on January 7, 2019 by guest

Size: px
Start display at page:

Download "on January 7, 2019 by guest"

Transcription

1 JOURNAL OF VIROLOGY, Nov. 2000, p Vol. 74, No X/00/$ Copyright 2000, American Society for Microbiology. All Rights Reserved. Pathogenic Simian/Human Immunodeficiency Virus SHIV KU Inoculated into Immunized Macaques Caused Infection, but Virus Burdens Progressively Declined with Time PETER S. SILVERSTEIN, 1 GLENN A. MACKAY, 1 SAMPA MUKHERJEE, 1 ZHUANG LI, 1 MICHAEL PIATAK, JR., 2 JEFFREY D. LIFSON, 2 OPENDRA NARAYAN, 1 AND ANIL KUMAR 1 * Marion Merrell Dow Laboratory of Viral Pathogenesis, Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas 66160, 1 and AIDS Vaccine Program, SAIC Frederick, National Cancer Institute Frederick Cancer Research and Development Center, Frederick, Maryland Received 20 June 2000/Accepted 23 August 2000 Using the simian immunodeficiency virus/human immunodeficiency virus (SHIV)-macaque model of AIDS, we had shown in a previous report that a live, nonpathogenic strain of SHIV, further attenuated by deletion of the vpu gene and inoculated orally into adult macaques, had effectively prevented AIDS following vaginal inoculation with pathogenic SHIV KU. Examination of lymph nodes from the animals at 18 weeks postchallenge had shown that all six animals were persistently infected with challenge virus. We report here on a 2-year follow-up study on the nature of the persistent infections in these animals. DNA of the vaccine virus was present in the lymph nodes at all time points tested, as far as 135 weeks postchallenge. In contrast, the DNA of SHIV KU became undetectable in one animal by week 55 and in three others by week 63. These four macaques have remained negative for SHIV KU DNA as far as the last time point examined at week 135. Quantification of the total viral DNA concentration in lymph nodes during the observation period showed a steady decline. All animals developed neutralizing antibody and cytotoxic-t-lymphocyte responses to SHIV KU that persisted throughout the observation period. Vaccine-like viruses were isolated from two animals, and a SHIV KU -like virus was isolated from one of the two macaques that remained positive for SHIV KU DNA. There was no evidence of recombination between the vaccine and the challenge viruses. Thus, immunization with the live vaccine not only prevented disease but also contributed to the steady decline in the virus burdens in the animals. Downloaded from * Corresponding author. Mailing address: Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS Phone: (913) Fax: (913) akumar@kumc.edu. Despite the pressing need for a vaccine against human immunodeficiency virus (HIV), evaluation of the efficacy of any vaccine candidate in human beings will require several years of observation. The reasons for this are that the incubation period and the clinical course of HIV-induced disease are both unpredictably variable and that the immunological correlates of protection against HIV infection and/or disease are not known (16). Evaluation of the concept that a vaccine against HIV could be developed despite the formidable biological properties of this virus has been greatly expedited using macaque models of HIV pathogenesis (5, 6, 8, 9, 22, 23). Use of the simian immunodeficiency virus (SIV) model has pioneered this field and demonstrated that deletion of accessory genes from genomes of pathogenic viruses greatly attenuated the virulence of the latter agents and that use of such viruses as live vaccines induced protection against disease caused by pathogenic strains of SIV (10 13, 31). The SIV/HIV (SHIV)-macaque model was created on the rationale that this virus is biologically closer to HIV than to SIV because it has the envelope of HIV type 1 (HIV-1) (17). Use of the SHIV-macaque system to model vaccine efficacy required the availability of a predictably pathogenic strain of SHIV that could serve as challenge. SHIV KU satisfied this condition because it not only causes both the loss of CD4 T cells and AIDS but it is also pathogenic after inoculation on the vaginal mucosal surface, thus providing a model of sexually transmitted HIV. Following leads on vaccine development in the SIV system, we deleted the accessory gene vpu from a strain of nonpathogenic SHIV and used this virus as a vaccine to test the concept that an orally inoculated vaccine could induce protection against AIDS caused by vaginally inoculated pathogenic SHIV KU (10). In a previous report we had shown that orally inoculated vaccine virus, vpushiv PPc (V-II), caused a transiently productive subclinical systemic infection in all of the macaques and that that this resulted in infection and establishment of vaccine virus DNA in the lymph nodes of the animals. Intravaginal challenge of the six immunized and four nonimmunized animals with SHIV KU resulted in infection in all 10 animals with this virus. However, whereas the nonimmunized animals rapidly developed productive infection followed by the onset of AIDS, the infection with SHIV KU in the six immunized animals was maintained at a minimally productive level that had no pathological effects. Nevertheless, DNA of challenge virus persisted in the lymph nodes of all six animals when examined at 18 weeks postchallenge (10). In this study we report on the nature of the persistent infection caused by the two viruses. All six immunized animals have remained healthy, and all developed and maintained strong antibody and cytotoxic-t-lymphocyte (CTL) responses against both viruses. DNA of the vaccine virus has persisted in lymph nodes of all six animals throughout the observation period. However, whereas the DNA of the challenge virus was also present in all six animals at 18 weeks postchallenge, concentrations of this DNA in the animals progressively decreased with time, and by 63 weeks postchallenge it had become un- on January 7, 2019 by guest 10489

2 10490 SILVERSTEIN ET AL. J. VIROL. detectable in four of the six animals. This suggested that the immunization had not only prevented AIDS but also contributed to the gradual decline in the level of challenge virus. MATERIALS AND METHODS Amplification of vpu/gp120 sequences for detection of viral sequences in DNA isolated from PBMC and lymph node. DNA was isolated from lymph node tissue or peripheral blood mononuclear cells (PBMC) as previously described (10). The vpu/gp120 regions of viral genomes were amplified by nested PCR that produces fragments diagnostic for either vaccine virus DNA or challenge virus DNA as previously described (10). After the second round of amplification, challenge virus DNA, which has an intact vpu gene, yields a fragment of 431 bp, while vaccine virus DNA yields a fragment of 371 bp. Amplification of each sample was repeated four times in separate reactions to ensure accuracy. This assay is capable of detecting one viral genome in 10 3 cells. Detection of viral sequences using a nested PCR assay specific for challenge virus DNA was accomplished as described above. However, for the second round of PCR the antisense oligonucleotide primer used was 5 -CACAAAATAGAGTGGTGGTTGCTTCCT-3. This primer is complementary to nucleotides 6361 to 6387 of the HIV (HxB2) genome. This region corresponds to the vpu gene and is present in the challenge virus SHIV KU but is absent from the vaccine virus ( vpushiv PPc ). Thus, when this oligonucleotide is used in the assay described above, challenge virus DNA yields a fragment of 197 bp after the second round of amplification, while vaccine virus DNA does not yield any detectable fragment. This assay is capable of detecting one viral genome in 10 4 cells. Virus recovery from lymph nodes. Lymph node cells were obtained from mesenteric lymph node tissue, and CD4 cells were negatively selected using immunomagnetic beads as previously described (9). The CD4 cells were counted and stimulated with phytohemagglutinin (1 g/ml) for 48 h, and cells were cocultured with 10 6 C8166 indicator cells. After cocultivation for 7 days, the cultures were examined for cytopathic effect (CPE). Cells and supernatant from cultures exhibiting CPE were pooled and used to inoculate fresh C8166 cells. DNA from infected cultures was then isolated using a QIAAMP kit (Qiagen, Valencia, Calif.) according to the directions provided by the manufacturer. Amplification of gp120 and nef sequences for sequencing. PCR amplifications of gp120 and nef sequences were performed as previously described (27). Three separate amplifications were performed for each isolate. PCR products from each amplification reaction were purified on an agarose gel and cloned in PGEM-T (Promega, Madison, Wis.). Plasmids containing gp120 and nef were sequenced using the Bigdye Terminator Cycle Sequencing Ready Reaction Kit with AmpliTaq DNA polymerase, FS (PE Applied Biosystems, Foster City, Calif.). Reactions were run utilizing an Applied Biosystems 377 Prism XL automated DNA sequencer. Sequences from the 5 and 3 ends were determined with T7 and SP6 primers. Internal sequences were determined using primers 7265 (5 -CTCCCTGGTCCTCTCTGG-3 ) and 7181 (5 -TAAAACCATAATAGTAC AGC-3 ). Challenge virus DNA was distinguished from vaccine virus DNA by the deletion in vpu. A minimum of one clone from each of the three separate amplifications of DNA from each viral isolate was sequenced. Consensus sequences are based on a minimum of three independently derived clones. Lymphocyte proliferation assay. PBMC from different macaques were cultured in triplicate in the presence or absence of UV-irradiated and heat-inactivated SHIV KU for 4 days (14). Cells were then pulsed with 1 Ci of [ 3 H]thymidine for 18 h before being subjected to scintillation spectroscopy. Stimulation indices were calculated as mean counts per minute (cpm), i.e., the cpm in stimulated wells/mean cpm in control wells. Generation of bulk CTL population and chromium release assay. CTL activity was determined as described elsewhere (14, 15, 28). Briefly, CTL effectors were prepared by stimulating PBMC from different macaques with UV-irradiated autologous CD4 T cells infected with SHIV KU. Targets were autologous CD4 T cells infected with SHIV KU or autologous B cells infected with recombinant vaccinia virus expressing HIV env, SIV gag,orsiv pol and then labeled with 51 Cr. Effector/target ratios ranging from 100:1 to 10:1 were used in a 4-h chromium release assay. The percent specific cytoxicity was calculated as follows: [(test release spontaneous release)/(maximum release spontaneous release)] 100. Quantitative PCR of viral DNA. Quantification of viral DNA in tissue samples was performed using a modification of a procedure described previously (25). Serum neutralization assay. Neutralizing antibody titer in plasma was determined as previously described (10). Briefly, twofold serial dilutions of heatinactivated plasma were prepared in RPMI 1640 containing 10% fetal bovine serum, 5 mm NaHCO 3 and 50 g of gentamicin (R-10) per ml in quadruplicate in a flat-bottom 96-well plate. Suspensions of eight 50% tissue culture infective doses of either VII or SHIV KU virus in R-10 were added to each well, and the plates were incubated at 37 C for 1 h, followed by the addition of 10 4 C8166 cells to each well. After 1 week of incubation at 37 C, each well was scored for CPE, and the 50% neutralization titer was calculated by the Karber method (10). Nucleotide sequence accession numbers. The sequence data reported here have been assigned GenBank accession no. AY (V PEy -gp120), AY (V PEy -Nef), AY (SHIV KU/8124 -gp120), AY (SHIV KU/8124 -Nef), AY (V PWl -gp120), and AY (V PWl -Nef). FIG. 1. PCR detection of challenge virus DNA in the lymph nodes of animals inoculated with VII and challenged with SHIV KU. At week 18 postchallenge, lymph node biopsies were performed, and DNA was isolated and then amplified using a nested-pcr assay specific for DNA from SHIV KU. Aliquots of the PCR were electrophoresed on a 2% agarose gel, and DNA was visualized by ethidium bromide staining. Lanes: 1, amplification of DNA isolated from lymph node of the control animal 4267; 2, amplification of V-DNA; 3 to 8, amplification of DNA isolated from the lymph nodes of animals PEy, 42106, 7024, PWl, 8124, and PWv, respectively; 9, 1-kb molecular size marker. The arrow on the left marked C indicates the positions of the fragments amplified from C-DNA. RESULTS As reported previously (10), the vaccine virus, V-II ( vpushiv PPc ), had replicated productively in all six animals tested after oral inoculation. This productive phase of infection lasted 10 to 12 weeks, after which virus replication became restricted, as indicated by a lack of detectable infection in PBMC after this period. The animals were challenged vaginally with SHIV KU 7 months after immunization. Infectious PBMC were sporadically detected in four of the animals during the first-20-week postchallenge period but, beyond this time point, virus was not isolated again for the duration of the observation period of 135 weeks. Viral RNA in plasma has been below the limit of detection throughout the observation period (results not shown). Analysis of DNA from lymph node biopsies at 18 weeks postchallenge showed a full-length vpu gene in all six animals, indicating that all animals had become infected with challenge virus. Further, a nested PCR assay, using an oligonucleotide primer whose sequence is complementary to a region of vpu found only in challenge virus DNA, confirmed that all animals were infected with SHIV KU (Fig. 1). Using PCR primers that distinguish between the full-length and truncated vpu genes of challenge and vaccine viruses, respectively, we examined PBMC or lymph nodes of the animals at four intervals between weeks 55 and 103 postchallenge to determine the identity of the virus DNA present at these periods. Two animals, 8124 and 42106, showed the presence of DNA from both vaccine and challenge viruses throughout the monitoring period (Table 1 and data not shown). The DNA PCR data from the other four animals are shown in Fig. 2 and are summarized in Table 1. The data showed that the DNAs of both viruses were present in three of the four animals at week 55. The fourth animal, PWv, showed the presence of only vaccine virus DNA (V-DNA) at this time point, even though challenge virus DNA (C-DNA) had been detected at week 18 (10). Examination of lymph nodes biopsied at week 63 showed that challenge virus DNA had become undetectable in three additional animals, PEy, PWl, and Examination of lymph nodes biopsied at week 103 showed results identical to those seen at week 63, confirming that challenge virus DNA had become undetectable in four of the six animals. Quantitative analysis of viral DNA concentrations in lymph nodes using real-time PCR demonstrated that total viral DNA concentra-

3 VOL. 74, 2000 VACCINE-MEDIATED DECLINE OF PATHOGENIC SHIV TABLE 1. Detection of V-DNA and C-DNA and virus recovery in macaques immunized with vaccine II and challenged with SHIV KU Macaque PBMC (wk 55) Lymph node (wk 63) Viral DNA in a : PBMC (wk 91) Lymph node (wk 103) Identity of recovered virus b PEy C/V V V V V PWl C/V V V V V 7024 C/V V V V NR PWv V V V V NR C/V C/V C/V C/V NR 8124 C/V C/V C/V C/V C a C/V, V-DNA and C-DNA; V, V-DNA only; C, C-DNA only. b Virus recovery was performed at week 63. NR, no virus could be recovered at week 63. tions declined in all six animals between weeks 63 and 135 (Table 2). Virus isolation attempts were performed on lymph node cells after depletion of CD8 T cells and activation of the remainder with mitogen, followed by cocultivation with indicator cell cultures. Using this method, at week 63, we isolated vaccine-like virus from animals PWl and PEy and challenge-like virus from animal Infectious virus could not be isolated from lymph node tissue of animal 7024, PWv, or These data are summarized in Table 1. Sequence analysis of the vpu/gp120 region of the viruses recovered from animals PWl and PEy (V PWl and V PEy, respectively) demonstrated that they have the vpu deletion and gp120 sequences characteristic of vaccine virus. Examination of Fig. 3 shows that there are 10 and 7 amino acid differences in V PWl - gp120 and V PEy -gp120, respectively, compared to the corresponding sequence of the vaccine virus that was used for inoculation. In V PWl -gp120 there was also a deletion of five amino acids which had been caused by the elimination of one copy of a directly repeated nucleotide sequence. Four of these amino acid replacements are identical in V PEy -gp120 and V PWl -gp120 (M278T, R3041, G470P, and D474N). The amino acid changes at amino acids 278, 304, and 470 resulted in the replacement of an amino acid characteristic of vaccine virus with an amino acid characteristic of challenge virus. Interspersed among these changes were amino acids characteristic of vaccine virus (i.e., at amino acids 65, 175, 412, and 476). Thus, it is much more likely that the amino acid changes were caused by a constellation of amino acid replacements rather than by recombination events between challenge virus and vaccine virus. Analysis of the nef gene of the viruses showed five and six amino acid substitutions, respectively, in V PWl, and V PEy (Fig. 4). V PWl -Nef has a K105E substitution, while V PEy -Nef had a K105Q substitution. Amino acid 105 is within the SH3 domain that has been previously identified in Nef. The other changes seen in Nef were animal specific. DNA sequence analysis revealed that the virus isolated from macaque 8124, SHIV KU-8124, has an intact vpu gene, along with a gp120 region that is closely related to SHIV KU (Fig. 3), demonstrating that this virus was derived from the challenge virus. This virus has three amino acid differences compared to the sequence from the challenge virus. Two of these changes are in V1, a region that is subject to frequent amino acid changes in challenge-derived viruses (P. Silverstein, unpublished observations). The third change, a T278M replacement, caused the loss of a potential glycosylation site. SHIV KU Nef has four amino acid changes compared to the corresponding sequence in SHIV KU (Fig. 4). FIG. 2. PCR analysis of viral sequences present in PBMC and lymph nodes of animals inoculated with VII and challenged with SHIV KU. Lymph node biopsies and PBMC isolations were performed, DNA isolated, and then amplified using nested PCR. Aliquots of the PCR were electrophoresed on a 2% agarose gel, and DNA was visualized by ethidium bromide staining. (A) PCR analysis of DNA isolated from PBMC 55 weeks after challenge. Lanes: 1, 1-kb molecular size marker; 2, 100-bp molecular size marker; 3, challenge viruspositive control; 4, vaccine virus-positive control; 5 to 8, amplification of DNA from animals PEy, PWl, 7024, and PWv, respectively; 9, DNA control; 10, 100-bp molecular size marker. Arrows on the left marked V or C indicate the positions of DNA fragments amplified from vaccine virus or challenge virus, respectively. (B) PCR analysis of DNA isolated from lymph node tissue 63 weeks after challenge. Lanes are in the same order as in panel A. (C) PCR analysis of DNA isolated from PBMC 91 weeks after challenge. Lanes are in the same order as in panel A. (D) PCR analysis of DNA isolated from lymph node 103 weeks after challenge. Lanes are in the same order as in panel A. Sequentially collected plasma samples from vaccinated and control animals were tested for the presence of neutralizing antibodies against vaccine and challenge viruses. The results of these experiments are shown in Fig. 5. None of the nonimmunized animals challenged with SHIV KU developed detectable neutralizing antibodies to the virus (results not shown). However, three vaccinated animals developed high titers of neutralizing antibodies against vaccine virus that peaked after challenge (reciprocal neutralizing antibody titers of 160 to 1,280). The other three animals developed high titers of neutralizing antibodies that peaked the week of challenge (reciprocal neutralizing antibody titers of 160 to 640). These anti- Time postchallenge (wk) TABLE 2. Viral DNA loads (SHIV DNA copies/10 5 cell equivalents) in vaccinated macaques SHIV DNA copies/10 5 cell Eq in macaque: PEy PWl 7024 PWv ,600 20,000 5,800 12, ,900 2,600 5,100 1, , a 1, a This animal died at 117 weeks postchallenge of causes unrelated to SHIV infection.

4 10492 SILVERSTEIN ET AL. J. VIROL. FIG. 3. Alignment of the consensus amino acid sequences of gp120 from the recovered viruses, the challenge virus, and the vaccine virus. The predicted protein sequences of gp120 from the viruses recovered from macaques PEy, PWl, and 8124 are aligned with the gp120 sequences of the vaccine, V-II, and challenge, SHIV KU, viruses. The underlined letters above the alignment indicate whether that amino acid of the recovered virus from PEy or PWl is characteristic of the vaccine or challenge virus. The underlined letters below the alignment indicate whether that amino acid of the virus recovered from animal 8124 is characteristic of vaccine or challenge virus. x denotes the deletion of an amino acid. FIG. 4. Alignment of the consensus amino acid sequences of Nef from SHIV KU, VII, and the viruses recovered from animals PEy, PWl, and The predicted protein sequences of Nef from SHIV KU and the viruses recovered from macaques PEy and PWl are aligned with the Nef sequence of the vaccine virus, V-II. bodies persisted throughout the 104-week postchallenge observation period. Prechallenge serum from two macaques, PWl and PWv, neutralized SHIV KU (antibody titers of 1/20 and 1/10, respectively). However, antibodies from four of six vaccinates did not neutralize pathogenic virus SHIV KU, indicating that at least some of the neutralization epitopes on vaccine virus were different from those on the challenge virus. After challenge with pathogenic virus, all six vaccinates developed SHIV KU -specific neutralizing antibodies that persisted throughout the observation period. T-cell proliferative responses in immunized animals were also assessed at different time points starting at 47 weeks after challenge (Table 3). All vaccinated macaques showed persistent T-helper responses to both the vaccine and the challenge viruses. Macaque 8124, from which challenge-like virus was isolated at week 63, exhibited the best T-helper response. Animals from which vaccine-like viruses were isolated (PEy and PWl) had T-helper responses similar to those seen in animals from which virus could not be isolated. To determine whether inhibition of SHIV KU replication in animals correlated with the presence of cell-mediated immune responses, we examined PBMC from each animal for the presence of cytotoxic T lymphocytes against SHIV KU. Tests performed several times between weeks 55 and 103 have shown the same basic pattern illustrated in Table 4. These data showed that all of the animals had developed challenge virusspecific CTLs. This activity was directed against the Gag, Pol, and Env proteins. All of the control animals exhibited high virus burdens (viral RNA, 10 5 copies/ml), massive loss of CD4 T cells, and a lack of detectable neutralizing antibodies against SHIV KU. Three of the four control animals died within 6 months of

5 VOL. 74, 2000 VACCINE-MEDIATED DECLINE OF PATHOGENIC SHIV Downloaded from FIG. 5. Titers of neutralizing antibodies to vaccine and challenge viruses in immunized animals. Sequential samples of sera were collected and tested for the presence of neutralizing antibody to VII (Œ) and SHIV KU ( ). challenge, but a fourth animal survived for 83 weeks. The longest-surviving control animal, PDb, developed virus-specific CTLs against Gag, Pol, and Env (Fig. 6). By week 59 the CTLs against Gag and Pol disappeared, and by week 75 the CTLs to Env also became undetectable. This animal never developed a significant level of T-helper response to either vaccine or challenge viruses and ultimately succumbed to disease at week 82. The lack of T-helper response at these three time points may have been due to the fact that this animal had already experienced a precipitous decline in CD4 T cells (10). This sequence of pathogenesis events is very similar to that in human beings succumbing to infection with HIV-1. We determined whether the changes in the genomes of the vaccine-like viruses isolated from animals PWl and PEy and the SHIV KU -like virus isolated from animal 8124 had any effect on their susceptibility to neutralization. Virus neutralization tests using plasma collected at the late time points showed that, whereas the antibodies in the plasma of animals PWl and PEy neutralized the vaccine virus at high dilutions, the same antibodies were only minimally effective against the new viruses (Fig. 7A and B). In contrast, the SHIV KU variant isolated from animal 8124, which had undergone a much smaller number of changes, was neutralized at approximately the same dilutions of plasma that neutralized SHIV KU (Fig. 7C). Similarly, we wished to determine whether genetic changes in the two vaccine virus variants affected the ability of CTLs to TABLE 3. Proliferative T-cell responses to vaccine and challenge viral antigens in macaques at various times following challenge Macaque Antigen a Stimulation index at: Wk 47 Wk 59 Wk 72 Wk 79 Wk 91 Wk vpushiv PPc ND b ND SHIV KU vpushiv PPc ND ND SHIV KU vpushiv PPc ND ND SHIV KU PEy vpushiv PPc ND ND SHIV KU PWl vpushiv PPc ND ND SHIV KU PWv vpushiv PPc ND ND SHIV KU PDb vpushiv PPc ND ND ND ND SHIV KU ND ND a A total of 20 l of UV-irradiated and heat-inactivated virus was added to each well and used as a source of antigen. b ND, not determined. on January 7, 2019 by guest

6 10494 SILVERSTEIN ET AL. J. VIROL. TABLE 4. Virus-specific CTL activities in macaques immunized with V-II and challenged with SHIV KU Macaque % Specific lysis against a : SHIV KU HIV env SIV gag SIV pol PEy PWl PWV a Macaques inoculated with V-II were tested for CTL activity at week 96 postchallenge. The specific lysis values shown here are at an effector/target ratio of 40:1. SHIV KU -specific lysis was determined by infecting concanavalin A-stimulated CD8 depleted PBMC with SHIV KU and using these cells as targets in a 4-h chromium release assay. The Env, Gag, and Pol specific lysis values were determined by infecting herpesvirus papio-transformed B-LCL with recombinant vaccinia virus expressing one of these proteins and then using these cells as targets in a 4-h chromium release assay. lyse cells infected with these new agents. The results demonstrated that cells infected with V PEy or V PWl were lysed at a significantly lower efficiency than cells infected with the original vaccine (Fig. 8). DISCUSSION The studies reported here extend findings in our first report that oral immunization of six macaques with the live virus vaccine V-II ( vpushiv PPc ) resulted in protection from productive replication and disease caused by vaginally inoculated SHIV KU (10). All six animals became infected with the challenge virus, but all vaccinates have controlled replication of the virus. This protection has lasted more than 2 years. The animals have remained healthy and are free of infectious cells in the peripheral blood. They do not have detectable viral RNA in plasma and have not sustained any loss of CD4 T cells. This control over SHIV KU is associated with persistent neutralizing antibodies and cell-mediated immune responses to this virus. The present report focuses on the viruses that have persisted in these vaccinates. DNA of the vaccine virus (V-DNA) has persisted in all six animals throughout the observation period of 135 weeks, and infectious vaccine virus was isolated from lymph node of two of these animals at week 63. DNA of SHIV KU was also present in the lymph nodes of the six animals in the first few months following challenge but, during the subsequent period, this DNA became undetectable in four of the six animals, and challenge virus could not be isolated from lymph node cells. Of the remaining two animals in which SHIV KU DNA persisted, infectious SHIV KU has been isolated from the lymph node of only one. These results suggest that the vaccine had not only contributed to the suppression of replication of SHIV KU but also to the gradual reduction in total viral load. The validity of the claim for the reduction in challenge virus DNA burden is supported by the fact that a single set of primers was used to amplify both species of DNA. Because the primers bound to identical sites in both DNA species and the sequences of the amplified fragments were similar, the amplification of one species served as an internal control for the amplification of the other species. Thus, the detection of V-DNA and challenge virus DNA (C-DNA) in PBMC from week 55 and the detection of only V-DNA at later time points reflected a decline in the DNA concentration of the challenge virus relative to the DNA concentration of vaccine virus. This relative change could have been brought about by either a decline in the level of C-DNA or an increase in the level of V-DNA. If the change in the relative levels of C-DNA and V-DNA was due to a decline in the level of C-DNA, then a decline in total DNA load in these tissues would have been expected. However, if the change in the relative levels of C- DNA and V-DNA was due to an increase in V-DNA, then an increase in total viral DNA load would have been expected. In order to distinguish between these two possibilities, total viral DNA load in the animals was quantitated using a real-time PCR assay. Between weeks 63 and 135, the viral DNA load in three of the four animals, i.e., PEy, PWl, and PWv, declined dramatically. The fourth animal, 7024, died of causes unrelated to SHIV infection at week 117. However, the viral DNA load in this animal had also declined between weeks 63 and 103. Thus, the change in the ratio of C-DNA to V-DNA was due to a decrease in the concentration of the DNA of the challenge virus. These data introduce the possibility that a vaccine could contribute to the reduction of viral DNA load, as well as protection from disease. In our previous report (10), only C-DNA was detected in FIG. 6. CTL activity in a nonimmunized macaque, PDb, that survived for 83 weeks after challenge with SHIV KU. PBMC collected from PDb at weeks 45, 59, or 75 postchallenge were stimulated in vitro with UV-irradiated autologous CD4 T cells infected with SHIV KU. The growing PBMC were used as effectors in a 4-h chromium release assay and were tested against mock-infected T cells, B-LCLs infected with wild-type vaccinia virus, and recombinant vaccinia virus expressing HIV Env, SIV Gag, and SIV Pol. The CTL activity shown here is the specific lysis obtained at an effector/target ratio of 40:1.

7 VOL. 74, 2000 VACCINE-MEDIATED DECLINE OF PATHOGENIC SHIV FIG. 7. Comparative analysis of neutralizing antibodies (Neut. Ab.) against challenge and reemergent viruses in macaques immunized with VII and challenged with SHIV KU. Three of the six macaques yielded replication-competent virus at week 63. Two of the recovered viruses were derived from vaccine virus (animals PEy and PWl), while one animal yielded virus derived from SHIV KU (animal 8124). Sequentially collected plasma samples from these three macaques were tested for their ability to neutralize both the parental and the recovered viruses. Titers of neutralizing antibodies in serum samples collected from animals PWl (A), PEy (B), and 8124 (C) are shown. three vaccinates at 18 weeks after challenge, but V-DNA was readily detected at later time points. This could have been due to the reactivation of latent reservoirs of vaccine virus by infection with the challenge virus. The reactivation then resulted in increased levels of V-DNA. Another possible explanation for this is that the lack of detection of V-DNA was due to a transient increase in the level of C-DNA associated with the initial burst of replication of challenge virus. Because the PCR assay used to detect V-DNA and C-DNA is similar to competitive PCR, an increase in the level of C-DNA would outcompete V-DNA in the PCR, allowing the detection of only C- DNA. As the replication of challenge virus was brought under control and the level of C-DNA started to decline, V-DNA once again became readily detectable. Nucleotide sequence analysis of the env and nef genes of the persistent viruses obtained from the animals in this vaccine group has shown changes unique to each agent. Viruses recovered from PEy and PWl have an M-T change at amino acid 278 of gp120, resulting in the gain of a glycosylation site in these viruses. It is well established that glycosylation of gp120 plays an important role in immune recognition of the virus (20, 24). Aside from the gain of a glycosylation site at amino acid 278, gp120 of PWl had a deletion of five amino acids between amino acids 396 and 400 that eliminated a glycosylation site. These amino acid replacements and deletions might have been capable of causing major conformational changes in the gp120 molecule and may have contributed to the altered neutralization pattern of these agents. Although there are reports of viral recombination in animals dually infected with two strains of SIV mac (3, 30), we found no evidence of recombination between vaccine and challenge viruses in the vaccinated animals. Viral gp120 sequences from the vaccine-like viruses recovered from PEy and PWl have clusters of amino acid replacements that could have arisen by recombination between vaccine and challenge sequences. However, multiple recombination events would have to be invoked to explain the interspersion of vaccine-like and challenge-like sequences. Such a high level of recombination within a region less than 2 kb in length is extremely unlikely. In the vaccine-like viruses that were recovered from these animals, there are two amino acid changes in Nef that PEy and PWl have in common. One of these amino acid replacements is the K-Q/E change at amino acid 105 that is in an SH3 domain. These domains have been shown to regulate proteinprotein interactions. Thus, this amino acid change may cause alterations in protein-protein interactions that are either quantitatively or qualitatively different than those seen with the parental vaccine virus. This study provided a good opportunity to examine the role of neutralizing antibodies in protection against challenge virus. Four of the animals did not have neutralizing antibodies to SHIV KU at the time of challenge. These antibodies appeared after challenge. In the two animals that did have SHIV KU neutralizing antibodies, the titers were low. These increased after challenge. Therefore, neutralizing antibodies appeared to play a minimal role in controlling the initial burst of SHIV KU replication. It is also of interest that all animals displayed significantly different neutralization titers to SHIV KU and VII, despite the fact that the gp120 sequences of these viruses are closely related. It is possible that one or more amino acid substitutions directly altered a major neutralization epitope(s) resulting in the change in neutralization titer. One of the differences between the gp120 sequences of SHIV KU and VII is the presence of a potential glycosylation site at amino acid 278. As discussed above, glycosylation has been shown to play an important role in the conformation of this protein and thus may be responsible for the difference in neutralization titer. In contrast to neutralizing antibodies, CTLs may have played a major role in the control of replication of SHIV KU. Although we do not have prechallenge CTL data on these animals, other animals immunized in an identical manner with this vaccine developed CTLs against proteins of this challenge virus within 6 months of immunization. These CTLs have persisted for more than 1 year (A. Kumar et al., unpublished observations). In addition, the longest-surviving control animal in this study was the only macaque to produce cell-mediated immune responses against the challenge virus. Taken together, these two lines of evidence suggest that CTLs have played a major role in suppressing virus replication and perhaps in the

8 10496 SILVERSTEIN ET AL. J. VIROL. FIG. 8. Comparative CTL activities of two animals against parental and recovered virus. PBMC collected at week 130 postchallenge were stimulated with UV-irradiated autologous CD4 T cells infected with either vaccine or reemergent virus. Two weeks after cocultivation, growing PBMCs were used as effectors in a 4-h chromium release assay and then tested at effector/target ratios of 40:1, 20:1, and 10:1. The targets included mock-infected, vaccine-infected, and CD4 T cells infected with reemergent virus. progressive elimination of infected cells. Our data are in agreement with other data that suggest a role for CTLs in the control of HIV replication in humans (2, 18, 19) and SIV replication in macaques (12, 13, 31). Further, the recent demonstration that depletion of CD8 T cells in SIV-infected macaques resulted in loss of CTLs and in higher viral burdens provided strong support for the importance of the role of CTLs in suppression of virus replication (7, 26). In the present study, all six vaccinated macaques had virusspecific CTLs that were present almost 2 years after challenge with pathogenic virus SHIV KU. In all six animals, CTLs were directed against the envelope as well as the core proteins of the virus. CTL activity was mediated by classical CD8 major histocompatibility complex-restricted CTLs. Although we did not find any distinctive bias regarding the induction of CTLs to individual proteins, Gag-specific CTL activity was higher than that induced to Env or Pol. We also did not find any difference in the immune responses between the animals from which virus was recovered from lymph node (i.e., animals PEy, PWl, and 8124) and those animals from which virus was not recovered (i.e., animals PWv, 7024, and 42106). This study demonstrated that the immunized animals have maintained high levels of humoral and cell-mediated immune responses despite the lack of detectable replication in PBMC and a lack of detectable viral RNA in plasma. However, these immune responses correlated with the persistence of viral DNA derived from vaccine virus. This DNA could represent a reservoir of latent virus that is capable of sporadic reactivation. Reactivation from this viral reservoir could provide the persistent source of antigen that is thought to be necessary for the maintenance of high levels of immune responses. In view of the fact that virus-specific CTLs and CD4 T-helper cells, as well as neutralizing antibodies, were present in the face of the progressive decline in levels of challenge virus, it is likely that both arms of the immune response are responsible for the progressive loss of this virus. This study also demonstrated that, although both V-DNA and the immune responses to vaccine virus were persistent, there must have been a substantial decline in the level of V- DNA. In animals PEy and PWl, this decrease occurred despite the reduction in effectiveness of the immune response to the newly isolated variants compared to the response to the parental vaccine viruses. This raises the possibility that mechanisms other than the cellular and humoral immune responses may have been responsible for the maintenance of low viral loads of the vaccine virus. This alternate mechanism of virus control might include soluble factors that have been shown to be inhibitory to virus replication (1, 4, 21, 29). Alternatively, although the effectiveness of the immune responses to the new variants is reduced, they may be sufficiently potent to inhibit the replication of a virus that is already attenuated. ACKNOWLEDGMENTS We thank Fenglan Jia for excellent technical assistance. We also thank Norman Letvin, Harvard Medical School, Boston, Mass., for providing herpesvirus papio stocks; D. Panicali and G. Mazzara, Therion Biologics, Cambridge, Mass., for recombinant vaccinia viruses; and the University of Kansas Medical Center Biotechnology Support Facility for assistance in DNA sequencing. This work was supported by National Institutes of Health grants AI-29382, RR-13152, RR-06753, and NS and NCI contract number NO1-CO REFERENCES 1. Garzino-Demo, A., R. B. Moss, J. B. Margolick, F. Cleghorn, A. Sill, W. A. Blattner, F. Cocchi, D. J. Carlo, A. L. DeVico, and R. C. Gallo Spontaneous and antigen-induced production of HIV-inhibitory beta-chemokines are associated with AIDS-free status. Proc. Natl. Acad. Sci. USA 96: Goulder, P. J., S. Rowland-Jones, A. McMichael, and B. D. Walker Anti-HIV cellular immunity: recent advances towards vaccine design. AIDS 13:S121 S Gundlach, B. R., M. G. Lewis, S. Sopper, T. Schnell, J. Sodroski, C. Stahl- Hennig, and K. Uberla Evidence for recombination of live, attenuated immunodeficiency virus vaccine with challenge virus to a more virulent strain. J. Virol. 74: Heeney, J. L., V. J. Teeuwsen, M. van Gils, W. M. Bogers, M. De Giuli, A. Radaelli, S. Barnett, B. Morein, L. Akerblom, Y. Wang, T. Lehner, and D. Davis Beta-chemokines and neutralizing antibody titers correlate with sterilizing immunity generated in HIV-1-vaccinated macaques. Proc. Natl. Acad. Sci. USA 95: Hirsch, V. M., G. Dapolito, P. R. Johnson, W. R. Elkins, W. T. London, R. J. Montali, S. Goldstein, and C. Brown Induction of AIDS by simian immunodeficiency virus from an African green monkey: species-specific variation in pathogenicity correlates with the extent of in vivo replication. J. Virol. 69: Igarashi, T., Y. Endo, G. Englund, R. Sadjadpour, T. Matano, C. Buckler, A. Buckler-White, R. Plishka, T. Theodore, R. Shibata, and M. Martin Emergence of a highly pathogenic simian/human immunodeficiency virus in a rhesus macaque treated with anti-cd8 mab during a primary infection with a nonpathogenic virus. Proc. Natl. Acad. Sci. USA 96: 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: Joag, S. V., I. Adany, Z. Li, L. Foresman, D. M. Pinson, C. Wang, E. B. Stephens, R. Raghavan, and O. Narayan Animal model of mucosally transmitted human immunodeficiency virus type 1 disease: intravaginal and oral deposition of simian/human immunodeficiency virus in macaques results in systemic infection, elimination of CD4 T cells, and AIDS. J. Virol. 71: Joag, S. V., Z. Li, L. Foresman, E. B. Stephens, L. J. Zhao, I. Adany, D. M. Pinson, H. M. McClure, and O. Narayan Chimeric simian/human immunodeficiency virus that causes progressive loss of CD4 T cells and AIDS in pig-tailed macaques. J. Virol. 70: Joag, S. V., Z. Q. Liu, E. B. Stephens, M. S. Smith, A. Kumar, Z. Li, C. Wang,

9 VOL. 74, 2000 VACCINE-MEDIATED DECLINE OF PATHOGENIC SHIV D. Sheffer, F. Jia, L. Foresman, I. Adany, J. Lifson, H. M. McClure, and O. Narayan Oral immunization of macaques with attenuated vaccine virus induces protection against vaginally transmitted AIDS. J. Virol. 72: Johnson, R. P., and R. C. Desrosiers Protective immunity induced by live attenuated simian immunodeficiency virus. Curr. Opin. Immunol. 10: Johnson, R. P., R. L. Glickman, J. Q. Yang, A. Kaur, J. T. Dion, M. J. Mulligan, and R. C. Desrosiers Induction of vigorous cytotoxic T- lymphocyte responses by live attenuated simian immunodeficiency virus. J. Virol. 71: Johnson, R. P., J. D. Lifson, S. C. Czajak, K. S. Cole, K. H. Manson, R. Glickman, J. Yang, D. C. Montefiori, R. Montelaro, M. S. Wyand, and R. C. Desrosiers Highly attenuated vaccine strains of simian immunodeficiency virus protect against vaginal challenge: inverse relationship of degree of protection with level of attenuation. J. Virol. 73: Kumar, A., J. Lifson, P. S. Silverstein, F. Jia, D. Sheffer, Z. Li, and O. Narayan Evaluation of immune responses induced by HIV-1 gp120 in rhesus macaques: effect of vaccination on challenge with homologous and heterologous strains of simian human immunodeficiency viruses. Virology 274: Kumar, A., H. Stipp, D. Sheffer, and O. Narayan Use of herpesvirus saimiri-immortalized macaque CD4 T cell clones as stimulators and targets of CTL responses in macaque/aids models. J. Immunol. Methods 230: Letvin, N. L Progress in the development of an HIV-1 vaccine. Science 280: Li, J., C. I. Lord, W. Haseltine, N. L. Letvin, and J. Sodroski Infection of cynomolgus monkeys with a chimeric HIV-1/SIVmac virus that expresses the HIV-1 envelope glycoproteins. J. Acquir. Immune Defic. Syndr. 5: McMichael, A. J., and C. A. O Callaghan A new look at T cells. J. Exp. Med. 187: Ogg, G. S., X. Jin, S. Bonhoeffer, P. R. Dunbar, M. A. Nowak, S. Monard, J. P. Segal, Y. Cao, S. L. Rowland-Jones, V. Cerundolo, A. Hurley, M. Markowitz, D. D. Ho, D. F. Nixon, and A. J. McMichael Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 279: Overbaugh, J., and L. M. Rudensey Alterations in potential sites for glycosylation predominate during evolution of the simian immunodeficiency virus envelope gene in macaques. J. Virol. 66: Pal, R., A. Garzino-Demo, P. D. Markham, J. Burns, M. Brown, R. C. Gallo, and A. L. DeVico Inhibition of HIV-1 infection by the beta-chemokine MDC. Science 278: Reimann, K. A., J. T. Li, R. Veazey, M. Halloran, I. W. Park, G. B. Karlsson, J. Sodroski, and N. L. Letvin A chimeric simian/human immunodeficiency virus expressing a primary patient human immunodeficiency virus type 1 isolate env causes an AIDS-like disease after in vivo passage in rhesus monkeys. J. Virol. 70: Reimann, K. A., J. T. Li, G. Voss, C. Lekutis, K. Tenner-Racz, P. Racz, W. Lin, D. C. Montefiori, D. E. Lee-Parritz, Y. Lu, R. G. Collman, J. Sodroski, and N. L. Letvin An env gene derived from a primary human immunodeficiency virus type 1 isolate confers high in vivo replicative capacity to a chimeric simian/human immunodeficiency virus in rhesus monkeys. J. Virol. 70: Reitter, J. N., R. E. Means, and R. C. Desrosiers A role for carbohydrates in immune evasion in AIDS. Nat. Med. 4: Rossio, J. L., M. T. Esser, K. Suryanarayana, D. K. Schneider, J. W. J. Bess, G. M. Vasquez, T. A. Wiltrout, E. Chertova, M. K. Grimes, Q. Sattentau, L. O. Arthur, L. E. Henderson, and J. D. Lifson Inactivation of human immunodeficiency virus type 1 infectivity with preservation of conformational and functional integrity of virion surface proteins. J. Virol. 72: Schmitz, J. E., M. J. Kuroda, S. Santra, V. G. Sasseville, M. A. Simon, M. A. Lifton, P. Racz, K. Tenner-Racz, M. Dalesandro, B. J. Scallon, J. Ghrayeb, M. A. Forman, D. C. Montefiori, E. P. Rieber, N. L. Letvin, and K. A. Reimann Control of viremia in simian immunodeficiency virus infection by CD8 lymphocytes. Science 283: Stephens, E. B., S. Mukherjee, M. Sahni, W. Zhuge, R. Raghavan, D. K. Singh, K. Leung, B. Atkinson, Z. Li, S. V. Joag, Z. Q. Liu, and O. Narayan A cell-free stock of simian-human immunodeficiency virus that causes AIDS in pig-tailed macaques has a limited number of amino acid substitutions in both SIVmac and HIV-1 regions of the genome and has offered cytotropism. Virology 231: Stipp, H., A. Kumar, and O. Narayan Characterization of immune escape viruses from a macaque immunized with live-virus vaccine and challenged with SHIVKu. AIDS Res. Hum. Retrovir. 16: Wang, Y., L. Tao, E. Mitchell, W. M. Bogers, C. Doyle, C. A. Bravery, L. A. Bergmeier, C. G. Kelly, J. L. Heeney, and T. Lehner Generation of CD8 suppressor factor and beta chemokines, induced by xenogeneic immunization, in the prevention of simian immunodeficiency virus infection in macaques. Proc. Natl. Acad. Sci. USA 95: Wooley, D. P., R. A. Smith, S. Czajak, and R. C. Desrosiers Direct demonstration of retroviral recombination in a rhesus monkey. J. Virol. 71: Wyand, M. S., K. Manson, D. C. Montefiori, J. D. Lifson, R. P. Johnson, and R. C. Desrosiers Protection by live, attenuated simian immunodeficiency virus against heterologous challenge. J. Virol. 73:

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

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

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

Virology. Longitudinal study to assess the safety and efficacy of a live-attenuated SHIV vaccine in long term immunized rhesus macaques

Virology. Longitudinal study to assess the safety and efficacy of a live-attenuated SHIV vaccine in long term immunized rhesus macaques Virology 383 (2009) 103 111 Contents lists available at ScienceDirect Virology journal homepage: www.elsevier.com/locate/yviro Longitudinal study to assess the safety and efficacy of a live-attenuated

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

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

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

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

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

Supporting Information

Supporting Information Supporting Information Walker et al. 1.173/pnas.111753118 - JR-CSF 1 3 1 4 1 5 1 6 1 7 1 8 blank beads protein A beads JR-FL - 1 3 1 4 1 5 1 6 1 7 1 8 - MGRM-C26 1 3 1 4 1 5 1 6 1 7 1 8 reciprocal serum

More information

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute Understanding HIV Transmission Utilizing Transmitted/Founder Viruses Brandon Keele SAIC-Frederick National Cancer Institute AIDS Vaccine 2011 15 September 2011 Overview Several years ago, the CHAVI sought

More information

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

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Apr. 2001, p. 3753 3765 Vol. 75, No. 8 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.8.3753 3765.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Route of Simian

More information

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

Immune Responses and Viral Replication in Long-Term Inapparent Carrier Ponies Inoculated with Equine Infectious Anemia Virus JOURNAL OF VIROLOGY, July 2000, p. 5968 5981 Vol. 74, No. 13 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Immune Responses and Viral Replication in Long-Term

More information

Original Article. Kwofie TB, Miura T 1. Abstract. Introduction

Original Article. Kwofie TB, Miura T 1. Abstract. Introduction Original Article Increased Virus Replication and Cytotoxicity of Non pathogenic Simian Human Immuno Deficiency Viruses NM 3rN After Serial Passage in a Monkey Derived Cell Line Kwofie TB, Miura T 1 Departments

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

JOURNAL OF VIROLOGY, Mar. 1999, p Vol. 73, No. 3. Copyright 1999, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Mar. 1999, p Vol. 73, No. 3. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Mar. 1999, p. 1853 1859 Vol. 73, No. 3 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Acute Effects of Pathogenic Simian-Human Immunodeficiency

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

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

GENETIC STUDY OF VPU VARIANTS AMONG THE HIV-1 INFECTED INTRAVENOUS DRUG USERS OF MANIPUR PREDICTS HIGHLY PATHOGENIC VIRUS

GENETIC STUDY OF VPU VARIANTS AMONG THE HIV-1 INFECTED INTRAVENOUS DRUG USERS OF MANIPUR PREDICTS HIGHLY PATHOGENIC VIRUS GENETIC STUDY OF VPU VARIANTS AMONG THE HIV-1 INFECTED INTRAVENOUS DRUG USERS OF MANIPUR PREDICTS HIGHLY PATHOGENIC VIRUS Adhikarimayum Lakhikumar Sharma 1, Thiyam Ramsing Singh 1, Takhellambam Chanu Machathoibi

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

Supporting Information

Supporting Information Supporting Information Sui et al..7/pnas.997 Pre-CLP CM9 LA9 SL Tat# Pol Vif % Tetramer + CD + CD + Vac+IL- +IL- Vac Fig. S. Frequencies of six different CD + CD + Mamu-A*-tetramer + cells were measured

More information

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

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques SUPPORTING INFORMATION FOR: Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques Lia Vassena, 1,2 Huiyi Miao, 1 Raffaello Cimbro,

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

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

JOURNAL OF VIROLOGY, Dec. 2000, p Vol. 74, No. 24. Copyright 2000, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Dec. 2000, p Vol. 74, No. 24. Copyright 2000, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Dec. 2000, p. 11935 11949 Vol. 74, No. 24 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. Protection of Macaca nemestrina from Disease

More information

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

Equine infectious anaemia virus proteins with epitopes most frequently recognized by cytotoxic T lymphocytes from infected horses Journal of General Virology (2000), 81, 2735 2739. Printed in Great Britain... SHORT COMMUNICATION Equine infectious anaemia virus proteins with epitopes most frequently recognized by cytotoxic T lymphocytes

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

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

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T cells, the RNA genomes with all modifications are generated

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

Received 4 December 2001/Accepted 29 April 2002

Received 4 December 2001/Accepted 29 April 2002 JOURNAL OF VIROLOGY, Aug. 2002, p. 8433 8445 Vol. 76, No. 16 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.16.8433 8445.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. The Relationship

More information

Because HIV-1 establishes infections only in humans and

Because HIV-1 establishes infections only in humans and Emergence of a highly pathogenic simian human immunodeficiency virus in a rhesus macaque treated with anti-cd8 mab during a primary infection with a nonpathogenic virus Tatsuhiko Igarashi*, Yasuyuki Endo*,

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

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

Updated information and services can be found at:

Updated information and services can be found at: REFERENCES CONTENT ALERTS Temporal Analyses of Virus Replication, Immune Responses, and Efficacy in Rhesus Macaques Immunized with a Live, Attenuated Simian Immunodeficiency Virus Vaccine Ruth I. Connor,

More information

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

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Medical Virology Immunology Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Human blood cells Phases of immune responses Microbe Naïve

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

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

Anti-SIV Cytolytic Molecules in Pigtail Macaques

Anti-SIV Cytolytic Molecules in Pigtail Macaques AIDS RESEARCH AND HUMAN RETROVIRUSES Volume 24, Number 8, 2008 Mary Ann Liebert, Inc. DOI: 10.1089/aid.2008.0081 Anti-SIV Cytolytic Molecules in Pigtail Macaques Erik Rollman, Stephen J. Turner, Katherine

More information

Removal of N-Linked Glycosylation Sites in the V1 Region of Simian Immunodeficiency Virus gp120 Results in Redirection of B-Cell Responses to V3

Removal of N-Linked Glycosylation Sites in the V1 Region of Simian Immunodeficiency Virus gp120 Results in Redirection of B-Cell Responses to V3 JOURNAL OF VIROLOGY, Feb. 2004, p. 1525 1539 Vol. 78, No. 3 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.3.1525 1539.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Removal of

More information

The emergence and characterization of macrophage-tropic SIV/HIV chimeric viruses (SHIVs) present in CD4 T cell-depleted rhesus monkeys

The emergence and characterization of macrophage-tropic SIV/HIV chimeric viruses (SHIVs) present in CD4 T cell-depleted rhesus monkeys The emergence and characterization of macrophage-tropic SIV/HIV chimeric viruses (SHIVs) present in CD4 T cell-depleted rhesus monkeys Tatsuhiko Igarashi,* Hiromi Imamichi, Charles R. Brown,* Vanessa M.

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

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

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers Adam R. Hersperger Department of Microbiology University of Pennsylvania Evidence for

More information

Manish Sagar, 1,2 Xueling Wu, 2 Sandra Lee, 3 and Julie Overbaugh 2 *

Manish Sagar, 1,2 Xueling Wu, 2 Sandra Lee, 3 and Julie Overbaugh 2 * JOURNAL OF VIROLOGY, Oct. 2006, p. 9586 9598 Vol. 80, No. 19 0022-538X/06/$08.00 0 doi:10.1128/jvi.00141-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Human Immunodeficiency

More information

hemagglutinin and the neuraminidase genes (RNA/recombinant viruses/polyacrylamide gel electrophoresis/genetics)

hemagglutinin and the neuraminidase genes (RNA/recombinant viruses/polyacrylamide gel electrophoresis/genetics) Proc. Natl. Acad. Sci. USA Vol. 73, No. 6, pp. 242-246, June 976 Microbiology Mapping of the influenza virus genome: Identification of the hemagglutinin and the neuraminidase genes (RNA/recombinant viruses/polyacrylamide

More information

Immunoprophylaxis against AIDS in macaques with a lentiviral DNA vaccine

Immunoprophylaxis against AIDS in macaques with a lentiviral DNA vaccine Virology 351 (2006) 444 454 www.elsevier.com/locate/yviro Immunoprophylaxis against AIDS in macaques with a lentiviral DNA vaccine ZhenQian Liu a, Dinesh K. Singh a,1, Darlene Sheffer a, Marilyn S. Smith

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

HIV 101: Fundamentals of HIV Infection

HIV 101: Fundamentals of HIV Infection HIV 101: Fundamentals of HIV Infection David H. Spach, MD Professor of Medicine University of Washington Seattle, Washington Learning Objectives After attending this presentation, learners will be able

More information

Vaccination with Attenuated Simian Immunodeficiency Virus by DNA Inoculation

Vaccination with Attenuated Simian Immunodeficiency Virus by DNA Inoculation JOURNAL OF VIROLOGY, Dec. 2001, p. 11930 11934 Vol. 75, No. 23 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.23.11930 11934.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Vaccination

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

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

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

A Noninfectious Simian/Human Immunodeficiency Virus DNA Vaccine That Protects Macaques against AIDS

A Noninfectious Simian/Human Immunodeficiency Virus DNA Vaccine That Protects Macaques against AIDS JOURNAL OF VIROLOGY, Mar. 2005, p. 3419 3428 Vol. 79, No. 6 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.6.3419 3428.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. A Noninfectious

More information

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR Supplemental Materials and Methods Plasmids and viruses To generate pseudotyped viruses, the previously described recombinant plasmids pnl4-3-δnef-gfp or pnl4-3-δ6-drgfp and a vector expressing HIV-1 X4

More information

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

Emergence and Kinetics of Simian Immunodeficiency Virus-Specific CD8 T Cells in the Intestines of Macaques during Primary Infection JOURNAL OF VIROLOGY, Nov. 2001, p. 10515 10519 Vol. 75, No. 21 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.21.10515 10519.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Emergence

More information

Virus Genetic Diversity

Virus Genetic Diversity Virus Genetic Diversity Jin-Ching Lee, Ph.D. 李 jclee@kmu.edu.tw http://jclee.dlearn.kmu.edu.t jclee.dlearn.kmu.edu.tw TEL: 2369 Office: N1024 Faculty of Biotechnology Kaohsiung Medical University Outline

More information

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Joseph J. Mattapallil 1, Daniel C. Douek 2, Brenna Hill 2, Yoshiaki Nishimura 3, Malcolm Martin 3 & Mario

More information

The Swarm: Causes and consequences of HIV quasispecies diversity

The Swarm: Causes and consequences of HIV quasispecies diversity The Swarm: Causes and consequences of HIV quasispecies diversity Julian Wolfson Dept. of Biostatistics - Biology Project August 14, 2008 Mutation, mutation, mutation Success of HIV largely due to its ability

More information

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Anne Piantadosi 1,2[, Bhavna Chohan 1,2[, Vrasha Chohan 3, R. Scott McClelland 3,4,5, Julie Overbaugh 1,2* 1 Division of Human

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

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

Prevention of infection 2 : immunisation. How infection influences the host : viruses. Peter Prevention of infection 2 : immunisation How infection influences the host : viruses Peter Balfe, p.balfe@bham.ac.uk @pbalfeuk Let s have some LO s just for fun 1. Define the Immune response to viruses,

More information

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

Department of Surgery, Duke University, Durham, North Carolina; and 7 Southern Research Institute, Frederick, Maryland MAJOR ARTICLE Highly Attenuated Rabies Virus Based Vaccine Vectors Expressing Simian-Human Immunodeficiency Virus 89.6P Env and Simian Immunodeficiency Virus mac239 Gag Are Safe in Rhesus Macaques and

More information

HIV-1 Viral Load Real Time (RG)

HIV-1 Viral Load Real Time (RG) -1 Viral Load Real Time (RG) Real Time RT-PCR type 1 RNA quantification assay MSP Reg. pending Valdense 3616. 11700. Montevideo. Uruguay. phone (598) 2 336 83 01. Fax (598) 2 336 71 60. Info@atgen.com.uy

More information

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

Vaccination of Cats with Attenuated Feline Immunodeficiency Virus Proviral DNA Vaccine Expressing Gamma Interferon JOURNAL OF VIROLOGY, Jan. 2007, p. 465 473 Vol. 81, No. 2 0022-538X/07/$08.00 0 doi:10.1128/jvi.00815-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Vaccination of Cats with

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

Investigation of the genetic differences between bovine herpesvirus type 1 variants and vaccine strains

Investigation of the genetic differences between bovine herpesvirus type 1 variants and vaccine strains Investigation of the genetic differences between bovine herpesvirus type 1 variants and vaccine strains Name: Claire Ostertag-Hill Mentor: Dr. Ling Jin Bovine herpesvirus Bovine herpesvirus-1 (BHV-1) Pathogen

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 J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01.

NIH Public Access Author Manuscript J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01. NIH Public Access Author Manuscript Published in final edited form as: J Acquir Immune Defic Syndr. 2012 September 1; 61(1): 19 22. doi:10.1097/qai.0b013e318264460f. Evaluation of HIV-1 Ambiguous Nucleotide

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

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

Received May 1, 2002; returned to author for revision May 20, 2002; accepted June 3, 2002 Virology 301, 365 373 (2002) doi:10.1006/viro.2002.1598 A Simian Immunodeficiency Virus Nef Peptide Is a Dominant Cytotoxic T Lymphocyte Epitope in Indian-Origin Rhesus Monkeys Expressing the Common MHC

More information

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

Magnitude and Diversity of Cytotoxic-T-Lymphocyte Responses Elicited by Multiepitope DNA Vaccination in Rhesus Monkeys JOURNAL OF VIROLOGY, Sept. 2003, p. 10113 10118 Vol. 77, No. 18 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.18.10113 10118.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Magnitude

More information

ALTHOUGH disease develops within 10 years in

ALTHOUGH disease develops within 10 years in 228 THE NEW ENGLAND JOURNAL OF MEDICINE Jan. 26, 1995 BRIEF REPORT: ABSENCE OF INTACT nef SEQUENCES IN A LONG-TERM SURVIVOR WITH NONPROGRESSIVE HIV-1 INFECTION FRANK KIRCHHOFF, PH.D., THOMAS C. GREENOUGH,

More information

CD8 memory, immunodominance, and antigenic escape

CD8 memory, immunodominance, and antigenic escape 2704 D. Wodarz and M. A. Nowak Eur. J. Immunol. 2000. 30: 2704 2712 CD8 memory, immunodominance, and antigenic escape Dominik Wodarz and Martin A. Nowak 1 Institute for Advanced Study, Princeton, USA Previous

More information

MID 36. Cell. HIV Life Cycle. HIV Diagnosis and Pathogenesis. HIV-1 Virion HIV Entry. Life Cycle of HIV HIV Entry. Scott M. Hammer, M.D.

MID 36. Cell. HIV Life Cycle. HIV Diagnosis and Pathogenesis. HIV-1 Virion HIV Entry. Life Cycle of HIV HIV Entry. Scott M. Hammer, M.D. Life Cycle Diagnosis and Pathogenesis Scott M. Hammer, M.D. -1 Virion Entry Life Cycle of Entry -1 virion -1 Virus virion envelope Cell membrane receptor RELEASE OF PROGENY VIRUS REVERSE Co- TRANSCRIPTION

More information

SEROLOGICAL DIAGNOSIS OF VIRAL INFECTIONS:

SEROLOGICAL DIAGNOSIS OF VIRAL INFECTIONS: SEROLOGICAL DIAGNOSIS OF VIRAL INFECTIONS: POSSIBILITIES OF SEROLOGICAL DIAGNOSIS TYPES OF SEROLOGICAL REACTIONS SEROLOGICAL REACTIONS Ag-Ab reactions used for the detection of unknown Ag or Ab, in vitro

More information

Effect of Humoral Immune Responses on Controlling Viremia during Primary Infection of Rhesus Monkeys with Simian Immunodeficiency Virus

Effect of Humoral Immune Responses on Controlling Viremia during Primary Infection of Rhesus Monkeys with Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Feb. 2003, p. 2165 2173 Vol. 77, No. 3 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.3.2165 2173.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Effect of

More information

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

Simian-Human Immunodeficiency Infection Is the Course Set in the Acute Phase? Simian-Human Immunodeficiency Infection Is the Course Set in the Acute Phase? Janka Petravic, Miles P. Davenport* Complex Systems in Biology Group, Centre for Vascular Research, University of New South

More information

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

VIRAL TITER COUNTS. The best methods of measuring infectious lentiviral titer VIRAL TITER COUNTS The best methods of measuring infectious lentiviral titer FLUORESCENCE CYCLES qpcr of Viral RNA SUMMARY Viral vectors are now routinely used for gene transduction in a wide variety of

More information

DATA SHEET. Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter calf thymus DNA.

DATA SHEET. Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter calf thymus DNA. Viral Load DNA >> Standard PCR standard 0 Copies Catalog Number: 1122 Lot Number: 150298 Release Category: A Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter

More information

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

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2 JOURNAL OF VIROLOGY, Jan. 2000, p. 573 579 Vol. 74, No. 1 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. The S2 Gene of Equine Infectious Anemia Virus Is

More information

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

Emergence of cytotoxic T lymphocyte escape mutations in nonpathogenic simian immunodeficiency virus infection Eur. J. Immunol. 2001. 31: 3207 3217 CTL escape in nonpathogenic SIV infection 3207 Emergence of cytotoxic T lymphocyte escape mutations in nonpathogenic simian immunodeficiency virus infection Amitinder

More information

, virus identified as the causative agent and ELISA test produced which showed the extent of the epidemic

, virus identified as the causative agent and ELISA test produced which showed the extent of the epidemic 1 Two attributes make AIDS unique among infectious diseases: it is uniformly fatal, and most of its devastating symptoms are not due to the causative agent Male to Male sex is the highest risk group in

More information

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay Background ImQuest BioSciences has developed and qualified a single-plate method to expedite the screening of antiviral agents against

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

Downloaded by on April 28, 2018 https://pubs.acs.org Publication Date: April 24, 1984 doi: /bk

Downloaded by on April 28, 2018 https://pubs.acs.org Publication Date: April 24, 1984 doi: /bk 1 Virus-Receptor Interactions BERNARD N. FIELDS Department of Microbiology and Molecular Genetics, Harvard Medical School, and Department of Medicine (Infectious Disease), Brigham and Women's Hospital,

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

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

ADCC Develops Over Time during Persistent Infection with Live-Attenuated SIV and Is Associated with Complete Protection against SIV(mac)251 Challenge University of Massachusetts Medical School escholarship@umms Preventive and Behavioral Medicine Publications and Presentations Preventive and Behavioral Medicine 8-2012 ADCC Develops Over Time during Persistent

More information

5. Over the last ten years, the proportion of HIV-infected persons who are women has: a. Increased b. Decreased c. Remained about the same 1

5. Over the last ten years, the proportion of HIV-infected persons who are women has: a. Increased b. Decreased c. Remained about the same 1 Epidemiology 227 April 24, 2009 MID-TERM EXAMINATION Select the best answer for the multiple choice questions. There are 60 questions and 9 pages on the examination. Each question will count one point.

More information

Diagnostic Methods of HBV and HDV infections

Diagnostic Methods of HBV and HDV infections Diagnostic Methods of HBV and HDV infections Zohreh Sharifi,ph.D Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine Hepatitis B-laboratory diagnosis Detection

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

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

What is the role of animal models in studying protective titres and the need for establishing surrogates/correlates of protection? What is the role of animal models in studying protective titres and the need for establishing surrogates/correlates of protection? Alan D.T. Barrett Department of Pathology and Sealy Center for Vaccine

More information

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish SUPPLEMENTARY INFOATION Divergent TLR7/9 signaling and type I interferon production distinguish pathogenic and non-pathogenic AIDS-virus infections Judith N. Mandl, Ashley P. Barry, Thomas H. Vanderford,

More information

Original Article Development and Sequence Analysis of a Cold-Adapted Strain of Influenza A/New Caledonia/20/1999(H1N1) Virus

Original Article Development and Sequence Analysis of a Cold-Adapted Strain of Influenza A/New Caledonia/20/1999(H1N1) Virus Iranian Journal of Virology 2011;5(4): 6-10 2011, Iranian Society for Virology Original Article Development and Sequence Analysis of a Cold-Adapted Strain of Influenza A/New Caledonia/20/1999(H1N1) Virus

More information

New Insights on Mechanisms of Foamy Macrophage (FM) Induction and Persistence

New Insights on Mechanisms of Foamy Macrophage (FM) Induction and Persistence New Insights on Mechanisms of Foamy Macrophage (FM) Induction and Persistence Marian Laderoute, Ph.D. Medical Sciences -Immunology Lab Director Immune System Management Clinic & Lab 80 Aberdeen Street,

More information

Virus-specific cytotoxic T-lymphocyte responses select for amino-acid variation in simian immunodeficiency virus Env and Nef

Virus-specific cytotoxic T-lymphocyte responses select for amino-acid variation in simian immunodeficiency virus Env and Nef Virus-specific cytotoxic T-lymphocyte responses select for amino-acid variation in simian immunodeficiency virus Env and Nef DAVID T. EVANS 1, DAVID H. O CONNOR 1, PEICHENG JING 1, JOHN L. DZURIS 2, JOHN

More information

Received 30 December 2006/Accepted 25 February 2007

Received 30 December 2006/Accepted 25 February 2007 JOURNAL OF VIROLOGY, May 2007, p. 5202 5211 Vol. 81, No. 10 0022-538X/07/$08.00 0 doi:10.1128/jvi.02881-06 Copyright 2007, American Society for Microbiology. All Rights Reserved. Long-Term Control of Simian

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

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys Dan H. Barouch, M.D., Ph.D. Center for Virology and Vaccine Research Beth Israel Deaconess Medical Center Ragon Institute of MGH, MIT,

More information

Selection on the Human Immunodeficiency Virus Type 1 Proteome following Primary Infection

Selection on the Human Immunodeficiency Virus Type 1 Proteome following Primary Infection JOURNAL OF VIROLOGY, Oct. 2006, p. 9519 9529 Vol. 80, No. 19 0022-538X/06/$08.00 0 doi:10.1128/jvi.00575-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Selection on the Human

More information

Nature Medicine: doi: /nm.4322

Nature Medicine: doi: /nm.4322 1 2 3 4 5 6 7 8 9 10 11 Supplementary Figure 1. Predicted RNA structure of 3 UTR and sequence alignment of deleted nucleotides. (a) Predicted RNA secondary structure of ZIKV 3 UTR. The stem-loop structure

More information