Вы находитесь на странице: 1из 14

Human Vaccines & Immunotherapeutics

ISSN: 2164-5515 (Print) 2164-554X (Online) Journal homepage: http://www.tandfonline.com/loi/khvi20

Therapeutic vaccines against HIV infection

Felipe Garca, Agathe Len, Josep M. Gatell, Montserrat Plana & Teresa
Gallart

To cite this article: Felipe Garca, Agathe Len, Josep M. Gatell, Montserrat Plana &
Teresa Gallart (2012) Therapeutic vaccines against HIV infection, Human Vaccines &
Immunotherapeutics, 8:5, 569-581, DOI: 10.4161/hv.19555

To link to this article: http://dx.doi.org/10.4161/hv.19555

Published online: 01 May 2012.

Submit your article to this journal

Article views: 499

View related articles

Citing articles: 26 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=khvi20

Download by: [112.203.94.168] Date: 16 August 2016, At: 15:55


REVIEW
Human Vaccines & Immunotherapeutics 8:5, 569581; May 2012; G 2012 Landes Bioscience

Therapeutic vaccines against HIV infection


Felipe Garca,* Agathe Len, Josep M. Gatell, Montserrat Plana and Teresa Gallart
Hospital Clinic-HIVACAT; IDIBAPS; University of Barcelona; Barcelona, Spain

Keywords: therapeutic vaccine, dendritic cell, HIV, functional cure, recombinant virus, DNA

Resistance to medication, adverse effects in the medium-to- adherence to medication for life, side effects4 and the risk of
long-term and cost all place important limitations on lifelong development of resistance remain unsolved problems. Further-
adherence to combined antiretroviral therapy (cART). In this more, the HIV epidemic has severely hampered economic growth
context, new therapeutic alternatives to cART for life in HIV- in developing countries. Although the number of HIV-infected
infected patients merit investigation. Some data suggest that patients under treatment in these countries has increased steadily
strong T cell-mediated immunity to HIV can indeed limit virus in recent years, and although the number of new HIV infections
replication and protect against CD4 depletion and disease has fallen, the demand for treatment continues to rise.5 In fact, the
progression. The combination of cART with immune therapy number of new infections is still higher than the number of new
to restore and/or boost immune-specific responses to HIV has patients on treatment. For economic, social, cultural and political

2012 Landes Bioscience.


been proposed, the ultimate aim being to achieve a functional
reasons, widespread treatment is difficult to implement in these
cure. In this scenario, new, induced, HIV-specific immune
responses would be able to control viral replication to
countries. Given this situation, there is a need for new therapeutic
undetectable levels, mimicking the situation of the minority alternatives to cART for life in HIV-infected patients.6
of patients who control viral replication without treatment and Two such alternatives which have been proposed are eradica-
do not progress to AIDS. Classical approaches such as whole tion and functional cure.7 There is one report of an HIV-infected
inactivated virus or recombinant protein initially proved useful patient in whom viral replication remained absent, despite dis-

Do not distribute.
as therapeutic vaccines. Overall, however, the ability of these continuation of antiretroviral therapy, after transplantation with
early vaccines to increase HIV-specific responses was very CCR5D32/D32 stem cells.8 Recently published follow-ups of this
limited and study results were discouraging, as no consistent patient strongly suggest that cure of HIV has been achieved.9
immunogenicity was demonstrated and there was no clear Intensification of cART,10 stem cell therapy,11 gene therapy12 and
impact on viral load. Recent years have seen the development the elimination of latently infected cells are other alternatives to
of new approaches based on more innovative vectors such as
eradication which are currently being assessed.7 However, it is
DNA, recombinant virus or dendritic cells. Most clinical trials
of these new vectors have demonstrated their ability to
unlikely that therapeutic strategies leading to eradication will
induce HIV-specific immune responses, although they show emerge as a valid alternative to cART, and in fact, there are no
very limited efficacy in terms of controlling viral replication. biological models to support this approach. Conversely, the con-
However, some preliminary results suggest that dendritic cell- cept of functional cure, defined as control of HIV replication
based vaccines are the most promising candidates. To improve without cART, is based on the observation that a small propor-
the effectiveness of these vaccines, a better understanding of tion of HIV-infected patients (referred to as long-term non-
the mechanisms of protection, virological control and immune progressors or elite controllers) are able to control HIV replication
deterioration is required; without this knowledge, an effi- to below detectable levels, and without abnormalities in CD4 T-
cacious therapeutic vaccine will remain elusive. cell counts, for more than 25 y. It has been proposed that the
strong HIV-specific immune responses observed in these patients
could account for this natural functional cure. Specifically,
control of viral replication has been associated with HLA types
Introduction B*57:01, B*27:05 and B*14, and it has been proposed that the
viral peptides presented by these HLA molecules induce strong
Combined antiretroviral therapy (cART) has become one the best HIV-specific CD8 T-cell responses which are responsible for viral
tools to control HIV infection at both the individual1 and social control.13,14 These findings prompted the development of new
level.2,3 Indeed, cART has achieved impressive reductions in strategies based on the use of anti-HIV vaccines to induce high
morbidity and mortality among patients with advanced human frequencies and breadth of response of HIV-specific CD8 cyto-
immunodeficiency virus (HIV).1 However, despite these advances toxic T lymphocytes (CTLs), together with strong CD4 T-cell
in infection control, the cost of medication, the mandatory high help, in HIV-infected patients.15,16 Recent reports suggest that
a strong T cell-mediated immunity to HIV can indeed limit
virus replication and protect against CD4 depletion and disease
*Correspondence to: Felipe Garca; Email: fgarcia@clinic.ub.es
progression.17 Here we will review the basis of the therapeutic
Submitted: 11/17/11; Revised: 01/21/12; Accepted: 02/01/12 vaccine strategy and the different candidates tested in human
http://dx.doi.org/10.4161/hv.19555 clinical trials.

www.landesbioscience.com Human Vaccines & Immunotherapeutics 569


Pathogenic Basis for a Therapeutic Vaccine Design models30 that a specific helper T response against HIV is crucial in
obtaining an optimal, specific CTL response which can control
HIV-specific responses against HIV infection in antiretroviral viral replication. This is consistent with other reported data on
naive patients. The main question to be answered here is whether chronic viral infections in murine models.31 Finally, studies in
the immune system can contain viral replication without cART, animal models have shown that high levels of neutralizing anti-
even if only for limited periods. One major problem is that the bodies can block infection regardless of the route of exposure
types of immunological responses which are able to control viral to the virus.32 It has also been reported that plasma virus con-
replication are not known, although it does seem that the tinually and rapidly evolves to escape neutralization, indicating
relevance of the different response types changes with the stage of that neutralizing antibodies exert a level of selective pressure. It is
HIV infection. Barouch et al. recently published the results of a probable that neutralizing antibody responses account for the
preventive vaccine which suggest that protection against infection extensive variation in the envelope gene observed in the early
is mediated mainly by antibodies against env, whereas post- months after primary HIV infection.33
infection control of viral replication is associated with cellular Despite the relevance of HIV-specific immune responses in
responses against gag.18 Recent data reported by Pickers group in both the acute and chronic stages of infection these responses
relation to SIV-infected rhesus macaques suggest that the induc- are not, in most patients, able to control viral replication to
tion of effector memory T-cell responses (and not central memory undetectable levels and prevent the infection from progressing to
T-cell responses) with a CMV-vectored vaccine is essential for AIDS. Many authors hypothesize that selective escape from CD8+
control of viral replication.19 T-cell and other immune responses is the main reason for the
Anti-HIV-1 CTL responses have been detected in all studied failure to control HIV replication.34-42 Some studies suggest that

2012 Landes Bioscience.


cases of acute HIV infection, and it is believed that they reduce selective escape from CD8+ T-cell responses represents a major
the peak plasma viral load (PVL) to the stabilization level (set- driving force of HIV-1 sequence diversity. Allen et al.35 assessed
point) of PVL which is reached at the end of the acute phase.20,21 the relationship between viral evolution and adaptive CD8+ T-cell
It has therefore been hypothesized that CTL play an essential role responses in four HIV-infected patients who were studied for five
in controlling viral replication during acute infection. However, years after acute infection. Nearly two-thirds of the amino acid
it is not clear that neutralizing antibodies contribute to the con- mutations identified in non-envelope antigens were attributable

Do not distribute.
trol of viral replication during acute infection, since a delay in to CD8+ T-cell selective pressures. The preferential selection of
the development of these antibodies is observed which is not individual residues for mutation and the repeated selection for
associated with changes in viral replication.22 Finally, and in identical mutations observed in this study suggest that there are
contrast to CTL and neutralizing antibodies, a clonal depletion significant biochemical constraints on viral evolution. In addi-
of HIV-specific CD4+ T cells occurs during acute HIV infection. tion, some mutations induced by immune-mediated selective
It has been suggested that the lack of response of these cells is pressures have an impact on viral replication capacity.39,43 These
key to explaining the failure to control viral replication during findings help to decide which epitopes should be used in a
this phase.20,21 In fact, massive infection and loss of memory therapeutic vaccine in order to avoid or limit the viral escape to
CD4+ T cells occur in multiple tissues during acute infection,23 HIV CTL response.
and HIV mainly infects HIV-specific CD4+ T cells.24 These In addition to viral escape, the lack of control of viral replica-
findings may provide a basis for explaining why a therapeutic tion in HIV infection could be secondary to functional defects
vaccine-induced transient activation of HIV-specific CD4but of immune responses, which may be explained in terms of
not CD8T cells might have a detrimental effect on HIV alterations to the immune system. Although CD4+ and CD8+
replication,25 and could offer important clues for the future design cells capable of secreting interferon gamma (IFN-gamma) can
of therapeutic vaccines. be found in most HIV-infected patients, proliferative CD4
Some data suggest that strong T cell-mediated immunity to responses are normally absent29,44,45 and CD8+ cells have defective
HIV can indeed limit virus replication and protect against CD4 cytolytic activity.46,47,48 One explanation for these functional
depletion and disease progression in chronic HIV-infected deficits of CD4 and CD8 responses could be that the antigen-
patients. Although in most infected patients replication leads to presenting functions of the dendritic cells have deteriorated in
the progressive destruction of the immune system and evolves these patients, which may contribute to the functional defects
inevitably toward AIDS, a small number of immunologically observed in the Th1 and CTL cellular responses.49,50 The absence
privileged individuals, or long-term non-progressors (LTNP), of a correct proliferation and expansion of CD4+ responses may,
have a potent and sustained response of anti-HIV-1 CTL, Th in turn, influence the lack of cytolytic activity of CD8+ cells.44,51
cells and neutralizing HIV-1 antibodies. This is associated with In animal models there is a clear deficit in the secretion of
control of viral replication and the presence of very low or cytokines by CD4+ cells, which starts when PVL peaks in primary
undetectable viral concentrations in plasma in the absence of infection.52 Lastly, the selective infection of HIV-specific CD4+
cART.20 Direct data on the critical role of the CTL response cells in infected patients may explain why these responses are
in controlling viral replication have been obtained in both the quickly lost in HIV infection.24
infection model with macaques devoid of CD8+ T lympho- In summary, it seems clear that both cellular and humoral
cytes,26,27 and in the immunodeficient murine model.28 In addi- immune responses are critical for correct control of HIV infection.
tion, there is clear evidence from both human17,29 and animal Viral escape and functional defects in these immune responses are

570 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


likely to be the main causes of sustained viral replication in antiretroviral naive patients with a normal CD4 T-cell count
HIV infection, and constitute the two main problems that a could be at higher risk of suffering non-AIDS events (cancer,
therapeutic vaccine should solve. cardiovascular or neurological events) than the general popu-
Influence of cART on HIV-specific immune responses. The lation.65-67 In addition, it seems that the early initiation of
benefit of cART on the immune system is obtained through antiretroviral therapy significantly improves survival, as compared
an increase in the absolute number of circulating naive CD4 with deferred therapy.68 Furthermore, the discontinuation of
T lymphocytes, a concomitant reduction in the number of T antiretroviral therapy in order to reduce drug-related adverse
lymphocytes with activation markers, and restoration of the effects and to improve the patients quality of life has been
response to memory antigens.53 However, despite the clinical shown to be associated with increased morbidity and mortality.69
efficacy of cART54,55 this treatment by itself is unable to eradicate It has been suggested that immune activation due to viral load
the infection, even if it were administered for more than 60 y.56,57 rebound after ART interruption would explain this increase in
This is mainly due to the fact that therapy cannot eliminate latent the rates of AIDS-defining and non-AIDS-defining malignancies
HIV-1 in the form of integrated proviral DNA, as well as to the or cardiovascular events among patients who discontinued
existence of low levels of viral replication, which makes even cART. Untreated HIV infection is characterized by a profound
cell-to-cell infection possible.58,59 Furthermore, cART is incapable and continuous state of immune activation manifested by an
of restoring the immune-specific response to HIV,17,45 and, in increased turnover of B and T lymphocytes, natural killer cells
fact, it leads to a fall in the specific CTL response due to the and a high level of pro-inflammatory cytokines such as IL-7 and
lack of antigenic exposure.60 Some reports have shown that the TNF-a.70 Another relevant feature is the elevation of activated
helper proliferative response to HIV p24 Ag presented by some CD8+ T-cells expressing the DR+/CD38+ phenotype, which is

2012 Landes Bioscience.


cART patients does not reflect an improvement in the immune considered as a surrogate marker of disease progression.71,72
phenotype or function of CD4 or CD8 cells, but in fact is Systemic immune activation could lead, first, to exhaustion of
secondary to the small increases in viremia typically observed in the immune system, including lymph node fibrosis,73,74 retention
patients taking cART.61 This would explain the rapid rebound of effector T cells in lymph nodes,75,76 clonal exhaustion and
of viral load that occurs within days or weeks of suspending thymic dysfunction. In addition, it could contribute to the
cART, even after several years of effective therapy.62,63 This spreading of HIV infection by generating more targets for HIV,

Do not distribute.
rebound occurs even if cART is initiated in very early-stage HIV- thereby allowing ongoing HIV replication.24 In summary, partial
infected patients, in whom the immune system is theoretically control of viral replication remains deleterious for HIV-infected
still well preserved (circulating CD4+ T lymphocytes above patients, and the objective of a therapeutic vaccine should be
500 cells/mL; PVL: 5,00010,000 copies/ml). Similarly, these complete remission of infection. Nonetheless, if a therapeutic
viral dynamics occur even when immune restoration is practically vaccine were shown to be capable of partially controlling viral
complete in terms of the homeostasis of T lymphocytes and their load this would be a proof of concept justifying further
subpopulations, as well as in terms of the capacity for response investigation of new candidates.
to polyclonal stimuli and memory antigens with cART.62,64 A second objective has recently been proposed for therapeutic
Taken together these data suggest that the first challenge for a vaccines.7 Based on data related to the eradication of HIV in the
therapeutic vaccine is to demonstrate that it is possible to induce Berlin patient8,9 it has been suggested that the combination
de novo HIV-specific responses that are able to control viral of cART intensification with a vaccine able to induce effective
replication, albeit only partially. If this is the case, it would be HIV-specific responses could purge the HIV reservoir. This
a proof of concept that would justify further investigation of strategy, together with others using different drugs or gene
different immunotherapies. therapy, is already being assessed in clinical trials.7
Types of therapeutic vaccines. Most of the therapeutic vac-
Therapeutic Vaccines cines tested have previously been used as preventive candidates.
Classical approaches such as whole inactivated virus (REMUNE)
Objectives of a therapeutic vaccine. The main objective of a or recombinant protein (gp120) were initially shown to be useful.
therapeutic vaccine is to induce or increase immune responses In general, however, the capacity of these early vaccines to increase
against HIV infection using a planned exposure to HIV viral HIV-specific responses was very limited and study results were
antigens. In the ideal scenario, these responses would control viral discouraging, as no consistent immunogenicity was demonstrated
replication to an undetectable level, mimicking the situation of and there was no clear impact on viral load.77,78-81,82-89 Recent
the minority of patients who control viral replication without years have seen the development of new approaches based on
treatment and do not progress to AIDS. This has been termed more innovative vectors such as DNA, recombinant virus or
functional cure and it is observed in most other infections (e.g., dendritic cells. Most of the clinical trials involving these new
herpes or tuberculosis). Previous research has suggested that vectors have been exploratory and with a low number of patients.
partial control of viral replication could be enough to delay the In fact, in randomized studies no therapeutic vaccine has yet to
initiation of cART in antiretroviral naive patients or to offer demonstrate lasting and potent effectiveness in terms of con-
drug holidays in already-treated patients. However, the current trolling viral load, improving CD4 T-cell count or delaying
knowledge of HIV pathogenesis precludes this partial remission clinical progression. These weak results have precluded further
of infection from being a conceivable clinical objective. Even development. Although a great number of candidates have been

www.landesbioscience.com Human Vaccines & Immunotherapeutics 571


Table 1. Therapeutic vaccine clinical trials
References Immunogen Virological responses Immunogenicity responses
Whole inactivated vaccine
8790 Remune: inactivated whole virus. No clinical benefits Induction of gag-specific helper responses
Subunit vaccines
91100 Recombinant envelope protein No clinical benefits Improvement in specific lymphocyte proliferation
101103 HIV p24-like peptides (Vacc-4x) No clinical benefits Induction of HIV-associated specific responses
in 90% of patients
104106 Tat protein Not assessed Induction of Tat-specific T helper cell responses
Consistent decrease in cellular and biochemical
activation markers
Persistent increases in regulatory T cells
Stable increases in the percentage and absolute
numbers of both CD4 T cells and B lymphocytes
Reduced percentage of CD8 T cells and NK
lymphocytes
107 A combination of Nef-Tat fusion protein and envelope Not assessed Induction of strong gp120-specific
glycoprotein gp120 formulated with AS02A adjuvant CD4+ T-cell responses.
Induction of HIV-1-specific CD4+ T cells and CD8+

2012 Landes Bioscience.


T-cell proliferation
Vaccines using DNA as a vector
111, 112 DNA constructs encoding the nef, No clinical benefits Induction of HIV-1-specific cellular responses
rev or tat regulatory genes of HIV-1
113 DNA encoding env/rev and gag/pol genes Reduction of viral blips Poor capacity to boost virus-specific CD4
and CD8 T-cell responses

Do not distribute.
114 DNA encoding HIVA, an immunogen comprising Not assessed Poor capacity to boost virus-specific CD4
HIV-1 clade A p24/p17 fused to a string of cytotoxic and CD8 T-cell responses
T-cell epitopes
115, 116 DermaVir, a single plasmid DNA (pDNA) immunogen No clinical benefits Induction of HIV-specific immune responses
expressing 15 HIV antigens formulated in a
nanoparticle developed for topical application
Viral vector vaccines
124 ALVAC-HIV vCP1452 [expresses HIV-1 env and gag No clinical benefits Significantly increased HIV-1-specific CD4(+)
gene products and nef and pol gene products and CD8(+) T-cell responses
encompassing known human HLA-A2restricted
cytotoxic T cell (CTL) epitopes from these genes]
with Remune
77, 125 ALVAC-HIV vCP1452 and recombinant gp160 No clinical benefits Significant increases in anti-gp120
or anti-p24 antibody titers,
Transient augmentation of T-cell
proliferation responses to gp160 and/or p24.
Increased HIV-1-specific CD8 T cells
129 ALVAC-HIV vCP1452 No clinical benefits Modest increases in the median
lymphoproliferative response
25, 130 ALVAC vCP1452 Increased virus production Weakly immunogenic, inducing activated
following treatment interruption CD4 T-cell responses
131 ALVAC vCP1452 with Remune Tended to delay viral load rebound Induction of HIV-specific cells,
Associated with a longer time to not correlated with viral load rebound
meet pre-set criteria to restart ART
126, 132 ALVAC-HIV vCP1433 (expresses several HIV genes 24% of the Vac-IL-2 group Vaccine-elicited immunological responses
for gp120, gp41, 55 polyprotein, for a portion of pol lowered their viral set-point predictive of virological control
encoding the protease and for genes expressing compared with 5% of controls
cytotoxic T lymphocyte peptides from pol and nef)
with Lipo-6T vaccine (a mixture of the tetanus toxoid
TT-830843 class II-restricted universal CD4 epitope
combined with some peptides of Gag, Nef and Pol) and
followed by three cycles of subcutaneous interleukin-2

572 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


Table 1. Therapeutic vaccine clinical trials (continued)
Viral vector vaccines
128 ALVAC-HIV vCP1433 Prolongation of treatment Significantly increased HIV-p24-specific
discontinuation lymphoproliferative responses,
HIV-gag-specific CD8 T cells boosted
in 55% of patients tested
133 Fowl pox vector containing gag/pol sequences or No clinical benefits No differences in anti-HIV-specific immune
fowl pox vector containing gag/pol sequences and responses between placebo and vaccine study arms
a gene which encodes human interferon gamma
140 HIV-1 nef-expressing MVA After interruption of cART viremia Recognition of new CTL epitopes
remained below the pre-cART
viral load in 9/14 patients
141143 MVA expressing HIV-1 gag p24, p17 Not assessed Significantly increased the frequencies of
and a CD8+ T cell epitope string CD8+ and CD4+ T cells secreting IFN-gamma
Enhanced proliferative responses to gag peptides
144, 145 Recombinant adenovirus serotype 5 HIV-1 gag Reduction of 0.5 log10 Induction of HIV-1 gag-specific CD4 cells correlated
of PVL after a 16-week ATI o with control of viral replication in vivo
Dendritic cell-based vaccines
162, Heterologous peptides or ALVAC-HIV-1 vCP1452167 No change in median VL Induced novel epitope-specific CD8 T cells

2012 Landes Bioscience.


165168 and/or CD4 T cells
163, 164, Autologous whole inactivated HIV or RNA encoding Median VL drop 0.50.7 log, Increased HIV-specific CD8/CD4 (ELISPOT/ICS)
169171 CD40L and autologous HIV-1 antigens169,171 maintained 48 weeks and correlated with VL changes

used in clinical trials, this review will focus on those summarized A number of clinical trials using recombinant gp120 or gp160

Do not distribute.
in Table 1. as immunogens have been performed both in combination and
Remune. The Remune vaccine initially aroused considerable without antiretrovirals.91-100 Overall, these trials have not demon-
expectation. This is a vaccine of an inactivated whole virus in strated clinical benefit, although an improvement in specific
which the envelope protein has been removed during the process lymphocyte proliferation is observed in some of them.
of inactivation, which is performed by synthesis and formulated Vacc-4x consists of four synthetic peptides corresponding to
with incomplete Freund adjuvant. The vaccine stems from a virus conserved domains of the HIV-1 protein p24 that preferentially
originally obtained in Zaire and contains a type-A envelope and include HLA-A2 restricted elements. To ensure optimal exposure
type-G gag. It has been administered to more than 3,000 people of the immunogen to antigen-presenting cells (APCs) the vaccine
with an antiviral-controlled virus. The results showed that it was was given intradermally together with granulocyte-macrophage
able to induce gag-specific helper responses which were sometimes colony-stimulating factor (GM-CSF).101 Forty HIV-infected
very potent, but did not suggest a capacity for immunological patients on cART were vaccinated with either low-dose or high-
control of viral replication.87-90 The best and most extensive study dose Vacc-4x over 26 weeks. HIV-associated specific responses
aimed to determine whether the addition of Remune would were safely induced in 90% of patients in a dose-dependent
confer added clinical efficacy to that achievable by cART. This manner and were influenced by the HLA haplotype.103 There-
was a multicenter, double-blind, placebo-controlled, randomized after, cART was discontinued in patients for 14 weeks. Patients
trial conducted at 77 centers in the United States, and involved with the strongest delayed-type hypersensitivity (DTH) responses
2527 cART-treated, HIV-infected patients with a CD4 T-cell before treatment interruption tended to achieve lower actual
level between 300 106/L and 549 106/L. There were no HIV RNA levels.101 Finally, long-term HIV-specific immune
differences in HIV progression-free survival, changes in HIV responses and clinical outcomes were evaluated. Vacc-4x-induced
RNA or CD4 cell counts when comparing Remune and placebo cellular immune responses were unchanged 1.5 y after completing
recipients.89 immunization, and 62% of patients were still off cART.102
Subunit vaccines. Multiple strategies have been used to design Based on efficacy studies in animal models108 and those
therapeutic HIV vaccines based on peptides or recombinant suggesting that the presence of an anti-Tat immune response
proteins, including vaccination with a recombinant envelope correlates with a slower progression to disease,109 some authors have
protein,91-100 HIV p24-like peptides (Vacc-4x),101-103 a Tat proposed that a preventive and therapeutic Tat protein-based
protein104-106 and a combination of Nef-Tat fusion protein and vaccine deserves to be investigated. The first randomized, double
envelope glycoprotein gp120 formulated with AS02A adjuvant.107 blind, placebo-controlled phase I vaccine trial based on the native
The basis for the use of these candidates is that a therapeutic Tat protein was conducted in HIV-infected asymptomatic
HIV vaccine that induces HIV-1-specific CD4+ T-cell responses individuals.106 The vaccine was well tolerated and induced and/or
could improve antiviral immunity, resulting in delayed disease maintained Tat-specific T helper cell responses in all subjects, with
progression. a wide spectrum of functional anti-Tat antibodies. In addition,

www.landesbioscience.com Human Vaccines & Immunotherapeutics 573


2.5 y after the last immunization Tat-specific humoral and cellular Viral vector vaccines. Given the poor results obtained with
immune responses were still detectable in vaccinees.105 These data Remune, subunit vaccines and vaccines using DNA as the vector
provided the basis for a phase II study in 87 successfully-treated it has been proposed that outcomes could be improved by using
HIV-infected patients, the results of which have recently been viral vectors as immunogens. Live recombinant vaccines are made
reported.104 It was confirmed that immunization with Tat was safe of a live viral or bacterial vector that is engineered to carry the
and immunogenic. In addition, there was a consistent decrease genes that encode HIV antigens. The antigens are presented on
in cellular and biochemical activation markers and persistent the surface of the cell as externalized peptides in the context of
increases in the number of regulatory T cells. This was associated MHC class I molecules, thus priming for CD8+ T-cell responses.
with stable increases in the percentage and absolute numbers of The best studied vaccine vectors in humans are the pox viruses.117
both CD4 T cells and B lymphocytes, as well as with a reduction Vaccinia virus engineered with HIV-1 genes has been shown to
in the percentage of CD8 T cells and NK lymphocytes. The induce virus-specific cellular and humoral immune responses in
authors concluded that Tat immunization in patients on cART immunized macaques, as well as protection against simian
could help to restore immune homeostasis. The improvement immunodeficiency virus (SIV) infection when immunization
in immune system function (as opposed to functional cure) is a with such constructs has been followed with boosting by
benefit that some other investigators have claimed as an alternative recombinant proteins. However, due to the development of
primary end-point for therapeutic vaccines.90,110 However, the life-threatening disseminated vaccinia infections in immuno-
clinical relevance of the recovery of these parameters is not clear. suppressed individuals there is a reluctance to use replication-
Until this issue is clarified, functional cure remains the best objec- competent vaccinia virus as a vector system in large human trials.
tive for a therapeutic vaccine. Particular attention has, however, been focused on pox viruses

