Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2012 Aug 1;1(5):699–716. doi: 10.4161/onci.20696

Trial Watch

Experimental Toll-like receptor agonists for cancer therapy

Lorenzo Galluzzi 1,2,, Erika Vacchelli 2,3,4,, Alexander Eggermont 2, Wolf Hervé Fridman 1,5,6,7, Jerome Galon 1,5,6,7,8, Catherine Sautès-Fridman 1,6,8, Eric Tartour 5,7,9, Laurence Zitvogel 2,10, Guido Kroemer 1,4,6,7,11,*
PMCID: PMC3429574  PMID: 22934262

Abstract

Toll-like receptors (TLRs) are prototypic pattern recognition receptors (PRRs) best known for their ability to activate the innate immune system in response to conserved microbial components such as lipopolysaccharide and double-stranded RNA. Accumulating evidence indicates that the function of TLRs is not restricted to the elicitation of innate immune responses against invading pathogens. TLRs have indeed been shown to participate in tissue repair and injury-induced regeneration as well as in adaptive immune responses against cancer. In particular, TLR4 signaling appears to be required for the efficient processing and cross-presentation of cell-associated tumor antigens by dendritic cells, which de facto underlie optimal therapeutic responses to some anticancer drugs. Thus, TLRs constitute prominent therapeutic targets for the activation/intensification of anticancer immune responses. In line with this notion, long-used preparations such as the Coley toxin (a mixture of killed Streptococcus pyogenes and Serratia marcescens bacteria) and the bacillus Calmette-Guérin (BCG, an attenuated strain of Mycobacterium bovis originally developed as a vaccine against tuberculosis), both of which have been associated with consistent anticancer responses, potently activate TLR2 and TLR4 signaling. Today, besides BCG, only one TLR agonist is FDA-approved for therapeutic use in cancer patients: imiquimod. In this Trial Watch, we will briefly present the role of TLRs in innate and cognate immunity and discuss the progress of clinical studies evaluating the safety and efficacy of experimental TLR agonists as immunostimulatory agents for oncological indications.

Keywords: agatolimod, CpG-7909, MYD88, resiquimod, TRIF, VTX-2337

Introduction

The Toll gene was first identified in 1985 as a controller of the dorsal-ventral embryonic polarity in the fruit fly Drosophila melanogaster.1,2 A few years later, Toll was shown to encode a transmembrane protein with a large extracellular domain containing at least 15 repeats of a 24 residue-long leucine-rich sequence.3 However, it was not until 1996 that Jules Hoffmann’s laboratory characterized the critical role of Toll in the response of D. melanogaster to fungal infections.4 In 1991, Gay and Keith discovered that the intracellular tails of Drosophila Toll and of the human receptor for interleukin-1 (IL-1) share consistent homology,5 yet the first human ortholog of Toll (initially named TIL for Toll/Interleukin 1 receptor-like) was discovered as late as in 1994,6 and mapped to chromosome 4p14 in 1996.7 In 1997, another member of the Toll-like receptor (TLR) family, namely TLR4, was shown to control the expression of genes involved in innate immunity.8 One year later, two independent laboratories demonstrated that TLR2 and TLR4 mediate the activation of innate immunity in response to bacterial lipopolysaccharide (LPS).9,10 Since then, 13 different TLRs have been characterized in mammals (10 in humans), and members of the TLR family have been identified in organisms as evolutionary distant from mammals as plants and pufferfish.11-13 The discovery of TLRs has granted to Jules Hoffmann and Bruce Beutler the 2011 Nobel prize in Medicine or Physiology.

TLRs are single membrane-spanning proteins that are devoid of enzymatic activity but can bind so-called pathogen-associated molecular patterns (PAMPs), i.e., conserved microbial components including bacterial (e.g., LPS and other constituents of the cell wall) and viral (e.g., double-stranded RNA, dsRNA) products.14 As TLRs recognize microbial products irrespective of their pathogenic potential, it has recently been suggested to abandon the term PAMP in favor of the more correct variant MAMP, standing for “microbe-associated molecular pattern.”15 According to currently accepted models, TLRs operate as homo- or hetero-dimers and are expressed either at the plasma membrane (TLRs that mainly bind proteo-lipidic MAMPs, i.e., TLR1, TLR2, TLR4, TLR5, TLR6 and TLR10) or in endosomes (TLRs that detect microbial nucleic acids, i.e., TLR3, TLR7, TLR8, TLR9).16 TLR10, which is the only orphan receptor among human TLRs, has also been shown to co-localize with TLR2 at phagosomes,17-19 suggesting that it may share with TLR2 the ability to bind acylated lipopeptides.20 Conclusive data on this issue, however, have not yet been reported.

TLR11–13 are not encoded in the human genome. In mice, TLR11–13 are constitutively expressed in the central nervous system and undergo several-fold induction in response to cysticercosis.21 TLR11 reportedly recognizes a profilin-like protein expressed by Toxoplasma gondii and has been localized at the endoplasmic reticulum.22-24 TLR13 also appears to be localized intracellularly, where it would specifically detect the vesicular stomatitis virus.25 So far, the ligand specificity and intracellular localization of TLR12 remain unexplored.

Ligand-bound TLR dimers can recruit the adaptor proteins MYD88 and TIRAP (also known as MYD88 adaptor-like, MAL), in turn driving the assembly of a multiprotein signaling complex that—among other effects—activates the transcriptional factor NFκB (which regulates a large number of pro-inflammatory cytokines).26,27 MYD88 is employed as an obligate or facultative adaptor by all human TLRs but TLR3. TLR3 dimers signal indeed via a MYD88-independent molecular cascade, involving the protein TICAM1 (also known as TRIF) and resulting in the activation of the transcription factor IRF7 (which regulates the expression of Type I interferons, IFNs, and other IFN-inducible genes).28-30 Such a MYD88-independent signaling pathway can also be engaged by TLR4, as demonstrated by the fact that TICAM1-deficient mice exhibit defects in both TLR3- and TLR4-mediated IRF activation.29 Of note, mice lacking TICAM2 (a TICAM1-related protein also known as TRAM), exhibit defective cytokine production in response to TLR4 (but not TLR3) ligands, indicating that TICAM1 is required—but not sufficient—to transduce MYD88-independent signals elicited by TLR4 (but not by TLR3).31 Recent data demonstrate that MYD88 and TIRAP actively inhibit TLR3 signaling,32,33 corroborating the notion that TLR3 constitutes a rather peculiar member of the TLR family, sharing limited functional properties with its homologs. Of note, most TLRs are predominantly expressed in immune cells (e.g., monocytes, mature macrophages, mast cells and dendritic cells, DCs) and/or in cells involved in the first-line defense against infection (e.g., intestinal epithelial cells) (Table 1).

Table 1. Expression, localization and ligand specificity of human TLRs.

TLR MAMP DAMP Localization Expression Ref.
TLR1
Lipoproteins
Triacylated lipopeptides
Zymosan
Unknown
Plasma membrane
B cells
Mature macrophages
Monocytes
260,261
TLR2
Acylated lipopeptides
Amphotericin B
Atypical LPS
Byglican
Forins
Glyco(phospho)lipids
Lipoteichoic acid
Peptidoglycan
Phenol soluble modulin
Zymosan
Heat-shock proteins
Surfactant protein A
Uric acid
Plasma membrane
DCs
Mast cells
Mature macrophages
Monocytes
34-36,
39-41,
60-62,
262
TLR3
Unknown
dsRNA
Endosome
B cells
DCs
Epithelial cells
Endothelial cells
16,91-93
TLR4
Glycolipids
LPS
Biglycan
Fibrinogen
Fibronectin
Heparan sulfate
HMGB1
Heat-shock proteins
Hyaluronic acid
Surfactant protein A
Uric acid
Plasma membrane
DCs
IECs
Mast cells
Mature macrophages
Monocytes
10,16,
35-38,
40,47,64
TLR5
Flagellin
Unknown
Plasma membrane
DCs
IECs
Mature macrophages
Monocytes
16,235-237
TLR6
Diacylated lipopeptides
Lipoproteins
Lipoteichoic acid
Zymosan
Unknown
Plasma membrane
B cells
Mast cells
Mature macrophages
Monocytes
Neutrophils
61,62,
263,264
TLR7
ssRNA of microbial origin
Unknown
Endosome
B cells
Mature macrophages
Monocytes
Plasmacytoid DCs
16,154,155
TLR8
ssRNA of microbial origin
Unknown
Endosome
Mast cells
Mature macrophages
Monocytes
16,154,155
TLR9
Unmethylated CpG-rich DNA
Unknown
Endosome
B cells
Mature macrophages
Monocytes
Plasmacytoid DCs
16,178,
179,265
TLR10 Acylated lipopeptides Unknown Plasma membrane B cells
Plasmacytoid DCs
17-20

Abbreviations: DAMP, damage-associated molecular pattern; DC, dendritic cell; dsRNA, double-stranded RNA; HMGB1, high mobility group B1; IEC, intestinal epithelial cell; LPS, lipopolysaccharide; MAMP, microbe-associated molecular pattern; ODN, oligodeoxynucleotide; ssRNA, single-stranded RNA.

Importantly, TLRs (in particular TLR2 and TLR4) bind not only to microbial products but also to a wide array of endogenous damage-associated molecular patterns (DAMPs), i.e., molecules that are released or exposed by stressed or dying cells and act as endogenous danger signals to promote sterile inflammation.34 These products encompass heat-shock proteins (e.g., HSP60, HSP70),35,36 the non-histone chromatin-binding protein HMGB1,37,38 uric acid,39 the surfactant protein A,40 as well as several components or breakdown products of the extracellular matrix.41 Specific DAMPs, including (though perhaps not limited to) the endoplasmic reticulum protein calreticulin (CRT),42,43 ATP,44-46 HMGB137 and HSP70,47 are generated—in a highly defined spatiotemporal pattern—by cancer cells as they succumb to radiotherapy or to a restricted group of chemotherapeutic agents (i.e., mitoxantrone, cyclophosphamide, oxaliplatin and doxorubicin).48,49 By binding to pattern-recognition receptors (PRRs) expressed on the surface of cells from the immune system (notably DCs), such a spatiotemporally defined combination of signals stimulates tumor-specific immune responses that, at least in some settings, underlie the success of anticancer therapy.49,50 In this context, TLR4 appears to bind both HSP70 and HMGB1, thus enhancing the processing of tumor antigens by DCs and their cross-presentation to T cells.37,47,51 In line with these observations, loss-of function polymorphisms of TLR4 have been shown to negatively influence the response of breast cancer patients to adjuvant radiotherapy or anthracyclin-based chemotherapy.37 Thus, TLRs are crucial not only for the orchestration of innate responses to infectious agents but also for the optimal activation of the immune system against cancer.

Given these premises, it was no surprise to discover that long-used remedies such as the Coley toxin (a mixture of killed Streptococcus pyogenes and Serratia marcescens bacteria) and the bacillus Calmette-Guérin (BCG, an attenuated strain of Mycobacterium bovis initially developed as an anti-tuberculosis vaccine), both of which have been associated with consistent anticancer responses, are potent activators of TLR2 and TLR452,53 that operate in a MYD88-dependent fashion.54 Along similar lines, imiquimod (a small imidazoquinoline derivative originally developed as a topic antiviral agent) was found to activate the TLR7-MYD88 axis, leading to the production of IFNα, tumor necrosis factor α (TNFα) and IL-6, as late as in 2002, five years after its initial approval by FDA for the treatment of actinic keratosis (a precancerous lesion of the skin).55 The use of the Coley toxin has been discontinued in the early 1960s, due to concerns following the thalidomide case (thalidomide was withdrawn 11 y after its approval as it was found to be highly teratogenic, leading to more than 10,000 children born with deformities worldwide).56 Conversely, both BCG and imiquimod are currently approved by FDA, the former for the immunotherapy of in situ bladder carcinoma and the latter against actinic keratosis, superficial basal cell carcinoma, and external genital warts (condylomata acuminata).57 Importantly, monophosphoryl lipid A (MPL), a derivative of the LPS of Salmonella minnesota that functions as a potent agonist or the TLR4-TICAM1 signaling axis,58 is approved by FDA for use in humans as part of Cervarix®, a vaccine against human papillomavirus 16 and 18 (the etiological agents of about 70% of cervical carcinoma cases).59

In this Trial Watch, we will review the results of recently completed clinical trials and discuss the progress of ongoing studies that have employed/are employing experimental TLR agonists as immunostimulatory agents for oncological indications.

TLR2 and TLR4 Agonists

TLR2 and TLR4 are predominantly expressed on the surface of monocytes, mature macrophages and DCs.16 Assisted by CD36, homodimeric TLR2 specifically recognizes products from Gram-positive bacteria, including lipoteichoic acid (LTA).60 As an alternative, TLR2 can heterodimerize with TLR1 or TLR6, thereby acquiring the capacity to bind triacylated lipopeptides, such as the synthetic molecule Pam3CSK4, and biacylated lipopeptides, like the natural occurring macrophage-activating lipopeptide 2 (MALP-2), respectively.61,62 Contrarily to TLR2, TLR4 binds components from Gram-negative bacteria, mainly LPS, with the assistance of CD14.63 This process is highly facilitated by the LPS-binding protein (LBP), an acute phase soluble protein that binds LPS and favors its interaction with the TLR4/CD14 complex.64

SMP-105 is a TLR2-specific agonist consisting of cell wall skeleton components (mainly mycolic acid and peptidoglycans) isolated from M. bovis (strain BCG Tokyo).65 Peritoneal exudate cells from Tlr2−/− and Myd88−/− (but not Tlr4−/−) mice fail to secrete TNFα and IL-6 in response to SMP-105, confirming the strict TLR selectivity of this agent.66 Mice receiving inactivated Lewis lung carcinoma cells together with SMP-105 develop an intense immune response featuring high levels of IFNγ-producing tumor-specific cytotoxic lymphocytes (CTLs).66 Such a response has been shown to efficiently control the growth of implanted tumors.66 Thus, SMP-105 exerts potent immunostimulatory effects in vivo. Nevertheless, the development of SMP-105 has not yet reached the clinic.

The anticancer properties of LPS, the natural ligand of TLR2 and TLR4 also known as endotoxin, have been discovered as early as in the 1960s,67,68 when the existence of TLRs was not even suspected. Since then, dozens of studies have confirmed that the administration of LPS, either alone or in combination with other anticancer agents, efficiently induce tumor regression in murine models.69-71 Early clinical trials sought to determine the safety of LPS, with particular attention to the fact that circulating LPS might cause the sequential release into the circulation of large amounts of pro-inflammatory cytokines, a potentially lethal process that is known as endotoxic shock.72,73 These studies determined that low doses of LPS are relatively safe, in particular if preceded by a bolus of the non-steroidal anti-inflammatory drug ibuprofene, even though virtually all patients receiving LPS showed increased circulating levels of pro-inflammatory cytokines including TNFα and IL-1.74,75 A few years later, the combination ibuprofen + LPS (purified from Salmonella abortus equi) was tested in colorectal cancer patients, showing encouraging results, as well as in non-small cell lung carcinoma (NSCLC) patients, for whom the therapeutic benefit was marginal.76 Today, official sources list only three clinical trials investigating the efficacy of LPS in oncological indications: one as part of a complex cell-based vaccination approach to treat Ewing sarcoma, rhabdomyosarcoma and neuroblastoma patients (NCT00923351), one in which LPS is co-administered with a peptide vaccine and a mineral oil-based adjuvant to melanoma patients (NCT01585350), and one in which LPS is combined with peptide-pulsed DCs for the treatment of hematological malignancies (NCT00923910) (source www.clinicaltrials.gov).

Picibanil (OK-432) is a lyophilized preparation of group A Streptococcus (namely Streptococcus pyogenes) that has been approved for use as an anticancer adjuvant as early as in 1975, yet only by the Japanese Ministry of Health and Welfare.41,77 Picibanil was soon found to induce potent Th1 immune responses in vivo, in mice, but its ability to bind and activate TLR4 on the surface of DCs has been unveiled only a few years ago.78,79 Since its approval by local authorities, picibanil has been extensively used in Japan for the treatment of cervical, gastric and oral squamous cell carcinoma.80-82 In the US and Europe, picibanil remains an investigational drug, and has recently been tested for its ability to boost immune responses in cancer patients treated with whole protein-based vaccines (NCT00291473). The results of this study, however, have not yet been divulgated. In addition, picibanil is currently being investigated in combination with a preoperative intratumoral injection of DCs in patients with resectable pancreatic cancer (NCT00795977), and associated to DCs and chemotherapy (cyclophosphamide + docetaxel) in head and neck squamous cell carcinoma (HNSCC) patients (NCT01149902) (source www.clinicaltrials.gov).

OM-174 (also known as CRX-527) is a water soluble, diphosphorylated and triacetylated form of lipid-A purified from Escherichia coli.83 OM-174, which functions as a mixed TLR2/TLR4 agonist,84 has been shown to exacerbate the efficacy of cyclophosphamide against murine melanoma B16 cells growing in immunocompetent mice,85 and to exert radiosensitizing effects in vivo, in an IFNγ-dependent fashion.86 Relatively recent publications indicate that the safety and efficacy of OM-174 have been tested in cancer patients.16,87 However, we were unable to find additional sources that would report the results of these trials. Now, the clinical development of OM-174 has been discontinued (source www.clinicaltrials.gov).

In summary, while many pure TLR4 and mixed TLR2/TLR4 agonists (including BCG, MPL and picibanil) have already been approved for use in humans by governmental agencies, the clinical development of pure TLR2 agonists as adjuvants to boost anticancer immune responses now appears to stand at an impasse. At least in part, this may be due to the fact that TLR2 agonists promote tumor infiltration by FOXP3+ T cells (Tregs) and stimulate the production of IL-10, de facto exerting local and systemic immunosuppressive effects.88,89 Along similar lines, the possibility to employ TLR4 antagonists for cancer-unrelated indications (mainly sepsis) raised initial enthusiasms that often have been deceived.16 One notable exception is provided by the small chemical ibudilast (AV411),90 which is currently being tested in (a few) Phase I/II clinical trials for the treatment of addiction withdrawal and headache (source www.clinicaltrials.gov).

Table 2 summarizes recent clinical progress of experimental TLR2/TLR4 agonists for oncological indications.

Table 2. Clinical trials evaluating TLR2/TLR4 agonists in oncological indications*.

Compound Indication Trials Phase Status Notes Ref.
LPS
Ewing's sarcoma
Neuroblastoma
Rhabdomyosarcoma
1
I-II
Suspended
Combined with 8H9 mAb, IL-4 and KLH-pulsed DCs vaccine
NCT00923351
Hematological malignancies
1
I-II
Recruiting
Combined with IL-4, KLH and a WT1-pulsed DC-based vaccine
NCT00923910
Melanoma
1
I
Not yet recruiting
Combined with a
multipeptide vaccine
NCT01585350
Picibanil Pancreatic cancer
Head and neck carcinoma
1
1
I-II
I
Active,
not recruiting
Recruiting
Combined with preoperative intratumoral injections of DCs
Combined with chemo-immunotherapy
NCT00795977
NCT01149902

Abbreviations: DC, dendritic cell; IL-4, interleukin 4; KLH, keyhole limpet hemocyanin; mAb, monoclonal antibody; LPS, lipopolysaccharide. *Started after January, 1st 2008.

TLR3 Agonists

TLR3 is an endosomal TLR primarily expressed by B cells, myeloid DCs as well as by epithelial and endothelial cells.16 At an acidic pH (such as that found in the endosomal lumen), TLR3 binds 40–50 nucleotide-long dsRNA molecules, interacting almost exclusively with the sugar phosphate backbone rather than with bases.91-93 Intriguingly, the interaction of dsRNA with TLR3 does not trigger conformational changes, but only facilitate/stabilize homodimerization, de facto bringing the intracellular tails of TLR3 monomers in close proximity of each other.93 Of note, in some epithelial cell types including breast cancer cells, ligand-bound TLR3 can stimulate the intrinsic pathway of apoptosis94-96 or trigger a chemokine cascade that orchestrates the recruitment of CTLs to the tumor bed.97,98

Ampligen (also known as rintatolimod) consists in polyriboinosinic polyribocytidylic acid (polyI:C), a synthetic dsRNA molecule originally developed in the late 1960s to exacerbate antiviral responses.99 In vitro, Ampligen not only optimally activates DCs (be them derived from healthy donors or cancer patients) to promote cytotoxic Th1 responses,100 but also exerts direct cytostatic/cytotoxic effects against a wide spectrum of tumor cells.101,102 In vivo, Ampligen has been shown to suppress the growth of a large panel of neoplasms, in both (at least partially) immunodeficient103-106 and immunocompetent models.107-110 In the latter scenario, Ampligen appears to operate in a multimodal fashion, encompassing the activation of natural killer (NK) cells107 the conversion of tumor-promoting macrophages into their tumor-suppressing counterparts,110 as well as direct cytostatic/cytotoxic effects on cancer cells.111 Of note, all these responses have been shown to be abolished in Ticam1−/−, but not in Myd88−/−, mice.107,110 Recently, Shir et al. have demonstrated that an epidermal growth factor receptor (EGFR)-targeted variant of Ampligen is highly efficient in activating (implanted) human immune cells to destroy pre-established disseminated EGFR-overexpressing tumors, with no adverse toxic effects.112 This suggests that specifically targeting TLR3 ligands to tumor cells might increase their therapeutic potential. Early clinical trials in oncological settings demonstrate that Ampligen is well tolerated, even over long administration periods (> 1 y), and that patients normally do not develop neutralizing antibodies.113 Today, Ampligen is being tested in combination with a peptide vaccine in HER2-positive breast cancer patients (NCT01355393), with autologous tumor cell lysates in patients affected by reproductive tract neoplasms (NCT01312389), or with celecoxib, IFNα and Hiltonol (see below) in resectable colorectal cancer patients (NCT01545141) (source www.clinicaltrials.gov).

Hiltonol (also known as poly-ICLC) is a particular formulation of polyI:C that includes carboxymethylcellulose and poly-l-lysine as stabilizing agents.114 Hiltonol appears to be 5 to 10 times more resistant to degradation by serum hydrolases than Ampligen, and exhibits a denaturation temperature about 40°C higher.114 In line with this notion, Hiltonol has been shown to significantly elevate the levels of circulating IFNγ in non-human primates in conditions in which an equivalent dose of Ampligen failed to do so,114 and to exert potent immunostimulatory effects in multiple preclinical models, including mice, rats, rabbits, monkeys and chimpanzees.114-118 Such encouraging preclinical results generated a consistent wave of Phase I/II clinical trials that assessed the safety and efficacy of Hiltonol (often as a standalone agent or combined with vaccination protocols) in multiple, very heterogeneous oncological indications, encompassing hematological malignancies,119-122 brain tumors,120,123-128 melanoma,129 ovarian carcinoma,130 renal cancer121,131 and other advanced neoplasms.132-135 With the exception of a single study, reporting substantial toxicity and no clinical benefits in ovarian carcinoma patients,130 these trials concluded that Hiltonol is safe and moderately efficient in boosting anticancer immune responses. More recently, Hiltonol has been investigated as a single agent or combined to radiotherapy in B- and T-cell lymphoma patients (NCT00880867), in combination with a chimeric vaccine and resiquimod (see below) in patients affected by bladder carcinoma (NCT01094496),136 as well as together with a peptide vaccine in patients bearing neoplasms of the reproductive tract. The former two studies have been terminated for unreported reasons, whereas the latter has been completed, though the results have not yet been divulgated. Nowadays, the safety and efficacy of Hiltonol are being investigated in around 20 Phase I/II clinical trials. In all but two cases, one in which Hiltonol is used as a single agent in glioma patients (NCT1188096) and one in which it is combined with celecoxib, IFNα and Ampligen (see above) in resectable colorectal cancer patients (NCT01545141), Hiltonol is employed as an adjuvant to peptide cell-based anticancer vaccines (source www.clinicaltrials.gov). Major oncological indications in which Hiltonol is being tested include brain malignancies (6 trials), melanoma (4 trials), breast carcinoma (2 trials) and colorectal cancer (2 trials).

The high molecular mass dsRNA mimic IPH-3102 has been shown to promote NFκB activation and to sustain Type I IFN responses in vitro, hence exerting cytotoxic effects against melanoma and breast cancer cells. In addition, IPH-3102 mediates consistent immunostimulatory effects in vivo, in mice.16,137 Still, IPH-3102 has not yet entered (and presumably will never enter) clinical trials.

Polyadenylic polyuridylic acid (polyA:U) is a synthetic dsRNA molecule that has been shown to exert immunostimulatory effects in vivo as early as in 1967.138 Still, polyA:U has not been recognized as a TLR3 agonist until 2008, when polyA:U was reported to stimulate myeloid DCs to produce IL-12.139 When combined with anticancer vaccines, poly(A:U) promotes Th1 responses that control tumor growth and are associated with the establishment of immunological memory.140 However, TLR3 is expressed not only in immune cells but also in epithelial, endothelial and cancer cells,96,141-143 Thus, poly(A:U) can exert, at least in some settings, pro-tumor functions,144 raising the need for compensatory strategies that would uncouple the immunostimulatory (and therapeutically beneficial) effects of TLR3 agonists from their pro-tumorigenic potential.97 During the last three decades, poly(A:U) has been demonstrated to be safe but variably efficient for the adjuvant therapy of resectable breast cancer (as a single agent or in combination with radiotherapy),145,146 locally advanced gastric cancer (combined with 5-fluorouracil and doxorubicin),147,148 resectable colorectal carcinoma (as a standalone agent),149 and other not better specified solid malignancies.150,151 Of note, the therapeutic benefits of poly(A:U) in combination with locoregional radiotherapy as an adjuvant treatment of breast cancer patients, as reported by Laplanche et al.,146 have recently been shown to be restricted to patients bearing TLR3+ tumors.98 This indicates that TLR3 expression levels might constitute a predictive biomarker for the efficacy of poly(A:U), at least in some oncological setting. Today, the development of poly(A:U) appears to have come at an impasse, as there are no clinical trials evaluating its efficacy for oncological as well as for cancer-unrelated indications (source www.clinicaltrials.gov).

Table 3 summarizes recent clinical progress of experimental TLR3 agonists for oncological indications.

Table 3. Clinical trials evaluating TLR3 agonists in oncological indications*.

Compound Indication Trials Phase Status Notes Ref.
Ampligen
Breast cancer
1
I-II
Recruiting
Combined with GM-CSF and a
HER-2-based peptide vaccine
NCT01355393
Colorectal cancer
1
I-II
Not yet recruiting
Combined with celecoxib,
Hiltonol and IFNα2b
NCT01545141
Reproductive tract cancer
1
I-II
Active, not recruiting
Combined with tumor cell lysates and multiple adjuvants
NCT01312389
Hiltonol Advanced cancer
1
I-II
Recruiting
Combined with CDX-1401 ± resiquimod
NCT00948961
B- and T-cell lymphoma
1
I
Terminated
As single agent or combined
with local radiotherapy
NCT00880867
Bladder cancer
1
II
Terminated
Combined with CDX-1307 vaccine, chemotherapy, GM-CSF and resiquimod
NCT01094496
Brain tumors
6
n.a.
Recruiting
Combined with a HLA-A2-restricted glioma antigen-based vaccine
NCT01130077
0
Active, not recruiting
NCT00795457
0
Active, not recruiting
NCT00874861
I-II
Active, not recruiting
Combined with DC-based vaccine
NCT00766753
II
Recruiting
As single agent
NCT01188096
II
Recruiting
Combined with autologous
tumor cell lysate-pulsed DCs
NCT01204684
Breast cancer
2
n.a.
Not yet recruiting
Combined with a multipeptide vaccine
NCT01532960
0
Recruiting
Combined with MUC-1-based vaccine
NCT00986609
Colorectal cancer
2
I-II
Not yet recruiting
Combined with celecoxib,
Ampligen and IFNα2b
NCT01545141
II
Recruiting
Combined with MUC-1-based vaccine
NCT00773097
Melanoma
4
I
Not yet recruiting
Combined with a multipeptide vaccine
NCT01585350
I
Recruiting
Combined with a CD40 agonist
and various peptides
NCT01008527
I-II
Recruiting
Combined with a NY-ESO-1-based vaccine ± oil adjuvant
NCT01079741
II
Recruiting
Combined with a multipeptide vaccine
NCT01437605
Myeloma
1
II
Recruiting
Combined with autologous SCT, lenalidomide and a multipeptide vaccine
NCT01245673
Pancreatic cancer
1
I
Recruiting
Combined with peptide-pulsed DCs
NCT01410968
Prostate cancer
1
n.a.
Recruiting
Combined with a multipeptide vaccine
NCT00694551
Reproductive tract cancer 1 I-II Completed Combined with NY-ESO-1 overlapping peptide and oil adjuvant NCT00616941

Abbreviations: DC, dendritic cell; GM-CSF, granulocyte macrophage colony-stimulating factor; HER2, human epidermal growth factor receptor 2; IFN, interferon; n.a., not available; SCT, stem cell transplantation. *Started after January, 1st 2008.

TLR7 and TLR8 Agonists

TLR7 and TLR8 are predominantly expressed at the endosomal membrane of monocytes and macrophages, plasmacytoid DCs (constituting one peculiar subset of DCs that operate at the interface between innate and adaptive immunity),152 and mast cells.16 Moreover, both these endosomal TLRs are highly expressed by bronchial epithelial cells (especially TLR7) and NSCLC cells (especially TLR8), in situ.153 TLR7 and TLR8 bind single-stranded RNA molecules of viral origin, the species specificity relying on the elevated content of uridine and guanosine nucleosides that characterize viral—as compared with mammalian—RNAs.154,155 Binding is inhibited by specific chemical modifications of the RNA molecule (such as the 2′-O-methylation of uridine and guanosine nucleosides).156 This is critical as it allows RNA interference to be used in vivo in the absence of massive DC activation and inflammation.156 Of note, TLR7 stimulation has been associated with the development of tolerance, which can be circumvented by the use of appropriate treatment schedules.157

Resiquimod (R-848) is a small imidazoquinoline that exerts potent immunostimulatory effects, both in vitro158-160 and in vivo,161,162 by operating as a TLR7/TLR8 agonist.55 Thus, similar to its FDA-approved structural analog imiquimod, topical resiquimod efficiently enhances immune responses to local antigens, leading to a consistent increase in the frequency of antigen-specific CTLs.163 Resiquimod has initially been investigated for the therapy of herpes simplex virus, yielding promising results,164 and hepatitis C virus, in this case being associated with dose-limiting toxicity similar to that of IFNα.165 The development of resiquimod as an anti-herpetic agent has been suspended due to lack of efficacy in Phase III clinical trials.16 However, following encouraging results obtained in patients affected by actinic keratosis,166 and perhaps with an influence from the clinical success of imiquimod, resiquimod has recently been tested on patients affected by common warts (which are caused by human papillomaviruses). The results of these six studies, all of which are listed as completed by official sources, have not yet been published. In addition, resiquimod has been evaluated in combination with a chimeric vaccine and Hiltonol (see above) in patients affected by bladder carcinoma (NCT01094496),136 a study that has been terminated for unreported reasons. Today, resiquimod is under investigation as a single agent for the treatment of actinic keratosis (NCT01583816), cutaneous T-cell lymphoma (NCT01497795) and melanoma (NCT00470379), as well as in combination with multiple vaccination strategies in melanoma patients (NCT00960752) and individuals bearing not better specified NY-ESO-1-expressing tumors (NCT00948961, NCT00821652) (source www.clinicaltrials.gov).

852A is another member of the imidazoquinoline group that functions as a TLR7 agonist.167 852A potently stimulates plasmacytoid DCs to produce IFNα as well as several other immunostimulatory cytokines, in vitro and in vivo, thus driving Th1 immune responses that—at least in some models—can exert antitumor effects.167-169 Early clinical trials that investigated the safety and pharmacokinetic profile of 852A reported no major side effects,170 and pointed to subcutaneous delivery as an acceptable means for obtaining systemic responses.171 More recently, intravenous delivery has been preferred as a method to systemically activate the immune system in cancer patients,172 even though prolonged schedules have been associated with a high incidence of mild-to-severe side effects (NCT00319748).173 In addition, the anticancer potential of 852A (always employed as a standalone therapy) has been investigated in unresectable melanoma patients (NCT00091689, NCT00189332), in individuals affected by Barrett's esophagus with high-grade dysplasia (NCT00386594), in patients with hematological malignancies (NCT00276159), as well as in subjects affected by not better specified refractory solid tumors (NCT00276159). Though official sources list these studies as completed, their results have not yet been disclosed (source www.clinicaltrials.gov).

VTX-2337 is a small molecule that specifically binds and activates TLR8.174 The preclinical characterization of VTX-2337 appears to be scarce. Indeed, we found a single scientific work reporting that VTX-2337 stimulates the production TNFα and IL-12 from monocytes and myeloid DCs, as well as the secretion of IFNγ by NK cells, in vitro.174 Nevertheless, VTX-2337 has already entered Phase I/II clinical trials for oncological indications. In particular, the safety and dose-dependent pharmacological profile of VTX-2337 have been demonstrated in a cohort of patients affected by advanced solid tumors (NCT00688415) (ref. 175 and Annual ASCO Meeting 2011, Abstract #2537). A Phase I/II clinical trial in which VTX-2337 was supposed to be used in combination with radiotherapy for the treatment of low-grade B-cell lymphoma has been recently terminated, due to insufficient rates of recruitment (NCT01289210). A similar trial (NCT01396018) is still listed as active by official sources, though the last update is nearly one year old. Thus, VTX-2337 is currently being evaluated in two Phase I/II studies only: in combination with the anti-EGFR antibody cetuximab for the treatment of advanced, recurrent or metastatic HNSCC (NCT01334177) and associated to doxorubicin in patients affected by tumors of the reproductive tract or peritoneal cavity (NCT01294293).

The clinical development of the TLR7 agonists AZD8848 (DSP-3025) and ANA773, as well as that of the TLR8 agonist VTX-1463, which until recently were being tested for the treatment of allergic asthma, allergic rhinitis and hepatitis C virus infection,16,176,177 has been discontinued (source www.clinicaltrials.gov).

TLR7 and TLR8 agonists have recently been shown to increase the resistance of cultured NSCLC cells to chemotherapy, an effect that ensued the activation of a MYD88-NFκB signaling axis resulting in the upregulation of anti-apoptotic BCL-2 proteins.153 Of note, NSCLC cells treated in vitro with TLR7 and TLR8 agonists exhibit a gene expression signature that is very similar to that of untreated NSCLC cells in situ, suggesting that the latter might be chronically stimulated by (endogenous?) TLR ligands.153 These observations suggest that the use of TLR7/TLR8 agonists as adjuvants in anticancer therapy might be beneficial to patients affected by specific neoplasms but detrimental to others, notably to patients whose tumors express high amounts of TLR7 and/or TLR8.

Table 4 summarizes recent clinical progress of experimental TLR7/TLR8 agonists for oncological indications.

Table 4. Clinical trials evaluating TLR7/TLR8 agonists in oncological indications*.

Compound Indication Trials Phase Status Notes Ref.
TLR7/ TLR8 agonists
Resiquimod
Actinic keratosis
1
II
Not yet recruiting
As single agent
NCT01583816
Advanced cancer
1
I-II
Recruiting
Combined with CDX-1401 ± Hiltonol
NCT00948961
Bladder cancer
1
II
Terminated
Combined with CDX-1307 vaccine, chemotherapy, GM-CSF and Hiltonol
NCT01094496
CTCL
1
I-II
Recruiting
As single agent
NCT01497795
Melanoma
1
II
Recruiting
Combined with a gp100- and
MAGE-3-based vaccine
NCT00960752
Various neoplasms
1
I
Unknown
Combined with a NY-ESO-1-based vaccine
NCT00821652
TLR8 agonists
VTX-2337 Advanced cancer
1
I
Completed
As single agent
NCT00688415
B-cell lymphoma
2
I-II
Recruiting
Combined with local radiotherapy
NCT01396018
Terminated
NCT01289210
HNC
1
I
Recruiting
Combined with cetuximab
NCT01334177
Reproductive tract tumors 1 I Recruiting Combined with doxorubicin NCT01294293

Abbreviations: CTCL, cutaneous T-cell lymphoma; HNC, head and neck carcinoma; GM-CSF, granulocyte macrophage colony-stimulating factor. *Started after January, 1st 2008.

TLR9 Agonists

TLR9 is mainly found in the endosomal compartment of B cells, monocytes, macrophages and plasmacytoid DCs.16 The main ligand of TLR9 is bacterial/viral DNA, differing from its mammalian counterpart for the elevated frequency of unmethylated CpG dinucleotides.178 Indeed, whereas mammalian DNA has no immunostimulatory activity, the administration of bacterial/viral DNA induces a potent Th1 immune response in vivo, which is entirely abrogated in Tlr9−/− mice.178 Of note, a TLR9 variant that is ectopically expressed at the plasma membrane has been shown to recognize not only bacterial/viral but also mammalian DNA,179 indicating that the basis for the discrimination between self and exogenous DNA mainly resides in the subcellular localization of TLR9 rather than in biochemical features (i.e., the methylation state and frequency of CpG islands) of its ligand.

CpG-28, CpG-685 (GNKG168) and CpG-7909 (PF-3512676, Agatolimod, Promune®) are class B unmethylated CpG oligonucleotides, i.e., CpG oligonucleotides that—due to precise structural features such a length comprised between 18 and 28 nucleotides, a fully phosphorothioated backbone and the presence of one or more 5′-RYCGYR-3′ 6-mers (R = A or G; Y = T or C)—strongly stimulate B cells and monocytes.180 In some cell types including malignant B cells, TLR9 stimulation by type B CpG oligonucleotides induces significant morphologic, phenotypic and functional alterations that are followed by a signal transducer and activator of transcription 1 (STAT1)-dependent program of apoptotic cell death.181 In line with this notion, human leukemic cells that are pretreated in vitro with type B CpG oligonucleotides lose the ability to generate overt leukemia upon inoculation into immunodeficient mice.181 Moreover, the administration of type B CpG oligonucleotides (alone or combined with chemotherapeutics or peptide vaccines) to tumor-bearing rodents reportedly exerts potent anticancer effects.181-185 Initial Phase I/II clinical trials to test the safety and efficacy of CpG-7909 for oncological indications were launched in April 2000.186 Approximately in the same period, CpG-7909 begun to be extensively investigated as an adjuvant for cancer-unrelated indications (mainly antiviral vaccines), showing no severe side effects and encouraging efficacy.187-189 In humans, the subcutaneous (more than the intravenous) administration of CpG-7909 activates both myeloid and plasmacytoid DCs,190 resulting in a potent systemic Th1 response that can boost anticancer immunity.191-193 During the last decade, the safety and anticancer potential of CpG-7909 (as a standalone agent or in combination with chemotherapy and/or vaccination approaches) have been investigated in a large number of Phase I/II clinical trials, including studies with leukemia,194 lymphoma,195-198 basal cell carcinoma,199 melanoma,199-202 esophageal squamous cell carcinoma,203 NSCLC,204-206 renal cell carcinoma,207 and prostate cancer patients.208 In a peculiar approach, CpG-7909 has been tested as an adjuvant for oncological indications in combination with MPL and QS-21, a water soluble saponin extracted from the South American tree Quillaja saponaria Molina.209 Such a formulation, known as AS15 or GSK2302025A, has been evaluated for its capacity to boost immune responses in melanoma patients receiving recombinant MAGE-A3 (see refs. 210, 211 and Annual ASCO Meeting 2008, Abstracts 9045 and 9065). In the vast majority of cases, CpG-7909 and AS15 were found to be safe and to exert moderate anticancer effects, de facto supporting subsequent clinical trials based on larger patient cohorts. One of these Phase III studies has already been terminated, demonstrating that the addition of CpG-7909 to gemcitabine + cisplatin in NSCLC patients improves neither progression-free nor overall survival.212 Following the disclosure of these negative results, the evaluation of CpG-7909 in NSCLC patients has been discontinued. Nevertheless, CpG-7909 remains under investigation in a few Phase I/II clinical trials, evaluating it in combination with radiotherapy in B-cell lymphoma patients (NCT00880581), combined with the anti-HER2 antibody trastuzumab in breast cancer patients (NCT00824733), and as an adjuvant to anticancer vaccination protocols in patients bearing esophageal malignancies (NCT00669292), or not better specified NY-ESO-1-expressing tumors (NCT00819806). Along similar lines, AS15 is now being extensively tested in multiple myeloma (NCT01380145), melanoma (NCT01149343, NCT01266603, NCT01425749, NCT01437605) and bladder cancer (NCT01435356, NCT01498172) patients, always in combination with MAGE-A3-based vaccines, as well as in metastatic breast cancer patients (NCT00952692), combined with a peptide vaccine targeting HER2. At odds with the large number of studies performed on CpG-7909 and AS15, the safety and efficacy of CpG-685 (as a single agent) are currently being investigated in a single Phase I clinical trial, enrolling patients with relapsed/refractory B-cell chronic lymphocytic leukemia (NCT01035216). Although the results of Phase I/II clinical trials testing CpG-28 in recurrent glioblastoma patients seemed encouraging,213,214 today official sources list no active studies that would evaluate this compound in oncological settings (source www.clinicaltrials.gov).

By virtue of synthetic CpR dinucleotides, the immunomodulatory oligonucleotide 2055 (IMO-2055, also known as EMD1201081) mimics unmethylated CpG sequences and hence functions as a potent TLR9 agonist.215,216 In vivo, IMO-2055 induces Th1 immune responses that are associated with consistent antitumor effects, as demonstrated in murine models of syngenic colon carcinoma and melanoma.217 In addition, IMO-2055 has recently been shown to impair EGFR signaling and to synergize—in an immune system-independent fashion—with cetuximab for the treatment of human colon carcinoma xenografts.218,219 In patients with advanced NSCLC, the use of IMO-2055 in combination with gemcitabine + carboplatin has been associated with no hematological toxicity and signs of efficacy (ref. 220 and Annual AACR Meeting 2006, Abstract #P3–022). Subsequent trials have investigated the safety and efficacy of IMO-2055 as a single agent in patients with recurrent/metastatic renal clear cell carcinoma (NCT00729053), combined with cetuximab + FOLFIRI (folinic acid + 5-fluorouracil + irinotecan) in colorectal cancer patients (NCT00719199) or with cetuximab + 5-fluorouracil + cisplatin in subjects with recurrent/metastatic HNSCC (NCT01360827), and in association with the EGFR inhibitor erlotinib221 + bevacizumab (an anti-angiogenic monoclonal antibody) in NSCLC patients (NCT00633529). The results of these studies, however, have not yet been disclosed. Today, official sources list one single study based on IMO-2055 as active, in which the drug is evaluated (in combination with cetuximab) for the treatment of cetuximab-naïve subjects with recurrent/metastatic HNSCC (NCT01040832) (source www.clinicaltrials.gov).

The immunostimulatory sequence (ISS) 1018 is a single-stranded, 22 bp-long, phosphorothioate oligonucleotide prepared by standard solid-phase chemistry techniques (sequence 5′-TGACTGTGAACGTTCGAGATGA-3′).222 The administration of 1018 ISS to simian immunodeficiency virus (SIV)-infected macaques reportedly results in massive activation of plasmacytoid DCs, IL-12 production and accumulation of IFNγ-secreting SIV-specific T cells.223 Along similar lines, ISS 1018 has been shown to trigger an IL-12-dependent cytotoxic immune response that efficiently prevents the growth of spontaneous B-cell lymphomas in SJL mice.224 Following early clinical trials that demonstrated its safety,225 ISS 1018 has been extensively tested as part of a vaccine against the hepatitis B virus (HEPLISAVTM).226 Moreover, ISS 1018 has been shown to be safe and moderately effective in follicular non-Hodgkin lymphoma patients treated with the anti-CD20 antibody rituximab (NCT00251394).227-229 Of note, the results of Phase I clinical trial in which ISS 1018 was combined with irinotecan + cetuximab to treat metastatic colorectal carcinoma patients have not been released yet (NCT00403052) (source www.clinicaltrials.gov).

MGN-1703 and MGN-1706 belong to the class of DNA-based double stem-loop immunomodulators (dSLIMs), i.e., IMOs that contain a phosphodiester, rather than a phosphorothioate, backbone and fold in a dumbbell-like covalently-closed structure.230 dSLIMs stimulate cultured peripheral blood mononuclear cells (PBMCs) from healthy donors to secrete Th1 cytokines, including IFNγ, IL-2 and IL-12.230 Moreover, dSLIMs have been shown to enhance the antileukemic effects of peptide vaccines combined with the granulocyte monocyte colony stimulating factor (GM-CSF), in vivo.231 Seventeen metastatic colorectal cancer patients receiving dSLIMs in combination with chemotherapy and a peptide vaccine developed no severe toxicity, yet in this setting the adjuvant failed to boost anticancer immune responses.232 More recently, the safety of MGN-1703 has been confirmed in patients with advanced colorectal carcinoma showing disease control after first-line standard chemotherapy (see refs. 233, 234 and Annual ASCO Meeting 2011, Abstract 618; Annual ASCO Meeting 2012, Abstract 633). This study, which intends to evaluate not only the safety but also the efficacy of MGN-1703 as a single agent for maintenance therapy, is still ongoing (NCT01208194) (source www.clinicaltrials.gov).

The clinical development of additional TLR9 agonists including AVE0675, IMO-2125, QAX-935 and SAR-21609 and SD-101, which until recently were being tested for the treatment of asthma/allergy or hepatitis C virus infection,16,176,177 has been discontinued. On the contrary, DIMS0150 (kappaproct) is under investigation in a Phase III study involving patients with ulcerative colitis (NCT01493960) (source www.clinicaltrials.gov).

Table 5 summarizes recent clinical progress of experimental TLR9 agonists for oncological indications.

Table 5. Clinical trials evaluating TLR9 agonists in oncological indications*.

Compound Indication Trials Phase Status Notes Ref.
Agatolimod
B-cell lymphoma
1
II
Recruiting
Combined with radiotherapy
NCT00880581
Bladder cancer
2
I
Enrolling by invitation
Combined with BCG, MPL and recMAGE-A3
NCT01498172
II
Recruiting
Combined with MPL and recMAGE-A3
NCT01435356
Breast cancer
2
I-II
Active, not recruiting
Combined with MPL, a HER2-based vaccine and lapatinib
NCT00952692
II
Recruiting
Combined with trastuzumab
NCT00824733
Esophageal cancer
1
I-II
Recruiting
Combined with a
multipeptide vaccine
NCT00669292
Melanoma
4
0
Recruiting
Combined with MPL and recMAGE-A3
NCT01425749
I
Combined with MPL
NCT01149343
II
Combined with IL-2, MPL and
recMAGE-A3
NCT01266603
II
Combined with MPL, recMAGE-A3 ± Hiltonol
NCT01437605
Multiple myeloma
1
I
Recruiting
Combined with MPL and recMAGE-A3
Recruiting
NY-ESO-1-expressing tumors
1
I-II
Recruiting
Combined with a multipeptide vaccine ± cyclophosphamide
NCT00819806
GNKG168
B-CLL
1
I
Recruiting
As single agent
NCT01035216
IMO-2055
Colorectal cancer
1
I
Terminated
Combined with cetuximab
and FOLFIRI
NCT00719199
Head and neck carcinoma
2
I
Terminated
Combined with 5-FU,
cisplatin and cetuximab
NCT01360827
II
Active, not recruiting
Combined with cetuximab
NCT01040832
NSCLC
1
I
Completed
Combined with erlotinib
and bevacizumab
NCT00633529
RCC
1
II
Completed
As single agent
NCT00729053
MGN-1703 Colorectal cancer 1 II Recruiting As single agent NCT01208194

Abbreviations: B-CLL, B-cell chronic lymphocytic leukemia; FOLFIRI, folinic acid, 5-FU, irinotecan; 5-FU, fluorouracil; IL-2, interleukin 2; MPL, monophosphoryl lipid A; NSCLC, non-small cell lung carcinoma; RCC, renal cell carcinoma; recMAGE-A3, recombinant MAGE-A3. *Started after January, 1st 2008.

Others

TLR5 is mainly expressed at the surface of intestinal epithelial cells, monocyte, macrophages and DCs.16 The only known natural ligand for TLR5 is flagellin, a component of bacterial flagella.235,236 In particular, TLR5 recognizes a short, highly conserved domain of flagellin that participates in intermolecular interactions within flagellar protofilaments and hence is required for bacterial motility.237 Recent crystallographic data indicate that TLR5 exhibits a peculiar ligand binding mode, even though it dimerizes following ligand binding similar to all other TLRs.236,238

CBLB502 is a peptide derivative of Salmonella enterica flagellin that has been developed as a TLR5 agonist.239 Similar to full-length flagellin, CBLB502 potently activates NFκB in vitro, hence exerting consistent anti-apoptotic effects.239 In line with this notion, CBLB502 attenuates injury in a murine model of ischemic renal failure240 and protects mice as well as primates from the life-threatening consequences of total body irradiation.239,241 Importantly, CBLB502 has been shown not to interfere with tumor radiosensitivity, at least in mice models,239 suggesting that CBLB502 might be employed as a radioprotective agent in cancer patients undergoing radiation therapy. Clinical trials that would evaluate this possibility have not yet been initiated, but the safety and efficacy of CBLB502 (used as a standalone agent) are being investigated in a single Phase I study enrolling patients with advanced unresectable solid tumors (NCT01527136). Intriguingly, full-length flagellin appears to inhibit the growth of immunogenic cancers in mice, but only if administered 8–10 d after tumor implantation, whereas it would exert pro-tumor effects if administered earlier.242 Although a few trials have recently investigated the efficacy of flagellin fused to a portion of the influenza virus matrix protein 2 (VAX102) as a vaccine against flu,243,244 when this paper was submitted (April 2012), no clinical trials were investigating the possibility to use full length flagellin as an adjuvant for anticancer therapy (source www.clinicaltrials.gov).

Cadi-05 (Immuvac), which consists of autoclaved Mycobacterium indicus pranii (also known as Mycobacterium w), has been originally developed as a vaccine against leprosy, an indication for which it is nowadays approved by FDA.245 Cadi-05 functions as a polyTLR agonist and has been shown to trigger potent anticancer immune responses in vivo, featuring increased levels of circulating IFNγ as well as the accumulation of tumor antigen-specific CD4+ and CD8+ T cells.246,247 In immunocompetent mouse models of myeloma, thymoma and melanoma, the intravenous administration of Cadi-05 (as a single agent) a few days after tumor implantation was sufficient to suppress neoplastic growth.246,247 The anticancer properties of Cadi-05 mainly rely on IFNγ, as they were abrogated in Ifng−/− mice.247 The use of Cadi-05 as an adjuvant seems promising, at least according to early reports on invasive bladder cancer patients treated with radiotherapy248 and NSCLC patients receiving radiotherapy in combination with cisplatin and etoposide.249 Subsequent studies have confirmed the safety and efficacy of Cadi-05 in this latter indication (ref. 250 and Annual AACR Meeting 2006, Abstract #P3–022) as well as in other oncological settings, including advanced solid tumors refractory to therapy (Annual ASCO Meeting 2008, Abstract 14019). In addition, Cadi-05 has recently been investigated as a standalone agent for the treatment of superficial transitional cell carcinomas of the bladder (NCT00694798), as a single agent in Stage III-IV melanoma patients (NCT00675727), combined with docetaxel in patients affected by hormone refractory metastatic prostate cancer (NCT00525408), and associated to paclitaxel + cisplatin in NSCLC patients (NCT00680940). The results of these studies, however, are still unpublished. At the time of submission of this Trial Watch (April 2012), only one Phase II clinical trial was ongoing to evaluate the efficacy of Cadi-05 in patients bearing superficial transitional cell carcinomas of the bladder (NCT00694915) (source www.clinicaltrials.gov).

Similar to Cadi-05, IMM-101 consists of inactivated bacteria, and more precisely of heat-killed Mycobacterium obuense.251 IMM-101 has been shown to indirectly activate γδ T cells (via a particular subset of myeloid DCs), resulting in the secretion of Th1 cytokines including IFNγ and TNFα and in the activation of antitumor immune responses.252 Although the preclinical characterization of IMM-101 is scarce, many other preparations that are similar in concept (including SRL172, which consists of heat-inactivated Mycobacterium vaccae) have been extensively investigated in humans for a variety of diseases, including cancer.253 SRL172 has been associated with minimal toxicity, potent immunostimulatory effects and—in some instances—a promising anticancer profile.254-256 These observations prompted for a Phase I clinical study to evaluate IMM-101 in stage III/IV melanoma patients (NCT01308762), which confirmed IMM-101 to be safe and potentially efficient.257 Today, the clinical benefits of SRL172 are being investigated in a few trials on cancer-unrelated indications (mainly tuberculosis). On the contrary, IMM-101 is being tested—combined with radiation therapy—in patients with previously treated colorectal cancer (NCT01539824), and—in combination with gemcitabine—in patients affected by advanced pancreatic cancer (NCT01303172) (source www.clinicaltrials.gov).

Table 6 summarizes recent clinical progress of experimental TLR5 and polyTLR agonists for oncological indications.

Table 6. Clinical trials evaluating TLR5 and polyTLR agonists in oncological indications*.

Compound Indication Trials Phase Status Notes Ref.
CBLB502
Adult solid tumors
1
I
Recruiting
As single agent
NCT01527136
CADI-05
Bladder cancer
2
I
Completed
As single agent
NCT00694798
II
Active, not recruiting
As single agent
NCT00694915
Melanoma
1
I/II
Terminated
As single agent
NCT00675727
NSCLC
1
II
Completed
Combined with cisplatin and paclitaxel
NCT00680940
Prostate cancer
1
II
Terminated
Combined with docetaxel
NCT00525408
IMM-101 Colorectal cancer
1
II
Recruiting
Combined with SBRT
NCT01539824
Melanoma
2
I
Completed
As single agent
NCT01308762
I/II
Enrolling by invitation
As single agent
NCT01559818
Pancreatic cancer 1 II Recruiting Combined with gemcitabine NCT01303172

Abbreviations: NSCLC, non-small cell lung carcinoma; SBRT, stereotactic body radiotherapy. *Started after January, 1st 2008.

Concluding Remarks

A vast amount of preclinical and clinical data indicate that TLR agonists exert potent immunostimulatory functions, in vivo. This said, with the notable exceptions of BCG, MPL and imiquimod (which are approved by FDA and the European Medicines Agency) and perhaps picibanil (which is approved by the Japanese Ministry of Health and Welfare), the clinical progress of TLR agonists for use as standalone agents or adjuvants in cancer therapy appears to have reached an impasse. The development of many TLR agonists has been discontinued due to a lack of efficacy in Phase III studies, and the number of clinical trials currently testing this rather large class of compounds (including no less than 30 distinct molecules) for oncological indications is small (< 50 trials, all phases confounded, excluding BGC-, MPL-, imiquimod and picibanil-based studies). This points to some decline in the interest of researchers and clinicians for TLR agonists, at least in the oncological setting. One element that has surely contributed to (though perhaps not entirely determined) this decline is the limited availability of clinical grade TLR agonists, pushing academic investigators to prefer surrogate compounds, such as clinically approved prophylactic vaccines.258,259 Our hope is that the results of ongoing clinical trials will invert this tendency and eventually lead to the development (and approval) of optimal immunochemotherapeutic regimens that fully exploit the immunostimulatory potential of TLR agonists against cancer.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipes labelisées), AXA Chair for Longevity Research, Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Médicale, Agence National de la Recherche, the European Commission (Apo-Sys, ArtForce, ChemoRes. Death-Train) and the LabEx Immuno-Oncology.

Glossary

Abbreviations:

CTL

cytotoxic lymphocyte

DAMPs

damage-associated molecular patterns

DC

dendritic cell

dSLIM

DNA-based double stem-loop immunomodulator

dsRNA

double-stranded RNA

EGFR

epidermal growth factor receptor

GM-CSF

granulocyte monocyte colony-stimulating factor

HNSCC

head and neck squamous cell carcinoma

IFN

interferon

IL

interleukin

IMO

immunomodulatory oligonucleotide

ISS

immunostimulatory sequence

LBP

LPS-binding protein

LPS

lipopolysaccharide

LTA

lipoteichoic acid

MAL

MYD88 adapter-like

MALP-2

macrophage-activating lipopeptide 2

MAMPs

microbe-associated molecular patterns

MPL

monophosphoryl lipid A

NSCLC

non-small cell lung carcinoma

PAMPs

pathogen-associated molecular patterns

PBMC

peripheral blood mononuclear cell

polyA:U

polyadenylic polyuridylic acid

polyI:C

polyriboinosinic polyribocytidylic acid

PRR

pattern recognition receptor

SIV

simian immunodeficiency virus

STAT1

signal transducer and activator of transcription 1

TIL

Toll/Interleukin 1 receptor-like

TNFα

tumor necrosis factor α

TLR

Toll-like receptor, Treg, FOXP3+ immunosuppressive T cell

Footnotes

References

  • 1.Anderson KV, Bokla L, Nüsslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: the induction of polarity by the Toll gene product. Cell. 1985;42:791–8. doi: 10.1016/0092-8674(85)90275-2. [DOI] [PubMed] [Google Scholar]
  • 2.Anderson KV, Jürgens G, Nüsslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: genetic studies on the role of the Toll gene product. Cell. 1985;42:779–89. doi: 10.1016/0092-8674(85)90274-0. [DOI] [PubMed] [Google Scholar]
  • 3.Hashimoto C, Hudson KL, Anderson KV. The Toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell. 1988;52:269–79. doi: 10.1016/0092-8674(88)90516-8. [DOI] [PubMed] [Google Scholar]
  • 4.Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86:973–83. doi: 10.1016/S0092-8674(00)80172-5. [DOI] [PubMed] [Google Scholar]
  • 5.Gay NJ, Keith FJ. Drosophila Toll and IL-1 receptor. Nature. 1991;351:355–6. doi: 10.1038/351355b0. [DOI] [PubMed] [Google Scholar]
  • 6.Nomura N, Miyajima N, Sazuka T, Tanaka A, Kawarabayasi Y, Sato S, et al. Prediction of the coding sequences of unidentified human genes. I. The coding sequences of 40 new genes (KIAA0001-KIAA0040) deduced by analysis of randomly sampled cDNA clones from human immature myeloid cell line KG-1. DNA Res. 1994;1:27–35. doi: 10.1093/dnares/1.1.27. [DOI] [PubMed] [Google Scholar]
  • 7.Taguchi T, Mitcham JL, Dower SK, Sims JE, Testa JR. Chromosomal localization of TIL, a gene encoding a protein related to the Drosophila transmembrane receptor Toll, to human chromosome 4p14. Genomics. 1996;32:486–8. doi: 10.1006/geno.1996.0150. [DOI] [PubMed] [Google Scholar]
  • 8.Medzhitov R, Preston-Hurlburt P, Janeway CA., Jr A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388:394–7. doi: 10.1038/41131. [DOI] [PubMed] [Google Scholar]
  • 9.Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, et al. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature. 1998;395:284–8. doi: 10.1038/26239. [DOI] [PubMed] [Google Scholar]
  • 10.Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–8. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 11.Song WY, Wang GL, Chen LL, Kim HS, Pi LY, Holsten T, et al. A receptor kinase-like protein encoded by the rice disease resistance gene, Xa21. Science. 1995;270:1804–6. doi: 10.1126/science.270.5243.1804. [DOI] [PubMed] [Google Scholar]
  • 12.Gómez-Gómez L, Boller T. FLS2: an LRR receptor-like kinase involved in the perception of the bacterial elicitor flagellin in Arabidopsis. Mol Cell. 2000;5:1003–11. doi: 10.1016/s1097-2765(00)80265-8. [DOI] [PubMed] [Google Scholar]
  • 13.Roach JC, Glusman G, Rowen L, Kaur A, Purcell MK, Smith KD, et al. The evolution of vertebrate Toll-like receptors. Proc Natl Acad Sci U S A. 2005;102:9577–82. doi: 10.1073/pnas.0502272102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21:335–76. doi: 10.1146/annurev.immunol.21.120601.141126. [DOI] [PubMed] [Google Scholar]
  • 15.Koropatnick TA, Engle JT, Apicella MA, Stabb EV, Goldman WE, McFall-Ngai MJ. Microbial factor-mediated development in a host-bacterial mutualism. Science. 2004;306:1186–8. doi: 10.1126/science.1102218. [DOI] [PubMed] [Google Scholar]
  • 16.Hennessy EJ, Parker AE, O’Neill LA. Targeting Toll-like receptors: emerging therapeutics? Nat Rev Drug Discov. 2010;9:293–307. doi: 10.1038/nrd3203. [DOI] [PubMed] [Google Scholar]
  • 17.Chuang T, Ulevitch RJ. Identification of hTLR10: a novel human Toll-like receptor preferentially expressed in immune cells. Biochim Biophys Acta. 2001;1518:157–61. doi: 10.1016/s0167-4781(00)00289-x. [DOI] [PubMed] [Google Scholar]
  • 18.Guan Y, Ranoa DR, Jiang S, Mutha SK, Li X, Baudry J, et al. Human TLRs 10 and 1 share common mechanisms of innate immune sensing but not signaling. J Immunol. 2010;184:5094–103. doi: 10.4049/jimmunol.0901888. [DOI] [PubMed] [Google Scholar]
  • 19.Hasan U, Chaffois C, Gaillard C, Saulnier V, Merck E, Tancredi S, et al. Human TLR10 is a functional receptor, expressed by B cells and plasmacytoid dendritic cells, which activates gene transcription through MyD88. J Immunol. 2005;174:2942–50. doi: 10.4049/jimmunol.174.5.2942. [DOI] [PubMed] [Google Scholar]
  • 20.Govindaraj RG, Manavalan B, Lee G, Choi S. Molecular modeling-based evaluation of hTLR10 and identification of potential ligands in Toll-like receptor signaling. PLoS One. 2010;5:e12713. doi: 10.1371/journal.pone.0012713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mishra BB, Gundra UM, Teale JM. Expression and distribution of Toll-like receptors 11-13 in the brain during murine neurocysticercosis. J Neuroinflammation. 2008;5:53. doi: 10.1186/1742-2094-5-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang D, Zhang G, Hayden MS, Greenblatt MB, Bussey C, Flavell RA, et al. A toll-like receptor that prevents infection by uropathogenic bacteria. Science. 2004;303:1522–6. doi: 10.1126/science.1094351. [DOI] [PubMed] [Google Scholar]
  • 23.Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL, Serhan CN, Hayden MS, et al. TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science. 2005;308:1626–9. doi: 10.1126/science.1109893. [DOI] [PubMed] [Google Scholar]
  • 24.Pifer R, Benson A, Sturge CR, Yarovinsky F. UNC93B1 is essential for TLR11 activation and IL-12-dependent host resistance to Toxoplasma gondii. J Biol Chem. 2011;286:3307–14. doi: 10.1074/jbc.M110.171025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Shi Z, Cai Z, Sanchez A, Zhang T, Wen S, Wang J, et al. A novel Toll-like receptor that recognizes vesicular stomatitis virus. J Biol Chem. 2011;286:4517–24. doi: 10.1074/jbc.M110.159590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fitzgerald KA, Palsson-McDermott EM, Bowie AG, Jefferies CA, Mansell AS, Brady G, et al. Mal (MyD88-adapter-like) is required for Toll-like receptor-4 signal transduction. Nature. 2001;413:78–83. doi: 10.1038/35092578. [DOI] [PubMed] [Google Scholar]
  • 27.Medzhitov R, Preston-Hurlburt P, Kopp E, Stadlen A, Chen C, Ghosh S, et al. MyD88 is an adaptor protein in the hToll/IL-1 receptor family signaling pathways. Mol Cell. 1998;2:253–8. doi: 10.1016/S1097-2765(00)80136-7. [DOI] [PubMed] [Google Scholar]
  • 28.Hoebe K, Du X, Georgel P, Janssen E, Tabeta K, Kim SO, et al. Identification of Lps2 as a key transducer of MyD88-independent TIR signalling. Nature. 2003;424:743–8. doi: 10.1038/nature01889. [DOI] [PubMed] [Google Scholar]
  • 29.Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science. 2003;301:640–3. doi: 10.1126/science.1087262. [DOI] [PubMed] [Google Scholar]
  • 30.Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature. 2005;434:772–7. doi: 10.1038/nature03464. [DOI] [PubMed] [Google Scholar]
  • 31.Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K, Kaisho T, et al. TRAM is specifically involved in the Toll-like receptor 4-mediated MyD88-independent signaling pathway. Nat Immunol. 2003;4:1144–50. doi: 10.1038/ni986. [DOI] [PubMed] [Google Scholar]
  • 32.Siednienko J, Gajanayake T, Fitzgerald KA, Moynagh P, Miggin SM. Absence of MyD88 results in enhanced TLR3-dependent phosphorylation of IRF3 and increased IFN-β and RANTES production. J Immunol. 2011;186:2514–22. doi: 10.4049/jimmunol.1003093. [DOI] [PubMed] [Google Scholar]
  • 33.Siednienko J, Halle A, Nagpal K, Golenbock DT, Miggin SM. TLR3-mediated IFN-β gene induction is negatively regulated by the TLR adaptor MyD88 adaptor-like. Eur J Immunol. 2010;40:3150–60. doi: 10.1002/eji.201040547. [DOI] [PubMed] [Google Scholar]
  • 34.Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10:826–37. doi: 10.1038/nri2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, et al. Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002;277:15028–34. doi: 10.1074/jbc.M200497200. [DOI] [PubMed] [Google Scholar]
  • 36.Kol A, Lichtman AH, Finberg RW, Libby P, Kurt-Jones EA. Cutting edge: heat shock protein (HSP) 60 activates the innate immune response: CD14 is an essential receptor for HSP60 activation of mononuclear cells. J Immunol. 2000;164:13–7. doi: 10.4049/jimmunol.164.1.13. [DOI] [PubMed] [Google Scholar]
  • 37.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  • 38.Hannani D, Sistigu A, Kepp O, Galluzzi L, Kroemer G, Zitvogel L. Prerequisites for the antitumor vaccine-like effect of chemotherapy and radiotherapy. Cancer J. 2011;17:351–8. doi: 10.1097/PPO.0b013e3182325d4d. [DOI] [PubMed] [Google Scholar]
  • 39.Liu-Bryan R, Pritzker K, Firestein GS, Terkeltaub R. TLR2 signaling in chondrocytes drives calcium pyrophosphate dihydrate and monosodium urate crystal-induced nitric oxide generation. J Immunol. 2005;174:5016–23. doi: 10.4049/jimmunol.174.8.5016. [DOI] [PubMed] [Google Scholar]
  • 40.Guillot L, Balloy V, McCormack FX, Golenbock DT, Chignard M, Si-Tahar M. Cutting edge: the immunostimulatory activity of the lung surfactant protein-A involves Toll-like receptor 4. J Immunol. 2002;168:5989–92. doi: 10.4049/jimmunol.168.12.5989. [DOI] [PubMed] [Google Scholar]
  • 41.Rakoff-Nahoum S, Medzhitov R. Toll-like receptors and cancer. Nat Rev Cancer. 2009;9:57–63. doi: 10.1038/nrc2541. [DOI] [PubMed] [Google Scholar]
  • 42.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
  • 43.Kepp O, Galluzzi L, Giordanetto F, Tesniere A, Vitale I, Martins I, et al. Disruption of the PP1/GADD34 complex induces calreticulin exposure. Cell Cycle. 2009;8:3971–7. doi: 10.4161/cc.8.23.10191. [DOI] [PubMed] [Google Scholar]
  • 44.Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–7. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
  • 45.Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–8. doi: 10.4161/cc.8.22.10026. [DOI] [PubMed] [Google Scholar]
  • 46.Martins I, Michaud M, Sukkurwala AQ, Adjemian S, Ma Y, Shen S, et al. Premortem autophagy determines the immunogenicity of chemotherapy-induced cancer cell death. Autophagy. 2012;8:8. doi: 10.4161/auto.19009. [DOI] [PubMed] [Google Scholar]
  • 47.Chen T, Guo J, Han C, Yang M, Cao X. Heat shock protein 70, released from heat-stressed tumor cells, initiates antitumor immunity by inducing tumor cell chemokine production and activating dendritic cells via TLR4 pathway. J Immunol. 2009;182:1449–59. doi: 10.4049/jimmunol.182.3.1449. [DOI] [PubMed] [Google Scholar]
  • 48.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunol. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell. 2010;140:798–804. doi: 10.1016/j.cell.2010.02.015. [DOI] [PubMed] [Google Scholar]
  • 50.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
  • 51.Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 2007;220:47–59. doi: 10.1111/j.1600-065X.2007.00573.x. [DOI] [PubMed] [Google Scholar]
  • 52.Heldwein KA, Liang MD, Andresen TK, Thomas KE, Marty AM, Cuesta N, et al. TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J Leukoc Biol. 2003;74:277–86. doi: 10.1189/jlb.0103026. [DOI] [PubMed] [Google Scholar]
  • 53.Uehori J, Matsumoto M, Tsuji S, Akazawa T, Takeuchi O, Akira S, et al. Simultaneous blocking of human Toll-like receptors 2 and 4 suppresses myeloid dendritic cell activation induced by Mycobacterium bovis bacillus Calmette-Guérin peptidoglycan. Infect Immun. 2003;71:4238–49. doi: 10.1128/IAI.71.8.4238-4249.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Akazawa T, Masuda H, Saeki Y, Matsumoto M, Takeda K, Tsujimura K, et al. Adjuvant-mediated tumor regression and tumor-specific cytotoxic response are impaired in MyD88-deficient mice. Cancer Res. 2004;64:757–64. doi: 10.1158/0008-5472.CAN-03-1518. [DOI] [PubMed] [Google Scholar]
  • 55.Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol. 2002;3:196–200. doi: 10.1038/ni758. [DOI] [PubMed] [Google Scholar]
  • 56.Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, et al. Identification of a primary target of thalidomide teratogenicity. Science. 2010;327:1345–50. doi: 10.1126/science.1177319. [DOI] [PubMed] [Google Scholar]
  • 57.Hoffman ES, Smith RE, Renaud RC., Jr From the analyst’s couch: TLR-targeted therapeutics. Nat Rev Drug Discov. 2005;4:879–80. doi: 10.1038/nrd1880. [DOI] [PubMed] [Google Scholar]
  • 58.Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science. 2007;316:1628–32. doi: 10.1126/science.1138963. [DOI] [PubMed] [Google Scholar]
  • 59.Agosti JM, Goldie SJ. Introducing HPV vaccine in developing countries--key challenges and issues. N Engl J Med. 2007;356:1908–10. doi: 10.1056/NEJMp078053. [DOI] [PubMed] [Google Scholar]
  • 60.Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, et al. CD36 is a sensor of diacylglycerides. Nature. 2005;433:523–7. doi: 10.1038/nature03253. [DOI] [PubMed] [Google Scholar]
  • 61.Shimizu T, Kida Y, Kuwano K. A dipalmitoylated lipoprotein from Mycoplasma pneumoniae activates NF-kappa B through TLR1, TLR2, and TLR6. J Immunol. 2005;175:4641–6. doi: 10.4049/jimmunol.175.7.4641. [DOI] [PubMed] [Google Scholar]
  • 62.Farhat K, Riekenberg S, Heine H, Debarry J, Lang R, Mages J, et al. Heterodimerization of TLR2 with TLR1 or TLR6 expands the ligand spectrum but does not lead to differential signaling. J Leukoc Biol. 2008;83:692–701. doi: 10.1189/jlb.0807586. [DOI] [PubMed] [Google Scholar]
  • 63.Jiang Q, Akashi S, Miyake K, Petty HR. Lipopolysaccharide induces physical proximity between CD14 and toll-like receptor 4 (TLR4) prior to nuclear translocation of NF-kappa B. J Immunol. 2000;165:3541–4. doi: 10.4049/jimmunol.165.7.3541. [DOI] [PubMed] [Google Scholar]
  • 64.Schröder NW, Opitz B, Lamping N, Michelsen KS, Zähringer U, Göbel UB, et al. Involvement of lipopolysaccharide binding protein, CD14, and Toll-like receptors in the initiation of innate immune responses by Treponema glycolipids. J Immunol. 2000;165:2683–93. doi: 10.4049/jimmunol.165.5.2683. [DOI] [PubMed] [Google Scholar]
  • 65.Uenishi Y, Fujita Y, Kusunose N, Yano I, Sunagawa M. Comprehensive analysis of mycolic acid subclass and molecular species composition of Mycobacterium bovis BCG Tokyo 172 cell wall skeleton (SMP-105) J Microbiol Methods. 2008;72:149–56. doi: 10.1016/j.mimet.2007.11.016. [DOI] [PubMed] [Google Scholar]
  • 66.Murata M. Activation of Toll-like receptor 2 by a novel preparation of cell wall skeleton from Mycobacterium bovis BCG Tokyo (SMP-105) sufficiently enhances immune responses against tumors. Cancer Sci. 2008;99:1435–40. doi: 10.1111/j.1349-7006.2008.00832.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Prigal SJ. Development in mice of prolonged non-specific resistance to sarcoma implant and Staphylococcus infection following repository injection of lipopolysaccharide. Nature. 1961;191:1111–2. doi: 10.1038/1911111a0. [DOI] [PubMed] [Google Scholar]
  • 68.Mizuno D, Yoshioka O, Akamatu M, Kataoka T. Antitumor effect of intracutaneous injection of bacterial lipopolysaccharide. Cancer Res. 1968;28:1531–7. [PubMed] [Google Scholar]
  • 69.Yamamura Y, Proctor JW, Fischer BC, Harnaha JB, Mahvi TA. Collaboration between specific anti-tumor immunity and chemotherapeutic agents. Int J Cancer. 1980;25:417–23. doi: 10.1002/ijc.2910250317. [DOI] [PubMed] [Google Scholar]
  • 70.Chicoine MR, Zahner M, Won EK, Kalra RR, Kitamura T, Perry A, et al. The in vivo antitumoral effects of lipopolysaccharide against glioblastoma multiforme are mediated in part by Toll-like receptor 4. Neurosurgery. 2007;60:372–80, discussion 381. doi: 10.1227/01.NEU.0000249280.61761.2E. [DOI] [PubMed] [Google Scholar]
  • 71.Davis MB, Vasquez-Dunddel D, Fu J, Albesiano E, Pardoll D, Kim YJ. Intratumoral administration of TLR4 agonist absorbed into a cellular vector improves antitumor responses. Clin Cancer Res. 2011;17:3984–92. doi: 10.1158/1078-0432.CCR-10-3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, et al. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature. 1987;330:662–4. doi: 10.1038/330662a0. [DOI] [PubMed] [Google Scholar]
  • 73.Ohlsson K, Björk P, Bergenfeldt M, Hageman R, Thompson RC. Interleukin-1 receptor antagonist reduces mortality from endotoxin shock. Nature. 1990;348:550–2. doi: 10.1038/348550a0. [DOI] [PubMed] [Google Scholar]
  • 74.Engelhardt R, Mackensen A, Galanos C. Phase I trial of intravenously administered endotoxin (Salmonella abortus equi) in cancer patients. Cancer Res. 1991;51:2524–30. [PubMed] [Google Scholar]
  • 75.Mackensen A, Galanos C, Engelhardt R. Treatment of cancer patients with endotoxin induces release of endogenous cytokines. Pathobiology. 1991;59:264–7. doi: 10.1159/000163659. [DOI] [PubMed] [Google Scholar]
  • 76.Otto F, Schmid P, Mackensen A, Wehr U, Seiz A, Braun M, et al. Phase II trial of intravenous endotoxin in patients with colorectal and non-small cell lung cancer. Eur J Cancer. 1996;32A:1712–8. doi: 10.1016/0959-8049(96)00186-4. [DOI] [PubMed] [Google Scholar]
  • 77.Okamoto H, Shoin S, Koshimura S, Shimizu R. Studies on the anticancer and streptolysin S-forming abilities of hemolytic streptococci. Jpn J Microbiol. 1967;11:323–6. doi: 10.1111/j.1348-0421.1967.tb00350.x. [DOI] [PubMed] [Google Scholar]
  • 78.Okamoto M, Oshikawa T, Tano T, Ahmed SU, Kan S, Sasai A, et al. Mechanism of anticancer host response induced by OK-432, a streptococcal preparation, mediated by phagocytosis and Toll-like receptor 4 signaling. J Immunother. 2006;29:78–86. doi: 10.1097/01.cji.0000192106.32206.30. [DOI] [PubMed] [Google Scholar]
  • 79.Hironaka K, Yamaguchi Y, Okita R, Okawaki M, Nagamine I. Essential requirement of toll-like receptor 4 expression on CD11c+ cells for locoregional immunotherapy of malignant ascites using a streptococcal preparation OK-432. Anticancer Res. 2006;26(5B):3701–7. [PubMed] [Google Scholar]
  • 80.Kikkawa F, Kawai M, Oguchi H, Kojima M, Ishikawa H, Iwata M, et al. Randomised study of immunotherapy with OK-432 in uterine cervical carcinoma. Eur J Cancer. 1993;29A:1542–6. doi: 10.1016/0959-8049(93)90291-M. [DOI] [PubMed] [Google Scholar]
  • 81.Maehara Y, Okuyama T, Kakeji Y, Baba H, Furusawa M, Sugimachi K. Postoperative immunochemotherapy including streptococcal lysate OK-432 is effective for patients with gastric cancer and serosal invasion. Am J Surg. 1994;168:36–40. doi: 10.1016/S0002-9610(05)80068-6. [DOI] [PubMed] [Google Scholar]
  • 82.Sato M, Harada K, Yoshida H, Yura Y, Azuma M, Iga H, et al. Therapy for oral squamous cell carcinoma by tegafur and streptococcal agent OK-432 in combination with radiotherapy: association of the therapeutic effect with differentiation and apoptosis in the cancer cells. Apoptosis. 1997;2:227–38. doi: 10.1023/A:1026428918301. [DOI] [PubMed] [Google Scholar]
  • 83.Brandenburg K, Lindner B, Schromm A, Koch MH, Bauer J, Merkli A, et al. Physicochemical characteristics of triacyl lipid A partial structure OM-174 in relation to biological activity. Eur J Biochem. 2000;267:3370–7. doi: 10.1046/j.1432-1327.2000.01370.x. [DOI] [PubMed] [Google Scholar]
  • 84.Lembo A, Pelletier M, Iyer R, Timko M, Dudda JC, West TE, et al. Administration of a synthetic TLR4 agonist protects mice from pneumonic tularemia. J Immunol. 2008;180:7574–81. doi: 10.4049/jimmunol.180.11.7574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.D’Agostini C, Pica F, Febbraro G, Grelli S, Chiavaroli C, Garaci E. Antitumour effect of OM-174 and cyclophosphamide on murine B16 melanoma in different experimental conditions. Int Immunopharmacol. 2005;5:1205–12. doi: 10.1016/j.intimp.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 86.De Ridder M, Verovski VN, Chiavaroli C, Van den Berge DL, Monsaert C, Law K, et al. The radiosensitizing effect of immunoadjuvant OM-174 requires cooperation between immune and tumor cells through interferon-gamma and inducible nitric oxide synthase. Int J Radiat Oncol Biol Phys. 2006;66:1473–80. doi: 10.1016/j.ijrobp.2006.07.1381. [DOI] [PubMed] [Google Scholar]
  • 87.Jeannin J-F. Lipid A in cancer therapy. New York Austin, Texas: Springer Science+Business Media;; Landes Bioscience , 2009
  • 88.Yamazaki S, Okada K, Maruyama A, Matsumoto M, Yagita H, Seya T. TLR2-dependent induction of IL-10 and Foxp3+ CD25+ CD4+ regulatory T cells prevents effective anti-tumor immunity induced by Pam2 lipopeptides in vivo. PLoS One. 2011;6:e18833. doi: 10.1371/journal.pone.0018833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Netea MG, Sutmuller R, Hermann C, Van der Graaf CA, Van der Meer JW, van Krieken JH, et al. Toll-like receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T cells. J Immunol. 2004;172:3712–8. doi: 10.4049/jimmunol.172.6.3712. [DOI] [PubMed] [Google Scholar]
  • 90.Ledeboer A, Hutchinson MR, Watkins LR, Johnson KW. Ibudilast (AV-411). A new class therapeutic candidate for neuropathic pain and opioid withdrawal syndromes. Expert Opin Investig Drugs. 2007;16:935–50. doi: 10.1517/13543784.16.7.935. [DOI] [PubMed] [Google Scholar]
  • 91.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;413:732–8. doi: 10.1038/35099560. [DOI] [PubMed] [Google Scholar]
  • 92.Choe J, Kelker MS, Wilson IA. Crystal structure of human toll-like receptor 3 (TLR3) ectodomain. Science. 2005;309:581–5. doi: 10.1126/science.1115253. [DOI] [PubMed] [Google Scholar]
  • 93.Liu L, Botos I, Wang Y, Leonard JN, Shiloach J, Segal DM, et al. Structural basis of toll-like receptor 3 signaling with double-stranded RNA. Science. 2008;320:379–81. doi: 10.1126/science.1155406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev. 2007;87:99–163. doi: 10.1152/physrev.00013.2006. [DOI] [PubMed] [Google Scholar]
  • 95.Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–20. doi: 10.1038/cdd.2011.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Salaun B, Coste I, Rissoan MC, Lebecque SJ, Renno T. TLR3 can directly trigger apoptosis in human cancer cells. J Immunol. 2006;176:4894–901. doi: 10.4049/jimmunol.176.8.4894. [DOI] [PubMed] [Google Scholar]
  • 97.Conforti R, Ma Y, Morel Y, Paturel C, Terme M, Viaud S, et al. Opposing effects of toll-like receptor (TLR3) signaling in tumors can be therapeutically uncoupled to optimize the anticancer efficacy of TLR3 ligands. Cancer Res. 2010;70:490–500. doi: 10.1158/0008-5472.CAN-09-1890. [DOI] [PubMed] [Google Scholar]
  • 98.Salaun B, Zitvogel L, Asselin-Paturel C, Morel Y, Chemin K, Dubois C, et al. TLR3 as a biomarker for the therapeutic efficacy of double-stranded RNA in breast cancer. Cancer Res. 2011;71:1607–14. doi: 10.1158/0008-5472.CAN-10-3490. [DOI] [PubMed] [Google Scholar]
  • 99.Baron S, Bogomolova NN, Billiau A, Levy HB, Buckler CE, Stern R, et al. Induction of interferon by preparations of synthetic single-stranded RNA. Proc Natl Acad Sci U S A. 1969;64:67–74. doi: 10.1073/pnas.64.1.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.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;27:107–15. doi: 10.1016/j.vaccine.2008.10.024. [DOI] [PubMed] [Google Scholar]
  • 101.Strayer DR, Watson P, Carter WA, Brodsky I. Antiproliferative effect of mismatched double-stranded RNA on fresh human tumor cells analyzed in a clonogenic assay. J Interferon Res. 1986;6:373–9. doi: 10.1089/jir.1986.6.373. [DOI] [PubMed] [Google Scholar]
  • 102.Strayer DR, Weisband J, Carter WA, Black P, Nidzgorski F, Cook AW. Growth of astrocytomas in the human tumor clonogenic assay and sensitivity to mismatched dsRNA and interferons. Am J Clin Oncol. 1987;10:281–4. doi: 10.1097/00000421-198708000-00003. [DOI] [PubMed] [Google Scholar]
  • 103.Carter WA, Hubbell HR, Krueger LJ, Strayer DR. Comparative studies of ampligen (mismatched double-stranded RNA) and interferons. J Biol Response Mod. 1985;4:613–20. [PubMed] [Google Scholar]
  • 104.Carter WA, Strayer DR, Hubbell HR, Brodsky I. Preclinical studies with Ampligen (mismatched double-stranded RNA) J Biol Response Mod. 1985;4:495–502. [PubMed] [Google Scholar]
  • 105.Hubbell HR, Pequignot EC, Todd J, Raymond LC, Mayberry SD, Carter WA, et al. Augmented antitumor effect of combined human natural interferon-alpha and mismatched double-stranded RNA treatment against a human malignant melanoma xenograft. J Biol Response Mod. 1987;6:525–36. [PubMed] [Google Scholar]
  • 106.Bucur N, Mizuno M, Wakabayashi T, Yoshida J. Growth inhibition of experimental glioma by human interferon-beta superinduced by cationic liposomes entrapping polyinosilic:polycytidilic acid. Neurol Med Chir (Tokyo) 1998;38:469–73, discussion 473-4. doi: 10.2176/nmc.38.469. [DOI] [PubMed] [Google Scholar]
  • 107.Akazawa T, Ebihara T, Okuno M, Okuda Y, Shingai M, Tsujimura K, et al. Antitumor NK activation induced by the Toll-like receptor 3-TICAM-1 (TRIF) pathway in myeloid dendritic cells. Proc Natl Acad Sci U S A. 2007;104:252–7. doi: 10.1073/pnas.0605978104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chin AI, Miyahira AK, Covarrubias A, Teague J, Guo B, Dempsey PW, et al. Toll-like receptor 3-mediated suppression of TRAMP prostate cancer shows the critical role of type I interferons in tumor immune surveillance. Cancer Res. 2010;70:2595–603. doi: 10.1158/0008-5472.CAN-09-1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Harrison SD, Jr., Stevens JJ, Waud WR, Dykes DJ, Schmid SM, Griswold DP., Jr. Evaluation of combinations of interferons and cytotoxic drugs in murine tumor models in vivo. J Biol Response Mod. 1990;9:395–400. [PubMed] [Google Scholar]
  • 110.Shime H, Matsumoto M, Oshiumi H, Tanaka S, Nakane A, Iwakura Y, et al. Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proc Natl Acad Sci U S A. 2012;109:2066–71. doi: 10.1073/pnas.1113099109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cheng YS, Xu F. Anticancer function of polyinosinic-polycytidylic acid. Cancer Biol Ther. 2011;10:1219–23. doi: 10.4161/cbt.10.12.13450. [DOI] [PubMed] [Google Scholar]
  • 112.Shir A, Ogris M, Roedl W, Wagner E, Levitzki A. EGFR-homing dsRNA activates cancer-targeted immune response and eliminates disseminated EGFR-overexpressing tumors in mice. Clin Cancer Res. 2011;17:1033–43. doi: 10.1158/1078-0432.CCR-10-1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Brodsky I, Strayer DR, Krueger LJ, Carter WA. Clinical studies with ampligen (mismatched double-stranded RNA) J Biol Response Mod. 1985;4:669–75. [PubMed] [Google Scholar]
  • 114.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;132:434–9. doi: 10.1093/infdis/132.4.434. [DOI] [PubMed] [Google Scholar]
  • 115.Stephen EL, Hilmas DE, Mangiafico JA, Levy HB. Swine influenza virus vaccine: potentiation of antibody responses in rhesus monkeys. Science. 1977;197:1289–90. doi: 10.1126/science.408923. [DOI] [PubMed] [Google Scholar]
  • 116.Levy HB, Lvovsky E. Topical treatment of vaccinia virus infection with an interferon inducer in rabbits. J Infect Dis. 1978;137:78–81. doi: 10.1093/infdis/137.1.78. [DOI] [PubMed] [Google Scholar]
  • 117.Olsen GA, Kern ER, Overall JC., Jr Effect of treatment with exogenous interferon, polyriboinosinic-polyribocytidylic acid, or polyriboinosinic-polyribocytidylic acid-poly-L-lysine complex on Herpesvirus hominis infections in mice. J Infect Dis. 1978;137:428–36. doi: 10.1093/infdis/137.4.428. [DOI] [PubMed] [Google Scholar]
  • 118.Machida H, Takezawa J, Kuninaka A, Yoshino H, Nakamura O, Takakura K. Interferon induction and therapy of brain tumors in rats by poly(ICLC) Microbiol Immunol. 1982;26:353–8. doi: 10.1111/j.1348-0421.1982.tb00185.x. [DOI] [PubMed] [Google Scholar]
  • 119.Levine AS, Levy HB. Phase I-II trials of poly IC stabilized with poly-L-lysine. Cancer Treat Rep. 1978;62:1907–12. [PubMed] [Google Scholar]
  • 120.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;45:5904–9. [PubMed] [Google Scholar]
  • 121.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;19:89–92. doi: 10.1023/A:1006458232384. [DOI] [PubMed] [Google Scholar]
  • 122.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;4:518–24. [PubMed] [Google Scholar]
  • 123.Nakamura O, Shitara N, Matsutani M, Takakura K, Machida H. Phase I-II trials of poly(ICLC) in malignant brain tumor patients. J Interferon Res. 1982;2:1–4. doi: 10.1089/jir.1982.2.1. [DOI] [PubMed] [Google Scholar]
  • 124.Okada H. Brain tumor immunotherapy with type-1 polarizing strategies. Ann N Y Acad Sci. 2009;1174:18–23. doi: 10.1111/j.1749-6632.2009.04932.x. [DOI] [PubMed] [Google Scholar]
  • 125.Butowski N, Chang SM, Junck L, DeAngelis LM, Abrey L, Fink K, et al. A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05) J Neurooncol. 2009;91:175–82. doi: 10.1007/s11060-008-9693-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Butowski N, Lamborn KR, Lee BL, Prados MD, Cloughesy T, DeAngelis LM, et al. A North American brain tumor consortium phase II study of poly-ICLC for adult patients with recurrent anaplastic gliomas. J Neurooncol. 2009;91:183–9. doi: 10.1007/s11060-008-9705-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Rosenfeld MR, Chamberlain MC, Grossman SA, Peereboom DM, Lesser GJ, Batchelor TT, et al. A multi-institution phase II study of poly-ICLC and radiotherapy with concurrent and adjuvant temozolomide in adults with newly diagnosed glioblastoma. Neuro Oncol. 2010;12:1071–7. doi: 10.1093/neuonc/noq071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Grossman SA, Ye X, Piantadosi S, Desideri S, Nabors LB, Rosenfeld M, et al. NABTT CNS Consortium Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States. Clin Cancer Res. 2010;16:2443–9. doi: 10.1158/1078-0432.CCR-09-3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.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;4:664–8. [PubMed] [Google Scholar]
  • 130.Rettenmaier MA, Berman ML, DiSaia PJ. Treatment of advanced ovarian cancer with polyinosinic-polycytidylic lysine carboxymethylcellulose (poly(ICLC)) Gynecol Oncol. 1986;24:359–61. doi: 10.1016/0090-8258(86)90313-6. [DOI] [PubMed] [Google Scholar]
  • 131.Droller MJ. Immunotherapy of metastatic renal cell carcinoma with polyinosinic-polycytidylic acid. J Urol. 1987;137:202–6. doi: 10.1016/s0022-5347(17)43953-x. [DOI] [PubMed] [Google Scholar]
  • 132.Krown SE, Kerr D, Stewart WE, 2nd, Field AK, Oettgen HF. Phase I trials of poly(I,C) complexes in advanced cancer. J Biol Response Mod. 1985;4:640–9. [PubMed] [Google Scholar]
  • 133.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;4:650–5. [PubMed] [Google Scholar]
  • 134.Ewel CH, Urba WJ, Kopp WC, Smith JW, 2nd, Steis RG, Rossio JL, et al. Polyinosinic-polycytidylic acid complexed with poly-L-lysine and carboxymethylcellulose in combination with interleukin 2 in patients with cancer: clinical and immunological effects. Cancer Res. 1992;52:3005–10. [PubMed] [Google Scholar]
  • 135.Morse MA, Chapman R, Powderly J, Blackwell K, Keler T, Green J, et al. Phase I study utilizing a novel antigen-presenting cell-targeted vaccine with Toll-like receptor stimulation to induce immunity to self-antigens in cancer patients. Clin Cancer Res. 2011;17:4844–53. doi: 10.1158/1078-0432.CCR-11-0891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Morse MA, Bradley DA, Keler T, Laliberte RJ, Green JA, Davis TA, et al. CDX-1307: a novel vaccine under study as treatment for muscle-invasive bladder cancer. Expert Rev Vaccines. 2011;10:733–42. doi: 10.1586/erv.11.20. [DOI] [PubMed] [Google Scholar]
  • 137.Panter G, Kuznik A, Jerala R. Therapeutic applications of nucleic acids as ligands for Toll-like receptors. Curr Opin Mol Ther. 2009;11:133–45. [PubMed] [Google Scholar]
  • 138.Braun W, Nakano M. Antibody formation: stimulation by polyadenylic and polycytidylic acids. Science. 1967;157:819–21. doi: 10.1126/science.157.3790.819. [DOI] [PubMed] [Google Scholar]
  • 139.Sugiyama T, Hoshino K, Saito M, Yano T, Sasaki I, Yamazaki C, et al. Immunoadjuvant effects of polyadenylic:polyuridylic acids through TLR3 and TLR7. Int Immunol. 2008;20:1–9. doi: 10.1093/intimm/dxm112. [DOI] [PubMed] [Google Scholar]
  • 140.Bot A, Smith D, Phillips B, Bot S, Bona C, Zaghouani H. Immunologic control of tumors by in vivo Fc gamma receptor-targeted antigen loading in conjunction with double-stranded RNA-mediated immune modulation. J Immunol. 2006;176:1363–74. doi: 10.4049/jimmunol.176.3.1363. [DOI] [PubMed] [Google Scholar]
  • 141.Kleinman ME, Yamada K, Takeda A, Chandrasekaran V, Nozaki M, Baffi JZ, et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008;452:591–7. doi: 10.1038/nature06765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Salaun B, Lebecque S, Matikainen S, Rimoldi D, Romero P. Toll-like receptor 3 expressed by melanoma cells as a target for therapy? Clin Cancer Res. 2007;13:4565–74. doi: 10.1158/1078-0432.CCR-07-0274. [DOI] [PubMed] [Google Scholar]
  • 143.Yu L, Chen S. Toll-like receptors expressed in tumor cells: targets for therapy. Cancer Immunol Immunother. 2008;57:1271–8. doi: 10.1007/s00262-008-0459-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Huang B, Zhao J, Li H, He KL, Chen Y, Chen SH, et al. Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res. 2005;65:5009–14. doi: 10.1158/0008-5472.CAN-05-0784. [DOI] [PubMed] [Google Scholar]
  • 145.Lacour J, Lacour F, Spira A, Michelson M, Petit JY, Delage G, et al. Adjuvant treatment with polyadenylic-polyuridylic acid (Polya.Polyu) in operable breast cancer. Lancet. 1980;2:161–4. doi: 10.1016/S0140-6736(80)90057-4. [DOI] [PubMed] [Google Scholar]
  • 146.Laplanche A, Alzieu L, Delozier T, Berlie J, Veyret C, Fargeot P, et al. Polyadenylic-polyuridylic acid plus locoregional radiotherapy versus chemotherapy with CMF in operable breast cancer: a 14 year follow-up analysis of a randomized trial of the Fédération Nationale des Centres de Lutte contre le Cancer (FNCLCC) Breast Cancer Res Treat. 2000;64:189–91. doi: 10.1023/A:1006498121628. [DOI] [PubMed] [Google Scholar]
  • 147.Youn JK, Kim BS, Min JS, Lee KS, Choi HJ, Lee YB, et al. Adjuvant treatment of operable stomach cancer with polyadenylic.polyuridylic acid in addition to chemotherapeutic agents: a preliminary report. Int J Immunopharmacol. 1990;12:289–95. doi: 10.1016/0192-0561(90)90084-Z. [DOI] [PubMed] [Google Scholar]
  • 148.Jeung HC, Moon YW, Rha SY, Yoo NC, Roh JK, Noh SH, et al. Phase III trial of adjuvant 5-fluorouracil and adriamycin versus 5-fluorouracil, adriamycin, and polyadenylic-polyuridylic acid (poly A:U) for locally advanced gastric cancer after curative surgery: final results of 15-year follow-up. Ann Oncol. 2008;19:520–6. doi: 10.1093/annonc/mdm536. [DOI] [PubMed] [Google Scholar]
  • 149.Lacour J, Laplanche A, Malafosse M, Gallot D, Julien M, Rotman N, et al. Polyadenylic-polyuridylic acid as an adjuvant in resectable colorectal carcinoma: a 6 1/2 year follow-up analysis of a multicentric double blind randomized trial. Eur J Surg Oncol. 1992;18:599–604. [PubMed] [Google Scholar]
  • 150.Ducret JP, Caillé P, Sancho Garnier H, Amiel JL, Michelson M, Hovanessian AG, et al. A phase I clinical tolerance study of polyadenylic-polyuridylic acid in cancer patients. J Biol Response Mod. 1985;4:129–33. [PubMed] [Google Scholar]
  • 151.Witt PL, Zahir S, Ritch PS, McAuliffe TM, Ewel CH, Borden EC. Phase I/IB study of polyadenylic-polyuridylic acid in patients with advanced malignancies: clinical and biologic effects. J Interferon Cytokine Res. 1996;16:631–5. doi: 10.1089/jir.1996.16.631. [DOI] [PubMed] [Google Scholar]
  • 152.Reizis B, Colonna M, Trinchieri G, Barrat F, Gilliet M. Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Nat Rev Immunol. 2011;11:558–65. doi: 10.1038/nri3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Cherfils-Vicini J, Platonova S, Gillard M, Laurans L, Validire P, Caliandro R, et al. Triggering of TLR7 and TLR8 expressed by human lung cancer cells induces cell survival and chemoresistance. J Clin Invest. 2010;120:1285–97. doi: 10.1172/JCI36551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303:1529–31. doi: 10.1126/science.1093616. [DOI] [PubMed] [Google Scholar]
  • 155.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;303:1526–9. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
  • 156.Judge AD, Bola G, Lee AC, MacLachlan I. Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo. Mol Ther. 2006;13:494–505. doi: 10.1016/j.ymthe.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 157.Bourquin C, Hotz C, Noerenberg D, Voelkl A, Heidegger S, Roetzer LC, et al. Systemic cancer therapy with a small molecule agonist of toll-like receptor 7 can be improved by circumventing TLR tolerance. Cancer Res. 2011;71:5123–33. doi: 10.1158/0008-5472.CAN-10-3903. [DOI] [PubMed] [Google Scholar]
  • 158.Ahonen CL, Gibson SJ, Smith RM, Pederson LK, Lindh JM, Tomai MA, et al. Dendritic cell maturation and subsequent enhanced T-cell stimulation induced with the novel synthetic immune response modifier R-848. Cell Immunol. 1999;197:62–72. doi: 10.1006/cimm.1999.1555. [DOI] [PubMed] [Google Scholar]
  • 159.Wagner TL, Ahonen CL, Couture AM, Gibson SJ, Miller RL, Smith RM, et al. Modulation of TH1 and TH2 cytokine production with the immune response modifiers, R-848 and imiquimod. Cell Immunol. 1999;191:10–9. doi: 10.1006/cimm.1998.1406. [DOI] [PubMed] [Google Scholar]
  • 160.Caron G, Duluc D, Frémaux 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;175:1551–7. doi: 10.4049/jimmunol.175.3.1551. [DOI] [PubMed] [Google Scholar]
  • 161.Tomai MA, Imbertson LM, Stanczak TL, Tygrett LT, Waldschmidt TJ. The immune response modifiers imiquimod and R-848 are potent activators of B lymphocytes. Cell Immunol. 2000;203:55–65. doi: 10.1006/cimm.2000.1673. [DOI] [PubMed] [Google Scholar]
  • 162.Vasilakos JP, Smith RM, Gibson SJ, Lindh JM, Pederson LK, Reiter MJ, et al. Adjuvant activities of immune response modifier R-848: comparison with CpG ODN. Cell Immunol. 2000;204:64–74. doi: 10.1006/cimm.2000.1689. [DOI] [PubMed] [Google Scholar]
  • 163.Chang BA, Cross JL, Najar HM, Dutz JP. Topical resiquimod promotes priming of CTL to parenteral antigens. Vaccine. 2009;27:5791–9. doi: 10.1016/j.vaccine.2009.07.062. [DOI] [PubMed] [Google Scholar]
  • 164.Mark KE, Corey L, Meng TC, Magaret AS, Huang ML, Selke S, et al. Topical resiquimod 0.01% gel decreases herpes simplex virus type 2 genital shedding: a randomized, controlled trial. J Infect Dis. 2007;195:1324–31. doi: 10.1086/513276. [DOI] [PubMed] [Google Scholar]
  • 165.Pockros PJ, Guyader D, Patton H, Tong MJ, Wright T, McHutchison JG, et al. Oral resiquimod in chronic HCV infection: safety and efficacy in 2 placebo-controlled, double-blind phase IIa studies. J Hepatol. 2007;47:174–82. doi: 10.1016/j.jhep.2007.02.025. [DOI] [PubMed] [Google Scholar]
  • 166.Szeimies RM, Bichel J, Ortonne JP, Stockfleth E, Lee J, Meng TC. A phase II dose-ranging study of topical resiquimod to treat actinic keratosis. Br J Dermatol. 2008;159:205–10. doi: 10.1111/j.1365-2133.2008.08615.x. [DOI] [PubMed] [Google Scholar]
  • 167.Birmachu W, Gleason RM, Bulbulian BJ, Riter CL, Vasilakos JP, Lipson KE, et al. Transcriptional networks in plasmacytoid dendritic cells stimulated with synthetic TLR 7 agonists. BMC Immunol. 2007;8:26. doi: 10.1186/1471-2172-8-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Inglefield JR, Dumitru CD, Alkan SS, Gibson SJ, Lipson KE, Tomai MA, et al. TLR7 agonist 852A inhibition of tumor cell proliferation is dependent on plasmacytoid dendritic cells and type I IFN. J Interferon Cytokine Res. 2008;28:253–63. doi: 10.1089/jir.2007.0097. [DOI] [PubMed] [Google Scholar]
  • 169.Dumitru CD, Antonysamy MA, Tomai MA, Lipson KE. Potentiation of the anti-tumor effects of imidazoquinoline immune response modifiers by cyclophosphamide. Cancer Biol Ther. 2010;10:155–65. doi: 10.4161/cbt.10.2.12163. [DOI] [PubMed] [Google Scholar]
  • 170.Dudek AZ, Yunis C, Harrison LI, Kumar S, Hawkinson R, Cooley S, et al. First in human phase I trial of 852A, a novel systemic toll-like receptor 7 agonist, to activate innate immune responses in patients with advanced cancer. Clin Cancer Res. 2007;13:7119–25. doi: 10.1158/1078-0432.CCR-07-1443. [DOI] [PubMed] [Google Scholar]
  • 171.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;47:962–9. doi: 10.1177/0091270007303766. [DOI] [PubMed] [Google Scholar]
  • 172.Dummer R, Hauschild A, Becker JC, Grob JJ, Schadendorf D, Tebbs V, et al. An exploratory study of systemic administration of the toll-like receptor-7 agonist 852A in patients with refractory metastatic melanoma. Clin Cancer Res. 2008;14:856–64. doi: 10.1158/1078-0432.CCR-07-1938. [DOI] [PubMed] [Google Scholar]
  • 173.Geller MA, Cooley S, Argenta PA, Downs LS, Carson LF, Judson PL, et al. Toll-like receptor-7 agonist administered subcutaneously in a prolonged dosing schedule in heavily pretreated recurrent breast, ovarian, and cervix cancers. Cancer Immunol Immunother. 2010;59:1877–84. doi: 10.1007/s00262-010-0914-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Lu H, Dietsch GN, Matthews MA, Yang Y, Ghanekar S, Inokuma M, et al. VTX-2337 is a novel TLR8 agonist that activates NK cells and augments ADCC. Clin Cancer Res. 2012;18:499–509. doi: 10.1158/1078-0432.CCR-11-1625. [DOI] [PubMed] [Google Scholar]
  • 175.Cohen PA, Northfelt DW, Weiss GJ, Von Hoff DD, Manjarrez K, Dietsch G, et al. Phase I clinical trial of VTX-2337, a selective toll-like receptor 8 (TLR8) agonist, in patients with advanced solid tumors. J Clin Oncol 2011; 29:abstr 2537.
  • 176.Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov. 2006;5:471–84. doi: 10.1038/nrd2059. [DOI] [PubMed] [Google Scholar]
  • 177.Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27:161–7. doi: 10.1038/sj.onc.1210911. [DOI] [PubMed] [Google Scholar]
  • 178.Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000;408:740–5. doi: 10.1038/35047123. [DOI] [PubMed] [Google Scholar]
  • 179.Barton GM, Kagan JC, Medzhitov R. Intracellular localization of Toll-like receptor 9 prevents recognition of self DNA but facilitates access to viral DNA. Nat Immunol. 2006;7:49–56. doi: 10.1038/ni1280. [DOI] [PubMed] [Google Scholar]
  • 180.Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA, Teasdale R, et al. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 1995;374:546–9. doi: 10.1038/374546a0. [DOI] [PubMed] [Google Scholar]
  • 181.Liang X, Moseman EA, Farrar MA, Bachanova V, Weisdorf DJ, Blazar BR, et al. Toll-like receptor 9 signaling by CpG-B oligodeoxynucleotides induces an apoptotic pathway in human chronic lymphocytic leukemia B cells. Blood. 2010;115:5041–52. doi: 10.1182/blood-2009-03-213363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Carpentier AF, Chen L, Maltonti F, Delattre JY. Oligodeoxynucleotides containing CpG motifs can induce rejection of a neuroblastoma in mice. Cancer Res. 1999;59:5429–32. [PubMed] [Google Scholar]
  • 183.Carpentier AF, Xie J, Mokhtari K, Delattre JY. Successful treatment of intracranial gliomas in rat by oligodeoxynucleotides containing CpG motifs. Clin Cancer Res. 2000;6:2469–73. [PubMed] [Google Scholar]
  • 184.Vicari AP, Luu R, Zhang N, Patel S, Makinen SR, Hanson DC, et al. Paclitaxel reduces regulatory T cell numbers and inhibitory function and enhances the anti-tumor effects of the TLR9 agonist PF-3512676 in the mouse. Cancer Immunol Immunother. 2009;58:615–28. doi: 10.1007/s00262-008-0586-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Meng Y, Carpentier AF, Chen L, Boisserie G, Simon JM, Mazeron JJ, et al. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. Int J Cancer. 2005;116:992–7. doi: 10.1002/ijc.21131. [DOI] [PubMed] [Google Scholar]
  • 186.Paul S. Technology evaluation: CpG-7909, Coley. Curr Opin Mol Ther. 2003;5:553–9. [PubMed] [Google Scholar]
  • 187.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;24:693–701. doi: 10.1007/s10875-004-6244-3. [DOI] [PubMed] [Google Scholar]
  • 188.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;22:3136–43. doi: 10.1016/j.vaccine.2004.01.058. [DOI] [PubMed] [Google Scholar]
  • 189.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;46:1310–4. doi: 10.1086/533467. [DOI] [PubMed] [Google Scholar]
  • 190.Molenkamp BG, van Leeuwen PA, Meijer S, Sluijter BJ, Wijnands PG, Baars A, et al. Intradermal CpG-B activates both plasmacytoid and myeloid dendritic cells in the sentinel lymph node of melanoma patients. Clin Cancer Res. 2007;13:2961–9. doi: 10.1158/1078-0432.CCR-07-0050. [DOI] [PubMed] [Google Scholar]
  • 191.Krieg AM, Efler SM, Wittpoth M, Al Adhami MJ, Davis HL. Induction of systemic TH1-like innate immunity in normal volunteers following subcutaneous but not intravenous administration of CPG 7909, a synthetic B-class CpG oligodeoxynucleotide TLR9 agonist. J Immunother. 2004;27:460–71. doi: 10.1097/00002371-200411000-00006. [DOI] [PubMed] [Google Scholar]
  • 192.Speiser DE, Liénard 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;115:739–46. doi: 10.1172/JCI23373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Molenkamp BG, Sluijter BJ, van Leeuwen PA, Santegoets SJ, Meijer S, Wijnands PG, et al. Local administration of PF-3512676 CpG-B instigates tumor-specific CD8+ T-cell reactivity in melanoma patients. Clin Cancer Res. 2008;14:4532–42. doi: 10.1158/1078-0432.CCR-07-4711. [DOI] [PubMed] [Google Scholar]
  • 194.Zent CS, Smith BJ, Ballas ZK, Wooldridge JE, Link BK, Call TG, et al. Phase I clinical trial of CpG oligonucleotide 7909 (PF-03512676) in patients with previously treated chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53:211–7. doi: 10.3109/10428194.2011.608451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Link BK, Ballas ZK, Weisdorf D, Wooldridge JE, Bossler AD, Shannon M, et al. Oligodeoxynucleotide CpG 7909 delivered as intravenous infusion demonstrates immunologic modulation in patients with previously treated non-Hodgkin lymphoma. J Immunother. 2006;29:558–68. doi: 10.1097/01.cji.0000211304.60126.8f. [DOI] [PubMed] [Google Scholar]
  • 196.Leonard JP, Link BK, Emmanouilides C, Gregory SA, Weisdorf D, Andrey J, et al. Phase I trial of toll-like receptor 9 agonist PF-3512676 with and following rituximab in patients with recurrent indolent and aggressive non Hodgkin’s lymphoma. Clin Cancer Res. 2007;13:6168–74. doi: 10.1158/1078-0432.CCR-07-0815. [DOI] [PubMed] [Google Scholar]
  • 197.Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol. 2010;28:4324–32. doi: 10.1200/JCO.2010.28.9793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Kim YH, Girardi M, Duvic M, Kuzel T, Link BK, Pinter-Brown L, et al. Phase I trial of a Toll-like receptor 9 agonist, PF-3512676 (CPG 7909), in patients with treatment-refractory, cutaneous T-cell lymphoma. J Am Acad Dermatol. 2010;63:975–83. doi: 10.1016/j.jaad.2009.12.052. [DOI] [PubMed] [Google Scholar]
  • 199.Hofmann MA, Kors C, Audring H, Walden P, Sterry W, Trefzer U. Phase 1 evaluation of intralesionally injected TLR9-agonist PF-3512676 in patients with basal cell carcinoma or metastatic melanoma. J Immunother. 2008;31:520–7. doi: 10.1097/CJI.0b013e318174a4df. [DOI] [PubMed] [Google Scholar]
  • 200.Pashenkov M, Goëss G, Wagner C, Hörmann M, Jandl T, Moser A, et al. Phase II trial of a toll-like receptor 9-activating oligonucleotide in patients with metastatic melanoma. J Clin Oncol. 2006;24:5716–24. doi: 10.1200/JCO.2006.07.9129. [DOI] [PubMed] [Google Scholar]
  • 201.Weber JS, Zarour H, Redman B, Trefzer U, O’Day S, van den Eertwegh AJ, et al. Randomized phase 2/3 trial of CpG oligodeoxynucleotide PF-3512676 alone or with dacarbazine for patients with unresectable stage III and IV melanoma. Cancer. 2009;115:3944–54. doi: 10.1002/cncr.24473. [DOI] [PubMed] [Google Scholar]
  • 202.Tarhini AA, Leng S, Moschos SJ, Yin Y, Sander C, Lin Y, et al. Safety and Immunogenicity of Vaccination With MART-1 (26-35, 27L), gp100 (209-217, 210M), and Tyrosinase (368-376, 370D) In Adjuvant With PF-3512676 and GM-CSF In Metastatic Melanoma. J Immunother. 2012;35:359–66. doi: 10.1097/CJI.0b013e31825481fe. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Iwahashi M, Katsuda M, Nakamori M, Nakamura M, Naka T, Ojima T, et al. Vaccination with peptides derived from cancer-testis antigens in combination with CpG-7909 elicits strong specific CD8+ T cell response in patients with metastatic esophageal squamous cell carcinoma. Cancer Sci. 2010;101:2510–7. doi: 10.1111/j.1349-7006.2010.01732.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Manegold C, Gravenor D, Woytowitz D, Mezger J, Hirsh V, Albert G, et al. Randomized phase II trial of a toll-like receptor 9 agonist oligodeoxynucleotide, PF-3512676, in combination with first-line taxane plus platinum chemotherapy for advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008;26:3979–86. doi: 10.1200/JCO.2007.12.5807. [DOI] [PubMed] [Google Scholar]
  • 205.Yamada K, Nakao M, Fukuyama C, Nokihara H, Yamamoto N, Sekine I, et al. Phase I study of TLR9 agonist PF-3512676 in combination with carboplatin and paclitaxel in patients with advanced non-small-cell lung cancer. Cancer Sci. 2010;101:188–95. doi: 10.1111/j.1349-7006.2009.01361.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Hirsh V, Paz-Ares L, Boyer M, Rosell R, Middleton G, Eberhardt WE, et al. Randomized phase III trial of paclitaxel/carboplatin with or without PF-3512676 (Toll-like receptor 9 agonist) as first-line treatment for advanced non-small-cell lung cancer. J Clin Oncol. 2011;29:2667–74. doi: 10.1200/JCO.2010.32.8971. [DOI] [PubMed] [Google Scholar]
  • 207.Thompson JA, Kuzel T, Drucker BJ, Urba WJ, Bukowski RM. Safety and efficacy of PF-3512676 for the treatment of stage IV renal cell carcinoma: an open-label, multicenter phase I/II study. Clin Genitourin Cancer. 2009;7:E58–65. doi: 10.3816/CGC.2009.n.025. [DOI] [PubMed] [Google Scholar]
  • 208.Karbach J, Neumann A, Atmaca A, Wahle C, Brand K, von Boehmer L, et al. Efficient in vivo priming by vaccination with recombinant NY-ESO-1 protein and CpG in antigen naive prostate cancer patients. Clin Cancer Res. 2011;17:861–70. doi: 10.1158/1078-0432.CCR-10-1811. [DOI] [PubMed] [Google Scholar]
  • 209.Kensil CR, Kammer R. QS-21: a water-soluble triterpene glycoside adjuvant. Expert Opin Investig Drugs. 1998;7:1475–82. doi: 10.1517/13543784.7.9.1475. [DOI] [PubMed] [Google Scholar]
  • 210.Kruit WH, Suciu S, Dreno B, Chiarion-Sileni V, Mortier L, Robert C, et al. Immunization with recombinant MAGE-A3 protein combined with adjuvant systems AS15 or AS02B in patients with unresectable and progressive metastatic cutaneous melanoma: A randomized open-label phase II study of the EORTC Melanoma Group (16032- 18031). J Clin Oncol 2008; 26:abstr 9065.
  • 211.Louhaed J, Gruselle O, Gaulis S, Coche T, Eggermont AM, Kruit WH, et al. Expression of defined genes identified by pretreatment tumor profiling: Association with clinical responses to the GSK MAGE-A3 immunotherapeutic in metastatic melanoma patients (EORTC 16032-18031). J Clin Oncol 2008; 26:abstr 9045.
  • 212.Manegold C, van Zandwijk N, Szczesna A, Zatloukal P, Au JS, Blasinska-Morawiec M, et al. A phase III randomized study of gemcitabine and cisplatin with or without PF-3512676 (TLR9 agonist) as first-line treatment of advanced non-small-cell lung cancer. Ann Oncol. 2012;23:72–7. doi: 10.1093/annonc/mdr030. [DOI] [PubMed] [Google Scholar]
  • 213.Carpentier A, Laigle-Donadey F, Zohar S, Capelle L, Behin A, Tibi A, et al. Phase 1 trial of a CpG oligodeoxynucleotide for patients with recurrent glioblastoma. Neuro Oncol. 2006;8:60–6. doi: 10.1215/S1522851705000475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Carpentier A, Metellus P, Ursu R, Zohar S, Lafitte F, Barrié M, et al. Intracerebral administration of CpG oligonucleotide for patients with recurrent glioblastoma: a phase II study. Neuro Oncol. 2010;12:401–8. doi: 10.1093/neuonc/nop047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Agrawal S, Kandimalla ER. Synthetic agonists of Toll-like receptors 7, 8 and 9. Biochem Soc Trans. 2007;35:1461–7. doi: 10.1042/BST0351461. [DOI] [PubMed] [Google Scholar]
  • 216.Kandimalla ER, Bhagat L, Li Y, Yu D, Wang D, Cong YP, et al. Immunomodulatory oligonucleotides containing a cytosine-phosphate-2′-deoxy-7-deazaguanosine motif as potent toll-like receptor 9 agonists. Proc Natl Acad Sci U S A. 2005;102:6925–30. doi: 10.1073/pnas.0501729102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Wang D, Li Y, Yu D, Song SS, Kandimalla ER, Agrawal S. Immunopharmacological and antitumor effects of second-generation immunomodulatory oligonucleotides containing synthetic CpR motifs. Int J Oncol. 2004;24:901–8. [PubMed] [Google Scholar]
  • 218.Damiano V, Caputo R, Garofalo S, Bianco R, Rosa R, Merola G, et al. TLR9 agonist acts by different mechanisms synergizing with bevacizumab in sensitive and cetuximab-resistant colon cancer xenografts. Proc Natl Acad Sci U S A. 2007;104:12468–73. doi: 10.1073/pnas.0705226104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Damiano V, Garofalo S, Rosa R, Bianco R, Caputo R, Gelardi T, et al. A novel toll-like receptor 9 agonist cooperates with trastuzumab in trastuzumab-resistant breast tumors through multiple mechanisms of action. Clin Cancer Res. 2009;15:6921–30. doi: 10.1158/1078-0432.CCR-09-1599. [DOI] [PubMed] [Google Scholar]
  • 220.Malik S, Hwang J, Cotarla I, Sullivan T, Kerr R, Marshall J. Initial phase 1 results of gemcitabine, carboplatin and IMO-2055, a toll like receptor 9 (TLR9) agonist, in patients (pts) with advanced solid tumors. J Thorac Oncol. 2007;2:S726–7. doi: 10.1097/01.JTO.0000284088.13204.12. [DOI] [Google Scholar]
  • 221.Boehrer S, Adès L, Braun T, Galluzzi L, Grosjean J, Fabre C, et al. Erlotinib exhibits antineoplastic off-target effects in AML and MDS: a preclinical study. Blood. 2008;111:2170–80. doi: 10.1182/blood-2007-07-100362. [DOI] [PubMed] [Google Scholar]
  • 222.Immunostimulatory DNA–Dynavax Immunostimulatory DNA--Dynavax. AIC, Amb a 1 immunostimulatory conjugate, HBV-ISS, ISS 1018, ISS DNA, ISS DNA--dynavax, ISS1, ISS2. Drugs R D. 2002;3:193–6. doi: 10.2165/00126839-200203030-00011. [DOI] [PubMed] [Google Scholar]
  • 223.Teleshova N, Kenney J, Jones J, Marshall J, Van Nest G, Dufour J, et al. CpG-C immunostimulatory oligodeoxyribonucleotide activation of plasmacytoid dendritic cells in rhesus macaques to augment the activation of IFN-gamma-secreting simian immunodeficiency virus-specific T cells. J Immunol. 2004;173:1647–57. doi: 10.4049/jimmunol.173.3.1647. [DOI] [PubMed] [Google Scholar]
  • 224.Ponzio NM, Cutro S, Hu J, Marzouk A, Marshall JD. CpG oligodeoxynucleotide-induced immunity prevents growth of germinal center-derived B lymphoma cells. Int Immunopharmacol. 2006;6:2057–68. doi: 10.1016/j.intimp.2006.08.008. [DOI] [PubMed] [Google Scholar]
  • 225.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;21:2461–7. doi: 10.1016/S0264-410X(03)00045-8. [DOI] [PubMed] [Google Scholar]
  • 226.Barry M, Cooper C. Review of hepatitis B surface antigen-1018 ISS adjuvant-containing vaccine safety and efficacy. Expert Opin Biol Ther. 2007;7:1731–7. doi: 10.1517/14712598.7.11.1731. [DOI] [PubMed] [Google Scholar]
  • 227.Friedberg JW, Kim H, McCauley M, Hessel EM, Sims P, Fisher DC, et al. Combination immunotherapy with a CpG oligonucleotide (1018 ISS) and rituximab in patients with non-Hodgkin lymphoma: increased interferon-alpha/beta-inducible gene expression, without significant toxicity. Blood. 2005;105:489–95. doi: 10.1182/blood-2004-06-2156. [DOI] [PubMed] [Google Scholar]
  • 228.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch—Monoclonal antibodies in cancer therapy. Oncoimmunol. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Friedberg JW, Kelly JL, Neuberg D, Peterson DR, Kutok JL, Salloum R, et al. Phase II study of a TLR-9 agonist (1018 ISS) with rituximab in patients with relapsed or refractory follicular lymphoma. Br J Haematol. 2009;146:282–91. doi: 10.1111/j.1365-2141.2009.07773.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Schmidt M, Anton K, Nordhaus C, Junghans C, Wittig B, Worm M. Cytokine and Ig-production by CG-containing sequences with phosphorodiester backbone and dumbbell-shape. Allergy. 2006;61:56–63. doi: 10.1111/j.1398-9995.2005.00908.x. [DOI] [PubMed] [Google Scholar]
  • 231.Köchling J, Prada J, Bahrami M, Stripecke R, Seeger K, Henze G, et al. Anti-tumor effect of DNA-based vaccination and dSLIM immunomodulatory molecules in mice with Ph+ acute lymphoblastic leukaemia. Vaccine. 2008;26:4669–75. doi: 10.1016/j.vaccine.2008.06.094. [DOI] [PubMed] [Google Scholar]
  • 232.Weihrauch MR, Ansén S, Jurkiewicz E, Geisen C, Xia Z, Anderson KS, et al. Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP-1 peptide and irinotecan, 5-fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res. 2005;11:5993–6001. doi: 10.1158/1078-0432.CCR-05-0018. [DOI] [PubMed] [Google Scholar]
  • 233.Tschaika M, Schmoll H, Scheithauer W, Mayer F, Schroff M, Schmidt M, et al. Preliminary results of an ongoing phase II/III clinical study of the TLR9 agonist MGN1703 in patients with advanced colorectal carcinoma with disease control after first-line induction therapy (IMPACT Study). J Clin Oncol 2011; 29:abstr 618.
  • 234.Tschaika M, Schmoll H, Riera-Knorrenschild J, Nitsche D, Trojan J, Kröning H, et al. IMPACT study: A phase II-III clinical study with the immunomodulator MGN1703 in patients with advanced colorectal carcincoma. J Clin Oncol 2012; 30:abstr 633.
  • 235.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;410:1099–103. doi: 10.1038/35074106. [DOI] [PubMed] [Google Scholar]
  • 236.Yoon SI, Kurnasov O, Natarajan V, Hong M, Gudkov AV, Osterman AL, et al. Structural basis of TLR5-flagellin recognition and signaling. Science. 2012;335:859–64. doi: 10.1126/science.1215584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Smith KD, Andersen-Nissen E, Hayashi F, Strobe K, Bergman MA, Barrett SL, et al. Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility. Nat Immunol. 2003;4:1247–53. doi: 10.1038/ni1011. [DOI] [PubMed] [Google Scholar]
  • 238.Lu J, Sun PD. The structure of the TLR5-flagellin complex: a new mode of pathogen detection, conserved receptor dimerization for signaling. Sci Signal. 2012;5:pe11. doi: 10.1126/scisignal.2002963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Burdelya LG, Krivokrysenko VI, Tallant TC, Strom E, Gleiberman AS, Gupta D, et al. An agonist of toll-like receptor 5 has radioprotective activity in mouse and primate models. Science. 2008;320:226–30. doi: 10.1126/science.1154986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Fukuzawa N, Petro M, Baldwin WM, 3rd, Gudkov AV, Fairchild RLA. A TLR5 agonist inhibits acute renal ischemic failure. J Immunol. 2011;187:3831–9. doi: 10.4049/jimmunol.1003238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Burdelya LG, Gleiberman AS, Toshkov I, Aygun-Sunar S, Bapardekar M, Manderscheid-Kern P, et al. Toll-like receptor 5 agonist protects mice from dermatitis and oral mucositis caused by local radiation: implications for head-and-neck cancer radiotherapy. Int J Radiat Oncol Biol Phys. 2012;83:228–34. doi: 10.1016/j.ijrobp.2011.05.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Sfondrini L, Rossini A, Besusso D, Merlo A, Tagliabue E, Mènard S, et al. Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer. J Immunol. 2006;176:6624–30. doi: 10.4049/jimmunol.176.11.6624. [DOI] [PubMed] [Google Scholar]
  • 243.Talbot HK, Rock MT, Johnson C, Tussey L, Kavita U, Shanker A, et al. Immunopotentiation of trivalent influenza vaccine when given with VAX102, a recombinant influenza M2e vaccine fused to the TLR5 ligand flagellin. PLoS One. 2010;5:e14442. doi: 10.1371/journal.pone.0014442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Turley CB, Rupp RE, Johnson C, Taylor DN, Wolfson J, Tussey L, et al. Safety and immunogenicity of a recombinant M2e-flagellin influenza vaccine (STF2.4xM2e) in healthy adults. Vaccine. 2011;29:5145–52. doi: 10.1016/j.vaccine.2011.05.041. [DOI] [PubMed] [Google Scholar]
  • 245.Hancock RE, Nijnik A, Philpott DJ. Modulating immunity as a therapy for bacterial infections. Nat Rev Microbiol. 2012;10:243–54. doi: 10.1038/nrmicro2745. [DOI] [PubMed] [Google Scholar]
  • 246.Ahmad F, Mani J, Kumar P, Haridas S, Upadhyay P, Bhaskar S. Activation of anti-tumor immune response and reduction of regulatory T cells with Mycobacterium indicus pranii (MIP) therapy in tumor bearing mice. PLoS One. 2011;6:e25424. doi: 10.1371/journal.pone.0025424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Rakshit S, Ponnusamy M, Papanna S, Saha B, Ahmed A, Nandi D. Immunotherapeutic efficacy of Mycobacterium indicus pranii in eliciting anti-tumor T cell responses: critical roles of IFNγ. Int J Cancer. 2012;130:865–75. doi: 10.1002/ijc.26099. [DOI] [PubMed] [Google Scholar]
  • 248.Chaudhuri P, Mukhopadhyay S. Bladder preserving approach for muscle invasive bladder cancer--role of mycobacterium w. J Indian Med Assoc. 2003;101:559–60. [PubMed] [Google Scholar]
  • 249.Sur PK, Dastidar AG. Role of mycobacterium w as adjuvant treatment of lung cancer (non-small cell lung cancer). J Indian Med Assoc 2003; 101:118, 20. [PubMed]
  • 250.Pant MC, Chakraborty BS, Patel N, Bisht SS, Verma VP, Singh S, et al. Role of Cadi-05 as an adjuvant therapy in advanced non small cell lung cancer. J Thorac Oncol. 2007;2:S616. doi: 10.1097/01.JTO.0000283779.80210.45. [DOI] [Google Scholar]
  • 251.Yuksel ZS, Buber E, Kocagoz T, Alp A, Saribas Z, Acan NL. Mycobacterial strains that stimulate the immune system most efficiently as candidates for the treatment of bladder cancer. J Mol Microbiol Biotechnol. 2011;20:24–8. doi: 10.1159/000324331. [DOI] [PubMed] [Google Scholar]
  • 252.Fowler DW, Copier J, Wilson N, Dalgleish AG, Bodman-Smith MD. Mycobacteria activate γδ T-cell anti-tumour responses via cytokines from type 1 myeloid dendritic cells: a mechanism of action for cancer immunotherapy. Cancer Immunol Immunother. 2012;61:535–47. doi: 10.1007/s00262-011-1121-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Stanford J, Stanford C, Stansby G, Bottasso O, Bahr G, Grange J. The common mycobacterial antigens and their importance in the treatment of disease. Curr Pharm Des. 2009;15:1248–60. doi: 10.2174/138161209787846838. [DOI] [PubMed] [Google Scholar]
  • 254.Maraveyas A, Baban B, Kennard D, Rook GA, Westby M, Grange JM, et al. Possible improved survival of patients with stage IV AJCC melanoma receiving SRL 172 immunotherapy: correlation with induction of increased levels of intracellular interleukin-2 in peripheral blood lymphocytes. Ann Oncol. 1999;10:817–24. doi: 10.1023/A:1008307821189. [DOI] [PubMed] [Google Scholar]
  • 255.Patel PM, Sim S, O’Donnell DO, Protheroe A, Beirne D, Stanley A, et al. An evaluation of a preparation of Mycobacterium vaccae (SRL172) as an immunotherapeutic agent in renal cancer. Eur J Cancer. 2008;44:216–23. doi: 10.1016/j.ejca.2007.11.003. [DOI] [PubMed] [Google Scholar]
  • 256.Stanford JL, Stanford CA, O’Brien ME, Grange JM. Successful immunotherapy with Mycobacterium vaccae in the treatment of adenocarcinoma of the lung. Eur J Cancer. 2008;44:224–7. doi: 10.1016/j.ejca.2007.08.021. [DOI] [PubMed] [Google Scholar]
  • 257.Stebbing J, Dalgleish A, Gifford-Moore A, Martin A, Gleeson C, Wilson G, et al. An intra-patient placebo-controlled phase I trial to evaluate the safety and tolerability of intradermal IMM-101 in melanoma. Ann Oncol. 2012;23:1314–9. doi: 10.1093/annonc/mdr363. [DOI] [PubMed] [Google Scholar]
  • 258.de Vries IJ, Tel J, Benitez-Ribas D, Torensma R, Figdor CG. Prophylactic vaccines mimic synthetic CpG oligonucleotides in their ability to modulate immune responses. Mol Immunol. 2011;48:810–7. doi: 10.1016/j.molimm.2010.12.022. [DOI] [PubMed] [Google Scholar]
  • 259.Schreibelt G, Benitez-Ribas D, Schuurhuis D, Lambeck AJ, van Hout-Kuijer M, Schaft N, et al. Commonly used prophylactic vaccines as an alternative for synthetically produced TLR ligands to mature monocyte-derived dendritic cells. Blood. 2010;116:564–74. doi: 10.1182/blood-2009-11-251884. [DOI] [PubMed] [Google Scholar]
  • 260.Jin MS, Kim SE, Heo JY, Lee ME, Kim HM, Paik SG, et al. Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell. 2007;130:1071–82. doi: 10.1016/j.cell.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 261.Takeuchi O, Sato S, Horiuchi T, Hoshino K, Takeda K, Dong Z, et al. Cutting edge: role of Toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol. 2002;169:10–4. doi: 10.4049/jimmunol.169.1.10. [DOI] [PubMed] [Google Scholar]
  • 262.Razonable RR, Henault M, Lee LN, Laethem C, Johnston PA, Watson HL, et al. Secretion of proinflammatory cytokines and chemokines during amphotericin B exposure is mediated by coactivation of toll-like receptors 1 and 2. Antimicrob Agents Chemother. 2005;49:1617–21. doi: 10.1128/AAC.49.4.1617-1621.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Kang JY, Nan X, Jin MS, Youn SJ, Ryu YH, Mah S, et al. Recognition of lipopeptide patterns by Toll-like receptor 2-Toll-like receptor 6 heterodimer. Immunity. 2009;31:873–84. doi: 10.1016/j.immuni.2009.09.018. [DOI] [PubMed] [Google Scholar]
  • 264.Takeuchi O, Kawai T, Mühlradt PF, Morr M, Radolf JD, Zychlinsky A, et al. Discrimination of bacterial lipoproteins by Toll-like receptor 6. Int Immunol. 2001;13:933–40. doi: 10.1093/intimm/13.7.933. [DOI] [PubMed] [Google Scholar]
  • 265.Krieg AM. CpG motifs: the active ingredient in bacterial extracts? Nat Med. 2003;9:831–5. doi: 10.1038/nm0703-831. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES