Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2022

Open Access 01-12-2022 | Cancer Immunotherapy | Review

Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy

Authors: Yi Wang, Kai Conrad Cecil Johnson, Margaret E. Gatti-Mays, Zihai Li

Published in: Journal of Hematology & Oncology | Issue 1/2022

Login to get access

Abstract

Immune checkpoint inhibitors targeting programmed cell death protein 1, programmed death-ligand 1, and cytotoxic T-lymphocyte-associated protein 4 provide deep and durable treatment responses which have revolutionized oncology. However, despite over 40% of cancer patients being eligible to receive immunotherapy, only 12% of patients gain benefit. A key to understanding what differentiates treatment response from non-response is better defining the role of the innate immune system in anti-tumor immunity and immune tolerance. Teleologically, myeloid cells, including macrophages, dendritic cells, monocytes, and neutrophils, initiate a response to invading pathogens and tissue repair after pathogen clearance is successfully accomplished. However, in the tumor microenvironment (TME), these innate cells are hijacked by the tumor cells and are imprinted to furthering tumor propagation and dissemination. Major advancements have been made in the field, especially related to the heterogeneity of myeloid cells and their function in the TME at the single cell level, a topic that has been highlighted by several recent international meetings including the 2021 China Cancer Immunotherapy workshop in Beijing. Here, we provide an up-to-date summary of the mechanisms by which major myeloid cells in the TME facilitate immunosuppression, enable tumor growth, foster tumor plasticity, and confer therapeutic resistance. We discuss ongoing strategies targeting the myeloid compartment in the preclinical and clinical settings which include: (1) altering myeloid cell composition within the TME; (2) functional blockade of immune-suppressive myeloid cells; (3) reprogramming myeloid cells to acquire pro-inflammatory properties; (4) modulating myeloid cells via cytokines; (5) myeloid cell therapies; and (6) emerging targets such as Siglec-15, TREM2, MARCO, LILRB2, and CLEVER-1. There is a significant promise that myeloid cell-based immunotherapy will help advance immuno-oncology in years to come.
Literature
1.
go back to reference Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–9.PubMedCrossRef Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–9.PubMedCrossRef
2.
3.
go back to reference Guc E, Pollard JW. Redefining macrophage and neutrophil biology in the metastatic cascade. Immunity. 2021;54(5):885–902.PubMedCrossRef Guc E, Pollard JW. Redefining macrophage and neutrophil biology in the metastatic cascade. Immunity. 2021;54(5):885–902.PubMedCrossRef
5.
go back to reference Wculek SK, et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24.PubMedCrossRef Wculek SK, et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24.PubMedCrossRef
7.
go back to reference Dunn GP, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8.PubMedCrossRef Dunn GP, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8.PubMedCrossRef
9.
go back to reference Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45.PubMedCrossRef Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45.PubMedCrossRef
10.
go back to reference Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer. 2016;16(7):447–62.PubMedCrossRef Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer. 2016;16(7):447–62.PubMedCrossRef
11.
go back to reference Cassetta L, et al. Human tumor-associated macrophage and monocyte transcriptional landscapes reveal cancer-specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell. 2019;35(4):588.PubMedPubMedCentralCrossRef Cassetta L, et al. Human tumor-associated macrophage and monocyte transcriptional landscapes reveal cancer-specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell. 2019;35(4):588.PubMedPubMedCentralCrossRef
12.
14.
go back to reference Cheng S, et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell. 2021;184(3):792-809 e23.PubMedCrossRef Cheng S, et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell. 2021;184(3):792-809 e23.PubMedCrossRef
16.
go back to reference Zhang Q, et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell. 2019;179(4):829-845 e20.PubMedCrossRef Zhang Q, et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell. 2019;179(4):829-845 e20.PubMedCrossRef
17.
go back to reference Klemm F, et al. Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells. Cell. 2020;181(7):1643-1660 e17.PubMedPubMedCentralCrossRef Klemm F, et al. Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells. Cell. 2020;181(7):1643-1660 e17.PubMedPubMedCentralCrossRef
18.
go back to reference Zilionis R, et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50(5):1317-1334 e10.PubMedPubMedCentralCrossRef Zilionis R, et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50(5):1317-1334 e10.PubMedPubMedCentralCrossRef
19.
go back to reference Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196(3):254–65.PubMedCrossRef Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196(3):254–65.PubMedCrossRef
20.
go back to reference Zhang QW, et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS ONE. 2012;7(12): e50946.PubMedPubMedCentralCrossRef Zhang QW, et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS ONE. 2012;7(12): e50946.PubMedPubMedCentralCrossRef
23.
go back to reference Schulz C, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336(6077):86–90.PubMedCrossRef Schulz C, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336(6077):86–90.PubMedCrossRef
25.
28.
go back to reference Zhu Y, et al. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity. 2017;47(2):323-338 e6.PubMedPubMedCentralCrossRef Zhu Y, et al. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity. 2017;47(2):323-338 e6.PubMedPubMedCentralCrossRef
30.
go back to reference Klug F, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24(5):589–602.PubMedCrossRef Klug F, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24(5):589–602.PubMedCrossRef
31.
32.
go back to reference Nathan CF, et al. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983;158(3):670–89.PubMedCrossRef Nathan CF, et al. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983;158(3):670–89.PubMedCrossRef
33.
go back to reference Stein M, et al. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med. 1992;176(1):287–92.PubMedCrossRef Stein M, et al. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med. 1992;176(1):287–92.PubMedCrossRef
34.
35.
go back to reference Davis MJ, et al. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection. MBio. 2013;4(3):e00264-e313.PubMedPubMedCentralCrossRef Davis MJ, et al. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection. MBio. 2013;4(3):e00264-e313.PubMedPubMedCentralCrossRef
37.
go back to reference Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. 2021;21(2):71–88.PubMedCrossRef Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. 2021;21(2):71–88.PubMedCrossRef
38.
go back to reference Cecchini MG, et al. Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse. Development. 1994;120(6):1357–72.PubMedCrossRef Cecchini MG, et al. Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse. Development. 1994;120(6):1357–72.PubMedCrossRef
39.
go back to reference Kitamura T, et al. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J Exp Med. 2015;212(7):1043–59.PubMedPubMedCentralCrossRef Kitamura T, et al. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J Exp Med. 2015;212(7):1043–59.PubMedPubMedCentralCrossRef
40.
41.
go back to reference Arwert EN, et al. A unidirectional transition from migratory to perivascular macrophage is required for tumor cell intravasation. Cell Rep. 2018;23(5):1239–48.PubMedPubMedCentralCrossRef Arwert EN, et al. A unidirectional transition from migratory to perivascular macrophage is required for tumor cell intravasation. Cell Rep. 2018;23(5):1239–48.PubMedPubMedCentralCrossRef
42.
go back to reference Lin EY, et al. Vascular endothelial growth factor restores delayed tumor progression in tumors depleted of macrophages. Mol Oncol. 2007;1(3):288–302.PubMedPubMedCentralCrossRef Lin EY, et al. Vascular endothelial growth factor restores delayed tumor progression in tumors depleted of macrophages. Mol Oncol. 2007;1(3):288–302.PubMedPubMedCentralCrossRef
43.
44.
go back to reference Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–12.PubMedCrossRef Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–12.PubMedCrossRef
45.
go back to reference Ruivo CF, et al. The biology of cancer exosomes: insights and new perspectives. Cancer Res. 2017;77(23):6480–8.PubMedCrossRef Ruivo CF, et al. The biology of cancer exosomes: insights and new perspectives. Cancer Res. 2017;77(23):6480–8.PubMedCrossRef
47.
go back to reference Morrissey SM, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33(10):2040-2058 e10.PubMedCrossRef Morrissey SM, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33(10):2040-2058 e10.PubMedCrossRef
49.
go back to reference Hwang WL, et al. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J Hematol Oncol. 2019;12(1):10.PubMedPubMedCentralCrossRef Hwang WL, et al. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J Hematol Oncol. 2019;12(1):10.PubMedPubMedCentralCrossRef
50.
51.
go back to reference Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206(6):1327–37.PubMedPubMedCentralCrossRef Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206(6):1327–37.PubMedPubMedCentralCrossRef
52.
54.
55.
go back to reference Li J, et al. Co-inhibitory molecule B7 superfamily member 1 expressed by tumor-infiltrating myeloid cells induces dysfunction of anti-tumor CD8(+) T cells. Immunity. 2018;48(4):773–7865.PubMedCrossRef Li J, et al. Co-inhibitory molecule B7 superfamily member 1 expressed by tumor-infiltrating myeloid cells induces dysfunction of anti-tumor CD8(+) T cells. Immunity. 2018;48(4):773–7865.PubMedCrossRef
57.
go back to reference Rodriguez PC, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64(16):5839–49.PubMedCrossRef Rodriguez PC, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64(16):5839–49.PubMedCrossRef
58.
go back to reference Ruffell B, et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 2014;26(5):623–37.PubMedPubMedCentralCrossRef Ruffell B, et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 2014;26(5):623–37.PubMedPubMedCentralCrossRef
60.
go back to reference Roux C, et al. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc Natl Acad Sci U S A. 2019;116(10):4326–35.PubMedPubMedCentralCrossRef Roux C, et al. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc Natl Acad Sci U S A. 2019;116(10):4326–35.PubMedPubMedCentralCrossRef
62.
go back to reference Zhang M, et al. A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients. J Ovarian Res. 2014;7:19.PubMedPubMedCentralCrossRef Zhang M, et al. A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients. J Ovarian Res. 2014;7:19.PubMedPubMedCentralCrossRef
63.
go back to reference Merad M, et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563–604.PubMedCrossRef Merad M, et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563–604.PubMedCrossRef
64.
go back to reference Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science. 2008;322(5904):1097–100.PubMedPubMedCentralCrossRef Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science. 2008;322(5904):1097–100.PubMedPubMedCentralCrossRef
65.
go back to reference Jongbloed SL, et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010;207(6):1247–60.PubMedPubMedCentralCrossRef Jongbloed SL, et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010;207(6):1247–60.PubMedPubMedCentralCrossRef
66.
go back to reference Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26(5):638–52.PubMedPubMedCentralCrossRef Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26(5):638–52.PubMedPubMedCentralCrossRef
67.
go back to reference Spranger S, et al. Tumor-residing batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31(5):711-723 e4.PubMedPubMedCentralCrossRef Spranger S, et al. Tumor-residing batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31(5):711-723 e4.PubMedPubMedCentralCrossRef
68.
go back to reference Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.PubMedPubMedCentralCrossRef Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.PubMedPubMedCentralCrossRef
69.
go back to reference Roberts EW, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30(2):324–36.PubMedPubMedCentralCrossRef Roberts EW, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30(2):324–36.PubMedPubMedCentralCrossRef
72.
go back to reference Duong E, et al. Type I interferon activates MHC class I-dressed CD11b(+) conventional dendritic cells to promote protective anti-tumor CD8(+) T cell immunity. Immunity. 2022;55(2):308-323 e9.PubMedCrossRef Duong E, et al. Type I interferon activates MHC class I-dressed CD11b(+) conventional dendritic cells to promote protective anti-tumor CD8(+) T cell immunity. Immunity. 2022;55(2):308-323 e9.PubMedCrossRef
73.
go back to reference Valdez Y, et al. Major histocompatibility complex class II presentation of cell-associated antigen is mediated by CD8alpha+ dendritic cells in vivo. J Exp Med. 2002;195(6):683–94.PubMedPubMedCentralCrossRef Valdez Y, et al. Major histocompatibility complex class II presentation of cell-associated antigen is mediated by CD8alpha+ dendritic cells in vivo. J Exp Med. 2002;195(6):683–94.PubMedPubMedCentralCrossRef
76.
77.
go back to reference Dunn GP, et al. A critical function for type I interferons in cancer immunoediting. Nat Immunol. 2005;6(7):722–9.PubMedCrossRef Dunn GP, et al. A critical function for type I interferons in cancer immunoediting. Nat Immunol. 2005;6(7):722–9.PubMedCrossRef
78.
go back to reference Fuertes MB, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J Exp Med. 2011;208(10):2005–16.PubMedPubMedCentralCrossRef Fuertes MB, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J Exp Med. 2011;208(10):2005–16.PubMedPubMedCentralCrossRef
79.
go back to reference Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17(10):1142–9.PubMedCrossRef Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17(10):1142–9.PubMedCrossRef
80.
go back to reference Deng L, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41(5):843–52.PubMedPubMedCentralCrossRef Deng L, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41(5):843–52.PubMedPubMedCentralCrossRef
81.
82.
go back to reference Garris CS, et al. Successful anti-PD-1 cancer immunotherapy requires T Cell-dendritic cell crosstalk involving the cytokines IFN-gamma and IL-12. Immunity. 2018;49(6):1148-1161 e7.PubMedPubMedCentralCrossRef Garris CS, et al. Successful anti-PD-1 cancer immunotherapy requires T Cell-dendritic cell crosstalk involving the cytokines IFN-gamma and IL-12. Immunity. 2018;49(6):1148-1161 e7.PubMedPubMedCentralCrossRef
83.
go back to reference Chemnitz JM, et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173(2):945–54.PubMedCrossRef Chemnitz JM, et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173(2):945–54.PubMedCrossRef
84.
go back to reference Clement M, et al. CD31 is a key coinhibitory receptor in the development of immunogenic dendritic cells. Proc Natl Acad Sci U S A. 2014;111(12):E1101–10.PubMedPubMedCentralCrossRef Clement M, et al. CD31 is a key coinhibitory receptor in the development of immunogenic dendritic cells. Proc Natl Acad Sci U S A. 2014;111(12):E1101–10.PubMedPubMedCentralCrossRef
85.
86.
go back to reference Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16(7):407–20.PubMedCrossRef Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16(7):407–20.PubMedCrossRef
87.
go back to reference de Mingo Pulido A, et al. TIM-3 regulates CD103(+) dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell. 2018;33(1):60-74 e6.PubMedPubMedCentralCrossRef de Mingo Pulido A, et al. TIM-3 regulates CD103(+) dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell. 2018;33(1):60-74 e6.PubMedPubMedCentralCrossRef
88.
go back to reference de Mingo Pulido A, et al. The inhibitory receptor TIM-3 limits activation of the cGAS-STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake. Immunity. 2021;54(6):1154-1167 e7.PubMedPubMedCentralCrossRef de Mingo Pulido A, et al. The inhibitory receptor TIM-3 limits activation of the cGAS-STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake. Immunity. 2021;54(6):1154-1167 e7.PubMedPubMedCentralCrossRef
90.
92.
go back to reference Rodrigues PF, et al. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat Immunol. 2018;19(7):711–22.PubMedCrossRef Rodrigues PF, et al. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat Immunol. 2018;19(7):711–22.PubMedCrossRef
94.
go back to reference Conrad C, et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. 2012;72(20):5240–9.PubMedPubMedCentralCrossRef Conrad C, et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. 2012;72(20):5240–9.PubMedPubMedCentralCrossRef
95.
go back to reference Labidi-Galy SI, et al. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011;71(16):5423–34.PubMedCrossRef Labidi-Galy SI, et al. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011;71(16):5423–34.PubMedCrossRef
96.
go back to reference Le Mercier I, et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res. 2013;73(15):4629–40.PubMedCrossRef Le Mercier I, et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res. 2013;73(15):4629–40.PubMedCrossRef
97.
go back to reference Sisirak V, et al. Impaired IFN-alpha production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 2012;72(20):5188–97.PubMedCrossRef Sisirak V, et al. Impaired IFN-alpha production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 2012;72(20):5188–97.PubMedCrossRef
98.
go back to reference Sisirak V, et al. Breast cancer-derived transforming growth factor-beta and tumor necrosis factor-alpha compromise interferon-alpha production by tumor-associated plasmacytoid dendritic cells. Int J Cancer. 2013;133(3):771–8.PubMedCrossRef Sisirak V, et al. Breast cancer-derived transforming growth factor-beta and tumor necrosis factor-alpha compromise interferon-alpha production by tumor-associated plasmacytoid dendritic cells. Int J Cancer. 2013;133(3):771–8.PubMedCrossRef
99.
go back to reference Terra M, et al. Tumor-derived TGFbeta alters the ability of plasmacytoid dendritic cells to respond to innate immune signaling. Cancer Res. 2018;78(11):3014–26.PubMedCrossRef Terra M, et al. Tumor-derived TGFbeta alters the ability of plasmacytoid dendritic cells to respond to innate immune signaling. Cancer Res. 2018;78(11):3014–26.PubMedCrossRef
100.
go back to reference Grenader T, et al. Derived neutrophil lymphocyte ratio is predictive of survival from intermittent therapy in advanced colorectal cancer: a post hoc analysis of the MRC COIN study. Br J Cancer. 2016;114(6):612–5.PubMedPubMedCentralCrossRef Grenader T, et al. Derived neutrophil lymphocyte ratio is predictive of survival from intermittent therapy in advanced colorectal cancer: a post hoc analysis of the MRC COIN study. Br J Cancer. 2016;114(6):612–5.PubMedPubMedCentralCrossRef
101.
102.
go back to reference Guthrie GJ, et al. The systemic inflammation-based neutrophil-lymphocyte ratio: experience in patients with cancer. Crit Rev Oncol Hematol. 2013;88(1):218–30.PubMedCrossRef Guthrie GJ, et al. The systemic inflammation-based neutrophil-lymphocyte ratio: experience in patients with cancer. Crit Rev Oncol Hematol. 2013;88(1):218–30.PubMedCrossRef
103.
go back to reference Peng B, et al. Prognostic significance of the neutrophil to lymphocyte ratio in patients with non-small cell lung cancer: a systemic review and meta-analysis. Int J Clin Exp Med. 2015;8(3):3098–106.PubMedPubMedCentral Peng B, et al. Prognostic significance of the neutrophil to lymphocyte ratio in patients with non-small cell lung cancer: a systemic review and meta-analysis. Int J Clin Exp Med. 2015;8(3):3098–106.PubMedPubMedCentral
104.
go back to reference Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol. 2019;16(10):601–20.PubMedCrossRef Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol. 2019;16(10):601–20.PubMedCrossRef
105.
go back to reference Templeton AJ, et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst. 2014;106(6):dju124.PubMedCrossRef Templeton AJ, et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst. 2014;106(6):dju124.PubMedCrossRef
106.
go back to reference Terashima T, et al. Blood neutrophil to lymphocyte ratio as a predictor in patients with advanced hepatocellular carcinoma treated with hepatic arterial infusion chemotherapy. Hepatol Res. 2015;45(9):949–59.PubMedCrossRef Terashima T, et al. Blood neutrophil to lymphocyte ratio as a predictor in patients with advanced hepatocellular carcinoma treated with hepatic arterial infusion chemotherapy. Hepatol Res. 2015;45(9):949–59.PubMedCrossRef
107.
go back to reference Valero C, et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021;12(1):729.PubMedPubMedCentralCrossRef Valero C, et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021;12(1):729.PubMedPubMedCentralCrossRef
108.
go back to reference Bagley SJ, et al. Pretreatment neutrophil-to-lymphocyte ratio as a marker of outcomes in nivolumab-treated patients with advanced non-small-cell lung cancer. Lung Cancer. 2017;106:1–7.PubMedCrossRef Bagley SJ, et al. Pretreatment neutrophil-to-lymphocyte ratio as a marker of outcomes in nivolumab-treated patients with advanced non-small-cell lung cancer. Lung Cancer. 2017;106:1–7.PubMedCrossRef
109.
110.
go back to reference Zaragoza J, et al. High neutrophil to lymphocyte ratio measured before starting ipilimumab treatment is associated with reduced overall survival in patients with melanoma. Br J Dermatol. 2016;174(1):146–51.PubMedCrossRef Zaragoza J, et al. High neutrophil to lymphocyte ratio measured before starting ipilimumab treatment is associated with reduced overall survival in patients with melanoma. Br J Dermatol. 2016;174(1):146–51.PubMedCrossRef
111.
go back to reference Rao HL, et al. Increased intratumoral neutrophil in colorectal carcinomas correlates closely with malignant phenotype and predicts patients’ adverse prognosis. PLoS ONE. 2012;7(1): e30806.PubMedPubMedCentralCrossRef Rao HL, et al. Increased intratumoral neutrophil in colorectal carcinomas correlates closely with malignant phenotype and predicts patients’ adverse prognosis. PLoS ONE. 2012;7(1): e30806.PubMedPubMedCentralCrossRef
112.
go back to reference Droeser RA, et al. High myeloperoxidase positive cell infiltration in colorectal cancer is an independent favorable prognostic factor. PLoS ONE. 2013;8(5): e64814.PubMedPubMedCentralCrossRef Droeser RA, et al. High myeloperoxidase positive cell infiltration in colorectal cancer is an independent favorable prognostic factor. PLoS ONE. 2013;8(5): e64814.PubMedPubMedCentralCrossRef
113.
go back to reference Galdiero MR, et al. Occurrence and significance of tumor-associated neutrophils in patients with colorectal cancer. Int J Cancer. 2016;139(2):446–56.PubMedCrossRef Galdiero MR, et al. Occurrence and significance of tumor-associated neutrophils in patients with colorectal cancer. Int J Cancer. 2016;139(2):446–56.PubMedCrossRef
114.
go back to reference Berry RS, et al. High levels of tumor-associated neutrophils are associated with improved overall survival in patients with stage II colorectal cancer. PLoS ONE. 2017;12(12): e0188799.PubMedPubMedCentralCrossRef Berry RS, et al. High levels of tumor-associated neutrophils are associated with improved overall survival in patients with stage II colorectal cancer. PLoS ONE. 2017;12(12): e0188799.PubMedPubMedCentralCrossRef
115.
go back to reference Wikberg ML, et al. Neutrophil infiltration is a favorable prognostic factor in early stages of colon cancer. Hum Pathol. 2017;68:193–202.PubMedCrossRef Wikberg ML, et al. Neutrophil infiltration is a favorable prognostic factor in early stages of colon cancer. Hum Pathol. 2017;68:193–202.PubMedCrossRef
117.
go back to reference Jaillon S, et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer. 2020;20(9):485–503.PubMedCrossRef Jaillon S, et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer. 2020;20(9):485–503.PubMedCrossRef
118.
go back to reference Powell DR, Huttenlocher A. Neutrophils in the tumor microenvironment. Trends Immunol. 2016;37(1):41–52.PubMedCrossRef Powell DR, Huttenlocher A. Neutrophils in the tumor microenvironment. Trends Immunol. 2016;37(1):41–52.PubMedCrossRef
119.
go back to reference Keeley EC, Mehrad B, Strieter RM. Chemokines as mediators of tumor angiogenesis and neovascularization. Exp Cell Res. 2011;317(5):685–90.PubMedCrossRef Keeley EC, Mehrad B, Strieter RM. Chemokines as mediators of tumor angiogenesis and neovascularization. Exp Cell Res. 2011;317(5):685–90.PubMedCrossRef
120.
go back to reference Casbon AJ, et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci U S A. 2015;112(6):E566–75.PubMedPubMedCentralCrossRef Casbon AJ, et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci U S A. 2015;112(6):E566–75.PubMedPubMedCentralCrossRef
121.
go back to reference Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16(7):431–46.PubMedCrossRef Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16(7):431–46.PubMedCrossRef
122.
go back to reference Colotta F, et al. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood. 1992;80(8):2012–20.PubMedCrossRef Colotta F, et al. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood. 1992;80(8):2012–20.PubMedCrossRef
123.
124.
go back to reference Dumitru CA, Lang S, Brandau S. Modulation of neutrophil granulocytes in the tumor microenvironment: mechanisms and consequences for tumor progression. Semin Cancer Biol. 2013;23(3):141–8.PubMedCrossRef Dumitru CA, Lang S, Brandau S. Modulation of neutrophil granulocytes in the tumor microenvironment: mechanisms and consequences for tumor progression. Semin Cancer Biol. 2013;23(3):141–8.PubMedCrossRef
125.
go back to reference Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci U S A. 2006;103(33):12493–8.PubMedPubMedCentralCrossRef Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci U S A. 2006;103(33):12493–8.PubMedPubMedCentralCrossRef
127.
go back to reference Gershkovitz M, et al. TRPM2 mediates neutrophil killing of disseminated tumor cells. Cancer Res. 2018;78(10):2680–90.PubMedCrossRef Gershkovitz M, et al. TRPM2 mediates neutrophil killing of disseminated tumor cells. Cancer Res. 2018;78(10):2680–90.PubMedCrossRef
129.
go back to reference Koga Y, et al. Neutrophil-derived TNF-related apoptosis-inducing ligand (TRAIL): a novel mechanism of antitumor effect by neutrophils. Cancer Res. 2004;64(3):1037–43.PubMedCrossRef Koga Y, et al. Neutrophil-derived TNF-related apoptosis-inducing ligand (TRAIL): a novel mechanism of antitumor effect by neutrophils. Cancer Res. 2004;64(3):1037–43.PubMedCrossRef
130.
132.
go back to reference Yang L, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020;583(7814):133–8.PubMedCrossRef Yang L, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020;583(7814):133–8.PubMedCrossRef
133.
go back to reference Xiao Y, et al. Cathepsin C promotes breast cancer lung metastasis by modulating neutrophil infiltration and neutrophil extracellular trap formation. Cancer Cell. 2021;39(3):423-437 e7.PubMedCrossRef Xiao Y, et al. Cathepsin C promotes breast cancer lung metastasis by modulating neutrophil infiltration and neutrophil extracellular trap formation. Cancer Cell. 2021;39(3):423-437 e7.PubMedCrossRef
135.
go back to reference Teijeira A, et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 2020;52(5):856-871 e8.PubMedCrossRef Teijeira A, et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 2020;52(5):856-871 e8.PubMedCrossRef
136.
go back to reference Schauer C, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511–7.PubMedCrossRef Schauer C, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511–7.PubMedCrossRef
139.
go back to reference Javeed N, et al. Immunosuppressive CD14(+)HLA-DR(lo/neg) monocytes are elevated in pancreatic cancer and “primed” by tumor-derived exosomes. Oncoimmunology. 2017;6(1): e1252013.PubMedCrossRef Javeed N, et al. Immunosuppressive CD14(+)HLA-DR(lo/neg) monocytes are elevated in pancreatic cancer and “primed” by tumor-derived exosomes. Oncoimmunology. 2017;6(1): e1252013.PubMedCrossRef
140.
go back to reference Plebanek MP, et al. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun. 2017;8(1):1319.PubMedPubMedCentralCrossRef Plebanek MP, et al. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun. 2017;8(1):1319.PubMedPubMedCentralCrossRef
141.
go back to reference Song X, et al. Cancer cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J Biol Chem. 2016;291(16):8453–64.PubMedPubMedCentralCrossRef Song X, et al. Cancer cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J Biol Chem. 2016;291(16):8453–64.PubMedPubMedCentralCrossRef
144.
145.
go back to reference Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485–98.PubMedPubMedCentralCrossRef Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485–98.PubMedPubMedCentralCrossRef
150.
go back to reference Netherby CS, et al. The granulocyte progenitor stage is a key target of IRF8-mediated regulation of myeloid-derived suppressor cell production. J Immunol. 2017;198(10):4129–39.PubMedCrossRef Netherby CS, et al. The granulocyte progenitor stage is a key target of IRF8-mediated regulation of myeloid-derived suppressor cell production. J Immunol. 2017;198(10):4129–39.PubMedCrossRef
151.
go back to reference Mohamed E, et al. The unfolded protein response mediator PERK governs myeloid cell-driven immunosuppression in tumors through inhibition of STING signaling. Immunity. 2020;52(4):668-682 e7.PubMedPubMedCentralCrossRef Mohamed E, et al. The unfolded protein response mediator PERK governs myeloid cell-driven immunosuppression in tumors through inhibition of STING signaling. Immunity. 2020;52(4):668-682 e7.PubMedPubMedCentralCrossRef
152.
go back to reference Tartour E, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 2011;30(1):83–95.PubMedCrossRef Tartour E, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 2011;30(1):83–95.PubMedCrossRef
153.
go back to reference Shojaei F, et al. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc Natl Acad Sci U S A. 2009;106(16):6742–7.PubMedPubMedCentralCrossRef Shojaei F, et al. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc Natl Acad Sci U S A. 2009;106(16):6742–7.PubMedPubMedCentralCrossRef
154.
go back to reference Corzo CA, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182(9):5693–701.PubMedCrossRef Corzo CA, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182(9):5693–701.PubMedCrossRef
157.
159.
go back to reference Kumar V, et al. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity. 2016;44(2):303–15.PubMedPubMedCentralCrossRef Kumar V, et al. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity. 2016;44(2):303–15.PubMedPubMedCentralCrossRef
160.
go back to reference Corzo CA, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53.PubMedPubMedCentralCrossRef Corzo CA, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53.PubMedPubMedCentralCrossRef
161.
go back to reference Rodriguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–91.PubMedPubMedCentralCrossRef Rodriguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–91.PubMedPubMedCentralCrossRef
162.
go back to reference Huang B, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66(2):1123–31.PubMedCrossRef Huang B, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66(2):1123–31.PubMedCrossRef
163.
go back to reference Pan PY, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70(1):99–108.PubMedCrossRef Pan PY, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70(1):99–108.PubMedCrossRef
164.
go back to reference Hoechst B, et al. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. 2011;117(24):6532–41.PubMedCrossRef Hoechst B, et al. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. 2011;117(24):6532–41.PubMedCrossRef
165.
go back to reference Grossman JG, et al. Recruitment of CCR2(+) tumor associated macrophage to sites of liver metastasis confers a poor prognosis in human colorectal cancer. Oncoimmunology. 2018;7(9): e1470729.PubMedPubMedCentralCrossRef Grossman JG, et al. Recruitment of CCR2(+) tumor associated macrophage to sites of liver metastasis confers a poor prognosis in human colorectal cancer. Oncoimmunology. 2018;7(9): e1470729.PubMedPubMedCentralCrossRef
166.
go back to reference Bonapace L, et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 2014;515(7525):130–3.PubMedCrossRef Bonapace L, et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 2014;515(7525):130–3.PubMedCrossRef
167.
go back to reference Salcedo R, et al. Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression. Blood. 2000;96(1):34–40.PubMedCrossRef Salcedo R, et al. Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression. Blood. 2000;96(1):34–40.PubMedCrossRef
168.
go back to reference Stamatovic SM, et al. CCL2 regulates angiogenesis via activation of Ets-1 transcription factor. J Immunol. 2006;177(4):2651–61.PubMedCrossRef Stamatovic SM, et al. CCL2 regulates angiogenesis via activation of Ets-1 transcription factor. J Immunol. 2006;177(4):2651–61.PubMedCrossRef
169.
go back to reference ClinicalTrials.gov., An Open-Label, Multicenter, Phase 2 Study of Single-Agent CNTO 888 (an Anti-CCL2 Monoclonal Antibody) for the Treatment of Subjects With Metastatic Castrate-Resistant Prostate Cancer. , in Identifier NCT00992186. ClinicalTrials.gov., An Open-Label, Multicenter, Phase 2 Study of Single-Agent CNTO 888 (an Anti-CCL2 Monoclonal Antibody) for the Treatment of Subjects With Metastatic Castrate-Resistant Prostate Cancer. , in Identifier NCT00992186.
170.
go back to reference Brana I, et al. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study. Target Oncol. 2015;10(1):111–23.PubMedCrossRef Brana I, et al. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study. Target Oncol. 2015;10(1):111–23.PubMedCrossRef
171.
go back to reference Nywening TM, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17(5):651–62.PubMedPubMedCentralCrossRef Nywening TM, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17(5):651–62.PubMedPubMedCentralCrossRef
172.
go back to reference Noel M, et al. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2020;38(3):800–11.PubMedCrossRef Noel M, et al. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2020;38(3):800–11.PubMedCrossRef
176.
go back to reference Zhu Y, et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74(18):5057–69.PubMedPubMedCentralCrossRef Zhu Y, et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74(18):5057–69.PubMedPubMedCentralCrossRef
177.
go back to reference Murray LJ, et al. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Clin Exp Metastasis. 2003;20(8):757–66.PubMedCrossRef Murray LJ, et al. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Clin Exp Metastasis. 2003;20(8):757–66.PubMedCrossRef
179.
go back to reference Autio KA, et al. Immunomodulatory activity of a colony-stimulating factor-1 receptor inhibitor in patients with advanced refractory breast or prostate cancer: a phase I study. Clin Cancer Res. 2020;26(21):5609–20.PubMedPubMedCentralCrossRef Autio KA, et al. Immunomodulatory activity of a colony-stimulating factor-1 receptor inhibitor in patients with advanced refractory breast or prostate cancer: a phase I study. Clin Cancer Res. 2020;26(21):5609–20.PubMedPubMedCentralCrossRef
180.
go back to reference Dowlati A, et al. LY3022855, an anti-colony stimulating factor-1 receptor (CSF-1R) monoclonal antibody, in patients with advanced solid tumors refractory to standard therapy: phase 1 dose-escalation trial. Invest New Drugs. 2021;39(4):1057–71.PubMedCrossRef Dowlati A, et al. LY3022855, an anti-colony stimulating factor-1 receptor (CSF-1R) monoclonal antibody, in patients with advanced solid tumors refractory to standard therapy: phase 1 dose-escalation trial. Invest New Drugs. 2021;39(4):1057–71.PubMedCrossRef
181.
go back to reference Papadopoulos KP, et al. First-in-human study of AMG 820, a monoclonal anti-colony-stimulating factor 1 receptor antibody, in patients with advanced solid tumors. Clin Cancer Res. 2017;23(19):5703–10.PubMedCrossRef Papadopoulos KP, et al. First-in-human study of AMG 820, a monoclonal anti-colony-stimulating factor 1 receptor antibody, in patients with advanced solid tumors. Clin Cancer Res. 2017;23(19):5703–10.PubMedCrossRef
182.
go back to reference Falchook GS, et al. A phase 1a/1b trial of CSF-1R inhibitor LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Invest New Drugs. 2021;39(5):1284–97.PubMedCrossRef Falchook GS, et al. A phase 1a/1b trial of CSF-1R inhibitor LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Invest New Drugs. 2021;39(5):1284–97.PubMedCrossRef
184.
go back to reference West RB, et al. A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor cells. Proc Natl Acad Sci U S A. 2006;103(3):690–5.PubMedPubMedCentralCrossRef West RB, et al. A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor cells. Proc Natl Acad Sci U S A. 2006;103(3):690–5.PubMedPubMedCentralCrossRef
185.
go back to reference Tap WD, et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet. 2019;394(10197):478–87.PubMedPubMedCentralCrossRef Tap WD, et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet. 2019;394(10197):478–87.PubMedPubMedCentralCrossRef
186.
go back to reference Benner B, et al. Pexidartinib, a novel small molecule CSF-1R inhibitor in use for tenosynovial giant cell tumor: a systematic review of pre-clinical and clinical development. Drug Des Devel Ther. 2020;14:1693–704.PubMedPubMedCentralCrossRef Benner B, et al. Pexidartinib, a novel small molecule CSF-1R inhibitor in use for tenosynovial giant cell tumor: a systematic review of pre-clinical and clinical development. Drug Des Devel Ther. 2020;14:1693–704.PubMedPubMedCentralCrossRef
188.
go back to reference Che J, et al. Targeting CXCR1/2: The medicinal potential as cancer immunotherapy agents, antagonists research highlights and challenges ahead. Eur J Med Chem. 2020;185: 111853.PubMedCrossRef Che J, et al. Targeting CXCR1/2: The medicinal potential as cancer immunotherapy agents, antagonists research highlights and challenges ahead. Eur J Med Chem. 2020;185: 111853.PubMedCrossRef
189.
go back to reference Schott AF, et al. Phase Ib pilot study to evaluate reparixin in combination with weekly paclitaxel in patients with HER-2-negative metastatic breast cancer. Clin Cancer Res. 2017;23(18):5358–65.PubMedPubMedCentralCrossRef Schott AF, et al. Phase Ib pilot study to evaluate reparixin in combination with weekly paclitaxel in patients with HER-2-negative metastatic breast cancer. Clin Cancer Res. 2017;23(18):5358–65.PubMedPubMedCentralCrossRef
190.
go back to reference Goldstein LJ, et al. A randomized, placebo-controlled phase 2 study of paclitaxel in combination with reparixin compared to paclitaxel alone as front-line therapy for metastatic triple-negative breast cancer (fRida). Breast Cancer Res Treat. 2021;190(2):265–75.PubMedPubMedCentralCrossRef Goldstein LJ, et al. A randomized, placebo-controlled phase 2 study of paclitaxel in combination with reparixin compared to paclitaxel alone as front-line therapy for metastatic triple-negative breast cancer (fRida). Breast Cancer Res Treat. 2021;190(2):265–75.PubMedPubMedCentralCrossRef
191.
go back to reference Kemp DM, et al. Ladarixin, a dual CXCR1/2 inhibitor, attenuates experimental melanomas harboring different molecular defects by affecting malignant cells and tumor microenvironment. Oncotarget. 2017;8(9):14428–42.PubMedPubMedCentralCrossRef Kemp DM, et al. Ladarixin, a dual CXCR1/2 inhibitor, attenuates experimental melanomas harboring different molecular defects by affecting malignant cells and tumor microenvironment. Oncotarget. 2017;8(9):14428–42.PubMedPubMedCentralCrossRef
193.
go back to reference Bilusic M, et al. Phase I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J Immunother Cancer. 2019;7(1):240.PubMedPubMedCentralCrossRef Bilusic M, et al. Phase I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J Immunother Cancer. 2019;7(1):240.PubMedPubMedCentralCrossRef
195.
go back to reference Habets TH, et al. Fractionated radiotherapy with 3 x 8 Gy induces systemic anti-tumour responses and abscopal tumour inhibition without modulating the humoral anti-tumour response. PLoS ONE. 2016;11(7): e0159515.PubMedPubMedCentralCrossRef Habets TH, et al. Fractionated radiotherapy with 3 x 8 Gy induces systemic anti-tumour responses and abscopal tumour inhibition without modulating the humoral anti-tumour response. PLoS ONE. 2016;11(7): e0159515.PubMedPubMedCentralCrossRef
196.
go back to reference Chakravarty PK, et al. Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res. 1999;59(24):6028–32.PubMed Chakravarty PK, et al. Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res. 1999;59(24):6028–32.PubMed
197.
go back to reference Fong L, et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci U S A. 2001;98(15):8809–14.PubMedPubMedCentralCrossRef Fong L, et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci U S A. 2001;98(15):8809–14.PubMedPubMedCentralCrossRef
198.
go back to reference Oba T, et al. Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s. Nat Commun. 2020;11(1):5415.PubMedPubMedCentralCrossRef Oba T, et al. Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s. Nat Commun. 2020;11(1):5415.PubMedPubMedCentralCrossRef
199.
go back to reference Freedman RS, et al. Pilot study of Flt3 ligand comparing intraperitoneal with subcutaneous routes on hematologic and immunologic responses in patients with peritoneal carcinomatosis and mesotheliomas. Clin Cancer Res. 2003;9(14):5228–37.PubMed Freedman RS, et al. Pilot study of Flt3 ligand comparing intraperitoneal with subcutaneous routes on hematologic and immunologic responses in patients with peritoneal carcinomatosis and mesotheliomas. Clin Cancer Res. 2003;9(14):5228–37.PubMed
200.
go back to reference Ohri N, et al. FLT3 ligand (CDX-301) and stereotactic radiotherapy for advanced non-small cell lung cancer. J Clin Oncol. 2020;38(15_suppl):9618–9618.CrossRef Ohri N, et al. FLT3 ligand (CDX-301) and stereotactic radiotherapy for advanced non-small cell lung cancer. J Clin Oncol. 2020;38(15_suppl):9618–9618.CrossRef
203.
204.
go back to reference Li H, et al. Fragment-based drug design and drug repositioning using multiple ligand simultaneous docking (MLSD): identifying celecoxib and template compounds as novel inhibitors of signal transducer and activator of transcription 3 (STAT3). J Med Chem. 2011;54(15):5592–6.PubMedPubMedCentralCrossRef Li H, et al. Fragment-based drug design and drug repositioning using multiple ligand simultaneous docking (MLSD): identifying celecoxib and template compounds as novel inhibitors of signal transducer and activator of transcription 3 (STAT3). J Med Chem. 2011;54(15):5592–6.PubMedPubMedCentralCrossRef
205.
206.
go back to reference Jonker DJ, et al. Napabucasin versus placebo in refractory advanced colorectal cancer: a randomised phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(4):263–70.PubMedCrossRef Jonker DJ, et al. Napabucasin versus placebo in refractory advanced colorectal cancer: a randomised phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(4):263–70.PubMedCrossRef
208.
go back to reference Bekaii-Saab T, et al. 1466P Napabucasin+ nab-paclitaxel with gemcitabine in patients (pts) with metastatic pancreatic adenocarcinoma (mPDAC): results from the phase III CanStem111P study. Ann Oncol. 2021;32:S1084–5.CrossRef Bekaii-Saab T, et al. 1466P Napabucasin+ nab-paclitaxel with gemcitabine in patients (pts) with metastatic pancreatic adenocarcinoma (mPDAC): results from the phase III CanStem111P study. Ann Oncol. 2021;32:S1084–5.CrossRef
209.
go back to reference Matozaki T, et al. Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009;19(2):72–80.PubMedCrossRef Matozaki T, et al. Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009;19(2):72–80.PubMedCrossRef
210.
go back to reference Pengam S, et al. SIRPalpha/CD47 axis controls the maintenance of transplant tolerance sustained by myeloid-derived suppressor cells. Am J Transplant. 2019;19(12):3263–75.PubMedCrossRef Pengam S, et al. SIRPalpha/CD47 axis controls the maintenance of transplant tolerance sustained by myeloid-derived suppressor cells. Am J Transplant. 2019;19(12):3263–75.PubMedCrossRef
212.
go back to reference Sim J, et al. Discovery of high affinity, pan-allelic, and pan-mammalian reactive antibodies against the myeloid checkpoint receptor SIRPalpha. MAbs. 2019;11(6):1036–52.PubMedPubMedCentralCrossRef Sim J, et al. Discovery of high affinity, pan-allelic, and pan-mammalian reactive antibodies against the myeloid checkpoint receptor SIRPalpha. MAbs. 2019;11(6):1036–52.PubMedPubMedCentralCrossRef
213.
go back to reference Chao MP, et al. Therapeutic targeting of the macrophage immune checkpoint CD47 in myeloid malignancies. Front Oncol. 2019;9:1380.PubMedCrossRef Chao MP, et al. Therapeutic targeting of the macrophage immune checkpoint CD47 in myeloid malignancies. Front Oncol. 2019;9:1380.PubMedCrossRef
216.
go back to reference Andrejeva G, et al. Novel SIRPalpha antibodies that induce single-agent phagocytosis of tumor cells while preserving T cells. J Immunol. 2021;206(4):712–21.PubMedPubMedCentralCrossRef Andrejeva G, et al. Novel SIRPalpha antibodies that induce single-agent phagocytosis of tumor cells while preserving T cells. J Immunol. 2021;206(4):712–21.PubMedPubMedCentralCrossRef
217.
go back to reference Chen YC, et al. Progress of CD47 immune checkpoint blockade agents in anticancer therapy: a hematotoxic perspective. J Cancer Res Clin Oncol. 2022;148(1):1–14.PubMedCrossRef Chen YC, et al. Progress of CD47 immune checkpoint blockade agents in anticancer therapy: a hematotoxic perspective. J Cancer Res Clin Oncol. 2022;148(1):1–14.PubMedCrossRef
218.
go back to reference Kauder SE, et al. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS ONE. 2018;13(8): e0201832.PubMedPubMedCentralCrossRef Kauder SE, et al. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS ONE. 2018;13(8): e0201832.PubMedPubMedCentralCrossRef
219.
go back to reference Weiskopf K, et al. Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies. Science. 2013;341(6141):88–91.PubMedCrossRef Weiskopf K, et al. Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies. Science. 2013;341(6141):88–91.PubMedCrossRef
221.
go back to reference Sikic BI, et al. First-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol. 2019;37(12):946–53.PubMedPubMedCentralCrossRef Sikic BI, et al. First-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol. 2019;37(12):946–53.PubMedPubMedCentralCrossRef
222.
go back to reference Lakhani NJ, et al. Evorpacept alone and in combination with pembrolizumab or trastuzumab in patients with advanced solid tumours (ASPEN-01): a first-in-human, open-label, multicentre, phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2021;22(12):1740–51.PubMedCrossRef Lakhani NJ, et al. Evorpacept alone and in combination with pembrolizumab or trastuzumab in patients with advanced solid tumours (ASPEN-01): a first-in-human, open-label, multicentre, phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2021;22(12):1740–51.PubMedCrossRef
226.
go back to reference Kristiansen G, et al. CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res. 2003;9(13):4906–13.PubMed Kristiansen G, et al. CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res. 2003;9(13):4906–13.PubMed
227.
go back to reference Overdevest JB, et al. CD24 offers a therapeutic target for control of bladder cancer metastasis based on a requirement for lung colonization. Cancer Res. 2011;71(11):3802–11.PubMedPubMedCentralCrossRef Overdevest JB, et al. CD24 offers a therapeutic target for control of bladder cancer metastasis based on a requirement for lung colonization. Cancer Res. 2011;71(11):3802–11.PubMedPubMedCentralCrossRef
228.
go back to reference Altevogt P, et al. Novel insights into the function of CD24: A driving force in cancer. Int J Cancer. 2021;148(3):546–59.PubMedCrossRef Altevogt P, et al. Novel insights into the function of CD24: A driving force in cancer. Int J Cancer. 2021;148(3):546–59.PubMedCrossRef
229.
go back to reference Arber N, et al. A Novel Anti-CD24 Monoclonal Antibody: Unarmed, Conjugated and Bi-specific, for Targeting Human Malignancies: 204. Official journal of the American College of Gastroenterology | ACG, 2017. 112: p. S106. Arber N, et al. A Novel Anti-CD24 Monoclonal Antibody: Unarmed, Conjugated and Bi-specific, for Targeting Human Malignancies: 204. Official journal of the American College of Gastroenterology | ACG, 2017. 112: p. S106.
230.
go back to reference Han Y, et al. CD24 targeting bi-specific antibody that simultaneously stimulates NKG2D enhances the efficacy of cancer immunotherapy. J Cancer Res Clin Oncol. 2019;145(5):1179–90.PubMedCrossRef Han Y, et al. CD24 targeting bi-specific antibody that simultaneously stimulates NKG2D enhances the efficacy of cancer immunotherapy. J Cancer Res Clin Oncol. 2019;145(5):1179–90.PubMedCrossRef
231.
go back to reference Sun F, et al. Anti-CD24 antibody-nitric oxide conjugate selectively and potently suppresses hepatic carcinoma. Cancer Res. 2019;79(13):3395–405.PubMedCrossRef Sun F, et al. Anti-CD24 antibody-nitric oxide conjugate selectively and potently suppresses hepatic carcinoma. Cancer Res. 2019;79(13):3395–405.PubMedCrossRef
232.
go back to reference Maliar A, et al. Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice. Gastroenterology. 2012;143(5):1375-1384 e5.PubMedCrossRef Maliar A, et al. Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice. Gastroenterology. 2012;143(5):1375-1384 e5.PubMedCrossRef
235.
go back to reference Li TT, Ogino S, Qian ZR. Toll-like receptor signaling in colorectal cancer: carcinogenesis to cancer therapy. World J Gastroenterol. 2014;20(47):17699–708.PubMedPubMedCentralCrossRef Li TT, Ogino S, Qian ZR. Toll-like receptor signaling in colorectal cancer: carcinogenesis to cancer therapy. World J Gastroenterol. 2014;20(47):17699–708.PubMedPubMedCentralCrossRef
236.
237.
go back to reference Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27(2):161–7.PubMedCrossRef Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27(2):161–7.PubMedCrossRef
239.
go back to reference Kuo TC, et al. Abstract 1721: TAC-001, a toll-like receptor 9 (TLR9) agonist antibody conjugate targeting B cells, promotes anti-tumor immunity and favorable safety profile following systemic administration in preclinical models. Cancer Res. 2021;81(13_supplement):1721–1721.CrossRef Kuo TC, et al. Abstract 1721: TAC-001, a toll-like receptor 9 (TLR9) agonist antibody conjugate targeting B cells, promotes anti-tumor immunity and favorable safety profile following systemic administration in preclinical models. Cancer Res. 2021;81(13_supplement):1721–1721.CrossRef
240.
go back to reference Wang D, et al. Modulation of the tumor microenvironment by intratumoral administration of IMO-2125, a novel TLR9 agonist, for cancer immunotherapy. Int J Oncol. 2018;53(3):1193–203.PubMed Wang D, et al. Modulation of the tumor microenvironment by intratumoral administration of IMO-2125, a novel TLR9 agonist, for cancer immunotherapy. Int J Oncol. 2018;53(3):1193–203.PubMed
241.
go back to reference Yuan S, et al. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol Lett. 2018;15(4):5271–9.PubMedPubMedCentral Yuan S, et al. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol Lett. 2018;15(4):5271–9.PubMedPubMedCentral
242.
go back to reference Kohrt HE, et al. Dose-escalated, intratumoral TLR9 agonist and low-dose radiation induce abscopal effects in follicular lymphoma. Blood. 2014;124(21):3092–3092.CrossRef Kohrt HE, et al. Dose-escalated, intratumoral TLR9 agonist and low-dose radiation induce abscopal effects in follicular lymphoma. Blood. 2014;124(21):3092–3092.CrossRef
243.
go back to reference Hirsh V, et al. Randomized phase III trial of paclitaxel/carboplatin with or without PF-3512676 (Toll-like receptor 9 agonist) as first-line treatment for advanced non-small-cell lung cancer. J Clin Oncol. 2011;29(19):2667–74.PubMedCrossRef Hirsh V, et al. Randomized phase III trial of paclitaxel/carboplatin with or without PF-3512676 (Toll-like receptor 9 agonist) as first-line treatment for advanced non-small-cell lung cancer. J Clin Oncol. 2011;29(19):2667–74.PubMedCrossRef
244.
go back to reference Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 1998;16:111–35.PubMedCrossRef Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 1998;16:111–35.PubMedCrossRef
246.
go back to reference Beatty GL, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331(6024):1612–6.PubMedPubMedCentralCrossRef Beatty GL, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331(6024):1612–6.PubMedPubMedCentralCrossRef
248.
249.
go back to reference Yu X, et al. Complex interplay between epitope specificity and isotype dictates the biological activity of anti-human CD40 antibodies. Cancer Cell. 2018;33(4):664-675.e4.PubMedPubMedCentralCrossRef Yu X, et al. Complex interplay between epitope specificity and isotype dictates the biological activity of anti-human CD40 antibodies. Cancer Cell. 2018;33(4):664-675.e4.PubMedPubMedCentralCrossRef
250.
go back to reference Vonderheide RH, et al. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin Oncol. 2007;25(7):876–83.PubMedCrossRef Vonderheide RH, et al. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin Oncol. 2007;25(7):876–83.PubMedCrossRef
251.
go back to reference Rüter J, et al. Immune modulation with weekly dosing of an agonist CD40 antibody in a phase I study of patients with advanced solid tumors. Cancer Biol Ther. 2010;10(10):983–93.PubMedPubMedCentralCrossRef Rüter J, et al. Immune modulation with weekly dosing of an agonist CD40 antibody in a phase I study of patients with advanced solid tumors. Cancer Biol Ther. 2010;10(10):983–93.PubMedPubMedCentralCrossRef
252.
go back to reference Bajor DL, et al. Long-term outcomes of a phase I study of agonist CD40 antibody and CTLA-4 blockade in patients with metastatic melanoma. Oncoimmunology. 2018;7(10): e1468956.PubMedPubMedCentralCrossRef Bajor DL, et al. Long-term outcomes of a phase I study of agonist CD40 antibody and CTLA-4 blockade in patients with metastatic melanoma. Oncoimmunology. 2018;7(10): e1468956.PubMedPubMedCentralCrossRef
253.
go back to reference Ribas A, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31(5):616–22.PubMedPubMedCentralCrossRef Ribas A, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31(5):616–22.PubMedPubMedCentralCrossRef
254.
go back to reference O’Hara MH, et al. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: an open-label, multicentre, phase 1b study. Lancet Oncol. 2021;22(1):118–31.PubMedCrossRef O’Hara MH, et al. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: an open-label, multicentre, phase 1b study. Lancet Oncol. 2021;22(1):118–31.PubMedCrossRef
255.
go back to reference Wainberg ZA, et al. Open-label, phase I study of nivolumab combined with nab-paclitaxel plus gemcitabine in advanced pancreatic cancer. Clin Cancer Res. 2020;26(18):4814–22.PubMedCrossRef Wainberg ZA, et al. Open-label, phase I study of nivolumab combined with nab-paclitaxel plus gemcitabine in advanced pancreatic cancer. Clin Cancer Res. 2020;26(18):4814–22.PubMedCrossRef
256.
go back to reference Barlesi F, et al. 291 Phase Ib study of selicrelumab (CD40 agonist) in combination with atezolizumab (anti-PD-L1) in patients with advanced solid tumors. J Immunother Cancer. 2020;8(Suppl 3):A178–A178. Barlesi F, et al. 291 Phase Ib study of selicrelumab (CD40 agonist) in combination with atezolizumab (anti-PD-L1) in patients with advanced solid tumors. J Immunother Cancer. 2020;8(Suppl 3):A178–A178.
257.
go back to reference Weiss SA, et al. A phase I study of APX005M and cabiralizumab with or without nivolumab in patients with melanoma, kidney cancer, or non-small cell lung cancer resistant to anti-PD-1/PD-L1. Clin Cancer Res. 2021;27(17):4757–67.PubMedPubMedCentralCrossRef Weiss SA, et al. A phase I study of APX005M and cabiralizumab with or without nivolumab in patients with melanoma, kidney cancer, or non-small cell lung cancer resistant to anti-PD-1/PD-L1. Clin Cancer Res. 2021;27(17):4757–67.PubMedPubMedCentralCrossRef
258.
260.
go back to reference Davis RJ, et al. Anti-PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kδ/γ. Cancer Res. 2017;77(10):2607–19.PubMedPubMedCentralCrossRef Davis RJ, et al. Anti-PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kδ/γ. Cancer Res. 2017;77(10):2607–19.PubMedPubMedCentralCrossRef
261.
go back to reference Lonetti A, et al. PI3K pan-inhibition impairs more efficiently proliferation and survival of T-cell acute lymphoblastic leukemia cell lines when compared to isoform-selective PI3K inhibitors. Oncotarget. 2015;6(12):10399–414.PubMedPubMedCentralCrossRef Lonetti A, et al. PI3K pan-inhibition impairs more efficiently proliferation and survival of T-cell acute lymphoblastic leukemia cell lines when compared to isoform-selective PI3K inhibitors. Oncotarget. 2015;6(12):10399–414.PubMedPubMedCentralCrossRef
262.
265.
go back to reference Sullivan RJ, et al. Initial results from first-in-human study of IPI-549, a tumor macrophage-targeting agent, combined with nivolumab in advanced solid tumors. J Clin Oncol. 2018;36(15_suppl):3013–3013.CrossRef Sullivan RJ, et al. Initial results from first-in-human study of IPI-549, a tumor macrophage-targeting agent, combined with nivolumab in advanced solid tumors. J Clin Oncol. 2018;36(15_suppl):3013–3013.CrossRef
266.
go back to reference Cohen E, et al. 352 Updated clinical data from the squamous cell carcinoma of the head and neck (SCCHN) expansion cohort of an ongoing Ph1/1b Study of eganelisib (formerly IPI-549) in combination with nivolumab. J Immunother Cancer. 2020;8(Suppl 3):A214–5. Cohen E, et al. 352 Updated clinical data from the squamous cell carcinoma of the head and neck (SCCHN) expansion cohort of an ongoing Ph1/1b Study of eganelisib (formerly IPI-549) in combination with nivolumab. J Immunother Cancer. 2020;8(Suppl 3):A214–5.
267.
go back to reference Postow M, et al. 434 Updated clinical data from the melanoma expansion cohort of an ongoing Ph1/1b study of eganelisib (formerly IPI-549) in combination with nivolumab. J Immunother Cancer. 2020;8(Suppl 3):A264–5. Postow M, et al. 434 Updated clinical data from the melanoma expansion cohort of an ongoing Ph1/1b study of eganelisib (formerly IPI-549) in combination with nivolumab. J Immunother Cancer. 2020;8(Suppl 3):A264–5.
268.
go back to reference Hatem S, et al. Abstract P5-16-02: Updated efficacy, safety and translational data from MARIO-3, a phase II open-label study evaluating a novel triplet combination of eganelisib (IPI-549), atezolizumab (atezo), and nab-paclitaxel (nab-pac) as first-line (1L) therapy for locally advanced or metastatic triple-negative breast cancer (TNBC). Cancer Res. 2022;82(4):P5-16-02-P5-16-02.CrossRef Hatem S, et al. Abstract P5-16-02: Updated efficacy, safety and translational data from MARIO-3, a phase II open-label study evaluating a novel triplet combination of eganelisib (IPI-549), atezolizumab (atezo), and nab-paclitaxel (nab-pac) as first-line (1L) therapy for locally advanced or metastatic triple-negative breast cancer (TNBC). Cancer Res. 2022;82(4):P5-16-02-P5-16-02.CrossRef
269.
go back to reference Tomczak P, et al. Preliminary analysis of a phase II, multicenter, randomized, active-control study to evaluate the efficacy and safety of eganelisib (IPI 549) in combination with nivolumab compared to nivolumab monotherapy in patients with advanced urothelial carcinoma. J Clinic Oncol. 2021;39(6_suppl):436–436.CrossRef Tomczak P, et al. Preliminary analysis of a phase II, multicenter, randomized, active-control study to evaluate the efficacy and safety of eganelisib (IPI 549) in combination with nivolumab compared to nivolumab monotherapy in patients with advanced urothelial carcinoma. J Clinic Oncol. 2021;39(6_suppl):436–436.CrossRef
270.
go back to reference Allard D, Turcotte M, Stagg J. Targeting A2 adenosine receptors in cancer. Immunol Cell Biol. 2017;95(4):333–9.PubMedCrossRef Allard D, Turcotte M, Stagg J. Targeting A2 adenosine receptors in cancer. Immunol Cell Biol. 2017;95(4):333–9.PubMedCrossRef
271.
go back to reference Antonioli L, et al. Switching off CD73: a way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov Today. 2017;22(11):1686–96.PubMedCrossRef Antonioli L, et al. Switching off CD73: a way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov Today. 2017;22(11):1686–96.PubMedCrossRef
272.
go back to reference Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature. 2001;414(6866):916–20.PubMedCrossRef Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature. 2001;414(6866):916–20.PubMedCrossRef
273.
go back to reference Cekic C, et al. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res. 2014;74(24):7250–9.PubMedPubMedCentralCrossRef Cekic C, et al. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res. 2014;74(24):7250–9.PubMedPubMedCentralCrossRef
274.
go back to reference Ghalamfarsa G, et al. CD73 as a potential opportunity for cancer immunotherapy. Expert Opin Ther Targets. 2019;23(2):127–42.PubMedCrossRef Ghalamfarsa G, et al. CD73 as a potential opportunity for cancer immunotherapy. Expert Opin Ther Targets. 2019;23(2):127–42.PubMedCrossRef
275.
go back to reference Jeffrey JL, Lawson KV, Powers JP. Targeting metabolism of extracellular nucleotides via inhibition of ectonucleotidases CD73 and CD39. J Med Chem. 2020;63(22):13444–65.PubMedCrossRef Jeffrey JL, Lawson KV, Powers JP. Targeting metabolism of extracellular nucleotides via inhibition of ectonucleotidases CD73 and CD39. J Med Chem. 2020;63(22):13444–65.PubMedCrossRef
277.
278.
go back to reference Mirza M, et al. 1195 Results of NSGO-OV-UMB1/ENGOT-OV30 study: a phase II study of durvalumab and oleclumab in patients with relapsed ovarian cancer (OC). Int J Gynecol Cancer. 2021;31(Suppl 3):A376–A376. Mirza M, et al. 1195 Results of NSGO-OV-UMB1/ENGOT-OV30 study: a phase II study of durvalumab and oleclumab in patients with relapsed ovarian cancer (OC). Int J Gynecol Cancer. 2021;31(Suppl 3):A376–A376.
279.
go back to reference Robert F, et al. Preliminary safety, pharmacokinetics (PK), pharmacodynamics (PD) and clinical efficacy of uliledlimab (TJ004309), a differentiated CD73 antibody, in combination with atezolizumab in patients with advanced cancer. J Clinic Oncol. 2021;39(15_suppl):2511–2511.CrossRef Robert F, et al. Preliminary safety, pharmacokinetics (PK), pharmacodynamics (PD) and clinical efficacy of uliledlimab (TJ004309), a differentiated CD73 antibody, in combination with atezolizumab in patients with advanced cancer. J Clinic Oncol. 2021;39(15_suppl):2511–2511.CrossRef
280.
go back to reference Perrot I, et al. Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep. 2019;27(8):2411-2425.e9.PubMedCrossRef Perrot I, et al. Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep. 2019;27(8):2411-2425.e9.PubMedCrossRef
282.
go back to reference Hauser RA, et al. Preladenant as an adjunctive therapy with levodopa in parkinson disease: two randomized clinical trials and lessons learned. JAMA Neurol. 2015;72(12):1491–500.PubMedCrossRef Hauser RA, et al. Preladenant as an adjunctive therapy with levodopa in parkinson disease: two randomized clinical trials and lessons learned. JAMA Neurol. 2015;72(12):1491–500.PubMedCrossRef
283.
go back to reference Churov A, Zhulai G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum Immunol. 2021;82(4):270–8.PubMedCrossRef Churov A, Zhulai G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum Immunol. 2021;82(4):270–8.PubMedCrossRef
284.
go back to reference Chandra D, et al. Abstract A87: The A2AR antagonist AZD4635 prevents adenosine-mediated immunosuppression in tumor microenvironment and enhances antitumor immunity partly by enhancing CD103+ dendritic cells. Cancer Immunol Res. 2020;8(3_supplement):A87–A87.CrossRef Chandra D, et al. Abstract A87: The A2AR antagonist AZD4635 prevents adenosine-mediated immunosuppression in tumor microenvironment and enhances antitumor immunity partly by enhancing CD103+ dendritic cells. Cancer Immunol Res. 2020;8(3_supplement):A87–A87.CrossRef
285.
go back to reference Fong L, et al. Safety and clinical activity of adenosine A2a receptor (A2aR) antagonist, CPI-444, in anti-PD1/PDL1 treatment-refractory renal cell (RCC) and non-small cell lung cancer (NSCLC) patients. J Clin Oncol. 2017;35(15_suppl):3004–3004.CrossRef Fong L, et al. Safety and clinical activity of adenosine A2a receptor (A2aR) antagonist, CPI-444, in anti-PD1/PDL1 treatment-refractory renal cell (RCC) and non-small cell lung cancer (NSCLC) patients. J Clin Oncol. 2017;35(15_suppl):3004–3004.CrossRef
286.
go back to reference Powderly J, et al. 1206P-Phase I evaluation of AB928, a novel dual adenosine receptor antagonist, combined with chemotherapy or AB122 (anti-PD-1) in patients (pts) with advanced malignancies. Ann Oncol. 2019;30: v493.CrossRef Powderly J, et al. 1206P-Phase I evaluation of AB928, a novel dual adenosine receptor antagonist, combined with chemotherapy or AB122 (anti-PD-1) in patients (pts) with advanced malignancies. Ann Oncol. 2019;30: v493.CrossRef
287.
go back to reference Yu J, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783–97.PubMedCrossRef Yu J, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783–97.PubMedCrossRef
288.
go back to reference Nakamura T, et al. Expression of indoleamine 2, 3-dioxygenase and the recruitment of Foxp3-expressing regulatory T cells in the development and progression of uterine cervical cancer. Cancer Sci. 2007;98(6):874–81.PubMedCrossRef Nakamura T, et al. Expression of indoleamine 2, 3-dioxygenase and the recruitment of Foxp3-expressing regulatory T cells in the development and progression of uterine cervical cancer. Cancer Sci. 2007;98(6):874–81.PubMedCrossRef
289.
go back to reference Munn DH, et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114(2):280–90.PubMedPubMedCentralCrossRef Munn DH, et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114(2):280–90.PubMedPubMedCentralCrossRef
290.
go back to reference Long GV, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20(8):1083–97.PubMedCrossRef Long GV, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20(8):1083–97.PubMedCrossRef
291.
292.
go back to reference Platten M, et al. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019;18(5):379–401.PubMedCrossRef Platten M, et al. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019;18(5):379–401.PubMedCrossRef
293.
295.
go back to reference Baardman J, et al. Metabolic-epigenetic crosstalk in macrophage activation. Epigenomics. 2015;7(7):1155–64.PubMedCrossRef Baardman J, et al. Metabolic-epigenetic crosstalk in macrophage activation. Epigenomics. 2015;7(7):1155–64.PubMedCrossRef
296.
go back to reference Chang YC, et al. Epigenetic control of MHC class II expression in tumor-associated macrophages by decoy receptor 3. Blood. 2008;111(10):5054–63.PubMedCrossRef Chang YC, et al. Epigenetic control of MHC class II expression in tumor-associated macrophages by decoy receptor 3. Blood. 2008;111(10):5054–63.PubMedCrossRef
297.
go back to reference Skov S, et al. Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res. 2005;65(23):11136–45.PubMedCrossRef Skov S, et al. Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res. 2005;65(23):11136–45.PubMedCrossRef
298.
go back to reference Liu M, et al. Understanding the epigenetic regulation of tumours and their microenvironments: opportunities and problems for epigenetic therapy. J Pathol. 2017;241(1):10–24.PubMedCrossRef Liu M, et al. Understanding the epigenetic regulation of tumours and their microenvironments: opportunities and problems for epigenetic therapy. J Pathol. 2017;241(1):10–24.PubMedCrossRef
299.
go back to reference Wang HF, et al. Histone deacetylase inhibitors deplete myeloid-derived suppressor cells induced by 4T1 mammary tumors in vivo and in vitro. Cancer Immunol Immunother. 2017;66(3):355–66.PubMedCrossRef Wang HF, et al. Histone deacetylase inhibitors deplete myeloid-derived suppressor cells induced by 4T1 mammary tumors in vivo and in vitro. Cancer Immunol Immunother. 2017;66(3):355–66.PubMedCrossRef
300.
go back to reference Dokmanovic M, Clarke C, Marks PA. Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res. 2007;5(10):981–9.PubMedCrossRef Dokmanovic M, Clarke C, Marks PA. Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res. 2007;5(10):981–9.PubMedCrossRef
301.
go back to reference Chen X, et al. Epigenetic strategies synergize with PD-L1/PD-1 targeted cancer immunotherapies to enhance antitumor responses. Acta Pharm Sin B. 2020;10(5):723–33.PubMedCrossRef Chen X, et al. Epigenetic strategies synergize with PD-L1/PD-1 targeted cancer immunotherapies to enhance antitumor responses. Acta Pharm Sin B. 2020;10(5):723–33.PubMedCrossRef
302.
go back to reference San José-Enériz E et al. HDAC Inhibitors in Acute Myeloid Leukemia. Cancers (Basel), 2019. 11(11). San José-Enériz E et al. HDAC Inhibitors in Acute Myeloid Leukemia. Cancers (Basel), 2019. 11(11).
304.
go back to reference Craddock CF, et al. Outcome of azacitidine therapy in acute myeloid leukemia is not improved by concurrent vorinostat therapy but is predicted by a diagnostic molecular signature. Clin Cancer Res. 2017;23(21):6430–40.PubMedCrossRef Craddock CF, et al. Outcome of azacitidine therapy in acute myeloid leukemia is not improved by concurrent vorinostat therapy but is predicted by a diagnostic molecular signature. Clin Cancer Res. 2017;23(21):6430–40.PubMedCrossRef
305.
go back to reference Mann BS, et al. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007;12(10):1247–52.PubMedCrossRef Mann BS, et al. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007;12(10):1247–52.PubMedCrossRef
306.
go back to reference Juo YY, et al. Epigenetic therapy for solid tumors: from bench science to clinical trials. Epigenomics. 2015;7(2):215–35.PubMedCrossRef Juo YY, et al. Epigenetic therapy for solid tumors: from bench science to clinical trials. Epigenomics. 2015;7(2):215–35.PubMedCrossRef
307.
go back to reference Vansteenkiste J, et al. Early phase II trial of oral vorinostat in relapsed or refractory breast, colorectal, or non-small cell lung cancer. Invest New Drugs. 2008;26(5):483–8.PubMedCrossRef Vansteenkiste J, et al. Early phase II trial of oral vorinostat in relapsed or refractory breast, colorectal, or non-small cell lung cancer. Invest New Drugs. 2008;26(5):483–8.PubMedCrossRef
308.
go back to reference Christmas BJ, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol Res. 2018;6(12):1561–77.PubMedPubMedCentralCrossRef Christmas BJ, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol Res. 2018;6(12):1561–77.PubMedPubMedCentralCrossRef
309.
go back to reference Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18(3):219–34.PubMedCrossRef Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18(3):219–34.PubMedCrossRef
310.
go back to reference Golomb HM, et al. Alpha-2 interferon therapy of hairy-cell leukemia: a multicenter study of 64 patients. J Clin Oncol. 1986;4(6):900–5.PubMedCrossRef Golomb HM, et al. Alpha-2 interferon therapy of hairy-cell leukemia: a multicenter study of 64 patients. J Clin Oncol. 1986;4(6):900–5.PubMedCrossRef
312.
go back to reference Ohkuri T, et al. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol Res. 2014;2(12):1199–208.PubMedPubMedCentralCrossRef Ohkuri T, et al. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol Res. 2014;2(12):1199–208.PubMedPubMedCentralCrossRef
313.
315.
go back to reference Lara PN Jr, et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J Clin Oncol. 2011;29(22):2965–71.PubMedCrossRef Lara PN Jr, et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J Clin Oncol. 2011;29(22):2965–71.PubMedCrossRef
317.
go back to reference Amouzegar A et al. STING Agonists as Cancer Therapeutics. Cancers (Basel), 2021. 13(11). Amouzegar A et al. STING Agonists as Cancer Therapeutics. Cancers (Basel), 2021. 13(11).
318.
319.
go back to reference Zhang X, et al. PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int J Clin Oncol. 2017;22(6):1026–33.PubMedCrossRef Zhang X, et al. PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int J Clin Oncol. 2017;22(6):1026–33.PubMedCrossRef
320.
go back to reference Zhou J, Zhang S, Guo C. Crosstalk between macrophages and natural killer cells in the tumor microenvironment. Int Immunopharmacol. 2021;101(Pt B): 108374.PubMedCrossRef Zhou J, Zhang S, Guo C. Crosstalk between macrophages and natural killer cells in the tumor microenvironment. Int Immunopharmacol. 2021;101(Pt B): 108374.PubMedCrossRef
321.
go back to reference Alberts DS, et al. Randomized phase 3 trial of interferon gamma-1b plus standard carboplatin/paclitaxel versus carboplatin/paclitaxel alone for first-line treatment of advanced ovarian and primary peritoneal carcinomas: results from a prospectively designed analysis of progression-free survival. Gynecol Oncol. 2008;109(2):174–81.PubMedCrossRef Alberts DS, et al. Randomized phase 3 trial of interferon gamma-1b plus standard carboplatin/paclitaxel versus carboplatin/paclitaxel alone for first-line treatment of advanced ovarian and primary peritoneal carcinomas: results from a prospectively designed analysis of progression-free survival. Gynecol Oncol. 2008;109(2):174–81.PubMedCrossRef
322.
go back to reference Bourgeois-Daigneault MC, et al. Oncolytic vesicular stomatitis virus expressing interferon-γ has enhanced therapeutic activity. Mol Ther Oncolytics. 2016;3:16001.PubMedPubMedCentralCrossRef Bourgeois-Daigneault MC, et al. Oncolytic vesicular stomatitis virus expressing interferon-γ has enhanced therapeutic activity. Mol Ther Oncolytics. 2016;3:16001.PubMedPubMedCentralCrossRef
323.
go back to reference Kerkar SP, et al. IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors. J Clin Invest. 2011;121(12):4746–57.PubMedPubMedCentralCrossRef Kerkar SP, et al. IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors. J Clin Invest. 2011;121(12):4746–57.PubMedPubMedCentralCrossRef
324.
go back to reference Watkins SK, et al. IL-12 rapidly alters the functional profile of tumor-associated and tumor-infiltrating macrophages in vitro and in vivo. J Immunol. 2007;178(3):1357–62.PubMedCrossRef Watkins SK, et al. IL-12 rapidly alters the functional profile of tumor-associated and tumor-infiltrating macrophages in vitro and in vivo. J Immunol. 2007;178(3):1357–62.PubMedCrossRef
325.
go back to reference Brunda MJ, et al. Antitumor activity of interleukin 12 in preclinical models. Cancer Chemother Pharmacol. 1996;38(Suppl):S16-21.PubMedCrossRef Brunda MJ, et al. Antitumor activity of interleukin 12 in preclinical models. Cancer Chemother Pharmacol. 1996;38(Suppl):S16-21.PubMedCrossRef
326.
go back to reference Leonard JP, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90(7):2541–8.PubMed Leonard JP, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90(7):2541–8.PubMed
327.
go back to reference Portielje JE, et al. Repeated administrations of interleukin (IL)-12 are associated with persistently elevated plasma levels of IL-10 and declining IFN-gamma, tumor necrosis factor-alpha, IL-6, and IL-8 responses. Clin Cancer Res. 2003;9(1):76–83.PubMed Portielje JE, et al. Repeated administrations of interleukin (IL)-12 are associated with persistently elevated plasma levels of IL-10 and declining IFN-gamma, tumor necrosis factor-alpha, IL-6, and IL-8 responses. Clin Cancer Res. 2003;9(1):76–83.PubMed
328.
go back to reference Lasek W, Zagożdżon R, Jakobisiak M. Interleukin 12: still a promising candidate for tumor immunotherapy? Cancer Immunol Immunother. 2014;63(5):419–35.PubMedPubMedCentralCrossRef Lasek W, Zagożdżon R, Jakobisiak M. Interleukin 12: still a promising candidate for tumor immunotherapy? Cancer Immunol Immunother. 2014;63(5):419–35.PubMedPubMedCentralCrossRef
329.
go back to reference Anwer K, et al. Phase I trial of a formulated IL-12 plasmid in combination with carboplatin and docetaxel chemotherapy in the treatment of platinum-sensitive recurrent ovarian cancer. Gynecol Oncol. 2013;131(1):169–73.PubMedCrossRef Anwer K, et al. Phase I trial of a formulated IL-12 plasmid in combination with carboplatin and docetaxel chemotherapy in the treatment of platinum-sensitive recurrent ovarian cancer. Gynecol Oncol. 2013;131(1):169–73.PubMedCrossRef
330.
go back to reference Thaker PH, et al. GEN-1 immunotherapy for the treatment of ovarian cancer. Future Oncol. 2019;15(4):421–38.PubMedCrossRef Thaker PH, et al. GEN-1 immunotherapy for the treatment of ovarian cancer. Future Oncol. 2019;15(4):421–38.PubMedCrossRef
331.
go back to reference Thaker PH, et al. GEN-1 in combination with neoadjuvant chemotherapy for patients with advanced epithelial ovarian cancer: a phase i dose-escalation study. Clin Cancer Res. 2021;27(20):5536–45.PubMedPubMedCentralCrossRef Thaker PH, et al. GEN-1 in combination with neoadjuvant chemotherapy for patients with advanced epithelial ovarian cancer: a phase i dose-escalation study. Clin Cancer Res. 2021;27(20):5536–45.PubMedPubMedCentralCrossRef
332.
go back to reference Barton KN, et al. Phase I trial of oncolytic adenovirus-mediated cytotoxic and interleukin-12 gene therapy for the treatment of metastatic pancreatic cancer. Mol Ther Oncolytics. 2021;20:94–104.PubMedCrossRef Barton KN, et al. Phase I trial of oncolytic adenovirus-mediated cytotoxic and interleukin-12 gene therapy for the treatment of metastatic pancreatic cancer. Mol Ther Oncolytics. 2021;20:94–104.PubMedCrossRef
333.
go back to reference Hewitt SL, et al. Intratumoral IL12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin Cancer Res. 2020;26(23):6284–98.PubMedCrossRef Hewitt SL, et al. Intratumoral IL12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin Cancer Res. 2020;26(23):6284–98.PubMedCrossRef
335.
go back to reference Ham B, et al. The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res. 2016;11(1):1–27.PubMedPubMedCentral Ham B, et al. The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res. 2016;11(1):1–27.PubMedPubMedCentral
336.
go back to reference Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10(1):45–65.PubMedCrossRef Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10(1):45–65.PubMedCrossRef
337.
go back to reference Cai X, et al. Inflammatory factor TNF-α promotes the growth of breast cancer via the positive feedback loop of TNFR1/NF-κB (and/or p38)/p-STAT3/HBXIP/TNFR1. Oncotarget. 2017;8(35):58338–52.PubMedPubMedCentralCrossRef Cai X, et al. Inflammatory factor TNF-α promotes the growth of breast cancer via the positive feedback loop of TNFR1/NF-κB (and/or p38)/p-STAT3/HBXIP/TNFR1. Oncotarget. 2017;8(35):58338–52.PubMedPubMedCentralCrossRef
341.
go back to reference Zhang YW, et al. Expression of tumor necrosis factor receptor 2 in human non-small cell lung cancer and its role as a potential prognostic biomarker. Thorac Cancer. 2019;10(3):437–44.PubMedPubMedCentralCrossRef Zhang YW, et al. Expression of tumor necrosis factor receptor 2 in human non-small cell lung cancer and its role as a potential prognostic biomarker. Thorac Cancer. 2019;10(3):437–44.PubMedPubMedCentralCrossRef
343.
go back to reference Ham B, et al. TNF receptor-2 facilitates an immunosuppressive microenvironment in the liver to promote the colonization and growth of hepatic metastases. Cancer Res. 2015;75(24):5235–47.PubMedCrossRef Ham B, et al. TNF receptor-2 facilitates an immunosuppressive microenvironment in the liver to promote the colonization and growth of hepatic metastases. Cancer Res. 2015;75(24):5235–47.PubMedCrossRef
344.
go back to reference Chen L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cell Mol Life Sci. 2018;75(11):2045–58.PubMedCrossRef Chen L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cell Mol Life Sci. 2018;75(11):2045–58.PubMedCrossRef
345.
go back to reference Tu S, et al. Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell. 2008;14(5):408–19.PubMedPubMedCentralCrossRef Tu S, et al. Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell. 2008;14(5):408–19.PubMedPubMedCentralCrossRef
347.
go back to reference Lee C, et al. Inflammasome as a promising therapeutic target for cancer. Life Sci. 2019;231: 116593.PubMedCrossRef Lee C, et al. Inflammasome as a promising therapeutic target for cancer. Life Sci. 2019;231: 116593.PubMedCrossRef
348.
go back to reference Ridker PM, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10105):1833–42.PubMedCrossRef Ridker PM, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10105):1833–42.PubMedCrossRef
349.
go back to reference Novartis top-line results for CANOPY-1 phase III study support further evaluation of canakinumab in lung cancer. News release. Novartis. [cited 2021 October 25]; https://bit.ly/3pyfhvt Novartis top-line results for CANOPY-1 phase III study support further evaluation of canakinumab in lung cancer. News release. Novartis. [cited 2021 October 25]; https://​bit.​ly/​3pyfhvt
350.
go back to reference Paz-Ares L, et al. 1194MO Canakinumab (CAN)+ docetaxel (DTX) for the second-or third-line (2/3L) treatment of advanced non-small cell lung cancer (NSCLC): CANOPY-2 phase III results. Ann Oncol. 2021;32:S953–4.CrossRef Paz-Ares L, et al. 1194MO Canakinumab (CAN)+ docetaxel (DTX) for the second-or third-line (2/3L) treatment of advanced non-small cell lung cancer (NSCLC): CANOPY-2 phase III results. Ann Oncol. 2021;32:S953–4.CrossRef
351.
go back to reference Liu Q, et al. Targeting interlukin-6 to relieve immunosuppression in tumor microenvironment. Tumour Biol. 2017;39(6):1010428317712445.PubMed Liu Q, et al. Targeting interlukin-6 to relieve immunosuppression in tumor microenvironment. Tumour Biol. 2017;39(6):1010428317712445.PubMed
353.
go back to reference San-Miguel J, et al. Phase 2 randomized study of bortezomib-melphalan-prednisone with or without siltuximab (anti-IL-6) in multiple myeloma. Blood. 2014;123(26):4136–42.PubMedPubMedCentralCrossRef San-Miguel J, et al. Phase 2 randomized study of bortezomib-melphalan-prednisone with or without siltuximab (anti-IL-6) in multiple myeloma. Blood. 2014;123(26):4136–42.PubMedPubMedCentralCrossRef
354.
go back to reference Orlowski RZ, et al. A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-IL-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma. Am J Hematol. 2015;90(1):42–9.PubMedPubMedCentralCrossRef Orlowski RZ, et al. A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-IL-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma. Am J Hematol. 2015;90(1):42–9.PubMedPubMedCentralCrossRef
355.
go back to reference Dijkgraaf EM, et al. A phase I trial combining carboplatin/doxorubicin with tocilizumab, an anti-IL-6R monoclonal antibody, and interferon-α2b in patients with recurrent epithelial ovarian cancer. Ann Oncol. 2015;26(10):2141–9.PubMedCrossRef Dijkgraaf EM, et al. A phase I trial combining carboplatin/doxorubicin with tocilizumab, an anti-IL-6R monoclonal antibody, and interferon-α2b in patients with recurrent epithelial ovarian cancer. Ann Oncol. 2015;26(10):2141–9.PubMedCrossRef
358.
go back to reference Oft M. IL-10: master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res. 2014;2(3):194–9.PubMedCrossRef Oft M. IL-10: master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res. 2014;2(3):194–9.PubMedCrossRef
359.
go back to reference Neven B, et al. A Mendelian predisposition to B-cell lymphoma caused by IL-10R deficiency. Blood. 2013;122(23):3713–22.PubMedCrossRef Neven B, et al. A Mendelian predisposition to B-cell lymphoma caused by IL-10R deficiency. Blood. 2013;122(23):3713–22.PubMedCrossRef
360.
go back to reference Berg DJ, et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest. 1996;98(4):1010–20.PubMedPubMedCentralCrossRef Berg DJ, et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest. 1996;98(4):1010–20.PubMedPubMedCentralCrossRef
361.
362.
go back to reference Emmerich J, et al. IL-10 directly activates and expands tumor-resident CD8(+) T cells without de novo infiltration from secondary lymphoid organs. Cancer Res. 2012;72(14):3570–81.PubMedCrossRef Emmerich J, et al. IL-10 directly activates and expands tumor-resident CD8(+) T cells without de novo infiltration from secondary lymphoid organs. Cancer Res. 2012;72(14):3570–81.PubMedCrossRef
363.
go back to reference Hecht JR, et al. Randomized phase III study of FOLFOX alone or with pegilodecakin as second-line therapy in patients with metastatic pancreatic cancer that progressed after gemcitabine (SEQUOIA). J Clin Oncol. 2021;39(10):1108–18.PubMedPubMedCentralCrossRef Hecht JR, et al. Randomized phase III study of FOLFOX alone or with pegilodecakin as second-line therapy in patients with metastatic pancreatic cancer that progressed after gemcitabine (SEQUOIA). J Clin Oncol. 2021;39(10):1108–18.PubMedPubMedCentralCrossRef
365.
go back to reference Huang CY, et al. Recent progress in TGF-β inhibitors for cancer therapy. Biomed Pharmacother. 2021;134: 111046.PubMedCrossRef Huang CY, et al. Recent progress in TGF-β inhibitors for cancer therapy. Biomed Pharmacother. 2021;134: 111046.PubMedCrossRef
367.
go back to reference Rocconi RP, et al. Gemogenovatucel-T (Vigil) immunotherapy as maintenance in frontline stage III/IV ovarian cancer (VITAL): a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Oncol. 2020;21(12):1661–72.PubMedCrossRef Rocconi RP, et al. Gemogenovatucel-T (Vigil) immunotherapy as maintenance in frontline stage III/IV ovarian cancer (VITAL): a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Oncol. 2020;21(12):1661–72.PubMedCrossRef
368.
go back to reference Teixeira AF, Ten Dijke P, Zhu HJ. On-target anti-TGF-β therapies are not succeeding in clinical cancer treatments: what are remaining challenges? Front Cell Dev Biol. 2020;8:605.PubMedPubMedCentralCrossRef Teixeira AF, Ten Dijke P, Zhu HJ. On-target anti-TGF-β therapies are not succeeding in clinical cancer treatments: what are remaining challenges? Front Cell Dev Biol. 2020;8:605.PubMedPubMedCentralCrossRef
369.
go back to reference Giaccone G, et al. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. Eur J Cancer. 2015;51(16):2321–9.PubMedCrossRef Giaccone G, et al. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. Eur J Cancer. 2015;51(16):2321–9.PubMedCrossRef
370.
go back to reference Strauss J, et al. 957O Long-term follow-up of patients (pts) with human papillomavirus (HPV)–associated malignancies treated with bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1. Ann Oncol. 2021;32:S829.CrossRef Strauss J, et al. 957O Long-term follow-up of patients (pts) with human papillomavirus (HPV)–associated malignancies treated with bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1. Ann Oncol. 2021;32:S829.CrossRef
372.
go back to reference Liu BL, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10(4):292–303.PubMedCrossRef Liu BL, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10(4):292–303.PubMedCrossRef
373.
go back to reference Goldsmith K, et al. Infected cell protein (ICP)47 enhances herpes simplex virus neurovirulence by blocking the CD8+ T cell response. J Exp Med. 1998;187(3):341–8.PubMedPubMedCentralCrossRef Goldsmith K, et al. Infected cell protein (ICP)47 enhances herpes simplex virus neurovirulence by blocking the CD8+ T cell response. J Exp Med. 1998;187(3):341–8.PubMedPubMedCentralCrossRef
375.
go back to reference Buonaguro L, et al. Translating tumor antigens into cancer vaccines. Clin Vaccine Immunol. 2011;18(1):23–34.PubMedCrossRef Buonaguro L, et al. Translating tumor antigens into cancer vaccines. Clin Vaccine Immunol. 2011;18(1):23–34.PubMedCrossRef
377.
go back to reference Glavan TM, Pavelic J. The exploitation of Toll-like receptor 3 signaling in cancer therapy. Curr Pharm Des. 2014;20(42):6555–64.PubMedCrossRef Glavan TM, Pavelic J. The exploitation of Toll-like receptor 3 signaling in cancer therapy. Curr Pharm Des. 2014;20(42):6555–64.PubMedCrossRef
378.
379.
go back to reference Sabbatini P, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18(23):6497–508.PubMedCrossRef Sabbatini P, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18(23):6497–508.PubMedCrossRef
380.
go back to reference Schuler-Thurner B, et al. Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. J Exp Med. 2002;195(10):1279–88.PubMedPubMedCentralCrossRef Schuler-Thurner B, et al. Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. J Exp Med. 2002;195(10):1279–88.PubMedPubMedCentralCrossRef
381.
go back to reference Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–22.PubMedCrossRef Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–22.PubMedCrossRef
382.
go back to reference Higano CS, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115(16):3670–9.PubMedCrossRef Higano CS, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115(16):3670–9.PubMedCrossRef
383.
go back to reference Keskin DB, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565(7738):234–9.PubMedCrossRef Keskin DB, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565(7738):234–9.PubMedCrossRef
385.
go back to reference Schadendorf D, et al. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol. 2006;17(4):563–70.PubMedCrossRef Schadendorf D, et al. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol. 2006;17(4):563–70.PubMedCrossRef
387.
go back to reference Besse B, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4): e1071008.PubMedCrossRef Besse B, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4): e1071008.PubMedCrossRef
388.
389.
go back to reference Escudier B, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10.PubMedPubMedCentralCrossRef Escudier B, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10.PubMedPubMedCentralCrossRef
390.
go back to reference Zitvogel L, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.PubMedCrossRef Zitvogel L, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.PubMedCrossRef
391.
go back to reference Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef
394.
go back to reference Tumino N, et al. Polymorphonuclear myeloid-derived suppressor cells impair the anti-tumor efficacy of GD2.CAR T-cells in patients with neuroblastoma. J Hematol Oncol. 2021;14(1):191.PubMedPubMedCentralCrossRef Tumino N, et al. Polymorphonuclear myeloid-derived suppressor cells impair the anti-tumor efficacy of GD2.CAR T-cells in patients with neuroblastoma. J Hematol Oncol. 2021;14(1):191.PubMedPubMedCentralCrossRef
396.
go back to reference Lai J, et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol. 2020;21(8):914–26.PubMedCrossRef Lai J, et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol. 2020;21(8):914–26.PubMedCrossRef
399.
400.
go back to reference Kang M, et al. Nanocomplex-mediated in vivo programming to chimeric antigen receptor-M1 macrophages for cancer therapy. Adv Mater. 2021;33(43): e2103258.PubMedCrossRef Kang M, et al. Nanocomplex-mediated in vivo programming to chimeric antigen receptor-M1 macrophages for cancer therapy. Adv Mater. 2021;33(43): e2103258.PubMedCrossRef
401.
go back to reference Hiruma Y, et al. Impaired osteoclast differentiation and function and mild osteopetrosis development in Siglec-15-deficient mice. Bone. 2013;53(1):87–93.PubMedCrossRef Hiruma Y, et al. Impaired osteoclast differentiation and function and mild osteopetrosis development in Siglec-15-deficient mice. Bone. 2013;53(1):87–93.PubMedCrossRef
402.
go back to reference Kameda Y, et al. Siglec-15 is a potential therapeutic target for postmenopausal osteoporosis. Bone. 2015;71:217–26.PubMedCrossRef Kameda Y, et al. Siglec-15 is a potential therapeutic target for postmenopausal osteoporosis. Bone. 2015;71:217–26.PubMedCrossRef
403.
go back to reference Angata T, et al. Siglec-15: an immune system Siglec conserved throughout vertebrate evolution. Glycobiology. 2007;17(8):838–46.PubMedCrossRef Angata T, et al. Siglec-15: an immune system Siglec conserved throughout vertebrate evolution. Glycobiology. 2007;17(8):838–46.PubMedCrossRef
404.
go back to reference Takamiya R, et al. The interaction between Siglec-15 and tumor-associated sialyl-Tn antigen enhances TGF-β secretion from monocytes/macrophages through the DAP12-Syk pathway. Glycobiology. 2013;23(2):178–87.PubMedCrossRef Takamiya R, et al. The interaction between Siglec-15 and tumor-associated sialyl-Tn antigen enhances TGF-β secretion from monocytes/macrophages through the DAP12-Syk pathway. Glycobiology. 2013;23(2):178–87.PubMedCrossRef
406.
407.
go back to reference Sun J, et al. Siglec-15 as an emerging target for next-generation cancer immunotherapy. Clin Cancer Res. 2021;27(3):680–8.PubMedCrossRef Sun J, et al. Siglec-15 as an emerging target for next-generation cancer immunotherapy. Clin Cancer Res. 2021;27(3):680–8.PubMedCrossRef
408.
go back to reference Shum E, et al. 490 clinical benefit through siglec-15 targeting with NC318 antibody in subjects with Siglec-15 positive advanced solid tumors. J Immunother Cancer. 2021;9(Suppl 2):A520–1.CrossRef Shum E, et al. 490 clinical benefit through siglec-15 targeting with NC318 antibody in subjects with Siglec-15 positive advanced solid tumors. J Immunother Cancer. 2021;9(Suppl 2):A520–1.CrossRef
410.
411.
412.
go back to reference Yao Y, et al. TREM-2 serves as a negative immune regulator through Syk pathway in an IL-10 dependent manner in lung cancer. Oncotarget. 2016;7(20):29620–34.PubMedPubMedCentralCrossRef Yao Y, et al. TREM-2 serves as a negative immune regulator through Syk pathway in an IL-10 dependent manner in lung cancer. Oncotarget. 2016;7(20):29620–34.PubMedPubMedCentralCrossRef
413.
go back to reference Jahchan NS, et al. Abstract LB071: Tuning the tumor myeloid microenvironment (TME) by targeting TREM2+ tumor-associated macrophages to overcome resistance to immune checkpoint inhibitors. Cancer Res. 2021;81(13_Supplement):LB071–LB071.CrossRef Jahchan NS, et al. Abstract LB071: Tuning the tumor myeloid microenvironment (TME) by targeting TREM2+ tumor-associated macrophages to overcome resistance to immune checkpoint inhibitors. Cancer Res. 2021;81(13_Supplement):LB071–LB071.CrossRef
414.
go back to reference Zhang X, et al. High TREM2 expression correlates with poor prognosis in gastric cancer. Hum Pathol. 2018;72:91–9.PubMedCrossRef Zhang X, et al. High TREM2 expression correlates with poor prognosis in gastric cancer. Hum Pathol. 2018;72:91–9.PubMedCrossRef
415.
go back to reference Suzuki H, et al. A role for macrophage scavenger receptors in atherosclerosis and susceptibility to infection. Nature. 1997;386(6622):292–6.PubMedCrossRef Suzuki H, et al. A role for macrophage scavenger receptors in atherosclerosis and susceptibility to infection. Nature. 1997;386(6622):292–6.PubMedCrossRef
416.
go back to reference Shi B, et al. The scavenger receptor MARCO expressed by tumor-associated macrophages are highly associated with poor pancreatic cancer prognosis. Front Oncol. 2021;11: 771488.PubMedPubMedCentralCrossRef Shi B, et al. The scavenger receptor MARCO expressed by tumor-associated macrophages are highly associated with poor pancreatic cancer prognosis. Front Oncol. 2021;11: 771488.PubMedPubMedCentralCrossRef
417.
go back to reference Chen AX, et al. Single-cell characterization of macrophages in glioblastoma reveals MARCO as a mesenchymal pro-tumor marker. Genome Med. 2021;13(1):88.PubMedPubMedCentralCrossRef Chen AX, et al. Single-cell characterization of macrophages in glioblastoma reveals MARCO as a mesenchymal pro-tumor marker. Genome Med. 2021;13(1):88.PubMedPubMedCentralCrossRef
418.
go back to reference Jeremiasen M, et al. Tumor-associated CD68(+), CD163(+), and MARCO(+) macrophages as prognostic biomarkers in patients with treatment-naïve gastroesophageal adenocarcinoma. Front Oncol. 2020;10: 534761.PubMedPubMedCentralCrossRef Jeremiasen M, et al. Tumor-associated CD68(+), CD163(+), and MARCO(+) macrophages as prognostic biomarkers in patients with treatment-naïve gastroesophageal adenocarcinoma. Front Oncol. 2020;10: 534761.PubMedPubMedCentralCrossRef
419.
go back to reference La Fleur L, et al. Expression of scavenger receptor MARCO defines a targetable tumor-associated macrophage subset in non-small cell lung cancer. Int J Cancer. 2018;143(7):1741–52.PubMedCrossRef La Fleur L, et al. Expression of scavenger receptor MARCO defines a targetable tumor-associated macrophage subset in non-small cell lung cancer. Int J Cancer. 2018;143(7):1741–52.PubMedCrossRef
420.
go back to reference Georgoudaki AM, et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 2016;15(9):2000–11.PubMedCrossRef Georgoudaki AM, et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 2016;15(9):2000–11.PubMedCrossRef
421.
go back to reference Eisinger S, et al. Targeting a scavenger receptor on tumor-associated macrophages activates tumor cell killing by natural killer cells. Proc Natl Acad Sci U S A. 2020;117(50):32005–16.PubMedPubMedCentralCrossRef Eisinger S, et al. Targeting a scavenger receptor on tumor-associated macrophages activates tumor cell killing by natural killer cells. Proc Natl Acad Sci U S A. 2020;117(50):32005–16.PubMedPubMedCentralCrossRef
422.
go back to reference Xing Q, et al. Scavenger receptor MARCO contributes to macrophage phagocytosis and clearance of tumor cells. Exp Cell Res. 2021;408(2): 112862.PubMedCrossRef Xing Q, et al. Scavenger receptor MARCO contributes to macrophage phagocytosis and clearance of tumor cells. Exp Cell Res. 2021;408(2): 112862.PubMedCrossRef
423.
go back to reference Yoshiizumi K, et al. Studies on scavenger receptor inhibitors. Part 1: synthesis and structure-activity relationships of novel derivatives of sulfatides. Bioorg Med Chem. 2002;10(8):2445–60.PubMedCrossRef Yoshiizumi K, et al. Studies on scavenger receptor inhibitors. Part 1: synthesis and structure-activity relationships of novel derivatives of sulfatides. Bioorg Med Chem. 2002;10(8):2445–60.PubMedCrossRef
425.
go back to reference Colonna M, et al. Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules. J Immunol. 1998;160(7):3096–100.PubMed Colonna M, et al. Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules. J Immunol. 1998;160(7):3096–100.PubMed
426.
427.
go back to reference Zhang Y, Zheng J. Functions of immune checkpoint molecules beyond immune evasion. Adv Exp Med Biol. 2020;1248:201–26.PubMedCrossRef Zhang Y, Zheng J. Functions of immune checkpoint molecules beyond immune evasion. Adv Exp Med Biol. 2020;1248:201–26.PubMedCrossRef
428.
go back to reference Chen HM, et al. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest. 2018;128(12):5647–62.PubMedPubMedCentralCrossRef Chen HM, et al. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest. 2018;128(12):5647–62.PubMedPubMedCentralCrossRef
429.
go back to reference Carbone C, et al. An angiopoietin-like protein 2 autocrine signaling promotes EMT during pancreatic ductal carcinogenesis. Oncotarget. 2015;6(15):13822–34.PubMedCrossRef Carbone C, et al. An angiopoietin-like protein 2 autocrine signaling promotes EMT during pancreatic ductal carcinogenesis. Oncotarget. 2015;6(15):13822–34.PubMedCrossRef
430.
go back to reference Siu LL, et al. First-in-class anti-immunoglobulin-like transcript 4 myeloid-specific antibody MK-4830 abrogates a PD-1 resistance mechanism in patients with advanced solid tumors. Clin Cancer Res. 2022;28(1):57–70.PubMedCrossRef Siu LL, et al. First-in-class anti-immunoglobulin-like transcript 4 myeloid-specific antibody MK-4830 abrogates a PD-1 resistance mechanism in patients with advanced solid tumors. Clin Cancer Res. 2022;28(1):57–70.PubMedCrossRef
431.
432.
go back to reference Karikoski M, et al. Clever-1/stabilin-1 controls cancer growth and metastasis. Clin Cancer Res. 2014;20(24):6452–64.PubMedCrossRef Karikoski M, et al. Clever-1/stabilin-1 controls cancer growth and metastasis. Clin Cancer Res. 2014;20(24):6452–64.PubMedCrossRef
433.
go back to reference Virtakoivu R, et al. Systemic blockade of clever-1 elicits lymphocyte activation alongside checkpoint molecule downregulation in patients with solid tumors: results from a phase I/II clinical trial. Clin Cancer Res. 2021;27(15):4205–20.PubMedCrossRef Virtakoivu R, et al. Systemic blockade of clever-1 elicits lymphocyte activation alongside checkpoint molecule downregulation in patients with solid tumors: results from a phase I/II clinical trial. Clin Cancer Res. 2021;27(15):4205–20.PubMedCrossRef
434.
go back to reference Wang B, et al. Microlocalization and clinical significance of stabilin-1(+) macrophages in treatment-naïve patients with urothelial carcinoma of the bladder. World J Urol. 2020;38(3):709–16.PubMedCrossRef Wang B, et al. Microlocalization and clinical significance of stabilin-1(+) macrophages in treatment-naïve patients with urothelial carcinoma of the bladder. World J Urol. 2020;38(3):709–16.PubMedCrossRef
435.
go back to reference Bono P, et al. LBA38 Bexmarilimab, a novel macrophage re-programmer shows promising anti-tumour activity in phase I/II trial in several last line solid tumour types. Ann Oncol. 2021;32:S1313.CrossRef Bono P, et al. LBA38 Bexmarilimab, a novel macrophage re-programmer shows promising anti-tumour activity in phase I/II trial in several last line solid tumour types. Ann Oncol. 2021;32:S1313.CrossRef
Metadata
Title
Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy
Authors
Yi Wang
Kai Conrad Cecil Johnson
Margaret E. Gatti-Mays
Zihai Li
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2022
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-022-01335-y

Other articles of this Issue 1/2022

Journal of Hematology & Oncology 1/2022 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