Coordinated Activation of Toll-Like Receptor8 (TLR8) and NLRP3 by the TLR8 Agonist, VTX-2337, Ignites Tumoricidal Natural Killer Cell Activity

Gregory N Dietsch, Hailing Lu, Yi Yang, Chihiro Morishima, Laura Q Chow, Mary L Disis, Robert M Hershberg, Gregory N Dietsch, Hailing Lu, Yi Yang, Chihiro Morishima, Laura Q Chow, Mary L Disis, Robert M Hershberg

Abstract

VTX-2337 (USAN: motolimod) is a selective toll-like receptor 8 (TLR8) agonist, which is in clinical development as an immunotherapy for multiple oncology indications, including squamous cell carcinoma of the head and neck (SCCHN). Activation of TLR8 enhances natural killer cell activation, increases antibody-dependent cell-mediated cytotoxicity, and induces Th1 polarizing cytokines. Here, we show that VTX-2337 stimulates the release of mature IL-1β and IL-18 from monocytic cells through coordinated actions on both TLR8 and the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome complex. In vitro, VTX-2337 primed monocytic cells to produce pro-IL-1β, pro-IL-18, and caspase-1, and also activated the NLRP3 inflammasome, thereby mediating the release of mature IL-1β family cytokines. Inhibition of caspase-1 blocked VTX-2337-mediated NLRP3 inflammasome activation, but had little impact on production of other TLR8-induced mediators such as TNFα. IL-18 activated natural killer cells and complemented other stimulatory pathways, including FcγRIII and NKG2D, resulting in IFNγ production and expression of CD107a. NLRP3 activation in vivo was confirmed by a dose-related increase in plasma IL-1β and IL-18 levels in cynomolgus monkeys administered VTX-2337. These results are highly relevant to clinical studies of combination VTX-2337/cetuximab treatment. Cetuximab, a clinically approved, epidermal growth factor receptor-specific monoclonal antibody, activates NK cells through interactions with FcγRIII and facilitates ADCC of tumor cells. Our preliminary findings from a Phase I open-label, dose-escalation, trial that enrolled 13 patients with recurrent or metastatic SCCHN show that patient NK cells become more responsive to stimulation by NKG2D or FcγRIII following VTX-2337 treatment. Together, these results indicate that TLR8 stimulation and inflammasome activation by VTX-2337 can complement FcγRIII engagement and may augment clinical responses in SCCHN patients treated with cetuximab.

Trial registration: ClinicalTrials.gov NCT01334177.

