Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Vaccine. Author manuscript; available in PMC 2012 Apr 12.
Published in final edited form as:
PMCID: PMC3000864
NIHMSID: NIHMS232779
PMID: 20713100

TLR-Based Immune Adjuvants

Abstract

This work describes the nature and strength of the immune response induced by various Toll-like receptor ligands and their ability to act as vaccine adjuvants. It reviews the various ligands capable of triggering individual TLRs, and then focuses on the efficacy and safety of those agents for which clinical results are available.

Introduction

The innate immune system provides the host with a rapid mechanism for detecting infection by viral, microbial and fungal pathogens. Cells of the innate immune system express pattern-recognition receptors, including Toll-like receptors, that bind conserved molecules expressed by a wide variety of infectious agents. Triggering via TLR stimulates the production of pro-inflammatory cytokines/chemokines and type I IFNs that increase the host's ability to eliminate the pathogen [1-4]. This innate immune response also supports the subsequent development of adaptive immunity, and thus can be harnessed to accelerate and enhance the induction of vaccine-specific responses [5]. This review examines the types of response elicited by various TLR ligands, focusing on the efficacy and safety of those agents currently in clinical trial.

TLR 2 (and associated TLR dimers)

General overview

TLR2 interacts with a broad and structurally diverse range of ligands, including molecules expressed by microbes and fungi. Multiple TLR2 agonists have been identified, including natural and synthetic lipopeptides (e.g. Mycoplasma fermentas macrophage-activating lipopeptide (MALP-2)), peptidoglycans (PG such as those from S. aureus), lipopolysaccharides from various bacterial strains (LPS), polysaccharides (e.g. zymosan), glycosylphosphatidyl-inositol-anchored structures from gram positive bacteria (e.g. lipoteichoic acid (LTA) and lipo-arabinomannan from mycobacteria and lipomannas from M. tuberculosis) [6]. Certain viral determinants may also trigger via TLR2 [7]. Yet uncertainty exists concerning the mechanism(s) by which TLR2 recognizes such a wide array of ligands, leading some to suggest that contamination with lipopeptides (which trigger TLR2 at picomolar levels [6]) may underlie some of the reported activity. In this context, several groups report that highly purified/synthetic peptidogylcans are unable to trigger via TLR2 (in contrast to previous claims) yet retain their ability to stimulate via Nod1/2 [8-10]. In recognition of these concerns, this review will focus on the use of lipopeptides as vaccine adjuvants, as available data suggest they represent the most relevant of the TLR2 agonists being evaluated for that purpose.

TLR2 ligands

Since TLR2 is expressed on many different cell types (including dendritic cells, macrophages and lymphocytes) the mechanisms by which bacterial lipopeptides manifest their adjuvant properties are diverse. Preclinical testing indicates that lipopeptides co-administered with or physically linked to Ag can i) induce DC maturation leading to the up-regulation of co-stimulatory signals and Ag-presenting molecules (e.g. MHC class II, CD80, CD83, IFNg, IL-12) [11-13], ii) stimulate macrophages to release cytokines (e.g. TNF, IL-1, IL-6) [14;15], iii) promote the maturation and activation of B cells leading to increased production of Ag-specific IgG and IgM Abs [16;17] and iv) boost the generation of antigen specific CD8+ T cell (CTL) responses [18-20].

Two strategies are commonly utilized to generate TLR2-dependant lipopeptide vaccines: i) conjugating bacterial lipopetides or their synthetic analogues to peptide and ii) covalently linking palmitic acid to peptide antigens. Bacterial lipopeptides are structural components of cell walls. They consist of an acylated s-glycerylcysteine moiety to which a peptide can be conjugated via the cysteine residue. The bacterial lipopeptides most frequently used as vaccine adjuvants are MALP-2 and it's synthetic analogue di-palmitoyl-S-glyceryl cysteine (Pam2Cys) or tri-palmitoyl-S-glyceryl cysteine (Pam3Cys).

The alternative approach to generating TLR2-dependant lipopeptide vaccines involves modifying the antigenic peptide with Nε-palmitoyl-lysine [21]. Pre-clinical studies of these lipidated Ag constructs show that they i) induce the maturation of DCs, increasing the production of pro-inflammatory cytokines (e.g. IL-12, TNFα, IFNg) [21;22], ii) activate B cells to increase production of IgG Abs [21;23;24] and iii) enhance the generation of Ag specific CTL responses [21;25-28]. Thus, preclinical data support the conclusion that the immunogenicity of peptide-based vaccines is significantly improved by conjugating them to either bacterial lipopetides or palmitic acid moieties.

Clinical activity

Multiple TLR2 ligands have undergone clinical testing (see Table 1). The most extensively studied was Pam3Cys linked to outer surface protein A (OspA) of B. burgdorferi (the spirochete that causes Lyme disease). This Lyme disease vaccine (LYMErix™) was tested in over 20,000 volunteers [29;30]. The induction of protective immunity correlated with the development of Abs against an epitope on the C-terminus of OspA (protective IgG titer >1,400 EIA units/ml). Three doses of the vaccine induced protective immunity in >75% of subjects, and was licensed by the FDA in 1998 for general use. The manufacturer voluntarily withdrew this product 3 years later amidst media coverage of possible autoimmune side effects which led to a decline in sales [31;32]. Of note, neither the FDA nor the CDC found a connection between the vaccine and the development of autoimmunity [31].

Table 1

Overview of published clinical trials utilizing TLR2-dependant adjuvants

TreatmentDiseaseStudy PhaseStudy populationOutcome Reference
OspA + Pam3Cys +/- alumLyme diseasePhase IIIHealthy Adults>75% elicit seroprotection additional alum had no benefit[29;30]
OspA + Pam3CysLyme diseasePhase I/IIChildren < 15 years>90% elicit protective IgG response[232;233]
(T1BT)4 + Pam3CysMalariaPhase IHealthy AdultsAll volunteers developed Ag-specific IgGs (2,750GMT), comparable to earlier study using alum /Q21[33]
Lipo-6 +/- QS-21HIVPhase I/IIHealthy AdultsHIV-specific B and T cell responses in up to 93% and 86% of volunteers, respectively, QS-21 had no benefit[38;39;234,235]
Lipo-4HIVPhase IHealthy AdultsI.d. injection required only 20% of the i.m. dose and induced CTL response in up to 52% volunteers[236]
Lipo-6HIVPhase IIHIV-infected adultsNew CD4+ and CD8+ T cell responses in 70% and 57% of patients, respectively, leading to longer HAART interruption[237;238]
Lipo-6T+ IL-2/vCP1433HIVPhase IIHIV-infected adultsNo impact on HIV-specific CD4+ T cells and only transient effect on CTL[40]
Theradigm-HBVHBVPhase-IHealthy AdultsDose dependant HBV-specific CTL response persisting for > 9 months[35;36]
Theradigm-HBVHBVPhase IIHBV-infected adultsNo effective CTL response[37]
HPV-16 E7HPVPhase ICervical/vaginal cancer patientsNo effective CTL response[239]

OspA; outer-surface protein A, Pam3Cys; tri-palmitoyl-S-glyceryl cysteine, (T1BT)4; repeat T and B cell epitopes + universal T cell epitope of the P. falciparum CS protein, Lipo-4; mixture of 4 Nef/Pol/Nef lipopeptides, Lipo-6; mixture of 6 Nef/Gag/ Env lipopeptides, Lipo-6T; mixture of 6 Nef/Gag/Pol lipopeptides, vCP1433; recombinant canarypox vector, Theradigm-HBV; contains a HTL epitope (tetanus toxin peptide) covalently linked to a CTL epitope (HBV core peptide) palmitoylated at the N terminus, HPV-16 E7; lipidated HPV-16 E7 peptide epitope linked to PADRE (HTL epitope)

Pam3Cys was also tested as an adjuvant in combination with a peptide vaccine targeting malaria. The vaccine contained multiple B cell epitopes plus a universal T cell epitope derived from the Plasmodium falciparum circumsporozoite protein (CSP) [33]. Following three immunizations, all volunteers (N = 10) developed detectable levels of peptide specific IgG Abs with titers ranging from 160 to 20,240 (2,750 GMT). The addition of Pam3Cys resulted in the production of epitope-specific IgG1, IgG3 and IgG4 Abs, while an earlier study using alum and QS21 as adjuvants induced only IgG1 and IgG3 isotypes [34]. The peptide-specific IgG titers and the CSP responses induced by the vaccine formulated with Pam3Cys or alum/QS21 were similar.

A palmitic acid conjugated vaccine was recently tested in subjects with chronic Hepatitis B infection. That vaccine (Theradigm-HBV) consisted of a CTL peptide from the HBV core antigen plus a helper T lymphocyte (HTL) peptide which was palmitoylated at the N-terminus [35]. The addition of palmitic acid significantly improved the immunogenicity of the CTL and HTL epitopes which were otherwise poorly immunogenic. In a phase I trial involving 26 healthy volunteers, four doses of this vaccine induced a dose-dependent HBV-specific CTL response that persisted for more than 9 months [36]. However CTL responses were not induced in patients chronically infected with HBV [37].

Vaccines targeting HIV using palmitic acid extended peptide antigens were also evaluated in phase I and II trials [38]. Those vaccines contained a mixture of up to six different HIV Ags (including Nef, Gag and Env) that were modified to express a single palmitoyl chain at their C terminus to serve as an endogenous adjuvant. After 4 immunizations, 93% of volunteers generated IgG Abs and 86% showed a specific CTL response (Ag alone had no effect). Both responses persisted for at least 2 years in a majority of participants, indicating that long-lasting memory was generated [39]. Unfortunately, this vaccine failed to boost HIV-specific CTL responses in HIV infected subjects (N = 43) [40].

The TLR2 ligands described above are easily incorporated during peptide synthesis, yielding a well-defined and totally synthetic vaccine. These lipid adjuvants consistently boosted both humoral and cell mediated responses to peptide-based vaccines in normal healthy volunteers. The magnitude and duration of the Ab responses suggest that they would be sufficient to prevent many infectious diseases. However the ability of such vaccines to promote CTL responses was less impressive, nor were they effective in post-exposure settings. Thus, their utility for the prevention/treatment of chronic infectious diseases (such as HPV or HIV) is uncertain.

Safety

Clinical trial results indicate that lipopetide vaccines are generally safe. The LYMErix™ vaccine was well tolerated in clinical trials involving over 20,000 individuals followed for up to 6 years. Local adverse events, in particular redness and/or swelling at the injection site, were mild to moderate and generally lasted only 2-3 days. Less than 4% of the vaccinees reported systemic complaints, primarily myalgia or fever, and no hypersensitivity reactions or abnormal laboratory results were observed [29;30;41]. By 2001, over 1.4 million doses of LYMErix™ had been administered in the United States. The Vaccine Adverse Events Reporting System database included 905 reports of mild self-limiting reactions and 59 reports of arthritis associated with vaccination [42].

The safety of LYMErix™ differed somewhat from the effect of lipopetide vaccines used in anti-HIV trials. A meta-analysis of 10 such trials involving a total of 200 healthy volunteers and 48 HIV infected subjects showed that 18 individuals (including a number of normals) experienced grade 3 systemic events related to vaccination, including asthenia, fever, headache and arthralgia [38], a reactogenicity profile that raises concern about the safety of this form of adjvuant.

TLR3

General overview

TLR3 is expressed within the endosomal compartment of conventional dendritic cells and macrophages and is present on the surface membrane of non-immune cells including epithelial cells [2]. TLR3 is triggered by double-stranded RNA (dsRNA) produced during the replication of most viruses. The interaction of TLR3 with dsRNA initiates a TRIF-dependent signaling cascade that progresses through the activation of NF-kB, MAP kinases and IRF3 and culminates in the production of inflammatory cytokines and type I IFNs [43;44]. Thus, TLR3 is distinct from all other TLRs in that it does not utilize the MyD88-dependent pathway for signaling.

A potential advantage of TLR3 agonists is that triggering via this signaling cascade facilitates Ag cross-presentation, in which CD8+ T cells are primed by exogenous Ag presented in the context of MHC class I molecules, improving the generation of cytotoxic T cells [45;46]. Cross-presentation is also enhanced by the type I IFNs induced following TLR3 stimulation [47]. Thus, TLR3 ligands are considered excellent candidates as adjuvants for vaccines targeting the induction of a strong cellular immune response [48].

TLR3 ligands and their immunogenicity

Poly I:C (polyriboinosinic:polyribocytidylic acid) is a synthetic analog of dsRNA and the archetypal TLR3 ligand [49]. Since poly I:C interacts with additional receptors (including retinoic acid-inducible gene I, melanoma differentiation-associated gene 5 and double-stranded RNA-dependent protein kinase), it's adjuvanticity cannot be uniquely ascribed to TLR3 activation [50;51]. In a murine model of influenza virus infection, intranasal administration of poly I:C with an HA-based influenza vaccine induced a strong IgA anti-HA response in the nasal mucosa and IgG response in serum, whereas vaccination without poly I:C had little effect. The addition of poly I:C protected mice from lethal nasal or pulmonary viral challenge [52]. Other studies in murine models show that poly I:C can enhance the efficacy of peptide-based cancer vaccines by promoting tumor specific T cell responses [53-56].

Despite promising findings in mice, poly I:C had limited success inducing IFNs or mediating anti-tumor activity in primates (including human) due to degradation by serum nucleases [57;58]. Higher doses of poly I:C caused severe safety problems, including shock, renal failure and coagulopathies in phase I-II clinical trials of cancer patients [59]. To improve the activity and safety of TLR3 ligands, derivatives of poly I:C were produced, of which poly ICLC and poly I:C12U (also known as Ampligen®) are the most widely studied. Although clinical trials involving these adjuvants have been initiated, no published data on their activity and safety is available.

Poly ICLC is a synthetic double-stranded polyriboinosinic-polyribocytidylic acid stabilized with poly-L-lysine carboxymethyl cellulose. It is 5 -10 times more resistant to hydrolysis by primate serum than poly I:C [60]. In a murine CNS tumor model, poly ICLC improved IFNg production, enhanced CTL responses by 2-4 fold and improved survival 2-fold vs vaccine alone [61]. A Study of human papillomavirus (HPV) infection in rhesus macaques showed that administering HPV antigen plus poly ICLC increased IFNg secreting cell numbers by 2-fold and enhanced HPV-specific CD4+ T cell responses by 5-fold when compared to vaccine alone [62]. Inclusion of poly ICLC boosted the titer of HPV-binding Ab by up to 1,000-fold.

Synthetically modifying poly I:C to introduce mismatched bases (uracil and guanine) generates TLR3 ligands whose immunostimulatory potential is similar to poly I:C, but with less toxicity [63]. Poly I:C12U is a representative example of such synthetic mismatched dsRNA. In pre-clinical studies, poly I:C12U stimulated human monocyte-derived DC to mature and produce IL-12 (and decrease their production of IL-10) [64]. These activated DC promoted antigen-specific CTL responses and the differentiation of CD4+ T cells toward a Th1 phenotype [65]. In an influenza virus H5N1 infection model, intranasal administration of H5N1 vaccine with poly I:C12U increased antigen-specific IgA production by nearly 3-fold [66]. Viral titers following challenge were significantly reduced, and nearly all mice were rescued from lethal H5N1 virus infection.

Safety

Initial clinical studies involving poly ICLC showed that high dose therapy was associated with mild to severe side effects including hypotension, fever, and anemia. Once the dose and frequency of delivery were optimized, this TLR3 agonist was well tolerated in humans [67-74]. Several phase I-II clinical trials evaluating the activity and safety of poly ICLC as a cancer vaccine adjuvant are ongoing.

Poly I:C12U was well-tolerated in several long-term phase I-II clinical trials involving cancer patients [75]. This reduced toxicity when compared to poly I:C may reflect the accelerated hydrolysis and shorter plasma half-life of poly I:C12U [75;76]. Available data suggest that this agent is the most promising TLR3 ligand under clinical development. Studies of poly I:C12U as monotherapy against viral infection and phase II and III clinical trials examining their use in HIV and chronic fatigue syndrome are underway [77;78]. Use of this TLR3 ligand as a vaccine adjuvant in humans is less advanced, with a phase I clinical trial in patients with ovarian cancer scheduled to begin in the near future [65].

TLR4

General overview

TLR4 is expressed by cells of the innate immune system, including conventional dendritic cells and macrophages. It is also expressed by many non-immune cells including fibroblasts and epithelial cells [2;79;80]. Triggering via TLR4 induces a signaling cascade that utilizes both the MyD88- and TRIF-dependent pathways, leading to NF-kB and IRF3/7 activation, respectively [2;81]. Among TLRs, only TLR3 and TLR4 stimulate the production of type I IFNs via TRIF [2;81]. TLR4 activation typically induces robust IL-12p70 production and strongly enhances Th1 type cellular and humoral immune responses [82;83].

TLR4 ligands

A diversity of ligands reportedly interact with TLR4, including lipopolysaccharides (LPS), mannans (Candida albicans), glycoinositolphospholipids (Trypanosoma), viral envelope proteins (RSV and MMTV) and endogenous antigens including fibrinogen and heat-shock proteins [2;81]. LPS, which is found in the outer membrane of gram negative bacteria, is the most widely studied of the TLR4 ligands, and virtually all clinical trials involving TLR4 adjuvants examine derivatives of LPS. LPS is a complex molecule, and it is the lipid A portion composed of polyacylated diglucosamine lipids that mediates interactions with TLR4 [84;85]. Although the immunostimulatory capacity of LPS has been known for decades, the intact molecule is highly toxic, preventing it's use as a vaccine adjuvant [86]. Fortunately, the monophosphoryl lipid A (MPLA) component of LPS (purified from the cell wall of Salmonella minnesota R595 and detoxified by mild hydrolytic treatment) is considerably less toxic yet maintains immunostimulatory activity [87-89]. Studies in rabbits, guinea pigs, dogs and horses show that MPLA is 1,000-fold less toxic than LPS, a finding that paved the way towards clinical trials of TLR4 agonists [90].

Clinical activity

Numerous clinical studies examining the adjuvant activity of TLR4 ligands combined with vaccines targeting a wide variety of pathogens and tumor antigens have been conducted (Table 2). In general, adding MPLA to vaccines typically boosted serum Ab titers by 10 - 20 fold when compared to vaccine alone (Table 2). MPLA preferentially induces the production of IgG2a Abs and supports the generation of Th1 immunity in mice [91;92]. Human vaccine trials indicate that MPLA has a safety profile similar to that of alum [82;92]. Even very high doses of MPLA (up to 100 mg/m2 delivered i.v.) are safe, inducing neither renal nor hepatic toxicity [93].

Table 2

Overview of published clinical trials utilizing TLR4-dependant adjuvants

A. Infectious Diseases
TreatmentDiseaseStudy PhaseStudy populationOutcome Reference
AS04 + HbsAgHepatitis BPhase III>1,300 healthy adultsNearly 100% seroprotection and 2-fold higher Ab titers than control vaccine[102]
AS04 + HbsAgHepatitis BPhase II165 hemodialysis patientsSeroprotection elicited more rapidly and effectively[103;240]
AS02 (MPL + QS21) + rHBVHepatitis BPhase I/II30 healthy adultsInduced strong Ab and CTL response[241]
AS02 (MPL + QS21) + HbsAgHepatitis BPhase I20 liver transplant recipients80% of patients had a strong Ab response allowing them to suspend HBIG therapy[242]
RC-529 + alum + HbsAgHepatitis BPhase III285 healthy adultsSeroprotection at day 90 was 99% for RC-529 and 84% for controls (p<0.0001)[117]
AS04 + HPV-16/18 VLPsGenital HPVPhase III>18,500 healthy women93% prophylactic efficacy against HPV-induced intraepithelial neoplasias
AS04 + HPV-16/18 VLPsGenital HPVPhase III>650 healthy women100% seropositivity after 3 doses[108]
AS04 + HPV-16/18 VLPsGenital HPVPhase II>1,100 healthy women100% protection for 1 yr, remained seropositive for >7 yr[105;106;243;244]
AS04 + HPV-16/18 VLPsGenital HPVPhase II129 healthy womenAb titer higher (p<0.05) for up to 2 yr[104]
AS04 + HSV glycoprotein DGenital HSVPhase III>2,700 healthy adults74% protection for HSV-1/2 seronegative subjects[245]
AS04 + gp350EBVPhase II181 healthy adults99% with anti-gp350 Abs for >18 mos[246]
AS04 + LEISH-F1LeishmaniasisPhase I/II80 healthy adults
An external file that holds a picture, illustration, etc.
Object name is nihms-232779-t0001.jpg
[247]
AS02 (MPL + QS21) + NefTat / gp120W61DHIVPhase I84 healthy adultsHIV-specific lymphoproliferation[248]
AS02 (MPL + QS21) + RTS,SMalariaPhase II306 healthy men71% efficacy against infection for 9 wk[249]
AS02 (MPL + QS21) + RTS,SMalariaPhase II>2,000 healthy childrenInfection reduced 29% for 21 mos[250;251]
AS02 (MPL + QS21) + Mtb72fTuberculosisPhase I12 healthy adults
An external file that holds a picture, illustration, etc.
Object name is nihms-232779-t0002.jpg
[252]
B. Cancer
Treatment Disease Study Phase Study population Outcome Reference
Detox adjuvant + lysates from two melanoma cell linesMelanomaPhase II139 melanoma patientsClinical response rates were CR 3%, PR 5%, MR 4%, and PD 65%[111]
Detox adjuvant + lysates from two melanoma cell linesMelanomaPhase III140 melanoma patientsNo difference in response rates or survival[253]
Detox adjuvant + lysates from two melanoma cell linesMelanomaPhase III553 melanoma patients5-year relapse-free survival rate was 83% in patients who matched more than 2 of the M51[254]
Detox adjuvant + Stn-KLHBreast cancerPhase II23 breast cancer patientsAll patients developed Abs to STn[255]
MPL + BLP25 + three lipids2Lung cancerPhase II171 NSCLC patientsMedian survival time extended to 17.4 vs 13.0 months (p=0.065)[256]
MPL + BLP25 + three lipids2Prostate cancerPhase II16 prostate cancer patients50% had stable PSA[257]
MPL + QS21) + MAGE-3 proteinMAGE-3 positive metastatic cancerPhase I/II57 cancer patients96% developed anti-MAGE-3 Abs[258;259]
AS02 (MPL + QS21) + MAGE-3 proteinMAGE-3 positive lung cancerPhase II17 NSCLC patientsElicited strong MAGE-3 specific Ab and CD4+ T-cell response[260]

MPL; monophosphoryl lipid A, alum; aluminum hydroxide or aluminum phosphate, LEISH -F1; recombinant Leishmania polyprotein (formerly known as Leish-111f), NefTat; comprised of Nef (derived from HIVLAI) and Tat (derived from HIVBH10), RTS,S; fusion protein of the carboxyl-terminal half of the P. falciparum (strain NF54, laboratory clone 3D7) circumsporozoite protein, Mtb72f; recombinant protein comprising two antigens (Mtb39a and Mtb32a) expressed in M. tuberculosis and in BCG,

Extensive clinical studies were conducted using MPL® (also known as AS04), consisting of MPLA absorbed onto aluminum hydroxide or aluminum phosphate [94;95]. The combination of AS04 plus hepatitis B vaccine (FENDrix®) was studied in immunocompromised individuals (including the elderly, those with immunodeficiency diseases and patients on hemodialysis) [96;97]. FENDrix® was well tolerated and induced higher seroprotection rates and Ab titers than Engerix-B® ( the licensed HBV vaccine) in multiple clinical studies [98-102]. In a phase III study [102], the safety and efficacy of 2 doses of FENDrix® was compared to 3 doses of Engerix-B® in >1,300 healthy individuals. One month after the first injection, the seropositivity rate (anti-HBs titer >1 mIU/ml) was 76.8% in the FENDrix® group versus 37.3% in the Engerix-B® group, and seroprotection rates (titer >10 mIU/ml ) were 34.1% versus 13.1%, respectively. After the final vaccination, 98.5% of FENDrix® subjects were seroprotected vs 96.8% in the Engerix-B® group. The GMTs elicited by FENDrix® were more than two-fold higher than those elicited by Engerix-B®, a difference that persisted through 36 months [97].

In hemodialysis patients, seroprotection was achieved in 91% of subjects immunized with 4 doses of FENDrix® vs 84% in recipients of Engerix-B® [103]. This differences persisted at 36 months post vaccination (73% versus 52%, respectively). Antibody concentrations in the FENDrix® group were higher at all time points, and the vaccine was well tolerated, resulting in licensure by the EU in 2005.

AS04 was also used as an adjuvant with Cervarix®, a conjugate vaccine that uses virus-like particles to induce immunity against HPV-16 and HPV-18 [94]. When compared to the same vaccine adjuvanted with alum, Cervarix® induced significantly higher anti-HPV16 and anti-HPV18 L1 Ab responses (1.6 - 3.2 fold, p<0.05) at all time points [104]. 3.5 years after Cervarix® vaccination, Ab titers were 17 - 30 fold higher than those induced by natural HPV16 or HPV 18 infection [105;106]. The efficacy of Cervarix® was examined in randomized phase II and III clinical studies. In the phase II trial, Cervarix® provided 100% protection against HPV-16/18 infection for >12 months [105]. In the phase III trial (which included women with and without previous HPV exposure), the vaccine reduced the frequency of HPV-associated cervical intraepithelial neoplasia grade 2 by 92.9% [107]. Additional Phase II/III clinical data established the safety and efficacy of Cervarix®, and this AS04-adjuvanted vaccine was approved for use in the EU and the US [107][108;109].

AS04 has also been studied in combination with cancer vaccines. Melacine® consists of the lysates from two allogeneic melanoma cell lines combined with DETOX®, an adjuvant containing AS04 plus cell wall material from Mycobacterium phlei [110]. This vaccine was tested in a phase II multi-center clinical trial of melanoma patients with stage III/IV disease [111]. 12% of participants showed some response to immunotherapy, and Melacine® was approved for the treatment of metastatic melanoma by the Canadian FDA [112]. AS04 is also a component of Stimuvax®, a complex mixture of lipopeptides, proteins, and lipids used in patients with adenocarcinomas including non-small cell lung cancer [113;114]. In a phase II trial, Stimuvax® improved median survival time by ≈33% (p=0.069) without inducing significant toxicity. A phase III trial to confirm these findings has been initiated [115].

AS04 is heterogenous, in that it contains several different MPLA species that vary in length and degree/type of fatty acid acylation. Building upon the success of MPLA/AS04, a new class of synthetic lipid A mimetics, the aminoalkyl glucosaminide 4-phosphates (AGPs) was developed [83;92][116]. In contrast to the complex mixture of lipid A congeners found in MPLA, AGPs can be synthesized at high purity as single chemical entities and modified to alter biological/pharmacodynamic activity. RC-529 (also known as Ribi.529) is a leading AGP that is structurally similar to the hexa-acyl component of MPL®. RC-529 was co-administered with the HBV vaccine (Supervax®) and compared to Engerix-B®. Supervax® induced significantly greater seroprotection, achieving 99% seroprotection at 3 months vs 84% for the control group (p< 0.0001). Anti-HBV titers were significantly higher at all time points for the Supervax® group. Based on these findings and an acceptable safety profile, the RC-529 conjugated HBV vaccine was approved for use in Argentina [117] .

Safety

In general, adverse events (typically mild - moderate) were more frequent in subjects receiving AS04 or RC-529 adjuvanted vaccines vs alum. In studies of FENDrix®, the frequency of pain, erythema and swelling exceeded that of Engerix-B®, [102]. The safety of the AS04 adjuvanted HPV-16/18 vaccine was summarized in a meta-analysis of 11 clinical trials. Local and systemic adverse events were higher in recipients of the AS04 vs alum adjuvanted vaccine. However compliance with the three-dose schedule did not differ between groups nor did the rates of serious adverse events (2.8% vs 3.1%), medically significant conditions (19.4% vs 21.4%), new onset of chronic diseases (1.7% for both) or new onset of autoimmune diseases (0.4% vs 0.3%) [118]. The safety profile of this TLR4 adjuvant was sufficiently to allow licensure in vaccines used in the US, Europe and Argentina.

In clinical studies of Supervax®, the incidence of injection site pain was significantly increased when compared to the alum-adjuvanted HBV vaccine (70% vs 42%, p<0.0001). These local reactions were typically mild to moderate, and resolved within 3 days. The incidence of systemic adverse events was low, and did not differ significantly between groups. No serious systemic adverse events related to vaccination were reported.

TLR5

General overview

TLR5 is triggered by a region of the flagellin molecule expressed by nearly all motile bacteria [119;120]. TLR5 is found on the surface of many types of immune cells, including monocytes, mDCs, Langerhans cells, T cells and NK cells [121-126]. When used as an adjuvant, flagellin is typically fused to a recombinant vaccine Ag. In that form, flagellin directly induces DC maturation, triggering the up-regulation of co-stimulatory signals and Ag-presenting molecules (CD80, CD83, CD86, MHC class II, TNFα, IL-8, IL-1β, CCL2, CCL5) [125]. The effect of fusing flagellin to protein-based vaccines was examined using the fluorescent protein EGFP as a model system. Almost 50% of APCs internalized flagellin-EGFP vs 3% internalizing EGFP alone. Flagellin-EGFP also stimulated APCs to produce 20-fold more TNFα than EGFP (p<0.001), and uniquely induced Ag-specific CTL responses in vivo [127]. When used as an adjvuant, flagellin stimulates monocytes to produce the cytokines IL-10 and TNFa [123], NK cells to produce IFNg and α-defensins, and T cells to proliferate and produce cytokines and chemokines (e.g. IL-10, IL-8 and IFNg) [122].

TLR5 ligands and their pre-clinical activity

The adjuvant properties of flagellin-Ag complexes were investigated in several animal models. When coupled to ovalbumin, flagellin induced an IgG Ab response 10-fold higher than OVA co-administered with alum [128]. Fusing the poorly immunogenic M2e protein from the influenza virus with recombinant Salmonella Typhimurium flagellin (STF2) yielded a vaccine that induced a 10-fold higher Ag-specific IgG response than M2e formulated in alum [129]. Other studies showed that flagellin-Ag vaccines induced protective Ab responses against Yersinia pestis [130], West Nile virus [131], vaccinia virus [132] and Pseudomonas aeruginosa [133;134]. These protective responses were characterized by high titered Ag-specific IgG (IgG1, IgG2a) responses that were generally two logs higher than Ag alone or Ag + flagellin (not fused) (p<0.05). Since TLR5 is not expressed on B cells [124]) this enhanced Ab production was presumably mediated by improved APC function.

There is no published information on the immunogenicity or safety of flagellin-based vaccines in humans, although several clinical trials are ongoing. No injection site inflammation or severe systemic adverse effects were detected in studies of mice and non-human primates [130;135].

TLR7/8

General overview

TLRs 7 and 8 are phylogenetically and structurally related [136]. Both recognize single stranded (ss) RNA sequences containing poly-U or GU-rich sequences and are activated by synthetic imidazoquinolines including imiquimod (R-837) and resiquimod (R-848) as well as by guanosine analogues such as loxoribine [136-139]. TLR 7/8 molecules are localized to the endosomal compartments of human immune cells including DCs, monocytes, macrophages, lymphocytes, Langerhans cells, and NK cells [124;140;141]. Because of their numerous similarities, TLRs 7/8 will be handled together in this section.

The interaction between TLRs 7/8 and their cognate ligands activates DC to i) enhance expression of co-stimulatory molecules (e.g. CD80, CD86, CD40) [142], ii) migrate [143] and iii) produce pro-inflammatory cytokines including IFNα, TNFα and/or IL-12 (the later facilitating induction of Th1-type responses). In this context, TLR7-specific activation preferentially triggers IFNα secretion by pDC whereas TLR8-specific activation preferentially induces IL-12 production by mDCs [140;144]. TLR 7/8 also promote the maturation of Langerhans cells and their migration from the skin to the lymph nodes [145;146].

In addition to activating DCs, TLR 7/8 stimulate B cells to secrete Ig and produce cytokines (e.g. IL-6, TNFα) [147] while triggering NK cells to produce IFNg [148]. TLR 7/8 ligation stimulates T cells to proliferate and produce IFNg, IL-2 and IL-10. Memory T cells are particularly sensitive to this form of TLR-mediated activation [122], and TLR7/8 ligands may reduce the immunosuppression mediated by CD4+ T regulatory cells [149].

TLR7/8 ligands and their pre-clinical activity

Most pre-clinical studies involving TLR7/8 agonists utilized imiquimod (which predominantly activates TLR7) or resiquimod (which triggers both TLR7 and 8). Compounds that selectively activate TLR7 (852A) or TLR8 (3M-002) [140;150] have been identified but there is insufficient published information to assess their utility as vaccine adjuvants. It is currently unclear whether ligands targeting TLR7 vs TLR8 will be superior vaccine adjuvants. Resolving this issue through pre-clinical studies is complicated, as there is disagreement in the literature concerning the ability of mice to respond to TLR8 ligands that are active in Man [136;151-153]. Studies utilizing monocytes and APCs collected from human newborns and adults indicate that the Th1-response induced by TLR8 stimulation substantially exceeds that induced by other TLR ligands (including TLR7). This finding raises the possibility that TLR8 ligands might be of particular utility in vaccines targeting newborns [154;155].

Pre-clinical studies indicate that imidazoquinolines can improve both the magnitude and quality of Ag-specific T cell and Ab responses. In mice and macaques, i.m. and s.c. delivery of TLR7/8 agonists with the HIV-1 Gag protein enhanced Ag-specific IgG and CTL responses, particularly when the adjuvant was conjugated to Ag [156-159]. In a guinea pig model of genital HSV infection, an HSV-2 glycoprotein based vaccine combined with imiquimod enhanced Ab production by up to 10-fold (p <0.001) and reduced the number of lesions and frequency of genital HSV recurrences by >80% (p<0.001). Interpretation of these findings was complicated by the observation that imiquimod alone reduced HSV recurrences by 62%, so that no significant reduction for the vaccine + imiquimod vs imiquimod alone groups was observed [160].

TLR7/8 agonists have also been used as adjuvants in combination with cancer vaccines. In a genetically engineered mouse model, a DNA vaccine encoding HER2/neu adjuvanted with imiquimod significantly delayed the development of spontaneous mammary tumors and reduced their incidence by 65% when compared to the DNA vaccine alone. These effects were accompanied by a significant increase in Ag-specific Ab production (3 fold, p <0.05), a switch from IgG1 to IgG2a Ab isotype, and a 30% increase in CTL activity (p <0.05) [161].

Clinical activity

Imiquimod (Aldara, 3M) is the only TLR7/8 agonist to undergo extensive clinical testing (Table 3). It is licensed for topical use to treat warts caused by HPV, basal cell carcinoma, and actinic keratosis [162-165]. Topical imiquimod was tested in combination with several cancer vaccines (Table 3). When used with a vaccine composed of several melanoma peptides plus Flt3 ligand (hematopoietic growth factor), imiquimod increased the fraction of patients who developed peptide-specific CTL responses. However, no effect on disease progression was observed in this study of 27 patients with stage II-IV post surgical disease [166].

Table 3

Overview of published clinical trials utilizing TLR7/8-dependant adjuvants

TreatmentDiseaseStudy PhaseStudy populationOutcome Reference
K562 cells transfected with GM-CSF +/- imiquimodCMLPhase IIPatients with CMLReduction of tumor burden and improved molecular response. Additional imiquimod had no benefit.[261]
HPV16 E6E7L2 fusion protein + imiquimodVulval cancerPhase IIPatients with VIN (grade II/III) HPV+/-63% were lesion responders with local CD4+ and CD8+ T cell infiltration. No control group without imiquimod included.[262]
Prostate specific peptides + montanide +/- imiquimodProstate cancerPhase I/IIPatients with PC (stage II/III)Imiquimod-adjuvanted vaccine slowed the PSA rise better than other adjuvants.[168]
NY-ESO-1 + imiquimodMelanomaPhase IPatients with stage IIB-III melanoma50% developed Ag-specific Ab and/or CD4+ responses. No CD8+ responses.[167]
melanoma peptides (Tyr, Mel, Ny-ESO-1) +Flt3/ +/- imiquimodMelanomaPhase IPatients with stage II-IV melanoma62.5% of patients developed Ag-specific CD8+ T cells when imiquimod was applied, in contrast to 25% without imiquimod.[166]

1 M5; any of 5 HLA class I Ags (HLA-A2, HLA-A28, HLA-B44, HLA-B45, and HLA-C3), 2 the three lipids are cholesterol, dimyristoyl phosphatidylglycerol and dipalmitoyl phosphatidylcholine, Detox adjuvant; includes monophosphoryl lipid A and cell wall skeleton from Mycobacterium phlei, STn-KLH; STn is a cancer-associated core region carbohydrate antigen of epithelial mucin and STn linked to keyhole limpet haemocyanin (KLH), BLP25; 25-amino acid sequence that provides MUC1 antigen, NSCLC; non small cell lung cancer, BSC; best supportive care, PSA; prostate specific antigen, MAGE-3; melanoma antigen-3, PC; prostate carcinoma, VIN; vulval intraepithelial neoplasia, PSA ; prostate specific antigen, NY-ESO-1; cancer-testis Ag, Flt3; hematopoietic growth factor.

Another vaccine used the NY-ESO-1 protein in combination with topically administered imiquimod for the treatment of malignant melanoma (stage II-III). Applying imiquimod cream to the vaccine site stimulated novel Ab and/or CD4+ T cell responses in approximately half of the trial participants. Biopsies of imiquimod treated skin showed a significant enhancement of mononuclear cell infiltrates including T cells, APCs and NK cells vs untreated skin [167].

A phase I-II trial compared topical imiquimod to 3 other adjuvants (GM-CSF, hyperthemia, mucin-1-mRNA/protamine complex ) in combination with a multi-peptide vaccine (HLA-A2 restricted TAA-eptiopes and MHC class II-binding peptides) in prostate cancer patients. Recipients of the imiquimod-adjuvanted vaccine showed the best response, as determined by a slowing in the rate of PSA rise. Unfortunately, this study did not evaluate immunological markers or disease progression, so no definitive conclusions can be drawn [168].

In summary, preclinical studies suggest that TLR7/8 ligands can boost both humoral and cell mediated responses to vaccines targeting infectious diseases and cancer. Only imiquimod was clinically evaluated as a vaccine adjuvant, where it boosted Ag-specific Ab and CD4 responses in cancer patients, although it is unclear whether this impacted disease progression.

Safety

Topical administration of imiquimod in human vaccine trials was well tolerated. Adverse events were uniformly mild, transient, and primarily involved local reactions (such as cutaneous erythema). No severe systemic events were reported. These findings are consistent with large clinical trials in which cutaneous imiquimod cream was used to treat diseases such as actinic keratosis [165].

In contrast, severe side effects were reported after oral or systemic use of imidazoquinoline as a monotherapy in humans [169-171]. In a phase I clinical trial of cancer patients, oral administration of 25 - 200 mg of imiquimod was associated with sustained dose-related hematological toxicity. At doses ≥50 mg, grade 3-4 lymphopenia developed in nearly half of all patients. At higher doses, hepatic and renal impairment were found [171]. Thus while safe for topical use, formulating imiquimod with vaccines designed for internal use is likely to be problematic.

TLR 9

General overview

TLR9 is expressed by human B cells and pDC. The receptor is localized within endo-lysosomal compartments and detects the unmethylated CpG motifs present at high frequency in bacterial (but not mammalian) DNA [172]. The recognition of CpG DNA by cells expressing TLR9 has a cascading effect on the immune system, leading to the maturation, differentiation, and/or proliferation of NK cells, T cells, B cells, monocytes and macrophages [124;172-179]. The resultant immune response is characterized primarily by the production of pro-inflammatory and Th-1 biased cytokines (including IL-1, IL-6, TNFα, IFNg and IL-12 [172;173;180-184].

TLR9 ligands

The immunostimulatory activity of bacterial DNA is mimicked by synthetic oligonucleotides (ODN) expressing CpG motifs [185-187]. These are typically composed of phosphorothioate nucleotides, which are considerably more resistant to nuclease digestion than native phosphodiester nucleotides, and thus have a substantially longer half-life in vivo [188]. Three major classes of CpG ODN have been described, each with distinct structural and biological properties. “K” type ODN (also referred to as “B” type) consist of multiple CpG motifs on a phosphorothioate backbone. They are strong modulators of B cell activation and induce the maturation of pDCs and monocytes [189-191]. “D” type ODN (also referred as “A” type) are constructed of a mixed phosphodiester/phosphothioate backbone, contain a single CpG motif flanked by palindromic sequences, and a poly G tail at the 3' and 5' termini, facilitating the formation of concatamers. “D” ODN excel are triggering pDC to produce IFNα [182][190]. “C” type ODN resemble “K” type in being composed entirely of phosphorothioate nucleotides, but resemble “D” type in containing palindromic CpG motifs. They have immunostimulatory properties found in both “K” and “D” type ODN, including the ability to activate B cells and stimulate the production of IFNα by pDC [188].

Preclinical studies conducted in rodents and non-human primates demonstrate that CpG ODN can accelerate [192], increase the magnitude [192-194], and prolong the duration of vaccine-specific Ab responses [194-196]. In addition, CpG ODN improve the response induced by mucosal vaccines [197-200] and enhance the immunogenicity of vaccines administered to immunocompromised populations [201-204]. CpG ODN promote vaccine immunogenicity by improving Ag uptake by professional APC (particularly pDC), triggering the functional maturation of APC, and generating a cytokine/chemokine microenvironment supportive of Ag-specific immunity [205]. These effects are optimized when vaccine and adjuvant are presented to the immune system in close spatial and temporal proximity [193;206;207].

Clinical activity

CpG ODN as adjuvants for vaccines targeting infectious diseases

Human clinical trials examining the adjuvant activity of TLR9 ligands have focused on “K” type ODN. Those trials evaluated CpG ODN combined with vaccines designed to prevent malaria [208-211], hepatitis B (HBV) [212-217] influenza [218] and anthrax [219] (Table 4). When co-administered with Engerix-B®, CpG ODN significantly improved the Ab response of healthy individuals through 48 weeks of follow-up when compared to the conventional vaccine. The geometric mean titer of anti-HBs antibody in volunteers treated with CpG ODN plus Engerix-B® was 13-fold higher after primary and 45-fold higher after secondary immunization than that induced by vaccine alone. Moreover, all participants immunized with the CpG-adjuvanted vaccine developed seroprotective titers by 2 wk after priming [215;216], compared to none of the control group. These findings were replicated in an independent clinical trial [217]. All patients receiving the CpG adjuvanted vaccine maintained titers in the seroprotective range for >1 year compared to 63% vaccinated with Engerix-B® alone [212;213]. The adjuvanted vaccine also induced protective Ab titers in HIV patients who were hyporesponsive to Engerix-B® alone [212].

Table 4

Overview of published clinical trials utilizing TLR9-dependant adjuvants

A. Infectious Diseases
TreatmentDiseaseStudy PhaseStudy populationOutcome Reference
CpG 7909 + AMA1/AlhydrogelHMalariaPhase IHealthy Adults5.5 fold increase in anti-AMA1 Ab titer[208]
CpG 7909 + AMA1/AlhydrogelHMalariaPhase ISemi-Immune Adults2 fold increase in Ab response,[209;210]
CpG 7909 + MSP142MalariaPhase IHealthy AdultsAb titers increased 8-fold after 3 doses[211]
CpG 7909 + Engerix-B®Hepatitis BPhase I/IIHIV-infected Adults100% serocoversion after 6 wk (vs 63% in controls), seroprotection maintained 5 yr[212;213]
CpG 1018 + HBsAgHepatitis BPhase IIIHealthy young adultsSeroprotection achieved faster and and with fewer doses than Engerix-B®.[217]
CpG 7909 +Hepatitis BPhase I/IIHealthy adultsSeroprotection achieved faster, and anti-HBs Ab titers 45-fold higher post boost vs Engerix-B®.[214;215] [216]
CpG 7909 + FluarixInfluenzaPhase IHealthy adultsImmune response not enhanced, but reduced vaccine doses without reduced immunogenicity required[218]
CpG 7909 + AVAAnthraxPhase IHealthy adultsAb response increased 7-fold, and accelerated by 3 wk.[219]
B. Cancer
Treatment Disease Study Phase # of subjects/group Outcome Reference
CpG 7909 + Melan-A/MART-1MelanomaPhase I8 patients with melanoma10-fold increase in Melan-A specific CD8+ T cells (>3%)[226]
CpG 7909 +NY-ESO-1 protein +/- MontanideMelanomaPhase I8 patients with melanomaIncreased CD8+ T cell response[223]
CpG 7909 +NY-ESO-1 protein + MontanideMelanoma
Breast cancer
Sarcoma
Ovarian cancer
Phase I18 patients with melanoma
14 cancer patients by 6 wk,
Increased Ab and CD4+ Th cell response
CD8+ T cell response by 12 wk
[227]
CpG 7909 +NY-ESO-1 protein + MontanideMelanoma
Lung cancer
Ovarian cancer
Breast cancer
Phase I13 patients with melanoma cancer patientsIncreased CD8+ T cell response and prolonged survival[228]
Sarcoma
CpG 1018 + GM-CSF + hTERT peptideSarcoma
Glioblastoma
Phase I17 cancer patientsCD8+ T cell response in only 1 subject[229]

AMA1; apical membrane antigen 1, MSP142; Merozoite surface protein 142, MART; Melanoma Antigen Recognized by T-cells, hTERT; human telomerase reverse transcriptase

CpG ODN were also tested as adjuvants in combination with AVA, the licensed anthrax vaccine. The adjuvanted vaccine boosted anthrax specific Ab responses of healthy subjects by 6-8 fold and accelerated the induction of immunity by approximately 3 wk [219]. Other trials showed that naive volunteers mounted a significantly stronger Ab response to the poorly immunogenic malaria vaccine candidates Apical Membrane Antigen 1 (AMA1) and Merozoite surface protein 142 (MSP142) when co-delivered with CpG ODN. The co-administration of CpG with AMA1 increased the GMT of anti-AMA1 Abs by 5.5 fold when compared to subjects receiving AMA1 alone [208]. This enhanced Ab response was achieved using a 4-fold lower concentration of AMA1 and was persistent: at 236 days after vaccination, those immunized with AMA1 + CpG ODN maintained serum Ab titers 4.6-fold higher than those vaccinated with just AMA1 [208]. While challenge studies were not conducted, sera from volunteers vaccinated with AMA1 + CpG ODN was >4-fold more effective at inhibiting the growth of P. falciparum 3D7 parasites in vitro that those vaccinated with AMA1 alone (p <.0001). Unfortunately, the impact of adding CpG ODN was insufficient to overcome the lack of immunogenicity of AMA1 when administered to semi-immune adults with a history of multiple previous plasmodium infections and circulating AMA1-specific Abs [209]. When co-administered with MSP142, CpG ODN boosted average Ab titers by 8 fold when compared to MSP142 alone measured 2 wk after third immunization [211]. The effect of CpG ODN on a vaccine containing both AMA1 and MSP1 is underway (ClinicalTrials.gov Identifier:NCT0088961). The effect of adding CpG ODN to the Fluarix influenza vaccine was less impressive. In that trial, CpG ODN reduced the dose of vaccine required to achieve a strong immune response, but did not increase the magnitude of the response [218].

CpG ODN as adjuvants for vaccines targeting cancer

The goal of most cancer vaccines is to generate large numbers of tumor-specific CTL, as cellular rather than humoral immunity is believed to play a central role in tumor eradication. In pre-clinical animal studies, CpG ODN enhanced the production of cytotoxic CD8+ T cells targeting tumor Ags [220;221]. This effect was observed when ODN were conjugated to or simply co-administered with tumor Ag [222;223]. Of considerable interest, CpG adjuvanted tumor vaccines effectively eliminated established cancers in murine models [224;225].

These findings supported clinical studies using CpG ODN as adjuvants for cancer vaccines (Table 4). In a trial examining the effect of a melanoma Ag A based vaccine (Melan-A; identical to MART-1), inclusion of CpG ODN plus incomplete Freund's adjuvant generated a stronger and more rapid CD8+ T cell response than the unadjuvanted vaccine. In that study, 4 immunizations with the adjuvanted vaccine resulted in >3% of circulating CD8+ T cells being Melan-A-specific: an order of magnitude higher than in patients treated with vaccine alone [226].

In a phase I trial of patients with stage III/IV NY-ESO-1Bexpressing melanoma, only a weak immune response was elicited by vaccination with the NY-ESO-1 peptide plus CpG ODN. When the co-adjuvant Montamide was added to the the NY-ESO peptide plus CpG ODN, the combination promoted the development of Ag-specific CD8+ T cells within 1 month [223]. A separate uncontrolled clinical trial used recombinant NY-ESO-1 protein plus CpG ODN and Montamide to vaccinate patients with different tumor types [227]. Tumor specific Ab responses rose significantly within 6 weeks while cross-primed NY-ESO-1-specific CD8+ T cells were detected in a subset of patients by 12 weeks.

A Phase I study involving 14 patients with different types of cancer detected new Ag-specific CD8+ T cell responses in 9 patients following combined vaccination with NY-ESO-1, CpG ODN and Montanide [228]. Six of these 9 patients lived an average of 39 months, far longer than their predicted survival of only 4 months. Whether vaccine-induced immunity was responsible for this improved clinical outcome could not be determined in this uncontrolled study. In contrast, when CpG ODN was co-administered with GM-CSF and a peptide corresponding to the immunodominant epitope from the tumor antigen hTERT (human telomerase reverse transcriptase), no beneficial effect on the CTL response of patients with sarcoma or glioblastoma was detected [229].

In toto, the clinical data indicates that CpG ODN are likely to find use as vaccine adjuvants, particularly for those vaccines targeting infectious diseases. This reflects the consistent ability of CpG ODN to boost Ag-specific humoral immunity in naive subjects. The utility of CpG ODN as tumor vaccines adjuvants is less clear, as the magnitude of the CTL response needed to clear an established tumor may exceed that which can be induced by the current generation of vaccines. This is particularly true given the ability of tumor cells to suppress or circumvent immune recognition. These concerns have focused attention on the use of CpG ODN in combination with other immunomodulatory agents, an area of research that deserves further attention.

Safety

Toxicity has not been observed in animal studies of CpG adjuvanted vaccines. At much higher doses (used for other purposes), or in combination with agents that induce the production of TNFα (such as LPS or D-galactosamine) [186;230;231], toxicity has been reported. Evidence from clinical trials indicates that CpG ODN are reasonably well tolerated when administered as vaccine adjuvants. However the frequency and severity of local adverse events (injection site reactions such as pain, swelling, induration, pruritus, and erythema) and systemic symptoms (including flu-like symptoms) were elevated. This higher frequency and severity of AEs is likely attributable to the immunostimulatory properties of CpG ODN. Most of these adverse events were mild-to-moderate, appeared within 24 hours of dosing, and persisted for only a few days.

Conclusions

Toll-like receptors differ from one-another in location (intra-cellular vs plasma membrane), use of accessory molecules to induce signaling (TIRAP, TRIF, TRAM and MyD88), the type of pathogen associated molecules they recognize (nucleic acids, polypeptides, lipopolysaccharides), and the type of response they induce (inflammatory, Th1 or Th2). These distinctions underlie inherent differences in the ability of speicifc TLR ligands to influence the nature of the adaptive immune response they support when used as vaccine adjuvants.

Clinical trials involving TLRs 2, 3, 4, 7/8 and 9 support the broad conclusion that TLR ligands can be safe and effective vaccine adjuvants, with vaccines already licensed in the US, Europe and Argentina containing such ligands. Sadly, there are no head-to-head pre-clinical (much less clinical) trials examining the relative adjuvanticity of different TLR agonists. Since vaccine development is a highly empirical process, and different types of response are required to protect against distinct pathogens/tumors, there is little justification for concluding that one particular ligand will be significantly more useful than other available alternatives. However as clinical data accumulate, evidence of safety and immunogenicity may shift the balance to favor incorporation of one or small subset of TLR agonists in future vaccines.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Janeway CA, Jr., Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.. 197–216. [PubMed] [Google Scholar]
2. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006 Feb 24;124(4):783–801. [PubMed] [Google Scholar]
3. Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007 Oct 18;449(7164):819–26. [PubMed] [Google Scholar]
4. Beutler BA. TLRs and innate immunity. Blood. 2009 Feb 12;113(7):1399–407. [PMC free article] [PubMed] [Google Scholar]
5. Pasare C, Medzhitov R. Toll-like receptors and acquired immunity. Semin Immunol. 2004 Feb;16(1):23–6. [PubMed] [Google Scholar]
6. Zahringer U, Lindner B, Inamura S, Heine H, Alexander C. TLR2 - promiscuous or specific? A critical re-evaluation of a receptor expressing apparent broad specificity1. Immunobiology. 2008;213(3-4):205–24. [PubMed] [Google Scholar]
7. Barbalat R, Lau L, Locksley RM, Barton GM. Toll-like receptor 2 on inflammatory monocytes induces type I interferon in response to viral but not bacterial ligands. Nat Immunol. 2009 Nov;10(11):1200–7. [PMC free article] [PubMed] [Google Scholar]
8. Inamura S, Fujimoto Y, Kawasaki A, Shiokawa Z, Woelk E, Heine H, et al. Synthesis of peptidoglycan fragments and evaluation of their biological activity. Org Biomol Chem. 2006 Jan 21;4(2):232–42. [PubMed] [Google Scholar]
9. Travassos LH, Girardin SE, Philpott DJ, Blanot D, Nahori MA, Werts C, et al. Toll-like receptor 2-dependent bacterial sensing does not occur via peptidoglycan recognition. EMBO Rep. 2004 Oct;5(10):1000–6. [PMC free article] [PubMed] [Google Scholar]
10. Girardin SE, Boneca IG, Carneiro LA, Antignac A, Jehanno M, Viala J, et al. Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan. Science. 2003 Jun 6;300(5625):1584–7. [PubMed] [Google Scholar]
11. Hertz CJ, Kiertscher SM, Godowski PJ, Bouis DA, Norgard MV, Roth MD, et al. Microbial lipopeptides stimulate dendritic cell maturation via Toll-like receptor 2. J Immunol. 2001 Feb 15;166(4):2444–50. [PubMed] [Google Scholar]
12. Chua BY, Healy A, Cameron PU, Stock O, Rizkalla M, Zeng W, et al. Maturation of dendritic cells with lipopeptides that represent vaccine candidates for hepatitis C virus. Immunol Cell Biol. 2003 Feb;81(1):67–72. [PubMed] [Google Scholar]
13. Chua BY, Zeng W, Lau YF, Jackson DC. Comparison of lipopeptide-based immunocontraceptive vaccines containing different lipid groups. Vaccine. 2007 Jan 2;25(1):92–101. [PubMed] [Google Scholar]
14. Muhlradt PF, Schade U. MDHM, a macrophage-stimulatory product of Mycoplasma fermentans, leads to in vitro interleukin-1 (IL-1), IL-6, tumor necrosis factor, and prostaglandin production and is pyrogenic in rabbits. Infect Immun. 1991 Nov;59(11):3969–74. [PMC free article] [PubMed] [Google Scholar]
15. Muhlradt PF, Kiess M, Meyer H, Sussmuth R, Jung G. Isolation, structure elucidation, and synthesis of a macrophage stimulatory lipopeptide from Mycoplasma fermentans acting at picomolar concentration. J Exp Med. 1997 Jun 2;185(11):1951–8. [PMC free article] [PubMed] [Google Scholar]
16. Rharbaoui F, Drabner B, Borsutzky S, Winckler U, Morr M, Ensoli B, et al. The Mycoplasma-derived lipopeptide MALP-2 is a potent mucosal adjuvant. Eur J Immunol. 2002 Oct;32(10):2857–65. [PubMed] [Google Scholar]
17. Borsutzky S, Kretschmer K, Becker PD, Muhlradt PF, Kirschning CJ, Weiss S, et al. The mucosal adjuvant macrophage-activating lipopeptide-2 directly stimulates B lymphocytes via the TLR2 without the need of accessory cells. J Immunol. 2005 May 15;174(10):6308–13. [PubMed] [Google Scholar]
18. Deres K, Schild H, Wiesmuller KH, Jung G, Rammensee HG. In vivo priming of virus-specific cytotoxic T lymphocytes with synthetic lipopeptide vaccine. Nature. 1989 Nov 30;342(6249):561–4. [PubMed] [Google Scholar]
19. Jackson DC, Lau YF, Le T, Suhrbier A, Deliyannis G, Cheers C, et al. A totally synthetic vaccine of generic structure that targets Toll-like receptor 2 on dendritic cells and promotes antibody or cytotoxic T cell responses. Proc Natl Acad Sci U S A. 2004 Oct 26;101(43):15440–5. [PMC free article] [PubMed] [Google Scholar]
20. Borsutzky S, Ebensen T, Link C, Becker PD, Fiorelli V, Cafaro A, et al. Efficient systemic and mucosal responses against the HIV-1 Tat protein by prime/boost vaccination using the lipopeptide MALP-2 as adjuvant. Vaccine. 2006 Mar 15;24(12):2049–56. [PubMed] [Google Scholar]
21. Zhu X, Ramos TV, Gras-Masse H, Kaplan BE, BenMohamed L. Lipopeptide epitopes extended by an Nepsilon-palmitoyl-lysine moiety increase uptake and maturation of dendritic cells through a Toll-like receptor-2 pathway and trigger a Th1-dependent protective immunity. Eur J Immunol. 2004 Nov;34(11):3102–14. [PubMed] [Google Scholar]
22. Hosmalin A, Andrieu M, Loing E, Desoutter JF, Hanau D, Gras-Masse H, et al. Lipopeptide presentation pathway in dendritic cells. Immunol Lett. 2001 Nov 1;79(1-2):97–100. [PubMed] [Google Scholar]
23. BenMohamed L, Gras-Masse H, Tartar A, Daubersies P, Brahimi K, Bossus M, et al. Lipopeptide immunization without adjuvant induces potent and long-lasting B, T helper, and cytotoxic T lymphocyte responses against a malaria liver stage antigen in mice and chimpanzees. Eur J Immunol. 1997 May;27(5):1242–53. [PubMed] [Google Scholar]
24. Daubersies P, Thomas AW, Millet P, Brahimi K, Langermans JA, Ollomo B, et al. Protection against Plasmodium falciparum malaria in chimpanzees by immunization with the conserved pre-erythrocytic liver-stage antigen 3. Nat Med. 2000 Nov;6(11):1258–63. [PubMed] [Google Scholar]
25. Deprez B, Sauzet JP, Boutillon C, Martinon F, Tartar A, Sergheraert C, et al. Comparative efficiency of simple lipopeptide constructs for in vivo induction of virus-specific CTL. Vaccine. 1996 Apr;14(5):375–82. [PubMed] [Google Scholar]
26. Loing E, Andrieu M, Thiam K, Schorner D, Wiesmuller KH, Hosmalin A, et al. Extension of HLA-A*0201-restricted minimal epitope by N epsilon-palmitoyl-lysine increases the life span of functional presentation to cytotoxic T cells. J Immunol. 2000 Jan 15;164(2):900–7. [PubMed] [Google Scholar]
27. Coutsinos Z, Villefroy P, Gras-Masse H, Guillet JG, Bourgault-Villada I. Evaluation of SIV-lipopeptide immunizations administered by the intradermal route in their ability to induce antigen specific T-cell responses in rhesus macaques. FEMS Immunol Med Microbiol. 2005 Mar 1;43(3):357–66. [PubMed] [Google Scholar]
28. Zhang X, Issagholian A, Berg EA, Fishman JB, Nesburn AB, BenMohamed L. Th-cytotoxic T-lymphocyte chimeric epitopes extended by Nepsilon-palmitoyl lysines induce herpes simplex virus type 1-specific effector CD8+ Tc1 responses and protect against ocular infection. J Virol. 2005 Dec;79(24):15289–301. [PMC free article] [PubMed] [Google Scholar]
29. Steere AC, Sikand VK, Meurice F, Parenti DL, Fikrig E, Schoen RT, et al. Vaccination against Lyme disease with recombinant Borrelia burgdorferi outer-surface lipoprotein A with adjuvant. Lyme Disease Vaccine Study Group. N Engl J Med. 1998 Jul 23;339(4):209–15. [PubMed] [Google Scholar]
30. Sigal LH, Zahradnik JM, Lavin P, Patella SJ, Bryant G, Haselby R, et al. A vaccine consisting of recombinant Borrelia burgdorferi outer-surface protein A to prevent Lyme disease. Recombinant Outer-Surface Protein A Lyme Disease Vaccine Study Consortium. N Engl J Med. 1998 Jul 23;339(4):216–22. [PubMed] [Google Scholar]
31. Nigrovic LE, Thompson KM. The Lyme vaccine: a cautionary tale. Epidemiol Infect. 2007 Jan;135(1):1–8. [PMC free article] [PubMed] [Google Scholar]
32. Abbott A. Lyme disease: uphill struggle. Nature. 2006 Feb 2;439(7076):524–5. [PubMed] [Google Scholar]
33. Nardin EH, Calvo-Calle JM, Oliveira GA, Nussenzweig RS, Schneider M, Tiercy JM, et al. A totally synthetic polyoxime malaria vaccine containing Plasmodium falciparum B cell and universal T cell epitopes elicits immune responses in volunteers of diverse HLA types. J Immunol. 2001 Jan 1;166(1):481–9. [PubMed] [Google Scholar]
34. Nardin EH, Oliveira GA, Calvo-Calle JM, Castro ZR, Nussenzweig RS, Schmeckpeper B, et al. Synthetic malaria peptide vaccine elicits high levels of antibodies in vaccinees of defined HLA genotypes. J Infect Dis. 2000 Nov;182(5):1486–96. [PubMed] [Google Scholar]
35. 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 Jan;95(1):341–9. [PMC free article] [PubMed] [Google Scholar]
36. Livingston BD, Crimi C, Grey H, Ishioka G, Chisari FV, Fikes J, et al. The hepatitis B virus-specific CTL responses induced in humans by lipopeptide vaccination are comparable to those elicited by acute viral infection. J Immunol. 1997 Aug 1;159(3):1383–92. [PubMed] [Google Scholar]
37. 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 Mar 1;162(5):3088–95. [PubMed] [Google Scholar]
38. Durier C, Launay O, Meiffredy V, Saidi Y, Salmon D, Levy Y, et al. Clinical safety of HIV lipopeptides used as vaccines in healthy volunteers and HIV-infected adults. AIDS. 2006 Apr 24;20(7):1039–49. [PubMed] [Google Scholar]
39. Gahery-Segard H, Pialoux G, Figueiredo S, Igea C, Surenaud M, Gaston J, et al. Long-term specific immune responses induced in humans by a human immunodeficiency virus type 1 lipopeptide vaccine: characterization of CD8+-T-cell epitopes recognized. J Virol. 2003 Oct;77(20):11220–31. [PMC free article] [PubMed] [Google Scholar]
40. 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 Sep;23(9):1105–13. [PubMed] [Google Scholar]
41. Poland GA, Jacobson RM. The prevention of Lyme disease with vaccine. Vaccine. 2001 Mar 21;19(17-19):2303–8. [PubMed] [Google Scholar]
42. Lathrop SL, Ball R, Haber P, Mootrey GT, Braun MM, Shadomy SV, et al. Adverse event reports following vaccination for Lyme disease: December 1998-July 2000. Vaccine. 2002 Feb 22;20(11-12):1603–8. [PubMed] [Google Scholar]
43. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001 Oct 18;413(6857):732–8. [PubMed] [Google Scholar]
44. Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006 May;13(5):816–25. [PubMed] [Google Scholar]
45. Schulz O, Diebold SS, Chen M, Naslund TI, Nolte MA, Alexopoulou L, et al. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature. 2005 Feb 24;433(7028):887–92. [PubMed] [Google Scholar]
46. Schroder M, Bowie AG. TLR3 in antiviral immunity: key player or bystander? Trends Immunol. 2005 Sep;26(9):462–8. [PubMed] [Google Scholar]
47. Le BA, Etchart N, Rossmann C, Ashton M, Hou S, Gewert D, et al. Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat Immunol. 2003 Oct;4(10):1009–15. [PubMed] [Google Scholar]
48. Ishii KJ, Akira S. Toll or toll-free adjuvant path toward the optimal vaccine development. J Clin Immunol. 2007 Jul;27(4):363–71. [PubMed] [Google Scholar]
49. Matsumoto M, Seya T. TLR3: interferon induction by double-stranded RNA including poly(I:C). Adv Drug Deliv Rev. 2008 Apr 29;60(7):805–12. [PubMed] [Google Scholar]
50. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001 Oct 18;413(6857):732–8. [PubMed] [Google Scholar]
51. Stowell NC, Seideman J, Raymond HA, Smalley KA, Lamb RJ, Egenolf DD, et al. Long-term activation of TLR3 by poly(I:C) induces inflammation and impairs lung function in mice. Respir Res. 2009;10:43. [PMC free article] [PubMed] [Google Scholar]
52. Ichinohe T, Watanabe I, Ito S, Fujii H, Moriyama M, Tamura S, et al. Synthetic double-stranded RNA poly(I:C) combined with mucosal vaccine protects against influenza virus infection. J Virol. 2005 Mar;79(5):2910–9. [PMC free article] [PubMed] [Google Scholar]
53. Pulko V, Liu X, Krco CJ, Harris KJ, Frigola X, Kwon ED, et al. TLR3-stimulated dendritic cells up-regulate B7-H1 expression and influence the magnitude of CD8 T cell responses to tumor vaccination. J Immunol. 2009 Sep 15;183(6):3634–41. [PMC free article] [PubMed] [Google Scholar]
54. Currie AJ, van der Most RG, Broomfield SA, Prosser AC, Tovey MG, Robinson BW. Targeting the effector site with IFN-alphabeta-inducing TLR ligands reactivates tumor-resident CD8 T cell responses to eradicate established solid tumors. J Immunol. 2008 Feb 1;180(3):1535–44. [PubMed] [Google Scholar]
55. Salem ML, Kadima AN, Cole DJ, Gillanders WE. Defining the antigen-specific T-cell response to vaccination and poly(I:C)/TLR3 signaling: evidence of enhanced primary and memory CD8 T-cell responses and antitumor immunity. J Immunother. 2005 May;28(3):220–8. [PubMed] [Google Scholar]
56. Celis E. Toll-like receptor ligands energize peptide vaccines through multiple paths. Cancer Res. 2007 Sep 1;67(17):7945–7. [PubMed] [Google Scholar]
57. McFarlin DE, Bever CT, Salazar AM, Levy HB. A preliminary trial of poly(I,C)-LC in multiple sclerosis. J Biol Response Mod. 1985 Oct;4(5):544–8. [PubMed] [Google Scholar]
58. Nordlund JJ, Wolff SM, Levy HB. Inhibition of biologic activity of poly I: poly C by human plasma. Proc Soc Exp Biol Med. 1970 Feb;133(2):439–44. [PubMed] [Google Scholar]
59. Robinson RA, DeVita VT, Levy HB, Baron S, Hubbard SP, Levine AS. A phase I-II trial of multiple-dose polyriboinosic-polyribocytidylic acid in patieonts with leukemia or solid tumors. J Natl Cancer Inst. 1976 Sep;57(3):599–602. [PubMed] [Google Scholar]
60. Levy HB, Baer G, Baron S, Buckler CE, Gibbs CJ, Iadarola MJ, et al. A modified polyriboinosinic-polyribocytidylic acid complex that induces interferon in primates. J Infect Dis. 1975 Oct;132(4):434–9. [PubMed] [Google Scholar]
61. Zhu X, Nishimura F, Sasaki K, Fujita M, Dusak JE, Eguchi J, et al. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J Transl Med. 2007;5:10. [PMC free article] [PubMed] [Google Scholar]
62. Stahl-Hennig C, Eisenblatter M, Jasny E, Rzehak T, Tenner-Racz K, Trumpfheller C, et al. Synthetic double-stranded RNAs are adjuvants for the induction of T helper 1 and humoral immune responses to human papillomavirus in rhesus macaques. PLoS Pathog. 2009 Apr;5(4):e1000373. [PMC free article] [PubMed] [Google Scholar]
63. Carter WA, Pitha PM, Marshall LW, Tazawa I, Tazawa S, Ts'o PO. Structural requirements of the rI n -rC n complex for induction of human interferon. J Mol Biol. 1972 Oct 14;70(3):567–87. [PubMed] [Google Scholar]
64. Adams M, Navabi H, Jasani B, Man S, Fiander A, Evans AS, et al. Dendritic cell (DC) based therapy for cervical cancer: use of DC pulsed with tumour lysate and matured with a novel synthetic clinically non-toxic double stranded RNA analogue poly [I]:poly [C(12)U] (Ampligen R). Vaccine. 2003 Jan 30;21(7-8):787–90. [PubMed] [Google Scholar]
65. Navabi H, Jasani B, Reece A, Clayton A, Tabi Z, Donninger C, et al. A clinical grade poly I:C-analogue (Ampligen) promotes optimal DC maturation and Th1-type T cell responses of healthy donors and cancer patients in vitro. Vaccine. 2009 Jan 1;27(1):107–15. [PubMed] [Google Scholar]
66. Ichinohe T, Kawaguchi A, Tamura S, Takahashi H, Sawa H, Ninomiya A, et al. Intranasal immunization with H5N1 vaccine plus Poly I:Poly C12U, a Toll-like receptor agonist, protects mice against homologous and heterologous virus challenge. Microbes Infect. 2007 Sep;9(11):1333–40. [PubMed] [Google Scholar]
67. Rettenmaier MA, Berman ML, DiSaia PJ. Treatment of advanced ovarian cancer with polyinosinic-polycytidylic lysine carboxymethylcellulose (poly(ICLC]. Gynecol Oncol. 1986 Jul;24(3):359–61. [PubMed] [Google Scholar]
68. Krown SE, Kerr D, Stewart WE, Field AK, Oettgen HF. Phase I trials of poly(I,C) complexes in advanced cancer. J Biol Response Mod. 1985 Dec;4(6):640–9. [PubMed] [Google Scholar]
69. Lampkin BC, Levine AS, Levy H, Krivit W, Hammond D. Phase II trial of poly(I,C)-LC, an interferon inducer, in the treatment of children with acute leukemia and neuroblastoma: a report from the Children's Cancer Study Group. J Biol Response Mod. 1985 Oct;4(5):531–7. [PubMed] [Google Scholar]
70. Lampkin BC, Levine AS, Levy H, Krivit W, Hammond D. Phase II trial of a complex polyriboinosinic-polyribocytidylic acid with poly-L-lysine and carboxymethyl cellulose in the treatment of children with acute leukemia and neuroblastoma: a report from the Children's Cancer Study Group. Cancer Res. 1985 Nov;45(11 Pt 2):5904–9. [PubMed] [Google Scholar]
71. Durie BG, Levy HB, Voakes J, Jett JR, Levine AS. Poly(I,C)-LC as an interferon inducer in refractory multiple myeloma. J Biol Response Mod. 1985 Oct;4(5):518–24. [PubMed] [Google Scholar]
72. Stevenson HC, Abrams PG, Schoenberger CS, Smalley RB, Herberman RB, Foon KA. A phase I evaluation of poly(I,C)-LC in cancer patients. J Biol Response Mod. 1985 Dec;4(6):650–5. [PubMed] [Google Scholar]
73. Hawkins MJ, Levin M, Borden EC. An Eastern Cooperative Oncology Group phase I-II pilot study of polyriboinosinic-polyribocytidylic acid poly-L-lysine complex in patients with metastatic malignant melanoma. J Biol Response Mod. 1985 Dec;4(6):664–8. [PubMed] [Google Scholar]
74. Giantonio BJ, Hochster H, Blum R, Wiernik PH, Hudes GR, Kirkwood J, et al. Toxicity and response evaluation of the interferon inducer poly ICLC administered at low dose in advanced renal carcinoma and relapsed or refractory lymphoma: a report of two clinical trials of the Eastern Cooperative Oncology Group. Invest New Drugs. 2001;19(1):89–92. [PubMed] [Google Scholar]
75. Brodsky I, Strayer DR, Krueger LJ, Carter WA. Clinical studies with ampligen (mismatched double-stranded RNA). J Biol Response Mod. 1985 Dec;4(6):669–75. [PubMed] [Google Scholar]
76. Carter WA, Strayer DR, Hubbell HR, Brodsky I. Preclinical studies with Ampligen (mismatched double-stranded RNA). J Biol Response Mod. 1985 Oct;4(5):495–502. [PubMed] [Google Scholar]
77. Jasani B, Navabi H, Adams M. Ampligen: a potential toll-like 3 receptor adjuvant for immunotherapy of cancer. Vaccine. 2009 May 26;27(25-26):3401–4. [PubMed] [Google Scholar]
78. Kanzler H, Barrat FJ, Hessel EM, Coffman RL. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat Med. 2007 May;13(5):552–9. [PubMed] [Google Scholar]
79. Samuelsson P, Hang L, Wullt B, Irjala H, Svanborg C. Toll-like receptor 4 expression and cytokine responses in the human urinary tract mucosa. Infect Immun. 2004 Jun;72(6):3179–86. [PMC free article] [PubMed] [Google Scholar]
80. Gribar SC, Anand RJ, Sodhi CP, Hackam DJ. The role of epithelial Toll-like receptor signaling in the pathogenesis of intestinal inflammation. J Leukoc Biol. 2008 Mar;83(3):493–8. [PubMed] [Google Scholar]
81. Kumar H, Kawai T, Akira S. Toll-like receptors and innate immunity. Biochem Biophys Res Commun. 2009 Oct 30;388(4):621–5. [PubMed] [Google Scholar]
82. Ulrich JT, Myers KR. Monophosphoryl lipid A as an adjuvant. Past experiences and new directions. Pharm Biotechnol. 1995;6:495–524. [PubMed] [Google Scholar]
83. Evans JT, Cluff CW, Johnson DA, Lacy MJ, Persing DH, Baldridge JR. Enhancement of antigen-specific immunity via the TLR4 ligands MPL adjuvant and Ribi.529. Expert Rev Vaccines. 2003 Apr;2(2):219–29. [PubMed] [Google Scholar]
84. Galanos C, Luderitz O, Rietschel ET, Westphal O, Brade H, Brade L, et al. Synthetic and natural Escherichia coli free lipid A express identical endotoxic activities. Eur J Biochem. 1985 Apr 1;148(1):1–5. [PubMed] [Google Scholar]
85. Rietschel ET, Brade H, Brade L, Brandenburg K, Schade U, Seydel U, et al. Lipid A, the endotoxic center of bacterial lipopolysaccharides: relation of chemical structure to biological activity. Prog Clin Biol Res. 1987;231:25–53. [PubMed] [Google Scholar]
86. Johndon AG, Gaines S, Landy M. Studies on the O antigen of Salmonella typhosa. V. Enhancement of antibody response to protein antigens by the purified lipopolysaccharide. J Exp Med. 1956 Feb 1;103(2):225–46. [PMC free article] [PubMed] [Google Scholar]
87. Johnson AG, Tomai M, Solem L, Beck L, Ribi E. Characterization of a nontoxic monophosphoryl lipid A. Rev Infect Dis. 1987 Sep;(9 Suppl 5):S512–S516. [PubMed] [Google Scholar]
88. Qureshi N, Mascagni P, Ribi E, Takayama K. Monophosphoryl lipid A obtained from lipopolysaccharides of Salmonella minnesota R595. Purification of the dimethyl derivative by high performance liquid chromatography and complete structural determination. J Biol Chem. 1985 May 10;260(9):5271–8. [PubMed] [Google Scholar]
89. Qureshi N, Takayama K, Ribi E. Purification and structural determination of nontoxic lipid A obtained from the lipopolysaccharide of Salmonella typhimurium. J Biol Chem. 1982 Oct 10;257(19):11808–15. [PubMed] [Google Scholar]
90. Johnson AG. Molecular adjuvants and immunomodulators: new approaches to immunization. Clin Microbiol Rev. 1994 Jul;7(3):277–89. [PMC free article] [PubMed] [Google Scholar]
91. Moore A, McCarthy L, Mills KH. The adjuvant combination monophosphoryl lipid A and QS21 switches T cell responses induced with a soluble recombinant HIV protein from Th2 to Th1. Vaccine. 1999 Jun 4;17(20-21):2517–27. [PubMed] [Google Scholar]
92. Baldridge JR, McGowan P, Evans JT, Cluff C, Mossman S, Johnson D, et al. Taking a Toll on human disease: Toll-like receptor 4 agonists as vaccine adjuvants and monotherapeutic agents. Expert Opin Biol Ther. 2004 Jul;4(7):1129–38. [PubMed] [Google Scholar]
93. Vosika GJ, Barr C, Gilbertson D. Phase-I study of intravenous modified lipid A. Cancer Immunol Immunother. 1984;18(2):107–12. [PMC free article] [PubMed] [Google Scholar]
94. Garcon N, Chomez P, Van MM. GlaxoSmithKline Adjuvant Systems in vaccines: concepts, achievements and perspectives. Expert Rev Vaccines. 2007 Oct;6(5):723–39. [PubMed] [Google Scholar]
95. Tagliabue A, Rappuoli R. Vaccine adjuvants: the dream becomes real. Hum Vaccine. 2008 Sep;4(5):347–9. [PubMed] [Google Scholar]
96. Hollinger FB. Factors influencing the immune response to hepatitis B vaccine, booster dose guidelines, and vaccine protocol recommendations. Am J Med. 1989 Sep 4;87(3A):36S–40S. [PubMed] [Google Scholar]
97. Kundi M. New hepatitis B vaccine formulated with an improved adjuvant system. Expert Rev Vaccines. 2007 Apr;6(2):133–40. [PubMed] [Google Scholar]
98. Thoelen S, Van DP, Mathei C, Leroux-Roels G, Desombere I, Safary A, et al. Safety and immunogenicity of a hepatitis B vaccine formulated with a novel adjuvant system. Vaccine. 1998 Apr;16(7):708–14. [PubMed] [Google Scholar]
99. Thoelen S, De CN, Tornieporth N. A prophylactic hepatitis B vaccine with a novel adjuvant system. Vaccine. 2001 Mar 21;19(17-19):2400–3. [PubMed] [Google Scholar]
100. Ambrosch F, Wiedermann G, Kundi M, Leroux-Roels G, Desombere I, Garcon N, et al. A hepatitis B vaccine formulated with a novel adjuvant system. Vaccine. 2000 Apr 14;18(20):2095–101. [PubMed] [Google Scholar]
101. Levie K, Gjorup I, Skinhoj P, Stoffel M. A 2-dose regimen of a recombinant hepatitis B vaccine with the immune stimulant AS04 compared with the standard 3-dose regimen of Engerix-B® in healthy young adults. Scand J Infect Dis. 2002;34(8):610–4. [PubMed] [Google Scholar]
102. Boland G, Beran J, Lievens M, Sasadeusz J, Dentico P, Nothdurft H, et al. Safety and immunogenicity profile of an experimental hepatitis B vaccine adjuvanted with AS04. Vaccine. 2004 Dec 2;23(3):316–20. [PubMed] [Google Scholar]
103. Tong NK, Beran J, Kee SA, Miguel JL, Sanchez C, Bayas JM, et al. Immunogenicity and safety of an adjuvanted hepatitis B vaccine in pre-hemodialysis and hemodialysis patients. Kidney Int. 2005 Nov;68(5):2298–303. [PubMed] [Google Scholar]
104. Giannini SL, Hanon E, Moris P, Van MM, Morel S, Dessy F, et al. Enhanced humoral and memory B cellular immunity using HPV16/18 L1 VLP vaccine formulated with the MPL/aluminium salt combination (AS04) compared to aluminium salt only. Vaccine. 2006 Aug 14;24(33-34):5937–49. [PubMed] [Google Scholar]
105. Romanowski B, de Borba PC, Naud PS, Roteli-Martins CM, De Carvalho NS, Teixeira JC, et al. Sustained efficacy and immunogenicity of the human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine: analysis of a randomised placebo-controlled trial up to 6.4 years. Lancet. 2009 Dec 12;374(9706):1975–85. [PubMed] [Google Scholar]
106. DeCarvalho N, Roteli-Martins CM, Teixeira J. Sustained levels of total and neutralizing antibodies and favourable long term safety with the HPV-16/18 AS04-adjuvanted vaccine (Cervarix):follow-up to 7.3 years. International Journal of Gynecology and Obstetrics. 2010;107(Supp 2):357. [Google Scholar]
107. Paavonen J, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter D, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009 Jul 25;374(9686):301–14. [PubMed] [Google Scholar]
108. Schwarz TF, Spaczynski M, Schneider A, Wysocki J, Galaj A, Perona P, et al. Immunogenicity and tolerability of an HPV-16/18 AS04-adjuvanted prophylactic cervical cancer vaccine in women aged 15-55 years. Vaccine. 2009 Jan 22;27(4):581–7. [PubMed] [Google Scholar]
109. Paavonen J, Jenkins D, Bosch FX, Naud P, Salmeron J, Wheeler CM, et al. Efficacy of a prophylactic adjuvanted bivalent L1 virus-like-particle vaccine against infection with human papillomavirus types 16 and 18 in young women: an interim analysis of a phase III double-blind, randomised controlled trial. Lancet. 2007 Jun 30;369(9580):2161–70. [PubMed] [Google Scholar]
110. Mitchell MS, Harel W, Kempf RA, Hu E, Kan-Mitchell J, Boswell WD, et al. Active-specific immunotherapy for melanoma. J Clin Oncol. 1990 May;8(5):856–69. [PubMed] [Google Scholar]
111. Elliott GT, McLeod RA, Perez J, Von Eschen KB. Interim results of a phase II multicenter clinical trial evaluating the activity of a therapeutic allogeneic melanoma vaccine (theraccine) in the treatment of disseminated malignant melanoma. Semin Surg Oncol. 1993 May;9(3):264–72. [PubMed] [Google Scholar]
112. Sondak VK, Sosman JA. Results of clinical trials with an allogenic melanoma tumor cell lysate vaccine: Melacine. Semin Cancer Biol. 2003 Dec;13(6):409–15. [PubMed] [Google Scholar]
113. North S, Butts C. Vaccination with BLP25 liposome vaccine to treat non-small cell lung and prostate cancers. Expert Rev Vaccines. 2005 Jun;4(3):249–57. [PubMed] [Google Scholar]
114. Palmer M, Parker J, Modi S, Butts C, Smylie M, Meikle A, et al. Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer. 2001 Aug;3(1):49–57. [PubMed] [Google Scholar]
115. Sangha R, North S. L-BLP25: a MUC1-targeted peptide vaccine therapy in prostate cancer. Expert Opin Biol Ther. 2007 Nov;7(11):1723–30. [PubMed] [Google Scholar]
116. Persing DH, Coler RN, Lacy MJ, Johnson DA, Baldridge JR, Hershberg RM, et al. Taking toll: lipid A mimetics as adjuvants and immunomodulators. Trends Microbiol. 2002;10(10 Suppl):S32–S37. [PubMed] [Google Scholar]
117. Dupont J, Altclas J, Lepetic A, Lombardo M, Vazquez V, Salgueira C, et al. A controlled clinical trial comparing the safety and immunogenicity of a new adjuvanted hepatitis B vaccine with a standard hepatitis B vaccine. Vaccine. 2006 Nov 30;24(49-50):7167–74. [PubMed] [Google Scholar]
118. Descamps D, Hardt K, Spiessens B, Izurieta P, Verstraeten T, Breuer T, et al. Safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine for cervical cancer prevention: a pooled analysis of 11 clinical trials. Hum Vaccin. 2009 May;5(5):332–40. [PubMed] [Google Scholar]
119. Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001 Apr 26;410(6832):1099–103. [PubMed] [Google Scholar]
120. Mizel SB, West AP, Hantgan RR. Identification of a sequence in human toll-like receptor 5 required for the binding of Gram-negative flagellin. J Biol Chem. 2003 Jun 27;278(26):23624–9. [PubMed] [Google Scholar]
121. Chalifour A, Jeannin P, Gauchat JF, Blaecke A, Malissard M, N'Guyen T, et al. Direct bacterial protein PAMP recognition by human NK cells involves TLRs and triggers alpha-defensin production. Blood. 2004 Sep 15;104(6):1778–83. [PubMed] [Google Scholar]
122. Caron G, Duluc D, Fremaux I, Jeannin P, David C, Gascan H, et al. Direct stimulation of human T cells via TLR5 and TLR7/8: flagellin and R-848 up-regulate proliferation and IFN-gamma production by memory CD4+ T cells. J Immunol. 2005 Aug 1;175(3):1551–7. [PubMed] [Google Scholar]
123. Farina C, Theil D, Semlinger B, Hohlfeld R, Meinl E. Distinct responses of monocytes to Toll-like receptor ligands and inflammatory cytokines. Int Immunol. 2004 Jun;16(6):799–809. [PubMed] [Google Scholar]
124. Hornung V, Rothenfusser S, Britsch S, Krug A, Jahrsdorfer B, Giese T, et al. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol. 2002 May 1;168(9):4531–7. [PubMed] [Google Scholar]
125. Means TK, Hayashi F, Smith KD, Aderem A, Luster AD. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol. 2003 May 15;170(10):5165–75. [PubMed] [Google Scholar]
126. Peiser M, Wanner R, Kolde G. Human epidermal Langerhans cells differ from monocyte-derived Langerhans cells in CD80 expression and in secretion of IL-12 after CD40 cross-linking. J Leukoc Biol. 2004 Sep;76(3):616–22. [PubMed] [Google Scholar]
127. Cuadros C, Lopez-Hernandez FJ, Dominguez AL, McClelland M, Lustgarten J. Flagellin fusion proteins as adjuvants or vaccines induce specific immune responses. Infect Immun. 2004 May;72(5):2810–6. [PMC free article] [PubMed] [Google Scholar]
128. Huleatt JW, Jacobs AR, Tang J, Desai P, Kopp EB, Huang Y, et al. Vaccination with recombinant fusion proteins incorporating Toll-like receptor ligands induces rapid cellular and humoral immunity. Vaccine. 2007 Jan 8;25(4):763–75. [PubMed] [Google Scholar]
129. Huleatt JW, Nakaar V, Desai P, Huang Y, Hewitt D, Jacobs A, et al. Potent immunogenicity and efficacy of a universal influenza vaccine candidate comprising a recombinant fusion protein linking influenza M2e to the TLR5 ligand flagellin. Vaccine. 2008 Jan 10;26(2):201–14. [PubMed] [Google Scholar]
130. Honko AN, Sriranganathan N, Lees CJ, Mizel SB. Flagellin is an effective adjuvant for immunization against lethal respiratory challenge with Yersinia pestis. Infect Immun. 2006 Feb;74(2):1113–20. [PMC free article] [PubMed] [Google Scholar]
131. McDonald WF, Huleatt JW, Foellmer HG, Hewitt D, Tang J, Desai P, et al. A West Nile virus recombinant protein vaccine that coactivates innate and adaptive immunity. J Infect Dis. 2007 Jun 1;195(11):1607–17. [PubMed] [Google Scholar]
132. Delaney KN, Phipps JP, Johnson JB, Mizel SB. A recombinant flagellin-poxvirus fusion protein vaccine elicits complement-dependent protection against respiratory challenge with vaccinia virus in mice. Viral Immunol. 2010 Apr;23(2):201–10. [PMC free article] [PubMed] [Google Scholar]
133. Weimer ET, Lu H, Kock ND, Wozniak DJ, Mizel SB. A fusion protein vaccine containing OprF epitope 8, OprI, and type A and B flagellins promotes enhanced clearance of nonmucoid Pseudomonas aeruginosa. Infect Immun. 2009 Jun;77(6):2356–66. [PMC free article] [PubMed] [Google Scholar]
134. Weimer ET, Ervin SE, Wozniak DJ, Mizel SB. Immunization of young African green monkeys with OprF epitope 8-OprI-type A- and B-flagellin fusion proteins promotes the production of protective antibodies against nonmucoid Pseudomonas aeruginosa. Vaccine. 2009 Nov 12;27(48):6762–9. [PubMed] [Google Scholar]
135. Honko AN, Mizel SB. Mucosal administration of flagellin induces innate immunity in the mouse lung. Infect Immun. 2004 Nov;72(11):6676–9. [PMC free article] [PubMed] [Google Scholar]
136. Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004 Mar 5;303(5663):1526–9. [PubMed] [Google Scholar]
137. Diebold SS, Kaisho T, Hemmi H, Akira S, Sousa Reis e. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004 Mar 5;303(5663):1529–31. [PubMed] [Google Scholar]
138. Lee J, Chuang TH, Redecke V, She L, Pitha PM, Carson DA, et al. Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: activation of Toll-like receptor 7. Proc Natl Acad Sci U S A. 2003 May 27;100(11):6646–51. [PMC free article] [PubMed] [Google Scholar]
139. Jurk M, Kritzler A, Schulte B, Tluk S, Schetter C, Krieg AM, et al. Modulating responsiveness of human TLR7 and 8 to small molecule ligands with T-rich phosphorothiate oligodeoxynucleotides. Eur J Immunol. 2006 Jul;36(7):1815–26. [PubMed] [Google Scholar]
140. Gorden KB, Gorski KS, Gibson SJ, Kedl RM, Kieper WC, Qiu X, et al. Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8. J Immunol. 2005 Feb 1;174(3):1259–68. [PubMed] [Google Scholar]
141. Zarember KA, Godowski PJ. Tissue expression of human Toll-like receptors and differential regulation of Toll-like receptor mRNAs in leukocytes in response to microbes, their products, and cytokines. J Immunol. 2002 Jan 15;168(2):554–61. [PubMed] [Google Scholar]
142. Gibson SJ, Lindh JM, Riter TR, Gleason RM, Rogers LM, Fuller AE, et al. Plasmacytoid dendritic cells produce cytokines and mature in response to the TLR7 agonists, imiquimod and resiquimod. Cell Immunol. 2002 Jul;218(1-2):74–86. [PubMed] [Google Scholar]
143. Lehner M, Morhart P, Stilper A, Petermann D, Weller P, Stachel D, et al. Efficient chemokine-dependent migration and primary and secondary IL-12 secretion by human dendritic cells stimulated through Toll-like receptors. J Immunother. 2007 Apr;30(3):312–22. [PubMed] [Google Scholar]
144. Ito T, Amakawa R, Kaisho T, Hemmi H, Tajima K, Uehira K, et al. Interferon-alpha and interleukin-12 are induced differentially by Toll-like receptor 7 ligands in human blood dendritic cell subsets. J Exp Med. 2002 Jun 3;195(11):1507–12. [PMC free article] [PubMed] [Google Scholar]
145. Burns RP, Jr., Ferbel B, Tomai M, Miller R, Gaspari AA. The imidazoquinolines, imiquimod and R-848, induce functional, but not phenotypic, maturation of human epidermal Langerhans’ cells. Clin Immunol. 2000 Jan;94(1):13–23. [PubMed] [Google Scholar]
146. Suzuki H, Wang B, Shivji GM, Toto P, Amerio P, Tomai MA, et al. Imiquimod, a topical immune response modifier, induces migration of Langerhans cells. J Invest Dermatol. 2000 Jan;114(1):135–41. [PubMed] [Google Scholar]
147. Bishop GA, Hsing Y, Hostager BS, Jalukar SV, Ramirez LM, Tomai MA. Molecular mechanisms of B lymphocyte activation by the immune response modifier R-848. J Immunol. 2000 Nov 15;165(10):5552–7. [PubMed] [Google Scholar]
148. Hart OM, thie-Morales V, O'Connor GM, Gardiner CM. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-gamma production. J Immunol. 2005 Aug 1;175(3):1636–42. [PubMed] [Google Scholar]
149. Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005 Aug 26;309(5739):1380–4. [PubMed] [Google Scholar]
150. Harrison LI, Astry C, Kumar S, Yunis C. Pharmacokinetics of 852A, an imidazoquinoline Toll-like receptor 7-specific agonist, following intravenous, subcutaneous, and oral administrations in humans. J Clin Pharmacol. 2007 Aug;47(8):962–9. [PubMed] [Google Scholar]
151. Jurk M, Heil F, Vollmer J, Schetter C, Krieg AM, Wagner H, et al. Human TLR7 or TLR8 independently confer responsiveness to the antiviral compound R-848. Nat Immunol. 2002 Jun;3(6):499. [PubMed] [Google Scholar]
152. Gorden KK, Qiu XX, Binsfeld CC, Vasilakos JP, Alkan SS. Cutting edge: activation of murine TLR8 by a combination of imidazoquinoline immune response modifiers and polyT oligodeoxynucleotides. J Immunol. 2006 Nov 15;177(10):6584–7. [PubMed] [Google Scholar]
153. Martinez J, Huang X, Yang Y. Toll-like receptor 8-mediated activation of murine plasmacytoid dendritic cells by vaccinia viral DNA. Proc Natl Acad Sci U S A. 2010 Apr 6;107(14):6442–7. [PMC free article] [PubMed] [Google Scholar]
154. Levy O, Zarember KA, Roy RM, Cywes C, Godowski PJ, Wessels MR. Selective impairment of TLR-mediated innate immunity in human newborns: neonatal blood plasma reduces monocyte TNF-alpha induction by bacterial lipopeptides, lipopolysaccharide, and imiquimod, but preserves the response to R-848. J Immunol. 2004 Oct 1;173(7):4627–34. [PubMed] [Google Scholar]
155. Angelone DF, Wessels MR, Coughlin M, Suter EE, Valentini P, Kalish LA, et al. Innate immunity of the human newborn is polarized toward a high ratio of IL-6/TNF-alpha production in vitro and in vivo. Pediatr Res. 2006 Aug;60(2):205–9. [PubMed] [Google Scholar]
156. Wille-Reece U, Wu CY, Flynn BJ, Kedl RM, Seder RA. Immunization with HIV-1 Gag protein conjugated to a TLR7/8 agonist results in the generation of HIV-1 Gag-specific Th1 and CD8+ T cell responses. J Immunol. 2005 Jun 15;174(12):7676–83. [PubMed] [Google Scholar]
157. Wille-Reece U, Flynn BJ, Lore K, Koup RA, Kedl RM, Mattapallil JJ, et al. HIV Gag protein conjugated to a Toll-like receptor 7/8 agonist improves the magnitude and quality of Th1 and CD8+ T cell responses in nonhuman primates. Proc Natl Acad Sci U S A. 2005 Oct 18;102(42):15190–4. [PMC free article] [PubMed] [Google Scholar]
158. Wille-Reece U, Flynn BJ, Lore K, Koup RA, Miles AP, Saul A, et al. Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primates. J Exp Med. 2006 May 15;203(5):1249–58. [PMC free article] [PubMed] [Google Scholar]
159. Otero M, Calarota SA, Felber B, Laddy D, Pavlakis G, Boyer JD, et al. Resiquimod is a modest adjuvant for HIV-1 gag-based genetic immunization in a mouse model. Vaccine. 2004 Apr 16;22(13-14):1782–90. [PubMed] [Google Scholar]
160. Harrison CJ, Miller RL, Bernstein DI. Reduction of recurrent HSV disease using imiquimod alone or combined with a glycoprotein vaccine. Vaccine. 2001 Feb 8;19(13-14):1820–6. [PubMed] [Google Scholar]
161. Smorlesi A, Papalini F, Orlando F, Donnini A, Re F, Provinciali M. Imiquimod and S-27609 as adjuvants of DNA vaccination in a transgenic murine model of HER2/neu-positive mammary carcinoma. Gene Ther. 2005 Sep;12(17):1324–32. [PubMed] [Google Scholar]
162. Beutner KR, Spruance SL, Hougham AJ, Fox TL, Owens ML, Douglas JM., Jr. Treatment of genital warts with an immune-response modifier (imiquimod). J Am Acad Dermatol. 1998 Feb;38(2 Pt 1):230–9. [PubMed] [Google Scholar]
163. Stanley MA. Imiquimod and the imidazoquinolones: mechanism of action and therapeutic potential. Clin Exp Dermatol. 2002 Oct;27(7):571–7. [PubMed] [Google Scholar]
164. Schulze HJ, Cribier B, Requena L, Reifenberger J, Ferrandiz C, Garcia DA, et al. Imiquimod 5% cream for the treatment of superficial basal cell carcinoma: results from a randomized vehicle-controlled phase III study in Europe. Br J Dermatol. 2005 May;152(5):939–47. [PubMed] [Google Scholar]
165. Lebwohl M, Dinehart S, Whiting D, Lee PK, Tawfik N, Jorizzo J, et al. Imiquimod 5% cream for the treatment of actinic keratosis: results from two phase III, randomized, double-blind, parallel group, vehicle-controlled trials. J Am Acad Dermatol. 2004 May;50(5):714–21. [PubMed] [Google Scholar]
166. Shackleton M, Davis ID, Hopkins W, Jackson H, Dimopoulos N, Tai T, et al. The impact of imiquimod, a Toll-like receptor-7 ligand (TLR7L), on the immunogenicity of melanoma peptide vaccination with adjuvant Flt3 ligand. Cancer Immun. 2004 Sep 23;4:9. [PubMed] [Google Scholar]
167. Adams S, O'Neill DW, Nonaka D, Hardin E, Chiriboga L, Siu K, et al. Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant. J Immunol. 2008 Jul 1;181(1):776–84. [PMC free article] [PubMed] [Google Scholar]
168. Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, et al. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate. 2009 Jun 15;69(9):917–27. [PubMed] [Google Scholar]
169. Gunzer M, Riemann H, Basoglu Y, Hillmer A, Weishaupt C, Balkow S, et al. Systemic administration of a TLR7 ligand leads to transient immune incompetence due to peripheral-blood leukocyte depletion. Blood. 2005 Oct 1;106(7):2424–32. [PubMed] [Google Scholar]
170. Goldstein D, Hertzog P, Tomkinson E, Couldwell D, McCarville S, Parrish S, et al. Administration of imiquimod, an interferon inducer, in asymptomatic human immunodeficiency virus-infected persons to determine safety and biologic response modification. J Infect Dis. 1998 Sep;178(3):858–61. [PubMed] [Google Scholar]
171. Savage P, Horton V, Moore J, Owens M, Witt P, Gore ME. A phase I clinical trial of imiquimod, an oral interferon inducer, administered daily. Br J Cancer. 1996 Nov;74(9):1482–6. [PMC free article] [PubMed] [Google Scholar]
172. Hemmi H, Takeuchi O, Kawai T, Sato S, Sanjo H, Matsumoto M, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000;408:740–5. [PubMed] [Google Scholar]
173. Takeshita F, Leifer CA, Gursel I, Ishii K, Takeshita S, Gursel M, et al. Cutting Edge: role of toll-like receptor 9 in CpG DNA-induced activation of human cells. J Immunol. 2001;167(7):3555–8. [PubMed] [Google Scholar]
174. Bauer S, Kirschning CJ, Hacker H, Redecke V, Hausmann S, Akira S, et al. Human TLR9 confers responsiveness to bacterial DNA via species -specific CpG motif recognition. Proc Natl Acad Sci U S A. 2001;98(16):9237–42. [PMC free article] [PubMed] [Google Scholar]
175. Gursel M, Verthelyi D, Gursel I, Ishii KJ, Klinman DM. Differential and competitive activation of human immune cells by distinct classes of CpG oligodeoxynucleotides. Journal of Leukocyte Biology. 2002;71:813–20. [PubMed] [Google Scholar]
176. Hayashi F, Means TK, Luster AD. Toll-like receptors stimulate human neutrophil function. Blood. 2003 Oct 1;102(7):2660–9. [PubMed] [Google Scholar]
177. Bauer M, Redecke V, Ellwart JW, Sherer B, Kremer JP, Wagner H, et al. Bacterial CpG DNA triggers activation and maturation of human CD11c(-), CD123(+) dendritic cells. J Immunol. 2001;166(8):5000–7. [PubMed] [Google Scholar]
178. Bauer M, Heeg K, Wagner H, Lipford GB. DNA activates human immune cells through a CpG sequence-dependent manner. Immunology. 1999;97(4):699–705. [PMC free article] [PubMed] [Google Scholar]
179. Auricchio G, Garg SK, Martino A, Volpe E, Ciaramella A, De Vito P, et al. Role of macrophage phospholipase D in natural and CpG-induced antimycobacterial activity. Cell Microbiol. 2003 Dec;5(12):913–20. [PubMed] [Google Scholar]
180. Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. 2002;20:709–60. 709–60. [PubMed] [Google Scholar]
181. Klinman DM, Yi A, Beaucage SL, Conover J, Krieg AM. CpG motifs present in bacterial DNA rapidly induce lymphocytes to secrete IL-6, IL-12 and IFNg. Proc Natl Acad Sci USA. 1996;93:2879–83. [PMC free article] [PubMed] [Google Scholar]
182. Ballas ZK, Rasmussen WL, Krieg AM. Induction of NK activity in murine and human cells by CpG motifs in oligodeoxynucleotides and bacterial DNA. J Immunol. 1996;157:1840–7. [PubMed] [Google Scholar]
183. Halpern MD, Kurlander RJ, Pisetsky DS. Bacterial DNA induces murine interferon-gamma production by stimulation of IL-12 and tumor necrosis factor-alpha. Cell Immunol. 1996;167:72–8. [PubMed] [Google Scholar]
184. Ishii KJ, Takeshita F, Gursel I, Gursel M, Conover J, Nussenzweig A, et al. Potential role of phosphatidylinositol 3 kinase, rather than DNA-dependent protein kinase, in CpG DNA-induced immune activation. J Exp Med. 2002 Jul 15;196(2):269–74. [PMC free article] [PubMed] [Google Scholar]
185. Wagner H. Bacterial CpG-DNA activates immune cells to signal “infectious danger”. Adv Immunol. 1999;73:329–68. [PubMed] [Google Scholar]
186. Sparwasser T, Meithke T, Lipford G, Borschert K, Hicker H, Heeg K, et al. Bacterial DNA causes septic shock. Nature. 1997;386:336–8. [PubMed] [Google Scholar]
187. Sparwasser T, Miethke T, Lipford G, Erdmann A, Hacker H, Heeg K, et al. Macrophages sense pathogens via DNA motifs: induction of tumor necrosis factor-a-mediated shock. Eur J Immunol. 1997;27:1671–9. [PubMed] [Google Scholar]
188. Vollmer J, Weeratna R, Payette P, Jurk M, Schetter C, Laucht M, et al. Characterization of three CpG oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur J Immunol. 2004 Jan;34(1):251–62. [PubMed] [Google Scholar]
189. Verthelyi D, Ishii KJ, Gursel M, Takeshita F, Klinman DM. Human peripheral blood cells differentially recognize and respond to two distinct CpG motifs. J Immunol. 2001;166:2372–7. [PubMed] [Google Scholar]
190. Krug A, Rothenfusser S, Hornung V, Jahrsdorfer B, Blackwell S, Ballas ZK, et al. Identification of CpG oligonucleotide sequences with high induction of IFNa/b in plasmacytoid dendritic cells. Eur J Immunol. 2001;31:2154–63. [PubMed] [Google Scholar]
191. Hartmann G, Krieg AM. Mechanism and function of a newly identified CpG DNA motif in human primary B cells. J Immunol. 2000 Jan 15;164(2):944–53. [PubMed] [Google Scholar]
192. Klinman DM, Currie D, Lee G, Grippe V, Merkel T. Systemic but not mucosal immunity induced by AVA prevents inhalational anthrax. Microbes Infect. 2007 Oct;9(12-13):1478–83. [PMC free article] [PubMed] [Google Scholar]
193. Xie H, Gursel I, Ivins BE, Singh M, O'Hagan DT, Ulmer JB, et al. CpG oligodeoxynucleotides adsorbed onto polylactide-co-glycolide microparticles improve the immunogenicity and protective activity of the licensed anthrax vaccine. Infect Immun. 2005 Feb;73(2):828–33. [PMC free article] [PubMed] [Google Scholar]
194. Klinman DM, xie H, Little sF, Currie D, Ivins BE. CpG oligonucleotides improve the protective immune response induced by the anthrax vaccination of rhesus macaques. Vaccine. 2004 Jul 29;22(21-22):2881–6. [PubMed] [Google Scholar]
195. Klinman DM, Tross D. A Single-Dose Combination Therapy that both Prevents and Treats Anthrax Infection. Vaccine. 2009 Apr 4; In Press. [PMC free article] [PubMed] [Google Scholar]
196. Tross D, Klinman DM. Effect of CpG oligonucleotides on vaccine-induced B cell memory. J Immunol. 2008 Oct 15;181(8):5785–90. [PMC free article] [PubMed] [Google Scholar]
197. Moldoveanu Z, Love-Homan L, Huang WQ, Krieg AM. CpG DNA, a novel immune enhancer for systemic and mucosal immunization with influenza virus. Vaccine. 1998;16:1216–24. [PubMed] [Google Scholar]
198. Horner AA, Raz E. Immunostimulatory-sequence DNA is an effective mucosal adjuvant. Curr Top Microbiol Immunol. 2000;247:185–98. [PubMed] [Google Scholar]
199. McCluskie MJ, Davis HL. Oral, intrarectal and intranasal immunizations using CpG and non-CpG oligodeoxynucleotides as adjuvants. Vaccine. 2000;19(4-5):413–22. [PubMed] [Google Scholar]
200. Prince GA, Mond JJ, Porter DD, Yim KC, Lan SJ, Klinman DM. Immunoprotective activity and safety of a respiratory syncytial virus vaccine: mucosal delivery of fusion glycoprotein with a CpG oligodeoxynucleotide adjuvant. J Virol. 2003 Dec;77(24):13156–60. [PMC free article] [PubMed] [Google Scholar]
201. Qin W, Jiang J, Chen Q, Yang N, Wang Y, Wei X, et al. CpG ODN enhances immunization effects of hepatitis B vaccine in aged mice. Cell Mol Immunol. 2004 Apr;1(2):148–52. [PubMed] [Google Scholar]
202. Kovarik J, Bozzotti P, Love-Homan L, Pihlgren M, Davis HL, Lambert PH, et al. CpG oligodeoxynucleotides can circumvent the Th2 polarization of neonatal responses to vaccines but may fail to fully redirect Th2 responses established by neonatal priming. J Immunol. 1999 Feb 1;162(3):1611–7. [PubMed] [Google Scholar]
203. Ito S, Ishii K, Shirot H, Klinman DM. CpG Oligodeoxynucleotides Improve the Survival of Pregnant and Fetal mice Following Listeria monocytogenes Infection. Infect Immune. 2004;72(6):3543–8. [PMC free article] [PubMed] [Google Scholar]
204. Verthelyi D, Gursel M, Kenney RT, Lifson JD, Liu S, Mican J, et al. CpG oligodeoxynucleotides protect normal and SIV-infected macaques from Leishmania infection. J Immunol. 2003 May 1;170(9):4717–23. [PubMed] [Google Scholar]
205. Klinman DM, Klaschik S, Sato T, Tross D. CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases. Adv Drug Deliv Rev. 2009 Mar 28;61(3):248–55. [PubMed] [Google Scholar]
206. Klinman DM, Barnhart KM, Conover J. CpG motifs as immune adjuvants. Vaccine. 1999;17:19–25. [PubMed] [Google Scholar]
207. Klinman DM. Adjuvant activity of CpG oligodeoxynucleotides. Int Rev Immunol. 2006 May;25(3-4):135–54. [PubMed] [Google Scholar]
208. Mullen GE, Ellis RD, Miura K, Malkin E, Nolan C, Hay M, et al. Phase 1 trial of AMA1-C1/Alhydrogel plus CPG 7909: an asexual blood-stage vaccine for Plasmodium falciparum malaria. PLoS One. 2008;3(8):e2940. [PMC free article] [PubMed] [Google Scholar]
209. Traore B, Kone Y, Doumbo S, Doumtabe D, Traore A, Crompton PD, et al. The TLR9 agonist CpG fails to enhance the acquisition of Plasmodium falciparum-specific memory B cells in semi-immune adults in Mali. Vaccine. 2009 Dec 9;27(52):7299–303. [PMC free article] [PubMed] [Google Scholar]
210. Sagara I, Ellis RD, Dicko A, Niambele MB, Kamate B, Guindo O, et al. A randomized and controlled Phase 1 study of the safety and immunogenicity of the AMA1-C1/Alhydrogel + CPG 7909 vaccine for Plasmodium falciparum malaria in semi-immune Malian adults. Vaccine. 2009 Dec 9;27(52):7292–8. [PMC free article] [PubMed] [Google Scholar]
211. Ellis RD, Martin LB, Shaffer D, Long CA, Miura K, Fay MP, et al. Phase 1 trial of the Plasmodium falciparum blood stage vaccine MSP1(42)-C1/Alhydrogel with and without CPG 7909 in malaria naive adults. PLoS One. 2010;5(1):e8787. [PMC free article] [PubMed] [Google Scholar]
212. Cooper CL, Davis HL, Angel JB, Morris ML, Elfer SM, Seguin I, et al. CPG 7909 adjuvant improves hepatitis B virus vaccine seroprotection in antiretroviral-treated HIV-infected adults. AIDS. 2005 Sep 23;19(14):1473–9. [PubMed] [Google Scholar]
213. Cooper CL, Angel JB, Seguin I, Davis HL, Cameron DW. CPG 7909 adjuvant plus hepatitis B virus vaccination in HIV-infected adults achieves long-term seroprotection for up to 5 years. Clin Infect Dis. 2008 Apr 15;46(8):1310–4. [PubMed] [Google Scholar]
214. Siegrist CA, Pihlgren M, Tougne C, Efler SM, Morris ML, AlAdhami MJ, et al. Co-administration of CpG oligonucleotides enhances the late affinity maturation process of human anti-hepatitis B vaccine response. Vaccine. 2004 Dec 16;23(5):615–22. [PubMed] [Google Scholar]
215. Cooper CL, Davis HL, Morris ML, Efler SM, Adhami MA, Krieg AM, et al. CPG 7909, an immunostimulatory TLR9 agonist oligodeoxynucleotide, as adjuvant to Engerix-B HBV vaccine in healthy adults: a double-blind phase I/II study. J Clin Immunol. 2004 Nov;24(6):693–701. [PubMed] [Google Scholar]
216. Halperin SA, Van Nest G, Smith B, Abtahi S, Whiley H, Eiden JJ. A phase I study of the safety and immunogenicity of recombinant hepatitis B surface antigen co-administered with an immunostimulatory phosphorothioate oligonucleotide adjuvant. Vaccine. 2003 Jun 2;21(19-20):2461–7. [PubMed] [Google Scholar]
217. Halperin SA, Dobson S, McNeil S, Langley JM, Smith B, Call-Sani R, et al. Comparison of the safety and immunogenicity of hepatitis B virus surface antigen co-administered with an immunostimulatory phosphorothioate oligonucleotide and a licensed hepatitis B vaccine in healthy young adults. Vaccine. 2006 Jan 9;24(1):20–6. [PubMed] [Google Scholar]
218. Cooper CL, Davis HL, Morris ML, Efler SM, Krieg AM, Li Y, et al. Safety and immunogenicity of CPG 7909 injection as an adjuvant to Fluarix influenza vaccine. Vaccine. 2004 Aug 13;22(23-24):3136–43. [PubMed] [Google Scholar]
219. Rynkiewicz D, Rathkopf M, Ransom J, et al. Marked enhancement of antibody response to anthrax vaccine adsorbed with CpG 7909 in healthy volunteers. ICAAC abstract LB-25. 2005 [PubMed] [Google Scholar]
220. Rothenfusser S, Hornung V, Ayyoub M, Britsch S, Towarowski A, Krug A, et al. CpG-A and CpG-B oligonucleotides differentially enhance human peptide-specific primary and memory CD8+ T-cell responses in vitro. Blood. 2004 Mar 15;103(6):2162–9. [PubMed] [Google Scholar]
221. Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov. 2006 Jun;5(6):471–84. [PubMed] [Google Scholar]
222. Tighe H, Takabayashi K, Schwartz D, Van Nest G, Tuck S, Eiden JJ, et al. Conjugation of immunostimulatory DNA to the short ragweed allergen amb a 1 enhances its immunogenicity and reduces its allergenicity. J Allergy Clin Immunol. 2000;106:124–34. [PubMed] [Google Scholar]
223. Fourcade J, Kudela P, ndrade Filho PA, Janjic B, Land SR, Sander C, et al. Immunization with analog peptide in combination with CpG and montanide expands tumor antigen-specific CD8+ T cells in melanoma patients. J Immunother. 2008 Oct;31(8):781–91. [PMC free article] [PubMed] [Google Scholar]
224. Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002 Jul 1;169(1):350–8. [PubMed] [Google Scholar]
225. Heckelsmiller K, Beck S, Rall K, Sipos B, Schlamp A, Tuma E, et al. Combined dendritic cell- and CpG oligonucleotide-based immune therapy cures large murine tumors that resist chemotherapy. Eur J Immunol. 2002 Nov;32(11):3235–45. [PubMed] [Google Scholar]
226. Speiser DE, Lienard D, Rufer N, Rubio-Godoy V, Rimoldi D, Lejeune F, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest. 2005 Mar;115(3):739–46. [PMC free article] [PubMed] [Google Scholar]
227. Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O'Neill D, et al. Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci U S A. 2007 May 22;104(21):8947–52. [PMC free article] [PubMed] [Google Scholar]
228. Karbach J, Gnjatic S, Bender A, Neumann A, Weidmann E, Yuan J, et al. Tumor-reactive CD8+ T-cell responses after vaccination with NY-ESO-1 peptide, CpG 7909 and Montanide ISA-51: association with survival. Int J Cancer. 2010 Feb 15;126(4):909–18. [PubMed] [Google Scholar]
220. Haining WN, Davies J, Kanzler H, Drury L, Brenn T, Evans J, et al. CpG oligodeoxynucleotides alter lymphocyte and dendritic cell trafficking in humans. Clin Cancer Res. 2008 Sep 1;14(17):5626–34. [PubMed] [Google Scholar]
230. Cowdery JS, Chace JH, Yi A-K, Krieg AM. Bacterial DNA induces NK cells to produce IFNgamma in vivo and increases the toxicity of lipopolysaccharides. J Immunology. 1996;156:4570–5. [PubMed] [Google Scholar]
231. Hartmann G, Krug A, Waller K, Endres S. Oligodeoxynucleotides enhance lipopolysaccharide-stimulated synthesis of TNF: dependence on phosphorothioate modification and reversal by heparin. Molecular Medicine. 1996;2:429–38. [PMC free article] [PubMed] [Google Scholar]
232. Beran J, De CN, Dieussaert I, Van HC. Reactogenicity and immunogenicity of a Lyme disease vaccine in children 2-5 years old. Clin Infect Dis. 2000 Dec;31(6):1504–7. [PubMed] [Google Scholar]
233. Feder HM, Jr., Beran J, Van HC, Abraham B, De CN, Buscarino C, et al. Immunogenicity of a recombinant Borrelia burgdorferi outer surface protein A vaccine against Lyme disease in children. J Pediatr. 1999 Nov;135(5):575–9. [PubMed] [Google Scholar]
234. Pialoux G, Gahery-Segard H, Sermet S, Poncelet H, Fournier S, Gerard L, et al. Lipopeptides induce cell-mediated anti-HIV immune responses in seronegative volunteers1. AIDS. 2001 Jul 6;15(10):1239–49. [PubMed] [Google Scholar]
235. Gahery-Segard H, Pialoux G, Charmeteau B, Sermet S, Poncelet H, Raux M, et al. Multiepitopic B- and T-cell responses induced in humans by a human immunodeficiency virus type 1 lipopeptide vaccine. J Virol. 2000 Feb;74(4):1694–703. [PMC free article] [PubMed] [Google Scholar]
236. Launay O, Durier C, Desaint C, Silbermann B, Jackson A, Pialoux G, et al. Cellular immune responses induced with dose-sparing intradermal administration of HIV vaccine to HIV-uninfected volunteers in the ANRS VAC16 trial. PLoS One. 2007;2(1):e725. [PMC free article] [PubMed] [Google Scholar]
237. Pialoux G, Quercia RP, Gahery H, Daniel N, Slama L, Girard PM, et al. Immunological responses and long-term treatment interruption after human immunodeficiency virus type 1 (HIV-1) lipopeptide immunization of HIV-1-infected patients: the LIPTHERA study. Clin Vaccine Immunol. 2008 Mar;15(3):562–8. [PMC free article] [PubMed] [Google Scholar]
238. Gahery H, Daniel N, Charmeteau B, Ourth L, Jackson A, Andrieu M, et al. New CD4+ and CD8+ T cell responses induced in chronically HIV type-1-infected patients after immunizations with an HIV type 1 lipopeptide vaccine. AIDS Res Hum Retroviruses. 2006 Jul;22(7):684–94. [PubMed] [Google Scholar]
239. Steller MA, Gurski KJ, Murakami M, Daniel RW, Shah KV, Celis E, et al. Cell-mediated immunological responses in cervical and vaginal cancer patients immunized with a lipidated epitope of human papillomavirus type 16 E7. Clin Cancer Res. 1998 Sep;4(9):2103–9. [PubMed] [Google Scholar]
240. Kong NC, Beran J, Kee SA, Miguel JL, Sanchez C, Bayas JM, et al. A new adjuvant improves the immune response to hepatitis B vaccine in hemodialysis patients. Kidney Int. 2008 Apr;73(7):856–62. [PubMed] [Google Scholar]
241. Vandepapeliere P, Rehermann B, Koutsoukos M, Moris P, Garcon N, Wettendorff M, et al. Potent enhancement of cellular and humoral immune responses against recombinant hepatitis B antigens using AS02A adjuvant in healthy adults. Vaccine. 2005 Apr 8;23(20):2591–601. [PubMed] [Google Scholar]
242. Bienzle U, Gunther M, Neuhaus R, Vandepapeliere P, Vollmar J, Lun A, et al. Immunization with an adjuvant hepatitis B vaccine after liver transplantation for hepatitis B-related disease. Hepatology. 2003 Oct;38(4):811–9. [PubMed] [Google Scholar]
243. Harper DM, Franco EL, Wheeler C, Ferris DG, Jenkins D, Schuind A, et al. Efficacy of a bivalent L1 virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and 18 in young women: a randomised controlled trial. Lancet. 2004 Nov 13;364(9447):1757–65. [PubMed] [Google Scholar]
244. Harper DM, Franco EL, Wheeler CM, Moscicki AB, Romanowski B, Roteli-Martins CM, et al. Sustained efficacy up to 4.5 years of a bivalent L1 virus-like particle vaccine against human papillomavirus types 16 and 18: follow-up from a randomised control trial. Lancet. 2006 Apr 15;367(9518):1247–55. [PubMed] [Google Scholar]
245. Stanberry LR, Spruance SL, Cunningham AL, Bernstein DI, Mindel A, Sacks S, et al. Glycoprotein-D-adjuvant vaccine to prevent genital herpes. N Engl J Med. 2002 Nov 21;347(21):1652–61. [PubMed] [Google Scholar]
246. Sokal EM, Hoppenbrouwers K, Vandermeulen C, Moutschen M, Leonard P, Moreels A, et al. Recombinant gp350 vaccine for infectious mononucleosis: a phase 2, randomized, double-blind, placebo-controlled trial to evaluate the safety, immunogenicity, and efficacy of an Epstein-Barr virus vaccine in healthy young adults. J Infect Dis. 2007 Dec 15;196(12):1749–53. [PubMed] [Google Scholar]
247. Velez ID, Gilchrist K, Martinez S, Ramirez-Pineda JR, Ashman JA, Alves FP, et al. Safety and immunogenicity of a defined vaccine for the prevention of cutaneous leishmaniasis. Vaccine. 2009 Dec 11;28(2):329–37. [PubMed] [Google Scholar]
248. Goepfert PA, Tomaras GD, Horton H, Montefiori D, Ferrari G, Deers M, et al. Durable HIV-1 antibody and T-cell responses elicited by an adjuvanted multi-protein recombinant vaccine in uninfected human volunteers. Vaccine. 2007 Jan 5;25(3):510–8. [PubMed] [Google Scholar]
249. Bojang KA, Milligan PJ, Pinder M, Vigneron L, Alloueche A, Kester KE, et al. Efficacy of RTS,S/AS02 malaria vaccine against Plasmodium falciparum infection in semi-immune adult men in The Gambia: a randomised trial. Lancet. 2001 Dec 8;358(9297):1927–34. [PubMed] [Google Scholar]
250. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Milman J, et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet. 2004 Oct 16;364(9443):1411–20. [PubMed] [Google Scholar]
251. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Aide P, et al. Duration of protection with RTS,S/AS02A malaria vaccine in prevention of Plasmodium falciparum disease in Mozambican children: single-blind extended follow-up of a randomised controlled trial. Lancet. 2005 Dec 10;366(9502):2012–8. [PubMed] [Google Scholar]
252. von EK, Morrison R, Braun M, Ofori-Anyinam O, De KE, Pavithran P, et al. The candidate tuberculosis vaccine Mtb72F/AS02A: Tolerability and immunogenicity in humans. Hum Vaccin. 2009 Jul;5(7):475–82. [PubMed] [Google Scholar]
253. Mitchell MS. Perspective on allogeneic melanoma lysates in active specific immunotherapy. Semin Oncol. 1998 Dec;25(6):623–35. [PubMed] [Google Scholar]
254. Sosman JA, Unger JM, Liu PY, Flaherty LE, Park MS, Kempf RA, et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol. 2002 Apr 15;20(8):2067–75. [PubMed] [Google Scholar]
255. Miles DW, Towlson KE, Graham R, Reddish M, Longenecker BM, Taylor-Papadimitriou J, et al. A randomised phase II study of sialyl-Tn and DETOX-B adjuvant with or without cyclophosphamide pretreatment for the active specific immunotherapy of breast cancer. Br J Cancer. 1996 Oct;74(8):1292–6. [PMC free article] [PubMed] [Google Scholar]
256. Butts C, Murray N, Maksymiuk A, Goss G, Marshall E, Soulieres D, et al. Randomized phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol. 2005 Sep 20;23(27):6674–81. [PubMed] [Google Scholar]
257. North SA, Graham K, Bodnar D, Venner P. A pilot study of the liposomal MUC1 vaccine BLP25 in prostate specific antigen failures after radical prostatectomy. J Urol. 2006 Jul;176(1):91–5. [PubMed] [Google Scholar]
258. Vantomme V, Dantinne C, Amrani N, Permanne P, Gheysen D, Bruck C, et al. Immunologic analysis of a phase I/II study of vaccination with MAGE-3 protein combined with the AS02B adjuvant in patients with MAGE-3-positive tumors. J Immunother. 2004 Mar;27(2):124–35. [PubMed] [Google Scholar]
259. Marchand M, Punt CJ, Aamdal S, Escudier B, Kruit WH, Keilholz U, et al. Immunisation of metastatic cancer patients with MAGE-3 protein combined with adjuvant SBAS-2: a clinical report. Eur J Cancer. 2003 Jan;39(1):70–7. [PubMed] [Google Scholar]
260. Atanackovic D, Altorki NK, Stockert E, Williamson B, Jungbluth AA, Ritter E, et al. Vaccine-induced CD4+ T cell responses to MAGE-3 protein in lung cancer patients. J Immunol. 2004 Mar 1;172(5):3289–96. [PubMed] [Google Scholar]
261. Smith BD, Kasamon YL, Kowalski J, Gocke C, Murphy K, Miller CB, et al. K562/GM-CSF immunotherapy reduces tumor burden in chronic myeloid leukemia patients with residual disease on imatinib mesylate. Clin Cancer Res. 2010 Jan 1;16(1):338–47. [PMC free article] [PubMed] [Google Scholar]
262. 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 Mar 30;102(7):1129–36. [PMC free article] [PubMed] [Google Scholar]
-