We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Published Online:

Indoleamine 2,3 dioxygenase (IDO), first discovered in the 1960s, is an enzyme that has become a highly investigated metabolic target in cancer research. IDO is the rate-limiting step in tryptophan metabolism catabolism into its byproducts – kynurenines. Both IDO and kynurenines have been implicated in altering the tumor microenvironment, allowing for a tolerogenesis by affecting T-cell maturation and proliferation, and more specifically by inducing differentiation into T regulatory cells. Two mechanisms have been suspected in creating this environment: tryptophan starvation and metabolite toxicity. IDO has been shown to be expressed not only in cancer cells but also in antigen-presenting cells. The exact mechanisms underlying the two different sites of expression have not been fully elucidated. To date, most literature has focused on the role of IDO in solid tumors; we provide a review of IDO and its impact on hematological malignancies – more specifically, acute myeloid leukemia. The pathophysiology of IDO will be discussed, including a summarization of the literature to date on how IDO expression effects prognosis and disease progression in acute myeloid leukemia, along with current IDO-specific therapeutics with future considerations.

Indoleamine 2,3 dioxygenase (IDO) is an enzyme that has become a highly investigated metabolic target in cancer research. IDO is the rate-limiting step in tryptophan metabolism catabolism into its byproducts – kynurenines. Both IDO and kynurenines have been implicated in altering the tumor microenvironment, allowing for a tolerogenesis by affecting T-cell maturation and proliferation, and more specifically by inducing differentiation into T regulatory cells. The exact mechanisms underlying the two different sites of expression have not been fully elucidated. The pathophysiology of IDO will be discussed, including a summarization of the literature to date on how IDO expression effects prognosis and disease progression in acute myeloid leukemia, along with current IDO-specific therapeutics with future considerations.

Papers of special note have been highlighted as: • of interest; •• of considerable interest

References

  • 1. Burnet M. Cancer: a biological approach. III. Viruses associated with neoplastic conditions. IV. Practical applications. Br. Med. J. 1(5023), 841–847 (1957).
  • 2. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3(11), 991–998 (2002).
  • 3. Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology 121(1), 1–14 (2007). •• This review provides an overview of the immune system and interactions with tumor environment.
  • 4. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases elimination, equilibrium and escape. Curr. Opin. Immunol. 27, 16–25 (2014).
  • 5. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 21(4), 687–692 (2015). • The authors provide a review of mechanisms that are important to understand the application of immunotherapy in cancer.
  • 6. Prendergast GC, Smith C, Thomas S et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol. Immunother. 63(7), 721–735 (2014). •• This review explores the importance of indoleamine 2,3-dioxygenase in immune regulation and the reasons for indoleamine 2,3-dioxygenase inhibition in cancer.
  • 7. Kuol N, Stojanovska L, Nurgali K, Apostolopoulos V. The mechanisms tumor cells utilize to evade the host’s immune system. Maturitas 105, 8–15 (2017).
  • 8. Yeung AW, Terentis AC, King NJ, Thomas SR. Role of indoleamine 2,3-dioxygenase in health and disease. Clin. Sci. (Lond.) 129(7), 601–672 (2015).
  • 9. Mellor AL, Munn DH. Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation? Immunol. Today 20(10), 469–473 (1999).
  • 10. Platten M, Wick W, Van den Eynde BJ. Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res. 72(21), 5435–5440 (2012).
  • 11. Boyland E, Williams DC. The metabolism of tryptophan. 2. The metabolism of tryptophan in patients suffering from cancer of the bladder. Biochem. J. 64(3), 578–582 (1956).
  • 12. Rose DP. Aspects of tryptophan metabolism in health and disease: a review. J. Clin. Pathol. 25(1), 17–25 (1972).
  • 13. Chang RQ, Li DJ, Li MQ. The role of indoleamine-2,3-dioxygenase in normal and pathological pregnancies. Am. J. Reprod. Immunol. 79(4), e12786 (2018).
  • 14. Wu H, Gong J, Liu Y. Indoleamine 2, 3-dioxygenase regulation of immune response (review). Mol. Med. Rep. 17(4), 4867–4873 (2018). • This article focuses on the role of indoleamine 2,3-dioxygenase in tryptophan metabolism and its role as an immunosuppressant.
  • 15. Munn DH, Mellor AL. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol. 37(3), 193–207 (2016). •• This review details the role of indoleamine 2,3-dioxygenase in controlling inflammation and tolerance. It provides the importance of indoleamine 2,3-dioxygenase in the tumor microenvironment.
  • 16. Matteoli G, Mazzini E, Iliev ID et al. Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut 59(5), 595–604 (2010).
  • 17. van der Marel AP, Samsom JN, Greuter M et al. Blockade of IDO inhibits nasal tolerance induction. J. Immunol. 179(2), 894–900 (2007).
  • 18. Ravishankar B, Liu H, Shinde R et al. The amino acid sensor GCN2 inhibits inflammatory responses to apoptotic cells promoting tolerance and suppressing systemic autoimmunity. Proc. Natl. Acad. Sci. USA. 112(34), 10774–10779 (2015).
  • 19. Ravishankar B, Liu H, Shinde R et al. Tolerance to apoptotic cells is regulated by indoleamine 2,3-dioxygenase. Proc. Natl. Acad. Sci. USA. 109(10), 3909–3914 (2012).
  • 20. Curti A, Pandolfi S, Valzasina B et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood 109(7), 2871–2877 (2007).
  • 21. Smith C, Chang MY, Parker KH et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov. 2(8), 722–735 (2012).
  • 22. Ferns DM, Kema IP, Buist MR, Nijman HW, Kenter GG, Jordanova ES. Indoleamine-2,3-dioxygenase (IDO) metabolic activity is detrimental for cervical cancer patient survival. Oncoimmunology 4(2), e981457 (2015).
  • 23. Adams S, Teo C, McDonald KL et al. Involvement of the kynurenine pathway in human glioma pathophysiology. PLoS ONE 9(11), e112945 (2014).
  • 24. Carvajal-Hausdorf DE, Mani N, Velcheti V, Schalper KA, Rimm DL. Objective measurement and clinical significance of IDO1 protein in hormone receptor-positive breast cancer. J. Immunother. Cancer 5(1), 81 (2017).
  • 25. Andersen MH. The targeting of immunosuppressive mechanisms in hematological malignancies. Leukemia 28(9), 1784–1792 (2014). • This article provides an overview of immune dysregulation in hematological malignancies and possibilities of targeting these pathways.
  • 26. Uyttenhove C, Pilotte L, Theate I et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 9(10), 1269–1274 (2003).
  • 27. Badawy AA. Kynurenine pathway of tryptophan metabolism: regulatory and functional aspects. Int. J. Tryptophan Res. 10, 1178646917691938 (2017).
  • 28. Lob S, Konigsrainer A, Rammensee HG, 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(6), 445–452 (2009).
  • 29. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J. Exp. Med. 189(9), 1363–1372 (1999).
  • 30. Munn DH, Sharma MD, Baban B et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 22(5), 633–642 (2005).
  • 31. Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3-dioxygenase and its therapeutic inhibition in cancer. Int. Rev. Cell. Mol. Biol. 336, 175–203 (2018).
  • 32. Terness P, Bauer TM, Rose L 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(4), 447–457 (2002).
  • 33. Fallarino F, Grohmann U, Vacca C et al. T cell apoptosis by kynurenines. Adv. Exp. Med. Biol. 527, 183–190 (2003).
  • 34. Li Q, Harden JL, Anderson CD, Egilmez NK. Tolerogenic phenotype of IFN-gamma-induced IDO+ dendritic cells is maintained via an autocrine IDO-kynurenine/AhR-IDO loop. J. Immunol. 197(3), 962–970 (2016).
  • 35. Forteza MJ, Polyzos KA, Baumgartner R et al. Activation of the regulatory T-cell/indoleamine 2,3-dioxygenase axis reduces vascular inflammation and atherosclerosis in hyperlipidemic mice. Front. Immunol. 9, 950 (2018).
  • 36. Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J. Immunol. 164(7), 3596–3599 (2000).
  • 37. Diehl L, den Boer AT, Schoenberger SP et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy. Nat. Med. 5(7), 774–779 (1999).
  • 38. Munn DH, Mellor AL. IDO and tolerance to tumors. Trends Mol. Med. 10(1), 15–18 (2004).
  • 39. Wang Y, Yang BH, Li H, Cao S, Ren XB, Yu JP. IDO(+) DCs and signalling pathways. Curr. Cancer Drug Targets 13(3), 278–288 (2013).
  • 40. Munn DH, Sharma MD, Lee JR et al. Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase. Science 297(5588), 1867–1870 (2002).
  • 41. Munn DH, Sharma MD, Hou D et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J. Clin. Invest. 114(2), 280–290 (2004).
  • 42. von Bubnoff D, Hanau D, Wenzel J et al. Indoleamine 2,3-dioxygenase-expressing antigen-presenting cells and peripheral T-cell tolerance: another piece to the atopic puzzle? J. Allergy Clin. Immunol. 112(5), 854–860 (2003).
  • 43. Du MX, Sotero-Esteva WD, Taylor MW. Analysis of transcription factors regulating induction of indoleamine 2,3-dioxygenase by IFN-gamma. J. Interferon Cytokine Res. 20(2), 133–142 (2000).
  • 44. Chen X, Kline DE, Kline J. Peripheral T-cell tolerance in hosts with acute myeloid leukemia. Oncoimmunology 2(8), e25445 (2013).
  • 45. Dhodapkar MV, Dhodapkar KM, Palucka AK. Interactions of tumor cells with dendritic cells: balancing immunity and tolerance. Cell Death Differ. 15(1), 39–50 (2008).
  • 46. von Bubnoff D, Scheler M, Wilms H, Fimmers R, Bieber T. Identification of IDO-positive and IDO-negative human dendritic cells after activation by various proinflammatory stimuli. J. Immunol. 186(12), 6701–6709 (2011).
  • 47. Corm S, Berthon C, Imbenotte M et al. Indoleamine 2,3-dioxygenase activity of acute myeloid leukemia cells can be measured from patients’ sera by HPLC and is inducible by IFN-gamma. Leuk. Res. 33(3), 490–494 (2009).
  • 48. Chamuleau ME, van de Loosdrecht AA, Hess CJ et al. High INDO (indoleamine 2,3-dioxygenase) mRNA level in blasts of acute myeloid leukemic patients predicts poor clinical outcome. Haematologica 93(12), 1894–1898 (2008).
  • 49. Mangaonkar A, Mondal AK, Fulzule S et al. A novel immunohistochemical score to predict early mortality in acute myeloid leukemia patients based on indoleamine 2,3 dioxygenase expression. Sci. Rep. 7(1), 12892 (2017). •• The authors discuss the role of an immunohistochemistry-based model to assess the prognostic impact of indoleamine 2,3-dioxygenase in acute myeloid leukemia patients.
  • 50. Curti A, Aluigi M, Pandolfi S et al. Acute myeloid leukemia cells constitutively express the immunoregulatory enzyme indoleamine 2,3-dioxygenase. Leukemia 21(2), 353–355 (2007).
  • 51. Chen XL, Guo JM, Zhang Y, Niu XN, Pei XH, Zhang WH. Expression of indoleamine 2,3-dioxygenase in acute leukemic cells and the clinical significance. Int. J. Clin. Exp. Med. 9(5), 8605–8609 (2016).
  • 52. Folgiero V, Goffredo BM, Filippini P et al. Indoleamine 2,3-dioxygenase 1 (IDO1) activity in leukemia blasts correlates with poor outcome in childhood acute myeloid leukemia. Oncotarget 5(8), 2052–2064 (2014).
  • 53. Arandi N, Ramzi M, Safaei F, Monabati A. Overexpression of indoleamine 2,3-dioxygenase correlates with regulatory T cell phenotype in acute myeloid leukemia patients with normal karyotype. Blood Res. 53(4), 294–298 (2018).
  • 54. Ye Q, Wang C, Xian J, Zhang M, Cao Y, Cao Y. Expression of programmed cell death protein 1 (PD-1) and indoleamine 2,3-dioxygenase (IDO) in the tumor microenvironment and in tumor-draining lymph nodes of breast cancer. Hum. Pathol. 75, 81–90 (2018).
  • 55. Lee JR, Dalton RR, Messina JL et al. Pattern of recruitment of immunoregulatory antigen-presenting cells in malignant melanoma. Lab. Invest. 83(10), 1457–1466 (2003).
  • 56. Xu J, Wei J, Zhu X et al. Increased plasma indoleamine 2,3-dioxygenase activity and interferon-gamma levels correlate with the severity of acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 19(2), 196–201 (2013).
  • 57. Melichar B, Solichova D, Freedman RS. Neopterin as an indicator of immune activation and prognosis in patients with gynecological malignancies. Int. J. Gynecol. Cancer 16(1), 240–252 (2006).
  • 58. Prommegger R, Widner B, Murr C, Unger A, Fuchs D, Salzer GM. Neopterin: a prognostic variable in operations for lung cancer. Ann. Thorac. Surg. 70(6), 1861–1864 (2000).
  • 59. Arpinati M, Curti A. Immunotherapy in acute myeloid leukemia. Immunotherapy 6(1), 95–106 (2014).
  • 60. Teague RM, Kline J. Immune evasion in acute myeloid leukemia: current concepts and future directions. J. Immunother. Cancer 1(13), 1 (2013).
  • 61. Kobayashi S, Kikuta A, Ito M et al. Loss of mismatched HLA in myeloid/NK cell precursor acute leukemia relapse after T cell-replete haploidentical hematopoietic stem cell transplantation. Pediatr. Blood Cancer 61(10), 1880–1882 (2014).
  • 62. Moon YW, Hajjar J, Hwu P, Naing A. Targeting the indoleamine 2,3-dioxygenase pathway in cancer. J. Immunother. Cancer 3, 51 (2015).
  • 63. Collin M. Immune checkpoint inhibitors: a patent review (2010–2015). Expert Opin. Ther. Pat. 26(5), 555–564 (2016).
  • 64. Hou DY, Muller AJ, Sharma MD et al. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 67(2), 792–801 (2007).
  • 65. Soliman HH, Jackson E, Neuger T 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(18), 8136–8146 (2014). • This article describes the use of indoleamine 2,3-dioxygenase inhibitor in combination with chemotherapy and providing the proof of concept.
  • 66. Brochez L, Chevolet I, Kruse V. The rationale of indoleamine 2,3-dioxygenase inhibition for cancer therapy. Eur. J. Cancer 76, 167–182 (2017).
  • 67. Muller AJ, Prendergast GC. Marrying immunotherapy with chemotherapy: why say IDO? Cancer Res. 65(18), 8065–8068 (2005).
  • 68. Lamble AJ, Lind EF. Targeting the immune microenvironment in acute myeloid leukemia: a focus on T cell immunity. Front. Oncol. 8, 213 (2018).