Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 6/2013

01-06-2013 | Original Article

Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin

Authors: Shivani Srivastava, David Pelloso, Hailin Feng, Larry Voiles, David Lewis, Zdenka Haskova, Margaret Whitacre, Stephen Trulli, Yi-Jiun Chen, John Toso, Zdenka L. Jonak, Hua-Chen Chang, Michael J. Robertson

Published in: Cancer Immunology, Immunotherapy | Issue 6/2013

Login to get access

Abstract

The antitumor activity of monoclonal antibodies is mediated by effector cells, such as natural killer (NK) cells, that express Fc receptors for immunoglobulin. Efficacy of monoclonal antibodies, including the CD20 antibody rituximab, could be improved by agents that augment the function of NK cells. Interleukin (IL)-18 is an immunostimulatory cytokine that has antitumor activity in preclinical models. The effects of IL-18 on NK cell function mediated through Fcγ receptors were examined. Human NK cells stimulated with immobilized IgG in vitro secreted IFN-γ as expected; such IFN-γ production was partially inhibited by blocking CD16 with monoclonal antibodies. IL-18 augmented IFN-γ production by NK cells stimulated with immobilized IgG or CD16 antibodies. NK cell IFN-γ production in response to immobilized IgG and/or IL-18 was inhibited by chemical inhibitors of Syk and several other kinases involved in CD16 signaling pathways. IL-18 augmented antibody-dependent cellular cytotoxicity (ADCC) of human NK cells against rituximab-coated Raji cells in vitro. IL-18 and rituximab acted synergistically to promote regression of human lymphoma xenografts in SCID mice. Inasmuch as IL-18 costimulates IFN-γ production and ADCC of NK cells activated through Fc receptors in vitro and augments antitumor activity of rituximab in vivo, it is an attractive cytokine to combine with monoclonal antibodies for treatment of human cancer.
Literature
2.
go back to reference Robertson MJ, Ritz J (1990) Biology and clinical relevance of human natural killer cells. Blood 76:2421–2438PubMed Robertson MJ, Ritz J (1990) Biology and clinical relevance of human natural killer cells. Blood 76:2421–2438PubMed
3.
go back to reference Leibson P (1997) Signal transduction during natural killer cell activation: inside the mind of a killer. Immunity 6:655–661PubMedCrossRef Leibson P (1997) Signal transduction during natural killer cell activation: inside the mind of a killer. Immunity 6:655–661PubMedCrossRef
4.
go back to reference Trotta R, Fettucciari K, Azzoni L, Abebe B, Puorro KA, Eisenlohr LC, Perussia B (2000) Differential role of p38 and c-Jun N-terminal kinase 1 mitogen-activated protein kinases in NK cell cytotoxicity. J Immunol 165:1782–1789PubMed Trotta R, Fettucciari K, Azzoni L, Abebe B, Puorro KA, Eisenlohr LC, Perussia B (2000) Differential role of p38 and c-Jun N-terminal kinase 1 mitogen-activated protein kinases in NK cell cytotoxicity. J Immunol 165:1782–1789PubMed
5.
go back to reference Roda JM, Parihar R, Magro C, Nuovo GJ, Tridandapani S, Carson WE (2006) Natural killer cells produce T cell-recruiting chemokines in response to antibody-coated target cells. Cancer Res 66(1):517–526PubMedCrossRef Roda JM, Parihar R, Magro C, Nuovo GJ, Tridandapani S, Carson WE (2006) Natural killer cells produce T cell-recruiting chemokines in response to antibody-coated target cells. Cancer Res 66(1):517–526PubMedCrossRef
6.
go back to reference Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, Watier H (2002) Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood 99:754–758PubMedCrossRef Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, Watier H (2002) Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood 99:754–758PubMedCrossRef
7.
go back to reference Musolino A, Naldi N, Bortesi B, Pezzuolo D, Capelletti M, Missale G, Laccabue D, Zerbini A, Camisa R, Bisagni G, Neri TM, Ardizzoni A (2008) Immunoglobulin G fragment c receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol 26(11):1789–1796PubMedCrossRef Musolino A, Naldi N, Bortesi B, Pezzuolo D, Capelletti M, Missale G, Laccabue D, Zerbini A, Camisa R, Bisagni G, Neri TM, Ardizzoni A (2008) Immunoglobulin G fragment c receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol 26(11):1789–1796PubMedCrossRef
8.
