195), as well as many tumor-associated antigens (181,184,186,196). Although the benefits of these vaccines are numerous, there has been some hesitation towards their practical use due to the high prevalence of pre-existing Ad5 immunity in the human population (160,197,198). Several tactics to evade pre-existing immunity towards Ad5 have been attempted, including alternative delivery methods (199,200), modification of viral surface proteins (163), and use of alternative Ad serotypes (176,193,201), each with varying levels of success. Another proposed strategy to combat this issue is to delete additional genes from the Adbased vectors. Traditional, or “first-generation” recombinant Ads are engineered to lack E1 and E3 gene regions (Ad5[E1-]), which substantially inhibits, but does not completely eliminate, viral replication and protein translation (182). Alternatively, second-generation recombinant Ads have a further deletion of the E2b gene region (Ad5[E1-,E2b-]), which normally encodes for the Ad5 Pol and PTP genes. In studies comparing these two generations of Ads, Ad5[E1-,E2b-] vaccines have been found to promote decreased expression of viral proteins, persist longer in vivo, and induce less viral-associated toxicity following in vivo administration (131,138,181). Furthermore, multiple murine, non-human primate, and human studies have shown that the antiAd5 pre-existing immunity that inhibits Ad5[E1-] efficacy can be circumvented through use of 24 Ad5[E1-,E2b-] platforms, allowing for induction of a robust immune response against the vaccine transgene target (181,183–186). Currently, the rationale for the responsive differences between these Ad5 vectors is not clear. For example, it has been consistently acknowledged that the large majority of immunity developed following Ad5 virus exposure is directed against Ad5 capsid proteins (e.g. hexon) (162,163), which are products of Ad5 late transcription units and, therefore, identical across both Ad5[E1-] and Ad5[E1-,E2b-] vaccines. While it has been reported that some human CD8+ T cells can recognize Ad5 E2b gene-derived epitopes, they are not as immunodominant as many other Ad5 capsid-based epitopes (166,202). It has also been observed that Ad5[E1-] viruses produce “leaky” expression of virus-derived proteins, while E2b-deleted vaccines do not (180,182). Consequently, it has been hypothesized that viral protein production from Ad5[E1-] vaccines accelerate anti-Ad5 memory responses towards the recombinant Ad-infected cells, thereby shortening transgene expression time and decreasing therapeutic efficacy. While this process may contribute partially to the in vivo differences between Ad5[E1-] and Ad5[E1-,E2b-] vaccines during long term infection, there is little evidence to suggest that this is the most influential factor responsible for their differential performances. Another possibility to consider is that the two Ad vector generations may produce divergent inductions of the innate immune response. Previous microarray studies have shown that Ad5[E1-] infection instigates a unique hepatocellular gene expression pattern as compared to fully-deleted Ad5 vaccines, which lack any viral genome-derived gene expression. Interestingly, the differences in immune-related gene expression peaked at six hours post-infection (hpi) and were greatly diminished by 72 hpi, suggesting that vaccine differences may be most profound during early innate stages of viral recognition (203). Acknowledging that additional viral gene 25 deletions within recombinant Ad5 vaccines can change the initial immune reactions towards them, we wished to compare the early immune responses that Ad5[E1-] and Ad5[E1-,E2b-] infection could induce. In this investigation, we initially examined previously gathered microarray data to compare murine liver gene expression following Ad5[E1-] or Ad5[E1-,E2b-] administration to delineate functional transcriptome differences. Additionally, we measured early amounts of cytokine secretion from multiple, independent samples of human peripheral blood mononuclear cells (hPBMCs) following Ad5[E1-] or Ad5[E1-,E2b-] infection and tested if such responses were independent of previous anti-Ad5 immunity. Finally, hPBMC expression of viral and innate immune genes was quantified following Ad5[E1-] or Ad[E1-,E2b-] treatment. Our studies revealed there to be significant differences between the innate responses to Ad5[E1-] and Ad5[E1-,E2b-] vector treatment both within in vivo murine and in vitro human studies. Furthermore, these findings suggest that the improved therapeutic value of Ad5[E1-,E2b-] over first-generation rAd5 vectors may be, in part, due to the alternative biological manners by which they modulate the innate immune response. 26 2.2 Methods 2.2.1 Microarray analysis Liver RNA microarray data from mice injected with Ad5[E1-] or Ad5[E1,E2b-] vectors was previously collected and preliminary findings were published (141). This data was submitted to NCBI’s Gene Omnibus Express as accession number GSE3739. Data was normalized using Genespring 6.2 software using Lowness normalization, with microarray values below 0.01 set to 0.01, as previously described (141). Novel analysis of this data was conducted using Expression Analysis Systematic Explorer (EASE v2.0) using developer’s standard protocols (204) with all gene ontology (GO) groups listed representing significant differences (p<0.05 with one-way ANOVA using a Benjamini-Hochberg false discover rate multiple test correction (BH FDR MTC)) between Ad5[E1-] and Ad5[E1-,E2b-] injected mice at a set time-point post-injection. Microarray data was also analyzed using QIAGEN’s Ingenuity Pathway Analysis software (IPA®, QIAGEN Redwood City, www.qiagen.com/ingenuity). Both EASE and IPA analysis used a variant of a one-tailed Fisher’s exact test, also called an “EASE score”, at a threshold of p<0.05 to denote significance between groups. 2.2.2 Recombinant adenovirus vector construction Transgene-lacking recombinant Ad5 vectors Ad5[E1-] (205) and Ad5[E1-,E2b-] (182) were built and propagated as previously described, using a pAdEasy-based system (182,205,206). Viral particle titers were determined by spectrophotometry. Ad5 gene region deletions were confirmed by PCR, as previously described (182). HD-Ad5 (FDAdhGAA) (207) was kindly provided by Dr. Philip Ng (Baylor College of Medicine). All viral vectors were tested and validated as having <0.01 EU (per injection dose) bacterial endotoxin contamination, as previously described (208). 27 2.2.3 Cell isolation, culture, and adenovirus infection Sera and hPBMCs were obtained from de-identified blood samples (Stanford Blood Bank, Palo Alto, CA) using Ficoll-Paque PLUS gradient (GE Healthcare Life Sciences, Piscataway, NJ), as described in manufacturer’s protocol. In experiments where sera was not required, de-identified buffy coats were used as a source of hPBMCs (Stanford Blood Bank) and were isolated in the same manner. hPBMCs were plated at 5x106 cells/well in 6-well plates and incubated in RPMI media supplemented with 2% fetal bovine serum (FBS), 1x PSF, 5 ng/mL IL12, and 10 ng/mL IL-18. HEK293 cells were seeded at 3.5x105 cells/well in 6-well plates in DMEM media supplemented with 10% FBS and 1x PSF. Ad5 vectors were administered in a volume of 50μL/well phosphate buffer saline (PBS), with mock treatment being PBS alone. 2.2.4 ELISA analysis Anti-Ad5 ELISAs were performed on sera as previously described (147). Briefly, 5x108 vp/well Ad5[E1-] was plated in a high-binding 96-well flat-bottom plate and incubated overnight at 4⁰C. Plates were rinsed with washing buffer (PBS containing 0.05% Tween) and incubated with blocking buffer (PBS containing 3% bovine serum albumin) for an hour at room temperature. Sera was diluted in PBS (1:200), plated, and incubated at room temperature for an hour. Wells were rinsed with washing buffer, coated with a horseradish peroxidase (HRP) conjugated rabbit anti-mouse antibody (Bio-Rad, Hercules, CA), and developed with tetramethylbenzidine (TMB) (Sigma-Aldrich, St. Louis, MO). The development reaction was halted by adding 1N phosphoric acid to wells. Plates were analyzed using an automatic microplate reader at 450nm absorbance. Cell media was collected and used to perform human IL-1β, IL-6, and TNFα ELISAs, as per manufacturer’s protocol (BD Biosciences, San Diego, CA). The protocol was basically 28 identical to that of the anti-Ad5 ELISA, with the exception that the plates were initially coated with an anti-IL-1β, anti-IL-6, or anti-TNFα capture antibody overnight and that the HRPconjugated antibodies used were also specific to the cytokine being measured. Sample concentrations of each cytokine were determined by normalization to a recombinant human IL1β, IL-6, or TNFα dose curve developed concurrently with experimental samples. 2.2.5 Cell staining and flow cytometry Fluorescent intracellular staining (ICS) was performed as described previously (189). Briefly, 1.5x106 hPBMCs were stimulated with 1x1010 vp UV-inactivated Ad5[E1-] for 30 minutes at 37⁰C. Cells were stained with Alexa Fluor488-CD3, Alexa Fluor700-CD8a, and CD16/32 Fc-block antibodies (BD Biosciences), fixed using 2% formaldehyde (Polysciences, Warrington, PA), permeabilized with 0.2% saponin (Sigma-Aldrich), and stained with PE-IFNγ (BD Biosciences) and violet fluorescent reactive dye for dead cell exclusion (Invitrogen, Carlsbad, CA). Fluorescence-activated cell sorting (FACS) buffer (PBS containing 2% FBS and sodium azide) was used to wash and resuspend cells between and after each treatment. Cell analysis was performed using an LSRII flow cytometer (BD Biosciences) with FlowJo software (Tree Star, San Carlos, CA). 2.2.6 RNA extraction, cDNA synthesis, and quantitative RT-PCR Cellular RNA and DNA was extracted using TRIzol reagent (Life Technologies, Grand Island, NY), as detailed in manufacturer’s manual. cDNA was synthesized by reverse transcription using SuperScript III First-Strand Synthesis SuperMix (Life Technologies) as per manufacturer’s instructions. Quantitative RT-PCR was performed and analyzed using SYBR Green PCR Mastermix (Life Technologies) on an ABI 7900HT Fast Real-Time PCR System (Life Technologies) as previously described (189,209). Primers used to detect expression of viral 29 and human genes were designed using the online PrimerBank database (http://pga.mgh.harvard.edu/primerbank) (Table 1). To measure relative gene expression, GAPDH and the comparative Ct method were used for all samples. Induction of gene expression was calculated as the relative change from the level of mock-treated cell transcripts to the level of rAd-treated cell transcripts. 30 Table 1 Quantitative RT-PCR primers for rAd5[E1-,E2b-] studies Primers were designed using PrimerBank online software. Primer sequences are written 5’ to 3’. 31 2.2.7 Statistical analysis The associations between cytokine production and anti-Ad5 IgG titers or anti-Ad5 CD8+ T cells were tested with the Pearson correlation coefficient. Statistical significance of all other data was determined using one-way ANOVAs with Newman-Keuls post-hoc corrections, unless otherwise stated (e.g. microarray data). Graphs in this chapter are presented as mean ± standard error. Statistical analyses were performed using GraphPad Prism (GraphPad Software, La Jolla, CA). 32 2.3 Results 2.3.1 Hepatocyte microarray analysis following first- or second-generation rAd infection Our group has previously published microarray studies examining murine hepatocyte rAd5 infection-induced transcriptome modifications, specifically, those observed when utilizing different generations of rAd vectors, analyzing several post-infection RNA collection timepoints, and utilizing C57BL/6-derived mouse strains (141). Briefly, adult C57BL/6 mice were IV injected through the retro-orbital sinus with 200μL PBS as a mock treatment, or a 200μL PBS suspension containing 1.5x1011 vp Ad5[E1-] or Ad5[E1-,E2b-]. Upon initial analysis, our results found the greatest divergence in rAd infection-triggered hepatocyte gene expression during early post-infection time-points (141). We observed that in vivo murine Ad5[E1-] infection significantly induced multiple gene groups to a greater extent than mock-treated animals at the 6 hpi time-point, including groups for innate immune signaling, RNA regulatory mechanisms, and apoptosis-related pathways. As compared to mock-treated animals, these same gene groups that Ad5[E1-] injection altered the expression of were also the same groups significantly induced by Ad5[E1-,E2b-]-treated mice (141). To determine which biological functions and/or diseases may be triggered by murine Ad5[E1-] or Ad5[E1-,E2b-] infection at a later time point, we analyzed the 3 days post-infection (dpi) time-point hepatocyte microarray data using IPA software, with mock-treated mouse data serving as the comparative baseline. Analysis revealed there to be many processes significantly activated by rAd5 infection, including many cellular biological functions (e.g. growth, proliferation, death, survival), immune cell trafficking, inflammation, humoral immunity, and cell-to-cell signaling (Figure 2). Also, the significant processes identified were very similar between Ad5[E1-]- and Ad5[E1-,E2b-]-injected mice, with there being only three out of fifteen 33 groups that Ad5[E1-], but not Ad5[E1,E2b-], administration triggered significantly over mock treatment (lymphoid tissue structure and development, hematopoiesis, and cell signaling). It should also be noted that the groups most significantly associated with Ad5[E1-] infection over mock treatment were the same as when Ad5[E1-,E2b-] and mock gene groups were compared. 34 Figure 2 Functional analysis of murine hepatocytes following in vivo Ad5[E1-] and Ad5[E1-,E2b-] infection 35 Figure 2 (cont’d) 36 Figure 2 (cont’d) 37 Figure 2 (cont’d) Previously gathered microarray data comparing mock-infected, Ad5[E1-]-infected, and Ad5[E1-,E2b-]-infected murine hepatocyte RNA was used for further analysis (GSE3739) (141). Biological functions induced by rAd5 infection as compared to mock infection were determined using Ingenuity Pathway Analysis software, with murine gene groups supplied by the included Knowledge Base database. Functional categories significantly upregulated by rAd5 treatment are listed on the y-axis. Significantly upregulated gene groups were determined by a variant righttailed Fischer’s exact test, as displayed on the x-axis as the negative log of the resulting p-value. The cutoff for significance is displayed as the dashed vertical line (p<0.05). Functional categories are listed most to least significant, according to Ad5[E1-] values. 38 While we did observe gene expression differences between Ad5[E1-] and Ad5[E1-,E2b-] treated samples, they remained ~96% homologous at 6 hpi and only became more homologous at the 3 dpi and 3 weeks post-injection (wpi) time points. To evaluate if these gene expression differences were not simply arbitrary findings, we normalized and analyzed this microarray data (GSE3739) by using EASE software to determine if these gene expression differences between Ad5[E1-] and Ad5[E1-,E2b-] treated mice were significantly clustered in recognized ontological categories. We found there to be many significant gene ontological differences between Ad5[E1]- and Ad5[E1-,E2b-]-infected mouse hepatocytes at 6 hpi and 3 dpi (22 and 20 groups, respectively), but no GO group differences at 3 wpi (Table 2). 39 Table 2 Ontological categorization of genes differentially and significantly expressed following in vivo Ad5[E1-] or Ad5[E1-,E2b-] infection 40 Table 2 (cont’d) 41 Table 2 (cont’d) 42 Table 2 (cont’d) 43 Table 2 (cont’d) 44 Table 2 (cont’d) 45 Table 2 (cont’d) 46 Table 2 (cont’d) Previously gathered microarray data comparing Ad5[E1-]- and Ad5[E1-,E2b-]-infected murine hepatocyte RNA was used for further analysis (GSE3739) (141). Significantly different GO groups were determined by a p<0.05 using one-way ANOVAs with a BH FDR MTC utilizing EASE software. 47 At 6 hpi, the GO groups most upregulated in murine hepatocytes following Ad5[E1-] infection, as compared to Ad5[E1-,E2b-] infection, were the immune and defense response groups. Interestingly, several of these individual genes are also important regulators of IFNinducible responses (e.g. CXCL10, IRF2, GBP2). Additionally, Ad5[E1-] infection significantly upregulated genes in GO groups involved with cell growth/maintenance, guanine-based nucleotide/nucleoside interactions, exonuclease activity, and catalase activity. Upregulated GO groups in the 6 hpi Ad5[E1-,E2b-]-treated samples as compared to the Ad5[E1-,E2b-]-treated samples include protein modification and adenine-based nucleotide/nucleoside interaction. Interestingly, all GO groups significantly promoted at 6 hpi by Ad5[E1-] infection over Ad5[E1-,E2b-] were not found to be significantly upregulated at 3 dpi; with the majority of upregulated GO groups conversely being related to cell death/apoptosis. GO groups upregulated by Ad5[E1-,E2b-] infection over Ad5[E1-] infection were more conserved, with nucleotide binding, phosphorylation, ATP binding, and adenyl nucleotide binding all being significantly greater at both 6 hpi and 3 dpi time points. Moreover, GO groups for phosphate/phosphorus metabolism, phosphorylation, and protein metabolism were also significantly upregulated by Ad5[E1-,E2b-] over Ad5[E1-] treated murine hepatocytes. These multifactorial analyses signify that differences between Ad5[E1-] and Ad5[E1-,E2b-] infection-induced gene expression appear to occur early after infection, with Ad5[E1-] infection over-expressed GO groups being substantially different from 6 hpi to 3 dpi. 2.3.2 Ad5[E1-,E2b-] vectors induce an increased pro-inflammatory cytokine response upon hPBMC infection Next, we wished to examine the early functional differences between Ad5[E1-] and Ad5[E1-,E2b-] infection. As some of the microarray findings pointed towards altered induction 48 of innate immune functions by cells exposed to the two different rAd5 vectors, we decided to test if exposure to Ad5[E1-] or Ad5[E1-,E2b-] vectors may induce altered levels of early innate immune cytokine secretion in humans. To allow for a more individualized and clinically relevant model, we obtained and utilized 17 unique, de-identified human blood samples. Upon receiving the samples, hPBMCs were isolated via Ficoll-Paque gradient, plated, and mock infected or infected with Ad5[E1-] or Ad5[E1-,E2b-] vectors. At 72 hpi, media was collected and used for ELISA measurement of IL-1β, TNFα, and IL-6. These particular pro-inflammatory cytokines were selected for quantification due to their significant production by hPBMCs following Ad infection and association with improved antigen-specific T cell expansion (150,210). Interestingly, mean statistical comparison of all samples revealed that Ad5[E1-,E2b-] exposure induced a greater amount of IL-1β (p<0.05) and TNFα (p<0.001) secretion than Ad5[E1-] treatment of the hPBMCs (Figure 3A). Furthermore, Ad5[E1-] treatment of hPBMCs did not appear to induce significantly higher levels of IL-1β or TNFα than mock-treated cells. Individually, the majority of hPBMC samples produced significantly higher IL-1β and TNFα levels when treated with Ad5[E1-,E2b-] as compared to Ad5[E1-], and none of the samples produced greater cytokine induction with Ad5[E1-] over Ad5[E1-,E2b-] infection (Table 3). There was no observed difference between Ad5[E1-]- and Ad5[E1-,E2b-]-induced secretion of IL-6 from hPBMCs, however, both triggered higher levels of IL-6 production than mock infection. Overall, these findings suggest that Ad5[E1-,E2b] infection drives a stronger proinflammatory response than Ad5[E1-], though both viruses can provoke significant hPBMC cytokine induction. 49 Figure 3 Ad5[E1-,E2b-] infection induces increased hPBMC pro-inflammatory cytokine secretion 50 Figure 3 (cont’d) hPBMCs and sera were isolated from 17 human blood samples by Ficoll-Paque PLUS gradient separation. Cells were plated at 5x106 cells/well in 6-well plates with RPMI media supplemented with 2% FBS, 1x PSF, 5 ng/mL IL-12, and 10 ng/mL IL-18 and either mock infected, or infected with 10,000 vp/cell Ad5[E1-] or Ad5[E1-,E2b-]. (A) At 72 hpi, cell media was collected and used to perform human IL-1β, TNFα, and IL-6 ELISAs to measure the amount of hPBMC cytokine secretion. Bars denote mean ± standard error. *, ** denote significant differences between that group and mock-treated samples (p<0.05, p<0.01 respectively). #, ### denote significant differences between Ad treatments (p<0.05, p<0.001 respectively). (B) Sera samples were used to measure pre-existing anti-Ad5 IgG by ELISA. The obtained (450nm) OD values were plotted against respective cytokine concentration values produced when hPBMCs from the same blood sample were infected with Ad5[E1-]. Correlation analysis between variables was performed by Pearson correlation coefficient test. (C) 1.5x106 hPBMCs per sample were stimulated with UV-inactivated Ad5[E1-]. Total percentages of IFNγ-producing CD3+, CD8+ hPBMCs were determined by ICS and flow cytometry analysis. The obtained percentages were plotted against respective cytokine concentrations produced when hPBMCs from the same blood sample were infected with Ad5[E1-]. Correlation analysis between variables was performed by Pearson correlation coefficient test. 51 Table 3 Individual hPBMC sample cytokine inductions 52 Table 3 (cont’d) 53 Table 3 (cont’d) 54 Table 3 (cont’d) hPBMCs were isolated from 17 human blood samples by Ficoll-Paque PLUS gradient separation. Cells were plated at 5x106 cells/well in 6-well plates in RPMI media supplemented with 2% FBS, 1x PSF, 5 ng/mL IL-12, and 10 ng/mL IL-18 and either mock infected, or infected with 10,000 vp/cell Ad5[E1-] or Ad5[E1-,E2b-]. For each sample, each treatment was repeated in triplicates. At 72 hpi, cell media was collected and used to perform human IL-1β, TNFα, and IL-6 ELISAs to measure the amount of hPBMC cytokine secretion. Values represent mean ± standard error. *, **, *** denote significant differences between that group and mock-treated samples (p<0.05, p<0.01, p<0.001 respectively). †, ††, ††† denote significant differences between Ad treatments (p<0.05, p<0.01, p<0.001 respectively). 55 2.3.3 Ad5-induced pro-inflammatory cytokine responses are not correlated to pre-existing Ad5 immunity in human blood samples One of the biggest apprehensions to Ad5 vaccine use has been the concern over naturallyoccurring, pre-existing Ad5 immunity in the general human population. Previous studies have shown that the presence of anti-Ad5 IgG antibodies or CMI can prevent development of transgene immunity following vaccination with Ad5[E1-] transgene-expressing vectors (165), yet this effect does not seem to be problematic during the use of Ad5[E1-,E2b-] vaccines (181,184–186,190,196). To investigate if Ad5[E1-] or Ad5[E1-,E2b-] infection-induced cytokine expression correlated to pre-existing anti-Ad5 IgG antibody levels, serum was collected from the previously specified human blood samples prior to hPBMC isolation. Anti-Ad5 IgG ELISA was performed and obtained values were plotted against the same subject’s hPBMC IL-1β, TNFα, and IL-6 secreted cytokine levels following Ad5[E1-] or Ad5[E1-,E2b-] infection. Correlation analysis revealed no association between pre-existing anti-Ad5 IgG levels and IL-1β, TNFα, or IL-6 secretion amounts, either with Ad5[E1-] (Figure 3B) or Ad5[E1-,E2b-] (Figure 4A) treatment. 56 Figure 4 Quantity of Ad5[E1-,E2b-]-induced hPBMC pro-inflammatory cytokine secretion is not correlated to pre- existing anti-Ad5 immunity 57 Figure 4 (cont’d) hPBMCs and sera were isolated from 17 human blood samples by Ficoll-Paque PLUS gradient separation. (A) Sera samples were used to measure pre-existing anti-Ad5 IgG by ELISA. The obtained (450nm) OD values were plotted against respective cytokine concentration values produced when hPBMCs from the same blood sample were infected with Ad5[E1-,E2b-], as seen in Figure 3. Correlation analysis between variables was performed by Pearson correlation coefficient test. (B) 1.5x106 hPBMCs per sample were stimulated with UV-inactivated Ad5[E1-]. Total percentages of IFNγ-producing CD3+, CD8+ hPBMCs were determined by ICS and flow cytometry analysis. The obtained percentages were plotted against respective cytokine concentrations produced when hPBMCs from the same blood sample were infected with Ad5[E1-,E2b-], as seen in Figure 3. Correlation analysis between variables was performed by Pearson correlation coefficient test. 58 Additionally, we wished to examine if pre-existing anti-Ad5 CMI could be correlated to Ad5[E1-] or Ad5[E1-,E2b-]-induced cytokine production, as CD8+ T lymphocytes have also been found to be an integral part of anti-Ad5 immunity (165). To do this, hPBMCs were stimulated with UV-inactivated Ad5[E1-] and analyzed by flow cytometry for quantification of IFNγ-expressing CD3+, CD8+ T cells, as previously described (189). CD3+, CD8+ cells which expressed intracellular IFNγ were considered anti-Ad5-specific CD8+ T cells and the percent of these cells were plotted against the samples’ respective IL-1β, TNFα, and IL-6 cytokine concentration amounts. Similar to the humoral response, correlation analysis revealed no association between percent of anti-Ad5-specific CD8+ T cells and any of the measured cytokine secretion amounts induced by Ad5[E1-] (Figure 3C) or Ad5[E1-,E2b-] (Figure 4B) infection. Several studies have shown that in instances where there are high levels of anti-Ad5 preexisting immunity, Ad5[E1-,E2b-] vaccines are able to induce much greater transgene-specific immunity than Ad5[E1-] vaccines (181,190,196). Consequently, we considered the possibility that the difference in Ad5[E1-,E2b-] over Ad5[E1-] vector pro-inflammatory cytokine induction may be correlated with the level of pre-existing anti-Ad5 antibodies and/or T cells. To examine these relationships, correlation analyses were completed as mentioned above, with the exception that the obtained values for anti-Ad5 immunity were plotted against a ratio of each subject’s Ad5[E1-,E2b-] to Ad5[E1-]-induced cytokine secretion. These analyses revealed that amounts of neither pre-existing anti-Ad5 IgG antibodies (Figure 5A) nor anti-Ad5 CD8+ T cells (Figure 5B) were correlated to cytokine expression ratios. Overall, these results suggest that while proinflammatory cytokine induction differs between Ad5[E1-] and Ad5[E1-,E2b-] vectors, this response seems to occur independent of pre-existing anti-Ad5 humoral or cellular immunity. 