A DNA/MVA-based candidate human immunodeficiency virus vaccine for Kenya induces multi-specific T cell responses in rhesus macaques

Similar documents
Magnitude and Diversity of Cytotoxic-T-Lymphocyte Responses Elicited by Multiepitope DNA Vaccination in Rhesus Monkeys

Received 14 July 2005/Accepted 3 September 2005

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

Received May 1, 2002; returned to author for revision May 20, 2002; accepted June 3, 2002

Immunological transitions in response to antigenic mutation during viral infection

Anti-SIV Cytolytic Molecules in Pigtail Macaques

Design and tests of an HIV vaccine

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

Comparison of Human Immunodeficiency Virus Antigens as Stimulants for Lymphocyte Proliferation Assays

Analysis of Total Human Immunodeficiency Virus (HIV)-Specific CD4 and CD8 T-Cell Responses: Relationship to Viral Load in Untreated HIV Infection

Emergence and Kinetics of Simian Immunodeficiency Virus-Specific CD8 T Cells in the Intestines of Macaques during Primary Infection

Received 19 March 2003/Accepted 28 May 2003

Efficacy of DNA and Fowlpox Virus Priming/Boosting Vaccines for Simian/Human Immunodeficiency Virus

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

Received 16 February 2004/Accepted 5 June 2004

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

Emergence of cytotoxic T lymphocyte escape mutations in nonpathogenic simian immunodeficiency virus infection

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

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

Comparative Efficacy of Subtype AE Simian-Human Immunodeficiency Virus Priming and Boosting Vaccines in Pigtail Macaques

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

staining and flow cytometry

Received 17 March 2003/Accepted 16 July 2003

Received 26 September 2003/Accepted 22 December 2003

Vaccine-Induced T Cells Control Reversion of AIDS Virus Immune Escape Mutants

Supporting Information

HIV-1 infection is characterized by an early peak of viremia

AND BRUCE D. WALKER 1

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

CD8 memory, immunodominance, and antigenic escape

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Potential cross reactions between HIV 1 specific T cells and the microbiome. Andrew McMichael Suzanne Campion

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers

Functionally Inert HIV-specific Cytotoxic T Lymphocytes Do Not Play a Major Role in Chronically Infected Adults and Children

Does Cytolysis by CD8 + T Cells Drive Immune Escape in HIV Infection?

Lecture 11. Immunology and disease: parasite antigenic diversity

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

DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04)

/JVI Updated information and services can be found at:

Heterologous Envelope Immunogens Contribute to AIDS Vaccine Protection in Rhesus Monkeys

Adjuvanting a DNA vaccine with a TLR9 ligand plus Flt3 ligand results in enhanced cellular immunity against the simian immunodeficiency virus

Why are validated immunogenicity assays important for HIV vaccine development?

Received 28 July 1997/Accepted 3 November 1997

JVI Accepts, published online ahead of print on 7 March 2007 J. Virol. doi: /jvi

Received 17 April 2003/Accepted 28 June 2003

Strategies for an HIV vaccine

Progress on new vaccine strategies against chronic viral infections

Subtype AE HIV-1 DNA and recombinant Fowlpoxvirus vaccines encoding five shared HIV-1 genes: safety and T cell immunogenicity in macaques

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

Optimizing Intracellular Flow Cytometry:

... Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA

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

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

Protection afforded by live attenuated SIV is associated with rapid killing kinetics of CTLs

Received 18 January 2005/Accepted 18 April 2005

Cellular Immunity in Aging and HIV: Correlates of Protection. Immune Senescence

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

Perspectives of AIDS Vaccine Development: T Cell-based Vaccine

MHC Tetramers and Monomers for Immuno-Oncology and Autoimmunity Drug Discovery

Introduction. University of Melbourne, Parkville, Vic., Australia, 2 Department of Pathology and Laboratory

Human Immunodeficiency Virus Type-1 Myeloid Derived Suppressor Cells Inhibit Cytomegalovirus Inflammation through Interleukin-27 and B7-H4

PRACTICE POINTS AIDS Vaccine 2001: Looking to the Future

How HIV Causes Disease Prof. Bruce D. Walker

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

The origins of acquired immunity against

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

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

RAISON D ETRE OF THE IMMUNE SYSTEM:

JOURNAL OF VIROLOGY, Sept. 1999, p Vol. 73, No. 9. Copyright 1999, American Society for Microbiology. All Rights Reserved.

T cell Vaccine Strategies for HIV, the Virus. With a Thousand Faces

Expansion of pre-existing, lymph node-localized CD8 + T cells during supervised treatment interruptions in chronic HIV-1 infection

HIV Anti-HIV Neutralizing Antibodies

This information is current as of August 20, Nina Malkevitch, L. Jean Patterson, Kristine Aldrich, Ersell

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

CD40L-Adjuvanted DNA/MVA SIV Vaccine Enhances Protection. Against Neutralization Resistant Mucosal SIV Infection

RESEARCH ARTICLE Engineering RENTA, a DNA prime-mva boost HIV vaccine tailored for Eastern and Central Africa

Received 31 March 2011/Accepted 19 July 2011

Human Immunodeficiency Virus Type 1 (HIV-1)-Specific CD8 -T-Cell Responses for Groups of HIV-1-Infected Individuals with Different HLA-B*35 Genotypes

Immunoprophylaxis against AIDS in macaques with a lentiviral DNA vaccine

Global Dysfunction of CD4 T-Lymphocyte Cytokine Expression in Simian-Human Immunodeficiency Virus/SIV-Infected Monkeys Is Prevented by Vaccination

Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals

Received 22 April 2002/Accepted 28 June 2002

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.

Are we targeting the right HIV determinants?

Principle of the FluoroSpot assay. Anti-tag mab-green. Streptavidin-Red. Detection mab-tag. Detection mab-biotin. Analyte. Analyte.

Received 19 September 2007/Accepted 21 November 2007

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

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

Antiviral therapy for HIV-infected patients has greatly improved

T-Pharmacytes for the Targeted Eradication of HIV Reservoirs

IOM Immunization Safety Review 11/12/2001. Immunological Competition and the Infant Immune Response to Vaccines

Mechanisms of antagonism of HIVspecific CD4+ T cell responses BSRI

C. Jane Dale 1,2, Anne Zhao 1,2, Stephen L. Jones 3, David B. Boyle 4, Ian A. Ramshaw 5, Stephen J. Kent 1,2. Introduction

Viral CTL Escape Mutants Are Generated in Lymph Nodes and Subsequently Become Fixed in Plasma and Rectal Mucosa during Acute SIV Infection of Macaques

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific

Review. Tomáš Hanke, Andrew J. McMichael and Lucy Dorrell

IL-12 and GM-CSF in DNA/MVA Immunizations against HIV-1 CRF12_BF Nef Induced T-Cell Responses With an Enhanced Magnitude, Breadth and Quality

HCV Vaccine where do we stand? U. Spengler University of Bonn

Transcription:

Journal of General Virology (2002), 83, 75 80. Printed in Great Britain... SHORT COMMUNICATION A DNA/MVA-based candidate human immunodeficiency virus vaccine for Kenya induces multi-specific T cell responses in rhesus macaques Edmund G.-T. Wee, Sandip Patel, Andrew J. McMichael and Toma s Hanke MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS, UK The minimum requirement for candidate human immunodeficiency virus (HIV) vaccines to enter clinical evaluation in humans should be their demonstrable immunogenicity in non-human primates: induction of antibodies neutralizing primary HIV isolates or elicitation of broad T cellmediated immune responses. Here, we showed in rhesus macaques that the very same vaccines that had entered clinical trials in Oxford and Nairobi, plasmid pthr.hiva DNA and recombinant modified vaccinia virus Ankara MVA.HIVA in a prime-boost protocol (Hanke & McMichael, Nature Medicine 6, 951 955, 2000), induced cellular immune responses specific for multiple HIV-derived epitopes. This was demonstrated by using the intracellular cytokine staining and ELISPOT assays detecting interferon-γ and pools of peptides employed in the clinical studies. These results have both boosted our expectations for the performance of these vaccines in humans and increased our confidence about the choice of these assays as the primary readouts in the on-going human trials. Close to 40 million people have been infected with human immunodeficiency virus type 1 (HIV-1), many of whom will develop AIDS. At least two out of three of these individuals live in sub-saharan Africa, where education programs have had virtually no effect in slowing the spread of HIV, and the highly active antiretroviral therapy available in industrialized countries is too costly and complex and in any case fails to clear the virus from the body. Moreover, 90% of the infected people are not aware of their HIV status. Under these circumstances, the best hope for controlling the HIV pandemic is effective prophylactic vaccination. While the inaccessibility of neutralizing epitopes on the Author for correspondence: Toma s Hanke. Fax 44 1865 222502. e-mail thanke molbiol.ox.ac.uk primary HIV isolates has been a major impediment to the development of envelope-based vaccines, considerable evidence has accumulated which indicates that CD8+ T cells are crucial for the control of HIV infection. First, the kinetics of the CD8+ cytotoxic T lymphocyte (CTL) response in acute and chronic HIV infections and inverse relationship with HIV virus load implied an anti-hiv effect in vivo (Borrow et al., 1994; Kent et al., 1997; Koup et al., 1994; Wilson et al., 2000). Second, CTL selected virus escape mutants (Borrow et al., 1994, 1997; Goulder et al., 1997; Haas et al., 1996; Koenig et al., 1995; Phillips et al., 1991; Price et al., 1997; Wilson et al., 1999; Wolinsky et al., 1996) and do so probably in most patients. Third, a temporary removal of CD8+ cells in acute and chronic simian immunodeficiency virus (SIV) infections by the in vivo use of anti-cd8 antibodies caused a marked rise in the virus load and failure to control acute viraemia (Jin et al., 1999; Schmitz et al., 1999). Fourth, CTL reduced and could eliminate HIV from cell cultures (Price et al., 1998; Stranford et al., 1999; Wagner et al., 1998; Yang et al., 1997; Zhang et al., 1996). Fifth, several vaccines induced protection in monkeys independent of virus-neutralizing antibodies (Amara et al., 2001; Barouch et al., 2000; Gallimore et al., 1995; Kent et al., 1998; Robinson et al., 1999). Sixth, the identification of exposed seronegative uninfected individuals with CTL responses against HIV raised the possibility that CTL responses have at least contributed to the prevention of infection (Rowland-Jones & McMichael, 1995). Finally, CTL have been shown to control many other virus infections in both mice and humans and could protect against new infections (Blaney et al., 1998; Fu et al., 1999; Schneider et al., 1998). This growing list of experimental data together with new potent technologies capable of inducing high levels of circulating virus-specific CD8+ T cells have strengthened the belief that a successful HIV vaccine is possible and within reach (Hanke, 2001). Murine models of chronic viral infections strongly support the requirement of virus-specific CD4+ T helper cells for efficient generation of anti-viral immune responses, including CTL (Matloubian et al., 1994; von Herrath et al., 1996). An analogous situation is seen in HIV-infected people, in whom a progressive depletion of CD4+ T cells weakens the responses to both HIV and opportunistic infections (Rosenberg et al., 0001-8058 2002 SGM HF

E. G.-T. Wee and others 1999). Thus, a protective HIV vaccine should induce HIVspecific T helper cells as well as CTL. Our original finding that a successive immunization with DNA- and modified vaccinia virus Ankara (MVA)-based vaccines expressing a common immunogen is a potent way of inducing CD8+ CTL (Hanke et al., 1998a; Schneider et al., 1998) has been since reinforced by us and others (Allen et al., 2000; Amara et al., 2001; Hanke et al., 1999a, b). The evaluations of the safety and immunogenicity of this approach have already commenced in healthy low-risk volunteers in Oxford and Nairobi, with the view of proceeding into a highrisk cohort in Kenya to test the vaccine efficacy. For this reason, the immunogen HIVA delivered by DNA and MVA was tailor-designed to match HIV clade A, the most prevalent local HIV strain. It consists of a consensus HIV clade A p24 p17 and a string of CTL epitopes (Hanke & McMichael, 2000), which includes the SIV p11c, C-M sequence presented by the rhesus monkey Mamu-A*01 molecule (Allen et al., 1998). Here, induction of multi-specific CD4+ and CD8+ T cell responses in rhesus macaques using clinical preparations of the pthr.hiva and MVA.HIVA vaccines in a prime-boost regimen was demonstrated. Furthermore, the elicited immune responses were measured using the same assays which are employed in the clinical trials, thus validating both the vaccine approach and detection of elicited immune responses in a pre-clinical setting in non-human primates. Five rhesus macaques (Macaca mulatta) positive for the Mamu-A*01 allele of the major histocompatibility complex (MHC) received initially a total of four successive immunizations: two using the plasmid pthr.hiva DNA and two with recombinant MVA.HIVA at 3 week intervals. In particular, two animals (Gaffa and Gilda) received 8 µg pthr.hiva epidermally using the Dermal XR particle delivery device (designated XR; PowderJect Vaccines, Inc.) or gene gun followed by needle-injected 5 10 p.f.u. of MVA.HIVA intradermally (i.d.) in 0 3 ml of 140 mm NaCl, 10 mm Tris HCl ph 7 7 solution, which is a dose similar to the immunizations in our previous macaque experiments employing a prototype multi-ctl epitope, here designated MXR (Hanke et al., 1999b). Three animals received human doses similar to those used or planned to be used in the clinical trials: one animal (Gloria) was vaccinated epidermally with 4 µg of pthr.hiva DNA and two animals (Gerry and Gege) were needle-injected intramuscularly (i.m.) with 500 µg of pthr.hiva in 0 5 ml of 140 mm NaCl, 0 5 mm Tris HCl ph 7 7 and 0 05 mm EDTA, followed for all three monkeys by 5 10 p.f.u. of MVA.HIVA i.d. in 0 1 ml of 140 mm NaCl and 10 mm Tris HCl ph 7 7. These doses were designated HXR and Him, respectively. Macaques Gloria and Gerry received an additional boost of 5 10 p.f.u. of MVA.HIVA i.d. on week 26. All immunizations and venipunctures were carried out under sedation with ketamine and the animals were clinically examined regularly. All procedures and care strictly conformed to the UK Home Office Guidelines. The combination of the pthr.hiva and MVA.HIVA vaccines focuses on the induction of cell-mediated immunity (Hanke & McMichael, 2000) and their immunogenicities in human volunteers are assessed in validated intracellular cytokine and ELISPOT assays estimating the frequencies of CD69+ IFN-γ+ cells and cells secreting IFN-γ, respectively. The intracellular cytokine assay is designed for detection of functional primarily CD4+, but also CD8+ cell populations, which upon antigen and antibody co-stimulation express IFNγ and activation marker CD69 (Maino & Picker, 1998; Suni et al., 1998). The assay was carried out according to the recommendations of the FastImmune Kit vendor (Becton Dickinson, cat. no. 340970) with a few modifications. Briefly, whole heparinized blood drawn from immunized macaques was incubated without any antigen, with staphylococcal enterotoxin B (SEB), HIV-1 p24 (IIIB) or pools 1 4 of 15- amino-acid (aa)-long peptides overlapping by 11 aa across the p24 p17 region of the HIVA immunogen plus a mixture of costimulatory anti-cd28 and anti-cd49d antibodies for an initial 2 h period at 37 C, 5% CO. Brefeldin A was then added to inhibit cytokine secretion and the samples were incubated for an additional 14 h before terminating the reaction with EDTA and the FACS fix solution. At this point, the blood was placed at 80 C until analysis. At convenience, samples were thawed, peripheral blood mononuclear cells (PBMC) permeabilized and incubated with mouse anti-huifn-γ FITCconjugated (Biosource International, cat. no. AHC4338), mouse anti-cd69 APC-conjugated (Becton Dickinson, cat. no. 340560) and mouse anti-hucd4 R-PE-conjugated (BD PharMingen, cat. no 556616) monoclonal antibodies. Addition of anti-hucd8 PerCP-conjugated (Becton Dickinson, cat. no.347314) antibodies allowed a parallel analysis of the CD8+ cellular responses. Arbitrarily, samples were considered positive if the percentage of stained cells increased after stimulation at least 3-fold compared to the no-antigen baseline. Thus, although the FACS analysis of blood from the immunized monkeys failed to demonstrate CD4+, but revealed CD8+ cell responses at week 12, all animals had in their circulation CD69- positive IFN-γ-producing CD4+ or CD8+ cells at week 30 (Table 1). Furthermore, the use of four peptide pools suggested that two animals responded to more than one and in one case at least four different epitopes (Table 1, Gilda, week 30). Note that approximately the first 2 and 1 2 of peptide pools correspond to p24 and that these pool responses on several occasions exceeded those of the p24 stimulation. This may be accounted for by differential processing of peptides versus protein antigens and the fact that p24 was of HIV-1 clade B. The IFN-γ release upon an HIVA-specific restimulation was assessed in an ELISPOT assay. The procedures and reagents of the MABTECH kit (cat. no. 3420M-2A) were used throughout. Briefly, PBMC were isolated on a Lymphoprep cushion and incubated at 37 C, 5% CO for 24 h with peptide p11c, C- M, of which the aa sequence is contained in the polyepitope portion of the HIVA immunogen (Hanke & McMichael, 2000). HG

Induction of broad T cell responses in monkeys Table 1. Antigen-specific induction of intracellular IFN-γ in PBMC CD4 PBMC CD8 PBMC Animal/dose* No Ag SEB p24 Pool 1 Pool 2 Pool 3 Pool 4 No Ag SEB p24 Pool 1 Pool 2 Pool 3 Pool 4 Week 12 Gaffa MXR 0 10 1 52 0 11 0 04 0 01 0 02 0 04 0 08 2 73 0 22 0 28 0 06 0 13 0 06 Gilda MXR 0 00 0 56 ND 0 00 0 00 0 01 0 01 0 09 2 13 ND 1 25 0 17 0 20 0 22 Gloria HXR 0 00 0 64 0 00 0 02 0 00 0 00 0 02 0 02 1 39 0 34 0 30 0 06 0 00 0 00 Gerry Him 0 00 0 50 0 03 0 00 0 00 ND 0 00 0 11 3 37 0 19 0 52 0 19 ND 0 09 Gege Him 0 00 1 63 0 00 0 00 0 00 0 00 0 00 0 08 2 64 0 13 0 30 0 18 0 14 0 18 Week 30 Gaffa MXR 0 08 1 09 0 00 0 02 0 00 0 00 0 22 0 11 2 39 0 09 0 07 0 03 0 04 0 37 Gilda MXR 0 08 1 48 0 26 0 22 0 20 0 29 0 16 0 00 2 19 0 25 0 19 0 14 0 08 0 14 Gloria HXR 0 00 1 79 0 01 0 00 0 04 0 00 0 03 0 01 2 08 0 16 0 15 0 11 0 02 0 06 Gerry Him 0 02 1 79 0 00 0 02 0 01 0 04 0 07 0 04 2 44 0 18 0 21 0 07 0 05 0 05 Gege Him 0 01 1 18 0 01 0 04 0 01 0 01 0 00 0 04 2 48 0 11 0 19 0 05 0 03 0 04 * Animals received a total of four immunizations with pthr.hiva (weeks 0 and 3) and MVA.HIVA (weeks 6 and 9). Data are shown as the percentage of CD4+ or CD8+ PBMC positive for CD69 and IFN-γ. Animals received additional boost with 5 10 p.f.u. of MVA.HIVA at week 26. ND, Not done. Table 2. Frequencies of IFN-γ-releasing PBMC upon specific peptide stimulation determined in an ELISPOT assay Week 24 In vitro Week 10 Animal/dose* restimulation Whole Whole CD8-depleted Gaffa MXR p11c, C-M 164 6 ND Pool 1 ND 200 ND Pool 2 ND 628 ND Pool 3 ND 473 ND Pool 4 ND 0 ND Gilda MXR p11c, C-M 132 10 0 Pool 1 ND 262 3 Pool 2 ND 652 20 Pool 3 ND 614 20 Pool 4 ND 103 0 Gloria HXR p11c, C-M 207 15 ND Pool 1 ND 135 ND Pool 2 ND 319 ND Pool 3 ND 1404 ND Pool 4 ND 1345 ND Gerry Him p11c, C-M 341 40 ND Pool 1 ND 40 ND Pool 2 ND 741 ND Pool 3 ND 704 ND Pool 4 ND 13 ND * Animals received a total of four immunizations with pthr.hiva (week 0 and 3) and MVA.HIVA (weeks 6 and 9). Assay was carried out on either whole or CD8+ cell-depleted PBMC. Frequencies are expressed as no. of SFU per 10 PBMC above no-peptide background. ND, Not done. HH

E. G.-T. Wee and others The released IFN-γ was captured by a monoclonal antibody immobilized on the bottom of assay wells, visualized by combination of a second monoclonal antibody coupled to an enzyme and a chromogenic substrate, and the spots were counted using the AID ELISpot Reader System (Autoimmun Diagnostika). It was found that 1 week after the last MVA.HIVA immunization four out of five vaccinated macaques responded to this Mamu-A*01-restricted epitope (Table 2). However, technical difficulties with the isolation of PBMC from Gege s blood might have occluded the detection of responses in this monkey. On week 34, only four animals were tested and all four showed responses against multiple epitopes. Magnetic-bead depletion of CD8+ cells from the PBMC of Gilda almost completely abolished the IFN-γ release suggesting that, at least in this macaque, the majority of IFNγ producing cells were CD8 positive. There was no incentive to further map HIV-1-derived epitopes in these macaques in which Mamu-A*01 was the only known MHC molecule. Thus, the vaccination induced multi-specific CD8+ T cell responses that lasted at least 21 weeks. No significant reactivity above background of PBMC with Mamu-A*01 p11c, C-M peptide tetrameric complexes was observed (not shown). However, 300 SFU per 10 PBMC seen in our macaques 1 week after the MVA.HIVA vaccination, i.e. at the peak of tetramer responses (Hanke et al., 1999b), would translate into 0 1% of CD8+ cells (Tan et al., 1999) which, for macaques, is at the limit of tetramer sensitivity. Note that this and our previous studies (Allen et al., 2000; Hanke et al., 1999b) showing high tetramer reactivity after the DNA MVA vaccination employed different immunogens [see Hanke et al. (1998b) and Hanke & McMichael (2000)], in which the Mamu- A*01-restricted p11c, C-M epitope is inserted between different adjacent aa sequences opening possibilities for differential processing and presentation. Indeed, in the present study, the p11c, C-M response was not dominant (Table 2) as it was shown for Mamu-A*01+ SIV-infected monkeys (Miller et al., 1991). Exactly matching CD8+ T cell responses were not detected by the intracellular cytokine and ELISPOT assays (Table 1 versus Table 2). However, although both these assays measure IFN-γ, they are not identical. Thus, the intracellular cytokine assay used a stimulation through CD28 CD49d plus the T cell receptor, the stimulation was carried out on whole blood, which although optimized by the vendor is likely to be less efficient than on isolated PBMC (hence the need for antibody co-stimulation), and the results are expressed as only CD69- positive cells showing intracellular IFN-γ. In contrast, the ELISPOT assay stimulated purified PBMC with peptide alone and the results included CD69-negative IFN-γ-releasing cells. Moreover, the assays were carried out at different time-points. In conclusion, we have confirmed in non-human primates the CD4+ and CD8+ T cell immunogenicities of the very vaccines that have entered clinical trials. Particularly encouraging is the fact that in all animals responses were elicited which were targeted at multiple HIV-derived epitopes and that these were detected by the readout assays employed in the ongoing human trials. The current work is being followed by a larger macaque study addressing the dosing and timing requirements of the two vaccine components for an efficient T cell induction. This line of experimentation is consistent with the principle of testing thoroughly candidate HIV vaccines in non-human primates before and or in parallel with their clinical evaluation. The authors would like to thank David Watkins for tissue typing the animals, Sarah Wolfensohn and Peter Thornton for their veterinary expertise, and Ian Jones NIBSC Centralized Facility for AIDS Reagents supported by EU Programme EVA contract (BMH4 97 2515) and MRC UK for generous gift of recombinant HIV-1 p24. Grant supports from MRC UK and International AIDS Vaccine Initiative are fully acknowledged. References Allen, T. M., Sidney, J., del Guercio, M.-F., Glickman, R. L., Lensmeyer, G. L., Wiebe, D. A., Pauza, C. D., Johnson, R. P., Sette, A. & Watkins, D. I. (1998). Characterization of the peptide-binding motif of a rhesus MHC class I molecule (Mamu-A*01) that binds an immunodominant CTL epitope from SIV. Journal of Immunology 160, 6062 6071. Allen, T. M., Vogel, T. U., Fuller, D. H., Monthe, B. R., Steffen, S., Boyson, J. E., Shipley, T., Fuller, J., Hanke, T., Sette, A., Altman, J. D., Moss, B., McMichael, A. J. & Watkins, D. I. (2000). Induction of AIDS virus-specific CTL activity in fresh, unstimulated peripheral blood lymphocytes from rhesus macaques vaccinated with a DNA prime modified vaccinia virus Ankara boost regimen. Journal of Immunology 164, 4968 4978. Amara, R. R., Villinger, F., Altman, J. D., Lydy, S. L., O Neil, S. P., Staprans, S. I., Montefiori, D. C., Xu, Y., Herndon, J. G., Wyatt, L. S., Candido, M. A., Kozyr, N. L., Earl, P. L., Smith, J. M., Ma, H. L., Grimm, B. D., Hulsey, M. L., Miller, J., McClure, H. M., McNicholl, J. M., Moss, B. & Robinson, H. L. (2001). Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA MVA vaccine. Science 292, 69 74. Barouch, D. H., Santra, S., Schmitz, J. E., Kuroda, M. J., Fu, T. M., Wagner, W., Bilska, M., Craiu, A., Zheng, X. X., Krivulka, G. R., Beaudry, K., Lifton, M. A., Nickerson, C. E., Trigona, W. L., Punt, K., Freed, D. C., Guan, L., Dubey, S., Casimiro, D., Simon, A., Davies, M. E., Chastain, M., Strom, T. B., Gelman, R. S., Montefiori, D. C., Lewis, M. G. & Letvin, N. L. (2000). Control of viremia and prevention of clinical AIDS in rhesus monkeys by cytokine-augmented DNA vaccination. Science 290, 486 492. Blaney, J. E. J., Nobusawa, E., Brehm, M. A., Bonneau, R. H., Mylin, L. M., Fu, T. M., Kawaoka, Y. & Tevethia, S. S. (1998). Immunization with a single major histocompatibility complex class I-restricted cytotoxic T-lymphocyte recognition epitope of herpes simplex virus type 2 confers protective immunity. Journal of Virology 72, 9567 9574. Borrow, P., Lewicki, H., Hahn, B. E., Shaw, G. M. & Oldstone, M. B. (1994). Virus-specific CD8 CTL activity associated with control of viremia in primary HIV-1 infection. Journal of Virology 68, 6103 6110. Borrow, P., Lewicki, H., Wei, X., Horwitz, M. S., Peffer, N., Meyers, H., Nelson, J. A., Gairin, J. E., Hahn, B. H., Oldstone, M. B. A. & Shaw, G. M. (1997). Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nature Medicine 3, 205 211. HI

Induction of broad T cell responses in monkeys Fu, T. M., Guan, L., Friedman, A., Schofield, T. L., Ulmer, J. B., Liu, M. A. & Donnelly, J. J. (1999). Dose dependence of CTL precursor frequency induced by a DNA vaccine and correlation with protective immunity against influenza virus challenge. Journal of Immunology 162, 4163 4170. Gallimore, A., Cranage, M., Cook, N., Almond, N., Bootman, J., Rud, R., Silvera, P., Dennis, M., Corcoran, T., Stott, J., McMichael, A. & Gotch, F. (1995). Early suppression of SIV replication by CD8 nef-specific cytotoxic T cells in vaccinated animals. Nature Medicine 1, 1167 1173. Goulder, P. J. R., Phillips, R. E., Colbert, R. A., McAdam, S., Ogg, G., Nowak, M. A., Giangrande, P., Luzzi, G., Morgan, B., Edwards, A., McMichael, A. J. & Rowland-Jones, S. (1997). Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nature Medicine 3, 212 217. Haas, G., Plikat, U., Debre, P., Lucchiari, M., Katlama, C., Dudoit, Y., Bonduelle, O., Bauer, M., Ihlenfeldt, H. G., Jung, G., Maier, B., Meyerhans, A. & Autran, B. (1996). Dynamics of viral variants in HIV- 1 Nef and specific cytotoxic T lymphocytes in vivo. Journal of Immunology 157, 4212 4221. Hanke, T. (2001). Prospect of a prophylactic vaccine for HIV. British Medical Bulletin 58 (in press). Hanke, T. & McMichael, A. J. (2000). Design and construction of an experimental HIV-1 vaccine for a year-2000 clinical trial in Kenya. Nature Medicine 6, 951 955. Hanke, T., Blanchard, T. J., Schneider, J., Hannan, C. M., Becker, M., Gilbert, S. C., Hill, A. V. S., Smith, G. L. & McMichael, A. (1998a). Enhancement of MHC class I-restricted peptide-specific T cell induction by a DNA prime MVA boost vaccination regime. Vaccine 16, 439 445. Hanke, T., Schneider, J., Gilbert, S. C., Hill, A. V. S. & McMichael, A. (1998b). DNA multi-ctl epitope vaccines for HIV and Plasmodium falciparum: Immunogenicity in mice. Vaccine 16, 426 435. Hanke, T., Neumann, V. C., Blanchard, T. J., Becker, M., Sweeney, P., Hill, A. V. S., Smith, G. L. & McMichael, A. (1999a). Effective induction of HIV-specific CTL by multi-epitope DNA using a gene gun in a combined vaccination regime. Vaccine 17, 589 596. Hanke, T., Samuel, R. V., Blanchard, T. J., Neumann, V. C., Allen, T. M., Boyson, J. E., Sharpe, A. S., Cook, N., Smith, G. L., Watkins, D. I., Cranage, M. P. & McMichael, A. (1999b). Effective induction of simian immunodeficiency virus-specific cytotoxic T lymphocytes in macaques by using a multiepitope gene and DNA prime-modified vaccinia virus Ankara boost vaccination regimen. Journal of Virology 73, 7524 7532. Jin, X., Bauer, D. E., Tuttleton, S. E., Lewin, S., Gettie, A., Blanchard, J., Irwin, C. E., Safrit, J. T., Mittler, J., Weinberger, L., Kostrikis, L. G., Zhang, L., Perelson, A. S. & Ho, D. D. (1999). Dramatic rise in plasma viremia after CD8+T cell depletion in simian immunodeficiency virusinfected macaques. Journal of Experimental Medicine 189, 991 998. Kent, S. J., Woodward, A. & Zhao, A. (1997). Human immunodeficiency virus type 1 (HIV-1)-specific T cell responses correlate with the control of acute HIV-1 infection in macaques. Journal of Infectious Diseases 176, 1188 1197. Kent, S. J., Zhao, A., Best, S. J., Chandler, J. D., Boyle, D. B. & Ramshaw, I. A. (1998). Enhanced T-cell immunogenicity and protective efficacy of a human immunodeficiency virus type 1 vaccine regimen consisting of consecutive priming with DNA and boosting with recombinant fowlpox virus. Journal of Virology 72, 10180 10188. Koenig, S., Conley, A. J., Brewah, Y. A., Jones, G. M., Leath, S., Boots, L. J., Davey, V., Pantaleo, G., Demarest, J. F. & Carter, C. and others (1995). Transfer of HIV-1-specific cytotoxic T lymphocytes to an AIDS patient leads to selection for mutant HIV variants and subsequent disease progression. Nature Medicine 1, 330 336. Koup, R. A., Safrit, J. T., Cao, Y., Andrews, C. A., McLeod, G., Borkowsky, W., Farthing, C. & Ho, D. D. (1994). Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. Journal of Virology 68, 4650 4655. Maino, V. C. & Picker, L. J. (1998). Identification of functional subsets by flow cytometry: intracellular detection of cytokine expression. Cytometry 34, 518 525. Matloubian, M., Concepcion, R. J. & Ahmed, R. (1994). CD4 T cells are required to sustain CD8 cytotoxic T-cell responses during chronic viral infection. Journal of Virology 68, 8056 8063. Miller, M. D., Yamamoto, H., Hughes, A. L., Watkins, D. I. & Letvin, N. L. (1991). Definition of an epitope and MHC class I molecule recognized by Gag-specific cytotoxic T lymphocytes in SIVmac-infected rhesus monkeys. Journal of Immunology 147, 320 329. Phillips, R. E., Rowland-Jones, S., Nixon, D. F., Gotch, F. M., Edwards, J. P., Ogunlesi, A. O., Elvin, J. G., Rothbard, J. A., Bangham, C. R., Rizza, C. R. & McMichael, A. J. (1991). Human immunodeficiency virus genetic variation that can escape cytotoxic T cell recognition. Nature 354, 453 459. Price, D. A., Goulder, P. J., Klenerman, P., Sewell, A. K., Easterbrook, P. J., Troop, M., Bangham, C. R. & Phillips, R. E. (1997). Positive selection of HIV-1 cytotoxic T lymphocyte escape variants during primary infection. Proceedings of the National Academy of Sciences, USA 94, 1890 1895. Price, D. A., Sewell, A. K., Dong, T., Tan, R., Goulder, P. J., Rowland- Jones, S. L. & Phillips, R. E. (1998). Antigen-specific release of betachemokines by anti-hiv-1 cytotoxic T lymphocytes. Current Biology 8, 355 358. Robinson, H. L., Montefiori, D. C., Johnson, R. P., Manson, K. H., Kalish, M. L., Lifson, J. D., Rizvi, T. A., Lu, S., Hu, S.-L., Mazzara, G. P., Panicali, D. L., Herndon, J. G., Glickman, R., Candido, M. A., Lydy, S. L., Wyand, M. S. & McClure, H. M. (1999). Neutralizing antibodyindependent containment of immunodeficiency virus challenges by DNA priming and recombinant pox virus booster immunizations. Nature Medicine 5, 526 534. Rosenberg, E. S., LaRosa, L., Flynn, T., Robbins, G. & Walker, B. D. (1999). Characterization of HIV-1-specific T-helper cells in acute and chronic infection. Immunology Letters 66, 89 93. Rowland-Jones, S. L. & McMichael, A. (1995). Immune responses in HIV-exposed seronegatives: have they repelled the virus? Current Opinion in Immunology 7, 448 455. Schmitz, J. E., Kuroda, M. J., Sasseville, V. G., Simon, M. A., Lifton, M. A., Racz, P., Tenner-Racz, K., Dalesandro, M., Scallon, B. J., Ghrayeb, J., Forman, M. A., Montefiori, D. C., Rieber, E. P., Letvin, N. L. & Reinmann, K. A. (1999). Control of viremia in simian immunodeficiency virus infection by CD8 lymphocytes. Science 283, 857 860. Schneider, J., Gilbert, S. C., Blanchard, T. J., Hanke, T., Robson, K. J., Hannan, C. M., Becker, M., Sinden, R., Smith, G. L. & Hill, A. V. S. (1998). Enhanced immunogenicity for CD8+ T cell induction and complete protective efficacy of malaria DNA vaccination by boosting with modified vaccinia virus Ankara. Nature Medicine 4, 397 402. Stranford, S. A., Skurnick, J., Louria, D., Osmond, D., Chang, S. Y., Sninsky, J., Ferrari, G., Weinhold, K., Lindquist, C. & Levy, J. A. (1999). Lack of infection in HIV-exposed individuals is associated with a strong CD8+ cell noncytotoxic anti-hiv response. Proceedings of the National Academy of Sciences, USA 96, 1030 1035. Suni, M. A., Picker, L. J. & Maino, V. C. (1998). Detection of antigenspecific T cell cytokine expression in whole blood by flow cytometry. Journal of Immunological Methods 212, 89 98. HJ

E. G.-T. Wee and others Tan, L. C., Gudgeon, N., Annels, N. E., Hansasuta, P., O Callaghan, C. A., Rowland-Jones, S., McMichael, A. J., Rickinson, A. B. & Callan, M. F. (1999). A re-evaluation of the frequency of CD8 T cells specific for EBV in healthy virus carriers. Journal of Immunology 162, 1827 1835. von Herrath, M. G., Yokoyama, M., Dockter, J., Oldstone, M. B. & Whitton, J. L. (1996). CD4-deficient mice have reduced levels of memory cytotoxic T lymphocytes after immunization and show diminished resistance to subsequent virus challenge. Journal of Virology 70, 1072 1079. Wagner, L., Yang, O. O., Garcia-Zepeda, E. A., Ge, Y., Kalams, S. A., Walker, B. D., Pasternack, M. S. & Luster, A. D. (1998). Betachemokines are released from HIV-1-specific cytolytic T cell granules complexed to proteoglycans. Nature 391, 908 911. Wilson, C. C., Brown, R. C., Korber, B. T., Wilkes, B. M., Ruhl, D. J., Sakamoto, D., Kunstman, K., Luzuriaga, K., Hanson, I. C., Widmayer, S. M., Wiznia, A., Clapp, S., Ammann, A. J., Koup, R. A., Wolinsky, S. M. & Walker, B. D. (1999). Frequent detection of escape from cytotoxic T- lymphocyte recognition in perinatal human immunodeficiency virus (HIV) type 1 transmission: the Ariel Project for the prevention of transmission of HIV from mother to infant. Journal of Virology 73, 3975 3985. Wilson, J. D., Ogg, G. S., Allen, R. L., Davis, C., Shaunak, S., Downie, J., Dyer, W., Workman, C., Sullivan, S., McMichael, A. J. & Rowland-Jones, S. L. (2000). Direct visualization of HIV-1-specific cytotoxic T lymphocytes during primary infection. AIDS 14, 225 233. Wolinsky, S. M., Korber, B. T., Neumann, A. U., Daniels, M., Kunstman, K. J., Whetsell, A. J., Furtado, M. R., Cao, Y., Ho, D. D. & Safrit, J. T. (1996). Adaptive evolution of human immunodeficiency virus-type 1 during the natural course of infection. Science 272, 537 542. Yang, O. O., Kalams, S. A., Trocha, A., Cao, H., Luster, A., Johnson, R. P. & Walker, B. D. (1997). Suppression of human immunodeficiency virus type 1 replication by CD8 cells: evidence for HLA class I- restricted triggering of cytolytic and noncytolytic mechanisms. Journal of Virology 71, 3120 3128. Zhang, C., Cui, Y., Houston, S. & Chang, L. J. (1996). Protective immunity to HIV-1 in SCID beige mice reconstituted with peripheral blood lymphocytes of exposed but uninfected individuals. Proceedings of the National Academy of Sciences, USA 93, 14720 14725. Received 23 August 2001; Accepted 25 September 2001 IA