Khám Phá Các Chất Ức Chế IDO1: Từ Thí Nghiệm Đến Lâm Sàng
Tóm tắt
Từ khóa
Tài liệu tham khảo
Prendergast, 2007, Cancer immunologists and cancer biologists: why we didn't talk then but need to now, Cancer Res, 67, 3500, 10.1158/0008-5472.CAN-06-4626
Prendergast, 2012, Immunological thought in the mainstream of cancer research: Past divorce, recent remarriage and elective affinities of the future, Oncoimmunology, 1, 793, 10.4161/onci.20909
Prendergast, 2013, Cancer immunotherapy: immune suppression and tumor growth, 2nd ed
Prendergast, 2014, Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer, Cancer Immunol Immunother, 63, 721, 10.1007/s00262-014-1549-4
Munn, 2016, IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance, Trends Immunol, 37, 193, 10.1016/j.it.2016.01.002
Muller, 2005, Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy, Nat Med, 11, 312, 10.1038/nm1196
Hou, 2007, Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses, Cancer Res, 67, 792, 10.1158/0008-5472.CAN-06-2925
Holmgaard, 2013, Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4, J Exp Med, 210, 1389, 10.1084/jem.20130066
Monjazeb, 2016, Blocking indolamine-2,3-dioxygenase rebound immune suppression boosts antitumor effects of radio-immunotherapy in murine models and spontaneous canine malignancies, Clin Cancer Res, 22, 4328, 10.1158/1078-0432.CCR-15-3026
Godin-Ethier, 2011, Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives, Clin Cancer Res, 17, 6985, 10.1158/1078-0432.CCR-11-1331
Smith, 2012, IDO is a nodal pathogenic driver of lung cancer and metastasis development, Cancer Discov, 2, 722, 10.1158/2159-8290.CD-12-0014
Mondal, 2016, IDO1 is an integral mediator of inflammatory neovascularization, EBioMedicine, 14, 74, 10.1016/j.ebiom.2016.11.013
Boyland, 1956, The metabolism of tryptophan. 2. The metabolism of tryptophan in patients suffering from cancer of the bladder, Biochem J, 64, 578, 10.1042/bj0640578
Yoshida, 1981, Induction of pulmonary indoleamine 2,3-dioxygenase by interferon, Proc Natl Acad Sci U S A, 78, 129, 10.1073/pnas.78.1.129
Munn, 1998, Prevention of allogeneic fetal rejection by tryptophan catabolism, Science, 281, 1191, 10.1126/science.281.5380.1191
Munn, 1999, Inhibition of T cell proliferation by macrophage tryptophan catabolism, J Exp Med, 189, 1363, 10.1084/jem.189.9.1363
Mellor, 1999, Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation?, Immunol Today, 20, 469, 10.1016/S0167-5699(99)01520-0
Muller, 2005, Marrying immunotherapy with chemotherapy: why say IDO?, Cancer Res, 65, 8065, 10.1158/0008-5472.CAN-05-2213
Muller, 2006, Targeting the mechanisms of tumoral immune tolerance with small-molecule inhibitors, Nat Rev Cancer, 6, 613, 10.1038/nrc1929
Munn, 2007, Indoleamine 2,3-dioxygenase and tumor-induced tolerance, J Clin Invest, 117, 1147, 10.1172/JCI31178
Prendergast, 2008, Immune escape as a fundamental trait of cancer: focus on IDO, Oncogene, 27, 3889, 10.1038/onc.2008.35
Uyttenhove, 2003, Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase, Nat Med, 9, 1269, 10.1038/nm934
Theate, 2015, Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues, Cancer Immunol Res, 3, 161, 10.1158/2326-6066.CIR-14-0137
Prendergast, 2009, BAR the door: cancer suppression by amphiphysin-like genes, Biochim Biophys Acta, 1795, 25
Ge, 1999, Mechanism for elimination of a tumor suppressor: aberrant splicing of a brain-specific exon causes loss of function of Bin1 in melanoma, Proc Natl Acad Sci U S A, 96, 9689, 10.1073/pnas.96.17.9689
Pineda-Lucena, 2005, A structure-based model of the c-Myc/Bin1 protein interaction shows alternative splicing of Bin1 and c-Myc phosphorylation are key binding determinants, J Mol Biol, 351, 182, 10.1016/j.jmb.2005.05.046
Karni, 2007, The gene encoding the splicing factor SF2/ASF is a proto-oncogene, Nat Struct Mol Biol, 14, 185, 10.1038/nsmb1209
Anczukow, 2012, The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation, Nat Struct Mol Biol, 19, 220, 10.1038/nsmb.2207
Golan-Gerstl, 2011, Splicing factor hnRNP A2/B1 regulates tumor suppressor gene splicing and is an oncogenic driver in glioblastoma, Cancer Res, 71, 4464, 10.1158/0008-5472.CAN-10-4410
Barekati, 2012, Methylation signature of lymph node metastases in breast cancer patients, BMC Cancer, 12, 244, 10.1186/1471-2407-12-244
Radpour, 2011, Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer, PLoS One, 6, e16080, 10.1371/journal.pone.0016080
Radpour, 2009, Methylation profiles of 22 candidate genes in breast cancer using high-throughput MALDI-TOF mass array, Oncogene, 28, 2969, 10.1038/onc.2009.149
Kuznetsova, 2007, Methylation of the BIN1 gene promoter CpG island associated with breast and prostate cancer, J Carcinog, 6, 9, 10.1186/1477-3163-6-9
McKenna, 2012, Epigenetic inactivation of the tumor suppressor BIN1 drives proliferation of SNF5-deficient tumors, Cell Cycle, 11, 1956, 10.4161/cc.20280
Muller, 2010, Non-hematopoietic expression of IDO is integrally required for inflammatory tumor promotion, Cancer Immunol Immunother, 59, 1655, 10.1007/s00262-010-0891-4
Prendergast, 2014, IDO in inflammatory programming and immune suppression in cancer, Tumor-induced immune suppression, 10.1007/978-1-4899-8056-4_11
Fallarino, 2002, T cell apoptosis by tryptophan catabolism, Cell Death Differ, 9, 1069, 10.1038/sj.cdd.4401073
Frumento, 2002, Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase, J Exp Med, 196, 459, 10.1084/jem.20020121
Terness, 2002, Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites, J Exp Med, 196, 447, 10.1084/jem.20020052
Munn, 2002, Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase, Science, 297, 1867, 10.1126/science.1073514
Munn, 2005, GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase, Immunity, 22, 633, 10.1016/j.immuni.2005.03.013
Della Chiesa, 2006, The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function, Blood, 108, 4118, 10.1182/blood-2006-03-006700
Mellor, 2004, Specific subsets of murine dendritic cells acquire potent T cell regulatory functions following CTLA4-mediated induction of indoleamine 2,3 dioxygenase, Int Immunol, 16, 1391, 10.1093/intimm/dxh140
Mellor, 2008, Creating immune privilege: active local suppression that benefits friends, but protects foes, Nat Rev Immunol, 8, 74, 10.1038/nri2233
Grohmann, 2003, Tolerance, DCs and tryptophan: much ado about IDO, Trends Immunol, 24, 242, 10.1016/S1471-4906(03)00072-3
Mellor, 2004, IDO expression by dendritic cells: tolerance and tryptophan catabolism, Nat Rev Immunol, 4, 762, 10.1038/nri1457
Fallarino, 2006, The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells, J Immunol, 176, 6752, 10.4049/jimmunol.176.11.6752
Opitz, 2011, An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor, Nature, 478, 197, 10.1038/nature10491
Muller, 2008, Chronic inflammation that facilitates tumor progression creates local immune suppression by inducing indoleamine 2,3 dioxygenase, Proc Natl Acad Sci U S A, 105, 17073, 10.1073/pnas.0806173105
Beatty, 2001, Regulation of tumor growth by IFN-gamma in cancer immunotherapy, Immunol Res, 24, 201, 10.1385/IR:24:2:201
Ancrile, 2007, Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis, Genes Dev, 21, 1714, 10.1101/gad.1549407
Dougan, 2011, A dual role for the immune response in a mouse model of inflammation-associated lung cancer, J Clin Invest, 121, 2436, 10.1172/JCI44796
Qin, 2000, CD4+ T cell–mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN gamma receptor expression by nonhematopoietic cells, Immunity, 12, 677, 10.1016/S1074-7613(00)80218-6
Qin, 2003, A critical requirement of interferon gamma-mediated angiostasis for tumor rejection by CD8+ T cells, Cancer Res, 63, 4095
Chevolet, 2015, Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma, Oncoimmunology, 4, e982382, 10.4161/2162402X.2014.982382
Chang, 2011, Cardiac and gastrointestinal liabilities caused by deficiency in the immune modulatory enzyme indoleamine 2,3-dioxygenase, Cancer Biol Ther, 12, 1050, 10.4161/cbt.12.12.18142
Balachandran, 2011, Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido, Nat Med, 17, 1094, 10.1038/nm.2438
Yeung, 2015, Role of indoleamine 2,3-dioxygenase in health and disease, Clin Sci, 129, 601, 10.1042/CS20140392
Rohrig, 2015, Challenges in the discovery of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors, J Med Chem, 58, 9421, 10.1021/acs.jmedchem.5b00326
Brochez, 2017, The rationale of indoleamine 2,3-dioxygenase inhibition for cancer therapy, Eur J Cancer, 76, 167, 10.1016/j.ejca.2017.01.011
Zhai, 2015, Molecular pathways: targeting IDO1 and other tryptophan dioxygenases for cancer immunotherapy, Clin Cancer Res, 21, 5427, 10.1158/1078-0432.CCR-15-0420
van Baren, 2015, Tumoral immune resistance mediated by enzymes that degrade tryptophan, Cancer Immunol Res, 3, 978, 10.1158/2326-6066.CIR-15-0095
Selvan, 2016, Indoleamine 2,3-dioxygenase (IDO): biology and target in cancer immunotherapies, Curr Cancer Drug Targets, 16, 755, 10.2174/1568009615666151030102250
Li, 2017, IDO1: an important immunotherapy target in cancer treatment, Int Immunopharmacol, 47, 70, 10.1016/j.intimp.2017.03.024
Cady, 1991, 1-methyl-DL-tryptophan, beta-(3-benzofuranyl)-DL-alanine (the oxygen analog of tryptophan), and beta-[3-benzo(b)thienyl]-DL-alanine (the sulfur analog of tryptophan) are competitive inhibitors for indoleamine 2,3-dioxygenase, Arch Biochem Biophys, 291, 326, 10.1016/0003-9861(91)90142-6
Peterson, 1994, Evaluation of functionalized tryptophan derivatives and related compounds as competitive inhibitors of indoleamine 2,3-dioxygenase, Med Chem Res, 3, 531
Friberg, 2002, Indoleamine 2,3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection, Int J Cancer, 101, 151, 10.1002/ijc.10645
Cheever, 2008, Twelve immunotherapy drugs that could cure cancers, Immunol Rev, 222, 357, 10.1111/j.1600-065X.2008.00604.x
Soliman, 2014, A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors, Oncotarget, 5, 8136, 10.18632/oncotarget.2357
Metz, 2012, IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: a novel IDO effector pathway targeted by D-1-methyl-tryptophan, Oncoimmunology, 1, 1460, 10.4161/onci.21716
Mautino MR, Kumar S, Zhuang H, Waldo J, Jaipuri F, Potturi H, et al A novel prodrug of indoximod with enhanced pharmacokinetic properties. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1–5; Washington, DC. Philadelphia (PA): AACR; 2017. Abstract nr 4076.
Colwell, 2017, Indoximod combo triggers responses in melanoma, Cancer Discov, 7, 542, 10.1158/2159-8290.CD-NB2017-056
Fu, 2011, The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy, Cancer Res, 71, 5445, 10.1158/0008-5472.CAN-11-1138
Xie, 2012, Tumor suppressor TSC1 is critical for T-cell anergy, Proc Natl Acad Sci U S A, 109, 14152, 10.1073/pnas.1119744109
Soliman, 2016, A phase I study of indoximod in patients with advanced malignancies, Oncotarget, 7, 22928, 10.18632/oncotarget.8216
Zakharia, 2017, Interim analysis of the Phase 2 clinical trial of the IDO pathway inhibitor indoximod in combination with pembrolizumab for patients with advanced melanoma, Cancer Res, 77, CT117, 10.1158/1538-7445.AM2017-CT117
Zahkaria Y . Combined inhibition of the IDO and PD-1 pathways improves the response rate for patients with advanced melanoma. In: Proceedings of the Third CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference; 2017 Sep 6–9; Mainz, Germany. Mainz, Germany: CIMT; 2017.
Jha, 2017, A phase II randomized, double-blind study of sipuleucel-T followed by IDO pathway inhibitor, indoximod, or placebo in the treatment of patients with metastatic castration resistant prostate cancer (mCRPC), J Clin Oncol, 35, 3066, 10.1200/JCO.2017.35.15_suppl.3066
Emadi A, Holtzman NG, Imran M, El-Chaer F, Koka M, Singh Z, et al Indoximod in combination with idarubicin and cytarabine for upfront treatment of patients with newly diagnosed acute myeloid leukemia (AML): Phase 1 report. In: Proceedings of the 22nd Congress of the European Hematology Association; 2017 Jun 22–25; Madrid, Spain. The Hague, the Netherlands: EHA; 2017. Abstract nr E912.
Bahary, Phase 2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus gemcitabine/nab-paclitaxel for the treatment of metastatic pancreas cancer: interim analysis, J Clin Oncol, 34
Gaspari, 2006, Structure-activity study of brassinin derivatives as indoleamine 2,3-dioxygenase inhibitors, J Med Chem, 49, 684, 10.1021/jm0508888
Banerjee, 2008, A key in vivo antitumor mechanism of action of natural product-based brassinins is inhibition of indoleamine 2,3-dioxygenase, Oncogene, 27, 2851, 10.1038/sj.onc.1210939
Kumar, 2008, Structure based development of phenylimidazole-derived inhibitors of indoleamine 2,3-dioxygenase, J Med Chem, 51, 4968, 10.1021/jm800512z
Kumar, 2008, Indoleamine 2,3-dioxygenase is the anticancer target for a novel series of potent naphthoquinone-based inhibitors, J Med Chem, 51, 1706, 10.1021/jm7014155
Sono, 1989, Enzyme kinetic and spectroscopic studies of inhibitor and effector interactions with indoleamine 2,3-dioxygenase. 1. Norharman and 4-phenylimidazole binding to the enzyme as inhibitors and heme ligands, Biochemistry, 28, 5392, 10.1021/bi00439a012
Metz, 2014, IDO2 is critical for IDO1-mediated T cell regulation and exerts a non-redundant function in inflammation, Int Immunol, 26, 357, 10.1093/intimm/dxt073
Sugimoto, 2006, Crystal structure of human indoleamine 2,3-dioxygenase: catalytic mechanism of O2 incorporation by a heme-containing dioxygenase, Proc Natl Acad Sci U S A, 103, 2611, 10.1073/pnas.0508996103
Rohrig, 2012, Rational design of 4-aryl-1,2,3-triazoles for indoleamine 2,3-dioxygenase 1 inhibition, J Med Chem, 55, 5270, 10.1021/jm300260v
Mautino, 2013, NLG919, a novel indoleamine-2,3-dioxygenase (IDO)-pathway inhibitor drug candidate for cancer therapy, Cancer Res, 73, 491, 10.1158/1538-7445.AM2013-491
Nayak A, Hao Z, Sadek R, Dobbins R, Marshall L, Vahanian NN, et al Phase 1a study of the safety, pharmacokinetics, and pharmacodynamics of GDC-0919 in patients with recurrent/advanced solid tumors. In: Proceedings of the ECCO-ESMO 2015; 2015 Sep 25; Vienna, Austria. Lugano, Switzerland: ESMO; 2015.
Peng, 2016, Important Hydrogen Bond Networks in Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitor Design Revealed by Crystal Structures of Imidazoleisoindole Derivatives with IDO1, J Med Chem, 59, 282, 10.1021/acs.jmedchem.5b01390
Spahn, 2015, Improved anti-tumor immunity and efficacy upon combination of the IDO1 inhibitor GDC-0919 with anti-PD-L1 blockade versus anti-PD-L1 alone in preclinical tumor models, J Immunother Canc, 3, 303, 10.1186/2051-1426-3-S2-P303
Burris, 2017, A Phase 1b dose escalation study of combined inhibition of IDO1 (GDC-0919) and PD-L1 (atezolizumab) in patients with locally advanced or metastatic solid tumors, J Clin Oncol, 35, 10.1200/JCO.2017.35.15_suppl.105
Halford, 2015, Drug candidates unveiled at ‘First-Time Disclosures’ symposium in Boston, Chem Eng News, 93, 38, 10.1021/cen-09338-scitech1
Yue, 2017, INCB24360 (epacadostat), a highly potent and selective indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor for immuno-oncology, ACS Med Chem Lett, 8, 486, 10.1021/acsmedchemlett.6b00391
Yue, 2009, Discovery of potent competitive inhibitors of indoleamine 2,3-dioxygenase with in vivo pharmacodynamic activity and efficacy in a mouse melanoma model, J Med Chem, 52, 7364, 10.1021/jm900518f
Lipinski, 1997, Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings, Adv Drug Deliv Rev, 23, 3, 10.1016/S0169-409X(96)00423-1
Veber, 2002, Molecular properties that influence the oral bioavailability of drug candidates, J Med Chem, 45, 2615, 10.1021/jm020017n
Liu, 2010, Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity, Blood, 115, 3520, 10.1182/blood-2009-09-246124
Spranger, 2014, Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment, J Immunother Cancer, 2, 3, 10.1186/2051-1426-2-3
Beatty, 2017, First-in-human phase 1 study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies, Clin Cancer Res, 23, 3269, 10.1158/1078-0432.CCR-16-2272
Kristeleit, 2017, A randomised, open-label, phase 2 study of the IDO1 inhibitor epacadostat (INCB024360) versus tamoxifen as therapy for biochemically recurrent (CA-125 relapse)-only epithelial ovarian cancer, primary peritoneal carcinoma, or fallopian tube cancer, Gynecol Oncol, 146, 484, 10.1016/j.ygyno.2017.07.005
Rose, 2017, Epacadostat shows value in two SCCHN trials, Cancer Disc, 7, OF2, 10.1158/2159-8290.CD-NB2017-100
Gangadhar, 2016, Epacadostat plus pembrolizumab in patients with advanced melanoma and select solid tumors: updated phase 1 results from ECHO-202/KEYNOTE-037, Ann Oncol, 27, 379, 10.1093/annonc/mdw379.06
Perez, 2017, Epacadostat plus nivolumab in patients with advanced solid tumors: preliminary phase I/II results of ECHO-204, J Clin Oncol, 35, 3003, 10.1200/JCO.2017.35.15_suppl.3003
Siu LL . BMS-986205, an optimized indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, is well tolerated with potent pharmacodynamic (PD) activity, alone and in combination with nivolumab (nivo) in advanced cancers in a Phase 1/2A trial. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1–5; Washington, DC. Philadelphia (PA): AACR; 2017. Abstract nr CT116.
Cheng, 2014, Discovery and structure-activity relationships of phenyl benzenesulfonylhydrazides as novel indoleamine 2,3-dioxygenase inhibitors, Bioorg Med Chem Lett, 24, 3403, 10.1016/j.bmcl.2014.05.084
Lin, 2016, Phenyl benzenesulfonylhydrazides exhibit selective indoleamine 2,3-dioxygenase inhibition with potent in vivo pharmacodynamic activity and antitumor efficacy, J Med Chem, 59, 419, 10.1021/acs.jmedchem.5b01640
Platten, 2012, Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion, Cancer Res, 72, 5435, 10.1158/0008-5472.CAN-12-0569
Platten, 2014, Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors, Front Immunol, 5, 673
Salter, 1995, The effects of a novel and selective inhibitor of tryptophan 2,3-dioxygenase on tryptophan and serotonin metabolism in the rat, Biochem Pharmacol, 49, 1435, 10.1016/0006-2952(95)00006-L
Dolusic, 2011, Tryptophan 2,3-dioxygenase (tdo) inhibitors. 3-(2-(Pyridyl)ethenyl)indoles as potential anticancer immunomodulators, J Med Chem, 54, 5320, 10.1021/jm2006782
Pilotte, 2012, Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase, Proc Natl Acad Sci U S A, 109, 2497, 10.1073/pnas.1113873109
Pantouris, 2014, Antitumour agents as inhibitors of tryptophan 2,3-dioxygenase, Biochem Biophys Res Commun, 443, 28, 10.1016/j.bbrc.2013.11.037
Wu, 2015, Identification of substituted naphthotriazolediones as novel tryptophan 2,3-dioxygenase (TDO) inhibitors through structure-based virtual screening, J Med Chem, 58, 7807, 10.1021/acs.jmedchem.5b00921
Abdel-Magid, 2017, Targeting the inhibition of tryptophan 2,3-dioxygenase (TDO-2) for cancer treatment, ACS Med Chem Lett, 8, 11, 10.1021/acsmedchemlett.6b00458
Kanai, 2009, Tryptophan 2,3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice, Mol Brain, 2, 8, 10.1186/1756-6606-2-8
Bessede, 2014, Aryl hydrocarbon receptor control of a disease tolerance defence pathway, Nature, 511, 184, 10.1038/nature13323
Larkin, 2016, Tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase 1 make separate, tissue-specific contributions to basal and inflammation-induced kynurenine pathway metabolism in mice, Biochim Biophys Acta, 1860, 2345, 10.1016/j.bbagen.2016.07.002
D'Amato, 2015, A TDO2-AhR signaling axis facilitates anoikis resistance and metastasis in triple-negative breast cancer, Cancer Res, 75, 4651, 10.1158/0008-5472.CAN-15-2011
Prendergast, 2014, IDO2 in immunomodulation and autoimmune disease, Front Immunol, 5, 585, 10.3389/fimmu.2014.00585
Lob, 2009, IDO1 and IDO2 are expressed in human tumors: levo- but not dextro-1-methyl tryptophan inhibits tryptophan catabolism, Cancer Immunol Immunother, 58, 153, 10.1007/s00262-008-0513-6
Witkiewicz, 2009, Genotyping and expression analysis of IDO2 in human pancreatic cancer: a novel, active target, J Am Coll Surg, 208, 781, 10.1016/j.jamcollsurg.2008.12.018
Eldredge, 2013, Concurrent whole brain radiotherapy and short-course chloroquine in patients with brain metastases: a pilot trial, J Radiat Oncol, 2, 10.1007/s13566-013-0111-x
Li, 2016, Establishment of a human indoleamine 2, 3-dioxygenase 2 (hIDO2) bioassay system and discovery of tryptanthrin derivatives as potent hIDO2 inhibitors, Eur J Med Chem, 123, 171, 10.1016/j.ejmech.2016.07.013
Trabanelli, 2014, The SOCS3-independent expression of IDO2 supports the homeostatic generation of T regulatory cells by human dendritic cells, J Immunol, 192, 1231, 10.4049/jimmunol.1300720
Merlo, 2014, IDO2 Is a critical mediator of autoantibody production and inflammatory pathogenesis in a mouse model of autoimmune arthritis, J Immunol, 92, 2082, 10.4049/jimmunol.1303012
Metz, 2007, Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target of the antitumor indoleamine 2,3-dioxygenase inhibitory compound D-1-methyl-tryptophan, Cancer Res, 67, 7082, 10.1158/0008-5472.CAN-07-1872
Merlo, 2016, IDO2 modulates T cell-dependent autoimmune responses through a B cell-intrinsic mechanism, J Immunol, 196, 4487, 10.4049/jimmunol.1600141
Merlo, 2017, Therapeutic antibody targeting of indoleamine-2,3-dioxygenase (IDO2) inhibits autoimmune arthritis, Clin Immunol, 179, 8, 10.1016/j.clim.2017.01.016
Affara, 2014, B cells regulate macrophage phenotype and response to chemotherapy in squamous carcinomas, Cancer Cell, 25, 809, 10.1016/j.ccr.2014.04.026
Schioppa, 2011, B regulatory cells and the tumor-promoting actions of TNF-alpha during squamous carcinogenesis, Proc Natl Acad Sci U S A, 108, 10662, 10.1073/pnas.1100994108
Gunderson, 2016, Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer, Cancer Discov, 6, 270, 10.1158/2159-8290.CD-15-0827
Lee, 2016, Hif1a deletion reveals pro-neoplastic function of B cells in pancreatic neoplasia, Cancer Discov, 6, 256, 10.1158/2159-8290.CD-15-0822
Bankoti, 2010, Functional and phenotypic effects of AhR activation in inflammatory dendritic cells, Toxicol Appl Pharmacol, 246, 18, 10.1016/j.taap.2010.03.013
Simones, 2011, Consequences of AhR activation in steady-state dendritic cells, Toxicol Sci, 119, 293, 10.1093/toxsci/kfq354
Rohrig, 2016, 1,2,3-Triazoles as inhibitors of indoleamine 2,3-dioxygenase 2 (IDO2), Bioorg Med Chem Lett, 26, 4330, 10.1016/j.bmcl.2016.07.031
Austin, 2010, Biochemical characteristics and inhibitor selectivity of mouse indoleamine 2,3-dioxygenase-2, Amino Acids, 39, 565, 10.1007/s00726-010-0475-9
Bakmiwewa, 2012, Identification of selective inhibitors of indoleamine 2,3-dioxygenase 2, Bioorg Med Chem Lett, 22, 7641, 10.1016/j.bmcl.2012.10.010
Pantouris, 2014, Human indoleamine 2,3-dioxygenase-2 has substrate specificity and inhibition characteristics distinct from those of indoleamine 2,3-dioxygenase-1, Amino Acids, 46, 2155, 10.1007/s00726-014-1766-3