59 Figure 5 Ratio of Ad5[E1-,E2b-] to Ad5[E1-] induced cytokine production is not correlated to pre-existing anti-Ad5 immunity 60 Figure 5 (cont’d) Using data described in Figure 3, ratios of cytokine secretion from Ad5[E1-,E2b-]-treated to Ad5[E1-]-treated hPBMCs derived from the same blood sample were calculated. Previously obtained pre-existing anti-Ad5 IgG OD values (A) and percentages of Ad5-specific CD8+ T cells (B) were plotted against these respective ratios produced from the same blood sample. Correlation analysis between variables was performed by Pearson correlation coefficient test. 61 2.3.4 Ad5[E1-,E2b-] infection produces dampened Ad gene expression Next, we wished to determine if lack of E2b gene function was the specific factor causing Ad5-induced cytokine production differences between Ad5[E1-] and Ad5[E1-,E2b-] vectors. To do this, we infected HEK293 cells with equivalent doses of Ad5[E1-], Ad5[E1-,E2b-], or HDAd5 (207) (a helper-dependent adenovirus; gutted for all viral genes) vectors. HEK293 cells constitutively produce the Ad5 E1 protein (129), allowing for Ad5[E1-], but not Ad5[E1,E2b-] or HD-Ad5, vectors to be replication-competent. Subsequently, we found that HEK293 cells expressed significantly greater amounts of E4, VAI, VAII, and hexon RNA when infected with Ad5[E1-] as compared to Ad5[E1-,E2b-] (p<0.001) (Figure 6A). These differences were so robust that they statistically rendered Ad5[E1-,E2b-] viral gene transcript levels not different from mock or HD-Ad5 infected cells, with the exception of early gene E4 (p<0.001). Neither mock nor HD-Ad5 showed any notable Ad gene expression within HEK293 cells. 62 Figure 6 Ad5[E1-,E2b-] infection produces dampened Ad gene expression 63 Figure 6 (cont’d) 64 Figure 6 (cont’d) (A) HEK293 cells were plated at 3.5x106 cells/well in 6-well plates with DMEM media supplemented with 10% FBS and 1x PSF and either mock infected, or infected with 200 vp/cell Ad5[E1-], Ad5[E1-,E2b-], or HD-Ad5. RNA was collected at 24 hpi for RT-PCR quantification of Ad gene expression. (B) hPBMCs were plated at 5x106 cells/well in 6-well plates with RPMI media supplemented with 2% FBS, 1x PSF, 5 ng/mL IL-12, and 10 ng/mL IL-18 and either mock infected or infected with 10,000 vp/cell Ad5[E1-], Ad5[E1-,E2b-], or HD-Ad5. RNA was collected at 6 hpi for RT-PCR quantification of Ad gene expression. Bars denote mean ± standard error. *, **, *** denote significant differences between that group and mock-treated samples (p<0.05, p<0.01, p<0.001, respectively). #, ##, ### denote significant differences between Ad treatments (p<0.05, p<0.01, p<0.001, respectively). 65 To determine if these viral gene expression differences would also be seen following infection of hPBMCs (which do not express the Ad5 E1 gene), these cells were plated and mock infected or infected with equivalent doses of Ad5[E1-], Ad5[E1-,E2b-], or HD-Ad. RNA collected from these samples revealed that hPBMC Ad5[E1-,E2b-] infection induced less E4 (p<0.001), hexon (p<0.01), VAI (p<0.01), and VAII (p<0.05) gene transcription than Ad5[E1-] treatment (Figure 6B). Again, the amount of gene expression induced by Ad5[E1-] vectors statistically skewed the analysis, resulting in expression levels from Ad5[E1-,E2b-]-treated hPBMCs to appear no different than the control-treated hPBMCs; with the exception of VAI (p<0.05 for mock, p<0.01 for HD-Ad5). Mock and HD-Ad5 infected hPBMCs yielded no expression of any Ad-specific genes in comparison to Ad5[E1-] and Ad5[E1-,E2b-]-infected samples. These dissimilarities in Ad5 gene expression are in agreement with past studies which show that Ad5[E1-,E2b-] infection produces substantially less “leaky” Ad5 protein expression than Ad5[E1-] vectors (180,182), suggesting that regulation of this expression likely occurs at the transcriptional level. To further verify that these Ad gene expression variations were not due to unequal amounts of Ad5 viral particles being introduced and/or entering the hPBMCs, sample DNA and RNA were collected simultaneously and relative viral genome quantification was conducted by quantitative PCR measurement of Ad5 hexon gene DNA, as described previously (209). Comparison of viral DNA from Ad5[E1-] and Ad5[E1-,E2b-]-treated cells revealed no differences in the amount of viral genomes present in either group (Figure 7). 66 Figure 7 Ad5[E1-] and Ad5[E1-,E2b-] viral genome quantification following human PBMC infection hPBMCs were plated at 5x106 cells/well in 6-well plates with RPMI media supplemented with 2% FBS, 1x PSF, 5 ng/mL IL-12, and 10 ng/mL IL-18 and either mock infected or infected with 10,000 vp/cell Ad5[E1-] or Ad5[E1-,E2b-]. At 6 hpi, DNA was collected simultaneously and from the same samples as RNA using TRIzol gradient separation. DNA was used to detect the presence of Ad5 genomes using quantitative RT-PCR with Ad5 hexon primers. 67 2.3.5 Ad5[E1-] and Ad5[E1-,E2b-] vectors induce distinct innate immune gene expression profiles To examine the induction of human immunity-related genes by different Ad5 vector platforms, hPBMCs were either mock infected, or infected with Ad5[E1-], Ad5[E1-,E2b-], or HD-Ad5 vectors at equivalent doses. RNA was isolated from infected hPBMC samples and used to measure expression of early innate immune response genes. Interestingly, exposure to the Ad5[E1-,E2b-] vector induced significantly more IFIT1 and IL-12β gene expression than the Ad5[E1-] vector (p<0.001, p<0.01 respectively) (Figure 8). IFIT1 and IL-12β expression triggered by Ad5[E1-,E2b-] infection was also significantly greater than that triggered by HDAd5 (p<0.05, p<0.001 respectively) or mock (p<0.001) infection. Conversely, Ad5[E1-] did not induce significantly higher amounts of IFIT1 or IL-12β gene expression than the other treatment groups; actually inducing statistically lower quantities of IFIT1 than the HD-Ad5 control vector (p<0.01). Infection of hPBMCs with Ad5[E1-,E2b-] also promoted greater RIG-I gene expression than noted in mock-infected cells (p<0.05), although no other differences between treatment groups were found to be statistically significant. Alternatively, cells infected with Ad5[E1-] and HD-Ad5 did produce significantly higher levels of IFNβ gene expression than mock (p<0.01, p<0.05 respectively) and, while not statistically notable, averaged greater amounts of IFNβ expression than cells treated with the Ad5[E1-,E2b-] vector. 68 Figure 8 Ad5 vectors promote distinct innate immune gene expression patterns dependent on vector generation hPBMCs were plated at 5x106 cells/well in 6-well plates with RPMI media supplemented with 2% FBS, 1x PSF, 5 ng/mL IL-12, and 10 ng/mL IL-18 and either mock infected or infected with 10,000 vp/cell Ad5[E1-], Ad5[E1-,E2b-], or HD-Ad5. RNA was collected at 6 hpi for RTPCR quantification of Ad gene expression. Bars denote mean ± standard error. *, **, *** denote significant differences between that group and mock-treated samples (p<0.05, p<0.01, p<0.001 respectively). #, ##, ### denote significant differences between Ad treatments (p<0.05, p<0.01, p<0.001 respectively). 69 2.4 Discussion Recombinant adenovirus vectors are promising, highly characterized platforms on which to create vaccines. First-generation rAd5 vectors possess the ability to promote strong immunologic responses against their expressed transgenes, but these same properties also trigger host anti-viral responses which can lead to increased toxicity, limited persistence, and decreased efficacy in the face of pre-existing anti-Ad immunity (131,132,138,167,180). Alternatively, second-generation rAds, possessing additional viral gene deletions, have been found to negate these detrimental effects. Back-to-back comparisons of traditional Ad5[E1-] vectors to E2bdeleted, Ad5[E1,E2b-] vectors have shown that the latter produces virtually no hepatotoxicity after IV administration (138), persists and expresses transgenes for longer durations than Ad5[E1-] vectors (131,180), and remains efficacious in hosts with substantial pre-existing antiAd5 immunity (181,183–186). Up until know, it has been largely assumed that the rationale for these rAd5 differences is that Ad5[E1-,E2b-] vectors do not produce substantial amounts of viral proteins upon infection, thereby more adeptly evading host anti-viral responses (180,182). While this may serve as a contributor towards these divergent responses, multiple rAd5 studies have shown that anti-Ad5 immunity alone is not predictive of a rAd5-based vaccine’s efficacy (186,194,211). To investigate what other biological mechanisms may be causing the split in Ad5[E1-] versus Ad5[E1-,E2b-] responses, we analyzed our group’s previously gathered and published hepatocyte RNA microarray data of mice that were injected with either Ad5[E1-] or Ad5[E1,E2b-] vectors. Our previously published manuscript revealed that the most hepatocyte transcriptome changes between mock and rAd5-treated mice occurred at the earliest time-point studied (6 hpi), so at that time we focused on the ontological differences that occurred between 70 these specific treatment groups (141). In our current studies, we analyzed the gene ontology groups that were expressed at a higher level at 3 dpi by Ad5[E1-] and Ad5[E1-,E2b-] injection as compared to mock. We found that several gene groups were expressed differentially, and that essentially all of the groups significantly overexpressed by Ad5[E1-]-injected mice were also overexpressed by mice administered Ad5[E1-,E2b-], when compared to mock-treated animals. However, when comparing the rAd5 groups head-to-head, we were still able to identify notable ontological differences, particularly at 6 hpi. While Ad5[E1-] administration seemed to promote a more global response at the 6 hpi time-point, these effects appeared to burn out quickly, so that the majority of overexpressed groups were absent at 3 dpi, as compared to Ad5[E1-,E2b-] injection. Instead, 3 dpi hepatocyte samples revealed that Ad5[E1-] treatment caused overexpression of gene groups focused on cell death/apoptosis responses, as compared to Ad5[E1-,E2b-] vectors. These results are experimentally consistent with past findings that Ad5[E1-] vectors induce more liver toxicity and inflammation than Ad5[E1-,E2b-] vectors (138). In contrast, several GO groups significantly upregulated by Ad5[E1-,E2b-] over Ad5[E1-] infection were conserved at both 6 hpi and 3 dpi time-points, which may be due to Ad5[E1-,E2b] vectors’ ability to persist beyond that which is seen with Ad5[E1-] vectors (131,180). While these microarray findings confirmed that there are substantial gene expression differences that Ad5[E1-] and Ad5[E1-,E2b-] promote, we understood these findings alone were not enough to confirm how these vaccine platforms may perform divergently in clinical models. Thus, the experimental model we chose to functionally compare Ad5[E1-] and Ad5[E1-,E2b-] effects were human PBMCs. As we chose to harvest the hPBMCs from multiple human blood samples, this not only improved the statistical power of our findings, but also allowed for a more diverse population to be tested. 71 Gene expression analysis of hPBMCs following rAd5 treatment lead us to find that Ad5[E1-,E2b-] infection promoted induction of genes that are strongly tied with Th1 and NFκB pathway activation, while Ad5[E1-] triggered higher amounts of type I interferon signaling, which may be a reason for Ad5[E1-,E2b-]’s superior vaccine efficacy and longer viral persistence in vivo. For example, it is known that NFκB activation and IL-12 expression enhances DC activation, promotes CD4+ T cell Th1 differentiation, and boosts IgG antibody production; all beneficial immune actions during the development of a substantial vaccination response (144,83). Our hPBMC cytokine secretion results also fall in-line with this trend; whereas, the pro-inflammatory cytokines more potently induced by Ad5[E1-,E2b-] than Ad[E1-] vectors, IL-1β and TNFα, are also important instigators of antigen-dependent CD4+ T cell clonal expansion (210). Alternatively, it has been found that increased IFNβ production following viral vaccine administration can inhibit mucosal phagocyte IL-12 responses, thereby decreasing efficiency of vaccine-induced phagocyte killing and class II MHC expression (195). These results further corroborate with previous microarray comparisons between Ad5[E1-] and HDAd5, showing there to be many significant interferon-mediated gene expression differences between these vectors at early post-infection times (203). The mechanism by which Ad5[E1-,E2b-] vectors produce this beneficial immune response requires further investigation, although the observed decreases in vector-derived gene expression when compared to Ad5[E1-] vectors may be a pivotal factor. For example, two of these differentially expressed viral genes were VAI and VAII, the Ad5 VARNA genes. Ad VARNAs have been characterized as potent dsRNA activators of type I interferon expression (152), yet also act as NFκB signaling inhibitors (212,213). Moreover, Ad VARNAs interact with RNAi-related enzymes to inhibit endogenous RNAi activities while simultaneously promoting 72 VARNA-derived siRNA and miRNA synthesis (154,214,215); production of which have been shown to impact many aspects of cell biology, including anti-viral immune function (216,217). Products from other rAd5 genes have also been shown to directly affect human cellular signaling pathways; for example, hexon protein has been described as interacting with dynein-dependent transport and the coagulation cascade (218,219). The decreased expression of Ad gene products by Ad5[E1-,E2b-] vaccines may minimize these types of effects, some of which (e.g. NFκB inhibition) are known to be directly detrimental to the development of transgene-specific immunity (83). The other postulation that can be reached from these results is that Ad5[E1-] vaccines may be elaborating Ad-derived immune evasion strategies. This is further supported by the fact that Ad5[E1-] hPBMC infection not only induced a smaller pro-inflammatory, Th1 response than Ad5[E1-,E2b-] infection, but also produced significantly less IFIT1 expression than a fullydeleted Ad5-based vector which lacks any Ad gene expression (Figure 8). In summary, Ad5[E1-]- and Ad5[E1-,E2b-]-based vaccines were found to each induce unique, early innate responses along with differences in viral-derived gene expression during hPBMC infection. These findings help to shed light on the immune balancing act that recombinant virus vectors face when being used for vaccination; triggering innate immunity can promote transgene-specific memory responses and vaccine efficacy, yet it can also activate antiviral responses and predispose them to pre-existing anti-vector immune responses that diminish vaccine efficacy. While adaptive immune responses caused by previous Ad5 exposure may still contribute to the lower efficacy of Ad5[E1-] as compared to Ad5[E1-,E2b-] vaccines, these results suggest that this is unlikely to be the only factor and that early adenoviral biological activities may also play a role in these differences. Better understanding of the immune 73 responses towards innovative recombinant Ad vectors, such as the Ad5[E1-,E2b-] platform, may allow for further optimization of safe and effective virus-based vaccines. Acknowledgements A.A. was supported by the Michigan State University Osteopathic Heritage Foundation. 74 Chapter 3: Utilization of a recombinant adenovirus vector expressing an Eimeria antigenderived TLR agonist as a vaccine adjuvant. Portions of this chapter are derived from the previously published research articles: Appledorn DM, Aldhamen YA, DePas W, Seregin SS, Liu C-JJ, Schuldt N, Quach D, Quiroga D, Godbehere S, Zlatkin I, Kim S, McCormick JJ, Amalfitano A. A new adenovirus based vaccine vector expressing an Eimeria tenella derived TLR agonist improves cellular immune responses to an antigenic target. PloS One. 2010; 5(3):e9579. Quiroga D – contributed towards experiments using ELISpot assays for measurement of splenocyte transgene response in mice injected with recombinant adenovirus vectors Seregin SS, Aldhamen YA, Appledorn DM, Aylsworth CF, Godbehere S, Liu C-JJ, Quiroga D, Amalfitano A. TRIF is a critical negative regulator of TLR agonist mediated activation of dendritic cells in vivo. PloS One. 2011; 6(7):e22064. Quiroga D – contributed towards experiments using ICS analysis of DC and NK cells treated with rEA 75 3.1 Introduction Vaccination protocols often call for not only an antigenic component, but also for an adjuvant to promote long-term immunity and memory towards the specific antigen. In turn, substances that are agonists of the TLR system have been connected to the development of many consequential adaptive immune responses (220,221). When the appropriate PAMP binds to a TLR, it triggers downstream activation of NFκB and other signaling pathways, which in turn promotes the upregulation of many pro-inflammatory genes and increase of surface MHC and co-stimulatory molecule expression (23). Clinical trials have been conducted using a variety of TLR agonists as adjuvant agents (222), but finding a compound which is both potent and safe has proven to be a difficult task. One promising TLR agonist candidate to use as an adjuvant immunostimulant is the 19 kDa protein, recombinant Eimeria antigen (rEA) (223). rEA was originally isolated following examination of the bovine-derived gut protozoa, Eimeria tenella; an organism found to be critically involved in the creation of a tumor-free, Th1-skewed environment (223). Introduction of this protein into animal models has resulted in improved survival rates following Toxoplasma gondii, flaviviruses, and phleboviruses challenges (224–227). rEA has also been tested for use as a therapeutic anti-tumor treatment, with studies confirming rEA’s ability to stimulate DC IL-12 release as well as lower circulating levels of the TAA, CA-125, in patients that were carrying CA-125 expressing tumors (223,228). Importantly, administration of sizable rEA protein concentrations resulted in no discernable animal or human patient side effects (223,228). As rEA shares a high amount of homology with T. gondii profilin-like protein and induces similar DC activation patterns, it has been assumed that rEA also induces its proinflammatory, anti-microbial, and anti-tumor effects at least in part through the mouse TLR11 76 pathway (229). However, this cannot be the route by which humans recognize rEA as the human TLR11 gene contains stop codons, thereby keeping it from encoding a functional protein (230). In this chapter, we wished to ascertain if rEA protein could be signaling through human TLR or TLR-related pathways by determining if its immunogenic effects are dependent upon TLR adaptor proteins, MyD88 and TRIF, and the immune cells it is exposed to (231). As previously characterized TLR agonists have been shown to be potent adjuvants (93,220,222), we also evaluated rEA’s ability to improve CMI and CTL functions against targeted antigens when it is being expressed by a rAd vector (187). Together, these findings assist in the understanding and characterization of a novel TLR ligand and potential vaccine adjuvant. 77 3.2 Methods 3.2.1 Animal care and procedures All animal procedures were conducted under the approval of Michigan State University’s Institutional Animal Care and Use Committee. Eight-week old male Balb/c and wild-type (WT) C57BL/6 mice were purchased from The Jackson Laboratory (Bar Harbor, ME). MyD88-KO and TRIF-KO mice were generously provided by Dr. Shizuo Akira. MyD88/TRIF-DKO mice were bred at Michigan State University. All knock-out mice were bred on a C57BL/6 background. For intraperitoneal (IP) injections, mice were administered 100μL PBS (pH 7.4) dilutions of purified rEA protein (NCBI Accession: AY745810), as previously described (187). For IV injections, mice were administered 200μL PBS dilutions of rAd5 virus or rEA protein via retro-orbital sinus. For intramuscular (IM) injections, mice were administered 20μL PBS dilutions of rAd5 virus or 50 μL PBS dilutions of rEA protein within the tibialis anterior muscle of the right hindlimb. 3.2.2 Recombinant adenovirus vector construction The rEA gene open reading frame was inserted directly upstream of a GFP cassette within a CMV-driven pShuttle cassette, creating the pAdTrack-rEA shuttle. pAdTrack-rEA was linearized and recombined within BJ 5183 cells with the pAdEasy1 Ad5 vector, as previously described (205). The resulting pAd5-rEA plasmid was linearized with PacI restriction enzyme, transfected into HEK293 cells for vector replication, and purified using cesium gradient separation, as previously described (205,206). During these processes, direct sequencing and restriction enzyme mapped were used to verify correct plasmid sequences. rAd5-GFP was built and propagated using the pAdEasy1 Ad5 system, as previously described (205). rAd5-GFP was used as a control vector as it was previously confirmed to have 78 no significant impact on measurable adaptive immune responses to co-administered antigens (191). All vectors underwent direct sequencing to verify correct transgene insertion and were found to be replication-incompetent via E1 gene region deletion by PCR, as previously described (232). Viral particle titers were determined by spectrophotometry. All viral vectors were tested and validated as having <0.01 EU (per injection dose) bacterial endotoxin contamination, as previously described (208). To verify that in vitro transduction efficiencies of rAd5-GFP and rAd5-rEA vectors were equivalent, HEK293 cells were infected with 1x1010 vp/mL and percentages of GFP-positive cells were compared (and found to be similar at 24 hpi) by FACS analysis using an LSRII flow cytometer and FlowJo software (Tree Star) (187). 3.2.3 Splenocyte processing Splenocytes were obtained and processed as previously described (142). Briefly, mouse spleens were homogenized through physical disruption by passage through a 40 μm sieve. Splenocytes were treated with red blood cell (RBC) lysis buffer, then washed and resuspended in RPMI media supplemented with 10% FBS and 1x PSF (penicillin, streptomycin, and fungizone). 3.2.4 Cell staining and flow cytometry Prepared splenocytes were measured for DC and NK cell activation and maturation markers, as previously described (172). For DC staining, 1x106 splenocytes per sample were treated with 4 mg/mL PECy7-Cd11c, APC-CD80, Pacific Blue-CD86, and PerCpCy5.5CD3/CD19/NK1.1 (dump channel) (BD Biosciences) on ice for 30 minutes, then washed with FACS buffer and sorted using an LSRII flow cytometer and FlowJo software. 79 For intracellular NK cell staining, 2x106 splenocytes per sample were treated with 1 μg/mL Brefeldin A (suspended in DMEM supplemented with 10% FBS and 1x PSF) for 4 hours at 37⁰C. Splenocytes were washed twice with FACS buffer, incubated with rat anti-mouse CD16/CD32 Fcγ block (BD Biosciences) for 15 minutes, and stained with 8 mg/mL APC-CD3 and PE-Cy7-NK1.1 (BD Biosciences) for 30 minutes on ice. Following fluorescent antibody staining, splenocytes were washed with FACS buffer, fixed on ice with a 20 minute treatment of 2% formaldehyde (Polysciences), permeabilized at room temperature for 20 minutes with 0.5% saponin (Sigma-Aldrich), and intracellularly-stained on ice for two hours with 8 mg/mL FITCIFNγ. Finally, cells were washed with FACS buffer and sorted using an LSRII flow cytometer and FlowJo software. 3.2.5 Cytokine and chemokine analysis Plasma cytokine and chemokine concentrations were measured via 23-plex mouse multiplex-based assay as indicated by the manufacturer’s instructions (Bio-Rad) by use of a Luminex 100 machine (Luminex, Austin, TX), as previously described (232). 3.2.6 Enzyme-linked immunosorbent spot (ELISpot) assay analysis ELISpot assay analyses were performed using a Ready-Set-GO! ELISpot mouse IFNγ kit (eBioscience, San Diego, CA), as previously described (233). In brief, splenocytes (5x106 cells/well) from individual mice were incubated on capture antibody pre-treated plates with 4 mg/mL GFP (DTLVNRIEL), LacZ (DAPIYTNV), or Gag (AMQMLKETI) peptides (Genscript, Piscataway, NJ). Staining of plates was performed as described in manufacturer’s protocol. Plate well photography and spot-forming cell (SFC) quantification was performed by an automated ELISpot reader (Cellular Technology, Cleveland, OH). 80 3.2.7 Statistical analysis Flow cytometry data was analyzed using two-tailed homoscedastic Student’s t-tests to compare differences between mock- and rEA-treated mice in each individual knockout group, as well as between WT mice and other knockout groups following mock or rEA treatment. All other experiments where statistical significance was determined was analyzed using a one-way ANOVA with a Newman-Keuls post-hoc correction. Graphs in this chapter are presented as mean ± standard error. Statistical analyses were performed using GraphPad Prism (GraphPad Software). 81 3.3 Results 3.3.1 Regulation of rEA-induced dendritic cell responses by TLR adaptor proteins In characterizing the type of innate immune response that rEA stimulation would induce, we wished to test if the presence of MyD88 and/or TRIF adaptor proteins would affect its immunostimulatory activities. To do this, we IP injected C57BL/6-background WT, MyD88-KO, TRIF-KO, and MyD88/TRIF-DKO mice with mock (PBS alone) or 100ng purified rEA protein. Previous studies have shown that rEA can induce DC activation by 6 hpi (187), therefore, mice were sacrificed for splenocytes at this time for flow cytometry analysis of DCs (CD3-, CD11c+, CD19- splenocytes), which were analyzed for expression of DC activation markers CD80 and CD86. In WT and TRIF-KO mice, we found that the percentage of DCs expressing CD80 and CD86 were higher following rEA treatment than with mock (p<0.01) (Figure 9, 10). Interestingly, rEA-treated TRIF-KO mice had a greater percent of activated DCs than rEAtreated WT mice (p<0.001). Alternatively, rEA treatment of MyD88-KO and MyD88/TRIFDKO mice did not result in a significant increase in the amount of CD80- or CD86-expressing DCs as compared to mock-treated animals. 82 Figure 9 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF 83 Figure 9 (cont’d) C57BL/6 WT (n = 3-4), MyD88-KO (n = 3), TRIF-KO (n = 3-4), and MyD88/TRIFDKO (n = 4) mice were mock injected or injected with 100 ng rEA protein. At 6 hpi, splenocytes were harvested, stained with fluorescent antibodies, and sorted by flow cytometry. Independent groups of WT mice were used for each knockout genotype comparison. Bars denote mean ± standard error. ** denotes a significant difference from mock-treated animals (p<0.01). 84 Figure 10 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF (histogram plots) Representative histograms of rEA-treated DC samples detailed in Figure 9. 85 The concentration of CD80 and CD86 surface expression per cell also followed this trend, with WT and TRIF-KO mice both possessing a significant increase in the proportion of activated DCs upon rEA administration as opposed to mock treated (Figure 11). rEA-treated TRIF-KO mice also produced significantly greater amounts of CD86 expression per cell than rEA-treated WT mice (p<0.001). Again, MyD88-KO and MyD88/TRIF-DKO mice did not have this induction of DC maturation following rEA treatment, with rEA-treated MyD88-KO mice producing significantly lower amounts of CD80 and CD86 expression per cell than rEA-treated WT mice (p<0.01). 86 Figure 11 rEA-mediated dendritic cell activation is differentially regulated by MyD88 and TRIF (MFI) 87 Figure 11 (cont’d) C57BL/6 WT (n = 3-4), MyD88-KO (n = 3), TRIF-KO (n = 3-4), and MyD88/TRIFDKO (n = 4) mice were mock injected or injected with 100 ng rEA protein. At 6 hpi, splenocytes were harvested, stained with fluorescent antibodies, and sorted by flow cytometry. Independent groups of WT mice were used for each knockout genotype comparison. Mean fluorescent intensity (MFI) values are displayed and are representative of the indicated marker concentration per cell. Bars denote mean ± standard error. ** denotes a significant difference from mocktreated animals (p<0.01 respectively). 88 3.3.2 Regulation of rEA-induced natural killer cell responses by TLR adaptor proteins Finding such striking differences in rEA-stimulated mouse DC activation, depending upon the presence of certain TLR adaptor proteins, prompted further investigation into the function of other immune cells such as NK cells. To study rEA protein effects, C57BL/6background WT, MyD88-KO, TRIF-KO, and MyD88/TRIF-DKO mice were IP injected with mock (PBS alone) or 100ng purified rEA protein. We have previously shown that rEA can promote significant NK cell activation at 6 hpi (187), therefore, mice were sacrificed at the this time-point for splenocyte-based flow cytometry analysis of NK cells (CD3-, NK1.1+ splenocytes) which were activated to express intracellular IFNγ. Similar to our DC results, WT and TRIF-KO mice produced a higher percentage of NK cells expressing IFNγ after rEA treatment than with mock treatment (p<0.01) (Figure 12, 13), whereas MyD88-KO and MyD88/TRIF-DKO mice showed no rEA-triggered increases in NK cell activation. 89 Figure 12 rEA-mediated NK cell activation is differentially regulated by MyD88 and TRIF 90 Figure 12 (cont’d) C57BL/6 WT (n = 3-4), MyD88-KO (n = 3), TRIF-KO (n = 3-4), and MyD88/TRIFDKO (n = 4) mice were mock injected or injected with 100 ng rEA protein. At 6 hpi, splenocytes were harvested, stained with fluorescent antibodies, and sorted by flow cytometry. Independent groups of WT mice were used for each knockout genotype comparison. MFI values displayed are representative of the concentration of the indicated marker per cell. Bars denote mean ± standard error. ** denotes a significant difference from mock-treated animals (p<0.01). 91 Figure 13 rEA-mediated NK cell activation is differentially regulated by MyD88 and TRIF (histogram plots) Representative histograms of rEA-treated DC samples detailed in Figure 12. 92 In WT and TRIF-KO mice, NK cell IFNγ MFI values were also increased following rEA treatment, but were left unchanged in MyD88-KO and MyD88/TRIF-DKO mice (p<0.01) (Figure 12). It should also be noted that the amount by which NK cell activation was increased following rEA protein treatment was elevated in WT mice (from 1% to 20-30% IFNγ-expressing NK cells), but even higher levels were noted in TRIF-KO mice following rEA administration (from 1% to >40% IFNγ-expressing NK cells). Conversely, MyD88 and MyD88/TRIF-DKO mice did not demonstrate an increase in the IFNγ MFI values of NK cells following rEA exposures. 3.3.3 Murine cytokine response to intramuscular or intraperitoneal rEA protein injections Since its original synthesis, rEA’s adjuvant properties have been studied in mice via IP injection, in part to be synonymous with tumor injection sites but also to take advantage of the robust immune responses that can be triggered through this route (187,223). However, if rEA is to be used for vaccine applications, IM administration would serve as a more convenient and consistent inoculation route. To measure if the innate immune responses triggered by IP-injected rEA protein differs from IM-injected rEA protein, C57BL/6 mice were injected with IP or IM with a total of 100 ng rEA protein. In previous IP studies, rEA injections were delivered in a 100 μL volume. Unfortunately, 100 μL injections could not be physically or ethically IM injected into a single, unilateral site on one of the animal’s tibialis anterior muscles, therefore, we split this volume into two 50 μL injections that were administered bilaterally. Mice were sacrificed for serum collection at 6 hpi and analyzed for serum cytokine and chemokine concentrations by multiplex assay. Of the 23 cytokines and chemokines measured by this assay, all but one (IL-10) were expressed at similar levels, independent of the rEA protein administration route (Figure 93 14). In general, we found that rEA injection, by either route, induced a broad spectrum of cytokine and chemokine production, including IL-2, IL-4, IL-10, IL-17, eotaxin, IFN-γ, RANTES, and TNFα, consistent with previous in vivo rEA findings (187,223). 94 Figure 14 rEA protein promotes cytokine and chemokine induction mainly independent of injection route 95 Figure 14 (cont’d) 96 Figure 14 (cont’d) C57BL/6 mice (n = 2) were mock injected, or injected with 100 ng rEA protein IM or IP. At 6 hpi, serum was collected and measured for cytokine and chemokine concentrations by multiplex assay. *, ** denotes a significant difference from mock-treated animals (p<0.05, p<0.01, respectively). # denotes a significant difference between IM- and IP-route injected animals (p<0.05). 97 3.3.4 An rEA-expressing recombinant Ad5 vector enhances co-expressed transgenespecific immunity After verifying rEA protein’s immunostimulatory characteristics, we wished to study how administration of rEA would affect the development of vaccine-based immunity. To do this, we wished to constitutively express rEA by utilization of an rEA-expressing rAd5 vector (rAd5rEA) (Figure 15). In addition to expressing the rEA gene, we built rAd5-rEA to also express green fluorescent protein (GFP) as a way to measure how it may induce immunity to a coexpressed transgene. This also allowed us to directly compare rAd5-rEA’s immunogenic effects to rAd5-GFP; a useful control vector which has been found to have little effect on the promotion of long-term memory responses when introduced alongside antigens of interest (191). 98 Figure 15 rAd5-rEA construction The open reading frame of the human rEA gene was inserted into a CMV expression cassette which also contained an upstream GFP expression cassette, resulting in the creation of pAdTrack-rEA. The plasmid was then linearized by PmeI restriction enzyme processing and recombined with pAdEasyI Ad5 (an Ad5 genomic backbone lacking E1 and E3 genes) inside of BJ 5183 cells. The resulting pAd5-rEA was linearized by PacI restriction enzyme processing and transfected into HEK293 cells, where the vector was able to replicate to large titers. rAd5rEA purification was conducted using cesium gradient separation and directly sequenced to confirm transgene rEA’s presence. This figure is an amended version of our previously published version, courtesy of Dr. Schuldt (191). 99 We studied the adjuvant effects of rAd5-rEA by IV (Figure 16A) or IM (Figure 16B) injecting C57BL/6 mice with mock, 1x1010 vp rAd5-GFP, or 1x1010 vp rAd5-rEA. Mice were sacrificed at 7 dpi for collection of splenocytes to use for ELISpot detection of GFP-specific CMI responses. We found that both rAd5-GFP and rAd5-rEA treated mice were able to induce a significant response against GFP (p<0.01), while also lacking a response towards the nonspecific LacZ peptide; showing that mouse CMI following vaccination was specific to the introduced transgene. Most importantly, the quantity of GFP-directed immunity was significantly greater in mice administered rAd5-rEA than rAd5-GFP (p<0.01), showing that rEA expression promotes transgene immunity. This difference was approximately four-fold higher in IV-injected animals and two-fold higher in IM-injected animals, however, the total quantities of GFPresponsive splenocytes in IV and IM rAd5-rEA treated mice were found to be mainly equivalent. 100 Figure 16 Improved cell-mediated immunity towards co-expressed transgene following rAd5-rEA vaccination C57BL/6 mice (n = 2-3) were IV (A) or IM (B) injected with 1x1010 vp rAd5-GFP or rAd5-rEA and were sacrificed for splenocyte collection at seven (A) or fourteen (B) days postinjection and examined by IFN-γ ELISpot to count peptide-specific SPCs. Bars denote mean ± standard error. ## denotes a significant difference from naïve animals (p<0.01). ** denotes a significant difference between rAd5-GFP and rAd5-rEA treated animals (p<0.01) Naïve rAd5-GFP rAd5-rEA Naïve rAd5-GFP rAd5-rEA 101 3.3.5 Addition of rAd5-rEA improves CMI against a co-injected rAd-expressed transgene For an adjuvant to be useful, it must be able to promote a vigorous response against a pathogenic antigen. To investigate the adjuvant properties rAd5-rEA can contribute for increased pathogen-specific peptide recognition, we injected Balb/c mice with 1x106 or 1x108 total vp of an equivalent combination of a rAd vector expressing the HIV-specific Gag gene (rAd5-Gag) and either rAd5-GFP or rAd5-rEA. Splenocytes were collected at 14 dpi, stimulated with one of two HIV Gag epitopes (#304769 or AMQMLKETI), and analyzed by IFN-γ ELISpot. We found that the 1x106 vp dose treatment of rAd5-Gag + rAd5-rEA induced significantly greater amounts of Gag-specific immunity towards both epitopes over 1x106 vp of rAd5-Gag + rAd5-GFP (p<0.001) (Figure 17). Also, CMI against the AMQMLKETI epitope was significantly higher in animals treated with 1x108 vp rAd5-Gag + rAd5-rEA than with 1x108 vp rAd5-Gag + rAd5-GFP (p<0.05). 102 Figure 17 rAd5-rEA induces heightened HIV Gag-specific CMI responses Balb/c mice (n = 3) were IM injected with 1x106 or 1x108 total vp of an equivalent combination of rAd5-Gag and rAd5-GFP or rAd5-Gag and rAd5-rEA. At 14 hpi, splenocytes were harvested and examined by IFN-γ ELISpot to measure Gag-specific immunity against two Gag-derived epitopes (#304769 and AMQMLKETI). Bars denote mean ± standard error. *, *** denote a significant difference between treatment groups (p<0.05, p<0.001, respectively). rAd5-Gag + rAd5-GFP rAd5-Gag + rAd5-rEA rAd5-Gag + rAd5-GFP rAd5-Gag + rAd5-rEA 103 3.4 Discussion Multiple past studies have shown how TLR stimulation can serve as an important factor in the development of long-term adaptive immune responses (86,220,234). With this in mind, we chose to investigate the properties of a novel TLR agonist, rEA. Previous investigations have validated that rEA is both safe to administer as well as adept at inducing innate immune and antitumor effects in humans (223,228), however, the exact pathways by which rEA exerts these actions is unclear. Therefore, we chose to test how mice that lacked TLR adaptor protein expression (MyD88 and/or TRIF) would respond to rEA protein stimulation. We found that both DCs and NK cells from WT and TRIF-KO mice were strongly activated following rEA treatment and that mice lacking TRIF sometimes experienced even greater levels of rEA-dependent activation than WT animals. On the other hand, rEA-induced activation of DC and NK cells was completely ablated in MyD88-KO and MyD88/TRIF-DKO mice. These findings indicate that rEA requires MyD88 to trigger activation of important innate immune components, while TRIF acts as a negative regulator of this response. Other experiments completed by our group also revealed that monocytes, NKT, T, and B cells were activated by MyD88 and inhibited by TRIF in the same manner (231). These studies also included measurement of cytokines and chemokines following rEA treatment, which showed that the pro-inflammatory response induced in WT mice (expression of IL-2, IL-6, IL12, G-CSF, and IFNγ) was even more pronounced in TRIF-KO mice and was diminished in MyD88-KO and MyD88/TRIF-DKO mice (231), further confirming rEA’s mechanism of immune activation. These systematic findings are important in validating the hypothesis that rEA is a TLR agonist; this conjecture was based on rEA’s high level of protein sequence homology with the TLR11 agonist Toxoplasma gondii profilin (224,225,229). As with rEA, profilin-TLR11 104 signaling and promotion of DC IL-12 production has been identified as MyD88 dependent (229). Past studies have also shown that the transition of TLR signaling to strong adaptive responses requires the presence of MyD88 (235), whereas TRIF has yet to be described in this fashion. It is additionally notable that the removal of TRIF allows for a more immunogenic effect following rEA administration. In future studies, inhibition of TRIF may be a useful tool in triggering innate immunity by other PAMPs as well. So to transition rEA into the clinical vaccine delivery field, it would be helpful to prove that rEA can be effective by multiple routes, including intramuscularly. In general, we found that administration of rEA protein by IP or IM route did not change its potency or pattern of cytokine signaling. With this in mind, we built an rEA- and GFP-expressing rAd vector (rAd5-rEA) to see if we could develop a way to deliver rEA more constitutively and immunogenically than rEA protein alone. Our group’s previous studies explored the differential cytokine responses that rEA protein, rAd5-GFP, rAd5-GFP with rEA protein, and rAd5-rEA treatments created upon in vivo injection. We found that rAd5-rEA injection promotes pro-inflammatory cytokine production at significantly greater levels than any of the other aforementioned treatments (187). Additionally, these studies found that DC, NK, and NKT cells are also more readily activated by rAd5-rEA than rAd5-GFP vector injection (187). To test if rAd5-rEA immunostimulatory effects could translate into potent adjuvant activities, we measured CMI towards rAd5-rEA’s co-expressed transgene, GFP, and a transgene expressed by a co-injected rAd5 vector, HIV Gag. In both instances, rAd5-rEA increased the amount of transgene-specific immunity over rAd5-GFP control vector treatment. Such boosting of Gag-directed immunity is of particular interest as studies of HIV long-term progenitors (patients that retain healthy immune function and T cell counts even without anti-HIV therapies) 105 have shown them to consistently possess strong anti-Gag CTL responses (236). Analysis of the failed rAd5 Merck HIV STEP clinical trial revealed that vaccine recipients who had strong inductions of TLR-specific signaling and Th1-dominant gene expression pathways were also more likely to mount substantial anti-HIV CTL responses (237). Additionally, it should also be noted that these studies were conducted in two different strains of mice, C57BL/6 and Balb/c. It has been established that C57BL/6 mice have an intrinsically Th1-skewed immune responses while Balb/c mice favor Th2-skewed immune processes (238,239); consequently demonstrating that we can develop sizeable transgene memory responses in both immunologically diverse in vivo mouse models. These findings also support the possibility of effective clinical translation with the hope that the inherent genetic differences between individual humans may not impact the efficacy of rEA-based treatments. In summation, these studies identified that rEA’s effects on portions of the innate immune response are MyD88-dependent and TRIF-inhibited. Also, by creating an rEAexpressing rAd5 vector, we showed that administration of rAd5-rEA has the capability to induce large amounts of cytokines and to promote transgene-specific immunity when administered concurrently with a transgene-expressing vector. We believe the next appropriate studies to test rAd5-rEA efficacy would be to further characterize the adaptive immunity that it fosters and examine if this response would allow for a multi-faceted immune induction against a vaccine antigenic target of interest. Acknowledgements 106 We wish to thank the personnel in Michigan State University’s laboratory animal support facilities for their assistance in the humane care of animals utilized in this work. A.A. was supported by the Michigan State University Osteopathic Heritage Foundation. 107 Chapter 4: Strengthened tumor antigen immune recognition by inclusion of a recombinant Eimeria antigen in therapeutic cancer vaccination This chapter is the edited version of a research article under review for publication; originally submitted June 25th, 2014 and currently undergoing revisions. Authors: Quiroga D, Aldhamen YA, Appledorn DM, Godbehere S, Amalfitano A 108 4.1 Introduction Immune responses against cancerous cells are often absent as a result of systemic T cell- mediated tolerance against them and their respective tumor-associated antigens (TAA). To mount a substantial immune response against malignancies, successful immunotherapy trials have shown that a therapeutic vaccine must be able to initiate and propagate a strong, TAAspecific cytotoxic immune response (240). Carcinoembryonic antigen (CEA) has been identified as a particularly promising TAA target due to its ubiquitous overexpression in colorectal and other carcinomas, as well as its relatively high level of immunogenicity (241). Various methods have been employed to use CEA as a vaccine transgene, with several resulting in induction of CEA-targeted CTL responses and anti-CEA antibody production (242–245). Recently, a phase I/II clinical trial in which colorectal cancer patients, who had failed an average of three prior chemotherapeutic regimens, were administered adenovirus vectors expressing an enhanced, modified version of CEA (rAd5-CEA) (186,196). This CEA-expressing adenovirus vector multiplied anti-CEA CMI responses several fold over patient pre-vaccination baseline. In addition, patient survival rates at one year were found to be greater than those previously reported in demographically similar colorectal cancer patient populations (246) and were most improved in the treatment group given the highest dose of rAd5-CEA. As this and many other cancer immunotherapy trials have revealed, there is a substantial positive correlation between the amount of TAA-directed CMI produced and rates of patient survival. It has also been recognized that even immunotherapy-naïve patient tumor microenvironments often contain TAA-presenting cells, yet they do not readily activate a CTL response against the antigen when present (247,248). Therefore, in addition to a TAA 109 vaccination, successful treatment may also require an adjuvant to promote long-term immunity and memory towards the cancer cells. Several adjuvants have been shown to improve both the degree and breadth of antigen specific cellular immune responses, particularly those that trigger TLR- and other PRR-mediated innate immune responses (77,88,220,221,249). Recombinant Eimeria antigen (rEA) is a notable activator of TLR and non-TLR innate immune signaling pathways. rEA was initially identified within the Eimeria tenella protozoan as an inducer of high IL-12 levels in the bovine intestine, an effect that promoted an overall anti-tumorigenic environment (223). Injection of the rEA protein has since been shown to prolong survival of tumor-carrying mice and induce a safe, cytokine-dependent decrease in the CA-125 tumor marker within advanced malignancy patients (223,228). In previous studies, we have created an rEA-expressing rAd vector (rAd5-rEA) and shown that in vivo delivery of this agent can promote a Th1-skewed, pro-inflammatory response greater than rEA protein or a non-specific rAd, as measured by heightened cytokine responses (e.g. IFNγ, TNFα, IL-12(p70)), activation of innate immune cells (e.g. NK, NKT, DC), and greater transgene memory responses against a co-injected HIV derived (HIV-Gag) antigen (187). Moreover, we have found that rEA can directly promote human NK effector cell activation and stimulate human peripheral blood mononuclear cell (PBMC) cytolytic tumor cell killing (250). Based on these findings, we wished to investigate if co-administration of rAd5-CEA and rAd5rEA could further improve anti-CEA immunity. Additionally, we explored the spectrum, quantity, and relationship of T and B cell-facilitated adaptive immune responses that rAd5-rEA introduces to a vaccine regimen targeting a human-relevant TAA. 110 4.2 Methods 4.2.1 Recombinant adenovirus vector construction Recombinant Ad5 vectors rAd5-CEA (196), rAd5-rEA (187), and rAd5-GFP (205) were built and propagated as previously described. rAd5-GFP was used as a control vector as it was previously confirmed to have no significant impact on measurable adaptive immune responses to co-administered antigens (191). The cDNA sequence of human CAP1(6D)-modified CEA was produced and generously supplied from Duke University (251). Vectors underwent recombination and viral propagation as previously described (206). All vectors underwent direct sequencing to verify correct transgene insertion and were found to be replication-incompetent via E1 gene region deletion by PCR, as previously described (232). Viral particle titers were determined by spectrophotometry and SDS-polyacrylamide gel electrophoresis following silver stain or western blotting. 4.2.2 Animal care and procedures All animal procedures were conducted under the approval of Michigan State University’s Institutional Animal Care and Use Committee. Eight-week old male C57BL/6 mice were purchased from The Jackson Laboratory and injected IM into the tibialis anterior of the right hindlimb with 20μL phosphate-buffered saline solution (PBS, pH 7.4) containing a total of 1x108 to 1x1010 vp, including rAd5-CEA and an equivalent vp dose of either rAd5-GFP or rAd5-rEA. Splenocytes and blood plasma were obtained and processed as previously described (142). 4.2.3 ELISpot assay analysis ELISpot assay analyses were performed using Ready-Set-GO! ELISpot mouse IFNγ and IL-2 kits (eBioscience), as previously described (233). In brief, splenocytes (5 x 106 cells/well) from individual mice were incubated on capture antibody pre-treated plates with individual CEA 111 peptides or a CEA peptide pool. Individual CEA peptides were 15 aa in size and covered the CEA498-676 sequence with a CAP1(6D) modification in 10aa overlaps using a PepTrack peptide library (JPT Peptide Technologies, Berlin, Germany). The CEA peptide pool of CAP1(6D)modified CEA sequence peptides was a generous gift from Dr. Michael Morse at Duke Medical Center (186). Staining of plates was performed as described in manufacturer’s protocol. Plate well photography and SFC quantification was performed by an automated ELISpot reader (Cellular Technology). 4.2.4 Cell staining and flow cytometry Fluorescent ICS was performed as previously described (189). In brief, 2 x 106 splenocytes were stained with APC-Cy7-CD3, Alexa Fluor700-CD8a, and CD16/32 Fc-block antibodies, fixed using 2% formaldehyde (Polysciences), permeabilized using 0.2% saponin (Sigma-Aldrich), stained for intracellular cytokines using APC-granzyme B, FITC-IFNγ, PEperforin, PE-Cy7-TNFα (BD Biosciences), and PerCpCy5.5-IL-2 (BioLegend, San Diego, CA), and stained for dead cell exclusion using violet fluorescent reactive dye (Invitrogen). Cells were analyzed with a LSRII flow cytometer (BD Biosciences) using FlowJo software (Tree Star). 