2012 Landes Bioscience.


A clinical trial of the gp120/NefTat/AS02A therapeutic with limited in vivo replicative capacity and which, therefore, are
vaccine in chronically HIV-1-infected volunteers on suppressive non-pathogenic in animal models and humans118,119; examples
ART found the vaccine to be safe, well tolerated and able include MVA and NYVAC, which carry deletions of the genes
to induce strong gp120-specific CD4+ T-cell responses, and a associated with pathogenicity, or avian pox viruses (canary pox
higher number of vaccinees developed both HIV-1-specific and fowl pox). These vectors are restricted to the early stages of
CD4+ T-cell responses and CD8+ T-cell proliferation.107 virus replication in human cells but initiate protein synthesis

Do not distribute.
Therapy interruption to determine whether these immunological and thus elicit specific immune responses to the recombinant
responses might lead to improved control of HIV-1viral load antigen.118,120-123
was not performed. The best studied vector has been canary pox
Vaccines using DNA as a vector. DNA vaccines are based on (ALVAC).25,77,124-128,129-131 The international QUEST study124
bacterial plasmids that express the desired antigen in situ, which was performed on patients who started treatment during the
leads to induction of an immune response of the Th1 type. The acute phase. After a mean of two years of virological control,
first trial, reported in 1998, included nine asymptomatic HIV-1- 79 patients were randomized to receive either immunization
infected patients who were immunized with DNA constructs with ALVAC-HIV vCP1452, with ALVAC-HIV vCP1452 plus
encoding the nef, rev or tat regulatory genes of HIV-1.111 The Remune, or placebo. ALVAC-HIV vCP1452 expresses HIV-1
vaccination induced HIV-1-specific cellular responses, but was env and gag gene products and nef and pol gene products
not able to control viral replication or change CD4+ T-cell counts. encompassing known human HLA-A2restricted cytotoxic T-cell
These results were confirmed with the same candidate in patients (CTL) epitopes from these genes. The choice of vaccine
on cART.112 A further two candidates have been tested in HIV- candidates for this study was dictated by their safety records in
infected patients on cART, the first encoding env/rev and gag/pol HIV-1-infected subjects and by immunogenicity. The combina-
genes113 and the second encoding HIVA, an immunogen com- tion of vaccines in a single treatment arm was selected for its
prising HIV-1 clade A p24/p17 fused to a string of cytotoxic potential to enhance CD4 T helper cells (Remune) and CTLs
T-cell epitopes.114 These candidates were safe but their capacity (ALVAC-HIV vCP1452). After 24 weeks immunization, cART
to boost virus-specific CD4 and CD8 T-cell responses was poor. was interrupted. There was no difference between the groups
Given these results, novel means of vaccine delivery have been in terms of viral rebound dynamics or viral load figures. These
proposed for further improvement of DNA vaccine potency. results confirm the findings of a previous small randomized
DermaVir, a single plasmid DNA (pDNA) immunogen expres- pilot study of HIV-infected patients treated with cART during
sing 15 HIV antigens, has been formulated in a nanoparticle acute infection and who received ALVAC-HIV vCP1452 and
with polyethylenimine-mannose and glucose, and has been recombinant gp160.77,125
developed for topical application to deliver DNA-encoded Similar results with ALVAC-HIV vCP1452 have been
antigens into Langerhans cells.115 This candidate has recently obtained in chronic HIV-infected patients. The ACTG 5068
been tested in the context of structured therapy interruption in a included 99 subjects receiving successful cART who were
pilot clinical trial.116 Although HIV DNA immunization induces randomized to undergo continued cART, structured therapy
broader and higher magnitudes of HIV-specific immune res- interruptions (STIs), ALVAC-HIV vCP1452 immunization, or
ponses compared with structured therapy interruptions alone, STIs and ALVAC-HIV vCP1452 immunization. Subjects in the
this candidate did not have any effect on the dynamics of viral two STI arms had significantly enhanced immunological control
rebound, set-point viral load or CD4 T-cell counts. of HIV replication compared with subjects in the two arms

574 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


without STIs. ALVAC-HIV vCP1452 did not affect viral load measured by ELISPOT, or in cytolytic responses after vaccina-
measures.129 A separate placebo-controlled study conducted in tion and before interruption of treatment. Treatment was not
chronically-infected patients suggested that the immunogenicity interrupted in ten patients. There were no differences in the
conferred by the ALVAC vCP1452 candidate vaccine alone could control of viral replication between the placebo group and the
be deleterious, since it was associated with higher virus production group vaccinated with gag/pol. However, patients immunized
following treatment interruption.130 Further research suggested with gag/pol and interferon gamma had better control of viral
that ALVAC vCP1452 was only weakly immunogenic, it failing replication, with a mean viral load of 0.8 log10 less than the
to elicit significant HIV-specific CD8 T-cell responses and other two groups, although the differences between groups were
inducing activated CD4 T-cell responses that could favor greater not significant. The absence of immune responses in the two
viral rebound after treatment interruption.25 These results appear vaccinated groups is disappointing, while the response in the
to conflict with those of a clinical trial reported recently by interferon group is surprising and merits further investigation.
Angel et al.131 This was a randomized, placebo-controlled, double- The modified vaccinia virus Ankara (MVA) was developed
blind study in which effectively-treated HIV-infected individuals toward the end of the smallpox eradication campaign in the
with high CD4 T-cell counts were randomized to receive 1970s, the aim being to obtain a vaccine with a better safety
ALVAC vCP1452 with Remune, ALVAC vCP1452 alone or profile than the ones in use at that time. MVA was dereived
placebo over 20 weeks. At week 24, participants interrupted from vaccinia by over 570 passages in chicken embryo fibroblast
cART. ALVAC with or without Remune did not lower the viral cells (CEF).134 The complete genomic sequence has been
load set-point, although it tended to delay viral load rebound sequenced and has a length of 178 kb. It consists of 193 open
and was associated with a greater time to meet pre-set criteria to reading frames (ORFs), corresponding to 177 genes, of which

2012 Landes Bioscience.


restart ART. In contrast to the findings reported by Autran 25 are split and/or have suffered mutations resulting in truncated
et al.130 and Papagno et al.25 the induction of helper CD4 T-cell proteins. These numerous mutations, affecting host interactive
responses was not associated with a worse virological outcome. proteins and some structural proteins, explain the attenuated
At all events, further research is required to determine whether phenotype of MVA.119 There is clinical experience with MVA
the best strategy to induce viral control in chronic HIV-infected as a vaccine against smallpox in over 120,000 people.118,120
patients should be based on inducing robust and broad CD8 During extensive field studies, including with high-risk patients,

Do not distribute.
T-cell responses but only modestly activating CD4 T-cell no side effects were associated with the use of the MVA
responses. vaccine.118,120 The combination of a very good safety profile and
The best results for a viral vector have been reported with the ability to deliver antigens in a highly immunogenic way
ALVAC vCP1433 in combination with Lipo-6T vaccines, makes MVA suitable as a vaccine vector. It can stimulate anti-
followed by three cycles of subcutaneous interleukin-2.126,132 body and T-cell responses even in the presence of pre-existing
ALVAC-HIV vCP1433 expresses several HIV genes for gp120, antibodies,135 and it has been shown to be effective in primates
gp41, 55 polyprotein, for a portion of pol encoding the protease and humans for several viruses, including SIV and SHIV.121,136-139
and for genes expressing cytotoxic T-lymphocyte peptides from Harrer et al.140 investigated the safety and immunogenicity of
pol and nef. The Lipo-6T vaccine is a mixture of the tetanus an HIV-1 nef-expressing MVA in 14 cART-treated HIV-
toxoid TT-830843 class II-restricted universal CD4 epitope infected patients. Vaccination with MVA-nef was safe, was
combined with certain peptides of Gag, Nef and Pol. Chronic associated with recognition of new CTL epitopes, and after
HIV-infected patients were randomized to receive either cART interruption of cART viremia remained below the pre-cART viral
alone (n = 37) or four doses of ALVAC-HIV vCP1433 and Lipo- load in 9/14 patients. In another pilot study, vaccination of 16
6T followed by three cycles of IL-2 (n = 34). Vaccination HIV-1-infected cART-treated individuals with MVA expressing
improved the proliferative response against p24 Ag, as well as HIV-1 gag p24, p17 and a CD8+ T-cell epitope string was
CD8 T-cell responses. After cART interruption, 24% of the not only safe but also significantly increased the frequencies of
Vac-IL-2 group lowered their viral set-point, compared with 5% CD8+ and CD4+ T cells secreting IFN-gamma and enhanced
of controls. Vaccine-elicited immunological responses were pre- proliferative responses to gag peptides.141-143
dictive of virological control. Vaccination with the same product Finally, in ACTG A5197, HIV-1-infected individuals who
(ALVAC-HIV vCP1433) in a French clinical trial was safe, received a recombinant adenovirus serotype 5 HIV-1 gag thera-
immunogenic and associated with prolongation of treatment peutic vaccine had a 0.5 log10 lower PVL after a 16-week ATI,
discontinuation.128 compared with those who received placebo.144,145
A pilot clinical trial of a therapeutic vaccination based on a In summary, although most viral vector vaccines have been
fowl pox vector and involving patients with primary infection able to induce HIV-specific immune responses in clinical trials
has been reported by Emery et al.133 After a mean of four years they have shown very limited efficacy in terms of controlling viral
of cART, 35 patients with controlled viral replication were replication. New therapeutic strategies are therefore warranted.
randomized to be vaccinated with a fowl pox vector free of HIV Dendritic cell-based vaccines. Dendritic cells (DC) are the
sequences, a vector containing gag/pol sequences, or a vector most potent professional antigen-processing and presenting cells
containing gag/pol sequences and a gene which encodes human (APC), with the unique ability to induce both primary and
interferon gamma. Surprisingly, there were few differences secondary immune responses of CD4+ and CD8+ T lympho-
between the groups in terms of the presence of CTL cells, as cytes.146,147 A wide variety of data in experimental in vivo and in

www.landesbioscience.com Human Vaccines & Immunotherapeutics 575


vitro systems have shown that DC are able to engulf exogenous immunized patients was small (ranging from 0.30 to 0.60 log10)
soluble proteins, tumor cell lysates, and inactivated virus and but clinically significant when compared with the placebo group,
virus-infected apoptotic cells, as well as presenting antigens and it was maintained up to week 48. However, this small
not only in the MHC-class II pathway to helper CD4+ T cells decrease in VL in immunized patients was inversely correlated
(Th)146 but also in the MHC-class I pathway to cytotoxic with a weak increase in HIV-1-specific CD8+ T-cell responses.
CD8+ T lymphocytes (CTL), a phenomenon also known as The other two trials involved antiretroviral-treated patients.
cross-presentation or cross-priming148-150. The first was an open, randomized (2:1) clinical trial using heat-
In vitro culture of human blood monocytes (MO) with GM- inactivated autologous virus in cART-treated HIV-1-infected
CSF and IL-4 gives rise to myeloid DC.151,152 Some data suggest patients, and reported partial and transient control of viral
that these human myeloid DC not only activate naive CD4+ replication after interruption of antiretroviral therapy.164 In this
T cells but also promote their differentiation into Th1 cells, study, autologous DC vaccine pulsed ex vivo with heat-inactivated
preventing the differentiation into Th2.153 In addition, antigen- autologous HIV-1 elicited only weak Th1- and HIV-1-specific
pulsed DC secrete antigen-presenting particles (termed exosomes) CD8+ T-cell responses. Recently, Routy et al.,171 in an uncon-
which express functional MHC-class I and II and T-cell co- trolled study, reported the final results of an immunotherapy
stimulatory molecules, and they are able to prime CTL in vivo regimen consisting of MD-DC electroporated with mRNA
and eradicate or suppress the growth of established murine encoding autologous HIV-1 antigens (Gag, Nef, Rev, Vpr); this
tumors.154 Autologous DC pulsed in vitro with a variety of was administered monthly in 33 subjects in four intradermal
inactivated pathogens have also been shown to induce a potent doses in combination with cART, followed by two more doses
protective immunity in murine models of human infections.154-158 during a 12-week cART interruption (STI). They found that

2012 Landes Bioscience.


All these data indicate that in vitro-generated DC may be the eight out of 24 subjects (33%) met the criteria for the primary
most potent cellular adjuvant for a vaccine preparation designed endpoint of three instances of viral load , 1,000 copies/mL,
to induce effective Th1 and CTL responses to tumor antigens while 11 out of 24 subjects (46%) met the criteria for the
and intracellular pathogens. secondary endpoint of three instances of viral load , 10,000
It has been reported that an inactivated SIV-pulsed therapeutic copies/mL. At week 12 of the STI, 16/24 subjects responded with
dendritic-cell vaccine was able to elicit effective and lasting SIV- a mean reduction in viral load of 1.2 log compared with their

Do not distribute.
specific cellular and humoral immunity. This study found that pre-cART viral load value. These data are impressive and are
after three immunizations administered at two-week intervals the similar to those reported by Lu et al.163 with HIV-1-infected
SIV cellular DNA and the SIV plasma RNA levels decreased by patients who were vaccinated when treated with cART. These
50-fold and 1,000-fold, respectively. This decrease in viral load data need to be confirmed in a randomized, placebo-controlled
was negatively correlated with SIV-specific T-cell responses. study. A review of DC-based vaccine clinical trials has recently
Neutralizing antibody responses also increased significantly and been published.172
remained increased throughout the follow-up period.159 Finally,
two studies in a hu-PBL-SCID mouse model suggest that mice Conclusions
immunized with inactivated virus-pulsed dendritic cells elicit a
potent immune response against HIV, which protects the animals Our knowledge of the immunological control of HIV viral
from virus challenge.160,161 replication and the causes of cellular and humoral immune
There are at least ten published clinical trials of DC immuno- deterioration is limited, and we lack immunological methods
therapy for HIV infection in humans.162-169,170,171 Most of them able to demonstrate an efficacious immune control of HIV in
found that DC immunotherapy elicited immunological res- vivo. The efficacy of immune therapy and therapeutic vaccines
ponses, even though the design of the trials and the HIV antigens has been modest in the best of cases, although some pre-
used to pulse DC were very different. It should be noted, liminary results with dendritic cell-based vaccines seem promis-
however, that virological responses were only observed in four of ing. Efforts must now be redoubled in order to improve our
these trials.163,164,170,171 understanding of the mechanisms of protection, virological
Two of these four trials were performed in untreated control and immune deterioration, as without this knowledge
patients. The first was a preliminary, non-controlled, non- an efficacious therapeutic vaccine remains a long way off. Given
randomized study that showed striking results for a MD-DC- the toxicity and long-term efficacy problems associated with
based vaccine loaded with high doses of chemically-inactivated current drugs the search for therapeutic vaccines must be seen as
(with aldrithiol-2) autologous virus in untreated HIV patients.163 a priority line of investigation.
Plasma viral load (PVL) decreased by 90% for at least one year
in eight of 18 patients. This decrease in PVL was associated Acknowledgments
with strong and sustained HIV-1-specific cellular responses. The This study was partially supported by the following grants: FIS
second trial was a double-blind, placebo-controlled study using PS09/01297, TRA-094, EC10153, FIS PI10/02984, SAF2006
the same schedule and immunizing the same type of patients 26667-E, RIS*, HIVACAT**, ORVACS***. F.G. was recipient
(HIV-1-infected patients off cART).170 Only a modest virological of a research grant from IDIBAPS****, Barcelona, Spain. M.P.
response, which was maintained for at least 48 weeks in the was supported by contract FIS 03/0072 from the Fundaci
vaccinated group, was observed.170 This change in VL in the Privada Clnic per a la Recerca Biomdica, in collaboration with

576 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


the Spanish Department of Health. *Red Temtica Cooperativa Catalonia. ***ORVACS: Objectif recherche vaccin sida.
de Grupos de Investigacin en Sida del Fondo de Investigacin ****IDIBAPS: Institut dInvestigacions Biomdiques August Pi
Sanitaria (FIS). **HIVACAT: HIV development program in I Sunyer.

References 15. Autran B, Carcelain G. AIDS. Boosting immunity to 28. de Quiros JC, Shupert WL, McNeil AC, Gea-
HIVcan the virus help? Science 2000; 290:946-9; Banacloche JC, Flanigan M, Savage A, et al.
1. Palella FJ, Delaney KM, Moorman AC, Loveless
PMID:11184735; http://dx.doi.org/10.1126/science. Resistance to replication of human immunodeficiency
MO, Fuhrer J, Satten GA, et al. Declining morbidity
290.5493.946 virus challenge in SCID-Hu mice engrafted with
and mortality among patients with advanced human
16. Autran B, Carcelain G, Combadiere B, Debre P. peripheral blood mononuclear cells of nonprogressors
immunodeficiency virus infection. In: 1998. pp. 853-
Therapeutic vaccines for chronic infections. Science is mediated by CD8(+) T cells and associated with a
860.
2004; 305:205-8; PMID:15247470; http://dx.doi.org/ proliferative response to p24 antigen. J Virol 2000;
2. Montaner JS. Treatment as preventiona double hat- 74:2023-8; PMID:10644376; http://dx.doi.org/10.
10.1126/science.1100600
trick. Lancet 2011; 378:208-9; PMID:21763923; 1128/JVI.74.4.2023-2028.2000
http://dx.doi.org/10.1016/S0140-6736(11)60821-0 17. Letvin NL, Walker BD. Immunopathogenesis and
immunotherapy in AIDS virus infections. Nat Med 29. Day CL, Walker BD. Progress in defining CD4 helper
3. Montaner JS, Lima VD, Barrios R, Yip B, Wood E, cell responses in chronic viral infections. J Exp Med
2003; 9:861-6; PMID:12835706; http://dx.doi.org/
Kerr T, et al. Association of highly active antiretro- 2003; 198:1773-7; PMID:14676292; http://dx.doi.
10.1038/nm0703-861
viral therapy coverage, population viral load, and yearly org/10.1084/jem.20031947
new HIV diagnoses in British Columbia, Canada: a 18. Barouch DH, Liu J, Li H, Maxfield LF, Abbink P,
Lynch DM, et al. Vaccine protection against acquisi- 30. Hel Z, Nacsa J, Tryniszewska E, Tsai WP, Parks
population-based study. Lancet 2010; 376:532-9;
tion of neutralization-resistant SIV challenges in rhesus RW, Montefiori DC, et al. Containment of simian
PMID:20638713; http://dx.doi.org/10.1016/S0140-
monkeys. Nature 2012; 482:89-93; PMID:22217938; immunodeficiency virus infection in vaccinated
6736(10)60936-1
http://dx.doi.org/10.1038/nature10766 macaques: correlation with the magnitude of virus-
4. Martinez E, Mocroft A, Garca-Viejo M, Prez-Cuevas J, specific pre- and postchallenge CD4+ and CD8+ T cell
Blanco J, Mallolas J, et al. A prospective cohort study on 19. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes
CM, Coyne-Johnson L, et al. Profound early control of responses. J Immunol 2002; 169:4778-87; PMID:
the risk for lipodystrophy in HIV-1 infected patients 12391187
treated with protease inhibitor-containing regimens. highly pathogenic SIV by an effector memory T-cell
vaccine. Nature 2011; 473:523-7; PMID:21562493; 31. Kaech SM, Ahmed R. Immunology. CD8 T cells

2012 Landes Bioscience.


Lancet 2001; 357:592-8; PMID:11558485; http://dx.
http://dx.doi.org/10.1038/nature10003 remember with a little help. Science 2003; 300:263-5;
doi.org/10.1016/S0140-6736(00)04056-3
20. Rosenberg ES, Billingsley JM, Caliendo AM, Boswell PMID:12690179; http://dx.doi.org/10.1126/science.
5. Sidib M. Newsmaker interview: Michel Sidib. New 1084511
HIV infections drop, but treatment demands rise. SL, Sax PE, Kalams SA, et al. Vigorous HIV-1-specific
CD4+ T cell responses associated with control of 32. Nishimura Y, Igarashi T, Haigwood NL, Sadjadpour
Interview by Jon Cohen. [Abstract]. Science 2010;
viremia. [see comments]. Science 1997; 278:1447-50; R, Donau OK, Buckler C, et al. Transfer of neutraliz-
330:1301; PMID:21127221
PMID:9367954; http://dx.doi.org/10.1126/science. ing IgG to macaques 6 h but not 24 h after SHIV
6. Garca F, Ruiz L, Lopez-Bernaldo de Quirs J, Moreno infection confers sterilizing protection: implications for
278.5342.1447
S, Domingo P. INMUNOTERAPIA Y VACUNAS HIV-1 vaccine development. Proc Natl Acad Sci U S A
TERAPEUTICAS EN LA INFECCIN POR VIH. 21. Rosenberg ES, Altfeld M, Poon SH, Phillips MN,

Do not distribute.
Wilkes BM, Eldridge RL, et al. Immune control of 2003; 100:15131-6; PMID:14627745; http://dx.doi.
Enf Inf Microb Clin 2005; 23(Suppl 2):84-94; http:// org/10.1073/pnas.2436476100
dx.doi.org/10.1016/S0213-005X(05)75164-8 HIV-1 after early treatment of acute infection. Nature
2000; 407:523-6; PMID:11029005; http://dx.doi.org/ 33. Richman DD, Wrin T, Little SJ, Petropoulos CJ.
7. Cohen J. The emerging race to cure HIV infections. Rapid evolution of the neutralizing antibody response
10.1038/35035103
Science 2011; 332:784-5, 787-9; PMID:21566173; to HIV type 1 infection. Proc Natl Acad Sci U S A
http://dx.doi.org/10.1126/science.332.6031.784 22. Moore JP, Cao Y, Ho DD, Koup RA. Development of
the anti-gp120 antibody response during seroconver- 2003; 100:4144-9; PMID:12644702; http://dx.doi.
8. Htter G, Nowak D, Mossner M, Ganepola S, Mssig org/10.1073/pnas.0630530100
sion to human immunodeficiency virus type 1. J Virol
A, Allers K, et al. Long-term control of HIV by CCR5 34. Gao X, Bashirova A, Iversen AK, Phair J, Goedert JJ,
1994; 68:5142-55; PMID:8035514
Delta32/Delta32 stem-cell transplantation. N Engl J Buchbinder S, et al. AIDS restriction HLA allotypes
Med 2009; 360:692-8; PMID:19213682; http://dx. 23. Mattapallil JJ, Douek DC, Hill B, Nishimura Y,
Martin M, Roederer M. Massive infection and loss of target distinct intervals of HIV-1 pathogenesis. Nat
doi.org/10.1056/NEJMoa0802905 Med 2005; 11:1290-2; PMID:16288280; http://dx.
memory CD4+ T cells in multiple tissues during acute
9. Allers K, Htter G, Hofmann J, Loddenkemper C, doi.org/10.1038/nm1333
SIV infection. Nature 2005; 434:1093-7; PMID:
Rieger K, Thiel E, et al. Evidence for the cure of HIV 35. Allen TM, Altfeld M, Geer SC, Kalife ET, Moore C,
15793563; http://dx.doi.org/10.1038/nature03501
infection by CCR5D32/D32 stem cell transplantation. Osullivan KM, et al. Selective escape from CD8+
Blood 2011; 117:2791-9; PMID:21148083; http://dx. 24. Douek DC, Brenchley JM, Betts MR, Ambrozak DR,
Hill BJ, Okamoto Y, et al. HIV preferentially infects T-cell responses represents a major driving force
doi.org/10.1182/blood-2010-09-309591 of human immunodeficiency virus type 1 (HIV-1)
HIV-specific CD4+ T cells. Nature 2002; 417:95-8;
10. Buzn MJ, Massanella M, Llibre JM, Esteve A, Dahl sequence diversity and reveals constraints on HIV-1
PMID:11986671; http://dx.doi.org/10.1038/417095a
V, Puertas MC, et al. HIV-1 replication and immune evolution. J Virol 2005; 79:13239-49; PMID:
dynamics are affected by raltegravir intensification of 25. Papagno L, Alter G, Assoumou L, Murphy RL, Garcia
F, Clotet B, et al. ORVACS Study Group. Com- 16227247; http://dx.doi.org/10.1128/JVI.79.21.
HAART-suppressed subjects. Nat Med 2010; 16:460-5; 13239-13249.2005
PMID:20228817; http://dx.doi.org/10.1038/nm.2111 prehensive analysis of virus-specific T-cells provides
clues for the failure of therapeutic immunization 36. Allen TM, Yu XG, Kalife ET, Reyor LL, Lichterfeld
11. Kambal A, Mitchell G, Cary W, Gruenloh W, Jung Y, M, John M, et al. De novo generation of escape
with ALVAC-HIV vaccine. AIDS 2011; 25:27-36;
Kalomoiris S, et al. Generation of HIV-1 resistant and variant-specific CD8+ T-cell responses following
PMID:21076273; http://dx.doi.org/10.1097/QAD.
functional macrophages from hematopoietic stem cell- cytotoxic T-lymphocyte escape in chronic human
0b013e328340fe55
derived induced pluripotent stem cells. Mol Ther immunodeficiency virus type 1 infection. J Virol
2011; 19:584-93; PMID:21119622; http://dx.doi.org/ 26. Schmitz JE, Kuroda MJ, Santra S. Control of viremia
in simian immunodeficiency virus infection by CD8+ 2005; 79:12952-60; PMID:16188997; http://dx.doi.
10.1038/mt.2010.269 org/10.1128/JVI.79.20.12952-12960.2005
lymphocytes. In: 1999. pp. 857-860.
12. Johnson PR, Schnepp BC, Zhang J, Connell MJ, 37. Feeney ME, Tang Y, Pfafferott K, Roosevelt KA,
Greene SM, Yuste E, et al. Vector-mediated gene 27. Metzner KJ, Jin X, Lee FV, Gettie A, Bauer DE, Di
Mascio M, et al. Effects of in vivo CD8(+) T cell Draenert R, Trocha A, et al. HIV-1 viral escape in
transfer engenders long-lived neutralizing activity and infancy followed by emergence of a variant-specific
protection against SIV infection in monkeys. Nat Med depletion on virus replication in rhesus macaques
immunized with a live, attenuated simian immuno- CTL response. J Immunol 2005; 174:7524-30;
2009; 15:901-6; PMID:19448633; http://dx.doi.org/ PMID:15944251
10.1038/nm.1967 deficiency virus vaccine. J Exp Med 2000; 191:1921-
31; PMID:10839807; http://dx.doi.org/10.1084/jem.
13. McMichael AJ, Jones EY. Genetics. First-class control of
191.11.1921
HIV-1. Science 2010; 330:1488-90; PMID:21148380;
http://dx.doi.org/10.1126/science.1200035
14. Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI,
Walker BD, et al. International HIV Controllers
Study. The major genetic determinants of HIV-1
control affect HLA class I peptide presentation. Science
2010; 330:1551-7; PMID:21051598; http://dx.doi.
org/10.1126/science.1195271

www.landesbioscience.com Human Vaccines & Immunotherapeutics 577


38. Altfeld M, Allen TM, Kalife ET, Frahm N, Addo 51. McNeil AC, Shupert WL, Iyasere CA, Hallahan CW, 64. Plana M, Garca F, Gallart T, Tortajada C, Soriano A,
MM, Mothe BR, et al. The majority of currently Mican JA, Davey RT, Jr., et al. High-level HIV-1 Palou E, et al. Immunological benefits of antiretroviral
circulating human immunodeficiency virus type 1 viremia suppresses viral antigen-specific CD4(+) T cell therapy in very early stages of asymptomatic chronic
clade B viruses fail to prime cytotoxic T-lymphocyte proliferation. Proc Natl Acad Sci U S A 2001; HIV-1 infection. AIDS 2000; 14:1921-33; PMID:
responses against an otherwise immunodominant 98:13878-83; PMID:11717444; http://dx.doi.org/10. 10997396; http://dx.doi.org/10.1097/00002030-
HLA-A2-restricted epitope: implications for vaccine 1073/pnas.251539598 200009080-00007
design. J Virol 2005; 79:5000-5; PMID:15795285; 52. McKay PF, Barouch DH, Schmitz JE, Veazey RS, 65. Reekie J, Gatell JM, Yust I, Bakowska E, Rakhmanova
http://dx.doi.org/10.1128/JVI.79.8.5000-5005.2005 Gorgone DA, Lifton MA, et al. Global dysfunction of A, Losso M, et al. EuroSIDA in EuroCoord. Fatal and
39. Allen TM, Altfeld M, Yu XG, OSullivan KM, CD4 T-lymphocyte cytokine expression in simian- nonfatal AIDS and non-AIDS events in HIV-1-
Lichterfeld M, Le Gall S, et al. Selection, transmission, human immunodeficiency virus/SIV-infected monkeys positive individuals with high CD4 cell counts
and reversion of an antigen-processing cytotoxic is prevented by vaccination. J Virol 2003; 77:4695- according to viral load strata. AIDS 2011; 25:2259-
T-lymphocyte escape mutation in human immuno- 702; PMID:12663776; http://dx.doi.org/10.1128/ 68; PMID:21918422; http://dx.doi.org/10.1097/
deficiency virus type 1 infection. J Virol 2004; JVI.77.8.4695-4702.2003 QAD.0b013e32834cdb4b
78:7069-78; PMID:15194783; http://dx.doi.org/10. 53. Autran B, Carcelain G, Li TS, Blanc C, Mathez D, 66. Mocroft A, Reiss P, Gasiorowski J, Ledergerber B,
1128/JVI.78.13.7069-7078.2004 Tubiana R, et al. Positive effects of combined anti- Kowalska J, Chiesi A, et al. EuroSIDA Study Group.
40. Draenert R, Le Gall S, Pfafferott KJ, Leslie AJ, Chetty retroviral therapy on CD4+ T cell homeostasis and Serious fatal and nonfatal non-AIDS-defining illnesses
P, Brander C, et al. Immune selection for altered function in advanced HIV disease. [see comments]. in Europe. J Acquir Immune Defic Syndr 2010;
antigen processing leads to cytotoxic T lymphocyte Science 1997; 277:112-6; PMID:9204894; http://dx. 55:262-70; PMID:20700060; http://dx.doi.org/10.
escape in chronic HIV-1 infection. J Exp Med 2004; doi.org/10.1126/science.277.5322.112 1097/QAI.0b013e3181e9be6b
199:905-15; PMID:15067030; http://dx.doi.org/10. 54. Perelson AS, Neumann AU, Markowitz M, Leonard 67. Lodwick RK, Sabin CA, Porter K, Ledergerber B, van
1084/jem.20031982 JM, Ho DD. HIV-1 dynamics in vivo: virion clearance Sighem A, Cozzi-Lepri A, et al. Study Group on Death
41. Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo rate, infected cell life-span, and viral generation time. Rates at High CD4 Count in Antiretroviral Naive
MM, Feeney M, et al. HIV evolution: CTL escape Science 1996; 271:1582-6; PMID:8599114; http:// Patients. Death rates in HIV-positive antiretroviral-
mutation and reversion after transmission. Nat Med dx.doi.org/10.1126/science.271.5255.1582 naive patients with CD4 count greater than 350 cells
2004; 10:282-9; PMID:14770175; http://dx.doi.org/ 55. Perelson AS, Essunger P, Cao Y. Decay characteristics per microL in Europe and North America: a pooled
10.1038/nm992 of HIV-1 infected compartments during combination cohort observational study. Lancet 2010; 376:340-5;

2012 Landes Bioscience.


42. Goulder PJ, Brander C, Tang Y, Tremblay C, Colbert therapy. In: 1997. pp. 188-191. PMID:20638118; http://dx.doi.org/10.1016/S0140-
RA, Addo MM, et al. Evolution and transmission of 56. Finzi D, Blankson J, Siliciano JD, Margolick JB, 6736(10)60932-4
stable CTL escape mutations in HIV infection. Nature Chadwick K, Pierson T, et al. Latent infection of 68. Kitahata MM, Gange SJ, Abraham AG, Merriman B,
2001; 412:334-8; PMID:11460164; http://dx.doi.org/ CD4+ T cells provides a mechanism for lifelong Saag MS, Justice AC, et al. NA-ACCORD Investi-
10.1038/35085576 persistence of HIV-1, even in patients on effective gators. Effect of early versus deferred antiretroviral
43. Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo combination therapy. Nat Med 1999; 5:512-7; PMID: therapy for HIV on survival. N Engl J Med 2009;
MM, Feeney M, et al. HIV evolution: CTL escape 10229227; http://dx.doi.org/10.1038/8394 360:1815-26; PMID:19339714; http://dx.doi.org/10.
mutation and reversion after transmission. Nat Med 57. Blankson JN, Persaud D, Siliciano RF. The challenge 1056/NEJMoa0807252
2004; 10:282-9; PMID:14770175; http://dx.doi.org/ of viral reservoirs in HIV-1 infection. Annu Rev Med 69. El-Sadr WM, Lundgren JD, Neaton JD, Gordin F,

Do not distribute.
10.1038/nm992 2002; 53:557-93; PMID:11818490; http://dx.doi.org/ Abrams D, Arduino RC, et al. Strategies for Manage-
44. Pitcher CJ, Quittner C, Peterson DM, Connors M, 10.1146/annurev.med.53.082901.104024 ment of Antiretroviral Therapy (SMART) Study
Koup RA, Maino VC, et al. HIV-1-specific CD4+ 58. Zhang L, Ramratnam B, Tenner-Racz K, He Y, Group. CD4+ count-guided interruption of anti-
T cells are detectable in most individuals with active Vesanen M, Lewin S, et al. Quantifying residual retroviral treatment. N Engl J Med 2006; 355:2283-
HIV-1 infection, but decline with prolonged viral HIV-1 replication in patients receiving combination 96; PMID:17135583; http://dx.doi.org/10.1056/
suppression. Nat Med 1999; 5:518-25; PMID: antiretroviral therapy. N Engl J Med 1999; 340: NEJMoa062360
10229228; http://dx.doi.org/10.1038/8400 1605-13; PMID:10341272; http://dx.doi.org/10. 70. Kahn JO, Walker BD. Current concepts: acute
45. Plana M, Garcia F, Gallart MT, Miro JM, Gatell JM. 1056/NEJM199905273402101 human immunodeficiency virus type 1 infection. In:
Lack of T-cell proliferative response to HIV-1 antigens 59. Furtado MR, Callaway DS, Phair JP, Kunstman KJ, 1998. pp. 33-39.
after one year of HAART in very early HIV-1 disease. Stanton JL, Macken CA, et al. Persistence of HIV-1 71. Mocroft A, Bofill M, Lipman M, Medina E,
Lancet 1998; 352:1194-5; PMID:9777842; http://dx. transcription in peripheral-blood mononuclear cells Borthwick N, Timms A, et al. CD8+,CD38+
doi.org/10.1016/S0140-6736(05)60532-6 in patients receiving potent antiretroviral therapy. lymphocyte percent: a useful immunological marker
46. Yokomaku Y, Miura H, Tomiyama H, Kawana- [see comments]. N Engl J Med 1999; 340:1614-22; for monitoring HIV-1-infected patients. J Acquir
Tachikawa A, Takiguchi M, Kojima A, et al. PMID:10341273; http://dx.doi.org/10.1056/ Immune Defic Syndr Hum Retrovirol 1997; 14:158-
Impaired processing and presentation of CTL epitopes NEJM199905273402102 62; PMID:9052725; http://dx.doi.org/10.1097/
are major escape mechanisms from CTL immune 60. Gamberg J, Barrett L, Bowmer MI, Howley C, Grant 00042560-199702010-00009
pressure in Human Immunodeficiency Virus type 1 M. Factors related to loss of HIV-specific cytotoxic T 72. Bofill M, Mocroft A, Lipman M, Medina E, Borthwick
infection. J Virol 2004; 78:1324-32; PMID: lymphocyte activity. AIDS 2004; 18:597-604; PMID: NJ, Sabin CA, et al. Increased numbers of primed
14722287; http://dx.doi.org/10.1128/JVI.78.3.1324- 15090764; http://dx.doi.org/10.1097/00002030- activated CD8+CD38+CD45RO+ T cells predict the
1332.2004 200403050-00003 decline of CD4+ T cells in HIV-1-infected patients.
47. Seder RA, Ahmed R. Similarities and differences in 61. Lange CG, Xu Z, Patterson BK, Medvik K, Harnisch AIDS 1996; 10:827-34; PMID:8828739; http://dx.doi.
CD4+ and CD8+ effector and memory T cell B, Asaad R, et al. Proliferation responses to HIVp24 org/10.1097/00002030-199607000-00005
generation. Nat Immunol 2003; 4:835-42; PMID: during antiretroviral therapy do not reflect improved 73. Diaz A, Garca F, Mozos A, Caballero M, Len A,
12942084; http://dx.doi.org/10.1038/ni969 immune phenotype or function. AIDS 2004; 18: Martinez A, et al. Lymphoid tissue collagen deposition
48. Yang OO, Sarkis PT, Trocha A, Kalams SA, Johnson 605-13; PMID:15090765; http://dx.doi.org/10.1097/ in HIV-infected patients correlates with the imbalance
RP, Walker BD. Impacts of avidity and specificity 00002030-200403050-00004 between matrix metalloproteinases and their inhibitors.
on the antiviral efficiency of HIV-1-specific CTL. J 62. Garca F, Plana M, Vidal C, Cruceta A, OBrien WA, J Infect Dis 2011; 203:810-3; PMID:21343147;
Immunol 2003; 171:3718-24; PMID:14500671 Pantaleo G, et al. Dynamics of viral load rebound and http://dx.doi.org/10.1093/infdis/jiq129
49. Donaghy H, Gazzard B, Gotch F, Patterson S. immunological changes after stopping effective anti- 74. Diaz A, Als L, Len A, Mozos A, Caballero M,
Dysfunction and infection of freshly isolated blood retroviral therapy. AIDS 1999; 13:F79-86; PMID: Martinez A, et al. Study Group of Lymphoid Tissue
myeloid and plasmacytoid dendritic cells in patients 10449278; http://dx.doi.org/10.1097/00002030- immunopathogenesis in HIV infection. Factors asso-
infected with HIV-1. Blood 2003; 101:4505-11; 199907300-00002 ciated with collagen deposition in lymphoid tissue in
PMID:12576311; http://dx.doi.org/10.1182/blood- 63. Ruiz L, Martinez-Picado J, Romeu J, Paredes R, Zayat long-term treated HIV-infected patients. AIDS 2010;
2002-10-3189 MK, Marfil S, et al. Structured treatment interruption 24:2029-39; PMID:20588162; http://dx.doi.org/10.
50. Chehimi J, Campbell DE, Azzoni L, Bacheller D, in chronically HIV-1 infected patients after long-term 1097/QAD.0b013e32833c3268
Papasavvas E, Jerandi G, et al. Persistent decreases in viral suppression. [Abstract 354 ] [Abstract]. AIDS
blood plasmacytoid dendritic cell number and func- 2000; 14:397-403; PMID:10770542; http://dx.doi.
tion despite effective highly active antiretroviral org/10.1097/00002030-200003100-00013
therapy and increased blood myeloid dendritic cells
in HIV-infected individuals. J Immunol 2002; 168:
4796-801; PMID:11971031

578 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


75. Garrido M, Mozos A, Martnez A, Garca F, Serafn A, 87. Robbins GK, Addo MM, Troung H, Rathod A, 98. Valentine FT, Kundu S, Haslett PA, Katzenstein D,
Morente V, et al. HIV-1 upregulates intercellular Habeeb K, Davis B, et al. Augmentation of HIV-1- Beckett L, Spino C, et al. A randomized, placebo-
adhesion molecule-1 gene expression in lymphoid specific T helper cell responses in chronic HIV-1 controlled study of the immunogenicity of human
tissue of patients with chronic HIV-1 infection. infection by therapeutic immunization. AIDS 2003; immunodeficiency virus (HIV) rgp160 vaccine in
J Acquir Immune Defic Syndr 2007; 46:268-74; 17:1121-6; PMID:12819512; http://dx.doi.org/10. HIV-infected subjects with . or = 400/mm3 CD4
PMID:17786132; http://dx.doi.org/10.1097/QAI. 1097/00002030-200305230-00002 T lymphocytes (AIDS Clinical Trials Group Protocol
0b013e318142c74c 88. Lederman MM, Douek DC. Sometimes help may 137). J Infect Dis 1996; 173:1336-46; PMID:
76. Mozos A, Garrido M, Carreras J, Plana M, Diaz A, not be enough. AIDS 2003; 17:1249-51; PMID: 8648205; http://dx.doi.org/10.1093/infdis/173.6.
Alos L, et al. Redistribution of FOXP3-positive 12819528; http://dx.doi.org/10.1097/00002030- 1336
regulatory T cells from lymphoid tissues to peripheral 200305230-00018 99. Wahren B, Bratt G, Persson C, Levn B, Hinkula J,
blood in HIV-infected patients. J Acquir Immune 89. Kahn JO, Cherng DW, Mayer K, Murray H, Lagakos Gilljam G, et al. Improved cell-mediated immune
Defic Syndr 2007; 46:529-37; PMID:18193494; S. Evaluation of HIV-1 immunogen, an immunologic responses in HIV-1-infected asymptomatic individuals
http://dx.doi.org/10.1097/QAI.0b013e31815b69ae modifier, administered to patients infected with HIV after immunization with envelope glycoprotein gp160.
77. Markowitz M, Jin X, Hurley A, Simon V, Ramratnam having 300 to 549 x 10(6)/L CD4 cell counts: A J Acquir Immune Defic Syndr 1994; 7:220-9; PMID:
B, Louie M, et al. Discontinuation of antiretroviral randomized controlled trial. JAMA 2000; 284:2193- 7906300
therapy commenced early during the course of human 202; PMID:11056590; http://dx.doi.org/10.1001/ 100. Wright PF, Lambert JS, Gorse GJ, Hsieh RH,
immunodeficiency virus type 1 infection, with or jama.284.17.2193 McElrath MJ, Weinhold K, et al. Immunization with
without adjunctive vaccination. J Infect Dis 2002; 90. Lichterfeld M, Kaufmann DE, Yu XG, Mui SK, envelope MN rgp120 vaccine in human immuno-
186:634-43; PMID:12195350; http://dx.doi.org/10. Addo MM, Johnston MN, et al. Loss of HIV-1- deficiency virus-infected pregnant women. J Infect Dis
1086/342559 specific CD8+ T cell proliferation after acute HIV-1 1999; 180:1080-8; PMID:10479134; http://dx.doi.
78. MacGregor RR, Ginsberg R, Ugen KE, Baine Y, Kang infection and restoration by vaccine-induced HIV-1- org/10.1086/314985
CU, Tu XM, et al. T-cell responses induced in normal specific CD4+ T cells. J Exp Med 2004; 200:701-12; 101. Kran AM, Sommerfelt MA, Srensen B, Nyhus J,
volunteers immunized with a DNA-based vaccine PMID:15381726; http://dx.doi.org/10.1084/jem. Baksaas I, Bruun JN, et al. Reduced viral burden
containing HIV-1 env and rev. AIDS 2002; 16:2137- 20041270 amongst high responder patients following HIV-1 p24
43; PMID:12409734; http://dx.doi.org/10.1097/ 91. Gorse GJ, Simionescu RE, Patel GB. Cellular immune peptide-based therapeutic immunization. Vaccine
00002030-200211080-00005 responses in asymptomatic human immunodeficiency 2005; 23:4011-5; PMID:15963359; http://dx.doi.

2012 Landes Bioscience.


79. MacGregor RR, Boyer JD, Ciccarelli RB, Ginsberg virus type 1 (HIV-1) infection and effects of vac- org/10.1016/j.vaccine.2005.03.010
RS, Weiner DB. Safety and immune responses to a cination with recombinant envelope glycoprotein of 102. Kran AM, Srensen B, Sommerfelt MA, Nyhus J,
DNA-based human immunodeficiency virus (HIV) HIV-1. Clin Vaccine Immunol 2006; 13:26-32; Baksaas I, Kvale D. Long-term HIV-specific responses
type I env/rev vaccine in HIV-infected recipients: PMID:16425996; http://dx.doi.org/10.1128/CVI.13. and delayed resumption of antiretroviral therapy after
follow-up data. J Infect Dis 2000; 181:406; PMID: 1.26-32.2006 peptide immunization targeting dendritic cells. AIDS
10608800; http://dx.doi.org/10.1086/315199 92. Birx DL, Loomis-Price LD, Aronson N, Brundage J, 2006; 20:627-30; PMID:16470131; http://dx.doi.org/
80. Schooley RT, Spino C, Kuritzkes D, Walker BD, Davis C, Deyton L, et al. Efficacy testing of recom- 10.1097/01.aids.0000210620.75707.ac
Valentine FA, Hirsch MS, et al. Two double-blinded, binant human immunodeficiency virus (HIV) gp160 103. Kran AM, Srensen B, Nyhus J, Sommerfelt MA,
randomized, comparative trials of 4 human immuno- as a therapeutic vaccine in early-stage HIV-1-infected Baksaas I, Bruun JN, et al. HLA- and dose-dependent

Do not distribute.
deficiency virus type 1 (HIV-1) envelope vaccines in volunteers. rgp160 Phase II Vaccine Investigators. J immunogenicity of a peptide-based HIV-1 immuno-
HIV-1-infected individuals across a spectrum of disease Infect Dis 2000; 181:881-9; PMID:10720508; http:// therapy candidate (Vacc-4x). AIDS 2004; 18:1875-
severity: AIDS Clinical Trials Groups 209 and 214. J dx.doi.org/10.1086/315308 83; PMID:15353973; http://dx.doi.org/10.1097/
Infect Dis 2000; 182:1357-64; PMID:11023459; 93. Eron JJ, Jr., Ashby MA, Giordano MF, Chernow M, 00002030-200409240-00003
http://dx.doi.org/10.1086/315860 Reiter WM, Deeks SG, et al. Randomised trial of 104. Ensoli B, Bellino S, Tripiciano A, Longo O,
81. Smith D, Gow I, Colebunders R, Weller I, MNrgp120 HIV-1 vaccine in symptomless HIV-1 Francavilla V, Marcotullio S, et al. Therapeutic
Tchamouroff S, Weber J, et al. 006 Study Group. infection. Lancet 1996; 348:1547-51; PMID:8950881; immunization with HIV-1 Tat reduces immune
Therapeutic vaccination (p24-VLP) of patients with http://dx.doi.org/10.1016/S0140-6736(96)05283-X activation and loss of regulatory T-cells and improves
advanced HIV-1 infection in the pre-HAART era does 94. Pontesilli O, Guerra EC, Ammassari A, Tomino C, immune function in subjects on HAART. PLoS One
not alter CD4 cell decline. HIV Med 2001; 2:272-5; Carlesimo M, Antinori A, et al. Phase II controlled trial 2010; 5:e13540; PMID:21085635; http://dx.doi.org/
PMID:11737409; http://dx.doi.org/10.1046/j.1468- of post-exposure immunization with recombinant 10.1371/journal.pone.0013540
1293.2001.00080.x gp160 versus antiretroviral therapy in asymptomatic 105. Longo O, Tripiciano A, Fiorelli V, Bellino S, Scoglio
82. Trauger RJ, Daigle AE, Giermakowska W, Moss RB, HIV-1-infected adults. VaxSyn Protocol Team. AIDS A, Collacchi B, et al. Phase I therapeutic trial of the
Jensen F, Carlo DJ. Safety and immunogenicity of 1998; 12:473-80; PMID:9543445; http://dx.doi.org/ HIV-1 Tat protein and long term follow-up. Vaccine
a gp120-depleted, inactivated HIV-1 immunogen: 10.1097/00002030-199805000-00008 2009; 27:3306-12; PMID:19208456; http://dx.doi.
results of a double-blind, adjuvant controlled trial. J 95. Sitz KV, Ratto-Kim S, Hodgkins AS, Robb ML, Birx org/10.1016/j.vaccine.2009.01.090
Acquir Immune Defic Syndr Hum Retrovirol 1995; DL. Proliferative responses to human immunodefi- 106. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R,
10(Suppl 2):S74-82; PMID:7552517 ciency virus type 1 (HIV-1) gp120 peptides in HIV-1- Narciso P, et al. The therapeutic phase I trial of the
83. Eron JJ, Jr., Ashby MA, Giordano MF, Chernow M, infected individuals immunized with HIV-1 rgp120 or recombinant native HIV-1 Tat protein. AIDS 2008;
Reiter WM, Deeks SG, et al. Randomised trial of rgp160 compared with nonimmunized and uninfected 22:2207-9; PMID:18832884; http://dx.doi.org/10.
MNrgp120 HIV-1 vaccine in symptomless HIV-1 controls. J Infect Dis 1999; 179:817-24; PMID: 1097/QAD.0b013e32831392d4
infection. Lancet 1996; 348:1547-51; PMID: 10068576; http://dx.doi.org/10.1086/314685 107. Lichterfeld M, Gandhi RT, Simmons RP, Flynn T,
8950881; http://dx.doi.org/10.1016/S0140-6736(96) 96. Redfield RR, Birx DL, Ketter N, Tramont E, Polonis Sbrolla A, Yu XG, et al. Induction of Strong HIV-1-
05283-X V, Davis C, et al. Military Medical Consortium for specific CD4+ T Cell Responses using an HIV-1
84. Sandstrm E, Wahren B. Therapeutic immunisation Applied Retroviral Research. A phase I evaluation of gp120/NefTat Vaccine adjuvanted with AS02A in
with recombinant gp160 in HIV-1 infection: a the safety and immunogenicity of vaccination with ARV Treated HIV-1-Infected Individuals. J Acquir
randomised double-blind placebo-controlled trial. recombinant gp160 in patients with early human Immune Defic Syndr 2011
Nordic VAC-04 Study Group. Lancet 1999; 353: immunodeficiency virus infection. N Engl J Med 108. Cafaro A, Titti F, Fracasso C, Maggiorella MT,
1735-42; PMID:10347985; http://dx.doi.org/10. 1991; 324:1677-84; PMID:1674589; http://dx.doi. Baroncelli S, Caputo A, et al. Vaccination with DNA
1016/S0140-6736(98)06493-9 org/10.1056/NEJM199106133242401 containing tat coding sequences and unmethylated
85. Peters BS, Cheingsong-Popov R, Callow D, Foxall R, 97. Schooley RT, Spino C, Kuritzkes D, Walker BD, CpG motifs protects cynomolgus monkeys upon
Patou G, Hodgkin K, et al. A pilot phase II study of Valentine FA, Hirsch MS, et al. Two double-blinded, infection with simian/human immunodeficiency virus
the safety and immunogenicity of HIV p17/p24:VLP randomized, comparative trials of 4 human immuno- (SHIV89.6P). Vaccine 2001; 19:2862-77; PMID:
(p24-VLP) in asymptomatic HIV seropositive subjects. deficiency virus type 1 (HIV-1) envelope vaccines in 11282197; http://dx.doi.org/10.1016/S0264-410X
J Infect 1997; 35:231-5; PMID:9459393; http://dx. HIV-1-infected individuals across a spectrum of disease (01)00002-0
doi.org/10.1016/S0163-4453(97)92814-0 severity: AIDS Clinical Trials Groups 209 and 214. J
86. MacGregor RR, Boyer JD, Ugen KE, Lacy KE, Infect Dis 2000; 182:1357-64; PMID:11023459;
Gluckman SJ, Bagarazzi ML, et al. First human trial http://dx.doi.org/10.1086/315860
of a DNA-based vaccine for treatment of human
immunodeficiency virus type 1 infection: safety and
host response. J Infect Dis 1998; 178:92-100; PMID:
9652427

www.landesbioscience.com Human Vaccines & Immunotherapeutics 579


109. Allen TM, OConnor DH, Jing P, Dzuris JL, Moth 122. Cebere I, Dorrell L, McShane H, Simmons A, 132. Lvy Y, Durier C, Lascaux AS, Meiffrdy V, Gahry-
BR, Vogel TU, et al. Tat-specific cytotoxic T lympho- McCormack S, Schmidt C, et al. Phase I clinical trial Sgard H, Goujard C, et al. ANRS 093 Study Group.
cytes select for SIV escape variants during resolution of safety of DNA- and modified virus Ankara-vectored Sustained control of viremia following therapeutic
primary viraemia. Nature 2000; 407:386-90; PMID: human immunodeficiency virus type 1 (HIV-1) immunization in chronically HIV-1-infected individuals.
11014195; http://dx.doi.org/10.1038/35036559 vaccines administered alone and in a prime-boost AIDS 2006; 20:405-13; PMID:16439874; http://dx.
110. Fernandez-Cruz E, Moreno S, Navarro J, Clotet B, regime to healthy HIV-1-uninfected volunteers. doi.org/10.1097/01.aids.0000206504.09159.d3
Bouza E, Carbone J, et al. STIR-2102 TEAM. Vaccine 2006; 24:417-25; PMID:16176847; http:// 133. Emery S, Kelleher AD, Workman C, Puls RL, Bloch
Therapeutic immunization with an inactivated HIV- dx.doi.org/10.1016/j.vaccine.2005.08.041 M, Baker D, et al. Influence of IFNgamma co-
1 Immunogen plus antiretrovirals versus antiretroviral 123. Goonetilleke N, Moore S, Dally L, Winstone N, expression on the safety and antiviral efficacy of
therapy alone in asymptomatic HIV-infected subjects. Cebere I, Mahmoud A, et al. Induction of multi- recombinant fowlpox virus HIV therapeutic vaccines
Vaccine 2004; 22:2966-73; PMID:15297045; http:// functional human immunodeficiency virus type 1 following interruption of antiretroviral therapy. Hum
dx.doi.org/10.1016/j.vaccine.2004.03.040 (HIV-1)-specific T cells capable of proliferation in Vaccin 2007; 3:260-7; PMID:18340117
111. Calarota S, Bratt G, Nordlund S, Hinkula J, healthy subjects by using a prime-boost regimen of 134. Stickl H, Hochstein-Mintzel V, Mayr A, Huber HC,
Leandersson AC, Sandstrm E, et al. Cellular cytotoxic DNA- and modified vaccinia virus Ankara-vectored Schfer H, Holzner A. [MVA vaccination against
response induced by DNA vaccination in HIV-1- vaccines expressing HIV-1 Gag coupled to CD8+ T-cell smallpox: clinical tests with an attenuated live vaccinia
infected patients. Lancet 1998; 351:1320-5; PMID: epitopes. J Virol 2006; 80:4717-28; PMID:16641265; virus strain (MVA) (authors transl)]. Dtsch Med
9643795; http://dx.doi.org/10.1016/S0140-6736(97) http://dx.doi.org/10.1128/JVI.80.10.4717-4728.2006 Wochenschr 1974; 99:2386-92; PMID:4426258;
09440-3 124. Kinloch-de Loes S, Hoen B, Smith DE, Autran B, http://dx.doi.org/10.1055/s-0028-1108143
112. Hejdeman B, Bostrm AC, Matsuda R, Calarota S, Lampe FC, Phillips AN, et al. QUEST Study Group. 135. Ramrez JC, Gherardi MM, Esteban M. Biology of
Lenkei R, Fredriksson EL, et al. DNA immunization Impact of therapeutic immunization on HIV-1 viremia attenuated modified vaccinia virus Ankara recom-
with HIV early genes in HIV type 1-infected patients after discontinuation of antiretroviral therapy initiated binant vector in mice: virus fate and activation of B-
on highly active antiretroviral therapy. AIDS Res Hum during acute infection. J Infect Dis 2005; 192:607-17; and T-cell immune responses in comparison with the
Retroviruses 2004; 20:860-70; PMID:15320990; PMID:16028129; http://dx.doi.org/10.1086/432002 Western Reserve strain and advantages as a vaccine. J
http://dx.doi.org/10.1089/0889222041725190 125. Jin X, Ramanathan M, Jr., Barsoum S, Deschenes GR, Virol 2000; 74:923-33; PMID:10623755; http://dx.
113. MacGregor RR, Boyer JD, Ugen KE, Tebas P, Higgins Ba L, Binley J, et al. Safety and immunogenicity of doi.org/10.1128/JVI.74.2.923-933.2000
TJ, Baine Y, et al. Plasmid vaccination of stable HIV- ALVAC vCP1452 and recombinant gp160 in newly 136. Barouch DH, Santra S, Kuroda MJ, Schmitz JE,

2012 Landes Bioscience.


positive subjects on antiviral treatment results in human immunodeficiency virus type 1-infected patients Plishka R, Buckler-White A, et al. Reduction of
enhanced CD8 T-cell immunity and increased control treated with prolonged highly active antiretroviral simian-human immunodeficiency virus 89.6P viremia
of viral blips. Vaccine 2005; 23:2066-73; PMID: therapy. J Virol 2002; 76:2206-16; PMID:11836398; in rhesus monkeys by recombinant modified vaccinia
15755572; http://dx.doi.org/10.1016/j.vaccine.2005. http://dx.doi.org/10.1128/jvi.76.5.2206-2216.2002 virus Ankara vaccination. J Virol 2001; 75:5151-8;
01.010 126. Lvy Y, Gahry-Sgard H, Durier C, Lascaux AS, PMID:11333896; http://dx.doi.org/10.1128/JVI.75.
114. Dorrell L, Yang H, Iversen AK, Conlon C, Suttill A, Goujard C, Meiffrdy V, et al. ANRS 093 Study 11.5151-5158.2001
Lancaster M, et al. Therapeutic immunization of Group. Immunological and virological efficacy of a 137. Ourmanov I, Brown CR, Moss B, Carroll M, Wyatt L,
highly active antiretroviral therapy-treated HIV-1- therapeutic immunization combined with interleukin- Pletneva L, et al. Comparative efficacy of recombinant
infected patients: safety and immunogenicity of an 2 in chronically HIV-1 infected patients. AIDS 2005; modified vaccinia virus Ankara expressing simian

Do not distribute.
HIV-1 gag/poly-epitope DNA vaccine. AIDS 2005; 19:279-86; PMID:15718838 immunodeficiency virus (SIV) Gag-Pol and/or Env
19:1321-3; PMID:16052088; http://dx.doi.org/10. 127. Johnson DC, McFarland EJ, Muresan P, Fenton T, in macaques challenged with pathogenic SIV. J Virol
1097/01.aids.0000180104.65640.16 McNamara J, Read JS, et al. Safety and immunogeni- 2000; 74:2740-51; PMID:10684290; http://dx.doi.
115. Lisziewicz J, Trocio J, Xu J, Whitman L, Ryder A, city of an HIV-1 recombinant canarypox vaccine in org/10.1128/JVI.74.6.2740-2751.2000
Bakare N, et al. Control of viral rebound through newborns and infants of HIV-1-infected women. J 138. Amara RR, Villinger F, Staprans SI, Altman JD,
therapeutic immunization with DermaVir. AIDS Infect Dis 2005; 192:2129-33; PMID:16288378; Montefiori DC, Kozyr NL, et al. Different patterns of
2005; 19:35-43; PMID:15627031; http://dx.doi.org/ http://dx.doi.org/10.1086/498163 immune responses but similar control of a simian-
10.1097/00002030-200501030-00004 128. Tubiana R, Carcelain G, Vray M, Gourlain K, Dalban human immunodeficiency virus 89.6P mucosal chal-
116. Gudmundsdotter L, Wahren B, Haller BK, Boberg A, C, Chermak A, et al. for the Vacciter Study group. lenge by modified vaccinia virus Ankara (MVA) and
Edbck U, Bernasconi D, et al. Amplified antigen- Therapeutic immunization with a human immuno- DNA/MVA vaccines. J Virol 2002; 76:7625-31;
specific immune responses in HIV-1 infected indivi- deficiency virus (HIV) type 1-recombinant canarypox PMID:12097576; http://dx.doi.org/10.1128/JVI.76.
duals in a double blind DNA immunization and vaccine in chronically HIV-infected patients: The 15.7625-7631.2002
therapy interruption trial. Vaccine 2011; 29:5558-66; Vacciter Study (ANRS 094). Vaccine 2005; 23: 139. Cosma A, Nagaraj R, Bhler S, Hinkula J, Busch DH,
PMID:21300092; http://dx.doi.org/10.1016/j.vaccine. 4292-301; PMID:15927325; http://dx.doi.org/10. Sutter G, et al. Therapeutic vaccination with MVA-
2011.01.064 1016/j.vaccine.2005.04.013 HIV-1 nef elicits Nef-specific T-helper cell responses
117. Pantaleo G, Esteban M, Jacobs B, Tartaglia J. Poxvirus 129. Jacobson JM, Pat Bucy R, Spritzler J, Saag MS, Eron in chronically HIV-1 infected individuals. Vaccine
vector-based HIV vaccines. Curr Opin HIV AIDS JJ, Jr., Coombs RW, et al. National Institute of Allergy 2003; 22:21-9; PMID:14604567; http://dx.doi.org/
2010; 5:391-6; PMID:20978379; http://dx.doi.org/ and Infectious Diseases-AIDS Clinical Trials Group 10.1016/S0264-410X(03)00538-3
10.1097/COH.0b013e32833d1e87 5068 Protocol Team. Evidence that intermittent 140. Harrer E, Buerle M, Ferstl B, Chaplin P, Petzold B,
118. Stickl H, Hochstein-Mintzel V, Mayr A, Huber HC, structured treatment interruption, but not immuniza- Mateo L, et al. Therapeutic vaccination of HIV-1-
Schfer H, Holzner A. [MVA vaccination against tion with ALVAC-HIV vCP1452, promotes host infected patients on HAART with a recombinant HIV-1
smallpox: clinical tests with an attenuated live vaccinia control of HIV replication: the results of AIDS nef-expressing MVA: safety, immunogenicity and influ-
virus strain (MVA) (authors transl)]. Dtsch Med Clinical Trials Group 5068. J Infect Dis 2006; ence on viral load during treatment interruption. Antivir
Wochenschr 1974; 99:2386-92; PMID:4426258; 194:623-32; PMID:16897661; http://dx.doi.org/10. Ther 2005; 10:285-300; PMID:15865223
http://dx.doi.org/10.1055/s-0028-1108143 1086/506364 141. Dorrell L, Yang H, Ondondo B, Dong T, di Gleria K,
119. Antoine G, Scheiflinger F, Dorner F, Falkner FG. The 130. Autran B, Murphy RL, Costagliola D, Tubiana R, Suttill A, et al. Expansion and diversification of virus-
complete genomic sequence of the modified vaccinia Clotet B, Gatell J, et al. ORVACS Study Group. specific T cells following immunization of human
Ankara strain: comparison with other orthopoxviruses. Greater viral rebound and reduced time to resume immunodeficiency virus type 1 (HIV-1)-infected
Virology 1998; 244:365-96; PMID:9601507; http:// antiretroviral therapy after therapeutic immunization individuals with a recombinant modified vaccinia virus
dx.doi.org/10.1006/viro.1998.9123 with the ALVAC-HIV vaccine (vCP1452). AIDS Ankara/HIV-1 Gag vaccine. J Virol 2006; 80:4705-16;
120. Stickl HA. Smallpox vaccination and its consequences: 2008; 22:1313-22; PMID:18580611; http://dx.doi. PMID:16641264; http://dx.doi.org/10.1128/JVI.80.
first experiences with the highly attenuated smallpox org/10.1097/QAD.0b013e3282fdce94 10.4705-4716.2006
vaccine MVA. Prev Med 1974; 3:97-101; PMID: 131. Angel JB, Routy JP, Tremblay C, Ayers D, Woods R,
4205586; http://dx.doi.org/10.1016/0091-7435(74) Singer J, et al. A randomized controlled trial of HIV
90066-8 therapeutic vaccination using ALVAC with or without
121. Mwau M, Cebere I, Sutton J, Chikoti P, Winstone N, Remune. AIDS 2011; 25:731-9; PMID:21330911;
Wee EG, et al. A human immunodeficiency virus 1 http://dx.doi.org/10.1097/QAD.0b013e328344cea5
(HIV-1) clade A vaccine in clinical trials: stimulation
of HIV-specific T-cell responses by DNA and recom-
binant modified vaccinia virus Ankara (MVA) vaccines
in humans. J Gen Virol 2004; 85:911-9; PMID:
15039533; http://dx.doi.org/10.1099/vir.0.19701-0

580 Human Vaccines & Immunotherapeutics Volume 8 Issue 5


142. Ondondo BO, Yang H, Dong T, di Gleria K, Suttill A, 153. Rissoan MC, Soumelis V, Kadowaki N, Grouard G, 164. Garca F, Lejeune M, Climent N, Gil C, Alcami J,
Conlon C, et al. Immunisation with recombinant Briere F, de Waal Malefyt R, et al. Reciprocal control Morente V, et al. Therapeutic immunization with
modified vaccinia virus Ankara expressing HIV-1 gag of T helper cell and dendritic cell differentiation. dendritic cells loaded with inactivated autologous
in HIV-1-infected subjects stimulates broad func- Science 1999; 283:1183-6; PMID:10024247; http:// HIV-1 in chronic HIV-1 infected patients. J Infect Dis
tional CD4+ T cell responses. Eur J Immunol 2006; dx.doi.org/10.1126/science.283.5405.1183 2005; 195:1680-5; http://dx.doi.org/10.1086/429340
36:2585-94; PMID:17013989; http://dx.doi.org/10. 154. Filgueira L, Nestl FO, Rittig M, Joller HI, Groscurth 165. Ide F, Nakamura T, Tomizawa M, Kawana-Tachikawa
1002/eji.200636508 P. Human dendritic cells phagocytose and process A, Odawara T, Hosoya N, et al. Peptide-loaded
143. Dorrell L, Williams P, Suttill A, Brown D, Roberts J, Borrelia burgdorferi. J Immunol 1996; 157:2998- dendritic-cell vaccination followed by treatment inter-
Conlon C, et al. Safety and tolerability of recombinant 3005; PMID:8816408 ruption for chronic HIV-1 infection: a phase 1 trial. J
modified vaccinia virus Ankara expressing an HIV-1 gag/ 155. Mbow ML, Zeidner N, Panella N, Titus RG, Piesman Med Virol 2006; 78:711-8; PMID:16628588; http://
multiepitope immunogen (MVA.HIVA) in HIV-1- J. Borrelia burgdorferi-pulsed dendritic cells induce a dx.doi.org/10.1002/jmv.20612
infected persons receiving combination antiretroviral protective immune response against tick-transmitted 166. Connolly NC, Whiteside TL, Wilson C, Kondragunta
therapy. Vaccine 2007; 25:3277-83; PMID:17257714; spirochetes. Infect Immun 1997; 65:3386-90; PMID: V, Rinaldo CR, Riddler SA. Therapeutic immuniza-
http://dx.doi.org/10.1016/j.vaccine.2007.01.005 9234802 tion with human immunodeficiency virus type 1
144. Schooley RT, Spritzler J, Wang H, Lederman MM, 156. Su H, Messer R, Whitmire W, Fischer E, Portis JC, (HIV-1) peptide-loaded dendritic cells is safe and
Havlir D, Kuritzkes DR, et al. AIDS Clinical Trials Caldwell HD. Vaccination against chlamydial genital induces immunogenicity in HIV-1-infected individuals.
Group 5197 Study Team. AIDS clinical trials group tract infection after immunization with dendritic cells Clin Vaccine Immunol 2008; 15:284-92; PMID:
5197: a placebo-controlled trial of immunization of pulsed ex vivo with nonviable Chlamydiae. J Exp Med 17942609; http://dx.doi.org/10.1128/CVI.00221-07
HIV-1-infected persons with a replication-deficient 1998; 188:809-18; PMID:9730883; http://dx.doi.org/ 167. Gandhi RT, ONeill D, Bosch RJ, Chan ES, Bucy RP,
adenovirus type 5 vaccine expressing the HIV-1 core 10.1084/jem.188.5.809 Shopis J, et al. AIDS Clinical Trials Group A5130 team.
protein. J Infect Dis 2010; 202:705-16; PMID: 157. Brossart P, Goldrath AW, Butz EA, Martin S, Bevan A randomized therapeutic vaccine trial of canarypox-
20662716; http://dx.doi.org/10.1086/655468 MJ. Virus-mediated delivery of antigenic epitopes into HIV-pulsed dendritic cells vs. canarypox-HIV alone
145. Li JZ, Brumme ZL, Brumme CJ, Wang H, Spritzler J, dendritic cells as a means to induce CTL. J Immunol in HIV-1-infected patients on antiretroviral therapy.
Robertson MN, et al. AIDS Clinical Trials Group 1997; 158:3270-6; PMID:9120283 Vaccine 2009; 27:6088-94; PMID:19450647; http://
A5197 Study Team. Factors associated with viral 158. Svensson M, Stockinger B, Wick MJ. Bone marrow- dx.doi.org/10.1016/j.vaccine.2009.05.016
rebound in HIV-1-infected individuals enrolled in a derived dendritic cells can process bacteria for MHC-I 168. Kloverpris H, Karlsson I, Bonde J, Thorn M, Vinner

2012 Landes Bioscience.


therapeutic HIV-1 gag vaccine trial. J Infect Dis 2011; and MHC-II presentation to T cells. J Immunol 1997; L, Pedersen AE, et al. Induction of novel CD8+ T-cell
203:976-83; PMID:21402549; http://dx.doi.org/10. 158:4229-36; PMID:9126984 responses during chronic untreated HIV-1 infection
1093/infdis/jiq143 159. Lu W, Wu X, Lu Y, Guo W, Andrieu JM. Therapeutic by immunization with subdominant cytotoxic T-
146. Banchereau J, Steinman RM. Dendritic cells and the dendritic-cell vaccine for simian AIDS. Nat Med 2003; lymphocyte epitopes. AIDS 2009; 23:1329-40;
control of immunity. Nature 1998; 392:245-52; 9:27-32; PMID:12496959; http://dx.doi.org/10.1038/ PMID:19528789; http://dx.doi.org/10.1097/QAD.
PMID:9521319; http://dx.doi.org/10.1038/32588 nm806 0b013e32832d9b00
147. Bottomly K. T cells and dendritic cells get intimate. 160. Lapenta C, Santini SM, Logozzi M, Spada M, Andreotti 169. Routy JP, Boulassel MR, Yassine-Diab B, Nicolette C,
Science 1999; 283:1124-5; PMID:10075571; http:// M, Di Pucchio T, et al. Potent immune response against Healey D, Jain R, et al. Immunologic activity and
dx.doi.org/10.1126/science.283.5405.1124 HIV-1 and protection from virus challenge in hu-PBL- safety of autologous HIV RNA-electroporated dendritic

Do not distribute.
148. Kovacsovics-Bankowski M, Rock KL. A phagosome- SCID mice immunized with inactivated virus-pulsed cells in HIV-1 infected patients receiving antiretroviral
to-cytosol pathway for exogenous antigens presented dendritic cells generated in the presence of IFN-alpha. J therapy. Clin Immunol 2010; 134:140-7; PMID:
on MHC class I molecules. Science 1995; 267:243-6; Exp Med 2003; 198:361-7; PMID:12874266; http:// 19889582; http://dx.doi.org/10.1016/j.clim.2009.09.
PMID:7809629; http://dx.doi.org/10.1126/science. dx.doi.org/10.1084/jem.20021924 009
7809629 161. Yoshida A, Tanaka R, Murakami T, Takahashi Y, 170. Garca F, Climent N, Assoumou L, Gil C, Gonzlez
149. Rock KL. A new foreign policy: MHC class I mole- Koyanagi Y, Nakamura M, et al. Induction of N, Alcam J, et al. DCV2/MANON07- AIDS Vaccine
cules monitor the outside world. Immunol Today protective immune responses against R5 human Research Objective Study Group. A therapeutic
1996; 17:131-7; PMID:8820271; http://dx.doi.org/ immunodeficiency virus type 1 (HIV-1) infection in dendritic cell-based vaccine for HIV-1 infection. J
10.1016/0167-5699(96)80605-0 hu-PBL-SCID mice by intrasplenic immunization Infect Dis 2011; 203:473-8; PMID:21233310; http://
150. Rock KL, Clark K. Analysis of the role of MHC class with HIV-1-pulsed dendritic cells: possible involve- dx.doi.org/10.1093/infdis/jiq077
II presentation in the stimulation of cytotoxic T ment of a novel factor of human CD4(+) T-cell origin. 171. Routy JP, Angel J, Vezina S, Tremblay C, Loutfy M,
lymphocytes by antigens targeted into the exogenous J Virol 2003; 77:8719-28; PMID:12885891; http:// Gill J, et al. Final Analysis of a Phase 2 Study of an
antigen-MHC class I presentation pathway. J dx.doi.org/10.1128/JVI.77.16.8719-8728.2003 Autologous DC Immunotherapy (AGS-004) Showed
Immunol 1996; 156:3721-6; PMID:8621907 162. Kundu SK, Engleman E, Benike C, Shapero MH, Positive Outcomes in Primary Endpoint of Viral Load
151. Peters JH, Gieseler R, Thiele B, Steinbach F. Dendritic Dupuis M, van Schooten WC, et al. A pilot clinical Control, and Favorable Safety and Immunogenicity
cells: from ontogenetic orphans to myelomonocytic trial of HIV antigen-pulsed allogeneic and autologous Profile, in Subjects Undergoing Structured Treatment
descendants. Immunol Today 1996; 17:273-8; PMID: dendritic cell therapy in HIV-infected patients. AIDS Interruption of ART (Abstract 385) [Abstract]. 18th
8962630; http://dx.doi.org/10.1016/0167-5699(96) Res Hum Retroviruses 1998; 14:551-60; PMID: Conference on Retroviruses and Opportunistic
80544-5 9591709; http://dx.doi.org/10.1089/aid.1998.14.551 Infections Boston February 27-March 2 2011.
152. Fan Z, Huang XL, Zheng L, Wilson C, Borowski L, 163. Lu W, Arraes LC, Ferreira WT, Andrieu JM. 172. Garca F, Routy JP. Challenges in dendritic cells-based
Liebmann J, et al. Cultured blood dendritic cells retain Therapeutic dendritic-cell vaccine for chronic HIV-1 therapeutic vaccination in HIV-1 infection Workshop
HIV-1 antigen-presenting capacity for memory CTL infection. Nat Med 2004; 10:1359-65; PMID: in dendritic cell-based vaccine clinical trials in HIV-1.
during progressive HIV-1 infection. J Immunol 1997; 15568033; http://dx.doi.org/10.1038/nm1147 Vaccine 2011; 29:6454-63; PMID:21791232
159:4973-82; PMID:9366424

www.landesbioscience.com Human Vaccines & Immunotherapeutics 581

Вам также может понравиться