Received 22 April 2009/Accepted 19 June 2009

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

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1.

Novel Heterologous Prime-Boost Vaccine Strategies for HIV. Dan Barouch April 18, 2012

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

Updated information and services can be found at:

Received 30 December 2006/Accepted 25 February 2007

It has been 25 years since HIV-1 was identified as the causative

Received 19 March 2003/Accepted 28 May 2003

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

SUPPLEMENTARY INFORMATION

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

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

Heterologous Envelope Immunogens Contribute to AIDS Vaccine Protection in Rhesus Monkeys

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

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

The global spread of the human immunodeficiency virus (HIV)

An AIDS vaccine: Why is it so difficult?

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Antibody-Dependent Cell-Mediated Cytotoxicity in Simian Immunodeficiency Virus-Infected Rhesus Monkeys

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

Received 26 September 2003/Accepted 22 December 2003

Efficacy of Multivalent Adenovirus-Based Vaccine against Simian Immunodeficiency Virus Challenge

Received 18 January 2005/Accepted 18 April 2005

Received 16 February 2004/Accepted 5 June 2004

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

Received 17 April 2003/Accepted 28 June 2003

The effect of pre-existing immunity to antigens of adenovirus of the human serotype 5 on

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

Low-Dose Mucosal Simian Immunodeficiency Virus Infection Restricts Early Replication Kinetics and Transmitted Virus Variants in Rhesus Monkeys

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

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys

NIH Public Access Author Manuscript Nat Rev Microbiol. Author manuscript; available in PMC 2014 November 19.

Why are validated immunogenicity assays important for HIV vaccine development?

Is an HIV Vaccine Possible?

A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys

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

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

Strategies for an HIV vaccine

Received 14 April 2005/Accepted 7 August 2005

Mosaic vaccines elicit CD8 + T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys

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

Supporting Information

Received 17 March 2003/Accepted 16 July 2003

Received 31 March 2009/Returned for modification 12 June 2009/Accepted 5 July 2009

Immunogens and Antigen Processing: Report from a Global HIV Vaccine Enterprise Working Group

In Vivo Electroporation Enhances the Immunogenicity of ADVAX, a DNA-based HIV-1 Vaccine Candidate, in Healthy Volunteers

Enhanced Breadth of CD4 T-Cell Immunity by DNA Prime and Adenovirus Boost Immunization to Human Immunodeficiency Virus Env and Gag Immunogens

HIV and Challenges of Vaccine Development

Department of Surgery, Duke University, Durham, North Carolina; and 7 Southern Research Institute, Frederick, Maryland

HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case cohort analysis

Perspectives of AIDS Vaccine Development: T Cell-based Vaccine

HIV Anti-HIV Neutralizing Antibodies

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

Received 13 November 2006/Accepted 20 March 2007

EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV. (Summary of the recommendations from an Enterprise Working Group)

AIDSVaccine2010 Atlanta, Georgia Willy Bogers. NIH HIVRad Grant nr 5P01AI066287

Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys

/JVI Updated information and services can be found at:

Vaccine Delivery and TLR Ligands Influence the Quality of T cell Responses in NHP. Robert A. Seder, M.D. Vaccine Research Center NIAID, NIH

Received 29 May 2008/Accepted 19 August 2008

Significant Protection against High-Dose Simian Immunodeficiency Virus Challenge Conferred by a New Prime-Boost Vaccine Regimen

Received 23 March 2005/Accepted 2 May 2005

A global approach to HIV-1 vaccine development

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

Simian-Human Immunodeficiency Infection Is the Course Set in the Acute Phase?

Therapeutic strategies for immune reconstitution in acquired immunodeficiency syndrome

Hepatitis C genotype 1 mosaic vaccines are immunogenic in mice and induce higher T cell

Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89.

Combinatorial Vaccines for AIDS and other Infectious Diseases

Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses

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

Received 22 April 2002/Accepted 28 June 2002

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

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

The humoral immune responses to IBV proteins.

Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques

How HIV Causes Disease Prof. Bruce D. Walker

Received 19 September 2007/Accepted 21 November 2007

Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge

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

Received 6 December 2007/Returned for modification 21 December 2007/Accepted 14 January 2008

Are we targeting the right HIV determinants?

Novel Vaccine Products for Planned Phase I Immunogenicity Studies in Infants

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

Low dose penile SIVmac251 exposure of rhesus macaques infected with adenovirus 5 then

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

Received 14 July 2005/Accepted 3 September 2005

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

staining and flow cytometry

HIV-1 Vaccine Development: Recent Advances in the CTL Platform Spotty Business ACCEPTED

Immunoprophylaxis against AIDS in macaques with a lentiviral DNA vaccine

HIV/AIDS: vaccines and alternate strategies for treatment and prevention

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

NIH Public Access Author Manuscript J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01.

Progress on new vaccine strategies against chronic viral infections

Efficacy Results from the Step Study

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

HIV Pathogenesis and Vaccine Development

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

Transcription:

JOURNAL OF VIROLOGY, Sept. 2009, p. 9584 9590 Vol. 83, No. 18 0022-538X/09/$08.00 0 doi:10.1128/jvi.00821-09 Copyright 2009, American Society for Microbiology. All Rights Reserved. Protective Efficacy of a Single Immunization of a Chimeric Adenovirus Vector-Based Vaccine against Simian Immunodeficiency Virus Challenge in Rhesus Monkeys Dan H. Barouch, 1,7 * Jinyan Liu, 1 Diana M. Lynch, 1 Kara L. O Brien, 1 Annalena La Porte, 1 Nathaniel L. Simmons, 1 Ambryice M. Riggs, 1 Sarah Clark, 1 Peter Abbink, 1 David C. Montefiori, 2 Gary Landucci, 3 Donald N. Forthal, 3 Steven G. Self, 4 Angela Carville, 5 Keith Mansfield, 5 and Jaap Goudsmit 6 Division of Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215 1 ; Duke University Medical Center, Durham, North Carolina 27710 2 ; University of California, Irvine School of Medicine, Irvine, California 92697 3 ; Department of Biostatistics, University of Washington, Seattle, Washington 98109 4 ; New England Primate Research Center, Southborough, MA 01772 5 ; Crucell Holland BV, 2301 CA, Leiden, The Netherlands 6 ; and Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02214 7 Received 22 April 2009/Accepted 19 June 2009 Rare serotype and chimeric recombinant adenovirus (rad) vectors that evade anti-ad5 immunity are currently being evaluated as potential vaccine vectors for human immunodeficiency virus type 1 and other pathogens. We have recently reported that a heterologous rad prime-boost regimen expressing simian immunodeficiency virus (SIV) Gag afforded durable partial immune control of an SIV challenge in rhesus monkeys. However, single-shot immunization may ultimately be preferable for global vaccine delivery. We therefore evaluated the immunogenicity and protective efficacy of a single immunization of chimeric rad5 hexon hypervariable region 48 (rad5hvr48) vectors expressing SIV Gag, Pol, Nef, and Env against a homologous SIV challenge in rhesus monkeys. Inclusion of Env resulted in improved control of peak and set point SIV RNA levels following challenge. In contrast, DNA vaccine priming did not further improve the protective efficacy of rad5hvr48 vectors in this system. Heterologous prime-boost vaccine regimens have proven substantially more immunogenic than single vector immunizations in a variety of experimental models, but a single-shot vaccine would presumably be ideal for eventual global delivery. The potential utility of single-shot vaccines against pathogenic simian immunodeficiency virus (SIV) challenges in rhesus monkeys has not been well characterized. We therefore evaluated the protective efficacy of a single immunization of recombinant chimeric adenovirus type 5 (rad5) hexon hypervariable region 48 (rad5hvr48) vectors (15) expressing SIV Gag, Pol, Nef, and Env against a pathogenic SIV challenge in rhesus monkeys. These vectors contain the HVRs of the rare Ad48 serotype and have been shown to evade dominant Ad5 hexonspecific neutralizing antibodies (NAbs) (15). We also assessed the potential utility of inclusion of Env as an immunogen (6, 7, 17) and the degree to which DNA vaccine priming would enhance the protective efficacy afforded by a single rad5hvr48 immunization (2, 7, 18, 21). Thirty adult rhesus monkeys (n 6/group) lacking the Mamu-A*01, Mamu-B*17, and Mamu-B*08 class I alleles were primed with plasmid DNA vaccines and boosted with rad5hvr48 vectors as follows: (1) adjuvanted DNA prime, rad5hvr48 boost; (2) DNA prime, rad5hvr48 boost; (3) rad5hvr48 alone; (4) rad5hvr48 alone (excluding Env); * Corresponding author. Mailing address: E/CLS-1047, Division of Vaccine Research, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215. Phone: (617) 735-4485. Fax: (617) 725-4527. E-mail: dbarouch@bidmc.harvard.edu. Published ahead of print on 24 June 2009. and (5) sham controls. Monkeys in groups 1 to 3 received vectors expressing SIVmac239 Gag, Pol, Nef, and Env, whereas monkeys in group 4 received vectors expressing only Gag, Pol, and Nef. The DNA vaccine adjuvants in group 1 were plasmids expressing the rhesus chemokine MIP-1 and Flt3L, which have been shown to increase recruitment of dendritic cells and to improve DNA vaccine immunogenicity (20). Monkeys were primed intramuscularly with a total dose of 4 mg of DNA vaccines at weeks 0, 4, and 8. All animals then received a single intramuscular immunization of 4 10 10 viral particles (vp) of rad5hvr48 at week 24. At week 52, animals were challenged intravenously (i.v.) with 100 monkey infectious doses of SIVmac251 (7, 10). Vaccine-elicited immune responses. We monitored vaccineelicited, SIV-specific cellular immune responses prior to challenge by gamma interferon (IFN- ) enzyme-linked immunospot (ELISPOT) assays following stimulation with SIV Gag, Pol, Nef, and Env peptide pools (8). As shown in Fig. 1A, monkeys that received a single immunization of the rad vaccines alone at week 24 developed IFN- ELISPOT responses to the encoded SIV antigens by week 26. As expected, animals that were primed with the DNA vaccines developed higher responses than those elicited by the rad alone vaccines (16, 18). The adjuvanted DNA vaccines elicited a trend toward higher peak responses compared with the unadjuvanted DNA vaccines, but these differences were not sustained at week 52. As shown in Fig. 1B, DNA priming augmented both CD8 and CD4 T-lymphocyte responses, as determined by cell- 9584

VOL. 83, 2009 NOTES 9585 FIG. 1. Immunogenicity of vaccine regimens. Rhesus monkeys were primed at weeks 0, 4, and 8 with adjuvanted DNA vaccines (DNA*) or unadjuvanted DNA vaccines and were boosted at week 24 with a single immunization of rad5hvr48 expressing SIV Gag, Pol, Nef, and Env (GPNE). One group of monkeys received rad5hvr48 vectors expressing only SIV Gag, Pol, and Nef (GPN). (A) SIV-specific IFN- ELISPOT assays were performed at weeks 0, 10, 24, 26, and 52 following immune priming. (B) SIV-specific CD8 (top panel) and CD4 (bottom panel) T-lymphocyte responses were evaluated at week 28 by CD4-depleted and CD8-depleted ELISPOT assays, respectively. Data represent mean responses with standard errors. (C) The functionality of SIV-specific CD8 and CD4 central memory CM (CD28 CD95 ) and effector memory EM (CD28 CD95 ) T-lymphocyte responses to all antigens elicited by DNA/rAd5HVR48 regimens and rad5hvr48 alone regimens was assessed by eight-color ICS assays. Proportions of IFN-, TNF-, and IL-2 responses are depicted individually and in all possible combinations for each cellular subpopulation. depleted IFN- ELISPOT assays (P 0.01 and P 0.001, respectively [Wilcoxon rank-sum tests]). Multiparameter intracellular cytokine staining (ICS) assays (8, 13, 14) confirmed these findings but did not reveal other consistent phenotypic differences in terms of IFN-, tumor necrosis factor alpha (TNF- ), and interleukin 2 (IL-2) secretion in CD8 and CD4 central memory (CM [CD28 CD95 ]) and effector memory (EM [CD28 CD95 ]) T-lymphocyte subpopula-

9586 NOTES J. VIROL. tions (Fig. 1C). The ICS assays utilized SIV Gag, Pol, Nef, and Env peptide pools and the following monoclonal antibodies: anti-cd3-alexa700 (SP34), anti-cd4-amcyan (L200), anti- CD8-allophycocyanin-Cy7 (anti-cd8-apc-cy7) (SK1), anti-cd28- PerCP-Cy5.5 (L293), anti-cd95-phycoerythrin (anti-cd95-pe) (DX2), anti-ifn- PE-Cy7 (B27), anti-il-2 APC (MQ1-17H12), and anti-tnf- fluorescein isothiocyanate (anti-tnf- FITC) (Mab11). Immune responses following SIV challenge. Six months after the rad boost immunization, monkeys were challenged i.v. with the essentially homologous virus SIVmac251 (7, 10). As shown in Fig. 2A, all vaccinated animals developed rapid and potent anamnestic SIV-specific cellular immune responses by week 2 following challenge, as determined by IFN- ELISPOT assays. SIV-specific humoral immune responses were evaluated by luciferase-based pseudovirus neutralization assays (12) and antibody-dependent cell-mediated virus inhibition (ADCVI) assays (5). All animals developed virus-specific NAbs against laboratory-adapted SIVmac251 but not against primary isolate SIVmac251 following challenge (Fig. 2B). Animals that received vaccines containing SIV Env (groups 1 to 3) also developed more rapid kinetics of ADCVI than animals that received sham controls or vaccines that did not include Env (Fig. 2C). We next monitored the CD4 T-lymphocyte dynamics in these animals following challenge (10) utilizing the following monoclonal antibodies: anti-cd3-alexa700 (SP34), anti-cd4- AmCyan (L200), anti-cd8-apc-cy7 (SK1), anti-cd28- PerCP-Cy5.5 (L293), anti-cd95-apc (DX2), anti-ccr5-pe (3A9), anti-hla-dr PE-Cy7 (L243), and anti-ki67-fitc (B56). As shown in Fig. 3, control animals exhibited slightly decreased levels of CD4 CM T lymphocytes, substantially decreased levels of CCR5 CD4 CM T lymphocytes, and markedly increased levels of Ki67 proliferation of CCR5 CD4 CM T lymphocytes following SIV challenge, consistent with our prior results (10). In contrast, animals vaccinated with rad alone had fewer dramatic perturbations of these CD4 T-lymphocyte subsets. Protective efficacy. To assess the protective efficacy of these vaccine regimens, we monitored SIV RNA levels and survival in these animals following challenge, as depicted in Fig. 4. SIV RNA levels for each individual animal (Fig. 4A) as well as peak (day 14) (Fig. 4B) and set point (median days 112 to 392) (Fig. 4C) SIV RNA levels are shown. The rad alone vaccine expressing only Gag, Pol, and Nef afforded only minimal control of SIV replication, but the rad alone vaccine expressing Gag, Pol, Nef, and Env resulted in a 1.34 log reduction of peak SIV RNA levels (Fig. 4B) (P 0.001 [Wilcoxon rank-sum test]) and a 1.08 log reduction of set point SIV RNA levels compared with sham controls (Fig. 4C) (P 0.05). These data suggest that the addition of Env improved control of SIV replication following challenge in this study. The rad alone vaccines also afforded trends toward improved survival compared with sham controls for 500 days following the challenge (Fig. 4D) (P 0.04 and P 0.07 [unadjusted two-sided log-rank tests comparing rad-gag/pol/nef/env and rad-gag/pol/nef regimens, respectively, versus sham controls]). Moreover, in a prespecified exploratory analysis, the combined rad alone vaccinated animals (n 12) exhibited significantly improved survival compared with the sham controls (P 0.003 [two-sided log-rank test]). The monkeys that were primed with DNA vaccines or adjuvanted DNA vaccines prior to the rad immunization, however, did not exhibit improved protection compared with the animals that received rad alone vaccines. The DNA/rAd regimens afforded 0.94 to 1.11 log reductions of peak SIV RNA levels (Fig. 4B) (P 0.004 [Wilcoxon rank-sum test]) but no significant reductions of set point SIV RNA levels (Fig. 4C) (P nonsignificant) and only marginal improvements of survival (Fig. 4D) (P nonsignificant) compared with the sham controls. These data show that DNA vaccine priming did not improve the protective efficacy afforded by rad5hvr48 vectors in this study. These results are consistent with previous SIV challenge studies that have similarly shown no significant and durable benefit of DNA vaccine priming prior to rad5 boosting in terms of set point SIV RNA levels or survival following challenge (2, 7). Casimiro et al. (2) reported that neither DNA/rAd5 regimens nor rad5 alone regimens expressing SIV Gag resulted in significant reductions of set point SIV RNA levels following SIV challenge of Mamu-A*01-positive rhesus monkeys, although a transient 0.8 log reduction of peak SIV RNA levels was noted in the DNA/rAd5 group. Similarly, Letvin et al. (7) reported that neither DNA/rAd5 regimens nor rad5 alone regimens expressing SIV Gag, Pol, and Env resulted in significant reductions of set point SIV RNA levels following SIV challenge of Mamu-A*01-negative monkeys, although a transient 1.1 log reduction of peak SIV RNA levels and a survival advantage were observed in a post hoc analysis of all vaccinated groups combined. One possible explanation for these observations is that the DNA vaccines utilized in these studies were simply not sufficiently potent to enhance protective efficacy in this stringent challenge model. Improved DNA vaccine delivery technologies, such as in vivo electroporation (9, 11) and the use of genetic adjuvants (1, 3), may therefore prove useful in this regard. An alternative possibility is that DNA priming skewed the phenotypes of the vaccine-elicited cellular immune responses. It has been reported that DNA priming prior to rad5 boosting increased virus-specific CD4 T-lymphocyte responses in both rhesus monkeys (16) and humans (4), and our data are consistent with these observations (Fig. 1B). It is therefore possible that SIV-specific CD4 T-lymphocyte responses that were augmented by DNA priming may have resulted in increased numbers of viral targets for infection (19). Our current data, however, are insufficient to address this possibility conclusively; thus, further studies will be required to address these hypotheses. The present findings show that a single-shot immunization with rad5hvr48 vectors afforded a detectable but relatively modest level of protective efficacy against a homologous SIV challenge in rhesus monkeys. Overall, the protective efficacy observed in this study was less impressive than that seen in a previous study in which we utilized a heterologous rad26 prime, rad5 boost regimen expressing SIV Gag (10). A direct comparison of the results of these two studies, however, is not possible as a result of the different vectors and inserts utilized. Nevertheless, it appears likely that a single-shot vaccine regimen may not afford as robust protection as an optimal heterologous rad prime-boost regimen. In our previous study of the rad26/rad5 regimen, we observed a significant inverse corre-

VOL. 83, 2009 NOTES 9587 FIG. 2. Cellular and humoral immune responses following challenge. (A) IFN- ELISPOT assays, (B) SIV-specific neutralizing antibody assays against T-cell laboratory-adapted (TCLA) and primary isolate SIVmac251, and (C) ADCVI assays at a 1:100 dilution were performed at multiple time points following challenge. GPNE, SIV Gag, Pol, Nef, and Env; GPN, SIV Gag, Pol, and Nef; SFC, spot-forming cells; PBMC, peripheral blood mononuclear cells. lation between the magnitude and breadth of Gag-specific cellular immune responses and set point viral loads (10). In contrast, we did not detect statistically significant correlations between vaccine-elicited immune responses and set point viral loads in the present study. This may reflect the fact that DNA priming could have opposing effects in terms of augmenting potentially useful cellular immune responses but also potentially increasing viral targets for infection, thus complicating analyses of immune correlates of protection. In the present study, peak and set point SIV RNA levels

9588 NOTES J. VIROL. FIG. 3. CD4 T-lymphocyte dynamics following challenge. (A) CM (CD28 CD95 ) CD4 T lymphocytes and (B) CCR5 CD4 CM T lymphocytes were assessed at multiple time points following challenge. (C) Ki67 staining of CD4 CCR5 CM T lymphocytes was also determined. Data represent mean responses with standard errors. GPNE, SIV Gag, Pol, Nef, and Env; GPN, SIV Gag, Pol, and Nef. were lower in animals that received rad5hvr48 vectors expressing Gag, Pol, Nef, and Env than in those that received rad5hvr48 vectors expressing only Gag, Pol, and Nef. These data indicate that inclusion of Env resulted in improved control of SIV replication in this study. We suspect that Env-specific cellular immune responses likely contributed to the improved protective efficacy, since Env-specific NAbs against primary isolate SIVmac251 were not observed either prior to challenge or following challenge. More rapid kinetics of Env-specific ADCVI activity, however, was observed in the groups administered vaccine containing Env; thus, it is possible that nonneutralizing Env-specific antibodies also contributed to protection. A limitation is that the challenge virus was essentially homologous to the antigens contained in the vac-

VOL. 83, 2009 NOTES 9589 FIG. 4. Protective efficacy of vaccine regimens. Monkeys were challenged i.v. with SIVmac251, and protective efficacy was monitored by SIV RNA levels (A to C) and clinical disease progression and mortality following challenge (D). (A to C) SIV RNA levels are depicted longitudinally for each group (A), and peak (day 14) (B) and set point (median days 112 to 392) (C) SIV RNA levels are summarized for each group. Red asterisks indicate mortality. The numbers at the bottom of the graphs indicate mean SIV RNA levels in each group. Black asterisks indicate significant differences compared with sham controls as assessed by two-tailed Wilcoxon rank-sum tests. (D) Survival curve for 500 days following challenge. GPNE, SIV Gag, Pol, Nef, and Env; GPN, SIV Gag, Pol, and Nef. cine. Thus, future studies utilizing larger numbers of animals will be required to evaluate the potential utility of Env against heterologous SIV challenges. The protective efficacy of single immunizations of optimal vaccine vectors should also continue to be explored. We thank R. Dolin, N. Letvin, D. Roberts, A. Nanda, P. Swanson, J. Arthur, F. Stephens, and M. Pensiero for generous advice, assistance, and reagents. We acknowledge support from the National Institutes of Health to D.H.B. (grants AI058727, AI066305, AI066924, and AI078526). We declare no competing financial interests.

9590 NOTES J. VIROL. REFERENCES 1. Barouch, D. H., S. Santra, J. E. Schmitz, M. J. Kuroda, T. M. Fu, W. Wagner, M. Bilska, A. Craiu, X. X. Zheng, G. R. Krivulka, K. Beaudry, M. A. Lifton, C. E. Nickerson, W. L. Trigona, K. Punt, D. C. Freed, L. Guan, S. Dubey, D. Casimiro, A. Simon, M. E. Davies, M. Chastain, T. B. Strom, R. S. Gelman, D. C. Montefiori, M. G. Lewis, E. A. Emini, J. W. Shiver, and N. L. Letvin. 2000. Control of viremia and prevention of clinical AIDS in rhesus monkeys by cytokine-augmented DNA vaccination. Science 290:486 492. 2. Casimiro, D. R., F. Wang, W. A. Schleif, X. Liang, Z.-Q. Zhang, T. W. Tobery, M. E. Davies, A. B. McDermott, D. H. O Connor, A. Fridman, A. Bagchi, L. G. Tussey, A. J. Bett, A. C. Finnefrock, T.-M. Fu, A. Tang, K. A. Wilson, M. Chen, H. C. Perry, G. J. Heidecker, D. C. Freed, A. Carella, K. S. Punt, K. J. Sykes, L. Huang, V. I. Ausensi, M. Bachinsky, U. Sadasivan-Nair, D. I. Watkins, E. A. Emini, and J. W. Shiver. 2005. Attenuation of simian immunodeficiency virus SIVmac239 infection by prophylactic immunization with dna and recombinant adenoviral vaccine vectors expressing Gag. J. Virol. 79:15547 15555. 3. Chong, S. Y., M. A. Egan, M. A. Kutzler, S. Megati, A. Masood, V. Roopchard, D. Garcia-Hand, D. C. Montefiori, J. Quiroz, M. Rosati, E. B. Schadeck, J. D. Boyer, G. N. Pavlakis, D. B. Weiner, M. Sidhu, J. H. Eldridge, and Z. R. Israel. 2007. Comparative ability of plasmid IL-12 and IL-15 to enhance cellular and humoral immune responses elicited by a SIVgag plasmid DNA vaccine and alter disease progression following SHIV(89.6P) challenge in rhesus macaques. Vaccine 25:4967 4982. 4. Cox, K. S., J. H. Clair, M. T. Prokop, K. J. Sykes, S. A. Dubey, J. W. Shiver, M. N. Robertson, and D. R. Casimiro. 2008. DNA gag/adenovirus type 5 (Ad5) gag and Ad5 gag/ad5 gag vaccines induce distinct T-cell response profiles. J. Virol. 82:8161 8171. 5. Forthal, D. N., G. Landucci, and E. S. Daar. 2001. Antibody from patients with acute human immunodeficiency virus (HIV) infection inhibits primary strains of HIV type 1 in the presence of natural-killer effector cells. J. Virol. 75:6953 6961. 6. Letvin, N. L., Y. Huang, B. K. Chakrabarti, L. Xu, M. S. Seaman, K. Beaudry, B. Korioth-Schmitz, F. Yu, D. Rohne, K. L. Martin, A. Miura, W. P. Kong, Z. Y. Yang, R. S. Gelman, O. G. Golubeva, D. C. Montefiori, J. R. Mascola, and G. J. Nabel. 2004. Heterologous envelope immunogens contribute to AIDS vaccine protection in rhesus monkeys. J. Virol. 78:7490 7497. 7. Letvin, N. L., J. R. Mascola, Y. Sun, D. A. Gorgone, A. P. Buzby, L. Xu, Z. Y. Yang, B. Chakrabarti, S. S. Rao, J. E. Schmitz, D. C. Montefiori, B. R. Barker, F. L. Bookstein, and G. J. Nabel. 2006. Preserved CD4 central memory T cells and survival in vaccinated SIV-challenged monkeys. Science 312:1530 1533. 8. Liu, J., B. A. Ewald, D. M. Lynch, M. Denholtz, P. Abbink, A. A. Lemckert, A. Carville, K. G. Mansfield, M. J. Havenga, J. Goudsmit, and D. H. Barouch. 2008. Magnitude and phenotype of cellular immune responses elicited by recombinant adenovirus vectors and heterologous prime-boost regimens in rhesus monkeys. J. Virol. 82:4844 4852. 9. Liu, J., R. Kjeken, I. Mathiesen, and D. H. Barouch. 2008. Recruitment of antigen-presenting cells to the site of inoculation and augmentation of human immunodeficiency virus type 1 DNA vaccine immunogenicity by in vivo electroporation. J. Virol. 82:5643 5649. 10. Liu, J., K. L. O Brien, D. M. Lynch, N. L. Simmons, A. La Porte, A. M. Riggs, P. Abbink, R. T. Coffey, L. E. Grandpre, M. S. Seaman, G. Landucci, D. N. Forthal, D. C. Montefiori, A. Carville, K. G. Mansfield, M. J. Havenga, M. G. Pau, J. Goudsmit, and D. H. Barouch. 2009. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457:87 91. 11. Luckay, A., M. K. Sidhu, R. Kjeken, S. Megati, S. Y. Chong, V. Roopchand, D. Garcia-Hand, R. Abdullah, R. Braun, D. C. Montefiori, M. Rosati, B. K. Felber, G. N. Pavlakis, I. Mathiesen, Z. R. Israel, J. H. Eldridge, and M. A. Egan. 2007. Effect of plasmid DNA vaccine design and in vivo electroporation on the resulting vaccine-specific immune responses in rhesus macaques. J. Virol. 81:5257 5269. 12. Montefiori, D. 2004. Evaluating neutralizing antibodies against HIV, SIV and SHIV in luciferase reporter gene assays. Curr. Prot. Immunol., unit 12.11. doi: 10.1002/0471142735.im1211s64. 13. Okoye, A., M. Meier-Schellersheim, J. M. Brenchley, S. I. Hagen, J. M. Walker, M. Rohankhedkar, R. Lum, J. B. Edgar, S. L. Planer, A. Legasse, A. W. Sylwester, M. Piatak, Jr., J. D. Lifson, V. C. Maino, D. L. Sodora, D. C. Douek, M. K. Axthelm, Z. Grossman, and L. J. Picker. 2007. Progressive CD4 central memory T cell decline results in CD4 effector memory insufficiency and overt disease in chronic SIV infection. J. Exp. Med. 204: 2171 2185. 14. Pitcher, C. J., S. I. Hagen, J. M. Walker, R. Lum, B. L. Mitchell, V. C. Maino, M. K. Axthelm, and L. J. Picker. 2002. Development and homeostasis of T cell memory in rhesus macaque. J. Immunol. 168:29 43. 15. Roberts, D. M., A. Nanda, M. J. Havenga, P. Abbink, D. M. Lynch, B. A. Ewald, J. Liu, A. R. Thorner, P. E. Swanson, D. A. Gorgone, M. A. Lifton, A. A. Lemckert, L. Holterman, B. Chen, A. Dilraj, A. Carville, K. G. Mansfield, J. Goudsmit, and D. H. Barouch. 2006. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature 441: 239 243. 16. Santra, S., M. S. Seaman, L. Xu, D. H. Barouch, C. I. Lord, M. A. Lifton, D. A. Gorgone, K. R. Beaudry, K. Svehla, B. Welcher, B. K. Chakrabarti, Y. Huang, Z. Y. Yang, J. R. Mascola, G. J. Nabel, and N. L. Letvin. 2005. Replication-defective adenovirus serotype 5 vectors elicit durable cellular and humoral immune responses in nonhuman primates. J. Virol. 79:6516 6522. 17. Shiver, J. W., and E. A. Emini. 2004. Recent advances in the development of HIV-1 vaccines using replication-incompetent adenovirus vectors. Annu. Rev. Med. 55:355 372. 18. Shiver, J. W., T. M. Fu, L. Chen, D. R. Casimiro, M. E. Davies, R. K. Evans, Z. Q. Zhang, A. J. Simon, W. L. Trigona, S. A. Dubey, L. Huang, V. A. Harris, R. S. Long, X. Liang, L. Handt, W. A. Schleif, L. Zhu, D. C. Freed, N. V. Persaud, L. Guan, K. S. Punt, A. Tang, M. Chen, K. A. Wilson, K. B. Collins, G. J. Heidecker, V. R. Fernandez, H. C. Perry, J. G. Joyce, K. M. Grimm, J. C. Cook, P. M. Keller, D. S. Kresock, H. Mach, R. D. Troutman, L. A. Isopi, D. M. Williams, Z. Xu, K. E. Bohannon, D. B. Volkin, D. C. Montefiori, A. Miura, G. R. Krivulka, M. A. Lifton, M. J. Kuroda, J. E. Schmitz, N. L. Letvin, M. J. Caulfield, A. J. Bett, R. Youil, D. C. Kaslow, and E. A. Emini. 2002. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 415:331 335. 19. Staprans, S. I., A. P. Barry, G. Silvestri, J. T. Safrit, N. Kozyr, B. Sumpter, H. Nguyen, H. McClure, D. Montefiori, J. I. Cohen, and M. B. Feinberg. 2004. Enhanced SIV replication and accelerated progression to AIDS in macaques primed to mount a CD4 T cell response to the SIV envelope protein. Proc. Natl. Acad. Sci. USA 101:13026 13031. 20. Sumida, S. M., P. F. McKay, D. M. Truitt, M. G. Kishko, J. C. Arthur, M. S. Seaman, S. S. Jackson, D. A. Gorgone, M. A. Lifton, N. L. Letvin, and D. H. Barouch. 2004. Recruitment and expansion of dendritic cells in vivo potentiate the immunogenicity of plasmid DNA vaccines. J. Clin. Investig. 114: 1334 1342. 21. Wilson, N. A., J. Reed, G. S. Napoe, S. Piaskowski, A. Szymanski, J. Furlott, E. J. Gonzalez, L. J. Yant, N. J. Maness, G. E. May, T. Soma, M. R. Reynolds, E. Rakasz, R. Rudersdorf, A. B. McDermott, D. H. O Connor, T. C. Friedrich, D. B. Allison, A. Patki, L. J. Picker, D. R. Burton, J. Lin, L. Huang, D. Patel, G. Heindecker, J. Fan, M. Citron, M. Horton, F. Wang, X. Liang, J. W. Shiver, D. R. Casimiro, and D. I. Watkins. 2006. Vaccineinduced cellular immune responses reduce plasma viral concentrations after repeated low-dose challenge with pathogenic simian immunodeficiency virus SIVmac239. J. Virol. 80:5875 5885.