Elsevier

Vaccine

Volume 29, Issue 16, 5 April 2011, Pages 2918-2932
Vaccine

A combination HIV vaccine based on Tat and Env proteins was immunogenic and protected macaques from mucosal SHIV challenge in a pilot study

https://doi.org/10.1016/j.vaccine.2011.02.006Get rights and content

Abstract

HIV native Tat and V2 loop-deleted Env (EnvΔV2) proteins already proved safe and immunogenic in phase I clinical testing as single vaccine components. Further, a phase II vaccine trial with Tat showed intensification of the therapeutic effects of HAART in successfully treated HIV-infected individuals. Here a pilot study assessed the immunogenicity and protective efficacy of an HIV/AIDS vaccine based on the combination of Tat and EnvΔV2 proteins in cynomolgus macaques against homologous intrarectal challenge with 35 MID50 (monkey infectious dose 50) of an R5 simian-human immunodeficiency virus (SHIVSF162P4cy).

Upon challenge, three of four macaques immunized with Tat and EnvΔV2, and two of three monkeys immunized with EnvΔV2 alone were protected from infection. In contrast, all three control animals, which had been either administered with the adjuvants only or left untreated, and an additional monkey immunized with Tat alone became systemically infected. Protection of the macaques vaccinated with EnvΔV2 or Tat/EnvΔV2 correlated with higher peak titers of pre-challenge neutralizing antibodies obtained during the immunization period (between 70 and 3 weeks before challenge) and with anti-Env V3 loop binding antibodies assessed 3 weeks before challenge.

Compared to EnvΔV2 alone, the Tat and EnvΔV2 combined vaccine elicited faster antibody responses (IgM) with a trend, early in the vaccination schedule, after the second immunization including EnvΔV2, towards broader anti-Env IgG epitope specificity and a higher ratio of neutralizing to Env-binding antibody titers. As the number of immunizations increased, vaccination with EnvΔV2 approached the immune response assessed after two inocula with the Tat/EnvΔV2 combined vaccine, even though some differences remained between groups, as indicated by anti-Env IgG epitope mapping. In fact, three weeks before challenge, plasma IgG of animals in the EnvΔV2 group showed a trend towards stronger specificity for the V1 loop and V5 loop-C5 regions of Env, whereas the Tat/EnvΔV2 group displayed an overall higher reactivity for epitopes within the Env V3 loop throughout the immunization period.

Although differences in terms of protection rate were not found between the EnvΔV2 or Tat/EnvΔV2 vaccination groups in this pilot study, vaccination with Tat/EnvΔV2 appeared to accelerate the induction of potentially protective antibody responses to Env. In particular, antibodies to the Env V3 loop, whose levels at pre-challenge correlated with protection, were already higher early in the vaccination schedule in monkeys immunized with Tat/EnvΔV2 as compared to EnvΔV2 alone.

Further studies including larger vaccination groups and fewer immunizations with these two vaccine candidates are needed to confirm these findings and to assess whether the Tat/EnvΔV2 vaccine may afford superior protection against infection.

Introduction

Currently, HIV/AIDS vaccine candidates based on the combination of structural and regulatory HIV proteins may hold the best promise for developing a protective vaccine [1]. In fact, the overall disappointing results of HIV/AIDS vaccine phase IIb/III clinical trials conducted to date [2], [3] can be, at least in part, ascribed to the inadequacy of classical vaccine strategies based on HIV structural antigens (Env, and/or Gag, and Pol), which failed to provide protection [4]. On the other hand, less traditional vaccine approaches with HIV regulatory proteins (Tat, Rev, and Nef) were able to contain virus replication and preventing disease onset and progression in preclinical studies [1], [4]. More recently, novel vaccine modalities encompassed both structural and regulatory HIV proteins to generate antibody (Ab) and cell-mediated immunity against multiple HIV components that are key for HIV entry/replication. Such combined vaccines are intended to block/control early and late HIV infection-related events, to contain acute virus infection and/or protect the host from disease progression [1], [4], [5]. The exploitation of HIV regulatory proteins’ immunomodulatory properties [6], [7] represents an added value of this combined vaccine approach [1].

We describe here the rational design and evaluation in macaques of an innovative HIV/AIDS vaccine based on the combination of HIV Tat and Env proteins, aimed at inducing protective immunity capable of neutralizing the virus as well as Tat biological activities [8], [9], [10], [11]. In particular, this study was conceived as a pilot study to evaluate the effects of native Tat protein addition to an Env-based vaccine. To this aim, the two pivotal experimental groups consisted of macaques immunized with Env only or with Tat and Env. Control groups included animals inoculated with the adjuvants only or left untreated (naïve), and one animal vaccinated with the native Tat protein alone.

An oligomeric, SF162 strain-derived, V2 loop-deleted Env (EnvΔV2) was chosen for its capacity of eliciting neutralizing antibodies (nAbs) against primary HIV isolates [12]. The deletion of the V2 loop allows exposure of conserved neutralization-sensitive epitopes to increase the breadth of vaccine-elicited Ab responses, thus potentially circumventing the issue of HIV Env intra- and inter-clade variability [13].

The inclusion of biologically active, HIIIB strain-derived Tat protein in the vaccine regimen was intended to generate immune responses against the early viral product Tat and to exploit Tat immunomodulatory properties.

Tat is key for virus replication, cell-to-cell virus transmission, and HIV pathogenicity, and can enter both infected and uninfected cells, modulating the expression of cellular genes [14]. Biologically active Tat promotes monocyte-derived dendritic cells maturation towards a Th-1 polarizing phenotype as well as their antigen-presenting activity, leading to a more efficient presentation of both allogeneic and exogenous soluble antigens, in vitro [15], [16]. By modulating the composition and activity of the immunoproteasome, Tat changes the hierarchy of CTL epitopes in favor of subdominant and cryptic ones and shows vaccine-adjuvant properties [17], [18], [19], [20].

Tat is highly conserved in its immunodominant domains representing, in principle, an ideal target for a broadly effective vaccine [21]. Because of its multiple functions, immune responses against biologically active Tat can contribute to the control of HIV infection and/or disease progression. Of note, anti-Tat cellular or humoral immunity was reported to correlate, in the course of natural infection, with early virus control [22], [23] or with asymptomatic infection and long-term non-progression to AIDS [24], [25], respectively. Furthermore, a retrospective analysis conducted on 112 cynomolgus macaques indicated that vaccination with the biologically active Tat protein reduced the rate of infection acquisition against challenge with 10 MID50 of pathogenic SHIV89.6P, contained acute CD4+ T cell depletion against 15 MID50 of virus, and contained CD4+ T cell loss in the chronic phase of infection, regardless of the challenge dose [26]. Recently, the HIV Tat vaccine has shown to be safe and immunogenic in preventive and therapeutic phase I trials [27], [28] and to intensify the therapeutic effects of HAART in successfully treated HIV-infected individuals, making Tat a promising target of an HIV/AIDS vaccine [29].

The experimental design of this pilot study consisted of multiple subcutaneous and intranasal immunizations with proteins in adjuvants, followed by assessment of immune responses at both the systemic and the mucosal levels. The Alum adjuvant (aluminum phosphate), which is commonly used in licensed human vaccines [30], was employed for subcutaneous immunizations, whereas adjuvant LT-K63 was given intranasally [31]. LT-K63 is an Escherichia coli heat-labile enterotoxin mutant, which proved generally safe and effective as an intranasal adjuvant both in animals and in humans [31], [32], [33]. In particular, multiple LT-K63 intranasal administrations in animals did not induce histological inflammatory changes in the respiratory tract or olfactory bulbs and in the meninges [31]. When co-administered with a large number of immunogens by different routes in mice, LT-K63 was able to enhance immune responses [31]. In female rhesus macaques immunized with the HIV Env protein and LT-K63 adjuvant either intramuscularly, or intramuscularly and intranasally, protection was achieved against intravaginal simian-human immunodeficiency virus (SHIV) challenge [33]. LT-K63 also showed an overall good safety profile and intranasal adjuvanticity for influenza in humans [32]. Nonetheless, more recently (after the completion of our monkey study), two phase I clinical trials [34] on nasal subunit vaccines against HIV and tuberculosis employing LT-K63 as an adjuvant confirmed previous concerns on the association of LT-K63-containing nasal influenza vaccine and Bell's palsy [35].

Efficacy of the Tat/EnvΔV2 or EnvΔV2 alone vaccines was evaluated against homologous intrarectal challenge with an R5 SHIV.

Section snippets

Macaques housing, immunizations, and virus challenge

The male Mauritian cynomolgus monkeys (Macaca fascicularis) employed in this study, negative for simian immunodeficiency virus (SIV), STLV-1, simian type-D retroviruses, and simian Herpes B virus infections, were housed at the National AIDS Center, Istituto Superiore di Sanità (ISS), according to the European guidelines for non-human primate care (ECC, Directive No. 86-609, Nov. 24, 1986). Animal experiments were approved by the Quality and Safety Committee for Animal Trials of the ISS. All

Anti-Tat binding antibodies and epitope mapping

All Tat-containing vaccine regimens elicited bAb responses against Tat, with peak titers ranging between 12,800 and 25,600. Two immunizations with Tat were sufficient to generate measurable anti-Tat IgM (Fig. 2a) and IgG (Fig. 2b) in all vaccinees. Before challenge, low-titer (titer: 25), Tat-specific IgA were found in the plasma of 3 (animals AC252, AC017, and AC259) of the 5 monkeys immunized with Tat or Tat and EnvΔV2 combined, respectively (data not shown).

Pre-challenge mucosal anti-Tat IgG

Discussion

In this study we showed that a vaccine based on the combination of HIV EnvΔV2 and native Tat proteins generated more rapid IgM responses and broader IgG epitope specificity than EnvΔV2 alone. Protection from mucosal infection was achieved in monkeys vaccinated with the Tat/EnvΔV2 combined vaccine or with EnvΔV2. These findings extend upon those reported using the EnvΔV2 protein alone, where vaccine protection was also observed in a somewhat different model in which female rhesus macaques were

Acknowledgements

The authors thank Piergiorgio Pupino Carbonelli and the monkey facility staff of the National AIDS Center (Istituto Superiore di Sanità) for animal care and for all animal procedures, Claudia Rovetto for performing Western blot assays, Iole Macchia, Michela Sabbatucci, and Giulia Cencioni for assistance with the ELISpot assays, Pasqualina Leone, Gaia Sciaranghella, and Zuleika Michelini for help with the assessment of Ab responses, Domenico Fulgenzi, Daniela Compagnoni, Viviana Buffa, Martina

References (76)

  • C. Buckner et al.

    Priming B cell-mediated anti-HIV envelope responses by vaccination allows for the long-term control of infection in macaques exposed to a R5-tropic SHIV

    Virology

    (2004)
  • M.T. Maggiorella et al.

    Long-term protection against SHIV89.6P replication in HIV-1 Tat vaccinated cynomolgus monkeys

    Vaccine

    (2004)
  • O. Longo et al.

    Phase I therapeutic trial of the HIV-1 Tat protein and long term follow-up

    Vaccine

    (2009)
  • V.R. Polonis et al.

    Recent advances in the characterization of HIV-1 neutralization assays for standardized evaluation of the antibody response to infection and vaccination

    Virology

    (2008)
  • R.H. Florese et al.

    Comparative study of Tat vaccine regimens in Mauritian cynomolgus and Indian rhesus macaques: influence of Mauritian MHC haplotypes on susceptibility/resistance to SHIV(89.6P) infection

    Vaccine

    (2008)
  • M.P. Girard et al.

    HIV/AIDS vaccines: a need for new concepts?

    Int Rev Immunol

    (2008)
  • G.P. Bansal et al.

    Future paths for HIV vaccine research: exploiting results from recent clinical trials and current scientific advances

    Curr Opin Mol Ther

    (2010)
  • N.L. Letvin et al.

    Prospects for vaccine protection against HIV-1 infection and AIDS

    Annu Rev Immunol

    (2002)
  • T. Demberg et al.

    A replication-competent adenovirus-human immunodeficiency virus (Ad-HIV) tat and Ad-HIV env priming/Tat and envelope protein boosting regimen elicits enhanced protective efficacy against simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques

    J Virol

    (2007)
  • B. Ensoli et al.

    Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation

    J Virol

    (1993)
  • H.K. Chang et al.

    Regulation of cellular gene expression and function by the human immunodeficiency virus type 1 Tat protein

    J Biomed Sci

    (1995)
  • B. Ensoli et al.

    Candidate HIV-1 Tat vaccine development: from basic science to clinical trials

    AIDS

    (2006)
  • S.W. Barnett et al.

    The ability of an oligomeric human immunodeficiency virus type 1 (HIV-1) envelope antigen to elicit neutralizing antibodies against primary HIV-1 isolates is improved following partial deletion of the second hypervariable region

    J Virol

    (2001)
  • I.K. Srivastava et al.

    Changes in the immunogenic properties of soluble gp140 human immunodeficiency virus envelope constructs upon partial deletion of the second hypervariable region

    J Virol

    (2003)
  • G. Barillari et al.

    Angiogenic effects of extracellular human immunodeficiency virus type 1 Tat protein and its role in the pathogenesis of AIDS-associated Kaposi's sarcoma

    Clin Microbiol Rev

    (2002)
  • E. Fanales-Belasio et al.

    Native HIV-1 Tat protein targets monocyte-derived dendritic cells and enhances their maturation, function, and antigen-specific T cell responses

    J Immunol

    (2002)
  • E. Fanales-Belasio et al.

    HIV-1 Tat addresses dendritic cells to induce a predominant Th1-type adaptive immune response that appears prevalent in the asymptomatic stage of infection

    J Immunol

    (2009)
  • R. Gavioli et al.

    HIV-1 tat protein modulates the generation of cytotoxic T cell epitopes by modifying proteasome composition and enzymatic activity

    J Immunol

    (2004)
  • A.L. Remoli et al.

    Intracellular HIV-1 Tat protein represses constitutive LMP2 transcription increasing proteasome activity by interfering with the binding of IRF-1 to STAT1

    Biochem J

    (2006)
  • S. Buttò et al.

    Sequence conservation and antibody cross-recognition of clade B human immunodeficiency virus (HIV) type 1 Tat protein in HIV-1-infected Italians, Ugandans, and South Africans

    J Infect Dis

    (2003)
  • C.A. Van Baalen et al.

    Kinetics of antiviral activity by human immunodeficiency virus type 1-specific cytotoxic T lymphocytes (CTL) and rapid selection of CTL escape virus in vitro

    J Virol

    (1998)
  • J. Cao et al.

    Comprehensive analysis of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-secreting CD8+ T cells in primary HIV-1 infection

    J Virol

    (2003)
  • G. Rezza et al.

    The presence of anti-Tat antibodies is predictive of long-term nonprogression to AIDS or severe immunodeficiency: findings in a cohort of HIV-1 seroconverters

    J Infect Dis

    (2005)
  • A. Cafaro et al.

    Impact of viral dose and major histocompatibility complex class IB haplotype on viral outcome in mauritian cynomolgus monkeys vaccinated with Tat upon challenge with simian/human immunodeficiency virus SHIV89.6P

    J Virol

    (2010)
  • B. Ensoli et al.

    The therapeutic phase I trial of the recombinant native HIV-1 Tat protein

    AIDS

    (2008)
  • B. Ensoli et al.

    Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART

    PLoS One

    (2010)
  • S.L. Hem et al.

    Relationship between physical and chemical properties of aluminum-containing adjuvants and immunopotentiation

    Expert Rev Vaccines

    (2007)
  • S. Peppoloni et al.

    Mutants of the Escherichia coli heat-labile enterotoxin as safe and strong adjuvants for intranasal delivery of vaccines

    Expert Rev Vaccines

    (2003)
  • Cited by (17)

    • Impact of antibody quality and anamnestic response on viremia control post-challenge in a combined Tat/Env vaccine regimen in rhesus macaques

      2013, Virology
      Citation Excerpt :

      The low-titers and poor quality of antibody responses suggest the adjuvant used may have been suboptimal. The change to use of alum adjuvant was made in order to correspond to an earlier Tat plus Env combination study in cynomolgus macaques (Ferrantelli et al., 2011), and to match a future Tat plus Env vaccine trial. Alum has long been used as an adjuvant and promotes immune responses independently of toll-like-receptor (TLR) mechanisms (De Gregorio et al., 2009).

    • Production of human antibodies by in vitro immunization using a fusion protein containing the transcriptional transactivator of HIV-1

      2013, Journal of Immunological Methods
      Citation Excerpt :

      The Tat protein was selected because of several biological effects linked to the immune response (Albini et al., 1998; Buonaguro et al., 1992; Fanales-Belasio et al., 2002; Izmailova et al., 2003), further suggesting that Tat may contribute to the triggering of the immune response. Tat was shown to raise an immune response in the absence of adjuvant (Kittiworakarn et al., 2006; Gadzinski et al., 2012) in mouse and this property could be used for a human vaccination protocol (Ensoli et al., 2009; Ferrantelli et al., 2011). Tat can also bind heparan sulfate proteoglycans, thus increasing the ability of an antigen to stimulate T cells (Léonetti et al., 2010).

    View all citing articles on Scopus
    View full text