SHORT COMMUNICATION. RAJIV KHANNA,*,2,3 SCOTT R. BURROWS,*,2 PATTY M. STEIGERWALD-MULLEN, SCOTT A. THOMSON,* MICHAEL G. KURILLA, and DENIS J.

Similar documents
RAJIV KHANNA, 1 * SCOTT R. BURROWS, 1 ANNE NEISIG, 2 JACQUES NEEFJES, 2 DENIS J. MOSS, 1 AND SHARON L. SILINS 1

Received 11 August 1997/Accepted 20 November 1997

Number of Patients Susceptibility Reference HLA-A* (278 EBV+) Increased incidence of. [1] EBV + chl 516 (192 EBV+) Increased incidence of

The Journal of Experimental Medicine

Received 24 July 2001/Accepted 12 November 2001

A-type and B-type Epstein Barr virus differ in their ability to spontaneously enter the lytic cycle

Decreased EBNA-1-specific CD8 T cells in patients with Epstein Barr virus-associated

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

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

Clinical implications of Epstein-Barr virus strain diversity

Yamaguchi University School of Medicine, Kogushi, Ube, Yamaguchi. growth in low serum, anchorage-independent growth in soft

Epstein-Barr virus, a -herpesvirus widespread in human

MATERIALS AND METHODS

EBV infection B cells and lymphomagenesis. Sridhar Chaganti

Definition of an Epitope on NS3 Recognized by Human CD4 Cytotoxic T Lymphocyte Clones Cross-Reactive for Dengue Virus Types 2, 3, and 4

Type 2 Cytokines Predominate in the Human CD4 T-Lymphocyte Response to Epstein-Barr Virus Nuclear Antigen 1

Recognition of a cervical cancer derived tumor cell line by a human papillomavirus type 16 E specific CD8 T cell clone

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16

Multi-Virus-Specific T cell Therapy for Patients after HSC and CB Transplant

Administration of Tumor-Specific Cytotoxic T Lymphocytes Engineered to Resist TGF-ß to Patients with EBV-Associated Lymphomas

Epstein Barr Virus Unit, Queensland Institute of Medical Research, The Bancroft Centre, 300 Herston Road, Brisbane, Queensland 4029, Australia

Novel Approach to the Formulation of an Epstein-Barr Virus Antigen-Based Nasopharyngeal Carcinoma Vaccine

Infection of autoreactive B lymphocytes with EBV, causing chronic autoimmune diseases

Translating EBV Immunology from Bench to Bedside: Somnium Animadverto A Dream Realized

Cellular & Molecular Immunology 229

Infection of Autoreactive B Lymphocytes with EBV, Causing Chronic Autoimmune Diseases

Human CD4 T Lymphocytes Consistently Respond to the Latent Epstein-Barr Virus Nuclear Antigen EBNA1

Human Cytomegalovirus Latency-Associated Proteins Elicit Immune-Suppressive IL-10 Producing CD4 + T Cells

1 Introduction. Rajiv Khanna 1, Sharon L. Silins 1, Zhiping Weng 2, David Gatchell 2, Scott R. Burrows 1 and Leanne Cooper 1

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Determining virological, serological and immunological parameters of EBV infection in the development of PTLD

Immunology Lecture 4. Clinical Relevance of the Immune System

Cover Page. The handle holds various files of this Leiden University dissertation.

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

Epstein- Barr Virus In Iraqi Patients With Nasopharangeal Carcinoma

RAISON D ETRE OF THE IMMUNE SYSTEM:

Human CD8 T Cell Responses to EBV EBNA1: HLA Class I Presentation of the (Gly-Ala) Containing Protein Requires Exogenous Processing

Antigen Presentation to T lymphocytes

Evolution of Two Types of Rhesus Lymphocryptovirus Similar to Type 1 and Type 2 Epstein-Barr Virus

Tumor Immunology. Tumor (latin) = swelling

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved.

Critical role of EBNA1-specific CD4 + T cells in the control of mouse Burkitt lymphoma in vivo

Restriction of Epstein-Barr Virus-Specific Cytotoxic T Cells by HLA-A,-B, and-c Molecules

Dr. Yi-chi M. Kong August 8, 2001 Benjamini. Ch. 19, Pgs Page 1 of 10 TRANSPLANTATION

Product Datasheet. HLA ABC Antibody (W6/32) NB Unit Size: 0.25 mg. Store at -20C. Avoid freeze-thaw cycles. Reviews: 1 Publications: 22

General information. Cell mediated immunity. 455 LSA, Tuesday 11 to noon. Anytime after class.

Direct killing of Epstein-Barr virus (EBV) infected B cells by CD4 T cells directed against the EBV lytic protein BHRF1

By Amy P. Sing, Richard F. Ambinder, Doley J. Hong, Michael Jensen, Wendy Batten, Effie Petersdorf, and Philip D. Greenberg

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

Conservation of Epstein-Barr Virus Cytotoxic T-Cell Epitopes in Posttransplant Lymphomas

C. Incorrect! MHC class I molecules are not involved in the process of bridging in ADCC.

EBV Infection. > Cellular Immune Response Profiling. > Humoral Immune Response Profiling EBV. ImmunoTools

Inhibition of Antigen Presentation by the Glycine/Alanine Repeat Domain Is Not Conserved in Simian Homologues of Epstein-Barr Virus Nuclear Antigen 1

Epstein-Barr virus and immunity

Expression of Epstein-Barr Virus Gene and Clonality of Infiltrated T Lymphocytes in Epstein-Barr Virus-associated Gastric Carcinoma

Cellular Pathology of immunological disorders

Immunotherapy in Treating Nasopharyngeal Carcinoma. Dora Kwong Department of Clinical Oncology Queen Mary Hospital The University of Hong Kong

Mucosal Immune System

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

K R N Baumforth, L S Young, K J Flavell, C Constandinou, P G Murray

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

Epstein-Barr Virus Polypeptides: Identification of Early Proteins and Their Synthesis and Glycosylation

Adaptive Immune System

Tumors arise from accumulated genetic mutations. Tumor Immunology (Cancer)

Developing Understanding of CMI. Dr Tom Wilkinson Associate Professor of Respiratory Medicine Faculty of Medicine University of Southampton UK

Role of De Novo Protein Synthesis in Target Cells Recognized by Cytotoxic T Lymphocytes Specific for Vesicular Stomatitis Virus

Identification of the Site of Epstein-Barr Virus Persistence In Vivo as a Resting B Cell

Vaccine Design: A Statisticans Overview

Downloaded by on April 28, Publication Date: April 24, 1984 doi: /bk

LAB 1, Immunology. Laboratory manual Immunology and Infection Biology Biomedicine course Autumn 2007

Minor H Antigen-Specific T Cells -The Black Box of the GVL Effect

Principles of Adaptive Immunity

1. Overview of Adaptive Immunity

Practical Solution: presentation to cytotoxic T cells. How dendritic cells present antigen. How dendritic cells present antigen

Defensive mechanisms include :

The Major Histocompatibility Complex (MHC)

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Third line of Defense

Epstein Barr virus (EBV) establishes a life-long, persistent,

CHAPTER 18: Immune System

NIH Public Access Author Manuscript Cytotherapy. Author manuscript; available in PMC 2012 May 1.

Persistent Infections

Supplemental Table 1. CD8 + and CD4 + T cell epitopes identified in EBV lytic and latent cycle proteins. CD8 + T cell epitopes.

Immune surveillance hypothesis (Macfarlane Burnet, 1950s)

Immunodeficiency. (2 of 2)

COURSE: Medical Microbiology, MBIM 650/720 - Fall TOPIC: Antigen Processing, MHC Restriction, & Role of Thymus Lecture 12

Citation Acta medica Nagasakiensia. 1991, 36

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

Jefferson Digital Commons

Third line of Defense. Topic 8 Specific Immunity (adaptive) (18) 3 rd Line = Prophylaxis via Immunization!

Mechanisms of antagonism of HIVspecific CD4+ T cell responses BSRI

The Innate Immune Response

Lecture 6. Burr BIO 4353/6345 HIV/AIDS. Tetramer staining of T cells (CTL s) Andrew McMichael seminar: Background

The present approach to this question draws upon information

Profiling HLA motifs by large scale peptide sequencing Agilent Innovators Tour David K. Crockett ARUP Laboratories February 10, 2009

Epstein-Barr Virus and the Somatic Hypermutation of Immunoglobulin Genes in Burkitt s Lymphoma Cells

Lecture 11. Immunology and disease: parasite antigenic diversity

7.012 Quiz 3 Answers

Transcription:

VIROLOGY 214, 633 637 (1995) SHORT COMMUNICATION Isolation of Cytotoxic T Lymphocytes from Healthy Seropositive Individuals Specific for Peptide Epitopes from Epstein Barr Virus Nuclear Antigen 1: Implications for Viral Persistence and Tumor Surveillance 1 RAJIV KHANNA,*,2,3 SCOTT R. BURROWS,*,2 PATTY M. STEIGERWALD-MULLEN, SCOTT A. THOMSON,* MICHAEL G. KURILLA, and DENIS J. MOSS* *Queensland Institute of Medical Research, The Bancroft Centre, 300 Herston Road, Brisbane, Australia 4029; and Department of Pathology and Microbiology, University of Virginia, Charlottesville, Virginia 22901 Received July 24, 1995; accepted October 11, 1995 The question of whether Epstein Barr nuclear antigen 1 (EBNA1) includes cytotoxic T lymphocyte (CTL) epitopes has generated considerable scientific interest, primarily due to its important implications for the overall biology of Epstein Barr virus (EBV). Earlier studies have suggested that EBV-associated malignancies that express only EBNA1 escape virus-specific immune surveillance since this antigen is not a target for CTL recognition. In the present report we have used a modified protocol to demonstrate that EBNA1 includes sequences which can be recognized by both polyclonal and clonal CTLs. CD4 / CTL clones were isolated from a healthy, seropositive donor that recognized the peptide epitope TSLYNLRRGTALA from EBNA1 in association with HLA DR1. Interestingly, these CTLs are unable to lyse EBV-infected B cells suggesting that EBNA1 may not be endogenously processed and/or presented to the host CTL response. Despite recent suggestions that glycine alanine repeat sequences within EBNA1 can inhibit endogenous processing, target cells infected with recombinant vaccinia vectors encoding truncated EBNA1 proteins, without these repeat sequences, were not recognized by this CTL clone. Thus it seems that the presence of glycine alanine repeats is not responsible for inhibiting the processing of the class II-restricted epitope defined in this study. These results substantiate the view that EBV-infected normal and malignant cells, where viral gene expression is limited to EBNA1, can resist CTL-mediated immune surveillance in vivo. 1995 Academic Press, Inc. Epstein Barr virus (EBV) is a member of the human herpesvirus family that maintains a life-long latent asso- ciation with B lymphocytes and a permissive association with stratified epithelium in the oropharynx (1, 2). Two types of EBV (1 and 2, also referred to as A and B) are recognized that show DNA sequence divergence within the BAM H1 WYH and E regions of the genome (3, 4). There is now convincing evidence that EBV-specific memory CD8 / and CD4 / cytotoxic T lymphocytes (CTL) are responsible for controlling the level of EBV-positive B lymphocytes which all healthy, seropositive individuals carry for life following primary infection with the virus (5 7). Previous studies have defined the relative distribution of CTL epitopes within EBV latent proteins recognized by T cells clones from healthy immune individuals expressing an array of MHC class I alleles (8 10). Using 1 A part of this work was presented at the CIBA Foundation Symposium on Vaccines against Virally Induced Cancers; March 15 17, 1994. 2 These two authors contributed equally in this work and their order should be considered arbitrary. 3 To whom reprint requests should be addressed. Fax: 61-7-362 0106; E-mail: rajivk@qimr.edu.au. recombinant vaccinia encoding each of the EBV nuclear antigens (EBNA) and latent membrane proteins (LMPs), numerous CTL epitopes were mapped within EBNA2-6 and LMP, while no CTL response was detected against EBNA1. The question of whether EBNA1 includes CTL epitopes has generated considerable scientific interest, primarily due to its important implications for the overall biology of EBV. First, it has been proposed that the EBV-associated tumor, Burkitt s lymphoma (BL), escapes virus-specific immune surveillance by restricting latent gene expression to EBNA1 (11). This view is reinforced by earlier studies which showed that BL patients retain detectable EBV-specific T cell surveillance, indicating that CTL dys- function is an unlikely cause of the outgrowth of these EBV-infected tumors in vivo (12). Second, it has recently been shown that EBV-infected B cells, expressing EBNA1 and LMP2a only, are capable of maintaining the latent viral infection in vivo (13). Third, an earlier study carried out by Trivedi and colleagues clearly showed that mammary carcinoma cells transfected with the EBNA1 gene were not rejected by histocompatible mice, whereas LMP-transfected cells were regularly rejected (14). Thus 633 0042-6822/95 $12.00 Copyright 1995 by Academic Press, Inc. All rights of reproduction in any form reserved.

634 SHORT COMMUNICATION ratio of 20:1). These polyclonal CTLs were tested against autologous phytohemagglutinin (PHA) blasts presensitized with an overlapping net of EBNA1 peptides (15-mer overlapping by 10 amino acids) as described earlier (15). Polyclonal CTLs from donors LC and DM, stimulated with autologous LCLs, recognized four different peptides from EBNA1. CTLs from donor LC showed reactivity against peptide 49 (RRGTALAIPQCRLTP), while CTLs from donor DM recognized autologous PHA blasts in the presence of peptides 30 (EPDVPPGAIEQGPAD), 47 (GGSKTSLYNLRRGTA), and 48 (SLYNLRRGTALAIPQ) (Figs. 1a and 1b). In contrast, CTLs from donors CM and NB showed no significant reactivity against any of the EBNA1 peptides (data not shown). These experiments were repeated with two different blood samples and consistently the same set of peptides was recognized by CTLs from donors DM and LC. To confirm the data obtained with polyclonal effectors, CTL clones were generated (16) from donors DM and LC by seeding UM cells or purified CD4 / T cells, after stimulation with either autologous LCLs or EBNA1 peptides (peptides 30, 47, and 48 for donor DM and peptide 49 for donor LC). CD4 / T cells were purified from fresh UM cells using a Lymphokwik reagent (One Lambda Inc., CA). Colonies were harvested after 3 days and amplified in culture with biweekly restimulation with ril-2 and autologous LCL precoated with EBNA1 peptide(s). Two CTL clones were isolated from donor DM (referred to as DM2 and DM6) following stimulation with peptide 48 (SLYNLR- FIG. 1. Identification of CTLs specific for EBNA1 peptides in healthy RGTALAIPQ). Clone DM2 was isolated from UM cells, seropositive individuals. Polyclonal CTLs from donor DM (a) and LC while DM6 was isolated using purified CD4 / T cells. (b) were used as effectors to test for reactivity against autologous These CTL clones showed strong T cell T cell killing PHA blasts presensitized with overlapping EBNA1 peptides. Results are expressed as percentage specific lysis observed in a standard 5- (17) in the presence of peptides GGSKTSLYNLRRGTA hr chromium-release assay. An effector to target ratio of 20:1 was used in the assay. it is plausible that some virus-infected B cells maintain a nonimmunogenic phenotype by down-regulating the critical latent proteins needed for CTL recognition. The present study was specifically designed to consider the possibility of detecting an EBNA1-specific CTL response in healthy EBV-seropositive individuals, since earlier studies have essentially drawn their conclusions on the basis of negative evidence. Unfractionated mononuclear (UM) cells (10 6 /ml) from EBV-seropositive donors LC (HLA A1, B8, B18, DR3, DR11), NB (A2, A24, B7, B35, DR11), CM (HLA A11, A24, B7, B44, DR1, DR4), and DM (A24, A29, B44, Bw47, DR1, DR7) were cultivated with irradiated (8000 R) autologous lymphoblastoid cell lines (LCLs) transformed with the type 1 EBV isolate, B95.8 (responder to stimulator ratio of 50:1) in 2-ml culture wells (Linbro) in RPMI 1640/10% FCS. After 7 days, cultures were restimulated with autolo- gous LCLs and on Day 10 the cells were used as effectors in a standard 51 Cr-release assay (effector:target FIG. 2. MHC restriction analysis for CTL clones. Peptide-coated DM PHA blasts were precoated with peptide 48 (SLYNLRRGTALAIPQ; 1 mg/ml) and then exposed to CTL clones (DM2 and DM6) in the presence of anti-class I (W6/32) or anti-class II (L243) antibody (1:20 diluted ascites). An effector to target ratio of 5:1 was used in the assay. Data from one representative experiment of three are shown.

SHORT COMMUNICATION 635 FIG. 3. Minimalizing the EBNA1 epitope. DM anti-cd40 B cell blasts were presensitized with individual overlapping peptides and then exposed to CTL clone DM2. The peptide sequence shown in a box was identified as the minimal epitope sequence required for maximal lysis. An effector to target ratio of 5:1 was used in the assay. Data from one representative experiment of two are shown. and SLYNLRRGTALAIPQ, suggesting that both these were synthesized and screened for recognition by DM2. overlapping sequences might include an EBNA1 epitope. As shown in Fig. 3, 10 different peptides were tested and No CTL clones specific for EBNA1 peptides were iso- compared for the concentration required for half-maximal lated from donors LC or DM following stimulation with lysis of autologous anti-cd40 B cell blasts. These B cell peptides 30 and 49. blasts were generated as described previously (18). Characterization (by FACS analysis) revealed that DM2 Based on this analysis, peptide TSLYNLRRGTALA was and DM6 were ú99.0% CD3 /, CD4 / cells, a phenotype the minimal peptide with maximal activity, inducing halfmaximal suggesting that these clones might be MHC class IIrestricted. lysis at a concentration of 0.069 mg/ml. To examine this possibility, autologous PHA An important aspect of the characterization of the CTL blasts were presensitized with peptide 48 and then exposed clone was to determine whether DM2 could recognize to the CTL clones in the presence of anti-class I autologous LCLs transformed with a range of virus (W6/32) or anti-class II (L243) antibody. As shown in Fig. strains. These LCLs were established either spontane- 2, recognition of peptide 48-coated PHA blasts was ously (19) or by exogenous virus transformation of peripheral blocked by L243 but was not significantly affected by B cells using type 1 or 2 EBV isolates (16). Surprisblocked W6/32 antibody. This result confirmed that these CTL ingly, no LCLs were recognized by DM2 (Fig. 4). The lack clones are MHC class II-restricted. of CTL recognition by DM2 was not due to any defect in To minimalize this CTL epitope, overlapping peptides the expression of MHC class II alleles, since all LCLs FIG. 4. Recognition of DM LCLs transformed with either type 1 or type 2 EBV isolates by CTL clone DM2. An effector to target ratio of 5:1 was used in the assay. Data from one representative experiment of four are shown.

636 SHORT COMMUNICATION FIG. 5. HLA class II restriction analysis for CTL clone DM2. Allogeneic EBV-transformed LCLs, some sharing an MHC class II antigen with donor DM, were exposed to the CTL clone DM2 in the absence or presence of TSLYNLRRGTALA peptide. An effector to target ratio of 5:1 was used in the assay. Data from one representative experiment of two are shown. were efficiently recognized by the clone if presensitized Indeed, the sequence conforms to the previously pubwith the TSLYNLRRGTALA peptide. This result suggests lished HLA DR1 binding motif (20). that LCLs are unable to process and present the EBNA1 To determine whether the lack of CTL lysis of autolo- CTL epitope endogenously. gous LCLs by CTL clone DM2 was due to insufficient To define the HLA restriction of the CTL clone DM2, levels of EBNA1 expressed in these cells, DM/B95.8 a panel of allogeneic LCLs, some sharing an MHC class LCLs and DM CD40 B cell blasts were infected with II allele with donor DM, was presensitized with TSLYNL- recombinant vaccinia constructs encoding the full-length RRGTALA and tested in a standard 51 Cr-release assay. EBNA1 gene (Vacc.EBNA1) and vaccinia encoding T7 As shown in Fig. 5, peptide-sensitized LCLs from RNA polymerase (Vacc.T7) (8). Despite high levels of DR1/ve donors, but not DR10ve donors, were recog- EBNA1 expression in these cells as determined by immunized by CTL clone DM2. These results suggest that noblotting (data not shown), no lysis was seen in the peptide TSLYNLRRGTALA is restricted through HLA DR1. presence of CTL clone DM2 (Fig. 6). These results confirm that, although EBNA1 includes a CTL epitope, this epitope is not processed and presented endogenously by EBV-infected cells. Levitskaya and colleagues have recently demonstrated that repeat sequences of glycine alanine within EBNA1 can inhibit presentation of class I epitopes (21). To explore the possibility that the lack of endogenous processing of the TSLYNLRRGTALA epitope relates to the presence of these repeat sequences, DM target cells were infected with two recombinant vaccinia vectors (Vacc.E1DGA and E1DCGA). Each of these constructs encodes a truncated EBNA1 protein without the glycine alanine repeats but including this epitope (P. M. Steigewald-Mullen and M. G. Kurilla, submitted for publication). Interestingly, target cells expressing these proteins were FIG. 6. Functional analysis of CTL clone DM2 using autologous LCLs or CD40 B cells infected with recombinant vaccinia expressing fullthat the presence of glycine alanine repeats is not re- not recognized by CTL clone DM2 (Fig. 6). Thus it seems length EBNA1 (Vacc.EBNA1), truncated EBNA1 proteins with glycine alanine repeat sequences deleted (Vacc.E1DGA and Vacc.E1DCGA), sponsible for inhibiting the processing of the class IIor with the nuclear localization signal sequence deleted (Vacc.E1DSS), restricted epitope defined in this study. Another possible as target cells. TSLYNLRRGTALA peptide-coated LCLs and CD40 B reason for the inability of infected cells to endogenously cells were used as controls. An effector to target ratio of 5:1 was used in the assay. Data from one representative experiment of four are process this epitope is that EBNA1 localizes to the nu- shown. cleus, thus limiting class II presentation. However, this

SHORT COMMUNICATION 637 seems unlikely since target cells were also not recog- REFERENCES nized by DM2 CTLs after infection with a recombinant 1. Yao, Q. Y., Ogan, P., Rowe, M., Wood, M., and Rickinson, A. B., Int. vaccinia (Vacc.E1DSS) encoding the EBNA1 protein with J. Cancer 43, 67 71 (1989). the nuclear localization signal sequence deleted (Fig. 6). 2. Greenspan, J. S., Greenspan, D., Lennette, E. T., Abrams, D. I., Furthermore, we have recently identified an epitope from Conant, M. A., Petersen, V., and Freese, U. K., N. Engl. J. Med. 313, 1564 1571 (1985). another EBV nuclear antigen (EBNA2) which is efficiently 3. Sculley, T. B., Apolloni, A., Stumm, R., Moss, D. J., Mueller-Lantzsch, processed endogenously through the class II pathway N., Misko, I. S., and Cooper, D. A., Virology 171, 401 408 (1989). (manuscript in preparation). 4. Sample, J., and Kieff, E., J. Virol. 64, 1667 1674 (1990). The present study clearly demonstrates that EBNA1 5. Moss, D. J., Rickinson, A. B., and Pope, J. H., Int. J. Cancer 22, 662 includes sequences which can be recognized by both 668 (1978). 6. Moss, D. J., Rickinson, A. B., and Pope, J. H., Int. J. Cancer 23, 618 polyclonal and clonal CTLs (Figs. 1 and 2). Interestingly, 625 (1979). the HLA DR1-restricted CTL clones are unable to recog- 7. Misko, I. S., Soszynski, T. D., Kane, R. G., and Pope, J. H., Clin. nize EBV-infected B cells, suggesting that EBNA1 may Immunol. Immunopathol. 32, 285 297 (1984). have evolved a mechanism to avoid endogenous pro- 8. Khanna, R., Burrows, S. R., Kurilla, M. G., Jacob, C. A., Misko, I. S., cessing and/or presentation through the class II path- Sculley, T. B., Kieff, E., and Moss, D. J., J. Exp. Med. 176, 169 176 (1992). way. The presence of memory CTLs specific for EBNA1 9. Murray, R. J., Kurilla, M. G., Brooks, J. M., Thomas, W. A., Rowe, peptide epitopes and their activation in vitro following M., Kieff, E., and Rickinson, A. B., J. Exp. Med. 176, 157 168 LCL stimulation (Fig. 1), however, suggests that EBNA1 (1992). protein from EBV-infected cells might be exogenously 10. Brooks, J. M., Murray, R. J., Thomas, W. A., Kurilla, M. G., and endocytosed by professional antigen-presenting cells Rickinson, A. B., J. Exp. Med. 178, 879 887 (1993). 11. Rowe, M., Rowe, D. T., Gregory, C. D., Young, L. S., Farrell, P. J., and presented in association with MHC class II moleand Rupani, H., EMBO J. 6, 2743 2751 (1987). cules. Alternatively, epitopes generated via extracellular 12. Rooney, C. M., Rickinson, A. B., Moss, D. J., Lenoir, G. M., and cleavage of EBNA1 by serum proteases might be respon- Epstein, M. A., IARC Sci. Publ. 249 264 (1985). sible for the activation of CTLs. It is also possible that 13. Chen, F., Zou, J. Z., di Renzo, L., Winberg, G., Hu, F., Klein, E., Klein, these CTLs have been activated in vivo by EBV-infected E., and Ernberg, I., J. Virol. 69, 3752 3758 (1995). 14. Trivedi, P., Masucci, M. G., Winberg, G., and Klein, G., Int. J. Cancer cells other than B cells. Indeed, earlier studies have 48, 794 800 (1991). shown that the efficiency of endogenous processing of 15. Burrows, S. R., Gardner, J., Khanna, R., Steward, T., Moss, D. J., class II epitopes is often dependent on the cell lineage Rodda, S., and Suhrbier, A., J. Gen. Virol. 75, 2489 2493 (1994). of the antigen-presenting cell (22). The results of the 16. Moss, D. J., Misko, I. S., Burrows, S. R., Burman, K., McCarthy, R., present study substantiate the view that EBV can persist and Sculley, T. B., Nature 331, 719 721 (1988). 17. Burrows, S. R., Suhrbier, A., Khanna, R., and Moss, D. J., Immunolin normal and malignant cells in vivo in an immunologiogy 76, 174 175 (1992). cally silent state. 18. Khanna, R., Jacob, C. A., Burrows, S. R., and Moss, D. J., J. Immunol. Methods 164, 41 49 (1993). 19. Rickinson, A. B., Rowe, M., Hart, I. J., Yao, Q. Y., Henderson, L. E., ACKNOWLEDGMENTS Rabin, H., and Epstein, M. A., Cell Immunol. 87, 646 658 (1984). 20. Chicz, R. M., Urban, R. G., Lane, W. S., Gorga, J. C., Stern, L. J., We acknowledge the Cetus Corporation for the generous gift of ril- Vignali, D. A., and Strominger, J. L., Nature 358, 764 768 (1992). 2. This work was supported by grants from Queensland Cancer Fund 21. Levitskaya, J., Coram, M., Levitsky, V., Imreh, S., Steigerwald-Mullen, and the National Cancer Institute (U.S.A.; CA-52250-04). M.G.K. is a P. M., Klein, G., Kurilla, M. G., and Masucci, M. G., Nature scholar of the Lucille P. Markey Charitable Trust. P.M.S.M. is supported 375, 685 688 (1995). by the Medical Scientist Training Program of the University of Virginia 22. Moreno, J., Vignali, D. A., Nadimi, F., Fuchs, S., Adorini, L., and Health Sciences Center. Hammerling, G. J., J. Immunol. 147, 3306 3313 (1991).