Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 May 10.
Published in final edited form as: Virology. 2009 Mar 18;387(2):273–284. doi: 10.1016/j.virol.2009.02.014

Control of viremia and maintenance of intestinal CD4+ memory T cells in SHIV162P3 infected macaques after pathogenic SIVMAC251 challenge

Bapi Pahar a, Andrew A Lackner a, Michael Piatak Jr b, Jeffrey D Lifson b, Xiaolei Wang a, Arpita Das c, Binhua Ling a, David C Montefiori d, Ronald S Veazey a
PMCID: PMC2674129  NIHMSID: NIHMS96763  PMID: 19298994

Abstract

Recent HIV vaccine failures have prompted calls for more preclinical vaccine testing in nonhuman primates. However, similar to HIV infection of humans, developing a vaccine that protects macaques from infection following pathogenic SIVMAC251 challenge has proven difficult, and current vaccine candidates at best, only reduce viral loads after infection. Here we demonstrate that prior infection with a chimeric simian-human immunodeficiency virus (SHIV) containing an HIV envelope gene confers protection against intravenous infection with the heterologous, highly pathogenic SIVMAC251 in rhesus macaques. Although definitive immune correlates of protection were not identified, preservation and/or restoration of intestinal CD4+ memory T cells were associated with protection from challenge and control of viremia. These results suggest that protection against pathogenic lentiviral infection or disease progression is indeed possible, and may correlate with preservation of mucosal CD4+ T cells.

Keywords: Antigen specific T cells, Neutralizing antibody, Mucosal immunity, ELISPOT, Rhesus macaque, SIV, SHIV, Vaccine

INTRODUCTION

Although it has been over 25 years since the beginning of the AIDS epidemic, an effective vaccine remains elusive. To date, all HIV-1 vaccine candidates tested in humans have failed to prevent infection or meaningfully impact viral replication (Koff et al., 2006). Furthermore, most vaccine approaches evaluated in non-human primate models, including protein subunits, inactivated particles, DNA, and recombinant live vectors, alone or in various combinations, have shown at best only limited, partial efficacy, particularly in studies involving stringent challenges with pathogenic SIV strains such as SIVMAC239, SIVMAC251 or SIVSME660 (Casimiro et al., 2005; Egan et al., 2000; Horton et al., 2002; Koff et al., 2006; Larke et al., 2005). The recent disappointing failure of the STEP clinical trial of the Merck Ad5 vectored candidate vaccine, despite induction of readily measurable virus specific cellular immune responses, emphasizes we still do not understand the basis of protective immunity against HIV infection (Cohen, 2007; Watkins, 2008).

In contrast, use of live attenuated viruses as vaccines has demonstrated more robust protection from rigorous homologous and heterologous viral challenges in macaques, thus providing critical proof-of-concept for the feasibility of an HIV vaccine to prevent or limit HIV infection (Abel et al., 2003; Connor et al., 1998; Daniel et al., 1992; Koff et al., 2006; Kumar et al., 2002; Marthas et al., 1992; Mori et al., 2001; Wyand et al., 1996). However, most of these studies utilized relatively insensitive methods for viral detection, or protection was defined as a reduction of ~1 log plasma viral load compared to controls. Moreover, relatively few studies have investigated the potential mechanisms of this protection with contemporary assays for virus-specific cellular immune assays or antiviral antibody. Abel et al have demonstrated that “immunization” with the attenuated virus SHIV89.6 can confer protection in a significant fraction of animals against subsequent intravaginal (IVAG) challenge with the highly pathogenic SIVMAC239 (Abel et al., 2003), demonstrating that protection can be achieved against mucosal transmission of a highly pathogenic partially heterologous virus that is sequence divergent in env and regulatory genes. However, since the challenge was IVAG, it is not clear whether this protection was attributable to mucosal or systemic immunity. A model that demonstrated protection against intravenous (IV) challenge would provide even more evidence that protection against highly pathogenic SIV challenge is achievable, and would allow assessment of the role of systemic immune responses in such control. Our recent study showed that rhesus macaques were highly susceptible to vaginal transmission of SHIV162P3, yet ~90% had low to undetectable plasma viremia within 70–90 days of infection (Pahar et al., 2007). We hypothesized that these animals may thus be “immunized” against subsequent challenge with SIVMAC251, which could provide a model for assessing correlates of immunity to this highly pathogenic challenge. Alternatively, we hypothesized that prior infection could immunocompromise animals, and result in an exacerbated course of infection in response to SIVMAC251 challenge, thus providing a model for studying HIV “superinfection”, defined as when a currently HIV-infected person becomes infected with a second strain of HIV from a subsequent exposure.

While superinfection of HIV-1 infected patients with a second strain of HIV-1 has implications for vaccine development (Gottlieb et al., 2007), superinfection can be difficult to prove, as it is often difficult to distinguish whether viral variants are the result of subsequent infection or simply represent mutations of the initial infecting virus (Gottlieb et al., 2007). Furthermore, the consequences of superinfection remain unclear (Gottlieb et al., 2007). Nonhuman primate models provide an opportunity to address these questions in an experimentally tractable fashion. Here we demonstrate that macaques primarily infected with the minimally pathogenic virus SHIV162P3 conferred partial to complete protection against subsequent IV challenge with the highly pathogenic SIVMAC251. Furthermore. We demonstrate that mucosal CD4+ memory T cells were maintained following pathogenic challenge, suggesting that mucosal immune responses play a role in this protection. Finally, these results indicate that immunity to infection with SIV/HIV may indeed be possible, and additional studies with these and similar models of protection may provide insights into the immune correlates of protection from HIV infection.

RESULTS

“Immunization” with SHIV162P3

Rhesus macaques of Indian (InRh) or Chinese (ChRh) were atraumatically inoculated intravaginally with 300 TCID50 SHIV162P3 and confirmed to be infected, as evidenced by high peak plasma viremia between 14 and 21 days post inoculation ranging from 0.45 to 6.3 x 106 viral RNA (vRNA) copies/ml plasma, proving that none of the animals were genetically “resistant” to SHIV/SIV infection (Figs. 1 and 2). Viral loads were undetectable in 6 out of 14 SHIV162P3 inoculated animals by 70 days post inoculation (Fig. 2). All macaques were followed for 89 to 345 days post inoculation, until plasma viral load (VL) declined to levels from undetectable to 7,600 vRNA copies/ml of plasma. This initial infection is subsequently referred to as the “immunization” phase.

Figure 1.

Figure 1

Experimental design of the SHIV162P3 inoculation and SIVMAC251 challenge. SHIV infected macaques were subsequently intravenously challenged with different dosages of pathogenic SIVMAC251 on the days post inoculation indicated in three separate experiments. For controls, four naïve Chinese origin macaques were concurrently inoculated with SIVMAC251 (highlighted in red).

Figure 2.

Figure 2

Plasma viral RNA levels in macaques after primary SHIV162P3 infection, prior to pathogenic SIVMAC251 challenge. All animals had high primary viremia 14–21 days after vaginal inoculation, which declined to low or undetectable levels by 70 days of infection. Viral loads were undetectable in 6 out of the 14 animals by 70 days post inoculation.

In a series of three sequential experiments, all “immunized” macaques were then intravenously challenged with SIVMAC251. The three experiments were designed to determine whether the dose, source, or passage of the inoculum contributed to protection from rechallenge. In the first experiment (Group A), seven previously “immunized” macaques (four ChRh and three InRh) were IV challenged with a high dose (100 TCID50) of SIVMAC251. On the day of challenge, the InRh macaques (AK46, R191 and T552) had higher residual plasma VLs from the prior SHIV162P3 immunization (0.03–0.76 x 104 vRNA copies/ml of plasma) compared to the four ChRh macaques, two of which had undetectable (AJ61 and DT93) or very low levels (340–650 vRNA copies/ml plasma) on the day of challenge (Fig. 3A). As a concurrent control, a naïve (non-immunized) ChRh macaque (V780) was IV inoculated with 100 TCID50 of SIVMAC251.

Figure 3.

Figure 3

Plasma viral RNA levels in macaques following SIVMAC251 challenge. Plasma viremia is reported as log10 vRNA copies per ml of plasma (detection limit 30 vRNA copies/ml). Plasma viral loads from individual experiments (A) and mean +/− SE of the mean viral loads for each of the different groups (B) are shown. Note there was significantly higher viremia in non-immunized, control macaques (red lines) compared to immunized macaques during early (day 14), steady state (day 42) and late (day 282) stages of infection following SIVMAC251 challenge. Plasma viral load from the 3 InRh in Group A is presented as a dashed line. (B) Mean plasma viral loads from 4 historical naive ChRh intravenously infected with 100TCID50 SIVMAC251 are shown as dashed purple line.

Following challenge, one ChRh macaque (DT93) showed no measurable viremia (<30 vRNA copies/ml) through >640 days of follow-up. The other 3 ChRh macaques showed excellent control of viral replication, with plasma viremia 2–3 logs lower than the naïve control throughout primary infection. All seven macaques immunized by prior infection with SHIV162P3 showed greater levels of resistance or control of the high dose IV SIVMAC251 challenge, at least through 154 days of challenge (Fig. 3A). In addition, five of the seven macaques did not demonstrate a “peak” of viremia in the first weeks following challenge. Only two macaques (InRh macaque T552 and ChRh macaque AJ61) showed any increased viremia after SIVMAC251 challenge. Plasma viremia declined or remained low in one macaque (DT55) throughout 154 days after challenge and increased thereafter (Fig. 3A). These results suggest that substantial control of viremia following a high dose IV challenge with a pathogenic, heterologous virus can be achieved through prior immunization with an attenuated/weakly pathogenic SHIV.

The second experiment (Group B) was designed to determine if greater control could be achieved following a lower IV challenge. Four ChRh macaques previously “immunized” by SHIV162P3 infection, and one SIV-naïve ChRh control were IV inoculated with 10 TCID50 of the same SIVMAC251 challenge stock used in Group A. On the day of challenge, all four “immunized” macaques had undetectable (FA10) or low levels of plasma viremia (40–340 vRNA copies/ml). Following IV SIVMAC251 challenge, three out of four immunized macaques had no significant increase in plasma viremia over pre-challenge levels (i.e., no peak viremia), and their vRNA was intermittently undetectable in plasma (<30 vRNA copies/ml) on multiple occasions (Figs. 3A and B). Plasma VLs in CF58 remained less than 1000 copies/ml after challenge through day 42, but then gradually climbed to approximately 10,000 copies/ml by day 285 and this animal developed AIDS 2.5 years after SIV challenge (Fig. 3A). The three remaining SHIV immunized and SIVMAC251 challenged macaques in this group have remained healthy through 3 years of follow-up, and have had undetectable plasma viremia. In contrast, the naïve ChRh control macaque had high peak and persistent set point plasma viremia indistinguishable from the control in Group A, and both controls were subsequently euthanized with signs and lesions of AIDS. Combined, the results of studies in Group A (high IV dose) and B (low IV dose) demonstrated that resistance to IV superinfection (1 macaque in group A) and/or remarkable control of plasma viremia (3 macaques in Group B) could be conferred by prior infection with a virus having a divergent envelope. Moreover, even in animals that were persistently infected, plasma viremia remained 1–3 logs lower during the early phase of infection than in control macaques inoculated with the same lot, route, and dose of virus.

To rule out potential artifacts of immune control resulting from using ChRh vs. InRh hosts, the third experiment (Group C) utilized an inoculum passaged in ChRh. Plasma VLs have been shown to be lower in ChRh than InRh macaques inoculated with SIV grown in InRh peripheral blood mononuclear cells (PBMC) (Ling et al., 2002), and thus the differences we observed could be due to adaptive “fitness” of the particular stocks for InRh macaque cells. Thus, we challenged five ChRh macaques intravenously with 1 ml plasma containing ~2.0 x106 vRNA copies/ml of SIVMAC251 which was obtained from a ChRh macaque on a serial passage study. This experiment was also important to compare the results of virus obtained from plasma compared to in vitro expanded viral stocks in generating infection and potential differences in protection from this challenge.

Of the 5 ChRh macaque animals inoculated with ChRh macaque-derived plasma virus, three were previously “immunized” with SHIV162P3 and two were naïve controls. Of the three “immunized” macaques, two had undetectable plasma viremia (< 30 vRNA copies/ml), while one (L375) had 1600 copies/ml remaining at the time of challenge. Following challenge, both animals with undetectable viremia on the day of challenge continued to control viremia to less than 1000 copies/ml after challenge. The third (L375) also demonstrated VLs 1–2 logs lower than the naïve controls until day 190 post challenge (p.c.) (Fig. 3A) but then plasma VL increased to levels similar to naïve challenged macaques. In contrast, both naïve controls demonstrated uncontrolled viral replication and 1 log higher vRNA plasma copies compared to that of the controls in Groups A and B (Fig. 3A).

Highly significant differences in peak and post peak VLs were detected between controls and immunized challenged macaques (p = 0.015 to 0.003, Fig. 3B). When the InRh were excluded from the analysis, these differences were even more significant. Prior immunization with SHIV162P3 resulted in a 1.8 to 3.7-log reduction in plasma VLs in all immunized macaques compared to naive controls through 156 days p.c. regardless of whether animals were challenged with high or low IV doses, or SIVMAC infected plasma (Fig. 3B).

We also examined cell-associated viral loads in intestinal CD4+ T cells on a subset of the animals where sufficient cells were available (Fig. 4A). Copies of SIVgag RNA were low (FA14, 50 copies of SIVgag RNA) to undetectable in both SHIV immunized and naïve challenged macaques (Fig. 4A) on the day of SIVMAC251 challenge. By 21 days post challenge all 3 immunized macaques had low (FA14, 90 copies of SIVgag RNA) to undetectable levels of virus whereas, naïve challenged macaques (FG46 and FA06) had much higher levels in intestinal tissues (Fig. 4A). Similar differences in intestinal VLs were detected in both SHIV immunized and naïve challenged macaques after 56 and 104 days of SIVMAC251 challenge (Fig. 4A).

Figure 4.

Figure 4

(A) Cell-associated viral RNA levels in jejunum LPL in macaques following SIVMAC251 challenge (detection limit 30 vRNA copies). There was low to undetectable viremia in intestinal tissues at the day of SIVMAC251 challenge in SHIV immunized macaques. By 21, 56 and 104 days post SIVMAC251 challenge increased viral replication was noticed in naïve challenged macaques, whereas SHIV162P3 immunized macaques had low to undetectable tissue viral load. Mean tissue VLs are shown with a line in each plot. (B) Kaplan-Meier survival curves for the immunized and non-immunized macaques in all three groups after SIVMAC251 challenge demonstrating significantly increased survival of immunized macaques compared to controls. Survival comparisons were performed using a log-rank test.

There was also a highly significant difference in survival between immunized and naïve macaques (p=0.009) (Fig. 4B). Of the 14 “immunized” macaques challenged with SIV, 5 remain alive and healthy (3 years since challenge); five were euthanized for unrelated reasons 2 years after infection (with no detectable plasma viremia or clinical signs related to SIV infection). Only 4 immunized animals were euthanized with AIDS defining illnesses (DT55, T552, CF58 and L375). In contrast, all 4 of the SIV naïve control ChRh macaques were euthanized due to AIDS-defining illnesses (opportunistic infection, lymphoma, and/or SIV encephalitis) within 14 months of challenge.

Maintenance of systemic and intestinal CD4+ T cells in immunized macaques

Although animals in groups A–C were challenged with SIVMAC251 at very different time points after SHIV162P3 immunization, all were challenged at time points when plasma VLs had declined to low or undetectable levels. To assess whether the limitation of viral replication observed in SHIV162P3 immunized animals following SIV challenge reflected a paucity of viral target cells induced by the initial SHIV162P3 infection, percentages of total CD4+ T cells were compared in peripheral blood, intestine (jejunum) and axillary lymph node (LN) in naïve and SHIV immunized macaques immediately prior to, and subsequent to SIV challenge. Prior to challenge, percentages of total CD4+ cells in blood were stable in all animals (mean CD4+ T cell % in group A, B and C was 51.7% (range 28.7–63.6), 56.4% (range 49.5–67.2) and 54.7% (range 49.3–62.2) respectively), reflecting the low level of plasma viremia at the time of challenge. Total CD4+ T cell populations in LN also were relatively normal in immunized macaques at the time of challenge. Following SIVMAC251 challenge, percentages of CD4+ T cell counts dropped significantly (P<0.01) in the peripheral blood of all naïve controls, yet remained stable in all immunized animals through at least 119 days p.c. (Fig. 5A). However, six macaques (AK46, CF58, DT55, L375, R191 and T552) had substantial drops in CD4+ T cells 250–402 days after challenge, which inversely correlated with increasing VLs at these timepoints. Similarly, there were decreased percentages of CD4+ T cells in the LN of naïve macaques, yet CD4+ T cells remained fairly stable in immunized macaques (Fig. 5B).

Figure 5.

Figure 5

Dynamics of CD4+ T cell percentages in peripheral blood (A) and axillary lymph node (B) of immunized and non-immunized macaques following SIVMAC251 challenge. Note all non-immunized (naïve) challenged macaques had a rapid decrease in CD4+ T cells by day 14 after challenge in both peripheral blood and LN (red lines) whereas CD4+ T cells were preserved in immunized macaques. InRh macaques are delineated by dashed lines.

Surprisingly, percentages of CD4+ T cells in the intestine at the time of challenge were highly variable in immunized animals, yet they did not appear to correlate with levels of plasma viremia. Several SHIV immunized macaques had lower levels of CD4+ T cells in intestinal lamina propria lymphocytes (LPL) compared to naïve controls at the time of challenge. Selective depletion of intestinal CD4+ T lymphocytes following SHIV162P3 infection has been previously reported (Hsu et al., 2003). Intestinal CD4+ T cell percentages were similar in groups A (mean 25%, ranged from 10.0–42.1%) and B (mean 26.7%, ranged from 12.3–39.7%) whereas naive macaques V780 and V302 had around 33% intestinal CD4+ T cells at the time of SIVMAC251 challenge (Figs. 6A and B). However, intestinal CD4+ T cells in group C were lower compared to animals in both A and B but not significantly different (p=0.62) (Fig. 6B). The two naïve controls in group C had higher (~43%) intestinal CD4+ T cells at the day of challenge (Fig. 6B). Following SIVMAC251 challenge, essentially all SHIV-immunized macaques maintained near baseline levels of intestinal CD4+ T cells through at least 42 days of infection, whereas all naïve macaques demonstrated marked and persistent reductions in intestinal CD4+ T cells (Figs. 6A and B). In fact, percentages of intestinal CD4+ T cells increased by day 7 and 14 in several SHIV-immunized macaques (Fig. 6B). The main exceptions were in immunized macaques DT93 and R191, which showed moderate drops in intestinal CD4+ T cells by 14 days post challenge, yet these returned to baseline levels in the following weeks (Fig 6B). Similarly, macaques CF58 and FA05 had a transient drop in percentages of intestinal CD4+ T cells 7 days after challenge, yet these animals also recovered to levels higher than at the time of SIV challenge, indicating eventual control of replication in mucosal sites (Fig. 6B). Memory (CD45RA) CD4+CCR5+ T cells in the intestine, the major early targets for SIV/HIV infection and replication (Brenchley et al., 2004; Mattapallil et al., 2005; Veazey et al., 2000a; Veazey et al., 2000b; Wang et al., 2007) were also relatively maintained in SHIV-infected macaques at the time of challenge (Figs. 7A and B) suggesting that the limitation of viral replication observed following pathogenic challenge was not attributable to a paucity of viral target cells. Overall, intestinal CD4+ T cell percentages from all naive control macaques (groups AC) demonstrated marked and significant losses by 14 days after challenge with SIVMAC251 (p=0.007), whereas immunized macaques did not change significantly. Similarly, a highly significant difference (p=0.009) in the availability of CD4+CCR5+ target cells was detected between SHIV immunized and naïve challenged macaques by 14 days after SIV challenge indicating that immunized animals had a bona fide “resistance” to infection, associated with preservation/restoration of mucosal viral target cells (Figs. 6 and 7).

Figure 6.

Figure 6

(A) A representative dot plots showing CD4+ T cells as a percentage of total T cells (gated through CD3+ lymphocytes) in macaques (Group B) after SIVMAC251 challenge. Note that restoration and/or preservation of intestinal CD4+ T cells was observed in a SHIV immunized macaque (FA08) compared to the non-immunized macaque (V302), in which there was rapid and sustained loss of intestinal CD4+ T cells within 14 days of challenge. (B) Changes in intestinal CD4+ T cells of immunized and non-immunized macaques following challenge with SIVMAC251. Note that in all groups, there was a rapid decrease in intestinal CD4+ T cells in all non-immunized (highlighted in red) compared to immunized macaques by day 14-post challenge. Intestinal CD4+ T cells of InRh are shown as dashed lines.

Figure 7.

Figure 7

(A) Percentages of memory (CCR5+CD45RAneg) CD4+ T cells in the intestine of a representative SHIV immunized (FA08) and non-immunized, naive (V302) macaque before (day 0) and 14 days after SIVMAC251 challenge. Note the marked and rapid decrease in intestinal target cells (CD3+CD4+CCR5+CD45RAneg) typical of naïve macaques challenged with SIVMAC251, whereas these cells are maintained in the immunized macaque. (B) Percentages of intestinal CD4+CCR5+ cells (gated through CD3+ lymphocytes) are shown for groups A–C. Note a rapid decrease in CD4+CCR5+ T cells occurs in all non-immunized challenged macaques (highlighted in red) within 14 days of SIVMAC251 challenge, whereas target cells are preserved in immunized macaques.

Investigations into potential “immune” control of SIV challenge

Several studies have suggested that neutralizing antibodies (Nabs) and/or T cell responses can play a role in controlling viral infection (Ferrantelli et al., 2004a; Ferrantelli et al., 2004b; Koup et al., 1994; Mascola et al., 2005; Schmitz et al., 1999). Nabs against the immunizing SHIV162P3, the SIVMAC251 challenge, as well as to laboratory-adapted SIVMAC251 were measured for all macaques from groups A, B and C. However, neither SHIV-immunized or naïve challenged macaques produced significant levels of Nab to SHIV or SIVMAC251 in any of the groups examined. Only one macaque (T552) had a transient low level of Nabs to SIVMAC251 4 weeks after infection, but Nab titers ranged from <20 to 67 in all other SHIV vaccinated macaques. Similarly, low to undetectable levels of Nabs were detected to the SIVMAC251 infection in naive challenged macaques.

On the day of SIVMAC251 challenge, antigen specific IFN-γ responses to tat, gag, nef, and env were examined in peripheral blood, LN and intestine of all SHIV162P3 immunized macaques in groups A & C. Overall, 90% of SHIV immunized animals demonstrated positive responses to SHIV-gag followed by HIV-env (70%) and HIV-tat (20%) antigens (Table 1). Despite these responses, neither the magnitude nor the presence of HIV-env and/or SIV-gag IFN-γ responses present at the time of challenge correlated with the level of viral control following SIVMAC251 challenge.

Table 1.

Antigen specific IFN-γ responses on the day of SIVMAC251 challenge from groups A and C macaquesa

Group Macaques Blood Lymph Node Jejunum LPL
A AJ61 negative 568 negative

AK46 negative 395 115
55 45

DT55 negative negative 95Inline graphic
115
300

DT91 negative 98 negative

DT93 253 negative 70
48

T552 138Inline graphic negative negative
300
110

R191 448 negative negative
210

V780 negative negative negative

C FA14 negative 302 608
100 90
115

L375 207 122 720
45 715
57

P015 negative negative negative

FG46 negative negative negative

FA06 negative negative negative
a

Macaques in red were unvaccinated (naïve) followed by SIVMAC251 challenge. Other macaques were infected with SHIV followed by SIVMAC251 challenge. Values represent responses to specific peptide pools; Inline graphic = Tat, ➂ = Nef, ➄ = Gag, ➉ =gp120, and ❷ = gp41. Numbers indicate spot forming cells per one million PBMCs after medium-only control values were subtracted.

Following challenge, IFN-γ responses in blood were monitored by ELISPOT for >118 days in groups A and C, and in selected animals, through 84 days of infection in lymph nodes (Tables 2 and 3). In general, gag-specific responses were higher in immunized macaques followed by tat, nef, gp120 and gp41 responses (Tables 2 and 3). SIV-env specific IFN-γ responses were detected in PBMC as early as 21 days after challenge, but only in one macaque (AK46). Similarly, 7 out of 10 immunized macaques had detectable antigen specific IFN-γ responses by day 14 after challenge in LN. However, there was no correlation between control of plasma vRNA and IFN-γ responses in any of the immunized or control macaques (r2 ranged from 0.006 to 0.86 and p=0.06 to 0.89). We have also performed one way analysis of variance of the number of IFN-γ secreting cells in both vaccinated and unvaccinated challenged macaques in all the time point of this study but no significance difference in the number of IFN-γ positive cells at any time point (P>0.05) has been established.

Table 2.

Antigen specific IFN-γ responses in SIVMAC251 challenged macaques in peripheral blooda

Groups Monkey Peptide Pools Days post challenge
7 14 21 42 56 118 352
A AJ61 Tat 48 ND
Nef ND ND ND ND ND ND ND
Gag 195 180 350 420 118 ND

AK46 Tat 153 58 148 50 112
Nef ND ND ND ND ND ND
Gag 540 170 625 130 362 278 230
gp120 513 211

DT55 Nef ND ND ND ND ND ND 210
Gag 50 326 180 255 310 360
gp120 52 62 98

DT91 Nef ND ND ND ND ND ND ND
Gag 90 90 60 ND
gp120 ND
gp41 ND

DT93 Nef ND ND ND ND ND ND
Gag 103 118 82

T552 Nef ND ND ND ND ND ND 93
Gag 90 196 275 113
gp120 83 53
gp41 70

R191 Nef ND ND ND ND ND ND
Gag 53 222 428 338
gp120 135 96 175 178

V780 Tat 50
Nef ND ND ND ND ND ND 73
Gag 88

C FA14 Nef 140 117 ND 135 ND
Gag 102 65 357 420 ND 347 ND

L375 Tat ND 238 ND
Nef 67 ND 217 ND
Gag 152 247 182 450 ND 612 ND
gp120 ND 105 ND

FG46 gp120 ND 62 ND
a

Peptide–specific responses are depicted for PBMCs in one million cells, medium control subtracted. Although all peptide pools (tat, nef, gag and env) were tested for groups A and C macaques as indicated, only positive responses are shown. Macaques with red fonts depicted unvaccinated challenged macaques. ND= not determined.

Table 3.

Antigen specific IFN-γ responses in SIVMAC251 challenged macaques in lymphocytes isolated from lymph nodea

Groups Monkey Peptide Pools Days post challenge
14 28 35 84
A AJ61 Nef ND ND ND ND
Gag 252 500 ND 424
gp120 650 ND 168

AK46 Nef ND ND ND ND
Gag 1027 ND 366
gp120 100 ND

DT55 Nef ND ND ND ND
Gag 40 50 ND 110

DT93 Nef ND ND ND ND
Gag 105 ND 162

T552 Nef ND ND ND ND
Gag 220 ND

V780 Nef ND ND ND
Gag 120 ND

C FA14 Nef 405 ND 300 ND
Gag 372 ND 767 ND

L375 Nef 55 ND 117 ND
Gag 602 ND 1265 ND

P015 Nef ND 160 ND
Gag ND 260 ND

FG46 Gag ND 175 ND
a

Peptide–specific responses are reported for lymphocytes isolated from lymph nodes in one million cells, medium control subtracted. Although all peptide pools (tat, nef, gag and env) were tested for groups A and C macaques as indicated, only positive responses are shown. Macaques with red fonts are depicted as unvaccinated challenged. ND = not determined.

DISCUSSION

Correlates of immunity or resistance to HIV infection remain unknown. This has hampered efforts to produce an effective vaccine. The aim of a vaccine would be to prevent infection, ameliorate disease course, and/or reduce virus transmission. The risk of transmission is greatest when viremia is high, especially during acute, and uncontrolled chronic infection. Infected individuals with plasma VLs of <1,700 copies/ml fail to transmit the virus to their HIV-negative partners (Gray et al., 2003; Gray et al., 2001). An effective HIV vaccine should at least be able to limit viral replication to low levels in which transmission is unlikely (Wilson et al., 2006). Although some vaccine candidates have shown modest reductions in VLs in macaques after SIV challenge, these were usually performed under ideal conditions using homologous viral challenges or against less than optimal SHIV’s (Casimiro et al., 2005; Koff et al., 2006; Letvin et al., 2006; Mattapallil et al., 2006; Watkins et al., 2008; Wilson et al., 2006). However, a more recent study has shown partially effective control of viremia by a live attenuated SIV followed by IV challenge with a heterologous SIV (Reynolds et al., 2008). Moreover, the studies here also demonstrate that significant levels of protection from an IV heterologous challenge with a highly pathogenic strain of SIV in macaques is indeed possible, at least in a proportion of macaques. Although the immunogen used in this study was a replication-competent SHIV known to cause AIDS in a proportion of macaques, the results demonstrate that the SHIV immunization prolonged the survival and/or delayed disease progression after challenge with SIVMAC251. Furthermore, the fact that control of viremia persisted for months after levels of the immunizing virus had subsided, combined with the observation that mucosal viral target cells are at least partially restored prior to challenge, and more importantly, maintained after pathogenic challenge, suggests this control is associated with immune responses rather than a phenomenon of target cell depletion. Low to undetectable viral replication in intestinal tissues also suggested that maintenance/restoration of CD4+CCR5+ memory T cell targets was associated with control of the SIV challenge.

Unfortunately, we could not determine the correlates of immunity in these animals. Neither levels of plasma Nabs, nor virus specific systemic or mucosal IFN-γ responses correlated with protection. However, the fact that the envelope of the immunizing virus markedly differs from the challenge virus indicated that protection was not associated with immune responses directed against envelope. This may be an additional source of optimism, as it is unlikely that a vaccine can be engineered to generate responses to all of the diverse HIV envelope sequences encountered in endemic areas. Thus, vaccines might not have to be matched for all possible envelope sequences in order to confer protection.

One caveat that must be considered is whether this “protection” is indeed due to inducible immune responses or instead, viral competition for viral binding or integration sites. In this and prior studies, “protection” appears to be associated with persistence of low levels of virus or antigen in tissues. Conceivably, integrated virus could somehow prevent “superinfection” of target cells by either competition for integration sites, or by mediating cellular machinery to prevent subsequent infection or replication within infected cells. In fact, the lower levels of memory CD4+CCR5+ target cells in intestinal tissues of SHIV162P3 immunized animals at the time of SIV challenge suggested that there may be persistent viral replication in the gut, even though our analysis of cell-associated virus did not demonstrate significant levels of viral RNA in these animals (Fig. 4A).

However, there is some evidence that this protection is mediated by adaptive immune responses. The fact that “immunized” animals maintained memory CD4+CCR5+ T cells in intestinal tissues following SIV challenge, in contrast to naïve controls who showed a significantly greater degree of intestinal CD4 depletion, suggests that the protection observed is not due to a limitation of target cells necessary to establish productive superinfection. Furthermore, all controls became infected, maintained high VLs, and progressed to AIDS, proving that this is not an artifact of macaques of Chinese origin, which have a higher incidence of long term nonprogression than macaques of Indian origin (Ling et al., 2007). In addition, this protection is not attributed to immune responses directed against foreign host major histocompatibility complex (MHC) proteins incorporated into the viruses, as the effects were the same whether the viral inoculum was grown in InRh macaque cells or passaged in ChRh. Finally, four immunized macaques did demonstrate transient slight (FA05, CF58) to moderate (DT93, R191) drops in mucosal CD4+ T cells immediately following SIV challenge, suggesting virus was reaching intestinal CD4+ T cells, and that there was an initial struggle in mucosal sites for control of local viral replication (Fig. 6). Unlike infected naïve controls, all four of these animals eventually restored mucosal CD4+ T cells to levels higher than at the time of challenge (Fig. 6). In summary, these studies provide evidence that protection against a highly pathogenic strain of SIV may be achieved, and suggest that this protection may be associated with adaptive mucosal immune responses (Casimiro et al., 2005; Kawada et al., 2007; Wilson et al., 2006).

These studies also argue against the hypothesis that superinfection results in a more severe disease progression (Altfeld et al., 2002; Gottlieb et al., 2007; Jost et al., 2002; Smith et al., 2004; van der Kuyl et al., 2005). The challenge virus used in these studies (SIVMAC251) is among the most pathogenic of viruses in macaques, and if infection with one virus impairs immune responses against a second virus, we should have observed an enhancement of disease progression following SIV challenge. However, VLs were clearly diminished in the superinfected animals compared to controls, indicating that the primary infection confers some type of protection against superinfection and disease progression. In summary, this model may prove useful for understanding superinfection as well as correlates of immunity to HIV infection in humans. Although we are not promoting the use of attenuated viruses as vaccine candidates, we propose that such models may eventually help define correlates of immunity to infection, which could eventually guide HIV vaccine development.

MATERIALS AND METHODS

Macaques

Fifteen adult female Chinese rhesus (ChRh) and three Indian rhesus (InRh) (AK46, R191 and T552) macaques (Macaca mulatta) were housed at the Tulane National Primate Research Center in accordance with the regulations of the American Association for Accreditation of Laboratory Animal Care International. All animals were healthy and free from SIV, STLV and Type D retrovirus at the time the study was initiated.

Immunization

A series of three experiments was performed to determine whether macaques previously infected with SHIV162P3 were susceptible to subsequent “superinfection” with pathogenic SIV (Fig. 1). SHIV162P3 is a chimeric virus constructed from a SIVMAC239 template, yet it contains functional HIV-1 tat, rev and env genes (Harouse et al., 2001; Harouse et al., 1999). Although IV inoculation of macaques with high doses of this virus occasionally results in persistent viremia and AIDS (Harouse et al., 2001; Harouse et al., 1999), in our experience, vaginal inoculation with SHIV162P3 usually results in transient viremia and apparent “clearance” of virus within 70–90 days of infection (Pahar et al., 2007). To determine whether macaques previously infected with SHIV162P3 were susceptible to IV “superinfection” with pathogenic SIV, a total of fourteen infected macaques (11 of ChRh and 3 InRh) and 4 naïve controls (all ChRh) were IV challenged with SIVMAC251. For the IVAG primary infections (“immunization” phase) macaques used as placebo controls in microbicide experiments were first treated with Depo Provera (depomedroxyprogesterone acetate) and 30–33 days later, sedated and intravaginally inoculated with 300 TCID50 SHIV162P3 obtained from the NIH AIDS Research & Reagent Program (Pahar et al., 2007; Veazey et al., 2003). All 14 animals became infected as evidenced by high peak viremia 14–21 days after inoculation (Fig. 2). Macaques were then followed for 89–345 days post inoculation, until plasma viral loads declined to low (<7,600 vRNA copies/ml) or undetectable levels. In three separate experiments, infected animals and controls were IV challenged with a high (group A) and low (groupB) dose of SIVMAC251 and plasma from a SIV-infected macaque of ChRh origin (group C).

In the first experiment (group A), seven SHIV162P3 infected (4 ChRh and 3 InRh) and one-naïve ChRh macaques were IV challenged with 100 TCID50 SIVMAC251. In subsequent experiments all macaques (immunized and controls) were of Chinese origin. In the second experiment (group B), 4 SHIV162P3 infected and 1-naïve macaques were IV challenged with a lower dose (10 TCID50) of SIVMAC251. The SIVMAC251 challenge stock used in both A and B was grown in Indian origin macaque PBMC, and thus to rule out the possibility that the results observed were an artifact of using virus grown in MHC-mismatched hosts, animals in the third experiment were challenged with plasma obtained in primary infection from a ChRh macaque infected with SIVMAC251. In this experiment (group C), three SHIV infected and two naive macaques were IV challenged with 1ml of plasma containing 2 x 106 vRNA copies/ml.

Quantitation of plasma and cell associated viral RNA

Plasma VL during prechallenge time points was quantified by a bDNA signal amplification assay, version 4.0, specific for SIV, from Bayer Inc., which has a threshold detection limit of 125 copies/ml of plasma (Pahar et al., 2007). After SIVMAC251 challenge, plasma VL and cell-associated SIVgag RNA copies were determined by a sensitive real-time RT-PCR method as previously described (Cline et al., 2005) (limit of detection as used in the present study, 30 RNA copies/ml).

Isolation of lymphocytes from blood, intestine and lymph nodes

Lymphocytes from the peripheral blood, jejunum and axillary LN were isolated as described earlier (Veazey et al., 2000a; Veazey et al., 2000b; Wang et al., 2008). In brief, intestinal biopsies were obtained by endoscopy and digested with serial incubations with EDTA and collagenase with rapid agitation. To enrich for lymphocytes, cells were centrifuged over discontinuous Percoll (Sigma) density gradients, washed, and resuspended in complete RPMI media containing 5% FCS (RPMI-5). Intestinal lymphocytes were >90% viable as assessed by Trypan blue dye exclusion.

Lymphocytes were isolated from axillary LN by mincing with paired scalpel blades and pressing them through nylon cell mesh. Cells were washed twice and resuspended in RPMI-5.

T cell immunophenotyping

T cell immunophenotyping was performed using CD3-FITC/PerCP (SP34–2), CD4-APC (L200), CD8-PE/PerCP (RPA-T8), CCR5 PE (3A9) and CD45RA FITC (5A9), all from BD Biosciences. For surface staining, 100ul of EDTA whole blood was incubated with titrated mAb for 30 min on ice. Red blood cells were lysed with 1x FACS lysing buffer (BD Biosciences) using a whole blood lysis technique as previously described (Pahar, Lackner, and Veazey, 2006).

Intestinal and LN lymphocytes were adjusted to 107 cells/ml and 100ul aliquots were incubated with appropriately diluted antibodies for 30 min on ice, washed, and fixed in 2% paraformaldehyde. Cells were protected from light at 4°C and acquisition was completed within 24 h of staining. Data were acquired on a FACS Calibur flow cytometer (BD Biosciences, SanJose, CA) using Cell Quest software. At least 20,000 events were collected by gating on lymphocytes, and data were analyzed using FlowJo software (TreeStar Inc.) version 8.5.3.

ELISPOT assay

The number of antigen-specific IFN-γ spot forming cells (SFC) was measured with a commercial ELISPOT assay specific for rhesus IFN-γ (U-Cytech, Utrecht, The Netherlands), using fresh heparinized PBMC or lymphocytes isolated from LNs or intestine as previously described (Pahar et al., 2003; Pahar et al., 2007; Van Rompay et al., 2004). Peptides (15-mers with 11 amino acids overlap) derived from the HIV-tat (cat. 5138), SIV-tat (cat. 6207), SIV-nef (cat. 8762), SIV-gag (cat. 6204), SIV-env (cat. 6883) and SHIV162P3 env (cat. 7619) were obtained from the NIH AIDS Research & Reference Reagent Program. Peptide pools were prepared so that each protein was equally represented. One pool was made for HIV-tat (peptide 1–23), SIV-tat (peptide 1–30) and SIV-nef (peptide 1–64) peptides and three peptide pools (gagA: peptide 1–33; gagB: peptide 34–89; gagC: peptide 90–125) were made to encompass the entire gag region of SIV. Similarly four (envA: peptide 1-60; envB; peptide 61–124; envC: peptide 125–168; envD: peptide 169–211) and three (envA: peptide 1–60; envB; peptide 61–124; envC: peptide 125–218) peptide pools were prepared to encompass the entire env region of SHIV162P3 and SIV-env regions, respectively. For all positive control wells PMA (50ng/ml) and Ionomycin (1ug/ml) were used. Negative controls had no antigen/mitogen stimulation. Pooled peptides were added to cells at a final concentration of 1ug/ml for each individual peptide.

The numbers of spot forming cells were counted using the KS Elispot system (Zellnet Consulting Inc.). The mean number of spots per duplicate or triplicate well was calculated and the results were considered positive if the number of SFC for 2 x 105 cells was ≥ 10 per well and greater than two-fold that of the negative control (medium only) wells plus 2 standard deviations.

Virus neutralization assay

Neutralization of SHIV162P3 and primary SIVMAC251 was measured as a function of reduction in luciferase reporter gene expression after a single round of infection in TZM-bl cells as described (Montefiori, 2004). TZM-bl cells were obtained from the NIH AIDS Research and Reference Reagent Program. Briefly, 200 TCID50 of virus was incubated with serial three-fold dilutions of serum in triplicate in a total volume of 150ul for 1 h at 37°C in 96-well flat-bottom cell culture plates (Costar). Freshly trypsinized cells (10,000 cells in 100ul of growth medium containing 75ug/ml DEAE dextran) were added to each well. Positive and negative controls were kept with this assay. After incubation, 100ul of cells was transferred to 96-well black solid plates (Costar) for measurements of luminescence using Bright Glo substrate solution as described by the supplier (Promega). Neutralization titers were defined as the serum dilution at which relative luminescence units (RLU) were reduced 50% compared to virus control wells after subtraction of background RLUs. Assay stocks of primary SIVMAC251 and SHIV162P3 were prepared in human PBMC.

Neutralizing antibodies against TCLA SIVMAC251 were assessed in CEMx174 cells as described previously (Montefiori et al., 1996). Briefly 50 μl of cell-free virus (5,000 TCID50) was added to multiple dilutions of test serum in 100 μl of growth medium in triplicate wells of 96-well microtiter plates and incubated for 1 hr at 37°C. Cells (7.5 x 104) in 100 μl of growth medium were added and incubated until extensive syncytium formation and nearly complete cell-killing were evident microscopically in virus control wells. Cell densities were reduced and medium replaced after 3 days incubation in cases where it took longer than 3 days to reach the assay end-point. Viable cells were stained with Finter’s neutral red in poly-L-lysine coated plates. Percent protection from virus-induced cell-killing was determined by calculating the difference in absorption (A540) between test wells (cells + serum sample + virus) and virus control wells (cells + virus), dividing this result by the difference in absorption between cell control wells (cells only) and virus control wells and multiplying by 100. Nab titers are expressed as the reciprocal of the serum dilution required to protect 50% of cells from virus-induced killing. This cut-off corresponds to an approximate 90% reduction in p24 antigen synthesis. An assay stock of TCLA-SIVMAC251 was prepared in H9 cells.

Statistical analysis

Graphical presentation and statistical analysis of the results were performed by one-way analysis of variance or the Student t test using the GraphPad Prism 4.0 (GraphPad Software Inc., SanDiego, CA). Plasma VLs comparison between immunized and control challenged macaque was determined by nonparametric Mann-Whitney t test. Correlation between plasma VL and antigen specific cytokine responses was determined using Spearman rank correlation test using Prism software. For all statistical analysis, results were considered significant if p<0.05.

Acknowledgments

We thank Maryjane Dodd, Kelsi Rasmussen, Janell LeBlanc, Linda Green, Nancy Parr, Maury Duplantis, Melinda Martin for their technical assistance. We would also like to thank all veterinarian and animal care staff of the Dept. of Veterinary Medicine at Tulane National Primate Research Center for their expertise in biopsy and collection of tissue samples. The following reagent was obtained through the AIDS Research and Reference Reagent Program, NIAID, NIH: SHIV162P3 was contributed by Drs. Janet Harouse, Cecilia Cheng-Mayer, and Ranajit Pal, and the TZM-bl cells were contributed by John Kappes and Xiaoyun Wu and with support from NIH, NIAID, and DAIDS. The work was supported in part by NIH grants AI49080, P01AI051649, U19AI065413 and RR00164 and in part with federal funds from the National Cancer Institute, National Institutes of Health, under contract N01-CO-12400 (J.D.L.). The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abel K, Compton L, Rourke T, Montefiori D, Lu D, Rothaeusler K, Fritts L, Bost K, Miller CJ. Simian-human immunodeficiency virus SHIV89.6-induced protection against intravaginal challenge with pathogenic SIVmac239 is independent of the route of immunization and is associated with a combination of cytotoxic T-lymphocyte and alpha interferon responses. J Virol. 2003;77(5):3099–118. doi: 10.1128/JVI.77.5.3099-3118.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Altfeld M, Allen TM, Yu XG, Johnston MN, Agrawal D, Korber BT, Montefiori DC, O’Connor DH, Davis BT, Lee PK, Maier EL, Harlow J, Goulder PJ, Brander C, Rosenberg ES, Walker BD. HIV-1 superinfection despite broad CD8+ T-cell responses containing replication of the primary virus. Nature. 2002;420(6914):434–9. doi: 10.1038/nature01200. [DOI] [PubMed] [Google Scholar]
  3. Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, Nguyen PL, Khoruts A, Larson M, Haase AT, Douek DC. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200(6):749–59. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Casimiro DR, Wang F, Schleif WA, Liang X, Zhang ZQ, Tobery TW, Davies ME, McDermott AB, O’Connor DH, Fridman A, Bagchi A, Tussey LG, Bett AJ, Finnefrock AC, Fu TM, Tang A, Wilson KA, Chen M, Perry HC, Heidecker GJ, Freed DC, Carella A, Punt KS, Sykes KJ, Huang L, Ausensi VI, Bachinsky M, Sadasivan-Nair U, Watkins DI, Emini EA, Shiver JW. Attenuation of simian immunodeficiency virus SIVmac239 infection by prophylactic immunization with dna and recombinant adenoviral vaccine vectors expressing Gag. J Virol. 2005;79(24):15547–55. doi: 10.1128/JVI.79.24.15547-15555.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Cline AN, Bess JW, Piatak M, Jr, Lifson JD. Highly sensitive SIV plasma viral load assay: practical considerations, realistic performance expectations, and application to reverse engineering of vaccines for AIDS. J Med Primatol. 2005;34(5–6):303–12. doi: 10.1111/j.1600-0684.2005.00128.x. [DOI] [PubMed] [Google Scholar]
  6. Cohen J. AIDS research. Promising prevention interventions perform poorly in trials. Science. 2007;317(5837):440. doi: 10.1126/science.317.5837.440. [DOI] [PubMed] [Google Scholar]
  7. Connor RI, Montefiori DC, Binley JM, Moore JP, Bonhoeffer S, Gettie A, Fenamore EA, Sheridan KE, Ho DD, Dailey PJ, Marx PA. Temporal analyses of virus replication, immune responses, and efficacy in rhesus macaques immunized with a live, attenuated simian immunodeficiency virus vaccine. J Virol. 1998;72(9):7501–9. doi: 10.1128/jvi.72.9.7501-7509.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Daniel MD, Kirchhoff F, Czajak SC, Sehgal PK, Desrosiers RC. Protective effects of a live attenuated SIV vaccine with a deletion in the nef gene. Science. 1992;258:1938–1941. doi: 10.1126/science.1470917. [DOI] [PubMed] [Google Scholar]
  9. Egan MA, Charini WA, Kuroda MJ, Schmitz JE, Racz P, Tenner-Racz K, Manson K, Wyand M, Lifton MA, Nickerson CE, Fu T, Shiver JW, Letvin NL. Simian immunodeficiency virus (SIV) gag DNA-vaccinated rhesus monkeys develop secondary cytotoxic T-lymphocyte responses and control viral replication after pathogenic SIV infection. J Virol. 2000;74(16):7485–95. doi: 10.1128/jvi.74.16.7485-7495.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ferrantelli F, Kitabwalla M, Rasmussen RA, Cao C, Chou TC, Katinger H, Stiegler G, Cavacini LA, Bai Y, Cotropia J, Ugen KE, Ruprecht RM. Potent cross-group neutralization of primary human immunodeficiency virus isolates with monoclonal antibodies--implications for acquired immunodeficiency syndrome vaccine. J Infect Dis. 2004a;189(1):71–4. doi: 10.1086/380102. [DOI] [PubMed] [Google Scholar]
  11. Ferrantelli F, Rasmussen RA, Buckley KA, Li PL, Wang T, Montefiori DC, Katinger H, Stiegler G, Anderson DC, McClure HM, Ruprecht RM. Complete protection of neonatal rhesus macaques against oral exposure to pathogenic simian-human immunodeficiency virus by human anti-HIV monoclonal antibodies. J Infect Dis. 2004b;189(12):2167–73. doi: 10.1086/420833. [DOI] [PubMed] [Google Scholar]
  12. Gottlieb GS, Nickle DC, Jensen MA, Wong KG, Kaslow RA, Shepherd JC, Margolick JB, Mullins JI. HIV type 1 superinfection with a dual-tropic virus and rapid progression to AIDS: a case report. Clin Infect Dis. 2007;45(4):501–9. doi: 10.1086/520024. [DOI] [PubMed] [Google Scholar]
  13. Gray RH, Li X, Wawer MJ, Gange SJ, Serwadda D, Sewankambo NK, Moore R, Wabwire-Mangen F, Lutalo T, Quinn TC. Stochastic simulation of the impact of antiretroviral therapy and HIV vaccines on HIV transmission; Rakai, Uganda. Aids. 2003;17(13):1941–51. doi: 10.1097/00002030-200309050-00013. [DOI] [PubMed] [Google Scholar]
  14. Gray RH, Wawer MJ, Brookmeyer R, Sewankambo NK, Serwadda D, Wabwire-Mangen F, Lutalo T, Li X, vanCott T, Quinn TC. Probability of HIV-1 transmission per coital act in monogamous, heterosexual, HIV-1-discordant couples in Rakai, Uganda. Lancet. 2001;357(9263):1149–53. doi: 10.1016/S0140-6736(00)04331-2. [DOI] [PubMed] [Google Scholar]
  15. Harouse JM, Gettie A, Eshetu T, Tan RC, Bohm R, Blanchard J, Baskin G, Cheng-Mayer C. Mucosal transmission and induction of simian AIDS by CCR5-specific simian/human immunodeficiency virus SHIV(SF162P3) J Virol. 2001;75(4):1990–5. doi: 10.1128/JVI.75.4.1990-1995.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Harouse JM, Gettie A, Tan RC, Blanchard J, Cheng-Mayer C. Distinct pathogenic sequela in rhesus macaques infected with CCR5 or CXCR4 utilizing SHIVs. Science. 1999;284(5415):816–9. doi: 10.1126/science.284.5415.816. [DOI] [PubMed] [Google Scholar]
  17. Horton H, Vogel TU, Carter DK, Vielhuber K, Fuller DH, Shipley T, Fuller JT, Kunstman KJ, Sutter G, Montefiori DC, Erfle V, Desrosiers RC, Wilson N, Picker LJ, Wolinsky SM, Wang C, Allison DB, Watkins DI. Immunization of rhesus macaques with a DNA prime/modified vaccinia virus Ankara boost regimen induces broad simian immunodeficiency virus (SIV)-specific T-cell responses and reduces initial viral replication but does not prevent disease progression following challenge with pathogenic SIVmac239. J Virol. 2002;76(14):7187–202. doi: 10.1128/JVI.76.14.7187-7202.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Hsu M, Harouse JM, Gettie A, Buckner C, Blanchard J, Cheng-Mayer C. Increased mucosal transmission but not enhanced pathogenicity of the CCR5-tropic, simian AIDS-inducing simian/human immunodeficiency virus SHIV(SF162P3) maps to envelope gp120. J Virol. 2003;77(2):989–98. doi: 10.1128/JVI.77.2.989-998.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Jost S, Bernard MC, Kaiser L, Yerly S, Hirschel B, Samri A, Autran B, Goh LE, Perrin L. A patient with HIV-1 superinfection. N Engl J Med. 2002;347(10):731–6. doi: 10.1056/NEJMoa020263. [DOI] [PubMed] [Google Scholar]
  20. Kawada M, Tsukamoto T, Yamamoto H, Takeda A, Igarashi H, Watkins DI, Matano T. Long-term control of simian immunodeficiency virus replication with central memory CD4+ T-cell preservation after nonsterile protection by a cytotoxic T-lymphocyte-based vaccine. J Virol. 2007;81(10):5202–11. doi: 10.1128/JVI.02881-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Koff WC, Johnson PR, Watkins DI, Burton DR, Lifson JD, Hasenkrug KJ, McDermott AB, Schultz A, Zamb TJ, Boyle R, Desrosiers RC. HIV vaccine design: insights from live attenuated SIV vaccines. Nat Immunol. 2006;7(1):19–23. doi: 10.1038/ni1296. [DOI] [PubMed] [Google Scholar]
  22. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol. 1994;68(7):4650–5. doi: 10.1128/jvi.68.7.4650-4655.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Kumar A, Mukherjee S, Shen J, Buch S, Li Z, Adany I, Liu Z, Zhuge W, Piatak M, Jr, Lifson J, McClure H, Narayan O. Immunization of macaques with live simian human immunodeficiency virus (SHIV) vaccines conferred protection against AIDS induced by homologous and heterologous SHIVs and simian immunodeficiency virus. Virology. 2002;301(2):189–205. doi: 10.1006/viro.2002.1544. [DOI] [PubMed] [Google Scholar]
  24. Larke N, Murphy A, Wirblich C, Teoh D, Estcourt MJ, McMichael AJ, Roy P, Hanke T. Induction of human immunodeficiency virus type 1-specific T cells by a bluetongue virus tubule-vectored vaccine prime-recombinant modified virus Ankara boost regimen. J Virol. 2005;79(23):14822–33. doi: 10.1128/JVI.79.23.14822-14833.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Letvin NL, Mascola JR, Sun Y, Gorgone DA, Buzby AP, Xu L, Yang ZY, Chakrabarti B, Rao SS, Schmitz JE, Montefiori DC, Barker BR, Bookstein FL, Nabel GJ. Preserved CD4+ central memory T cells and survival in vaccinated SIV-challenged monkeys. Science. 2006;312(5779):1530–3. doi: 10.1126/science.1124226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Ling B, Veazey RS, Hart M, Lackner AA, Kuroda M, Pahar B, Marx PA. Early restoration of mucosal CD4 memory CCR5 T cells in the gut of SIV-infected rhesus predicts long term non-progression. Aids. 2007;21(18):2377–85. doi: 10.1097/QAD.0b013e3282f08b32. [DOI] [PubMed] [Google Scholar]
  27. Ling B, Veazey RS, Luckay A, Penedo C, Xu K, Lifson JD, Marx PA. SIV(mac) pathogenesis in rhesus macaques of Chinese and Indian origin compared with primary HIV infections in humans. Aids. 2002;16(11):1489–96. doi: 10.1097/00002030-200207260-00005. [DOI] [PubMed] [Google Scholar]
  28. Marthas ML, Miller CJ, Sutjipto S, Higgins J, Torten J, Lohman BL, Unger RE, Ramos RA, Kiyono H, McGhee JR, et al. Efficacy of live-attenuated and whole-inactivated simian immunodeficiency virus vaccines against vaginal challenge with virulent SIV. J Med Primatol. 1992;21(2–3):99–107. [PubMed] [Google Scholar]
  29. Mascola JR, Sambor A, Beaudry K, Santra S, Welcher B, Louder MK, Vancott TC, Huang Y, Chakrabarti BK, Kong WP, Yang ZY, Xu L, Montefiori DC, Nabel GJ, Letvin NL. Neutralizing antibodies elicited by immunization of monkeys with DNA plasmids and recombinant adenoviral vectors expressing human immunodeficiency virus type 1 proteins. J Virol. 2005;79(2):771–9. doi: 10.1128/JVI.79.2.771-779.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Mattapallil JJ, Douek DC, Buckler-White A, Montefiori D, Letvin NL, Nabel GJ, Roederer M. Vaccination preserves CD4 memory T cells during acute simian immunodeficiency virus challenge. J Exp Med. 2006;203(6):1533–41. doi: 10.1084/jem.20060657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature. 2005;434(7037):1093–7. doi: 10.1038/nature03501. [DOI] [PubMed] [Google Scholar]
  32. Montefiori D. Evaluating neutralizing antibodies against HIV, SIV and SHIV in luciferase reporter gene assays. In: Coligan KAJ, Margulies D, Shevach E, Strober W, Coico R, editors. Current protocols in immunology. John Wiley & Sons; New York, NY: 2004. [DOI] [PubMed] [Google Scholar]
  33. Montefiori DC, Baba TW, Li A, Bilska M, Ruprecht RM. Neutralizing and infection-enhancing antibody responses do not correlate with the differential pathogenicity of SIVmac239delta3 in adult and infant rhesus monkeys. J Immunol. 1996;157(12):5528–35. [PubMed] [Google Scholar]
  34. Mori K, Yasutomi Y, Ohgimoto S, Nakasone T, Takamura S, Shioda T, Nagai Y. Quintuple deglycosylation mutant of simian immunodeficiency virus SIVmac239 in rhesus macaques: robust primary replication, tightly contained chronic infection, and elicitation of potent immunity against the parental wild-type strain. J Virol. 2001;75(9):4023–8. doi: 10.1128/JVI.75.9.4023-4028.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Pahar B, Lackner AA, Veazey RS. Intestinal double-positive CD4+CD8+ T cells are highly activated memory cells with an increased capacity to produce cytokines. Eur J Immunol. 2006;36(3):583–92. doi: 10.1002/eji.200535520. [DOI] [PubMed] [Google Scholar]
  36. Pahar B, Li J, Rourke T, Miller CJ, McChesney MB. Detection of antigen-specific T cell interferon gamma expression by ELISPOT and cytokine flow cytometry assays in rhesus macaques. J Immunol Methods. 2003;282(1–2):103–15. doi: 10.1016/j.jim.2003.08.003. [DOI] [PubMed] [Google Scholar]
  37. Pahar B, Wang X, Dufour J, Lackner AA, Veazey RS. Virus-specific T cell responses in macaques acutely infected with SHIV(sf162p3) Virology. 2007;363(1):36–47. doi: 10.1016/j.virol.2007.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Reynolds MR, Weiler AM, Weisgrau KL, Piaskowski SM, Furlott JR, Weinfurter JT, Kaizu M, Soma T, Leon EJ, MacNair C, Leaman DP, Zwick MB, Gostick E, Musani SK, Price DA, Friedrich TC, Rakasz EG, Wilson NA, McDermott AB, Boyle R, Allison DB, Burton DR, Koff WC, Watkins DI. Macaques vaccinated with live-attenuated SIV control replication of heterologous virus. J Exp Med. 2008;205(11):2537–50. doi: 10.1084/jem.20081524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, Lifton MA, Racz P, Tenner-Racz K, Dalesandro M, Scallon BJ, Ghrayeb J, Forman MA, Montefiori DC, Rieber EP, Letvin NL, Reimann KA. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science. 1999;283(5403):857–60. doi: 10.1126/science.283.5403.857. [DOI] [PubMed] [Google Scholar]
  40. Smith DM, Wong JK, Hightower GK, Ignacio CC, Koelsch KK, Daar ES, Richman DD, Little SJ. Incidence of HIV superinfection following primary infection. Jama. 2004;292(10):1177–8. doi: 10.1001/jama.292.10.1177. [DOI] [PubMed] [Google Scholar]
  41. van der Kuyl AC, Kozaczynska K, van den Burg R, Zorgdrager F, Back N, Jurriaans S, Berkhout B, Reiss P, Cornelissen M. Triple HIV-1 infection. N Engl J Med. 2005;352(24):2557–9. doi: 10.1056/NEJM200506163522420. [DOI] [PubMed] [Google Scholar]
  42. Van Rompay KK, Singh RP, Brignolo LL, Lawson JR, Schmidt KA, Pahar B, Canfield DR, Tarara RP, Sodora DL, Bischofberger N, Marthas ML. The clinical benefits of tenofovir for simian immunodeficiency virus-infected macaques are larger than predicted by its effects on standard viral and immunologic parameters. J Acquir Immune Defic Syndr. 2004;36(4):900–14. doi: 10.1097/00126334-200408010-00003. [DOI] [PubMed] [Google Scholar]
  43. Veazey RS, Mansfield KG, Tham IC, Carville AC, Shvetz DE, Forand AE, Lackner AA. Dynamics of CCR5 expression by CD4(+) T cells in lymphoid tissues during simian immunodeficiency virus infection. J Virol. 2000a;74(23):11001–7. doi: 10.1128/jvi.74.23.11001-11007.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Veazey RS, Shattock RJ, Pope M, Kirijan JC, Jones J, Hu Q, Ketas T, Marx PA, Klasse PJ, Burton DR, Moore JP. Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat Med. 2003;9(3):343–6. doi: 10.1038/nm833. [DOI] [PubMed] [Google Scholar]
  45. Veazey RS, Tham IC, Mansfield KG, DeMaria M, Forand AE, Shvetz DE, Chalifoux LV, Sehgal PK, Lackner AA. Identifying the target cell in primary simian immunodeficiency virus (SIV) infection: highly activated memory CD4(+) T cells are rapidly eliminated in early SIV infection in vivo. J Virol. 2000b;74(1):57–64. doi: 10.1128/jvi.74.1.57-64.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Wang X, Das A, Lackner AA, Veazey RS, Pahar B. Intestinal double-positive CD4+CD8+ T cells of neonatal rhesus macaques are proliferating, activated memory cells and primary targets for SIVMAC251 infection. Blood. 2008;112(13):4981–90. doi: 10.1182/blood-2008-05-160077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Wang X, Rasmussen T, Pahar B, Poonia B, Alvarez X, Lackner AA, Veazey RS. Massive infection and loss of CD4+ T cells occurs in the intestinal tract of neonatal rhesus macaques in acute SIV infection. Blood. 2007;109(3):1174–81. doi: 10.1182/blood-2006-04-015172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Watkins DI. Basic HIV Vaccine Development. Top HIV Med. 2008;16(1):7–8. [PubMed] [Google Scholar]
  49. Watkins DI, Burton DR, Kallas EG, Moore JP, Koff WC. Nonhuman primate models and the failure of the Merck HIV-1 vaccine in humans. Nat Med. 2008;14(6):617–21. doi: 10.1038/nm.f.1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Wilson NA, Reed J, Napoe GS, Piaskowski S, Szymanski A, Furlott J, Gonzalez EJ, Yant LJ, Maness NJ, May GE, Soma T, Reynolds MR, Rakasz E, Rudersdorf R, McDermott AB, O’Connor DH, Friedrich TC, Allison DB, Patki A, Picker LJ, Burton DR, Lin J, Huang L, Patel D, Heindecker G, Fan J, Citron M, Horton M, Wang F, Liang X, Shiver JW, Casimiro DR, Watkins DI. Vaccine-induced cellular immune responses reduce plasma viral concentrations after repeated low-dose challenge with pathogenic simian immunodeficiency virus SIVmac239. J Virol. 2006;80(12):5875–85. doi: 10.1128/JVI.00171-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Wyand MS, Manson KH, Garcia-Moll M, Montefiori D, Desrosiers RC. Vaccine protection by a triple deletion mutant of simian immunodeficiency virus. J Virol. 1996;70(6):3724–33. doi: 10.1128/jvi.70.6.3724-3733.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES