Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2018 Oct 11;7(12):e1526250. doi: 10.1080/2162402X.2018.1526250

Trial Watch: Toll-like receptor agonists in cancer immunotherapy

Melody Smith a, Elena García-Martínez b, Michael R Pitter a, Jitka Fucikova c,d, Radek Spisek c,d, Laurence Zitvogel e,f,g,h, Guido Kroemer i,j,k,l,m,n,o, Lorenzo Galluzzi i,p,q,
PMCID: PMC6279325  PMID: 30524908

ABSTRACT

Toll-like receptor (TLR) agonists demonstrate therapeutic promise as immunological adjuvants for anticancer immunotherapy. To date, three TLR agonists have been approved by US regulatory agencies for use in cancer patients. Additionally, the potential of hitherto experimental TLR ligands to mediate clinically useful immunostimulatory effects has been extensively investigated over the past few years. Here, we summarize recent preclinical and clinical advances in the development of TLR agonists for cancer therapy.

KEYWORDS: Ampligen®, bacillus Calmette-Guérin, Hiltonol®, imiquimod, motolimod, SD-101

Introduction

Toll-like receptors (TLRs) are one of the families of pattern recognition receptors (PRRs) operating in the innate immunity, which also encompass RIG-I-like receptors (RLRs) and NOD-like receptors (NLRs).1,2 PRRs share the ability to recognize relatively conserved microbial components, which are generally referred to as microbe- or pathogen-associated molecular patterns (MAMPs or PAMPs), as well as endogenous danger signals commonly known as damage-associated molecular patterns (DAMPs).311 Common TLR-activating MAMPs include viral and bacterial nucleic acids (which can signal trough TLR3 or TLR9), flagellin (a TLR5 agonist), as well as lipopolysaccharide (LPS), lipoteichoic acid, and mannans (which signal through TLR2 or TLR4).1234 Endogenous nucleic acids and the nuclear non-histone protein high mobility group box 1 (HGMB1) are prototypic TLR-activating DAMPs.8,9,3545

Toll was initially identified and characterized for its antifungal activity in Drosophila melanogaster, and TLRs are evolutionarily conserved from Caenorhabditis elegans through mammals.4655 Thus far, 13 TLRs have been identified in mammals (TLR1-TLR13), 10 of which are encoded in the human genome (TLR1-TLR10).40,56 Notably, human TLR11 is a pseudogene, and human cells lack Tlr12 and Tlr13.40,56,57

TLRs are type I integral membrane glycoproteins characterized by an extracellular domain with a leucine-rich-repeat (LRR) motif and a cytoplasmic signaling domain, which is homologous to the interleukin 1 receptor (IL1R) and is classified as the Toll/IL-1R homology (TIR) domain.58 TLRs either reside in the plasma membrane (TLR1, TLR2, TLR4, TLR5, TLR6 and TLR10) or in endosomal membranes (TLR3, TLR7, TLR8, and TLR9).59 As a family, TLRs are expressed by multiple immune cells, including macrophages, dendritic cells (DCs), B cells, and natural killer (NK) cells, as well as by non-immune cells including epithelial cells, fibroblasts and malignant cells.6062 Thus, besides controlling the activation, maturation and immunological functions of immune cells (notably cytokine secretion),6369 TLR signaling can influence tumor metabolism, proliferation and dissemination.60,62,7075

Several TLR ligands demonstrated potential therapeutic efficacy against malignant disorders.70,76 Imiquimod, also called Aldara® (imiquimod 5% cream as commercialized by 3M Pharmaceuticals) or R-837, is a TLR7 agonist that is approved by the US Food and Drug Administration (FDA) for the treatment of actinic keratosis, external genital/perianal warts (condylomata acuminata), and superficial basal cell carcinoma.7779 The structure of imiquimod is similar to an adenosine nucleoside analogue of the imidazoquinoline family, and it also inhibits Hedgehog signaling by stimulating the adenosine receptor.80 Bacillus Calmette-Guérin (BCG) is an attenuated form of Mycobacterium bovis, which is immunologically related to M. tuberculosis. It was initially used as a vaccine against tuberculosis, but it is now FDA-approved for the treatment of non-invasive transitional cell carcinoma of the bladder.77,81,82 Although the mechanism of action of BCG is not entirely understood, TLR2, TLR4, and TLR9 have all been implicated in the host response to this TLR agonist.83 Finally, monophosphoryl lipid A (MPL) is a derivative of Salmonella minnesota LPS that is employed as an adjuvant in Cervarix®,84,85 a prophylactic vaccine against human papillomavirus (HPV) type 16 and 18, which are strongly associated with cervical carcinoma.86 In this setting, MPL, which mostly signals through TLR2 and TLR4, forms part of so-called “Adjuvant System 04” (AS04), together with aluminum salt.14,87

Here, we discuss recent preclinical and clinical progress on the development of TLR agonists for cancer therapy.

Update on the development of TLR agonists for cancer therapy

Completed clinical studies

Since the publication of the latest Trial Watch dealing with this topic (September 2015),88, various clinical trials investigating the safety and therapeutic profile of TLR agonists in cancer patients have been completed. Only three of these studies, however, reported results to the National Library of Medicine accessible at https://clinicaltrials.gov/ or https://www.ncbi.nlm.nih.gov/pubmed/. The remaining studies have been presented during national or international meetings in the form of oral or written abstracts.

Levy and colleagues (Stanford University, Stanford, CA, USA) in collaboration with the National Cancer Institute (NCI) investigated the side effects and the dose-limiting toxicity of ipilimumab, an anti-CTLA-4 monoclonal antibody, in combination with the TLR9 agonist SD-10189 and radiation therapy90 in patients with recurrent low-grade B-cell lymphoma (NCT02254772). These results were first presented as a poster at the American Society of Clinical Oncology (ASCO) in 201591 and have been featured in several peer-reviewed journal articles.92,93 In this Phase II study, nine participants received intratumoral injections of 10 mg ipilimumab on day 2 of week 1 and 1 mg/week of SD-101 for up to 5 weeks. On days 1 and 2 of the study, participants underwent local radiation therapy. The safety and tolerability of the treatment was assessed over the course of 10 weeks. Tumor response to the treatment and lesion growth were evaluated over the course of 2 years. Of the nine participants included in the study, one experienced at least one serious adverse event (AE). All participants experienced other Grade 3 AEs including fatigue, fever, gastrointestinal disorders or chills. Of the nine participants, seven completed the study. Six out of the seven patients (85.7%) had progressive disease (PD), developing new lesions or significant increases to existing lesion sites. Only one participant had stable disease (SD). The results from this study suggest that combining the intratumoral administration of ipilimumab with SD-101 and radiation at these dose levels does not constitute a promising therapeutic option.

Novartis Pharmaceuticals (Arlington Heights, Illinois, USA) tested the safety, tolerability and efficacy of the investigational drug, LFX453, against placebo in treating the pre-cancerous HPV-induced external genital warts (EGWs) in circumcised men in parallel with an additional open label arm using Aldara®94,95 (NCT02482428). In this Phase II study, 88 participants were randomized and separated across five treatment interventions. Of the two experimental arms, one received LFX453 0.1% nanomedicinal cream (NMC) and the other LFX453 0.15% liquid crystal cream (LCC). The placebo comparator arms received vehicle. Across these groups, treatment was applied twice daily for up to 12 weeks. Aldara® was applied 3 times per week for a maximum of 16 weeks to participants in the final active comparator arm. The treatment efficacy in clearing warts was assessed for up to 14 weeks and the safety and tolerability were assessed for up to 30 weeks. Participants were also evaluated as to whether they had a clearance rate of at least 75% reduction in counts of EGWs by the end of treatment (EOT) at weeks 12 or 16. Between the groups that received NMC (n = 24), LCC (n = 22), combined vehicle (n = 20) and Aldara® (n = 22), only one individual (4.16%) from the NMC group had complete clearance of disease at week 14. By week 30, 3 participants from the NMC group (12.5%), 5 from the LLC group (22.7%), 3 from the vehicle to NMC group (30%), 2 from the vehicle to LCC group (20%) and the 10 participants given Aldara® (45.5%) experienced AEs only. Two subjects from the NMC group (8.3%), 1 from the LCC group (4.5%), none from the combined vehicle group and 3 from the Aldara® group (13.6%) had a partial clearance rate of at least 75% reduction in EGWs. These results support the potential efficacy LFX453 NMC after further testing and refinement.88

Griffiths et al. (Roswell Park Cancer Institute, Buffalo, NY, USA) investigated the safety and therapeutic efficacy of treating myelodysplastic syndrome or acute myeloid leukemia with a DEC-205/NY-ESO-1 fusion protein (CDX-1401)96,97 with an adjuvant, Hiltonol®, a TLR3 agonist,98,99 and combined with standard decitabine-based chemotherapy100 (NCT01834248). In this Phase I study, organized into 4 cycles of chemotherapy and 5 vaccinations, 9 participants receiving 20 mg/m2/day decitabine i.v. per 5 days were treated with s.c. and i.d. injections of CDX-1401 along with Hiltonol® s.c. on days 14 and 15 of cycle 1 and day 15 on cycles 2–4. Treatment was repeated every 28 days for a total of 4 cycles as long as there was no disease progression or unacceptable toxicity. Any incidence of toxicity over the course of a 30-day period following the last dose of the study treatment was assessed according to the NCI Common Terminology Criteria for Adverse Events. Immune and molecular responses were monitored for up to 16 weeks. Patients were followed at days 30, 60, 90 and 180 to evaluate their response to the treatment. The most frequent AEs, which were attributed to chemotherapy or the underlying hematological malignancy, included cytopenia (Grade 3/4), elevated liver enzymes (Grade 3), fatigue (Grade 2), edema (Grade 2/3) and diarrhea (Grade 1/2). Patients also developed localized skin reactions to the vaccine. Only 7 participants completed the study, 2 of whom experienced SAEs. In 6 out of 7 (85.7%) and in 4 out of 7 (57.1%) patients, NY-ESO-1-specific CD4+ and CD8+ T-lymphocytes, respectively, could be documented. NY-ESO-1-expressing myeloid cells isolated from patients at different time points during chemotherapy were able to activate a cytotoxic response from autologous NY-ESO-1-specific lymphocytes. These data indicate that Hiltonol®-adjuvanted vaccinations against NY-ESO-1 can drive an antigen-specific immune response, highlighting the therapeutic potential of antigen-specific immunotherapies combined with potent TLR agonists.101

The results of a large portion of the clinical studies completed between 2016 and 2018 have not been publically posted on https://clinicaltrials.gov/ and have not been published in the peer-reviewed literature. Most of these studies, however, have been presented at annual meetings, and only abstracts or preliminary results are publically available. Although momentarily this precludes obtaining robust insights into the progress and versatility of TLR agonists for cancer therapy, an introduction to the aims and designs of these studies enables understanding of the current and future directions of these immunotherapeutic agents.

GlaxoSmithKline (Berlin, Germany) investigated the safety, tolerability, pharmacokinetic and pharmacodynamic profile of GSK1795091, a TLR4 agonist,102,103 in 42 healthy subjects (NCT02798978). This three-armed randomized, double-blinded, placebo-controlled Phase I study was split into two parts. Part 1 assessed the safety of ascending single doses (starting at 7 ng) of GSK1795091 i.v., while Part 2 was a parallel evaluation of two cohorts that received the drug as in Part 1 but at different time points during the trial. In Part 1, participants were given either the drug or placebo on day 1 administered as an i.v. bolus for 2 to 5 min, followed by an i.v. bolus of 10 mL saline. In Part 2, cohort 1 received an i.v. injection of GSK1795091 on day 1 and a dose on day 8, one week later. Cohort 2 received an i.v. injection of the drug on day 1 and a second dose on day 15. The measures for determining the safety of the drug include the number of AEs, oral body temperature, blood pressure and respiratory rate assessed for up to 11 weeks. In assessing the pharmacokinetics and pharmacodynamics of GSK1795091, measures included the maximum observed drug concentration (Cmax), the time occurrence of Cmax, the terminal half-life, immune cell and plasma cytokine phenotype, the white blood cell count and the clearance of GSK1795091 in Parts 1 and 2 for up to 144 hours. These results will determine the design of future clinical trials in which GSK1795091 will be administered together with immunomodulators in patients with cancer.

Fox et al. (LSU Stanley S. Scott Cancer Center, New Orleans, LA, USA) in collaboration with the NCI and Mayo Clinic hypothesized that a DRibble-based vaccine could induce an immune response against tumor-associated antigens (TAAs) in patients afflicted with Stage III non-small cell lung carcinoma (NSCLC) and that cyclophosphamide plus a HPV vaccine combined with the Dribble-based vaccine alone, with the Dribble-based vaccine plus Aldara® or with the Dribble-based vaccine plus colony stimulating factor 2 (CSF2; best known as GM-CSF) could have therapeutic effects (NCT01909752). DRibbles are tumor cell-derived defective ribosomal products and short-lived proteins packed within autophagosomes.104108 To each of the three arms of this randomized Phase II study, cyclophosphamide 300 mg/m2 was administered as a single dose 3 days prior to vaccination along with a 0.5 mL i.m. injection of the HPV vaccine at the time of the first and third Dribble-based vaccinations. In the first arm of 12 participants, the Dribble-based vaccine was administered every three weeks for 43 weeks. The second arm received the same vaccine and, in addition, applied one 250 mg packet of Aldara (containing 12.5 mg of imiquimod) to a 4 × 5 cm outlined area of skin (including the vaccine site) daily starting at week 4 (with the second Dribble-based vaccine) and for four days following each vaccine cycle. The third arm received the Dribble-based vaccine and was administered GM-CSF 50 μg/day via a CADD-MSTM 3 Ambulatory infusion pump starting at week 4. The goals of the study were to determine which treatment combination elicited the greatest antibody response in a 95-day time frame, and to evaluate safety within a 43-week period, progression-free survival (PFS), and the correlation between PFS and immune responses within a 2-year period. Preliminary results showed that compared to vaccination alone or vaccination plus GM-CSF, vaccination plus imiquimod significantly increased the number of antibody responses. This study was presented at the 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC) 2016.109

In collaboration with Oncovir, Britten and colleagues (Medical University of South Carolina, Charleston, SC, USA) tested the safety and tolerability of Hiltonol® in combination with a DC-based vaccine to patients with locally advanced unresectable pancreatic ductal adenocarcinoma (NCT01677962). Alongside, investigators aimed at elucidating whether combining DCs with Hiltonol® could serve as a therapeutic vaccine against the disease. In this Phase I study, 12 participants received i.t. injections on day 0 and day 14, followed by standard of care procedures for the remainder of the study. AEs were monitored until day 56 (the last day of treatment).110,111

Peereboom and collaborators (Cleveland Clinic, Cleveland, OH, USA) investigated the therapeutic profile of SL-701, a peptide-based vaccine,112 adjuvanted with imiquimod plus GM-CSF (Stage 1), or with Hiltonol® and bevacizumab, an FDA-approved monoclonal antibody specific for vascular endothelial growth factor A (VEGFA)113,114 (Stage 2) in patients with relapsed/refractory HLA-A2+ glioblastoma (GBM) (NCT02078648). Primary objectives were safety, tolerability, investigator assessed objective response rate (ORR), and 12-month OS rate. Alongside, SL-701-specific CD8+ T-cell frequency was monitored by flow cytometry. At reporting, 74 patients were treated, most frequent AEs related to treatment being fatigue (22%) and injection site reaction (18%). Amongst 46 patients enrolled in Stage 1, 1 partial response (PR) and 15 instances of SD were documented. Amongst 28 patients, enrolled in Stage 2, 2 CRs, 4 PRs and 19 instances of SDs were seen. OS at 12 months was 43% in Stage 2 and 37% in Stage 1. These data suggest that adjuvanted SL-701, alone or combined with bevacizumab is well tolerated and mediates clinically relevant antitumor activity.115

Additional studies completed between 2016 and 2018 include NCT01957878, NCT02266147 and NCT02404389. To the best of our knowledge, the results of these studies have not yet been communicated to the public.

Preclinical and translational advances

A considerable body of preclinical and translational findings on the use of TLR agonists for cancer therapy has been disseminated since the publication of the latest Trial Watch dealing with this topic88 (source https://www.ncbi.nlm.nih.gov/pubmed/). Among these studies, we found of particular interest the work of: (1) Molgora and colleagues (Humanitas Clinical and Research Center, Rozzano, Italy), who demonstrated that interleukin-1 receptor 8 (IL1R8, also known as TIR8 or SIGIRR), which is known to impair the signal transduction cascades elicited by several TLRs as well as by various interleukin receptors,116,117 negatively regulates the activity of NK cells as an immunosuppressive checkpoint;118 (2) Lou and co-authors (Institute for Molecular Bioscience. The University of Queensland, Brisbane, Australia), who showed that SLP adaptor and CSK interacting membrane protein (SCIMP), a transmembrane adaptor protein, facilitates TLR4 signaling upon direct binding to its TIR domain;119 (3) Acharya and collaborators (Benaroya Research Institute, Seattle, Washington, USA), who reported a new regulatory circuitry of B cell activation that involves the integrin-dependent recruitment of microtubule-associated proteins 1 light chain 3 (MAP1LC3, best known as LC3)120 to TLR-containing endosomes;121 (4) Combes et al. (Aix Marseille Université, Marseille, France), who reported that lysosomal associated membrane protein family member 5 (LAMP5, also known as BAD-LAMP) negatively regulates TLR9-driven type I interferon (IFN)122 production in the tumor microenvironment by plasmacytoid DCs (pDC);123 (5) New and colleagues (University of Oxford, Oxford, United Kingdom), who demonstrated that the sensitivity of hematologic cancers to histone deacetylase (HDAC) inhibitors is influenced by myeloid differentiation 88 (MYD88), a transducer of TLR signaling;124127 (6) Zhang and collaborators (The University of Tokyo, Tokyo, Japan), who harnessed crystallography to identify two different ligand-binding sites that regulate TLR7;128 (7) Okazaki et al. (USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA), who suggested that the TLR7 rs3853839 G/G variant may constitute a positive prognostic factor as it is associated with a PFS benefit for patients with metastatic colorectal carcinoma (CRC) treated with cetuximab-based chemotherapy;128 (8) Takeda et al. (Hokkaido University Graduate School of Medicine, Sapporo, Japan), who demonstrated that the combination of a TLR3 agonist (ARNAX),129,130 a PD-L1-specific immune checkpoint blocker and a TAA-derived vaccine may be used to overcome resistance to PD-1-targeting therapies, at least in mice;131 (9) Caronni and colleagues (International Centre for Genetic Engineering and Biotechnology, Trieste, Italy), who found that lactic acid blocks the ability of DCs to produce type I IFN in response to TLR3 and STING agonists132 in a mouse model of lung cancer;133 confirming previous reports on the major influence of local metabolism on the immune functions of the tumor microenvironment;134137 (10) Dooduijn and collaborators (Leiden University Medical Center Leiden, The Netherlands), who showed that TLR7/TLR8 agonism, compared to TLR3 and TLR9 agonism, drives an NK cell-dependent immune response that can eradicate tumors that have escaped immunosurveillance following MHC Class I downregulation,138 which is in line with data from other groups demonstrating the ability of TLR7 ligands to trigger NK cell-dependent tumor control;75,139142 (11) Klein and co-authors (University Hospital Essen, Essen, Germany), who showed that mice lacking TLR3, TLR7 and TLR9 are able to reject syngeneic wild-type malignant cells upon the activation of a tumor-targeting immune response involving both CD4+ and CD8+ T lymphocytes, suggesting that endosomal TLRs may operate as part of immunological checkpoints, at least in some settings;143 (12) Rashedi et al. (University of Toronto, Toronto, Canada), who demonstrated that mesenchymal stromal cells recruit TREG cells144,145 upon TLR3 or TLR4 activation, as a consequence of Notch signaling modulation146 and Delta-like 1 (DL1) upregulation.147 (13) Hotz et al. (University of Fribourg, Fribourg, Switzerland), who demonstrated that the sequential administration of polyriboinosinic polyribocytidylic acid (poly I:C, a TLR3 agonist)148 and R848 (a TLR7 agonist)149 24 hours apart activates both MYD88-dependent and -independent pathways that culminate with a DC-driven, NK cell, and CTL-dependent anticancer immune response;75 (14) Le Noci and collaborators (Università degli Studi di Milano, Milan, Italy), who reported that combining a nebulized anti-MDSC antibody (RB6-8C5)150,151 with aerosolized CpG oligodeoxynucleotides (which operate as TLR9 agonists)152,153 and polyI:C results in the downregulation of multiple immunosuppressive molecules, including interleukin 10 (IL10)154,155, ultimately resulting in the activation of tumor-infiltrating NK cells that mediate robust therapeutic responses against lung metastases from syngeneic melanoma;156158 (15) Müller and colleagues (University of Oslo, Oslo, Norway), who found that interferon gamma (IFNG, best known as IFN-γ) synergizes with various TLR agonists to trigger nitric oxide and pro-inflammatory cytokine production by M1 macrophages, coupled with robust anticancer activity;159 (16) Camargo and co-workers (University of Campinas, São Paulo, Brazil), who showed that BCG and imiquimod suppress chemical bladder tumorigenesis, while decreasing markers of proliferation (Ki67)160 and increasing markers of cell death (TUNEL)45,161,162 and autophagy;163165 as well as (17) Kim et al. (University of Minnesota, Minneapolis, MN, USA), who found enhanced tumor-targeting CTL activity following administration of nanoparticles containing mixed TLR7/TLR8 agonists.166

This large amount of preclinical and translation literature corroborates the notion that TLR agonism is a promising strategy for the development of combinatorial anticancer regimens based on the reactivation of immunosurveillance.

Recently initiated clinical trials

Since the submission of the latest Trial Watch dealing with this topic (September 2015),88 no less than 66 clinical studies involving the administration of TLR agonists to cancer patients have been initiated (source http://clinicaltrials.gov/). The majority of these trials involve the FDA-approved molecules imiquimod (17 studies) and BCG (11 studies), as well as the hitherto experimental TLR3 agonist Hiltonol®, a particular formulation of polyI:C that includes carboxymethylcellulose and poly-L-lysine as stabilizing agents,156,167,168 (20 studies), and the TLR9 agonist SD-101 (9 studies) (Table 1).

Table 1.

Clinical trials recently started to investigate the therapeutic profile of TLR agonists in cancer patients.*.

Molecule Indication(s) Phase Status Route Notes Ref.
Ampligen™ Colorectal carcinoma I Recruiting Intravenous In combination with celecoxib and recombinant interferon-α2b for patients with metastatic disease to the liver NCT03403634
II Withdrawn Intravenous In combination with DC-based vaccination, interferon-α2b and celecoxib NCT02615574
BCG Bladder carcinoma I Recruiting Intravesical In combination with rapamycin NCT02753309
Recruiting Intravesical In combination with pembrolizumab NCT02808143
Recruiting Intravesical Following re-resection as compared to BCG with no re-resection NCT03266900
I/II Recruiting Intravesical In combination with durvalumab NCT03317158
I/II Recruiting Intravesical Atezolizumab alone or in combination with BCG NCT02792192
I/II Recruiting Intravesical In combination with ALT803, an IL-15 superagonist NCT02138734
II Recruiting Intravesical In combination with ALT803 NCT03022825
II Recruiting Intravesical Nivolumab or nivolumab/BMS-986205 alone or in combination with BCG NCT03519256
III Recruiting Intravesical Intradermal In different strains NCT03091660
Urological tumors II Not yet Recruiting Intravesical In combination with pembrolizumab NCT03345134
Melanoma II Not yet Recruiting Intravesical In combination with cyclophosphamide, IL-2 and a melanoma-specific vaccine NCT00477906
DUK-CPG-001 Hematological neoplasms II Withdrawn Intravenous In combination with a DC vaccine NCT02115126
G100 Solid tumors I Active, not Recruiting Intratumoral In combination with a NY-ESO-1-targeting vaccine NCT02387125
Follicular lymphoma I/II Active, not Recruiting Intratumoral Alone or combined with pembrolizumab NCT02501473
Hiltonol™ Colorectal carcinoma I/II Recruiting Intramuscular In combination with pembrolizumab NCT02834052
Breast carcinoma I Recruiting Intramuscular In combination with a peptide vaccine and durvalumab NCT02826434
Recruiting n.a. In combination with a peptide vaccine and pembrolizumab NCT03362060
Gynecological tumors I Recruiting Intramuscular In combination with a CA 125 monoclonal antibody NCT03162562
I/IIb Recruiting Subcutaneous In combination with a DC-based vaccine, guadecitabine and atezolizumab NCT03206047
Lung cancer I Recruiting Subcutaneous In combination with a MUC1-vaccine NCT03300817
Recruiting Subcutaneous In combination with NEO-PV-01, pembrolizumab and chemotherapy NCT03380871
Solid tumors I Recruiting n.a. In combination with a personalized vaccine NCT02721043
Active, not recruiting n.a. In combination with bevacizumab and a peptide vaccine NCT02754362
Recruiting Subcutaneous In combination with NEO-PV-01 nivolumab NCT02897765
Not yet recruiting Subcutaneous In conjunction with a combination of the following agents: NEO-PV-01, APX005M, ipilimumab, and nivolumab NCT03597282
I/II Recruiting Intratumoral Intramuscular In combination with durvalumab ± tremelimumab NCT02643303
Recruiting Intramuscular In combination with anti-MUC1 and anti-influenza vaccines plus tadalafil NCT02544880
Withdrawn Subcutaneous In combination with CDX-1401 and pembrolizumab NCT02661100
II Recruiting Intramuscular In combination with a peptide vaccine NCT02873819
Multiple myeloma I Active, not recruiting n.a. In combination with a peptide vaccine and durvalumab ± lenalidomide NCT02886065
Acute myeloid leukemia I Recruiting Subcutaneous In combination with DEC-205/NY-ESO-1 fusion protein CDX-1401, decitabine, and nivolumab NCT03358719
Glioma I Recruiting Subcutaneous In combination with a cancer cell lysate vaccine before and after or only after surgery NCT02549833
Recruiting Subcutaneous In combination with a peptide vaccine ± varlilumab NCT02924038
Recruiting n.a. In combination with a peptide vaccine NCT02960230
Prostate carcinoma I Recruiting Intramuscular Followed by radical prostatectomy NCT03262103
Imiquimod Basal cell carcinoma II Not yet recruiting Topical In combination with neoadjuvant sonidegib followed by surgery or imiquimod NCT03534947
III Active, not recruiting Topical Combined with curettage surgery as compared to surgery alone NCT02242929
Squamous cell carcinoma I Not yet recruiting Topical In combination with 5-fluorouracil NCT03370406
Melanoma I Recruiting Topical In combination with pembrolizumab NCT03276832
Anal carcinoma III Recruiting Topical Compared to 5-fluorouracil or observation NCT02059499
Cervical intraepithelial lesions n.a Recruiting Topical Alone or upon tumor resection NCT02917746
n.a Recruiting Topical In combination with a DNA-based vaccine NCT03206138
I Not yet recruiting Topical In combination with 5-fluorouracil NCT03196180
II Recruiting Topical In combination with tumor resection NCT03233412
Recruiting Topical Alone or in combination with HPV vaccination NCT02864147
III Recruiting Topical Alone as compared to resection NCT02669459
Genital warts II Recruiting Topical In combination with a DNA-based vaccine NCT03180684
Cutaneous T cell lymphoma I Recruiting Topical In combination with doxycycline NCT03116659
Chronic lymphocytic lymphoma II Recruiting Topical In combination with a peptide-based vaccine and lenalidomide NCT02802943
NSCLC I Not yet recruiting Topical In combination with a DRibble-based vaccine, DC/CIK cells and GM-CSF NCT03057340
Anal HPV lesions III Not yet recruiting Topical Adjuvant therapy for anal HPV lesions NCT03289260
Prostate carcinoma I Recruiting Topical Combined with a peptide-based
vaccination and chemotherapy
NCT02234921
Motolimod Solid tumors Ib Unknown Subcutaneous In combination with cetuximab ± nivolumab NCT02124850
Ib Active, not recruiting Subcutaneous In combination with cyclophosphamide NCT02650635
Ib Active, not Recruiting Subcutaneous In combination with cyclophosphamide NCT02650635
SD-101 Lymphoma I Not yet recruiting Intratumoral In combination with an anti-OX40 antibody, BMS-986178, and RT NCT03410901
I Terminated Intratumoral In combination with RT after allogeneic hematopoietic cell transplantation NCT01745354
Advanced malignancies I/Ib Terminated Intratumoral In combination with an anti-IL10 agent NCT02731742
Solid tumors Ib/II Recruiting Intratumoral In combination with pembrolizumab NCT02521870
Follicular lymphoma Ib/II Recruiting Intratumoral In combination with ibrutinib and RT NCT02927964
Advanced solid tumors & lymphoma I/II Recruiting Intratumoral In combination with epacadostat and RT NCT03322384
lymphoma I/II Completed Intratumoral In combination with ipilimumab and RT NCT02254772
I/II Completed Intratumoral In combination with RT NCT02266147
Prostate carcinoma II Suspended Intratumoral In combination with pembrolizumab and RT NCT03007732

In particular, imiquimod is being tested (1) in combination with neoadjuvant sonidegib followed by surgery or imiquimod for the treatment of basal cell carcinoma (NCT03534947); (2) in combination with curettage surgery as compared to surgery alone in patients with basal cell carcinoma (NCT02242929); (3) in combination with 5-fluorouracil for the treatment of squamous cell carcinoma (NCT03370406); (4) in combination with pembrolizumab, an FDA-approved immune checkpoint blocker specific for PD-1,169 in patients with melanoma157 (NCT03276832); (5) compared to 5-flurouracil-based chemotherapy or observation for the treatment of anal carcinoma (NCT02059499); (6) as a standalone immunotherapeutic agent or following large loop excision of the transformation zone (LLETZ) in patients with cervical intraepithelial neoplasms (CINs) (NCT02917746); (7) in combination with a DNA-based vaccine, GX-188E170 (NCT03206138); (8) in combination with 5-flurouracil for the treatment of patients with high-grade cervical intraepithelial neoplasia (NCT03196180); (9) in combination with conization of the uterine cervix based on loop electrosurgical excision procedure (LEEP) as compared to LEEP alone for patients with CIN (NCT03233412); (10) as standalone therapy or in combination with a nonavalent HPV-specific vaccine for patients with CINs (NCT02864147); (11) as a single agent compared to LLETZ for patients with CINs (NCT02669459); (12) in combination with a DNA vaccine, VGX-3100,171174 for patients with HPV-16 and/or HPV-18-related high grade squamous intraepithelial lesion (HSIL) of the vulva (NCT03180684); (13) in combination with doxycycline for the treatment of cutaneous T cell lymphoma (NCT03116659); (14) in combination with a peptide vaccine, iVAC-L-CLL01,175,176 and the immunomodulatory agent lenalidomide177-180 in patients with chronic lymphocytic lymphoma (NCT02802943); (15) combined with a DRibble-based vaccine181 and DC-activated cytokine-induced killer (DC/CIK) cells and GM-CSF in NSCLC patients (NCT03057340); (16) as adjuvant therapy for patients with anal HPV lesions (NCT03289260); and (17) in combination with DPV-001, another DRibble-based vaccine,107,182184 in patients with advanced prostate carcinoma (NCT02234921).

BCG is being investigated in clinical settings: (1) in combination with rapamycin185 for bladder carcinoma (NCT02753309); (2) in combination with pembrolizumab186,187 for the treatment of bladder carcinoma (NCT02808143); (3) following re-resection as compared to BCG with no re-resection188 for the treatment of bladder carcinoma (NCT03266900); (4) in combination with durvalumab189-191 for the treatment of bladder carcinoma (NCT03317158); (5) in combination with atezolizumab192-194 for the treatment of bladder carcinoma (NCT02792192); (6) in combination with ALT-803, an IL-15 superagonist195 for the treatment of bladder carcinoma (NCT02138734); (7) in a multicenter trial of BCG in combination with ALT-803195,196 for treatment of BCG-unresponsive, high-risk, non-muscle invasive bladder carcinoma (NCT03022825); (8) as nivolumab or nivolumab/BMS-986205 (an investigational IDO1 inhibitor) alone or in combination with BCG197-199 for the treatment of BCG-unresponsive, high-risk, non-muscle invasive bladder carcinoma (NCT03519256); (9) different strains of BCG200,201 for the treatment of bladder carcinoma (NCT03091660); (10) in combination with pembrolziumab202,203 for the treatment of urological tumors (NCT03345134); and (11) in combination with cyclophosphamide, IL-2, and a melanoma-specific vaccine204 for the treatment of melanoma171 (NCT00477906).

Additionally, Hiltonol® is being evaluated: (1) in combination with pembrolizumab in patients with CRC (NCT02834052); (2) in combination with PVX-410, a peptide vaccine,168,205,206 and pembrolizumab or durvalumab, a PD-L1-targeting immune checkpoint blocker,207 in patients with breast carcinoma (NCT02826434; NCT03362060); (3) in combination with oregovomab, a CA-125-targeting monoclonal antibody,208 for the treatment of recurrent, advanced ovarian carcinoma (NCT03162562); (4) in combination with CDX-1401, guadecitabine and atezolizumab, another PD-L1-targeting immune checkpoint blocker,209211 in patients with recurrent ovarian, fallopian tube, or primary peritoneal cancer (NCT03206047); (5) combined with a peptide vaccine specific for mucin 1 (MUC1) for the prevention of lung cancer in current and former smokers who are at high risk (NCT03300817); (6) in combination with a neoantigen vaccine, NEO-PV-01,131,212 pembrolizumab and chemotherapy for patients with lung cancer (NCT03380871); (7) in combination with PGV001, a peptide vaccine,213 for the treatment of non-hematologic malignancies in the adjuvant setting (NCT02721043); (8) in combination with bevacizumab and a peptide vaccine for patients with recurrent glioblastoma (NCT02754362); (9) in combination with NEO-PV-01 and nivolumab, another PD-1-specific immune checkpoint blocker, in patients with melanoma, lung cancer, and bladder cancer (NCT02897765); (10) in combination with durvalumab and tremelimumab, a CTLA-4-targeting immune checkpoint blocker, for the treatment of advanced, measurable, biopsy-accessible cancers (NCT02643303); (11) in conjunction with a combination of the following agents- NEO-PV-01, APX005M, ipilimumab, and nivolumab214 for the treatment of advanced melanoma146 (NCT03597282); (12) in combination with MUC1- and influenza-specific vaccines along with tadalafil, an inhibitor of phosphodiesterase 5 (PDE5)215217 in patients with head and neck squamous cell carcinoma (NCT02544880); (13) in combination with CDX-1401 and pembrolizumab in patients with previously treated, advanced solid tumor (NCT02661100); (14) in combination with a peptide vaccine, GL-0817,218 and cyclophosphamide to the prevent the recurrence of squamous cell carcinoma of the oral cavity (NCT02873819); (15) in combination with PVX-41 and durvalumab with or without lenalidomide in patients with multiple myeloma (NCT02886065); (16) in combination with DEC-205/NY-ESO-1 fusion protein CDX-1401, decitabine, and nivolumab101 for the treatment of myelodysplastic syndrome or acute myeloid leukemia (NCT03358719); (17) in combination with GBM6-AD, a cancer cell-based vaccine,23 in patients with Grade II glioma (NCT02549833); (18) in combination with varlilumab and a peptide vaccine, IMA950,219221 in patients with low-grade glioma (NCT02924038); (19) in combination with H3.3K27M, a peptide vaccine,222,223 in children with newly diagnosed diffuse intrinsic pontine glioma and other newly diagnosed HLA-A2+ H3.3K27M+ positive gliomas (NCT02960230); and (20) as standalone therapy for prostate carcinoma in the neoadjuvant setting (NCT03262103).

Furthermore, SD-101 is being studied in clinical trials: (1) in combination with an anti-OXO40 antibody,224,225 BMS-986178, and RT for the treatment of lymphoma (NCT03410901); (2) in combination with RT90 after allogeneic hematopoietic cell transplant for the treatment of lymphoma (NCT01745354); (3) in combination with an anti-IL-10 agent226 for the treatment of advanced malignancies (NCT02731742); (4) in combination with pembrolizumab89,227,228 for the treatment of solid tumors (NCT02521870); (5) in combination with ibrutinib229,230 and RT for the treatment of follicular lymphoma (NCT02927964); (6) in combination with epacadostat, an inhibitor of indoleamine 2,3-dioxygenase-1,231,232 and RT for the treatment of advanced solid tumors and lymphoma (NCT03322384); (7) in combination with ipilimumab and RT for the treatment of lymphoma (NCT02254772); (8) in combination with RT229,233 for the treatment of lymphoma (NCT02266147); and (9) in combination with pembrolizumab90,234 and RT for the treatment of prostate carcinoma (NCT03007732).

The status of the following clinical trials discussed in our previous Trial Watches dealing with TLR agonists,88,235237 has changed during the last 35 months: NCT02501473, NCT02320305, NCT02180698, NCT02134925, NCT02149225, NCT02281682, NCT02454634, NCT02242929, NCT02293707, NCT02015104, NCT02035657, NCT01926496, NCT02061449, NCT01970358, and NCT02077868, which are now listed as “Active, not recruiting”; NCT02432378, NCT02427581, NCT02334735, NCT02394132, NCT02431559, NCT02521870, NCT02452697 and NCT02059499, which are currently listed as “Recruiting participants”; NCT02385188, which is listed as “Enrolling by invitation”; NCT02326168, NCT02333474, NCT02404389, NCT02482428, NCT02254772, NCT02266147, NCT01957878, NCT02078648, NCT01909752, and NCT01920191, which are listed as “Completed”; NCT02202044, NCT02510950, NCT02413827, NCT02332889, NCT02329171, NCT01902771, and NCT01984892, which have been “Terminated”; as well as NCT02495636, which has been “Withdrawn” (source http://clinicaltrials.gov/).

Concluding remarks

During the last 35 months (September 2015 – August 2018), more than 60 clinical trials have been initiated to investigate the potential therapeutic efficacy of TLR agonists in patients with cancer (Table 1). These metrics are notable for a decrease in the rate of initiation of clinical trials testing TLR agonists as immunotherapeutics for cancer as compared to the previous 15 months (May 2014 – August 2015), during which approximately 50 clinical trials were initiated. Although it is difficult to attribute such a decrease to one or more specific factors, we suspect that the extraordinary clinical achievement of other immunotherapeutics including immune checkpoint blockers and CAR-expressing T cells238-244 may have caused some refocusing in the attention of oncologists and pharmaceutical companies to investigate the ongoing role of TLR agonists. Currently available preclinical and clinical data strongly suggest that successful anticancer immunotherapy in a large fraction of patients requires combinatorial approaches. In this setting, TLR agonists present an opportunity to boost the immune response in patients in an effort to contribute to better clinical outcomes.

Acknowledgments

M. Smith would like to acknowledge funding from the American Society for Blood and Marrow Transplantation (ASBMT) New Investigator Award, the Burroughs Wellcome Fund Postdoctoral Enrichment Program, and the Damon Runyon Physician-Scientist Award. LG is supported by an intramural startup from the Department of Radiation Oncology of Weill Cornell Medical College (New York, US), and by donations from Sotio a.s. (Prague, Czech Republic), Phosplatin LLC (New York, US) and the Luke Heller TECPR2 Foundation (Boston, US).

Abbreviations

AE adverse event

AS04 Adjuvant System 04

ASCO American Society of Clinical Oncology

BCG bacillus Calmette-Guérin

CIN cervical intraepithelial neoplasm

CRC colorectal carcinoma

CTL cytotoxic T-lymphocyte

DAMP damage-associated molecular pattern

DC dendritic cell

ESMO European Society for Medical Oncology

FDA Food and Drug Administration

GBM glioblastoma multiforme

HDAC histone deacetylase

HPV human papillomavirus

i.d. intradermal

i.v. intravenous

LEEP loop electrosurgical excision procedure

LLETZ large loop excision of the transformation zone

LPS lipopolysaccharide

MDSC myeloid-derived suppressor cell

MPL monophosphoryl lipid A

NMC nanomedicinal cream

NCI National Cancer Institute

NK natural killer

NLR NOD-like receptor

NSCLC non-small cell lung carcinoma

ODN oligodeoxynucleotide

ORR objective response rate

OS overall survival

PAMP pathogen-associated molecular pattern

PD progressive disease

PFS progression-free survival

polyI:C polyriboinosinic polyribocytidylic acid

PRR pattern recognition receptor

RLR RIG-I-like receptor

SAE serious adverse event

s.c. subcutaneous

SD stable disease

TLR Toll-like receptor.

References

  • 1.Kawai T, Akira S.. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol. 2009;21:317–337. doi: 10.1093/intimm/dxp017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 3.Fucikova J, Moserova I, Urbanova L, Bezu L, Kepp O, Cremer I, Salek C, Strnad P, Kroemer G, Galluzzi L, et al. Prognostic and Predictive Value of DAMPs and DAMP-Associated Processes in Cancer. Front Immunol. 2015;6:402. doi: 10.3389/fimmu.2015.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Garg AD, Galluzzi L, Apetoh L, Baert T, Birge RB, Bravo-San Pedro JM, Breckpot K, Brough D, Chaurio R, Cirone M, et al. Molecular and translational classifications of DAMPs in immunogenic cell death. Front Immunol. 2015;6:588. doi: 10.3389/fimmu.2015.00588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. doi: 10.1038/nri.2016.107. [DOI] [PubMed] [Google Scholar]
  • 6.Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–305. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
  • 7.Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10:826–837. doi: 10.1038/nri2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013;59:583–594. doi: 10.1016/j.jhep.2013.03.033. [DOI] [PubMed] [Google Scholar]
  • 9.Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol. 2012;13:780–788. doi: 10.1038/nrm3479. [DOI] [PubMed] [Google Scholar]
  • 10.Vacchelli E, Ma Y, Baracco EE, Zitvogel L, Kroemer G. Yet another pattern recognition receptor involved in the chemotherapy-induced anticancer immune response: formyl peptide receptor-1. Oncoimmunology. 2016;5:e1118600. doi: 10.1080/2162402X.2015.1118600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology. 2017;6:e1386829. doi: 10.1080/2162402X.2017.1386829. [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;165:3541–3544. [DOI] [PubMed] [Google Scholar]
  • 13.Nw S, Opitz B, Lamping N, Ks M, Zahringer U, Ub G, 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;165:2683–2693. [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;316:1628–1632. 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;433:523–527. 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;410:1099–1103. 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;4:1247–1253. doi: 10.1038/ni1011. [DOI] [PubMed] [Google Scholar]
  • 18.Lu J, Pd S. The structure of the TLR5-flagellin complex: a new mode of pathogen detection, conserved receptor dimerization for signaling. Sci Signal. 2012;5:pe11. [PMC free article] [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;335:859–864. 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;408:740–745. 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;303:1529–1531. 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;303:1526–1529. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
  • 23.Garg AD, Vara Perez M, Schaaf M, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: dendritic cell-based anticancer immunotherapy. Oncoimmunology. 2017;6:e1328341. doi: 10.1080/2162402X.2017.1328341. [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;413:732–738. 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;309:581–585. 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;320:379–381. 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;287:43417–43423. doi: 10.1074/jbc.M112.411009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Oldenburg M, Kruger 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;337:1111–1115. doi: 10.1126/science.1220363. [DOI] [PubMed] [Google Scholar]
  • 29.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;277:15028–15034. doi: 10.1074/jbc.M200497200. [DOI] [PubMed] [Google Scholar]
  • 30.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;164:13–17. [DOI] [PubMed] [Google Scholar]
  • 31.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;174:5016–5023. [DOI] [PubMed] [Google Scholar]
  • 32.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;168:5989–5992. [DOI] [PubMed] [Google Scholar]
  • 33.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;16:713–717. doi: 10.1038/nm.2150. [DOI] [PubMed] [Google Scholar]
  • 34.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;13:1042–1049. doi: 10.1038/nm1638. [DOI] [PubMed] [Google Scholar]
  • 35.Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010;2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81:1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 37.Evankovich J, Billiar T, Tsung A. Toll-like receptors in hepatic ischemia/reperfusion and transplantation. Gastroenterol Res Pract. 2010;2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ. 2015;22:58–73. doi: 10.1038/cdd.2014.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Langefeld T, Mohamed W, Ghai R, Chakraborty T. Toll-like receptors and NOD-like receptors: domain architecture and cellular signalling. Adv Exp Med Biol. 2009;653:48–57. [DOI] [PubMed] [Google Scholar]
  • 40.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]
  • 41.Barton GM, Kagan JC. A cell biological view of Toll-like receptor function: regulation through compartmentalization. Nat Rev Immunol. 2009;9:535–542. 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 GAO, 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;56:1971–1982. 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;464:104–107. doi: 10.1038/nature08780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–875. doi: 10.1038/nrc3380. [DOI] [PubMed] [Google Scholar]
  • 45.Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541. doi: 10.1038/s41418-017-0012-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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;270:1804–1806. [DOI] [PubMed] [Google Scholar]
  • 47.Gomez-Gomez L, Boller T. FLS2: an LRR receptor-like kinase involved in the perception of the bacterial elicitor flagellin in Arabidopsis. Mol Cell. 2000;5:1003–1011. [DOI] [PubMed] [Google Scholar]
  • 48.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 U S A. 2005;102:9577–9582. doi: 10.1073/pnas.0502272102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Anderson KV, Bokla L, Nusslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: the induction of polarity by the Toll gene product. Cell. 1985;42:791–798. [DOI] [PubMed] [Google Scholar]
  • 50.Anderson KV, Jurgens G, Nusslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: genetic studies on the role of the Toll gene product. Cell. 1985;42:779–789. [DOI] [PubMed] [Google Scholar]
  • 51.Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 52.Beutler B. Inferences, questions and possibilities in Toll-like receptor signalling. Nature. 2004;430:257–263. doi: 10.1038/nature02761. [DOI] [PubMed] [Google Scholar]
  • 53.Hoffmann JA. The immune response of Drosophila. Nature. 2003;426:33–38. doi: 10.1038/nature02021. [DOI] [PubMed] [Google Scholar]
  • 54.Janeway CA Jr., Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. doi: 10.1146/annurev.immunol.20.083001.084359. [DOI] [PubMed] [Google Scholar]
  • 55.Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86:973–983. [DOI] [PubMed] [Google Scholar]
  • 56.Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34:637–650. doi: 10.1016/j.immuni.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 57.Lauw FN, Caffrey DR, Golenbock DT. Of mice and man: TLR11 (finally) finds profilin. Trends Immunol. 2005;26:509–511. doi: 10.1016/j.it.2005.08.006. [DOI] [PubMed] [Google Scholar]
  • 58.Bowie A, O’Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal generators for pro-inflammatory interleukins and microbial products. J Leukoc Biol. 2000;67:508–514. [DOI] [PubMed] [Google Scholar]
  • 59.Ve T, Gay NJ, Mansell A, Kobe B, Kellie S. Adaptors in toll-like receptor signaling and their potential as therapeutic targets. Curr Drug Targets. 2012;13:1360–1374. [DOI] [PubMed] [Google Scholar]
  • 60.Huang B, Zhao J, Unkeless JC, Feng ZH, Xiong H. TLR signaling by tumor and immune cells: a double-edged sword. Oncogene. 2008;27:218–224. doi: 10.1038/sj.onc.1210904. [DOI] [PubMed] [Google Scholar]
  • 61.Su X, Ye J, Hsueh EC, Zhang Y, Hoft DF, Peng G. Tumor microenvironments direct the recruitment and expansion of human Th17 cells. J Immunol. 2010;184:1630–1641. doi: 10.4049/jimmunol.0902813. [DOI] [PubMed] [Google Scholar]
  • 62.Ye J, Ma C, Hsueh EC, Dou J, Mo W, Liu S, Han B, Huang Y, Zhang Y, Varvares MA, et al. TLR8 signaling enhances tumor immunity by preventing tumor-induced T-cell senescence. EMBO Mol Med. 2014;6:1294–1311. doi: 10.15252/emmm.201403918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, Cross JR, Jung E, Thompson CB, Jones RG, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010;115:4742–4749. doi: 10.1182/blood-2009-10-249540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Pantel A, Teixeira A, Haddad E, Wood EG, Steinman RM, Longhi MP. Direct type I IFN but not MDA5/TLR3 activation of dendritic cells is required for maturation and metabolic shift to glycolysis after poly IC stimulation. PLoS Biol. 2014;12:e1001759. doi: 10.1371/journal.pbio.1001759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25:771–784. doi: 10.1038/cr.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Qualls JE, Subramanian C, Rafi W, Smith AM, Balouzian L, DeFreitas AA, Shirey KA, Reutterer B, Kernbauer E, Stockinger S, et al. Sustained generation of nitric oxide and control of mycobacterial infection requires argininosuccinate synthase 1. Cell Host Microbe. 2012;12:313–323. doi: 10.1016/j.chom.2012.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Geeraerts X, Bolli E, Fendt SM, Van Ginderachter JA. Macrophage metabolism as therapeutic target for cancer, atherosclerosis, and obesity. Front Immunol. 2017;8:289. doi: 10.3389/fimmu.2017.00289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Everts B, Pearce EJ. Metabolic control of dendritic cell activation and function: recent advances and clinical implications. Front Immunol. 2014;5:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dong H, Bullock TN. Metabolic influences that regulate dendritic cell function in tumors. Front Immunol. 2014;5:24. doi: 10.3389/fimmu.2014.00024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Smits EL, Ponsaerts P, Berneman ZN, Van Tendeloo VF. The use of TLR7 and TLR8 ligands for the enhancement of cancer immunotherapy. Oncologist. 2008;13:859–875. doi: 10.1634/theoncologist.2008-0097. [DOI] [PubMed] [Google Scholar]
  • 71.Ridnour LA, Cheng RY, Switzer CH, Heinecke JL, Ambs S, Glynn S, Young HA, Trinchieri G, Wink DA. Molecular pathways: toll-like receptors in the tumor microenvironment–poor prognosis or new therapeutic opportunity. Clin Cancer Res. 2013;19:1340–1346. doi: 10.1158/1078-0432.CCR-12-0408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yu L, Wang L, Chen S. Dual character of Toll-like receptor signaling: pro-tumorigenic effects and anti-tumor functions. Biochim Biophys Acta. 2013;1835:144–154. doi: 10.1016/j.bbcan.2012.10.006. [DOI] [PubMed] [Google Scholar]
  • 73.Veyrat M, Durand S, Classe M, Glavan TM, Oker N, Kapetanakis NI, Jiang X, Gelin A, Herman P, Casiraghi O, et al. Stimulation of the toll-like receptor 3 promotes metabolic reprogramming in head and neck carcinoma cells. Oncotarget. 2016;7:82580–82593. doi: 10.18632/oncotarget.12892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Karki K, Pande D, Negi R, Khanna S, Khanna RS, Khanna HD. Correlation of serum toll like receptor 9 and trace elements with lipid peroxidation in the patients of breast diseases. J Trace Elem Med Biol. 2015;30:11–16. doi: 10.1016/j.jtemb.2014.12.003. [DOI] [PubMed] [Google Scholar]
  • 75.Hotz C, Treinies M, Mottas I, Rotzer LC, Oberson A, Spagnuolo L, Perdicchio M, Spinetti T, Herbst T, Bourquin C. Reprogramming of TLR7 signaling enhances antitumor NK and cytotoxic T cell responses. Oncoimmunology. 2016;5:e1232219. doi: 10.1080/2162402X.2016.1232219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kanzler H, Barrat FJ, Hessel EM, Coffman RL. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat Med. 2007;13:552–559. doi: 10.1038/nm1589. [DOI] [PubMed] [Google Scholar]
  • 77.Hoffman ES, Smith RE, Renaud RC Jr.. From the analyst’s couch: TLR-targeted therapeutics. Nat Rev Drug Discov. 2005;4:879–880. doi: 10.1038/nrd1880. [DOI] [PubMed] [Google Scholar]
  • 78.Wiedemann GM, Jacobi SJ, Chaloupka M, Krachan A, Hamm S, Strobl S, Baumgartner R, Rothenfusser S, Duewell P, Endres S, et al. A novel TLR7 agonist reverses NK cell anergy and cures RMA-S lymphoma-bearing mice. Oncoimmunology. 2016;5:e1189051. doi: 10.1080/2162402X.2016.1189051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hanna E, Abadi R, Abbas O. Imiquimod in dermatology: an overview. Int J Dermatol. 2016;55:831–844. doi: 10.1111/ijd.13235. [DOI] [PubMed] [Google Scholar]
  • 80.Wolff F, Loipetzberger A, Gruber W, Esterbauer H, Aberger F, Frischauf AM. Imiquimod directly inhibits Hedgehog signalling by stimulating adenosine receptor/protein kinase A-mediated GLI phosphorylation. Oncogene. 2013;32:5574–5581. doi: 10.1038/onc.2013.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.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;74:277–286. [DOI] [PubMed] [Google Scholar]
  • 82.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-Guerin peptidoglycan. Infect Immun. 2003;71:4238–4249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Redelman-Sidi G, Glickman MS, Bochner BH. The mechanism of action of BCG therapy for bladder cancer–a current perspective. Nat Rev Urol. 2014;11:153–162. doi: 10.1038/nrurol.2014.15. [DOI] [PubMed] [Google Scholar]
  • 84.Paavonen J, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter D, Kitchener H, Castellsague X, Teixeira JC, Skinner SR, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009;374:301–314. doi: 10.1016/S0140-6736(09)61248-4. [DOI] [PubMed] [Google Scholar]
  • 85.Srivastava AK, Yolcu ES, Dinc G, Sharma RK. Shirwan H. SA-4-1BBL/MPL as a novel immune adjuvant platform to combat cancer. Oncoimmunology. 2016;5:e1064580. doi: 10.1080/2162402X.2015.1064580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Szarewski A. Cervarix(R): a bivalent vaccine against HPV types 16 and 18, with cross-protection against other high-risk HPV types. Expert Rev Vaccines. 2012;11:645–657. doi: 10.1586/erv.12.42. [DOI] [PubMed] [Google Scholar]
  • 87.Didierlaurent AM, Morel S, Lockman L, Giannini SL, Bisteau M, Carlsen H, Kielland A, Vosters O, Vanderheyde N, Schiavetti F, et al. AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J Immunol. 2009;183:6186–6197. doi: 10.4049/jimmunol.0901474. [DOI] [PubMed] [Google Scholar]
  • 88.Iribarren K, Bloy N, Buque A, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Špíšek R, Zitvogel L, et al. Trial Watch: immunostimulation with Toll-like receptor agonists in cancer therapy. Oncoimmunology. 2016;5:e1088631. doi: 10.1080/2162402X.2015.1088631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wang S, Campos J, Gallotta M, Gong M, Crain C, Naik E, Coffman RL, Guiducci C. Intratumoral injection of a CpG oligonucleotide reverts resistance to PD-1 blockade by expanding multifunctional CD8+ T cells. Proc Natl Acad Sci U S A. 2016;113:E7240–e9. doi: 10.1073/pnas.1608555113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Vacchelli E, Bloy N, Aranda F, Buque A, Cremer I, Demaria S, Eggermont A, Formenti SC, Fridman WH, Fucikova J, et al. Trial Watch: immunotherapy plus radiation therapy for oncological indications. Oncoimmunology. 2016;5:e1214790. doi: 10.1080/2162402X.2016.1214790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Khodadoust MS, Chu MP, Czerwinski D, McDonald K, Long S, Kohrt HE, Hoppe RT, Advani RH, Lowsky T, Levy R. Phase I/II study of intratumoral injection of SD-101, an immunostimulatory CpG, and intratumoral injection of ipillumumab, an anti-CTLA-4 monoclonal antibody, in combination with local radiation in low-grade B-cell lymphomas. J Clin Oncol. 2015;33:TPS8604–TPS. [Google Scholar]
  • 92.Hammerich L, Binder A, Brody JD. In situ vaccination: cancer immunotherapy both personalized and off-the-shelf. Mol Oncol. 2015;9:1966–1981. doi: 10.1016/j.molonc.2015.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ok CY, Young KH. Checkpoint inhibitors in hematological malignancies. J Hematol Oncol. 2017;10:103. doi: 10.1186/s13045-017-0474-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Edwards L, Ferenczy A, Eron L, Baker D, Owens ML, Fox TL, Hougham AJ, Schmitt KA. Self-administered topical 5% imiquimod cream for external anogenital warts. HPV Study Group. Human PapillomaVirus. Arch Dermatol. 1998;134:25–30. [DOI] [PubMed] [Google Scholar]
  • 95.Chen FP. Efficacy of imiquimod 5% cream for persistent human papillomavirus in genital intraepithelial neoplasm. Taiwan J Obstet Gynecol. 2013;52:475–478. doi: 10.1016/j.tjog.2013.10.004. [DOI] [PubMed] [Google Scholar]
  • 96.Riedmann EM. CDX-1401 combined with TLR agonist: positive phase 1 results. Hum Vaccin Immunother. 2012;8:1742. [PubMed] [Google Scholar]
  • 97.Dhodapkar MV, Sznol M, Zhao B, Wang D, Carvajal RD, Keohan ML, Chuang E, Sanborn RE, Lutzky J, Powderly J, et al. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci Transl Med. 2014;6:232ra51. doi: 10.1126/scitranslmed.3008068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Patchett AL, Tovar C, Corcoran LM, Lyons AB, Woods GM. The toll-like receptor ligands Hiltonol((R)) (polyICLC) and imiquimod effectively activate antigen-specific immune responses in Tasmanian devils (Sarcophilus harrisii). Dev Comp Immunol. 2017;76:352–360. doi: 10.1016/j.dci.2017.07.004. [DOI] [PubMed] [Google Scholar]
  • 99.Rodriguez-Ruiz ME, Perez-Gracia JL, Rodriguez I, Alfaro C, Onate C, Perez G, Gil-Bazo I, Benito A, Inogés S, López-Diaz de Cerio A, et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann Oncol. 2018;29:1312–1319. doi: 10.1093/annonc/mdy089. [DOI] [PubMed] [Google Scholar]
  • 100.Lainey E, Wolfromm A, Marie N, Enot D, Scoazec M, Bouteloup C, Leroy C, Micol J-B, De Botton S, Galluzzi L, et al. Azacytidine and erlotinib exert synergistic effects against acute myeloid leukemia. Oncogene. 2013;32:4331–4342. doi: 10.1038/onc.2012.469. [DOI] [PubMed] [Google Scholar]
  • 101.Griffiths EA, Srivastava P, Matsuzaki J, Brumberger Z, Wang ES, Kocent J, Miller A, Roloff GW, Wong HY, Paluch BE, et al. NY-ESO-1 vaccination in combination with decitabine induces antigen-specific T-lymphocyte responses in patients with myelodysplastic syndrome. Clin Cancer Res. 2018;24:1019–1029. doi: 10.1158/1078-0432.CCR-17-1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Alderson MR, McGowan P, Baldridge JR, Probst P. TLR4 agonists as immunomodulatory agents. J Endotoxin Res. 2006;12:313–319. doi: 10.1179/096805106X118753. [DOI] [PubMed] [Google Scholar]
  • 103.Dowling JK, Mansell A. Toll-like receptors: the swiss army knife of immunity and vaccine development. Clin Transl Immunology. 2016;5:e85. doi: 10.1038/cti.2016.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Dong H, Su H, Chen L, Liu K, Hu HM, Yang W, Mou Y. Immunocompetence and mechanism of the DRibble-DCs vaccine for oral squamous cell carcinoma. Cancer Manag Res. 2018;10:493–501. doi: 10.2147/CMAR.S155914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yu G, Li Y, Cui Z, Morris NP, Weinberg AD, Fox BA, Urba WJ, Wang L, Hu H-M. Combinational Immunotherapy with Allo-DRibble Vaccines and Anti-OX40 Co-Stimulation leads to generation of cross-reactive effector T cells and tumor regression. Sci Rep. 2016;6:37558. doi: 10.1038/srep37558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Li J, Xing Y, Zhou Z, Yao W, Cao R, Li T, Xu M, Wu J. Microbial HSP70 peptide epitope 407-426 as adjuvant in tumor-derived autophagosome vaccine therapy of mouse lung cancer. Tumour Biol. 2016;37:15097–15105. doi: 10.1007/s13277-016-5309-2. [DOI] [PubMed] [Google Scholar]
  • 107.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;17:7047–7057. doi: 10.1158/1078-0432.CCR-11-0951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ren H, Zhao S, Li W, Dong H, Zhou M, Cao M, Hu H-M, Wang L-X. Therapeutic antitumor efficacy of B cells loaded with tumor-derived autophagasomes vaccine (DRibbles). J Immunother. 2014;37:383–393. doi: 10.1097/CJI.0000000000000051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Althammer SSK, Rebelatto M, Tan TH, Wiestler T, Schmidt G, Higgs B, Li X, Shi L, Jin X, Antal J, et al. Combinatorial CD8+ and PD-L1+ cell densities correlate with response and improved survival in non-small cell lung cancer (NSCLC) patients treated with durvalumab. MD, USA: Society for Immunotherapy of Cancer National Harbor; 2016. [Google Scholar]
  • 110.Azuma M, Takeda Y, Nakajima H, Sugiyama H, Ebihara T, Oshiumi H, Matsumoto M, Seya T. Biphasic function of TLR3 adjuvant on tumor and spleen dendritic cells promotes tumor T cell infiltration and regression in a vaccine therapy. Oncoimmunology. 2016;5:e1188244. doi: 10.1080/2162402X.2016.1188244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mehrotra S, Britten CD, Chin S, Garrett-Mayer E, Cloud CA, Li M, Scurti G, Salem ML, Nelson MH, Thomas MB, et al. Vaccination with poly(IC:LC) and peptide-pulsed autologous dendritic cells in patients with pancreatic cancer. J Hematol Oncol. 2017;10:82. doi: 10.1186/s13045-017-0459-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chiang CL, Coukos G, Kandalaft LE. Whole Tumor Antigen Vaccines: where Are We? Vaccines (Basel). 2015;3:344–372. doi: 10.3390/vaccines3020344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Schmitz-Winnenthal FH, Hohmann N, Schmidt T, Podola L, Friedrich T, Lubenau H, Springer M, Wieckowski S, Breiner KM, Mikus G, et al. A phase 1 trial extension to assess immunologic efficacy and safety of prime-boost vaccination with VXM01, an oral T cell vaccine against VEGFR2, in patients with advanced pancreatic cancer. Oncoimmunology. 2018;7:e1303584. doi: 10.1080/2162402X.2017.1303584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Garzon-Muvdi T, Theodros D, Luksik AS, Maxwell R, Kim E, Jackson CM, Belcaid Z, Ganguly S, Tyler B, Brem H, et al. Dendritic cell activation enhances anti-PD-1 mediated immunotherapy against glioblastoma. Oncotarget. 2018;9:20681–20697. doi: 10.18632/oncotarget.25061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A, Normolle DP, Connelly AK, Dibridge S, Mason G, Whiteside TL, et al. Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomas. Neuro Oncol. 2016;18:1157–1168. doi: 10.1093/neuonc/now026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Qin J, Qian Y, Yao J, Grace C, Li X. SIGIRR inhibits interleukin-1 receptor- and toll-like receptor 4-mediated signaling through different mechanisms. J Biol Chem. 2005;280:25233–25241. doi: 10.1074/jbc.M501363200. [DOI] [PubMed] [Google Scholar]
  • 117.Gong J, Wei T, Stark RW, Jamitzky F, Heckl WM, Anders HJ, Lech M, Rössle SC. Inhibition of Toll-like receptors TLR4 and 7 signaling pathways by SIGIRR: a computational approach. J Struct Biol. 2010;169:323–330. doi: 10.1016/j.jsb.2009.12.007. [DOI] [PubMed] [Google Scholar]
  • 118.Molgora M, Bonavita E, Ponzetta A, Riva F, Barbagallo M, Jaillon S, Popović B, Bernardini G, Magrini E, Gianni F, et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature. 2017;551:110–114. doi: 10.1038/nature24293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Luo L, Bokil NJ, Wall AA, Kapetanovic R, Lansdaal NM, Marceline F, Burgess BJ, Tong SJ, Guo Z, Alexandrov K, et al. SCIMP is a transmembrane non-TIR TLR adaptor that promotes proinflammatory cytokine production from macrophages. Nat Commun. 2017;8:14133. doi: 10.1038/ncomms14133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, et al. Molecular definitions of autophagy and related processes. Embo J. 2017;36:1811–1836. doi: 10.15252/embj.201796697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Acharya M, Sokolovska A, Tam JM, Conway KL, Stefani C, Raso F, Mukhopadhyay S, Feliu M, Paul E, Savill J, et al. alphav Integrins combine with LC3 and atg5 to regulate Toll-like receptor signalling in B cells. Nat Commun. 2016;7:10917. doi: 10.1038/ncomms10917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol. 2015;15:405–414. doi: 10.1038/nri3845. [DOI] [PubMed] [Google Scholar]
  • 123.Combes A, Camosseto V, N’Guessan P, Arguello RJ, Mussard J, Caux C, Bendriss-Vermare N, Pierre P, Gatti E. BAD-LAMP controls TLR9 trafficking and signalling in human plasmacytoid dendritic cells. Nat Commun. 2017;8:913. doi: 10.1038/s41467-017-00695-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.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;424:743–748. doi: 10.1038/nature01889. [DOI] [PubMed] [Google Scholar]
  • 125.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;301:640–643. doi: 10.1126/science.1087262. [DOI] [PubMed] [Google Scholar]
  • 126.Scheeren FA, Kuo AH, van Weele LJ, Cai S, Glykofridis I, Sikandar SS, Zabala M, Qian D, Lam JS, Johnston D, et al. A cell-intrinsic role for TLR2-MYD88 in intestinal and breast epithelia and oncogenesis. Nat Cell Biol. 2014;16:1238–1248. doi: 10.1038/ncb3058. [DOI] [PubMed] [Google Scholar]
  • 127.New M, Sheikh S, Bekheet M, Olzscha H, Thezenas ML, Care MA, Fotheringham S, Tooze RM, Kessler B, La Thangue NB. TLR Adaptor Protein MYD88 mediates sensitivity to HDAC Inhibitors via a Cytokine-Dependent Mechanism. Cancer Res. 2016;76:6975–6987. doi: 10.1158/0008-5472.CAN-16-0504. [DOI] [PubMed] [Google Scholar]
  • 128.Zhang Z, Ohto U, Shibata T, Krayukhina E, Taoka M, Yamauchi Y, Tanji H, Isobe T, Uchiyama S, Miyake K, et al. Structural analysis reveals that toll-like receptor 7 Is a dual receptor for guanosine and single-stranded RNA. Immunity. 2016;45:737–748. doi: 10.1016/j.immuni.2016.09.011. [DOI] [PubMed] [Google Scholar]
  • 129.Takeda Y, Yoshida S, Takashima K, Ishii-Mugikura N, Shime H, Seya T, Matsumoto M. Vaccine immunotherapy with ARNAX induces tumor-specific memory T cells and durable anti-tumor immunity in mouse models. Cancer Sci. 2018;109:2119–2129. doi: 10.1111/cas.13649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Matsumoto M, Takeda Y, Tatematsu M, Seya T. Toll-Like receptor 3 signal in dendritic cells benefits cancer immunotherapy. Front Immunol. 2017;8:1897. doi: 10.3389/fimmu.2017.01897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Takeda Y, Kataoka K, Yamagishi J, Ogawa S, Seya T, Matsumoto M. A TLR3-specific adjuvant relieves innate resistance to PD-L1 blockade without cytokine toxicity in tumor vaccine immunotherapy. Cell Rep. 2017;19:1874–1887. doi: 10.1016/j.celrep.2017.05.015. [DOI] [PubMed] [Google Scholar]
  • 132.Galluzzi L, Vanpouille-Box C, Bakhoum SF, Demaria S. SnapShot: CGAS-STING Signaling. Cell. 2018;173:276–e1. doi: 10.1016/j.cell.2018.03.015. [DOI] [PubMed] [Google Scholar]
  • 133.Caronni N, Simoncello F, Stafetta F, Guarnaccia C, Ruiz-Moreno JS, Opitz B, Galli T, Proux-Gillardeaux V, Benvenuti F. Downregulation of membrane trafficking proteins and lactate conditioning determine loss of dendritic cell function in lung cancer. Cancer Res. 2018;78:1685–1699. doi: 10.1158/0008-5472.CAN-17-1307. [DOI] [PubMed] [Google Scholar]
  • 134.Bantug GR, Galluzzi L, Kroemer G, Hess C. The spectrum of T cell metabolism in health and disease. Nat Rev Immunol. 2018;18:19–34. doi: 10.1038/nri.2017.99. [DOI] [PubMed] [Google Scholar]
  • 135.Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107:2013–2021. doi: 10.1182/blood-2005-05-1795. [DOI] [PubMed] [Google Scholar]
  • 136.Iraporda C, Errea A, Romanin DE, Cayet D, Pereyra E, Pignataro O, Sirard JC, Garrote GL, Abraham AG, Rumbo M. Lactate and short chain fatty acids produced by microbial fermentation downregulate proinflammatory responses in intestinal epithelial cells and myeloid cells. Immunobiology. 2015;220:1161–1169. doi: 10.1016/j.imbio.2015.06.004. [DOI] [PubMed] [Google Scholar]
  • 137.Hoque R, Farooq A, Ghani A, Gorelick F, Mehal WZ. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology. 2014;146:1763–1774. doi: 10.1053/j.gastro.2014.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Doorduijn EM, Sluijter M, Salvatori DC, Silvestri S, Maas S, Arens R, Ossendorp F, van der Burg SH, van Hall T. CD4(+) T Cell and NK cell interplay key to regression of MHC Class I(low) Tumors upon TLR7/8 Agonist Therapy. Cancer Immunol Res. 2017;5:642–653. doi: 10.1158/2326-6066.CIR-16-0334. [DOI] [PubMed] [Google Scholar]
  • 139.Cho JH, Lee HJ, Ko HJ, Yoon BI, Choe J, Kim KC, Hahn T-W, Han JA, Choi SS, Jung YM, et al. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget. 2017;8:24932–24948. doi: 10.18632/oncotarget.15326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Fournier C, Martin F, Zitvogel L, Kroemer G, Galluzzi L, Apetoh L. Trial Watch: adoptively transferred cells for anticancer immunotherapy. Oncoimmunology. 2017;6:e1363139. doi: 10.1080/2162402X.2017.1363139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lopez-Soto A, Gonzalez S, Galluzzi L. Soluble NKG2D ligands limit the efficacy of immune checkpoint blockade. Oncoimmunology. 2017;6:e1346766. doi: 10.1080/2162402X.2017.1346766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Lopez-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of Metastasis by NK cells. Cancer Cell. 2017;32:135–154. doi: 10.1016/j.ccell.2017.06.009. [DOI] [PubMed] [Google Scholar]
  • 143.Klein JC, Moses K, Zelinskyy G, Sody S, Buer J, Lang S, Helfrich I, Dittmer U, Kirschning CJ, Brandau S. Combined toll-like receptor 3/7/9 deficiency on host cells results in T-cell-dependent control of tumour growth. Nat Commun. 2017;8:14600. doi: 10.1038/ncomms14600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Ghannam S, Pene J, Moquet-Torcy G, Jorgensen C, Yssel H. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol. 2010;185:302–312. doi: 10.4049/jimmunol.0902007. [DOI] [PubMed] [Google Scholar]
  • 145.Duffy MM, Ritter T, Ceredig R, Griffin MD. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011;2:34. doi: 10.1186/scrt75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Uzhachenko R, Shanker A. Notching tumor: signaling through notch receptors improves antitumor T cell immunity. Oncoimmunology. 2016;5:e1122864. doi: 10.1080/2162402X.2015.1122864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Rashedi I, Gomez-Aristizabal A, Wang XH, Viswanathan S, Keating A. TLR3 or TLR4 activation enhances mesenchymal stromal cell-mediated treg induction via notch signaling. Stem Cells. 2017;35:265–275. doi: 10.1002/stem.2485. [DOI] [PubMed] [Google Scholar]
  • 148.Bagchi A, Herrup EA, Warren HS, Trigilio J, Shin HS, Valentine C, Hellman J. MyD88-dependent and MyD88-independent pathways in synergy, priming, and tolerance between TLR agonists. J Immunol. 2007;178:1164–1171. [DOI] [PubMed] [Google Scholar]
  • 149.Hart OM, Athie-Morales V, O’Connor GM, Gardiner CM. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-gamma production. J Immunol. 2005;175:1636–1642. [DOI] [PubMed] [Google Scholar]
  • 150.Xu D, Wang P, Yang J, Qian Q, Li M, Wei L, Xu W. Gr-1+ cells other Than Ly6G+ neutrophils limit virus replication and promote myocardial inflammation and fibrosis following coxsackievirus B3 infection of mice. Front Cell Infect Microbiol. 2018;8:157. doi: 10.3389/fcimb.2018.00026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ma C, Kapanadze T, Gamrekelashvili J, Manns MP, Korangy F, Greten TF. Anti-Gr-1 antibody depletion fails to eliminate hepatic myeloid-derived suppressor cells in tumor-bearing mice. J Leukoc Biol. 2012;92:1199–1206. doi: 10.1189/jlb.0212059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Adamus T, Kortylewski M. The revival of CpG oligonucleotide-based cancer immunotherapies. Contemp Oncol (Pozn). 2018;22:56–60. doi: 10.5114/wo.2018.73887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Guan X, Chen J, Hu Y, Lin L, Sun P, Tian H, Chen X. Highly enhanced cancer immunotherapy by combining nanovaccine with hyaluronidase. Biomaterials. 2018;171:198–206. doi: 10.1016/j.biomaterials.2018.04.039. [DOI] [PubMed] [Google Scholar]
  • 154.Mondragon L, Kroemer G, Galluzzi L. Immunosuppressive gammadelta T cells foster pancreatic carcinogenesis. Oncoimmunology. 2016;5:e1237328. doi: 10.1080/2162402X.2016.1237328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines–from host defence to tissue homeostasis. Nat Rev Immunol. 2014;14:783–795. doi: 10.1038/nri3766. [DOI] [PubMed] [Google Scholar]
  • 156.Le Noci V, Sommariva M, Tortoreto M, Zaffaroni N, Campiglio M, Tagliabue E, Balsari A, Sfondrini L. Reprogramming the lung microenvironment by inhaled immunotherapy fosters immune destruction of tumor. Oncoimmunology. 2016;5:e1234571. doi: 10.1080/2162402X.2016.1234571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Pol J, Kroemer G, Galluzzi L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology. 2016;5:e1115641. doi: 10.1080/2162402X.2015.1115641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Vacchelli E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: oncolytic viruses for cancer therapy. Oncoimmunology. 2013;2:e24612. doi: 10.4161/onci.24612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Muller E, Christopoulos PF, Halder S, Lunde A, Beraki K, Speth M, Øynebråten I, Corthay A. Toll-Like Receptor Ligands and Interferon-gamma Synergize for Induction of Antitumor M1 Macrophages. Front Immunol. 2017;8:1383. doi: 10.3389/fimmu.2017.01383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.He Y, Wang N, Zhou X, Wang J, Ding Z, Chen X, Deng Y. Prognostic value of ki67 in BCG-treated non-muscle invasive bladder cancer: a meta-analysis and systematic review. BMJ Open. 2018;8:e019635. doi: 10.1136/bmjopen-2017-019635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Galluzzi L, Lopez-Soto A, Kumar S, Kroemer G. Caspases connect cell-death signaling to organismal homeostasis. Immunity. 2016;44:221–231. doi: 10.1016/j.immuni.2016.01.020. [DOI] [PubMed] [Google Scholar]
  • 162.Martins I, Galluzzi L, Kroemer G. Hormesis, cell death and aging. Aging (Albany NY). 2011;3:821–828. doi: 10.18632/aging.100380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Camargo JA, Passos GR, Ferrari KL, Billis A, Saad MJA, Reis LO. Intravesical immunomodulatory imiquimod enhances bacillus calmette-guerin downregulation of nonmuscle-invasive bladder cancer. Clin Genitourin Cancer. 2018;16:e587–e93. doi: 10.1016/j.clgc.2017.10.019. [DOI] [PubMed] [Google Scholar]
  • 164.Rybstein MD, Bravo-San Pedro JM, Kroemer G, Galluzzi L. The autophagic network and cancer. Nat Cell Biol. 2018;20:243–251. [DOI] [PubMed] [Google Scholar]
  • 165.Sica V, Galluzzi L, Bravo-San Pedro JM, Izzo V, Maiuri MC, Kroemer G. Organelle-Specific Initiation of Autophagy. Mol Cell. 2015;59:522–539. doi: 10.1016/j.molcel.2015.07.021. [DOI] [PubMed] [Google Scholar]
  • 166.Kim H, Niu L, Larson P, Kucaba TA, Murphy KA, James BR, Ferguson DM, Griffith TS, Panyam J. Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials. 2018;164:38–53. doi: 10.1016/j.biomaterials.2018.02.034. [DOI] [PubMed] [Google Scholar]
  • 167.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;132:434–439. [DOI] [PubMed] [Google Scholar]
  • 168.Ammi R, De Waele J, Willemen Y, Van Brussel I, Schrijvers DM, Lion E, Smits ELJ. Poly(I:C) as cancer vaccine adjuvant: knocking on the door of medical breakthroughs. Pharmacol Ther. 2015;146:120–131. doi: 10.1016/j.pharmthera.2014.09.010. [DOI] [PubMed] [Google Scholar]
  • 169.Zeichner JA, Patel RV, Birge MB. Treatment of Basal cell carcinoma with curettage followed by imiquimod 3.75% cream. J Clin Aesthet Dermatol. 2011;4:39–43. [PMC free article] [PubMed] [Google Scholar]
  • 170.Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology. 2017;6:e1398878. doi: 10.1080/2162402X.2017.1398878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Macedo R, Rochefort J, Guillot-Delost M, Tanaka K, Le Moignic A, Noizat C, Baillou C, Mateo V, Carpentier AF, Tartour E, et al. Intra-cheek immunization as a novel vaccination route for therapeutic vaccines of head and neck squamous cell carcinomas using plasmo virus-like particles. Oncoimmunology. 2016;5:e1164363. doi: 10.1080/2162402X.2016.1164363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078–2088. doi: 10.1016/S0140-6736(15)00239-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Morrow MP, Kraynyak KA, Sylvester AJ, Shen X, Amante D, Sakata L, Parker L, Yan J, Boyer J, Roh C, Humeau L, et al. Augmentation of cellular and humoral immune responses to HPV16 and HPV18 E6 and E7 antigens by VGX-3100. Mol Ther Oncolytics. 2016;3:16025. doi: 10.1038/mto.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Morrow MP, Kraynyak KA, Sylvester AJ, Dallas M, Knoblock D, Boyer JD, Yan J, Vang R, Khan AS, Humeau L, et al. Clinical and immunologic biomarkers for histologic regression of high-grade cervical dysplasia and clearance of HPV16 and HPV18 after Immunotherapy. Clin Cancer Res. 2018;24:276–294. doi: 10.1158/1078-0432.CCR-17-2335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Kowalewski DJ, Schuster H, Backert L, Berlin C, Kahn S, Kanz L, Salih HR, Rammensee H-G, Stevanovic S, Stickel JS. HLA ligandome analysis identifies the underlying specificities of spontaneous antileukemia immune responses in chronic lymphocytic leukemia (CLL). Proc Natl Acad Sci U S A. 2015;112:E166–75. doi: 10.1073/pnas.1416389112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Mauldin IS, Wages NA, Stowman AM, Wang E, Olson WC, Deacon DH, Smith KT, Galeassi N, Teague JE, Smolkin ME, et al. Topical treatment of melanoma metastases with imiquimod, plus administration of a cancer vaccine, promotes immune signatures in the metastases. Cancer Immunol Immunother. 2016;65:1201–1212. doi: 10.1007/s00262-016-1880-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Vo D-N, Alexia C, Allende-Vega N, Morschhauser F, Houot R, Menard C, Tarte K, Cartron G, Villalba M. NK cell activation and recovery of NK cell subsets in lymphoma patients after obinutuzumab and lenalidomide treatment. Oncoimmunology. 2018;7:e1409322. doi: 10.1080/2162402X.2017.1409322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ray A, Tian Z, Das DS, Coffman RL, Richardson P, Chauhan D, Anderson KC. A novel TLR-9 agonist C792 inhibits plasmacytoid dendritic cell-induced myeloma cell growth and enhance cytotoxicity of bortezomib. Leukemia. 2014;28:1716–1724. doi: 10.1038/leu.2014.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Itchaki G, Brown JR. Lenalidomide in the treatment of chronic lymphocytic leukemia. Expert Opin Investig Drugs. 2017;26:633–650. doi: 10.1080/13543784.2017.1313230. [DOI] [PubMed] [Google Scholar]
  • 180.Palma M, Hansson L, Mulder TA, Adamson L, Nasman-Glaser B, Eriksson I, Heimersson K, Ryblom H, Mozaffari F, Svensson A, et al. Lenalidomide as immune adjuvant to a dendritic cell vaccine in chronic lymphocytic leukemia patients. Eur J Haematol. 2018;101:68–77. doi: 10.1111/ejh.13065. [DOI] [PubMed] [Google Scholar]
  • 181.Sanborn RE, Ross HJ, Aung S, Acheson A, Moudgil T, Puri S, Hilton T, Fisher B, Coffey T, Paustian C, et al. A pilot study of an autologous tumor-derived autophagosome vaccine with docetaxel in patients with stage IV non-small cell lung cancer. J Immunother Cancer. 2017;5:103. doi: 10.1186/s40425-017-0306-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Shariat SF, Sadeghi F, Slawin KM. Vaccine-based immunotherapy for prostate cancer. Rev Urol. 2000;2:222–227. [PMC free article] [PubMed] [Google Scholar]
  • 183.Madan RA, Gulley JL, Arlen PM. PSA-based vaccines for the treatment of prostate cancer. Expert Rev Vaccines. 2006;5:199–209. doi: 10.1586/14760584.5.2.199. [DOI] [PubMed] [Google Scholar]
  • 184.Page DB, Hulett TW, Hilton TL, Hu HM, Urba WJ, Fox BA. Glimpse into the future: harnessing autophagy to promote anti-tumor immunity with the DRibbles vaccine. J Immunother Cancer. 2016;4:25. doi: 10.1186/s40425-016-0130-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Jagannath C, Bakhru P. Rapamycin-induced enhancement of vaccine efficacy in mice. Methods Mol Biol. 2012;821:295–303. doi: 10.1007/978-1-61779-430-8_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Inman BA, Sebo TJ, Frigola X, Dong H, Bergstralh EJ, Frank I, Fradet Y, Lacombe L, Kwon ED. PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer. 2007;109:1499–1505. doi: 10.1002/cncr.22588. [DOI] [PubMed] [Google Scholar]
  • 187.Bellmunt J, Powles T, Vogelzang NJ. A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: the future is now. Cancer Treat Rev. 2017;54:58–67. doi: 10.1016/j.ctrv.2017.01.007. [DOI] [PubMed] [Google Scholar]
  • 188.Klaassen Z, Kamat AM, Kassouf W, Gontero P, Villavicencio H, Bellmunt J, van Rhijn BWG, Hartmann A, Catto JWF, Kulkarni GS, et al. Treatment strategy for newly diagnosed T1 high-grade bladder urothelial carcinoma: new insights and updated recommendations. Eur Urol. 2018; doi: 10.1016/j.eururo.2018.06.024. [DOI] [PubMed] [Google Scholar]
  • 189.Startsev VY. The role of combined method in organ-sparing treatment of muscle-invasive bladder cancer recurrences. Arch Ital Urol Androl. 2002;74:54–56. [PubMed] [Google Scholar]
  • 190.Mehta K, Patel K, Parikh RA. Immunotherapy in genitourinary malignancies. J Hematol Oncol. 2017;10:95. doi: 10.1186/s13045-017-0457-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Siefker-Radtke A, Curti B. Immunotherapy in metastatic urothelial carcinoma: focus on immune checkpoint inhibition. Nat Rev Urol. 2018;15:112–124. doi: 10.1038/nrurol.2017.190. [DOI] [PubMed] [Google Scholar]
  • 192.Donin NM, Lenis AT, Holden S, Drakaki A, Pantuck A, Belldegrun A, Chamie K. Immunotherapy for the Treatment of Urothelial Carcinoma. J Urol. 2017;197:14–22. doi: 10.1016/j.juro.2016.02.3005. [DOI] [PubMed] [Google Scholar]
  • 193.Liu SV, Camidge DR, Gettinger SN, Giaccone G, Heist RS, Hodi FS, Ready NE, Zhang W, Wallin J, Funke R, et al. Long-term survival follow-up of atezolizumab in combination with platinum-based doublet chemotherapy in patients with advanced non-small-cell lung cancer. Eur J Cancer. 2018;101:114–122. doi: 10.1016/j.ejca.2018.06.033. [DOI] [PubMed] [Google Scholar]
  • 194.Horn L, Gettinger SN, Gordon MS, Herbst RS, Gandhi L, Felip E, Sequist LV, Spigel DR, Antonia SJ, Balmanoukian A, et al. Safety and clinical activity of atezolizumab monotherapy in metastatic non-small-cell lung cancer: final results from a phase I study. Eur J Cancer. 2018;101:201–209. doi: 10.1016/j.ejca.2018.06.031. [DOI] [PubMed] [Google Scholar]
  • 195.Gomes-Giacoia E, Miyake M, Goodison S, Sriharan A, Zhang G, You L, Egan JO, Rhode PR, Parker AS, Chai KX, et al. Intravesical ALT-803 and BCG treatment reduces tumor burden in a carcinogen induced bladder cancer rat model; a role for cytokine production and NK cell expansion. PLoS One. 2014;9:e96705. doi: 10.1371/journal.pone.0096705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Huang J, Schisler J, Wong HC, Rosser CJ, Sterbis J. Intravesical ALT-803 for BCG-unresponsive Bladder Cancer - A Case Report. Urol Case Rep. 2017;14:15–17. doi: 10.1016/j.eucr.2017.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Bidnur S, Savdie R, Black PC. Inhibiting immune checkpoints for the treatment of bladder cancer. Bladder Cancer. 2016;2:15–25. doi: 10.3233/BLC-150026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Davarpanah NN, Yuno A, Trepel JB, Apolo AB. Immunotherapy: a new treatment paradigm in bladder cancer. Curr Opin Oncol. 2017; doi: 10.1097/CCO.0000000000000366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Hakenberg OW. Nivolumab for the treatment of bladder cancer. Expert Opin Biol Ther. 2017;17:1309–1315. doi: 10.1080/14712598.2017.1353076. [DOI] [PubMed] [Google Scholar]
  • 200.Rentsch CA, Birkhauser FD, Biot C, Gsponer JR, Bisiaux A, Wetterauer C, Lagranderie M, Marchal G, Orgeur M, Bouchier C, et al. Bacillus Calmette-Guerin strain differences have an impact on clinical outcome in bladder cancer immunotherapy. Eur Urol. 2014;66:677–688. doi: 10.1016/j.eururo.2014.02.061. [DOI] [PubMed] [Google Scholar]
  • 201.Boehm BE, Cornell JE, Wang H, Mukherjee N, Oppenheimer JS, Svatek RS. Efficacy of bacillus calmette-guerin strains for treatment of nonmuscle invasive bladder cancer: a systematic review and network meta-analysis. J Urol. 2017;198:503–510. doi: 10.1016/j.juro.2017.01.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Carmignani L, Bianchi R, Cozzi G, Grasso A, Macchione N, Marenghi C, Melegari S, Rosso M, Tondelli E, Maggioni A. Intracavitary immunotherapy and chemotherapy for upper urinary tract cancer: current evidence. Rev Urol. 2013;15:145–153. [PMC free article] [PubMed] [Google Scholar]
  • 203.Rijnders M, de Wit R, Boormans JL, Lolkema MPJ, van der Veldt AAM. Systematic review of immune checkpoint inhibition in urological cancers. Eur Urol. 2017;72:411–423. doi: 10.1016/j.eururo.2017.06.012. [DOI] [PubMed] [Google Scholar]
  • 204.Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, Gailani F, Riley L, Conlon K, Pockaj B, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–2127. doi: 10.1056/NEJMoa1012863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Galvez-Cancino F, Lopez E, Menares E, Diaz X, Flores C, Caceres P, Hidalgo S, Chovar O, Alcántara-Hernández M, Borgna V, et al. Vaccination-induced skin-resident memory CD8(+) T cells mediate strong protection against cutaneous melanoma. Oncoimmunology. 2018;7:e1442163. doi: 10.1080/2162402X.2018.1442163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Khong H, Overwijk WW. Adjuvants for peptide-based cancer vaccines. J Immunother Cancer. 2016;4:56. doi: 10.1186/s40425-016-0160-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kang J, Galluzzi L. PD-L1 blockade for urothelial carcinoma. Oncoimmunology. 2017;6:e1334028. doi: 10.1080/2162402X.2017.1334028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Ehlen TG, Hoskins PJ, Miller D, Whiteside TL, Nicodemus CF, Schultes BC, Swenerton KD. A pilot phase 2 study of oregovomab murine monoclonal antibody to CA125 as an immunotherapeutic agent for recurrent ovarian cancer. Int J Gynecol Cancer. 2005;15:1023–1034. doi: 10.1111/j.1525-1438.2005.00483.x. [DOI] [PubMed] [Google Scholar]
  • 209.Crosby EJ, Wei J, Yang XY, Lei G, Wang T, Liu CX, Agarwal P, Korman AJ, Morse MA, Gouin K, et al. Complimentary mechanisms of dual checkpoint blockade expand unique T-cell repertoires and activate adaptive anti-tumor immunity in triple-negative breast tumors. Oncoimmunology. 2018;7:e1421891. doi: 10.1080/2162402X.2017.1421891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Knepper TC, Saller J, Walko CM. Novel and expanded oncology drug approvals of 2016-PART 1: new options in solid tumor management. Oncology (Williston Park). 2017;31:110–121. [PubMed] [Google Scholar]
  • 211.Zhu X, Lang J. Programmed death-1 pathway blockade produces a synergistic antitumor effect: combined application in ovarian cancer. J Gynecol Oncol. 2017;28:e64. doi: 10.3802/jgo.2017.28.e64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Aldous AR, Dong JZ. Personalized neoantigen vaccines: A new approach to cancer immunotherapy. Bioorg Med Chem. 2018;26:2842–2849. doi: 10.1016/j.bmc.2017.10.021. [DOI] [PubMed] [Google Scholar]
  • 213.Rubinsteyn A, Kodysh J, Hodes I, Mondet S, Aksoy BA, Finnigan JP, Bhardwaj N, Hammerbacher J. Computational Pipeline for the PGV-001 Neoantigen Vaccine Trial. Front Immunol. 2017;8:1807. doi: 10.3389/fimmu.2017.01807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Grenier JM, Yeung ST, Khanna KM. Combination immunotherapy: taking cancer vaccines to the next level. Front Immunol. 2018;9:610. doi: 10.3389/fimmu.2018.00610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Buque A, Bloy N, Aranda F, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Spisek R, Tartour E, et al. Trial Watch-Small molecules targeting the immunological tumor microenvironment for cancer therapy. Oncoimmunology. 2016;5:e1149674. doi: 10.1080/2162402X.2016.1149674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov. 2016;15:235–247. doi: 10.1038/nrd.2015.35. [DOI] [PubMed] [Google Scholar]
  • 217.Adams JL, Smothers J, Srinivasan R, Hoos A. Big opportunities for small molecules in immuno-oncology. Nat Rev Drug Discov. 2015;14:603–622. doi: 10.1038/nrd4596. [DOI] [PubMed] [Google Scholar]
  • 218.Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Xu YY, Kalos M, Cai L, Fang H-B, Weiss BM, Badros A, et al. Combination immunotherapy after ASCT for multiple myeloma using MAGE-A3/Poly-ICLC immunizations followed by adoptive transfer of vaccine-primed and costimulated autologous T cells. Clin Cancer Res. 2014;20:1355–1365. doi: 10.1158/1078-0432.CCR-13-2817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Dutoit V, Migliorini D, Ranzanici G, Marinari E, Widmer V, Lobrinus JA, Momjian S, Costello J, Walker PR, Okada H, et al. Antigenic expression and spontaneous immune responses support the use of a selected peptide set from the IMA950 glioblastoma vaccine for immunotherapy of grade II and III glioma. Oncoimmunology. 2018;7:e1391972. doi: 10.1080/2162402X.2017.1391972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Rampling R, Peoples S, Mulholland PJ, James A, Al-Salihi O, Twelves CJ, McBain C, Jefferies S, Jackson A, Stewart W, et al. A cancer research UK first time in human phase I trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in patients with newly diagnosed glioblastoma. Clin Cancer Res. 2016;22:4776–4785. doi: 10.1158/1078-0432.CCR-16-0506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.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;135:1042–1054. doi: 10.1093/brain/aws042. [DOI] [PubMed] [Google Scholar]
  • 222.Gnjatic S, Sawhney NB, Bhardwaj N. Toll-like receptor agonists: are they good adjuvants? Cancer J. 2010;16:382–391. doi: 10.1097/PPO.0b013e3181eaca65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Scutti JAB. Importance of immune monitoring approaches and the use of immune checkpoints for the treatment of diffuse intrinsic pontine glioma: from bench to clinic and vice versa (Review). Int J Oncol. 2018;52:1041–1056. doi: 10.3892/ijo.2018.4283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Crittenden M, Kohrt H, Levy R, Jones J, Camphausen K, Dicker A, Demaria S, Formenti S. Current clinical trials testing combinations of immunotherapy and radiation. Semin Radiat Oncol. 2015;25:54–64. doi: 10.1016/j.semradonc.2014.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Linch SN, McNamara MJ, Redmond WL. OX40 agonists and combination immunotherapy: putting the pedal to the metal. Front Oncol. 2015;5:34. doi: 10.3389/fonc.2015.00034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Sato-Kaneko F, Yao S, Ahmadi A, Zhang SS, Hosoya T, Kaneda MM, Varner JA, Pu M, Messer KS, Guiducci C, et al. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight. 2017;2:e93397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Wu X, Giobbie-Hurder A, Connolly EM, Li J, Liao X, Severgnini M, Zhou J, Rodig S, Hodi FS. Anti-CTLA-4 based therapy elicits humoral immunity to galectin-3 in patients with metastatic melanoma. Oncoimmunology. 2018;7:e1440930. doi: 10.1080/2162402X.2018.1440930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, et al. Trial watch: immune checkpoint blockers for cancer therapy. Oncoimmunology. 2017;6:e1373237. doi: 10.1080/2162402X.2017.1373237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, Kim YH, Hoppe RT, Knox SJ, Shin LK, et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol. 2010;28:4324–4332. doi: 10.1200/JCO.2010.28.9793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Sagiv-Barfi I, Kohrt HE, Burckhardt L, Czerwinski DK, Levy R. Ibrutinib enhances the antitumor immune response induced by intratumoral injection of a TLR9 ligand in mouse lymphoma. Blood. 2015;125:2079–2086. doi: 10.1182/blood-2014-08-593137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Ito H, Ando T, Arioka Y, Saito K, Seishima M. Inhibition of indoleamine 2,3-dioxygenase activity enhances the anti-tumour effects of a Toll-like receptor 7 agonist in an established cancer model. Immunology. 2015;144:621–630. doi: 10.1111/imm.12413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Brochez L, Chevolet I, Kruse V. The rationale of indoleamine 2,3-dioxygenase inhibition for cancer therapy. Eur J Cancer. 2017;76:167–182. doi: 10.1016/j.ejca.2017.01.011. [DOI] [PubMed] [Google Scholar]
  • 233.Kim YH, Gratzinger D, Harrison C, Brody JD, Czerwinski DK, Ai WZ, Morales A, Abdulla F, Xing L, Navi D, et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study. Blood. 2012;119:355–363. doi: 10.1182/blood-2011-05-355222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Stone L. Pembrolizumab effective in PD-L1-positive disease. Nat Rev Urol. 2018; doi: 10.1038/s41585-018-0070-5. [DOI] [PubMed] [Google Scholar]
  • 235.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautes-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]
  • 236.Vacchelli E, Eggermont A, Sautes-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]
  • 237.Helminen O, Huhta H, Lehenkari PP, Saarnio J, Karttunen TJ, Kauppila JH. Nucleic acid-sensing toll-like receptors 3, 7 and 8 in esophageal epithelium, barrett’s esophagus, dysplasia and adenocarcinoma. Oncoimmunology. 2016;5:e1127495. doi: 10.1080/2162402X.2015.1127495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013;342:1432–1433. doi: 10.1126/science.342.6165.1432. [DOI] [PubMed] [Google Scholar]
  • 239.Sadelain M, Riviere I, Riddell S. Therapeutic T cell engineering. Nature. 2017;545:423–431. doi: 10.1038/nature22395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–1365. doi: 10.1126/science.aar6711. [DOI] [PubMed] [Google Scholar]
  • 241.Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372:2521–2532. doi: 10.1056/NEJMoa1503093. [DOI] [PubMed] [Google Scholar]
  • 242.Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082. [DOI] [PubMed] [Google Scholar]
  • 243.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon R-A, Reed K, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–133. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Poole RM. Pembrolizumab: first global approval. Drugs. 2014;74:1973–1981. doi: 10.1007/s40265-014-0314-5. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES