X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

Size: px
Start display at page:

Download "X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved."

Transcription

1 JOURNAL OF VIROLOGY, July 2001, p Vol. 75, No X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved. Induction of Neutralizing Antibodies and Gag-Specific Cellular Immune Responses to an R5 Primary Isolate of Human Immunodeficiency Virus Type 1 in Rhesus Macaques DAVID C. MONTEFIORI, 1 * JEFFREY T. SAFRIT, 2 SHARI L. LYDY, 2 ASHLEY P. BARRY, 2 MIROSLAWA BILSKA, 1 HA T. T. VO, 1 MICHEL KLEIN, 3 JAMES TARTAGLIA, 4 HARRIET L. ROBINSON, 2 AND BENJAMIN ROVINSKI 4 Department of Surgery, Duke University Medical Center, Durham, North Carolina ; Yerkes Regional Primate Research Center, Atlanta, Georgia ; Aventis Pasteur, Marcy l Etoile, France 3 ; and Aventis Pasteur, Willowdale, Ontario, Canada 4 Received 6 December 2000/Accepted 23 March 2001 The ability to generate antibodies that cross-neutralize diverse primary isolates is an important goal for human immunodeficiency virus type 1 (HIV-1) vaccine development. Most of the candidate HIV-1 vaccines tested in humans and nonhuman primates have failed in this regard. Past efforts have focused almost entirely on the envelope glycoproteins of a small number of T-cell line-adapted strains of the virus as immunogens. Here we assessed the immunogenicity of noninfectious virus-like particles (VLP) consisting of Gag, Pro (protease), and Env from R5 primary isolate HIV-1 Bx08. Immunogens were delivered to rhesus macaques in the form of either purified VLP, recombinant DNA and canarypox (ALVAC) vectors engineered to express VLP, or a combination of these products. Seroconversion to Gag and Pro was detected in all of the immunized animals. Antibodies that could neutralize HIV-1 Bx08 were detected in animals that received (i) coinoculations with DNA Bx08 and VLP Bx08, (ii) DNA Bx08 followed by ALVAC Bx08 boosting, and (iii) VLP Bx08 alone. The neutralizing antibodies were highly strain specific despite the fact that they did not appear to be directed to linear epitopes in the V3 loop. Virus-specific cellular immune responses also were generated, as judged by the presence of Gag-specific gamma interferon (IFN- )-producing cells. These cellular immune responses required the inclusion of DNA Bx08 in the immunization modality, since few or no IFN- -producing cells were detected in animals that received either VLP Bx08 or ALVAC Bx08 alone. The results demonstrate the feasibility of generating neutralizing antibodies and cellular immune responses that target an R5 primary HIV-1 isolate by vaccination in primates. Accumulating evidence supports a positive role for neutralizing antibodies and CD8 cytotoxic T lymphocytes (CTL) in the antiviral immune response to human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) (33, 44). The ability to generate antibodies that neutralize a broad spectrum of primary HIV-1 isolates has proven to be one of the more difficult challenges for vaccine development (44). Major targets for neutralizing antibodies are the surface gp120 and transmembrane gp41 envelope glycoproteins of the virus (12). Antibodies that bind these viral glycoproteins with adequate affinity and appropriate specificity can prevent HIV-1 from entering host cells (32, 41, 45, 71, 78, 83, 84) and have provided potent protection against infection in passiveantibody experiments in animal models (2, 20, 22, 35, 38, 76). Attempts at inducing cross-reactive neutralizing antibodies through vaccination have, however, had limited success. Most attempts have focused on the envelope glycoproteins of a small number of T-cell line-adapted (TCLA) strains of virus. Although the envelope glycoproteins of those strains generate high titers of neutralizing antibodies, those antibodies mostly target strain-specific epitopes in the third variable cysteinecysteine loop (V3 loop) of gp120 (10, 45) and have been highly * Corresponding author. Mailing address: Department of Surgery, Box 2926, Duke University Medical Center, Durham, NC Phone: (919) Fax: (919) monte@acpub.duke.edu. specific for TCLA strains of virus (23, 50). Importantly, the antibodies have failed to neutralize primary isolates (4, 5, 10, 37) and often possess little or no detectable neutralizing activity against heterologous TCLA strains (5, 10). The B-cell response leading to HIV-1-specific neutralizing antibody production could be fundamentally different for primary isolates compared to TCLA strains. For example, whereas TCLA strains are highly sensitive to neutralization by V3 loop-specific antibodies (26, 68), the V3 loop on primary isolates is occluded by N-linked glycans and tertiary folds on the native gp120 molecule (30, 62, 73 75, 90, 91), making it a poor target for neutralizing antibodies (9, 77, 86). The envelope glycoproteins of TCLA strains and many, but not all, primary isolates also exhibit different coreceptor preferences for virus entry. Generally speaking, primary isolates utilize either CCR5 (R5), CXCR4 (X4), or both coreceptors (R5/X4) whereas all TCLA strains have the X4 phenotype (6, 7, 67). For reasons that are poorly understood, most transmitted strains of HIV-1 have an R5 phenotype (63, 87) and, although no association has been found between coreceptor preference and neutralization sensitivity (31, 43, 81), it seems prudent to target R5 strains when designing candidate HIV-1 vaccines. Much of what is known about the neutralization epitopes on primary isolates comes from studies of three human monoclonal antibodies (MAbs), immunoglobulin G1b12 (IgG1b12), 2G12, and 2F5. Each of these MAbs can neutralize diverse 5879

2 5880 MONTEFIORI ET AL. J. VIROL. primary isolates (12) and, when combined, exhibit synergistic neutralizing activity (36). A vaccine that generates antibodies equal to the combination of these specificities is highly desirable and deserving of intense investigation. Unfortunately, the corresponding epitopes have proven to be poorly immunogenic in natural infections and in experimentally immunized animals. Other neutralization epitopes are present on primary isolates that are less conserved. The latter epitopes account for the sporadic, low-level neutralization of heterologous isolates by sera from infected individuals (46, 58, 89). They might also account for the potent neutralization of primary isolates by autologous serum samples that are obtained months or years after the time of virus isolation (29, 48, 56, 58, 89), which would require the cognate epitopes to be immunogenic and well exposed on the virus surface. The presence of neutralization epitopes that are both immunogenic and antigenic on primary isolates affords opportunities for vaccine development that have received little attention. Part of the reason why the latter epitopes have received little attention for vaccine development relates to their high degree of variability, which may not be suitable for cross-reactive neutralizing antibody induction. Given the paucity of information on the immunogenicity of primary isolate Env, however, it would be premature to conclude that no monovalent Env will generate cross-reactive neutralizing antibodies. Also, should monovalent primary isolate Env be capable of generating antibodies that neutralize the autologous primary isolate, it might ultimately make it feasible to formulate a composite (polyvalent) Env vaccine that will target a broad spectrum of primary isolates. Few studies have examined the immunogenicity of primaryisolate envelope glycoproteins. VanCott et al. (85) showed that immunization with the gp120 protein from the R5 primary isolate HIV-1CM235 generated antibodies in baboons that neutralized TCLA strains, including the TCLA version of the vaccine strain, but not the autologous primary isolate. Similar results were obtained by Beddows et al. (4), who immunized healthy, HIV-1-seronegative volunteers with the gp120 protein from R5/X4 strain HIV-1W61D. Only Berman et al. (8) have succeeded at generating antibodies that neutralized an R5 primary isolate; this was done by immunizing rabbits with the gp120 protein from HIV-1CM244. In addition to neutralizing antibodies, increasing evidence supports a critical role for virus-specific CD8 T cells in protective immunity against HIV-1 (11, 15, 55, 61, 64, 69, 70, 92). CD8 T-cell responses to HIV-1 can be both cytotoxic (11, 15, 55, 61, 69, 92) and virus suppressive (80, 88). Recent reports of the effect of CD8 cell depletion on viremia and CD4 cell counts in SIV- or SIV-HIV-infected macaques (27, 40, 72) has given additional in vivo relevance to the importance of the CD8 antiviral response in the control of this viral infection. Even more recently, vaccine-induced, virus-specific CTL strongly correlated with viremia control and the preservation of CD4 lymphocytes in the highly pathogenic SIV-HIV 89.6P macaque model (3). The immunogenicity of Gag, Pro, and Env from R5 primary isolate HIV-1 Bx08 was investigated here in rhesus monkeys. Immunogens were delivered in the form of either purified noninfectious virus-like particles (VLP), recombinant DNA and canarypox vectors engineered to express VLP, or a combination of delivery modes. Our results reveal the priming of Gag-specific cellular responses by DNA-based immunization. We also demonstrate that envelope glycoprotein from an R5 primary isolate can be used to generate antibodies in nonhuman primates that are capable of neutralizing the autologous primary isolate. MATERIALS AND METHODS Cells and viruses. MT-2 is a CD4 human lymphoblastoid cell line that is highly permissive to cytopathic infection with TCLA strains of HIV-1 (24). Stocks of HIV-1 strains IIIB, MN, and SF2 were produced in H9 cells as previously described (47). MT-2 and H9 cells were maintained at 37 C in 5% CO 2 in growth medium consisting of RPMI 1640 medium supplemented with 12% heat-inactivated fetal bovine serum (FBS) and 50 g of gentamicin/ml. Peripheral blood mononuclear cells (PBMC) were purified from buffy coats of healthy HIV-1-seronegative individuals and stimulated for 1 day with phytohemagglutinin P as described previously (46). Growth medium for PBMC consisted of RPMI 1640 medium supplemented with 20% heat-inactivated FBS, 4% interleukin-2, and 50 g of gentamicin/ml. Monkey kidney Vero cells used for Bx08 VLP production were grown and passaged biweekly in Dulbecco s modified Eagle s medium (Flow Laboratories, McLean, Va.) supplemented with 10% heat-inactivated FBS, glutamine (2 mm), penicillin (50 IU/ml), and streptomycin (50 g/ml). Stably transfected cells secreting VLP were maintained in the presence of 0.5 mg of Geneticin (Gibco BRL, Grand Island, N.Y.) per ml. Clade B primary isolate Bx08 was isolated from an HIV-1-infected French individual 8 months postseroconversion (48) and was kindly provided by H. J. A. Fleury (Bordeaux, France). The other clade B primary isolates used in this study were Bal, JR-FL, P15, and P27. Bal and JR-FL were obtained from the National Institutes of Health AIDS Research and Reference Reagent Program (Bal was contributed by Suzanne Gartner, Mikulas Popovic, and Robert Gallo; JR-FL was contributed by Irvin Chen). Bal was isolated from explanted human infant lung tissue (21), JR-FL was isolated from frontal lobe brain tissue of a patient with AIDS dementia (53), and P15 and P27 were isolated during early seroconversion by PBMC coculture (58). Bal, JR-FL, P15, and P27 all use the CCR5 coreceptor (10, 18). Stocks of primary isolates were produced in PBMC as previously described (46). All virus stocks were made cell free by m (pore size) filtration and stored in aliquots at 80 C until use. Coreceptor usage of Bx08 was assessed in MT-2, U87-CD4-CCR5, and U87-CD4-CXCR4 cells (25) by using p24 production as a measure of infection. scd4 and MAbs. Recombinant soluble human CD4 (scd4) containing the full-length extracellular domain of human CD4 was obtained from Progenics Pharmaceuticals (Tarrytown, N.Y.). Human MAbs IgG1b12, 2G12, and 2F5 have been shown to neutralize diverse primary isolates (14, 19, 28, 59, 82). IgG1b12 recognizes an epitope in the CD4-binding domain of gp120 that is sensitive to mutations in V2 and C3 (42). 2G12 recognizes an epitope comprising residues within C2-V4 of gp120 that involved sites of N glycosylation (82). 2F5 recognizes a linear epitope in the ectodomain of gp41 having the amino acid sequence ELDKWA (51). DNA vectors. The nucleotide and amino acid numbering used throughout for HIV was that of Myers et al. (52). All pseudovirion-expressing vectors were constructed from p83-19 (57, 65), which was, in turn, derived from plasmid pmthivd25 (66). Plasmid p83-19 was engineered to incorporate several mutations to enhance safety, such as deletion of a 25-bp DNA fragment (nucleotides 753 to 777; LAI sequence) containing viral RNA packaging sequences (1, 34) and deletion of a 1.9-kbp DNA fragment containing most of the pol gene, effectively eliminating the coding sequences for reverse transcriptase and integrase. However, the deletion left intact the gene encoding the viral protease (pro), resulting in the expression of particles with processed Gag antigens. In p83-19, transcription of the HIV-1 coding sequences is regulated by the inducible human metallothionein II a (MT) promoter and a simian virus 40 polyadenylation sequence. In order to produce pseudovirions with the Env glycoprotein from the Bx08 primary isolate, the HIV-1 LAI env gene was replaced with that of HIV-1 Bx08. To this end, a 2,440-bp fragment containing the entire gp160-encoding gene of HIV-1 Bx08 was amplified by PCR from cells infected with this isolate. The PCR product was then used to replace the corresponding region in p83-19, giving rise to plasmid p133b1. In order to produce a plasmid DNA vector for immunization studies, a construct was prepared incorporating the env gene from HIV-1 Bx08 under the control of the cytomegalovirus promoter. The construct, pcmv3bx08, is derived from plasmid pcmvgdtat vpr Bx08, which was, in turn, engineered from p83-19 after several modifications. These included (i) replacement of the MT

3 VOL. 75, 2001 IMMUNOGENICITY OF HIV-1 VLP IN MACAQUES 5881 TABLE 1. Vaccination groups and schedules Macaque group no. Inoculation(s) at wk 0 and wk 4 Inoculation(s) at wk 24 and wk mg of DNA Bx08,50 g ofvlp Bx08, 100 g of QS21 Same as wk 0 and wk g ofdna Bx08,50 g ofvlp Bx08, 100 g of QS21 Same as wk 0 and wk mg of DNA Bx08 ALVAC Bx g ofdna Bx08, ALVAC Bx g ofvlp Bx08, 100 g of QS21 Same as wk 0 and wk 4 6 Control DNA ALVAC Bx08 promoter with a 1.6-kbp DNA fragment containing the human cytomegalovirus immediate-early gene promoter, enhancer, and intron A sequences; (ii) deletion of the coding sequences for HIV-1 Tat and Vpr; (iii) replacement of the HIV-1 gp120 Bx08 signal peptide sequence with the human herpes simplex virus glycoprotein D signal peptide sequence; (iv) replacement of the Amp r -encoding gene with the Kan r -encoding gene; and (v) replacement of the simian virus 40 polyadenylation sequence with that of the bovine growth hormone-encoding gene. The nucleotide sequences of all constructs were confirmed by DNA sequencing. Production of noninfectious VLP for immunogenicity studies. Vero cells were transfected at passage 141 with plasmid p133b1 by the calcium phosphate method as specified previously (57), and stable transfectants expressing noninfectious HIV-1-like particles were established. VLP were isolated from the supernatants of a stable clone (DD19) after induction of the Vero cells with heavy metals (57). Expression of Gag and Env was verified by Western blot assay using, respectively, an anti-hiv-1 p24 mouse MAb (NEA-9306; Dupont Canada, Inc., Markham, Ontario, Canada) and MAb K3A, which recognizes gp120 envelope proteins from different clades (kindly supplied by Ronald Kennedy). Fully assembled, Env-containing particles were isolated from the supernatants of the stably engineered Vero DD19 cell clone by ultracentrifugation through a glycerol cushion and purified by sucrose gradient fractionation (66). The p24 contents of the various particle species were determined by a p24-specific enzyme immunoassay (Coulter Immunology, Hialeah, Fla.). The cell line secreted approximately 800 to 950 g of p24 per liter. Construction of recombinant canary poxvirus vcp1579. Recombinant vcp1579 was engineered to express HIV-1 LAI Gag and Pro, together with HIV- 1 Bx08 gp120 fused to a 28-amino-acid fragment of the transmembrane domain of HIV-1 LAI gp41, and a synthetic polypeptide containing Pol and Nef CTL epitopes. This recombinant is similar to vcp1452 (10, 79), except that the gene fragment encoding gp120 LAI was replaced with its cognate sequences from HIV- 1 Bx08. Due to the absence of the gp41 ectodomain, the expressed envelope glycoprotein would be expected to consist of membrane-anchored gp120. The construct was generated in several steps. Briefly, the vector-modifying sequences from the pmpc6h6k3e3 insertion vector, encoding E3L and K3L, were inserted into the C6 locus of recombinant vcp1566. Recombinant vcp1566 was, in turn, generated by insertion of an expression cassette encoding a synthetic polypeptide containing Pol and Nef CTL epitopes into the C5 locus of vcp1453. Recombinant vcp1453 was obtained after coinsertion of the genes encoding the HIV-1 Bx08 Env and HIV-1 LAI Gag-Pro products into the ALVAC genome at the C3 locus. Macaque immunizations. Rhesus macaques were divided into six groups with four animals in each group, as summarized in Table 1. Briefly, animals in groups 1 and 2 were inoculated with either a high dose (3 mg) or a low dose (600 g) of DNA Bx08, respectively, together with 50 g ofvlp Bx08 in QS21 (100 g) at weeks 0, 4, 24, and 44. Animals in groups 3 and 4 were inoculated with either the high dose or the low dose of DNA Bx08, respectively, at weeks 0 and 4 and were boosted with ALVAC Bx08 at weeks 24 and 44 ( PFU at week 24 and PFU at week 44). Animals in group 5 were inoculated with 50 g ofvlp Bx08 in QS21 adjuvant at weeks 0, 4, 24, and 44. Animals in group 6 received 3 mg of control DNA at weeks 0 and 4 and were boosted with ALVAC Bx08 at weeks 24 and 44 (same doses as above). All immunogens were administered in a volume of 2 ml intramuscularly. The QS21 adjuvant was prepared and filter sterilized before filling in of single-dose vials and stored at 20 C. Western blot. Antibodies specific for HIV-1 antigens were assessed with a commercial HIV-1 Western blot kit (Genetic Systems, Sanofi Diagnostics Pasteur, Inc.) that is based on the LAI strain of HIV-1. Enzyme-linked immunosorbent assay. Bx08-V3 peptide-specific binding antibodies were assessed in Nunc (Roskilde, Denmark) Immunoplates (MaxiSorb F96) using alkaline phosphatase-conjugated goat anti-monkey IgG as described previously (16). Plasma samples were assayed in duplicate at a 1:50 dilution, and values are given as the average A 405. The Bx08-V3 peptide had the amino acid sequence TRPNNNTRKSIHIGPGRAFYTTGDIIGDIR and was made by Syn- Pep Corporation (Dublin, Calif.). The peptide was judged to be 91.5% pure by high-pressure liquid chromatography analysis. This same peptide was used in neutralization competition assays as described below. Neutralizing antibody assays. Neutralization of HIV-1 Bx08 and other primary isolates was assessed in human PBMC by using a reduction in p24 Gag antigen synthesis as described previously (46, 58). Briefly, % tissue culture-infective dose of virus were incubated with various dilutions of serum samples for 1 h at 37 C before the addition of PBMC. Cells were washed three times with 200 l of growth medium and resuspended in 200 l of fresh interleukin-2 growth medium 1 day later. Culture supernatants were assessed for p24 content at a time when p24 synthesis in virus control wells (no test sample) was in a linear phase of increase, which is when optimum sensitivity is achieved in this assay (93). Neutralization titers were defined as either the dilution of serum or concentration of scd4 and MAbs at which p24 synthesis was reduced by 80% relative to that of a negative control. These titers refer to conditions under which serum samples were incubated with virus before the addition of cells. All serum samples were heat inactivated for 1 h at 56 C prior to assay. V3 peptide neutralization competition assays were performed by preincubating serum samples with either phosphate-buffered saline (PBS) or Bx08-V3 peptide (50 g/ml [final concentration]) for 1 h at 37 C. The serum samples were then assayed at a 1:3 dilution on the virus as described above. Neutralization of HIV-1 strains IIIB, MN, and SF2 was measured in an MT-2 cell-killing assay by using neutral red to quantify the fraction of cells that survived virus-induced cytopathic effects (47). Briefly, % tissue culture-infective doses of virus were incubated with multiple dilutions of serum samples for 1 h at 37 C before the addition of cells. The incubation was continued until extensive syncytium formation had just occurred in wells that contained no serum sample (usually 4 to 6 days). Neutralizing antibody titers are defined as the serum dilution (before the addition of cells) at which 50% of the cells were protected from virus-induced killing. A 50% reduction in cell-killing corresponds to an approximately 90% reduction in viral Gag antigen synthesis in this assay (10, 54). Each set of assays included a positive control serum that had been assayed multiple times and had a known average titer. ELISpot assay for IFN- release from antigen-specific PBMC. PBMC were separated from whole blood by Lymphocyte Separation Medium (Cellgro), washed twice, counted, and viably frozen in freezing medium for time course studies. Cells were thawed, washed twice, resuspended at /ml, and stimulated for 48 h with 2 g of HIV-1 HXB2 Gag peptide pools (National Institutes of Health AIDS Research and Reference Reagent Program) per ml. The peptides were 20-mers overlapping by 10 amino acids and were divided into two pools of peptides with 25 peptides per pool. Pool 1 represented amino acids 1 to 260, while pool 2 represented amino acids 251 to 500. Ovalbumin peptide was used as a negative control, and 50 ng of phorbol myristate acetate (Sigma) per ml plus 1 g of ionomycin (Sigma) per ml served as a positive control. Ninety-sixwell mixed cellulose ester filtration plates (Millipore) were coated with 50 l of a5- g/ml concentration of mouse anti-human IFN- MAb 1-D1K (Biosource International) overnight at 4 C. Plates were then washed four times with PBS and blocked with RPMI (Cellgro) medium supplemented with L-glutamine, penicillin, and 10% heat-inactivated FBS (HyClone) for 1 h at 37 C. Prestimulated cells were then added to the coated plate in 100 l of R10. Duplicate wells containing either or stimulated PBMC were plated for each sample and incubated for 16 to 20 h at 37 C. Plates were washed four times with PBS (Cellgro) and then four times with PBS containing 0.05% Tween 20 (Sigma) to remove all cells and debris from the plates. Next, 100 l of1- g/ml biotinylated anti-ifn- MAb 1-D1K (Mabtech) diluted in PBS was added. After 2 h, the plates were washed four times with PBS containing 0.1% Tween 20 and incubated with 100 l of Vectastain ABC (Vector Laboratories) per well for 1 h. Following four washes with PBS plus 0.1% Tween 20, Stable DAB (Research Genetics) was added to the wells and monitored for color development (approx-

4 5882 MONTEFIORI ET AL. J. VIROL. imately 5 min). The Stable DAB reaction was stopped by washing the wells three times with H 2 O. Wells were counted visually under a dissecting microscope (Olympus) after the plates had dried. RESULTS Biologic and immunologic properties of HIV-1 Bx08. Coreceptor usage was determined by infection of cells that coexpressed CD4 and either CCR5 or CXCR4. HIV-1 Bx08 was able to infect U87-CD4-CCR5 cells but not U87-CD4-CXCR4 cells or MT-2 cells (data not shown). This outcome is consistent with an R5 phenotype for this virus. As a control for the integrity of the cells used for X4 phenotype classification, HIV- 1 IIIB was able to infect U87-CD4-CXCR4 and MT-2 cells but not U87-CD4-CCR5 cells. The neutralization sensitivity of HIV-1 Bx08 was assessed with scd4 and human MAbs in the PBMC assay. The virus was sensitive to all four reagents. The minimum concentration of scd4 required to inhibit HIV-1 Bx08 was 9.9 g/ml. This concentration of scd4 is approximately 500 to 5,000 times higher than that required to inhibit TCLA strains (10, 17, 49) but was moderately low compared to other primary isolates, most of which resist inhibition by this same preparation of scd4 at concentrations of up to 50 g/ml (10). The virus was also neutralized by the MAbs, where the 80% inhibition doses were 11.5 (IgG1b12), 1.2 (2G12), and 27.9 (2F5) g/ml. HIV-1 seroconversion. HIV-1-specific IgG was assessed by Western blot assay at multiple time points throughout the immunization schedule. Figure 1A shows that all of the immunized animals possessed Gag-specific antibodies by week 6 (2 weeks after the second inoculation). Gag-specific seroconversion was strongest in animals that received VLP Bx08 either alone (group 5) or in combination with the high and low doses of DNA Bx08 (groups 1 and 2, respectively). Gag-specific seroconversion was variable and less potent at this time in animals that received the high and low doses of DNA Bx08 (groups 3 and 4, respectively). A similar pattern was noted for Pro-specific antibodies (p31), with the exception of very low levels of anti- Pro antibodies in animals inoculated with the low dose of DNA Bx08 (group 4). Little or no Env-specific antibody was detected at this time. Western blot reactivities had intensified by 2 weeks following the final immunization (week 46) where, in addition to anti-gag antibodies, all immunized animals showed evidence of Pro-specific seroconversion (Fig. 1B). We noted that the anti-gag and anti-pro reactivity was relatively weak in animals RAd5 (group 3) and REl5 (group 4). Env-specific antibodies (for gp160 and gp41 but not for gp120) were detected at this time in animals in groups 1, 2, and 5 only. The lack of detection of gp120-specific antibodies in these animals could be due to genetic and antigenic dissimilarities in the gp120 of Bx08 compared to the IIIB gp120 antigen used in the Western blot assay strips. All of the animals immunized with control DNA and boosted with ALVAC Bx08 were seronegative for Env at week 46 (Fig. 1B). A boosting effect at weeks 26 and 46 (2 weeks after the third and fourth inoculations, respectively) was seen in each animal, where evidence of seroconversion remained present 12 weeks after the final inoculation (data not shown). Neutralization of HIV-1 Bx08 with sera from immunized macaques. Neutralization of HIV-1 Bx08 was assessed in human PBMC with serum collected preimmunization and after the second, third, and fourth (final) immunizations. These assays were performed without knowledge of the animal group assignments. Figure 2A shows that neutralizing antibodies were detected in three of the four animals in group 1 (high-dose DNA Bx08 plus VLP Bx08 ), four of the four animals in group 2 (low-dose DNA Bx08 plus VLP Bx08 ), three of the four animals in group 3 (high-dose DNA Bx08 plus ALVAC Bx08 boosting), and three of the four animals in group 5 (VLP Bx08 ). These neutralization titers ranged from 1:2 to 1:48. The highest titer was achieved in an animal (RYl5) immunized four times with VLP Bx08. Notably, a 1:2 dilution of sera from several animals in groups 1, 2, and 5 achieved 90% neutralization and serum from at least one animal in each of the latter groups achieved 99 to 100% neutralization. The potency of neutralization at a 1:2 serum dilution did not correlate with the 80% neutralization titer. As shown in Fig. 2B, no HIV-1 Bx08 -specific neutralization was detected with sera from animals immunized with the low dose of DNA Bx08 plus ALVAC Bx08 (group 4) or control DNA plus ALVAC Bx08 (group 6). Sera from these animals reduced p24 synthesis by 113% to 76% at the lowest dilution tested (1:2), which is within the normal variation range of this assay. Although an 80% reduction in p24 Gag antigen synthesis (fivefold decrease in infectivity) has proven to be the most reliable minimum cutoff for a true positive (10), two animals in the group that received the low dose of DNA Bx08 plus ALVAC Bx08 (REl5 and RHg5) had neutralization values of 76 and 75%, respectively, after final boosting. Absence of cross-reactive neutralizing antibodies. Crossneutralizing activity was assessed with three TCLA strains (IIIB, MN, and SF2) and four heterologous R5, clade B primary isolates. In only two cases were neutralizing antibodies detected with a TCLA strain as measured in the MT-2 cell assay (data not shown). These cases occurred 2 weeks after final boosting and included sera from animal RLj5 in group 3 and animal REl5 in group 4, both having weak neutralizing activity against HIV-1 MN (titers of 1:29 and 1:50, respectively). Nine serum samples that neutralized HIV-1 Bx08 were further tested for the ability to neutralize heterologous R5 primary isolates in the PBMC assay. The results of the latter assays were all negative (Table 2). Antibody specificity for the V3 loop. Serum samples obtained 2 weeks after final boosting (week 46) were tested for the presence antibodies that could bind a Bx08-V3 peptide. Sera from animals in groups 1, 2, and 5 and, to a lesser extent, groups 3 and 4 tested positive by enzyme-linked immunosorbent assay (Fig. 3). The detection of V3-specific antibodies did not always predict the ability to neutralize HIV-1 Bx08. For example, at least three serum samples that neutralized HIV- 1 Bx08 had no detectable V3-specific antibodies (RZh5, RMg5, and RWh5). In addition, two samples that were positive for V3-specific antibodies (RSg5 and RNl5) failed to neutralize HIV-1 Bx08. The frequent detection of antibodies to the V3 loop of Bx08 is strong evidence of the presence of gp120- specific antibodies that were not detected by the Western blot assay (Fig. 1). Two serum samples that contained antibodies reactive with the Bx08 V3 peptide and neutralized HIV-1 Bx08 were tested for neutralizing activity in the presence and absence of Bx08 V3 peptide. Similar experiments have shown this to be a very

5 VOL. 75, 2001 IMMUNOGENICITY OF HIV-1 VLP IN MACAQUES 5883 FIG. 1. Western blot analysis of HIV-1 antigen-specific antibodies. Serum samples were assayed at a 1:100 dilution. Panels: A, sera obtained at week 6; B, sera obtained at week 46. wks, weeks; HIV, HIV negative; HIV, weakly HIV positive; HIV, strongly HIV positive. effective means by which to determine whether a portion of the neutralizing antibodies are V3 specific (10, 16). The results of these assays showed that the Bx08 V3 peptide (50 g/ml) was not able to outcompete the neutralizing activity of these serum samples (Fig. 4). Analysis of IFN- secretion. PBMC from the vaccinated and control animals were tested for the ability to secrete IFN- upon stimulation with two pools of overlapping peptides rep- resenting the Gag sequence of HIV-1HXB2. Peptide pool 1 spanned amino acids 1 to 260, while peptide pool 2 spanned amino acids 251 to 500. The number of IFN- spots per 106 PBMC for each animal at each time point is shown in Fig. 5, and the average value for each group is shown in Fig. 6. We measured Gag-specific responses 2 weeks after the second immunization, a time when near-peak cellular responses would be expected, and then 3 months later (16 weeks), when mem-

6 5884 MONTEFIORI ET AL. J. VIROL. FIG. 2. Neutralization of HIV-1 Bx08 by sera from immunized animals. Bar height represents the percent reduction in p24 relative to the amount of p24 produced in the presence of the corresponding preimmunization serum from each animal as assayed at a 1:2 dilution. Values above the bars are the highest serum dilutions at which p24 production was reduced by 80%. Panels: A, animal groups with detectable neutralizing antibodies; B, animal groups with no detectable neutralizing antibodies.

7 VOL. 75, 2001 IMMUNOGENICITY OF HIV-1 VLP IN MACAQUES 5885 FIG. 3. Bx08-V3 peptide-binding antibodies. Serum samples obtained 2 weeks after the fourth immunization (week 46) were tested at a 1:50 dilution for antibodies reactive with a peptide corresponding to the V3 loop of HIV-1 Bx08. Bar height represents the average A 450 of duplicate tests. All duplicate values agreed within 5% of the average. The value obtained with serum from an HIV-1-naive macaque is shown by a dashed line. Values greater than twice that of the negative control were considered to be positive. Serum samples that neutralized HIV-1 Bx08 are indicated by asterisks above the bars. OD, optical density; grp, group. ory responses from the first two immunizations would be in effect. The PBMC were then screened 2 weeks after each of the next two immunizations (weeks 26 and 46) to determine boosting effects and finally at week 56 (3 months following the final immunization) for immunologic memory for the HIV-1 antigen. The presence of 20 ELISpots in an assay was considered a positive test. All groups of animals primed with DNA Bx08 showed clear positive Gag-specific ELISpot responses following the third immunization, while the groups of animals receiving only VLP Bx08 or ALVAC Bx08 failed to score positive. Following the Animal a TABLE 2. Neutralization of heterologous R5 primary HIV-1 isolates Titer of NAb to HIV-1 Bx08 b % Reduction in p24 c with HIV-1 isolate: Bal JR-FL P15 P27 RY15 1: RKh5 1: RBf5 1: RQj5 1: RJh5 1: RZh5 1: RWh5 1: RHg5 1: RBu4 1: a Serum samples were obtained at week 46. b Titers are reproduced from Fig. 2A and represent the serum dilutions at which p24 production was reduced by 80% relative to that obtained with preimmunization serum. NAb, neutralizing antibody. c Serum samples were assayed at a 1:4 dilution with virus in the PBMC blast assay. Percent reduction in p24 was calculated relative to the amount of p24 produced in the presence of pooled preimmunization serum samples from animals RBf5, RSg5, RHg5, RRj5, and RU15. Amounts of p24 produced in the presence of pooled negative serum sample: Bal, 64 ng/ml; JR-FL, 36 ng/ml; P15, 60 ng/ml; P27, 38 ng/ml. first two inoculations with DNA Bx08, the groups receiving the low dose of DNA Bx08 had more animals with 20 ELISpots (five of eight animals) than did the groups receiving the high dose of DNA Bx08 (none of eight animals). Following the first boost with either VLP Bx08 or ALVAC Bx08, all DNA Bx08 - primed groups contained animals that were positive, with 12 of FIG. 4. Inability to detect neutralizing antibodies specific for the HIV-1 Bx08 V3 loop. Two serum samples that contained antibodies reactive with a Bx08 V3 peptide and that neutralized HIV-1 Bx08 were tested for neutralizing activity in the presence and absence of the Bx08 V3 peptide. Samples were preincubated with either peptide (50 g/ml) or an equal volume of sterile PBS for 1hat37 C and assayed at a 1:3 dilution. Both postimmunization serum samples were obtained at week 46 (2 weeks after final boosting). Dark-shaded bars, no V3 peptide; light-shaded bars, with V3 peptide; pre, preimmunization serum; post, postimmunization serum. Each error bar represents the standard deviation of the average of triplicate values.

8 5886 MONTEFIORI ET AL. J. VIROL. FIG. 5. Number of HIV-1 Gag-specific IFN- -producing cells detected by ELISpot assay with PBMC from individual animals. Solid bars represent cells stimulated with peptide pool 1, covering the amino-terminal half of Gag. Hatched bars represent cells stimulated with peptide pool 2, covering the carboxyl-terminal half of Gag. Values on the y axis are numbers of IFN- -producing cells per million cells. ND, not done.

9 VOL. 75, 2001 IMMUNOGENICITY OF HIV-1 VLP IN MACAQUES 5887 FIG. 6. Average number of IFN- -producing cells in each group of animals. The values below the x axis are the fractions of animals that were positive for Bx08-specific neutralizing antibodies at weeks 26 and 46. Downloaded from 16 animals scoring 20 ELISpots. Animals that scored positive typically did so with both peptide pools (28 of 42 positive samples). The second boost did not markedly increase the magnitude or frequency of the ELISpot responses. At the end of the experiment, the frequency of Gag-specific ELISpotpositive animals was only slightly lower (11 of 16 animals) than at 2 weeks after the first boost (12 of 16 animals). Boosting with ALVAC Bx08 was no more effective than boosting with DNA Bx08 combined with VLP Bx08. By comparison, a typical response to corresponding SIV Gag peptide pools using cells from SIV-infected rhesus macaques is 50 to 150 ELISpots, with most animals responding to both peptide pools (data not shown). DISCUSSION Despite a growing desire to generate antibodies that are capable of neutralizing primary isolates, very few primary-isolate Envs have been investigated as potential immunogens for inclusion in an HIV-1 vaccine. Here we show that noninfectious VLP containing the surface envelope glycoprotein of an R5 primary isolate were capable of generating antibodies that neutralized the vaccine strain of virus. The antibodies could be generated by (i) coinoculation with DNA Bx08 and VLP Bx08, (ii) priming with DNA Bx08 and boosting with ALVAC Bx08, and (iii) inoculation with VLP Bx08 alone. Most notable was the fact that, in certain cases, neutralizing antibodies were induced by recombinant vectors without the need for protein boosting a milestone that has not been reported previously with respect to primary-isolate neutralization by macaque serum. Unlike the neutralizing antibodies generated by most of the candidate HIV-1 Env vaccines tested to date, the neutralizing antibodies generated here did not appear to be directed to linear epitopes in the V3 loop of gp120. Two lines of evidence support this conclusion. First, some serum samples with neutralizing activity had little or no antibody reactive with a Bx08 V3 loop peptide. Second, the neutralizing activity of two serum samples with strong reactivity to the Bx08 V3 loop peptide was not reduced in the presence of a high concentration (50 g/ml) of the V3 loop peptide. These results suggest that the neutralizing antibodies targeted one or more epitopes outside the V3 loop. We note, however, that our experiments with linear peptides do not eliminate the possibility that the epitope(s) consisted of a conformational structure within V3 that was not represented by the linear peptide. A disadvantage of the neutralizing antibodies seen here was their minimal cross-reactivity. Specifically, the antibodies had little or no neutralizing activity against TCLA strains and heterologous R5 primary isolates. The neutralizing antibody response generated by these candidate vaccines needs to be improved to target a broad spectrum of HIV-1 antigenic variants. We also noted that the Bx08-specific neutralizing antibodies were low in titer (1:2 to 1:48); however, the small magnitude of this response may be less of a concern than the limited strain specificity. For example, results of recent passiveimmunization experiments with nonhuman primates indicate on November 18, 2018 by guest

10 5888 MONTEFIORI ET AL. J. VIROL. that preexisting neutralization titers of 1:10 or less may provide complete or partial protection against intravenous and mucosal challenge with simian-human immunodeficiency virus (35, 38, 76). Given that natural HIV-1 transmission appears to be virus dose dependent (60), where not all exposures result in infection (39), it is conceivable that a 5- or 10-fold reduction in virus infectivity by minimally diluted serum in vitro will correspond to an undiluted potency in vivo that is effective prophylactically. A number of recombinant vaccine vectors are also known to prime B cells for a rapid secondary neutralizing antibody response generated by HIV-1 Env protein boosting (23, 79), and at least one example exists of a dramatic secondary neutralizing antibody response after a virus challenge of macaques that had been primed with recombinant modified vaccinia virus strain Ankara-SIV vaccines (54). It may be expected that DNA vectors expressing VLP will prime for similar secondary neutralizing antibody production in response to HIV-1 infection. We also detected HIV-1 Gag-specific cellular immune responses in the groups of animals that were inoculated with DNA Bx08. All groups of animals receiving this immunogen scored positive for IFN- -producing cells, and with the exception of a weak positive response to Gag pool 1 in one animal (RWh5, week 56), no IFN- -producing cells were detected in animals inoculated with either VLP Bx08 or ALVAC Bx08 alone. Thus, as judged by the best overall immune responses (i.e., IFN- -producing cells and HIV-1 Bx08 neutralizing antibody), coinoculation with a low dose of DNA Bx08 plus VLP Bx08 was the superior immunization modality investigated (Fig. 6). The Gag peptides used for PBMC stimulation were not completely matched to the vaccine strain, making it possible that some strain-specific reactivities went undetected in our assay. Despite this, it is noteworthy that IFN- production could be stimulated in many cases with two overlapping peptide pools covering the amino-terminal and carboxyl-terminal halves of Gag as evidence that at least two specificities of the cellular response were induced by vaccination. While we consider the combined humoral and cellular immune responses elicited by these vaccine candidates to be encouraging, we also readily acknowledge the limited benefit that can be expected from neutralizing antibodies that are so narrow in specificity. Nonetheless, our results point to several vaccine strategies that may be useful for delivering a more appropriate immunogen for cross-reactive neutralizing antibody induction once that immunogen has been identified. We also did not determine whether the structure of Bx08 Env incorporated into the VLP is optimal for neutralizing antibody induction. For example, it has been argued that the native oligomeric (trimeric) structure of gp120-gp41 heterodimers must be preserved for optimal immunogenicity (13). Although it is possible that the Bx08 VLP expressed by our DNA vector contained native oligomeric envelope glycoproteins, we have not confirmed this. Furthermore, the envelope glycoproteins expressed by ALVAC Bx08 consisted of membrane-anchored gp120 lacking the gp41 ectodomain, which may not represent the most suitable structure for optimal immunogenicity. Additional studies are required to determine the full potential of VLP as an immunogen for neutralizing antibody induction. ACKNOWLEDGMENTS We thank H. J. A. Fleury for HIV-1 Bx08 and Dennis Burton, Herman Katinger, and John Mascola for MAbs. We also thank Ginger James, Lauren Rodrigues, Heike Marshall, and Lisa Murdin for excellent technical assistance. This work was supported by funding from Aventis Pasteur and the National Institutes of Health (AI-85343). REFERENCES 1. Aldovini, A., and R. Young Mutations of RNA and protein sequences involved in human immunodeficiency virus type 1 packaging result in production of noninfectious virus. J. Virol. 64: Baba, T. W., V. Liska, R. Hofmann-Lehmann, J. Vlasak, W. Xu, S. Ayehunie, L. A. Cavacini, M. R. Posner, H. Katinger, G. Stiegler, B. J. Bernacky, T. A. Rizvi, R. Schmidt, L. R. Hill, M. E. Keeling, Y. Lu, J. E. Wright, T.-C. Chou, and R. M. Ruprecht Human neutralizing monoclonal antibodies of the IgG1 subtype protect against mucosal simian-human immunodeficiency virus infection. Nat. Med. 6: 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 Control of viremia and prevention of clinical AIDS in rhesus monkeys by cytokine-augmented DNA vaccination. Science 290: Beddows, S., S. Lister, R. Cheingsong, C. Bruck, and J. Weber Comparison of the antibody repertoire generated in healthy volunteers following immunization with a monomeric recombinant gp120 construct derived from a CCR5/CXCR4-using human immunodeficiency virus type 1 isolate with sera from naturally infected individuals. J. Virol. 73: Belshe, R. B., G. J. Gorse, M. J. Mulligan, T. G. Evans, M. C. Keefer, J.-L. Excler, A.-M. Duliege, J. Tartaglia, W. I. Cox, J. McNamara, K.-L. Hwang, A. Bradney, D. Montefiori, and K. J. Weinhold Induction of immune responses to HIV-1 canarypox virus (ALVAC) HIV-1 and gp120 SF-2 recombinant vaccines in uninfected volunteers. AIDS 12: Berger, E. A HIV entry and tropism: the chemokine receptor connection. AIDS 11(Suppl. A):S3 S Berger, E. A., R. W. Doms, E. M. Fenyö, B. T. M. Korber, D. R. Littman, J. P. Moore, Q. J. Sattentau, H. Schuitemaker, J. Sodrsoki, and R. A. Weiss HIV-1 phenotypes classified by co-receptor usage. Nature 391: Berman, P. W., W. Huang, L. Riddle, A. M. Gray, T. Wrin, J. Vennari, A. Johnson, M. Klaussen, H. Prashad, C. Köhne, C. dewit, and T. J. Gregory Development of bivalent (B/E) vaccines able to neutralize CCR5- dependent viruses from the United States and Thailand. Virology 265: Bou-Habib, D. C., G. Roderiquez, T. Oravecz, P. W. Berman, P. Lusso, and M. A. Norcross Cryptic nature of envelope V3 region epitopes protects primary monocytotropic human immunodeficiency virus type 1 from antibody neutralization. J. Virol. 68: Bures, R., A. Gaitan, T. Zhu, C. Graziosi, K. M. McGrath, J. Tartaglia, P. Caudrelier, R. El Habib, M. Klein, A. Lazzarin, D. M. Stablein, M. Deers, L. Corey, M. L. Greenberg, D. H. Schwartz, and D. C. Montefiori Immunization with recombinant canarypox vectors expressing membrane-anchored gp120 followed by gp160 protein boosting fails to generate antibodies that neutralize R5 primary isolates of human immunodeficiency virus type 1. AIDS Res. Hum. Retrovir. 16: Burrow, P., H. Lewicki, B. H. Hahn, G. M. Shaw, and M. B. A. Oldstone Virus-specific CD8 cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J. Virol. 68: Burton, D. R., and D. C. Montefiori The antibody response in HIV-1 infection. AIDS 11(Suppl. A):S87 S Burton, D. R A vaccine for HIV type 1: the antibody perspective. Proc. Natl. Acad. Sci. USA 94: Burton, D. R., J. Pyati, R. Koduri, S. J. Sharp, G. B. Thornton, P. W. H. I. Parren, L. S. W. Sawyer, R. M. Hendry, N. Dunlop, P. L. Nara, M. Lamacchia, E. Garratty, E. R. Stiehm, Y. J. Bryson, Y. Cao, J. P. Moore, D. D. Ho, and C. F. Barbas III Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 266: Cao, Y., L. Qin, L. Zhang, J. Safrit, and D. D. Ho Virologic and immunologic characterization of long-term survivors of HIV-1 infection. N. Engl. J. Med. 332: Crawford, J. M., P. L. Earl, B. Moss, K. A. Reimann, M. S. Wyand, K. H. Manson, M. Bilska, J. T. Zhou, C. D. Pauza, P. W. H. I. Parren, D. R. Burton, J. G. Sodroski, N. L. Letvin, and D. C. Montefiori Characterization of primary isolate-like variants of simian-human immunodeficiency virus. J. Virol. 73: Daar, E. S., X. L. Li, T. Moudgil, and D. D. Ho High concentrations of recombinant soluble CD4 are required to neutralize primary human immunodeficiency type 1 isolates. Proc. Natl. Acad. Sci. USA 87: Deng, H., D. Unutmaz, V. N. KewalRamani, and D. R. Littman

11 VOL. 75, 2001 IMMUNOGENICITY OF HIV-1 VLP IN MACAQUES 5889 Expression cloning of new receptors used by simian and human immunodeficiency viruses. Nature 388: D Souza, M. P., D. Livnat, J. A. Bradac, and S. H. Bridges Evaluation of monoclonal antibodies to human immunodeficiency virus type 1 primary isolates by neutralization assays: performance criteria for selecting candidate antibodies for clinical trials. J. Infect. Dis. 175: Foresman, L., F. Jia, Z. Li, C. Wang, E. B. Stephens, M. Sahni, O. Narayan, and S. V. Joag Neutralizing antibodies administered before, but not after, virulent SHIV prevent infection in macaques. AIDS Res. Hum. Retrovir. 14: Gartner, S., P. Markovits, D. M. Markovitz, M. H. Kaplan, R. C. Gallo, and M. Popovic The role of mononuclear phagocytes in HTLV/LAV infection. Science 233: Gaudin, M.-C., P. W. H. I. Parren, R. Weir, C. F. Barbas, D. R. Burton, and R. A. Koup Passive immunization with a human monoclonal antibody protects hu-pbl-scid mice against challenge by primary isolates of HIV-1. Nat. Med. 3: Graham, B. S Serologic responses to candidate AIDS vaccines. AIDS Res. Hum. Retrovir. 10(Suppl. 2):S145 S Harada, S., Y. Koyanagi, and N. Yamamoto Infection of HTLV-III/ LAV in HTLV-I-carrying cells MT-2 and MT-4 and application in a plaque assay. Science 229: Hill, C. M., H. Deng, D. Unutmaz, V. N. Kewalramani, L. Bastiani, M. K. Gorny, S. Zolla-Pazner, and D. R. Littman Envelope glycoproteins from human immunodeficiency virus types 1 and 2 and simian immunodeficiency virus can use human CCR5 as a coreceptor for viral entry and make direct CD4-dependent interactions with this chemokine receptor. J. Virol. 71: Javaherian, K., A. J. Langlois, C. McDanal, K. L. Ross, L. I. Eckler, C. L. Jellis, A. T. Profy, J. R. Rusche, D. P. Bolognesi, S. D. Putney, and T. J. Matthews Principal neutralizing domain of the human immunodeficiency virus type 1 envelope protein. Proc. Natl. Acad. Sci. USA 86: Jin, X., D. E. Bauer, S. E. Tuttleton, S. Lewin, A. Gettie, J. Blanchard, C. E. Irwin, J. T. Safrit, J. Mittler, L. Weinberger, L. G. Kostrikis, L. Zhang, A. S. Perelson, and D. D. Ho Dramatic rise in plasma viremia after CD8 T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 189: Kessler, J. A., II, P. M. McKenna, E. A. Emini, C. P. Chan, M. D. Patel, S. K. Gupta, G. E. Mark III, C. F. Barbas III, D. R. Burton, and A. J. Conley The recombinant human monoclonal antibody IgG1b12 neutralizes diverse human immunodeficiency virus type 1 primary isolates. AIDS Res. Hum. Retrovir. 13: Koup, R. A., J. T. Safrit, Y. Cao, C. A. Andrews, G. McLeod, W. Borkowsky, C. Farthing, and D. D. Ho Temporal association of cellular immune responses with initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 68: Kwon, P. D., R. Wyatt, J. Robinson, R. W. Sweet, J. Sodroski, and W. A. Hendrickson Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 393: LaCasse, R. A., K. E. Follis, T. Moudgil, M. Trahey, J. M. Binley, V. Planelles, S. Zolla-Pazner, and J. H. Nunberg Coreceptor utilization by human immunodeficiency virus type 1 is not a primary determinant of neutralization sensitivity. J. Virol. 72: LaCasse, R. A., K. E. Follis, M. Trahey, J. D. Scarborough, D. R. Littman, and J. H. Nunberg Fusion-competent vaccines: broad neutralization of primary isolates of HIV. Science 283: Letvin, N. L Progress in the development of an HIV-1 vaccine. Science 280: Lever, A., H. Gottlinger, W. Haseltine, and J. Sodroski Identification of a sequence required for efficient packaging of human immunodeficiency virus type 1 RNA into virions. J. Virol. 63: Mascola, J. R., M. G. Lewis, G. Stiegler, D. Harris, T. C. VanCott, D. Hayes, M. K. Louder, C. R. Brown, C. V. Sapan, S. S. Frankel, Y. Lu, M. L. Robb, H. Katinger, and D. L. Birx Protection of macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J. Virol. 73: Mascola, J. R., M. K. Louder, T. C. VanCott, C. V. Sapan, J. S. Lambert, L. R. Muenz, B. Bunow, D. L. Birx, and M. L. Robb Potent and synergistic neutralization of human immunodeficiency virus (HIV) type 1 primary isolates by hyperimmune anti-hiv immunoglobulin combined with monoclonal antibodies 2F5 and 2G12. J. Virol. 71: Mascola, J. R., S. W. Snyder, O. S. Weislow, S. M. Belay, R. B. Belshe, D. H. Schwartz, M. L. Clements, R. Dolin, B. S. Graham, G. J. Gorse, M. C. Keefer, M. J. McElrath, M. C. Walker, K. F. Wagner, J. G. McNeil, F. E. McCutchan, and D. S. Burke Immunization with envelope subunit vaccine products elicits neutralizing antibodies against laboratory-adapted but not primary isolates of human immunodeficiency virus type 1. J. Infect. Dis. 173: Mascola, J. R., G. Stiegler, T. C. VanCott, H. Katinger, C. B. Carpenter, C. E. Hanson, H. Beary, D. Hayes, S. S. Frankel, D. L. Birx, and M. G. Lewis Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat. Med. 6: Mastro, T. D., and D. Kitayaporn HIV type 1 transmission probabilities: estimates from epidemiologic studies. AIDS Res. Hum. Retrovir. 14(Suppl. 3):S223 S Matano, T., R. Shibata, C. Siemon, M. Connors, H. C. Lane, and M. A. Martin Administration of an anti-cd8 monoclonal antibody interferes with the clearance of chimeric simian/human immunodeficiency virus during primary infections of rhesus macaques. J. Virol. 72: McDougal, J. S., M. S. Kennedy, S. L. Orloff, J. K. A. Nicholson, and T. J. Spira Mechanism of human immunodeficiency virus type 1 (HIV-1) neutralization: irreversible inactivation of infectivity by anti-hiv-1 antibody. J. Virol. 70: Mo, H., L. Stamatatos, J. E. Ip, C. F. Barbas, P. W. H. I. Parren, D. R. Burton, J. P. Moore, and D. D. Ho Human immunodeficiency virus type 1 mutants that escape neutralization by human monoclonal antibody IgG1b12. J. Virol. 71: Montefiori, D. C., R. G. Collman, T. R. Fouts, J. Y. Zhou, M. Bilska, J. A. Hoxie, J. P. Moore, and D. P. Bolognesi Evidence that antibodymediated neutralization of human immunodeficiency virus type 1 is independent of coreceptor usage. J. Virol. 72: Montefiori, D. C., and T. G. Evans Toward an HIV-1 vaccine that generates potent, broadly cross-reactive neutralizing antibodies. AIDS Res. Hum. Retrovir. 15: Montefiori, D. C., B. S. Graham, J. T. Zhou, J. Y. Zhou, R. Bucco, L. A. Cavacini, M. R. Posner, and The NIH AIDS Vaccine Clinical Trials Network V3-specific neutralizing antibodies in sera from HIV-1 gp160- immunized volunteers block virus fusion and act synergistically with human monoclonal antibody to the conformation-dependent CD4 binding region of gp120. J. Clin. Investig. 92: Montefiori, D. C., G. Pantaleo, L. M. Fink, J. T. Zhou, J. Y. Zhou, M. Bilska, G. D. Miralles, and A. S. Fauci Neutralizing and infection-enhancing antibody responses to human immunodeficiency virus type 1 in long-term nonprogressors. J. Infect. Dis. 173: Montefiori, D. C., W. E. Robinson, Jr., S. S. Schuffman, and W. M. Mitchell Evaluation of antiviral drugs and neutralizing antibodies to human immunodeficiency virus by a rapid and sensitive microtiter infection assay. J. Clin. Microbiol. 26: Moog, C., H. J. A. Fleury, I. Pellegrin, A. Kirn, and A. M. Aubertin Autologous and heterologous neutralizing antibody responses following initial seroconversion in human immunodeficiency virus type 1-infected individuals. J. Virol. 71: Moore, J. P., J. A. McKeating, Y. X. Huang, A. Ashkenazi, and D. D. Ho Virions of primary human immunodeficiency virus type 1 isolates resistant to soluble CD4 (scd4) neutralization differ in scd4 binding and glycoprotein gp120 retention from scd4-sensitive isolates. J. Virol. 66: Mulligan, M. J., and J. Weber Human trials of HIV-1 vaccines. AIDS 13(Suppl. A):S105 S Muster, T., F. Steindl, M. Purtscher, A. Trkola, A. Klima, G. Himmler, F. Ruker, and H. Katinger A conserved neutralization epitope on gp41 of human immunodeficiency virus type 1. J. Virol. 67: Myers, G., J. A. Berzofsky, A. B. Rabson, T. F. Smith, and F. Wong-Staal (ed.) Human retroviruses and AIDS. Theoretical Biology and Biophysics Group T-10. Los Alamos National Laboratory, Los Alamos, N.Mex. 53. O Brien, W. A., Y. Koyanagi, A. Namazie, J. Q. Zhao, A. Diagne, K. Idler, J. A. Zack, and I. S. Chen HIV-1 tropism for mononuclear phagocytes can be determined by regions of gp120 outside the CD4-binding domain. Nature 348: Ourmanov, I., M. Bilska, V. H. Hirsch, and D. C. Montefiori Recombinant modified vaccinia virus Ankara expressing the surface gp120 of simian immunodeficiency virus (SIV) primes for a rapid neutralizing antibody response to SIV infection in macaques. J. Virol. 74: Pantaleo, G., S. Menzo, M. Vaccarezza, C. Graziosi, O. J. Cohen, J. F. Demarest, W. E. Biddison, D. Montefiori, J. M. Orenstein, C. Fox, L. K. Schrager, J. B. Margolick, R. Detels, S. Buchbinder, J. V. Giorgi, C. R. Rinaldo, J. P. Phair, and A. S. Fauci Studies in subjects with long-term nonprogressive human immunodeficiency virus infection. N. Engl. J. Med. 332: Pellegrin, I., E. Legrand, D. Neau, P. Bonot, B. Masquelier, J.-L. Pellegrin, J.-M. Ragneud, N. Bernard, and H. J. A. Fleury Kinetics of appearance of neutralizing antibodies in 12 patients with primary or recent HIV-1 infection and relationship with plasma and cellular viral loads. J. Acquir. Immun. Defic. Syndr. Hum. Retrovirol. 11: Persson, R. H., S. X. Cao, G. Cates, F. L. Yao, M. H. Klein, and B. Rovinski Modifications of HIV-1 retrovirus-like particles to enhance safety and immunogenicity. Biologicals 26: Pilgrim, A. K., G. Pantaleo, O. J. Cohen, L. M. Fink, J. Y. Zhou, J. T. Zhou, D. P. Bolognesi, A. S. Fauci, and D. C. Montefiori Neutralizing antibody responses to human immunodeficiency virus type 1 in primary infection and long-term nonprogressive infection. J. Infect. Dis. 176:

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

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Feb. 1999, p. 1740 1745 Vol. 73, No. 2 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Comparison of the Antibody Repertoire Generated

More information

HIV Anti-HIV Neutralizing Antibodies

HIV Anti-HIV Neutralizing Antibodies ,**/ The Japanese Society for AIDS Research The Journal of AIDS Research : HIV HIV Anti-HIV Neutralizing Antibodies * Junji SHIBATA and Shuzo MATSUSHITA * Division of Clinical Retrovirology and Infectious

More information

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

EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV. (Summary of the recommendations from an Enterprise Working Group) AIDS Vaccine 07, Seattle, August 20-23, 2007 EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV (Summary of the recommendations from an Enterprise Working Group) The Working Group Reston, Virginia,

More information

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

GOVX-B11: A Clade B HIV Vaccine for the Developed World GeoVax Labs, Inc. 19 Lake Park Drive Suite 3 Atlanta, GA 3 (678) 384-72 GOVX-B11: A Clade B HIV Vaccine for the Developed World Executive summary: GOVX-B11 is a Clade B HIV vaccine targeted for use in

More information

Received 13 July 2000/Accepted 27 January 2001

Received 13 July 2000/Accepted 27 January 2001 JOURNAL OF VIROLOGY, May 2001, p. 4165 4175 Vol. 75, No. 9 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.9.4165 4175.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Vaccine-Elicited

More information

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

Regional Clustering of Shared Neutralization Determinants on Primary Isolates of Clade C Human Immunodeficiency Virus Type 1 from South Africa JOURNAL OF VIROLOGY, Mar. 2002, p. 2233 2244 Vol. 76, No. 5 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.5.2233 2244.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. Regional Clustering

More information

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

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center Antibody Dependent Cellular Cytotxic activity: Past and Future Guido Ferrari, M.D. Duke University Medical Center Mechanism of Antibody Dependent Cellular Cytotoxicity (ADCC) ADCC Effector Cells (NK, monocytes/macrophages,

More information

HIV and Challenges of Vaccine Development

HIV and Challenges of Vaccine Development Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health HIV and Challenges of Vaccine Development Richard A. Koup, MD INTEREST

More information

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART BIOE 301 LECTURE 10 MITALI BANERJEE A VACCINE FOR HIV HIV HAART Visit wikipedia.org and learn the mechanism of action of the five classes of antiretroviral drugs. (1) Reverse transcriptase inhibitors (RTIs)

More information

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

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study Note: I have added some clarifying comments to the slides -- please click on Comments under View to see them. Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a

More information

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood tree illustrating CRF01_AE gp120 protein sequence relationships between 107 Envs sampled in the RV144 trial

More information

Challenges in Designing HIV Env Immunogens for Developing a Vaccine

Challenges in Designing HIV Env Immunogens for Developing a Vaccine b514_chapter-13.qxd 12/4/2007 3:39 PM Page 327 Chapter 13 Challenges in Designing HIV Env Immunogens for Developing a Vaccine Indresh K. Srivastava* and R. Holland Cheng Summary HIV continues to be a major

More information

ILNOUR OURMANOV, 1 MIROSLAWA BILSKA, 2 VANESSA M. HIRSCH, 1

ILNOUR OURMANOV, 1 MIROSLAWA BILSKA, 2 VANESSA M. HIRSCH, 1 JOURNAL OF VIROLOGY, Mar. 2000, p. 2960 2965 Vol. 74, No. 6 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Recombinant Modified Vaccinia Virus Ankara Expressing

More information

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

Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses Recombinant Baculovirus Derived HIV-1 Virus-Like Particles Elicit Potent Neutralizing Antibody Responses Weimin Liu University of Alabama at Birmingham Introduction and Rationale Virus-like particles (VLPs)

More information

2005 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.

2005 LANDES BIOSCIENCE. DO NOT DISTRIBUTE. [Human Vaccines 1:2, 45-60; March/April 2005]; 2005 Landes Bioscience Review Role of Neutralizing Antibodies in Protective Immunity Against HIV Indresh K. Srivastava* Jeffrey B. Ulmer Susan W. Barnett

More information

Identification of Mutation(s) in. Associated with Neutralization Resistance. Miah Blomquist

Identification of Mutation(s) in. Associated with Neutralization Resistance. Miah Blomquist Identification of Mutation(s) in the HIV 1 gp41 Subunit Associated with Neutralization Resistance Miah Blomquist What is HIV 1? HIV-1 is an epidemic that affects over 34 million people worldwide. HIV-1

More information

Progress on new vaccine strategies against chronic viral infections

Progress on new vaccine strategies against chronic viral infections Progress on new vaccine strategies against chronic viral infections Jay A. Berzofsky,, Masaki Terabe, Igor M. Belyakov J Clin Invest. 2004;114(4):450-462. https://doi.org/10.1172/jci22674. Review Among

More information

Received 5 October 2001/Accepted 6 December 2001

Received 5 October 2001/Accepted 6 December 2001 JOURNAL OF VIROLOGY, Mar. 2002, p. 2123 2130 Vol. 76, No. 5 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.5.2123 2130.2002 Determination of a Statistically Valid Neutralization Titer in Plasma That Confers

More information

The humoral immune responses to IBV proteins.

The humoral immune responses to IBV proteins. The humoral immune responses to IBV proteins. E. Dan Heller and Rosa Meir The Hebrew University of Jerusalem, Israel COST FA1207 meeting WG2 + WG3, Budapest, Jan. 2015 1 IBV encodes four major structural

More information

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

AIDSVaccine2010 Atlanta, Georgia Willy Bogers. NIH HIVRad Grant nr 5P01AI066287 HIV-1 envelope-cd4 receptor complexes elicit broad T- and B- cell immune responses as well as cross-reactive neutralizing antibodies in Rhesus macaques NIH HIVRad Grant nr 5P01AI066287 AIDSVaccine2010

More information

Received 9 June 1997/Accepted 7 October 1997

Received 9 June 1997/Accepted 7 October 1997 JOURNAL OF VIROLOGY, Jan. 1998, p. 286 293 Vol. 72, No. 1 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology Antibody-Dependent Cellular Cytotoxicity Directed against Cells Expressing

More information

Strategies for an HIV vaccine

Strategies for an HIV vaccine Strategies for an HIV vaccine Norman L. Letvin J Clin Invest. 2002;110(1):15-27. https://doi.org/10.1172/jci15985. Perspective The development of an HIV vaccine poses an unprecedented challenge to the

More information

Are we targeting the right HIV determinants?

Are we targeting the right HIV determinants? QuickTime et un décompresseur TIFF (non compressé) sont requis pour visionner cette image. AIDS Vaccine 2009 October 22 nd 2009 - Paris Are we targeting the right HIV determinants? Françoise BARRÉ-SINOUSSI

More information

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

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES 1 of 7 I. Viral Origin. A. Retrovirus - animal lentiviruses. HIV - BASIC PROPERTIES 1. HIV is a member of the Retrovirus family and more specifically it is a member of the Lentivirus genus of this family.

More information

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

Comparison of Human Immunodeficiency Virus Antigens as Stimulants for Lymphocyte Proliferation Assays CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY, May 2002, p. 525 529 Vol. 9, No. 3 1071-412X/02/$04.00 0 DOI: 10.1128/CDLI.9.3.525 529.2002 Copyright 2002, American Society for Microbiology. All Rights

More information

Why are validated immunogenicity assays important for HIV vaccine development?

Why are validated immunogenicity assays important for HIV vaccine development? Why are validated immunogenicity assays important for HIV vaccine development? There is a need to compare immunogenicity of products in the pipeline, when similar or different in class when developed by

More information

RAISON D ETRE OF THE IMMUNE SYSTEM:

RAISON D ETRE OF THE IMMUNE SYSTEM: RAISON D ETRE OF THE IMMUNE SYSTEM: To Distinguish Self from Non-Self Thereby Protecting Us From Our Hostile Environment. Innate Immunity Acquired Immunity Innate immunity: (Antigen nonspecific) defense

More information

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

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) BACKGROUND Human Immunodeficiency Virus ( HIV ) can be divided into two major types, HIV type 1 (HIV-1) and HIV type 2 (HIV-2). HIV-1 is related to

More information

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

Biomedical Engineering for Global Health. Lecture 10 HIV/AIDS vaccine development Biomedical Engineering for Global Health Lecture 10 HIV/AIDS vaccine development Review of lecture 9 How do vaccines work? Types ofvaccines: Review of lecture 9 Are vaccines effective? -Edward Jenner s

More information

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

JOURNAL OF VIROLOGY, Oct. 1999, p Vol. 73, No. 10. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Oct. 1999, p. 8201 8215 Vol. 73, No. 10 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Role of Immune Responses against the Envelope

More information

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

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells Tumor Immunotherapy Autologous virus Inactivation Inactivated virus Lymphopheresis Culture? Monocyte s Dendritic cells Immunization Autologous vaccine Development of Tumor Vaccines Types of Tumor Vaccines

More information

MICHAEL W. CHO, 1 * MYUNG K. LEE, 1 CHIN H. CHEN, 2 TOM MATTHEWS, 3

MICHAEL W. CHO, 1 * MYUNG K. LEE, 1 CHIN H. CHEN, 2 TOM MATTHEWS, 3 JOURNAL OF VIROLOGY, Oct. 2000, p. 9749 9754 Vol. 74, No. 20 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Identification of gp120 Regions Targeted by a

More information

How HIV Causes Disease Prof. Bruce D. Walker

How HIV Causes Disease Prof. Bruce D. Walker How HIV Causes Disease Howard Hughes Medical Institute Massachusetts General Hospital Harvard Medical School 1 The global AIDS crisis 60 million infections 20 million deaths 2 3 The screen versions of

More information

HVTN P5 Vaccine Trials

HVTN P5 Vaccine Trials HVTN P5 Vaccine Trials Erica Andersen-Nissen, PhD Director, Cape Town HVTN Immunology Laboratory Considerations for a Pan-African HIV Vaccine Development Agenda Kigali, Rwanda 16-17 March 2015 HVTN Mission

More information

New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS. Dublin, Ireland. June 24, AIDS Vaccines.

New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS. Dublin, Ireland. June 24, AIDS Vaccines. New Preventive Technology: Providing New Options to Stop the Spread of HIV/AIDS Dublin, Ireland June 24, 2004 AIDS Vaccines - An R&D Briefing This paper has been prepared by the International AIDS Vaccine

More information

Received 17 April 2003/Accepted 28 June 2003

Received 17 April 2003/Accepted 28 June 2003 JOURNAL OF VIROLOGY, Oct. 2003, p. 10348 10356 Vol. 77, No. 19 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.19.10348 10356.2003 Cellular Immunity Elicited by Human Immunodeficiency Virus Type 1/ Simian Immunodeficiency

More information

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma Excerpt from MACS&more Vol 8 1/2004 Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma L. Davis Lupo and Salvatore T. Butera HIV and Retrovirology Branch,

More information

RAISON D ETRE OF THE IMMUNE SYSTEM:

RAISON D ETRE OF THE IMMUNE SYSTEM: RAISON D ETRE OF THE IMMUNE SYSTEM: To Distinguish Self from Non-Self Thereby Protecting Us From Our Hostile Environment. Innate Immunity Adaptive Immunity Innate immunity: (Antigen - nonspecific) defense

More information

HIV Vaccine. 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง

HIV Vaccine. 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง HIV Vaccine 15 September 2016 นพ.นคร เปรมศร ผ อ านวยการส าน กงาน โครงการศ กษาว คซ นเอดส ทดลอง GLOBAL STATISTICS 2015 17 million people were accessing antiretroviral therapy 36.7 million people globally

More information

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

Development of Broadly Reactive HIV-1/AIDS Virus-like Particle Vaccines. Sean Patrick McBurney. B.S. Microbiology, University of Pittsburgh, 2004 Development of Broadly Reactive HIV-1/AIDS Virus-like Particle Vaccines by Sean Patrick McBurney B.S. Microbiology, University of Pittsburgh, 2004 Submitted to the Graduate Faculty of School of Medicine

More information

Received 26 September 2003/Accepted 22 December 2003

Received 26 September 2003/Accepted 22 December 2003 JOURNAL OF VIROLOGY, Apr. 2004, p. 3930 3940 Vol. 78, No. 8 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.8.3930 3940.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Highly Effective

More information

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

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED Viv Peut Kent Laboratory, University of Melbourne, Australia WHY ENVELOPE? Env subject to both humoral and cellular immune responses Perhaps

More information

In Vitro Antigen Challenge of Human Antibody Libraries for Vaccine Evaluation: the Human Immunodeficiency Virus Type 1 Envelope

In Vitro Antigen Challenge of Human Antibody Libraries for Vaccine Evaluation: the Human Immunodeficiency Virus Type 1 Envelope JOURNAL OF VIROLOGY, Dec. 1996, p. 9046 9050 Vol. 70, No. 12 0022-538X/96/$04.00 0 Copyright 1996, American Society for Microbiology In Vitro Antigen Challenge of Human Antibody Libraries for Vaccine Evaluation:

More information

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people Micro 301 HIV/AIDS Shiu-Lok Hu hus@uw.edu December 3, 2012 Since its discovery 31 years ago Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people In 2011 34.0 million [31.4 35.9 million]

More information

Novel Vaccine Products for Planned Phase I Immunogenicity Studies in Infants

Novel Vaccine Products for Planned Phase I Immunogenicity Studies in Infants Office of AIDS Research Novel Vaccine Products for Planned Phase I Immunogenicity Studies in Infants L. Jean Patterson, PhD Office of AIDS Research, NIH February 7, 2017 Office of AIDS Research OAR Responsibilities

More information

Human Immunodeficiency Virus type 1 (HIV-1) gp120 / Glycoprotein 120 ELISA Pair Set

Human Immunodeficiency Virus type 1 (HIV-1) gp120 / Glycoprotein 120 ELISA Pair Set Human Immunodeficiency Virus type 1 (HIV-1) gp120 / Glycoprotein 120 ELISA Pair Set Catalog Number : SEK11233 To achieve the best assay results, this manual must be read carefully before using this product

More information

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Catalog Number : SEK11695 To achieve the best assay results, this manual must be read carefully before using this product

More information

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

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies? Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Department of Health and Human Services Should There be Further Efficacy

More information

Dissecting the Neutralizing Antibody Specificities of Broadly Neutralizing Sera from Human Immunodeficiency Virus Type 1-Infected Donors

Dissecting the Neutralizing Antibody Specificities of Broadly Neutralizing Sera from Human Immunodeficiency Virus Type 1-Infected Donors JOURNAL OF VIROLOGY, June 2007, p. 6548 6562 Vol. 81, No. 12 0022-538X/07/$08.00 0 doi:10.1128/jvi.02749-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Dissecting the Neutralizing

More information

Updated information and services can be found at:

Updated information and services can be found at: REFERENCES CONTENT ALERTS Neutralizing Antibodies Elicited by Immunization of Monkeys with DNA Plasmids and Recombinant Adenoviral Vectors Expressing Human Immunodeficiency Virus Type 1 Proteins John R.

More information

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

J. D. Trujillo,* N. M. Kumpula-McWhirter, K. J. Hötzel, M. Gonzalez, and W. P. Cheevers JOURNAL OF VIROLOGY, Sept. 2004, p. 9190 9202 Vol. 78, No. 17 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.17.9190 9202.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Glycosylation

More information

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections Amy Chung Dr. Ivan Stratov Prof. Stephen Kent ADCC process consists of Target cell QuickTime and a TIFF (Uncompressed) FcγR decompressor

More information

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

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity Innate & adaptive Immunity Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity Helen Horton PhD Seattle Biomedical Research Institute Depts of Global Health & Medicine, UW Cellular

More information

SIV p27 ANTIGEN CAPTURE ASSAY

SIV p27 ANTIGEN CAPTURE ASSAY SIV p27 ANTIGEN CAPTURE ASSAY Enzyme Immunoassay for the detection of Simian Immunodeficiency Virus (SIV) p27 in tissue culture media Catalog #5436 and #5450 Version 6; 12/2012 ABL PRODUCTS AND SERVICES

More information

Glycosylation of the ENV Spike of Primate Immunodeficiency Viruses and Antibody Neutralization

Glycosylation of the ENV Spike of Primate Immunodeficiency Viruses and Antibody Neutralization Current HIV Research, 2004, 2, 243-254 243 Glycosylation of the ENV Spike of Primate Immunodeficiency Viruses and Antibody Neutralization Cheryl A. Pikora *1,2 1 Department of Infectious Diseases, Children

More information

University of Massachusetts Medical School Michael Vaine University of Massachusetts Medical School

University of Massachusetts Medical School Michael Vaine University of Massachusetts Medical School University of Massachusetts Medical School escholarship@umms GSBS Student Publications Graduate School of Biomedical Sciences 8-23-2008 Improved induction of antibodies against key neutralizing epitopes

More information

human Total Cathepsin B Catalog Number: DY2176

human Total Cathepsin B Catalog Number: DY2176 human Total Cathepsin B Catalog Number: DY2176 This DuoSet ELISA Development kit contains the basic components required for the development of sandwich ELISAs to measure natural and recombinant human Total

More information

Received 9 August 2004/Accepted 26 October 2004

Received 9 August 2004/Accepted 26 October 2004 JOURNAL OF VIROLOGY, Mar. 2005, p. 3358 3369 Vol. 79, No. 6 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.6.3358 3369.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Protection

More information

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

DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04) DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04) Rashmi Jalah Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, National Cancer Institute

More information

MedChem 401~ Retroviridae. Retroviridae

MedChem 401~ Retroviridae. Retroviridae MedChem 401~ Retroviridae Retroviruses plus-sense RNA genome (!8-10 kb) protein capsid lipid envelop envelope glycoproteins reverse transcriptase enzyme integrase enzyme protease enzyme Retroviridae The

More information

Review Article. AIDS vaccine: Present status and future challenges

Review Article. AIDS vaccine: Present status and future challenges Review Article AIDS vaccine: Present status and future challenges P. K. Nigam, Manjula Kerketta Department of Dermatology and Venereology, Pt. JNM Medical College and associated Dr. BRAM Hospital, Raipur,

More information

ACTG Laboratory Technologist Committee Revised Version 2.0 ACTG Lab Man HIV Syncytium-Inducing (MT-2) assay 29 April 2004

ACTG Laboratory Technologist Committee Revised Version 2.0 ACTG Lab Man HIV Syncytium-Inducing (MT-2) assay 29 April 2004 HIV SYNCYTIUM-INDUCING (MT-2) ASSAY 1. BACKGROUND and CLINICAL SIGNIFICANCE Host and viral factors may play a role in determining the way in which an individual responds to anti-retroviral therapy. Presence

More information

Under the Radar Screen: How Bugs Trick Our Immune Defenses

Under the Radar Screen: How Bugs Trick Our Immune Defenses Under the Radar Screen: How Bugs Trick Our Immune Defenses Session 7: Cytokines Marie-Eve Paquet and Gijsbert Grotenbreg Whitehead Institute for Biomedical Research HHV-8 Discovered in the 1980 s at the

More information

Human Immunodeficiency Virus

Human Immunodeficiency Virus Human Immunodeficiency Virus Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Viruses and hosts Lentivirus from Latin lentis (slow), for slow progression of disease

More information

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

Development of a Universal T Cell Vaccine. Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom Development of a Universal T Cell Vaccine Tomáš Hanke Weatherall Institute of Molecular Medicine University of Oxford United Kingdom Development of HIV-1 vaccines Induction of cell-mediated responses Immunogens

More information

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

The author hereby certifies that the use of any copyrighted material in the dissertation entitled: The author hereby certifies that the use of any copyrighted material in the dissertation entitled: Characterization and Enhanced Processing of Soluble, Oligomeric gp140 Envelope Glycoproteins Derived from

More information

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors VIROLOGY Engineering Viral Genomes: Retrovirus Vectors Viral vectors Retrovirus replicative cycle Most mammalian retroviruses use trna PRO, trna Lys3, trna Lys1,2 The partially unfolded trna is annealed

More information

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

It has been 25 years since HIV-1 was identified as the causative Vol 4j2 October 8jdoi:.38/nature732 Challenges in the development of an HIV-1 vaccine Dan H. Barouch 1 The development of a safe and effective human immunodeficiency virus (HIV)-1 vaccine is a critically

More information

Received 29 August 2002/Accepted 3 December 2002

Received 29 August 2002/Accepted 3 December 2002 JOURNAL OF VIROLOGY, Mar. 2003, p. 3099 3118 Vol. 77, No. 5 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.5.3099 3118.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Simian-Human

More information

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

Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239 Immunization with Single-Cycle SIV Significantly Reduces Viral Loads After an Intravenous Challenge with SIV mac 239 Bin Jia 1, Sharon K. Ng 1, M. Quinn DeGottardi 1, Michael Piatak Jr. 2, Eloísa Yuste

More information

Introduction. Abstract

Introduction. Abstract Virology 337 (2005) 136 148 www.elsevier.com/locate/yviro Neutralization sensitivity of HIV-1 Env-pseudotyped virus clones is determined by co-operativity between mutations which modulate the CD4-binding

More information

A PROJECT ON HIV INTRODUCED BY. Abdul Wahab Ali Gabeen Mahmoud Kamal Singer

A PROJECT ON HIV INTRODUCED BY. Abdul Wahab Ali Gabeen Mahmoud Kamal Singer A PROJECT ON HIV INTRODUCED BY Abdul Wahab Ali Gabeen Mahmoud Kamal Singer Introduction: Three groups of nations have been identified in which the epidemiology of HIV(Human Immunodeficiency Virus) varies:

More information

HIV Immunopathogenesis. Modeling the Immune System May 2, 2007

HIV Immunopathogenesis. Modeling the Immune System May 2, 2007 HIV Immunopathogenesis Modeling the Immune System May 2, 2007 Question 1 : Explain how HIV infects the host Zafer Iscan Yuanjian Wang Zufferey Abhishek Garg How does HIV infect the host? HIV infection

More information

Lecture 11. Immunology and disease: parasite antigenic diversity

Lecture 11. Immunology and disease: parasite antigenic diversity Lecture 11 Immunology and disease: parasite antigenic diversity RNAi interference video and tutorial (you are responsible for this material, so check it out.) http://www.pbs.org/wgbh/nova/sciencenow/3210/02.html

More information

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

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 1. NIH Public Access Author Manuscript Published in final edited form as: Nature. 2009 January 1; 457(7225): 87 91. doi:10.1038/nature07469. Immune Control of an SIV Challenge by a T Cell-Based Vaccine in

More information

Received 29 May 2001/Accepted 3 October 2001

Received 29 May 2001/Accepted 3 October 2001 JOURNAL OF VIROLOGY, Jan. 2002, p. 292 302 Vol. 76, No. 1 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.1.292 302.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. ALVAC-SIV-gag-pol-env-Based

More information

A Quarterly Update on HIV Prevention Research. Vol. 8 No. 2

A Quarterly Update on HIV Prevention Research. Vol. 8 No. 2 What is it? What could it do? Key Facts Antibodies Passive immunization is the transfer of pre-made antibodies to a person. Passive immunization using today's pre-made antibodies can involve infusion delivered

More information

Gene Vaccine Dr. Sina Soleimani

Gene Vaccine Dr. Sina Soleimani Gene Vaccine Dr. Sina Soleimani Human Viral Vaccines Quality Control Laboratory (HVVQC) Titles 1. A short Introduction of Vaccine History 2. First Lineage of Vaccines 3. Second Lineage of Vaccines 3. New

More information

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

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures HIV-1 evolution in response to immune selection pressures BISC 441 guest lecture Zabrina Brumme, Ph.D. Assistant Professor, Faculty of Health Sciences Simon Fraser University http://www3.niaid.nih.gov/topics/hivaids/understanding/biology/structure.htm

More information

Boosts Following Priming with gp120 DNA

Boosts Following Priming with gp120 DNA Neutralizing Antibody Responses Induced with V3-scaffold Protein Boosts Following Priming with gp120 DNA Susan Zolla-Pazner NYU School of Medicine Problems with Whole Env Immunogens Poor induction of Abs

More information

HIV-1 p24 Antigen ELISA 2.0 Catalog Number:

HIV-1 p24 Antigen ELISA 2.0 Catalog Number: INTENDED USE The RETRO-TEK HIV-1 p24 Antigen ELISA 2.0 is an enzyme linked immunoassay used to detect Human Immunodeficiency Virus Type 1 (HIV-1) p24 antigen in cell culture media. It can be used to monitor

More information

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

Viral vaccines. Lec. 3 أ.د.فائزة عبد هللا مخلص Lec. 3 أ.د.فائزة عبد هللا مخلص Viral vaccines 0bjectives 1-Define active immunity. 2-Describe the methods used for the preparation of attenuated live & killed virus vaccines. 3- Comparison of Characteristics

More information

A Cell Line-Based Neutralization Assay for Primary Human Immunodeficiency Virus Type 1 Isolates That Use either the CCR5 or the CXCR4 Coreceptor

A Cell Line-Based Neutralization Assay for Primary Human Immunodeficiency Virus Type 1 Isolates That Use either the CCR5 or the CXCR4 Coreceptor JOURNAL OF VIROLOGY, Nov. 1999, p. 8966 8974 Vol. 73, No. 11 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. A Cell Line-Based Neutralization Assay for Primary

More information

HIV INFECTION: An Overview

HIV INFECTION: An Overview HIV INFECTION: An Overview UNIVERSITY OF PAPUA NEW GUINEA SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL MBBS II SEMINAR VJ

More information

Isolation, Propagation, and Titration of Human Immunodeficiency Virus Type 1 From Peripheral Blood of Infected Individuals

Isolation, Propagation, and Titration of Human Immunodeficiency Virus Type 1 From Peripheral Blood of Infected Individuals Isolation of HIV-1 From PBMC of Infected Individuals 17 2 Isolation, Propagation, and Titration of Human Immunodeficiency Virus Type 1 From Peripheral Blood of Infected Individuals Hanneke Schuitemaker

More information

Received 24 October 2004/Accepted 11 January 2005

Received 24 October 2004/Accepted 11 January 2005 JOURNAL OF VIROLOGY, June 2005, p. 6957 6968 Vol. 79, No. 11 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.11.6957 6968.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Cryptic Nature

More information

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.

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. Title Studies of SARS virus vaccines Author(s) 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. 39-43 Issued

More information

Received 19 March 2003/Accepted 28 May 2003

Received 19 March 2003/Accepted 28 May 2003 JOURNAL OF VIROLOGY, Aug. 2003, p. 8729 8735 Vol. 77, No. 16 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.16.8729 8735.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Plasmid

More information

Supplementary Material

Supplementary Material Supplementary Material Nuclear import of purified HIV-1 Integrase. Integrase remains associated to the RTC throughout the infection process until provirus integration occurs and is therefore one likely

More information

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

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1. NIH Public Access Author Manuscript Published in final edited form as: Nat Med. 2010 March ; 16(3): 319 323. doi:10.1038/nm.2089. Mosaic HIV-1 Vaccines Expand the Breadth and Depth of Cellular Immune Responses

More information

We are IntechOpen, the first native scientific publisher of Open Access books. International authors and editors. Our authors are among the TOP 1%

We are IntechOpen, the first native scientific publisher of Open Access books. International authors and editors. Our authors are among the TOP 1% We are IntechOpen, the first native scientific publisher of Open Access books 3,350 108,000 1.7 M Open access books available International authors and editors Downloads Our authors are among the 151 Countries

More information

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

Immunization with single-cycle SIV significantly reduces viral loads after an intravenous challenge with SIV(mac)239 University of Massachusetts Medical School escholarship@umms Preventive and Behavioral Medicine Publications and Presentations Preventive and Behavioral Medicine 1-23-2009 Immunization with single-cycle

More information

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

A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys A centralized gene-based HIV-1 vaccine elicits broad cross-clade cellular immune responses in rhesus monkeys Sampa Santra, Bette T. Korber, Mark Muldoon, Dan H. Barouch, Gary J. Nabel, Feng Gao, Beatrice

More information

MID-TERM EXAMINATION

MID-TERM EXAMINATION Epidemiology 227 May 2, 2007 MID-TERM EXAMINATION Select the best answer for the multiple choice questions. There are 75 questions and 11 pages on the examination. Each question will count one point. Notify

More information

Received 8 October 1997/Accepted 5 January 1998

Received 8 October 1997/Accepted 5 January 1998 JOURNAL OF VIROLOGY, Apr. 1998, p. 3248 3258 Vol. 72, No. 4 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology In Vivo Replication Capacity Rather Than In Vitro Macrophage Tropism

More information

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

Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89. Replicating measles-shiv vaccine induces long term preservation of central memory CD4 cells in the gut of macaques challenged with SHIV89.6P Frédéric Tangy Viral Genomics and Vaccination Laboratory Measles

More information

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

Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP 1 Learning Objectives Recognize hazards associated with viral vectors in research and animal

More information

Title: Neutralization resistance of HIV-1 virological synapse-mediated infection is. Running Title: Virological-synapse neutralization resistance

Title: Neutralization resistance of HIV-1 virological synapse-mediated infection is. Running Title: Virological-synapse neutralization resistance JVI Accepts, published online ahead of print on 2 May 2012 J. Virol. doi:10.1128/jvi.00230-12 Copyright 2012, American Society for Microbiology. All Rights Reserved. 1 2 Title: Neutralization resistance

More information

Influenza A H1N1 HA ELISA Pair Set

Influenza A H1N1 HA ELISA Pair Set Influenza A H1N1 HA ELISA Pair Set for H1N1 ( A/Puerto Rico/8/1934 ) HA Catalog Number : SEK11684 To achieve the best assay results, this manual must be read carefully before using this product and the

More information

DNA Vaccines against Human Immunodeficiency Virus Type 1 in the Past Decade

DNA Vaccines against Human Immunodeficiency Virus Type 1 in the Past Decade CLINICAL MICROBIOLOGY REVIEWS, Apr. 2004, p. 370 389 Vol. 17, No. 2 0893-8512/04/$08.00 0 DOI: 10.1128/CMR.17.2.370 389.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. DNA

More information