go back to reference Bibeau F, Lopez-Crapez E, Di Fiore F, Thezenas S, Ychou M, Blanchard F, Lamy A, Penault-Llorca F, Frebourg T, Michel P, Sabourin J-C, Boissiere-Michot F (2009) Impact of FcγRIIa-FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol 27(7):1122–1129PubMedCrossRef Bibeau F, Lopez-Crapez E, Di Fiore F, Thezenas S, Ychou M, Blanchard F, Lamy A, Penault-Llorca F, Frebourg T, Michel P, Sabourin J-C, Boissiere-Michot F (2009) Impact of FcγRIIa-FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol 27(7):1122–1129PubMedCrossRef
9.
go back to reference Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H (2001) Interleukin-18 regulates both Th1 and Th2 responses. Ann Rev Immunol 19:423–474CrossRef Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H (2001) Interleukin-18 regulates both Th1 and Th2 responses. Ann Rev Immunol 19:423–474CrossRef
10.
go back to reference Osaki T, Peron J-M, Cai Q, Okamura H, Robbins PD, Kurimoto M, Lotze MT, Tahara H (1998) IFN-γ-inducing factor/IL-18 administration mediates IFN-γ- and IL-12-independent antitumor effects. J Immunol 160:1742–1749PubMed Osaki T, Peron J-M, Cai Q, Okamura H, Robbins PD, Kurimoto M, Lotze MT, Tahara H (1998) IFN-γ-inducing factor/IL-18 administration mediates IFN-γ- and IL-12-independent antitumor effects. J Immunol 160:1742–1749PubMed
11.
go back to reference Hashimoto W, Osaki T, Okamura H, Robbins PD, Kurimoto M, Nagata S, Peron J-M, Lotze MT, Tahara H (1999) Differential antitumor effects of administration of recombinant IL-18 or recombinant IL-12 are mediated primarily by Fas–Fas ligand- and perforin-induced tumor apoptosis, respectively. J Immunol 163:583–589PubMed Hashimoto W, Osaki T, Okamura H, Robbins PD, Kurimoto M, Nagata S, Peron J-M, Lotze MT, Tahara H (1999) Differential antitumor effects of administration of recombinant IL-18 or recombinant IL-12 are mediated primarily by Fas–Fas ligand- and perforin-induced tumor apoptosis, respectively. J Immunol 163:583–589PubMed
12.
go back to reference Robertson MJ, Mier JW, Logan T, Atkins M, Koon H, Koch KM, Kathman S, Pandite LN, Oei C, Kirby LC, Jewell RC, Bell WN, Thurmond LM, Weisenbach J, Roberts S, Dar MM (2006) Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer. Clin Cancer Res 12(14):4265–4273PubMedCrossRef Robertson MJ, Mier JW, Logan T, Atkins M, Koon H, Koch KM, Kathman S, Pandite LN, Oei C, Kirby LC, Jewell RC, Bell WN, Thurmond LM, Weisenbach J, Roberts S, Dar MM (2006) Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer. Clin Cancer Res 12(14):4265–4273PubMedCrossRef
13.
go back to reference Robertson MJ, Kirkwood JM, Logan TF, Koch KM, Kathman S, Kirby LC, Bell WN, Thurmond LM, Weisenbach J, Dar MM (2008) A dose-escalation study of recombinant human interleukin-18 using two different schedules of administration in patients with cancer. Clin Cancer Res 14(11):3462–3469PubMedCrossRef Robertson MJ, Kirkwood JM, Logan TF, Koch KM, Kathman S, Kirby LC, Bell WN, Thurmond LM, Weisenbach J, Dar MM (2008) A dose-escalation study of recombinant human interleukin-18 using two different schedules of administration in patients with cancer. Clin Cancer Res 14(11):3462–3469PubMedCrossRef
14.
go back to reference Robertson MJ, Abonour R, Hromas R, Nelson RP, Fineberg NS, Cornetta K (2005) Augmented high-dose regimen of cyclophosphamide, carmustine, and etoposide with autologous hematopoietic stem cell transplantation for relapsed and refractory aggressive non-Hodgkin’s lymphoma. Leuk Lymphoma 46:1477–1487PubMedCrossRef Robertson MJ, Abonour R, Hromas R, Nelson RP, Fineberg NS, Cornetta K (2005) Augmented high-dose regimen of cyclophosphamide, carmustine, and etoposide with autologous hematopoietic stem cell transplantation for relapsed and refractory aggressive non-Hodgkin’s lymphoma. Leuk Lymphoma 46:1477–1487PubMedCrossRef
15.
go back to reference Robertson MJ, Caligiuri MA, Manley TJ, Levine H, Ritz J (1990) Human natural killer cell adhesion molecules: differential expression after activation and participation in cytolysis. J Immunol 145:3194–3201PubMed Robertson MJ, Caligiuri MA, Manley TJ, Levine H, Ritz J (1990) Human natural killer cell adhesion molecules: differential expression after activation and participation in cytolysis. J Immunol 145:3194–3201PubMed
16.
go back to reference Chang H-C, Han L, Goswami R, Nguyen ET, Pelloso D, Robertson MJ, Kaplan MH (2009) Impaired development of human Th1 cells in patients with deficient expression of STAT4. Blood 113(23):5887–5890PubMedCrossRef Chang H-C, Han L, Goswami R, Nguyen ET, Pelloso D, Robertson MJ, Kaplan MH (2009) Impaired development of human Th1 cells in patients with deficient expression of STAT4. Blood 113(23):5887–5890PubMedCrossRef
17.
go back to reference Parihar R, Dierksheide J, Hu Y, Carson WE (2002) IL-12 enhances the natural killer cell cytokine response to Ab-coated target cells. J Clin Invest 110(7):983–992PubMed Parihar R, Dierksheide J, Hu Y, Carson WE (2002) IL-12 enhances the natural killer cell cytokine response to Ab-coated target cells. J Clin Invest 110(7):983–992PubMed
18.
go back to reference Roda JM, Parihar R, Lehman A, Mani A, Tridandapani S, Carson WE (2006) Interleukin-21 enhances NK cell activation in response to antibody-coated targets. J Immunol 177:120–129PubMed Roda JM, Parihar R, Lehman A, Mani A, Tridandapani S, Carson WE (2006) Interleukin-21 enhances NK cell activation in response to antibody-coated targets. J Immunol 177:120–129PubMed
19.
go back to reference Fehniger TA, Shah MH, Turner MJ, VanDeusen JB, Whitman SP, Cooper MA, Suzuki K, Wechser M, Goodsaid F, Caligiuri MA (1999) Differential cytokine and chemokine gene expression by human NK cells following activation with IL-18 or IL-15 in combination with IL-12: implications for the innate immune response. J Immunol 162:4511–4520PubMed Fehniger TA, Shah MH, Turner MJ, VanDeusen JB, Whitman SP, Cooper MA, Suzuki K, Wechser M, Goodsaid F, Caligiuri MA (1999) Differential cytokine and chemokine gene expression by human NK cells following activation with IL-18 or IL-15 in combination with IL-12: implications for the innate immune response. J Immunol 162:4511–4520PubMed
20.
go back to reference Robertson MJ, Chang H-C, Pelloso D, Kaplan MH (2005) Impaired interferon-γ production as a consequence of STAT4 deficiency after autologous hematopoietic stem cell transplantation for lymphoma. Blood 106:963–970PubMedCrossRef Robertson MJ, Chang H-C, Pelloso D, Kaplan MH (2005) Impaired interferon-γ production as a consequence of STAT4 deficiency after autologous hematopoietic stem cell transplantation for lymphoma. Blood 106:963–970PubMedCrossRef
21.
go back to reference Robertson MJ, Pelloso D, Abonour R, Hromas RA, Nelson RP Jr, Wood L, Cornetta K (2002) Interleukin-12 immunotherapy after autologous stem cell transplantation for hematologic malignancies. Clin Cancer Res 8:3383–3393PubMed Robertson MJ, Pelloso D, Abonour R, Hromas RA, Nelson RP Jr, Wood L, Cornetta K (2002) Interleukin-12 immunotherapy after autologous stem cell transplantation for hematologic malignancies. Clin Cancer Res 8:3383–3393PubMed
22.
go back to reference Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard J, Wei M, Mao H, Byrd JC, Perrotti D, Caligiuri MA (2008) TGF-β utilizes SMAD3 to inhibit CD16-mediated IFN-γ production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181:3784–3792PubMed Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard J, Wei M, Mao H, Byrd JC, Perrotti D, Caligiuri MA (2008) TGF-β utilizes SMAD3 to inhibit CD16-mediated IFN-γ production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181:3784–3792PubMed
23.
go back to reference Perussia B, Trinchieri G, Jackson A, Warner NL, Faust J, Rumpold H, Kraft D, Lanier LL (1984) The Fc receptor for IgG on human natural killer cells: phenotypic, functional, and comparative studies with monoclonal antibodies. J Immunol 133(1):180–189PubMed Perussia B, Trinchieri G, Jackson A, Warner NL, Faust J, Rumpold H, Kraft D, Lanier LL (1984) The Fc receptor for IgG on human natural killer cells: phenotypic, functional, and comparative studies with monoclonal antibodies. J Immunol 133(1):180–189PubMed
24.
go back to reference Metes D, Ernst LK, Chambers WH, Sulica A, Herberman RB, Morel PA (1998) Expression of functional CD32 molecules on human NK cells is determined by an allelic polymorphism of the FcγRIIC gene. Blood 91(7):2369–2380PubMed Metes D, Ernst LK, Chambers WH, Sulica A, Herberman RB, Morel PA (1998) Expression of functional CD32 molecules on human NK cells is determined by an allelic polymorphism of the FcγRIIC gene. Blood 91(7):2369–2380PubMed
25.
go back to reference Metes D, Manciulea M, Petrusca D, Rabinowich H, Ernst LK, Popescu I, Calugaru A, Sulica A, Chambers WH, Herberman RB, Morel PA (1999) Ligand binding specificities and signal transduction pathways of Fcγ receptor IIc isoforms: the CD32 isoforms expressed by human NK cells. Eur J Immunol 29:2842–2852PubMedCrossRef Metes D, Manciulea M, Petrusca D, Rabinowich H, Ernst LK, Popescu I, Calugaru A, Sulica A, Chambers WH, Herberman RB, Morel PA (1999) Ligand binding specificities and signal transduction pathways of Fcγ receptor IIc isoforms: the CD32 isoforms expressed by human NK cells. Eur J Immunol 29:2842–2852PubMedCrossRef
26.
go back to reference Cassatella MA, Anegon I, Cuturi MC, Griskey P, Trinchieri G, Perussia B (1989) Fcg R (CD16) interaction with ligand induces Ca2 + mobilization and phosphoinositide turnover in human natural killer cells: role of Ca2 + in Fcγ R (CD16)-induced transcription and expression of lymphokine genes. J Exp Med 169:549–567PubMedCrossRef Cassatella MA, Anegon I, Cuturi MC, Griskey P, Trinchieri G, Perussia B (1989) Fcg R (CD16) interaction with ligand induces Ca2 + mobilization and phosphoinositide turnover in human natural killer cells: role of Ca2 + in Fcγ R (CD16)-induced transcription and expression of lymphokine genes. J Exp Med 169:549–567PubMedCrossRef
27.
go back to reference Mailliard RB, Alber SM, Shen H, Watkins SC, Kirkwood JM, Herberman RB, Kalinski P (2005) IL-18-induced CD83 + CCR7 + NK helper cells. J Exp Med 202(7):941–953PubMedCrossRef Mailliard RB, Alber SM, Shen H, Watkins SC, Kirkwood JM, Herberman RB, Kalinski P (2005) IL-18-induced CD83 + CCR7 + NK helper cells. J Exp Med 202(7):941–953PubMedCrossRef
28.
go back to reference Schroder K, Hertzog PJ, Ravasi T, Hume DA (2004) Interferon-γ: an overview of signals, mechanisms and functions. J Leukoc Biol 75:163–189PubMedCrossRef Schroder K, Hertzog PJ, Ravasi T, Hume DA (2004) Interferon-γ: an overview of signals, mechanisms and functions. J Leukoc Biol 75:163–189PubMedCrossRef
29.
go back to reference Kondadasula SV, Roda JM, Parihar R, Yu J, Lehman A, Caligiuri MA, Tridandapani S, Burry RW, Carson WE (2008) Colocalization of the IL-12 receptor and FcγRIIIa to natural killer cell lipid rafts leads to activation of ERK and enhanced production of interferon-g. Blood 111(8):4173–4183PubMedCrossRef Kondadasula SV, Roda JM, Parihar R, Yu J, Lehman A, Caligiuri MA, Tridandapani S, Burry RW, Carson WE (2008) Colocalization of the IL-12 receptor and FcγRIIIa to natural killer cell lipid rafts leads to activation of ERK and enhanced production of interferon-g. Blood 111(8):4173–4183PubMedCrossRef
30.
go back to reference Mavropoulos A, Sully G, Cope AP, Clark A (2004) Stabilization of IFN-γ mRNA by MAPK p38 in IL-12- and IL-18-stimulated human NK cells. Blood 105:282–288PubMedCrossRef Mavropoulos A, Sully G, Cope AP, Clark A (2004) Stabilization of IFN-γ mRNA by MAPK p38 in IL-12- and IL-18-stimulated human NK cells. Blood 105:282–288PubMedCrossRef
31.
go back to reference Nakahira M, Ahn H-J, Park W-R, Gao P, Tomura M, Park C-S, Hamaoka T, Ohta T, Kurimoto M, Fujiwara H (2002) Synergy of IL-12 and IL-18 for IFN-γ gene expression: IL-12-induced STAT4 contributes to IFN-γ promoter activation by up-regulating the binding activity of IL-18-induced activator protein 1. J Immunol 168:1146–1153PubMed Nakahira M, Ahn H-J, Park W-R, Gao P, Tomura M, Park C-S, Hamaoka T, Ohta T, Kurimoto M, Fujiwara H (2002) Synergy of IL-12 and IL-18 for IFN-γ gene expression: IL-12-induced STAT4 contributes to IFN-γ promoter activation by up-regulating the binding activity of IL-18-induced activator protein 1. J Immunol 168:1146–1153PubMed
32.
go back to reference Zhang S, Kaplan MH (2000) The p38 mitogen-activated protein kinase is required for IL-12-induced IFN-γ expression. J Immunol 165:1375–1380 Zhang S, Kaplan MH (2000) The p38 mitogen-activated protein kinase is required for IL-12-induced IFN-γ expression. J Immunol 165:1375–1380
33.
go back to reference Robertson MJ (2002) Role of chemokines in the biology of natural killer cells. J Leukoc Biol 71:173–183PubMed Robertson MJ (2002) Role of chemokines in the biology of natural killer cells. J Leukoc Biol 71:173–183PubMed
34.
go back to reference Overdijk MB, Verploegen S, Buijsse AO, Vink T, Leusen JHW, Bleeker WK, Parren PWHI (2012) Crosstalk between human IgG isotypes and murine effector cells. J Immunol 189:3430–3438PubMedCrossRef Overdijk MB, Verploegen S, Buijsse AO, Vink T, Leusen JHW, Bleeker WK, Parren PWHI (2012) Crosstalk between human IgG isotypes and murine effector cells. J Immunol 189:3430–3438PubMedCrossRef
35.
go back to reference Daniel D, Yang B, Lawrence DA, Totpal K, Balter I, Lee WP, Gogineni A, Cole MJ, Yee SF, Ross S, Ashkenazi A (2007) Cooperation of the proapoptotic receptor agonist rhApo2L/TRAIL with the CD20 antibody rituximab against non-Hodgkin lymphoma xenografts. Blood 110:4037–4046PubMedCrossRef Daniel D, Yang B, Lawrence DA, Totpal K, Balter I, Lee WP, Gogineni A, Cole MJ, Yee SF, Ross S, Ashkenazi A (2007) Cooperation of the proapoptotic receptor agonist rhApo2L/TRAIL with the CD20 antibody rituximab against non-Hodgkin lymphoma xenografts. Blood 110:4037–4046PubMedCrossRef
36.
go back to reference Hernandez-Ilizaliturri FJ, Reddy N, Holkova B, Ottman E, Czuczman MS (2005) Immunomodulatory drug CC-5013 or CC-4047 and rituximab enhance antitumor activity in a severe combined immunodeficient mouse lymphoma model. Clin Cancer Res 11(16):5984–5992PubMedCrossRef Hernandez-Ilizaliturri FJ, Reddy N, Holkova B, Ottman E, Czuczman MS (2005) Immunomodulatory drug CC-5013 or CC-4047 and rituximab enhance antitumor activity in a severe combined immunodeficient mouse lymphoma model. Clin Cancer Res 11(16):5984–5992PubMedCrossRef
37.
go back to reference Wigginton JM, Lee J-K, Wiltrout TA, Alvord WG, Hixon JA, Subleski J, Back TC, Wiltrout RH (2002) Syngergistic enhancement of ineffective endogenous antitumor immune response and induction of IFN-γ and Fas-ligand-dependent tumor eradication by combined administration of IL-18 and IL-2. J Immunol 169:4467–4474PubMed Wigginton JM, Lee J-K, Wiltrout TA, Alvord WG, Hixon JA, Subleski J, Back TC, Wiltrout RH (2002) Syngergistic enhancement of ineffective endogenous antitumor immune response and induction of IFN-γ and Fas-ligand-dependent tumor eradication by combined administration of IL-18 and IL-2. J Immunol 169:4467–4474PubMed
38.
go back to reference Jonak ZL, Trulli S, Maier C, McCabe FL, Kirkpatrick R, Johanson K, Ho YS, Elefante L, Chen Y-J, Herzyk D, Lotze MT, Johnson RK (2002) High-dose recombinant interleukin-18 induces an effective Th1 immune response to murine MOPC-315 plasmacytoma. J Immunother 25(Suppl. 1):S20–S27PubMedCrossRef Jonak ZL, Trulli S, Maier C, McCabe FL, Kirkpatrick R, Johanson K, Ho YS, Elefante L, Chen Y-J, Herzyk D, Lotze MT, Johnson RK (2002) High-dose recombinant interleukin-18 induces an effective Th1 immune response to murine MOPC-315 plasmacytoma. J Immunother 25(Suppl. 1):S20–S27PubMedCrossRef
39.
go back to reference Robertson MJ, Bauman J, Gardner O, Jonak Z, Struemper H, Germaschewski F, Koch KM, Murray S, Weisenbach J, Toso J (2011) A phase I trial evaluating the safety and biological activity of iboctadekin (rhIL-18) in combination with rituximab in patients with CD20 + B cell non-Hodgkin’s lymphoma. Blood 118:1579–1580 (Abstract 3697)CrossRef Robertson MJ, Bauman J, Gardner O, Jonak Z, Struemper H, Germaschewski F, Koch KM, Murray S, Weisenbach J, Toso J (2011) A phase I trial evaluating the safety and biological activity of iboctadekin (rhIL-18) in combination with rituximab in patients with CD20 + B cell non-Hodgkin’s lymphoma. Blood 118:1579–1580 (Abstract 3697)CrossRef
Metadata
Title
Effects of interleukin-18 on natural killer cells: costimulation of activation through Fc receptors for immunoglobulin
Authors
Shivani Srivastava
David Pelloso
Hailin Feng
Larry Voiles
David Lewis
Zdenka Haskova
Margaret Whitacre
Stephen Trulli
Yi-Jiun Chen
John Toso
Zdenka L. Jonak
Hua-Chen Chang
Michael J. Robertson
Publication date
01-06-2013
Publisher
Springer-Verlag
Published in
Cancer Immunology, Immunotherapy / Issue 6/2013
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-013-1403-0

Other articles of this Issue 6/2013

Cancer Immunology, Immunotherapy 6/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine