The effect of pre-existing immunity to antigens of adenovirus of the human serotype 5 on

Similar documents
Received 13 November 2006/Accepted 20 March 2007

The effect of adenovirus-specific antibodies on adenoviral vector induced, transgene product specific T cell responses

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study

Recombinant adeno-associated virus vectors induce functionally impaired transgene product specific CD8 + T cells in mice

Supporting Information

Received 22 April 2009/Accepted 19 June 2009

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 1.

Recombinant adeno-associated virus vectors induce functionally impaired transgene product specific CD8 + T cells in mice

ADCC Assay Protocol Vikram Srivastava 1, Zheng Yang 1, Ivan Fan Ngai Hung 2, Jianqing Xu 3, Bojian Zheng 3 and Mei- Yun Zhang 3*

Why are validated immunogenicity assays important for HIV vaccine development?

Peptide stimulation and Intracellular Cytokine Staining

Modulation of DNA Vaccine-Elicited CD8 T-Lymphocyte Epitope Immunodominance Hierarchies

HIV and Challenges of Vaccine Development

GOVX-B11: A Clade B HIV Vaccine for the Developed World

Primary Adult Naïve CD4+ CD45RA+ Cells. Prepared by: David Randolph at University of Alabama, Birmingham

Novel Heterologous Prime-Boost Vaccine Strategies for HIV. Dan Barouch April 18, 2012

Detailed Analysis of the CD8 T-Cell Response following Adenovirus Vaccination

Updated information and services can be found at:

Rapid antigen-specific T cell enrichment (Rapid ARTE)

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1.

Direct ex vivo characterization of human antigen-specific CD154 + CD4 + T cells Rapid antigen-reactive T cell enrichment (Rapid ARTE)

Development of a Universal T Cell Vaccine. Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood

Strategies for an HIV vaccine

MATERIALS. Peptide stimulation and Intracellular Cytokine Staining- EFFECTOR 45RA PANEL

Feb 11, Gene Therapy. Sam K.P. Kung Immunology Rm 417 Apotex Center

Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY. Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p.

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

Detailed step-by-step operating procedures for NK cell and CTL degranulation assays

Supplementary Figures

A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys

Received 31 March 2009/Returned for modification 12 June 2009/Accepted 5 July 2009

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity

Received 19 March 2003/Accepted 28 May 2003

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual)

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set

The humoral immune responses to IBV proteins.

SUPPLEMENTARY INFORMATION

Supplementary Materials for

Combinatorial Vaccines for AIDS and other Infectious Diseases

Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89.

Received 16 February 2004/Accepted 5 June 2004

HIV Anti-HIV Neutralizing Antibodies

Supporting Information

Human Immunodeficiency Virus type 1 (HIV-1) gp120 / Glycoprotein 120 ELISA Pair Set

Cell isolation. Spleen and lymph nodes (axillary, inguinal) were removed from mice

Characterization of a Single-Cycle Rabies Virus-Based Vaccine Vector

staining and flow cytometry

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center

Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses

Technical Resources. BD Immunocytometry Systems. FastImmune Intracellular Cytokine Staining Procedures

Alternate Antibody-Based Therapeutic Strategies To Purge the HIV Cell Reservoir

In vitro bactericidal assay Fig. S8 Gentamicin protection assay Phagocytosis assay

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

Gladstone Institutes, University of California (UCSF), San Francisco, USA

HIV-1 Virus-like Particle Budding Assay Nathan H Vande Burgt, Luis J Cocka * and Paul Bates

Gene Vaccine Dr. Sina Soleimani

MagniSort Mouse CD4 Naive T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Supplementary Data 1. Alanine substitutions and position variants of APNCYGNIPL. Applied in

Vaccines Based on Novel Adeno-Associated Virus Vectors Elicit Aberrant CD8 T-Cell Responses in Mice

<20 <20 <20 < <20 <20 <20 <20. Mock

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP

Choosing Between Lentivirus and Adeno-associated Virus For DNA Delivery

Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to

CONTENTS. STUDY DESIGN METHODS ELISA protocol for quantitation of mite (Dermatophagoides spp.) Der p 1 or Der f 1

Supporting Information

Mosaic vaccines elicit CD8 + T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys

Higher priming doses enhance HIV-specific humoral but not cellular. responses in a randomized, double-blind phase Ib clinical trial of

Viral vaccines. Lec. 3 أ.د.فائزة عبد هللا مخلص

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma

Third line of Defense

Constitutive Reporter Lentiviral Vectors Expressing Fluorescent Proteins

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies?

Optimizing Intracellular Flow Cytometry:

CHAPTER 4 IMMUNOLOGICAL TECHNIQUES

Antibody-Dependent Cell-Mediated Cytotoxicity in Simian Immunodeficiency Virus-Infected Rhesus Monkeys

Development of a VP6 subunit rotavirus vaccine A dual role of VP6 as a vaccine antigen and an adjuvant

Min Levine, Ph. D. Influenza Division US Centers for Disease Control and Prevention. June 18, 2015 NIBSC

New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS. Dublin, Ireland. June 24, AIDS Vaccines.

Harnessing (and Controlling) L. monocytogenes to Treat. and Prevent Human Disease. Thomas W. Dubensky, Jr., Ph.D. Cerus Corporation Concord, CA

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2*

MagniSort Mouse CD4 T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Midi Plant Genomic DNA Purification Kit

7/14/2014. Multiple immune effector mechanisms contribute to protection influenza. What is a correlate of protection?

J. D. Trujillo,* N. M. Kumpula-McWhirter, K. J. Hötzel, M. Gonzalez, and W. P. Cheevers

RAISON D ETRE OF THE IMMUNE SYSTEM:

La risposta immune all infezione da virus ebola. Chiara Agrati, PhD

PBMC from each patient were suspended in AIM V medium (Invitrogen) with 5% human

Studying Repeated Immunization in an Animal Model. Kanta Subbarao Laboratory of Infectious Diseases, NIAID

Supporting Online Material for

Challenges in Development and Validation of an Intracellular Cytokine Staining assay

Novel Vaccine Products for Planned Phase I Immunogenicity Studies in Infants

Protocol for Gene Transfection & Western Blotting

EVALUATION OF THE EFFECTIVENESS OF A 7% ACCELERATED HYDROGEN PEROXIDE-BASED FORMULATION AGAINST CANINE PARVOVIRUS

Supplemental Information. T Cells Enhance Autoimmunity by Restraining Regulatory T Cell Responses via an Interleukin-23-Dependent Mechanism

Preclinical Studies in Gene Delivery Biodistribution & Host Response Examples from AAV

In vitro human regulatory T cell expansion

Supplementary Material

Transcription:

JVI Accepts, published online ahead of print on April 00 J. Virol. doi:./jvi.0-0 Copyright 00, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. 1 1 1 1 1 1 1 1 1 0 1 The effect of pre-existing immunity to antigens of adenovirus of the human serotype on immune responses of nonhuman primates to vaccine regimens based on human or chimpanzee-derived adenovirus vectors running title: Immune responses of monkeys to chimpanzee adenoviral vectors McCoy, Kimberly 1, Tatsis, Nia 1, Korioth-Schmitz, Birgit, Lasaro, Marcio O 1, Hensley, Scott E 1, Lin, Shih-Wen 1, Li, Yan 1, Giles-Davis, Wynetta 1, Cun, Ann 1, Zhou, Dongming 1, Xiang, Zhiquan 1, Letvin, Norman L and Ertl, Hildegund CJ 1 * 1 Wistar Institute, Philadelphia, PA Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA These authors contributed equally abstract word count: paper word count: * Corresponding author: The Wistar Institute 01 Spruce St Philadelphia PA 1 Phone: 1-- Fax: 1--0 Email: ertl@wistar.upenn.edu Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 ABSTRACT In this study we compared a prime boost regimen with two serologically distinct replication-defective adenovirus (Ad) vectors derived from chimpanzee serotypes C and C1 expressing gag, pol, gp and nef of HIV-1 with a regimen in which replication-defective Ad vectors of the human serotype (AdHu) were given twice. Experiments were conducted in rhesus macaques that had or had not been pre-exposed to antigens of AdHu. There was no significant difference in T cell responses tested from peripheral blood of the different groups although responses were overall highest in non- pre-exposed animals immunized with the chimpanzee Ad vectors. Pre-existing immunity to AdHu completely inhibited induction of transgene product-specific antibodies by the AdHu vectors without affecting antibody responses to the chimpanzee vectors. Upon euthanasia, T cell responses were tested from a number of tissues. Pre-existing immunity to AdHu, commonly found in humans, changed the homing pattern of vaccine-induced T cells. In AdHu pre-exposed animals vaccinated with the chimpanzee Ad vectors frequencies of transgene-specific T cells were higher in spleens than in blood and in most pre-exposed animals vaccinated either with AdHu vectors or chimpanzee adenovirus vectors frequencies of such T cells were exceptionally high in livers. The latter results indicate that analysis of T cell responses solely from blood mononuclear cells of vaccine recipients may not suffice to compare the potency of different vaccine regimens. INTRODUCTION A multitude of different vaccine prototypes for HIV-1 have undergone early stage clinical trials and more are in pre-clinical testing (,,,, 1). Among those, E1-deleted adenoviral vectors of the human serotype (AdHu) are promising candidates that have been tested in phase II trials (1). E1- deleted adenoviral (Ad) recombinants of common human serotypes such as AdHu were initially Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 developed for gene therapy (, 1). Their advantages as gene transfer vehicles are numerous. They are well characterized, easy to grow, infect a wide range of cell types including resting cells, and induce high levels of transgene expression. Due to deletion of E1, which renders the virus replication- defective, the constructs are well tolerated unless given at excessive doses. The disadvantages of such constructs for permanent replacement of missing or faulty genes include the hosts vigorous immune responses against both the Ad antigens and the transgene products (, ). Furthermore pre-exposure to the homologous serotype of Ad, many of which infect nearly all humans during childhood, induces serotype-specific virus neutralizing antibodies (VNAs) (0), which interfere with successful delivery of the gene therapy vehicles (1,). The high immunogenicity of E1-deleted Ad recombinants of the common AdHu disallowed their successful use for sustained gene therapy but led to their development as vaccine carriers. E1- deleted AdHu vaccines were shown to induce unsurpassed B and CD + T cell responses in experimental animals, including rodents (1,, 1), canines () and primates (1,, ) and they are currently being tested in human clinical trials for antigens of HIV-1 by Merck. Ad vectors are being tested in additional clinical trials by the Vaccine Research Center (VRC) using DNA vaccine priming regimens followed by AdHu vector booster immunizations. However, the same problems that plagued gene therapists using E1-deleted AdHu recombinants may hamper the use of such constructs as vaccines in humans, especially in humans residing in developing countries. Most humans are pre-immune to common human serotypes of Ad such as serotypes,,, or 1. In the US, depending on the age of the study population and the sensitivity of the assay, 0 0% of humans carry readily detectable VNAs to AdHu virus (). Seroprevalence rates to AdHu virus are markedly higher in human populations from developing countries (). In animal models including nonhuman primates, VNAs generated upon pre-exposure Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 to AdHu virus have been shown previously to strongly impair the B and T cell responses to the transgene product of a vaccine based on an E1-deleted Ad of the same serotype (1,, 0). A similar impairment was observed in human volunteers that were tested by Merck in a clinical trial with their AdHu vaccine to HIV-1 (1). To circumvent impairment of vaccine efficacy by pre-existing neutralizing antibodies, we developed vaccine vectors based on Ads that had been isolated from chimpanzees. Vectors described here were derived from different viruses, i.e., AdC and AdC1 that represent two distinct serotypes (1,). AdC1 was constructed as a chimera with some of the early genes of AdC, a chimpanzee virus that is related to AdC, to allow for its growth on cell lines that provide the E1 of AdHu. AdC has close sequence homology with AdHu virus and thus belongs to subgroup E of adenovirideae. AdC1 virus does not bind the coxsackie adenovirus receptor (CAR) that is used by most human Ad serotypes, but attaches to CD (), which is used by the B subgroup of human Ad viruses such as AdHu and AdHu (, ). Here we tested prime boost regimens using a mixture of vectors expressing gag, pol, nef and gp of HIV-1 Clade B in small groups of AdHu pre-exposed and non-pre-exposed nonhuman primates (NHPs). We compared a heterologous prime boost regimen using AdC vectors followed by AdC1/C vectors to a homologous prime boost regimen in which AdHu vectors were used twice. The data show that pre-existing immunity to AdHu vectors alters homing of transgene productspecific T cells elicited by chimpanzee and human Ad vectors. Furthermore the data shows that preexposure to AdHu vectors abrogates the antibody responses to the transgene product expressed by AdHu but not by the chimpanzee Ad vectors. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 MATERIAL AND METHODS Production of Vectors. Ad vectors were derived from the human serotype (AdHu), chimpanzee serotypes (AdC) or 1 (AdC1). Vectors were E1- or E1- and E-deleted and generated from viral molecular clones by viral rescue on HEK cells. The AdC1 vector could not be propagated on HEK cells that provide the E1 of AdHu virus in trans. To allow for growth of this vector on HEK cells a chimeric vector was constructed between AdC1 (ATCC VR-0 ) and AdC (a chimpanzee adenovirus that is related to AdC, ATCC VR-1 ). In the chimeric AdC1/C vector map units 1- (left handed ITR) and 1- (right handed ITR) were replaced with sequences 1- and - of AdC virus. E1-deleted Ads were grown on HEK cells, purified by two rounds of buoyant density ultracentrifugation on CsCl gradients followed by column purification (Bio-Gel P-DG). Vectors were diluted in PBS supplemented with % glycerol and stored at 0 o C. Titration of Vectors. Content of virus particles (vps) was determined by spectrophotometry at 0 nm and 0 nm with the latter determining purity of the preparation. Quality Control of Vectors. Vector batches were checked for replication competent Ad (RCA) on A cells. None of the batches used had detectable contamination with RCA. Batches were tested for sterility and LPS contamination (limulus test). Vector Inserts. An AdHu vector expressing alpha-1-antitrypsin (A1AT) was used for preexposure of animals. The vaccine vectors were constructed to express a codon-optimized gag of HIV- 1 clade B (generous gift from G Pavlakis at the NIH/NCI, Fredrick, MD), a codon-humanized HIV-1 clade B gp (provided by G Nabel at the NIH/VRC, Bethesda, MD, USA), the end of polymerase of HIV-1 clade B encoding amino acids 1 0 and a fusion of HIV-1 nef (containing a TPA leader sequence and a GA mutation) and the end of polymerase (encoding the C-terminal amino Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 acids). Both HIV pol and TPAnef- pol were synthesized using human codons by Geneart (Regensburg, FRG). Vectors expressing the rabies virus glycoprotein (rab.gp) were used as controls. Vectors expressing enhanced green fluorescent protein (EGFP) were used for neutralization assays. A list of vectors and their pertinent features is shown in Table 1. Non-Human Primates. Two to three year-old Chinese origin Macaca mulatta were purchased from PrimateProducts Inc. and housed at the Richards Primate Facility at the University of PA. Sera were pre-screened for neutralizing antibodies to AdHu, AdC and AdC1 virus. Animals that were selected for the study did not have detectable titers of neutralizing antibodies to these viruses. Immunization of Non-Human Primates. The 1 animals were divided into groups (Table ). Animals of group and were pre-exposed to AdHu vector. These animals were injected with x virus particles (vp) of AdHu expressing A1AT. Animals were bled weeks later and neutralizing antibody titers to AdHu were determined. Thirty-four days after pre-exposure, animals were vaccinated. Animals of groups 1 and were immunized with AdC vectors expressing gag, gp, pol or TPAnef- pol. Animals of groups and were immunized with the corresponding AdHu vectors. Each vector was given at. x vp/animal, thus each NHP received a total of vp of vector. Vectors expressing HIVgag and HIV pol were inoculated into one leg and those expressing HIVTPAnef- pol and HIVgp into the opposite leg. The control animal of group was primed with a total of vp of AdCrabgp vector ( x vp per leg). NHPs were boosted days later with a second dose of vector. Animals of groups 1 and were boosted with AdC1/C vectors expressing gag, gp, pol or TPAnef- pol. Animals of groups and were boosted with the corresponding AdHu vectors. Vectors expressing HIVgag and HIV pol were inoculated into the left leg and HIVTPAnef- pol and HIVgp into the right leg. Again each vector was given at. x vp/animal. The control animal of group was boosted with vp of AdC1/Crab.gp vector. All Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 vaccinations were performed by injecting animals with vector diluted in 1 ml of saline into the right or left quadriceps muscles. Sample Collection. Animals were bled from the cephalic vein under light anesthesia. Blood was collected into Vacutainer cell collection tubes (CPT) with or without heparin (BD Biosciences). Upon euthanasia animals were exsanguinated. The peritoneal cavity was rinsed with 00 ml of Hank s Buffered Saline Solution (HBSS) (Cellgro) to isolate lymphoid cells. Tissues including lymph nodes, spleen, liver, peritoneal lavage were collected into HBSS. Isolation of Peripheral Blood Mononuclear Cells (PBMCs). Lymphocytes were collected in CPT tubes. The plasma layer was frozen at -0ºC. PBMCs were treated with ACK Lysis Buffer (Amersham) for minutes at room temperature and washed twice with HBSS and twice with RPMI complete media (RPMIc): RPMI- supplemented with % fetal bovine serum (FBS, Tissue Culture Biological), mm Hepes, Penicillin-Streptomycin and Gentamycin (Cellgro). Isolation of Lymphocytes from Tissues. Lymphocytes were isolated from peritoneal lavage by passing the lavage fluid through a 0 µm nylon filter and then washing cells x with HBSS and x with RPMIc. Cells were isolated from spleens and lymph nodes by dissecting the tissues into small sections and grinding them against a stainless steel mesh. Cells were filtered through a 0 µm nylon filter (BD Falcon) and washed. Liver lymphocytes were isolated by grinding the tissue in HBSS supplemented with % FBS. The resulting suspension was spun at 00 rpm for minutes at ºC. The supernatant was isolated and spun at 0 rpm for minutes. The resulting pellet was washed twice with DPBS plus % FBS. Lymphocytes were obtained using Percoll gradient centrifugation and washed twice with RPMI. Preservation of Lymphocytes. Cells were tested immediately after isolation by ELISPOT assays. Remaining cells were frozen in 0% FBS and % dimethyl sulfoxide (DMSO, Sigma) first at Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1-0 ºC and then cryopreserved in liquid nitrogen. When using frozen PBMCs, cells were thawed in a ºC water bath, diluted 1: in RPMI, and then washed twice with RMPI plus units/ml of RNAse free DNAse (Roche). Intracellular cytokine staining (ICS) was conducted on cells that had been frozen. Synthetic Peptides. HIV clade B consensus sequence gag, pol, nef and env peptides, 1mers overlapping by amino acids, were obtained from NIH Research and Reference Reagents Program and pooled into either or peptide pools for ICS and ELISPOT assays respectively. For ICS this resulted in pools for the gag peptides, pools for pol peptides, 1 pool for nef and pools for env (corresponding to gp). For ELISPOT assays 1 pool was used for gag, pools for pol, 1 pool for nef and pools for env (gp). Enzyme Linked Immunospot Assay (ELISpot). The assay was conducted as described (1). Briefly, PBMC were added to wells of MultiScreen-IP white plates (Millipore) at. x cells per well. For each tissue, pools were tested in triplicate (except where cell number was limiting). Each 1-mer peptide was used at a final concentration of µg/ml. Plates were incubated either with or without peptide for 1 1 hours at ºC, % CO. Spots were counted using the C.T.L. Series A Analyzer and ImmunoSpot. (Cellular Technology Ltd, Cleveland, OH). The minimum spot size was set to 0.001 mm, and the maximum spot size was set to 0.00 mm. Data has been summarized as spots per cells, so a dilution factor was used to multiply the raw values obtained from the ImmunoSpot count. There were three criteria for positive samples: 1) For every cells stimulated with peptides at least spots had to be detected, ) the number of spots in peptide stimulated wells had to be at least times the number of spots in unstimulated control wells, and ) the number of spots in peptide stimulated wells had to be standard deviations above the mean of Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 the control wells. Data shown on graphs represent values of peptide stimulated wells from which background values were subtracted. Intracellular Cytokine Staining (ICS). PBMCs ( cells) were stimulated in RPMI medium for h with αcd, αcdd, and Brefeldin A ( µg/ml each), with or without 1 µg/ml/peptide of the HIV- 1 peptide pools at ºC, % CO. After incubation, cells were stained with anti-human-cd PerCP- Cy. for 0 min at o C. After washing, fixing and permeabilization, cells were stained with anti- human-ifn-γ-apc, anti-human-il--pe and anti-monkey-cd-fitc for 0 min at o C. Cells were washed twice, resuspended in 1% formalin in PBS, and analyzed by FACS. Cells were acquired on a Cyan LX flow cytometer (DakoCytomation), and FACS data were analyzed by using Summit software (DakoCytomation). All antibodies were purchased from BD Bioscience except for anti-cd, which was purchased from Biosource. Titration of Antibodies to Gag. Nunc Immuno plates (MaxiSorb F, Denmark) were coated with 0 µl of a solution containing 1 µg of gag/ml in carbonate buffer (1 mm Na CO, mm NaHCO, ph.) overnight at o C. Plates were blocked overnight with PBS supplemented with % bovine serum albumin (BSA). Plates were washed with PBS and serial dilutions of monkey sera were added in borate buffer (0.1 M boric acid, mm sodium borate, mm NaCl, 0.0% v/v Tween-0) with % BSA and incubated at o C for hr. Wells were washed with PBS and 0 µl of a 1:00 dilution of alkaline-phosphatase-conjugated, goat anti-monkey IgG (Sigma Chemical Company, St. Louis, MO) was added and incubated at o C for hours. Wells were washed and incubated with 0 µl of substrate buffer containing p-nitrophenylphosphate disodium hexahydrate in diethanolamine. After 0 min at room temperature absorbance was read at 0 nm. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

Adenovirus Neutralization Assay. Heat-inactivated (0 min, o C) NHP plasma was tested on HEK cells for neutralization of Ads using vectors expressing EGFP in a plaque reduction assay starting with a 1:0 dilution of sample (0). Statistics. Significance was determined by one-tailed Student s t-tests and ANOVA analysis performed with Microsoft Excel. Significance was set at p 0.0. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 RESULTS Pre-screening, pre-exposure and immunization of animals. A trial was conducted in - year-old Chinese Macaca mulatta to assess the immunogenicity of replication-defective chimpanzee- origin Ad vectors in comparison to replication-defective AdHu vectors. NHPs were tested prior to purchase for VNAs to the Ad vectors. Sixteen animals that were negative were selected. One of those animals (R000) had antibodies that by ELISA bound to AdHu, AdC, and AdC1/C. Eight animals were immunized i.m. with x vp of an AdHuA1AT vector. Animals were bled weeks later and tested for VNA titers to AdHu virus. Animals had VNA titers ranging from 1:0 1:0. It should be pointed out that in human adults average titers to AdHu virus range from 1:0 1:00 using the assay system that was employed for studies presented here. Vaccination of NHPs. All of the animals were vaccinated weeks after injection of the AdHuA1AT vector (Table ). Each of the vaccines to HIV-1 was composed of an equal mixture of vectors expressing different sequences of HIV-1 clade B. Specifically, vectors expressed gag, gp, the part of pol and a fusion polypeptide composed of nef and the part of pol. Two groups of animals, i.e., groups 1 and were primed with vaccines based on the chimpanzee-origin AdC virus. These animals were boosted with the AdC1/C chimpanzee Ad vector expressing the same transgenes. The other two experimental groups were immunized twice with AdHu vectors expressing the same transgenes. This immunization schedule was chosen to allow for comparison with results from other research teams that have focused on the use of AdHu vectors and that have used such vectors in homologous prime boost regimens (1). The control animal was immunized with vps of chimpanzee Ad vectors expressing the rabies virus glycoprotein. All of the vectors prior to their use in NHPs were tested in vitro for expression of the transgene product (except for the vector expressing HIVTPAnefpol for which no suitable antibody was available). In addition, all of the Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 vectors (of the same batch that was used in NHPs) were tested in dose escalation experiments in mice to confirm their immunogenicity (data not shown). HIV-1 specific T cell responses in blood. PBMCs were collected twice before vaccination and tested for T cell responses to the different HIV-1 peptide pools by ELISPOT and ICS. Specific responses could not be detected by either assay. After each vaccine dose, animals were bled at different time points and frequencies of T cells were determined by an ELISPOT for IFN-γ (Figure 1) and by ICS for CD, CD, CD, IL- and IFN-γ (Figure ). By weeks after priming, / animals of group 1 that was not pre-exposed and received the AdC vectors responded and animals had frequencies above 00 spots/ PBMCs (Figure 1). In group, which received the AdC vectors after AdHu pre-exposure, / NHPs developed T cells frequencies that were comparable to those of group 1. In group, which received the AdHu vectors without pre-exposure, / animals were tested and only 1 developed a response; CD + T cell frequencies in this animal were below frequencies achieved by the high responders of the two groups of NHPs that were vaccinated with the AdC vectors. From group, which was vaccinated with AdHu after pre-exposure, / animals were tested and all of them had low but detectable frequencies of HIV antigen-specific IFN-γ producing cells. Animals were retested weeks after priming by ELISPOT assay. T cell responses could be detected by weeks after priming in / animals of groups 1 (AdC vaccinated), 1/ animals in group (AdHu pre-exposed, AdC vaccinated), / animals in group (AdHu vaccinated) and / animals in group (AdHu pre-exposed, AdHu vaccinated). Responses in 1/ animals of group and / animals of group, the groups that were vaccinated with the AdHu vectors, were barely above background by week ( spots for a given peptide pool). Approximately 1 days after priming one of the animals (R000) of group 1 (AdC vaccinated) died (see below for further details). The surviving NHPs were boosted and their PBMCs Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 were retested weeks later. All of the animals of groups 1 scored positive by ELISPOT after booster immunization but only some animals showed an increase in frequencies compared to peak frequencies after the 1st immunization. After the AdC1/C boost of AdC primed NHPs, frequencies were lower in the AdHu pre-exposed group as compared to non-pre-exposed animals of group 1, suggesting that pre-exposure to AdHu had a negative effect on the recall immune response to the chimpanzee vectors. Nevertheless, this difference was statistically not significant. It was of interest to note that animals of groups and, that had received two doses of the AdHu vectors developed mainly T cells to gag and pol and only animals of group 1 and to a lesser extend group developed strong T cell responses to peptides of env. Responses to nef peptides were low in all of the animals. PBMCs were retested 1 weeks later and then again at the time of necropsy. In animals that had been vaccinated with the chimpanzee Ad vectors 1/ animals of the non-pre-exposed group 1 showed a decrease in frequencies over time while frequencies remained stable in the other NHPs. In the AdHu pre-exposed group frequencies increased in all animals. Upon the homologous booster immunization with AdHu vectors frequencies decreased in / animals of the non-pre- exposed group and / animals in the AdHu pre-exposed group. In general, frequencies of specific T cells in blood did not decrease markedly between weeks and 1 after the boost. Results obtained by ICS (Figure and Table ) showed a similar although not identical pattern to those obtained by ELISPOT assays. Two weeks after priming, most animals had low frequencies ( 0.%) of CD + IFN-γ producing T cells and only 1 animal of group, which had been pre-exposed prior to vaccination with AdC vectors had frequencies > 0.% (Figure ). After booster immunization frequencies increased to above 0.% in all of the non-pre-exposed animals vaccinated with the chimpanzee Ad vectors (group 1) and in / animals that were pre-exposed prior to vaccination with the same vectors (group ). In AdHu vaccinated animals frequencies above 0.% Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 were seen in / of the non-pre-exposed animals (group ) and in / of the pre-exposed (group ). Frequencies declined by weeks after booster immunization and at the time of necropsy only 1 animals of each groups but for the group that was pre-exposed prior to vaccination with the chimpanzee Ad vectors retained frequencies > 0.%. As had been observed using ELISPOT assays, responses were mainly directed against gag and pol peptides and responses against env were mainly detected after priming in non-pre-exposed chimpanzee Ad vector vaccinated NHPs (not shown). Frequencies of CD + T cells producing IL- were lower than those of CD + T cells producing IFN-γ (Figure ). After the nd immunization with the heterologous chimpanzee Ad vector / of the non-pre-exposed animals (group 1) and / of the AdHu pre-exposed animals (group ) had CD + IL- + frequencies above 0.% while after AdHu vaccination none of the non-pre-exposed (group ) and / of the pre-exposed animals (group ) scored positive above 0.%. Frequencies of CD + T cells producing IFN-γ or IL- (Figure ) were low and only 1 or animals of each of the different groups developed detectable CD + T cell responses after priming or booster immunization. Overall, although non-pre-exposed NHPs vaccinated with the chimpanzee Ad vectors mounted the highest CD + T cell responses for IFN-γ and IL-, differences between the groups did not reach significance. T cell responses in different tissues. Animals were euthanized weeks after the initial priming and animals were euthanized approximately every second week. Animals were chosen randomly from the different groups. Lymphocytes were harvested from a number of compartments and tested by ELISPOT assays (Figure A, B). From most animals specific T cells could be isolated from the spleens. It was remarkable to notice that frequencies in spleens failed to correlate with frequencies in blood. For example, the mean frequencies of responses from PBMCs upon vaccination with the chimpanzee Ad vectors were higher in non-pre-exposed than in pre-exposed animals, Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 suggesting that pre-existing immunity affected the T cell response to the chimpanzee Ad vectors. In contrast, in splenocytes mean frequencies were higher in pre-exposed animals. Neither difference reached statistical significance. Non-pre-exposed animals vaccinated with the chimpanzee Ad vectors / and / showed detectable frequencies of cells producing IFN-γ in inguineal or iliac lymph nodes respectively. Such responses were seen in 1/ and / pre-exposed, chimpanzee Ad vector vaccinated animals. Upon AdHu vaccination 1/ and 1/ of the non-pre-exposed and / and / of the pre-exposed animals responded. Some animals showed responses in one but not the other set of lymph nodes. HIV-antigen-specific T cells could only be detected in / animals of the non-preexposed, chimpanzee Ad vector vaccinated group and in 1 animal of the other groups in peritoneal lavage (Figure A). Lymphocytes isolated from liver showed extraordinarily high frequencies of HIV specific IFN-γ producing T cells ranging from,000-,000 cells/ lymphocytes (Figure B). Frequencies were especially high in animals that had been pre-exposed to the AdHu A1AT vector, i.e., pre-exposed animals vaccinated either with the chimpanzee Ad vectors or with AdHu vectors showed in general higher frequencies than non-pre-exposed animals given either vaccine regimen. Two animals did not follow this pattern, i.e., one non-pre-exposed AdHu vaccinated animal (R001) had a very high response in the liver and one pre-exposed animal vaccinated with the chimpanzee Ad vectors (R0000) had only a low response in liver. Overall, T cell frequencies in liver were higher in pre-exposed animals upon AdHu vaccination than upon vaccination with the chimpanzee Ad vectors. The majority of cells isolated from liver responded to the nef and env peptide pools. Comparing the groups none of the responses in spleens, blood or lymph nodes tissues reached a statistically significant difference (p > 0.0) except for the response in liver comparing nonpre-exposed, chimpanzee Ad vector immunized animals with those that had been pre-exposed prior to vaccination with AdHu vectors (p = 0.0). Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 Serum antibody responses. Sera were tested before vaccination and the weeks before and weeks after booster immunization for antibodies to gag. After priming, non-pre-exposed animals vaccinated with the AdHu vectors showed the highest antibody titers; titers were low to undetectable in animals that were vaccinated with AdC or that had been pre-exposed to AdHu prior to vaccination with the AdHu vector (Figure ); after booster immunization / and / of the chimpanzee Ad vector vaccinated animals that had not or had been pre-exposed to AdHu showed a significant increase in titers and there was no significant difference in the antibody response between the two groups although mean titers of pre-exposed animals were lower than those of animals that had not been pre-exposed. In AdHu vaccinated animals that had not been pre-exposed / showed an increase in antibodies to gag after the boost while none of the animals that had been pre-exposed to AdHu developed antibodies to gag after immunizations with the AdHu vectors. Animals were tested for neutralizing antibodies to HIV-1 to SF1 and BAL (D. Montefiori, Duke University, Durham, NC). All samples (before and after boost) were negative. Animals were also tested for binding antibodies to gp. Only three of the animals responded, i.e., one of those that were pre- exposed to AdHu and then immunized with the chimpanzee Ad vectors (R0000), one of the AdHu immunized animals that had not been pre-exposed (R001) and one of the AdHu pre- exposed animals vaccinated with the AdHu vectors (R000). Responses were low and although there was no significant difference in the magnitude of antibody titers between the responders of the different groups, animals immunized with the chimpanzee Ad vectors developed detectable titers after one immunization while those immunized with the AdHu vectors only generated detectable titers after the nd immunization (data not shown). Animals were tested for neutralizing antibody titers to the Ad vectors used for the experiment and upon pre-exposure and immunization, animals developed antibodies to the Ad vectors (Table ). Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 Antibodies to AdHu were boosted in pre-exposed animals after priming and in all of the AdHu vaccinated animals after the boost with AdHu vectors. Immunization with AdC or AdC1/C vectors failed to increase antibody titers to AdHu, again confirming lack of cross-reactivity of neutralizing antibodies to these vectors. In addition, animals were analyzed after pre-exposure and after the first immunization for binding antibodies to the Ad vector by an ELISA (data not shown). In the AdHu pre- exposed groups / animals developed antibodies that cross-reacted between the Ad vectors; animals (R000 and R001) developed antibodies that only bound to AdHu. In the non-pre- exposed group, 1/ animals that had been vaccinated with AdC vector developed antibodies that also bound to AdHu and AdC1/C, the other two animals of this group only had antibodies that only bound to AdC. The control animal that was vaccinated only with an AdC vector expressing an irrelevant antigen showed a response that was specific for AdC. In the AdHu pre-exposed group, all animals upon AdC vaccination showed an increase in titers to AdC; animal R00 remained negative for antibodies to AdC1. In the non-pre-exposed group that was vaccinated with the AdHu vectors / animals developed antibodies that showed cross-reactive binding between AdHu, AdC and AdC1 while one animal (R00) had antibodies that bound to AdHu and AdC1 but not to AdC. The pattern of serum reactivity in the AdHu pre-exposed group did not change after vaccination with AdHu expressing HIV antigens. These data show that while there is a lack of neutralizing antibody cross-reactivity among the human- and chimpanzee-derived adenoviruses, binding antibodies cross-react extensively. Adverse reactions. During sample collection weeks after priming one of the AdC vaccinated animals (group 1) developed hypothermia and died the following day. Data for this animal are not shown in any of the figures. During anesthesia serum had been collected and the animal s glucose level was found to be very low (< mg/ml). A necropsy followed and histological analyses of Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 tissue sections did not reveal any tissue pathology that could be associated with the vaccine vectors. It was concluded that the death of the animal was not related to the vaccine but rather by anesthesiarelated stress. DISCUSSION We developed Ad vectors of chimpanzee origin to prevent a potential dampening effect of pre- existing VNAs to common human serotypes of Ad on the potency of Ad vector-based vaccines. We generated vectors from several different serotypes to allow for heterologous prime boost regimens. Here, were tested two chimpanzee Ad vectors, i.e., AdC, which belongs to subfamily E of adenovirideae, and AdC1/C, in which AdC1 that belongs to subfamily B encodes the structural viral antigens. Adenoviruses of the B subfamily use CD as the attachment receptor. This receptor is also used by the measles vaccine, which causes a transient immunosuppression and a shift towards Th responses (1, 1), neither of which have been observed upon immunization of animals with CD binding adenovirus vectors (, ). Experiments were conducted in non-human primates that had or had not been pre-exposed to antigens of AdHu. Additional animals were immunized twice with AdHu vectors. Animals were pre- exposed with a high dose of an AdHu vector expressing an irrelevant antigen. As in previously described studies, AdHu vectors were given i.m. (1,0). This route of immunization elicits strong VNA responses to AdHu and presumably T cells including CD + T cells to antigens of AdHu (). Animals were pre-exposed to AdHu virus rather than a chimpanzee adenovirus to mimic humans that commonly carry neutralizing antibodies to AdHu but show low prevalence rates of neutralizing antibodies to the chimpanzee adenoviruses (0). While neutralizing antibodies generated upon post- Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 exposure were highly specific to AdHu, binding antibodies showed extensive cross-reactivity between the different Ad vectors we used in this study (not shown). All of the chimpanzee Ad vectors we generated had been tested previously in comparison to AdHu vectors in naïve mice and in mice that had been pre-exposed to AdHu (,, ). The results showed that in naïve mice the AdC vector induced transgene product-specific CD + T cells that were comparable or, depending on the insert, slightly higher than those induced by AdHu vectors. AdC1/C vectors were less immunogenic. In AdHu pre-exposed animals CD + T cells induced by the chimpanzee Ad vectors were slightly reduced and this was caused by CD + T cells that cross-react between antigens of human and chimpanzee Ad vectors (). In contrast, CD + T cells responses to AdHu vectors were strongly diminished or even abolished in mice pre-exposed to antigens of AdHu. AdHu vectors induced higher antibody responses compared to chimpanzee Ad vectors in naïve mice, but again such responses were strongly reduced upon pre-exposure of animals to AdHu, which had no effect on the induction of transgene product-specific antibodies by the chimpanzee vectors (). In the NHP study described here, pre-exposure of rhesus macaques to antigens of AdHu had an apparent effect on induction of transgene product-specific CD + T cells by the chimpanzee Ad vectors when PBMCs were analyzed. Pre-exposed animals mounted a less vigorous CD + T cell response comparing PBMCs from non-pre-exposed animals to those that had been pre-exposed. Although this difference did not reach statistical significance due to the small number of animals, there was a clear trend that could be observed by ELISPOT assays (which fail to discriminate between T cell subsets) and by ICS. For example animal #R0000, which had the highest prevaccination antibody titers to AdHu (1:0) of this group developed the poorest T cell response, while animal #R00 which had low antibody titers to AdHu (1:0) prior to vaccination mounted Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 a more robust T cell response. Animal #R00, which had anti AdHu antibody titers of 1:0 prior to vaccination also failed to develop a strong T cell response in blood. Nevertheless, testing of lymphocytes from different lymphatic and non lymphatic tissues upon euthanasia showed that #R0000 had very high frequencies of HIV-1 antigen-specific T cell in its spleen (> 00 spots / splenocytes) and both #R0000 and #R00 had high frequencies of such T cells in liver. Overall, responses in spleen and liver were higher in / NHPs in the AdHu pre-exposed group than in non-pre-exposed NHPs vaccinated with the chimpanzee vectors suggesting that pre-exposure to AdHu vectors affected a change in the homing pattern of the responding T cells. T cell responses to the AdHu vectors given twice were indistinguishable in blood and spleens between NHPs that had or had not been pre-exposed to AdHu. In both groups, a second immunization with AdHu vectors increased frequencies of HIV-1 antigen-specific CD + T cells producing IFN-γ in some but not all of the animals. It should be pointed out that animal #R000, which had the highest titers of antibodies upon pre-exposure (1:0) had very low frequencies of specific T cells in its spleen at the time of euthanasia, indicating that in AdHu-vaccinated NHPs, unlike those vaccinated with the chimpanzee Ad vectors, pre-exposure to AdHu did not cause a shift in distribution of vaccine induced T cells towards the spleen. Pre-exposure resulted in a marked increase in T cells that homed to the liver in AdHu-vaccinated NHPs. The mechanism of the pronounced homing of T cells towards the liver in AdHu pre-immunized animals vaccinated either with AdHu vectors (/) or the chimpanzee Ad vectors (/) is unclear. Ad vectors readily infect hepatocytes, and some of the vectors may have entered the blood stream upon intramuscular application and reached the liver thus attracting and retaining a T cell infiltrate. Adenoviruses acquired by natural infections persist for years, mainly in T cells (). We showed recently that E1- deleted Ad vectors persist for more than one year mainly at the site of inoculation and in T cells Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 0

1 1 1 1 1 1 1 1 0 1 (manuscript submitted). In mice, Ad vector genome can be detected in liver of intramuscularly immunized mice transiently for a few days after vector inoculation and is then cleared. We tested liver sections of the NHPs harvested at the time of euthanasia for presence of vector genomes by a nested PCR. Although we could detect vector sequences in some of the samples, there was no correlation between their presence and the frequencies of transgene product-specific T cells in these livers suggesting expression of the transgene through persisting Ad vectors within the liver tissue was unlikely to have contributed to the recruitment or retention of the specific T cells (data not shown). Also we would like to point out that the PCR analyses were conducted using unfractionated liver sections, which does not allow us to distinguish between vector genomes present in liver cells or in infiltrating lymphocytes. Neutralizing or non-neutralizing antibodies directed to antigens of adenovirus may have further promoted targeting of Fc-receptor positive hepatocytes. One of the AdHu vaccinated animals that had not been pre-exposed to antigens of AdHu developed very high frequencies of T cells in the liver (> 0,000 spots / mononuclear cell). This animal developed high neutralizing antibody titers to AdHu after the first vaccine dose and the shifted migration of T cells to the liver could have been caused by these antibodies. Nevertheless, as there was no correlation between the presence of cross-reactive binding antibodies to the Ad vectors and frequencies and migration patterns of transgene product-specific T cells, retargeting of vectors through antibodies fails to explain our data. The AdHu vectors induced a more potent transgene product-specific antibody response than the chimpanzee Ad vectors reproducing previous findings in mice (). Nevertheless, such a gagspecific antibody response could not be elicited in AdHu pre-exposed animals even after a second application of the AdHu vector. Immunization with a single moderate dose of the AdC vector expressing gag failed to elicit detectable antibodies to gag. Such antibodies were induced in / of Downloaded from http://jvi.asm.org/ on May 1, 01 by guest 1

1 1 1 1 1 1 1 1 0 1 the non-pre-exposed NHPs and in / of the AdHu pre-exposed NHP after booster immunization with the AdC1/C vector. Levels of antibodies were slightly higher in the non-pre-exposed group, nevertheless, this difference failed to reach statistical significance. None of the animals developed HIV-1 neutralizing antibodies after immunization (data not shown, assays most kindly conducted by Dr. D. Montefiori, Duke University, Durham, NC). In summary, the presented study allows for three conclusions. First, pre-exposure to an Ad virus, which induces CD + T cells and non-neutralizing antibodies that widely cross-react with other Ad serotypes including chimpanzee Ads, affects the homing pattern of transgene product-specific T cells induced by a heterologous Ad vector. In pre-exposed animals vaccinated with heterologous Ad serotypes, T cells home preferentially to the spleen and fewer T cells circulate through blood. In clinical phase I/II trials the potency of a vaccine is established by testing PBMCs for T cells to the vaccine antigen. Data presented here indicate that results obtained with PBMCs may be misleading. Second, in NHPs, pre-existing immunity to antigens of Ad results in a pronounced enrichment of T cells induced by vaccination with homologous or heterologous Ad vectors towards the liver. This had not been observed in mice (unpublished observation) and the underlying mechanism remains elusive. Third, pre-existing immunity to antigens of AdHu virus completely inhibits the transgene product- specific antibody response to an AdHu vaccine vector. It is one of the major goals of ongoing research efforts to design env immunogens that are capable to elicit broadly cross-reactive neutralizing antibodies to HIV-1. Although AdHu vectors can induce very potent antibody responses, data presented here conclusively show that their efficacy as B cell-inducing vaccines will most likely be strongly impaired in humans with even moderate titers of pre-existing VNAs to AdHu virus. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 0 1 0 1 REFERENCES 1. Casimiro, D. R., L. Chen, T. M. Fu, R. K. Evans, M. J. Caulfield, M. E. Davies, A. Tang, M. Chen, L. Huang, V. Harris, D. C. Freed, K. A. Wilson, S. Dubey, D. M. Zhu, D. Nawrocki, H. Mach, R. Troutman, L. Isopi, D. Williams, W. Hurni, Z. Xu, J. G. Smith, S. Wang, X. Liu, L. Guan, R. Long, W. Trigona, G. J. Heidecker, H. C. Perry, N. Persaud, T. J. Toner, Q. Su, X. Liang, R. Youil, M. Chastain, A. J. Bett, D. B. Volkin, E. A. Emini, and J. W. Shiver. 00. Comparative immunogenicity in rhesus monkeys of DNA plasmid, recombinant vaccinia virus, and replication-defective adenovirus vectors expressing a human immunodeficiency virus type 1 gag gene. J Virol :0-1.. Cohen, P. 00. Immunity's yin and yang. A successful vaccine must first avoid being eliminated by pre-existing immunity before it can promote a protective immune response. IAVI Rep :1-.. Crystal, R. G. 1. Gene therapy strategies for pulmonary disease. Am J Med :S-S.. DiPaolo, N., S. Ni, A. Gaggar, R. Strauss, S. Tuve, Z. Y. Li, D. Stone, D. Shayakhmetov, N. Kiviat, P. Toure, S. Sow, B. Horvat, and A. Lieber. 00. Evaluation of adenovirus vectors containing serotype fibers for vaccination. Mol Ther 1:-.. Durier, C., O. Launay, V. Meiffredy, Y. Saidi, D. Salmon, Y. Levy, J. G. Guillet, G. Pialoux, and J. P. Aboulker. 00. Clinical safety of HIV lipopeptides used as vaccines in healthy volunteers and HIV-infected adults. Aids 0:-.. Fitzgerald, J. C., G. P. Gao, A. Reyes-Sandoval, G. N. Pavlakis, Z. Q. Xiang, A. P. Wlazlo, W. Giles-Davis, J. M. Wilson, and H. C. Ertl. 00. A simian replication-defective adenoviral recombinant vaccine to HIV-1 gag. J Immunol :-.. Flynn, N. M., D. N. Forthal, C. D. Harro, F. N. Judson, K. H. Mayer, and M. F. Para. 00. Placebo-controlled phase trial of a recombinant glycoprotein vaccine to prevent HIV-1 infection. J Infect Dis :-.. Gaggar, A., D. M. Shayakhmetov, and A. Lieber. 00. CD is a cellular receptor for group B adenoviruses. Nat Med :-1.. Garnett, C. T., D. Erdman, W. Xu, and L. R. Gooding. 00. Prevalence and quantitation of species C adenovirus DNA in human mucosal lymphocytes. J Virol :0-1.. Goepfert, P. A., H. Horton, M. J. McElrath, S. Gurunathan, G. Ferrari, G. D. Tomaras, D. C. Montefiori, M. Allen, Y. L. Chiu, P. Spearman, J. D. Fuchs, B. A. Koblin, W. A. Blattner, S. Frey, M. C. Keefer, L. R. Baden, and L. Corey. 00. High-dose recombinant Canarypox vaccine expressing HIV-1 protein, in seronegative human subjects. J Infect Dis 1:1-.. Goonetilleke, N., S. Moore, L. Dally, N. Winstone, I. Cebere, A. Mahmoud, S. Pinheiro, G. Gillespie, D. Brown, V. Loach, J. Roberts, A. Guimaraes-Walker, P. Hayes, K. Loughran, C. Smith, J. De Bont, C. Verlinde, D. Vooijs, C. Schmidt, M. Boaz, J. Gilmour, P. Fast, L. Dorrell, T. Hanke, and A. J. McMichael. 00. Induction of multifunctional human immunodeficiency virus type 1 (HIV-1)-specific T cells capable of proliferation in healthy subjects by using a prime-boost regimen of DNA- and modified vaccinia virus Ankara-vectored vaccines expressing HIV-1 Gag coupled to CD+ T-cell epitopes. J Virol 0:1-. 1. Hahm, B., N. Arbour, and M. B. Oldstone. 00. Measles virus interacts with human SLAM receptor on dendritic cells to cause immunosuppression. Virology :-0. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest

1 1 1 1 1 1 1 1 0 1 0 1 0 1 1. He, Z., A. P. Wlazlo, D. W. Kowalczyk, J. Cheng, Z. Q. Xiang, W. Giles-Davis, and H. C. Ertl. 000. Viral recombinant vaccines to the E and E antigens of HPV-1. Virology 0:1-1. 1. Kerdiles, Y. M., C. I. Sellin, J. Druelle, and B. Horvat. 00. Immunosuppression caused by measles virus: role of viral proteins. Rev Med Virol 1:-. 1. Kozarsky, K. F., and J. M. Wilson. 1. Gene therapy: adenovirus vectors. Curr Opin Genet Dev :-0. 1. Kresge, K. J. 00. Renewed promise. Annual AIDS vaccine meeting highlights recent data from clinical trials and lessons on recruitment and retention of volunteers. IAVI Rep :1-0. 1. Letvin, N. L., J. R. Mascola, Y. Sun, D. A. Gorgone, A. P. Buzby, L. Xu, Z. Y. Yang, B. Chakrabarti, S. S. Rao, J. E. Schmitz, D. C. Montefiori, B. R. Barker, F. L. Bookstein, and G. J. Nabel. 00. Preserved CD+ central memory T cells and survival in vaccinated SIVchallenged monkeys. Science 1:-. 1. Mulligan, M. J., N. D. Russell, C. Celum, J. Kahn, E. Noonan, D. C. Montefiori, G. Ferrari, K. J. Weinhold, J. M. Smith, R. R. Amara, and H. L. Robinson. 00. Excellent safety and tolerability of the human immunodeficiency virus type 1 pga/js plasmid DNA priming vector vaccine in HIV type 1 uninfected adults. AIDS Res Hum Retroviruses :-. 1. Reyes-Sandoval, A. Fitzgerald, J.C., Grant, R., Roy, S. Xiang, Z.Q. Li, Y., Gao, G.P., Wilson, J.M., and Ertl, H.C.J. 00. Human immunodeficiency virus type 1-specific immune responses in primates upon sequential immunization with adenoviral vaccine carriers of human and simian serotypes. J.Virol. : -. 0. Roberts, D. M., A. Nanda, M. J. Havenga, P. Abbink, D. M. Lynch, B. A. Ewald, J. Liu, A. R. Thorner, P. E. Swanson, D. A. Gorgone, M. A. Lifton, A. A. Lemckert, L. Holterman, B. Chen, A. Dilraj, A. Carville, K. G. Mansfield, J. Goudsmit, and D. H. Barouch. 00. Hexon-chimaeric adenovirus serotype vectors circumvent pre-existing anti-vector immunity. Nature 1:-. 1. Roy, S., G. Gao, D. S. Clawson, L. H. Vandenberghe, S. F. Farina, and J. M. Wilson. 00. Complete nucleotide sequences and genome organization of four chimpanzee adenoviruses. Virology :1-.. Roy, S., Y. Zhi, G. P. Kobinger, J. Figueredo, R. Calcedo, J. R. Miller, H. Feldmann, and J. M. Wilson. 00. Generation of an adenoviral vaccine vector based on simian adenovirus 1. J Gen Virol :-.. Segerman, A., J. P. Atkinson, M. Marttila, V. Dennerquist, G. Wadell, and N. Arnberg. 00. Adenovirus type uses CD as a cellular receptor. J Virol :1-1.. Shiver, J. W., T. M. Fu, L. Chen, D. R. Casimiro, M. E. Davies, R. K. Evans, Z. Q. Zhang, A. J. Simon, W. L. Trigona, S. A. Dubey, L. Huang, V. A. Harris, R. S. Long, X. Liang, L. Handt, W. A. Schleif, L. Zhu, D. C. Freed, N. V. Persaud, L. Guan, K. S. Punt, A. Tang, M. Chen, K. A. Wilson, K. B. Collins, G. J. Heidecker, V. R. Fernandez, H. C. Perry, J. G. Joyce, K. M. Grimm, J. C. Cook, P. M. Keller, D. S. Kresock, H. Mach, R. D. Troutman, L. A. Isopi, D. M. Williams, Z. Xu, K. E. Bohannon, D. B. Volkin, D. C. Montefiori, A. Miura, G. R. Krivulka, M. A. Lifton, M. J. Kuroda, J. E. Schmitz, N. L. Letvin, M. J. Caulfield, A. J. Bett, R. Youil, D. C. Kaslow, and E. A. Emini. 00. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 1:1-.. Sullivan, N. J., T. W. Geisbert, J. B. Geisbert, L. Xu, Z. Y. Yang, M. Roederer, R. A. Koup, P. B. Jahrling, and G. J. Nabel. 00. Accelerated vaccination for Ebola virus haemorrhagic fever in non-human primates. Nature :1-. Downloaded from http://jvi.asm.org/ on May 1, 01 by guest