Elsevier

Vaccine

Volume 27, Issue 47, 5 November 2009, Pages 6605-6612
Vaccine

Induction of HIV-1 subtype B and AE-specific neutralizing antibodies in mice and macaques with DNA prime and recombinant gp140 protein boost regimens

https://doi.org/10.1016/j.vaccine.2009.08.016Get rights and content

Abstract

We developed highly expressing clade B and AE DNA and envelope protein (Env) vaccines for evaluation in mice and macaques as DNA prime/protein boost regimens. High levels of Env-specific antibodies were induced in mice, albeit with limited neutralizing activity in vitro. A combined clade B and AE regimen induced high titer Env-specific antibody in two pigtail macaques that neutralized several strains of HIV-1. However, upon mucosal challenge with SHIVSF162P3 no protection from infection was observed. Although the vaccines tested provide a platform for inducing robust humoral immunity, further refinements to broaden coverage against divergent strains and induce mucosal immunity are needed.

Introduction

Despite being the focus of much research effort, an effective vaccine to prevent HIV infection or ameliorate disease outcome after infection remains elusive. Env is the major viral target for neutralization by antibody. However Env has high sequence variability, extensive glycosylation and many of the more conserved Env structures are recessed, protecting the virus from neutralization. The first phase III clinical trial of an HIV vaccine used monomeric gp120 protein with the aim of generating neutralizing antibodies (nAb) to prevent infection. This vaccine was not protective [1], and it is now accepted that gp120 monomers induce only narrow nAb responses, primarily against CXCR4-tropic laboratory strains. Recently, efforts have focused on the potential of cytotoxic T lymphocyte (CTL) responses to control viremia and lower viral set point during chronic infection, thus preventing or delaying disease progression. Disappointingly, while a number of studies demonstrated the ability of vaccine-generated CTL responses to prevent or delay disease progression in non-human primate models, an adenoviral vector-based phase III clinical trial (STEP trial) recently failed to achieve these objectives [2]. Similarly, a previous approach from our group using a DNA prime and recombinant fowlpox virus boost failed to elicit significant adaptive cellular immune responses in human subjects [3]. In that study, a major truncation of Env (at amino acid 275) abrogated its ability to elicit anti-Env antibodies in macaques [4]. In the studies reported here we have tested immunogens with VLP-associated full-length Env and soluble Env analogues including the entire ectodomain (amino acids 1–683). VLP immunogens are recognized and taken up by antigen presenting cells, facilitating the generation of CD4 help and B-cell priming, whereas soluble immunogens can efficiently boost immune responses. VLP-producing plasmid DNA immunogens or purified VLP particles administered to macaques in the absence of subsequent heterologous boosting were able to mediate viral load reduction after homologous intravenous challenge, but not prevent infection [5], [6].

We have focused on generating broader humoral immune responses to Env. Env derived from both clade B and circulating recombinant form 01 AE (AE) strains were assessed in an attempt to increase the breadth of the immune responses elicited and tailor the vaccine for Thailand and other South East Asian countries where clade B and AE viruses are predominant. We tested our approach firstly in a murine model and then in a pilot study using pig-tailed macaques. Env was presented in DNA prime/trimeric protein boost regimens, as this has proven useful in eliciting strong and durable responses against Env and other immunogens, as reviewed by Lu [7]. DNA priming was designed to present Env in two different configurations: on the surface of virus like particles (VLP) and as a soluble Env analogue lacking the transmembrane and cytoplasmic domains (gp140). Our strategy elicited robust serum antibody responses that in the case of pig-tailed macaques showed some breadth of neutralization. However, immunization did not prevent infection after intrarectal challenge with a simian/human immunodeficiency virus expressing a heterologous HIV-1 Env (SHIVSF162P3), suggesting that eliciting broader immune responses in the mucosal compartment may be critical in future vaccine strategies.

Section snippets

DNA immunogens

The VLP-encoding plasmid pVLP was constructed by replacing the gag and pol genes of the plasmid pHIS-HIV-B [4] with those of pHIS-HIV-AE [8] using the restriction sites NotI and EcoRI, and restoring the truncated Env coding frame using an Nde1/AvrII fragment from the wild-type AD8 env gene [9] (Fig. 1A). To make plasmid vaccine vectors for the expression of soluble Env analogues, a BssHII/BlpI fragment of pDRenv2 [10] spanning the NL4.3 env gene was cloned into the same sites of pHIS-HIV-B,

In vitro expression from DNA vaccine vectors

A series of DNA and protein vaccines were designed to present native Env trimers and elicit nAb. The expression cassette of pVLP (Fig. 1A) retains approximately two-thirds of the proviral sequence. Multiple mutations or deletions of the gag, pol and nef coding regions and complete removal of the vpr and vif genes and both long terminal repeat sequences (all derived from the pHIS-HIV-B and pHIS-HIV-AE parental plasmids) ensured maximal safety. The expression cassettes of pVLP (Fig. 1A), pAD8-140

Discussion

In this study, we tested several modes of Env-based vaccine delivery in murine and non-human primate models. All plasmid vectors were tested in vitro by transfection of a human cell line, and high-level Env expression was confirmed. The VLP-encoding construct was very efficient at mediating secretion of VLP with high levels of incorporation of cleaved gp160, similar to that of virion-associated Env [20] indicating the potential utility of this vaccine platform. The murine system allowed us to

Acknowledgements

We thank Dr. Robert De Rose, Thakshila Amarasena, Caroline Fernandez, Jeanette Reece, Roberta Goli and Leanne Smith for expert technical assistance. This work was supported financially by the Australian NHMRC (Program Grant 299907 and Career Development Award 359233 to RJC) and the Australian Centre for HIV and Hepatitis Virology Research. The following reagents were obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH: pSVIII-93.TH976.17 (for

References (39)

  • M.S. Seaman et al.

    Standardized assessment of NAb responses elicited in rhesus monkeys immunized with single- or multi-clade HIV-1 envelope immunogens

    Virology

    (2007)
  • N.M. Flynn et al.

    Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection

    J Infect Dis

    (2005)
  • A.D. Kelleher et al.

    A randomized, placebo-controlled phase I trial of DNA prime, recombinant fowlpox virus boost prophylactic vaccine for HIV-1

    Aids

    (2006)
  • W. Akahata et al.

    DNA vaccination of macaques by a full-genome simian/human immunodeficiency virus type 1 plasmid chimera that produces non-infectious virus particles

    J Gen Virol

    (2003)
  • S. Lu

    Combination DNA plus protein HIV vaccines

    Springer Semin Immunopathol

    (2006)
  • T.S. Theodore et al.

    Construction and characterization of a stable full-length macrophage-tropic HIV type 1 molecular clone that directs the production of high titers of progeny virions

    AIDS Res Hum Retroviruses

    (1996)
  • J.L. Anderson et al.

    Both linear and discontinuous ribosome scanning are used for translation initiation from bicistronic human immunodeficiency virus type 1 Env mRNAs

    J Virol

    (2007)
  • F. Gao et al.

    Molecular cloning and analysis of functional envelope genes from human immunodeficiency virus type 1 sequence subtypes A through G

    J Virol

    (1996)
  • R.J. Center et al.

    The human immunodeficiency virus type 1 gp120 V2 domain mediates gp41-independent intersubunit contacts

    J Virol

    (2000)
  • Cited by (38)

    • Broad and ultra-potent cross-clade neutralization of HIV-1 by a vaccine-induced CD4 binding site bovine antibody

      2022, Cell Reports Medicine
      Citation Excerpt :

      Avi-Tagged trimers were biotinylated using BirA enzyme according to manufacturer’s instructions (Avidity, LLC). Monomeric AD8 gp120 was also produced as described previously.60–62 BG505 Unc.

    • Prime-Boost Vaccination: Impact on the HIV-1 Vaccine Field

      2015, Novel Approaches and Strategies for Biologics, Vaccines and Cancer Therapies
    • The magnitude of HIV-1 resistance to the CCR5 antagonist maraviroc may impart a differential alteration in HIV-1 tropism for macrophages and T-cell subsets

      2013, Virology
      Citation Excerpt :

      All Envs were cloned into the pSVIII-Env expression vector (Gao et al., 1996). Env-pseudotyped, GFP reporter viruses were produced by transfection of 293 T cells with pNL4-3Env-GFP (Center et al., 2009) and pSVIII-Env plasmids using Lipofectamine 2000 (Invitrogen) at a ratio of 4:1. Supernatants were harvested 48 h later and filtered through 0.45 µm filters.

    • Utility of the Sindbis replicon system as an Env-targeted HIV vaccine

      2013, Vaccine
      Citation Excerpt :

      RNA (10 μg/dose) in RNase free water was immunized i.m. DNA and RNA immunogens directed the expression of full length Env gp160. Soluble HIV-1NL4.3 Env (gp140) lacking the transmembrane domain [31] was formulated with Freund's incomplete adjuvant (FIA) or Freund's complete adjuvant (FCA) (both Sigma) as indicated for protein immunizations. 5 μg of gp140 protein in PBS was administered by the i.p. route and 5 μg was administered by the sub-cutaneous (s.c.) route simultaneously.

    • Role of monocytes in mediating HIV-specific antibody-dependent cellular cytotoxicity

      2012, Journal of Immunological Methods
      Citation Excerpt :

      The RFADCC assay was performed as previously described (Gómez-Román et al., 2006). Briefly, 1 × 106 CEM.NKr-CCR5 cells in 100 μl of RF10 medium were coated with 3 μg of purified HIVAD8 gp140 (clade B) (production of gp140 described elsewhere (Center et al., 2009)) for 1 h at room temperature. Uncoated CEM.NKr-CCR5 cells were treated identically but without adding gp140 protein.

    View all citing articles on Scopus
    View full text