4.2.5 In vivo cytotoxic T lymphocyte (CTL) assay In vivo CTL analysis was performed similarly to previously described studies (233). Briefly, mice were vaccinated with 5 x 109 vp rAd5-CEA and 5 x 109 vp of either rAd5-GFP or rAd5-rEA. At 21dpi, syngeneic splenocytes were pulsed with a CEA-specific peptide pool or the irrelevant HIV Gag peptide, AMQ (AMQMLKETI), for 1 hour at 37⁰C. Carboxyfluorescein succinimidyl ester (CFSE) was used to stain CEA-pulsed splenocytes at a 10μM concentration (CFSEHigh) and AMQ-pulsed splenocytes at a 1μM concentration (CFSELow). Immunized and naïve mice were injected in the left retro-orbital sinus with equal amounts of CEA- and AMQ- 112 pulsed splenocytes at a total volume of 8x106 cells. Twenty-four hours following splenocyte infusion, mice were sacrificed for splenocyte collection, washing, and FACS analysis by LSRII flow cytometer. FlowJo software was utilized to quantify amounts of CFSE-stained splenocytes. The percentage of specific CEA-pulsed splenocyte killing was determined using the following equation: % specific killing = 1 – ((% CFSEHigh /% CFSELow)immunized/(% CFSEHigh /% CFSELow)naive). 4.2.6 Anti-CEA and anti-Ad ELISA analysis ELISAs were performed as previously described (147). Briefly, 5 x 108 vp of a null Ad5 vector, 0.2 μg CEA peptide pool, or 0.2 μg individual CEA peptide epitopes were plated per well in a high-binding 96-well flat-bottom plate. Following overnight 4⁰C incubation, plates were rinsed with washing buffer (PBS containing 0.05% Tween) and incubated with blocking buffer (PBS containing 3% bovine serum albumin) for an hour at room temperature. Plasma was plated following 1:1600 dilution (or pooled and diluted 1:10 for measurement of individual CEA epitope IgG binding) in blocking buffer and incubated for an hour at room temperature. Wells were subsequently rinsed with washing buffer and coated with an HRP-conjugated rabbit antimouse antibody (Bio-Rad) diluted 1:4000 in washing buffer. TMB substrate (Sigma-Aldrich) was added to each well to initiate the spectrophotometric reaction, which was stopped with 1N phosphoric acid. Plates were analyzed using an automatic microplate reader at 450nm absorbance. For anti-CEA IgG antibody avidity binding ELISAs, assays were performed as previously described (252). Briefly, 0.2 μg CEA peptide pool was plated per well in a highbinding 96-well flat-bottom plate. Following overnight 4⁰C incubation, plates were rinsed with washing buffer and incubated with blocking buffer for an hour at room temperature. Serum was 113 diluted 1:20, plated in duplicates, and incubated for one hour at room temperature. Wells were subsequently rinsed with washing buffer and incubated with either PBS or PBS containing 8M urea at 37⁰C for 10 minutes. Following another wash, wells were coated with an HRPconjugated rabbit anti-mouse antibody (Bio-Rad) diluted 1:4000 in washing buffer. TMB substrate (Sigma-Aldrich) was added to each well to initiate the spectrophotometric reaction, which was stopped with 1N phosphoric acid. Plates were analyzed using an automatic microplate reader at 450nm absorbance. Individual samples’ anti-CEA IgG binding avidity ratios were calculated as: OD450[urea-treated sample] / OD450[untreated sample]. 4.2.7 Statistical analysis Statistical analysis comparing in vivo CTL CEA-specific killing between Ad treatment groups was performed using a Student’s t-test. The association between percent CEA-specific in vivo CTL killing and anti-CEA IgG titers within individual animals was tested with the Pearson correlation coefficient. All other experiment data where statistical significance was determined were analyzed using a one-way ANOVA with a Newman-Keuls post-hoc correction. Graphs in this chapter are presented as mean ± standard error. Statistical analyses were performed using GraphPad Prism (GraphPad Software). 114 4.3 Results 4.3.1 Increased rAd5-CEA transgene-specific CMI responses induced in the presence of rAd5-rEA We and others have verified that rAd5-CEA vaccination of murine and human subjects induces significant CEA-specific CMI (186,196,233). To examine if the addition of rAd5-rEA to a TAA-expressing vaccine would further promote anti-TAA CMI responses, we treated mice with rAd5-CEA and an equivalent dose of either rAd5-rEA or a GFP-expressing control adenovirus vector (rAd5-GFP). Mice were injected with a total viral dose of either 1x108, 1x109, or 1x1010 vp per mouse. Splenocytes were harvested at 14 days post-injection, processed, and analyzed for anti-CEA specific CMI by IFNγ and IL-2 ELISpot assays. Our data revealed the amount of CEA-specific, IFNγ-secreting splenocytes to be greater in mice injected with the rAd5-CEA + rAd5-rEA combination rather than those given rAd5-CEA + rAd5-GFP (Figure 18A). Specifically, both the 1x109 and 1x1010 vp rAd5-CEA + rAd5-rEA groups yielded statistically more IFNγ-secreting splenocytes than the 1x108 and 1x109 vp rAd5-CEA + rAd5GFP treated groups (p<0.01). IL-2 ELISpot analysis also revealed a significantly greater CEAspecific CMI response when comparing the 1x109 vp dose of rAd5-CEA + rAd5-rEA with the 1x108 (p<0.01) and 1x109 vp (p<0.05) rAd5-CEA + rAd5-GFP treated animals, as well as the 1x108 vp dose of rAd5-CEA + rAd5-rEA treated animals (p<0.05), showing a positive relationship between dose and rAd5-rEA-promoted CEA-specific CMI (Figure 18B). Interestingly, the 1x1010 vp dose showed no difference in the amount of cells secreting IL-2 in response to CEA stimulation. These findings indicate that co-injection of rAd5-rEA with rAd5CEA improves anti-CEA CMI in a mainly dose-dependent manner. 115 Figure 18 Strong CEA-specific cell-mediated immunity produced by rAd5-CEA and rAd5-rEA co-vaccination C57BL/6 mice (n = 2–3) were IM injected with rAd5-CEA and either rAd5-GFP or rAd5-rEA at equal viral particles per vector, totaling 1x108, 1x109, or 1x1010 vp. Splenocytes were collected from vaccinated and naïve mice at fourteen days post-injection and examined by IFN-γ (A) and IL-2 (B) ELISpot to count SPCs. Bars denote mean ± standard error. Solid and dashed lines denote a significant difference between treatment groups (p<0.05, p<0.01 respectively). 116 4.3.2 Improved cytotoxic T cell responses against CEA following high-dose rAd5-CEA + rAd5-rEA co-administration During the process of tumor cell killing, effector memory CD8+ T cells produce cytolytic and inflammatory cytokines in response to specific tumor-derived antigens (253,254). To quantify the amount of CEA-specific cytokine producing CD8+ T cells, splenocytes were collected from naïve or vaccinated mice administered rAd5-CEA combined with an equal dose of either rAd5-rEA or rAd5-GFP. Splenocytes were stimulated ex vivo with either a CEA peptide pool or non-specific HIV-Gag peptide. Splenocytes were analyzed by multicolor flow cytometry after subsequent fluorescent antibody extracellular staining against CD3 and CD8 and ICS against IFNγ, TNFα, IL-2, and perforin. Upon CEA stimulation, 1x1010 vp rAd5-CEA + rAd5-rEA co-injection induced a significantly higher frequency of IFNγ-expressing CD8+ T cells than those given rAd5-CEA and rAd5-GFP at all treatment doses, rAd5-CEA + rAd5-rEA at the 1x108 vp dose, and naïve mice (p<0.05) (Figure 19A). The highest dose of rAd5-CEA + rAd5-rEA also produced a higher frequency of TNFα-producing CD8+ T cells than 1x108 vp and 1x109 vp rAd5-CEA + rAd5GFP injected and naive mice (p<0.05) (Figure 19B). Quantity of intracellular perforin production within CD8+ T cells after CEA stimulation was significantly larger following 1x1010 vp rAd5CEA + rAd5-rEA administration in comparison to all other treatment groups (p<0.05 for 1x108 vp and 1x1010 vp rAd5-CEA + rAd5-GFP; p<0.01 for all other groups) (Figure 19C). Intracellular IL-2 within CD8+ T cells was also measured, but no significant differences were found between any treatment groups (data not shown). Stimulation with the irrelevant HIV-Gag peptides did not produce significantly different amounts of cytokine-producing CD8+ T cells between treatment groups (data not shown). These results suggest that the combination of rAd5- 117 CEA + rAd5-rEA at a 1x1010 vp dose induces substantially more CEA-triggered activation of cytotoxic CD8+ T lymphocytes. 118 Figure 19 High-dose rAd5-CEA and rAd5-rEA administration induces a robust, CEA- specific CD8+ cytotoxic T cell response C57BL/6 mice (n = 3) were IM injected with combination of rAd5-CEA and either rAd5GFP or rAd5-rEA at equal viral particles per vector, totaling 1x108, 1x109, or 1x1010 vp. Splenocytes were collected from vaccinated and naïve mice at fourteen days post-injection, permeabilized, and stained for CD8+ T cell IFNγ (A), TNFα (B), and perforin (C) expression. Bars denote mean ± standard error. *, ** denote significant differences between that group and naïve animals (p<0.05, p<0.01 respectively). Solid and dashed lines denote a significant difference between treatment groups (p<0.05, p<0.01 respectively). 119 4.3.3 Dose-dependent increase in anti-CEA IgG production following rAd5-CEA + rAd5rEA co-administration In addition to TAA-directed CMI, the generation of TAA-specific IgG antibodies is also an important aspect of effective cancer immunotherapy through participation in antibodydependent cell-mediated cytotoxicity (251). Curiously, in our clinical trial using solely rAd5CEA, we did not find notable changes in patient serum anti-CEA IgG following vaccination (186). To measure if the co-administration of rAd5-CEA + rAd5-rEA would induce greater antiCEA IgG titers than rAd5-CEA + rAd5-GFP, mice were co-injected with either combination at a total viral dose of 1x108, 1x109, or 1x1010 vp. Serum was collected at 14 days post-injection and used to perform an anti-CEA IgG ELISA. We observed that at the 1x1010 vp dose, mice treated with rAd5-CEA + rAd5-rEA had significantly greater anti-CEA IgG titers than all other rAd5CEA-treated mice (p<0.05) (Figure 20A). Serum was also used to measure anti-Ad5 IgG titers to evaluate if vaccinated mice developed an antibody response against the vector itself. As seen with anti-CEA IgG, 1x1010 vp rAd5-CEA + rAd5-rEA induced a significantly greater titer of anti-Ad5 IgG than any of the other vaccine-treated mice (p<0.01 for 1x1010 vp rAd5-CEA + rAd5-GFP and 1x109 vp rAd5-CEA + rAd5-rEA; p<0.001 for all other treatments) (Figure 20B), suggesting that rEA overexpression enhances B cell responses to all antigenic targets, including Ad-derived antigens. Moreover, the mice in the 1x109 vp rAd5-CEA + rAd5-rEA had higher amounts of anti-Ad5 IgG than both groups of 1x108 vp dosed mice (p<0.05). This data suggests that high doses of rAd5-CEA + rAd5-rEA successfully induce substantial anti-CEA IgG production, while also producing greater levels of antibodies against the Ad5 vector. 120 Figure 20 Anti-CEA IgG production is potently induced by rAd5-CEA and rAd5-rEA co-injection C57BL/6 mice (n = 2–3) were IM injected with rAd5-CEA and either rAd5-GFP or rAd5-rEA at equal viral particles per vector, totaling 108, 109, or 1010 vp. Serum was collected at fourteen days post-injection, diluted at a 1:1600 ratio, and measured for IgG antibody levels against CEA (A) and Ad5 (B) by ELISA with 450nm OD. Bars denote mean ± standard error. Solid, dashed, and dotted lines denote a significant difference between treatment groups (p<0.05, p<0.01, p<0.001 respectively). 121 4.3.4 rAd5-CEA + rAd5-rEA co-injection enhances breadth of T cell-recognized CEA peptide epitopes While it is important to have a strong memory response against a TAA, worth is also placed on the creation of an epitope-diverse response (255,256). Vaccine-induced recognition of a greater number of immunogenic antigen epitopes can possibly allow for superior protection in cases where some epitopes may be bypassed via tumor survival methods and intrinsic HLA haplotype differences (255,256). We have previously shown that the addition of rAd5-rEA to an HIV antigen-expressing Ad vaccine increased the breadth and quality of response against HIV antigen epitopes (187); therefore, we wished to investigate if rAd5-rEA could also improve the antigen epitope recognition of CEA. To measure the range of CEA immunogenic areas following vaccination, we collected splenocytes isolated from mice injected with 1x1010 total vp of either rAd5-CEA + rAd5-rEA or rAd5-CEA + rAd5-GFP. Splenocytes were stimulated ex vivo with individual, 15 aa-long CEA peptides that encompassed the entirety of one of CEA protein’s repeating domains in N- to Cterminal 10aa overlaps and were numbered subsequently in that order (Table 4). 122 Table 4 CEA peptide epitope sequences and assigned numbering The location of the epitopes’ amino acid sequence within the CEA protein (NP_004354.3) is indicated as counted from N- to C-terminus. Peptide Peptide Location in CEA protein sequence number1 AELPKPSISSNN sequence 498-512 2 PSISSNNSKPVE SKP 503-517 3 NNSKPVEDKD DKD 508-522 4 VEDKDAVAFTC AVAFT 513-527 5 AVAFTCEPEAQ EPEA 518-532 6 CEPEAQNTTYL NTTY 523-537 7 QNTTYLWWVN WWVN 528-542 8 LWWVNGQSLP GQSLP 533-547 9 GQSLPVSPRLQ VSPRL 538-552 10 VSPRLQLSNGN LSNG 543-557 11 QLSNGNRTLTL RTLT 548-562 12 NRTLTLFNVTR FNVT 553-567 13 LFNVTRNDARA NDAR 558-572 14 RNDARAYVCGI YVCG 563-577 15 AYVCGIQNSVS QNSV 568-582 ANRS 123 Table 4 (cont’d) Peptide Peptide Location in CEA protein sequence 16 number IQNSVSANRSD sequence 573-587 17 SANRSDPVTLD PVTL 578-592 18 DPVTLDVLYGP VLYG 583-597 19 DVLYGPDTPIIS DTPI 588-602 20 PDTPIISPPDSSY PPD 593-607 21 ISPPDSSYLSGA LS 598-612 22 SSYLSGADLNL DLN 603-617 23 GADLNLSCHSA SCHS 608-622 24 LSCHSASNPSP SNPS 613-627 25 ASNPSPQYSWR QYSW 618-632 26 PQYSWRINGIP INGI 623-637 27 RINGIPQQHTQ QQHT 628-642 28 PQQHTQVLFIA VLFI 633-647 29 QVLFIAKITPNN KITP 638-652 30 AKITPNNNGTY NGT 643-657 31 NNNGTYACFVS ACFV 648-662 32 YACFVSNLATG NLAT 653-667 33 SNLATGRNNSI RNNS 658-672 34 TGRNNSIVKSIT VKSI 662-676 VSA 124 Using an IFNγ ELISpot to measure CMI, we found several areas of epitope recognition following rAd5-CEA + rAd5-rEA vaccination (Figure 21), as it produced significantly higher levels of IFNγ-producing cells than naïve (p<0.05 for peptides #8, 24, 25; p<0.01 for peptide #31) and rAd5-CEA + rAd5-GFP vaccinated (p<0.05 for peptides #24 and 25; p<0.01 for peptide #8; p<0.001 for peptide #31) mice following splenocyte stimulation with four distinct peptide epitopes. 125 Figure 21 rAd5-CEA and rAd5-rEA co-vaccination induces CMI against multiple unique CEA peptide epitopes 126 Figure 21 (cont’d) C57BL/6 mice (n = 9) were IM injected with 5x109 vp of rAd5-CEA and 5x109 vp of either rAd5-GFP or rAd5-rEA. Splenocytes from vaccinated and naïve (n = 5) mice were collected at fourteen days post-injection and examined by IFN-γ ELISpot where splenocytes were divided and stimulated by a single CEA-derived peptide epitope per well (see Table 4 for sequences). ELISpot analysis was completed by quantification of SFCs. CEA peptides that induced greater amounts of SFCs in rAd5-CEA-treated animals than naïve animals are shown. Bars denote mean ± standard error. *, ** denote significant difference from naïve group (p<0.05, p<0.01 respectively). #, ##, ### denote significant difference from rAd5-CEA + rAd5-GFP treatment group (p<0.05, p<0.01, p<0.001 respectively). 127 Independent of the particular CEA epitope, rAd5-CEA + rAd5-GFP was unable to produce significantly higher amounts of IFNγ-producing cells than the other treatment groups and, for all treatment groups, most CEA epitopes produced SFC counts close to baseline (Figure 22). These results suggest that rAd5-rEA co-administration with rAd5-CEA allows for a diverse subset of CEA epitopes to be recognized by an adaptive CMI response. 128 Figure 22 Addition of rAd5-rEA to an rAd5-CEA vaccine regimen induces CMI against multiple CEA peptide regions C57BL/6 mice (n = 9) were IM injected with 5x109 vp of rAd5-CEA and 5x109 vp of either rAd5-GFP or rAd5-rEA. Splenocytes were collected at fourteen days post-injection and examined by IFN-γ ELISpot where splenocytes were divided and stimulated by a single CEA-derived peptide epitope per well (see Table 4 for sequences). ELISpot analysis was completed by quantification of SFCs. Bars denote mean ± standard error. 129 4.3.5 rAd5-CEA + rAd5-rEA co-injection enhances diversity of anti-CEA IgG antibodyidentified CEA peptide epitopes While researchers have identified several T cell receptor-recognized CEA epitopes (257– 259), there are presently none conclusively verified to be recognized by B cell-based receptors. In addition, previous vaccine prototypes targeting CEA have often been unable to induce measurable amounts of anti-CEA IgG antibodies within patients’ blood serum (186,260). We wished to see if the supplementation of rAd5-rEA to a rAd5-CEA injection would allow for a more varied and detectable repertoire of CEA-binding IgG antibodies. To measure anti-CEA IgG diversity, we collected serum isolated from mice that had been injected with 1x1010 total vp of either rAd5-CEA + rAd5-rEA or rAd5-CEA + rAd5-GFP. Serum was pooled within treatment groups and plated on wells that were coated with the aforementioned individual 15mer CEA peptides (Table 4). In identifying values above the baseline optical density (OD) of 0.2 (measured value in wells without serum plated), ELISA analysis revealed that both rAd5-CEA + rAd5-rEA and rAd5-CEA + rAd5-GFP treatments induced epitope recognition by anti-CEA IgG (Figure 23), while naïve mice did not. However, in observing the number of CEA peptide epitopes recognized by rAd5-CEA-vaccinated mouse serum IgG, there were clearly more unique epitopes bound by immunoglobulins derived from mice given rAd5-rEA as compared to rAd5GFP (28 and 2, respectively). Furthermore, in the instances where rAd5-CEA + rAd5-GFP vaccination did produce a significant amount of epitope-specific anti-CEA IgG, rAd5-CEA + rAd5-rEA vaccination always produced a substantially higher IgG titer against those same epitopes. 130 Figure 23 rAd5-CEA and rAd5-rEA co-injection induces creation of IgG antibodies against a diverse area of CEA peptide regions C57BL/6 mice (n = 7) were IM injected with 5x109 vp rAd5-CEA and 5x109 vp rAd5-GFP or rAd5-rEA. Serum from vaccinated and naïve (n = 5) mice was collected at 21 days post-injection. Serum dilutions (1:10) were pooled by treatment group and plated on wells containing a single CEA-derived peptide epitope per well (see Table 4 for sequences). IgG levels against individual CEA epitopes were measured by ELISA at 450nm OD. The inset graph demonstrates the amount of epitopes in each treatment group that produced an OD value greater than 0.2, as this was the baseline OD measured in wells without serum plated. 131 To test if the addition of rAd5-rEA to the rAd5-CEA vaccine was inducing creation of high affinity anti-CEA IgG antibodies, we next performed a urea-binding affinity ELISA using the same serum samples mentioned above. Assay results revealed that the anti-CEA IgG binding ability was high in both rAd5 treatment groups (Figure 24). Overall, these findings serve as a novel instance where adjuvant treatment alongside a CEA-targeted vaccine increased the breadth of durable anti-CEA B cell responses. 132 Figure 24 High anti-CEA IgG binding affinity following rAd5-CEA and rAd5-rEA co-injection C57BL/6 mice (n = 7) were IM injected with 5x109 vp rAd5-CEA and 5x109 vp rAd5-GFP or rAd5-rEA. Serum from vaccinated and naïve (n = 5) mice was collected at 21 days post-injection. Serum dilutions (1:20) were plated on wells that had been previously incubated with a CEA peptide pool. Following serum incubation, wells were briefly incubated with either PBS (untreated) or an 8M urea PBS solution (urea-treated). (A) IgG levels against individual CEA epitopes were measured by ELISA at 450nm OD. Bars denote mean ± standard error. * denotes significant difference between treatment group and naïve animals (p<0.05). #, ## denote significant difference between rAd5 treatment groups (p<0.05, p<0.01, respectively). (B) Avidity binding ratios were individually calculated and plotted as equaling (OD450[urea-treated sample] / OD450[untreated sample]). 133 4.3.6 Enhanced in vivo CEA-targeted CTL killing following rAd5-CEA + rAd5-rEA coadministration It is important to verify that the components of a multi-faceted, robust memory response can be amplified against a TAA, but these constituents must also be functionally tested to validate their practical anti-tumorigenic actions. To measure the amount of CEA-specific cell killing that could be produced after use of an adjuvant such as rEA, mice vaccinated twenty-one days prior with rAd5-CEA + rAd5-rEA or rAd5-CEA + rAd5-GFP, and Ad-naïve mice, were injected with equal amounts of CEA- or AMQ-pulsed syngeneic splenocytes, which had been labeled with high and low amounts of CFSE, respectively. Twenty-four hours following adoptive transfer of peptide-loaded, CFSE-labeled splenocytes, mice were sacrificed and splenocytes were prepared for measurement of the remaining CEA- (CFSEHigh) and AMQ- (CFSELow) pulsed cells. Flow cytometry analysis revealed that the ratios of CEA:AMQ cells were no different between naïve (Figure 25A) and rAd5-CEA + rAd5-GFP (Figure 25B) co-injected mice. In contrast, splenocytes derived from rAd5-CEA + rAd5-rEA treated animals possessed a notably smaller ratio of CEA:AMQ cells (Figure 25C), indicating more potent in vivo CEA-specific CTL activity. Calculation of the percentage of specific CEA cell killing revealed that all animals treated with rAd5-CEA + rAd5-rEA sustained some measurable level of specific cytotoxic lymphocyte killing, while none of the rAd5-CEA + rAd5-GFP co-injected animals could (p<0.001) (Figure 25D). 134 Figure 25 rAd5-CEA and rAd5-rEA co-injection induces significant CEA-specific in vivo cytolytic T cell killing activity 135 Figure 25 (cont’d) C57BL/6 mice (n = 7) were IM injected with 5x109 vp rAd5-CEA and 5x109 vp rAd5GFP or rAd5-rEA. At 21 days post-injection, syngeneic splenocytes were pulsed with CEAspecific peptides or AMQ and labeled with CFSEHigh (10μM) or CFSELow (1μM), respectively. Immunized and naïve (n = 5) mice were injected with equal amounts of CEA- and AMQ-pulsed splenocytes and, twenty-four hours following, mouse splenocytes were collected, washed, and analyzed via FACS. Population percentages of low-CFSE AMQ-pulsed splenocytes and highCFSE CEA-pulsed splenocytes were compared in naïve (A), rAd5-CEA + rAd5-GFP (B), and rAd5-CEA + rAd5-rEA (C) treatment groups. (D) Percentages of CEA-specific CTL killing was compared between vaccine groups by calculating % specific killing = 1 – ((%CEA/%AMQ)immunized/(%CEA/%AMQ)naive. (E) At the time of sacrifice, serum was collected from these mice, diluted (1:100), and used to perform an anti-CEA ELISA to measure anti-CEA IgG levels. The obtained (450nm) OD values from rAd5-CEA + rAd5-rEA were plotted against respective percentages of CEA-specific CTL killing from each individual animal. The line represents a calculated best-fit linear regression between variables. 136 To investigate if the extent of the anti-CEA T cell and B cell responses may be correlated, serum was collected at the same time as splenocyte collection and used to measure anti-CEA IgG by ELISA. Since rAd5-CEA + rAd5-GFP treatment produced no assessable CEAspecific killing, we only compared the killing percentages of rAd5-CEA + rAd5-rEA treated animals to their respective anti-CEA IgG titers. Linear regression analysis revealed a significant, positive linear correlation (p = 0.0148; r = 0.8523) between the percent of CEA-specific killing and anti-CEA IgG titers (Figure 25E). These findings suggest not only that the formulation of rAd5-CEA vaccine with rAd5-rEA allows for measurable, functional CEA-specific cell killing, but also that these actions correspond directly with levels of IgG antibodies against CEA. 4.4 Discussion Creating a strong enough memory response against an often immune-tolerated TAA has been a persistent issue of the cancer immunotherapy field since its inception (261). Alternatively, researchers have observed that immunogenic treatments that are too aggressive can induce severe systemic side effects, as was seen in the development of an inflammatory colitis following highdose autologous T cell transfer (254). Recombinant adenoviral delivery of tumor-associated antigens has become a popular immunotherapy method, leading to the exploration of many antigenic vaccine targets within numerous human clinical trials. This class of vectors serves as a desirable option due to their ability to efficiently and exceptionally promote T cell memory responses against expressed antigens, which is largely due to rAds’ ability to trigger multidimensional innate immune responses (142,148,220). Recently, we have reported that the use of an adenovirus vector encoding the gene CEA (rAd5-CEA) can be safely administered to human subjects and may prolong survival of patients with advanced, chemotherapy-resistant colorectal cancers by inducing potent anti-CEA-specific T cell immunity (186). In this current 137 study, we wished to investigate if the efficaciousness of this same vaccine could be further improved with the supplementation of a vector expressing the TLR agonist and anti-tumorigenic protein, rEA. The adjuvant properties and human safety profile of rEA have been previously established (187,223,228), therefore, we felt that the characterization of rEA’s immunomodulatory effects on a TAA-based vaccine were warranted. Our first round of experiments included dosage studies so that the optimal therapeutic viral dose could be estimated. The amount of total viral particles used ranged from 1x108 to 1x1010, which has been found to be appropriate for IM murine vaccination protocols in the past (187,189,196). Generally, we found that increasing doses of rAd5-CEA + rAd5-rEA resulted in a greater amount of CEA immune recognition, including stronger CMI (as quantified by the amount of IFNγ- and IL-2-secreting splenocytes), greater amounts of CD8+ cytotoxic T cell activation, and higher anti-CEA IgG titers. Considering these results, all future experiments were performed at the 1x1010 vp dose. While the optimal amount of rAd5-rEA is greater than what we had previously used to improve immunity against the HIV Gag protein (187), it is possible that CEA is less immunogenic than HIV Gag or, that immunization against human CEA, which shares significant homology to mouse CEACAM family proteins (262), may require a larger treatment dose to induce the antigen-directed immune responses towards a protein sequence that is similar one or more of the animal’s own intrinsic non-foreign, “self” proteins. The amount of anti-Ad IgG was also measured among the vaccinated animals, finding that the rAd5-CEA(6D) + rAd5-rEA combination also induced increased amounts of vectordirected antibodies within the animal’s serum. This is a notable finding due to the ongoing concern of pre-existing Ad5 immunity negatively affecting vaccination outcomes, which was most strikingly established following the early termination of the Merck HIV STEP trial (168). 138 While this issue cannot be ignored, it has been shown multiple times since that anti-Ad immunity does not directly correlate with vaccine inefficacy, particularly in cases using modified versions of Ad5 (186,194,211). Our rAd5-CEA(6D) vector is a second-generation, E2b-deleted virus and has been verified, both in murine and human subjects, to induce anti-CEA immunity despite the presence of Ad5 neutralizing antibodies (186,196). Moreover, the amount of anti-CEA CMI and survival time of rAd5-CEA(6D)-vaccinated patients were found to have no association with the serum titers of pre- or post-vaccination anti-Ad5 neutralizing antibodies (186); thus ameliorating some concern over whether or not a paired rAd5-CEA(6D) + rAd5-rEA treatment would be unsuccessful due to previous Ad5 exposure. It has been observed that T cells from both healthy and tumor-burdened patients can respond to a wide range of CEA derived antigens; it is a major therapeutic goal that vaccination against CEA may break down immunologic tolerance to these antigens (253,263). Vaccine trials where a single CEA epitope was targeted have reported having to exclude many patients who did not meet HLA-matching criteria (264), limiting the vaccine’s treatable population. Alternatively, a strong, multiregional repertoire of antigen epitopes could allow for a greater incidence of efficacy across different HLA backgrounds and against multiple variants of CEA-expressing malignancies (255,256). We tested if the adjuvant activities of rAd5-rEA could not only increase the response to individual CEA-derived antigens, but also broaden this type of response to greater numbers of CEA-derived antigens when paired with the rAd5-CEA vaccine. We found that inclusion of an rEA-expressing vector along with a CEA-targeting vaccine induced substantial CMI against several distinct CEA-derived peptides, including four documented epitopes previously shown to be directly recognized by three distinct HLA haplotypes (257– 259). Interestingly, these haplotypes include class I and II HLAs, suggesting that rAd5-CEA + 139 rAd5-rEA co-vaccination may have the potential to activate not only CEA-directed CD8+ T cells, but also effector CD4+ T cells, the numbers of which have been found to be strongly and positively correlated with improved clinical outcomes of advanced malignancy patients (265). However, verification of these notions will require further study in a humanized murine model or human subjects. While the importance of anti-TAA IgG development has been well established (241), many CEA-targeting vaccine prototypes have been found to be unable to induce anti-CEA IgG production above pre-treatment levels (186,260). In our work, while rAd5-CEA vaccination alone was a weak inducer of anti-CEA humoral immunity (186), the combination of rAd5-CEA with rAd5-rEA was able to create a significant amount of circulating serum anti-CEA IgG to several peptide epitopes of CEA. According to the Immune Epitope Database and Analysis Resource (www.iedb.org), there are over 100 confirmed immune epitopes derived from human CEA protein, but as of yet none have been confirmed by B cell response assay, making further investigation into the humoral promotion that rAd5-CEA + rAd5-rEA could provide even more important. Furthermore, rAd5-CEA + rAd5-rEA vaccinated animals also showed markedly higher levels of CEA-specific killing, with the amount of CEA-specific in vivo CTL killing in rAd5CEA + rAd5-rEA vaccinated animals being positively correlated with the amount of anti-CEA IgG detected in their serum. This suggests that induction of humoral and/or cell-mediated adaptive immune responses against TAAs such as CEA are not mutually exclusive, and can be promoted alongside each other using potent adjuvants such as rEA. The molecular mechanisms by which innate immune modulators, such as rEA, assist in the development of a strong immunological memory response are still in the process of being 140 understood (86). Many of the identified characteristics that make an adjuvant effective have been detected in rEA, such as promotion of NK cell activation, increased amounts of DC activation/maturation, induction of pro-inflammatory cytokines (e.g. IL-12(p70), TNFα), and an overall robust Th1-skewing response (187,223,231,250). rEA also promotes relatively unique agonist signaling mechanics in that it uses the TLR adapter protein MyD88 as a positive regulator, and yet another TLR adapter, TRIF, as a negative regulator (231). While there have been previous concerns regarding rEA’s immunogenicity as a profilin-like protein to be limited to mammals that express functional TLR11/12, clinical trials and ex vivo human studies have assuaged much of this concern, revealing there to be many TLR11/12-independent mechanisms by which rEA triggers human immunity (223,228,250). Moreover, it has been recently confirmed that human PBMC recognition of profilin is instead dependent on TLR5 (266); thereby suggesting rEA may be recognized by this PRR as well. We have also shown that rEA can directly activate human NK cells (250), a critical subtype of innate immune cells that perform anti-tumor immune activities (17). Furthermore, rEA has been verified as safe to administer to human patients, even those who may have advanced malignancies with compromised immune systems (223,228); an attribute that is lacking for many other innate immune agonists (267). With the knowledge that rEA is a robust innate stimulator, we explored the scope of adaptive immunity that it could affect. While we have previously shown rAd5-rEA to promote transgene immunity against the HIV Gag protein (187), this is the first time that co-injection of it with a TAA-expressing Ad has been shown to have similar benefits. Furthermore, the importance of gut microbiota (of which rEA is derived) and innate activation has been shown to be essential in the response to several chemotherapy treatments (268,269), suggesting the use of rAd5-CEA + rAd5-rEA alongside these medications may provide a substantially better clinical 141 outcome without making the treatment regimen more physically rigorous for the patient. As phase II trials have already shown significant preliminary success with the use of rAd5-CEA in advanced colorectal cancer patients, it is exceedingly relevant that an adjuvant which would enhance anti-tumor effect to an even greater extent be considered. All in all, rAd5-rEA has the possibility of serving as a conduit to a much larger and more diverse immune response against CEA and a multitude of other TAA-expressing malignancies. Acknowledgments We wish to thank the personnel in Michigan State University’s laboratory animal support facilities for their assistance in the humane care of animals utilized in this work. A.A. was supported by the Michigan State University Osteopathic Heritage Foundation. 142 Chapter 5: Recombinant adenoviruses expressing antigen-trimming protein ERAP1 trigger a robust innate immune response. Portions of this chapter are derived from the previously published research articles: Seregin SS, Rastall DP, Evnouchidou I, Aylsworth CF, Quiroga D, Kamal RP, GodbehereRoosa S, Blum CF, York IA, Stratikos E, Amalfitano A. Endoplasmic reticulum aminopeptidases-1 alleles associated with increased risk of ankylosing spondylitis reduce HLAB27 mediated presentation of multiple antigens. Autoimmunity. 2013; 46(8):497-508. Quiroga D – contributed towards HeLa cell transfection experiments and the measurement of ERAP1 expression following transfection 143 5.1 Introduction Endoplasmic reticulum aminopeptidase 1 (ERAP1) protein was first characterized for its ability to trim and process antigens, thereby preparing them for MHC molecule loading (270). Recently, it has also been implicated as the second-most pathologically-associated gene of the autoimmune disease, ankylosing spondylitis (AS), a condition which causes inflamed, painful vertebral joints and often disability (271). Approximately 0.5% of Americans will suffer from this disease in their lifetime, yet the current treatment options available to patients are limited and mainly palliative (271). Moreover, the pathogenesis of this disease is not well defined. It is known that over 90% of AS patients are HLA-B27-positive, with twin studies showing there to be approximately a 75% monozygotic to 27% dizygotic concordance rate (272). However, less than 5% of the HLA-B27 positive population ever develops AS, which suggests that other genes must be implicated in its development as well (272,273). Genome-wide association studies (GWAS) have been able to link certain ERAP1 polymorphisms to the decreased likelihood of an HLA-B27-positive individual developing AS, while other ERAP1 polymorphisms increased the likelihood of developing AS (271,274). The exact functional mechanism of how these genes may bias an individual towards or away from developing AS is unknown, although HLA-B27’s predisposition to misfold and allow for peptide aggregation, particularly when ERAP1 is absent; suggesting that the endoplasmic reticulum (ER)-based cellular stress reaction, known as the unfolded protein response (UPR), and/or other inflammatory responses may be to blame (275–277). ERAP1 has also been implicated in the pathogenesis of other diseases, such as hypertension, inflammatory bowel disease, and psoriasis (278–280). Besides antigen processing, ERAP1 is known to directly promote TNFα and IL-6 receptor shedding, therefore, 144 overexpression of ERAP1 may alter pro-inflammatory innate responses (281,282). Moreover, TLR and/or LPS and IFNγ stimulation of murine macrophage RAW264.7 cells has been found to promote ERAP1 extracellular secretion (283,284). Exposure of RAW264.7 cells to this secreted form of ERAP1 was also shown to promote cellular phagocytosis activity, further supporting the hypothesis that ERAP1 may have functional significance in the innate immune response (283). In these studies, we wished to investigate the effects that the presence of low-AS risk and high-AS risk ERAP1 alleles may have on UPR and pro-inflammatory signaling. To do this, we constructed a HLA-B27 restricted HeLa cell model to examine intrinsic differences overexpression of low- or high-AS risk ERAP1 allele may induce upon UPR gene expression. We also created rAd vectors to express the low- and high-AS risk ERAP1 alleles to study how UPR and pro-inflammatory genes may react to ERAP1 overexpression. Finally, we tested the function of macrophages exposed to ERAP1-expressing rAds in vivo and how low- or high-AS risk ERAP1 expression may alter their in vitro phagocytosis activity. Our findings suggest ERAP1 overexpression does promote UPR-related gene expression and that high-risk ERAP1 expression may induce a greater level of pro-inflammatory innate immune signaling; further implicating ERAP1 with AS pathogenesis and innate immune signaling. 145 5.2 Methods 5.2.1 Cell transfection and culture pTracer-CMV-RFP plasmids encoding either low- or high-AS risk human ERAP1 alleles were created using a QuikChange II XL Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA) as per manufacturer’s instructions and as previously described (270,276). The high-AS risk ERAP1 allele is the HeLa-endogenous sequence, with the low-AS risk ERAP1 allele differs from it at five amino acid positions: M349V, K528R, D575N, R725Q, and Q730E. All of these polymorphisms were previously identified in GWASs to be significantly associated with the increased risk of AS diagnosis (285,286). HeLa-Kb-B27/47 cells were cultured at 3x105 cell/well on 6 well plates in high-glucose DMEM media (Life Technologies) supplemented with 10% FBS, 100 μg/mL Geneticin (Life Technologies), 100 μg/mL Zeocin (Invivogen), and 500 μg/mL Hygromycin (Life Technologies). HeLa-Kb-B27/47 cells were transfected with 1 μg low AS-risk (ERAP1_low) or high-AS risk (ERAP1_high) ERAP1-containing plasmids using the HeLA-specific TransIT HeLaMonster transfection kit (Mirus Bio LLC, Madison, WI). Transfection efficiency was typically observed by flow cytometry as 5-10% RFP-positive cells (287). RNA and protein samples were collected at 24 hours post-transfection (hpt). THP-1 monocytes were cultured in DMEM media (Life Technologies) supplemented with 10% FBS and 1x PSF. Monocytes were differentiated to macrophages by application of 1 μg/mL phorbol myristate acetate (PMA) for twelve hours, after which media was changed and macrophages were left PMA-free for twelve hours prior to treatment. RAW264.7 macrophages were cultured in RPMI 1640 (Life Technologies) containing 10% FBS. 146 5.2.2 Recombinant adenovirus construction and cell infection ERAP1_low and ERAP1_high gene open reading frames were inserted within CMVdriven pShuttle cassettes, then linearized, recombined, and propagated within BJ 5183 cells using a pAdEasy system, as previously described (182,205,206). The resulting Ad5-ERAPlow and Ad5-ERAPhigh vector titers were quantified by spectrophotometry. Ad5 gene region deletions were confirmed by PCR, as previously described (182). All viral vectors were tested and validated as having <0.01 EU (per injection dose) bacterial endotoxin contamination, as previously described (208). 5.2.3 RNA extraction, cDNA synthesis, and quantitative RT-PCR Cellular RNA was extracted using TRIzol reagent (Life Technologies), as detailed in manufacturer’s manual. cDNA was synthesized by reverse transcription using SuperScript III First-Strand Synthesis SuperMix (Life Technologies) as per manufacturer’s instructions. Quantitative RT-PCR was performed and analyzed using SYBR Green PCR Mastermix (Life Technologies) on an ABI 7900HT Fast Real-Time PCR System (Life Technologies) as previously described (189,209). Primers used to detect cellular gene expression were designed using the online PrimerBank database (http://pga.mgh.harvard.edu/primerbank) (Table 5). To measure relative gene expression, GAPDH and the comparative Ct method were used for all samples. Induction of gene expression was calculated as the relative change from the level of mock-treated cell transcripts to the level of rAd-treated cell transcripts. 147 Table 5 Quantitative RT-PCR primers for ERAP1 studies Primers were designed using PrimerBank online software. Primer sequences are written 5’ to 3’. 148 5.2.4 Protein isolation and western blotting HeLa-Kb-B27/47 cells were mock transfected or transfected with pTracer-CMV-RFP, pTracer-CMV-RFP-ERAP1_Low, or pTracer-CMV-RFP-ERAP1_High. Protein was harvested using lysis buffer (20mM Tris-HCl, pH 7.4, 1mM EDTA, 150mM NaCl, 1% Triton X-100, 1x protease inhibitor) at 24 hpt. Protein was isolated by centrifugation and quantified by BCA, as previously described (231). Equal amounts of protein were run and separated electrophoretically using 7% polyacrylamide gels. Protein bands on the gels were transferred onto nitrocellulose membranes, then probed using a mouse anti-human tubulin monoclonal primary antibody (Sigma-Aldrich) and a polyclonal rabbit-anti-hERAP1 (Proteintech, Chicago, IL). Following incubation with primary antibodies, IRDYE800-conjugated goat anti-mouse IgG (Rockland) and Alexa Fluor 680-conjugated goat anti-rabbit IgG (Invitrogen) were used as detection secondary antibodies. Membranes were scanned for protein bands using a LI-COR Odyssey scanner and software (LICOR Biosciences, Lincoln, NE). Fluorescence of the samples’ tubulin bands were used to normalize ERAP1 protein levels. 5.2.5 In vitro phagocytosis assay Measurement of in vitro phagocytosis activity was completed using a phagocytosis assay kit (FITC) (Cayman Chemical, Ann Arbor, MI). Briefly, RAW264.7 macrophages were cultured at 3x104 cells/well and were either mock infected or infected with 20,000 vp/cell control rAd5, Ad-ERAPlow, or Ad-ERAPhigh. At 48 hpi, cells were washed with PBS and given fresh cell media containing 2.5 μL FITC-labeled bead stock per 1 mL media. 18 hours following bead application, cells were harvested, washed, and resuspended in FACS buffer. The quantity and 149 intensity of FITC-positive cells was measured by FACS analysis using an LSRII flow cytometer and FlowJo software (Tree Star). 5.2.6 Statistical analysis Statistical significance all other data was determined using one-way ANOVAs with Newman-Keuls post-hoc corrections, unless otherwise stated. Graphs in this chapter are presented as mean ± standard error. Statistical analyses were performed using GraphPad Prism (GraphPad Software). 150 5.3 Results 5.3.1 ERAP-1 expression following transfection of HeLa-Kb-B27/47 cells We utilized a previously described HeLa cell model (HeLa-Kb-B27/47) to stably express human HLA-B27, murine H-2Kb, and a TAP1 blocker (herpesvirus protein ICP47) to allow only for peptides that possess a specific ER-localization sequence to be substrates for ERAP1-based HLA-B27 antigen loading (276,287–290). To test the function of this system, we transfected HeLa-Kb-B27/47 cells with RFP (control plasmid), ERAP1_Low (low-AS risk ERAP1 allele), or ERAP1_High (high-AS risk ERAP1 allele) plasmids or performed a mock transfection on them. Following collection of RNA and protein samples at 24 hpt, we found there to be no significant differences in the amount of ERAP1 gene (Figure 26A) or protein expression (Figure 26B,C) following ERAP1_Low or ERAP1_High plasmid transfection. Both ERAP1 plasmids expressed approximately 700 times the ERAP1 RNA and 4 times the ERAP1 protein concentration as the endogenous HeLa-Kb-B27/47 ERAP1 amounts. This indicates that it is unlikely that ERAP1_Low or ERAP1_High alleles create more or less ERAP1 production or persistence at the transcriptional and translational levels. RFP, our control plasmid, failed to produce a significantly greater amount of ERAP1 RNA or protein than mock, indicating that the transfection process alone was not inducing significant amounts of ERAP1 expression. 151 Figure 26 ERAP1 expression following HeLa-Kb-B27/47 transfection 152 Figure 26 (cont’d) HeLa-Kb-B27/47 cells were mock transfected or transfected with plasmids expressing RFP, ERAP1_Low, or ERAP1_High alleles. RNA and protein were harvested from samples at 24 hpt. (A) ERAP1 mRNA levels were measured using RT-PCR quantification. (B,C) ERAP1 protein concentrations were measured by Western blot. Bars denote mean ± standard error. * denotes a significant difference between that group and mock-transfected cells (p<0.05). 153 We used this model to test if low- and high-AS risk ERAP1 alleles express different levels of genes previously identified as overexpressed by AS patients in GWAS studies. To test this, HeLa-Kb-B27/47 cells were mock transfected or transfected RFP, ERAP1_Low, or ERAP1_high plasmids, then harvested at 24 hpt to measure expression of other AS and UPRrelated genes. The genes we chose to test were the ones most strongly related to genes differentially expressed in mammalian models of HLA-B27-induced spondyloarthropathies, particularly after IFNγ-triggered UPR activation; these include viperin, CHOP, HLA-B, and IRF1 (291–293). We found that introduction of the RFP plasmid into HeLa-Kb-B27/47 cells resulted in significant inductions of viperin (p<0.05), CHOP (p<0.01), HLA-B (p<0.001), and IRF1 (p<0.01) over mock transfection (Figure 27). However, ERAP1_Low and ERAP1_High allele plasmid-transfected cells did not produce significantly different gene expression patterns from each other or from the control RFP plasmid-transfected cells; possibly suggesting that it may be possible the transfection procedure alone could induce these genes and mask any effects that the ERAP1 plasmids may have induced. 154 Figure 27 Expression of AS-related genes in ERAP1-transfected HeLa-Kb-B27/47 cells HeLa-Kb-B27/47 cells were mock transfected or transfected with plasmids expressing RFP, ERAP1_Low, or ERAP1_High alleles. RNA was harvested from samples at 24 hpt and analyzed by RT-PCR quantification for gene expression. *, **, *** denotes a significant difference between that group and mock-transfected cells (p<0.05, p<0.01, p<0.001, respectively). 155 5.3.2 Ad-ERAPlow and Ad-ERAPhigh activation of AS-related gene immune responses Considering the results we found with our HeLa-Kb-B27/47 system, we decided to test if ERAP1_low and ERAP1_high could promote altered responses in another cell system. We selected to use macrophage-differentiated THP-1 cells as a model to study in vitro ERAP1_Low and ERAP1_High differences as these cells are functionally more similar to in vivo human professional APCs. We also wished to allow for a longer-term and constitutive expression of the ERAP1 genes, therefore, we constructed rAd5[E1-] vectors expressing the low- and high-AS risk ERAP1 alleles (Ad-ERAPlow and Ad-ERAPhigh, respectively), as previously described (172,205,206,209). To test if low- and high-risk ERAP1 alleles could induce the expression of other ASrelated genes when introduced by rAd5 vectors, we mock infected or infected THP-1 macrophages for 24 hours with a control rAd5 vector, Ad-ERAPlow, or Ad-ERAPhigh. Gene expression of the viperin, CHOP, HLA-B, and IRF1 UPR-related genes, two additional UPRrelated genes (BiP and XBP1s), and IFIT1 and IFIT2 innate immune and UPR components, was measured as conducted previously (293–295,292). Compared to mock and control rAd5-treated cells, gene expression in Ad-ERAPhigh treated cells were able to induce significantly greater amounts of viperin (p<0.001, p<0.01, respectively) , BiP (p<0.01), CHOP (p<0.01), HLA-B (p<0.05 with mock-treated cells), IFIT1 (p<0.001), IFIT2 (p<0.001), IRF1 (p<0.001), and XBP1s (p<0.05) (Figure 28). Ad-ERAPlow treated cells were also able to induce several ASrelated gene expression over mock and control rAd5-treated cells, including viperin (p<0.01), BiP (p<0.01), CHOP (p<0.05, p<0.01, respectively), IFIT1 (p<0.001), IFIT2 (p<0.001), and IRF1 (p<0.001). It was also noted that the control rAd5 infection induced significantly greater 156 amounts of viperin (p<0.01), IFIT1 (p<0.01), IFIT2 (p<0.001), and IRF1 (p<0.01) gene expression than mock treatment, which is consistent with previous rAd5 studies (141,148). 157 Figure 28 Ad-ERAPlow and Ad-ERAPhigh induce AS-related gene expression 158 Figure 28 (cont’d) THP-1 macrophages were mock infected or infected with a control rAd5 vector, AdERAPlow, or Ad-ERAPhigh at 20,000 vp/cell. At 24 hpi, RNA was isolated for RT-PCR quantification of Ad gene expression. Bars denote mean ± standard error. *, **, *** denote significant differences between that group and mock-treated samples (p<0.05, p<0.01, p<0.001 respectively). #, ##, ### denote significant differences between Ad treatments (p<0.05, p<0.01, p<0.001 respectively). 159 5.3.3 Phagocytosis activity of RAW macrophages following Ad-ERAP stimulation Following the confirmation that Ad-ERAPlow and Ad-ERAPhigh vectors were able to promote production of genes which are upregulated in association with AS diagnosis, we wished to test if these vectors could also promote functional in vitro differences in macrophages. Previous studies have shown that exogenously-introduced ERAP1 protein can stimulate murine RAW264.7 macrophage phagocytosis activity (283). Therefore, we wished to test if endogenous introduction of low- and high-AS risk ERAP1 alleles via rAd5 would also induce phagocytosis actions in a macrophage cell line. RAW264.7 cells were either mock infected or infected with a control rAd5, Ad-ERAPlow, or Ad-ERAPhigh. The day after infection, FITC-labeled beads were applied to each sample. Following bead incubation, macrophages were collected and washed to measure the amount of cells that had internalized FITC-labeled beads and the amount of beads engulfed per cell. We found that bead application alone induced about a 28% rate of phagocytosis and that application of an rAd5 vector to these cells increased this rate to ~ 37%, while also increasing the amount of beads each cell was able to engulf, as measured by MFI (p<0.001) (Figure 29). Moreover, it was observed that a greater percentage of RAW264.7 macrophages treated with Ad-ERAPlow or Ad-ERAPhigh vectors, as opposed to mock treatment, were activated to phagocytose the FITC-labeled beads upon application (p<0.001). Ad-ERAPlow or Ad-ERAPhigh treated cells also phagocytosed a larger average amount of beads per cell (p<0.001). In comparing the amount of phagocytically-active macrophages and the concentration in which they engulf FITC-labeled beads, we found that Ad-ERAPhigh treatment resulted in significantly greater phagocytosis activity than Ad-ERAPlow (p<0.05, p<0.01, respectively). 160 These studies were also attempted using the THP-1 cell line, but due to PMA stimulation required to differentiate THP-1 monocytes to macrophages, these cells were immunologically activated prior to infection. This effect was so strong that even the mock-treated cells were stimulated so to allow for close to 100% of the applied FITC-beads to be engulfed (data not shown). 161 Figure 29 Ad-ERAPlow and Ad-ERAPhigh stimulate RAW macrophage phagocytosis activation 162 Figure 29 (cont’d) RAW264.7 macrophages (n=3) were mock infected or infected with 20,000 vp/cell control rAd5, Ad-ERAPlow, or Ad-ERAPhigh, then treated with FITC-labeled beads. At 18 hours post-bead application, cells were harvested to measure the quantity (percent of FITC+ cells) and intensity (MFI) of FITC-positive cells using FACS analysis. Bars denote mean ± standard error. *** denotes significant differences between that group and samples not treated with beads (p<0.001). ^^^ denotes significant differences between that group and the mock treatment group (p<0.001). #, ##, ### denote significant differences between Ad treatments (p<0.05, p<0.01, p<0.001 respectively). 163 5.4 Discussion Originally characterized as only a processor of MHC-antigen loading, human ERAP1 has been implicated in a variety of processes, from cytokine receptor shedding to anti-viral miRNA degradation (281,282,296). It seems that many of its disease-related roles are double-sided, in that ERAP1 can either increase or decrease the risk of pathogenesis; dependent upon the ERAP1 polymorphism that an individual carries (272,274,297,298). Recently, our group has also found that ERAP1 is able to skew the selection of which epitopes will be immunodominant in APC presentation and can alter the balance of which type of memory T cell response will be developed (299). Although ERAP1 clearly demonstrates association with polymorphismdependent immune alterations, the mechanism by which it acts on these systems is currently unknown. In this chapter, we used an HLA-B27-expressing HeLa cell model to test the expression of ERAP1 low- and high-AS risk variants following their introduction by plasmid transfection. We found that this method was able to induce about a 500-1000 fold induction of ERAP1 mRNA expression and about a 3-4 fold induction of ERAP1 protein expression, independent of which ERAP1 variant plasmid was used. These similarities reveal that it is unlikely that the differences between low- and high-AS risk ERAP1 alleles due to differences in the protein’s naturallyoccurring transcriptional or translational levels, but instead are caused by altered ERAP1 functions. Our group also conducted further experiments with the HeLa-Kb-B27/47 cell system in which low- and high-AS risk ERAP1 plasmids were co-transfected with selected peptide epitopes; revealing that the high-AS risk ERAP1 variant degrades HLA-B27-specific epitopes at a greater rate, is less efficient at loading those epitopes onto HLA-B27 molecules, and, overall, decreases HLA-B27 surface expression of to a greater extent than the low-risk ERAP1 variant 164 (287). The idea that high AS-risk ERAP1 alleles may have an increased rate of peptide processing to blame for functional differences between the variants is supported by previous preliminary studies that have suggested low-AS risk ERAP1 polymorphisms possess decelerated peptidase activity rates (274). The UPR is an important cellular stress response, induced by accumulation of dysfunctionally-folded proteins in the ER lumen, that allows for cellularly-resuscitative measures to take place and clear compartmental aggregates before proceeding towards apoptosis signaling (300). It is believed that neurological diseases characterized by over-accumulation of harmful/misfolded protein products (e.g. Alzheimer’s disease, Parkinson’s disease) could be linked to UPR inhibition (301,302), however, excessive UPR signaling may also be to blame in the inflammatory pathogenesis of other diseases, including spondyloarthropathies like AS (293,303,304). In studying the UPR in our HeLa-Kb-B27/47 cell system, we were able to see inductions of multiple UPR genes upon ERAP1_Low and ERAP1_High plasmid transfection. However, as these genes were also upregulated upon RFP control plasmid transfection, it was realized that due to the nature of the transfection procedure and/or the efficacy of this cell model, there may have been other ways to more opportunistically study the ERAP1-UPR interactions. Additionally, we recognized the importance of measuring low- and high-AS risk ERAP1 effects in a non-HLA-B27-dependent environment and, also, in an APC model so that the cells in which ERAP1’s antigen processing activities are most utilized may be examined (270,281,288). While several UPR-related genes were found to be upregulated by rAd expression of ERAP1, production of the high-AS risk allele allowed for a more robust induction of those UPR genes that have also been implicated in interferon-inducible innate immunity. Interestingly, Ad- 165 ERAPhigh treatment also increased the amount and breadth of in vivo macrophage activation as compared to Ad-ERAPlow treatment, although both did induce high levels of phagocytosis from baseline and the mean difference in the amount of cells that phagocytosed FITC-labeled beads between the two treatment groups was only about 2.5%. Whether this amount of variance is noteworthy, or if larger discrepancy may be observed in a human macrophage line or in vivo animal model, would require further validation. Like other gene products activated during innate immunity, ERAP1 expression is triggered by exposure to TLR agonists and pro-inflammatory cytokines (270,283,284). The understanding of ERAP1 as more than just a step along the antigen processing and loading path is still developing, and from our results, it seems likely that these “non-traditional” roles may play a larger part in how ERAP1 protects or predisposes an individual from/to specific, ERAP1 linked diseases. Acknowledgments We wish to thank the personnel in Michigan State University’s laboratory animal support facilities for their assistance in the humane care of animals utilized in this work. A.A. was supported by the Michigan State University Osteopathic Heritage Foundation. 166 Chapter 6: Summary and future directions 167 The modern development of vaccines cannot be undertaken without thorough consideration of each human immune response component and how vaccine formulations may manipulate them. From the initial physical barriers that microbes must pass to enter the human body, to epitope-specific adaptive targeting of pathogenic antigens; the interplay of each cell type, signaling pathway, and cytokine expressed combine to result in a complex immune system activation milieu. Thus, vaccine researchers are employed with the task of developing vaccine components that direct an individual towards the types of immune responses thought to be needed for an effective immune response against antigens present in harmful microbes and/or tumor cells (305). In this dissertation, we discussed the immunogenic properties that both recombinant adenovirus vaccine vectors and TLR agonist-based adjuvants possessed, while also introducing novel methods and mechanics by which these constituents may be effectivity utilized. In Chapter 2, we compared the biological and innate immune characteristics of firstgeneration Ad5[E1-] and second-generation Ad5[E1-,E2b-] vectors. These rAd5 vectors only differ by an additional deletion of the E2b transcriptional region; a sequence that encodes the necessary genes for Ad5 DNA polymerase and PTP synthesis. This small modification within the Ad5[E1-,E2b-] platform has been shown to result in several advantageous vaccine properties as compared to Ad5[E1-] vectors; including lower hepatotoxicity, longer in vivo persistence, and decreased expression of viral proteins (131,138,181,182). Perhaps most desirable, Ad5[E1-,E2b] vector activities are not as easily disabled by pre-existing anti-Ad5 immune responses as Ad5[E1-] vectors are; a phenomena often thought as being due to the “leaky” Ad5[E1-] viral protein expression that doesn’t substantially exist with Ad5[E1-,E2b-] vectors (181,183–186). 168 Looking back on these previous studies, Ad5[E1-] and Ad5[E1-,E2b-] vectors have mainly been compared for their divergences in adaptive immune responses. Alternatively, we wished to compare the early, innate responses induced by Ad5[E1-] and Ad5[E1-, E2b-]. Prior to experimentation, we analyzed previously published mouse liver RNA microarray data to determine if there were differences between animals given Ad5[E1-] or Ad5[E1-, E2b-] vectors and at which time-point after infection did they occur. While the large majority of the Ad5[E1-] and Ad5[E1-,E2b-] vector-induced transcriptomes were similar, there were several significant gene groups induced at a higher amount than the other vector. We found that most Ad5[E1-] and Ad5[E1-,E2b-] vector differences occurred within the first day of infection and mainly disappeared by 3 dpi, suggesting that the dissimilarities between rAds are more likely to be based upon an early, innate responses. To test this hypothesis, we gathered blood samples from 17 independent human donors. Utilizing a large number of blood samples not only increased our sample size, but also allowed us to obtain serum and T cells from individuals with varied amounts of anti-Ad5 antibodies and CTLs; therefore, these adaptive components could be compared side-to-side with the innate hPBMC products. In measuring the cytokines secreted from these blood donor samples after they were infected with mock, Ad5[E1-], or Ad5[E1-,E2b-] vectors, we found a surprisingly unified pro-inflammatory cytokine signaling pattern. For IL-1β and TNFα expression, Ad5[E1-,E2b-] vector infection served as a significantly greater stimulant than Ad5[E1-]. When these cytokine responses were plotted against the same donor’s anti-Ad5 IgG titer and anti-Ad5 CD8+ CTL levels, we found no association between the amount of pre-existing Ad5 immunity and the measured cytokine responses measured after rAd5 infection (by either Ad5[E1-] or Ad5[E1,E2b-] vectors). These data strongly argue that the functional differences between Ad5[E1-] and 169 Ad5[E1-, E2b-] are not completely attributed to adaptive anti-Ad5 responses, but also to more intrinsic innate response differences between the vectors. We further compared these first- and second-generation rAd vectors by measuring levels of Ad-specific gene expression and found that the Ad5[E1-] vectors produced tremendously greater amounts of both early and late Ad5 gene products than Ad5[E1-, E2b-] did. Interestingly, innate immune gene induction triggered by Ad5[E1-, E2b-] vector treatment was generally higher than Ad5[E1-] vector-treated cells. Moreover, cells infected with Ad5[E1-] vectors produced such blunted expression of the pro-inflammatory IFIT1 gene that the levels were significantly lower than that which was produced by cells infected with an rAd5 vector lacking any Ad genes (HD-Ad5). With this in mind, it appears that Ad5[E1-] vector infection may even promote inhibition of certain innate immune responses. . Future studies to characterize these potential inhibitory signals and how they can be modulated to create an immunogenic, but safe, viral platform would serve as important tools to more thoroughly understand how a beneficial vaccine response can be mounted. In upcoming investigations, determining the mechanism by which Ad5[E1-] and Ad5[E1-,E2b-] vectors differentially trigger innate immunity should be a prioritized goal. The methods and problemsolving approach by which such a task could be completed are diverse. For example, a more extensive immune gene and cytokine expression profile could be obtained further comparing Ad5[E1-] and Ad5[E1-,E2b-] infected hPBMCs; this may help to indicate which specific pathways are being utilized differently between vectors. Experiments could also be conducted to investigate if greater expression of certain Ad5 genes (e.g. VARNA, hexon) by Ad5[E1-] over Ad5[E1-,E2b-] may be leading to the early, pro-inflammatory dissimilarities. 170 In Chapter 3, we examined the characteristics a novel Eimeria tenella-derived TLR agonist, rEA, which has the ability to activate and mature many innate immune cells when delivered exogenously as a purified protein. From this work, we were able to conclude that TRIF-KO can promote rEA-stimulatory activities to a greater extent than even WT mice, suggesting that TRIF acts as a sort of inhibitor against rEA immunostimulatory effects. Alternatively, MyD88-KO and MyD88/TRIF-DKO animals completely lacked any rEAmediated innate stimulation, suggesting that MyD88 is essential for rEA early signaling. These findings support the current hypothesis that rEA is highly homologous to the TLR11 agonist, profilin; not only in protein sequence, but also in immune-induced responses (229). Similar to other TLR ligands, profilin-like protein recognition by TLR11 had previously been found to be a significant, MyD88-dependent process by which the innate and CMI could be stimulated (306). Since the publication of the Chapter 3 results, it has been found that while humans do not produce functional TLR11 (230), profilin proteins can be recognized by and trigger activation of human TLR5 (266). As rEA shares significant homology to profilin and other profilin-like proteins (223,229), this seems likely to be the MyD88-dependent pathway in which it promotes pro-inflammatory signals. Moreover, other researchers have found that TLR11 can be activated by the introduction of flagellin, which has traditionally been viewed as a TLR5 agonist (307). While TLR5 and TLR11 both signal through the MyD88 adaptor protein, TLR5 has been identified as a plasma membrane-bound receptor, while TLR11 is believed to reside mainly in endolysosomal compartments within the endoplasmic reticulum (308). Whether rEA actually is a human TLR5 ligand and if detection of rEA extracellularly versus within endolysosomal compartments may alter the type of immunity it produces are questions that would be beneficial to answer in future pre-clinical studies. For example, our group has also published findings in 171 which mice were injected with either rEA protein or the rAd5-rEA vector. Both groups were found to have significant inductions of many pro-inflammatory cytokines, however, rAd5-rEA vectors produced a much more significant quantity of response and stimulation (187). This may be due to the fact that vectors can promote a more robust production of its transgene and/or that, by being immunogenic itself, a transgene-expressing rAd5 may be expected to be more proinflammatory (148,171). However, it is also possible that, due to endosomal TLR11 positioning (308), internalized rAd5-rEA vectors have better access to this PRR than extracellularlyintroduced rEA protein. Whether these differences in TLR cellular locations may or may not be a factor in rEA-triggered immunity, it is, nevertheless, important to consider during future TLR agonist studies. We also worked to create an rEA-expressing rAd5 vector, which was found to promote co-injected transgene memory responses of such pathogens as the HIV Gag protein. In Chapter 4, rAd5-rEA was used alongside a therapeutic colorectal cancer vaccine that is currently in phase IIb clinical trials, rAd5-CEA (186). As CEA is a highly sensitive marker for colorectal cancer, is overexpressed in many other carcinomas, and possesses some natural immunogenicity, it has become one of immunotherapy researchers’ favorite TAA targets (241). While rAd5-CEA trials have shown promising inductions of anti-CEA CMI, as well as increased survival rates in the patients given higher doses of this vector (186), we believe this response could be heightened even more with the introduction of the adjuvant, rEA. Throughout our studies, murine injection of rAd5-CEA + rAd5-rEA was compared in a variety of ways to the control vaccine protocol, rAd5-CEA + rAd5-GFP. Both rAd5 vaccine combinations induced dose-dependent increases in the amount of CEA-specific CMI, CTL, and IgG production, but the rAd5-CEA + rAd5-rEA 172 treatment provided the most superior amounts of each of these factors when utilized at the highest dose (1 x1010 vp total). After establishing a greater intensity of anti-CEA cell-mediated and humoral adaptive immune responses following rAd5-CEA + rAd5-rEA vaccination, we characterized the diversity of the response. We found that rAd5-CEA + rAd5-rEA vaccination of mice promoted recognition of several CEA epitopes, whereas rAd5-CEA + rAd5-GFP produce a more narrow scope of antigenic epitopes, as seen with both CEA-directed CMI and CEA-neutralizing IgG antibodies. Finally, an in vivo CTL killing assay was also used to verify that rAd5-CEA + rAd5rEA vaccination effects could translate to functional recognition and killing of TAA-expressing cells. Considering these encouraging results, the next pre-clinical step to take this vaccineadjuvant combination treatment would be to an animal tumor model. Past rAd5-CEA studies have utilized a C57BL/6 murine-derived colon adenocarcinoma cell line that has been modified to express human CEA on its surface (MC38-cea2) to test its anti-tumor vaccine efficacy (196), so this would serve as an appropriate model to conduct such an experiment for rAd5-CEA + rAd5-rEA vaccination studies as well. Additionally, it may be advantageous to use human CEAtransgenic mice so to more accurately replicate the spatiotemporal pattern in which colorectal cancer patients typically express CEA and other TAAs (309,310). When developing new adjuvants and vaccine models for cancer immunotherapy, it is also important to consider the potential side effects of excessive immune stimulation or induced autoimmune reactions. While current rAd5-CEA clinical trial participants have not complained or presented with serious side effects, there have been cancer immunotherapy trials in the past in which severe autoimmune patient reactions have occurred, with one testing a CEA-specific 173 adoptive T cell transfer treatment that lead to an acute, autoimmune colitis (109,254). In future studies, this vaccination protocol and others should be monitored within in vivo models for signs of such exorbitant and toxic autoimmune effects. In Chapter 5, we briefly examined the implications of low- and high-AS risk ERAP1 alleles and how the small polymorphic differences between them may lead to altered UPR and/or innate immune responses by infecting cell cultures with ERAP1-expressing vectors specific to each allele. Within Ad-ERAPhigh vector-infected macrophage lines, we were able to measure significant upregulation of genes associated with both pro-inflammatory and unfolded protein responses over that seen in macrophages administered the Ad-ERAPlow vector. Also, AdERAPhigh vector infection promoted significantly greater levels of macrophage phagocytosis activity, further implicating that the high-AS risk allele may induce a stronger innate immune response than other ERAP1 polymorphisms. Our group is currently conducting extensive research using human ERAP1 transgenic mice and other applicable models that will be useful for the characterization of ERAP1’s novel immunologic roles. Additional clinically-relevant experiments that could be conducted to study AS-related ERAP1 alleles are ones in which blood and tissue samples are obtained from symptomatic AS patients and compared for innate immune and UPR-related gene and protein expression differences, as compared to samples from persons without AS. In summation, this dissertation includes several promising studies and results for the fields of vaccinology and cancer immunotherapy. We showed that first-generation rAd5 vectors, which have received considerable criticism in the past for some of their detrimental side effects and inefficacies, can be re-engineering into E2b-deleted second-generation rAd5 vectors that break past many of these barriers. We also extensively studied the properties of TLR agonist 174 rEA, both alone and alongside a cancer vaccine platform. Lastly, we touched on some of the work we completed regarding the antigen-processing ERAP1 protein and how it may modulate cellular stress and innate immune pathways. All together, these studies highlight many of the methods by which innate immunity directly and indirectly plays a role in the synthesis of a valuable, robust memory vaccine response. 175 BIBLIOGRAPHY 176 BIBLIOGRAPHY 1. J Philpott D, E Girardin S, J Sansonetti P. Innate immune responses of epithelial cells following infection with bacterial pathogens. Curr Opin Immunol. 2001; 13(4):410–416. 2. Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002; 20:197–216. 3. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124(4):783–801. 4. Girardin SE, Sansonetti PJ, Philpott DJ. Intracellular vs extracellular recognition of pathogens – common concepts in mammals and flies. Trends Microbiol. 2002; 10(4):193– 199. 5. Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007; 7(3):179–190. 6. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11(5):373–384. 7. Ting JP-Y, Trowsdale J. Genetic control of MHC class II expression. Cell. 2002; 109 Suppl:S21–33. 8. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008; 8(12):958–969. 9. Villadangos JA, Schnorrer P. Intrinsic and cooperative antigen-presenting functions of dendritic-cell subsets in vivo. Nat Rev Immunol. 2007; 7(7):543–555. 10. Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat Rev Immunol. 2008; 8(6):435–446. 11. Rescigno M, Sabatino A Di. Dendritic cells in intestinal homeostasis and disease. J Clin Invest. 2009; 119(9):2441–2450. 12. Leon B, Ardavin C. Monocyte-derived dendritic cells in innate and adaptive immunity. Immunol Cell Biol. 2008; 86(4):320–324. 13. Sun JC, Lopez-Verges S, Kim CC, DeRisi JL, Lanier LL. NK cells and immune “memory.” J Immunol. 2011; 186(4):1891–1897. 14. Paust S, Andrian UH von. Natural killer cell memory. Nat Immunol. 2011; 12(6):500– 508. 177 15. Lieberman N, Mandelboim O. The role of NK cells in innate immunity. Adv Exp Med Biol. 2000; 479:137–145. 16. Terunuma H, Deng X, Dewan Z, Fujimoto S, Yamamoto N. Potential role of NK cells in the induction of immune responses: implications for NK cell-based immunotherapy for cancers and viral infections. Int Rev Immunol. 2008; 27(3):93–110. 17. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011; 331(6013):44–49. 18. Makino Y, Kanno R, Ito T, Higashino K, Taniguchi M. Predominant expression of invariant V alpha 14+ TCR alpha chain in NK1.1+ T cell populations. Int Immunol. 1995; 7(7):1157–1161. 19. Zlotnik A, Godfrey DI, Fischer M, Suda T. Cytokine production by mature and immature CD4-CD8- T cells. Alpha beta-T cell receptor+ CD4-CD8- T cells produce IL-4. J Immunol. 1992; 149(4):1211–1215. 20. Hermans IF, Silk JD, Gileadi U, Masri SH, Shepherd D, Farrand KJ, et al. Dendritic cell function can be modulated through cooperative actions of TLR ligands and invariant NKT cells. J Immunol. 2007; 178(5):2721–2729. 21. Beutler BA. TLRs and innate immunity. Blood. 2009; 113(7):1399–1407. 22. Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF. A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci. 1998; 95(2):588–593. 23. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004; 4(7):499–511. 24. Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001; 410(6832):1099–1103. 25. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001; 413(6857):732–738. 26. Du X, Poltorak A, Wei Y, Beutler B. Three novel mammalian toll-like receptors: gene structure, expression, and evolution. Eur Cytokine Netw. 2000; 11(3):362–371. 27. Jin MS, Kim SE, Heo JY, Lee ME, Kim HM, Paik S-G, et al. Crystal Structure of the TLR1-TLR2 Heterodimer Induced by Binding of a Tri-Acylated Lipopeptide. Cell. 2007; 130(6):1071–1082. 28. Kang JY, Nan X, Jin MS, Youn S-J, Ryu YH, Mah S, et al. Recognition of Lipopeptide Patterns by Toll-like Receptor 2-Toll-like Receptor 6 Heterodimer. Immunity. 2009; 31(6):873–884. 178 29. Kawai T, Akira S. Signaling to NF-κB by Toll-like receptors. Trends Mol Med. 2007; 13(11):460–469. 30. Doyle SL, O’Neill LAJ. Toll-like receptors: from the discovery of NFkappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol. 2006; 72(9):1102–1113. 31. Boehme KW, Guerrero M, Compton T. Human cytomegalovirus envelope glycoproteins B and H are necessary for TLR2 activation in permissive cells. J Immunol. 2006; 177(10):7094–7102. 32. Leoni V, Gianni T, Salvioli S, Campadelli-Fiume G. Herpes simplex virus glycoproteins gH/gL and gB bind Toll-like receptor 2, and soluble gH/gL is sufficient to activate NF-κB. J Virol. 2012; 86(12):6555–6562. 33. Georgel P, Jiang Z, Kunz S, Janssen E, Mols J, Hoebe K, et al. Vesicular stomatitis virus glycoprotein G activates a specific antiviral Toll-like receptor 4-dependent pathway. Virology. 2007; 362(2):304–313. 34. Okumura A, Pitha PM, Yoshimura A, Harty RN. Interaction between Ebola virus glycoprotein and host toll-like receptor 4 leads to induction of proinflammatory cytokines and SOCS1. J Virol. 2010; 84(1):27–33. 35. Lester SN, Li K. Toll-like receptors in antiviral innate immunity. J Mol Biol. 2014; 426(6):1246–1264. 36. Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol. 2002; 3(2):196–200. 37. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000; 408(6813):740–745. 38. Guggemoos S, Hangel D, Hamm S, Heit A, Bauer S, Adler H. TLR9 Contributes to Antiviral Immunity during Gammaherpesvirus Infection. J Immunol. 2008; 180(1):438– 443. 39. Takeuchi O, Akira S. Innate immunity to virus infection. Immunol Rev. 2009; 227(1):75– 86. 40. Sumpter R, Loo Y-M, Foy E, Li K, Yoneyama M, Fujita T, et al. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol. 2005; 79(5):2689–2699. 41. Rothenfusser S, Goutagny N, DiPerna G, Gong M, Monks BG, Schoenemeyer A, et al. The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I. J Immunol. 2005; 175(8):5260–5268. 179 42. Fredericksen BL, Keller BC, Fornek J, Katze MG, Gale M. Establishment and maintenance of the innate antiviral response to West Nile Virus involves both RIG-I and MDA5 signaling through IPS-1. J Virol. 2008; 82(2):609–616. 43. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006; 441(7089):101–105. 44. Tortorella D, Gewurz BE, Furman MH, Schust DJ, Ploegh HL. Viral Subversion of the Immune System. Annu Rev Immunol. 2000; 18(1):861–926. 45. Jones TR, Wiertz EJ, Sun L, Fish KN, Nelson JA, Ploegh HL. Human cytomegalovirus US3 impairs transport and maturation of major histocompatibility complex class I heavy chains. Proc Natl Acad Sci. 1996; 93(21):11327–11333. 46. Piguet V, Schwartz O, Gall S Le, Trono D. The downregulation of CD4 and MHC-I by primate lentiviruses: a paradigm for the modulation of cell surface receptors. Immunol Rev. 1999; 168:51–63. 47. Mahr JA, Gooding LR. Immune evasion by adenoviruses. Immunol Rev. 1999; 168:121– 130. 48. Beck S, Barrell BG. Human cytomegalovirus encodes a glycoprotein homologous to MHC class-I antigens. Nature. 1988; 331(6153):269–272. 49. Smith CA, Hu FQ, Smith TD, Richards CL, Smolak P, Goodwin RG, et al. Cowpox virus genome encodes a second soluble homologue of cellular TNF receptors, distinct from CrmB, that binds TNF but not LT alpha. Virology. 1996; 223(1):132–147. 50. Benedict CA, Butrovich KD, Lurain NS, Corbeil J, Rooney I, Schneider P, et al. Cutting edge: a novel viral TNF receptor superfamily member in virulent strains of human cytomegalovirus. J Immunol. 1999; 162(12):6967–6970. 51. Smith GL, Chan YS. Two vaccinia virus proteins structurally related to the interleukin-1 receptor and the immunoglobulin superfamily. J Gen Virol. 1991; 72 ( Pt 3):511–518. 52. Foster GR, Ackrill AM, Goldin RD, Kerr IM, Thomas HC, Stark GR. Expression of the terminal protein region of hepatitis B virus inhibits cellular responses to interferons alpha and gamma and double-stranded RNA. Proc Natl Acad Sci. 1991; 88(7):2888–2892. 53. Isegawa Y, Ping Z, Nakano K, Sugimoto N, Yamanishi K. Human herpesvirus 6 open reading frame U12 encodes a functional beta-chemokine receptor. J Virol. 1998; 72(7):6104–6112. 54. Strockbine LD, Cohen JI, Farrah T, Lyman SD, Wagener F, DuBose RF, et al. The Epstein-Barr virus BARF1 gene encodes a novel, soluble colony-stimulating factor-1 receptor. J Virol. 1998; 72(5):4015–4021. 180 55. Rhee EG, Kelley RP, Agarwal I, Lynch DM, Porte A La, Simmons NL, et al. TLR4 Ligands Augment Antigen-Specific CD8+ T Lymphocyte Responses Elicited by a Viral Vaccine Vector. J Virol. 2010; 84(19):10413–10419. 56. Bessis N, GarciaCozar FJ, Boissier M-C. Immune responses to gene therapy vectors: influence on vector function and effector mechanisms. Gene Ther. 2004; 11 Suppl 1:S10– 17. 57. Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol. 2013; 13(5):309–320. 58. Davis SJ, Ikemizu S, Evans EJ, Fugger L, Bakker TR, Merwe PA van der. The nature of molecular recognition by T cells. Nat Immunol. 2003; 4(3):217–224. 59. Murphy KM, Reiner SL. The lineage decisions of helper T cells. Nat Rev Immunol. 2002; 2(12):933–944. 60. Wong P, Pamer EG. CD8 T cell responses to infectious pathogens. Annu Rev Immunol. 2003; 21:29–70. 61. Bluestone JA, Abbas AK. Natural versus adaptive regulatory T cells. Nat Rev Immunol. 2003; 3(3):253–257. 62. Rioux JD, Abbas AK. Paths to understanding the genetic basis of autoimmune disease. Nature. 2005; 435(7042):584–589. 63. Staveley-O’Carroll K, Sotomayor E, Montgomery J, Borrello I, Hwang L, Fein S, et al. Induction of antigen-specific T cell anergy: An early event in the course of tumor progression. Proc Natl Acad Sci. 1998; 95(3):1178–1183. 64. Lee PP, Yee C, Savage PA, Fong L, Brockstedt D, Weber JS, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med. 1999; 5(6):677–685. 65. Fearon DT, Carroll MC, Carroll MC. Regulation of B Lymphocyte Responses to Foreign and Self-Antigens by the CD19/CD21 Complex. Annu Rev Immunol. 2000; 18(1):393– 422. 66. Carrasco YR, Batista FD. B cell recognition of membrane-bound antigen: an exquisite way of sensing ligands. Curr Opin Immunol. 2006; 18(3):286–291. 67. Harwood NE, Batista FD. New Insights into the Early Molecular Events Underlying B Cell Activation. Immunity. 2008; 28(5):609–619. 68. Shapiro-Shelef M, Calame K. Regulation of plasma-cell development. Nat Rev Immunol. 2005; 5(3):230–242. 181 69. Pan-Hammarström Q, Zhao Y, Hammarström L. Class switch recombination: a comparison between mouse and human. Adv Immunol. 2007; 93:1–61. 70. Allen CDC, Okada T, Cyster JG. Germinal-Center Organization and Cellular Dynamics. Immunity. 2007; 27(2):190–202. 71. McHeyzer-Williams LJ, Malherbe LP, McHeyzer-Williams MG. Helper T cell-regulated B cell immunity. Curr Top Microbiol Immunol. 2006; 311:59–83. 72. Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med. 2000; 6(4):443–446. 73. Jin B, Sun T, Yu X-H, Yang Y-X, Yeo AET. The effects of TLR activation on T-cell development and differentiation. Clin Dev Immunol. 2012; 2012:836485. 74. Gelman AE, LaRosa DF, Zhang J, Walsh PT, Choi Y, Sunyer JO, et al. The Adaptor Molecule MyD88 Activates PI-3 Kinase Signaling in CD4+ T Cells and Enables CpG Oligodeoxynucleotide-Mediated Costimulation. Immunity. 2006; 25(5):783–793. 75. Geng D, Zheng L, Srivastava R, Asprodites N, Velasco-Gonzalez C, Davila E. When Tolllike receptor and T-cell receptor signals collide: a mechanism for enhanced CD8 T-cell effector function. Blood. 2010; 116(18):3494–3504. 76. Asprodites N, Zheng L, Geng D, Velasco-Gonzalez C, Sanchez-Perez L, Davila E. Engagement of Toll-like receptor-2 on cytotoxic T-lymphocytes occurs in vivo and augments antitumor activity. FASEB J. 2008; 22(10):3628–3637. 77. Yang Y, Huang C-T, Huang X, Pardoll DM. Persistent Toll-like receptor signals are required for reversal of regulatory T cell-mediated CD8 tolerance. Nat Immunol. 2004; 5(5):508–515. 78. Krebs P, Barnes MJ, Lampe K, Whitley K, Bahjat KS, Beutler B, et al. NK cell–mediated killing of target cells triggers robust antigen-specific T cell–mediated and humoral responses. Blood. 2009; 113(26):6593–6602. 79. Aldhamen YA, Seregin SS, Aylsworth CF, Godbehere S, Amalfitano A. Manipulation of EAT-2 expression promotes induction of multiple beneficial regulatory and effector functions of the human innate immune system as a novel immunomodulatory strategy. Int Immunol. 2014; 26(5):291–303. 80. Hermans IF, Silk JD, Gileadi U, Salio M, Mathew B, Ritter G, et al. NKT cells enhance CD4+ and CD8+ T cell responses to soluble antigen in vivo through direct interaction with dendritic cells. J Immunol. 2003; 171(10):5140–5147. 81. Cerundolo V, Silk JD, Masri SH, Salio M. Harnessing invariant NKT cells in vaccination strategies. Nat Rev Immunol. 2009; 9(1):28–38. 182 82. Kadowaki N, Ho S, Antonenko S, Malefyt RW, Kastelein RA, Bazan F, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001; 194(6):863–869. 83. Barton GM, Medzhitov R. Control of adaptive immune responses by Toll-like receptors. Curr Opin Immunol. 2002; 14(3):380–383. 84. Krug A, Towarowski A, Britsch S, Rothenfusser S, Hornung V, Bals R, et al. Toll-like receptor expression reveals CpG DNA as a unique microbial stimulus for plasmacytoid dendritic cells which synergizes with CD40 ligand to induce high amounts of IL-12. Eur J Immunol. 2001; 31(10):3026–3037. 85. Pulendran B, Li S, Nakaya HI. Systems Vaccinology. Immunity. 2010; 33(4):516–529. 86. Coffman RL, Sher A, Seder RA. Vaccine Adjuvants: Putting Innate Immunity to Work. Immunity. 2010; 33(4):492–503. 87. Nicholls EF, Madera L, Hancock REW. Immunomodulators as adjuvants for vaccines and antimicrobial therapy. Ann N Y Acad Sci. 2010; 1213:46–61. 88. Clements CJ, Griffiths E. The global impact of vaccines containing aluminium adjuvants. Vaccine. 2002; 20 Suppl 3:S24–33. 89. Marrack P, McKee AS, Munks MW. Towards an understanding of the adjuvant action of aluminium. Nat Rev Immunol. 2009; 9(4):287–293. 90. Kool M, Pétrilli V, Smedt T De, Rolaz A, Hammad H, Nimwegen M van, et al. Cutting Edge: Alum Adjuvant Stimulates Inflammatory Dendritic Cells through Activation of the NALP3 Inflammasome. J Immunol. 2008; 181(6):3755–3759. 91. Kool M, Soullié T, Nimwegen M van, Willart MAM, Muskens F, Jung S, et al. Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells. J Exp Med. 2008; 205(4):869–882. 92. Dubensky TW, Reed SG. Adjuvants for cancer vaccines. Semin Immunol. 2010; 22(3):155–161. 93. Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow S-N, Apter D, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a doubleblind, randomised study in young women. Lancet. 2009; 374(9686):301–314. 94. Ambrosch F, Wiedermann G, Kundi M, Leroux-Roels G, Desombere I, Garcon N, et al. A hepatitis B vaccine formulated with a novel adjuvant system. Vaccine. 2000; 18(20):2095–2101. 183 95. Jacques P, Moens G, Desombere I, Dewijngaert J, Leroux-Roels G, Wettendorff M, et al. The immunogenicity and reactogenicity profile of a candidate hepatitis B vaccine in an adult vaccine non-responder population. Vaccine. 2002; 20(31-32):3644–3649. 96. Didierlaurent AM, Morel S, Lockman L, Giannini SL, Bisteau M, Carlsen H, et al. AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J Immunol. 2009; 183(10):6186–6197. 97. Leroux-Roels I, Borkowski A, Vanwolleghem T, Dramé M, Clement F, Hons E, et al. Antigen sparing and cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet. 2007; 370(9587):580– 589. 98. Rouleau I, Serres G De, Drolet JP, Skowronski DM, Ouakki M, Toth E, et al. Increased risk of anaphylaxis following administration of 2009 AS03-adjuvanted monovalent pandemic A/H1N1 (H1N1pdm09) vaccine. Vaccine. 2013; 31(50):5989–5996. 99. Herran-Arita AK De la, Kornum BR, Mahlios J, Jiang W, Lin L, Hou T, et al. CD4+ T Cell Autoimmunity to Hypocretin/Orexin and Cross-Reactivity to a 2009 H1N1 Influenza A Epitope in Narcolepsy. Sci Transl Med. 2013; 5(216):216ra176–216ra176. 100. Wang R-F, Peng G, Wang HY. Regulatory T cells and Toll-like receptors in tumor immunity. Semin Immunol. 2006; 18(2):136–142. 101. Rharbaoui F, Drabner B, Borsutzky S, Winckler U, Morr M, Ensoli B, et al. The Mycoplasma-derived lipopeptide MALP-2 is a potent mucosal adjuvant. Eur J Immunol. 2002; 32(10):2857–2865. 102. Link C, Gavioli R, Ebensen T, Canella A, Reinhard E, Guzmán CA. The Toll-like receptor ligand MALP-2 stimulates dendritic cell maturation and modulates proteasome composition and activity. Eur J Immunol. 2004; 34(3):899–907. 103. Zaks K, Jordan M, Guth A, Sellins K, Kedl R, Izzo A, et al. Efficient immunization and cross-priming by vaccine adjuvants containing TLR3 or TLR9 agonists complexed to cationic liposomes. J Immunol. 2006; 176(12):7335–7345. 104. Honko AN, Sriranganathan N, Lees CJ, Mizel SB. Flagellin is an effective adjuvant for immunization against lethal respiratory challenge with Yersinia pestis. Infect Immun. 2006; 74(2):1113–1120. 105. Wille-Reece U, Flynn BJ, Loré K, Koup RA, Kedl RM, Mattapallil JJ, et al. HIV Gag protein conjugated to a Toll-like receptor 7/8 agonist improves the magnitude and quality of Th1 and CD8+ T cell responses in nonhuman primates. Proc Natl Acad Sci. 2005; 102(42):15190–15194. 106. Klinman DM. Immunotherapeutic uses of CpG oligodeoxynucleotides. Nat Rev Immunol. 2004; 4(4):249–258. 184 107. Kumar P, Tyagi R, Das G, Bhaskar S. Mycobacterium indicus pranii and Mycobacterium bovis BCG lead to differential macrophage activation in Toll-like receptor-dependent manner. Immunology. 2014; 143(2):258–268. 108. Wei MQ, Mengesha A, Good D, Anné J. Bacterial targeted tumour therapy-dawn of a new era. Cancer Lett. 2008; 259(1):16–27. 109. Caspi RR. Immunotherapy of autoimmunity and cancer: the penalty for success. Nat Rev Immunol. 2008; 8(12):970–976. 110. Russell WC. Adenoviruses: update on structure and function. J Gen Virol. 2009; 90(1):1– 20. 111. Saban SD, Silvestry M, Nemerow GR, Stewart PL. Visualization of alpha-helices in a 6angstrom resolution cryoelectron microscopy structure of adenovirus allows refinement of capsid protein assignments. J Virol. 2006; 80(24):12049–12059. 112. Rux JJ, Burnett RM. Adenovirus Structure. Hum Gene Ther. 2004; 15(12):1167–1176. 113. Cusack S. Adenovirus complex structures. Curr Opin Struct Biol. 2005; 15(2):237–243. 114. Roelvink PW. Identification of a Conserved Receptor-Binding Site on the Fiber Proteins of CAR-Recognizing Adenoviridae. Science. 1999; 286(5444):1568–1571. 115. McConnell MJ, Imperiale MJ. Biology of adenovirus and its use as a vector for gene therapy. Hum Gene Ther. 2004; 15(11):1022–1033. 116. Amalfitano A, Parks RJ. Separating fact from fiction: assessing the potential of modified adenovirus vectors for use in human gene therapy. Curr Gene Ther. 2002; 2(2):111–133. 117. Young CSH. The structure and function of the adenovirus major late promoter. Curr Top Microbiol Immunol. 2003; 272:213–249. 118. Leong K, Lee W, Berk AJ. High-level transcription from the adenovirus major late promoter requires downstream binding sites for late-phase-specific factors. J Virol. 1990; 64(1):51–60. 119. Sandkovsky U, Vargas L, Florescu DF. Adenovirus: current epidemiology and emerging approaches to prevention and treatment. Curr Infect Dis Rep. 2014; 16(8):416. 120. Russell KL, Broderick MP, Franklin SE, Blyn LB, Freed NE, Moradi E, et al. Transmission Dynamics and Prospective Environmental Sampling of Adenovirus in a Military Recruit Setting. J Infect Dis. 2006; 194(7):877–885. 121. Tatsis N, Ertl HCJ. Adenoviruses as Vaccine Vectors. Mol Ther. 2004; 10(4):616–629. 122. Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, et al. Insertional mutagenesis combined with acquired somatic mutations causes 185 leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest. 2008; 118(9):3143–3150. 123. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008; 118(9):3132–3142. 124. Philipson L, Pettersson RF. The coxsackie-adenovirus receptor--a new receptor in the immunoglobulin family involved in cell adhesion. Curr Top Microbiol Immunol. 2004; 273:87–111. 125. Mathias P, Wickham T, Moore M, Nemerow G. Multiple adenovirus serotypes use alpha v integrins for infection. J Virol. 1994; 68(10):6811–6814. 126. Zhang Y, Bergelson JM. Adenovirus Receptors. J Virol. 2005; 79(19):12125–12131. 127. Liu MA. Immunologic Basis of Vaccine Vectors. Immunity. 2010; 33(4):504–515. 128. Fonseca GJ, Thillainadesan G, Yousef AF, Ablack JN, Mossman KL, Torchia J, et al. Adenovirus Evasion of Interferon-Mediated Innate Immunity by Direct Antagonism of a Cellular Histone Posttranslational Modification. Cell Host Microbe. 2012; 11(6):597–606. 129. Graham FL, Smiley J, Russell WC, Nairn R. Characteristics of a Human Cell Line Transformed by DNA from Human Adenovirus Type 5. J Gen Virol. 1977; 36(1):59–72. 130. Everett R., Evans H., Hodges B., Ding E., Serra D., Amalfitano A. Strain-specific rate of shutdown of CMV enhancer activity in murine liver confirmed by use of persistent [E1−, E2b−] adenoviral vectors. Virology. 2004; 325(1):96–105. 131. Hu H, Serra D, Amalfitano A. Persistence of an [E1-, polymerase-] adenovirus vector despite transduction of a neoantigen into immune-competent mice. Hum Gene Ther. 1999; 10(3):355–364. 132. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao G, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab. 2003; 80(1-2):148–158. 133. Leppard KN. E4 gene function in adenovirus, adenovirus vector and adeno-associated virus infections. J Gen Virol. 1997; 78 ( Pt 9):2131–2138. 134. Gao GP, Yang Y, Wilson JM. Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy. J Virol. 1996; 70(12):8934–8943. 135. Gilbert R, Nalbantoglu J, Howell JM, Davies L, Fletcher S, Amalfitano A, et al. Dystrophin expression in muscle following gene transfer with a fully deleted (“gutted”) adenovirus is markedly improved by trans-acting adenoviral gene products. Hum Gene Ther. 2001; 12(14):1741–1755. 186 136. Parker E. Adenovirus DNA polymerase: domain organisation and interaction with preterminal protein. Nucleic Acids Res. 1998; 26(5):1240–1247. 137. Jong RN de. DNA binding properties of the adenovirus DNA replication priming protein pTP. Nucleic Acids Res. 2003; 31(12):3274–3286. 138. Everett RS, Hodges BL, Ding EY, Xu F, Serra D, Amalfitano A. Liver toxicities typically induced by first-generation adenoviral vectors can be reduced by use of E1, E2b-deleted adenoviral vectors. Hum Gene Ther. 2003; 14(18):1715–1726. 139. Kumar-Singh R, Chamberlain JS. Encapsidated adenovirus minichromosomes allow delivery and expression of a 14 kb dystrophin cDNA to muscle cells. Hum Mol Genet. 1996; 5(7):913–921. 140. Weaver EA, Nehete PN, Buchl SS, Senac JS, Palmer D, Ng P, et al. Comparison of replication-competent, first generation, and helper-dependent adenoviral vaccines. PloS One. 2009; 4(3):e5059. 141. Hartman ZC, Kiang A, Everett RS, Serra D, Yang XY, Clay TM, et al. Adenovirus Infection Triggers a Rapid, MyD88-Regulated Transcriptome Response Critical to AcutePhase and Adaptive Immune Responses In Vivo. J Virol. 2007; 81(4):1796–1812. 142. Appledorn DM, Patial S, McBride A, Godbehere S, Rooijen N Van, Parameswaran N, et al. 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. 2008; 181(3):2134–2144. 143. Appledorn DM, Patial S, Godbehere S, Parameswaran N, Amalfitano A. TRIF, and TRIFinteracting TLRs differentially modulate several adenovirus vector-induced immune responses. J Innate Immun. 2009; 1(4):376–388. 144. Morelli AE, Larregina AT, Ganster RW, Zahorchak AF, Plowey JM, Takayama T, et al. Recombinant adenovirus induces maturation of dendritic cells via an NF-kappaBdependent pathway. J Virol. 2000; 74(20):9617–9628. 145. Hartman ZC, Black EP, Amalfitano A. Adenoviral infection induces a multi-faceted innate cellular immune response that is mediated by the toll-like receptor pathway in A549 cells. Virology. 2007; 358(2):357–372. 146. Kiang A, Hartman ZC, Everett RS, Serra D, Jiang H, Frank MM, et al. Multiple innate inflammatory responses induced after systemic adenovirus vector delivery depend on a functional complement system. Mol Ther. 2006; 14(4):588–598. 147. Appledorn DM, McBride A, Seregin S, Scott JM, Schuldt N, Kiang A, et al. Complex interactions with several arms of the complement system dictate innate and humoral immunity to adenoviral vectors. Gene Ther. 2008; 15(24):1606–1617. 187 148. Hartman ZC, Appledorn DM, Amalfitano A. Adenovirus vector induced innate immune responses: Impact upon efficacy and toxicity in gene therapy and vaccine applications. Virus Res. 2008; 132(1–2):1–14. 149. Muruve DA, Barnes MJ, Stillman IE, Libermann TA. Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Hum Gene Ther. 1999; 10(6):965–976. 150. Higginbotham JN, Seth P, Blaese RM, Ramsey WJ. The release of inflammatory cytokines from human peripheral blood mononuclear cells in vitro following exposure to adenovirus variants and capsid. Hum Gene Ther. 2002; 13(1):129–141. 151. Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008; 452(7183):103–107. 152. Yamaguchi T, Kawabata K, Kouyama E, Ishii KJ, Katayama K, Suzuki T, et al. Induction of type I interferon by adenovirus-encoded small RNAs. Proc Natl Acad Sci. 2010; 107(40):17286–17291. 153. Minamitani T, Iwakiri D, Takada K. Adenovirus Virus-Associated RNAs Induce Type I Interferon Expression through a RIG-I-Mediated Pathway. J Virol. 2011; 85(8):4035– 4040. 154. Andersson MG, Haasnoot PCJ, Xu N, Berenjian S, Berkhout B, Akusjärvi G. Suppression of RNA interference by adenovirus virus-associated RNA. J Virol. 2005; 79(15):9556– 9565. 155. McKenna SA, Lindhout DA, Shimoike T, Aitken CE, Puglisi JD. Viral dsRNA Inhibitors Prevent Self-association and Autophosphorylation of PKR. J Mol Biol. 2007; 372(1):103– 113. 156. Launer-Felty K, Cole JL. Domain interactions in adenovirus VAI RNA mediate highaffinity PKR binding. J Mol Biol. 2014; 426(6):1285–1295. 157. Launer-Felty K, Wong CJ, Wahid AM, Conn GL, Cole JL. Magnesium-dependent interaction of PKR with adenovirus VAI. J Mol Biol. 2010; 402(4):638–644. 158. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003; 4(5):346–358. 159. Sun C, Zhang Y, Feng L, Pan W, Zhang M, Hong Z, et al. Epidemiology of adenovirus type 5 neutralizing antibodies in healthy people and AIDS patients in Guangzhou, southern China. Vaccine. 2011; 29(22):3837–3841. 160. Barouch DH, Kik SV, Weverling GJ, Dilan R, King SL, Maxfield LF, et al. International seroepidemiology of adenovirus serotypes 5, 26, 35, and 48 in pediatric and adult populations. Vaccine. 2011; 29(32):5203–5209. 188 161. Mast TC, Kierstead L, Gupta SB, Nikas AA, Kallas EG, Novitsky V, et al. International epidemiology of human pre-existing adenovirus (Ad) type-5, type-6, type-26 and type-36 neutralizing antibodies: Correlates of high Ad5 titers and implications for potential HIV vaccine trials. Vaccine. 2010; 28(4):950–957. 162. Sumida SM, Truitt DM, Lemckert AAC, Vogels R, Custers JHHV, Addo MM, et al. Neutralizing antibodies to adenovirus serotype 5 vaccine vectors are directed primarily against the adenovirus hexon protein. J Immunol. 2005; 174(11):7179–7185. 163. Roberts DM, Nanda A, Havenga MJE, Abbink P, Lynch DM, Ewald BA, et al. Hexonchimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature. 2006; 441(7090):239–243. 164. Bradley RR, Lynch DM, Iampietro MJ, Borducchi EN, Barouch DH. Adenovirus serotype 5 neutralizing antibodies target both hexon and fiber following vaccination and natural infection. J Virol. 2012; 86(1):625–629. 165. Sumida SM, Truitt DM, Kishko MG, Arthur JC, Jackson SS, Gorgone DA, et al. Neutralizing antibodies and CD8+ T lymphocytes both contribute to immunity to adenovirus serotype 5 vaccine vectors. J Virol. 2004; 78(6):2666–2673. 166. Joshi A, Tang J, Kuzma M, Wagner J, Mookerjee B, Filicko J, et al. Adenovirus DNA polymerase is recognized by human CD8+ T cells. J Gen Virol. 2009; 90(1):84–94. 167. McElrath MJ, Rosa SC De, Moodie Z, Dubey S, Kierstead L, Janes H, et al. HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case–cohort analysis. Lancet. 372(9653):1894–1905. 168. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet. 372(9653):1881–1893. 169. Sekaly R-P. The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? J Exp Med. 2008; 205(1):7–12. 170. Liu Q, Muruve DA. Molecular basis of the inflammatory response to adenovirus vectors. Gene Ther. 2003; 10(11):935–940. 171. Muruve DA. The innate immune response to adenovirus vectors. Hum Gene Ther. 2004; 15(12):1157–1166. 172. Seregin SS, Aldhamen YA, Appledorn DM, Hartman ZC, Schuldt NJ, Scott J, et al. Adenovirus capsid-display of the retro-oriented human complement inhibitor DAF reduces Ad vector–triggered immune responses in vitro and in vivo. Blood. 2010; 116(10):1669–1677. 189 173. Rodríguez A, Mintardjo R, Tax D, Gillissen G, Custers J, Pau MG, et al. Evaluation of a prime-boost vaccine schedule with distinct adenovirus vectors against malaria in rhesus monkeys. Vaccine. 2009; 27(44):6226–6233. 174. Tatsis N, Lasaro MO, Lin S-W, Haut LH, Xiang ZQ, Zhou D, et al. Adenovirus vectorinduced immune responses in nonhuman primates: responses to prime boost regimens. J Immunol. 2009; 182(10):6587–6599. 175. Seregin SS, Aldhamen YA, Appledorn DM, Zehnder J, Voss T, Godbehere S, et al. Use of DAF-displaying adenovirus vectors reduces induction of transgene- and vector-specific adaptive immune responses in mice. Hum Gene Ther. 2011; 22(9):1083–1094. 176. Barouch DH, Pau MG, Custers JHHV, Koudstaal W, Kostense S, Havenga MJE, et al. Immunogenicity of recombinant adenovirus serotype 35 vaccine in the presence of preexisting anti-Ad5 immunity. J Immunol. 2004; 172(10):6290–6297. 177. Seregin SS, Amalfitano A. Improving adenovirus based gene transfer: strategies to accomplish immune evasion. Viruses. 2010; 2(9):2013–2036. 178. Aldhamen YA, Seregin SS, Amalfitano A. Immune recognition of gene transfer vectors: focus on adenovirus as a paradigm. Front Immunol. 2011; 2:40. 179. Muruve DA, Cotter MJ, Zaiss AK, White LR, Liu Q, Chan T, et al. Helper-dependent adenovirus vectors elicit intact innate but attenuated adaptive host immune responses in vivo. J Virol. 2004; 78(11):5966–5972. 180. Hodges BL, Serra D, Hu H, Begy CA, Chamberlain JS, Amalfitano A. Multiply deleted [E1, polymerase-, and pTP-] adenovirus vector persists despite deletion of the preterminal protein. J Gene Med. 2000; 2(4):250–259. 181. Osada T, Yang XY, Hartman ZC, Glass O, Hodges BL, Niedzwiecki D, et al. Optimization of vaccine responses with an E1, E2b and E3-deleted Ad5 vector circumvents pre-existing anti-vector immunity. Cancer Gene Ther. 2009; 16(9):673–682. 182. Amalfitano A, Hauser MA, Hu H, Serra D, Begy CR, Chamberlain JS. Production and Characterization of Improved Adenovirus Vectors with the E1, E2b, and E3 Genes Deleted. J Virol. 1998; 72(2):926 –933. 183. Jones FR, Gabitzsch ES, Xu Y, Balint JP, Borisevich V, Smith J, et al. Prevention of influenza virus shedding and protection from lethal H1N1 challenge using a consensus 2009 H1N1 HA and NA adenovirus vector vaccine. Vaccine. 2011; 29(40):7020–7026. 184. Gabitzsch ES, Xu Y, Balcaitis S, Balint JP, Jones FR. An Ad5[E1-, E2b-]-HER2/neu vector induces immune responses and inhibits HER2/neu expressing tumor progression in Ad5 immune mice. Cancer Gene Ther. 2011; 18(5):326–335. 190 185. Gabitzsch ES, Xu Y, Balint JP Jr, Balcaitis S, Sanders-Beer B, Jones FR. Induction and comparison of SIV immunity in Ad5 naïve and Ad5 immune non-human primates using an Ad5 [E1-, E2b-] based vaccine. Vaccine. 2011; 29(45):8101–8107. 186. Morse MA, Chaudhry A, Gabitzsch ES, Hobeika AC, Osada T, Clay TM, et al. Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother. 2013; 62(8):1293–1301. 187. Appledorn DM, Aldhamen YA, Depas W, Seregin SS, Liu C-JJ, Schuldt N, et al. A new adenovirus based vaccine vector expressing an Eimeria tenella derived TLR agonist improves cellular immune responses to an antigenic target. PloS One. 2010; 5(3):e9579. 188. Appledorn DM, Aldhamen YA, Godbehere S, Seregin SS, Amalfitano A. Sublingual administration of an adenovirus serotype 5 (Ad5)-based vaccine confirms Toll-like receptor agonist activity in the oral cavity and elicits improved mucosal and systemic cellmediated responses against HIV antigens despite preexisting Ad5 immunity. Clin Vaccine Immunol. 2011; 18(1):150–160. 189. Aldhamen YA, Appledorn DM, Seregin SS, Liu CJ, Schuldt NJ, Godbehere S, et al. Expression of the SLAM Family of Receptors Adapter EAT-2 as a Novel Strategy for Enhancing Beneficial Immune Responses to Vaccine Antigens. J Immunol. 2011; 186(2):722–732. 190. Gabitzsch ES, Xu Y, Yoshida LH, Balint J, Gayle RB, Amalfitano A, et al. A preliminary and comparative evaluation of a novel Ad5 [E1-, E2b-] recombinant-based vaccine used to induce cell mediated immune responses. Immunol Lett. 2009; 122(1):44–51. 191. Schuldt NJ, Aldhamen YA, Appledorn DM, Seregin SS, Kousa Y, Godbehere S, et al. Vaccine platforms combining circumsporozoite protein and potent immune modulators, rEA or EAT-2, paradoxically result in opposing immune responses. PloS One. 2011; 6(8):e24147. 192. Shott JP, McGrath SM, Pau MG, Custers JHV, Ophorst O, Demoitié M-A, et al. Adenovirus 5 and 35 vectors expressing Plasmodium falciparum circumsporozoite surface protein elicit potent antigen-specific cellular IFN-gamma and antibody responses in mice. Vaccine. 2008; 26(23):2818–2823. 193. Schuldt NJ, Aldhamen YA, Godbehere-Roosa S, Seregin SS, Kousa YA, Amalfitano A. Immunogenicity when utilizing adenovirus serotype 4 and 5 vaccines expressing circumsporozoite protein in naïve and adenovirus (Ad5) immune mice. Malar J. 2012; 11:209. 194. Smaill F, Jeyanathan M, Smieja M, Medina MF, Thanthrige-Don N, Zganiacz A, et al. A human type 5 adenovirus-based tuberculosis vaccine induces robust T cell responses in humans despite preexisting anti-adenovirus immunity. Sci Transl Med. 2013; 5(205):205ra134. 191 195. Jeyanathan M, Damjanovic D, Shaler CR, Lai R, Wortzman M, Yin C, et al. Differentially imprinted innate immunity by mucosal boost vaccination determines antituberculosis immune protective outcomes, independent of T-cell immunity. Mucosal Immunol. 2013; 6(3):612–625. 196. Gabitzsch E, Xu Y, Balint J, Hartman Z, Lyerly HK, Jones F. Anti-tumor immunotherapy despite immunity to adenovirus using a novel adenoviral vector Ad5 [E1-, E2b-]-CEA. Cancer Immunol Immunother. 2010; 59(7):1131–1135. 197. O’Brien KL, Liu J, King SL, Sun Y-H, Schmitz JE, Lifton MA, et al. Adenovirus-specific immunity after immunization with an Ad5 HIV-1 vaccine candidate in humans. Nat Med. 2009; 15(8):873–875. 198. Frahm N, DeCamp AC, Friedrich DP, Carter DK, Defawe OD, Kublin JG, et al. Human adenovirus-specific T cells modulate HIV-specific T cell responses to an Ad5-vectored HIV-1 vaccine. J Clin Invest. 2012; 122(1):359–367. 199. Choi JH, Schafer SC, Zhang L, Juelich T, Freiberg AN, Croyle MA. Modeling PreExisting Immunity to Adenovirus in Rodents: Immunological Requirements for Successful Development of a Recombinant Adenovirus Serotype 5-Based Ebola Vaccine. Mol Pharm. 2013; 10(9):3342–3355. 200. Richardson JS, Abou MC, Tran KN, Kumar A, Sahai BM, Kobinger GP. Impact of Systemic or Mucosal Immunity to Adenovirus on Ad-Based Ebola Virus Vaccine Efficacy in Guinea Pigs. J Infect Dis. 2011; 204(suppl 3):S1032–S1042. 201. Thorner AR, Lemckert AAC, Goudsmit J, Lynch DM, Ewald BA, Denholtz M, et al. Immunogenicity of heterologous recombinant adenovirus prime-boost vaccine regimens is enhanced by circumventing vector cross-reactivity. J Virol. 2006; 80(24):12009–12016. 202. Haveman LM, Bierings M, Legger E, Klein MR, Jager W de, Otten HG, et al. Novel panDR-binding T cell epitopes of adenovirus induce pro-inflammatory cytokines and chemokines in healthy donors. Int Immunol. 2006; 18(11):1521–1529. 203. McCaffrey AP, Fawcett P, Nakai H, McCaffrey RL, Ehrhardt A, Pham T-TT, et al. The host response to adenovirus, helper-dependent adenovirus, and adeno-associated virus in mouse liver. Mol Ther J Am Soc Gene Ther. 2008; 16(5):931–941. 204. Hosack DA, Dennis G, Sherman BT, Lane HC, Lempicki RA. Identifying biological themes within lists of genes with EASE. Genome Biol. 2003; 4(10):R70. 205. He T-C, Zhou S, Costa LT da, Yu J, Kinzler KW, Vogelstein B. A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci. 1998; 95(5):2509–2514. 206. Ng P, Graham FL. Construction of first-generation adenoviral vectors. Methods Mol Med. 2002; 69:389–414. 192 207. Kiang A, Hartman ZC, Liao S, Xu F, Serra D, Palmer DJ, et al. Fully deleted adenovirus persistently expressing GAA accomplishes long-term skeletal muscle glycogen correction in tolerant and nontolerant GSD-II mice. Mol Ther. 2006; 13(1):127–134. 208. Cotter MJ, Zaiss AK, Muruve DA. Neutrophils Interact with Adenovirus Vectors via Fc Receptors and Complement Receptor 1. J Virol. 2005; 79(23):14622–14631. 209. Hodges BL, Evans HK, Everett RS, Ding EY, Serra D, Amalfitano A. Adenovirus Vectors with the 100K Gene Deleted and Their Potential for Multiple Gene Therapy Applications. J Virol. 2001; 75(13):5913–5920. 210. Pape KA, Khoruts A, Mondino A, Jenkins MK. Inflammatory cytokines enhance the in vivo clonal expansion and differentiation of antigen-activated CD4+ T cells. J Immunol. 1997; 159(2):591–598. 211. Steffensen MA, Jensen BAH, Holst PJ, Bassi MR, Christensen JP, Thomsen AR. Preexisting vector immunity does not prevent replication deficient adenovirus from inducing efficient CD8 T-cell memory and recall responses. PloS One. 2012; 7(4):e34884. 212. Schneider RJ, Safer B, Munemitsu SM, Samuel CE, Shenk T. Adenovirus VAI RNA prevents phosphorylation of the eukaryotic initiation factor 2 alpha subunit subsequent to infection. Proc Natl Acad Sci. 1985; 82(13):4321–4325. 213. Weber F, Wagner V, Kessler N, Haller O. Induction of interferon synthesis by the PKRinhibitory VA RNAs of adenoviruses. J Interferon Cytokine Res Off J Int Soc Interferon Cytokine Res. 2006; 26(1):1–7. 214. Sano M, Kato Y, Taira K. Sequence-specific interference by small RNAs derived from adenovirus VAI RNA. FEBS Lett. 2006; 580(6):1553–1564. 215. Aparicio O, Razquin N, Zaratiegui M, Narvaiza I, Fortes P. Adenovirus virus-associated RNA is processed to functional interfering RNAs involved in virus production. J Virol. 2006; 80(3):1376–1384. 216. Umbach JL, Kramer MF, Jurak I, Karnowski HW, Coen DM, Cullen BR. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature. 2008; 454(7205):780–783. 217. Sullivan CS, Grundhoff AT, Tevethia S, Pipas JM, Ganem D. SV40-encoded microRNAs regulate viral gene expression and reduce susceptibility to cytotoxic T cells. Nature. 2005; 435(7042):682–686. 218. Bremner KH, Scherer J, Yi J, Vershinin M, Gross SP, Vallee RB. Adenovirus Transport via Direct Interaction of Cytoplasmic Dynein with the Viral Capsid Hexon Subunit. Cell Host Microbe. 2009; 6(6):523–535. 219. Khare R, Reddy VS, Nemerow GR, Barry MA. Identification of adenovirus serotype 5 hexon regions that interact with scavenger receptors. J Virol. 2012; 86(4):2293–2301. 193 220. Ishii KJ, Akira S. Toll or toll-free adjuvant path toward the optimal vaccine development. J Clin Immunol. 2007; 27(4):363–371. 221. Wille-Reece U, Flynn BJ, Loré K, Koup RA, Miles AP, Saul A, et al. Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after primeboost immunization in nonhuman primates. J Exp Med. 2006; 203(5):1249–1258. 222. Toussi D, Massari P. Immune Adjuvant Effect of Molecularly-defined Toll-Like Receptor Ligands. Vaccines. 2014; 2(2):323–353. 223. Rosenberg B, Juckett DA, Aylsworth CF, Dimitrov NV, Ho S-C, Judge JW, et al. Protein from intestinal Eimeria protozoan stimulates IL-12 release from dendritic cells, exhibits antitumor properties in vivo and is correlated with low intestinal tumorigenicity. Int J Cancer. 2005; 114(5):756–765. 224. Hedhli D, Dimier-Poisson I, Judge JW, Rosenberg B, Mévélec MN. Protective immunity against Toxoplasma challenge in mice by coadministration of T. gondii antigens and Eimeria profilin-like protein as an adjuvant. Vaccine. 2009; 27(16):2274–2281. 225. Gowen BB, Smee DF, Wong M-H, Judge JW, Jung K-H, Bailey KW, et al. Recombinant Eimeria Protozoan Protein Elicits Resistance to Acute Phlebovirus Infection in Mice but Not Hamsters. Antimicrob Agents Chemother. 2006; 50(6):2023–2029. 226. Gowen BB, Judge JW, Wong M-H, Jung K-H, Aylsworth CF, Melby PC, et al. Immunoprophylaxis of Punta Toro virus (Phlebovirus, Bunyaviridae) infection in hamsters with recombinant Eimeria profilin-like antigen. Int Immunopharmacol. 2008; 8(8):1089– 1094. 227. Julander J, Judge J, Olsen A, Rosenberg B, Schafer K, Sidwell R. Prophylactic treatment with recombinant Eimeria protein, alone or in combination with an agonist cocktail, protects mice from Banzi virus infection. Antiviral Res. 2007; 75(1):14–19. 228. Rader JS, Aylsworth CF, Juckett DA, Mutch DG, Powell MA, Lippmann L, et al. Phase I Study and Preliminary Pharmacology of the Novel Innate Immune Modulator rBBX-01 in Gynecologic Cancers. Clin Cancer Res. 2008; 14(10):3089–3097. 229. Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL, Serhan CN, Hayden MS, et al. TLR11 Activation of Dendritic Cells by a Protozoan Profilin-Like Protein. Science. 2005; 308(5728):1626–1629. 230. Roach JC, Glusman G, Rowen L, Kaur A, Purcell MK, Smith KD, et al. The evolution of vertebrate Toll-like receptors. Proc Natl Acad Sci. 2005; 102(27):9577–9582. 231. Seregin SS, Aldhamen YA, Appledorn DM, Aylsworth CF, Godbehere S, Liu C-JJ, et al. TRIF is a critical negative regulator of TLR agonist mediated activation of dendritic cells in vivo. PloS One. 2011; 6(7):e22064. 194 232. Seregin SS, Appledorn DM, McBride AJ, Schuldt NJ, Aldhamen YA, Voss T, et al. Transient pretreatment with glucocorticoid ablates innate toxicity of systemically delivered adenoviral vectors without reducing efficacy. Mol Ther. 2009; 17(4):685–696. 233. Aldhamen YA, Seregin SS, Kousa YA, Rastall DPW, Appledorn DM, Godbehere S, et al. Improved cytotoxic T-lymphocyte immune responses to a tumor antigen by vaccines coexpressing the SLAM-associated adaptor EAT-2. Cancer Gene Ther. 2013; 20(10):564– 575. 234. Sen G, Khan AQ, Chen Q, Snapper CM. In vivo humoral immune responses to isolated pneumococcal polysaccharides are dependent on the presence of associated TLR ligands. J Immunol. 2005; 175(5):3084–3091. 235. Schnare M, Barton GM, Holt AC, Takeda K, Akira S, Medzhitov R. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001; 2(10):947–950. 236. Dyer WB, Zaunders JJ, Yuan FF, Wang B, Learmont JC, Geczy AF, et al. Mechanisms of HIV non-progression; robust and sustained CD4+ T-cell proliferative responses to p24 antigen correlate with control of viraemia and lack of disease progression after long-term transfusion-acquired HIV-1 infection. Retrovirology. 2008; 5:112. 237. Zak DE, Andersen-Nissen E, Peterson ER, Sato A, Hamilton MK, Borgerding J, et al. Merck Ad5/HIV induces broad innate immune activation that predicts CD8+ T-cell responses but is attenuated by preexisting Ad5 immunity. Proc Natl Acad Sci. 2012; 109(50):E3503–3512. 238. Watanabe H, Numata K, Ito T, Takagi K, Matsukawa A. Innate immune response in Th1and Th2-dominant mouse strains. Shock. 2004; 22(5):460–466. 239. Liu T, Matsuguchi T, Tsuboi N, Yajima T, Yoshikai Y. Differences in Expression of TollLike Receptors and Their Reactivities in Dendritic Cells in BALB/c and C57BL/6 Mice. Infect Immun. 2002; 70(12):6638–6645. 240. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. N Engl J Med. 2010; 363(5):411–422. 241. Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The Prioritization of Cancer Antigens: A National Cancer Institute Pilot Project for the Acceleration of Translational Research. Clin Cancer Res. 2009; 15(17):5323–5337. 242. Marshall JL, Gulley JL, Arlen PM, Beetham PK, Tsang K-Y, Slack R, et al. Phase I study of sequential vaccinations with fowlpox-CEA(6D)-TRICOM alone and sequentially with vaccinia-CEA(6D)-TRICOM, with and without granulocyte-macrophage colonystimulating factor, in patients with carcinoembryonic antigen-expressing carcinomas. J Clin Oncol. 2005; 23(4):720–731. 195 243. Bilusic M, Heery CR, Arlen PM, Rauckhorst M, Apelian D, Tsang KY, et al. Phase I trial of a recombinant yeast-CEA vaccine (GI-6207) in adults with metastatic CEA-expressing carcinoma. Cancer Immunol Immunother. 2014; 63(3):225–234. 244. Morse MA, Clay TM, Hobeika AC, Osada T, Khan S, Chui S, et al. Phase I study of immunization with dendritic cells modified with fowlpox encoding carcinoembryonic antigen and costimulatory molecules. Clin Cancer Res. 2005; 11(8):3017–3024. 245. Ullenhag GJ, Frödin J-E, Jeddi-Tehrani M, Strigård K, Eriksson E, Samanci A, et al. Durable carcinoembryonic antigen (CEA)-specific humoral and cellular immune responses in colorectal carcinoma patients vaccinated with recombinant CEA and granulocyte/macrophage colony-stimulating factor. Clin Cancer Res. 2004; 10(10):3273– 3281. 246. Grothey A, Cutsem E Van, Sobrero A, Siena S, Falcone A, Ychou M, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013; 381(9863):303–312. 247. Gerner MY, Casey KA, Mescher MF. Defective MHC class II presentation by dendritic cells limits CD4 T cell help for antitumor CD8 T cell responses. J Immunol. 2008; 181(1):155–164. 248. Stoitzner P, Green LK, Jung JY, Price KM, Atarea H, Kivell B, et al. Inefficient presentation of tumor-derived antigen by tumor-infiltrating dendritic cells. Cancer Immunol Immunother. 2008; 57(11):1665–1673. 249. Hartman ZC, Osada T, Glass O, Yang XY, Lei G, Lyerly HK, et al. Ligand-Independent Toll-like Receptor Signals Generated by Ectopic Overexpression of MyD88 Generate Local and Systemic Antitumor Immunity. Cancer Res. 2010; 70(18):7209–7220. 250. Aylsworth CF, Aldhamen YA, Seregin SS, Godbehere S, Amalfitano A. Activation of human natural killer cells by the novel innate immune modulator recombinant Eimeria antigen. Hum Immunol. 2013; 74(8):916–926. 251. Morse MA, Hobeika AC, Osada T, Berglund P, Hubby B, Negri S, et al. An alphavirus vector overcomes the presence of neutralizing antibodies and elevated numbers of Tregs to induce immune responses in humans with advanced cancer. J Clin Invest. 2010; 120(9):3234–3241. 252. Fialová L, Bartos A, Švarcová J, Malbohan I. Increased Intrathecal High-Avidity Anti-Tau Antibodies in Patients with Multiple Sclerosis. Villoslada P, editor. PLoS ONE. 2011; 6(11):e27476. 253. Zhu MZ, Marshall J, Cole D, Schlom J, Tsang KY. Specific cytolytic T-cell responses to human CEA from patients immunized with recombinant avipox-CEA vaccine. Clin Cancer Res. 2000; 6(1):24–33. 196 254. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan D-AN, Feldman SA, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011; 19(3):620–626. 255. Kawashima I, Hudson SJ, Tsai V, Southwood S, Takesako K, Appella E, et al. The multiepitope approach for immunotherapy for cancer: identification of several CTL epitopes from various tumor-associated antigens expressed on solid epithelial tumors. Hum Immunol. 1998; 59(1):1–14. 256. Lehmann F, Marchand M, Hainaut P, Pouillart P, Sastre X, Ikeda H, et al. Differences in the antigens recognized by cytolytic T cells on two successive metastases of a melanoma patient are consistent with immune selection. Eur J Immunol. 1995; 25(2):340–347. 257. Nukaya I, Yasumoto M, Iwasaki T, Ideno M, Sette A, Celis E, et al. Identification of HLA-A24 epitope peptides of carcinoembryonic antigen which induce tumor-reactive cytotoxic T lymphocyte. Int J Cancer. 1999; 80(1):92–97. 258. Ruiz M, Kobayashi H, Lasarte JJ, Prieto J, Borrás-Cuesta F, Celis E, et al. Identification and characterization of a T-helper peptide from carcinoembryonic antigen. Clin Cancer Res. 2004; 10(8):2860–2867. 259. Kobayashi H, Omiya R, Ruiz M, Huarte E, Sarobe P, Lasarte JJ, et al. Identification of an antigenic epitope for helper T lymphocytes from carcinoembryonic antigen. Clin Cancer Res. 2002; 8(10):3219–3225. 260. Kaufman HL, Lenz H-J, Marshall J, Singh D, Garett C, Cripps C, et al. Combination Chemotherapy and ALVAC-CEA/B7.1 Vaccine in Patients with Metastatic Colorectal Cancer. Clin Cancer Res. 2008; 14(15):4843–4849. 261. Yu Z, Theoret MR, Touloukian CE, Surman DR, Garman SC, Feigenbaum L, et al. Poor immunogenicity of a self/tumor antigen derives from peptide-MHC-I instability and is independent of tolerance. J Clin Invest. 2004; 114(4):551–559. 262. Barnett T, Goebel SJ, Nothdurft MA, Elting JJ. Carcinoembryonic antigen family: characterization of cDNAs coding for NCA and CEA and suggestion of nonrandom sequence variation in their conserved loop-domains. Genomics. 1988; 3(1):59–66. 263. Crosti M, Longhi R, Consogno G, Melloni G, Zannini P, Protti MP. Identification of novel subdominant epitopes on the carcinoembryonic antigen recognized by CD4+ T cells of lung cancer patients. J Immunol. 2006; 176(8):5093–5099. 264. Geynisman DM, Zha Y, Kunnavakkam R, Aklilu M, Catenacci DV, Polite BN, et al. A randomized pilot phase I study of modified carcinoembryonic antigen (CEA) peptide (CAP1-6D)/montanide/GM-CSF-vaccine in patients with pancreatic adenocarcinoma. J Immunother Cancer. 2013; 1(1):8. 265. Peguillet I, Milder M, Louis D, Vincent-Salomon A, Dorval T, Piperno-Neumann S, et al. High Numbers of Differentiated Effector CD4 T Cells Are Found in Patients with Cancer 197 and Correlate with Clinical Response after Neoadjuvant Therapy of Breast Cancer. Cancer Res. 2014; 74(8):2204–2216. 266. Salazar Gonzalez RM, Shehata H, O’Connell MJ, Yang Y, Moreno-Fernandez ME, Chougnet CA, et al. Toxoplasma gondii- Derived Profilin Triggers Human Toll-Like Receptor 5-Dependent Cytokine Production. J Innate Immun. 2014; 6(5):685–694. 267. Alexander C, Rietschel ET. Bacterial lipopolysaccharides and innate immunity. J Endotoxin Res. 2001; 7(3):167–202. 268. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013; 342(6161):967–970. 269. Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Science. 2013; 342(6161):971–976. 270. Saric T, Chang S-C, Hattori A, York IA, Markant S, Rock KL, et al. An IFN-gammainduced aminopeptidase in the ER, ERAP1, trims precursors to MHC class I-presented peptides. Nat Immunol. 2002; 3(12):1169–1176. 271. Australo-Anglo-American Spondyloarthritis Consortium (TASC), Reveille JD, Sims A-M, Danoy P, Evans DM, Leo P, et al. Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci. Nat Genet. 2010; 42(2):123–127. 272. Brown MA, Kennedy LG, MacGregor AJ, Darke C, Duncan E, Shatford JL, et al. Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and the environment. Arthritis Rheum. 1997; 40(10):1823–1828. 273. Khan MA. Epidemiology of HLA-B27 and Arthritis. Clin Rheumatol. 1996; 15 Suppl 1:10–12. 274. Evans DM, Spencer CCA, Pointon JJ, Su Z, Harvey D, Kochan G, et al. Interaction between ERAP1 and HLA-B27 in ankylosing spondylitis implicates peptide handling in the mechanism for HLA-B27 in disease susceptibility. Nat Genet. 2011; 43(8):761–767. 275. Mear JP, Schreiber KL, Münz C, Zhu X, Stevanović S, Rammensee HG, et al. Misfolding of HLA-B27 as a result of its B pocket suggests a novel mechanism for its role in susceptibility to spondyloarthropathies. J Immunol. 1999; 163(12):6665–6670. 276. Evnouchidou I, Kamal RP, Seregin SS, Goto Y, Tsujimoto M, Hattori A, et al. Cutting Edge: Coding single nucleotide polymorphisms of endoplasmic reticulum aminopeptidase 1 can affect antigenic peptide generation in vitro by influencing basic enzymatic properties of the enzyme. J Immunol. 2011; 186(4):1909–1913. 277. Colbert RA, DeLay ML, Klenk EI, Layh-Schmitt G. From HLA-B27 to spondyloarthritis: a journey through the ER. Immunol Rev. 2010; 233(1):181–202. 198 278. Bremander A, Petersson IF, Bergman S, Englund M. Population-based estimates of common comorbidities and cardiovascular disease in ankylosing spondylitis. Arthritis Care Res. 2011; 63(4):550–556. 279. Guerini FR, Cagliani R, Forni D, Agliardi C, Caputo D, Cassinotti A, et al. A functional variant in ERAP1 predisposes to multiple sclerosis. PloS One. 2012; 7(1):e29931. 280. Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consortium 2, Strange A, Capon F, Spencer CCA, Knight J, Weale ME, et al. A genomewide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010; 42(11):985–990. 281. Cui X, Hawari F, Alsaaty S, Lawrence M, Combs CA, Geng W, et al. Identification of ARTS-1 as a novel TNFR1-binding protein that promotes TNFR1 ectodomain shedding. J Clin Invest. 2002; 110(4):515–526. 282. Cui X, Rouhani FN, Hawari F, Levine SJ. An aminopeptidase, ARTS-1, is required for interleukin-6 receptor shedding. J Biol Chem. 2003; 278(31):28677–28685. 283. Goto Y, Ogawa K, Hattori A, Tsujimoto M. Secretion of endoplasmic reticulum aminopeptidase 1 is involved in the activation of macrophages induced by lipopolysaccharide and interferon-gamma. J Biol Chem. 2011; 286(24):21906–21914. 284. Goto Y, Ogawa K, Nakamura TJ, Hattori A, Tsujimoto M. TLR-mediated secretion of endoplasmic reticulum aminopeptidase 1 from macrophages. J Immunol. 2014; 192(9):4443–4452. 285. Chen R, Yao L, Meng T, Xu W. The association between seven ERAP1 polymorphisms and ankylosing spondylitis susceptibility: a meta-analysis involving 8,530 cases and 12,449 controls. Rheumatol Int. 2012; 32(4):909–914. 286. Lee YH, Choi SJ, Ji JD, Song GG. Associations between ERAP1 polymorphisms and ankylosing spondylitis susceptibility: a meta-analysis. Inflamm Res. 2011; 60(11):999– 1003. 287. Seregin SS, Rastall DPW, Evnouchidou I, Aylsworth CF, Quiroga D, Kamal RP, et al. Endoplasmic reticulum aminopeptidase-1 alleles associated with increased risk of ankylosing spondylitis reduce HLA-B27 mediated presentation of multiple antigens. Autoimmunity. 2013; 46(8):497–508. 288. York IA, Chang S-C, Saric T, Keys JA, Favreau JM, Goldberg AL, et al. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by trimming epitopes to 89 residues. Nat Immunol. 2002; 3(12):1177–1184. 289. York IA, Grant EP, Dahl AM, Rock KL. A mutant cell with a novel defect in MHC class I quality control. J Immunol. 2005; 174(11):6839–6846. 199 290. Hill A, Jugovic P, York I, Russ G, Bennink J, Yewdell J, et al. Herpes simplex virus turns off the TAP to evade host immunity. Nature. 1995; 375(6530):411–415. 291. Gotoh T, Oyadomari S, Mori K, Mori M. Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involving ATF6 and CHOP. J Biol Chem. 2002; 277(14):12343–12350. 292. Nakayama Y, Endo M, Tsukano H, Mori M, Oike Y, Gotoh T. Molecular mechanisms of the LPS-induced non-apoptotic ER stress-CHOP pathway. J Biochem. 2010; 147(4):471– 483. 293. Turner MJ, DeLay ML, Bai S, Klenk E, Colbert RA. HLA–B27 up-regulation causes accumulation of misfolded heavy chains and correlates with the magnitude of the unfolded protein response in transgenic rats: Implications for the pathogenesis of spondylarthritislike disease. Arthritis Rheum. 2007; 56(1):215–223. 294. Ke P-Y, Chen SS-L. Activation of the unfolded protein response and autophagy after hepatitis C virus infection suppresses innate antiviral immunity in vitro. J Clin Invest. 2011; 121(1):37–56. 295. Terenzi F, Hui DJ, Merrick WC, Sen GC. Distinct induction patterns and functions of two closely related interferon-inducible human genes, ISG54 and ISG56. J Biol Chem. 2006; 281(45):34064–34071. 296. Kim S, Lee S, Shin J, Kim Y, Evnouchidou I, Kim D, et al. Human cytomegalovirus microRNA miR-US4-1 inhibits CD8+ T cell responses by targeting the aminopeptidase ERAP1. Nat Immunol. 2011; 12(10):984–991. 297. Draenert R. Immune Selection for Altered Antigen Processing Leads to Cytotoxic T Lymphocyte Escape in Chronic HIV-1 Infection. J Exp Med. 2004; 199(7):905–915. 298. Cagliani R, Riva S, Biasin M, Fumagalli M, Pozzoli U, Caputo S Lo, et al. Genetic diversity at endoplasmic reticulum aminopeptidases is maintained by balancing selection and is associated with natural resistance to HIV-1 infection. Hum Mol Genet. 2010; 19(23):4705 –4714. 299. Rastall DPW, Aldhamen YA, Seregin SS, Godbehere S, Amalfitano A. ERAP1 functions override the intrinsic selection of specific antigens as immunodominant peptides, thereby altering the potency of antigen-specific cytolytic and effector memory T-cell responses. Int Immunol. 2014; . 300. Rutkowski DT, Kaufman RJ. A trip to the ER: coping with stress. Trends Cell Biol. 2004; 14(1):20–28. 301. Hoozemans JJM, Haastert ES van, Nijholt DAT, Rozemuller AJM, Eikelenboom P, Scheper W. The Unfolded Protein Response Is Activated in Pretangle Neurons in Alzheimer’s Disease Hippocampus. Am J Pathol. 2009; 174(4):1241–1251. 200 302. Holtz WA, O’Malley KL. Parkinsonian Mimetics Induce Aspects of Unfolded Protein Response in Death of Dopaminergic Neurons. J Biol Chem. 2003; 278(21):19367–19377. 303. Turner MJ, Sowders DP, DeLay ML, Mohapatra R, Bai S, Smith JA, et al. HLA-B27 misfolding in transgenic rats is associated with activation of the unfolded protein response. J Immuno. 2005; 175(4):2438–2448. 304. Hetz C, Chevet E, Harding HP. Targeting the unfolded protein response in disease. Nat Rev Drug Discov. 2013; 12(9):703–719. 305. Moser M, Leo O. Key concepts in immunology. Vaccine. 2010; 28:C2–C13. 306. Yarovinsky F, Kanzler H, Hieny S, Coffman RL, Sher A. Toll-like receptor recognition regulates immunodominance in an antimicrobial CD4+ T cell response. Immunity. 2006; 25(4):655–664. 307. Zhang D, Zhang G, Hayden MS, Greenblatt MB, Bussey C, Flavell RA, et al. A toll-like receptor that prevents infection by uropathogenic bacteria. Science. 2004; 303(5663):1522–1526. 308. Broz P, Monack DM. Newly described pattern recognition receptors team up against intracellular pathogens. Nat Rev Immunol. 2013; 13(8):551–565. 309. Hance KW, Zeytin HE, Greiner JW. Mouse models expressing human carcinoembryonic antigen (CEA) as a transgene: evaluation of CEA-based cancer vaccines. Mutat Res. 2005; 576(1-2):132–154. 310. Eades-Perner AM, Putten H van der, Hirth A, Thompson J, Neumaier M, Kleist S von, et al. Mice transgenic for the human carcinoembryonic antigen gene maintain its spatiotemporal expression pattern. Cancer Res. 1994; 54(15):4169–4176. 201