Elsevier

Vaccine

Volume 27, Issue 32, 9 July 2009, Pages 4337-4349
Vaccine

Human immunodeficiency virus-like particles with consensus envelopes elicited broader cell-mediated peripheral and mucosal immune responses than polyvalent and monovalent Env vaccines

https://doi.org/10.1016/j.vaccine.2009.04.032Get rights and content

Abstract

Envelope (Env) sequences from human immunodeficiency virus (HIV) strains can vary by 15–20% within a single clade and as much as 35% between clades. Previous AIDS vaccines based upon a single isolate often could not elicit protective immune responses against heterologous viral challenges. In order to address the vast sequence diversity in Env sequences, consensus sequences were constructed for clade B and clade C envelopes and delivered to the mouse lung mucosa on the surface of virus-like particles (VLP). Consensus sequences decrease the genetic difference between the vaccine strain and any given viral isolate. The elicited immune responses were compared to a mixture of VLPs with Envs from primary viral isolates. This polyvalent vaccine approach contains multiple, diverse Envs to increase the breadth of epitopes recognized by the immune response and thereby increase the potential number of primary isolates recognized. Both consensus and polyvalent clade B Env VLP vaccines elicited cell-mediated immune responses that recognized a broader number of clade B Env peptides than a control monovalent Env VLP vaccine in both the systemic and the mucosal immune compartments. All three clade C Env vaccine strategies elicited similar responses to clade C peptides. However, both the consensus B and C Env VLP vaccines were more effective at eliciting cross-reactive cellular immune responses to epitopes in other clades. This is the first study to directly compare the breadth of cell-mediated immune responses elicited by consensus and polyvalent Env vaccines.

Introduction

The development of an effective AIDS vaccine has been hampered by the intrinsic diversity among circulating populations of HIV-1 in various geographical locations. There is a need to develop vaccines that can elicit enduring protective immunity to variant HIV-1 strains. Based on viral genetic distances and positions in phylogenetic trees, HIV-1 is divided into three separate groups: M, O, and N [1]. Group O infections are limited to central Africa and Group N infections are found in a small number of patients in Cameroon [2]. The vast majority of viral isolates are designated as Group M and they are responsible for the AIDS pandemic continuing worldwide. Due to the diversity in amino acid sequences between isolates within Group M, this group is further subdivided into nine subtypes or clades (A–D, F–H, J and K) [1]. The diversity between isolates in different clades can vary by as much as 35%.

The ultimate goal of an AIDS vaccine is to elicit potent cellular and humoral immune responses that will result in enduring, broadly protective immunity. Over the past 20 years, a number of potential AIDS vaccines have elicited immune responses that decreased viral set-points and maintained CD4+ T cells in vaccinated animals, but sterilizing immunity has not been achieved [3], [4], [5], [6]. Often, this limited protection occurs when the challenge virus matches the viral proteins in the vaccine [3], [4], [5], [6]. However, these artificial challenge models are not reflected in human infections and therefore the diversity of HIV-1 isolates requires vaccine designs that elicit broadly reactive immunity.

The greatest diversity is localized to the viral envelope glycoproteins. This may reflect the primary role in eliciting host immune recognition and responses that result in progressive evolution of the envelope proteins during persistent infection. Interestingly, while Env variation is widely assumed to be a major obstacle to AIDS vaccine development, there is very little experimental data in animal or human lentivirus systems addressing this critical issue. One method previously used to address Env variation in AIDS vaccines is a polyvalent vaccine strategy [4], [7], [8], [9], [10], [11]. These vaccines consist of a mixture of divergent isolates of the same antigen administered simultaneously. Polyvalent vaccines function by presenting a wide range of epitopes that cover a majority of individual strains. Vaccines for pneumococcus, poliovirus, and influenza virus have used this strategy [12]. Polyvalent vaccines increase the strength and breadth of humoral immune responses compared to monovalent vaccines [8], [9], [10], [13], [14], [15], [16], [17], [18]. The increase in the number of immunogens, below a certain threshold, does not result in immune interference [19].

More recently, centralized sequences have been developed as an alternative vaccine strategy to address viral sequence diversity. These artificial sequences are designed using computational methods to minimize the distance between a vaccine strain and a primary wild-type isolate. The three main strategies for developing centralized vaccines are center of the tree, ancestral, and consensus (for review see [20], [21]). Each of these designs has advantages for vaccine development. Consensus sequences minimize the degree of sequence dissimilarity between vaccine immunogens and circulating virus strains by creating artificial sequences based upon the most common amino acid in each position in an alignment [22], [23], [24], [25], [26], [27]. Recently, vaccine strategies utilizing consensus or ancestral HIV-1 Gag and Env sequences have proven potent inducers of CTL activity [23], [28], [29], [30], [31]. These consensus Envs form native structures and facilitate infection via the CCR5 coreceptor [32], [33].

In this study, monovalent, polyvalent and consensus Env vaccines were compared for the ability to elicit broadly reactive immune responses following vaccination in mice. Each set of Env vaccines was generated from clade B or clade C envelope sequences. HIV-1 virus-like particles (VLP) were used to deliver these Env sequences to the immune system in their native trimeric structure on the surface of a viral particle and the elicited immune responses were compared for the breadth of Env reactivity.

Section snippets

Envelope gene sequences

The wild-type HIV-1 subtypes B and C and consensus full-length env gene sequence were derived from the most common amino acids found at each location within the Env gene from over 200 isolates for each clade (Los Alamos National Laboratory; www.lanl.gov). Contemporary subtypes B and C env genes were cloned by polymerase chain reaction (PCR) amplification from Envgp160 plasmids obtained from the AIDS Reagent and Reference Program (National Institutes of Health). Primers were designed to insert a

Selection of wild-type and design of consensus B or C env gene sequences

The aim of this study was to compare the breadth of immune responses elicited by wild-type (individually or in a mixture) or consensus envelope sequences. The consensus (Con) B or C Env sequences were generated by selecting the most common amino acid at each position of a full-length Env alignment derived from 200 subtype B or C viruses deposited in the 2005 HIV Sequence Database. Each set of subtype sequences was supplemented with env sequences used in a standardized neutralizing panel [50],

Discussion

The intrinsic diversity among circulating populations of HIV-1 in various geographical locations is one of the greatest challenges for developing an effective AIDS vaccine. There is a great need to develop vaccines that can elicit enduring protective immunity to variant HIV-1 strains. While variation is observed in all of the viral proteins, the greatest diversity is localized to the viral envelope glycoproteins, evidently reflecting the predominant role of these proteins in eliciting host

References (70)

  • M. Roederer et al.

    Optimized determination of T cell epitope responses

    J Immunol Methods

    (2003)
  • S.P. McBurney et al.

    Membrane embedded HIV-1 envelope on the surface of a virus-like particle elicits broader immune responses than soluble envelopes

    Virology

    (2007)
  • E.T. Crooks et al.

    A comparative immunogenicity study of HIV-1 virus-like particles bearing various forms of envelope proteins, particles bearing no envelope and soluble monomeric gp120

    Virology

    (2007)
  • P.B.E.S. Brandtzaeg et al.

    Regional specialization in the mucosal immune systen: what happens in the microcompartments?

    Immunology Today

    (1999)
  • H.B.I. Kaneko et al.

    Oral DNA vaccination promotes mucosal and systemic immune responses to HIV envelope glycoprotein

    Virology

    (2000)
  • J.R.M. McGhee

    J. In defense of mucosal surfaces: development of novel vaccines for IgA responses protective at the portals of entry of microbial pathogens

    Infect Dis Clin N Am

    (1990)
  • K. Tochikubo et al.

    Recombinant cholera toxin B subunit acts as an adjuvant for the mucosal and systemic responses of mice to mucosally co-administered bovine serum albumin

    Vaccine

    (1998)
  • D. Verthelyi et al.

    Immunoregulatory activity of CpG oligonucleotides in humans and nonhuman primates

    Clin Immunol

    (2003)
  • S.P. McBurney et al.

    Membrane embedded HIV-1 envelope on the surface of a virus-like particle elicits broader immune responses than soluble envelopes

    Virology

    (2007)
  • Q. Yao et al.

    Intranasal immunization with SIV virus-like particles (VLPs) elicits systemic and mucosal immunity

    Vaccine

    (2002)
  • D.L. Robertson et al.

    A Reference Guide to HIV-1 Classification

  • P.W. Berman et al.

    Protection of chimpanzees from infection by HIV-1 after vaccination with recombinant glycoprotein gp120 but not gp160

    Nature

    (1990)
  • M.W. Cho et al.

    Polyvalent envelope glycoprotein vaccine elicits a broader neutralizing antibody response but is unable to provide sterilizing protection against heterologous simian/human immunodeficiency virus infection in pigtailed macaques

    J Virol

    (2001)
  • P. Polacino et al.

    Limited breadth of the protective immunity elicited by simian immunodeficiency virus SIVmne gp160 vaccines in a combination immunization regimen

    J Virol

    (1999)
  • A. Azizi et al.

    Immunogenicity of a polyvalent HIV-1 candidate vaccine based on fourteen wild type gp120 proteins in golden hamsters

    BMC Immunol

    (2006)
  • E.J.H. Rollman et al.

    Multi-subtype gp160 DNA immunization induces broadly neutralizing anti-HIV antibodies

    Gene Ther

    (2004)
  • K. Slobod et al.

    HIV vaccine rationale, design and testing

    Curr HIV Res

    (2005)
  • W.-P. Kong et al.

    Immunogenicity of multiple gene and clade human immunodeficiency virus type 1 DNA vaccines

    J Virol

    (2003)
  • Lemiale F.B. Denys et al.

    Immunogenicity of recombinant envelope glycoproteins derived from T-Cell line-adapted isolates or primary HIV isolates: a comparative study using multivalent vaccine approaches

    JAIDS J Acquir Immune Defic Syndr

    (2001)
  • R. Pal et al.

    Polyvalent DNA prime and envelope protein boost HIV-1 vaccine elicits humoral and cellular responses and controls plasma viremia in rhesus macaques following rectal challenge with an R5 SHIV isolate

    J Med Primatol

    (2005)
  • M.S. Seaman et al.

    Multiclade human immunodeficiency virus Type 1 envelope immunogens elicit broad cellular and humoral immunity in rhesus monkeys

    J Virol

    (2005)
  • K.S. Slobod et al.

    Subcutaneous administration of a recombinant vaccinia virus vaccine expressing multiple envelopes of HIV-1

    Eur J Clin Microbiol Infect Dis

    (2004)
  • S. Lu

    Immunogenicity of Monovalent and Polyvalent Dna Prime/Protein Boost Formulation Encoding Primary Hiv-1 Env Antigens as Tested in Rabbits and Non-Human Primate Models

    (2004)
  • S.P. McBurney et al.

    Developing broadly reactive HIV-1/AIDS vaccines: a review of polyvalent and centralized HIV-1 vaccines

    Curr Pharm Des

    (2007)
  • H-XL Feng Gao et al.

    Centralized HIV-1 envelope immunogens and neutralizing antibodies

    Curr HIV Res

    (2007)
  • Cited by (29)

    • In vivo electroporation in DNA-VLP prime-boost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses

      2017, Vaccine
      Citation Excerpt :

      Previously we developed a novel system to produce HIV-1 VLP using stably transfected Drosophila S2 cell clones [40]. We demonstrated that the amount of HIV-1 VLP produced by S2 cell clones is comparable to that produced by insect cells infected with recombinant baculoviruses (RB) [41] and much higher than those produced by mammalian cells [42–47]. Unlike HIV-1 VLP produced by insect cells infected with RB, culture supernatants produced by stably transfected S2 cell clones are not contaminated with recombinant viruses, which makes downstream purification process simpler.

    • Immunization with a consensus epitope from human papillomavirus L2 induces antibodies that are broadly neutralizing

      2014, Vaccine
      Citation Excerpt :

      In contrast, the less well-conserved C-terminal region of this peptide is likely a type-specific neutralizing epitope. There are several examples of vaccines that use viral consensus sequences to successfully to induce antibodies with broader specificities [38–41]. To test whether this strategy would work against a region of HPV L2 that is a type-specific neutralizing epitope, we synthesized a peptide that represented the consensus sequence of aa65–85 from 12 high-risk HPV types and as 3 low-risk types (Fig. 4A).

    • Highly potent delivery method of gp160 envelope vaccine combining lentivirus-like particles and DNA electrotransfer

      2012, Journal of Controlled Release
      Citation Excerpt :

      Different VLP vaccination routes have been studied and it was demonstrated that intradermal immunization might be a preferable route for studies involving VLPs [26]. The VLP doses have also been studied and range between 20 and 100 μg [26–28]. Concerning DNA vaccines several delivery methods emerged in the past few years and electrotransfer appeared as one of the most efficient.

    • Human immunodeficiency virus (HIV) immunopathogenesis and vaccine development: A review

      2011, Vaccine
      Citation Excerpt :

      Fully synthetic genes derived from theoretically defined consensus or ancestral HIV envelope sequences [421–423], that were found to be superior to many wild-type immunogens for inducing cross-subtype T- and B-cell immune responses and antibodies that neutralized a spectrum of HIV Env pseudoviruses [424]. VLPs with consensus clade B or clade C Env sequences delivered to the mouse lung mucosa were found to elicit a broadened cell-mediated immune response [425]. An M-group CON-S and a clade B-based CON-B Env protein each elicited antibodies with good titers and breadth against tier-1 HIV-1 viruses [426], but not against isolates that are more difficult to neutralize (for a review, see [299]).

    View all citing articles on Scopus
    View full text