Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond

Abstract

l-Tryptophan (Trp) metabolism through the kynurenine pathway (KP) is involved in the regulation of immunity, neuronal function and intestinal homeostasis. Imbalances in Trp metabolism in disorders ranging from cancer to neurodegenerative disease have stimulated interest in therapeutically targeting the KP, particularly the main rate-limiting enzymes indoleamine-2,3-dioxygenase 1 (IDO1), IDO2 and tryptophan-2,3-dioxygenase (TDO) as well as kynurenine monooxygenase (KMO). However, although small-molecule IDO1 inhibitors showed promise in early-stage cancer immunotherapy clinical trials, a phase III trial was negative. This Review summarizes the physiological and pathophysiological roles of Trp metabolism, highlighting the vast opportunities and challenges for drug development in multiple diseases.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Tryptophan catabolism, key therapeutic targets and drugs in development.
Fig. 2: Tryptophan catabolism — key organs involved.
Fig. 3: Neuroactivity of tryptophan metabolites.
Fig. 4: Immunological effects of tryptophan metabolism.
Fig. 5: Different binding mechanisms of IDO1 inhibitors.

Similar content being viewed by others

References

  1. Cervenka, I., Agudelo, L. Z. & Ruas, J. L. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science 357, eaaf9794 (2017).

    PubMed  Google Scholar 

  2. Schwarcz, R. & Stone, T. W. The kynurenine pathway and the brain: challenges, controversies and promises. Neuropharmacology 112, 237–247 (2017).

    CAS  PubMed  Google Scholar 

  3. Le Floc’h, N., Otten, W. & Merlot, E. Tryptophan metabolism, from nutrition to potential therapeutic applications. Amino Acids 41, 1195–1205 (2011).

    PubMed  Google Scholar 

  4. Stone, T. W., Stoy, N. & Darlington, L. G. An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol. Sci. 34, 136–143 (2013).

    CAS  PubMed  Google Scholar 

  5. van der Goot, A. T. & Nollen, E. A. Tryptophan metabolism: entering the field of aging and age-related pathologies. Trends Mol. Med. 19, 336–344 (2013).

    PubMed  Google Scholar 

  6. Guillemin, G. J., Smythe, G., Takikawa, O. & Brew, B. J. Expression of indoleamine 2,3-dioxygenase and production of quinolinic acid by human microglia, astrocytes, and neurons. Glia 49, 15–23 (2005).

    PubMed  Google Scholar 

  7. Munn, D. H. & Mellor, A. L. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol. 37, 193–207 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Opitz, C. A., Wick, W., Steinman, L. & Platten, M. Tryptophan degradation in autoimmune diseases. Cell. Mol. Life Sci. 64, 2542–2563 (2007).

    CAS  PubMed  Google Scholar 

  9. Puccetti, P. et al. Accumulation of an endogenous tryptophan-derived metabolite in colorectal and breast cancers. PLOS ONE 10, e0122046 (2015).

    PubMed  PubMed Central  Google Scholar 

  10. Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).This paper identifies the AHR as a target of tumour-derived immunosuppressive Trp metabolism and a potential drug target in immuno-oncology.

    CAS  PubMed  Google Scholar 

  11. Vezzani, A., Gramsbergen, J. B., Speciale, C. & Schwarcz, R. Production of quinolinic acid and kynurenic acid by human glioma. Adv. Exp. Med. Biol. 294, 691–695 (1991).

    CAS  PubMed  Google Scholar 

  12. Roussel, G., Bessede, A., Klein, C., Maitre, M. & Mensah-Nyagan, A. G. Xanthurenic acid is localized in neurons in the central nervous system. Neuroscience 329, 226–238 (2016).

    CAS  PubMed  Google Scholar 

  13. Kennedy, P. J., Cryan, J. F., Dinan, T. G. & Clarke, G. Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 112, 399–412 (2017).

    CAS  PubMed  Google Scholar 

  14. Prendergast, G. C., Malachowski, W. J., Mondal, A., Scherle, P. & Muller, A. J. Indoleamine 2,3-dioxygenase and its therapeutic inhibition in cancer. Int. Rev. Cell Mol. Biol. 336, 175–203 (2018).

    PubMed  Google Scholar 

  15. Schwarcz, R., Bruno, J. P., Muchowski, P. J. & Wu, H. Q. Kynurenines in the mammalian brain: when physiology meets pathology. Nat. Rev. Neurosci. 13, 465–477 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Fazio, F. et al. Cinnabarinic acid and xanthurenic acid: Two kynurenine metabolites that interact with metabotropic glutamate receptors. Neuropharmacology 112, 365–372 (2017).

    CAS  PubMed  Google Scholar 

  17. Lovelace, M. D. et al. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology 112, 373–388 (2017).

    CAS  PubMed  Google Scholar 

  18. Munn, D. H. et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 281, 1191–1193 (1998).This paper is the first to demonstrate the role of Trp metabolism in immune tolerance.

    CAS  PubMed  Google Scholar 

  19. Mellor, A. L., Lemos, H. & Huang, L. Indoleamine 2,3-dioxygenase and tolerance: where are we now? Front. Immunol. 8, 1360 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Pilotte, L. et al. Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc. Natl Acad. Sci. USA 109, 2497–2502 (2012).This paper provides evidence for tumoural TDO as a drug target in immuno-oncology.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Della Chiesa, M. et al. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood 108, 4118–4125 (2006).

    PubMed  Google Scholar 

  22. Fallarino, F. et al. T cell apoptosis by tryptophan catabolism. Cell Death Differ. 9, 1069–1077 (2002).This important paper demonstrates that Trp metabolites actively suppress T cells.

    CAS  PubMed  Google Scholar 

  23. Frumento, G. et al. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J. Exp. Med. 196, 459–468 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Terness, P. et al. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J. Exp. Med. 196, 447–457 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Lee, G. K. et al. Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 107, 452–460 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Munn, D. H. et al. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J. Exp. Med. 189, 1363–1372 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Munn, D. H. et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 22, 633–642 (2005).This controversial paper demonstrates the ‘death by starvation’ mechanism of Trp degradation in regulating T cells.

    CAS  PubMed  Google Scholar 

  28. Sonner, J. K. et al. The stress kinase GCN2 does not mediate suppression of antitumor T cell responses by tryptophan catabolism in experimental melanomas. Oncoimmunology 5, e1240858 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Badawy, A. A., Namboodiri, A. M. & Moffett, J. R. The end of the road for the tryptophan depletion concept in pregnancy and infection. Clin. Sci. 130, 1327–1333 (2016).

    CAS  Google Scholar 

  30. Brenk, M. et al. Tryptophan deprivation induces inhibitory receptors ILT3 and ILT4 on dendritic cells favoring the induction of human CD4+CD25+Foxp3+T regulatory cells. J. Immunol. 183, 145–154 (2009).

    CAS  PubMed  Google Scholar 

  31. Chen, W., Liang, X., Peterson, A. J., Munn, D. H. & Blazar, B. R. The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J. Immunol. 181, 5396–5404 (2008).

    CAS  PubMed  Google Scholar 

  32. Chung, D. J. et al. Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells. Blood 114, 555–563 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Curti, A. et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+T regulatory cells. Blood 109, 2871–2877 (2007).

    CAS  PubMed  Google Scholar 

  34. Fallarino, F. et al. 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, 6752–6761 (2006).

    CAS  PubMed  Google Scholar 

  35. Hippen, K. L. et al. In vitro induction of human regulatory T cells using conditions of low tryptophan plus kynurenines. Am. J. Transplant. 17, 3098–3113 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Sharma, M. D. et al. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Invest. 117, 2570–2582 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mezrich, J. D. et al. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 185, 3190–3198 (2010).

    CAS  PubMed  Google Scholar 

  38. Stockinger, B., Di Meglio, P., Gialitakis, M. & Duarte, J. H. The aryl hydrocarbon receptor: multitasking in the immune system. Annu. Rev. Immunol. 32, 403–432 (2014).

    CAS  PubMed  Google Scholar 

  39. Marshall, N. B. & Kerkvliet, N. I. Dioxin and immune regulation: emerging role of aryl hydrocarbon receptor in the generation of regulatory T cells. Ann. NY Acad. Sci. 1183, 25–37 (2010).

    CAS  PubMed  Google Scholar 

  40. Quintana, F. J. & Sherr, D. H. Aryl hydrocarbon receptor control of adaptive immunity. Pharmacol. Rev. 65, 1148–1161 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. DiNatale, B. C. et al. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol. Sci. 115, 89–97 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Novikov, O. et al. An aryl hydrocarbon receptor-mediated amplification loop that enforces cell migration in ER-/PR-/Her2- human breast cancer cells. Mol. Pharmacol. 90, 674–688 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Lowe, M. M. et al. Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production. PLOS ONE 9, e87877 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Seok, S. H. et al. Trace derivatives of kynurenine potently activate the aryl hydrocarbon receptor (AHR). J. Biol. Chem. 293, 1994–2005 (2017).

    PubMed  PubMed Central  Google Scholar 

  45. Takenaka, M. C. & Quintana, F. J. Tolerogenic dendritic cells. Semin. Immunopathol. 39, 113–120 (2017).

    CAS  PubMed  Google Scholar 

  46. Li, Q., Harden, J. L., Anderson, C. D. & Egilmez, N. K. Tolerogenic phenotype of IFN-gamma-Induced IDO+dendritic cells is maintained via an autocrine IDO-kynurenine/AhR-IDO loop. J. Immunol. 197, 962–970 (2016).

    CAS  PubMed  Google Scholar 

  47. Nguyen, N. T. et al. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc. Natl Acad. Sci. USA 107, 19961–19966 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, J. H. & Lee, J. Indole as an intercellular signal in microbial communities. FEMS Microbiol. Rev. 34, 426–444 (2010).

    CAS  PubMed  Google Scholar 

  49. Favre, D. et al. Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Sci. Transl Med. 2, 32ra36 (2010).This paper demonstrates the importance of Trp metabolism in controlling HIV.

    PubMed  PubMed Central  Google Scholar 

  50. Shapiro, H., Thaiss, C. A., Levy, M. & Elinav, E. The cross talk between microbiota and the immune system: metabolites take center stage. Curr. Opin. Immunol. 30, 54–62 (2014).

    CAS  PubMed  Google Scholar 

  51. Zelante, T. et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).This paper identifies an important mechanism of how microbial Trp metabolism regulates intestinal barrier function.

    CAS  PubMed  Google Scholar 

  52. Gao, J. et al. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front. Cell. Infect. Microbiol. 8, 13 (2018).

    PubMed  PubMed Central  Google Scholar 

  53. Rothhammer, V. et al. Microglial control of astrocytes in response to microbial metabolites. Nature 557, 724–728 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Rothhammer, V., Mascanfroni, I. D. & Bunse, L. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 22, 586–597 (2016).This paper provides an important link between microbial Trp metabolism and brain inflammation.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Opitz, C. A. & Heiland, I. Dynamics of NAD-metabolism: everything but constant. Biochem. Soc. Trans. 43, 1127–1132 (2015).

    CAS  PubMed  Google Scholar 

  56. Chiarugi, A., Dolle, C., Felici, R. & Ziegler, M. The NAD metabolome—a key determinant of cancer cell biology. Nat. Rev. Cancer 12, 741–752 (2012).

    CAS  PubMed  Google Scholar 

  57. Shi, H. et al. NAD deficiency, congenital malformations, and niacin supplementation. N. Engl. J. Med. 377, 544–552 (2017).

    CAS  PubMed  Google Scholar 

  58. Bender, D. A. & Olufunwa, R. Utilization of tryptophan, nicotinamide and nicotinic acid as precursors for nicotinamide nucleotide synthesis in isolated rat liver cells. Br. J. Nutr. 59, 279–287 (1988).

    CAS  PubMed  Google Scholar 

  59. Braidy, N., Guillemin, G. J. & Grant, R. Effects of kynurenine pathway inhibition on NAD metabolism and cell viability in human primary astrocytes and neurons. Int. J. Tryptophan Res. 4, 29–37 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Grant, R. & Kapoor, V. Inhibition of indoleamine 2,3-dioxygenase activity in IFN-gamma stimulated astroglioma cells decreases intracellular NAD levels. Biochem. Pharmacol. 66, 1033–1036 (2003).

    CAS  PubMed  Google Scholar 

  61. Grant, R., Nguyen, S. & Guillemin, G. Kynurenine pathway metabolism is involved in the maintenance of the intracellular NAD concentration in human primary astrocytes. Int. J. Tryptophan Res. 3, 151–156 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Soultoukis, G. A. & Partridge, L. Dietary protein, metabolism, and aging. Annu. Rev. Biochem. 85, 5–34 (2016).

    CAS  PubMed  Google Scholar 

  63. van der Goot, A. T. et al. Delaying aging and the aging-associated decline in protein homeostasis by inhibition of tryptophan degradation. Proc. Natl Acad. Sci. USA 109, 14912–14917 (2012).This paper provides a novel mechanism of how Trp metabolism regulates neurodegeneration and ageing.

    PubMed  PubMed Central  Google Scholar 

  64. Frick, B., Schroecksnadel, K., Neurauter, G., Leblhuber, F. & Fuchs, D. Increasing production of homocysteine and neopterin and degradation of tryptophan with older age. Clin. Biochem. 37, 684–687 (2004).

    CAS  PubMed  Google Scholar 

  65. Pertovaara, M. et al. Indoleamine 2,3-dioxygenase activity in nonagenarians is markedly increased and predicts mortality. Mech. Ageing Dev. 127, 497–499 (2006).

    CAS  PubMed  Google Scholar 

  66. Marcos-Perez, D. et al. Frailty status in older adults is related to alterations in indoleamine 2,3-dioxygenase 1 and guanosine triphosphate cyclohydrolase I enzymatic pathways. J. Am. Med. Dir. Assoc. 18, 1049–1057 (2017).

    PubMed  Google Scholar 

  67. Sutphin, G. L. et al. Caenorhabditis elegans orthologs of human genes differentially expressed with age are enriched for determinants of longevity. Aging Cell 16, 672–682 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Oxenkrug, G. F. The extended life span of Drosophila melanogaster eye-color (white and vermilion) mutants with impaired formation of kynurenine. J. Neural Transm. 117, 23–26 (2010).

    CAS  PubMed  Google Scholar 

  69. Oxenkrug, G. F., Navrotskaya, V., Voroboyva, L. & Summergrad, P. Extension of life span of Drosophila melanogaster by the inhibitors of tryptophan-kynurenine metabolism. Fly 5, 307–309 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Edwards, C. et al. Mechanisms of amino acid-mediated lifespan extension in Caenorhabditis elegans. BMC Genet. 16, 8 (2015).

    PubMed  PubMed Central  Google Scholar 

  71. He, C. et al. Enhanced longevity by ibuprofen, conserved in multiple species, occurs in yeast through inhibition of tryptophan import. PLOS Genet. 10, e1004860 (2014).

    PubMed  PubMed Central  Google Scholar 

  72. Mirzaei, H., Suarez, J. A. & Longo, V. D. Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol. Metab. 25, 558–566 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Ooka, H., Segall, P. E. & Timiras, P. S. Histology and survival in age-delayed low-tryptophan-fed rats. Mech. Ageing Dev. 43, 79–98 (1988).

    CAS  PubMed  Google Scholar 

  74. Fang, E. F. et al. NAD(+) in aging: molecular mechanisms and translational implications. Trends Mol. Med. 23, 899–916 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhang, H. et al. NAD(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 352, 1436–1443 (2016).This paper links Trp metabolism to ageing.

    CAS  PubMed  Google Scholar 

  76. Kanai, M. et al. Tryptophan 2,3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice. Mol. Brain 2, 8 (2009).

    PubMed  PubMed Central  Google Scholar 

  77. Mellor, A. L. et al. Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. J. Immunol. 171, 1652–1655 (2003).

    CAS  PubMed  Google Scholar 

  78. Wyss-Coray, T. Ageing neurodegeneration and brain rejuvenation. Nature 539, 180–186 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Maddison, D. C. & Giorgini, F. The kynurenine pathway and neurodegenerative disease. Semin. Cell Dev. Biol. 40, 134–141 (2015).

    CAS  PubMed  Google Scholar 

  80. Chouraki, V. et al. Association of amine biomarkers with incident dementia and Alzheimer’s disease in the Framingham Study. Alzheimers Dement. 13, 1327–1336 (2017).

    PubMed  PubMed Central  Google Scholar 

  81. de Bie, J., Lim, C. K. & Guillemin, G. J. Kynurenines, gender and neuroinflammation; showcase schizophrenia. Neurotox. Res. 30, 285–294 (2016).

    PubMed  Google Scholar 

  82. Breda, C. et al. Tryptophan-2,3-dioxygenase (TDO) inhibition ameliorates neurodegeneration by modulation of kynurenine pathway metabolites. Proc. Natl Acad. Sci. USA 113, 5435–5440 (2016).This paper demonstrates the importance of TDO as a regulator of neurodegeneration.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Campesan, S. et al. The kynurenine pathway modulates neurodegeneration in a Drosophila model of Huntington’s disease. Curr. Biol. 21, 961–966 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Savitz, J. et al. Reduction of kynurenic acid to quinolinic acid ratio in both the depressed and remitted phases of major depressive disorder. Brain Behav. Immun. 46, 55–59 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Brundin, L. et al. An enzyme in the kynurenine pathway that governs vulnerability to suicidal behavior by regulating excitotoxicity and neuroinflammation. Transl Psychiatry 6, e865 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Bryleva, E. Y. & Brundin, L. Suicidality and activation of the kynurenine pathway of tryptophan metabolism. Curr. Top. Behav. Neurosci. 31, 269–284 (2017).

    PubMed  Google Scholar 

  87. Dantzer, R. Role of the kynurenine metabolism pathway in inflammation-induced depression: preclinical approaches. Curr. Top. Behav. Neurosci. 31, 117–138 (2017).

    PubMed  PubMed Central  Google Scholar 

  88. Fuertig, R. et al. Mouse chronic social stress increases blood and brain kynurenine pathway activity and fear behaviour: both effects are reversed by inhibition of indoleamine 2,3-dioxygenase. Brain Behav. Immun. 54, 59–72 (2016).

    CAS  PubMed  Google Scholar 

  89. Jeon, S. W. & Kim, Y. K. Inflammation-induced depression: Its pathophysiology and therapeutic implications. J. Neuroimmunol. 313, 92–98 (2017).

    CAS  PubMed  Google Scholar 

  90. Smith, A. K. et al. Association of a polymorphism in the indoleamine- 2,3-dioxygenase gene and interferon-alpha-induced depression in patients with chronic hepatitis C. Mol. Psychiatry 17, 781–789 (2012).

    CAS  PubMed  Google Scholar 

  91. Kegel, M. E. et al. Kynurenic acid and psychotic symptoms and personality traits in twins with psychiatric morbidity. Psychiatry Res. 247, 105–112 (2017).

    CAS  PubMed  Google Scholar 

  92. Sathyasaikumar, K. V. et al. Impaired kynurenine pathway metabolism in the prefrontal cortex of individuals with schizophrenia. Schizophr. Bull. 37, 1147–1156 (2011).

    PubMed  Google Scholar 

  93. Erhardt, S. et al. Kynurenic acid levels are elevated in the cerebrospinal fluid of patients with schizophrenia. Neurosci. Lett. 313, 96–98 (2001).

    CAS  PubMed  Google Scholar 

  94. DeAngeli, N. E. et al. Exposure to kynurenic acid during adolescence increases sign-tracking and impairs long-term potentiation in adulthood. Front. Behav. Neurosci. 8, 451 (2014).

    PubMed  Google Scholar 

  95. Pershing, M. L. et al. Elevated levels of kynurenic acid during gestation produce neurochemical, morphological, and cognitive deficits in adulthood: implications for schizophrenia. Neuropharmacology 90, 33–41 (2015).

    CAS  PubMed  Google Scholar 

  96. Kozak, R. et al. Reduction of brain kynurenic acid improves cognitive function. J. Neurosci. 34, 10592–10602 (2014).This paper links Trp metabolism to cognitive function.

    PubMed  PubMed Central  Google Scholar 

  97. Iaccarino, H. F., Suckow, R. F., Xie, S. & Bucci, D. J. The effect of transient increases in kynurenic acid and quinolinic acid levels early in life on behavior in adulthood: implications for schizophrenia. Schizophr. Res. 150, 392–397 (2013).

    PubMed  Google Scholar 

  98. Aoyama, N. et al. Association study between kynurenine 3-monooxygenase gene and schizophrenia in the Japanese population. Genes Brain Behav. 5, 364–368 (2006).

    CAS  PubMed  Google Scholar 

  99. Holtze, M. et al. Kynurenine 3-monooxygenase polymorphisms: relevance for kynurenic acid synthesis in patients with schizophrenia and healthy controls. J. Psychiatry Neurosci. 37, 53–57 (2012).

    PubMed  PubMed Central  Google Scholar 

  100. Lavebratt, C. et al. The KMO allele encoding Arg452 is associated with psychotic features in bipolar disorder type 1, and with increased CSF KYNA level and reduced KMO expression. Mol. Psychiatry 19, 334–341 (2014).

    CAS  PubMed  Google Scholar 

  101. Sellgren, C. M. et al. A genome-wide association study of kynurenic acid in cerebrospinal fluid: implications for psychosis and cognitive impairment in bipolar disorder. Mol. Psychiatry 21, 1342–1350 (2016).

    CAS  PubMed  Google Scholar 

  102. Yu, D. et al. The IDO inhibitor coptisine ameliorates cognitive impairment in a mouse model of Alzheimer’s disease. J. Alzheimers Dis. 43, 291–302 (2015).

    CAS  PubMed  Google Scholar 

  103. Woodling, N. S. et al. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer’s disease model mice. Brain 139, 2063–2081 (2016).This paper links a key anti-inflammatory pathway to neuronal Trp metabolism and neurodegeneration.

    PubMed  PubMed Central  Google Scholar 

  104. Wu, W. et al. Expression of tryptophan 2,3-dioxygenase and production of kynurenine pathway metabolites in triple transgenic mice and human Alzheimer’s disease brain. PLOS ONE 8, e59749 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Sathyasaikumar, K. V. et al. Xanthurenic acid formation from 3-hydroxykynurenine in the mammalian brain: neurochemical characterization and physiological effects. Neuroscience 367, 85–97 (2017).

    CAS  PubMed  Google Scholar 

  106. Pellicciari, R. et al. Modulators of the kynurenine pathway of tryptophan metabolism: synthesis and preliminary biological evaluation of (S)-4-(ethylsulfonyl)benzoylalanine, a potent and selective kynurenine aminotransferase II (KAT II) inhibitor. ChemMedChem 1, 528–531 (2006).

    CAS  PubMed  Google Scholar 

  107. Rossi, F. et al. Crystal structure-based selective targeting of the pyridoxal 5,-phosphate dependent enzyme kynurenine aminotransferase II for cognitive enhancement. J. Med. Chem. 53, 5684–5689 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Dounay, A. B. et al. Discovery of brain-penetrant, irreversible kynurenine aminotransferase II inhibitors for schizophrenia. ACS Med. Chem. Lett. 3, 187–192 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Nematollahi, A., Sun, G., Jayawickrama, G. S. & Church, W. B. Kynurenine aminotransferase isozyme inhibitors: a review. Int. J. Mol. Sci. 17, 946 (2016).

    PubMed Central  Google Scholar 

  110. Pellicciari, R. et al. Sequence variants in kynurenine aminotransferase II (KAT II) orthologs determine different potencies of the inhibitor S-ESBA. ChemMedChem 3, 1199–1202 (2008).

    CAS  PubMed  Google Scholar 

  111. Smith, J. R., Jamie, J. F. & Guillemin, G. J. Kynurenine-3-monooxygenase: a review of structure, mechanism, and inhibitors. Drug Discov. Today 21, 315–324 (2016).

    CAS  PubMed  Google Scholar 

  112. Hutchinson, J. P. et al. Structural and mechanistic basis of differentiated inhibitors of the acute pancreatitis target kynurenine-3-monooxygenase. Nat. Commun. 8, 15827 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Walker, A. L. et al. Development of a series of kynurenine 3-monooxygenase inhibitors leading to a clinical candidate for the treatment of acute pancreatitis. J. Med. Chem. 60, 3383–3404 (2017).

    CAS  PubMed  Google Scholar 

  114. Rover, S., Cesura, A. M., Huguenin, P., Kettler, R. & Szente, A. Synthesis and biochemical evaluation of N-(4-phenylthiazol-2-yl)benzenesulfonamides as high-affinity inhibitors of kynurenine 3-hydroxylase. J. Med. Chem. 40, 4378–4385 (1997).

    CAS  PubMed  Google Scholar 

  115. Zwilling, D. et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell 145, 863–874 (2011).This controversial paper links KMO as a drug target on monocytes to the treatment of neurodegeneration.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Justinova, Z. et al. Reducing cannabinoid abuse and preventing relapse by enhancing endogenous brain levels of kynurenic acid. Nat. Neurosci. 16, 1652–1661 (2013).This paper provides evidence for Trp metabolism as a mediator of drug addiction.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Pellicciari, R. et al. Modulation of the kynurine pathway of tryptophan metabolism in search for neuroprotective agents. Focus on kynurenine-3-hydroxylase. Adv. Exp. Med. Biol. 527, 621–628 (2003).

    CAS  PubMed  Google Scholar 

  118. Mole, D. J. et al. Kynurenine-3-monooxygenase inhibition prevents multiple organ failure in rodent models of acute pancreatitis. Nat. Med. 22, 202–209 (2016).This paper highlights KMO as a drug target in pancreatitis.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Toledo-Sherman, L. M. et al. Development of a series of aryl pyrimidine kynurenine monooxygenase inhibitors as potential therapeutic agents for the treatment of Huntington’s disease. J. Med. Chem. 58, 1159–1183 (2015).

    CAS  PubMed  Google Scholar 

  120. Beaumont, V. et al. The novel KMO inhibitor CHDI-340246 leads to a restoration of electrophysiological alterations in mouse models of Huntington’s disease. Exp. Neurol. 282, 99–118 (2016).

    CAS  PubMed  Google Scholar 

  121. Kim, H. T. et al. Structural basis for inhibitor-induced hydrogen peroxide production by kynurenine 3-monooxygenase. Cell Chem. Biol. 25, 426–438 (2018).

    CAS  PubMed  Google Scholar 

  122. Beconi, M. G. et al. Metabolism and pharmacokinetics of JM6 in mice: JM6 is not a prodrug for Ro-61-8048. Drug Metab. Dispos. 40, 2297–2306 (2012).

    CAS  PubMed  Google Scholar 

  123. Larkin, P. B. et al. 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, 2345–2354 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. McGaha, T. L. et al. Amino acid catabolism: a pivotal regulator of innate and adaptive immunity. Immunol. Rev. 249, 135–157 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Zelante, T., Pieraccini, G., Scaringi, L., Aversa, F. & Romani, L. Learning from other diseases: protection and pathology in chronic fungal infections. Semin. Immunopathol. 38, 239–248 (2016).

    CAS  PubMed  Google Scholar 

  126. Mellor, A. L. & Munn, D. H. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat. Rev. Immunol. 4, 762–774 (2004).

    CAS  PubMed  Google Scholar 

  127. Romani, L. Immunity to fungal infections. Nat. Rev. Immunol. 11, 275–288 (2011).

    CAS  PubMed  Google Scholar 

  128. Schmidt, S. V. & Schultze, J. L. New insights into IDO biology in bacterial and viral infections. Front. Immunol. 5, 384 (2014).

    PubMed  PubMed Central  Google Scholar 

  129. Agaugue, S., Perrin-Cocon, L., Coutant, F., Andre, P. & Lotteau, V. 1-Methyl-tryptophan can interfere with TLR signaling in dendritic cells independently of IDO activity. J. Immunol. 177, 2061–2071 (2006).

    CAS  PubMed  Google Scholar 

  130. Popov, A. & Schultze, J. L. IDO-expressing regulatory dendritic cells in cancer and chronic infection. J. Mol. Med. 86, 145–160 (2008).

    CAS  PubMed  Google Scholar 

  131. Volpi, C. et al. High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9-TRIF pathway. Nat. Commun. 4, 1852 (2013).

    PubMed  Google Scholar 

  132. Puccetti, P. On watching the watchers: IDO and type I/II IFN. Eur. J. Immunol. 37, 876–879 (2007).

    CAS  PubMed  Google Scholar 

  133. Hissong, B. D., Byrne, G. I., Padilla, M. L. & Carlin, J. M. Upregulation of interferon-induced indoleamine 2,3-dioxygenase in human macrophage cultures by lipopolysaccharide, muramyl tripeptide, and interleukin-1. Cell. Immunol. 160, 264–269 (1995).

    CAS  PubMed  Google Scholar 

  134. Fallarino, F. et al. Modulation of tryptophan catabolism by regulatory T cells. Nat. Immunol. 4, 1206–1212 (2003).

    CAS  PubMed  Google Scholar 

  135. Grohmann, U. et al. Reverse signaling through GITR ligand enables dexamethasone to activate IDO in allergy. Nat. Med. 13, 579–586 (2007).

    CAS  PubMed  Google Scholar 

  136. Carlin, J. M., Borden, E. C., Sondel, P. M. & Byrne, G. I. Interferon-induced indoleamine 2,3-dioxygenase activity in human mononuclear phagocytes. J. Leukoc. Biol. 45, 29–34 (1989).

    CAS  PubMed  Google Scholar 

  137. Murray, H. W. et al. Role of tryptophan degradation in respiratory burst-independent antimicrobial activity of gamma interferon-stimulated human macrophages. Infect. Immun. 57, 845–849 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Pfefferkorn, E. R. Interferon gamma blocks the growth of Toxoplasma gondii in human fibroblasts by inducing the host cells to degrade tryptophan. Proc. Natl Acad. Sci. USA 81, 908–912 (1984).This paper is the first to link interferon signalling to Trp metabolism as a mechanism of host defence.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Schmitz, J. L., Carlin, J. M., Borden, E. C. & Byrne, G. I. Beta interferon inhibits Toxoplasma gondii growth in human monocyte-derived macrophages. Infect. Immun. 57, 3254–3256 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. de Araujo, E. F. et al. The IDO-AhR axis controls Th17/Treg immunity in a pulmonary model of fungal infection. Front. Immunol. 8, 880 (2017).

    PubMed  PubMed Central  Google Scholar 

  141. Zhang, Y. J. et al. Tryptophan biosynthesis protects mycobacteria from CD4 T cell-mediated killing. Cell 155, 1296–1308 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Beatty, W. L., Belanger, T. A., Desai, A. A., Morrison, R. P. & Byrne, G. I. Tryptophan depletion as a mechanism of gamma interferon-mediated chlamydial persistence. Infect. Immun. 62, 3705–3711 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Mehta, S. J., Miller, R. D., Ramirez, J. A. & Summersgill, J. T. Inhibition of Chlamydia pneumoniae replication in HEp-2 cells by interferon-gamma: role of tryptophan catabolism. J. Infect. Dis. 177, 1326–1331 (1998).

    CAS  PubMed  Google Scholar 

  144. Lamas, B., Richard, M. L., Leducq, V. & Pham, H. P. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 22, 598–605 (2016).This paper demonstrates an important pathway regulating gut inflammation through microbial Trp metabolism.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Moura-Alves, P. et al. AhR sensing of bacterial pigments regulates antibacterial defence. Nature 512, 387–392 (2014).

    CAS  PubMed  Google Scholar 

  146. Bessede, A. et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511, 184–190 (2014).This paper highlights the relevance of Trp metabolism for regulating tolerance to bacterial infections.

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Schmidt, S. K. et al. Antimicrobial and immunoregulatory properties of human tryptophan 2,3-dioxygenase. Eur. J. Immunol. 39, 2755–2764 (2009).

    CAS  PubMed  Google Scholar 

  148. Cui, L. et al. Serum metabolome and lipidome changes in adult patients with primary dengue infection. PLOS Negl. Trop. Dis. 7, e2373 (2013).

    PubMed  PubMed Central  Google Scholar 

  149. Greco, F. A. et al. The Janus-faced nature of IDO1 in infectious diseases: challenges and therapeutic opportunities. Future Med. Chem. 8, 39–54 (2016).

    CAS  PubMed  Google Scholar 

  150. Yeung, A. W., Terentis, A. C., King, N. J. & Thomas, S. R. Role of indoleamine 2,3-dioxygenase in health and disease. Clin. Sci. 129, 601–672 (2015).

    CAS  Google Scholar 

  151. Ibana, J. A. et al. Inhibition of indoleamine 2,3-dioxygenase activity by levo-1-methyl tryptophan blocks gamma interferon-induced Chlamydia trachomatis persistence in human epithelial cells. Infect. Immun. 79, 4425–4437 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Adams, O. et al. Inhibition of human herpes simplex virus type 2 by interferon gamma and tumor necrosis factor alpha is mediated by indoleamine 2,3-dioxygenase. Microbes Infect. 6, 806–812 (2004).

    CAS  PubMed  Google Scholar 

  153. Bodaghi, B. et al. Role of IFN-gamma-induced indoleamine 2,3 dioxygenase and inducible nitric oxide synthase in the replication of human cytomegalovirus in retinal pigment epithelial cells. J. Immunol. 162, 957–964 (1999).

    CAS  PubMed  Google Scholar 

  154. Obojes, K., Andres, O., Kim, K. S., Daubener, W. & Schneider-Schaulies, J. Indoleamine 2,3-dioxygenase mediates cell type-specific anti-measles virus activity of gamma interferon. J. Virol. 79, 7768–7776 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Terajima, M. & Leporati, A. M. Role of indoleamine 2,3-dioxygenase in antiviral activity of interferon-gamma against vaccinia virus. Viral Immunol. 18, 722–729 (2005).

    CAS  PubMed  Google Scholar 

  156. Loughman, J. A. & Hunstad, D. A. Induction of indoleamine 2,3-dioxygenase by uropathogenic bacteria attenuates innate responses to epithelial infection. J. Infect. Dis. 205, 1830–1839 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Boasso, A. & Shearer, G. M. How does indoleamine 2,3-dioxygenase contribute to HIV-mediated immune dysregulation. Curr. Drug Metab. 8, 217–223 (2007).

    CAS  PubMed  Google Scholar 

  158. Asghar, K. et al. Indoleamine 2,3-dioxygenase expression and activity in patients with hepatitis C virus-induced liver cirrhosis. Exp. Ther. Med. 9, 901–904 (2015).

    PubMed  Google Scholar 

  159. Huang, L. et al. Induction and role of indoleamine 2,3 dioxygenase in mouse models of influenza a virus infection. PLOS ONE 8, e66546 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Zelante, T., Fallarino, F., Bistoni, F., Puccetti, P. & Romani, L. Indoleamine 2,3-dioxygenase in infection: the paradox of an evasive strategy that benefits the host. Microbes Infect. 11, 133–141 (2009).

    CAS  PubMed  Google Scholar 

  161. Montagnoli, C. et al. Immunity and tolerance to Aspergillus involve functionally distinct regulatory T cells and tryptophan catabolism. J. Immunol. 176, 1712–1723 (2006).

    CAS  PubMed  Google Scholar 

  162. Soliman, H. H. et al. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 5, 8136–8146 (2014).

    PubMed  PubMed Central  Google Scholar 

  163. Kasper, S. H., Bonocora, R. P., Wade, J. T., Musah, R. A. & Cady, N. C. Chemical inhibition of kynureninase reduces Pseudomonas aeruginosa quorum sensing and virulence factor expression. ACS Chem. Biol. 11, 1106–1117 (2016).

    CAS  PubMed  Google Scholar 

  164. Platten, M., Wick, W. & Van den Eynde, B. J. Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res. 72, 5435–5440 (2012).

    CAS  PubMed  Google Scholar 

  165. Tang, D. et al. P53 prevent tumor invasion and metastasis by down-regulating IDO in lung cancer. Oncotarget 8, 54548–54557 (2017).

    PubMed  PubMed Central  Google Scholar 

  166. Hsu, Y. L. et al. Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis. Oncotarget 7, 27584–27598 (2016).

    PubMed  PubMed Central  Google Scholar 

  167. Levina, V., Su, Y. & Gorelik, E. Immunological and nonimmunological effects of indoleamine 2,3-dioxygenase on breast tumor growth and spontaneous metastasis formation. Clin. Dev. Immunol. 2012, 173029 (2012).

    PubMed  PubMed Central  Google Scholar 

  168. Smith, C. et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov. 2, 722–735 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Pan, K. et al. Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 134, 1247–1253 (2008).

    CAS  PubMed  Google Scholar 

  170. Lee, A. et al. IDO1 and IDO2 non-synonymous gene variants: correlation with crohn’s disease risk and clinical phenotype. PLOS ONE 9, e115848 (2014).

    PubMed  PubMed Central  Google Scholar 

  171. Agliardi, C. et al. Indoleamine-2,3-dioxygenase(IDO)2 polymorphisms are not associated with multiple sclerosis in Italians. J. Neurol. Sci. 377, 31–34 (2017).

    CAS  PubMed  Google Scholar 

  172. Lippens, C. et al. IDO-orchestrated crosstalk between pDCs and Tregs inhibits autoimmunity. J. Autoimmun. 75, 39–49 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Kwidzinski, E. et al. Indolamine 2,3-dioxygenase is expressed in the CNS and down-regulates autoimmune inflammation. FASEB J. 19, 1347–1349 (2005).

    CAS  PubMed  Google Scholar 

  174. Sakurai, K., Zou, J. P., Tschetter, J. R., Ward, J. M. & Shearer, G. M. Effect of indoleamine 2,3-dioxygenase on induction of experimental autoimmune encephalomyelitis. J. Neuroimmunol. 129, 186–196 (2002).

    CAS  PubMed  Google Scholar 

  175. Yan, Y. et al. IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. J. Immunol. 185, 5953–5961 (2010).

    CAS  PubMed  Google Scholar 

  176. Criado, G., Simelyte, E., Inglis, J. J., Essex, D. & Williams, R. O. Indoleamine 2,3 dioxygenase-mediated tryptophan catabolism regulates accumulation of Th1/Th17 cells in the joint in collagen-induced arthritis. Arthritis Rheum. 60, 1342–1351 (2009).

    PubMed  Google Scholar 

  177. Ravishankar, B. et al. Tolerance to apoptotic cells is regulated by indoleamine 2,3-dioxygenase. Proc. Natl Acad. Sci. USA 109, 3909–3914 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Fallarino, F. et al. IDO mediates TLR9-driven protection from experimental autoimmune diabetes. J. Immunol. 183, 6303–6312 (2009).

    CAS  PubMed  Google Scholar 

  179. Scott, G. N. et al. The immunoregulatory enzyme IDO paradoxically drives B cell-mediated autoimmunity. J. Immunol. 182, 7509–7517 (2009).

    CAS  PubMed  Google Scholar 

  180. Cole, J. E. et al. Indoleamine 2,3-dioxygenase-1 is protective in atherosclerosis and its metabolites provide new opportunities for drug development. Proc. Natl Acad. Sci. USA 112, 13033–13038 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Godin-Ethier, J., Hanafi, L. A., Duvignaud, J. B., Leclerc, D. & Lapointe, R. IDO expression by human B lymphocytes in response to T lymphocyte stimuli and TLR engagement is biologically inactive. Mol. Immunol. 49, 253–259 (2011).

    CAS  PubMed  Google Scholar 

  182. Opitz, C. A. et al. The indoleamine-2,3-dioxygenase (IDO) inhibitor 1-methyl-D-tryptophan upregulates IDO1 in human cancer cells. PLOS ONE 6, e19823 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Merlo, L. M. & Mandik-Nayak, L. IDO2: a pathogenic mediator of inflammatory autoimmunity. Clin. Med. Insights Pathol. 9, 21–28 (2016).

    PubMed  PubMed Central  Google Scholar 

  184. Merlo, L. M. F. et al. IDO2 is a critical mediator of autoantibody production and inflammatory pathogenesis in a mouse model of autoimmune arthritis. J. Immunol. 192, 2082–2090 (2014).

    CAS  PubMed  Google Scholar 

  185. Metz, R. et al. IDO2 is critical for IDO1-mediated T cell regulation and exerts a non-redundant function in inflammation. Int. Immunol. 26, 357–367 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Yuasa, H. J. & Ball, H. J. Efficient tryptophan-catabolizing activity is consistently conserved through evolution of TDO enzymes, but not IDO enzymes. J. Exp. Zool. B 324, 128–140 (2015).

    CAS  Google Scholar 

  187. Maria, N. I. et al. Association of increased Treg cell levels with elevated indoleamine 2,3-dioxygenase activity and an imbalanced kynurenine pathway in interferon-positive primary Sjogren’s syndrome. Arthritis Rheumatol. 68, 1688–1699 (2016).

    CAS  PubMed  Google Scholar 

  188. Mancuso, R. et al. Indoleamine 2,3 dioxygenase (IDO) expression and activity in relapsing-remitting multiple sclerosis. PLOS ONE 10, e0130715 (2015).

    PubMed  PubMed Central  Google Scholar 

  189. Lim, C. K. et al. Kynurenine pathway metabolomics predicts and provides mechanistic insight into multiple sclerosis progression. Sci. Rep. 7, 41473 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Inglis, J. J. et al. The anti-allergic drug, N-(3,4,-dimethoxycinnamonyl) anthranilic acid, exhibits potent anti-inflammatory and analgesic properties in arthritis. Rheumatology 46, 1428–1432 (2007).

    CAS  PubMed  Google Scholar 

  191. Platten, M. et al. Treatment of autoimmune neuroinflammation with a synthetic tryptophan metabolite. Science 310, 850–855 (2005).

    CAS  PubMed  Google Scholar 

  192. Kaye, J. et al. Laquinimod arrests experimental autoimmune encephalomyelitis by activating the aryl hydrocarbon receptor. Proc. Natl Acad. Sci. USA 113, E6145–E6152 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Quintana, F. J. et al. An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 107, 20768–20773 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Yeste, A., Nadeau, M., Burns, E. J., Weiner, H. L. & Quintana, F. J. Nanoparticle-mediated codelivery of myelin antigen and a tolerogenic small molecule suppresses experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 109, 11270–11275 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Fazio, F. et al. Cinnabarinic acid, an endogenous agonist of type-4 metabotropic glutamate receptor, suppresses experimental autoimmune encephalomyelitis in mice. Neuropharmacology 81, 237–243 (2014).

    CAS  PubMed  Google Scholar 

  196. Lanz, T. V. et al. Suppression of Th1 differentiation by tryptophan supplementation in vivo. Amino Acids 49, 1169–1175 (2017).

    CAS  PubMed  Google Scholar 

  197. Orabona, C. et al. SOCS3 drives proteasomal degradation of indoleamine 2,3-dioxygenase (IDO) and antagonizes IDO-dependent tolerogenesis. Proc. Natl Acad. Sci. USA 105, 20828–20833 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Mondanelli, G. et al. The proteasome inhibitor bortezomib controls indoleamine 2,3-dioxygenase 1 breakdown and restores immune regulation in autoimmune diabetes. Front. Immunol. 8, 428 (2017).

    PubMed  PubMed Central  Google Scholar 

  199. Merlo, L. M. F. et al. Therapeutic antibody targeting of indoleamine-2,3-dioxygenase (IDO2) inhibits autoimmune arthritis. Clin. Immunol. 179, 8–16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Li, J. et al. 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, 171–179 (2016).

    CAS  PubMed  Google Scholar 

  201. Rohrig, U. F. et al. 1,2,3-Triazoles as inhibitors of indoleamine 2,3-dioxygenase 2 (IDO2). Bioorg. Med. Chem. Lett. 26, 4330–4333 (2016).

    CAS  PubMed  Google Scholar 

  202. Lanz, T. V. et al. Mouse mesenchymal stem cells suppress antigen-specific TH cell immunity independent of indoleamine 2,3-dioxygenase 1 (IDO1). Stem Cells Dev. 19, 657–668 (2010).

    CAS  PubMed  Google Scholar 

  203. Liu, Y. et al. Therapeutic potential of human umbilical cord mesenchymal stem cells in the treatment of rheumatoid arthritis. Arthritis Res. Ther. 12, R210 (2010).

    PubMed  PubMed Central  Google Scholar 

  204. Reading, J. L. et al. Clinical-grade multipotent adult progenitor cells durably control pathogenic T cell responses in human models of transplantation and autoimmunity. J. Immunol. 190, 4542–4552 (2013).

    CAS  PubMed  Google Scholar 

  205. Jalili, R. B. et al. Fibroblast cell-based therapy for experimental autoimmune diabetes. PLOS ONE 11, e0146970 (2016).

    PubMed  PubMed Central  Google Scholar 

  206. Pallotta, M. T. et al. Forced IDO1 expression in dendritic cells restores immunoregulatory signalling in autoimmune diabetes. J. Cell. Mol. Med. 18, 2082–2091 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Vavrincova-Yaghi, D. et al. Local gene therapy with indoleamine 2,3-dioxygenase protects against development of transplant vasculopathy in chronic kidney transplant dysfunction. Gene Ther. 23, 797–806 (2016).

    CAS  PubMed  Google Scholar 

  208. Muller, A. J., DuHadaway, J. B., Donover, P. S., Sutanto-Ward, E. & Prendergast, G. C. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat. Med. 11, 312–319 (2005).This paper shows IDO as a target of oncogenic signalling and highlights the potential of combining chemotherapy with IDO inhibition for the treatment of cancer.

    CAS  PubMed  Google Scholar 

  209. Uyttenhove, C. et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 9, 1269–1274 (2003).This paper is the first to demonstrate IDO as a drug target in immuno-oncology.

    CAS  PubMed  Google Scholar 

  210. Theate, I. et al. Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues. Cancer Immunol. Res. 3, 161–172 (2015).

    CAS  PubMed  Google Scholar 

  211. Platten, M., von Knebel Doeberitz, N., Oezen, I., Wick, W. & Ochs, K. Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors. Front. Immunol. 5, 673 (2014).

    PubMed  Google Scholar 

  212. D’Amato, N. C. et al. A TDO2-AhR signaling axis facilitates anoikis resistance and metastasis in triple-negative breast cancer. Cancer Res. 75, 4651–4664 (2015).

    PubMed  PubMed Central  Google Scholar 

  213. Giusti, R. M. et al. Differential patterns of serum biomarkers of immune activation in human T cell lymphotropic virus type I-associated myelopathy/tropical spastic paraparesis, and adult T-cell leukemia/lymphoma. Cancer Epidemiol. Biomarkers Res. 5, 699–704 (1996).

    CAS  Google Scholar 

  214. Huang, A. et al. Serum tryptophan decrease correlates with immune activation and impaired quality of life in colorectal cancer. Br. J. Cancer 86, 1691–1696 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Schroecksnadel, K., Winkler, C., Fuith, L. C. & Fuchs, D. Tryptophan degradation in patients with gynecological cancer correlates with immune activation. Cancer Lett. 223, 323–329 (2005).

    CAS  PubMed  Google Scholar 

  216. Weinlich, G., Murr, C., Richardsen, L., Winkler, C. & Fuchs, D. Decreased serum tryptophan concentration predicts poor prognosis in malignant melanoma patients. Dermatology 214, 8–14 (2007).

    PubMed  Google Scholar 

  217. Suzuki, Y. et al. Increased serum kynurenine/tryptophan ratio correlates with disease progression in lung cancer. Lung Cancer 67, 361–365 (2010).

    PubMed  Google Scholar 

  218. Zhai, L. et al. The kynurenine to tryptophan ratio as a prognostic tool for glioblastoma patients enrolling in immunotherapy. J. Clin. Neurosci. 22, 1964–1968 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  219. 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. 98, 874–881 (2007).

    CAS  PubMed  Google Scholar 

  220. Witkiewicz, A. et al. Expression of indoleamine 2,3-dioxygenase in metastatic pancreatic ductal adenocarcinoma recruits regulatory T cells to avoid immune detection. J. Am. Coll. Surg. 206, 849–854; discussion 854–846 (2008).

    PubMed  Google Scholar 

  221. Liu, X. Q. et al. Up-regulated expression of indoleamine 2,3-dioxygenase 1 in non-Hodgkin lymphoma correlates with increased regulatory T cell infiltration. Leuk. Lymphoma 55, 405–414 (2014).

    CAS  PubMed  Google Scholar 

  222. Brandacher, G. et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clin. Cancer Res. 12, 1144–1151 (2006).

    CAS  PubMed  Google Scholar 

  223. Inaba, T. et al. Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma. Gynecol. Oncol. 115, 185–192 (2009).

    CAS  PubMed  Google Scholar 

  224. Ino, K. et al. Inverse correlation between tumoral indoleamine 2,3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: its association with disease progression and survival. Clin. Cancer Res. 14, 2310–2317 (2008).

    CAS  PubMed  Google Scholar 

  225. Liu, Y. et al. Tumor-repopulating cells induce PD-1 expression in CD8(+) T cells by transferring kynurenine and AhR activation. Cancer Cell 33, 480–494 (2018).This paper demonstrates immunosuppressive Trp metabolism as a key regulator of immune checkpoints.

    CAS  PubMed  Google Scholar 

  226. Tummala, K. S. et al. Inhibition of de novo NAD(+) synthesis by oncogenic URI causes liver tumorigenesis through DNA damage. Cancer Cell 26, 826–839 (2014).

    CAS  PubMed  Google Scholar 

  227. Sahm, F. et al. The endogenous tryptophan metabolite and NAD+precursor quinolinic acid confers resistance of gliomas to oxidative stress. Cancer Res. 73, 3225–3234 (2013).

    CAS  PubMed  Google Scholar 

  228. Maleki Vareki, S. et al. Indoleamine 2,3-dioxygenase mediates immune-independent human tumor cell resistance to olaparib, gamma radiation, and cisplatin. Oncotarget 5, 2778–2791 (2014).

    PubMed  Google Scholar 

  229. Cheong, J. E., Ekkati, A. & Sun, L. A patent review of IDO1 inhibitors for cancer. Expert Opin. Ther. Pat. 28, 317–330 (2018).

    CAS  PubMed  Google Scholar 

  230. Prendergast, G. C., Malachowski, W. P., DuHadaway, J. B. & Muller, A. J. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res. 77, 6795–6811 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Rohrig, U. F., Majjigapu, S. R., Vogel, P., Zoete, V. & Michielin, O. Challenges in the discovery of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. J. Med. Chem. 58, 9421–9437 (2015).

    CAS  PubMed  Google Scholar 

  232. Gyulveszi, G. et al. RG70099: a novel, highly potent dual IDO1/TDO inhibitor to reverse metabolic suppression of immune cells in the tumor micro-environment. Cancer Res. 76 (Suppl.), LB–085 (2016).

    Google Scholar 

  233. Gullapalli, S. et al. EPL-1410, a novel fused heterocycle based orally active dual inhibitor of IDO1/TDO2, as a potential immune-oncology therapeutic. Cancer Res. 78 (Suppl.), 1701 (2018).

    Google Scholar 

  234. Salter, M., Hazelwood, R., Pogson, C. I., Iyer, R. & Madge, D. J. The effects of a novel and selective inhibitor of tryptophan 2,3-dioxygenase on tryptophan and serotonin metabolism in the rat. Biochem. Pharmacol. 49, 1435–1442 (1995).

    CAS  PubMed  Google Scholar 

  235. Salter, M. et al. The effects of an inhibitor of tryptophan 2,3-dioxygenase and a combined inhibitor of tryptophan 2,3-dioxygenase and 5-HT reuptake in the rat. Neuropharmacology 34, 217–227 (1995).

    CAS  PubMed  Google Scholar 

  236. Mautino, M. R. et al. NLG919, a novel indoleamine-2,3-dioxygenase (IDO)-pathway inhibitor drug candidate for cancer therapy. Cancer Res. 73 (Suppl.), 491 (2013).

    Google Scholar 

  237. Cady, S. G. & Sono, M. 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, 326–333 (1991).

    CAS  PubMed  Google Scholar 

  238. Hou, D. Y. et al. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 67, 792–801 (2007).

    CAS  PubMed  Google Scholar 

  239. Lob, S., Konigsrainer, A., Rammensee, H. G., Opelz, G. & Terness, P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat. Rev. Cancer 9, 445–452 (2009).

    PubMed  Google Scholar 

  240. Prendergast, G. C. et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol. Immunother. 63, 721–735 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Qian, F. et al. Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2,3-dioxygenase-mediated arrest of T cell proliferation in human epithelial ovarian cancer. Cancer Res. 69, 5498–5504 (2009).

    CAS  PubMed  Google Scholar 

  242. Davar, D. & Bahary, N. Modulating tumor immunology by inhibiting indoleamine 2,3-dioxygenase (IDO): recent developments and first clinical experiences. Target. Oncol. 13, 125–140 (2018).

    PubMed  Google Scholar 

  243. Beatty, G. L. et al. First-in-human phase I study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies. Clin. Cancer Res. 23, 3269–3277 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  244. Soliman, H. H. et al. A phase I study of indoximod in patients with advanced malignancies. Oncotarget 7, 22928–22938 (2016).

    PubMed  PubMed Central  Google Scholar 

  245. Nishino, M., Ramaiya, N. H., Hatabu, H. & Hodi, F. S. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat. Rev. Clin. Oncol. 14, 655–668 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Toulmonde, M. et al. Use of PD-1 targeting, macrophage infiltration, and IDO pathway activation in sarcomas: a phase 2 clinical trial. JAMA Oncol. 4, 93–97 (2017).This paper demonstrates immunosuppressive Trp metabolism as a resistance mechanism against immune checkpoint blockade in patients with sarcomas.

    PubMed Central  Google Scholar 

  247. Holmgaard, R. B., Zamarin, D., Munn, D. H., Wolchok, J. D. & Allison, J. P. Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J. Exp. Med. 210, 1389–1402 (2013).This paper demonstrates immunosuppressive Trp metabolism as a resistance mechanism against immune checkpoint blockade in a preclinical tumour model.

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Balachandran, V. P. et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat. Med. 17, 1094–1100 (2011).This paper provides an important link between key oncogenic signalling and immunosuppressive Trp metabolism.

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Seifert, A. M. et al. PD-1/PD-L1 blockade enhances T cell activity and antitumor efficacy of imatinib in gastrointestinal stromal tumors. Clin. Cancer Res. 23, 454–465 (2017).

    CAS  PubMed  Google Scholar 

  250. D’Angelo, S. P. et al. Combined KIT and CTLA-4 blockade in patients with refractory GIST and other advanced sarcomas: a phase Ib study of dasatinib plus ipilimumab. Clin. Cancer Res. 23, 2972–2980 (2017).

    PubMed  Google Scholar 

  251. Garber, K. A new cancer immunotherapy suffers a setback. Science 360, 588 (2018).

    CAS  PubMed  Google Scholar 

  252. Monjazeb, A. M. et al. 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, 4328–4340 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Opitz, C. A. et al. Toll-like receptor engagement enhances the immunosuppressive properties of human bone marrow-derived mesenchymal stem cells by inducing indoleamine-2,3-dioxygenase-1 via interferon-beta and protein kinase R. Stem Cells 27, 909–919 (2009).

    CAS  PubMed  Google Scholar 

  254. Lemos, H. et al. STING promotes the growth of tumors characterized by low antigenicity via IDO activation. Cancer Res. 76, 2076–2081 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Li, M. et al. The indoleamine 2,3-dioxygenase pathway controls complement-dependent enhancement of chemo-radiation therapy against murine glioblastoma. J. Immunother. Cancer 2, 21 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  256. Creelan, B. C. et al. Indoleamine 2,3-dioxygenase activity and clinical outcome following induction chemotherapy and concurrent chemoradiation in stage III non-small cell lung cancer. Oncoimmunology 2, e23428 (2013).

    PubMed  PubMed Central  Google Scholar 

  257. Wang, W. et al. IDO immune status after chemoradiation may predict survival in lung cancer patients. Cancer Res. 78, 809–816 (2017).

    PubMed  PubMed Central  Google Scholar 

  258. Uhlen, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    PubMed  Google Scholar 

  259. Ott, M. et al. Suppression of TDO-mediated tryptophan catabolism in glioblastoma cells by a steroid-responsive FKBP52-dependent pathway. Glia 63, 78–90 (2015).

    PubMed  Google Scholar 

  260. Lanz, T. V. et al. Tryptophan-2,3-Dioxygenase (TDO) deficiency is associated with subclinical neuroprotection in a mouse model of multiple sclerosis. Sci. Rep. 7, 41271 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Ochs, K. et al. Tryptophan-2,3-dioxygenase is regulated by prostaglandin E2 in malignant glioma via a positive signaling loop involving prostaglandin E receptor-4. J. Neurochem. 136, 1142–1154 (2015).

    Google Scholar 

  262. Lemieux, G. A. et al. Kynurenic acid is a nutritional cue that enables behavioral plasticity. Cell 160, 119–131 (2015).This paper provides an important link between dietary Trp metabolites and brain function.

    CAS  PubMed  PubMed Central  Google Scholar 

  263. Yoshida, R., Imanishi, J., Oku, T., Kishida, T. & Hayaishi, O. Induction of pulmonary indoleamine 2,3-dioxygenase by interferon. Proc. Natl Acad. Sci. USA 78, 129–132 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Yoshida, R., Oku, T., Imanishi, J., Kishida, T. & Hayaishi, O. Interferon: a mediator of indoleamine 2,3-dioxygenase induction by lipopolysaccharide, poly(I) X poly(C), and pokeweed mitogen in mouse lung. Arch. Biochem. Biophys. 249, 596–604 (1986).

    CAS  PubMed  Google Scholar 

  265. Grohmann, U. et al. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat. Immunol. 3, 1097–1101 (2002).

    CAS  PubMed  Google Scholar 

  266. Wang, Y. et al. Kynurenine is an endothelium-derived relaxing factor produced during inflammation. Nat. Med. 16, 279–285 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Yan, E. B. et al. Activation of the kynurenine pathway and increased production of the excitotoxin quinolinic acid following traumatic brain injury in humans. J. Neuroinflamm. 12, 110 (2015).

    Google Scholar 

  268. Formisano, S. et al. Central nervous system infection with Borna disease virus causes kynurenine pathway dysregulation and neurotoxic quinolinic acid production. J. Virol. 91, e00673–17 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Knyihar-Csillik, E. et al. Effect of 6-hydroxydopamine treatment on kynurenine aminotransferase-I (KAT-I) immunoreactivity of neurons and glial cells in the rat substantia nigra. Acta Neuropathol. 112, 127–137 (2006).

    CAS  PubMed  Google Scholar 

  270. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (1994).

    Google Scholar 

  271. Lewis-Ballester, A. et al. Structural insights into substrate and inhibitor binding sites in human indoleamine 2,3-dioxygenase 1. Nat. Commun. 8, 1693 (2017).

    PubMed  PubMed Central  Google Scholar 

  272. Peng, Y. H. et al. 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, 282–293 (2016).

    CAS  PubMed  Google Scholar 

  273. Nayak-Kapoor, A. et al. Phase Ia study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) in patients with recurrent advanced solid tumors. J. Immunother. Cancer 6, 61 (2018).

    PubMed  PubMed Central  Google Scholar 

  274. Crosignani, S. et al. Discovery of a novel and selective indoleamine 2,3-dioxygenase (IDO-1) inhibitor 3-(5-fluoro-1H-indol-3-yl)pyrrolidine-2,5-dione (EOS200271/PF-06840003) and its characterization as a potential clinical candidate. J. Med. Chem. 60, 9617–9629 (2017).

    CAS  PubMed  Google Scholar 

  275. Nelp, M. T. et al. Immune-modulating enzyme indoleamine 2,3-dioxygenase is effectively inhibited by targeting its apo-form. Proc. Natl Acad. Sci. USA 115, 3249–3254 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Sadok, I., Gamian, A. & Staniszewska, M. M. Chromatographic analysis of tryptophan metabolites. J. Sep. Sci. 40, 3020–3045 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Alegre, E., López, A. S. & González, A. Tryptophan metabolites interfere with the Ehrlich reaction used for the measurement of kynurenine. Anal. Biochem. 339, 188–189 (2005).

    CAS  PubMed  Google Scholar 

  278. Daubener, W. et al. A new, simple, bioassay for human IFN-gamma. J. Immunol. Methods 168, 39–47 (1994).

    CAS  PubMed  Google Scholar 

  279. Takikawa, O., Kuroiwa, T., Yamazaki, F. & Kido, R. Mechanism of interferon-gamma action. Characterization of indoleamine 2,3-dioxygenase in cultured human cells induced by interferon-gamma and evaluation of the enzyme-mediated tryptophan degradation in its anticellular activity. J. Biol. Chem. 263, 2041–2048 (1988).

    CAS  PubMed  Google Scholar 

  280. Seegers, N. et al. High-throughput fluorescence-based screening assays for tryptophan-catabolizing enzymes. J. Biomol. Screen. 19, 1266–1274 (2014).

    CAS  PubMed  Google Scholar 

  281. Tomek, P. et al. Formation of an N-formylkynurenine-derived fluorophore and its use for measuring indoleamine 2,3-dioxygenase 1 activity. Anal. Bioanal. Chem. 405, 2515–2524 (2013).

    CAS  PubMed  Google Scholar 

  282. Kaper, T. et al. Nanosensor detection of an immunoregulatory tryptophan influx/kynurenine efflux cycle. PLOS Biol. 5, e257 (2007).

    PubMed  PubMed Central  Google Scholar 

  283. Maliniemi, P. et al. Biological and clinical significance of tryptophan-catabolizing enzymes in cutaneous T cell lymphomas. Oncoimmunology 6, e1273310 (2017).

    PubMed  PubMed Central  Google Scholar 

  284. Konishi, M. et al. Impact of plasma kynurenine level on functional capacity and outcome in heart failure- results from studies investigating co-morbidities aggravating heart failure (SICA-HF). Circ. J. 81, 52–61 (2016).

    PubMed  Google Scholar 

  285. Reichetzeder, C. et al. Pre-interventional kynurenine predicts medium-term outcome after contrast media exposure due to coronary angiography. Kidney Blood Press. Res. 42, 244–256 (2017).

    CAS  PubMed  Google Scholar 

  286. Dewi, D. L. et al. Suppression of indoleamine-2,3-dioxygenase 1 expression by promoter hypermethylation in ER-positive breast cancer. Oncoimmunology 6, e1274477 (2017).

    PubMed  PubMed Central  Google Scholar 

  287. Ait-Belkacem, R. B. et al. Microenvironment tumor metabolic hnteractions highlighted by qMSI: application to the tryptophan-kynurenine pathway in immuno-oncology. SLAS Discov. 22, 1182–1192 (2017).

    CAS  PubMed  Google Scholar 

  288. Bosnyak, E. et al. Prognostic molecular and imaging biomarkers in primary glioblastoma. Clin. Nucl. Med. 42, 341–347 (2017).

    PubMed  PubMed Central  Google Scholar 

  289. Guastella, A. R. et al. Tryptophan PET imaging of the kynurenine pathway in patient-derived xenograft models of glioblastoma. Mol. Imaging 15, 1–11 (2016).

    CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the German Cancer Aid (70110392) and German Research Foundation (DFG PL-315/5-1) to M.P.; the Italian Association of Cancer Research (19903) and Telethon (GGP17094) to F.F.; the German Federal Ministry of Education and Research (BMBF) e:Med initiative (GlioPATH, 01ZX1402), the European Research Council (ERC) and the alumni chapter of Gooische Groningers facilitated by Ubbo Emmius Fonds to E.A.A.N; and funding from the European Union’s Horizon 2020 research and innovation programme under grant agreement No 754688 to C.A.O. The authors thank F. Sorgdrager for Fig. 2 and A. Sadik for help with Table 2.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael Platten.

Ethics declarations

Competing interests

M.P. has received royalties for patents on aryl hydrocarbon receptor inhibitors and tryptophan metabolites and has received honoraria for advisory board services and research support from Bayer. E.A.A.N., F.F., U.F.R. and C.A.O. declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

ClinicalTrials.gov: http://www.clinicaltrials.gov/

Glossary

Kynurenine (Kyn) pathway

(KP). The major pathway in the metabolism of the essential amino acid tryptophan, which contains many immunoactive and neuroactive intermediate metabolites.

Indoleamine-2,3-dioxygenase 1

(IDO1). The first enzyme discovered to initiate immunosuppressive kynurenine pathway metabolism. IDO1 and tryptophan-2,3-dioxygenase (TDO) represent a key intracellular immune checkpoint.

Kynurenine aminotransferases

(KATI–KATIII). KATs catalyse the conversion of kynurenine to kynurenic acid (KA) and of 3-hydroxykynurenine to KA and are a drug target for schizophrenia and cognitive impairment disorders.

Kynurenine monooxygenase

(KMO). A key kynurenine pathway (KP) enzyme and drug target that controls the conversion of kynurenine to neuroactive and immunoregulatory KP metabolites.

Quinolinic acid

(QA). A key kynurenine pathway metabolite accumulating in the brain during inflammation and mediating neuronal death through the activation of N-methyl-d-aspartate (NMDA) receptors, a process termed excitotoxicity.

NAD+

A key coenzyme in metabolic processes and redox reactions regulating cellular fitness, for which the kynurenine pathway is a major source.

Immune checkpoint inhibitors

Antibodies for cancer immunotherapy, which act by neutralizing inhibitory pathways in T cells, thereby enhancing antitumor immunity.

General control non-derepressible 2

(GCN2). A kinase leading to dysfunction of T cells and antigen-presenting cells in response to extreme l-tryptophan shortage (<1 µM).

Aryl hydrocarbon receptor

(AHR). A cytosolic transcription factor originally described as a mediator of xenobiotic detoxification but increasingly shown to mediate important functions of tryptophan metabolism by binding kynurenine and kynurenic acid.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Platten, M., Nollen, E.A.A., Röhrig, U.F. et al. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov 18, 379–401 (2019). https://doi.org/10.1038/s41573-019-0016-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-019-0016-5

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer