Institute of Virology, Erasmus University Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands 2

Similar documents
Virus neutralization reveals antigenic variation among feline immunodeficiency virus isolates

Received 27 May 1997/Accepted 7 August 1997

HIV Anti-HIV Neutralizing Antibodies

DNA Vaccination Affords Significant Protection against Feline Immunodeficiency Virus Infection without Inducing Detectable Antiviral Antibodies

Received 13 August 1997/Returned for modification 24 September 1997/Accepted 12 November 1997

Immunogenicity of an Anti-Clade B Feline Immunodeficiency Fixed-Cell Virus Vaccine in Field Cats

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

Neutralization sensitivity and accessibility of continuous B cell epitopes of the feline immunodeficiency virus envelope

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

Dynamics of lentiviral infection in vivo in the absence of adaptive immune responses

Human Immunodeficiency Virus

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved.

Materials and Methods

Lecture 11. Immunology and disease: parasite antigenic diversity

Equine infectious anaemia virus proteins with epitopes most frequently recognized by cytotoxic T lymphocytes from infected horses

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

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.

Comparison of the efficacy of early versus late viral proteins in vaccination against SIV

Principles of Vaccination

Received 29 August 2002/Accepted 3 December 2002

Envelope gene sequences encoding variable regions 3 and 4 are involved in macrophage tropism of feline immunodeficiency virus

Association of Plasma Viral RNA Load with Prognosis in Cats Naturally Infected with Feline Immunodeficiency Virus

Evolution of Replication Efficiency following Infection with a Molecularly Cloned Feline Immunodeficiency Virus of Low Virulence

IS AN AIDS VACCINE ALREADY AVAILABLE?

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2

Immune Responses and Viral Replication in Long-Term Inapparent Carrier Ponies Inoculated with Equine Infectious Anemia Virus

Production of Interferon Alpha by Dengue Virus-infected Human Monocytes

Expression of CXCR4 on feline peripheral blood mononuclear. cells: effect of feline immunodeficiency virus (FIV) infection

Prevention of infection 2 : immunisation. How infection influences the host : viruses. Peter

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Improving Influenza vaccines: Looking ahead

Evaluation of Feline Immunodeficiency Virus and Feline Leukemia

Genomic Alterations Associated with Persistent Infections by Equine Infectious Anaemia Virus, a Retrovirus

The humoral immune responses to IBV proteins.

RAISON D ETRE OF THE IMMUNE SYSTEM:

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

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

Feline Immunodeficiency Virus: an Interesting Model for AIDS Studies and an Important Cat Pathogen

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

RAISON D ETRE OF THE IMMUNE SYSTEM:

NEUTRALIZING ANTIBODY-MEDIATED CONTROL OF LENTIVIRUS INFECTION SANDRA D. TAYLOR

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

Construction and In Vitro Characterization of Attenuated Feline Immunodeficiency Virus Long Terminal Repeat Mutant Viruses

9/10/2018. Principles of Vaccination. Immunity. Antigen. September 2018

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

Supporting Information

Strategies for an HIV vaccine

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ

Are booster immunisations needed for lifelong hepatitis B immunity?

ABSTRACT. HIV-induced immunodeficiency may be mediated by the activation of

Appendix A A Technical Review of the Evidence for Adverse Reactions to HIV Vaccines

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

Preventive and therapeutic HIV vaccines. Markus Bickel Infektiologikum Frankfurt

Active and Passive Immunization for Avian Influenza Virus Infections

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Gene Vaccine Dr. Sina Soleimani

Detection of equine infectious anemia nucleic acid in asymptomatic carrier horses by nested PCR

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

Prokaryotic Biology. VIRAL STDs, HIV-1 AND AIDS

Received 13 July 2000/Accepted 27 January 2001

JANELLE MARISA NOVAK UNIVERSITY OF FLORIDA

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

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

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

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

Feline Panleukopenia Vaccine

SEROLOGICAL DIAGNOSIS OF VIRAL INFECTIONS:

The Cat with FIV: The Vaccine and Diagnostic Testing

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

Challenges in Designing HIV Env Immunogens for Developing a Vaccine

Ralf Wagner Paul-Ehrlich-Institut

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T

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

Department of Veterinary Pathology, University of Liverpool, Leahurst Campus, Neston, South Wirral, CH64 7TE, UNITED KINGDOM

Approaching a Cure Daniel R. Kuritzkes, MD

HIV INFECTION: An Overview

Differential Cell Tropism of Feline Immunodeficiency Virus Molecular Clones In Vivo

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

Inhibition of Feline Immunodeficiency Virus Infection in Vitro by Envelope Glycoprotein Synthetic Peptides

A Single Mutation within the V3 Envelope Neutralization Domain of Feline Immunodeficiency Virus Determines Its Tropism for CRFK Cells

Escaich Sonia, COO BIOSANTECH SA.

Regional Clustering of Shared Neutralization Determinants on Primary Isolates of Clade C Human Immunodeficiency Virus Type 1 from South Africa

Are we targeting the right HIV determinants?

Received 9 August 2004/Accepted 26 October 2004

Therapeutic Cancer Vaccines

on January 7, 2019 by guest

How HIV Causes Disease Prof. Bruce D. Walker

HIV/AIDS. Biology of HIV. Research Feature. Related Links. See Also

VIRAL TITER COUNTS. The best methods of measuring infectious lentiviral titer

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

MedChem 401~ Retroviridae. Retroviridae

Attribution: University of Michigan Medical School, Department of Microbiology and Immunology

Introduction. In the past 15 years, several technological advancements have open new perspectives and applications in the field of vaccinology.

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

The author hereby certifies that the use of any copyrighted material in the dissertation entitled:

Chapter 35 Active Reading Guide The Immune System

SV40 Detection and Transmission: Does SV40 Circulate in Human Communities?

Lentiviruses: HIV-1 Pathogenesis

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

Vaccination of Cats with Attenuated Feline Immunodeficiency Virus Proviral DNA Vaccine Expressing Gamma Interferon

Transcription:

Journal of General Virology (1999), 80, 761 765. Printed in Great Britain... SHORT COMMUNICATION Vaccination with experimental feline immunodeficiency virus vaccines, based on autologous infected cells, elicits enhancement of homologous challenge infection Jos A. Karlas, 1 Kees H. J. Siebelink, 1 Maartje A. v. Peer, 1 Willem Huisman, 1 Anne Marie Cuisinier, Guus F. Rimmelzwaan 1 and Albert D. M. E. Osterhaus 1 1 Institute of Virology, Erasmus University Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands Virbac Laboratories, BP 7, 06511, Carros, Cedex, France Cats were vaccinated with fixed autologous feline immunodeficiency virus (FIV)-infected cells in order to present viral proteins to the immune system of individual cats in an MHC-matched fashion. Upon vaccination, a humoral response against Gag was induced. Furthermore, virus-neutralizing antibodies were detected in a Crandell feline kidney cell-based neutralization assay, but not in a neutralization assay based on primary peripheral blood mononuclear cells. Despite the induction of these FIV-specific responses, vaccinated cats were not protected. Instead, accelerated virus replication was found, an observation similar to what previous experiments using other vaccine candidates have shown. Here, the results of the present study are discussed in the light of enhancement of lentivirus infections as a complicating factor in lentivirus vaccine development. Feline immunodeficiency virus (FIV) infection of the cat serves as a suitable model to study the pathogenesis of human immunodeficiency virus (HIV) infection and has been used to evaluate lentivirus vaccine development strategies (Willett et al., 1997). Several of these have been based on the use of envelope glycoproteins of FIV. Envelope-based lentivirus vaccines, however, may induce enhanced rather than decreased susceptibility to challenge infection or disease (Hosie et al., 199; Lombardi et al., 1994; Wang et al., 1994; Siebelink et al., 1995; Montelaro et al., 1996; Richardson et al., 1997). We have previously shown that cats vaccinated with recombinant envelope protein-based candidate vaccines exhibited an accelerated viraemia upon challenge infection, which was most likely mediated by virus-specific antibodies (Siebelink et al., Author for correspondence: Albert Osterhaus. Fax 31 10 4365145. e-mail Osterhaus viro.fgg.eur.nl 1995). Antibody-mediated enhancement of HIV-1 in vitro proved to be related to the syncytium-inducing phenotype of the virus, which is directly related to the envelope glycoprotein structure (Allan et al., 1990; Schutten et al., 1995; Kostrikis et al., 1996). In the present study, we have vaccinated cats with fixed autologous FIV-infected or uninfected peripheral blood mononuclear cells (PBMC) and challenged with the homologous molecular clone FIV-19k1. This approach was chosen to take advantage of the induction of cell-mediated immunity, since antigen presentation would take place in an MHCmatched fashion in each individual cat. We and others have recently shown that cellular immune responses against lentiviral proteins are of major importance in controlling lentivirus replication and disease (Flynn et al., 1995, 1996; Hosie et al., 1996; van Baalen et al., 1997; Ogg et al., 1998). Since we showed that paraformaldehyde is a relatively mild fixative for lentivirus membrane glycoproteins (Hulskotte et al., 1997), we speculate that the envelope protein in this candidate vaccine was indeed presented in a way that closely resembles the native form, which may not always be the case with subunit vaccines. The HIV-1 envelope proteins of primary isolates are organized in oligomeric structures, which may be necessary for the induction of effective neutralizing antibodies (Poignard et al., 1996). Also, when using this strategy we could exclude any Fig. 1. Kinetics of the antibody titre developed against the Gag protein. Cats no., 9 and 1 were vaccinated with fixed autologous FIV-19k1- infected PBMC. Cats no. 7 and 13 were vaccinated with fixed autologous uninfected PBMC. Small arrows, time-points of vaccination; large arrow, time-point of challenge. 0001-5897 1999 SGM HGB

J. A. Karlas and others VNA titer 10000 1000 100 <10 9 1 7 13 vaccinated controls Weeks postvaccination 0 4 6 8 16 18 Fig.. Virus-neutralizing antibody titres of the individual cats at different weeks post-vaccination as measured in the CrFK virus-neutralization assay. Cats no., 9 and 1 were vaccinated with fixed autologous FIV-19k1-infected PBMC. Cats no. 7 and 13 were vaccinated with fixed autologous uninfected PBMC. beneficial role of antibodies against cellular proteins in protection against infection as has been described in simian immunodeficiency virus vaccine studies (Stott, 1991; Osterhaus et al., 199). PBMC of five 6-month-old cats (nos, 7, 9, 1, 13) were obtained and cultured as described previously (Siebelink et al., 1995) and were infected after 7 days with 1000 CID of molecular clone FIV-19k1 (Siebelink et al., 199). In parallel, cultures with uninfected cells from each cat were maintained. Virus production was measured in an antigen ELISA (Siebelink et al., 1990). As soon as FIV antigen was detected in the culture supernatant, the percentage of infected cells was determined by an immunofluorescence assay, using serum of an experimentally FIV-infected specific-pathogen-free (SPF) cat. The number of infected PBMC for cats no., 9 and 1 was approximately 5%, whereas this was found to be approximately 1% for cats no. 7 and 13. This variability in the in vitro rate is common in our experience when infection is allowed to progress for a limited period of time. This observation is probably due to minor differences in culture conditions. Subsequently, infected as well as uninfected cells from all cats were inactivated with 1 5% paraformaldehyde as previously described (Yamamoto et al., 1991). Complete inactivation of the vaccines was confirmed in vitro by co-cultivation of the vaccines with autologous cells and in vivo by attempting to reisolate FIV from the PBMC of cats following two inoculations with fixed autologous FIV-infected cells. Neither method led to antigen production as measured in an antigen ELISA (Siebelink et al., 1990). Three cats (nos, 9 and 1) were vaccinated with fixed autologous FIV-19k1-infected cells. The two other cats (nos 7 and 13) served as controls and received fixed autologous non-infected cells. The cats were vaccinated six times at weeks 0,, 4, 6, 8 and 16 intravenously and subcutaneously, each time with 5 10 cells. Challenge infection was performed intramuscularly with 10 CID of molecular clone FIV-19k1 at week 18. Blood samples were collected at weekly intervals during the first 4 weeks and at week 6 and 10 post-challenge. We determined the development of Gag-specific antibodies in plasma by ELISA as described before (Siebelink et al., 1995). All cats vaccinated with fixed autologous FIV-infected cells developed a Gagspecific antibody response after three vaccinations (Fig. 1). On the day of challenge the titres of the three vaccinated cats varied from 400 to 1600, while the two controls remained sero-negative. Antibody responses measured against two Env peptides (SU and TM) were also determined as previously described (Siebelink et al., 1995) and were negative for all cats except at one time-point for cat no. 1, which showed an ELISA titre of 00 after three vaccinations (data not shown). Virus-neutralizing antibody titres were determined using two systems: the first is based on inhibition of infection of Crandell feline kidney (CrFK) cells with CrFK cell-adapted virus FIV- AM6c (Siebelink et al., 1995), and the second is based on inhibition of infection of primary PBMC with molecular clone FIV-19k1 (Siebelink et al., 1993). In the assay based on CrFK cells, virus-neutralizing antibodies could be detected in sera of the cats vaccinated with fixed autologous FIV-infected cells from the second vaccination onward, reaching titres of 640 180 on the day of challenge (Fig. ). FIV-neutralizing antibodies could not be demonstrated in the two control cats before challenge. No virus-neutralizing activity could be detected in the serum of any cat on the day of challenge in the assay based on primary PBMC. Gag-specific antibodies became detectable in the control cats at 10 weeks post-challenge, reaching a titre of 1600 in both cats. In the vaccinated cats, an anamnestic response against Gag was evident from weeks post-challenge onward (Fig. 1). These titres increased substantially in the following weeks, ranging from 800 to 13000, except for cat no. 1, which showed a lower titre rise upon challenge. Cell-associated virus loads were determined with an infectious centre test as previously described (Siebelink et al., 1995). In the cats vaccinated with fixed autologous FIVinfected cells, 10 36 and 30 80 FIV-infected cells per 10 PBMC were demonstrated and 3 weeks post-challenge HGC

Enhanced infection upon FIV vaccination Cat no. 9 1 7 13 Fig. 3. The upper part of the figure shows the numbers of FIV-infected cells per 10 6 PBMC at different time-points postchallenge. Serially diluted PBMC samples (10 3,3 10 3 and 10 4 cells) from the vaccinated and challenged cats were cocultivated with 10 5 ConA- and IL--stimulated PBMC from an SPF cat in ten duplicate wells. After 3 weeks the culture supernatants were tested for the presence of FIV antigen by ELISA. The numbers of infected cells in the PBMC were calculated by assuming that one infected cell would give rise to antigen production. The lower part represents the presence of proviral DNA in PBMC of the cats at different time-points post-challenge, as determined with a nested gag PCR. Primer pair FIVDP1 (5 GGCCCTCCACAGGCATATCC) and FIVDP (5 GCATTTTATATCCTGGTGAGCC) was used for the first reaction and primer pair FIVDP3 (5 GGCAAGAGAAGGACTAGGAGG) and FIVDP4 (5 GCACAGCTCGAGGAGACTTAGC) was used for the nested reaction., Negative in PCR;, positive in PCR; NT, not tested. Cats no., 9 and 1 were vaccinated with fixed autologous FIV-19k1-infected PBMC. Cats no. 7 and 13 were vaccinated with fixed autologous uninfected PBMC. respectively, whereas in the control cats no FIV-infected cells could be detected (Fig. 3). From 4 weeks post-challenge onward FIV-infected cells could also be determined in the two control cats. From 6 weeks post-challenge onward no differences in virus load between the two groups could be demonstrated. These observations were confirmed using a nested PCR based on the gag gene of FIV (Fig. 3). The cats vaccinated with fixed autologous FIV-19k1-infected PBMC contained proviral DNA in PBMC from weeks post-challenge onward, whereas proviral DNA in the control cats could be shown from 4 weeks post-challenge onward. To determine whether antibodies were the cause of the observed enhancement, we purified immunoglobulins from plasma of both groups of cats, collected on the day of challenge, by means of protein A affinity chromatography. Naı ve kittens were inoculated with 10 ml of whole plasma or an equivalent amount of purified immunoglobulins and subsequently challenged intramuscularly with 10 CID of molecular clone FIV-19k1. Blood samples were taken at the same time-points as described for the vaccinated cats. The virus loads of the cats were monitored using the infectious centre test and PCR. No differences in the kinetics of FIV infection and virus loads were observed between the four groups of cats. Although we did not show the induction of virus-specific MHC class I-restricted CTL, we may expect that in these experiments, presentation of the envelope protein of FIV in its natural form and presentation of antigenic peptides derived thereof by matched MHC molecules did indeed result in the induction of these CTL. Consequently, if we assume that such CTL contribute to protective immunity we would have expected that this type of immunization would have led to protective immunity against a homologous challenge infection with molecularly cloned FIV. Instead, an accelerated viraemia was observed after challenge infection. Enhancement of challenge infection was observed in a previous study, after vaccination of cats with FIV subunit vaccines (Siebelink et al., 1995) based on recombinant envelope protein incorporated into immune-stimulating complexes (iscom) (Rimmelzwaan et al., 1994). Vaccine-induced envelope protein-specific antibody titres, as measured in the virus-neutralization assay based on CrFK cells, were similar in both studies. However, antibody titres directed against two peptides derived from the envelope protein were much lower in the cats vaccinated with fixed autologous FIV-infected cells than in the cats vaccinated with FIV Env iscom, which may explain the failure to transfer the phenomenon of accelerated viraemia to naı ve kittens by means of plasma and purified immunoglobulins from this plasma in the present study. Besides the role of complement- and Fcreceptors in antibody-mediated enhancement, it has recently been described that the virus phenotype may also be involved (Schutten et al., 1995). This was shown with monoclonal antibodies and virtually identical virus isolates derived from one patient. Some monoclonal antibodies neutralized syncytium-inducing viruses, whereas the replication of a non- HGD

J. A. Karlas and others syncytium-inducing variant proved to be enhanced. A similar mechanism may play a role in the vaccination studies performed in the equine infectious anaemia virus model, where protection was found against homologous challenge after vaccination with recombinant envelope protein. In contrast, enhancement of infection was found in animals challenged with a heterologous virus (Montelaro et al., 1996). This principle may also have played a role in our previous study, in which cats were vaccinated with recombinant envelope protein incorporated into iscom and where a biological FIV isolate was used for the challenge infection. This isolate contains a swarm of viruses, including the highly homologous molecular clones FIV-19k1 and FIV-19k3 (Siebelink et al., 199). It is of interest to note that FIV-19k1 is neutralized by its homologous serum, while FIV-19k3 was enhanced by the same antiserum (K. Siebelink, personal communication), indicating that there is a delicate balance between enhancement and neutralization. In the present study, however, it is unlikely that this mechanism of heterotypic enhancement of infection played a role, since a homologous molecular clone was used for challenge. Recently immune activation has been suggested as a possible mechanism for enhanced replication of FIV in a vaccination study (Richardson et al., 1997). Furthermore, immune activation, including the activation of T-cells, seems to play a role in HIV- 1-infected individuals who are vaccinated against opportunistic pathogens and who show a rise in HIV-1 virus load in plasma as a consequence of this (Ho, 199; Staprans et al., 1995; Brichacek et al., 1996; Goletti et al., 1996). It may be speculated that in the present study, in which a challenge infection was carried out only weeks after the last booster vaccination, immune activation may also have played a role. To evaluate this possibility, we determined the induction of IFN-γ and IL- 4 in PBMC after vaccination and challenge in both groups of cats. No differences in cytokine expression were observed, suggesting that immune activation did not play a role in the enhanced virus replication. However, cytokines other than IFN-γ and IL-4 may have played a role in the observed enhancement. Recently, it has been described that IL-10 upregulates expression of CCR-5 (Sozzani et al., 1998), the chemokine receptor involved in infection of macrophagetropic variants of HIV-1 (Choe et al., 1996). This resulted in an enhanced in vitro infection of peripheral blood lymphocytes with HIV-1. Similar mechanisms, affecting primary or secondary receptors used by FIV, may be involved in the outcome of the study described here. The results obtained in the present experiments and in other studies referred to above show that enhancement of lentivirus infections can be a major problem in vaccine development. Several mechanisms can form the basis of this and it is important that the contribution of these mechanisms are addressed. The authors would like to thank Bettina Hansen for statistical analysis of the data, and Rob van Herwijnen from the European Veterinary Laboratory, Woerden, The Netherlands for the ELISAs. Furthermore, we thank Dr Ellen Hulskotte for fruitful discussions and Ger van der Water for continuous support. References Allan, J. S., Strauss, J. & Buck, D. W. (1990). Enhancement of SIV infection with soluble receptor molecules. Science 47, 1084 1088. Brichacek, B., Swindells, S., Janoff, E. N., Pirruccello, S. & Stevenson, M. (1996). Increased plasma human immunodeficiency virus type 1 burden following antigenic challenge with pneumococcal vaccine. Journal of Infectious Diseases 174, 1191 1199. Choe, H., Farzan, M., Sun, Y., Sullivan, N., Rollins, B., Ponath, P. D., Wu, L., Mackay, C. R., LaRosa, G., Newman, W., Gerard, N., Gerard, C. & Sodroski, J. (1996). The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85, 1135 1148. Flynn, J. N., Beatty, J. A., Cannon, C. A., Stephens, E. B., Hosie, M. J., Neil, J. C. & Jarrett, O. (1995). Involvement of gag- and env-specific cytotoxic T lymphocytes in protective immunity to feline immunodeficiency virus. AIDS Research and Human Retroviruses 11, 1107 1111. Flynn, J. N., Keating, P., Hosie, M. J., Mackett, M., Stephens, E. B., Beatty, J. A., Neil, J. C. & Jarrett, O. (1996). Env-specific CTL predominate in cats protected from feline immunodeficiency virus infection by vaccination. Journal of Immunology 157, 3658 3665. Goletti, D., Weissman, D., Jackson, R. W., Graham, N. M., Vlahov, D., Klein, R. S., Munsiff, S. S., Ortona, L., Cauda, R. & Fauci, A. S. (1996). Effect of Mycobacterium tuberculosis on HIV replication. Role of immune activation. Journal of Immunology 157, 171 178. Ho, D. D. (199). HIV-1 viraemia and influenza. Lancet 339, 1549. Hosie, M. J. & Flynn, J. N. (1996). Feline immunodeficiency virus vaccination: characterization of the immune correlates of protection. Journal of Virology 70, 7561 7568. Hosie, M. J., Osborne, R., Reid, G., Neil, J. C. & Jarrett, O. (199). Enhancement after feline immunodeficiency virus vaccination. Veterinary Immunology and Immunopathology 35, 191 197. Hulskotte, E. G. J., Dings, M. E. M., Norley, S. G. & Osterhaus, A. D. M. E. (1997). Chemical inactivation of recombinant vaccinia viruses and the effect on antigenicity and immunogenicity of recombinant simian immunodeficiency virus envelope glycoproteins. Vaccine 15, 1839 1845. Kostrikis, L. G., Cao, Y., Ngai, H., Moore, J. P. & Ho, D. D. (1996). Quantitative analysis of serum neutralisation of human immunodeficiency virus type 1 from subtypes A, B, C, D, E, F, and I: lack of direct correlation between neutralisation serotypes and genetic subtypes and evidence for prevalent serum-dependent infectivity enhancement. Journal of Virology 70, 445 458. Lombardi, S., Garzelli, C., Pistello, M., Massi, C., Matteucci, D., Baldinotti, F., Cammarota, G., da Prato, L., Bandecchi, P., Tozzini, F. & Bendinelli, M. (1994). A neutralising antibody-inducing peptide of the V3 domain of feline immunodeficiency virus envelope glycoprotein does not induce protective immunity. Journal of Virology 68, 8374 8379. Montelaro, R. C., Grund, C., Raabe, M., Woodson, B., Cook, R. F., Cook, S. & Issel, C. J. (1996). Characterization of protective and enhancing immune responses to equine infectious anemia virus resulting from experimental vaccines. AIDS Research and Human Retroviruses 1, 413 415. Ogg, G. S., Jin, X., Bonhoeffer, S., Dunbar, P. R., Nowak, M. A., Monard, S., Segal, J. P., Cao, Y., Rowland-Jones, S. L., Cerundolo, V., Hurley, A., Markowitz, M., Ho, D. D., Nixon, D. F. & McMichael, A. J. (1998). Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 79, 103 106. Osterhaus, A., de Vries, P. & Heeney, J. (199). AIDS vaccine developments. Nature 355, 684 685. HGE

Enhanced infection upon FIV vaccination Poignard, P., Klasse, P. J. & Sattentau, Q. J. (1996). Antibody neutralization of HIV-1. Immunology Today 17, 39 46. Richardson, J., Moraillon, A., Baud, S., Cuisinier, A. M., Sonigo, P. & Pancino, G. (1997). Enhancement of feline immunodeficiency virus (FIV) infection after DNA vaccination with the FIV envelope. Journal of Virology 71, 9640 9649. Rimmelzwaan, G. F., Siebelink, K. H. J., Huisman, R. C., Moss, B., Francis, M. J. & Osterhaus, A. D. M. E. (1994). Removal of the cleavage site of recombinant feline immunodeficiency virus envelope protein facilitates incorporation of the surface glycoprotein in immune-stimulating complexes. Journal of General Virology 75, 097 10. Schutten, M., Andeweg, A. C., Bosch, M. L. & Osterhaus, A. D. (1995). Enhancement of infectivity of a non-syncytium inducing HIV-1 by scd4 and by human antibodies that neutralize syncytium inducing HIV-1. Scandinavian Journal of Immunology 41, 18. Siebelink, C. H., Windrich, R. W., Chu, I., Groen, J., Weijer, K., UytdeHaag, F. G. & Osterhaus, A. D. (1990). An enzyme linked immunosorbent assay (ELISA) for the detection of feline immunodeficiency virus (FIV) antigen in cell culture and FIV specific antibodies in feline serum. Development in Biological Standardization 7, 189 196. Siebelink, K. H., Chu, I. H., Rimmelzwaan, G. F., Weijer, K., Osterhaus, A. D. & Bosch, M. L. (199). Isolation and partial characterization of infectious molecular clones of feline immunodeficiency virus obtained directly from bone marrow DNA of a naturally infected cat. Journal of Virology 66, 1091 1097. Siebelink, K. H., Rimmelzwaan, G. F., Bosch, M. L., Meloen, R. H. & Osterhaus, A. D. (1993). A single amino acid substitution in hypervariable region 5 of the envelope protein of feline immunodeficiency virus allows escape from virus neutralization. Journal of Virology 67, 0 08. Siebelink, K. H., Tijhaar, E., Huisman, R. C., Huisman, W., de Ronde, A., Darby, I. H., Francis, M. J., Rimmelzwaan, G. F. & Osterhaus, A. D. (1995). Enhancement of feline immunodeficiency virus infection after immunization with envelope glycoprotein subunit vaccines. Journal of Virology 69, 3704 3711. Sozzani, S., Ghezzi, S., Iannolo, G., Luini, W., Borsatti, A., Polentarutti, N., Sica, A., Locati, M., Mackay, C., Wells, T. N., Biswas, P., Vicenzi, E., Poli, G. & Mantovani, A. (1998). Interleukin 10 increases CCR5 expression and HIV infection in human monocytes. Journal of Experimental Medicine 187, 439 444. Staprans, S. I., Hamilton, B. L., Follansbee, S. E., Elbeik, T., Barbosa, P., Grant, R. M. & Feinberg, M. B. (1995). Activation of virus replication after vaccination of HIV-1-infected individuals. Journal of Experimental Medicine 18, 177 1737. Stott, E. J. (1991). Anti-cell antibody in macaques. Nature 353, 393. van Baalen, C. A., Pontesilli, O., Huisman, R. C., Geretti, A. M., Klein, M. R., de Wolf, F., Miedema, F., Gruters, R. A. & Osterhaus, A. D. M. E. (1997). Human immunodeficiency virus type 1 Rev- and Tat-specific cytotoxic T lymphocyte frequencies inversely correlate with rapid progression to AIDS. Journal of General Virology 78, 1913 1918. Wang, S. Z., Rushlow, K. E., Issel, C. J., Cook, R. F., Cook, S. J., Raabe, M. L., Chong, Y. H., Costa, L. & Montelaro, R. C. (1994). Enhancement of EIAV replication and disease by immunization with a baculovirusexpressed recombinant envelope surface glycoprotein. Virology 199, 47 51. Willett, B. J., Flynn, J. N. & Hosie, M. J. (1997). FIV infection of the domestic cat: an animal model for AIDS. Immunology Today 18, 18 189. Yamamoto, J. K., Okuda, T., Ackley, C. D., Louie, H., Pembroke, E., Zochlinski, H., Munn, R. J. & Gardner, M. B. (1991). Experimental vaccine protection against feline immunodeficiency virus. AIDS Research and Human Retroviruses 7, 911 9. Received 1 August 1998; Accepted October 1998 HGF