Skip to main content
Journal of the International AIDS Society logoLink to Journal of the International AIDS Society
. 2015 Nov 9;18(1):20497. doi: 10.7448/IAS.18.1.20497

Evaluating the efficacy of therapeutic HIV vaccines through analytical treatment interruptions

Gina M Graziani 1,*, Jonathan B Angel 1,2,§,*
PMCID: PMC4641978  PMID: 26561337

Abstract

Introduction

The development of an effective therapeutic HIV vaccine that induces immunologic control of viral replication, thereby eliminating or reducing the need for antiretroviral therapy (ART), would be of great value. Besides the obvious challenges of developing a therapeutic vaccine that would generate effective, sustained anti-HIV immunity in infected individuals is the issue of how to best assess the efficacy of vaccine candidates.

Discussion

This review discusses the various outcome measures assessed in therapeutic HIV vaccine clinical trials involving individuals receiving suppressive ART, with a particular focus on the role of analytical treatment interruption (ATI) as a way to assess the virologic control induced by an immunotherapy. This strategy is critical given that there are otherwise no readily available measures to determine the ability of a vaccine-induced immune response to effectively control HIV replication. The various outcome measures that have been used to assess vaccine efficacy in published therapeutic HIV vaccine clinical trials will also be discussed. Outcome measures have included the kinetics of viral rebound, the new viral set point and changes in the size of the viral reservoir. Clinically relevant outcomes such as the CD4 decline, the time to resume therapy or the time to meet the criterion to resume therapy, the proportion of participants who resume therapy and/or the development of clinical symptoms such as acute retroviral syndrome are also measures of vaccine efficacy.

Conclusions

Given the lack of consistency between therapeutic HIV vaccine trials in how efficacy is assessed, comparing vaccines has been difficult. It would, therefore, be beneficial to determine the most clinically relevant measure for use in future studies. Other recommendations for future clinical trials also include studying compartments in addition to blood and replacing ATIs with single-copy assays in situations in which the use of an ATI is not ideal.

Keywords: HIV, AIDS, analytical treatment interruption(s), clinical trials, outcome measure, therapeutic vaccine, vaccine efficacy, viral reservoir

Introduction

The idea that HIV-positive individuals might benefit from therapeutic immunization was first proposed by Jonas Salk in 1987 [1]. The discovery since then of long-term non-progressors and elite controllers whose immune systems naturally control HIV infection without the need for antiretroviral therapy (ART) provides evidence for effective host-mediated anti-HIV immunity, thus providing a rationale for the development of therapeutic vaccines (reviewed in Refs. [24]).

The development of an HIV therapeutic vaccine capable of inducing control of HIV replication such that ART could be eliminated is a major focus of HIV research [57]. While ART has transformed HIV infection into a chronic, manageable disease for most individuals who have access to it [8,9], ART is associated with a number of disadvantages and limitations. In addition to being a lifelong therapy [7,8,10], ART can be toxic [8,9], is potentially associated with the development of HIV drug resistance [9] and does not eliminate latent HIV in viral reservoirs [69]. Finally, the high cost of ART makes it unavailable to the majority of the world's HIV-positive individuals who live in resource-limited countries [811]. A therapeutic vaccine would, therefore, circumvent many of the limitations associated with ART.

Besides the obvious challenges of developing a therapeutic vaccine that would induce effective, sustained anti-HIV immunity in infected individuals is the issue of how to best assess the efficacy of vaccine candidates [12]. In many clinical trials of therapeutic HIV vaccines (Tables 1A1E), assessing efficacy involves comparing various outcome measures before and after an analytical treatment interruption (ATI), which is used to assess vaccine-induced, immune-mediated viral control [2,5]. While therapeutic HIV vaccine clinical trials typically include the CD4 count as a safety/clinical event, virologic outcome measures vary from trial to trial, making it challenging to compare the results of different vaccine studies.

Table 1A.

Summary of the outcome measures of efficacy assessed in therapeutic HIV vaccine clinical trials with analytical treatment interruptions: protein or peptide subunit vaccines

Vaccine Study design Primary outcome measure(s) Other relevant efficacy outcome measures Main findings References
Vacc-4x (a mixture of four p24-like peptides) Open, prospective RCT comparing low vaccine dose vs. high vaccine dose (no ATI during this phase of study)
  • Safety

  • CD4 T cell count

  • CD8 T cell count

The higher dose of the vaccine induced stronger HIV-specific DTH and CD4 and CD8 T cell responses than the lower dose. [136]
Observation period of 26 weeks following immunization period in Ref. [136] that included two ATIs, one of four weeks’ duration and one of 14 weeks
  • Viral load ratio (end of study viral load/pre-ART viral load set point)

  • Immunogenicity

  • CD4 T cell count

  • CD8 T cell count

Participants with the highest DTH responses before ATI had lower VL by the end of the study compared to participants with low DTH responses. [137]
Long-term observation (1.5 years) after immunization in Ref. [136]
  • Percentage of participants who resumed ART

  • Immunogenicity

  • CD4 T cell count

  • pVL

Participants with the greatest DTH responses following immunization were less likely to require ART resumption compared to low responders. [138]
Observation period four years after enrolling in Ref. [136]
  • Time until ART resumption

  • Immunogenicity

  • CD4 T cell count

  • CD8 T cell count

  • pVL

  • Percentage of participants who resumed ART

Participants with the greatest DTH responses following immunization resumed ART later than low responders. [139]
RCT
  • Percentage of participants who met the criteria to resume ART

  • Percent change in CD4 T cell count between the start of the ATI and the last CD4 T cell count before ART was resumed or the end of the study if ART was not resumed

  • Time to restart ART

  • CD4 T cell count

  • CD8 T cell count

  • Pre-ART viral load set point (when available)*

  • Viral load set point during ATI* *substudy

The vaccine had no effect on the proportion of participants who resumed ART or on changes in the CD4 T cell count during the ATI. However, vaccinated participants had significantly reduced viral load set points during ATI compared to controls. [111]
TUTI-16 (synthetic HIV-1 Tat epitope) RCT
  • Safety

  • Prevention of viral rebound following ATI

  • CD4 T cell count

The vaccine did not prevent viral rebound following ATI. [140]
LFn-p24C (subtype C HIV Gag protein p24 fused to a detoxified anthrax-derived polypeptide) Open label, single-arm study; phase 1A: three immunizations; phase 1B: booster + ATI
  • Safety

  • CD4 T cell count

  • Percentage of participants who did not experience viral rebound

Immunized participants had significantly higher CD4 T cell counts compared to historical controls 12 months after enrolment in phase 1A and 30% of participants did not experience any viral rebound following ATI in phase 1B. [141]

Table 1E.

Summary of the outcome measures of efficacy assessed in therapeutic HIV vaccine clinical trials with analytical treatment interruptions: autologous dendritic cell vaccines

Vaccine Study design Primary outcome measure(s) Other relevant efficacy outcome measures Main findings References
Autologous monocyte-derived dendritic cells loaded with heat-inactivated autologous HIV-1 RCT
  • Safety

  • Percentage of participants with a set point pVL decrease of 0.5 log10 HIV-1 RNA copies/ml after the second ATI

  • Dynamics of pVL rebound after the second ATI

  • compared to the first ATI

  • CD4 T cell count

  • CD8 T cell count

  • CD4/CD8 ratio

Vaccination resulted in transient, partial virologic control. [160]
RCT
  • Safety

  • Change in pVL set point during ATI (Week 24)

  • Percentage of participants with a decrease in pVL ≥ 1 log10 at Week 24

  • pVL set point changes at Weeks 12, 36 and 48 (during ATI)

  • Percentage of participants with a decrease in pVL≥1 log10 at Weeks 12, 36 and 48 (during ATI)

  • Percentage of participants with a decrease in pVL≥0.5 log10 at Weeks 12, 24, 36 and 48 (during ATI)

  • Percentage of participants who restarted ART

  • CD4 T cell count

Vaccination resulted in a significant but transient reduction in viral load during ATI, which was associated with increased HIV-1-specific T cell responses. [110]
Viral reservoir substudy of Ref. [110]
  • Total and integrated HIV-1 DNA in CD4 T cells

  • Immunogenicity

  • CD4 T cell count

  • CD8 T cell count

Vaccination had no effect on the size of the viral reservoir during the vaccination period, although vaccine-induced T cell responses transiently delayed the replenishment of the viral reservoir after ATI. [112]
Autologous monocyte-derived DCs loaded with seven HIV-1-derived CTL epitope peptides Single-arm study
  • Safety

  • Immunogenicity

  • CD4 T cell count

  • Serum HIV-1 RNA

  • Viral load rebound after ATI

  • Viral load set point during ATI

Vaccination was safe and immunogenic in some participants but did not reduce the viral load set point during ATI. [161]
ANRS HIV-LIPO-5 (autologous monocyte-derived DCs loaded with five HIV-1-antigen peptides [Gag(17–35), Gag(253–284), Nef(66–97), Nef(116–145) and Pol(325–355)], each covalently linked to a palmitoyl-lysylamide moiety) Single-arm study
  • Safety

  • ART resumption

  • CD4 T cell count

  • Serious non-AIDS events

  • AIDS-defining events

  • Maximum viral load during ATI

The vaccine was safe and induced HIV- specific CD4 T cell responses that were associated with a trend toward reduced maximum viral load during ATI. [162]
Autologous monocyte-derived DCs loaded with ALVAC-HIV vCP1452 RCT
  • Safety

  • Immunogenicity

  • Viral load set point during ATI

  • CD4 T cell count

  • Percentage of CD4 T cells

The mean viral load set point during ATI did not differ between the two vaccine groups (another vaccine group received ALVAC-HIV vCP1452 independently of DC). [152]
Autologous monocyte-derived DCs loaded with mRNA encoding HIV-1 Tat, Rev and Nef Single-arm study
  • Safety

  • CD4 T cell count

  • CD8 T cell count

  • Kinetics of viral rebound during ATI

  • Duration off ART

The vaccine was safe and immunogenic. Although 6/17 participants remained off ART 96 weeks post-ATI, there was no correlation between HIV-specific immune responses and time off ART. [163]

RCT: randomized controlled trial; ATI: analytical treatment interruption; VL: viral load; ART: antiretroviral therapy; pVL: plasma viral load; IFN-γ: IFN-gamma; ddI: didanosine; GM-CSF: granulocyte-macrophage colony-stimulating factor; Nef: HIV negative regulatory factor; Rev: HIV regulator of expression of virion proteins; Tat: HIV transactivator of transcription; Gag: HIV group antigens; Pol: HIV precursor of reverse transcriptase, protease and integrase; Env: HIV envelope; RT: HIV reverse transcriptase; DCs: dendritic cells; IL-2: interleukin-2; rFPV: recombinant fowlpox virus; IL-10: interleukin-10; CTL: cytotoxic T lymphocytes; PBMC: peripheral blood mononuclear cells; SCA: single-copy assay.

Table 1B.

Summary of the outcome measures of efficacy assessed in therapeutic HIV vaccine clinical trials with analytical treatment interruptions: inactivated HIV vaccines

Vaccine Study design Primary outcome measure(s) Other relevant efficacy outcome measures Main findings References
Remune® (inactivated HIV-1 particles) Open label, non-randomized, two-arm study (immunized vs. unimmunized)
  • Immunogenicity

  • pVL

  • CD4 T cell count

Immunization-induced HIV-specific immune responses that correlated with CD4 T cell counts and with viral control during ATI. [142]
Remune®+ART intensification with ddI, hydroxyurea and GM-CSF Proof-of-concept, single-arm study
  • Immunogenicity

  • Viral load decrease: the difference between the viral load plateaus of the first two ATIs

  • CD4 T cell count

  • Percentage of CD4 T cells

Following ART intensification+Remune®, HIV-specific IFN-γ secretion increased between the first two of three ATIs, while viral load decreased significantly, although there was no correlation between these two observations. [143]

Table 1C.

Summary of the outcome measures of efficacy assessed in therapeutic HIV vaccine clinical trials with analytical treatment interruptions: DNA vaccines

Vaccine Study design Primary outcome measure(s) Other relevant efficacy outcome measures Main findings References
DNA vaccine encoding the HIV-1 Nef, Rev and Tat proteins RCT Changes in immune responses of previously immunized HIV-positive participants following ART initiation.
  • CD4 T cell count

  • CD8 T cell count

  • pVL

In a substudy in which participants had undergone ATI, there was no significant change in HIV-specific responses during or after ATI. [144]
VRC-HIV DNA 009-00-VP (a four-plasmid mixture encoding modified envelope constructs from HIV-1 subtypes A, B and C and a subtype B Gag-Pol-Nef fusion protein) RCT; participants initiated ART during early HIV infection
  • Safety

  • Immunogenicity

  • CD4 T cell count

  • Viral load set point following ATI

The vaccine was safe but not immunogenic and had no effect on the viral set point during ATI. [104]
DNA vaccine consisting of seven plasmids encoding HIV-1 Gag (subtypes A and B), Env (subtypes A, B or C), RT or Rev RCT
  • HIV-specific epitope reactivity

  • Immunogenicity

  • Time to ART resumption

  • CD4 T cell count

  • Viral load rebound following ATI

  • Viral load set point following ATI

Although immunogenic, the vaccine did not affect the viral set point during ATI or the time to resume ART. [145]

Table 1D.

Summary of the outcome measures of efficacy assessed in therapeutic HIV vaccine clinical trials with analytical treatment interruptions: viral vectors

Vaccine Study design Primary outcome measure(s) Other relevant efficacy outcome measures Main findings References
MVA-Nef (modified vaccinia Ankara virus encoding the HIV-1 LAI Nef gene) Single-arm study
  • Safety

  • Immunogenicity

  • Time to viral rebound following ATI

  • Time to peak viremia following ATI

  • Peak viremia following ATI

  • Number of participants who resumed ART

The vaccine was safe and immunogenic but did not prevent viral rebound during ATI. However, in the majority of participants, viral load during ATI and CD4 T cell counts were improved compared to pre-ART levels. [146]
MVA.HIVA (modified vaccinia Ankara virus encoding clade A HIV-1 Gag p24/p17 and a multi-CTL epitope) Extension of a single-arm study by Dorrell et al. [147]; in this extension study, participants were boosted then underwent an ATI
  • IL-10 production

  • Immunogenicity

  • Criteria for ART resumption (pVL and CD4 T cell count)

Vaccination did not increase IL-10 levels. However, IL-10 levels did increase during ATI and were correlated with pVL. [148]
MVA-B (modified vaccinia Ankara virus encoding monomeric gp120 and the clade B fused Gag-Pol-Nef polyprotein)±disulfiram RCT
  • Safety and immunogenicity

  • Kinetics of viral load rebound following ATI

  • Time and criteria to resume ART

  • Cell-associated HIV-1 RNA

  • HIV-1 proviral DNA levels

The vaccine was safe and immunogenic but did not significantly affect viral load rebound after ATI or the size of the viral reservoir, whether given alone or with disulfiram. [149]
ALVAC-HIV vCP1452 (a recombinant canarypox virus encoding HIV-1 Env, Gag and protease and part of the Nef and RT proteins) RCT
  • pVL at the end of the ATI

  • CD4 T cell count

  • Percentage of CD4 T cells

  • Kinetics of viral load rebound

  • Viral load set point following ATI

ATI, but not vaccination, contributed to enhanced viral control. [150]
RCT
  • Immunogenicity

  • Time to resume ART (viral rebound >50 000 copies/ml following ATI)

  • CD4 T cell count

  • HIV-1 DNA in PBMCs

Although immunogenic, the vaccine-induced immune responses were associated with reduced time to resume ART and greater viral rebound. [151]
RCT
  • Safety

  • Immunogenicity

  • Viral load set point during ATI

  • CD4 T cell count

  • Percentage of CD4 T cells

The mean viral load set point during ATI did not differ between the two vaccine groups (second vaccine group received autologous DC loaded with ALVAC vCP1452). [152]
ALVAC vCP1452 + rgp160 Two-arm study (vaccinated participants from a previous study vs. unvaccinated participants); participants initiated ART during early HIV infection
  • Time to viral rebound after ATI

  • Initial rate of viral rebound after ATI

  • Peak viremia during ATI

  • CD4 T cell count

ATI was followed by viral rebound in all subjects and was not affected by vaccination. [105]
ALVAC-HIV vCP1452±IL-2 RCT
  • pVL at Weeks 11 and 12 post-ATI

  • Viral load set-point during ATI

  • Peak viral load during ATI

  • CD4 T cell count

  • CD8 T cell count

  • Disease progression, opportunistic infections or acute retroviral syndrome after ATI

Immunization with ALVAC resulted in a statistically significant reduction in viral rebound following ATI. The addition of IL-2 to ALVAC increased CD4 T cell counts but did not further reduce viral rebound. [153]
ALVAC-HIV vCP1452±Remune® RCT; participants initiated ART during acute HIV infection
  • Percentage of participants with pVL≤1000 HIV-1 RNA copies/ml at 24 weeks post-ATI

  • CD4 T cell count

  • CD8 T cell count

  • Cell-associated HIV-1 DNA and RNA

  • Viral load set point during ATI

  • Percentage of participants with pVL ≤400 HIV-1 RNA copies/ml during entire ATI period

  • Time to reach pVL > 1000 HIV-1 RNA copies/ml after ATI

Although immunogenic, the vaccines did not induce virologic control during ATI. [106]
RCT
  • Time to viral rebound >50 HIV-1 RNA copies/ml

  • Safety

  • CD4 T cell count

  • Viral load 12 weeks after ATI

  • Viral load set point following ATI

  • Time to ART resumption

  • Time to meet criteria to resume ART

ALVAC±Remune® was associated with an increased time to meet the predefined criteria to restart ART and tended to delay viral rebound, but did not reduce the viral set point during ATI. [120]
Viral reservoir substudy of Ref. [120]
  • Size of the viral reservoir

  • CD4 T cell count

ALVAC±Remune® did not affect the size of the viral reservoir. [154]
ALVAC-HIV vCP1433 (a recombinant canarypox virus encoding part of HIV-1 Env, Gag, protease and multiple immunodominant Nef and Pol CTL epitopes) Single-arm study
  • Percentage of participants who remained off ART 44 weeks after the initiation of the ATI among those having at least one HIV-specific T cell response during the vaccination period

  • CD4 T cell count

  • Percentage of participants who resumed ART (pVL > 50,000 copies/ml within eight weeks of ATI or two consecutive measurements > 10,000 copies/ml any time after eight weeks of ATI)

  • CD4 and/or CD8 HIV-specific immune responses

11% of the participants with at least one HIV-specific T cell response during vaccination remained off ART 44 weeks after the initiation of ATI. [155]
ALVAC-HIV vCP1433 + HIV Lipo6-Tfollowed by three cycles of IL-2. The Lipo-6T vaccine is a mixture of the tetanus toxoid TT-830–843 class II restricted universal CD4 T cell epitope and five HIV-1LAI peptides: Gag 17–35, Gag 235–284, Nef 66–97, Nef 116–145 and Pol 325–355. RCT
  • Percentage of participants who responded to both HIV p24 and at least one of 11 HIV peptides

  • CD4 T cell count

  • HIV-1 DNA in PBMC

  • HIV-specific CD8 T cell responses (IFN-γ production)

  • Percentage of participants experiencing virologic success following ATI

  • Viral load set point during ATI

  • Time to virologic failure

The vaccines induced both HIV-specific CD4 and CD8 T cell responses. Vaccine-induced immune responses predicted virologic control during ATI. [109]
RCT; participants initiated ART during acute HIV infection
  • Percentage of participants with a CD4 T cell response to HIV p24 or to one HIV peptide at Week 36

  • HIV-specific CD8 T cell responses

  • Percentage of participants with virologic success at study end

  • Time without ART

  • HIV-1 DNA in PBMC

  • CD4 T cell count

Vaccination did not induce CD4 T cell immune responses, had a transient impact on CD8 T cell IFN-γ responses and had no effect on viral rebound during ATI. [107]
rFPV vaccines (recombinant fowlpox virus that encodes HIV Gag/Pol±human IFN-γ) Extension study of an RCT by Emery et al. [156] in which participants who had initiated ART during acute HIV infection received placebo or rFPV±human IFN-γ; in this extension study, participants received a booster and then underwent ATI one week later.
  • Time-weighted mean area-under-the-curve change from baseline log pVL until ART resumption

  • Kinetics and rate of pVL rebound

  • Median time to ART resumption

Immunization with rFPV Gag/Pol + IFN-γ, but not with rFPV Gag/Pol or placebo, was associated with a trend toward reduced plasma viral load following ATI. [108]
Replication-defective adenovirus 5 HIV-1 Gag RCT
  • Time averaged area-under-the curve analysis of pVL during ATI

  • Viral load set point after ATI

  • CD4 T cell count

The vaccine did not significantly affect viral rebound kinetics during ATI. [157]
Follow-up study of Ref. [157]
  • pVL set point (mean of the ATI weeks 12 and 16 pVL)

  • Immunogenicity

The majority of the initial viral suppressors had been vaccinated; this suppression was transient. [158]
Retrospective analysis of Ref. [157]
  • Cell-associated HIV-1 DNA and RNA

  • Residual viremia (SCA)

  • Immunogenicity

Vaccination had a modest, transient impact on residual viremia. [159]

Discussion

The current state of non-HIV therapeutic vaccines

Only a few therapeutic vaccines are currently licensed worldwide and most of them are used to treat cancer [13]. In 2010, the US Food and Drug Administration (FDA) approved sipuleucel-T (Provenge®) to treat hormone-refractory prostate cancer [14]. A therapeutic vaccine for ovarian cancer has been approved in Dubai [13], while another one was recently given fast track designation by the FDA [15]. Two different therapeutic vaccines for renal cell carcinoma have been approved, one each in Russia and South Korea [13]. In the meantime, phase III clinical trials have been or are being conducted to assess the efficacy of candidate therapeutic vaccines against a variety of malignancies including cancers of the breast [16,17], pancreas [1820], liver [21], lung [2227], kidneys [28], skin [2932], prostate [33], stomach or oesophagus [34] and brain [3538].

Zostavax® is a therapeutic vaccine that reduces the frequency and severity of shingles, which is caused by the reactivation of the varicella zoster virus that causes chickenpox [39]. Zostavax is the first example of a vaccine with clinical efficacy against an established infection [40]. The success of this vaccine provides hope that it might be possible to induce clinically beneficial immunity against other viruses that establish chronic infections.

The development of therapeutic vaccines to treat other chronic infections in humans is an area of active research. The various pathogens, other than HIV, against which therapeutic vaccines are currently being or have been assessed in various clinical trials include cytomegalovirus [4143], hepatitis B [4461] and hepatitis C [6276] viruses, human papillomavirus [7790], herpes simplex virus 2 [9193], Mycobacterium tuberculosis [94,95], Trypanosoma cruzi [96] and Leishmania [97]. Encouraging results have been obtained in some of these trials [43,47,48,59,6366,68,76,7982,84,86].

The current state of therapeutic HIV vaccines assessed in clinical trials

The features that might make a therapeutic vaccine effective and the inherent challenges of making such a vaccine have been recently described in a number of excellent reviews [3,6,98102]. Minimally, a therapeutic vaccine should improve the benefits of existing ART regimens, simplify these regimens or allow for periodic ART interruption [6,98100]. Ideally, a therapeutic vaccine would eliminate the need for ART, either by eradicating virus (a sterilizing cure) or by inducing an immune response capable of controlling virus replication (a functional cure) [6,98100,102].

Therapeutic vaccination would be of particular value for HIV-positive individuals residing in resource-limited countries in which access to ART is limited [98]. In these settings, the HIV epidemic is fuelled by the higher rate of new infections relative to the rate at which newly infected individuals receive ART [98,99]. An effective therapeutic vaccination could, therefore, help control the epidemic. Therapeutic vaccines would also be invaluable to those who struggle with daily, lifelong ART compliance [98].

Over the course of more than two decades, more than four dozen therapeutic HIV vaccine candidates have been evaluated in clinical trials for safety, immunogenicity and, in some cases, for efficacy. The results of these trials have shown limited success (reviewed in Refs. [3,6,99102]) with respect to their ability to control HIV replication or maintain CD4 T cell counts in the absence of ART [99,102,103]. While the majority of these trials involved therapeutic vaccination of individuals who initiated ART during chronic HIV infection, vaccination of individuals who initiated ART during acute or early infection was also ineffective [104108]. In one of these studies, the dynamics of viral rebound following vaccination and ATI were similar to those observed in studies of chronically infected individuals who discontinued ART [105].

A few randomized, controlled clinical trials by Levy et al. [109], Garcia et al. [110] and Pollard et al. [111] have, however, produced somewhat encouraging results.

In a trial assessing the effects of receiving two vaccines, ALVAC-HIV vCP1433 and Lipo6-T, followed by IL-2 administration, Levy et al. [109] observed that a significantly greater proportion of vaccinated HIV-positive participants had a lower viral set point 12 weeks after stopping ART, compared to unvaccinated controls. The times to viral rebound and to resume therapy were also significantly delayed in the vaccinated participants.

Garcia et al. [110] observed that therapeutic vaccination of HIV-positive participants with an autologous dendritic cell vaccine loaded with autologous, inactivated HIV-1 resulted in a decrease in the viral load set point following ATI. Unfortunately, the decrease in the viral load induced by vaccination was transient. Furthermore, vaccination did not prevent the CD4 T cell count decline after interruption of ART. It was subsequently reported that, although no change was observed in the size of the viral reservoir during the vaccination period, vaccine-induced T cell responses transiently delayed the replenishment of the viral reservoir after ATI [112].

Pollard et al. [111] observed that the Vacc-4x vaccine, which contains a mixture of conserved Gag peptide domains, was able to significantly reduce the viral load following ATI, resulting in a new viral load set point. However, vaccination did not affect the change in the CD4 T cell count following ATI, nor did it affect the proportion of participants who resumed therapy or the time until therapy was resumed.

The role of ATIs in assessing the efficacy of therapeutic HIV vaccines

ART may be interrupted as part of a structured treatment interruption (STI) or as part of an ATI. The main goals of the STI are to reduce ART-associated burden (reviewed in Refs. [4,113115]) and/or to induce HIV “autoimmunization” (reviewed in Refs. [3,4,114,116]), whereas the purpose of the ATI is to assess the efficacy of an experimental therapeutic candidate [12]. STIs and ATIs are discussed in further detail below.

STIs have been used in the past to address the ART-associated issues of toxicity, cost and resistance (reviewed in Refs. [4,113115]). Another goal of the STI was to allow for viral rebound, resulting in “autoimmunization” with increased exposure to HIV antigens (reviewed in Refs. [3,4,114,116]). It was hypothesized that the resulting viremia would boost the anti-HIV immune response sufficiently to induce viral control, thus avoiding ART resumption. Unfortunately, the various clinical trials that assessed the immunological benefits of STIs failed to show any benefits (reviewed in Refs. [4,114]), while the SMART study showed that treatment interruptions can increase morbidity and mortality [117].

When ART is interrupted, plasma HIV RNA levels typically first become detectable within days or weeks [118120], reach a peak and then decrease to a steady state level, or viral set point [121]. Exceptions to the occurrence of viral rebound following therapy interruption do occur and may be more frequent in those who are treated during acute primary infection [122], although the exact immune mechanisms responsible for this degree of viral control are currently unknown.

The ATI is an intentional interruption of ART that is included in controlled clinical trials of therapeutic vaccines (reviewed in Refs. [2,4,5,115,123]). The ATI is a frequently used strategy for assessing HIV therapeutic vaccine efficacy [12]; it is considered by some to be the “gold standard” [5]. This strategy, which is used to assess the virologic control induced by an immunotherapy given while the individual is still taking suppressive ART [2,115,123] is necessary because there are currently no laboratory assays that measure the ability of the immune system to effectively control HIV replication [2,99,123]. In addition to assessing the kinetics of viral rebound [2,99], ATIs also allow for the assessment of a potentially new viral set point as well as CD4 T cell dynamics following treatment interruption [115,123].

The SMART study revealed that HIV-positive participants who interrupted ART experienced an increased risk of developing AIDS and non-AIDS events compared to participants who continued therapy [117]. However, it also showed that individuals having high CD4 counts (>500 cells/µl), high CD4 nadir (>200 cells/µl) and undetectable virus levels (<50 copies/ml) can safely undergo treatment interruptions in carefully monitored clinical trials without increasing their risk of death and non-AIDS events or developing viral resistance [115,124126]. It was recently shown that chronically infected individuals having undetectable virus levels and preserved CD4 counts, including those with low CD4 nadir, can also safely undergo treatment interruptions if the interruptions are short, that is if treatment is reinitiated upon detection of viral rebound [127].

Despite the safety of ATIs, the increased viral load that occurs following treatment interruption can occasionally be associated with the development of an acute retroviral syndrome [128,129] or thrombocytopenia [130], as well as with an increased risk of HIV transmission by individuals involved in high-risk activities [131].

Alternatives to ATIs

In studies that include an ATI, therapy is typically reintroduced either at the end of a fixed period of treatment interruption (e.g. 16 weeks), during which time a new viral set point is usually achieved, or when specific virologic, immunologic or clinical outcomes are met. A new, alternative treatment interruption strategy in clinical trials of HIV immunotherapies is the monitored antiretroviral pause (MAP), which reintroduces ART as soon as viral rebound occurs [123]. The advantage of the MAP is that, by reintroducing ART as soon as viral rebound occurs, the risk is reduced compared to the risk associated with an ATI. However, since the MAP does not allow a new viral set point to be established, this strategy cannot be used to determine whether the immunotherapy being tested improved virologic control by the immune system. Thus, whether an ATI or a MAP should be used in a clinical trial of an HIV immunotherapy depends on the scientific question being asked, with the MAP lending itself to assess therapies designed to measure the time to viral rebound, which may be a surrogate measure of the size of the viral reservoir, while the ATI should be used to assess therapies designed to improve immune control of HIV. It should be noted, however, that it has not yet been established whether the time to viral rebound following ATI is, in fact, a surrogate measure of the size of the viral reservoir [132].

Recently, single-copy reverse transcriptase (RT)-qPCR assays with single-copy sensitivity (i.e. the single-copy assay (SCA) for HIV-1 RNA) were used to detect virus in the plasma of individuals who had undergone myeloablation and autologous stem cell transplantation for the treatment of lymphoma [133]. Since these patients had undetectable plasma viremia by standard methods, it was hypothesized that their lymphoma treatment had resulted in HIV eradication; the results of the SCA, however, proved otherwise. Therefore, in this setting, SCA was used to guide the decision regarding whether to interrupt ART; because virus was detected using this assay, ART was not interrupted and the viral rebound that would have otherwise occurred was avoided. However, had the SCA failed to detect virus, then an ATI would have been warranted. The use of these highly sensitive assays has been suggested as an additional approach to the assessment of therapeutic vaccine efficacy [5]. The inclusion of such assays into future clinical trials of HIV immunotherapeutics could expedite these trials if only subjects with undetectable viral load by SCA proceeded on to ATI. However, such an alternative approach would need to be validated first by concurrent analysis in clinical trials in which it can be determined that SCA results predict virologic rebound following ATI [5]. In the interim, or until some other strategy is validated, treatment interruptions, the current gold standard for assessing therapeutic vaccine efficacy [5], will continue to play a crucial role in the evaluation of HIV therapeutic vaccines and should only be replaced with some other strategy if treatment interruption must be avoided.

It was recently shown that the ex vivo antiviral capacity of CD8+ T cells [134] predicts the rate of CD4 T cell loss in early HIV infection and is inversely correlated with viral load set point [135]. It has been suggested, therefore, that this assay [134] be included as a read-out in clinical trials of therapeutic vaccines. However, whether this accurately measures vaccine-induced immunologic control of viral replication remains to be established [135].

Read-outs of therapeutic HIV vaccine studies that incorporate ATIs

More than four dozen therapeutic HIV vaccine candidates have been evaluated in clinical trials for safety, immunogenicity and, in some cases, for efficacy. Tables 1A1E summarizes the outcome measures of efficacy that have been assessed in published vaccine trials that include ATI.

Since the correlates of viral suppression/immunological response that should be used to assess the therapeutic benefits of vaccines are not well defined [7,99,135,164166], the surrogate measure(s) used as the primary end point(s) to assess the clinical benefits of therapeutic vaccine candidates vary from trial to trial.

The virologic outcome measures assessed following vaccination and ATI may include the time to detectable viremia, the peak level of viremia, the new viral set point, the time to reach the new viral set point, the time to reach a certain viral load threshold, the viral load at a predefined time following ATI and changes in the size of the viral reservoir.

Of these read-outs, it has been suggested that the new viral set point is the most relevant clinical assessment of the antiviral efficacy of a therapeutic vaccine (reviewed in Ref. [5]). Whereas the new viral set point, the peak level of viremia and the time to rebound are all affected by the strength of the host's anti-HIV immune response, the peak level of viremia and the time to rebound may also be affected by the number of susceptible target cells and the size of the viral reservoir, respectively. It has also been suggested that the new viral set point established after immunotherapy and ATI should be the primary end point of clinical trials for assessing the effectiveness of anti-HIV immunotherapies; a difference of at least 0.5 log copies/ml between the experimental and control arms of a study is probably clinically significant, as determined by the results obtained in studies of antiretrovirals (reviewed in Ref. [2]). One disadvantage of using the new viral set point as a primary end point, however, is that it may miss important virologic and immunologic events that occur early in the immune response to the vaccine [165]. Another disadvantage is that the establishment of a new viral set point may be delayed, thus extending the length of treatment interruption and its associated risks.

Another primary end point that is commonly used in HIV therapeutic vaccine clinical trials is the time to detectable virus or the time to viral rebound (i.e. the time to achieve a viral load >50 copies/ml) following an ATI. However, an accurate assessment of this requires frequent viral load monitoring [165]. While this outcome would seem to be clinically relevant, its value is unknown since this measure does not appear to correlate with other virologic outcome measures. In a therapeutic vaccine trial of ALVAC-HIV vCP1452 by Jacobson et al., the time until viral rebound did not correlate with any of the other virologic measures assessed, such as the new viral load set point [150]. As a result, it has been suggested that the time to viral rebound is probably not an appropriate outcome measure for assessing the effectiveness of HIV therapeutic vaccines [2]. Similarly, our own study of ALVAC-HIV vCP1452 with or without Remune® failed to find a correlation between the time to viral rebound and the new viral set point, nor with the magnitude of the viral rebound [120]. A correlation was observed, however, between the time to viral rebound and the time to restart therapy, as well as the time to meet the criteria to do so. In the one other trial in which vaccination was found to delay the time to viral rebound (this trial involved ALVAC-HIV vCP1433 and Lipo-6T), no assessments were made for correlations with other virologic outcome measures [109].

The CD4 count, which is routinely used to determine the risk of opportunistic infection [167], is typically included in trials of HIV therapeutic vaccines. In addition to monitoring the change in the absolute CD4 count, changes in the percentage of CD4 T cells, the CD4:CD8 ratio, the time to decline to a predefined level or the change in the slope of the CD4 count have also been assessed in clinical trials of HIV therapeutic vaccines. However, this is not an ideal primary outcome as it requires waiting for a decline in the CD4 count.

In addition to virologic and immunologic outcomes, some studies of therapeutic HIV vaccines include the assessment of clinical outcomes. These outcomes include the development of clinical events, including symptoms of acute retroviral syndrome after ATI [128,129] or the time until either ART is resumed or the criteria for ART resumption is met. In addition, the proportion of participants who do or do not resume ART may also be determined.

Therapeutic HIV vaccines and their potential role in an HIV cure strategy

One of the research priorities recently identified by the International AIDS Society (IAS) Global Scientific Strategy “Towards an HIV Cure” working group is the development of a therapeutic HIV vaccine capable of boosting the immune system of the infected host to control HIV replication in the absence of ART, thus producing a functional cure [7] similar to that experienced naturally by long-term non-progressors and elite controllers (reviewed in Refs. [7,99,166]). According to this IAS working group, a therapeutic vaccine should be directed to conserved HIV epitopes and either 1) elicit a humoral response consisting of neutralizing anti-HIV antibodies that would a) prevent cell-to-cell transmission or b) recognize virus-producing cells for destruction by antibody-dependent cellular cytotoxicity; or 2) induce a strong cytotoxic cellular response for the destruction of cells producing virus before virus progeny is released [7]. These strategies should lead to a sustained reduction in the number of cells actively transcribing virus and induce an immune selective pressure that would lead to loss of viral fitness and replicative potential.

The persistence of the viral reservoir is considered to be the major obstacle to curing HIV infection [3,6,7,10]. In fact, when ART is interrupted, the viral rebound that occurs within days or weeks is the result of reseeding from viral reservoirs [11]. Furthermore, high levels of HIV DNA, a surrogate marker of the size of the viral reservoir, are correlated with quicker viral rebound following ART interruption [126]. The few trials of therapeutic vaccines that have assessed the change in the size of the viral reservoir did not observe any significant effect [106,112,149,154,159,168172]. Five of these studies assessed whether vaccination induced any changes in the size of the viral reservoir by measuring proviral DNA, either by co-culture assay [154,168,171] or by PCR [149,169]. One of the advantages of assessing changes in the size of the viral reservoir as a read-out of therapeutic vaccine efficacy is that this outcome measure can be made in trials that do not include an ATI [168], thus minimizing the risks that may be associated with treatment interruptions. Disadvantages of using this outcome measure, however, include the fact that no single assay accurately measures the size of the viral reservoir [173,174] and the lack of strong correlations between assays [174].

Conclusions and future directions

The development of a therapeutic HIV vaccine would be a valuable alternative to the use of expensive, toxic, lifelong ART regimens. The results of dozens of clinical trials performed over more than two decades to assess the safety, immunogenicity and, in many cases, the efficacy of various HIV therapeutic vaccines have been published, and more trials are underway. Besides the obvious challenges of developing a successful therapeutic vaccine is the issue of how to best assess the efficacy of vaccine candidates [12]. Currently, the inconsistent assessment of different outcome measures in different trials makes it difficult to compare the relative efficacies of the various vaccine candidates.

In its “Towards an HIV Cure” recommendations, the IAS recommends that future clinical trials of therapeutic HIV include studying compartments in addition to peripheral blood, such as the gastrointestinal tract and lymph nodes, both sites of HIV infection and immune responses [7]. Other suggestions for future trials include measuring immune control of viral replication by using highly sensitive SCA in situations in which the use of ATI is not ideal.

Given that immune activation predicts HIV disease progression independently of the CD4 count and viral load, it has also been recommended that, when assessing the efficacy of an HIV immunotherapy such as therapeutic vaccination, concurrent changes in immune activation markers, vaccine-specific responses and viral replication should be assessed during treatment interruption [175].

It is apparent that despite significant efforts made, the therapeutic vaccine candidates studied to date have been associated with limited clinical benefit [3,6,99]. Continued efforts will be required, therefore, to develop and test a safe and effective therapeutic HIV vaccine that will help end the global HIV epidemic. Future work may be influenced by promising prophylactic simian immunodeficiency virus (SIV) vaccines. In one study, virus levels became undetectable following initial viremia in half of the macaques vaccinated prior to SIV challenge [176], while in another SIV/macaque study, one-third of the monkeys that became infected following SIV challenge ultimately became elite controllers [177]. Thus, despite being designed as preventative SIV vaccines, both appeared to induce therapeutic benefits. These simian vaccines may, therefore, provide some valuable insight into the design of effective therapeutic HIV vaccines.

Finally, while the characteristics of a successful therapeutic HIV vaccine are currently unknown, standardizing which outcome measures should be used in future clinical trials to evaluate vaccine efficacy would certainly be beneficial.

Competing interests

JBA has done contract research for Argos Therapeutics, Inc., Sanofi Pasteur Ltd. and Immune Response Corp. GMG has no competing interests to declare.

Authors' contributions

GMG and JBA contributed equally to the preparation of the manuscript.

Author information

JBA is the Co-leader of the Vaccines and Immunotherapies (VIT) Core of the CTN (CIHR Canadian HIV Trials Network). GMG is the VIT Core Research Associate.

References

  • 1.Salk J. Prospects for the control of AIDS by immunizing seropositive individuals. Nature. 1987;327(6122):473–6. doi: 10.1038/327473a0. [DOI] [PubMed] [Google Scholar]
  • 2.Kutzler MA, Jacobson JM. Treatment interruption as a tool to measure changes in immunologic response to HIV-1. Curr Opin HIV AIDS. 2008;3(2):131–5. doi: 10.1097/COH.0b013e3282f54cde. [DOI] [PubMed] [Google Scholar]
  • 3.Carcelain G, Autran B. Immune interventions in HIV infection. Immunol Rev. 2013;254(1):355–71. doi: 10.1111/imr.12083. [DOI] [PubMed] [Google Scholar]
  • 4.Levy Y. Preparation for antiretroviral interruption by boosting the immune system. Curr Opin HIV AIDS. 2008;3(2):118–23. doi: 10.1097/COH.0b013e3282f5122a. [DOI] [PubMed] [Google Scholar]
  • 5.Lederman MM, Penn-Nicholson A, Stone SF, Sieg SF, Rodriguez B. Monitoring clinical trials of therapeutic vaccines in HIV infection: role of treatment interruption. Curr Opin HIV AIDS. 2007;2(1):56–61. doi: 10.1097/COH.0b013e3280119264. [DOI] [PubMed] [Google Scholar]
  • 6.Vanham G, Van GE. Can immunotherapy be useful as a “functional cure” for infection with human immunodeficiency virus-1? Retrovirology. 2012;9:72. doi: 10.1186/1742-4690-9-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.The International AIDS Society Scientific Working Group on HIV Cure. Towards an HIV cure; full recommendations. 1st ed [Internet]. 2012 Jul. [cited 2014 Aug 5]. Available from: http://www.iasociety.org/Web/WebContent/File/HIV_Cure_Full_recommendations_July_2012.pdf.
  • 8.Deeks SG, Lewin SR, Havlir DV. The end of AIDS: HIV infection as a chronic disease. Lancet. 2013;382(9903):1525–33. doi: 10.1016/S0140-6736(13)61809-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vella S, Schwartlander B, Sow SP, Eholie SP, Murphy RL. The history of antiretroviral therapy and of its implementation in resource-limited areas of the world. AIDS. 2012;26(10):1231–41. doi: 10.1097/QAD.0b013e32835521a3. [DOI] [PubMed] [Google Scholar]
  • 10.Deeks SG, Autran B, Berkhout B, Benkirane M, Cairns S, Chomont N, et al. Towards an HIV cure: a global scientific strategy. Nat Rev Immunol. 2012;12(8):607–14. doi: 10.1038/nri3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Joos B, Fischer M, Kuster H, Pillai SK, Wong JK, Boni J, et al. HIV rebounds from latently infected cells, rather than from continuing low-level replication. Proc Natl Acad Sci U S A. 2008;105(43):16725–30. doi: 10.1073/pnas.0804192105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Harari A, Rozot V, Cavassini M, Enders FB, Vigano S, Tapia G, et al. NYVAC immunization induces polyfunctional HIV-specific T-cell responses in chronically-infected, ART-treated HIV patients. Eur J Immunol. 2012;42(11):3038–48. doi: 10.1002/eji.201242696. [DOI] [PubMed] [Google Scholar]
  • 13.Boukhebza H, Bellon N, Limacher JM, Inchauspe G. Therapeutic vaccination to treat chronic infectious diseases: current clinical developments using MVA-based vaccines. Hum Vaccin Immunother. 2012;8(12):1746–57. doi: 10.4161/hv.21689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gomella LG, Gelpi-Hammerschmidt F, Kundavram C. Practical guide to immunotherapy in castration resistant prostate cancer: the use of sipuleucel-T immunotherapy. Can J Urol. 2014;21(2 Suppl 1):48–56. [PubMed] [Google Scholar]
  • 15.Immunovaccine Inc. Press release: immunovaccine receives FDA fast track designation for DPX-survivac for treatment of ovarian cancer [Internet] [cited 2014 Dec 10]. Available from: http://www.imvaccine.com/releases.php?releases_id=353.
  • 16.Galena Biopharma, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. PRESENT: prevention of recurrence in early-stage, node-positive breast cancer with low to intermediate HER2 expressions with NeuVax™Treatment. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01479244. [Google Scholar]
  • 17.Oncothyreon Canada Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A multi-center phase III randomized, controlled study of theratope vaccine for metastatic breast cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00003638. [Google Scholar]
  • 18.NewLink Genetics Corporation. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase III study of chemotherapy and chemoradiotherapy with or without HyperAcute®-Pancreas (Algenpantucel-L) immunotherapy in subjects with surgically resected pancreatic cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01072981. [Google Scholar]
  • 19.NewLink Genetics Corporation. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase III study of chemotherapy with or without Algenpantucel-L (HyperAcute®-Pancreas) immunotherapy in subjects with borderline resectable or locally advanced unresectable pancreatic cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01836432. [Google Scholar]
  • 20.Royal Liverpool University Hospital. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A prospective, phase III, controlled, multicentre, randomised clinical trial comparing combination gemcitabine and capecitabine therapy with concurrent and sequential chemoimmunotherapy using a Telomerase Vaccine in Locally Advanced and Metastatic Pancreatic Cancer [TELOVAC] [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT00425360. [Google Scholar]
  • 21.Galyna Kutsyna. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase III randomized, placebo-controlled clinical trial of V5 versus placebo in patients with advanced Hepatocellular Carcinoma (HCC) [cited 2015 Apr 2]. Available from: http://clinicaltrials.gov/ct2/show/NCT02232490. [Google Scholar]
  • 22.Transgene. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase IIb multicentric controlled study evaluating the therapeutic vaccine TG4010(MVA-MUC1-IL2) as an adjunct to standard chemotherapy in advanced non small cell lung cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00415818. [Google Scholar]
  • 23.Recombio SL. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A prospective, randomized, multicenter, open label phase III study of active specific immunotherapy with racotumomab plus best support treatment versus best support treatment in patients with advanced non-small cell lung cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01460472. [Google Scholar]
  • 24.Merck KGaA. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A multi-national, double-blind, placebo-controlled, randomized, phase III clinical trial of the cancer vaccine Stimuvax® (L-BLP25 or BLP25 Liposome Vaccine) in Asian subjects with stage III, unresectable, Non-small Cell Lung Cancer (NSCLC) who have demonstrated either stable disease or objective response following primary chemo-radiotherapy. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01015443. [Google Scholar]
  • 25.EMD Serono. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A multi-center phase III randomized, double-blind placebo-controlled study of the cancer vaccine Stimuvax® (L-BLP25 or BLP25 Liposome Vaccine) in Non-small Cell Lung Cancer (NSCLC) subjects with unresectable stage III disease. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00409188. [Google Scholar]
  • 26.Bioven Europe. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase 3 open-label, multicentre, randomised trial to establish safety & efficacy of an EGF cancer vaccine in inoperable, stage IV biomarker positive, wild type EGF-R NSCLC patients eligible to receive standard treatment and supportive care. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT02187367. [Google Scholar]
  • 27.NewLink Genetics Corporation. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. An open-label, randomized phase IIB/III active control study of second-line hyper-acute(R)-lung (Tergenpumatucel-L) immunotherapy versus docetaxel in progressive or relapsed non-small cell lung cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01774578. [Google Scholar]
  • 28.Immatics Biotechnologies GmbH. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A randomized, controlled phase III study investigating IMA901 multipeptide cancer vaccine in patients receiving sunitinib as first-line therapy for advanced/metastatic renal cell carcinoma. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01265901. [Google Scholar]
  • 29.Laboratorio Pablo Cassará SRL. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Randomized, comparative phase II/III study between treatment with CSF470 vaccine (allogeneic, irradiated) Plus BCG and MOLGRAMOSTIN (rhGM-CSF) as adjuvants and interferon-alfa 2b (IFN-ALPHA), in stages IIB, IIC and III post surgery cutaneous melanoma patients. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01729663. [Google Scholar]
  • 30.Bristol-Myers Squibb. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A randomized, double-blind, multicenter study comparing MDX-010 monotherapy, MDX-010 in combination with a melanoma peptide vaccine, and melanoma vaccine monotherapy in HLA-A2*0201-positive patients with previously treated unresectable stage III or IV melanoma. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/results/NCT00094653. [Google Scholar]
  • 31.National Cancer Institute (NCI) ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase III multi-institutional randomized study of immunization with the gp100: 209-217 (210M) peptide followed by high dose IL-2 vs. high dose IL-2 alone in patients with metastatic melanoma. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00019682. [Google Scholar]
  • 32.AVAX Technologies. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Comparison of M-Vax plus low dose interleukin-2 versus placebo vaccine plus low dose interleukin-2 in patients with stage IV melanoma. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00477906. [Google Scholar]
  • 33.Sotio AS. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A randomized, double blind, multicenter, parallel-group, phase III study to evaluate efficacy and safety of DCVAC/PCa versus placebo in men with metastatic castration resistant prostate cancer eligible for 1st line chemotherapy. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT02111577. [Google Scholar]
  • 34.Jonsson Comprehensive Cancer Center. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. An open label, sequential multi-center multi dose study of G17T immunogen in combination with cisplatin (CDDP) and 5-fluorouracil (5-FU) in subjects with metastatic or locally recurrent gastric or gastroesophageal cancer previously untreated with chemotherapy for advanced disease (Stage IV) [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00020787. [Google Scholar]
  • 35.NeuroVita Clinic. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Proteome-based personalized immunotherapy of malignant brain tumors. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01759810. [Google Scholar]
  • 36.NeuroVita Clinic. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Proteome-based personalized immunotherapy of brain metastases from lung cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01782287. [Google Scholar]
  • 37.NeuroVita Clinic. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Proteome-based personalized immunotherapy of brain metastases from breast cancer. [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT01782274. [Google Scholar]
  • 38.Northwest Biotherapeutics. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase III clinical trial evaluating DCVax®-L, autologous dendritic cells pulsed with tumor lysate antigen for the treatment of Glioblastoma Multiforme (GBM) [cited 2014 Aug 5]. Available from: http://clinicaltrials.gov/ct2/show/NCT00045968. [Google Scholar]
  • 39.Oxman MN. Zoster vaccine: current status and future prospects. Clin Infect Dis. 2010;51(2):197–213. doi: 10.1086/653605. [DOI] [PubMed] [Google Scholar]
  • 40.Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med. 2005;352(22):2271–84. doi: 10.1056/NEJMoa051016. [DOI] [PubMed] [Google Scholar]
  • 41.Astellas Pharma Global Development, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A randomized, double-blind, placebo-controlled, phase 3 trial to evaluate the protective efficacy and safety of a therapeutic vaccine, ASP0113, in cytomegalovirus (CMV)-seropositive recipients undergoing allogeneic, Hematopoietic Cell Transplant (HCT) [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01877655. [Google Scholar]
  • 42.Astellas Pharma Global Development, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. An open phase 2 trial to evaluate safety of a cytomegalovirus (CMV) therapeutic vaccine, ASP0113, in recipients undergoing allogeneic, Hematopoietic Cell Transplant (HCT) [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT01903928. [Google Scholar]
  • 43.Sabbaj S, Pass RF, Goepfert PA, Pichon S. Glycoprotein B vaccine is capable of boosting both antibody and CD4 T-cell responses to cytomegalovirus in chronically infected women. J Infect Dis. 2011;203(11):1534–41. doi: 10.1093/infdis/jir138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cavenaugh JS, Awi D, Mendy M, Hill AV, Whittle H, McConkey SJ. Partially randomized, non-blinded trial of DNA and MVA therapeutic vaccines based on hepatitis B virus surface protein for chronic HBV infection. PLoS One. 2011;6(2):e14626. doi: 10.1371/journal.pone.0014626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dikici B, Kalayci AG, Ozgenc F, Bosnak M, Davutoglu M, Ece A, et al. Therapeutic vaccination in the immunotolerant phase of children with chronic hepatitis B infection. Pediatr Infect Dis J. 2003;22(4):345–9. doi: 10.1097/01.inf.0000059443.49414.8b. [DOI] [PubMed] [Google Scholar]
  • 46.Jung MC, Gruner N, Zachoval R, Schraut W, Gerlach T, Diepolder H, et al. Immunological monitoring during therapeutic vaccination as a prerequisite for the design of new effective therapies: induction of a vaccine-specific CD4+ T-cell proliferative response in chronic hepatitis B carriers. Vaccine. 2002;20(29–30):3598–612. doi: 10.1016/s0264-410x(02)00309-2. [DOI] [PubMed] [Google Scholar]
  • 47.Senturk H, Tabak F, Akdogan M, Erdem L, Mert A, Ozaras R, et al. Therapeutic vaccination in chronic hepatitis B. J Gastroenterol Hepatol. 2002;17(1):72–6. doi: 10.1046/j.1440-1746.2002.02652.x. [DOI] [PubMed] [Google Scholar]
  • 48.Heintges T, Petry W, Kaldewey M, Erhardt A, Wend UC, Gerlich WH, et al. Combination therapy of active HBsAg vaccination and interferon-alpha in interferon-alpha nonresponders with chronic hepatitis B. Dig Dis Sci. 2001;46(4):901–6. doi: 10.1023/a:1010785325067. [DOI] [PubMed] [Google Scholar]
  • 49.Livingston BD, Alexander J, Crimi C, Oseroff C, Celis E, Daly K, et al. Altered helper T lymphocyte function associated with chronic hepatitis B virus infection and its role in response to therapeutic vaccination in humans. J Immunol. 1999;162(5):3088–95. [PubMed] [Google Scholar]
  • 50.Vitiello A, Ishioka G, Grey HM, Rose R, Farness P, LaFond R, et al. Development of a lipopeptide-based therapeutic vaccine to treat chronic HBV infection. I. Induction of a primary cytotoxic T lymphocyte response in humans. J Clin Invest. 1995;95(1):341–9. doi: 10.1172/JCI117662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dynavax Technologies Corporation. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase Ib dose-escalation study to assess the safety and tolerability of DV-601 in subjects with chronic hepatitis B on concurrent treatment with a nucleoside analogue. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01023230. [Google Scholar]
  • 52.Chang Gung Memorial Hospital. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Effect of hepatitis B vaccine in chronic hepatitis B patients with low serum HBsAg-a pilot study. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01817725. [Google Scholar]
  • 53.Genexine, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. An open-label, dose-escalating clinical study to evaluate the tolerability, immunogenicity and efficacy of HB-110 administered by electroporation (EP) in an add-on therapy with entecavir in chronic hepatitis B patients. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01641536. [Google Scholar]
  • 54.PowderMed. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase I, multi-centre, randomised, placebo-controlled, dose escalation study to assess local & systemic tolerability of therapeutic DNA plasmid pdpSC18 vaccine administered by particle mediated epidermal delivery using powderJect ND10 delivery system in subjects with chronic hepatitis B infection. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT00277576. [Google Scholar]
  • 55.Seoul National University Hospital. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase 4, to compare efficacy and safety of therapeutic vaccination with intensified schedule plus pegylated interferon dual therapy on seroclearance of HBS antigen in patients with complete virological response induced by entecavir. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT02097004. [Google Scholar]
  • 56.Vandepapeliere P, Lau GK, Leroux-Roels G, Horsmans Y, Gane E, Tawandee T, et al. Therapeutic vaccination of chronic hepatitis B patients with virus suppression by antiviral therapy: a randomized, controlled study of co-administration of HBsAg/AS02 candidate vaccine and lamivudine. Vaccine. 2007;25(51):8585–97. doi: 10.1016/j.vaccine.2007.09.072. [DOI] [PubMed] [Google Scholar]
  • 57.Yalcin K, Danis R, Degertekin H, Alp MN, Tekes S, Budak T. The lack of effect of therapeutic vaccination with a pre-S2/S HBV vaccine in the immune tolerant phase of chronic HBV infection. J Clin Gastroenterol. 2003;37(4):330–5. doi: 10.1097/00004836-200310000-00012. [DOI] [PubMed] [Google Scholar]
  • 58.Xu DZ, Wang XY, Shen XL, Gong GZ, Ren H, Guo LM, et al. Results of a phase III clinical trial with an HBsAg-HBIG immunogenic complex therapeutic vaccine for chronic hepatitis B patients: experiences and findings. J Hepatol. 2013;59(3):450–6. doi: 10.1016/j.jhep.2013.05.003. [DOI] [PubMed] [Google Scholar]
  • 59.Ren F, Hino K, Yamaguchi Y, Funatsuki K, Hayashi A, Ishiko H, et al. Cytokine-dependent anti-viral role of CD4-positive T cells in therapeutic vaccination against chronic hepatitis B viral infection. J Med Virol. 2003;71(3):376–84. doi: 10.1002/jmv.10509. [DOI] [PubMed] [Google Scholar]
  • 60.Lee YB, Lee JH, Kim YJ, Yoon JH, Lee HS. The effect of therapeutic vaccination for the treatment of chronic hepatitis B virus infection. J Med Virol. 2015;87(4):575–82. doi: 10.1002/jmv.24091. [DOI] [PubMed] [Google Scholar]
  • 61.Clinical Research Organization, Dhaka Bangladesh. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase III study of a therapeutic vaccine candidate containing hepatitis B virus (HBV) core antigen (HBcAg) and HBV surface antigen (HBsAg) for treatment of patients with chronic HBV infection. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT01374308. [Google Scholar]
  • 62.Schlaphoff V, Klade CS, Jilma B, Jelovcan SB, Cornberg M, Tauber E, et al. Functional and phenotypic characterization of peptide-vaccine-induced HCV-specific CD8+ T cells in healthy individuals and chronic hepatitis C patients. Vaccine. 2007;25(37–38):6793–806. doi: 10.1016/j.vaccine.2007.06.026. [DOI] [PubMed] [Google Scholar]
  • 63.Nevens F, Roskams T, Van VH, Horsmans Y, Sprengers D, Elewaut A, et al. A pilot study of therapeutic vaccination with envelope protein E1 in 35 patients with chronic hepatitis C. Hepatology. 2003;38(5):1289–96. doi: 10.1053/jhep.2003.50474. [DOI] [PubMed] [Google Scholar]
  • 64.Amador-Canizares Y, Martinez-Donato G, Alvarez-Lajonchere L, Vasallo C, Dausa M, Aguilar-Noriega D, et al. HCV-specific immune responses induced by CIGB-230 in combination with IFN-alpha plus ribavirin. World J Gastroenterol. 2014;20(1):148–62. doi: 10.3748/wjg.v20.i1.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Weiland O, Ahlen G, Diepolder H, Jung MC, Levander S, Fons M, et al. Therapeutic DNA vaccination using in vivo electroporation followed by standard of care therapy in patients with genotype 1 chronic hepatitis C. Mol Ther. 2013;21(9):1796–805. doi: 10.1038/mt.2013.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wedemeyer H, Schuller E, Schlaphoff V, Stauber RE, Wiegand J, Schiefke I, et al. Therapeutic vaccine IC41 as late add-on to standard treatment in patients with chronic hepatitis C. Vaccine. 2009;27(37):5142–51. doi: 10.1016/j.vaccine.2009.06.027. [DOI] [PubMed] [Google Scholar]
  • 67.Drane D, Maraskovsky E, Gibson R, Mitchell S, Barnden M, Moskwa A, et al. Priming of CD4+ and CD8+ T cell responses using a HCV core ISCOMATRIX vaccine: a phase I study in healthy volunteers. Hum Vaccin. 2009;5(3):151–7. doi: 10.4161/hv.5.3.6614. [DOI] [PubMed] [Google Scholar]
  • 68.Batdelger D, Dandii D, Jirathitikal V, Bourinbaiar AS. Open-label trial of therapeutic immunization with oral V-5 Immunitor (V5) vaccine in patients with chronic hepatitis C. Vaccine. 2008;26(22):2733–7. doi: 10.1016/j.vaccine.2008.03.021. [DOI] [PubMed] [Google Scholar]
  • 69.Transgene. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase II randomized, multicenter, open-label study of TG4040 (MVA-HCV) in combination with pegylated interferon Alfa-2a and ribavirin versus pegylated interferon Alfa-2a and ribavirin in treatment-naïve patients with chronic genotype 1 hepatitis C. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT01055821. [Google Scholar]
  • 70.GlobeImmune. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase 1 double-blind, placebo controlled, dose-escalation, multi-center therapeutic trial of the safety, immunogenicity, and efficacy of GI-5005; an inactivated recombinant saccharomyces cerevisiae expressing a hepatitis C virus NS3-core fusion protein, in patients with chronic hepatitis C infection. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT00124215. [Google Scholar]
  • 71.Pevion Biotech Ltd. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase I single-blind randomised placebo controlled dose escalating study of one virosome formulated CD4 and two virosomes formulated CD8 hepatitis C virus (HCV) vaccine components (PEV2A and PEV2B) administered to healthy adult volunteers. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT00445419. [Google Scholar]
  • 72.ReiThera Srl. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase Ib study to assess the safety and immunogenicity of a novel HCV vaccine, based on the sequential injection of Ad6NSmut and MVA-NSmut, given in combination with PEG-interferon Alfa plus ribavirin for re-treatment of chronic hepatitis C. [cited 2015 Apr 10]. Available from: http://clinicaltrials.gov/ct2/show/NCT01701336. [Google Scholar]
  • 73.ReiThera Srl. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase I study to assess the safety and immunogenicity of Ad6NSmut and AdCh3NSmut in patients with hepatitis C virus infection. [cited 2015 Apr 10]. Available from: http://clinicaltrials.gov/ct2/show/NCT01094873. [Google Scholar]
  • 74.GlobeImmune. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase 2 randomized, open label, multi-center, therapeutic trial of the efficacy, immunogenicity, and safety of GI-5005; an inactivated recombinant saccharomyces cerevisiae expressing a hepatitis C virus NS3-core fusion protein, combined with pegylated interferon plus ribavirin standard of care therapy versus standard of care alone, and GI-5005 salvage of standard of care failures, in patients with genotype 1 chronic hepatitis C infection. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT00606086. [Google Scholar]
  • 75.Valneva Austria GmbH. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Open-label, multicenter trial with IC41, a therapeutic HCV vaccine in patients with chronic HCV. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT00601770. [Google Scholar]
  • 76.Di Bisceglie AM, Janczweska-Kazek E, Habersetzer F, Mazur W, Stanciu C, Carreno V, et al. Efficacy of immunotherapy with TG4040, peg-interferon, and ribavirin in a phase 2 study of patients with chronic HCV infection. Gastroenterology. 2014;147(1):119–31. doi: 10.1053/j.gastro.2014.03.007. [DOI] [PubMed] [Google Scholar]
  • 77.Daayana S, Elkord E, Winters U, Pawlita M, Roden R, Stern PL, et al. Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br J Cancer. 2010;102(7):1129–36. doi: 10.1038/sj.bjc.6605611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Radulovic S, Brankovic-Magic M, Malisic E, Jankovic R, Dobricic J, Plesinac-Karapandzic V, et al. Therapeutic cancer vaccines in cervical cancer: phase I study of Lovaxin-C. J BUON. 2009;14(Suppl 1):S165–8. [PubMed] [Google Scholar]
  • 79.Van Doorslaer K, Reimers LL, Studentsov YY, Einstein MH, Burk RD. Serological response to an HPV16 E7 based therapeutic vaccine in women with high-grade cervical dysplasia. Gynecol Oncol. 2010;116(2):208–12. doi: 10.1016/j.ygyno.2009.05.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Albarran YC, de la Garza A, Cruz Quiroz BJ, Vazquez ZE, Diaz EI, Mendez FE, et al. MVA E2 recombinant vaccine in the treatment of human papillomavirus infection in men presenting intraurethral flat condyloma: a phase I/II study. BioDrugs. 2007;21(1):47–59. doi: 10.2165/00063030-200721010-00006. [DOI] [PubMed] [Google Scholar]
  • 81.Palefsky JM, Berry JM, Jay N, Krogstad M, Da CM, Darragh TM, et al. A trial of SGN-00101 (HspE7) to treat high-grade anal intraepithelial neoplasia in HIV-positive individuals. AIDS. 2006;20(8):1151–5. doi: 10.1097/01.aids.0000226955.02719.26. [DOI] [PubMed] [Google Scholar]
  • 82.Garcia-Hernandez E, Gonzalez-Sanchez JL, Andrade-Manzano A, Contreras ML, Padilla S, Guzman CC, et al. Regression of papilloma high-grade lesions (CIN 2 and CIN 3) is stimulated by therapeutic vaccination with MVA E2 recombinant vaccine. Cancer Gene Ther. 2006;13(6):592–7. doi: 10.1038/sj.cgt.7700937. [DOI] [PubMed] [Google Scholar]
  • 83.Vandepapeliere P, Barrasso R, Meijer CJ, Walboomers JM, Wettendorff M, Stanberry LR, et al. Randomized controlled trial of an adjuvanted human papillomavirus (HPV) type 6 L2E7 vaccine: infection of external anogenital warts with multiple HPV types and failure of therapeutic vaccination. J Infect Dis. 2005;192(12):2099–107. doi: 10.1086/498164. [DOI] [PubMed] [Google Scholar]
  • 84.Baldwin PJ, van der Burg SH, Boswell CM, Offringa R, Hickling JK, Dobson J, et al. Vaccinia-expressed human papillomavirus 16 and 18 e6 and e7 as a therapeutic vaccination for vulval and vaginal intraepithelial neoplasia. Clin Cancer Res. 2003;9(14):5205–13. [PubMed] [Google Scholar]
  • 85.Klencke B, Matijevic M, Urban RG, Lathey JL, Hedley ML, Berry M, et al. Encapsulated plasmid DNA treatment for human papillomavirus 16-associated anal dysplasia: a phase I study of ZYC101. Clin Cancer Res. 2002;8(5):1028–37. [PubMed] [Google Scholar]
  • 86.Lacey CJ, Thompson HS, Monteiro EF, O'Neill T, Davies ML, Holding FP, et al. Phase IIa safety and immunogenicity of a therapeutic vaccine, TA-GW, in persons with genital warts. J Infect Dis. 1999;179(3):612–18. doi: 10.1086/314616. [DOI] [PubMed] [Google Scholar]
  • 87.Academisch Medisch Centrum—Universiteit van Amsterdam (AMC-UvA) ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Therapeutic vaccination against human papillomavirus type 16 for the treatment of anal intraepithelial neoplasia in HIV+ men. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT01923116. [Google Scholar]
  • 88.University of Arkansas. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase I clinical trial of an HPV therapeutic vaccine. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01653249. [Google Scholar]
  • 89.Genticel. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A double-blind, randomised, placebo-controlled, phase II study to evaluate ProCervix efficacy to clear HPV 16 and HPV 18 infection in women with normal cytology or ASCUS/LSIL. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/NCT01957878. [Google Scholar]
  • 90.ISA Pharmaceuticals. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. Phase I/II study to determine the safety and immune modulating effects of the therapeutic human papilloma virus 16 (HPV16) E6/E7 long peptides vaccine (ISA101) at different doses with or without IFNα as combination therapy with carboplatin and paclitaxel in women with HPV16 positive advanced or recurrent cervical cancer. [cited 2014 Aug 7]. Available from: http://clinicaltrials.gov/ct2/show/record/NCT02128126. [Google Scholar]
  • 91.Genocea Biosciences, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase I/IIa, randomized, double-blind, dose-ranging, placebo-controlled study of the safety and immunogenicity of a HSV-2 vaccine containing matrix M-2 adjuvant in individuals with documented genital HSV-2 genital infection. [cited 2014 Aug 7]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01667341. [Google Scholar]
  • 92.Genocea Biosciences, Inc. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A randomized, double-blind, factorial study to compare the safety and efficacy of varying combinations of GEN-003 and matrix-M2 in subjects with genital HSV-2 infection. [cited 2014 Aug 7]. Available from: http://www.clinicaltrials.gov/ct2/show/NCT02114060. [Google Scholar]
  • 93.Wald A, Koelle DM, Fife K, Warren T, Leclair K, Chicz RM, et al. Safety and immunogenicity of long HSV-2 peptides complexed with rhHsc70 in HSV-2 seropositive persons. Vaccine. 2011;29(47):8520–9. doi: 10.1016/j.vaccine.2011.09.046. [DOI] [PubMed] [Google Scholar]
  • 94.Efremenko YV, Butov DA, Prihoda ND, Zaitzeva SI, Yurchenko LV, Sokolenko NI, et al. Randomized, placebo-controlled phase II trial of heat-killed mycobacterium vaccae (longcom batch) formulated as an oral pill (V7) Hum Vaccin Immunother. 2013;9(9):1852–6. doi: 10.4161/hv.25280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Nell AS, D'lom E, Bouic P, Sabate M, Bosser R, Picas J, et al. Safety, tolerability, and immunogenicity of the novel antituberculous vaccine RUTI: randomized, placebo-controlled phase II clinical trial in patients with latent tuberculosis infection. PLoS One. 2014;9(2):e89612. doi: 10.1371/journal.pone.0089612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dumonteil E, Bottazzi ME, Zhan B, Heffernan MJ, Jones K, Valenzuela JG, et al. Accelerating the development of a therapeutic vaccine for human Chagas disease: rationale and prospects. Expert Rev Vaccines. 2012;11(9):1043–55. doi: 10.1586/erv.12.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Infectious Disease Research Institute. ClinicalTrials.gov [Internet] Bethesda, MD: National Library of Medicine (US); 2000. A phase 2, randomized, open-label, controlled study to evaluate the efficacy, safety, and immunogenicity of the LEISH-F2+MPL-SE vaccine in the treatment of patients with cutaneous leishmaniasis. [cited 2015 Apr 2]. Available from: http://clinicaltrials.gov/ct2/show/NCT01011309. [Google Scholar]
  • 98.Ensoli B, Cafaro A, Monini P, Marcotullio S, Ensoli F. Challenges in HIV vaccine research for treatment and prevention. Front Immunol. 2014;5:417. doi: 10.3389/fimmu.2014.00417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Garcia F, Leon A, Gatell JM, Plana M, Gallart T. Therapeutic vaccines against HIV infection. Hum Vaccin Immunother. 2012;8(5):569–81. doi: 10.4161/hv.19555. [DOI] [PubMed] [Google Scholar]
  • 100.Fisher AK, Voronin Y, Jefferys R. Therapeutic HIV vaccines: prior setbacks, current advances, and future prospects. Vaccine. 2014;32(43):5540–5. doi: 10.1016/j.vaccine.2014.06.066. [DOI] [PubMed] [Google Scholar]
  • 101.Mylvaganam GH, Silvestri G, Amara RR. HIV therapeutic vaccines: moving towards a functional cure. Curr Opin Immunol. 2015;35:1–8. doi: 10.1016/j.coi.2015.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.de Goede AL, Vulto AG, Osterhaus AD, Gruters RA. Understanding HIV infection for the design of a therapeutic vaccine. Part II: vaccination strategies for HIV. Ann Pharm Fr. 2015;73(3):169–79. doi: 10.1016/j.pharma.2014.11.003. [DOI] [PubMed] [Google Scholar]
  • 103.Boffito M, Fox J, Bowman C, Fisher M, Orkin C, Wilkins E, et al. Safety, immunogenicity and efficacy assessment of HIV immunotherapy in a multi-centre, double-blind, randomised, placebo-controlled phase Ib human trial. Vaccine. 2013;31(48):5680–6. doi: 10.1016/j.vaccine.2013.09.057. [DOI] [PubMed] [Google Scholar]
  • 104.Rosenberg ES, Graham BS, Chan ES, Bosch RJ, Stocker V, Maenza J, et al. Safety and immunogenicity of therapeutic DNA vaccination in individuals treated with antiretroviral therapy during acute/early HIV-1 infection. PLoS One. 2010;5(5):2010. doi: 10.1371/journal.pone.0010555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Markowitz M, Jin X, Hurley A, Simon V, Ramratnam B, Louie M, et al. Discontinuation of antiretroviral therapy commenced early during the course of human immunodeficiency virus type 1 infection, with or without adjunctive vaccination. J Infect Dis. 2002;186(5):634–43. doi: 10.1086/342559. [DOI] [PubMed] [Google Scholar]
  • 106.Kinloch-De LS, Hoen B, Smith DE, Autran B, Lampe FC, Phillips AN, et al. Impact of therapeutic immunization on HIV-1 viremia after discontinuation of antiretroviral therapy initiated during acute infection. J Infect Dis. 2005;192(4):607–17. doi: 10.1086/432002. [DOI] [PubMed] [Google Scholar]
  • 107.Goujard C, Marcellin F, Hendel-Chavez H, Burgard M, Meiffredy V, Venet A, et al. Interruption of antiretroviral therapy initiated during primary HIV-1 infection: impact of a therapeutic vaccination strategy combined with interleukin (IL)-2 compared with IL-2 alone in the ANRS 095 Randomized Study. AIDS Res Hum Retroviruses. 2007;23(9):1105–13. doi: 10.1089/aid.2007.0047. [DOI] [PubMed] [Google Scholar]
  • 108.Emery S, Kelleher AD, Workman C, Puls RL, Bloch M, Baker D, et al. Influence of IFNgamma co-expression on the safety and antiviral efficacy of recombinant fowlpox virus HIV therapeutic vaccines following interruption of antiretroviral therapy. Hum Vaccin. 2007;3(6):260–7. doi: 10.4161/hv.4627. [DOI] [PubMed] [Google Scholar]
  • 109.Levy Y, Gahery-Segard H, Durier C, Lascaux AS, Goujard C, Meiffredy V, et al. Immunological and virological efficacy of a therapeutic immunization combined with interleukin-2 in chronically HIV-1 infected patients. AIDS. 2005;19(3):279–86. [PubMed] [Google Scholar]
  • 110.Garcia F, Climent N, Guardo AC, Gil C, Leon A, Autran B, et al. A dendritic cell-based vaccine elicits T cell responses associated with control of HIV-1 replication. Sci Transl Med. 2013;5(166):166ra2. doi: 10.1126/scitranslmed.3004682. [DOI] [PubMed] [Google Scholar]
  • 111.Pollard RB, Rockstroh JK, Pantaleo G, Asmuth DM, Peters B, Lazzarin A, et al. Safety and efficacy of the peptide-based therapeutic vaccine for HIV-1, Vacc-4x: a phase 2 randomised, double-blind, placebo-controlled trial. Lancet Infect Dis. 2014;14(4):291–300. doi: 10.1016/S1473-3099(13)70343-8. [DOI] [PubMed] [Google Scholar]
  • 112.Andres C, Plana M, Guardo AC, Alvarez-Fernandez C, Climent N, Gallart T, et al. HIV-1 reservoir dynamics after vaccination and antiretroviral therapy interruption are associated with dendritic cell vaccine-induced T cell responses. J Virol. 2015;89(18):9189–99. doi: 10.1128/JVI.01062-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Eron JJ. Managing antiretroviral therapy: changing regimens, resistance testing, and the risks from structured treatment interruptions. J Infect Dis. 2008;197(Suppl 3):S261–71. doi: 10.1086/533418. [DOI] [PubMed] [Google Scholar]
  • 114.Paton NI. Treatment interruption strategies: how great are the risks? Curr Opin Infect Dis. 2008;21(1):25–30. doi: 10.1097/QCO.0b013e3282f4069d. [DOI] [PubMed] [Google Scholar]
  • 115.Routy JP, Boulassel MR, Nicolette CA, Jacobson JM. Assessing risk of a short-term antiretroviral therapy discontinuation as a read-out of viral control in immune-based therapy. J Med Virol. 2012;84(6):885–9. doi: 10.1002/jmv.23297. [DOI] [PubMed] [Google Scholar]
  • 116.Doherty M, Ford N, Vitoria M, Weiler G, Hirnschall G. The 2013 WHO guidelines for antiretroviral therapy: evidence-based recommendations to face new epidemic realities. Curr Opin HIV AIDS. 2013;8(6):528–34. doi: 10.1097/COH.0000000000000008. [DOI] [PubMed] [Google Scholar]
  • 117.El-Sadr WM, Lundgren J, Neaton JD, Gordin F, Abrams D, Arduino RC, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355(22):2283–96. doi: 10.1056/NEJMoa062360. [DOI] [PubMed] [Google Scholar]
  • 118.Garcia F, Plana M, Vidal C, Cruceta A, O'Brien WA, Pantaleo G, et al. Dynamics of viral load rebound and immunological changes after stopping effective antiretroviral therapy. AIDS. 1999;13(11):F79–86. doi: 10.1097/00002030-199907300-00002. [DOI] [PubMed] [Google Scholar]
  • 119.Davey RT, Jr, Bhat N, Yoder C, Chun TW, Metcalf JA, Dewar R, et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc Natl Acad Sci U S A. 1999;96(26):15109–14. doi: 10.1073/pnas.96.26.15109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Angel JB, Routy JP, Tremblay C, Ayers D, Woods R, Singer J, et al. A randomized controlled trial of HIV therapeutic vaccination using ALVAC with or without Remune. AIDS. 2011;25(6):731–9. doi: 10.1097/QAD.0b013e328344cea5. [DOI] [PubMed] [Google Scholar]
  • 121.Hatano H, Vogel S, Yoder C, Metcalf JA, Dewar R, Davey RT, Jr, et al. Pre-HAART HIV burden approximates post-HAART viral levels following interruption of therapy in patients with sustained viral suppression. AIDS. 2000;14(10):1357–63. doi: 10.1097/00002030-200007070-00008. [DOI] [PubMed] [Google Scholar]
  • 122.Saez-Cirion A, Bacchus C, Hocqueloux L, Avettand-Fenoel V, Girault I, Lecuroux C, et al. Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI study. PLoS Pathog. 2013;9(3):e1003211. doi: 10.1371/journal.ppat.1003211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Anderson JL, Fromentin R, Corbelli GM, Ostergaard L, Ross AL. Progress towards an HIV cure: update from the 2014 International AIDS Society Symposium. AIDS Res Hum Retroviruses. 2015;31(1):36–44. doi: 10.1089/aid.2014.0236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Ananworanich J, Gayet-Ageron A, Le Braz M, Prasithsirikul W, Chetchotisakd P, Kiertiburanakul S, et al. CD4-guided scheduled treatment interruptions compared with continuous therapy for patients infected with HIV-1: results of the Staccato randomised trial. Lancet. 2005;368(9534):459–65. doi: 10.1016/S0140-6736(06)69153-8. [DOI] [PubMed] [Google Scholar]
  • 125.Maggiolo F, Airoldi M, Callegaro A, Martinelli C, Dolara A, Bini T, et al. CD4 cell-guided scheduled treatment interruptions in HIV-infected patients with sustained immunologic response to HAART. AIDS. 2009;23(7):799–807. doi: 10.1097/QAD.0b013e328321b75e. [DOI] [PubMed] [Google Scholar]
  • 126.Piketty C, Weiss L, Assoumou L, Burgard M, Melard A, Ragnaud JM, et al. A high HIV DNA level in PBMCs at antiretroviral treatment interruption predicts a shorter time to treatment resumption, independently of the CD4 nadir. J Med Virol. 2010;82(11):1819–28. doi: 10.1002/jmv.21907. [DOI] [PubMed] [Google Scholar]
  • 127.Rothenberger MK, Keele BF, Wietgrefe SW, Fletcher CV, Beilman GJ, Chipman JG, et al. Large number of rebounding/founder HIV variants emerge from multifocal infection in lymphatic tissues after treatment interruption. Proc Natl Acad Sci U S A. 2015;112(10):E1126–34. doi: 10.1073/pnas.1414926112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Colven R, Harrington RD, Spach DH, Cohen CJ, Hooton TM. Retroviral rebound syndrome after cessation of suppressive antiretroviral therapy in three patients with chronic HIV infection. Ann Intern Med. 2000;133(6):430–4. doi: 10.7326/0003-4819-133-6-200009190-00010. [DOI] [PubMed] [Google Scholar]
  • 129.Kilby JM, Goepfert PA, Miller AP, Gnann JW, Jr, Sillers M, Saag MS, et al. Recurrence of the acute HIV syndrome after interruption of antiretroviral therapy in a patient with chronic HIV infection: a case report. Ann Intern Med. 2000;133(6):435–8. doi: 10.7326/0003-4819-133-6-200009190-00011. [DOI] [PubMed] [Google Scholar]
  • 130.Ananworanich J, Phanuphak N, Nuesch R, Apateerapong W, Rojnuckarin P, Ubolyam S, et al. Recurring thrombocytopenia associated with structured treatment interruption in patients with human immunodeficiency virus infection. Clin Infect Dis. 2003;37(5):723–5. doi: 10.1086/376989. [DOI] [PubMed] [Google Scholar]
  • 131.Burman W, Grund B, Neuhaus J, Douglas J, Jr, Friedland G, Telzak E, et al. Episodic antiretroviral therapy increases HIV transmission risk compared with continuous therapy: results of a randomized controlled trial. J Acquir Immune Defic Syndr. 2008;49(2):142–50. doi: 10.1097/QAI.0b013e318183a9ad. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Stevenson M. CROI 2015: basic science review. Top Antivir Med. 2015;23(1):4–7. [PubMed] [Google Scholar]
  • 133.Cillo AR, Krishnan A, Mitsuyasu RT, McMahon DK, Li S, Rossi JJ, et al. Plasma viremia and cellular HIV-1 DNA persist despite autologous hematopoietic stem cell transplantation for HIV-related lymphoma. J Acquir Immune Defic Syndr. 2013;63(4):438–41. doi: 10.1097/QAI.0b013e31828e6163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Spentzou A, Bergin P, Gill D, Cheeseman H, Ashraf A, Kaltsidis H, et al. Viral inhibition assay: a CD8 T cell neutralization assay for use in clinical trials of HIV-1 vaccine candidates. J Infect Dis. 2010;201(5):720–9. doi: 10.1086/650492. [DOI] [PubMed] [Google Scholar]
  • 135.Yang H, Wu H, Hancock G, Clutton G, Sande N, Xu X, et al. Antiviral inhibitory capacity of CD8+ T cells predicts the rate of CD4+ T-cell decline in HIV-1 infection. J Infect Dis. 2012;206(4):552–61. doi: 10.1093/infdis/jis379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kran A-M, Sorensen B, Nyhus J, Sommerfelt MA, Baksaas I, Bruun JN, et al. HLA- and dose-dependent immunogenicity of a peptide-based HIV-1 immunotherapy candidate (Vacc-4x) AIDS. 2004;18(14):1875–83. doi: 10.1097/00002030-200409240-00003. [DOI] [PubMed] [Google Scholar]
  • 137.Kran AM, Sommerfelt MA, Sorensen B, Nyhus J, Baksaas I, Bruun JN, et al. Reduced viral burden amongst high responder patients following HIV-1 p24 peptide-based therapeutic immunization. Vaccine. 2005;23(31):4011–15. doi: 10.1016/j.vaccine.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 138.Kran AM, Sorensen B, Sommerfelt MA, Nyhus J, Baksaas I, Kvale D. Long-term HIV-specific responses and delayed resumption of antiretroviral therapy after peptide immunization targeting dendritic cells. AIDS. 2006;20(4):627–30. doi: 10.1097/01.aids.0000210620.75707.ac. [DOI] [PubMed] [Google Scholar]
  • 139.Kran AM, Sommerfelt MA, Baksaas I, Sorensen B, Kvale D. Delayed-type hypersensitivity responses to HIV Gag p24 relate to clinical outcome after peptide-based therapeutic immunization for chronic HIV infection. APMIS. 2012;120(3):204–9. doi: 10.1111/j.1600-0463.2011.02843.x. [DOI] [PubMed] [Google Scholar]
  • 140.Goldstein G, Damiano E, Donikyan M, Pasha M, Beckwith E, Chicca J. HIV-1 Tat B-cell epitope vaccination was ineffectual in preventing viral rebound after ART cessation: HIV rebound with current ART appears to be due to infection with new endogenous founder virus and not to resurgence of pre-existing Tat-dependent viremia. Hum Vaccin Immunother. 2012;8(10):1425–30. doi: 10.4161/hv.21616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kityo C, Bousheri S, Akao J, Ssali F, Byaruhanga R, Ssewanyana I, et al. Therapeutic immunization in HIV infected Ugandans receiving stable antiretroviral treatment: a Phase I safety study. Vaccine. 2011;29(8):1617–23. doi: 10.1016/j.vaccine.2010.12.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Moss RB, Brandt C, Giermakowska WK, Savary JR, Theofan G, Zanetti M, et al. HIV-specific immunity during structured antiviral drug treatment interruption. Vaccine. 2003;21(11–12):1066–71. doi: 10.1016/s0264-410x(02)00610-2. [DOI] [PubMed] [Google Scholar]
  • 143.Huang KH, Boisvert MP, Chung F, Loignon M, Zarowny D, Cyr L, et al. Longitudinal changes in HIV-specific IFN-gamma secretion in subjects who received Remune vaccination prior to treatment interruption. J Immune Based Ther Vaccines. 2006;4:7. doi: 10.1186/1476-8518-4-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Bostrom A-C, Hejdeman B, Matsuda R, Fredriksson M, Fredriksson E-L, Bratt G, et al. Long-term persistence of vaccination and HAART to human immunodeficiency virus (HIV) Vaccine. 2004;22(13–14):1683–91. doi: 10.1016/j.vaccine.2003.09.049. [DOI] [PubMed] [Google Scholar]
  • 145.Gudmundsdotter L, Wahren B, Haller BK, Boberg A, Edback U, Bernasconi D, et al. Amplified antigen-specific immune responses in HIV-1 infected individuals in a double blind DNA immunization and therapy interruption trial. Vaccine. 2011;29(33):5558–66. doi: 10.1016/j.vaccine.2011.01.064. [DOI] [PubMed] [Google Scholar]
  • 146.Harrer E, Bauerle M, Ferstl B, Chaplin P, Petzold B, Mateo L, et al. Therapeutic vaccination of HIV-1-infected patients on HAART with a recombinant HIV-1 nef-expressing MVA: safety, immunogenicity and influence on viral load during treatment interruption. Antivir Ther. 2005;10(2):285–300. [PubMed] [Google Scholar]
  • 147.Dorrell L, Yang H, Ondondo B, Dong T, Di GK, Suttill A, et al. Expansion and diversification of virus-specific T cells following immunization of human immunodeficiency virus type 1 (HIV-1)-infected individuals with a recombinant modified vaccinia virus Ankara/HIV-1 gag vaccine. J Virol. 2006;80(10):4705–16. doi: 10.1128/JVI.80.10.4705-4716.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Yang H, Guimaraes-Walker A, Hibbs S, Dong T, Stacey A, Borrow P, et al. Interleukin-10 responses to therapeutic vaccination during highly active antiretroviral therapy and after analytical therapy interruption. AIDS. 2009;23(16):2226–30. doi: 10.1097/QAD.0b013e328331a424. [DOI] [PubMed] [Google Scholar]
  • 149.Mothe B, Climent N, Plana M, Rosas M, Jimenez JL, Munoz-Fernandez MA, et al. Safety and immunogenicity of a modified vaccinia Ankara-based HIV-1 vaccine (MVA-B) in HIV-1-infected patients alone or in combination with a drug to reactivate latent HIV-1. J Antimicrob Chemother. 2015;70(6):1833–42. doi: 10.1093/jac/dkv046. [DOI] [PubMed] [Google Scholar]
  • 150.Jacobson JM, Bucy RP, Spritzler J, Saag MS, Eron JJ, Coombs RW, et al. Evidence that intermittent structured treatment interruption, but not immunization with ALVAC-HIV vCP1452, promotes host control of HIV replication: the results of AIDS clinical trials group 5068. J Infect Dis. 2006;194(5):623–32. doi: 10.1086/506364. [DOI] [PubMed] [Google Scholar]
  • 151.Autran B, Murphy RL, Costagliola D, Tubiana R, Clotet B, Gatell J, et al. Greater viral rebound and reduced time to resume antiretroviral therapy after therapeutic immunization with the ALVAC-HIV vaccine (vCP1452) AIDS. 2008;22(11):1313–22. doi: 10.1097/QAD.0b013e3282fdce94. [DOI] [PubMed] [Google Scholar]
  • 152.Gandhi RT, O'Neill D, Bosch RJ, Chan ES, Bucy RP, Shopis J, et al. A randomized therapeutic vaccine trial of canarypox-HIV-pulsed dendritic cells vs. canarypox-HIV alone in HIV-1-infected patients on antiretroviral therapy. Vaccine. 2009;27(43):6088–94. doi: 10.1016/j.vaccine.2009.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kilby JM, Bucy RP, Mildvan D, Fischl M, Santana-Bagur J, Lennox J, et al. A randomized, partially blinded phase 2 trial of antiretroviral therapy, HIV-specific immunizations, and interleukin-2 cycles to promote efficient control of viral replication (ACTG A5024) J Infect Dis. 2006;194(12):1672–6. doi: 10.1086/509508. [DOI] [PubMed] [Google Scholar]
  • 154.Angel J, Routy J-P, Graziani GM, Tremblay CL. The effect of therapeutic HIV vaccination with ALVAC-HIV with or without Remune on the size of the viral reservoir (a CTN 173 substudy) J Acquir Immune Defic Syndr. 2015;70:122–8. doi: 10.1097/QAI.0000000000000734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Tubiana R, Carcelain G, Vray M, Gourlain K, Dalban C, Chermak A, et al. Therapeutic immunization with a human immunodeficiency virus (HIV) type 1-recombinant canarypox vaccine in chronically HIV-infected patients: The Vacciter Study (ANRS 094) Vaccine. 2005;23(34):4292–301. doi: 10.1016/j.vaccine.2005.04.013. [DOI] [PubMed] [Google Scholar]
  • 156.Emery S, Workman C, Puls RL, Bloch M, Baker D, Bodsworth N, et al. Randomized, placebo-controlled, phase I/IIa evaluation of the safety and immunogenicity of fowlpox virus expressing HIV gag-pol and interferon-gamma in HIV-1 infected subjects. Hum Vaccin. 2005;1(6):232–8. doi: 10.4161/hv.1.6.2342. [DOI] [PubMed] [Google Scholar]
  • 157.Schooley RT, Spritzler J, Wang H, Lederman MM, Havlir D, Kuritzkes DR, et al. AIDS clinical trials group 5197: a placebo-controlled trial of immunization of HIV-1-infected persons with a replication-deficient adenovirus type 5 vaccine expressing the HIV-1 core protein. J Infect Dis. 2010;202(5):705–16. doi: 10.1086/655468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Li JZ, Brumme CJ, Lederman MM, Brumme ZL, Wang H, Spritzler J, et al. Characteristics and outcomes of initial virologic suppressors during analytic treatment interruption in a therapeutic HIV-1 gag vaccine trial. PLoS One. 2012;7(3):e34134. doi: 10.1371/journal.pone.0034134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Li JZ, Heisey A, Ahmed H, Wang H, Zheng L, Carrington M, et al. Relationship of HIV reservoir characteristics with immune status and viral rebound kinetics in an HIV therapeutic vaccine study. AIDS. 2014;28(18):2649–57. doi: 10.1097/QAD.0000000000000478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Garcia F, Lejeune M, Climent N, Gil C, Alcami J, Morente V, et al. Therapeutic immunization with dendritic cells loaded with heat-inactivated autologous HIV-1 in patients with chronic HIV-1 infection. J Infect Dis. 2005;191(10):1680–5. doi: 10.1086/429340. [DOI] [PubMed] [Google Scholar]
  • 161.Ide F, Nakamura T, Tomizawa M, Kawana-Tachikawa A, Odawara T, Hosoya N, et al. Peptide-loaded dendritic-cell vaccination followed by treatment interruption for chronic HIV-1 infection: a phase 1 trial. J Med Virol. 2006;78(6):711–18. doi: 10.1002/jmv.20612. [DOI] [PubMed] [Google Scholar]
  • 162.Levy Y, Thiebaut R, Montes M, Lacabaratz C, Sloan L, King B, et al. Dendritic cell-based therapeutic vaccine elicits polyfunctional HIV-specific T-cell immunity associated with control of viral load. Eur J Immunol. 2014;44:2802–10. doi: 10.1002/eji.201344433. [DOI] [PubMed] [Google Scholar]
  • 163.Allard SD, De Keersmaecker B, de Goede AL, Verschuren EJ, Koetsveld J, Reedijk ML, et al. A phase I/IIa immunotherapy trial of HIV-1-infected patients with Tat, Rev and Nef expressing dendritic cells followed by treatment interruption. Clin Immunol. 2012;142(3):252–68. doi: 10.1016/j.clim.2011.10.010. [DOI] [PubMed] [Google Scholar]
  • 164.Garcia F, Plana M, Climent N, Leon A, Gatell JM, Gallart T. Dendritic cell based vaccines for HIV infection: The way ahead. Hum Vaccin Immunother. 2013;9(11):2445–52. doi: 10.4161/hv.25876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Spaans JN, Routy JP, Tremblay C, Singer J, Bernard N, Gurunathan S, et al. Optimizing the efficiency of therapeutic HIV vaccine trials: a case for CTN 173. Trials Vaccinol. 2012;1:21–6. [Google Scholar]
  • 166.Pantaleo G, Levy Y. Vaccine and immunotherapeutic interventions. Curr Opin HIV AIDS. 2013;8(3):236–42. doi: 10.1097/COH.0b013e32835fd5cd. [DOI] [PubMed] [Google Scholar]
  • 167.Crowe SM, Carlin JB, Stewart KI, Lucas CR, Hoy JF. Predictive value of CD4 lymphocyte numbers for the development of opportunistic infections and malignancies in HIV-infected persons. J Acquir Immune Defic Syndr. 1991;4(8):770–6. [PubMed] [Google Scholar]
  • 168.Persaud D, Luzuriaga K, Ziemniak C, Muresan P, Greenough T, Fenton T, et al. Effect of therapeutic HIV recombinant poxvirus vaccines on the size of the resting CD4+ T-cell latent HIV reservoir. AIDS. 2011;25(18):2227–34. doi: 10.1097/QAD.0b013e32834cdaba. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Herasimtschuk A, Downey J, Nelson M, Moyle G, Mandalia S, Sikut R, et al. Therapeutic immunisation plus cytokine and hormone therapy improves CD4 T-cell counts, restores anti-HIV-1 responses and reduces immune activation in treated chronic HIV-1 infection. Vaccine. 2014;32(51):7005–13. doi: 10.1016/j.vaccine.2014.09.072. [DOI] [PubMed] [Google Scholar]
  • 170.Patterson BK, Carlo DJ, Kaplan MH, Marecki M, Pawha S, Moss RB. Cell-associated HIV-1 messenger RNA and DNA in T-helper cell and monocytes in asymptomatic HIV-1-infected subjects on HAART plus an inactivated HIV-1 immunogen. AIDS. 1999;13(13):1607–11. doi: 10.1097/00002030-199909100-00002. [DOI] [PubMed] [Google Scholar]
  • 171.Casazza JP, Bowman KA, Adzaku S, Smith EC, Enama ME, Bailer RT, et al. Therapeutic vaccination expands and improves the function of the HIV-specific memory T-cell repertoire. J Infect Dis. 2013;207(12):1829–40. doi: 10.1093/infdis/jit098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Achenbach CJ, Assoumou L, Deeks SG, Wilkin TJ, Berzins B, Casazza JP, et al. Effect of therapeutic intensification followed by HIV DNA prime and rAd5 boost vaccination on HIV-specific immunity and HIV reservoir (EraMune 02): a multicentre randomised clinical trial. Lancet HIV. 2015;2(3):e82–91. doi: 10.1016/S2352-3018(15)00026-0. doi: http://dx.doi.org/10.1016/S2352-3018(15)00026-0. Epub 2015 Feb 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Rouzioux C, Richman D. How to best measure HIV reservoirs? Curr Opin HIV AIDS. 2013;8(3):170–5. doi: 10.1097/COH.0b013e32835fc619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Eriksson S, Graf EH, Dahl V, Strain MC, Yukl SA, Lysenko ES, et al. Comparative analysis of measures of viral reservoirs in HIV-1 eradication studies 399. PLoS Pathog. 2013;9(2):e1003174. doi: 10.1371/journal.ppat.1003174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Smith PL, Tanner H, Dalgleish A. Developments in HIV-1 immunotherapy and therapeutic vaccination. F1000Prime Rep. 2014;6:43. doi: 10.12703/P6-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011;473(7348):523–7. doi: 10.1038/nature10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Barouch DH, Alter G, Broge T, Linde C, Ackerman ME, Brown EP, et al. HIV-1 vaccines. Protective efficacy of adenovirus/protein vaccines against SIV challenges in rhesus monkeys. Science. 2015;349(6245):320–4. doi: 10.1126/science.aab3886. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of the International AIDS Society are provided here courtesy of Wiley

RESOURCES