Conflict of interest statement

Competing Interests: Gregory N. Dietsch and Robert M. Hershberg are both employees and have ownership (shares) in VentiRx Pharmaceuticals, Seattle, WA, United States of America. Gregory N. Dietsch and Robert M. Hershberg have Patent applications (pending and actual) to which the authors are affiliated and from which the authors may benefit. Robert M. Hershberg is a Board Member of VentiRx Pharmaceuticals. Mary L. Disis is a paid consultant for VentiRx Pharmaceuticals, Seattle, WA, United States of America. The association of Gregory N. Dietsch, Robert M. Hershberg and Mary L. Disis with VentiRx Pharmaceuticals does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. CONSORT Flow Diagram for Phase…
Fig 1. CONSORT Flow Diagram for Phase 1 clinical study in adult patients with advanced recurrent squamous cell carcinomas of the head and neck (SCCHN).
Study subjects received VTX-2337 administered by the SC route on days 1, 8 and 15 of a 28-day treatment cycle. The first cohort received a 2.5 mg/m2 dose of VTX-2337 following cetuximab administration; this dose was escalated in subsequent cohorts using a 3+3 design to 3.0 mg/m2 and finally 3.5 mg/m2.
Fig 2. VTX-2337 induces mature IL-1β and…
Fig 2. VTX-2337 induces mature IL-1β and IL-18 via NLRP3 inflammasome activation in THP-1 cells.
(A-B) VTX-2337 induced pro-IL-1β and pro-IL-18 mRNA expression in THP-1 cells. Cells were cultured overnight in medium or VTX-2337 (3 or 10 μM), and expression of IL-1β and IL-18 mRNA, normalized to β-actin levels, were measured by real-time RT-PCR. (C) Western blot analysis of IL-1β and caspase-1 protein levels in THP-1 culture supernatants, and pro-IL-1β protein levels in cell lysates, following overnight activation with medium (Ø), or VTX-2337 (3 or 10 μM). (D) Pretreatment with caspase-1 inhibitor z-VAD blocks VTX-2337-induced secretion of mature IL-1β. Triplicate THP-1 cultures (10 μg/ml) were treated with VTX-2337. Levels of IL-1β in the presence or absence of z-VAD-FMK were assessed using HEK-Blue™ IL-1β cells (mean±sem). (E and F) Levels of secreted IL-1β and IL-18 following overnight VTX-2337 stimulation (0.03 to 25 μM in 1:3 serial dilutions) in wild type THP-1 (●) and NLRP3 deficient THP-1 (THP-1/NLRP3def) cells (○). Levels of IL-1β and IL-18 (mean±sem) in culture supernatant from triplicate wells were measured by ELISA. All results shown are representative of three independent experiments. **, p<0.01, ***, p<0.001 by unpaired Student t-test.
Fig 3. VTX-2337 induces IL-1β and IL-18…
Fig 3. VTX-2337 induces IL-1β and IL-18 in human PBMC.
(A-B) IL-1β and IL-18 secretion from VTX-2337 activated PBMC. Dose responses are shown for one representative donor, where data point represents the mean for duplicate culture wells at the indicated concentration of VTX-2337 after 24 h. (C-D) Summary graphs of VTX-2337-induced IL-1β and IL-18 in whole blood from 11 donors, as analyzed by TruCulture. Whole blood was incubated with medium (Ø) or VTX-2337 (0.3 or 1 μM) for 24 h, levels of secreted IL-1β and IL-18 were analyzed in a multiplexed assay. Each symbol represents data from an individual donor. The horizontal bar represents group average. **, p<0.01; ***, p<0.001 by Wilcoxon signed-rank test.
Fig 4. Caspase-1 activation and IL-18 induction…
Fig 4. Caspase-1 activation and IL-18 induction by VTX-2337 leads to NK cell activation.
(A-D) The effects of caspase-1 inhibition on IL-1β, IL-18, IFNγ, and TNFα production by PBMC. Duplicate PBMC cultures from four healthy donors (indicated as D1, D2, D3 and D4) were treated overnight with medium (Ø), VTX-2337 (1 μM), or VTX-2337 plus z-VAD-FMK (10 μg/ml). Levels of IL-1β, IL-18, IFNγ, and TNFα in culture supernatant were measured by ELISA. (E-F) The effects of IL-18 blockade on VTX-2337-induced IFNγ and CD107a expression in NK cells. Summary graphs show the percentage of IFNγ+ and CD107a+ NK cells from PBMC cultured with medium (Ø), VTX-2337 (0.5 μM), or VTX-2337 and anti-IL18 neutralizing antibody (20 μg/ml) for 9 donors where each symbol represents one individual donor. The horizontal bar represents group the mean for the treatment. Data shown are the summary of four independent experiments from the 9 donors, *, p<0.05 by Wilcoxon signed-rank test.
Fig 5. VTX-2337 enhances NK activation in…
Fig 5. VTX-2337 enhances NK activation in response to stimulation by K562 cells and anti-CD16 in vitro.
(A) FACS plots showing IFNγ and CD107a expression by NK cells following overnight cultured in medium or VTX-2337 (0.5 μM), and then left unstimulated, stimulated with either K562 cells, or plate-bound anti-CD16 mAb. (B-C) Summary graphs of FACS analyses of NK cell activation in PBMC from 11 donors. Each data point represents an individual donor. The horizontal bar represents group average. Each donor was analyzed in an independent experiment. *, p<0.05, **, p<0.01, ***, p<0.001 by Wilcoxon signed-rank test.
Fig 6. VTX-2337 administration to cynomolgus monkeys…
Fig 6. VTX-2337 administration to cynomolgus monkeys induces IL-1β and IL-18.
Male cynomolgus monkeys (n = 6 per group) received a subcutaneously injection of VTX-2337 (1 or 10 mg/kg). Plasma collected predose, and 6, 12, 24 and 96 h following dosing was assessed for levels of IL-1β, IL-18 and IL-6. Cytokine levels in pg/mL (mean±sem) at each time point for the two VTX-2337 dose levels are shown. At each VTX-2337 dose level, plasma cytokine levels at 6, 12, 24 and 96 h were compared to predose levels *, p<0.05, **, p<0.01, by t-test.
Fig 7. Administration of VTX-2337 to SCCHN…
Fig 7. Administration of VTX-2337 to SCCHN cancer patients enhances NK cell degranulation.
Representative FACS plots from two patients illustrate NK cell activation measured by increased CD107a expression 24 h following the administration of VTX-2337 (3.0 mg/m2) compared to pre-treatment. Freshly isolated PBMC from each patient were unstimulated (negative control), or were stimulated with K562-15mb-41BBL cells or plate-bound anti-CD16 mAb.

References

    1. Diefenbach A, Raulet DH. The innate immune response to tumors and its role in the induction of T-cell immunity. Immunol Rev. 2002;188: 9–21.
    1. Kaifu T, Escaliere B, Gastinel LN, Vivier E, Baratin M. B7-H6/NKp30 interaction: a mechanism of alerting NK cells against tumors. Cell Mol Life Sci. 2011;68: 3531–9. 10.1007/s00018-011-0802-7
    1. Lu H, Dietsch GN, Matthews MA, Yang Y, Ghanekar S, Inokuma M, et al. VTX-2337 is a novel TLR8 agonist that activates NK cells and augments ADCC. Clin Cancer Res. 2012;18: 499–509. 10.1158/1078-0432.CCR-11-1625
    1. Chen X, Trivedi PP, Ge B, Krzewski K, and Strominger JL. Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity. PNAS. 2007;104: 6329–34.
    1. López-Albaitero A, Lee SC, Morgan S, Grandis JR, Gooding WE, Ferrone S, Ferris RL. Role of polymorphic Fc gamma receptor IIIa and EGFR expression level in cetuximab mediated, NK cell dependent in vitro cytotoxicity of head and neck squamous cell carcinoma cells. Cancer Immunol Immunother. 2009;58(11): 1853–64. 10.1007/s00262-009-0697-4
    1. Srivastava RM, Lee SC, Andrade Filho PA, Lord CA, Jie HB, Davidson HC, et al. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res. 2013;19: 1858–72. 10.1158/1078-0432.CCR-12-2426
    1. Zhou Z, Zhang C, Zhang J, Tian Z. Macrophages help NK cells to attack tumor cells by stimulatory NKG2D ligand but protect themselves from NK killing by inhibitory ligand Qa-1. PLoSOne. 2012;7(5): e36928.
    1. Srivastava S, Pelloso D, Feng H, Voiles L, Lewis D, Haskova Z, Whitacre M, Trulli S, Chen Y, Toso J, Jonak Z, Chang H, Robertson M. Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin. Cancer Immunol Immunother.2013;62: 1073–1082. 10.1007/s00262-013-1403-0
    1. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9: 847–56. 10.1038/ni.1631
    1. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440: 237–41.
    1. Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009;10: 241–7. 10.1038/ni.1703
    1. Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007;7: 31–40.
    1. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15: 1170–8. 10.1038/nm.2028
    1. Lu H, Knutson KL, Gad E, Disis ML. The tumor antigen repertoire identified in tumor-bearing Neu transgenic mice predicts human tumor antigens. Cancer Res. 2006;66: 9754–61.
    1. Mueller SC, Marz R, Schmolz M, Drewelow B. Intraindividual long term stability and response corridors of cytokines in healthy volunteers detected by a standardized whole-blood culture system for bed-side application. BMC Med Res Methodol. 2012;12: 112 10.1186/1471-2288-12-112
    1. Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106: 376–83.
    1. Alter G, Malenfant JM, Altfeld M. CD107a as a functional marker for the identification of natural killer cell activity. J of Immunol Methods. 2004;294: 15–22.
    1. Liu J, Xu C, Hsu LC, Luo Y, Xiang R, Chuang TH. A five-amino-acid motif in the undefined region of the TLR8 ectodomain is required for species-specific ligand recognition. Mol Immunol. 2010;47: 1083–1090. 10.1016/j.molimm.2009.11.003
    1. Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012;13: 333–2. 10.1038/ni.2237
    1. Stephenson RM, Lim CM, Matthews M, Dietsch G, Hershberg R, Ferris RL. TLR8 stimulation enhances cetuximab-mediated natural killer cell lysis of head and neck cancer cells and dendritic cell cross-priming of EGFR-specific CD8 T cells. Cancer Immunol Immunother. 2013;62: 1347–57. 10.1007/s00262-013-1437-3
    1. Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183: 787–91. 10.4049/jimmunol.0901363
    1. Bruchard M, Mignot G, Derangere V, Chalmin F, Chevriaux A, Vegran F, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19: 57–64. 10.1038/nm.2999
    1. Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, Castellano R, et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 2011;121: 3609–22. 10.1172/JCI45816
    1. Schilling B, Halstead ES, Schuler P, Harasymczuk M, Egan JE, Whiteside TL. IRX-2, a novel immunotherapeutic, enhances and protects NK-cell functions in cancer patients. Cancer Immunol Immunother. 2012; 61: 1395–1405 10.1007/s00262-011-1197-x
    1. Stoll S, Jonuleit H, Schmitt E, Müller G, Yamauchi H, Kurimoto M, et al. Production of functional IL-18 by different subtypes of murine and human dendritic cells (DC): DC-derived IL-18 enhances IL-12-dependent Th1 development. Eur J Immunol. 1998;28: 3231–9.
    1. Spranger S, Javorovic M, Bürdek M, Wilde S, Mosetter B, Tippmer S, et al. Generation of Th1-polarizing dendritic cells using the TLR7/8 agonist CL075. J Immunol. 2010;185: 738–47. 10.4049/jimmunol.1000060
    1. Pufnock JS, Cigal M, Rolczynski LS, Andersen-Nissen E, Wolfl M, McElrath MJ, et al. Priming CD8+ T cells with dendritic cells matured using TLR4 and TLR7/8 ligands together enhances generation of CD8 + T cells retaining CD28. Blood. 2011;117: 6542–51. 10.1182/blood-2010-11-317966
    1. Roda JM, Parihar R, Carson WE 3rd. CpG-containing oligodeoxynucleotides act through TLR9 to enhance the NK cell cytokine response to antibody-coated tumor cells. J Immunol. 2005;175: 1619–27.
    1. 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: 1636–42.
    1. Wysocka M, Dawany N, Benoit B, Kossenkov AV, Troxel AB, Gelfand JM, et al. Synergistic enhancement of cellular immune responses by the novel Toll receptor 7/8 agonist 3M-007 and interferon-gamma: implications for therapy of cutaneous T-cell lymphoma. Leuk Lymphoma. 2011;52: 1970–9. 10.3109/10428194.2011.582202

Source: PubMed

Подписаться