Transcript
Page 1: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

1591

Review

ISSN 1743-588910.2217/NNM.12.131 © 2012 Future Medicine Ltd Nanomedicine (2012) 7(10), 1591–1610

Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by in vivo targeting to dendritic cells

BackgroundHighly active antiretroviral treatment (HAART) inhibits the replication of HIV and slows down disease progression, thus keeping viral RNA levels very low [1]. However, alternative treat-ment strategies are required as there are still a number of unresolved problems with HAART. These include high cost, persistent low-grade viral replication, incomplete normalization of T-cell functions, drug-related side effects (lipid metabolism problems: lipodystrophy and athero-sclerosis) and emerging primary HIV infections with multidrug-resistant viruses [2].

During the past two decades, a plethora of immunotherapeutic strategies and HIV vaccines have been developed to treat AIDS. Although these induce a certain level of immune response, they are not associated with significant clinical benefit or with an effective and sustained control of viral replication [3–5]. Nevertheless, a study by Lévy et al. demonstrated that immunization in combination with antiretroviral therapy and IL-2 improves HIV-specific immunity and lowers the viral set-point after withdrawal of HAART [6]. Lu et al. showed that ex vivo pulsed autologous monocyte-derived dendritic cells (DCs) with inactivated autologous virus induce a prolonged reduction of HIV RNA in a substantial num-ber of patients [7]. These results highlighted the potential of immunotherapy strategies targeting HIV antigens to DCs in HIV-infected patients.

The mutation capacity of HIV implies that an infected patient may harbor many genetic variants of the virus. This is undoubtedly a major obstacle for the development of a vaccine

for all variants. However, more than 90% of infections belong to HIV-1 group M [8], which includes at least nine genetically distinct sub-types (or clades). Of these, clade C (HIV-C) strains comprise 56% of all HIV infections worldwide. Therefore, HIV vaccines must include protection against this virus subtype.

While live-inactivated HIV vaccines are strongly immunogenic and can induce both cellular and humoral immune responses, their use remains a major concern because of the risk of HIV mutating back or recombining with a pathogenic virus. Although virus-inactivated vaccines are still being used because of the sim-plicity in obtaining an autologous virus, new HIV vaccine research has focused mainly on the administration of conserved recombinant viral proteins and peptides as single preparations or cocktails. Attention has also been devoted to DNA encoding several HIV antigens, recom-binant vectors and prime–boost strategies, using two vaccination approaches to immunize with the same antigen (Figure 1 shows the genome and virion organization with the potential antigens).

The expression of HIV genes in DCs could induce a response mediated through MHC class I molecules to generate cytotoxic T-lymphocytes (CTLs). In addition, HIV Tat protein stimu-lates the maturation of DCs and increases their antigen-presenting capacity, thereby promot-ing the induction of antigen-specific T-cell responses [9,10]. Recently, several DC-based HIV vaccines have produced encouraging results, thereby indicating that some HIV anti-gens – those more invariant or essential for viral

Current retroviral treatments have reduced AIDS to a chronic disease for most patients. However, given drug-related side effects, the emergence of drug-resistant strains and the persistence of viral replication, the development of alternative treatments is a pressing need. This review focuses on recent developments in HIV immunotherapy treatments, with particular emphasis on current vaccination strategies for optimizing the induction of an effective immune response by the recruitment of dendritic cells. In addition to cell-based therapies, targeted strategies aiming to deliver synthetic HIV peptides to dendritic cell-specific receptors in vivo will be discussed.

Keywords: carrier conjugates n cross-reactivity n delivery vehicle n HIV-1 n immunogenicity n nanoparticle n specific ligands n synthetic peptide vaccines n targeting n V3 loop

Luis J Cruz1,2*, Felix Rueda3, Paul Tacken1, Fernando Albericio2,4, Ruurd Torensma1 & Carl G Figdor1

1Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands 2Institute for Research in Biomedicine and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain 3Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain 4School of Chemistry, University of KwaZulu-Natal, 4041 Durban, South Africa *Author for correspondence: Endocrinology Research Lab and Molecular Imaging, Leiden University Medical Center, Building 1, C4-R67, Albinusdreef 2, 2333ZA Leiden, The Netherlands [email protected]

For reprint orders, please contact: [email protected]

Page 2: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1592 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

U3 R U5

LTR

gag

MACANCp6

PR RT IN

pol

vif

vpr rev env

vputat

SU TM

nefLTR

Surface (GP120) SU

Transmembrane(GP41) TM

Nucleocapsid (p24) NCMatrix MA

Integrase IN (p32)

Reverse transcriptase(p66)

p6, vif, vpr, nef

Protease PR(p11)

Transmembrane(GP41) TMSurface (GP120) SU

100 200 300 400 500 600 700

V1 V2 V3 V4 V5

Variable domains (gp120)

Transmembraneanchor

Fusion domainProteolyticcleavageCD4-binding domainV3 loop

1

Nanomedicine © Future Science Group (2012)

Figure 1. HIV-1 genome and virion organization: the potential use of virus proteins for vaccines. The genome of HIV has the characteristics of a lentivirus. It is composed of two LTR regions, which are involved in the replication and integration of the virus. Three regions, gag, pol and env, code for NC and MA, enzymatic proteins (protease, reverse transcriptase and integrase) and envelope proteins, respectively, and a set of accessory proteins (tat, nef, vpr, vif). Gag (p55) polyprotein is processed during maturation to MA (p17), capsid protein (p24), spacer peptide 1 (p2); NC (p7), spacer peptide 2 (p1) and p6. Pol codes for viral enzymes reverse transcriptase, integrase and HIV protease. The most frequently used proteins in HIV vaccine development are Env, envelope glycoproteins and glycoprotein (gp)120 (SU or gp120) in particular. The latter contains variable regions, such as the V3 loop. Consisting of approximately 30 amino acids, this loop is formed by two cysteine residues that form a disulfide bridge, and it has a conserved GPGRAF amino acid sequence at the perimeter. The remainder of the loop is more variable. Gp120 is known to bind with high affinity to the cellular protein CD4. After binding, this glycoprotein binds to cellular chemokine receptors, notably CCR5. The other is a transmembrane glycoprotein (TM or gp41), which contains highly hydrophobic residues in a short segment of the N-terminal (fusion) domain. This region of fusion is crucial because it allows the membrane of the virion to fuse with the cell membrane and thus allows virus entry into the cell. In addition to the fusion region, gp41 has a second hydrophobic region in the middle, which serves to anchor the protein to the lipid bilayer of the viral envelope. The gp120 and gp41 proteins are produced by fragmentation of a large precursor, gp160, and are found as oligomers on the viral particle. LTR: Long terminal repeat; MA: Matrix proteins; NC: Nucleocapsid; TM: Transmembrane glycoprotein.

Page 3: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1593future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

replication – generate specific T lymphocytes in subjects with long-term stable disease [11–13].

direct antigen vaccination using a series of presentation strategies for HIV-1Given that many of the vaccine candidates cur-rently available induce antigen-specific antibody responses and do not cross-react with heterolo-gous strains, improvements in HIV-1 vaccines are needed. To overcome the problem of HIV-1 variability, novel vaccines should be designed to induce an extensive range of reactive antibodies against multiple HIV-1 epitopes. In this regard, the authors and others have focused on achieving a good immunogen on the V3 loop of the HIV-1 envelope. This loop was chosen because it is the most variable region and is responsible for induc-tion. It is also the primary target of many neu-tralizing antibodies [14], and it is involved in vari-ous other aspects that affect infectivity. Thus, sequence changes in V3 affect chemokine recep-tor usage and therefore modulate the cell types infected [15]. Moreover, it appears that antibodies against the principal neutralizing domain of V3 exert control over viral load [16]. For the HIV-1 MN subtype B isolate, a high titer against V3 has been correlated with slow progression rates in seropositive hemophilic Japanese patients [17]. All these findings highlight the immunological relevance of the V3 loop which, in addition, includes both T-helper and CTL epitopes [18], thereby justifying its inclusion in a number of HIV-1 vaccine candidates [19]. However, in a recent report from the RV144 trial, in a vaccine composed of a recombinant canarypox vector expressing HIV-1, gag, pro and membrane-linked gp120 (vCP1521), combined with a bivalent gp120 protein boost (AIDSVAX B/E) (one of the few vaccines that has shown some protection against HIV-1), the T-cell responses were preferentially targeted to epitopes in the V2 region of the HIV-1 envelope, although the neutralizing antibodies were targeted to the V3 region [20,21]. Furthermore, recent results have shown the presence of neutralizing antibodies rec-ognizing quaternary structures in V2 and V3 [22]. These results demonstrate that regions other than V3 could also be important for the generation of new vaccines.

Current HIV vaccine models comprise either recombinant proteins [23] or synthetic HIV-1 multi-epitopic peptide constructs [24,25]; how-ever, naked DNA [26], plasmid DNA [27,28] and mRNA are gaining ground [29–31]. Multi-epitope polypeptide TAB9, containing six V3 epitopes

from isolates LR10, JY1, RF, MN, BRVA and IIIB, were selected for immunological response in animal models (Figure 2), and were subsequently used in a Phase I clinical trial. All immunized volunteers seroconverted, and antibodies were broadly reactive against the V3 peptides included in the protein. However, the immunogenicity of the recombinant protein induced high levels of reactivity directed against the recombinant protein itself, mainly at JY1 and LR150, and only weakly at MN [32]. Naked DNA technology has progressed in the last decade; however, suitable delivery routes continue to be elusive. Recently, Rosenberg et al. [33] demonstrated that the HIV-1 DNA vaccine is safe but poorly immunogenic in subjects treated with antiretrovirals during acute/early HIV-1 infection. However, a combi-nation of polypeptide and a DNA vaccine could boost the immune response as a result of a com-bination with T-helper epitopes [34]. Viral vectors achieve this objective efficiently; however, they present other kinds of problems, including safety issues. The discovery of specific and small anti-gens, such as those of the V3 loop, has allowed the development of synthetic peptides for HIV-1 vaccines (Figure 2) [35,36].

�n Peptides, multiple antigenic peptides & combinatorial libraries, & their conjugation to carrier proteins Given the variability of HIV-1, in a previous study, the authors focused their efforts on several V3-based multiple antigen peptides (MAPs) and peptides. This strategy enhances the immuno-genicity and cross-reactivity of these structures towards other V3 sequences [37]. To this end, several presentation strategies were examined, such as polymers [38], MAPs and peptides cou-pled to dextran microparticles [39]. Combined approaches, such as the coupling of MAPs to carrier proteins [40], were also tested [41].

Cocktails containing several peptide variants were used in an attempt to overcome viral vari-ability. The authors had previously shown that V3 cocktails with MAPs provide an efficient vaccination strategy [28]. In the context of that study, the following points of attention were raised: first, recognition of distinct V3 peptides is closely related to the degree of homology between them; second, conjugation allows the inclusion of more peptide variants in cocktails with a smaller mass of antigen; and third, a combination of con-jugated MAP and peptide significantly enhances cross-reactivity.

In fact, the authors’ results confirmed their hypothesis that the inclusion of V3 peptides,

Page 4: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1594 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

such as JY1 and RF subtype, in a dendrimeric structure, such as a MAP or MAP coupled to a carrier protein, increases the generation of broadly reactive antibodies [33].

The HBsAg–JY1–MAP conjugates elicited the highest titers against JY1 in comparison with all the other strategies tested. Carriers such as KLH and HBsAg form nanostructures with greater surfaces to incorporate MAP and peptides. Furthermore, these carriers offer more sites for the coupling of synthetic peptides. By contrast, carrier–MAP conjugates are more immunogenic than carrier–peptides [42].

An alternative approach aimed to cover the identification of a wide spectrum of V3 epitopic sequences is the generation of combinatorial

peptide libraries, called mixotopes [43,44]. Mixotopes comprise several peptides that rep-resent variable positions of the V3 loop, and act as model antigens for recognition of the immune system. Promising results using this type of strategy have been reported [45].

In contrast to the findings described by Resnick et al. [46], the authors demonstrated that the mixotope strategy does not generate higher levels of cross-reactivity against several V3 pep-tides than those of the consensus strategy. The mixotope library tends to target mainly the con-served regions of the V3 loop, which are overrep-resented compared with the variant ones. With regard to highly variable antigens, it could be concluded that a rationally designed consensus

=

Recombinant protein vaccine

N-terminus P64K LR150 JY1 RF MN BRVA IIIB

Multiple antigen peptide vaccine:

Loop V3:LR150, JY1,RF, MN, BRVA,IIB

Loop V3 Loop V3

Loop V3

Loop V3

Loop V3

Loop V3

Loop V3

Loop V3

KK

KK

KK

KK

T helper

T helper

T helper

T helper

MAP

MAP

MAPMAP

MAP

MAP MAP

KGC-S—–S-CGK

Complex macromolecules for vaccines

Proteins:Gp120 or gp40,P64k, KLH orHBsAg

COOH

COOHHOOC

HOOC

Conjugation

Peptides

Proteins

Proteins

Proteins

Nanomedicine © Future Science Group (2012)

Figure 2. Various strategies for HIV-1 peptide vaccine formulations: recombinant proteins, multiple antigen peptides and protein conjugates. (A) Recombinant protein, a multi-epitope polypeptide including the V3 region from six divergent HIV-1 isolates (LR150, JY1, RF, MN, BRVA and IIIB, in this order) fused to the amino terminus (47 amino acids) of P64K protein from Neisseria meningitidis. (B) The V3-MAP variant includes two monomers linked through a disulfide bond and with a 2:1 epitope ratio. A cathepsin-like enzyme cleavage site (-KK-) was inserted between B- and T-cell epitopes. Six V3-MAP8 constructions bearing divergent V3 HIV-1 sequences LR150, JY1, RF, MN, BRVA and IIIB as B-cell epitopes were synthesized. Monomers were properly assembled for heterodimeric MAP combinations. (C) Complex macromolecule vaccines are conjugates of peptides or MAPs with carrier proteins, such as Gp120, KLH or HBsAg. MAP: Multiple antigen peptide.

Page 5: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1595future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

sequence, such as the one derived for the V3 loop of gp120, is a valid approach for synthetic vaccine development, providing desirable cross-reactivity and therapeutic advantage.

However, early enthusiasm for exploring pep-tide vaccines against HIV-1 infection was based on the supposition that the V3 loop of HIV-1 was the main neutralizing determinant of the virus and the demonstration that this deter-minant is mimicked antigenically by synthetic peptides [47]. Two Phase III trials showed that bivalent, recombinant gp120 (rgp120) vaccines do not prevent HIV infection [48,49]. Although vaccine efficacy was not demonstrated, a separate analysis revealed an inverse correlation between vaccine-induced antibody activity and HIV infection rate [49]. The authors proposed that the vaccine-induced antibody was a marker of another – presumably innate – immune response that ultimately reduced the rate of infection. Recently, an HIV-1 lipopeptide vaccine tested in healthy adults in a Phase II trial proved immu-nogenic and safe at a range of doses. Moreover, it elicited HIV-specific sustained CD8 and CD4 T-cell responses in healthy adults [50].

However, the limited success of direct pep-tide vaccination strategies in Phase III prompted various research groups to develop cell-based HIV-vaccines using DCs.

strategies to activate HIV-1 antigen presentation in dCsThe functional characteristics of DCs allow them to capture antigens by several pathways. Thus, a number antigen-loading methods for DCs have been described [51]. For instance, synthetic peptides need mature DCs to be more effective, while major antigens, such as proteins and whole virus, are usually loaded into immature DCs.

In the case of HIV-1, where the genetic variation of the virus makes it difficult to fully identify and define the antigen epitopes of the virus infecting each patient, DCs can be loaded with the whole virus. This approach may reduce antigen escape by a loss of epitope variants. If the antigen is processed and presented through both MHC class I and II molecules, a more potent immune reaction is gen-erated because CD4+ T cells play a critical role in inducing and maintaining effective CTL responses [52]. The main disadvantage of this strategy is the potential risk associated with the use of inactivated whole virus (despite the safety of inactivating meth-ods, recombination with integrated HIV DNA may occur and may thereby generate infectious parti-cles). In addition, immune monitoring is hindered because the epitopes involved are unknown.

�n Inactivated HIV-1 virus Several methods to inactivate HIV-1 have been described, with different effects on the integrity of HIV-1 antigens: irradiation, heat, formalin and aldrithiol-2 (AT-2). All these approaches render the virus non-replicative before in vivo administration. Yoshida et al. tested a prophylac-tic vaccine consisting of autologous mature DCs (mDCs) loaded with either AT-2-inactivated HIV-1 or an irrelevant antigen, ovalbumin [53]. Although the study was performed in severe combined immunodeficiency (SCID) mice transplanted with human peripheral blood mononuclear cells, only mice immunized with DC-HIV-1 were protected against HIV-1 chal-lenge in vivo. In another study, 3 days after reconstitution with autologous human peripheral blood lymphocytes (PBLs), human PBL-SCID mice were injected with autologous DCs loaded with AT-2-inactivated HIV-1 [54]. Immunization induced human anti-HIV-1 antibodies and the generation of a human CD8+ T-cell response against HIV-1. In addition, immunized human PBL-SCID mice were more resistant to virus infection than control animals.

In rhesus monkeys infected with simian immunodeficiency virus (SIV), Lu et al. showed that a therapeutic vaccination performed with AT-2-inactivated SIV-pulsed DCs induced effec-tive and sustainable SIV-specific cellular and humoral immunity and a decrease in the viral load induced [55].

In a clinical trial carried out by the same group in 18 patients with chronic HIV-1 infection, the efficacy of a therapeutic AT-2-inactivated HIV-pulsed autologous DC vaccine was assessed [7,56]. A temporary decrease in plasma viral loads of 80% was observed. This reduction correlated with HIV-1-specific IL-2- or IFN-g-expressing CD4+ T cells and with HIV-1 gag-specific per-forin-expressing CD8+ effector cells. These data suggest that a robust virus-specific CD4+ Th1 response is required for inducing and maintain-ing virus-specific CD8+ effectors in order to control HIV-1 replication in vivo. Other clinical trials are currently underway with psoralen and UVB irradiation (312 nm) [57] or heat-inactivated HIV. Recently, results from 24 untreated patients with chronic HIV-1 infection enrolled in a dou-ble-blinded, controlled study of vaccination with three doses of autologous monocyte-derived DCs pulsed with heat-inactivated autologous HIV-1 were reported [58]. The vaccine was safe and well tolerated, and after vaccination, an increase in HIV-specific T-cell responses was observed. This enhancement tended to be inversely associated

Page 6: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1596 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

with the decrease in plasma viral load in vacci-nated patients, but not in the control group. No positive CD4 lymphoproliferative responses to HIV-1 p24 antigen were observed in any patient. However, as noted above, the use of live-inac-tivated HIV vaccines entails safety concerns. Therefore, the administration of recombinant proteins, peptides, DNA or mRNA encod-ing defined antigens is an attractive alterna-tive (Figure 3). Unfortunately, immunological responses induced by this approach are only low and partial and may allow the virus to escape recognition and neutralization. Therefore, new strategies should be tested with combinations of viral proteins or peptides showing low variability, DNA vaccines composed of several genes that encode HIV subunits, recombinant vectors and prime–boost strategies, which use two ways of administering the same antigen.

�n Protein antigen HIV-1 has several proteins that can be used as antigens for vaccination purposes (Figure 1). However, when administered alone, recombi-nant viral proteins are poor immunogens. An attractive alternative is to combine several HIV proteins into one vaccine. Liposomal prepara-tions containing several HIV-1 structural pro-teins (p24 gag, gp160 env or p55 gag) to load

DCs are delivered to and processed through the HLA class I pathway, and prime CD8+ T cells against HIV-1 in vitro [59]. However, some HIV proteins may induce adverse effects, which represent an important drawback; for instance, HIV-1 gp120 binds to the surface of CD4+ T cells, thus contributing to the immu-nopathogenesis of this virus. This envelope gly-coprotein influences various cell populations of the immune system, including hematopoietic progenitors, T and B lymphocytes, monocytes/macrophages and DCs, as well as neuronal cells [60]. Furthermore, despite displaying a typi-cal phenotypic maturation, immature human monocyte-derived DCs (iDCs) loaded with either AT-2-inactivated HIV-1 or gp120 show an impaired capacity to secrete cytokines or chemokines and to induce T-cell proliferation [61]. This immunosuppressive condition is not reverted by stimulation with lipopolysaccharide or CD40L. However, exogenous addition of IL-12 does restore the allostimulatory capacity of gp120-exposed DCs.

Extracellular Tat has crucial effects on immu-noregulatory mechanisms, driving iDC matu-ration, inducing MHC class I molecules and tipping the balance towards a Th1-inducing phenotype [62]. However, Tat also alters the proteasome function by modifying proteasome composition and enzymatic activity, thereby changing the processing of CTL epitopes. This favors the presentation of subdominant and cryptic epitopes [63]. Furthermore, Tat, directly or indirectly, also induces damaging effects in several organs and host systems, such as the myocardium, kidney, liver and CNS. HIV-1 Nef protein downregulates the expression of HLA class I [64,65] and II molecules [66]. Taken together, these observations indicate that caution should be taken regarding the use of full-length viral proteins for vaccination purposes.

�n Antigen-encoding DNA & RNA Among the methods available for HIV-1 anti-gen loading on DCs, genetic techniques for the introduction of RNA or DNA are gaining popularity owing to their flexibility and simplic-ity. The use of RNA isolated from pathogens or infected cells has a number of advantages. First, sufficient RNA can be generated from a small amount of sample by amplification tech-niques [67]. Second, target-restricted RNA can be enriched before DC loading by subtractive hybridization with RNA from uninfected cells. Pathogen-specific immune responses are thus augmented and the risk of autoimmunity is

DC vaccination strategy

IL-4GM-CSF

ImmatureDC Maturation

stimuli HIV-1 peptides,RNA, DNA,

recombinant protein

Injection in patient

Monocyte

Isolationfrom blood

Disadvantages:- Time consuming- High costs- Preparation of individual vaccine

Figure 3. HIV vaccination strategy with dendritic cells. DCs can be loaded with the whole protein when a pathogenic antigen has been defined but the peptide epitope has not yet been identified. This approach not only circumvents the need to identify peptide epitopes, but also expands the clinical application to patients who are excluded because of MHC restriction, a problem associated with the use of peptide epitopes. Adapted with permission from [170].

Page 7: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1597future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

reduced. However, the major drawback of the use of RNA is the instability of its products and the greater technical demands required [68]. Similar to RNA, it is possible to generate DNA encoding a target antigen that can be expressed in the antigen-presenting cell (APC). Thus, the antigen can be processed and presented in a way that closely resembles the processing of endog-enous proteins or virus, where protein-derived peptides are loaded onto MHC class I molecules to activate CD8+ T cells [69].

Because DNA is less prone to degradation than RNA, antigen presentation may last lon-ger. However, the generation of immunocom-petent DCs with target DNA continues to be difficult because of the limitations of DNA delivery systems. The most common method for introducing RNA into DCs is electropora-tion [70]. This approach allows the expression of exogenous antigens by DCs, without the risk of viral vectors. Furthermore, genetically modified recombinant vectors have proved to be the most suitable vehicle for DNA delivery into DCs. Recombinant vector vaccines use attenuated virus or bacteria as carriers of modified target antigens into the host. The target genetic infor-mation is incorporated into the vector genome and target proteins are produced. DCs loaded ex vivo with recombinant vectors, such as rep-lication-defective herpes simplex virus type 1 amplicons that express HIV gp120 (herpes sim-plex virus gp120MN/LAI), replication-defective adenovirus serotype 5 vectors expressing SIV Gag antigens p17 and p45 [71] and recombinant Lactococcus lactis vector expressing the V2-V4 loops of HIV Env on its cell surface (IL1403-pHIV) [72], induce HIV-specif ic immune responses in vivo. However, vector-specific neu-tralizing antibodies may limit the effectiveness of repeated booster injections. Therefore, the use of these vectors in clinical vaccination trials is still limited [73]. Furthermore, although the new generation of recombinant viruses offers greater safety, these viruses pose a certain degree of risk for immunocompromised patients. Despite this risk, some clinical trials have been performed with recombinant viral vectors. Autologous DCs from healthy volunteers were transfected ex vivo with ALVAC-HIV (vCP205), a canarypox-based vaccine containing env, gag and pol genes from HIV. Although only modest cytotoxic CD8 T-cell responses were presented, volunteers who received the DC autologous vaccine loaded in mDCs (which showed lower expression of the target genes than those loaded with iDCs) developed a broader and more durable immune

response than those who received ALVAC-HIV by intramuscular, intradermal or subcutaneous injection. Another possibility is the use of viral structural proteins that resemble virus particles or virus-like particles (VLPs), particularly HPV capsid proteins, which show the same capacity to induce an immune response [74] as the viral vector. Moreover, the affinity of the VLP for heterologous DNA allows its use in gene trans-fer of antigens [75], but this approach shows less efficiency than that achieved by a viral vector.

�n Exosomes as a tool for HIV-1 vaccinationIn 1998, DC-derived exosomes were shown to have immunogenic properties leading to tumor eradication in mice [76]. Exosomes are small membrane vesicles of approximately 30–100 nm in diameter. Exosome secretion is a process in which multivesicular bodies fuse with the cell membrane. Exosomes derived from tumor or APCs are released into the extracellular environ-ment and fuse with the membranes of neighbor-ing cells, thus delivering membrane and cyto-plasmic proteins from one cell to another. Such proteins, for example, MHC molecules, costim-ulatory molecules and heat-shock proteins, play key roles in immune responses [77]. DCs could be primed with antigens and then the DC-derived exosomes could be used as cell-free vaccines [78].

A lentiviral vector encoding HIV gag–pol without env would form viral cores in transduced DCs, but would release noninfectious particles. It was shown that accumulation of viral cores inside transduced DCs and the exosomes from these cells produce a second-round transduction of iDCs. Mice were immunized with transduced DCs to induce specific lymphocyte activation, and they produced antigen-specific T-cell responses and specific antibodies after immunization. This approach could provide a novel strategy for HIV

Table 1. receptors used to target antigens to dendritic cells.

Targeted receptor ref.

Mannose receptor [98,99]

Dec-205 [100–102]

LOX-1 [103,104]

Langerin [105]

DC-SIGN [107–111]

Clec9A [112,113]

CD40 [117–119]

FcR family [120–132]

Page 8: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1598 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

vaccination [79]. However, exosomes produced by T cells and DCs from HIV-1-infected patients have been associated with HIV-1 dissemina-tion, although a budding pathway is not shared by exosomes and HIV-1 [80]. Some studies have shown a previously undescribed mechanism of DC-mediated HIV-1 transmission and suggest that virus particle trafficking to multivesicu-lar endosomal bodies and subsequent exocyto-sis can provide an avenue for immune escape for HIV-1 particles captured by DCs [81], thus allowing viral internalization and final transin-fection of CD4+ T cells [82]. Furthermore, exo-somes released from HIV-1-infected cells harbor regulatory molecules that affect cell apoptosis (ANXA5 and LDHB) and proliferation (CD38) [83], and export Nef, which facilitates the depletion of CD4+ T cells, a hallmark of AIDS [84]. Given all these observations, although exosomes are a prom-ising tool for vaccination for tumors and infectious pathogens, caution is needed when considering them in the setting of HIV-1 infection.

Targeting of antigens to dCs in vivo�n Ligands to deliver protein or

peptides to DC receptorsDespite the promising results of vaccines using antigen loading on DCs ex vivo, the cost and technical demand of ex vivo generation of DCs makes this approach unsuitable for large-scale immunization programs. Ex vivo procedures require the isolation of rare naturally occur-ring DCs or the ex vivo generation of DCs from monocytes isolated from the patient. Both procedures are labor-intensive and require spe-cial equipment to collect the cells from blood. Moreover, it is costly to perform ex vivo culture of cells following good manufacturing proce-dures. The cell collection procedure takes hours and is an extra burden for the patient. These drawbacks of cellular therapies could be pre-vented by targeting antigens to DCs in vivo via specific surface receptors. These constructs are cost effective and can be used from the shelf [85].

Triggering the appropriate receptors might even lead to the activation and maturation of DCs and to the boosting of immune responses. The antigen delivery system will determine the route of entry into the cell and the pathways, mecha-nisms and efficiency of antigen processing and presentation via MHC class I and II molecules. Receptor ligands can be directly attached to pro-tein and peptide antigens for targeting aims or, alternatively, proteins, peptides or DNA encod-ing the antigen could be incorporated on other suitable drug delivery systems, such as liposomes,

nanoparticles and dendrimers [86]. Two categories of receptor ligands target antigens to DC recep-tors: first, ‘natural’ receptor ligands, including bacterial toxins, heat-shock proteins and sugar residues; and second, receptor-specific antibodies.

�n Targeting DC surface receptorsMany receptors on the surface of DCs induce intracellular signaling pathways upon interaction with the ligand. The general characteristics and qualities that make receptors amenable as vac-cine targets include specific and high expression on DCs, endocytic activity, capacity to induce antigen processing and presentation via MHC class I and II pathways, as well as a potential to trigger signaling pathways to stimulate an immune response. Receptors expressed by DCs displaying at least several of these properties include CD40, Fc-receptors (FcRs), Toll-like receptors (TLRs), C-type lectin receptors, Mac-1 (CD11b/CD18) and globotriaosylceramide (Gb3/CD77). Table 1

presents a summary of DC surface receptors that have been used to target antigens to DCs.

�n Targeting antigens to the MHC class I & II pathwaysAn efficient strategy for HIV vaccination should induce both humoral and cellular immune responses. To this end, antigens should be pro-cessed and presented by both MHC class I and II pathways. For antibodies, the B cell must also be reached, and this is difficult when the packed antigen is targeted to a DC, where it is unzipped inside this cell. In general, strategies targeting antigens to be released directly into the cytosol, or DCs transfected with DNA-encoding anti-gens will be efficiently presented via the MHC class I pathway. As cellular immune responses are considered crucial in reducing viral load, routing antigens into the MHC class I loading pathway appears to be an essential requisite for HIV-1 vaccines. However, targeting protein antigen to receptors on DCs will essentially result in MHC class II presentation; never-theless, antigens captured this way are able to escape from endosomes and can be presented by MHC class I by means of a cross-presentation mechanism (Figure 4) [87]. Although particulate antigens loaded by phagocytosis are more effi-ciently cross-presented than soluble antigens taken up via endocytosis, both soluble and par-ticulate antigens can be targeted to DC surface receptors for class I presentation [88].

Several bacterial toxins have been used to deliver antigens to be presented by MHC class I molecules. These toxins bind to DC receptors

Page 9: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1599future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

and can be directly translocated into the cyto-sol or cross the endosomal membrane to reach the cytosol, or the endoplasmic reticulum (ER) via a retrograde endosome–Golgi–ER pathway [89]. In this way, antigens coupled to toxins that target the cytosol and ER gain direct access to the classical MHC class I pathway and induce CTL responses in vivo [90].

Regarding the recognition of structurally conserved molecules derived from infectious agents, several TLRs, which are among the most potent activators of the immune response, are present on DCs [91]. TLR engagement alerts the immune system and leads to the activation of innate and adaptive immune responses. TLRs trigger DC maturation, stimulate the prolifera-tion of CD4+ and CD8+ T cells and modulate the suppressive function of Tregs [92]. These

immune-stimulating properties have resulted in the use of TLR agonists in cancer treatment, both as monotherapy to induce innate responses and as adjuvants for vaccines [93]. It is possible to target viral antigens to TLRs, either by conjuga-tion of the antigen to an appropriate TLR lig-and, or by using HIV-1 itself as a TLR agonist, as HIV ssRNA is a potent immune activator triggering TLR 7/8. However, TLR triggering in HIV-infected individuals is not without risks as the activation of certain TLRs, including TLR-2 and -9, induces HIV-1 replication and thereby disease progression [94]. Thus, while flagellin (TLR5 agonist) treatment enhances the repli-cation of CC chemokine receptor 5-tropic and CXC chemokine receptor-tropic HIV-1, treat-ment with oligodeoxynucleotide M362 (TLR9 agonist) suppresses these two viral variants [95].

Targeting antigens to MHC-I and -II

Target receptor

Direct entrance

MHC-I

Proteicantigens

MHC-II

Phagosome

MHC-I

Endosome

MHC-II

Virus orDNA

Proteosome

Peptides

Endoplasmic reticulum

Nanomedicine © Future Science Group (2012)

Figure 4. Targeting antigens to the MHC class I and II pathways. In order to transport and present exogenous antigens into the MHC class II pathway, endocytotic and phagocytotic endosomes fuse with vesicles that transport MHC class II molecules from the endoplasmic reticulum compartment. There are several vaccination strategies to target antigens across the cytoplasmic membrane to reach the MHC class I pathway. Furthermore, there is evidence that antigens escape from the phagosome and endosome compartments into the cytosol. The same happens with DNA vaccines encoding antigen that enter DCs via the endosomal or phagosomal compartment, where they can escape, thus resulting in the expression of the antigen in the cytosol. Finally, certain bacterial toxins linked to antigens target them via the endosome and Golgi to the ER through the retrograde pathway. In addition, endocytosed proteins can be transferred from the endosome to the ER, where they can reach the cytosol.

Page 10: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1600 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

Another strategy to target antigens to DCs comprises the attachment of sugar residues to the antigen. This procedure allows recognition by C-type lectin receptors on DCs, thereby resulting in presentation via the MHC class I and II pathways. C-type lectins belong to a family of lectins that share primary structural homology in their carbohydrate-recognition domain, which binds to sugar residues in a cal-cium-dependent manner. The choice of sugar residue determines the receptors targeted and thus the immunological outcome. For example, antigens coupled to oxidized mannan induce mostly cellular responses, whereas those cou-pled to reduced mannan produce humoral responses [96]. Expressed by DCs, these recep-tors are implicated in immunoregulatory pro-cesses, such as antigen capture, DC trafficking and DC–T-cell interactions [97]. Of the C-type lectin receptors used for antigen targeting for vaccination purposes, mannose receptor (MR), DEC-205 and DC-SIGN have received the most attention.

Certain MR ligands and antibodies induce an immunosuppressive response in DCs [98]. However, recent studies involving monoclonal antibodies specifically targeting antigens to MR conclusively demonstrate enhanced antigen uptake and presentation via the MHC class I and II pathways [99].

The characteristics associated with C-type lectin receptors have led many researchers to assay them in vaccine designs. Thus, DEC-205 is a type I C-type lectin receptor that is highly expressed on mDCs and to a much lesser extent on certain leukocyte subpopulations [100]. DCs cross-present HIV gag to CD8+ T cells, but when the antigen is delivered within an antibody to DEC-205 receptor, cross-presentation becomes 100-fold more efficient than nontargeted anti-gens [101]. Antibody–antigen conjugates target-ing tumor antigens to MR and HIV gag p24 to DEC-205 are currently being evaluated in clinical trials [102].

DC LOX-1 is a type II C-type lectin receptor expressed on several cell types, including macro-phages and peripheral blood myeloid DCs [103]. LOX-1 is a receptor for Hsp70, and targeting DCs with protein antigen conjugated to Hsp70 results in cross-presentation in vitro and in vivo [104]. Targeting vaccine antigen to Langerin+ DCs favors the induction of T-cell responses [105], whereas targeting LOX-1+ DCs enhances the induction of antibody responses [106].

DC-SIGN is a type II C-type lectin with a more restricted expression to professional APCs than that shown by DEC-205 and MRs. DC-SIGN is highly expressed on DCs residing in lymphoid tissues, mucosal surfaces and der-mal tissue, and on specialized macrophages in the placenta and lung. This expression profile repre-sents a major advantage of targeting this lectin over other C-type lectin receptors [107]. Although DC-SIGN has been associated mainly with the spread of HIV-1 in humans and most of the work reported concerns blocking HIV-1–DC-SIGN interactions, in vitro studies show efficient target-ing of liposomes [108], adenovirus [109] and pro-tein antigens [110] to DCs via DC-SIGN, which induces both naive and recall T-cell responses via MHC classes I and II molecules. In primary DCs, DC-SIGN-captured HIV virions are rapidly degraded, possibly in a lysosomal compartment. Thus, a fraction of the incoming viral material is processed by the proteasome, leading to activation of anti-HIV-specific cytotoxic T-lymphocytes by DC-SIGN-expressing cells [111].

Finally, the type II C-type lectin receptor CLEC9A was recently proposed as an interest-ing target for vaccination purposes. In humans and mice, CLEC9A expression is relatively restricted to a specific DC subset, which is desig-nated BDCA3+ in humans and CD8a+ in mice. Moreover, in mice, this subset is specialized at cross-presenting antigens to CD8+ T cells [112,113].

Antibodies, and not the natural receptor ligands, appear to be the most efficient system to target antigens to DCs, and they are more potent than ex vivo antigen-loading DCs [114]. The production of humanized and human anti-bodies has improved the clinical efficiency of these tools and has led to the approval of many antibody-based therapies for clinical use [115]. Numerous antibodies targeted to cell-surface receptors induce receptor-mediated endocytosis, a relatively inefficient process for the activation of cross-presentation. However, antibody-mediated targeting of protein and peptide antigens to DC surface receptors induces strong CTL responses in vivo [116].

Table 2. delivery systems to target dendritic cells.

Group delivery system ref.

Live vector Live-attenuated virusLive recombinant virus (including HIV-1) or bacteria (gene transfer)

[133][134,135]

Micro/nanoparticles Exosomes Virus-like particlesVirosomesLiposomesPolymer micro/nanoparticles

[76–78,81–84][136][137]

[138,139][140–143,146,147]

Page 11: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1601future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

A paradigmatic case is the targeting of anti-bodies to CD40 in DCs. This protein is expressed in many cell types, including macrophages, DCs and endothelial cells [117].

CD40 is an excellent candidate for HIV vac-cines based on DC targeting, as its activation not only causes maturation of APCs but also allows efficient presentation and cross-presentation of HIV epitopes, thereby stimulating strong recall T-cell responses [118]. In addition, studies with recombinant monoclonal antibodies (rAbs) for multiple internalizing human DC recep-tors, engineered by fusion via the heavy chain C-terminus to a string of five 19- to 32-amino acid long sequences from HIV-1 gag, Nef and Pol proteins (called rAb.HIV5pep), showed that anti-CD40.HIV5pep was strikingly more effec-tive. CD4+ T cells were found to produce multiple cytokines, while the CD8+ T cells produced cyto-kines, in addition to perforin, granzyme B and surface CD107a, and had CTL characteristics and proliferative capacity [119].

As an alternative to antibodies that recognize target receptors, those recognizing antigens could also be used for vaccination purposes. These antibodies form immune complexes with the antigen, which are captured by FcRs recog-nizing the constant fragment of the antibody. Antigens targeted to FcRs are presented via the MHC class I pathway, thereby simultane-ously inducing DC maturation and increasing the efficiency of T-cell activation by a factor of 1000–10,000 [120].

FcRs comprise a family of membrane pro-teins that are important mediators of immune responses, such as phagocytosis, antibody-dependent cell-mediated cytotoxicity, regula-tion of lymphocyte proliferation and antigen presentation [121]. DCs express receptors for IgG (FcgR), IgA (FcaR) and IgE (FceR).

Immune complexes of IgG and antigen induce DC maturation and the priming of peptide-spe-cific CD8+ T cells in vivo [122]. However, the bal-ance between activating and inhibitory signaling after engagement of the various FcRs on the DC dictates whether the uptake of the immune com-plexes results in efficient naive T-cell activation and protective immunity [123]. This can be over-come by targeting vectors containing antibodies that recognize a specific activating FcR. These vectors should be carefully designed as FcRs need to be cross-linked in order to induce inter-nalization. Therefore, single-chain Fv constructs are not suitable for FcR targeting unless they are themselves cross-linked. Moreover, single-chain Fv constructs might inhibit immune responses,

as continuous stimulation of FcaRI with single-chain Fv has been described to block activating responses of heterologous FcgRI and FceRI [124]. Targeting antigens specifically to FcgRI results in antigen presentation via the MHC class I and II pathway in vitro [125].

At least 1% of human antibodies recognize a-gal residues and the expression of a-gal epit-opes in HIV gp120 and influenza virus vaccines increases immunogenicity by approximately 100-fold. The immunogenicity of a microbial vaccine can be markedly increased by linked a-gal epitopes as a result of in vivo formation of immune complexes with anti-Gal and the effec-tive internalization of these complexes by APCs via Fc/FcgR interaction. The increased transport to lymph nodes and processing of anti-Gal com-plexed vaccines internalized by APCs results in effective activation of vaccine-specific CD4+ and CD8+ T cells and potent cellular and humoral immune responses [126]. In addition, rgp120 vac-cination can elicit antibodies with antiviral activity against clinical strains of HIV-1. However, this activity requires the presence of FcR-bearing effec-tor cells. This result provides further evidence that antibody-dependent cell-mediated virus inhibition can be harnessed to prevent HIV infection [127].

An example of the relevance of FcRs in HIV vaccines is that Fc/FcgR interactions play a criti-cal role in the biological function of an antibody. Moreover, these receptors may be essential in preventing or modulating lentiviral infection. Exploiting antibody responses that depend on Fc/FcgR interactions may help overcome some of the difficulties associated with HIV vaccine development by increasing the potency of the antibody response. Although the importance of generating optimal Fab–antigen interactions can-not be overestimated, improving Fc/FcgR interac-tions through adjuvants, by directly altering the Fc segment of monoclonal antibodies or by other strategies, provides another option for ameliorat-ing HIV vaccines and immunotherapies [128–130].

The effect of neutralizing antibodies that are specific for HIV-1 gp41 membrane-proximal external region were moderately stronger for IgG1 than for IgG3 subtypes, and were ineffective when using fab fragments. It was concluded that FcgRI and FcgRIIb facilitate antibody-mediated neutral-ization of HIV-1 by a mechanism that is depen-dent on the Fc region, IgG subclass and epitope specificity of the antibody [131].

The importance of FcgRs in AIDS pathogen-esis and the critical role of interactions between FcgRs and immune complexes in disease pro-gression are reflected by the observation that

Page 12: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1602 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

patients with IgG2 anti-p24 and who express the ‘high-affinity’ polymorphism of FcgRIIa exhibit lower HIV replication than those without the polymorphism [132].

�n Drug delivery systems to target DCs Drug delivery systems to target DCs can be subdivided into live vectors, micro- or nano-particles and receptor ligands (Table 2). Live infectious vectors and certain microparticles are particularly well suited to deliver DNA to DCs. For delivery of protein and peptide antigens to these cells, both microparticles/nanoparticles, liposomes and receptor ligands are used.

Live vectors to deliver DNA to DCsLive-attenuated viruses and bacteria potently induce cellular immune responses in HIV vac-cination therapy in animal models [133]. The live vectors drive expression of antigen encoded by DNA inside the cells they invade and the antigen then enters the classical MHC class I pathway.

Viruses in particular show promise as vehicles for targeted gene delivery. The incorporation of DC surface receptor ligands, mostly antibodies, into viruses allows them to use these receptors for cell entry and to specifically infect DCs [134]. A DC-targeted recombinant lentiviral vector was reported to effectively transfect DCs in vivo and to induce strong antigen-specific humoral and cellular responses. DC-specific targeting of the vector was accomplished by introducing an engineered gly-coprotein derived from Sindbis virus, which serves as a ligand for the DC-specific receptor DC-SIGN [135]. Several issues, including some related to safety, remain to be addressed regarding the use of live vectors as vaccines. Reversion of attenuated live vectors to virulent strains by mutation or genetic recombination cannot be excluded and may be especially problematic in immunocompromised individuals such as AIDS patients.

Micro/nanoparticles to deliver DNA, protein or peptides to DCsMicroparticles include lipid particles such as liposomes, polymer microparticles, VLPs [136] and virosomes [137]. Particulate delivery of anti-gens often increases their stability and bioavail-ability. Furthermore, microparticles are effi-ciently taken up by APCs through phagocytosis, thus resulting in antigen presentation.

Targeting of liposomes containing peptide antigen or DNA to professional APCs effectively enhances their capacity to induce CTL responses in vivo [138]. Several methods to target DCs with liposomes have been described [139].

As occurs with liposomes, polymer micro/nanoparticles allow the protection of antigenic proteins, peptides and DNA from immedi-ate degradation. Entrapment of HIV protein antigen in poly(lactide-co-glycolide) (PLGA) microparticles results in an effective vaccine that induces specific MHC class I- and class II-mediated responses in mice [140].

Some data provide proof of principle that both the antigen and DC maturation signal can be delivered as a complex with polyelectro-lyte capsules to stimulate virus-specific T cells both in vitro and in vivo. Other nanomaterials could be useful for in vivo immunization against HIV-1 and other infections [141,142].

Targeting nanoparticles to human DCs as an effective strategy to deliver HIV-1 antigenSpecific antigens for CTL response (such as gag, pol, env and nef antigens) and TLR ligands (such as Poly I:C, PAM and R848) can be incorpo-rated into biodegradable polymers, such as PLGA (Figure 5). This polymer is safely used in clinical practice. Combining TLR ligands (as adjuvant) and antigen in PLGA particles enhances the induction of CTL responses [143]. For optimal antigen presentation, it has been shown that the TLR ligands and antigen should be in the same compartment [144]. Recently, PLGA nanopar-ticles carrying a combination of TLR ligands were reported to protect mice and nonhuman primates against a variety of influenza strains and to induce long-lasting humoral memory [145]. Importantly, such PLGA nanoparticles can be modified to target DC-specific recep-tors on human DCs by introducing humanized antibodies recognizing the DC-specific recep-tor DC-SIGN [146,147]. Alongside antibodies, the HIV-1 gp120 protein might be used for targeting purposes. This glycoprotein binds to DC-SIGN and could thereby simultaneously serve as an antigen and the targeting moiety [148].

Immunological regulation in the setting of HIV-1 vaccination with dCsHIV-1 infection is associated with elevated immunological activation, which may cause T-cell exhaustion. Depending on the dura-tion and magnitude of antigenic activation of T cells during HIV-1 infection, some intrinsic and extrinsic factors are produced that influ-ence the severity of T-cell exhaustion. Recent observations have detailed the upregulation of inhibitory receptors, which are the major intrinsic factors of T-cell exhaustion. Among the

Page 13: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1603future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

extrinsic factors are the production of suppres-sive cytokines, T-cell priming by either nonpro-fessional APCs or myeloid-derived suppressor cells, a kind of tolerogenic DC, and alteration of Tregs [149]. In the setting of HIV infection, most immunological studies have shown an increase of Tregs within the T-cell pool, with respect to healthy individuals, but a decrease in the absolute number of T cells in all HIV-infected individuals. On the basis of evidence that Tregs suppress virus-specific immune responses, it has been proposed that these cells are deleterious during HIV infection. Conversely, some authors suggested that Tregs could be beneficial, par-ticularly during early HIV infection when effec-tor T cells are not yet activated, as these cells limit immune activation, thus controlling the availability of HIV targets as well as prevent-ing immune-based pathologies [150,151]. The fre-quency of Tregs in peripheral blood mononuclear cells and gut-associated lymphoid tissue using CD4+Foxp3+ and CD4+Foxp3+CD127Low/- as markers was found to be higher in HIV patients than in controls [152]. Conversely, other authors reported that while HIV ‘elite controllers’ and uninfected individuals had similar Treg num-bers and frequencies, the absolute numbers of Tregs declined in blood and gut-associated lym-phoid tissue in patients with chronic progressive HIV-1 infection, but was largely normalized by HAART. However, HIV-1 infection was not associated with an impairment of ex vivo sup-pressive function of flow-sorted Tregs in either HIV controllers or untreated chronic progres-sors [153,154]. It is possible that Tregs contribute to the low number of repopulating CD4 T cells in patients under HAART, since a higher ratio of Tregs to naive CD4 T cells was observed in these patients. The role of Tregs seems to be

related rather to the control of homeostasis and cell proliferation of naive CD4 T cells, than to immune activation [155]. Alterations in Treg fre-quency in HIV/AIDS are more directly related to the degree of CD4 depletion than to viremia [156]. The relative frequency of Tregs directly correlate with HIV viral load and inversely with CD4+ counts. However, the absolute number of these cells was reduced in HIV-infected patients compared with healthy controls (p < 0.0001), with the exception of elite controllers (p > 0.05). The loss of absolute Treg numbers coincided with increased markers of immune activation (p < 0.0006). The initiation of antiviral therapy significantly increased absolute Treg numbers (p < 0.0031). The authors found that the expres-sion of CD39, a newly defined ectonucleotidase with immunomodulatory functions in Tregs, correlated with progressive HIV disease, HIV viral load and immune activation. Of note, when Tregs were tested in peripheral blood mononu-clear cells of healthy volunteers, their in vitro capacity to suppress T-cell proliferation was limited to CD4+CD25highCD39+ T cells [157]. The sustained increase of the peripheral Treg pool in IL-2-treated HIV patients may account for the unexpected clinical observation that patients with the greatest expansion of CD4+ T cells had the highest relative risk of clinical progression to AIDS [158]. Although most HIV-specific CD8+ T cells lose proliferative capacity during chronic infection, T cells restricted by HLA-B*27 or HLA-B*57 allele groups do not. CD8+ T cells restricted by ‘protective’ HLA allele groups are not suppressed by Treg cells. HLA-B*27- and HLA-B*57-restricted effec-tors also evade Treg-cell-mediated suppression by directly killing Treg cells [159]. In monkeys, tissue myeloid DCs from SIV-infected animals

Target HIV-1 antigen to DC via DC-SIGN receptor

gag, pol, envand nefantigens

TLR ligands

Surface modification:PEGylated lipid layer

gp120

Targeting moietiesanti-DC-SIGN:

gp120 or Abs anti-DC-SIGN

DC-SIGN receptor

Adaptive immuneresponse

Antigen-presentingcell

T cell

Nanomedicine © Future Science Group (2012)

Figure 5. Targeting HIV-1 antigen to dendritic cells via dC-sIGN receptor. Schematic representation of a new strategy in the design of next-generation anti-HIV-1 vaccines.

Page 14: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1604 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

exhibit an enhanced capability to induce Tregs and may contribute to the accumulation of these cells in lymphoid tissues during progres-sive infection [160]. Furthermore, Nef protein is a potent factor for increasing Treg numbers in vitro [161]. In HIV-1-infected progressors and AIDS patients, the T-cell costimulatory mol-ecule programmed death-1 (PD-1) ligand-1 (B7-H1) is significantly upregulated on peripheral mDCs, but is maintained at a relatively low level in long-term nonprogressors. When HAART achieved complete immune reconstitution by full suppression of HIV-1 replication and sub-stantial increases of CD4 T-cell counts, a cor-relation with a decrease in B7-H1 expression was found. Interestingly, X4 HIV-1 isolates directly induce B7-H1 expression on mDCs in vitro, while the addition of antiviral agents hamper this B7-H1 upregulation [162]. Myeloid-derived suppressor cells (a kind of inhibitory DC) pres-ent elevated levels in chronic uncontrolled HIV infection. Such levels potentially contribute to the impaired T-cell responses characteristic in this progressive stage of the disease [163]. HAART seems to enhance the DC population, increasing counts at week 60 post-therapy and achieving similar levels to healthy controls, but is still lower than those of long-term nonprogres-sors. During HAART treatment, DC counts correlate directly with CD4 counts [164]. Taken together, all these results indicate that interac-tion of B7-H1–PD-1 with APCs and T cells correlates with the impairment of CD4+ T cells and CTL responses in vivo. This is associated with the progression of the disease in infected individuals. Blockade of this pathway may have therapeutic implications for HIV-infected patients [165].

In a murine model of AIDS, which may not fully mimic the disease in humans, following in vivo depletion of CD4 Tregs and/or selec-tive interruption of PD-1-negative signaling in the CD8 T-cell compartment, retroviral pathogenesis was significantly decreased upon the combined treatment of CD4 Treg deple-tion and PD-1 blockade, which worked in a synergistic fashion to substantially reduce the induction of murine AIDS [166]. A combina-tion of 4-1BBL and CD40L overexpression on DCs dramatically enhances CD4+ and CD8+ T-cell responses. Signaling through 4-1BB, but not through CD40, can alleviate the suppres-sive effect of Tregs on CD8+ T-cell proliferation. Thus, the combination of 4-1BBL and CD40L enhances HIV-specific CD8+ T-cell responses in a synergistic way [167]. However, the complexity

of immune regulation in HIV-1 infection, as shown above, demonstrates that applying Treg immunotherapeutic strategies tested in dis-eases other than HIV could involve the risk of inducing further progression of HIV infection. Therefore, extreme care must be taken when designing such studies [168].

ConclusionThe current developments in DC-based vaccines against HIV-1 infection or to treat AIDS patients hold promise for improving the control of HIV infection. The ex vivo production and activation of DCs is expensive and technically complex. Therefore, the use of this technique in broad-scale immunization programs is not viable. Vaccines that target DCs in vivo provide an interesting alternative with a dramatically reduced cost with respect to the in vitro obtention of DCs and activa-tion systems, thereby facilitating their application across the wide geographical and socioeconomic range of people infected by the virus.

As outlined in this review, the huge efforts devoted over recent decades to the design of effec-tive immunotherapies for HIV infection have failed to deliver effective vaccines. However, promising results have been obtained. In fact, the experiences from DC therapy studies in cancer show that there could be many setbacks before advances are made because of the numerous factors that potentially affect DC immunotherapy. Nevertheless, we should learn from experience in developing a DC-based immunotherapy for the fearsome enemy of cancer when pursuing a DC immunotherapeutic vaccine against the equally formidable foe, HIV [169].

It is to be hoped that a solution, although com-plex, will be found that takes into account virus variability, antigen design, the immunological state of the patients and the appropriate receptor targets that ensure DC activation, maturation and MHC class I and II presentation, possibly by several pathways, together with strong adju-vants that help the immunological response and maintenance. The administration route and the delivery systems of the vaccine are two addi-tional key factors that will determine whether an effective immunotherapy for HIV infection is achieved. Given the intense dedication of the hundreds of research groups cited in this review and many others, the authors hope the promise of DC-targeted therapy for HIV infection will become a reality in the not too distant future.

Future perspectiveThe history of modern medicine and possi-bly of humankind has changed thanks to the

Page 15: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1605future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

executive summary

� Dendritic cell (DC)-based vaccines for HIV have been shown to be the most effective strategy to induce an effective and prolonged immunological response; however, this response is still insufficient. Furthermore, the generation of DCs ex vivo is not suitable for broad vaccination programs owing to cost and technical difficulties.

� A series of presentation strategies for HIV-1 vaccinations, both with respect to antigens as well as to more appropriate vehicles, are presented.

� Several strategies to activate HIV-1 antigen presentation to DCs, by means of whole inactivated virus, proteins and peptides or by DNA or mRNA, are also presented.

� Appropriate ligands to target antigens in vivo to DCs and the best routes to gain access to MHC class-I and -II pathways, as well as the current delivery systems used are reviewed.

� Negative regulatory mechanisms associated with HIV-1 infection and that could impair the effectivity of the vaccines in development are described.

development of vaccines against multiple patho-gens. However, the variability of some viruses, such as HIV, has made the generation of effec-tive vaccines elusive. With current knowledge, it is easier to envisage a curative vaccine than a preventive one, because it is possible to charac-terize HIV variants in an infected patient, but very difficult to develop a vaccine covering all the HIV-1 subtypes or clades. However, these dif-ficulties were overcome in other highly variable viruses, such as influenza virus subtypes.

HIV-1 infection defines a complex and evolv-ing situation with marked differences between each stage of the disease and genetic background of the individuals, but future directions in vac-cine development should uncover several crucial aspects that are still poorly understood.

Depending on the duration and magnitude of antigenic activation, chronic HIV infection involves T-cell exhaustion, which is character-ized by a progressive loss of T-cell function. It is therefore important to identify/study the factors that are implicated in the maintenance of this situation and to control them for a better vac-cination schedule. Furthermore, as T-cell acti-vation is associated with an increase in viremia and disease progression, it would be appropri-ate to direct vaccines that specifically activate CD8 T cells, thus controlling CD4 activation by maintaining Tregs (since CD4 activation or a decreased number of Tregs have been associated with HIV-1 replication).

DCs have been shown to be the most suitable tools for the development of efficient immune responses against HIV-1; however, ex vivo gen-eration and manipulation of these cells for their use as vaccines is expensive, time-consuming and technically difficult. Therefore, new vac-cines should be designed to target DCs in vivo. This strategy should not imply a lower response, since targeted antigens in nanoparticles, nano-liposomes or polymers to DC receptors allows the use of several kinds of antigens and protects

them from degradation and dispersion in blood. In addition, some receptors on DCs trigger acti-vation and maturation signals to improve antigen presentation by the desired route. However, HIV infection has been associated with elevated levels of myeloid-derived suppressor cells, a type of DC that induces Treg development and potentially contributes to impaired T-cell responses. The expression of B7-H1 by DCs (a PD-1 ligand that induces apoptosis in CD8 T cells) and CTLA-4 by T cells (an inhibitory receptor that binds costimulatory molecules CD80 and CD86 but inhibits T-cell responses) could impair vaccine responses. Therefore, an appropriate choice of adjuvant could help to reverse this inhibitory sit-uation, and blockade of this molecule with anti-bodies may improve vaccine immune activation.

Finally, we recommend that efforts be focused on finding the vaccine administration route that, in addition to CD8 T-cell-specific activation, pro-duces high levels of neutralizing antibodies, since the combination of cellular and humoral activity is essential to achieve satisfactory results. Neutralizing antibodies generate immune complexes that activate DCs and reinforce memory T cells.

Although our understanding of the com-plex regulation associated with HIV-1 infec-tion is still in its infancy, our knowledge of the mechanisms involved advances daily. Thus, it is expected that the near future will bring about successful manipulation of natural antiviral activity and effective vaccination strategies.

Financial & competing interests disclosureThe authors have no relevant affiliations or financial involvement with any organization or entity with a finan-cial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

Page 16: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1606 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

referencesPapers of special note have been highlighted as:n of interestnn of considerable interest

1 Douek DC, Picker LJ, Koup RA. T cell dynamics in HIV-1 infection. Annu. Rev. Immunol. 21, 265–304 (2003).

2 Domingo P, Torres-Torronteras J, Pomar V et al. Uridine metabolism in HIV-1-infected patients: effect of infection, of antiretroviral therapy and of HIV-1/ART-associated lipodystrophy syndrome. PLoS One 5(11), e13896 (2010).

3 Berzofsky JA, Ahlers JD, Janik J et al. Progress on new vaccine strategies against chronic viral infections. J. Clin. Invest. 114, 450–462 (2004).

4 Egan MA. Current prospects for the development of a therapeutic vaccine for the treatment of HIV type 1 infection. AIDS Res. Hum. Retrovir. 20, 794–806 (2004).

5 Palmisano L, Vella S. A brief history of antiretroviral therapy of HIV infection: success and challenges. Ann. Ist Super Sanita. 47(1), 44–48 (2011).

6 Lévy Y, Gahéry-Ségard H, Durier C et al. Immunological and virological efficacy of a therapeutic immunization combined with interleukin-2 in chronically HIV-1 infected patients. AIDS 19, 279–286 (2005).

7 Lu W, Arraes LC, Ferreira WT, Andrieu JM. Therapeutic dendritic- cell vaccine for chronic HIV-1 infection. Nat. Med. 10, 1359–1365 (2004).

nn� One of the first promising results showing the activity of vaccines based on dendritic cells in reducing viral load in a significant number of patients.

8 Leitner T. Genetic subtypes of HIV-1. In: Human Retrovirus and AIDS: a Compilation of Nucleic Acids and Amino Acid Sequences. Myers G, Korber BT, Foley BT, Jeang KT, Mellors J, Wain-Hobson S (Eds). Los Alamos National Laboratory, NM, USA, III28 (1996).

9 Fanales-Belasio E, Moretti S, Nappi F 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. 168(1), 197–206 (2002).

10 Izmailova E, Bertley FM, Huang Q et al. HIV-1 Tat reprograms immature dendritic cells to express chemoattractants for activated T cells and macrophages. Nat. Med. 9(2), 191–197 (2003).

n� Shows the usefulness of Tat as one of the HIV proteins to be incorporated in vaccines, since it helps dendritic cell maturation and activation.

11 García F, Routy JP. Challenges in dendritic cells-based therapeutic vaccination in HIV-1 infection workshop in dendritic cell-based vaccine clinical trials in HIV-1. Vaccine 29(38), 6454–6463 (2011).

12 Patham B, Simmons GL, Subramanya S. Advances in dendritic cell-based vaccines for HIV. Curr. Med. Chem. 18(26), 3987–3994 (2011).

13 Goldstein G, Chicca II JJ. A universal anti-HIV-1 Tat epitope vaccine that is fully synthetic and self-adjuvanting. Vaccine 28, 1008–1014 (2010).

n� Shows the usefulness of Tat as one of the HIV proteins to be incorporated in vaccines, since it helps dendritic cell maturation and activation.

14 Wu L, Yang ZY, Xu L et al. Cross-clade recognition and neutralization by the V3 region from clade C human immunodeficiency virus-1 envelope. Vaccine 24, 4995–5002 (2006).

15 Lu L, Zhu Y, Diao J, Wang Z, Chen YH. V3 CTL epitope density in a single recombinant molecule antigen differentially affects the number and activity of primary and memory CD8+ T cells. Vaccine 26, 845–852 (2008).

16 Rubinstein A, Goldstein H, Pettoello-Mantovani M et al. Safety and immunogenicity of a V3 loop synthetic peptide conjugated to purified protein derivative in HIV-seronegative volunteers. AIDS 9, 243–251 (1995).

17 Yamanaka T, Fujimura Y, Ishimoto S et al. Correlation of titer of antibody to principal neutralizing domain of HIVMN strain with disease progression in Japanese hemophiliacs seropositive for HIV type 1. AIDS Res. Hum. Retroviruses 4, 317–326 (1997).

n� One of the first articles showing a correlation between the presence of neutralizing antibodies and disease progression.

18 Berzofsky JA. Development of artificial vaccines against HIV using defined epitopes. FASEB J. 5, 2412–2418 (1991).

19 Pantophlet R, Wrin T, Cavacini LA, Robinson JE, Burton DR. Neutralizing activity of antibodies to the V3 loop region of HIV-1 gp120 relative to their epitope fine specificity. Virology 381(2), 251–260 (2008).

20 de Souza MS, Ratto-Kim S, Chuenarom W et al. Thai Phase III trial (RV144) vaccine regimen induces T cell responses that preferentially target epitopes within the V2 region of HIV-1 envelope. J. Immunol. 188(10), 5166–5176 (2012).

nn� Update of the results of the first preventive HIV vaccine protocol that has shown some effectivity in an uninfected Thai population.

21 Montefiori DC, Karnasuta C, Huang Y et al. Magnitude and breadth of the neutralizing antibody response in the RV144 and Vax003 HIV-1 Vaccine Efficacy Trials. J. Infect. Dis. 206(3), 431–441 (2012).

nn� Update of the results of the first preventive HIV vaccine protocol that has shown some effectivity in an uninfected Thai population.

22 Haynes BF, Moody MA, Liao HX, Verkoczy L, Tomaras GD. B cell responses to HIV-1 infection and vaccination: pathways to preventing infection. Trends Mol. Med. 17(2), 108–116 (2011).

23 Spearman P, Kalams S, Elizaga M et al. Safety and immunogenicity of a CTL multiepitope peptide vaccine for HIV with or without GM-CSF in a Phase I trial. Vaccine 27(2), 243–249 (2009).

24 Bråve A, Hallengärd D, Malm M et al. Combining DNA technologies and different modes of immunization for induction of humoral and cellular anti-HIV-1 immune responses. Vaccine 27(2), 184–186 (2009).

25 Barouch DH, O’Brien KL, Simmons NL et al. Mosaic HIV-1 vaccines expand the breadth and depth of cellular immune responses in rhesus monkeys. Nat. Med. 16(3), 319–323 (2010).

26 Dittmer U, Werner T, Kraft AR. Co-immunization of mice with a retroviral DNA vaccine and GITRL-encoding plasmid augments vaccine-induced protection against retrovirus infection. Viral Immunol. 21(4), 459–467 (2008).

27 Schilling R, Heil A, Langner K et al. A multivalent HIV-vaccine: development of a plasmid DNA for the expression of HIV envelope glycoproteins with hypervariable V3-loop domains. Vaccine 24, 4648–4650 (2006).

28 Lu S. Immunogenicity of DNA vaccines in humans. Human Vaccines 4(6), 449–452 (2008).

29 Romain G, van Gulck E, Epaulard O et al. CD34-derived dendritic cells transfected ex vivo with HIV-Gag mRNA induce polyfunctional T-cell responses in nonhuman primates. Eur. J. Immunol. doi:10.1002/eji.201242478 (2012) (Epub ahead of print).

30 Allard SD, De Keersmaecker B, de Goede AL et al. A Phase I/IIa immunotherapy trial of HIV-1-infected patients with Tat, Rev and Nef expressing dendritic cells followed by treatment interruption. Clin. Immunol. 142(3), 252–268 (2012).

31 Van Gulck E, Vlieghe E, Vekemans M et al. mRNA-based dendritic cell vaccination induces potent antiviral T-cell responses in HIV-1-infected patients. AIDS 26(4), F1–12 (2012).

Page 17: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1607future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

1607www.futuremedicine.com

32 Toledo H, Baly A, Castro O et al. A Phase I clinical trial of a multi-epitope polypeptide TAB9 combined with Montanide ISA 720 adjuvant in non-HIV-1 infected human volunteers. Vaccine 19(30), 4328–4336 (2001).

33 Rosenberg ES, Graham BS, Chan ES et al. Safety and immunogenicity of therapeutic DNA vaccination in individuals treated with antiretroviral therapy during acute/early HIV-1 infection. PLoS One 5(5), e10555 (2010).

n� This team was one of the first to use DNA vaccination, and this reference updates the results of a Phase I/II, randomized, placebo-controlled, double-blinded trial showing the safety of this type of vaccine.

34 Jin X, Newman MJ, De-Rosa S et al. A novel HIV T helper epitope-based vaccine elicits cytokine-secreting HIV-specific CD4+ T cells in a Phase I clinical trial in HIV-uninfected adults. Vaccine 27(50), 7080–7086 (2009).

35 Cruz LJ, Iglesias E, Aguilar JC et al. A comparative study of different presentation strategies for an HIV peptide immunogen. Bioconjug. Chem. 15, 112–120 (2004).

36 Jones T. Vacc-4x, a therapeutic vaccine comprised of four engineered peptides for the potential treatment of HIV infection. Curr. Opin. Investig. Drugs 11(8), 964–970 (2010).

37 Iglesias E, Aguilar JC, Cruz LJ, Reyes O. Broader cross-reactivity after conjugation of V3 based multiple antigen peptides to HBsAg. Mol. Immunol. 42, 99–104 (2005).

n� With [38], shows the effectiveness of different constructs of synthetic peptides with several antigenic epitopes as vaccines.

38 Cruz LJ, Iglesias E, Garay HE, Besada V, Reyes O. Linear polymerization of a synthetic peptide of the V3 region from HIV-1 JY1 isolate using acetamidomethyl-protected thiol groups of cysteine residues. Biotecnol. Aplic. 17, 35–38 (2000).

39 Cruz LJ, Padron R, Gonzalez LJ et al. Peptide synthesis containing a B-cell and a T-cell epitope on dextran beads and evaluation of humoral response against bead-peptide construct. Lett. Pept. Sci. 7, 229–237 (2001).

40 Cruz LJ, Iglesias E, Aguilar JC et al. Study of different coupling agents in the conjugation of a V3-based synthetic MAP to carrier proteins. J. Pept. Sci. 7, 511–518 (2001).

41 Cruz LJ, Iglesias E, Aguilar JC et al. Different immune response of mice immunized with conjugates containing multiple copies of either consensus or mixotope versions of the V3 loop peptide from Human Immunodeficiency Virus type 1. Bioconjug. Chem. 15, 1110–1117 (2004).

42 Cruz LJ, Cabrales A, Iglesias E, Aguilar JC, González LJ, Reyes O. Enhanced immunogenicity and cross-reactivity of HIV-1 V3-peptide and multiple antigen peptides conjugated to distinct carrier proteins. Int. Immunopharmacol. 9(12), 1452–1459 (2009).

43 Gras-Masse H, Boutillon C, Dieses E, Deprez B, Tartar A. Confronting the degeneracy of convergent combinatorial immunogens, or ‘mixotopes’, with the specificity of recognition of the target sequences. Vaccine 15, 1568–1578 (1997).

44 Thammavongsa V, Schaefer M, Filzen T et al. Assembly and intracellular trafficking of HLA-B*3501 and HLA-B*3503. Immunogenetics 61(11–12), 703–716 (2009).

45 Smith AD, Geisler SC, Chen AA et al. Human rhinovirus type 14: human immunodeficiency virus type 1 (HIV-1) V3 loop chimeras from a combinatorial library induce potent neutralizing antibody responses against HIV-1. J. Virol. 72, 651–659 (1998).

46 Resnick DA, Smith AD, Gesiler SC et al. Chimeras from a human rhinovirus 14-human immunodeficiency virus type 1 (HIV-1) V3 loop seroprevalence library induce neutralizing responses against HIV-1. J. Virol. 4, 2406–2411 (1995).

47 Hart MK, Palker TJ, Haynes BF. Design of experimental synthetic peptide immunogens for prevention of HIV-1 and HTLV-I retroviral infections. Pharm. Biotechnol. 6, 821–845 (1995).

48 Pitisuttithum P, Gilbert P, Gurwith M et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J. Infect. Dis. 194, 1661–1671 (2006).

nn� Preventive vaccines showing good immunological response but not able to demonstrate prevention of HIV-1 infection.

49 Gilbert PB, Peterson ML, Follmann D et al. Correlation between immunologic responses to a recombinant glycoprotein 120 vaccine and incidence of HIV-1 infection in a Phase 3 HIV-1 preventive vaccine trial. J. Infect. Dis. 191, 666–677 (2005).

nn� Preventive vaccines showing good immunological response but not able to demonstrate prevention of HIV-1 infection.

50 Salmon-Céron D, Durier C, Desaint C et al. Immunogenicity and safety of an HIV-1 lipopeptide vaccine in healthy adults: a Phase 2 placebo-controlled ANRS trial. AIDS 24(14), 2211–2223 (2010).

nn� Preventive vaccines showing good immunological response but not able to demonstrate prevention of HIV-1 infection.

51 Zhou Y, Bosch ML, Salgaller ML. Current methods for loading dendritic cells with tumor antigen for the induction of antitumor immunity. J. Immunother. 25, 289–303 (2002).

52 Kalams SA, Walker BD. The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J. Exp. Med. 188, 2199–2204 (1998).

53 Yoshida A, Tanaka R, Murakami T et al. Induction of protective immune responses against R5 human immunodeficiency virus type 1 (HIV-1) infection in hu-PBL-SCID mice by intrasplenic immunization with HIV-1-pulsed dendritic cells: possible involvement of a novel factor of human CD4+ T-cell origin. J. Virol. 77(16), 8719–8728 (2003).

54 Lapenta C, Santini SM, Logozzi M et al. Potent immune response against HIV-1 and protection from virus challenge in hu-PBL-SCID mice immunized with inactivated virus-pulsed dendritic cells generated in the presence of IFN-alpha. J. Exp. Med. 198(2), 361–367 (2003).

55 Lu W, Wu X, Lu Y, Guo W, Andrieu JM. Therapeutic dendritic-cell vaccine for simian AIDS. Nat. Med. 9(1), 27–32 (2003).

56 Andrieu JM, Lu W. A dendritic cell-based vaccine for treating HIV infection: background and preliminary results. J. Intern. Med. 1(2), 123–131 (2007).

57 Whiteside TL, Piazza P, Reiter A et al. Production of a dendritic cell-based vaccine containing inactivated autologous virus for therapy of patients with chronic human immunodeficiency virus type 1 infection. Clin. Vaccine Immunol. 16(2), 233–240 (2009).

58 García F, Climent N, Assoumou L et al. A therapeutic dendritic cell-based vaccine for HIV-1 infection. J. Infect. Dis. 203(4), 473–478 (2011).

59 Huang XL, Fan Z, Zheng L et al. Priming of human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T cell responses by dendritic cells loaded with HIV-1 proteins. J. Infect. Dis. 187, 315–319 (2003).

60 Alexaki A, Wigdahl B. HIV-1 infection of bone marrow hematopoietic progenitor cells and their role in trafficking and viral dissemination. PLoS Pathogens 4(12), e1000215 (2008).

61 Fantuzzi L, Purificato C, Donato K, Belardelli F, Gessani S. Human immunodeficiency virus type 1 gp120 induces abnormal maturation and functional alterations of dendritic cells: a novel mechanism for AIDS pathogenesis. J. Virol. 78(18), 9763–9772 (2004).

Page 18: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1608 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

62 Fanales-Belasio E, Cafaro A, Cara A et al. HIV-1 Tatbased vaccines: from basic science to clinical trials. DNA Cell Biol. 21, 599–610 (2002).

63 Gavioli R, Gallerani E, Fortini C et al. HIV-1 tat protein modulates the generation of cytotoxic T cell epitopes by modifying proteasome composition and enzymatic activity. J. Immunol. 173, 3838–3843 (2004).

64 Kasper MR, Collins KL. Nef-mediated disruption of HLA-A2 transport to the cell surface in T-cells. J. Virol. 77, 3041–3049 (2003).

65 Yang OO, Nguyen PT, Kalams SA et al. Nef-mediated resistance of human immunodeficiency virus type 1 to antiviral cytotoxic T lymphocytes. J. Virol. 76, 1626–1631 (2002).

66 Stumptner-Cuvelette P, Morchoisne S, Dugast M et al. HIV-1 Nef impairs MHC class II antigen presentation and surface expression. Proc. Natl Acad. Sci. USA 98(21), 12144–12149 (2001).

67 Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J. Exp. Med. 184(2), 465–472 (1996).

68 Mitchell DA, Nair SK. RNA-transfected dendritic cells in cancer immunotherapy. J. Clin. Invest. 106, 1065–1069 (2000).

69 Tillman BW, Hayes TL, DeGruijl TD, Douglas JT, Curiel DT. Adenoviral vectors targeted to CD40 enhance the efficacy of dendritic cell-based vaccination against human papillomavirus 16-induced tumor cells in a murine model. Cancer Res. 60, 5456–5463 (2000).

70 Gilboa E, Vieweg J. Cancer immunotherapy with mRNA transfected dendritic cells. Immunol. Rev. 199, 251–263 (2004).

71 Brown K, Gao W, Alber S et al. Adenovirus-transduced dendritic cells injected into skin or lymph node prime potent simian immunodeficiency virus-specific T cell immunity in monkeys. J. Immunol. 171, 6875–6882 (2003).

72 Xin KQ, Hoshino Y, Toda Y et al. Immunogenicity and protective efficacy of orally administered recombinant Lactococcus lactis expressing surface-bound HIV Env. Blood 102, 223–228 (2003).

73 Kappes JC, Wu X. Safety considerations in vector development. Somat. Cell Mol. Genet. 26, 147–158 (2001).

74 Rudolf MP, Fausch SC, Da Silva DM, Kast WM. Human dendritic cells are activated by chimeric human papillomavirus type-16 viruslike particles and induce epitope-specific human T cell responses in vitro. J. Immunol. 166, 5917–5924 (2001).

75 Touze A, Coursaget P. In vitro gene transfer using human papillomavirus-like particles. Nucleic Acids Res. 26(5), 1317–1323 (1998).

76 Chaput N, Taïeb J, André F, Zitvogel L. The potential of exosomes in immunotherapy. Expert Opin. Biol. Ther. 5(6), 737–747, (2005).

77 Li X, Zhang Z, Beiter T, Schluesener HJ. Nanovesicular vaccines: exosomes. Arch. Immunol. Ther. Exp. (Warsz) 53(4), 329–235 (2005).

78 André F, Chaput N, Schartz NE et al. Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J. Immunol. 172(4), 2126–2136 (2004).

79 Wingard JB, Anderson B, Weissman D. Induction of HIV-specific T and B cell responses with a replicating and conditionally infectious lentiviral vaccine. Eur. J. Immunol. 38(5), 1310–1320 (2008).

80 Coren LV, Shatzer T, Ott DE. CD45 immunoaffinity depletion of vesicles from Jurkat T cells demonstrates that exosomes contain CD45: no evidence for a distinct exosome/HIV-1 budding pathway. Retrovirology 16, 50–64 (2008).

81 Wiley RD, Gummuluru S. Immature dendritic cell-derived exosomes can mediate HIV-1 transinfection. Proc. Natl Acad. Sci. USA 103(3), 738–743 (2006).

82 Nanjundappa RH, Wang R, Xie Y et al. GP120-specific exosome-targeted T cell-based vaccine capable of stimulating DC- and CD4+ T-independent CTL responses. Vaccine 29(19), 3538–3547 (2011).

83 Li M, Aliotta JM, Asara JM et al. Quantitative proteomic analysis of exosomes from HIV-1-infected lymphocytic cells. Proteomics 12(13), 2203–2211 (2012).

84 Lenassi M, Cagney G, Liao M et al. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 11(1), 110–122 (2010).

85 Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 7(10), 790–802 (2007).

86 Tang J, Yin R, Tian Y et al. A novel self-assembled nanoparticle vaccine with HIV-1 Tat(49–57)/HPV16 E7(49–57) fusion peptide and GM-CSF DNA elicits potent and prolonged CD8+ T cell-dependent anti-tumor immunity in mice. Vaccine 30(6), 1071–1082 (2011).

87 Jacobs JFM, Pereira CF, Tacken PJ et al. Ex vivo-generated dendritic cells for clinical trials versus in vivo targeting to dendritic cells: critical issues. In: The Biology of Dendritic Cells and HIV Infection. Gessani S, Belardelli F (Eds). Springer Science + Business Media, Rome, Italy (2007).

88 Tacken PJ, Torensma R, Figdor CG. Targeting antigens to dendritic cells in vivo. Immunobiology 211(6–8), 599–608 (2006).

89 Moron G, Dadaglio G, Leclerc C. New tools for antigen delivery to the MHC class I pathway. Trends Immunol. 25, 92–97 (2004).

90 Jeannin P, Renno T, Goetsch L et al. OmpA targets dendritic cells, induces their maturation and delivers antigen into the MHC class I presentation pathway. Nat. Immunol. 1(6), 502–509 (2000).

91 Kawai T, Akira1 S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 11(5), 373–384 (2010).

92 Liu G, Zhao Y. Toll-like receptors and immune regulation: their direct and indirect modulation on regulatory CD4+ CD25+ T cells. Immunology 122, 149–156 (2007).

93 Spaner DE, Masellis A. Toll-like receptor agonists in the treatment of chronic lymphocytic leukemia. Leukemia 21, 53–60 (2007).

94 Equils O, Schito ML, Karahashi H et al. Toll-like receptor 2 (TLR2) and TLR9 signaling results in HIV-long terminal repeat trans-activation and HIV replication in HIV-1 transgenic mouse spleen cells: implications of simultaneous activation of TLRs on HIV replication. J. Immunol. 170, 5159–5164 (2003).

95 Breckpot K, Escors D, Arce F et al. HIV-1 lentiviral vector immunogenicity is mediated by Toll-like receptor 3 (TLR3) and TLR7. J. Virol. 84(11), 5627–5636 (2010).

96 Apostolopoulos V, Pietersz GA, Gordon S, Martinez-Pomares L, McKenzie IF. Aldehyde-mannan antigen complexes target the MHC class I antigen-presentation pathway. Eur. J. Immunol. 30, 1714–1723 (2000).

97 Frison N, Taylor ME, Soilleux E et al. Oligolysine-based oligosaccharide clusters: selective recognition and endocytosis by the mannose receptor and dendritic cell-specific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin. J. Biol. Chem. 278, 23922–23929 (2003).

98 Nigou J, Zelle-Rieser C, Gilleron M, Thurnher M, Puzo G. Mannosylated lipoarabinomannans inhibit IL-12 production by human dendritic cells: evidence for a negative signal delivered through the mannose receptor. J. Immunol. 166, 7477–7485 (2001).

99 He LZ, Crocker A, Lee J et al. Antigenic targeting of the human mannose receptor induces tumor immunity. J. Immunol. 178(10), 6259–6267 (2007).

100 Guo M, Gong S, Maric S et al. A monoclonal antibody to the DEC-205 endocytosis

Page 19: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

www.futuremedicine.com 1609future science group

Enhancing immunogenicity & cross-reactivity of HIV-1 antigens by in vivo targeting to DCs Review

1609www.futuremedicine.com

receptor on human dendritic cells. Hum. Immunol. 61, 729–738 (2000).

101 Bozzacco L, Trumpfheller C, Huang Y et al. HIV gag protein is efficiently cross-presented when targeted with an antibody towards the DEC-205 receptor in Flt3 ligand-mobilized murine DC. Eur. J. Immunol. 40(1), 36–46 (2010).

102 Tacken PJ, Figdor CG. Targeted antigen delivery and activation of dendritic cells in vivo: steps towards cost effective vaccines. Semin. Immunol. 23(1), 12–20 (2011).

103 Thériault JR, Mambula SS, Sawamura T, Stevenson MA, Calderwood SK. Extracellular HSP70 binding to surface receptors present on antigen presenting cells and endothelial/epithelial cells. FEBS Lett. 579(9), 1951–1960 (2005).

104 Delneste Y, Magistrelli G, Gauchat J et al. Involvement of LOX-1 in dendritic cell-mediated antigen crosspresentation. Immunity 17, 353–362 (2002).

105 Idoyaga J, Lubkin A, Fiorese C et al. Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A. Proc. Natl Acad. Sci. USA 108(6), 2384–2389 (2011).

106 Martinon F, Dereuddre-Bosquet N, Méderlé-Mangeot I et al. Directing macaque immune responses with an anti-dendritic cell HIV Gag p24 fusion protein vaccine. Retrovirology 6(Suppl. 3), 346 (2009).

107 Soilleux EJ, Morris LS, Leslie G et al. Constitutive and induced expression of DC-SIGN on dendritic cell and macrophage subpopulations in situ and in vitro. J. Leukoc. Biol. 71, 445–457 (2002).

108 Gieseler RK, Marquitan G, Hahn MJ et al. DC-SIGN-specific liposomal targeting and selective intracellular compound delivery to human myeloid dendritic cells: implications for HIV disease. Scand. J. Immunol. 59, 415–424 (2004).

109 Korokhov N, de Gruijl TD, Aldrich WA et al. High efficiency transduction of dendritic cells by adenoviral vectors targeted to DC-SIGN. Cancer Biol. Ther. 4(3), 289–294 (2005).

110 Tacken PJ, de Vries IJ, Gijzen K et al. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood 106(4), 1278–1285 (2005).

111 Moris A, Nobile C, Buseyne F, Porrot F, Abastado J-P, Schwartz O. DC-SIGN promotes exogenous MHC-I-restricted HIV-1 antigen presentation. Blood 103, 2648–2654 (2004).

112 Poulin LF, Salio M, Griessinger E et al. Characterization of human DNGR-1+

BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J. Exp. Med. 207(6), 1261–1271 (2010).

113 Joffre OP, Sancho D, Zelenay S, Keller AM, Reis e Sousa C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A. Eur. J. Immunol. 40(5), 1255–1265 (2010).

114 Bonifaz LC, Bonnyay DP, Charalambous A et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J. Exp. Med. 199, 815–824 (2004).

115 Hudson PJ, Souriau C. Engineered antibodies. Nat. Med. 9, 129–134 (2003).

116 Steinman RM, Bonifaz L, Fujii S et al. The innate functions of dendritic cells in peripheral lymphoid tissues. Adv. Exp. Med. Biol. 560, 83–97 (2005).

117 Grousson J, Peguet-Navarro J. Functional relevance of CD40 expression in human epidermis. Cell Biol. Toxicol. 14, 345–350 (1998).

118 Loof N, Montes M, Flamar A et al. HIV-specific responses induced by anti-CD40 targeting antibodies. Retrovirology 6(Suppl. 3), O46 (2009).

119 Flamar A, Zurawski S, Cobb A et al. Targeting HIV peptides to human dendritic cells via CD40 elicits expansion of multi-epitope polyfunctional CD4+ and CD8+ T cells in HIV patients. Retrovirology 6(Suppl. 3), P286 (2009).

120 Serre K, Giraudo L, Leserman L, Machy P. Liposomes targeted to Fc receptors for antigen presentation by dendritic cells in vitro and in vivo. Methods Enzymol. 373, 100–118 (2003).

121 Ortiz-Stern A, Rosales C. Cross-talk between Fc receptors and integrins. Immunol. Lett. 90, 137–143 (2003).

122 Schuurhuis DH, Ioan-Facsinay A, Nagelkerken B et al. Antigen–antibody immune complexes empower dendritic cells to efficiently prime specific CD8+ CTL responses in vivo. J. Immunol. 168, 2240–2246 (2002).

123 Kalergis AM, Ravetch JV. Inducing tumor immunity through the selective engagement of activating Fcg receptors on dendritic cells. J. Exp. Med. 195, 1653–1659 (2002).

124 Pasquier B, Launay P, Kanamaru Y et al. Identification of FcalphaRI as an inhibitory receptor that controls inflammation: dual role of FcRgamma ITAM. Immunity 22, 31–42 (2005).

125 Wallace PK, Tsang KY, Goldstein J et al. Exogenous antigen targeted to FcgammaRI

on myeloid cells is presented in association with MHC class I. J. Immunol. Methods 248, 183–194 (2001).

126 Abdel-Motal UM, Wigglesworth K, Galili U. Mechanism for increased immunogenicity of vaccines that form in vivo immune complexes with the natural anti-Gal antibody. Vaccine 27(23), 3072–3082 (2009).

127 Forthal DN, Gilbert PB, Landucci G, Phan T. Recombinant gp120 vaccine-induced antibodies inhibit clinical strains of HIV-1 in the presence of Fc receptor-bearing effector cells and correlate inversely with HIV infection rate. J. Immunol. 178, 6596–6603 (2007).

128 Rapaka RR, Goetzman ES, Zheng M et al. Enhanced defense against Pneumocystis carinii mediated by a novel dectin-1 receptor Fc fusion protein. J. Immunol. 178, 3702–3712 (2007).

129 Zhang MY, Vu BK, Choudhary A et al. Cross-reactive human immunodeficiency virus type 1-neutralizing human monoclonal antibody that recognizes a novel conformational epitope on gp41and lacks reactivity against self-antigens. J. Virol. 82, 6869–6879 (2008).

130 Duval M, Posner MR, Cavacini LA. A bispecific antibody composed of a nonneutralizing antibody to the gp41 immunodominant region and an anti-CD89 antibody directs broad human immunodeficiency virus destruction by neutrophils. J. Virol. 82, 4671–4674 (2008).

131 Perez LG, Costa MR, Todd CA, Haynes BF, Montefiori DC. Utilization of immunoglobulin G Fc receptors by human immunodeficiency virus type 1: a specific role for antibodies against the membrane-proximal external region of gp41. J. Virol. 83(15), 7397–7410 (2009).

132 French MA, Tanaskovic S, Law MG et al. Vaccine-induced IgG2 anti-HIV p24 is associated with control of HIV in patients with a ‘high-affinity’ FcgRIIa genotype. AIDS 24(13), 1983–1990 (2010).

133 Rose NF, Marx PA, Luckay A et al. An effective AIDS vaccine based on live attenuated vesicular stomatitis virus recombinants. Cell 106, 539–549 (2001).

134 Hedley SJ, Auf der Maur A, Hohn S et al. An adenovirus vector with a chimeric fiber incorporating stabilized single chain antibody achieves targeted gene delivery. Gene Ther. 13(1), 88–94 (2006).

135 Yang L, Yang H, Rideout K et al. Engineered lentivector targeting of dendritic cells for in vivo immunization. Nat. Biotechnol. 26(3), 326–334 (2008).

136 Datta SA, Temeselew LG, Crist RM et al. On the role of the SP1 domain in HIV-1 particle assembly: a molecular switch? J. Virol. 85(9), 4111–4121 (2011).

Page 20: Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by               in vivo               targeting to dendritic cells

Nanomedicine (2012) 7(10)1610 future science group

Review Cruz, Rueda, Tacken, Albericio, Torensma & Figdor

137 Tanzi E, Esposito S, Bojanin J et al. Immunogenicity and effect of a virosomal influenza vaccine on viral replication and T-cell activation in HIV-infected children receiving highly active antiretroviral therapy. J. Med. Virol. 78(4), 440–445 (2006).

138 Hattori Y, Kawakami S, Suzuki S, Yamashita F, Hashida M. Enhancement of immune responses by DNA vaccination through targeted gene delivery using mannosylated cationic liposome formulations following intravenous administration in mice. Biochem. Biophys. Res. Commun. 317, 992–999 (2004).

139 Joshi MD, Unger WW, van Beelen AJ et al. DC-SIGN mediated antigen-targeting using glycan-modified liposomes: formulation considerations. Int. J. Pharm. 416(2), 426–432 (2011).

140 Moore A, McGuirk P, Adams S et al. Immunization with a soluble recombinant HIV protein entrapped in biodegradable microparticles induces HIV-specific CD8+ cytotoxic T lymphocytes and CD4+ Th1 cells. Vaccine 13, 1741–1749 (1995).

141 De Haes W, De Koker S, Pollard C et al. Polyelectrolyte capsules-containing HIV-1 p24 and Poly I:C modulate dendritic cells to stimulate HIV-1-specific immune responses. Mol. Ther. 18(7), 1408–1416 (2010).

142 Marradi M, Di Gianvincenzo P, Enríquez-Navas PM et al. Gold nanoparticles coated with oligomannosides of HIV-1 glycoprotein gp120 mimic the carbohydrate epitope of antibody 2G12. J. Mol. Biol. 410(5), 798–810 (2011).

143 Schlosser E, Mueller M, Fischer S et al. TLR ligands and antigen need to be coencapsulated into the same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses. Vaccine 26(13), 1626–1637 (2008).

144 Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat. Immunol. 6(8), 769–776 (2005).

145 Kasturi SP, Skountzou I, Albrecht RA et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature 470(7335), 543–547 (2011).

146 Cruz LJ, Tacken PJ, Fokkink R et al. Targeted PLGA nano – but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. J. Control Release 144(2), 118–126 (2010).

147 Tacken PJ, Zeelenberg IS, Cruz LJ et al. Targeted delivery of TLR ligands to human and mouse dendritic cells strongly enhances adjuvanticity. Blood 118(26), 6836–6844 (2011).

148 Gringhuis SI, den Dunnen J, Litjens M, van der Vlist M, Geijtenbeek TB. Carbohydrate-specific signaling through the DC-SIGN signalosome tailors immunity to Mycobacterium tuberculosis, HIV-1 and Helicobacter pylori. Nat. Immunol. 10(10), 1081–1088 (2009).

149 Jin HT, Jeong YH, Park HJ, Ha SJ. Mechanism of T cell exhaustion in a chronic environment. BMB Rep. 44(4), 217–231 (2011).

150 Rios CM, Velilla PA, Rugeles MT. Chronically HIV-1 infected patients exhibit low frequencies of CD25+ regulatory T cells. Open Virol. J. 6, 49–58 (2012).

151 Moreno-Fernandez ME, Rueda CM, Rusie LK, Chougnet CA. Regulatory T cells control HIV replication in activated T cells through a cAMP-dependent mechanism. Blood 117(20), 5372–5380 (2011).

152 Moreno-Fernandez ME, Presicce P, Chougnet CA. Homeostasis and function of regulatory T cells in HIV/SIV infection. J. Virol. doi: 10.1128/JVI.00993-12 (2012) (Epub ahead of print).

153 Angin M, Kwon DS, Streeck H et al. Preserved function of regulatory T cells in chronic HIV-1 infection despite decreased numbers in blood and tissue. J. Infect. Dis. 205(10), 1495–1500 (2012).

154 Presicce P, Orsborn K, King E, Pratt J, Fichtenbaum CJ, Chougnet CA. Frequency of circulating regulatory T cells increases during chronic HIV infection and is largely controlled by highly active antiretroviral therapy. PLoS One 6(12), e28118 (2011).

155 Méndez-Lagares G, del Pozo Balado Mdel M, Genebat González M et al. Severe immune dysregulation affects CD4+CD25hiFoxP3+ regulatory T cells in HIV-infected patients with low-level CD4 T-cell repopulation despite suppressive highly active antiretroviral therapy. J. Infect. Dis. 205(10), 1501–1509 (2012).

156 Foxall RB, Albuquerque AS, Soares RS et al. Memory and naive-like regulatory CD4+ T cells expand during HIV-2 infection in direct association with CD4+ T-cell depletion irrespectively of viremia. AIDS 25(16), 1961–1970 (2011).

157 Schulze Zur Wiesch J, Thomssen A, Hartjen P et al. Comprehensive analysis of frequency and phenotype of T regulatory cells in HIV infection: CD39 expression of FoxP3+ T regulatory cells correlates with progressive disease. J. Virol. 85(3), 1287–1297 (2011).

158 Weiss L, Letimier FA, Carriere M et al. In vivo expansion of naive and activated CD4+CD25+FOXP3+ regulatory T cell populations in interleukin-2-treated HIV patients. Proc. Natl Acad. Sci. USA 107(23), 10632–10637 (2010).

159 Elahi S, Dinges WL, Lejarcegui N et al. Protective HIV-specific CD8+ T cells evade Treg cell suppression. Nat. Med.17(8), 989–995 (2011).

160 Presicce P, Shaw JM, Miller CJ, Shacklett BL, Chougnet CA. Myeloid dendritic cells isolated from tissues of SIV-infected Rhesus macaques promote the induction of regulatory T cells. AIDS 26(3), 263–273 (2012).

161 Qin X, Yao J, Yang F, Nie J, Wang Y, Liu PC. Human immunodeficiency virus type 1 Nef in human monocyte-like cell line THP-1 expands treg cells via Toll-like receptor 2. J. Cell Biochem.112(12), 3515–3524 (2011).

162 Wang X, Zhang Z, Zhang S et al. B7-H1 up-regulation impairs myeloid DC and correlates with disease progression in chronic HIV-1 infection. Eur. J. Immunol. 38(11), 3226–3236 (2008).

163 Vollbrecht T, Stirner R, Tufman A et al. Chronic progressive HIV-1 infection is associated with elevated levels of myeloid-derived suppressor cells. AIDS 26(12), F31–37 (2012).

164 Zhang M, Zhang H, Zhang T, Ji Y, Jiao Y, Wu H. Longitudinal changes of peripheral blood DC subsets and regulatory T cells in Chinese Chronic HIV-1-infected patients during antiretroviral therapy. PLoS One 7(5), e37966 (2012).

165 Xu H, Wang X, Pahar B et al. Increased B7-H1 expression on dendritic cells correlates with programmed death 1 expression on T cells in simian immunodeficiency virus-infected macaques and may contribute to T cell dysfunction and disease progression. J. Immunol. 185(12), 7340–7348 (2010).

166 Li W, Green WR. Immunotherapy of murine retrovirus-induced acquired immunodeficiency by CD4 T regulatory cell depletion and PD-1 blockade. J. Virol. 85(24), 13342–13353 (2011).

167 De Keersmaecker B, Heirman C, Corthals J et al. The combination of 4-1BBL and CD40L strongly enhances the capacity of dendritic cells to stimulate HIV-specific T cell responses. J. Leukoc. Biol. 89(6), 989–999 (2011).

168 Macatangay BJ, Rinaldo CR. Regulatory T cells in HIV immunotherapy. HIV Ther. 4(6), 639–647 (2010).

n� Reviews the controversial role of T-regulatory cells.

169 Rinaldo CR. Dendritic cell-based human immunodeficiency virus vaccine. J. Intern. Med. 265, 138–158 (2008).

170 Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 7(10), 790–802 (2007).


Recommended