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

Similar documents
Recombinant Vaccinia Virus-Induced T-Cell Immunity: Quantitation of the Response to the Virus Vector and the Foreign Epitope

Viral Persistence Alters CD8 T-Cell Immunodominance and Tissue Distribution and Results in Distinct Stages of Functional Impairment

Memory CD4 T Cells Enhance Primary CD8 T-Cell Responses

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

SUPPLEMENTARY INFORMATION

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

Cationic Liposome-DNA Complexes as Immunomodulators and Vaccine Adjuvants

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

Signal 3 requirement for memory CD8 1 T-cell activation is determined by the infectious pathogen

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Therapeutic PD L1 and LAG 3 blockade rapidly clears established blood stage Plasmodium infection

Dendritic cell subsets and CD4 T cell immunity in Melanoma. Ben Wylie 1 st year PhD Candidate

Chapter 22: The Lymphatic System and Immunity

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Control of Virus-Specific CD8 T-Cell Exhaustion and Immune-Mediated Pathology by E3 Ubiquitin Ligase Cbl-b during Chronic Viral Infection

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands

Lineage relationship and protective immunity of memory CD8 T cell subsets

Critical Role for Alpha/Beta and Gamma Interferons in Persistence of Lymphocytic Choriomeningitis Virus by Clonal Exhaustion of Cytotoxic T Cells

TITLE: MODULATION OF T CELL TOLERANCE IN A MURINE MODEL FOR IMMUNOTHERAPY OF PROSTATIC ADENOCARCINOMA

Vaccine 22 (2004)

Memory NK cells during mousepox infection. Min Fang, Ph.D, Professor Institute of Microbiology, Chinese Academy of Science

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

Trends in vaccinology

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

Out-of-sequence signal 3 as a mechanism for virusinduced immune suppression of CD8 T cell responses

Supplemental Figure 1

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Dynamics of Memory T Cell Proliferation Under Conditions of Heterologous Immunity and Bystander Stimulation

T cell memory Friday, February 10, 17

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

SUPPLEMENTARY FIGURE 1

REVIEW Cell-mediated Immunity to Influenza Virus Infections: From the Perspective to the Vaccine Development against Highly Pathogenic Avian Influenza

Madhav V. Dhodapkar, Joseph Krasovsky, Ralph M. Steinman, and Nina Bhardwaj

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

Dendritic cells in cancer immunotherapy Aimin Jiang

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

Micro 204. Cytotoxic T Lymphocytes (CTL) Lewis Lanier

days days and gbt-i.cd Recipient 20

Lines of Defense. Immunology, Immune Response, and Immunological Testing. Immunology Terminology

Frequency, Specificity, and Sites of. Pulmonary Influenza Virus Infection. Christopher W. Lawrence, Rebecca M. Ream and Thomas J.

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

Received 19 March 2003/Accepted 28 May 2003

Shenghua Zhou, Rong Ou, Lei Huang, and Demetrius Moskophidis*

Naive and memory CD8 T cell responses after antigen stimulation in vivo

Supplementary Figure 1 Protease allergens induce IgE and IgG1 production. (a-c)

Direct quantitation of rapid elimination of viral antigen-positive lymphocytes by antiviral CD8 + T cells in vivo

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Supplementary Figure 1. Characterization of basophils after reconstitution of SCID mice

Medical Officer of Health Medical Director, Travel Health and Tropical Medicine Services Winnipeg Regional Health Authority

Anti-Gamma Interferon Antibodies Enhance the Immunogenicity of Recombinant Adenovirus Vectors

1. Overview of Adaptive Immunity

Human Effector and Memory CD8 + T Cell Responses to Smallpox and Yellow Fever Vaccines

NK cell flow cytometric assay In vivo DC viability and migration assay

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

CHAPTER-VII IMMUNOLOGY R.KAVITHA, M.PHARM, LECTURER, DEPARTMENT OF PHARMACEUTICS, SRM COLLEGE OF PHARMACY, SRM UNIVERSITY, KATTANKULATHUR.

Updated information and services can be found at:

Received 16 February 2004/Accepted 5 June 2004

Nature Immunology: doi: /ni Supplementary Figure 1. Gene expression profile of CD4 + T cells and CTL responses in Bcl6-deficient mice.

Therapeutic Cancer Vaccines

IMMUNOLOGICAL MEMORY. CD4 T Follicular Helper Cells. Memory CD8 T Cell Differentiation

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

B220 CD4 CD8. Figure 1. Confocal Image of Sensitized HLN. Representative image of a sensitized HLN

AGAINST VIRAL INFECTIONS. Identify the types of immunity involve in the mechanisms of protection against viral infections.

Estimating primate effector T cell responses to DNA vaccination

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 28.

Rapid Memory CD8 T-Lymphocyte Induction through Priming with Recombinant Mycobacterium smegmatis

Enumeration of Cytotoxic CD8 T Cells Ex Vivo during the Response to Listeria monocytogenes Infection

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

Herpes Simplex Virus Type 1-Specific Cytotoxic T-Lymphocyte Arming Occurs within Lymph Nodes Draining the Site of Cutaneous Infection

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York.

Kinetics of Primary and Memory Cytotoxic. T Lymphocyte Responses to Herpes. Simplex Virus 1 Infection: Granzyme B. Mediated CTL Activity

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Functionally Heterogeneous CD8 T-Cell Memory Is Induced by Sendai Virus Infection of Mice

Immunity to Viruses. Patricia Fitzgerald-Bocarsly September 25, 2008

Immunological memory.

Mucosal Immune System

CD40-Activated B Cells Can Efficiently Prime Antigen- Specific Naïve CD8 + T Cells to Generate Effector but Not Memory T cells

Third line of Defense

Immunodomination during Peripheral Vaccinia Virus Infection

Interferon regulatory factor-1 (IRF-1) shapes both innate and CD8+ T cell immune responses against West Nile virus infection

The origins of acquired immunity against

TITLE: A Potent Oncolytic Herpes Simplex Virus for the Therapy of Advanced Prostrate

Immunodominance in Virus-Induced CD8 T-Cell Responses Is Dramatically Modified by DNA Immunization and Is Regulated by Gamma Interferon

Immunity and Infection. Chapter 17

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.

I. Lines of Defense Pathogen: Table 1: Types of Immune Mechanisms. Table 2: Innate Immunity: First Lines of Defense

Supplementary Information:

Tumor Immunology: A Primer

Antiviral Cd8+ T Cell Responses in Neonatal Mice

The Immune System is the Third Line of Defense Against Infection. Components of Human Immune System

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

Pro5 MHC Pentamers. Dr. Jeremy Fry. Copyright ProImmune Limited All Rights Reserved

EBV Infection and Immunity. Andrew Hislop Institute for Cancer Studies University of Birmingham

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Upregulation of CD94/NKG2A receptors and Qa-1 b ligand during murine cytomegalovirus infection of salivary glands

Helminth worm, Schistosomiasis Trypanosomes, sleeping sickness Pneumocystis carinii. Ringworm fungus HIV Influenza

NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Supplemental Table I.

Transcription:

JOURNAL OF VIROLOGY, Dec. 2003, p. 13407 13411 Vol. 77, No. 24 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.24.13407 13411.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Detailed Analysis of the CD8 T-Cell Response following Adenovirus Vaccination Teng Chih Yang, Kelley Dayball, Yong Hong Wan, and Jonathan Bramson* Center for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada Received 9 May 2003/Accepted 28 August 2003 We examined CD8 T-cell expansion and function following intramuscular immunization with a recombinant adenovirus. This study has identified a number of properties which may explain the strong immunogenicity of adenovirus vectors: (i) the ability to deliver large amounts of antigen into the lymphoid tissues, (ii) the ability to induce rapid expansion and migration of CD8 T cells throughout the lymphatics, and (iii) the ability to produce a sustained, high-level CD8 T-cell response. Adenovirus (Ad) vectors have proven to be highly efficacious vaccine carriers in both murine and primate models (2, 8, 17 20). Although the ability of Ad vectors to elicit antigenspecific CD8 and CD4 T cells is well described, little is known about the kinetics or nature of the immune response following Ad immunization (12, 21, 22). The aim of the present study was to examine the kinetics of the antitransgene CD8 T-cell response following Ad immunization and characterize the relationship between CD8 T-cell activation and transgene expression. To this end, mice were immunized with an Ad expressing a model antigen, termed SIINFEKL-Luc, where the SIINFEKL epitope from chicken egg ovalbumin was added to the N terminus of firefly luciferase. This modified version of luciferase provides a sensitive measurement of gene expression in vivo along with a well-described CD8 T-cell epitope for tracking the CD8 T-cell response. The SIINFEKL-Luc cdna was inserted into the E1 region of an Ad with a deletion of E1 and E3 under the control of the murine cytomegalovirus promoter using a recently developed, high-efficiency system for virus rescue (16). Most reports of CD8 T-cell-dependent immunity produced by Ad vectors employed 51 Cr-release cytotoxicity assays. Unfortunately, these assays do not provide a quantitative measure of the CD8 T-cell population, and preferential outgrowth during the restimulation period may overestimate differences between two CD8 T-cell populations. We have employed K b /SIINFEKL tetramers (prepared at the Trudeau Institute, Sarnac Lake, N.Y.) to directly enumerate peptide-specific T cells as described in reference 7. Additionally, gamma interferon (IFN- ) production by antigen-specific CD8 T-cell populations was measured by stimulating lymphocytes directly ex vivo with SIINFEKL followed by intracellular staining for IFN- using the Cytofix/Cytoperm kit (BD Pharmingen) as described previously (10). Finally, cytotoxic function was monitored using the in vivo cytotoxic T-lymphocyte (CTL) assay, where naive lymphocytes were coated with either SIINFEKL (specific peptide) or KAVYNFATM (irrelevant peptide) and * Corresponding author. Mailing address: Department of Pathology and Molecular Medicine, McMaster University, Room HSC-4H21B, 1200 Main St. West, Hamilton, Ontario L8N 3Z5, Canada. Phone: (905) 525-9140. Fax: (905) 522 6750. E-mail: bramsonj@mcmaster.ca. labeled with 5 and 0.5 um CFSE (Molecular Probes), respectively, permitting the distinction between antigen-specific and antigen-nonspecific targets by flow cytometry. The two target cell populations were mixed at a 1:1 ratio and injected intravenously into mice. Four hours later, lymphocytes were isolated from lymphoid tissues and the presence of CFSE-labeled target cells was determined by flow cytometry. Specific lysis was determined using the calculation described by Coles et al. (6). An example of the results from these assays is provided in Fig. 1, and the results are summarized in Fig. 2 and 4. This research has complied with all relevant guidelines and institutional policies. In the following experiments, C57Bl/6 mice (Charles River Breeding Laboratories) were immunized with 10 8 PFU of Ad- SIINFEKL-Luc by intramuscular injection. Immune reactivity was measured within both the lymph nodes (LNs) and the spleen. The LNs were separated into the following groups based on their relative distance from the injection site: (i) local (popliteal and inguinal), (ii) regional (ileac), and (iii) distal (axial and brachial). The earliest evidence of CD8 T-cell activation came from the in vivo cytotoxicity assays. A 4-h assay period was chosen to minimize the migration of the effectors and target cells within the lymphatic system. Antigen-specific cytotoxicity was observed within all LNs that we investigated as early as 3 days following immunization (Fig. 2A). The greatest cytolytic activity was measured in the regional LN (30.5% 8.7%), followed by the local LN (13.3% 8.1%), distal LN (5.8% 3.0%), and spleen (4.2% 0.5%). Cytotoxic activity continued to increase in all sites over the course of the experiment, but the activity in the regional LN was always greatest. By day 6, the cytotoxic activity in the regional LN, local LN, and spleen was almost 100%, but the lytic activity in the distal LN at day 6 was only 75%. These results suggest that CTL develop initially within the local and distal LN, since the lytic activity in those sites at days 3 and 4 was two- to threefold greater than the lytic activity in the spleen and distal LN. However, we cannot exclude the possibility that differential migration of the target cells to these sites may influence the observed CTL activity. By 4 days following immunization, tetramer-positive CD8 cells were measurable in the regional LN and spleen (0.2% and 0.1%, respectively) (Fig. 2B). These results are somewhat dis- 13407

13408 NOTES J. VIROL. Downloaded from http://jvi.asm.org/ FIG. 1. Analysis of effector function within ileac lymph nodes at early times postimmunization. Cells were obtained from the regional lymph nodes 3 to 6 days following intramuscular immunization with 10 8 PFU of AdSIINFEKL-Luc. Upper panels, cells were stained directly with tetramer, and the number in the upper-right corner of each tetramer plot is the frequency of SIINFEKL-reactive CD8 T cells (determined using the gate shown)/total CD8 cells in the experimental group minus the frequency of SIINFEKL-reactive CD8 T cells/total CD8 cells in naïve controls; Middle panels, cells were immediately restimulated with SIINFEKL or KAVYNFATM peptide for intracellular cytokine staining, and the number in the upper-right corner is the frequency of IFN- -producing CD8 T cells (determined using the gate shown)/total CD8 following stimulation with SIINFEKL minus the frequency of IFN- -producing CD8 T cells/total CD8 cells following stimulation with KAVYNFATM; Lower panels, CFSE-labeled target cells were adoptively transferred into immunized recipient for in vivo CTL 4 h prior to harvesting tissues, and the number in the upper-right corner represents the percent specific lysis relative to naive controls. These histograms and density plots are representative of five mice, and the calculated values in all panels represent the means for the five mice. on April 11, 2018 by guest cordant with the findings of the in vivo cytotoxicity assay, since the specific lysis in the local LN was 48.4% 8.2% at day 4 compared to 19.2% 4.9% in the spleen, yet greater numbers of tetramer-positive cells were found in the spleen than in the local LN (Fig. 2A and B). While these results cannot be explained at this time, they may reflect a limitation in the tetramer-staining method when measuring low frequencies. By day 5, tetramer-positive cells were measurable in all sites, and the frequencies of tetramer-positive cells in the local LN and spleen (0.6% 0.2% and 0.9% 0.3%, respectively) were consistent with the levels of cytolytic activity (76.6% 9.2% and 73.3% 10.2%, respectively). The frequencies of tetramer-positive CD8 T cells within the local LN, regional LN, and spleen were comparable on days 5 and 6; however, as in the case of cytotoxic activity, the presence of tetramer-positive cells within the distal LN lagged behind that in the other nodes. The discrepancy between cytotoxicity and tetramer analyses likely reflects the difference in sensitivity between the two assays, where the in vivo cytotoxicity assay provides greater sensitivity. We interpret these data to suggest that antigenspecific CD8 T cells are first activated in the draining LNs and then migrate to other LNs and the spleen. These observations are consistent with those from previous studies of herpes simplex virus (HSV) infection, where the frequency of tetramer-positive cells within the draining LN and spleen expanded at similar rates but cytotoxicity was apparent in the LN

VOL. 77, 2003 NOTES 13409 FIG. 2. Analysis of effector function within lymph nodes and spleens at early times postimmunization. Cells were obtained from local, regional, and distal lymph nodes and spleens 3 to 6 days following intramuscular immunization with 10 8 PFU of AdSIINFEKL-Luc. On the day of each harvest, CFSE-labeled target cells were adoptively transferred into mice for the in vivo CTL assay, and tissues were harvested 4 h later. (A) In vivo CTL assay; (B) tetramer analysis with K b /SIINFEKL; (C) intracellular cytokine stain analysis for IFN- after 5hofpeptide stimulation ex vivo. Each histogram represents the mean standard error of the mean for five mice. before the spleen (6). Interestingly, antigen-specific CD8 T cells were not measurable in the distal LN by either assay following HSV infection, suggesting that the migration of CD8 T-cell effectors elicited by HSV may be different from the effectors elicited by Ad. Another notable difference between HSV and Ad is that the HSV doesn t appear to express antigen in the draining lymph nodes (11), whereas the Ad vectors do (5). IFN- -producing CD8 T cells were not found in any lymph nodes until 5 days after immunization, 24 h following the appearance of tetramer-positive CD8 T cells (Fig. 2C). This 24-hour delay likely reflects the time required for newly activated T cells to prepare their intracellular machinery for cytokine production. At day 6 following immunization, the fractions of IFN- -producing CD8 T cells within regional, local, and distal LN were 1.4% 0.3%, 1.1% 0.4%, and 0.6% 0.1%, respectively, comparable to the frequencies of tetramerpositive cells in the same sites on day 5 (1.2% 0.2%, 0.6% 0.2%, and 0.3% 0.2%, respectively) (Fig. 2B and C). By contrast, the frequency of IFN- -secreting CD8 T cells matched closely to the frequency of tetramer-positive CD8 T cells in the spleen at both days 5 and 6. At day 5, 0.9% 0.3% of CD8 T cells in the spleen were tetramer positive, and 1.5% 0.5% secreted IFN- in response to ex vivo stimulation. Likewise, at day 6, 2.3% 0.3% of the CD8 T cells in the spleen were tetramer positive, and 3.7% 0.7% secreted IFN- in response to ex vivo stimulation. These results suggest that the CD8 T-cell population in the spleen represents fully differentiated effectors, while the population in the lymph nodes represents CD8 T cells at earlier stages of differentiation. To determine the relationship between antigen expression and T-cell activation, muscle tissue, lymph nodes, and spleen were collected 24, 48, and 72 h following immunization and homogenized in cell culture lysis reagent (Promega). Luciferase activity was measured in cleared homogenates using the luciferase assay kit (Promega). As expected, the highest levels of total luciferase activity were found in the muscle tissue throughout the examination period (Fig. 3A). High levels of gene expression also were measured within the local and regional LN, although these levels were 10- to 50-fold lower than the levels in the muscle. When the luciferase activity in the local and regional LNs was normalized to the amount of protein in the homogenates (determined using the Bio-Rad DC protein assay) to account for differences in tissue size, the luciferase activity in the LN, in relation to the amount of tissue, was identical to that in the muscle (Fig. 3B). Little gene expression (less than 10-fold above background levels) was detectable in the distal nodes and the spleen. A recent publication demonstrated that the efficacy of plasmid DNA vaccines was increased by 100-fold over that of intramuscular injection when they were injected directly into the LN (14). Thus, the robust activity of the Ad vaccine may be related to the ability of this vector to directly deliver its payload into the LNs. To assess the durability of the CD8 T-cell response following Ad immunization, the frequency of antigen-specific CD8 T cells in the spleen was assessed over a period of 47 days. Tetramer-positive CD8 T cells accumulated rapidly in the spleen, reaching a maximum around 11 days postimmunization, where 20% of all CD8 T cells in the spleen were tetramer positive (Fig. 4). By day 14, only half of the peak number of tetramer-positive CD8 T cells remained in the

13410 NOTES J. VIROL. FIG. 3. Gene expression in vivo following intramuscular injection of AdSIINFEKL-Luc. Muscles, lymph nodes, and spleens were harvested 24 (open bars), 48 (grey bars), and 72 h (closed bars) following intramuscular injection with 10 8 PFU of AdSIINFEKL-Luc. Three groups of lymph nodes were harvested: local lymph nodes, regional lymph nodes, and distal lymph nodes. Luciferase activity was measured in tissue homogenates. (A) Luciferase activity per tissue. (B) Luciferase activity per tissue normalized to the amount of protein in the homogenate. Each histogram represents the mean [plusmn] standard error the mean for three to six samples. spleen, consistent with the rapid contraction of the CD8 T-cell response observed in other models. The rapid contraction phase was followed by a slower loss of cells, where 4.69% 0.65% of the CD8 T-cell population was tetramer positive at day 47 compared to 9.3% 1.4% at day 14. When IFN- production was employed to measure the frequency of SIINFEKL-reactive CD8 T cells in the spleen, the picture was slightly different. Similar to tetramer results, the frequency of IFN- -producing CD8 T cells in the spleen peaked around day 11. Unlike the tetramer-positive population, the IFN- -secreting population only contracted marginally from day 11 to day 14 (from 10.1% 1.4% to 9.3% 1.3%) and continued to wane slowly, leaving 3.4% 0.8% IFN- -secreting CD8 T cells at day 47 (Fig. 4). Additionally, FIG. 4. Expansion of SIINFEKL-specific CD8 T cells following immunization with AdSIINFEKL-Luc. Cells were obtained from spleens 3 to 47 days following intramuscular immunization with 10 8 PFU of AdSIINFEKL-Luc and stained directly with Kb/SIINFEKL tetramer and anti-cd8 (tetramer ve) or restimulated with SIINFEKL peptide for intracellular cytokine staining (IFN- ve). Each point represents the mean standard error of the mean for five to eight mice from two to three experiments. while the frequency of IFN- -secreting CD8 T cells was consistent with the frequency of tetramer-positive CD8 T cells throughout most of the analyses, at the peak of the response (days 9 and 11), the IFN- -secreting population was half of the tetramer-positive population (Fig. 4). The apparent discordance between IFN- and tetramer analyses at the peak time point may reflect either the presence of an antigen-specific, IFN- -negative population or an artifact of the tetramer staining. We are currently addressing this issue by investigating the expression of other cytokines (interleukin 2, interleukin 4, and tumor necrosis factor alpha) and combining intracellular cytokine staining techniques with tetramer staining. The kinetics of CD8 T-cell expansion and contraction observed following Ad immunization is somewhat distinct from observations using other infectious agents. Previous studies investigating a variety of agents (lymphocytic choriomeningitis virus, influenza, and Listeria monocytogenes) demonstrated that the CD8 T-cell response in the spleen peaked sharply around day 7 followed by a dramatic contraction that reduced the population size by 80 to 90% at day 21 (3, 4, 6, 9, 15). While the tetramer-positive population did exhibit an abrupt contraction phase, where 50% of the cells were lost between days 11 and 14, the IFN- -secreting population was much more stable and exhibited little change between days 11 (10.1%) and 21 (8.4%). Similar stability of the CD8 T-cell population measured by IFN- staining has been observed following HSV infection in the footpad (1, 13). Thus, the difference in contraction may simply reflect the impact of the immunization route, since the studies with lymphocytic choriomeningitis virus, influenza, and L. monocytogenes all employed the intraperitoneal route of delivery. We are currently investigating the impact of immunization route on the contraction of the CD8 T-cell response. These studies revealed a number of novel insights into the

VOL. 77, 2003 NOTES 13411 CD8 T-cell response following the introduction of recombinant Ad vectors and should provide a useful benchmark for individuals studying antitransgene immunity elicited following Ad injection. We thank Carole Evelegh for preparing the virus used in these experiments. This work was funded by the Canadian Institutes of Health Research (MOP-42433 to J.B.) and, in part, by grants from the Hamilton Health Sciences Corporation and St. Joseph s Hospital. J.B. is supported by an Rx & D Health Research Foundation/CIHR Career Award in Health Research. Y.H.W. is a CIHR New Investigator. T.C.Y. is the recipient of an Ontario Graduate Studentship. REFERENCES 1. Andersen, H., D. Dempsey, R. Chervenak, and S. R. Jennings. 2000. Expression of intracellular IFN-gamma in HSV-1-specific CD8 T cells identifies distinct responding subpopulations during the primary response to infection. J. Immunol. 165:2101 2107. 2. Babiuk, L. A., and S. K. Tikoo. 2000. Adenoviruses as vectors for delivering vaccines to mucosal surfaces. J. Biotechnol. 83:105 113. 3. Badovinac, V. P., B. B. Porter, and J. T. Harty. 2002. Programmed contraction of CD8( ) T cells after infection. Nat. Immunol. 3:619 626. 4. Bertram, E. M., A. Tafuri, A. Shahinian, V. S. Chan, L. Hunziker, M. Recher, P. S. Ohashi, T. W. Mak, and T. H. Watts. 2002. Role of ICOS versus CD28 in antiviral immunity. Eur. J. Immunol. 32:3376 3385. 5. Chan, S. Y., K. Li, J. R. Piccotti, M. C. Louie, T. A. Judge, L. A. Turka, E. J. Eichwald, and D. K. Bishop. 1999. Tissue-specific consequences of the antiadenoviral immune response: implications for cardiac transplants. Nat. Med. 5:1143 1149. 6. Coles, R. M., S. N. Mueller, W. R. Heath, F. R. Carbone, and A. G. Brooks. 2002. Progression of armed CTL from draining lymph node to spleen shortly after localized infection with herpes simplex virus 1. J. Immunol. 168:834 838. 7. Dobrzanski, M. J., J. B. Reome, and R. W. Dutton. 2001. Role of effector cell-derived IL-4, IL-5, and perforin in early and late stages of type 2 CD8 effector cell-mediated tumor rejection. J. Immunol. 167:424 434. 8. Gallichan, W. S., D. C. Johnson, F. L. Graham, and K. L. Rosenthal. 1993. Mucosal immunity and protection after intranasal immunization with recombinant adenovirus expressing herpes simplex virus glycoprotein B. J. Infect. Dis. 168:622 629. 9. Harrington, L. E., R. Most Rv, J. L. Whitton, and R. Ahmed. 2002. Recombinant vaccinia virus-induced T-cell immunity: quantitation of the response to the virus vector and the foreign epitope. J. Virol. 76:3329 3337. 10. Hassett, D. E., M. K. Slifka, J. Zhang, and J. L. Whitton. 2000. Direct ex vivo kinetic and phenotypic analyses of CD8 T-cell responses induced by DNA immunization. J. Virol. 74:8286 8291. 11. Jones, C. M., S. C. Cose, R. M. Coles, A. C. Winterhalter, A. G. Brooks, W. R. Heath, and F. R. Carbone. 2000. Herpes simplex virus type 1-specific cytotoxic T-lymphocyte arming occurs within lymph nodes draining the site of cutaneous infection. J. Virol. 74:2414 2419. 12. Jooss, K., H. C. Ertl, and J. M. Wilson. 1998. Cytotoxic T-lymphocyte target proteins and their major histocompatibility complex class I restriction in response to adenovirus vectors delivered to mouse liver. J. Virol. 72:2945 2954. 13. Kumaraguru, U., and B. T. Rouse. 2000. Application of the intracellular gamma interferon assay to recalculate the potency of CD8 T-cell responses to herpes simplex virus. J. Virol. 74:5709 5711. 14. Maloy, K. J., I. Erdmann, V. Basch, S. Sierro, T. A. Kramps, R. M. Zinkernagel, S. Oehen, and T. M. Kundig. 2001. Intralymphatic immunization enhances DNA vaccination. Proc. Natl. Acad. Sci. USA 98:3299 3303. 15. Murali-Krishna, K., J. D. Altman, M. Suresh, D. J. Sourdive, A. J. Zajac, J. D. Miller, J. Slansky, and R. Ahmed. 1998. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity 8:177 187. 16. Ng, P., R. J. Parks, D. T. Cummings, C. M. Evelegh, and F. L. Graham. 2000. An enhanced system for construction of adenoviral vectors by the twoplasmid rescue method. Hum. Gene Ther. 11:693 699. 17. Shi, Z., M. Zeng, G. Yang, F. Siegel, L. J. Cain, K. R. van Kampen, C. A. Elmets, and D. C. Tang. 2001. Protection against tetanus by needle-free inoculation of adenovirus-vectored nasal and epicutaneous vaccines. J. Virol. 75:11474 11482. 18. 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. 2002. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 415:331 335. 19. 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. 2003. Accelerated vaccination for Ebola virus haemorrhagic fever in non-human primates. Nature 424:681 684. 20. Sullivan, N. J., A. Sanchez, P. E. Rollin, Z. Y. Yang, and G. J. Nabel. 2000. Development of a preventive vaccine for Ebola virus infection in primates. Nature 408:605 609. 21. Tripathy, S. K., H. B. Black, E. Goldwasser, and J. M. Leiden. 1996. Immune responses to transgene-encoded proteins limit the stability of gene expression after injection of replication-defective adenovirus vectors. Nat Med. 2:545 550. 22. Yang, Y., Q. Su, and J. M. Wilson. 1996. Role of viral antigens in destructive cellular immune responses to adenovirus vector-transduced cells in mouse lungs. J. Virol. 70:7209 7212.