Characterization of a Live-Attenuated Retroviral Vaccine Demonstrates Protection via Immune Mechanisms

Similar documents
Gene Vaccine Dr. Sina Soleimani

Lecture 11. Immunology and disease: parasite antigenic diversity

Role of Natural Killer Cells in Resistance against Friend Retrovirus-Induced Leukemia

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

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

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

HIV Anti-HIV Neutralizing Antibodies

BBS 2711 Virology. Virus Vaccines

Role of Mink Cell Focus-Inducing Virus in Leukemias Induced by Friend Ecotropic Virus

Temporal Effects of Gamma Interferon Deficiency on the Course of Friend Retrovirus Infection in Mice

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

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Principles of Vaccination

Trends in vaccinology

125. Identification o f Proteins Specific to Friend Strain o f Spleen Focus forming Virus (SFFV)

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Pathogenesis of Simian Foamy Virus Infection in Natural and Experimental Hosts

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

FOCiS. Lecture outline. The immunological equilibrium: balancing lymphocyte activation and control. Immunological tolerance and immune regulation -- 1

Effects of Acute and Chronic Murine Norovirus Infections on Immune Responses and Recovery from Friend Retrovirus Infection

VIRUSES. Biology Applications Control. David R. Harper. Garland Science Taylor & Francis Group NEW YORK AND LONDON

IN VIVO STUDIES ON VIRAL VIRULENCE

Immunodeficiency. (2 of 2)

Brief Definitive Report

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

Effect of Complement and Viral Filtration on the

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

Supporting Information

HIV and Challenges of Vaccine Development

Fluid movement in capillaries. Not all fluid is reclaimed at the venous end of the capillaries; that is the job of the lymphatic system

Strategies for an HIV vaccine

RADIATION LEUKEMIA IN C57BL/6 MICE I. Lack of Serological Evidence for the Role of Endogenous Ecotropic Viruses in Pathogenesis*

Protective Efficacy of Nonneutralizing Monoclonal Antibodies in Acute Infection with Murine Leukemia Virus

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

RAISON D ETRE OF THE IMMUNE SYSTEM:

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

Supporting Information

Prolonged E55 Retrovirus Expression in Aged Mice Is Associated with a Decline in the Anti-Virus Immune Response

The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS. Ronald C Desrosiers University of Miami Miller School of Medicine

Tissue-Specific Replication of Friend and Moloney Murine Leukemia Viruses in Infected Mice

Third line of Defense

Are we targeting the right HIV determinants?

Immunization with single-cycle SIV significantly reduces viral loads after an intravenous challenge with SIV(mac)239

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

Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239

ISOLATION OF A SARCOMA VIRUS FROM A SPONTANEOUS CHICKEN TUMOR

By:Reham Alahmadi NOV The production of antibodies and vaccination technology

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

the xenotropic sequences in the region of the env gene. The to the env gene of mouse xenotropic type C virus.

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures

The humoral immune responses to IBV proteins.

Recombinant Protein Expression Retroviral system

Received 29 August 2002/Accepted 3 December 2002

Immunology Lecture 4. Clinical Relevance of the Immune System

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia

Adaptive Immunity. Lecture 14 Biology W3310/4310 Virology Spring Life is simple, but we insist on making it complicated CONFUCIUS

Biotechnology-Based Vaccines. Dr. Aws Alshamsan Department of Pharmaceutics Office: AA87 Tel:

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

Yellow Fever Vaccine: Direct Challenge of Monkeys Given Graded Doses of 17D

SPECIFIC ANTIINFECTIOUS IMMUNITY. colostral immunity. administration of antibodies VIRULENT VACCINE INACTIVATED VACCINE

Campbell's Biology: Concepts and Connections, 7e (Reece et al.) Chapter 24 The Immune System Multiple-Choice Questions

HIV cure: current status and implications for the future

Transplantation. Immunology Unit College of Medicine King Saud University

development of erythroid precursor cells (reviewed in reference 26). Erythroid burst-forming cells are elevated in animals

Chapter 1. Full file at

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

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

BIT 120. Copy of Cancer/HIV Lecture

Principles of Adaptive Immunity

Host Restriction of Friend Leukemia Virus. Role of the Viral Outer Coat (mice/fv-1 locus/vesicular stomatitis virus)

Chapter 22: The Lymphatic System and Immunity

LESSON 4.6 WORKBOOK. Designing an antiviral drug The challenge of HIV

Activation of Nonexpressed Endogenous Ecotropic Murine Leukemia Virus by Transfection of Genomic DNA into Embryo Cells

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.

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

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells

Human Immunodeficiency Virus

Development of Broadly Reactive HIV-1/AIDS Virus-like Particle Vaccines. Sean Patrick McBurney. B.S. Microbiology, University of Pittsburgh, 2004

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

ADCC Develops Over Time during Persistent Infection with Live-Attenuated SIV and Is Associated with Complete Protection against SIV(mac)251 Challenge

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

5 Cell recognition and the immune system Exam-style questions. AQA Biology. 1 Figure 1 shows a phagocyte attracted to a bacterium.

Phase of immune response

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

Biomedical Engineering for Global Health. Lecture 10 HIV/AIDS vaccine development

Received 4 December 2001/Accepted 29 April 2002

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED

Viral Genetics. BIT 220 Chapter 16

Mucosal Immune System

Host Defense Mechanisms Against Influenza Virus: Interaction of Influenza Virus with Murine Macrophages In Vitro

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

Xenotransplantation. Pig as an organ donor

Immunization. Immunization procedure called vaccination and the immunizing agent called vaccine (or serum in historical references)

3. Lymphocyte proliferation (fig. 15.4): Clones of responder cells and memory cells are derived from B cells and T cells.

What is the role of animal models in studying protective titres and the need for establishing surrogates/correlates of protection?

Heat-killed Lactobacillus casei

Supporting Information

Acquired Immunity 2. - Vaccines & Immunological Memory - Wataru Ise. WPI Immunology Frontier Research Center (IFReC) Osaka University.

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

Transcription:

JOURNAL OF VIROLOGY, Aug. 1998, p. 6554 6558 Vol. 72, No. 8 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology. All Rights Reserved. Characterization of a Live-Attenuated Retroviral Vaccine Demonstrates Protection via Immune Mechanisms ULF DITTMER,* DIANE M. BROOKS, AND KIM J. HASENKRUG Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana 59840 Received 16 March 1998/Accepted 11 May 1998 Live-attenuated retroviruses have been shown to be effective retroviral vaccines, but currently little is known regarding the mechanisms of protection. In the present studies, we used Friend virus as a model to analyze characteristics of a live-attenuated vaccine in protection against virus-induced disease. Highly susceptible mice were immunized with nonpathogenic Friend murine leukemia helper virus (F-MuLV), which replicates poorly in adult mice. Further attenuation of the vaccine virus was achieved by crossing the Fv-1 genetic resistance barrier. The minimum dose of vaccine virus required to protect 100% of the mice against challenge with pathogenic Friend virus complex was determined to be 10 3 focus-forming units of attenuated virus. Live vaccine virus was necessary for induction of immunity, since inactivated F-MuLV did not induce protection. To determine whether immune cells mediated protection, spleen cells from vaccinated donor mice were adoptively transferred into syngeneic recipients. The results indicated that immune mechanisms rather than viral interference mediated protection. Live-attenuated viruses are successfully used for preventing virus-induced diseases such as measles, mumps, rubella, and polio. Since the discovery of retroviruses such as human T-cell leukemia virus and human immunodeficiency virus (HIV) that cause diseases in humans, biomedical researchers have been interested in designing live-attenuated vaccines for retroviruses as well. In recent experiments, monkeys were protected by live-attenuated simian immunodeficiency virus (SIV) against challenge with pathogenic SIV isolates (1, 13, 36). However, the mechanism of protection is not understood, and there has even been a question regarding whether protection was immunologically based. While it is not theoretically necessary to understand how a vaccine works in order to use it, there are numerous safety concerns involved in the use of live-attenuated retroviruses as a vaccine. Thus, it would be beneficial to establish the basic parameters regarding protection by liveattenuated retroviruses, and the most useful animals for such studies are mice. Since there is currently no mouse model for HIV infection, we have initiated studies using Friend virus (FV), a murine retrovirus that causes immunosuppression and erythroleukemia in adult mice. Although there are major differences between the diseases caused by FV and HIV, it is possible that the basic requirements for vaccine protection against retroviruses are very similar. FV is a retroviral complex comprised of a replication-competent helper virus, Friend murine leukemia virus (F-MuLV), and a replication-defective spleen focus-forming virus (SFFV) (22). In susceptible adult animals, FV induces rapid polyclonal erythroblast proliferation (19, 24), followed within 3 to 4 weeks by the immortalization of erythroid cells (12, 28, 30, 38). FV infections cause profound splenomegaly, abnormally high hematocrits (greater than 80%), and lethal erythroleukemias in most strains of mice. In several different vaccination experiments, protective immunity against FV has been achieved by * Corresponding author. Mailing address: Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 S. 4th St., Hamilton, MT 59840. Phone: (406) 363-9312. Fax: (406) 363-9204. E-mail: udittmer@atlas.niaid.nih.gov. using killed virus with adjuvants (21, 29), isolated viral proteins (20, 21), recombinant viral vectors expressing FV proteins (15, 17), and live-attenuated vaccines (15, 25). In contrast to vaccination with vaccinia virus vectors, live-attenuated virus is highly protective, even in mouse strains that have poor immunological responsiveness to FV because of their major histocompatibility complex (MHC) haplotype (15). In previous experiments with live-attenuated FV, attenuation was achieved by crossing a host genetic resistance barrier called Fv-1. Successful replication of virus in Fv-1-resistant cells in vitro requires at least 100-fold more virus than in Fv-1-compatible cells (32). Likewise, induction of disease in Fv-1-incompatible mice requires much higher doses of virus than are required in Fv-1-compatible strains (25). Thus, inoculation of 1,500 focus-forming units (FFU) of N-tropic FV (FV-N) complex induces rapid erythroleukemia in DBA/2 (Fv- 1 n/n ) mice, but not in A.BY (Fv-1 b/b ) mice. In addition, after infection with FV-N, Fv-1 b/b mice are subsequently protected from challenge with B-tropic FV (FV-B) complex (15, 25). Although such vaccination has been shown to generate virusspecific B-cells, cytotoxic T cells (CTLs), and helper T cells (TH) (15), the role of these immune cells in protection has not been established. In fact, evidence indicates that in Fv-1-compatible neonatal mice, protection by live-attenuated vaccines occurs through viral interference rather than immunological mechanisms (11, 12, 26). The present paper establishes that only the F-MuLV helper component of the FV-N complex is required for effective vaccination and demonstrates that protection by F-MuLV in adult mice is primarily mediated by immune cells rather than viral interference. MATERIALS AND METHODS Mice. (B10.A A/Wy)F 1 mice 3 to 6 months of age at experimental onset were used. F 1 parental strain mice were obtained from the Jackson Laboratories. Breeding of F 1 strains was done at Rocky Mountain Laboratories. All mice were treated in accordance with National Institutes of Health regulations and the guidelines of the Animal Care and Use Committee of Rocky Mountain Laboratories. Virus vaccination and virus challenge. The FV-B complex used in these experiments was from uncloned virus stocks obtained from 10% spleen cell 6554

VOL. 72, 1998 LIVE-ATTENUATED FRIEND VIRUS VACCINE 6555 homogenates from BALB/c mice infected 9 days previously with polycythemiainducing FV stocks originally obtained from Frank Lilly (10, 15). The N-tropic F-MuLV (stock 29-51N) (6) was a 24-h supernatant from infected Mus dunni cells (23). Mice were vaccinated by intravenous injection of 0.5 ml of phosphatebuffered, balanced salt solution (PBBS) (9) containing 2% fetal bovine serum and 10 4 FFU of N-tropic F-MuLV vaccine virus. Heat inactivation of the F- MuLV vaccine was performed by 1hofincubation in a 56 C water bath. In virus challenge experiments, mice were injected intravenously with 0.5 ml of PBBS containing 2% fetal bovine serum and 1,500 spleen FFU (SFFU) of FV-B complex. Splenomegaly as a measure of Friend disease. Palpation for splenomegaly is the standard procedure used to monitor the progression of Friend disease (10, 15, 31) and was used in the following manner. At weekly intervals, each individual animal under general anesthesia was palpated in a blinded fashion and rated on a scale of 1 to 4 according to its spleen size. Normal (1 ) spleen weights range from 0.1 to 0.25 g. Spleens greater than twice normal size (more than 0.4 g), but not large enough to reach the ventral midline, were rated as 2. Spleens that weigh between 0.25 and 0.4 g were still rated as 1. If spleens were large enough to reach the ventral midline, they were rated as 3 (weight of between 0.8 and 1.6 g). Spleens which extended across the abdominal midline and caused protrusion of the abdominal wall were rated as 4 (weight greater than 1.6 g). Cross-checking of actual spleen weights with spleen sizes determined by palpations has demonstrated consistency in differentiation of spleens weighing in the normal range from those weighing greater than 0.4 g (2 ) (reference 5) and our unpublished data). Mice that have sustained splenomegaly by 8 weeks postinfection begin to die about 10 weeks postinfection, and most are dead by 16 weeks postinfection. In contrast, mice which recover from splenomegaly live normal life spans (5, 10). Viremia and virus-neutralizing antibody assays. For viremia assays, freshly frozen plasma samples were titrated by focal infectivity assays (35) on susceptible M. dunni cells pretreated with 4 g of Polybrene per ml. Cultures were incubated for 5 days, fixed with ethanol, stained with F-MuLV envelope-specific monoclonal antibody 720 (33), and developed with goat anti-mouse peroxidase-conjugated antisera (Cappel, West Chester, Pa.) and aminoethylcarbazol to detect foci. To test plasma samples for virus-neutralizing antibodies, heat-inactivated (56 C, 10 min) samples at titrated dilutions were incubated with virus stock in the presence of complement at 37 C as previously described (29). The samples were then plated as described for the viremia assay to determine the dilution at which 75% of the virus had been neutralized. Infectious center assays. Titrations of single-cell suspensions from persistently infected mouse spleens were plated onto susceptible M. dunni cells (23), cocultivated for 5 days, fixed with ethanol, stained with F-MuLV envelope-specific monoclonal antibody 720 (33), and developed with peroxidase-conjugated goat anti-mouse antibodies and aminoethylcarbazol to detect foci. Adoptive spleen cell transfer. For the transfer experiments, (B10.A A/Wy)F 1 mice (Fv-1 b/b ) were vaccinated by infection with 10 4 FFU of liveattenuated N-tropic F-MuLV. After 30 days, spleen cells from these mice were adoptively transferred to naive syngeneic animals via tail vein injection. Each mouse received 7.5 10 7 spleen cells in 0.75 ml of PBBS. Cell suspensions were depleted of erythroid cells and filtered through a nylon screen to remove chunks. The PBBS was supplemented with 15 U of heparin per ml. One day later, the recipients were challenged with 1,500 SFFU of FV-B complex. Controls received either (i) 7.5 10 7 spleen cells from naive mice, (ii) 7.5 10 7 spleen cells taken from animals 3 days post-f-mulv vaccination, or (iii) 10 4 FFU of N-tropic F-MuLV alone before challenge 1 day later. After FV-B challenge, the recipients were palpated weekly for virus-induced splenomegaly. Statistical comparisons of groups of mice that received spleen cells from F-MuLV-inoculated donor animals 3 or 30 days after vaccination were performed with Fisher s exact test of chi-square analysis. RESULTS Protection of Fv-1 b/b mice with N-tropic F-MuLV. Previous studies describing a live-attenuated FV vaccine used the FV- N complex composed of N-tropic F-MuLV helper virus and SFFV (15). However, FV-N is pathogenic, depending on the virus dose and mouse strain used, while F-MuLV alone is nonpathogenic in adult mice. We wished to determine if F- MuLV alone could be used to provide protection, thereby reducing the pathogenic potential of the vaccine and eliminating SFFV as an immunological variable. To this end, highly susceptible (B10.A A/Wy)F 1 (Fv-1 b/b ) mice were vaccinated by infection with 10 4 FFU of N-tropic F-MuLV. A control group of animals was inoculated with the same dose of heatinactivated virus. At 1 month postvaccination, the mice were challenged with pathogenic FV-B complex, and the animals were examined weekly for virus-induced splenomegaly. All 18 animals that were vaccinated with live F-MuLV were protected FIG. 1. F-MuLV vaccine-induced protection from FV-induced splenomegaly in FV-1 b/b mice. Age-matched (B10.A A/Wy)F 1 mice were vaccinated with N-tropic live-attenuated F-MuLV ( ) or heat inactivated F-MuLV ( ). The mice were challenged 1 month later with a high dose (1,500 SFFU) of FV-B complex and monitored for induction and progression of splenomegaly. For live F-MuLV-vaccinated mice, n 18; for inactivated F-MuLV-vaccinated mice, n 16; and for naive controls (F), n 20. from splenomegaly during the 8-week observation period after FV-B challenge (Fig. 1). Furthermore, they were also protected against a second FV-B challenge 3 months later (data not shown). In contrast, all 16 mice immunized with inactivated F-MuLV developed splenomegaly within 2 weeks after infection, and more than 80% of the animals had to be euthanized within 8 weeks because of FV-induced erythroleukemia (Fig. 1). Thus, the helper component alone was sufficient for protection against FV-induced disease. Furthermore, in vivo infection and, perhaps, replication were required to induce protective immunity. Viremia and virus-neutralizing antibodies in vaccinated mice. To determine if vaccination had limited the challenge infection sufficiently to prevent viremia, plasma samples from immunized animals were tested for the presence of free virus. Infectious virus was detected 7 days after FV-B challenge in the blood of unvaccinated animals and animals inoculated with inactivated virus (Fig. 2A). In contrast, animals vaccinated with live F-MuLV had no measurable viremia at the same time point. Since it has been previously shown that viremia in FV infection is controlled by virus-neutralizing antibodies (4, 8), the titers of neutralizing antibodies in the groups vaccinated with live or inactivated virus were determined. Virus-neutralizing antibody titers ranging from 1/40 to 1/160 were found in the sera from mice that had received the live vaccine virus (Fig. 2B). In contrast, mice immunized with inactivated virus had no detectable virus-neutralizing antibody titers. Thus, vaccination with live F-MuLV induced production of virus-neutralizing antibody and prevented the spread of virus to the blood. Determination of the minimal viral dose for efficient F- MuLV vaccination. To determine the minimal live-attenuated F-MuLV viral dose required to induce protection, (B10.A A/Wy)F 1 mice were inoculated with various amounts of the vaccine virus. Groups of animals vaccinated with either 10 4

6556 DITTMER ET AL. J. VIROL. TABLE 1. Detection of vaccine virus at various time points after F-MuLV inoculation No. of ICs/10 7 spleen cells at postvaccination day a 3 10 30 5 2 0 1 24 0 2 80 0 3 32 0 0 9 1 0 0 0 a Determined after inoculation with 10 4 FFU of F-MuLV. FIG. 2. Plasma viremia and virus-neutralizing antibodies in N-tropic F- MuLV-vaccinated and FV-B-challenged mice. (A) Plasma samples were taken at 1 week postchallenge to assay for the presence of plasma viremia. The limit of detection was 220 FFU/ml of plasma. (B) The same samples were also analyzed for F-MuLV-neutralizing antibody. The neutralizing antibody titer was considered to be the highest dilution at which greater than 75% of the input virus was neutralized. Six individual animals (F) from the groups of mice vaccinated with live or inactivated F-MuLV were investigated. or 10 3 FFU of F-MuLV were completely protected against splenomegaly induced by challenge with 1,500 FFU of FV-B (Fig. 3). In contrast, at 8 weeks postchallenge, mice vaccinated with 10 1 and 10 2 FFU of F-MuLV showed 71 and 57% splenomegaly, respectively. All unvaccinated mice and mice that received 10 0 FFU of the vaccine virus developed severe splenomegaly after FV-B challenge and had to be euthanized. Thus, full protection required immunization with 10 3 FFU of F- MuLV. The level of in vivo replication of the vaccine virus was determined by measuring infectious centers (ICs) in the spleens of F-MuLV-inoculated mice. At both 3 and 10 days postinoculation with 10 4 FFU of F-MuLV, fewer than 10 3 ICs per spleen were detected (Table 1). This level of in vivo infection was very low: approximately 10,000 times less than that in the spleens of mice infected with pathogenic FV-B (15). By 30 days postvaccination with F-MuLV, only one of six mice had detectable spleen virus (Table 1), indicating that immune responses had cleared most virus. Thus, the results indicated that only low-level replication of the F-MuLV vaccine virus was required to induce protection. Transfer of F-MuLV-primed spleen cells protected syngeneic recipients against FV-B challenge. To address the question of whether F-MuLV-induced protection was due to an immunological mechanism, we performed an adoptive cell transfer experiment. Spleen cells (7.5 10 7 ) from F-MuLVvaccinated or naive (B10.A A/Wy)F 1 mice were transferred into naive, syngeneic animals. One day later, the recipients were challenged with pathogenic FV-B. Six of nine animals receiving cells from vaccinated mice were protected against FV-induced early splenomegaly (Fig. 4). In addition, one of the three animals which had early splenomegaly later recovered from Friend disease. In contrast, all nine animals that received spleen cells from naive mice developed severe splenomegaly, and none of the mice recovered. A possible mechanism for protection by live-attenuated retroviruses is viral interference. This mechanism appeared unlikely, given that there was no detectable vaccine virus in the immune spleen cells that were transferred at 30 days post-f- MuLV vaccination (Table 1). However, as a control for passage of an interfering virus, spleen cells were transferred at 3 days postvaccination. At this time point, there was detectable vaccine virus in the spleen cells, but the immune response had not yet had time to develop. None of the mice that received spleen cells from donors at 3 days postvaccination were protected from FV-induced splenomegaly (Fig. 4). Likewise, mice that received the F-MuLV vaccine virus 1 day prior to FV-B challenge also were not protected. These results indicate that immune cells rather than viral interference mediated protection by the F-MuLV live-attenuated retroviral vaccine. FIG. 3. Titration of the F-MuLV vaccine virus. Age-matched (B10.A A/Wy)F 1 (FV-1 b/b ) mice were vaccinated with five 10-fold dilutions (10 0 to 10 4 FFU) of the N-tropic F-MuLV vaccine virus. The mice were challenged 1 month later with FV-B complex and monitored for induction and progression of splenomegaly. For vaccination with live F-MuLV, the inoculations and numbers of mice were as follows: 10 0 FFU ( ), n 7; 10 1 FFU ( ), n 7; 10 2 FFU (F), n 7; 10 3 FFU (E), n 7; 10 4 FFU ( ), n 18. For naive controls ({), n 20. DISCUSSION The current results demonstrate that the primary mechanism of protection by live-attenuated FV vaccination is immunologically mediated. There was no indication of an effect from viral interference, since vaccine virus without any immune cells did not protect adult mice against FV-induced disease. This was found for the inoculation of free virus 1 day prior to challenge, the same time point used to establish viral interference as the mechanism of protection in neonatal mice (11). To rule out the possibility that transfers of infected cells might

VOL. 72, 1998 LIVE-ATTENUATED FRIEND VIRUS VACCINE 6557 FIG. 4. Transfer of F-MuLV-primed spleen cells into syngeneic mice. Spleen cells (7.5 10 7 ) from F-MuLV-vaccinated or naive (B10.A A/Wy)F 1 mice were transferred to age-matched syngeneic animals. Nine animals in each group received either spleen cells from naive mice (Œ) or spleen cells from mice vaccinated with F-MuLV 30 days prior to transfer (F). One day after transfer, the recipients were challenged with FV-B complex and monitored for virusinduced splenomegaly. Eight controls were inoculated with the F-MuLV vaccine virus 1 day prior to FV-B challenge ( ). In addition, nine animals received spleen cells from animals which were F-MuLV vaccinated only 3 days prior to the transfer (E). The difference between the groups of animals that received spleen cells 3 days postvaccination and those who received them 30 days postvaccination was statistically very significant (P 0.009, by Fisher s exact test of chi-square analysis). transfer vaccine virus more efficiently than infection with free virus, we transferred cells at 3 days postvaccination. At this time point, there was detectable virus in the transferred cells, but significant numbers of immune cells had not been generated. There was no protection conferred by transfers of infected, but nonimmune spleen cells. In contrast, immune cells that harbored no detectable F-MuLV were protective. Although these cells might have harbored an undetectable lowlevel infection, the results from our control groups make it unlikely that this contributed significantly to their protective effect. It has been shown for Fv-4-induced resistance of mice against FV that high but not low levels of FV envelope expression induced protection by an interference mechanism (26). Thus, large numbers of vaccine virus-infected target cells seem to be necessary to induce resistance against FV infection via interference. However, a minor additive effect of interference together with the immune-mediated protection by live-attenuated retroviruses cannot be ruled out. Our results are significant in that they suggest it may be possible to achieve similar protection with vectors believed to be safer than live-attenuated viruses. However, this will require much more detailed knowledge about the immunological mechanisms required for protection. For example, previous attempts to vaccinate mice against FV with recombinant vaccinia virus vectors expressing viral proteins have resulted in only limited protection dependent on the MHC type of the mouse (7, 15, 18). Recombinant vaccinia virus vectors have also been relatively unsuccessful in the SIV model (14, 16), possibly because of the nonresponsiveness of some MHC types (3). Likewise, peptide vaccines in the FV model have shown only limited protection in certain MHC types (27). Any vaccine useful for human immunization against retroviral infections must be broadly protective in multiple MHC types, as are the live-attenuated viruses. Thus, it is important to determine which characteristics of the live-attenuated viruses account for their efficacy and differentiate them from less-effective vaccines, such as vaccinia virus vectors. In the FV model, one of the known differences between vaccination with live-attenuated viruses and that with vaccinia virus vectors is that the live-attenuated viruses elicit immunological effectors, including CTLs, virus-neutralizing antibody, and CD4 T cells, while vaccinia virus vectors generally only prime for CTL and antibody responses, but do elicit CD4 T cells (15). The reasons for this remain unclear, but may be related to the duration of antigen available for immune stimulation. In the present studies, which were done with highly susceptible animals with poorly responsive MHC haplotypes, it is possible that vaccine virus persisted at levels below the limit of detection of our assay and contributed to immune protection. With sensitive PCR techniques, persistent vaccine viruses have been found in macaques vaccinated with live-attenuated SIV (37), and a chronic source of antigen may be an essential aspect of long-term protection. It will be important to address this issue more carefully in future studies. Other attributes which may contribute to the efficacy of liveattenuated viruses as vaccines include the wide range of viral proteins which are expressed, the types of immune cells responding to the infection, the specific location of viral antigen within the immunological architecture, and the degree of vaccine virus replication. These issues have broad implications for the rational design of safe and effective retroviral vaccines and must be addressed. The system described here offers the opportunity to examine these basic issues and could lead to novel vaccination methods which exploit the advantages of live-attenuated retroviruses while reducing or eliminating potential dangers. While this is obviously the preferred outcome, it may also be found that only live-attenuated retroviruses stimulate the specific types of immune responses uniquely required for retroviral protection. The critical issue then becomes how to make a safe live-attenuated retrovirus. Many of the safety issues, such as reversion to virulence, insertional mutagenesis, and recombination with endogenous or exogenous retroviral sequences, are frequency dependent and are related to levels of infection and virus replication. Thus, it may be possible to reduce the risk to acceptable levels by determining the proper dose and precisely attenuating the vaccine virus such that replication is only high enough to stimulate protective immunity. Our data give a hint about what those minimum levels might be. In some animals, doses as low as 10 FFU of F-MuLV induced protection (Fig. 3). It is possible that a booster administration of the same dose might significantly improve protection while restricting replication levels. In the current experiments, only live vaccine virus induced protection. However, it remains possible that virus replication and spread were not requirements for protection and that there were only requirements for infection and protein synthesis. Future experiments with live replication-defective viruses may clarify this issue, but previous experiments have indicated that without replication, protection is limited by the MHC haplotype of the animal (34). Since we have now established immune cells as the effectors of protection, it will be possible in future studies to dissect which types of immune cells are necessary. Such experiments may reveal whether it is feasible to focus on a single lymphocyte subset alone to provide protection from retroviral infection.

6558 DITTMER ET AL. J. VIROL. ACKNOWLEDGMENT U.D. is supported by a fellowship from the Deutsche Forschungsgemeinschaft. REFERENCES 1. Almond, N., K. Kent, M. Cranage, E. Rud, B. Clarke, and E. J. Stott. 1995. Protection by attenuated simian immunodeficiency virus in macaques against challenge with virus-infected cells. Lancet 345:1342 1344. 2. Ben-David, Y., and A. Bernstein. 1991. Friend virus-induced erythroleukemia and the multistage nature of cancer. Cell 66:831 834. 3. Bontrop, R. E., N. Otting, H. Niphius, R. Noort, V. Teeuwsen, and J. L. Heeney. 1996. The role of major histocompatibility complex polymorphisms on SIV infection in rhesus macaques. Immunol. Lett. 51:35 38. 4. Britt, W. J., and B. Chesebro. 1983. Use of monoclonal anti-gp70 antibodies to mimic the effects of the Rfv-3 gene in mice with Friend virus-induced leukemia. J. Immunol. 130:2363 2367. 5. Chesebro, B., M. Bloom, K. Wehrly, and J. Nishio. 1979. Persistence of infectious Friend virus in spleens of mice after spontaneous recovery from virus-induced erythroleukemia. J. Virol. 32:832 837. 6. Chesebro, B., W. Britt, L. Evans, K. Wehrly, J. Nishio, and M. Cloyd. 1983. Characterization of monoclonal antibodies reactive with murine leukemia viruses: use in analysis of strains of Friend MCF and Friend ecotropic murine leukemia virus. Virology 127:134 148. 7. Chesebro, B., M. Miyazawa, and W. J. Britt. 1990. Host genetic control of spontaneous and induced immunity to Friend murine retrovirus infection. Annu. Rev. Immunol. 8:477 499. 8. Chesebro, B., and K. Wehrly. 1979. Identification of a non-h-2 gene (Rfv-3) influencing recovery from viremia and leukemia induced by Friend virus complex. Proc. Natl. Acad. Sci. USA 76:425 429. 9. Chesebro, B., and K. Wehrly. 1976. Studies on the role of the host immune response in recovery from Friend virus leukemia. II. Cell-mediated immunity. J. Exp. Med. 143:85 99. 10. Chesebro, B., K. Wehrly, and J. Stimpfling. 1974. Host genetic control of recovery from Friend leukemia virus-induced splenomegaly. Mapping of a gene within the major histocompatibility complex. J. Exp. Med. 140:1457 1467. 11. Corbin, A., and M. Sitbon. 1993. Protection against retroviral diseases after vaccination is conferred by interference to superinfection with attenuated murine leukemia viruses. J. Virol. 67:5146 5152. 12. Czub, M., F. J. McAtee, S. Czub, W. P. Lynch, and J. L. Portis. 1995. Prevention of retrovirus-induced neurological disease by infection with a nonneuropathogenic retrovirus. Virology 206:372 380. 13. Daniel, M. D., F. Kirchhoff, S. C. Czajak, P. K. Sehgal, and R. C. Desrosiers. 1992. Protective effects of a live attenuated SIV vaccine with a deletion in the nef gene. Science 258:1938 1941. 14. Daniel, M. D., G. P. Mazzara, M. A. Simon, P. K. Sehgal, T. Kodama, D. L. Panicali, and R. C. Desrosiers. 1994. High-titer immune responses elicited by recombinant vaccinia virus priming and particle boosting are ineffective in preventing virulent SIV infection. AIDS Res. Hum. Retroviruses 10:839 851. 15. Earl, P. L., B. Moss, R. P. Morrison, K. Wehrly, J. Nishio, and B. Chesebro. 1986. T-lymphocyte priming and protection against Friend leukemia by vaccinia-retrovirus env gene recombinant. Science 234:728 731. 16. Giavedoni, L. D., V. Planelles, N. L. Haigwood, S. Ahmad, J. D. Kluge, M. L. Marthas, M. B. Gardner, P. A. Luciw, and T. D. Yilma. 1993. Immune response of rhesus macaques to recombinant simian immunodeficiency virus gp130 does not protect from challenge infection. J. Virol. 67:577 583. 17. Hasenkrug, K. J., D. M. Brooks, J. Nishio, and B. Chesebro. 1996. Differing T-cell requirements for recombinant retrovirus vaccines. J. Virol. 70:368 372. 18. Hasenkrug, K. J., and B. Chesebro. 1997. Immunity to retroviral infection: the Friend virus model. Proc. Natl. Acad. Sci. USA 94:7811 7816. 19. Hoatlin, M. E., and D. Kabat. 1995. Host-range control of a retroviral disease: Friend erythroleukemia. Trends Microbiol. 3:51 57. 20. Hunsmann, G., J. Schneider, and A. Schulz. 1981. Immunoprevention of Friend virus-induced erythroleukemia by vaccination with viral envelope glycoprotein complexes. Virology 113:603 612. 21. Ishihara, C., M. Miyazawa, J. Nishio, and B. Chesebro. 1991. Induction of protective immunity to Friend murine leukemia virus in genetic nonresponders to virus envelope protein. J. Immunol. 146:3958 3963. 22. Kabat, D. 1989. Molecular biology of Friend viral erythroleukemia. Curr. Top. Microbiol. Immunol. 148:1 42. 23. Lander, M. R., and S. K. Chattopadhyay. 1984. A Mus dunni cell line that lacks sequences closely related to endogenous murine leukemia viruses and can be infected by ecotropic, amphotropic, xenotropic, and mink cell focusforming viruses. J. Virol. 52:695 698. 24. Li, J.-P., A. D. D Andrea, H. F. Lodish, and D. Baltimore. 1990. Activation of cell growth by binding of Friend spleen focus-forming virus gp55 glycoprotein to the erythropoietin receptor. Nature (London) 343:762 764. 25. Lilly, F. 1967. Susceptibility to two strains of Friend leukemia virus in mice. Science 155:461 462. 26. Limjoco, T. I., P. Dickie, H. Ikeda, and J. Silver. 1993. Transgenic Fv-4 mice resistant to Friend virus. J. Virol. 67:4163 4168. 27. Miyazawa, M., R. Fujisawa, C. Ishihara, Y. A. Takei, T. Shimizu, H. Uenishi, H. Yamagishi, and K. Kuribayashi. 1995. Immunization with a single T helper cell epitope abrogates Friend virus-induced early erythroid proliferation and prevents late leukemia development. J. Immunol. 155:748 758. 28. Moreau-Gachelin, F., A. Tavitian, and P. Tambourin. 1988. Spi-1 is a putative oncogene in virally induced murine erythroleukemia. Nature (London) 331:277 280. 29. Morrison, R. P., P. L. Earl, J. Nishio, D. L. Lodmell, B. Moss, and B. Chesebro. 1987. Different H-2 subregions influence immunization against retrovirus and immunosuppression. Nature (London) 329:729 732. 30. Munroe, D. G., J. W. Peacock, and S. Benchimol. 1990. Inactivation of the cellular p53 gene is a common feature of Friend virus-induced erythroleukemia: relationship of inactivation to dominant transforming alleles. Mol. Cell. Biol. 10:3307 3313. 31. Polsky, D., and F. Lilly. 1991. Suppression of H-2b-associated resistance to Friend erythroleukemia virus by a class I gene from the H-2d major histocompatibility complex haplotype. Proc. Natl. Acad. Sci. USA 88:9243 9247. 32. Pryciak, P. M., and H. E. Varmus. 1992. Fv-1 restriction and its effects on murine leukemia virus integration in vivo and in vitro. J. Virol. 66:5959 5966. 33. Robertson, M. N., M. Miyazawa, S. Mori, B. Caughey, L. H. Evans, S. F. Hayes, and B. Chesebro. 1991. Production of monoclonal antibodies reactive with a denatured form of the Friend murine leukemia virus gp70 envelope protein: use in a focal infectivity assay, immunohistochemical studies, electron microscopy and western blotting. J. Virol. Methods 34:255 271. 34. Ruan, K.-S., and F. Lilly. 1992. Approach to a retrovirus vaccine: immunization of mice against Friend virus disease with a replication-defective Friend murine leukemia virus. Proc. Natl. Acad. Sci. USA 89:12202 12206. 35. Sitbon, M., J. Nishio, K. Wehrly, D. Lodmell, and B. Chesebro. 1985. Use of a focal immunofluorescence assay on live cells for quantitation of retroviruses: distinction of host range classes in virus mixtures and biological cloning of dual-tropic murine leukemia viruses. Virology 141:110 118. 36. Stahl-Hennig, C., U. Dittmer, T. Nisslein, K. Pekrun, H. Petry, E. Jurkiewicz, D. Fuchs, H. Wachter, E. W. Rud, and G. Hunsmann. 1996. Attenuated SIV imparts immunity to challenge with pathogenic spleen-derived SIV but cannot prevent repair of the nef deletion. Immunol. Lett. 51:129 135. 37. Titti, F., L. Sernicola, A. Geraci, G. Panzini, S. Di Fabio, R. Belli, F. Monardo, A. Borsetti, M. T. Maggiorella, M. Koango-Mogtomo, F. Corrias, R. Zamarchi, A. Amadori, L. Chieco-Bianch, and P. Verani. 1997. Live attenuated simian immunodeficiency virus prevents super-infection by cloned SIVmac251 in cynomolgus monkeys. J. Gen. Virol. 78:2529 2539. 38. Wendling, F., and P. E. Tambourin. 1978. Oncogenicity of Friend-virusinfected cells: determination of origin of spleen colonies by the H-2 antigens as genetic markers. Int. J. Cancer 22:479 486.