Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2014 Aug 1;3:e29179. doi: 10.4161/onci.29179

Trial Watch

Toll-like receptor agonists in oncological indications

Fernando Aranda 1,2,3,4,, Erika Vacchelli 1,2,3,4,, Florine Obrist 1,2,3,4, Alexander Eggermont 1, Jérôme Galon 2,5,6, Catherine Sautès-Fridman 2,5,7,8, Isabelle Cremer 2,7,5,8, Jan Henrik ter Meulen 9, Laurence Zitvogel 1,10, Guido Kroemer 11,12,2,3,5,‡,*, Lorenzo Galluzzi 1,3,5,‡,*
PMCID: PMC4091055  PMID: 25083332

Abstract

Toll-like receptors (TLRs) are an evolutionarily conserved group of enzymatically inactive, single membrane-spanning proteins that recognize a wide panel of exogenous and endogenous danger signals. Besides constituting a crucial component of the innate immune response to bacterial and viral pathogens, TLRs appear to play a major role in anticancer immunosurveillance. In line with this notion, several natural and synthetic TLR ligands have been intensively investigated for their ability to boost tumor-targeting immune responses elicited by a variety of immunotherapeutic and chemotherapeutic interventions. Three of these agents are currently approved by the US Food and Drug Administration (FDA) or equivalent regulatory agencies for use in cancer patients: the so-called bacillus Calmette-Guérin, monophosphoryl lipid A, and imiquimod. However, the number of clinical trials testing the therapeutic potential of both FDA-approved and experimental TLR agonists in cancer patients is stably decreasing, suggesting that drug developers and oncologists are refocusing their interest on alternative immunostimulatory agents. Here, we summarize recent findings on the use of TLR agonists in cancer patients and discuss how the clinical evaluation of FDA-approved and experimental TLR ligands has evolved since the publication of our first Trial Watch dealing with this topic.

Keywords: BCG, CpG-7909, Hiltonol™, damage-associated molecular patterns, polyI:C, resiquimod

Introduction

Organisms as evolutionarily distant as plants, flies, fish and mammals detect invading pathogens via a panel of conserved, enzymatically inactive, single membrane-spanning proteins commonly known as Toll-like receptors (TLRs),1-3 owing to their high degree of homology with Drosophila melanogaster Toll.4-6 At present, 13 distinct TLRs have been identified in mice (Tlr1-Tlr13), 10 of which are also encoded by the human genome (TLR1-TLR10).7,8 Conversely, TLR11 is a pseudogene and human cells are devoid of Tlr12 and Tlr13 homologs.7-9

TLRs are mainly expressed by immune cells including monocytes, mature macrophages, mast cells and dendritic cells (DCs) as well as by other cells involved in the first-line defense against infection (e.g., intestinal epithelial cells).10,11 TLRs sense indeed a wide panel of conserved microbial components that are cumulatively referred to as “microbe-associated molecular patterns” (MAMPs), including specific nucleic acids and peculiar proteo-lipidic structures like lipopolysaccharide (LPS).12-28 Moreover, several TLRs have been shown to respond to so-called “damage-associated molecular patterns” (DAMPs), i.e., endogenous molecules that are released by stressed, dying or dead cells as a signal of danger.29-34 This is for instance the case of TLR2 and TLR4,35-40 both of which are expressed on the cell surface (similar to other TLRs that recognize proteo-lipidic components),8,41 as well as of TLR9,42,43 which is mainly expressed in the endosomal compartment (hence resembling other TLRs that detect nucleic acids).8,41

Upon binding to their ligands, most TLRs form dimers that recruit the adaptor proteins myeloid differentiation primary response 88 (MYD88) and toll-interleukin 1 receptor domain containing adaptor protein (TIRAP, also known as MAL), hence initiating the assembly of plasma-membrane proximal multiprotein signaling complex.44,45 Among several effects, such a supramolecular complex promotes the activation of the transcription factor NF-κB, hence driving the synthesis of multiple pro-inflammatory cytokines.44,45 At odds with other TLRs, all of which employ MYD88 as an obligate or facultative adaptor, TLR3 dimers trigger a MYD88-independent signaling pathway that critically relies on toll-like receptor adaptor molecule 1 (TICAM1, also known as TRIF) and does not trigger NF-κB activation.46-48 Rather, TICAM1-transduced signals activate interferon regulatory factor 7 (IRF7),49,50 hence regulating the expression of type I interferons (IFNs) and other IFN-responsive mediators.46-48 Of note, Ticam1−/− mice exhibit defects in both TLR3 and TLR4 signaling,47 suggesting that TLR4 can also employ TICAM1 as an adaptor, at least under some circumstances. Moreover, MYD88 and TIRAP have been shown to actively inhibit TLR3 signaling,51-53 lending further support to the notion that the functional profile of TLR3 is rather dissimilar from that of other members of the TLR family.8,41,54-56

Reflecting their ligand-binding profile, TLRs are not only crucial for the initiation of an innate immune response against bacterial and viral pathogens,7,8 but also participate in the organismal reaction to inflammatory conditions that usually do not involve a microbial component, including a wide array of neoplasms.43,57,58 In line with this notion, single nucleotide polymorphisms (SNPs) that alter the functionality of multiple TLRs have been shown to influence the natural progression of several tumors of non-viral etiology.59-77 Of note, TLR signaling is impaired in many neoplasms of viral etiology, including hepatitis B virus-associated hepatocellular carcinoma, human papillomavirus (HPV)-associated cervical carcinoma and Merkel cell carcinoma.78 At least in part, this stems from the ability of several viruses to avoid the activation of TLR9 by their own nucleic acids.78

Some TLRs have also been shown to play a critical role in the (re)activation of tumor-specific immune responses by a diverse array of chemo-, radio- and immunotherapeutic interventions.79-85 Accordingly, SNPs that reduce the activity of various TLRs have been shown to negatively impact the response of cancer patients to therapy in multiple scenarios.83,86-88 At least in part, this reflects the key role that some TLRs play in the perception of cell death as immunogenic.79,80 Indeed, cancer cells subjected to specific therapeutic regimens, including irradiation, some forms of photodynamic therapy as well as doxorubicin-, mitoxantrone-, and oxaliplatin-based chemotherapy, die while emitting a spatiotemporally defined combination of DAMPs that renders them capable of triggering a therapeutically relevant adaptive immune response.79,80

Three TLR agonists are currently licensed by the US Food and Drug Administration and equivalent regulatory agencies for use in cancer patients. First, the so-called bacillus Calmette-Guérin (BCG): an attenuated variant of Mycobacterium bovis originally conceived as an anti-tuberculosis vaccine that is nowadays approved as a standalone immunotherapeutic intervention in patients with non-invasive transitional cell carcinoma of the bladder.89 BCG appears to operate as a mixed TLR2/TLR4 agonist.90,91 Second, monophosphoryl lipid A (MPL): a derivative of Salmonella minnesota LPS that is currently employed as adjuvant in Cervarix®, a vaccine specific for HPV-16 and -18.92,93 MPL resembles BCG in its ability to trigger both TLR2 and TLR4 signaling.14 Third, imiquimod: an imidazoquinoline derivative and guanosine analog that is nowadays used for the topical therapy of actinic keratosis, superficial basal cell carcinoma and external genital/perianal warts (condylomata acuminata).89 At odds with BCG and MPL, imiquimod (formerly known as R837) mostly operates through TLR7.94-96 This said, Aldara® (imiquimod 5% cream as commercialized by 3M Pharmaceuticals) has been shown to mediate TLR7-independent immunostimulatory effects, perhaps owing to the pro-inflammatory activity of isostearic acid.97,98 Of note, picibanil – a lyophilized preparation of Streptococcus pyogenes that activates TLR2 and TLR4 – is not licensed by the US FDA but has been approved for use in cancer patients by the Japanese Ministry of Health and Welfare as early as in 1975.57,99

Along the lines of our monthly Trial Watch series,100,101 here we summarize recent key discoveries on the biological activity of TLRs and discuss the latest developments on the use of natural and synthetic TLR agonists as therapeutic agents in cancer patients.

Literature Update

To the best of our knowledge, the results of no more than 6 studies assessing the clinical activity of TLR agonists in oncological indications have been published in the peer-reviewed scientific literature since the submission of our latest Trial Watch dealing with this topic (May 2013)102 (source http://www.ncbi.nlm.nih.gov/pubmed).

Belani and colleagues investigated the ability of agatolimod (CpG-7909, PF-3512676, Promune®), an unmethylated CpG oligodeoxynucleotide that activates TLR9,103 to improve the therapeutic profile of the FDA-approved epidermal growth factor receptor (EGFR) inhibitor erlotinib104 in patients with advanced recurrent EGFR+ non-small cell lung carcinoma (NSCLC). In this Phase II clinical trial, patients were randomized 1:1 to receive 150 mg erlotinib per day, alone or combined with subcutaneous agatolimod (0.20 mg/kg once weekly). The study was terminated upon the enrollment of 43 patients as an unplanned interim analysis suggested that agatolimod was unlikely to significantly ameliorate the therapeutic activity of erlotinib. Median progression free-survival (PFS) in patients receiving erlotinib alone or erlotinib plus agatolimod was 1.7 and 1.6 mo, respectively (HR, 1.00; 95% CI, 0.5–2.0; P = 0.9335). The incidence of Grade 3–4 toxicities was similar in both study arms, the most adverse events being diarrhea, fatigue, decreased appetite and rash. Thus, agatolimod appears to be unable to improve PFS among EGFR+ NSCLC patients treated with erlotinib.105

Witzig and collaborators tested whether agatolimod would potentiate the therapeutic profile of 111In-ibritumomab tiuxetan and 90Y-ibritumomab tiuxetan (two FDA-approved, radionuclide-conjugated monoclonal antibodies targeting CD20),106,107 in patients with relapsed B-cell non-Hodgkin lymphoma (NHL). Thirty patients affected by relapsing, biopsy-proven CD20+ B-cell NHL and eligible were enrolled. These subjects received 250 mg/m2 rituximab (an FDA-approved, naked monoclonal antibody specific for CD20)108,109 on days 1, 8 and 15; 111In-ibritumomab tiuxetan on days 1 and 8; agatolimod (0.08, 0.16, 0.32 or 0.48 mg/kg) on days 6, 13, 20, 27; and 90Y-ibritumomab tiuxetan on day 15, all as intravenous injections. No dose-limiting toxicity was associated with the administration of agatolimod. Moreover, the authors observed an overall response rate (ORR) of 93% (28/30), including 63% (19/30) complete remissions, a median PFS of 42.7 mo and a median duration of response of 35 mo.110 These encouraging results warrant further investigation in large, randomized clinical studies.

Kruit and coworkers compared the immunostimulatory activity of two distinct preparations that trigger TLR signaling, namely AS02B and AS15, in 75 Stage III or IV M1a melanoma patients vaccinated with full-length melanoma antigen family A3 (MAGEA3). AS02B includes MPL, QS-21 (a water soluble saponin extracted from the South American tree Quillaja saponaria Molina),111 and SB62 (a commercial oil-in-water emulsion), hence activating TLR2 and TLR4.112-114 Conversely, AS15 contains MPL, QS-21 and agatolimod, thus operating as a mixed TLR2/TLR4/TLR9 agonist.11,102 The co-administration of recombinant MAGEA3 with AS02B and AS15 was equally well tolerated. However, the use of AS15 was associated with 4 objective responses, a 6-mo PFS rate of 25%, and a median overall survival (OS) of 33 mo, while that of AS02B resulted in 1 objective response, a 6-mo PFS rate of 14% and an OS of 19.9 mo. All patients developed anti-MAGEA3 antibodies, yet their levels were 3-fold higher in the AS15 arm, corroborating the superior immunostimulatory and clinical activity of this preparation.115,116 Interestingly, the authors also identified a 84-gene signature associated with clinical benefit among AS15-treated and less so AS02B-treated patients. The same signature, including several immunologically relevant genes (e.g., IFNγ-related genes, chemokine-coding genes), turned out to predict the likelihood of resected NSCLC patients to respond to full-length MAGEA3 plus AS15.117

Hartmann et al. conducted a prospective open-label Phase II trial to test the therapeutic profile of Hiltonol™, a particular formulation of polyriboinosinic polyribocytidylic acid (polyI:C, also known as Ampligen™) that includes carboxymethylcellulose and poly-l-lysine as stabilizing agents,118,119 in pediatric patients with newly diagnosed or recurrent brain tumors. In this setting, 47 children affected by variety of brain neoplasms were treated with Hiltonol™ as a standalone therapeutic intervention. The authors observed no dose-limiting toxicities. Moreover, 3 out of 12 patients with progressive high-grade glioma manifested an objective response to treatment, while 2 out of 4 patient affected by progressive low-grade glioma (LGG) experienced disease stabilization for 18–24 mo. These results prompted the authors to initiate a second study focusing on LGG patients. In this follow-up Phase II clinical trial, 5 out of 10 patients responded to treatment, 2 of which exhibiting stable disease for over 18 mo.120 These data indicate that Hiltonol™ may constitute a promising therapeutic option for pediatric LGG patients.

Dhodapkar and colleagues tested the safety, immunogenicity and clinical activity of CDX-1401, a DC-based vaccine targeting the tumor-associated antigen NY-ESO-1,121,122 adjuvanted with Hiltonol™ and resiquimod, an imiquimod-like molecule formerly called R848 that operates as a mixed TLR7/TLR8 agonist.96,123-125 This Phase I clinical trial enrolled a total of 45 patients with advanced malignancies refractory to available therapies. CDX-1401 promoted NY-ESO-1-specific immune responses in all patients bearing NY-ESO-1+ tumors,126 and the treatment was not associated with dose-limiting or Grade 3–4 toxicities. Moreover, 13 patients experienced disease stabilization (median duration: 6.7 mo) and 2 subjects had objective tumor regression (shrinkage of the target lesion of approximately 20%). A similar response was achieved by 6 out of 8 patients who also received immune checkpoint-blocking antibodies127 within 3 mo after vaccination. These results suggest that Hiltonol™ and resiquimod may adequately support the immunogenic potential of vaccines that target DCs in vivo, such as CDX-1401.

Paleja and collaborators investigated the expression levels of various TLRs on the peripheral blood lymphocytes of oral cancer patients, as well as the ability of these cells to respond to various TLR ligands. The TLR expression profile was altered in oral cancer patients as compared with healthy individuals. In particular, unconventional T-cell subsets including γδ T cells and natural killer (NK) T cells were found to express abnormally high levels of several TLRs. Moreover, the peripheral blood lymphocytes of oral cancer patients failed to respond normally to a panel of TLR agonists in terms of (1) TLR signaling, (2) proliferative burst, (3) IFNγ secretion, (4) upregulation of activation markers (e.g., CD25 and CD69), and (5) cytotoxic activity.128 These findings suggest that a deregulation of the TLR system may at least in part contribute to the systemic immunosuppression that often characterizes malignant conditions.

A large body of preclinical literature on the use of TLR agonists as immunostimulatory agents against cancer has been published during the last 13 mo (source http://www.ncbi.nlm.nih.gov/pubmed). Among all these studies, we have found of particular interest the work of (1) Newton and collaborators, who demonstrated that a monoclonal antibody specific for IL-10 receptor α (IL10Rα) consistently improves the immunostimulatory and therapeutic potential of BCG, most likely as it supports BCG-induced TH1 responses;129,130 (2) Perret and colleagues, who reported that CpG oligodeoxynucleotides and Ampligen, but not imiquimod and Quil-A® (a saponin-type adjuvant),131,132 are able to promote the accumulation of effector, rather than regulatory, T cells upon the subcutaneous administration of peptide-based anticancer vaccines, hence stimulating a therapeutically relevant TH1 immune response against established neoplasms;133 (3) Ali and coworkers, who not only identified the combination of granulocyte macrophage colony-stimulating factor (GM-CSF) and CpG oligodeoxynucleotides or Ampligen as the most efficient adjuvant for polymeric anticancer vaccines (in both prophylactic and therapeutic settings), but also elucidated the critical role played by basic leucine zipper transcription factor, ATF-like 3 (BATF3)-dependent DCs in this scenario;134 (4) Yamazaki et al., who found that a chemically defined TLR4 agonist, dendrophilin, can be used to restore the immunogenic potential of tumors lacking the endogenous TLR4 ligand high mobility group box 1 (HMGB1);135 (5) Tai and colleagues, who proved that the perioperative administration of Ampligen™ can limit the loss of NK cell functions that is generally associated with tumor resection, hence exerting a robust anti-metastatic effect;136 and (6) Huang and collaborators, who showed that imiquimod can promote aerobic glycolysis in cancer cells in a TLR7- and TLR8-independent fashion, a protective response centered around the upregulation of hypoxia-inducible factor 1α (HIF1α).137 In this setting, imiquimod and multiple pharmacological interventions that inhibit the glycolytic flux, including 2-deoxyglucose and 17-N-allylamino-17-demethoxygeldanamycin,138 were found to exert synergistic antitumor effects, in vitro and in vivo.137 These data indicate that the therapeutic activity of imiquimod may involve a cancer cell-intrinsic component.

Update on Ongoing Clinical Trials

When this Trial Watch was being prepared (May 2014), official sources listed no less than 14 clinical trials launched after May 1st, 2013 that would assess the immunostimulatory and therapeutic profile of TLR agonists in cancer patients (source http://www.clinicaltrials.gov). Seven of these studies involve FDA-approved molecules, i.e., imiquimod (6 trials) and BCG (1 trial), while the other 7 aim at investigating the safety and efficacy of hitherto experimental TLR ligands, i.e., Hiltonol™ (5 trials), the DNA-based double stem-loop immunomodulator139 MGN1703 (1 trial), and glucopyranosyl lipid adjuvant (GLA), a synthetic TLR4 agonist that – when formulated as a stable emulsion – promotes potent poly-functional TH1 responses upon intradermal administration140-142 (Table 1).

Table 1. Clinical trials recently launched to evaluate the safety and efficacy of TLR agonists in cancer patients.*.

Agent Indication(s) Status Phase Route Notes Ref.
BCG Bladder cancer Recruiting II Intravesical Combined with a CEA- and
MUC1-targeting vaccine
NCT02015104
GLA Merkel cell carcinoma Recruiting I/II Intratumoral As single agent NCT02035657
Imiquimod Actinic keratosis Recruiting IV Topical     As single agent   NCT01926496
Anal intraepithelial neoplasia Not yet recruiting III Anal     As single agent   NCT02059499
Glioblastoma Not yet recruiting I/II Topical Combined with GM-CSF and a synthetic peptide-based vaccine (SL-701)   NCT01957878
Low-grade HPV+ cervical lesions Recruiting II Topical Combined with a HPV-16-
and HPV-18-targeting vaccine
  NCT02078648
NSCLC Recruiting II Topical Combined with GM-CSF, cyclophosphamide and an autophagosome-derived vaccine   NCT01909752
Pediatric brain tumors Recruiting I Topical Combined with DCs loaded ex vivo
with autologous cancer cell lysates
  NCT01902771
Hiltonol™ B-cell lymphoma Recruiting I/II Intratumoral Combined with recombinant human FLT3L   NCT01976585
Cutaneous T-cell lymphoma Recruiting I Subcutaneous Combined with radiation
therapy and romidepsin
  NCT02061449
Glioblastoma Recruiting I/II Intramuscular Combined with multipeptide vaccine, radiation therapy and temozolomide   NCT01920191
Melanoma Recruiting I n.a. Combined with a personalized
peptide-based vaccine
  NCT01970358
Solid tumors Recruiting II Intratumoral
Intramuscular
    As single agent   NCT01984892
MGN1703 Metastatic CRC Not yet recruiting III n.a.     As single agent   NCT02077868

Abbreviations: BCG, bacillus Calmette-Guérin; CEA, carcinoembryonic antigen; CRC, colorectal carcinoma; DC, dendritic cell; GLA, glucopyranosyl lipid adjuvant; GM-CSF, granulocyte macrophage colony-stimulating factor; HPV, human papilloma virus; FLT3L, FLT3 ligand; n.a., not available; MUC1, mucin 1; NSCLC, non-small cell lung carcinoma. *between 2013, May 1st and the date of submission.

In particular, imiquimod is being tested (1) as a standalone topical intervention, in HIV+ patients with high-grade anal squamous skin lesions (NCT02059499); (2) in combination with ProCervix, a vaccine targeting HPV-16 and -18 (source http://www.genticel.com/products/procervix/),143-145 in HPV+ women who have not yet developed high-grade cervical lesions (NCT01957878); (3) together with GM-CSF as adjuvant to a synthetic peptide-based vaccine (SL-701),146 in subjects with recurrent glioblastoma multiforme (NCT02078648); (4) in combination with cyclophosphamide (an immunogenic alkylating agent)147,148 and GM-CSF to support an autophagosome-based vaccine derived from allogeneic cancer cells,149-151 administered to NSCLC patients (NCT01909752); and (5) as adjuvant to DCs loaded ex vivo with autologous cancer cell lysates, in children with high-grade brain tumors (NCT01902771). On a slightly different note, NCT01926496 has recently been initiated to prospectively evaluate the risk of subjects with actinic keratosis treated with Aldara® or ingenol mebutate (an FDA-approved substance commercialized as 0.015% or 0.05% cream under the trade name Picato®),152-154 to develop squamous cell carcinoma (NCT01926496). In addition, BCG is currently being evaluated as a standalone therapeutic intervention or combined with a viral vector encoding mucin 1 (MUC1), carcinoembryonic antigen (CEA) and three immunostimulatory molecules, i.e., CD80 (also known as B7–1), CD58 (also known as LFA-3) and intercellular adhesion molecule 1 (ICAM1),155-159 in adults with high-grade non-invasive bladder carcinoma who failed at least 1 course of BCG (NCT02015104).

The clinical profile of Hiltonol™ is currently being assessed (1) in subjects affected by solid tumors easily accessible by needle, receiving Hiltonol™ as a standalone therapeutic intervention i.t. (NCT01984892); (2) in melanoma patients, who receive Hiltonol™ to improve the immunogenicity of a personalized, neoantigen-targeting peptide-based vaccine (NCT01970358); (3) in individuals with glioblastoma, who are treated with Hiltonol™ as adjuvant to a multipeptide-based vaccine (IMA950)160 given in combination with temozolomide-based radiochemotherapy82,161 (NCT01920191); (4) in cutaneous T-cell lymphoma patients, receiving Hiltonol™ to boost the therapeutic potential of radiation therapy coupled to the histone deacetylase inhibitor romidepsin162-165 (NCT02061449); and (5) in B-cell lymphoma patients, who are concurrently treated with recombinant human FLT3 ligand (FLT3L, also known as CDX-301)166-168 i.t. plus low-dose radiation therapy in the attempt to recruit DCs to neoplastic lesions and thus promote clinically relevant anticancer immune responses (NCT01976585). MGN1703 is under investigation as a standalone agent for the maintenance of metastatic colorectal carcinoma patients experiencing objective responses in the course of induction chemotherapy (NCT02077868). Finally, GLA in stable emulsion is being tested as a standalone therapeutic intervention in biopsy-confirmed Merkel cell carcinoma patients who bear at least one injectable lesion (NCT02035657).

The following clinical studies discussed in our previous Trial Watches dealing with this topic10,11,102 have changed status during the past 13 mo: NCT00003715 and NCT01808950, which are now listed as “Terminated”; NCT01264731, which nowadays appears as “Suspended”; NCT00694551 and NCT00899574, which are listed as “Active, not recruiting” but are associated with preliminary results; as well as NCT01410968, NCT00923910, and NCT01171469, which are reported as “Completed” (source http://www.clinicaltrials.gov).

The reasons for the termination of NCT00003715 and NCT01808950 are not specified, while the temporary suspension of NCT01264731 appears to relate to the study staff being on medical leave. NCT00694551 aimed at evaluating the clinical profile of a peptide-based vaccine adjuvanted with Hiltonol™ in prostate carcinoma patients. At present, safety data referring to n = 29 patients treated with three different doses of the vaccine have been released, demonstrating that this regimen is very well tolerated. Efficacy data on NCT00694551 have not been disclosed yet. NCT00899574 was intended to assess the efficacy of Aldara® in breast cancer patients with chest wall recurrence or skin metastases. No serious adverse effects were documented. Moreover, the authors report an ORR of 20% (95% CI: 3–56) and an amelioration of pain or pruritus in 9 out of 10 patients analyzed. To the best of our knowledge, the results of NCT01410968, NCT00923910, and NCT01171469 have not been released yet (source http://www.clinicaltrials.gov).

Concluding Remarks

In spite of their robust and well-characterized immunostimulatory potential, the attention attracted by currently available TLR agonists for use as therapeutic agents in oncological indications is stably decreasing. One year ago, in the latest Trial Watch dealing with TLR ligands,102 we speculated that such a trend may reflect, at least in part, the limited availability of clinical-grade reagents. Several groups were indeed refocusing their efforts on alternative sources of TLR agonists, including commonly employed prophylactic vaccines.169,170 A few chemically defined TLR agonists have recently been shown to exert therapeutic effects in preclinical tumor models, including dendrophilin,135 yet the clinical interest in these agents appears to remain low. It is therefore tempting to speculate that oncologists may now be prioritizing other, relatively more specific, immunostimulatory interventions, such as immune checkpoint blockers. Several molecules that block immunosuppressive receptors expressed on activated immune cells, including the cytotoxic T lymphocyte-assocaited protein 4 (CTLA4)-specific antibody ipilimumab171-173 and the programmed cell death 1 (PDCD1)-targeting antibody nivolumab,174 have been shown to significantly boost the immunostimulatory potential of TLR agonists.175-177 In this scenario, it will be interesting to see not only whether TLR agonists and checkpoint-blocking antibodies can be combined in a safe and efficient manner in patients, but also whether the latter might one day replace the former as standalone adjuvants to active immunotherapeutic interventions.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipe labelisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

Glossary

Abbreviations:

BCG

bacillus Calmette-Guérin

CI

confidence interval

DAMP

damage-associated molecular pattern

DC

dendritic cell

EGFR

epidermal growth factor receptor

FDA

Food and Drug Administration

GLA

glucopyranosyl lipid adjuvant

GM-CSF

granulocyte macrophage colony-stimulating factor

HR

hazard ratio

HPV

human papillomavirus

IFN

interferon

LGG

low-grade glioma

LPS

lipopolysaccharide

MAGEA3

melanoma antigen family A3

MPL

monophosphoryl lipid A

MYD88

myeloid differentiation primary response 88

NHL

non-Hodgkin’s lymphoma

NK

natural killer

NSCLC

non-small cell lung carcinoma

ORR

overall response rate

OS

overall survival

PFS

progression-free survival

SNP

single nucleotide polymorphism

TICAM1

toll-like receptor adaptor molecule 1

TIRAP

toll-interleukin 1 receptor domain containing adaptor protein

TLR

toll-like receptor

10.4161/onci.29179

References

  • 1.Song WY, Wang GL, Chen LL, Kim HS, Pi LY, Holsten T, Gardner J, Wang B, Zhai WX, Zhu LH, 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]
  • 2.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]
  • 3.Roach JC, Glusman G, Rowen L, Kaur A, Purcell MK, Smith KD, Hood LE, Aderem A. The evolution of vertebrate Toll-like receptors. Proc Natl Acad Sci USA. 2005;102:9577–82. doi: 10.1073/pnas.0502272102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.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]
  • 5.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]
  • 6.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]
  • 7.Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34:637–50. doi: 10.1016/j.immuni.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 8.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]
  • 9.Lauw FN, Caffrey DR, Golenbock DT. Of mice and man: TLR11 (finally) finds profilin. Trends Immunol. 2005;26:509–11. doi: 10.1016/j.it.2005.08.006. [DOI] [PubMed] [Google Scholar]
  • 10.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. OncoImmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. OncoImmunology. 2012;1:894–907. doi: 10.4161/onci.20931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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]
  • 13.Schröder NW, Opitz B, Lamping N, Michelsen KS, Zähringer U, Göbel UB, Schumann RR. 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]
  • 14.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]
  • 15.Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, Sovath S, Shamel L, Hartung T, Zähringer U, et al. CD36 is a sensor of diacylglycerides. Nature. 2005;433:523–7. doi: 10.1038/nature03253. [DOI] [PubMed] [Google Scholar]
  • 16.Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, Eng JK, Akira S, Underhill DM, Aderem A. 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]
  • 17.Smith KD, Andersen-Nissen E, Hayashi F, Strobe K, Bergman MA, Barrett SL, Cookson BT, Aderem A. 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]
  • 18.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] [PubMed] [Google Scholar]
  • 19.Yoon SI, Kurnasov O, Natarajan V, Hong M, Gudkov AV, Osterman AL, Wilson IA. 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]
  • 20.Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000;408:740–5. doi: 10.1038/35047123. [DOI] [PubMed] [Google Scholar]
  • 21.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]
  • 22.Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. 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]
  • 23.Hotz C, Bourquin C. Systemic cancer immunotherapy with Toll-like receptor 7 agonists: Timing is everything. OncoImmunology. 2012;1:227–8. doi: 10.4161/onci.1.2.18169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.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]
  • 25.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]
  • 26.Liu L, Botos I, Wang Y, Leonard JN, Shiloach J, Segal DM, Davies DR. 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]
  • 27.Shi Z, Cai Z, Yu J, Zhang T, Zhao S, Smeds E, Zhang Q, Wang F, Zhao C, Fu S, et al. Toll-like receptor 11 (TLR11) prevents Salmonella penetration into the murine Peyer patches. J Biol Chem. 2012;287:43417–23. doi: 10.1074/jbc.M112.411009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Oldenburg M, Krüger A, Ferstl R, Kaufmann A, Nees G, Sigmund A, Bathke B, Lauterbach H, Suter M, Dreher S, et al. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science. 2012;337:1111–5. doi: 10.1126/science.1220363. [DOI] [PubMed] [Google Scholar]
  • 29.Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–5. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
  • 30.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]
  • 31.Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013;59:583–94. doi: 10.1016/j.jhep.2013.03.033. [DOI] [PubMed] [Google Scholar]
  • 32.Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol. 2012;13:780–8. doi: 10.1038/nrm3479. [DOI] [PubMed] [Google Scholar]
  • 33.Spel L, Boelens JJ, Nierkens S, Boes M. Antitumor immune responses mediated by dendritic cells: How signals derived from dying cancer cells drive antigen cross-presentation. OncoImmunology. 2013;2:e26403. doi: 10.4161/onci.26403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yi Y, Zhou Z, Shu S, Fang Y, Twitty C, Hilton TL, Aung S, Urba WJ, Fox BA, Hu HM, et al. Autophagy-assisted antigen cross-presentation: Autophagosome as the argo of shared tumor-specific antigens and DAMPs. OncoImmunology. 2012;1:976–8. doi: 10.4161/onci.20059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, Stevenson MA, Calderwood SK. 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.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]
  • 38.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]
  • 39.Loser K, Vogl T, Voskort M, Lueken A, Kupas V, Nacken W, Klenner L, Kuhn A, Foell D, Sorokin L, et al. The Toll-like receptor 4 ligands Mrp8 and Mrp14 are crucial in the development of autoreactive CD8+ T cells. Nat Med. 2010;16:713–7. doi: 10.1038/nm.2150. [DOI] [PubMed] [Google Scholar]
  • 40.Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA, Nacken W, Foell D, van der Poll T, Sorg C, et al. Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med. 2007;13:1042–9. doi: 10.1038/nm1638. [DOI] [PubMed] [Google Scholar]
  • 41.Barton GM, Kagan JC. A cell biological view of Toll-like receptor function: regulation through compartmentalization. Nat Rev Immunol. 2009;9:535–42. doi: 10.1038/nri2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Marques PE, Amaral SS, Pires DA, Nogueira LL, Soriani FM, Lima BH, Lopes GA, Russo RC, Avila TV, Melgaço JG, et al. Chemokines and mitochondrial products activate neutrophils to amplify organ injury during mouse acute liver failure. Hepatology. 2012;56:1971–82. doi: 10.1002/hep.25801. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464:104–7. doi: 10.1038/nature08780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Fitzgerald KA, Palsson-McDermott EM, Bowie AG, Jefferies CA, Mansell AS, Brady G, Brint E, Dunne A, Gray P, Harte MT, 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]
  • 45.Medzhitov R, Preston-Hurlburt P, Kopp E, Stadlen A, Chen C, Ghosh S, Janeway CA., Jr. 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]
  • 46.Hoebe K, Du X, Georgel P, Janssen E, Tabeta K, Kim SO, Goode J, Lin P, Mann N, Mudd S, 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]
  • 47.Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, Takeuchi O, Sugiyama M, Okabe M, Takeda K, 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]
  • 48.Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, Shimada N, Ohba Y, Takaoka A, Yoshida N, 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]
  • 49.Yanai H, Negishi H, Taniguchi T. The IRF family of transcription factors: Inception, impact and implications in oncogenesis. OncoImmunology. 2012;1:1376–86. doi: 10.4161/onci.22475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ksienzyk A, Neumann B, Kröger A. IRF-1 is critical for IFNγ mediated immune surveillance. OncoImmunology. 2012;1:533–4. doi: 10.4161/onci.19405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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]
  • 52.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]
  • 53.Zambirinis CP, Miller G. Signaling via MYD88 in the pancreatic tumor microenvironment: A double-edged sword. OncoImmunology. 2013;2:e22567. doi: 10.4161/onci.22567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lee CC, Avalos AM, Ploegh HL. Accessory molecules for Toll-like receptors and their function. Nat Rev Immunol. 2012;12:168–79. doi: 10.1038/nri3151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.O’Neill LA, Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 2007;7:353–64. doi: 10.1038/nri2079. [DOI] [PubMed] [Google Scholar]
  • 56.Seya T, Shime H, Takaki H, Azuma M, Oshiumi H, Matsumoto M. TLR3/TICAM-1 signaling in tumor cell RIP3-dependent necroptosis. OncoImmunology. 2012;1:917–23. doi: 10.4161/onci.21244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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]
  • 58.Jinushi M. The role of innate immune signals in antitumor immunity. OncoImmunology. 2012;1:189–94. doi: 10.4161/onci.1.2.18495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tahara T, Arisawa T, Wang F, Shibata T, Nakamura M, Sakata M, Hirata I, Nakano H. Toll-like receptor 2 -196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci. 2007;98:1790–4. doi: 10.1111/j.1349-7006.2007.00590.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mollaki V, Georgiadis T, Tassidou A, Ioannou M, Daniil Z, Koutsokera A, Papathanassiou AA, Zintzaras E, Vassilopoulos G. Polymorphisms and haplotypes in TLR9 and MYD88 are associated with the development of Hodgkin’s lymphoma: a candidate-gene association study. J Hum Genet. 2009;54:655–9. doi: 10.1038/jhg.2009.90. [DOI] [PubMed] [Google Scholar]
  • 61.Gast A, Bermejo JL, Claus R, Brandt A, Weires M, Weber A, Plass C, Sucker A, Hemminki K, Schadendorf D, et al. Association of inherited variation in Toll-like receptor genes with malignant melanoma susceptibility and survival. PLoS ONE. 2011;6:e24370. doi: 10.1371/journal.pone.0024370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Guirado M, Gil H, Saenz-Lopez P, Reinboth J, Garrido F, Cozar JM, Ruiz-Cabello F, Carretero R. Association between C13ORF31, NOD2, RIPK2 and TLR10 polymorphisms and urothelial bladder cancer. Hum Immunol. 2012;73:668–72. doi: 10.1016/j.humimm.2012.03.006. [DOI] [PubMed] [Google Scholar]
  • 63.Junjie X, Songyao J, Minmin S, Yanyan S, Baiyong S, Xiaxing D, Jiabin J, Xi Z, Hao C. The association between Toll-like receptor 2 single-nucleotide polymorphisms and hepatocellular carcinoma susceptibility. BMC Cancer. 2012;12:57. doi: 10.1186/1471-2407-12-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Carvalho A, Cunha C, Almeida AJ, Osório NS, Saraiva M, Teixeira-Coelho M, Pedreiro S, Torrado E, Domingues N, Gomes-Alves AG, et al. The rs5743836 polymorphism in TLR9 confers a population-based increased risk of non-Hodgkin lymphoma. Genes Immun. 2012;13:197–201. doi: 10.1038/gene.2011.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kim MK, Park SW, Kim SK, Park HJ, Eun YG, Kwon KH, Kim J. Association of Toll-like receptor 2 polymorphisms with papillary thyroid cancer and clinicopathologic features in a Korean population. J Korean Med Sci. 2012;27:1333–8. doi: 10.3346/jkms.2012.27.11.1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yang ZH, Dai Q, Gu YJ, Guo QX, Gong L. Cytokine and chemokine modification by Toll-like receptor polymorphisms is associated with nasopharyngeal carcinoma. Cancer Sci. 2012;103:653–8. doi: 10.1111/j.1349-7006.2012.02210.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Resler AJ, Malone KE, Johnson LG, Malkki M, Petersdorf EW, McKnight B, Madeleine MM. Genetic variation in TLR or NFkappaB pathways and the risk of breast cancer: a case-control study. BMC Cancer. 2013;13:219. doi: 10.1186/1471-2407-13-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Roszak A, Lianeri M, Sowińska A, Jagodziński PP. Involvement of Toll-like Receptor 9 polymorphism in cervical cancer development. Mol Biol Rep. 2012;39:8425–30. doi: 10.1007/s11033-012-1695-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Agúndez JA, García-Martín E, Devesa MJ, Carballo M, Martínez C, Lee-Brunner A, Fernández C, Díaz-Rubio M, Ladero JM. Polymorphism of the TLR4 gene reduces the risk of hepatitis C virus-induced hepatocellular carcinoma. Oncology. 2012;82:35–40. doi: 10.1159/000335606. [DOI] [PubMed] [Google Scholar]
  • 70.Castaño-Rodríguez N, Kaakoush NO, Goh KL, Fock KM, Mitchell HM. The role of TLR2, TLR4 and CD14 genetic polymorphisms in gastric carcinogenesis: a case-control study and meta-analysis. PLoS ONE. 2013;8:e60327. doi: 10.1371/journal.pone.0060327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kim HJ, Bae JS, Chang IH, Kim KD, Lee J, Shin HD, Lee JY, Kim WJ, Kim W, Myung SC. Sequence variants of Toll-like receptor 4 (TLR4) and the risk of prostate cancer in Korean men. World J Urol. 2012;30:225–32. doi: 10.1007/s00345-011-0690-3. [DOI] [PubMed] [Google Scholar]
  • 72.Zhang K, Zhou B, Wang Y, Rao L, Zhang L. The TLR4 gene polymorphisms and susceptibility to cancer: a systematic review and meta-analysis. Eur J Cancer. 2013;49:946–54. doi: 10.1016/j.ejca.2012.09.022. [DOI] [PubMed] [Google Scholar]
  • 73.Nischalke HD, Coenen M, Berger C, Aldenhoff K, Müller T, Berg T, Krämer B, Körner C, Odenthal M, Schulze F, et al. The toll-like receptor 2 (TLR2) -196 to -174 del/ins polymorphism affects viral loads and susceptibility to hepatocellular carcinoma in chronic hepatitis C. Int J Cancer. 2012;130:1470–5. doi: 10.1002/ijc.26143. [DOI] [PubMed] [Google Scholar]
  • 74.Wang X, Li J, Xie W, Zhang W, Chang Y. Toll-like receptor 2 gene polymorphisms and cancer susceptibility: a meta-analysis. Neoplasma. 2013;60:459–67. doi: 10.4149/neo_2013_060. [DOI] [PubMed] [Google Scholar]
  • 75.Theodoropoulos GE, Saridakis V, Karantanos T, Michalopoulos NV, Zagouri F, Kontogianni P, Lymperi M, Gazouli M, Zografos GC. Toll-like receptors gene polymorphisms may confer increased susceptibility to breast cancer development. Breast. 2012;21:534–8. doi: 10.1016/j.breast.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 76.Zou TH, Wang ZH, Fang JY. Positive association between Toll-like receptor 4 gene +896A/G polymorphism and susceptibility to gastric carcinogenesis: a meta-analysis. Tumour Biol. 2013;34:2441–50. doi: 10.1007/s13277-013-0795-y. [DOI] [PubMed] [Google Scholar]
  • 77.Chew V, Tow C, Huang C, Bard-Chapeau E, Copeland NG, Jenkins NA, Weber A, Lim KH, Toh HC, Heikenwalder M, et al. Toll-like receptor 3 expressing tumor parenchyma and infiltrating natural killer cells in hepatocellular carcinoma patients. J Natl Cancer Inst. 2012;104:1796–807. doi: 10.1093/jnci/djs436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hirsch I, Caux C, Hasan U, Bendriss-Vermare N, Olive D. Impaired Toll-like receptor 7 and 9 signaling: from chronic viral infections to cancer. Trends Immunol. 2010;31:391–7. doi: 10.1016/j.it.2010.07.004. [DOI] [PubMed] [Google Scholar]
  • 79.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
  • 80.Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–75. doi: 10.1038/nrc3380. [DOI] [PubMed] [Google Scholar]
  • 81.Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M, Sukkurwala AQ, Menger L, Zitvogel L, Kroemer G. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev. 2011;30:61–9. doi: 10.1007/s10555-011-9273-4. [DOI] [PubMed] [Google Scholar]
  • 82.Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Anticancer radioimmunotherapy. OncoImmunology. 2013;2:e25595. doi: 10.4161/onci.25595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.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]
  • 84.Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39:74–88. doi: 10.1016/j.immuni.2013.06.014. [DOI] [PubMed] [Google Scholar]
  • 85.Demaria S, Vanpouille-Box C, Formenti SC, Adams S. The TLR7 agonist imiquimod as an adjuvant for radiotherapy-elicited in situ vaccination against breast cancer. OncoImmunology. 2013;2:e25997. doi: 10.4161/onci.25997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, Mariette C, Chaput N, Mira JP, Delaloge S, 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]
  • 87.Ahmed A, Redmond HP, Wang JH. Links between Toll-like receptor 4 and breast cancer. OncoImmunology. 2013;2:e22945. doi: 10.4161/onci.22945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tittarelli A, González FE, Pereda C, Mora G, Muñoz L, Saffie C, García T, Díaz D, Falcón C, Hermoso M, et al. Toll-like receptor 4 gene polymorphism influences dendritic cell in vitro function and clinical outcomes in vaccinated melanoma patients. Cancer Immunol Immunother. 2012;61:2067–77. doi: 10.1007/s00262-012-1268-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.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]
  • 90.Heldwein KA, Liang MD, Andresen TK, Thomas KE, Marty AM, Cuesta N, Vogel SN, Fenton MJ. 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]
  • 91.Uehori J, Matsumoto M, Tsuji S, Akazawa T, Takeuchi O, Akira S, Kawata T, Azuma I, Toyoshima K, Seya T. 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]
  • 92.Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow SN, Apter D, Kitchener H, Castellsague X, Teixeira JC, Skinner SR, et al. HPV PATRICIA Study Group Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009;374:301–14. doi: 10.1016/S0140-6736(09)61248-4. [DOI] [PubMed] [Google Scholar]
  • 93.Lehtinen M, Paavonen J. Sound efficacy of prophylactic HPV vaccination: Basics and implications. OncoImmunology. 2012;1:995–6. doi: 10.4161/onci.20011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, Horiuchi T, Tomizawa H, Takeda K, Akira S. 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]
  • 95.Holcmann M, Drobits B, Sibilia M. How imiquimod licenses plasmacytoid dendritic cells to kill tumors. OncoImmunology. 2012;1:1661–3. doi: 10.4161/onci.22033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schön MP, Schön M. TLR7 and TLR8 as targets in cancer therapy. Oncogene. 2008;27:190–9. doi: 10.1038/sj.onc.1210913. [DOI] [PubMed] [Google Scholar]
  • 97.Walter A, Schäfer M, Cecconi V, Matter C, Urosevic-Maiwald M, Belloni B, Schönewolf N, Dummer R, Bloch W, Werner S, et al. Aldara activates TLR7-independent immune defence. Nat Commun. 2013;4:1560. doi: 10.1038/ncomms2566. [DOI] [PubMed] [Google Scholar]
  • 98.Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 2012;13:343–51. doi: 10.1038/ni.2224. [DOI] [PubMed] [Google Scholar]
  • 99.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]
  • 100.Aranda F, Vacchelli E, Obrist F, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. OncoImmunology. 2014;3:e27297. doi: 10.4161/onci.28344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Cremer I, et al. Trial Watch: Oncolytic viruses for cancer therapy. OncoImmunology. 2014;3 doi: 10.4161/onci.28694. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. OncoImmunology. 2013;2:e25238. doi: 10.4161/onci.25238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Murad YM, Clay TM, Lyerly HK, Morse MA. CPG-7909 (PF-3512676, ProMune): toll-like receptor-9 agonist in cancer therapy. Expert Opin Biol Ther. 2007;7:1257–66. doi: 10.1517/14712598.7.8.1257. [DOI] [PubMed] [Google Scholar]
  • 104.Boehrer S, Adès L, Braun T, Galluzzi L, Grosjean J, Fabre C, Le Roux G, Gardin C, Martin A, de Botton S, 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]
  • 105.Belani CP, Nemunaitis JJ, Chachoua A, Eisenberg PD, Raez LE, Cuevas JD, Mather CB, Benner RJ, Meech SJ. Phase 2 trial of erlotinib with or without PF-3512676 (CPG 7909, a Toll-like receptor 9 agonist) in patients with advanced recurrent EGFR-positive non-small cell lung cancer. Cancer Biol Ther. 2013;14:557–63. doi: 10.4161/cbt.24598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. OncoImmunology. 2013;2:e22789. doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. OncoImmunology. 2014;3:e27048. doi: 10.4161/onci.27048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Akhtar S, Maghfoor I. Rituximab plus CHOP for diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:1830-1, author reply 1830-1;; 10.1056/NEJM200206063462317 [DOI] [PubMed]
  • 109.Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:235–42. doi: 10.1056/NEJMoa011795. [DOI] [PubMed] [Google Scholar]
  • 110.Witzig TE, Wiseman GA, Maurer MJ, Habermann TM, Micallef IN, Nowakowski GS, Ansell SM, Colgan JP, Inwards DJ, Porrata LF, et al. A phase I trial of immunostimulatory CpG 7909 oligodeoxynucleotide and 90 yttrium ibritumomab tiuxetan radioimmunotherapy for relapsed B-cell non-Hodgkin lymphoma. Am J Hematol. 2013;88:589–93. doi: 10.1002/ajh.23460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.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]
  • 112.Cluff CW. Monophosphoryl lipid A (MPL) as an adjuvant for anti-cancer vaccines: clinical results. Adv Exp Med Biol. 2010;667:111–23. doi: 10.1007/978-1-4419-1603-7_10. [DOI] [PubMed] [Google Scholar]
  • 113.Vandepapelière P, Horsmans Y, Moris P, Van Mechelen M, Janssens M, Koutsoukos M, Van Belle P, Clement F, Hanon E, Wettendorff M, et al. Vaccine adjuvant systems containing monophosphoryl lipid A and QS21 induce strong and persistent humoral and T cell responses against hepatitis B surface antigen in healthy adult volunteers. Vaccine. 2008;26:1375–86. doi: 10.1016/j.vaccine.2007.12.038. [DOI] [PubMed] [Google Scholar]
  • 114.Vantomme V, Dantinne C, Amrani N, Permanne P, Gheysen D, Bruck C, Stoter G, Britten CM, Keilholz U, Lamers CH, et al. Immunologic analysis of a phase I/II study of vaccination with MAGE-3 protein combined with the AS02B adjuvant in patients with MAGE-3-positive tumors. J Immunother. 2004;27:124–35. doi: 10.1097/00002371-200403000-00006. [DOI] [PubMed] [Google Scholar]
  • 115.Kruit WH, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob JJ, et al. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma. J Clin Oncol. 2013;31:2413–20. doi: 10.1200/JCO.2012.43.7111. [DOI] [PubMed] [Google Scholar]
  • 116.Brichard VG, Godechal Q. MAGE-A3-specific anticancer immunotherapy in the clinical practice. OncoImmunology. 2013;2:e25995. doi: 10.4161/onci.25995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ulloa-Montoya F, Louahed J, Dizier B, Gruselle O, Spiessens B, Lehmann FF, Suciu S, Kruit WH, Eggermont AM, Vansteenkiste J, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol. 2013;31:2388–95. doi: 10.1200/JCO.2012.44.3762. [DOI] [PubMed] [Google Scholar]
  • 118.Levy HB, Baer G, Baron S, Buckler CE, Gibbs CJ, Iadarola MJ, London WT, Rice J. 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]
  • 119.Ming Lim C, Stephenson R, Salazar AM, Ferris RL. TLR3 agonists improve the immunostimulatory potential of cetuximab against EGFR(+) head and neck cancer cells. OncoImmunology. 2013;2:e24677. doi: 10.4161/onci.24677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Hartman LL, Crawford JR, Makale MT, Milburn M, Joshi S, Salazar AM, Hasenauer B, Vandenberg SR, Macdonald TJ, Durden DL. Pediatric Phase II Trials of Poly-ICLC in the Management of Newly Diagnosed and Recurrent Brain Tumors. J Pediatr Hematol Oncol. 2013 doi: 10.1097/MPH.0000000000000047. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. OncoImmunology. 2012;1:1111–34. doi: 10.4161/onci.21494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. OncoImmunology. 2013;2:e25771. doi: 10.4161/onci.25771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Mark KE, Corey L, Meng TC, Magaret AS, Huang ML, Selke S, Slade HB, Tyring SK, Warren T, Sacks SL, 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]
  • 124.Miller RL, Meng TC, Tomai MA. The antiviral activity of Toll-like receptor 7 and 7/8 agonists. Drug News Perspect. 2008;21:69–87. doi: 10.1358/dnp.2008.21.2.1188193. [DOI] [PubMed] [Google Scholar]
  • 125.Meyer T, Surber C, French LE, Stockfleth E. Resiquimod, a topical drug for viral skin lesions and skin cancer. Expert Opin Investig Drugs. 2013;22:149–59. doi: 10.1517/13543784.2013.749236. [DOI] [PubMed] [Google Scholar]
  • 126.Lai JP, Rosenberg AZ, Miettinen MM, Lee CC. NY-ESO-1 expression in sarcomas: A diagnostic marker and immunotherapy target. OncoImmunology. 2012;1:1409–10. doi: 10.4161/onci.21059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. OncoImmunology. 2014;3:e27297. doi: 10.4161/onci.27297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Paleja B, Anand A, Chaukar D, D’Cruz A, Chiplunkar S. Decreased functional response to Toll like receptor ligands in patients with oral cancer. Hum Immunol. 2013;74:927–36. doi: 10.1016/j.humimm.2013.04.018. [DOI] [PubMed] [Google Scholar]
  • 129.Newton MR, Askeland EJ, Andresen ED, Chehval VA, Wang X, Askeland RW, O’Donnell MA, Luo Y. Anti-interleukin-10R1 monoclonal antibody in combination with BCG is protective against bladder cancer metastasis in a murine orthotopic tumor model and demonstrates systemic specific antitumor immunity. Clin Exp Immunol. 2014 doi: 10.1111/cei.12315. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Luo Y. Blocking IL-10 enhances bacillus Calmette-Guérin induced T helper Type 1 immune responses and anti-bladder cancer immunity. OncoImmunology. 2012;1:1183–5. doi: 10.4161/onci.20640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sun HX, Xie Y, Ye YP. Advances in saponin-based adjuvants. Vaccine. 2009;27:1787–96. doi: 10.1016/j.vaccine.2009.01.091. [DOI] [PubMed] [Google Scholar]
  • 132.White K, Rades T, Kearns P, Toth I, Hook S. Immunogenicity of liposomes containing lipid core peptides and the adjuvant Quil A. Pharm Res. 2006;23:1473–81. doi: 10.1007/s11095-006-0272-z. [DOI] [PubMed] [Google Scholar]
  • 133.Perret R, Sierro SR, Botelho NK, Corgnac S, Donda A, Romero P. Adjuvants that improve the ratio of antigen-specific effector to regulatory T cells enhance tumor immunity. Cancer Res. 2013;73:6597–608. doi: 10.1158/0008-5472.CAN-13-0875. [DOI] [PubMed] [Google Scholar]
  • 134.Ali OA, Verbeke C, Johnson C, Sands RW, Lewin SA, White D, Doherty E, Dranoff G, Mooney DJ. Identification of immune factors regulating antitumor immunity using polymeric vaccines with multiple adjuvants. Cancer Res. 2014;74:1670–81. doi: 10.1158/0008-5472.CAN-13-0777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Yamazaki T, Hannani D, Poirier-Colame V, Ladoire S, Locher C, Sistigu A, Prada N, Adjemian S, Catani JP, Freudenberg M, et al. Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ. 2014;21:69–78. doi: 10.1038/cdd.2013.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tai LH, Zhang J, Scott KJ, de Souza CT, Alkayyal AA, Ananth AA, Sahi S, Adair RA, Mahmoud AB, Sad S, et al. Perioperative influenza vaccination reduces postoperative metastatic disease by reversing surgery-induced dysfunction in natural killer cells. Clin Cancer Res. 2013;19:5104–15. doi: 10.1158/1078-0432.CCR-13-0246. [DOI] [PubMed] [Google Scholar]
  • 137.Huang SW, Kao JK, Wu CY, Wang ST, Lee HC, Liang SM, Chen YJ, Shieh JJ. Targeting aerobic glycolysis and HIF-1alpha expression enhance imiquimod-induced apoptosis in cancer cells. Oncotarget 2014; 5:1352-62; [DOI] [PMC free article] [PubMed]
  • 138.Galluzzi L, Kepp O, Vander Heiden MG, Kroemer G. Metabolic targets for cancer therapy. Nat Rev Drug Discov. 2013;12:829–46. doi: 10.1038/nrd4145. [DOI] [PubMed] [Google Scholar]
  • 139.Köchling J, Prada J, Bahrami M, Stripecke R, Seeger K, Henze G, Wittig B, Schmidt M. 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]
  • 140.Fox CB, Moutaftsi M, Vergara J, Desbien AL, Nana GI, Vedvick TS, Coler RN, Reed SG. TLR4 ligand formulation causes distinct effects on antigen-specific cell-mediated and humoral immune responses. Vaccine. 2013;31:5848–55. doi: 10.1016/j.vaccine.2013.09.069. [DOI] [PubMed] [Google Scholar]
  • 141.Schneider LP, Schoonderwoerd AJ, Moutaftsi M, Howard RF, Reed SG, de Jong EC, Teunissen MB. Intradermally administered TLR4 agonist GLA-SE enhances the capacity of human skin DCs to activate T cells and promotes emigration of Langerhans cells. Vaccine. 2012;30:4216–24. doi: 10.1016/j.vaccine.2012.04.051. [DOI] [PubMed] [Google Scholar]
  • 142.Orr MT, Duthie MS, Windish HP, Lucas EA, Guderian JA, Hudson TE, Shaverdian N, O’Donnell J, Desbien AL, Reed SG, et al. MyD88 and TRIF synergistic interaction is required for TH1-cell polarization with a synthetic TLR4 agonist adjuvant. Eur J Immunol. 2013;43:2398–408. doi: 10.1002/eji.201243124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Prior S, Fleck RA, Gillett ML, Rigsby PR, Corbel MJ, Stacey GN, Xing DK. Evaluation of adenyl cyclase toxin constructs from Bordetella pertussis as candidate vaccine components in an in vitro model of complement-dependent intraphagocytic killing. Vaccine. 2006;24:4794–803. doi: 10.1016/j.vaccine.2006.03.065. [DOI] [PubMed] [Google Scholar]
  • 144.Mackova J, Stasikova J, Kutinova L, Masin J, Hainz P, Simsova M, Gabriel P, Sebo P, Nemeckova S. Prime/boost immunotherapy of HPV16-induced tumors with E7 protein delivered by Bordetella adenylate cyclase and modified vaccinia virus Ankara. Cancer Immunol Immunother. 2006;55:39–46. doi: 10.1007/s00262-005-0700-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Fayolle C, Ladant D, Karimova G, Ullmann A, Leclerc C. Therapy of murine tumors with recombinant Bordetella pertussis adenylate cyclase carrying a cytotoxic T cell epitope. J Immunol. 1999;162:4157–62. [PubMed] [Google Scholar]
  • 146.Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL, Brown CK, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with alpha-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–6. doi: 10.1200/JCO.2010.30.7744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ziccheddu G, Proietti E, Moschella F. The Janus face of cyclophosphamide: A sterile inflammatory response that potentiates cancer immunotherapy. OncoImmunology. 2013;2:e25789. doi: 10.4161/onci.25789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Waldron TJ, Quatromoni JG, Karakasheva TA, Singhal S, Rustgi AK. Myeloid derived suppressor cells: Targets for therapy. OncoImmunology. 2013;2:e24117. doi: 10.4161/onci.24117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Su S, Zhou H, Xue M, Liu JY, Ding L, Cao M, Zhou ZX, Hu HM, Wang LX. Anti-tumor efficacy of a hepatocellular carcinoma vaccine based on dendritic cells combined with tumor-derived autophagosomes in murine models. Asian Pac J Cancer Prev. 2013;14:3109–16. doi: 10.7314/APJCP.2013.14.5.3109. [DOI] [PubMed] [Google Scholar]
  • 150.Li Y, Wang LX, Pang P, Cui Z, Aung S, Haley D, Fox BA, Urba WJ, Hu HM. Tumor-derived autophagosome vaccine: mechanism of cross-presentation and therapeutic efficacy. Clin Cancer Res. 2011;17:7047–57. doi: 10.1158/1078-0432.CCR-11-0951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Twitty CG, Jensen SM, Hu HM, Fox BA. Tumor-derived autophagosome vaccine: induction of cross-protective immune responses against short-lived proteins through a p62-dependent mechanism. Clin Cancer Res. 2011;17:6467–81. doi: 10.1158/1078-0432.CCR-11-0812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Aditya S, Gupta S. Ingenol mebutate: A novel topical drug for actinic keratosis. Indian Dermatol Online J 2013; 4:246-9;; 10.4103/2229-5178.115538 [DOI] [PMC free article] [PubMed]
  • 153.Lebwohl M, Shumack S, Stein Gold L, Melgaard A, Larsson T, Tyring SK. Long-term follow-up study of ingenol mebutate gel for the treatment of actinic keratoses. JAMA Dermatol. 2013;149:666–70. doi: 10.1001/jamadermatol.2013.2766. [DOI] [PubMed] [Google Scholar]
  • 154.Lebwohl M, Swanson N, Anderson LL, Melgaard A, Xu Z, Berman B. Ingenol mebutate gel for actinic keratosis. N Engl J Med. 2012;366:1010–9. doi: 10.1056/NEJMoa1111170. [DOI] [PubMed] [Google Scholar]
  • 155.Morse MA, Niedzwiecki D, Marshall JL, Garrett C, Chang DZ, Aklilu M, Crocenzi TS, Cole DJ, Dessureault S, Hobeika AC, et al. A randomized phase II study of immunization with dendritic cells modified with poxvectors encoding CEA and MUC1 compared with the same poxvectors plus GM-CSF for resected metastatic colorectal cancer. Ann Surg. 2013;258:879–86. doi: 10.1097/SLA.0b013e318292919e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Vasir B, Zarwan C, Ahmad R, Crawford KD, Rajabi H, Matsuoka K, Rosenblatt J, Wu Z, Mills H, Kufe D, et al. Induction of antitumor immunity ex vivo using dendritic cells transduced with fowl pox vector expressing MUC1, CEA, and a triad of costimulatory molecules (rF-PANVAC) J Immunother. 2012;35:555–69. doi: 10.1097/CJI.0b013e31826a73de. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Mohebtash M, Tsang KY, Madan RA, Huen NY, Poole DJ, Jochems C, Jones J, Ferrara T, Heery CR, Arlen PM, et al. A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clin Cancer Res. 2011;17:7164–73. doi: 10.1158/1078-0432.CCR-11-0649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Madan RA, Arlen PM, Gulley JL. PANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther. 2007;7:543–54. doi: 10.1517/14712598.7.4.543. [DOI] [PubMed] [Google Scholar]
  • 159.Petrulio CA, Kaufman HL. Development of the PANVAC-VF vaccine for pancreatic cancer. Expert Rev Vaccines. 2006;5:9–19. doi: 10.1586/14760584.5.1.9. [DOI] [PubMed] [Google Scholar]
  • 160.Dutoit V, Herold-Mende C, Hilf N, Schoor O, Beckhove P, Bucher J, Dorsch K, Flohr S, Fritsche J, Lewandrowski P, et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain. 2012;135:1042–54. doi: 10.1093/brain/aws042. [DOI] [PubMed] [Google Scholar]
  • 161.Grossman SA, Ye X, Piantadosi S, Desideri S, Nabors LB, Rosenfeld M, Fisher J, 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]
  • 162.Akilov OE, Grant C, Frye R, Bates S, Piekarz R, Geskin LJ. Low-dose electron beam radiation and romidepsin therapy for symptomatic cutaneous T-cell lymphoma lesions. Br J Dermatol. 2012;167:194–7. doi: 10.1111/j.1365-2133.2012.10905.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Lansigan F, Foss FM. Current and emerging treatment strategies for cutaneous T-cell lymphoma. Drugs. 2010;70:273–86. doi: 10.2165/11532190-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 164.West AC, Smyth MJ, Johnstone RW. The anticancer effects of HDAC inhibitors require the immune system. OncoImmunology. 2014;3:e27414. doi: 10.4161/onci.27414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Adamopoulou E, Naumann U. HDAC inhibitors and their potential applications to glioblastoma therapy. OncoImmunology. 2013;2:e25219. doi: 10.4161/onci.25219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Lim SN, Kuhn S, Hyde E, Ronchese F. Combined TLR stimulation with Pam3Cys and Poly I: C enhances Flt3-ligand dendritic cell activation for tumor immunotherapy. J Immunother. 2012;35:670–9. doi: 10.1097/CJI.0b013e318270e135. [DOI] [PubMed] [Google Scholar]
  • 167.Hennies CM, Reboulet RA, Garcia Z, Nierkens S, Wolkers MC, Janssen EM. Selective expansion of merocytic dendritic cells and CD8DCs confers anti-tumour effect of Fms-like tyrosine kinase 3-ligand treatment in vivo. Clin Exp Immunol. 2011;163:381–91. doi: 10.1111/j.1365-2249.2010.04305.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Curran MA, Allison JP. Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res. 2009;69:7747–55. doi: 10.1158/0008-5472.CAN-08-3289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.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]
  • 170.Schreibelt G, Benitez-Ribas D, Schuurhuis D, Lambeck AJ, van Hout-Kuijer M, Schaft N, Punt CJ, Figdor CG, Adema GJ, de Vries IJ. 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]
  • 171.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Mavilio D, Lugli E. Inhibiting the inhibitors: Checkpoints blockade in solid tumors. OncoImmunology. 2013;2:e26535. doi: 10.4161/onci.26535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Robert C, Thomas L, Bondarenko I, O’Day S. M D JW, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364:2517-26;; 10.1056/NEJMoa1104621 [DOI] [PubMed]
  • 174.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–33. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res. 2013;73:6900–12. doi: 10.1158/0008-5472.CAN-13-1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Williams EL, Dunn SN, James S, Johnson PW, Cragg MS, Glennie MJ, Gray JC. Immunomodulatory monoclonal antibodies combined with peptide vaccination provide potent immunotherapy in an aggressive murine neuroblastoma model. Clin Cancer Res. 2013;19:3545–55. doi: 10.1158/1078-0432.CCR-12-3226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Mangsbo SM, Sandin LC, Anger K, Korman AJ, Loskog A, Tötterman TH. Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with CpG therapy. J Immunother. 2010;33:225–35. doi: 10.1097/CJI.0b013e3181c01fcb. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES