• Ei tuloksia

6�3�2 Extracellular vesicles in tryptophan catabolism

The importance of tryptophan catabolism in a wide variety of cancers is well established. Our metabolomic study revealed two EV-derived metabolites of the tryptophan pathway, reaching statistical significance based on the significance versus fold-change values (FC > 2, p ≤ 0.05) compared with controls. The prostate carcinoma cell line-derived EVs carrying tryptophan were significantly enriched (log2 (FC) = 2.8, p = 0.0007), and the colon carcinoma kynurenine was significantly decreased (log2 (FC) = -4.9, p = 0.04). Surprisingly, our results for these two metabolites were the opposite of the tryptophan and kynurenine concentrations detected in the serum or tissue samples of cancers. Accurately, tryptophan levels are commonly low and kynurenine levels are high, also detected in CRC and prostate carcinoma.318 This is in line with our study of CTCL, where plasma levels of tryptophan were significantly downregulated and kynurenine levels were upregulated.

The transport system of maintaining a high kynurenine level and a low tryptophan level in cancer cells is not fully understood.255 The drawback of our study is that only two metabolites from the kynurenine pathway reached the lower limit of quantification. However, the findings that kynurenine and tryptophan are packed into EVs will likely be important. The hypothesis that the Kyn/Trp balance regulation in cancers is EV-mediated may open a new perspective for exploring the transport mechanism further. Primarily, tryptophan was significantly enriched in cancer EVs, suggesting that the transport of tryptophan out of the cells may be EV-mediated. The opposite was true for kynurenine since kynurenine was significantly reduced in cancer EVs, supposed to maintain high kynurenine levels inside the cell. The phase III clinical trial of blocking the IDO1 with epacadostat did not reach significant improvement of melanoma patients, indicating limited

another mechanism, in addition to the IDO1, compensates the blocked IDO1 and maintains the Kyn/Trp ratio and promotes the immunosuppressive environment should be considered. Future studies should thus examine the role of EVs in IDO1/ TDO-mediated immunosuppression.

7. CONCLUSION

The findings presented in this thesis represent an essential contribution to understanding the role of TME in the pathogenesis of CTCL. Our efforts to analyze altered metabolites of the tryptophan pathway reveal the important role of IDO1 and TDO in different CTCL subtypes. In future, confirmation of the serum Kyn/Trp ratio as a biomarker of CTCL may be valuable. Furthermore, the strategy of combined IDO1/ TDO inhibition therapy may achieve superior results for CTCL treatment, as TDO acts as a compensatory enzyme for IDO1, both expressed in CTCLs.

The discovery that the human endogenous retrovirus origin syncytin-1 was overexpressed in CTCL cell lines and in cell-derived EVs may help answer the question of the evolutionary origin of EVs. Since human syncytin-1 has retained its receptor-mediated cell membrane fusion capacity, the syncytin-1-carrying EVs may facilitate cell-to-cell fusion in cancer cells and transfer tumor cell signals to recipient cells. Targeting and eradicating cancer-associated molecules exposed on the surface of cancer-EVs but absent in normal EVs, including syncytin-1, may be a valuable next-generation strategy for cancer treatment.

Our findings of metabolomic alterations in cancer-derived EVs, including CTCL, prostate carcinoma, and colon carcinoma, reflects the cancer-associated metabolic imbalance into EVs. We detected common metabolites of EVs, namely proline, succinate, folate, and creatinine, from all studies cancer types. These altered EV-transported metabolites may be useful in discovery of biomarkers and in shedding light on the complex metabolic network that is fundamental for cancer progression.

8. ACKNOWLEDGMENTS

This study was conducted at the Department of Dermatology and Allergology, University of Helsinki, and Helsinki University Hospital, Finland, during the years 2016–2021. I want to acknowledge the high-quality research facility and inspiring work environment at the Skin and Allergy hospital and Biomedicum Helsinki. I want to express my gratitude to the patients and volunteers who donated their samples to this research.

The study has been financially supported by grants from the Cancer Foundation Finland, the Finnish Dermatological Society, the Ida Montin Foundation, and the Doctoral School in Health Science. I am grateful for their support.

I am incredibly thankful to my supervisor Professor Annamari Ranki for the opportunity to conduct my doctoral thesis and for her support, mentorship, and patience during this project. Her experience, academic knowledge, positive energy, and kindness are admirable.

I thank my co-authors, collaborators, and colleagues. I am incredibly thankful to Francois Mallet for his excellent knowledge about the syncytin-1 protein.

He was always willing to answer my questions and provided valuable opinions during our project. I am grateful to the personnel of the EV core facility for their impressive knowledge and cooperation concerning extracellular vesicles. Without their help, this thesis would not be possible. Special thanks to Mari Palviainen, who guided me to the world of extracellular vesicles. It has been an excellent opportunity to work with Mari. Her inspiring, helpful, and optimistic attitude has made my journey marvelous. I appreciate your friendship! I thank Pia Siljander for the excellent collaboration. Her knowledge of extracellular vesicles is impressive. I thank Maija Puhka for the opportunity to learn so much about electron microscopy. I have spent wonderful times taking photographs from the EVs and enjoying your peaceful company. I thank Biomedicum Imaging Unit for advice and support. Special thanks to Pilvi Maliniemi for sharing your article with me. It was a pleasure to work with you!

I want to express my gratitude to all my lab members. Especially Annika, thank you for sharing your knowledge and cooperating during my projects. Alli, without you, it would be impossible to work in the lab. You take care of everything, and I am always grateful for your assistance. Thank you, Eira, for the excellent company, for example, at the faculty Christmas party! Thank you, Al Amin, for your peaceful attitude and company. Thank you Martta, Liisa, and Tea from the IAS. Tea, I want to thank you for your wonderful and inspiring attitude during our current project! Thank you, Inga, and Mirjam for your technical assistance.

It has been pleasant to work with you! Thank you, Juli, for your secretary’s help.

I appreciate your helpfulness. I want to thank previous lab members: Kaija, Dmitri, and Markus. I am also grateful to Professor emeritus Kaj Krohn for his valuable knowledge and opinions.

I want to thank my thesis committee members, Professor Tom Böhling and Docent Erja Kerkelä, for their support during my thesis. Special thanks to Professor Jyrki Heino and Docent Kirsi Rilla for reviewing my thesis. Your views have been valuable. I am profoundly grateful to Professor Veli-Matti Kähäri for being my opponent. I am also appreciative of Doctoral School in Health Science and Doctoral Programme in Interactive Life Science for excellent courses and support during my thesis. Thank you, Carol Ann Pelli, for the language editing.

My journey to this moment has been long, unusual, and marvelous. I want to express my gratitude to all inspiring and intelligent people during my career.

First of all, I want to thank Professor Helena Kääriäinen, who offered me the opportunity to work at the Department of Medical Genetics beginning of the year 1991. Since then, I wanted to study more molecular biology, and fortunately, I had wonderful opportunities to work in excellent research groups and take part in many colleagues’ PhD projects. I will never forget Professor Albert de la Chapelle and his impressive intelligence. I also have the honor of doing research with Professor Kristina Aittomäki, who greatly encouraged and inspired me. I want to especially thank Professor Lauri Aaltonen and his research group for an encouraging and wonderful working environment that inspired me to continue my studies. So many people I am grateful to, but it would take several pages to write all names here! I will never forget the Karyon Oy and its funny and lovely colleagues and I am grateful for their company and especially for the nice parties!

I want to thank Professor Iiris Hovatta for her excellent skill in supervising me in my pro graduate thesis. I had a wonderful time in your friendly and talented lab. The department of Equine and Small Animal Medicine: Merja, Kaisa and Taina, I want to thank you for the best possible friendship and support.

Rakkaimmat kiitokset perheelleni ja ystävilleni. Ilman teidän kannustavaa tukeanne väitöskirjani ei olisi valmistunut. Kiitos Elli, että sain käyttää maalaustasi kirjani kannessa. Kiitos Satu, Paula, Kiiki, Maria, Lotta, Heikki ja Tani-serkku sekä monet muut ystäväni kannustuksesta ja hauskoista yhteisistä rentoutushetkistä kiireenkin keskellä. Kiitos sielunsiskoni Taija ihanista keskusteluista sekä tuesta elämän monissa kiemuroissa. Olet esikuvani niin tieteen saralla kuin myös ihanana ihmisenä. Kiitos isä ja äiti kaikesta mahdollisesta tuesta ja välittämisestä koko elämäni aikana. Isälle vielä kiitos kiinnostuksesta biologiaa kohtaan. Samille kiitos olemalla oma itsesi sekä ”sparraavista” ajatuksistasi. Olet kyllä hyvä tyyppi!

Sara siskoni ja sielunkumppanini, eihän tästä elämästä tulisi mitään ilman sinua.

Kiitos tuntien keskusteluista sekä, että olet ollut läsnä ja tukenani koko elämäni ajan. Kiitos Emmille, kun olet jaksanut vastailla kysymyksiini ”kieliongelmissa”.

On ollut hienoa seurata opiskeluasi ja rohkeuttasi tehdä päätöksiä. Rakkaat

tottuneet siihen, että äiti opiskelee aina jotain ja suhtaudutte siihen kannustavasti ja ylpeydellä. Rakkaimmat kiitokset kaikesta! Ari, rakkauteni, kumppanini ja mentorini, olet auttanut minua selviämään tästä koitoksesta olemalla läsnä, kuuntelemalla, kannustamalla sekä ihan konkreettisesti monissa teknisissä pulmissa, kirjan taittamisessa sekä kuvien viimeistelyssä. Sanat eivät riitä osoittamaan kiitollisuuttani!

Kirsi Laukkanen Porvoo, August 2021

9. REFERENCES

1. Sung, H. et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 71, 209-249 (2021).

2. Arneth, B. Tumor Microenvironment. Medicina (Kaunas) 56 (2019).

3. Bussard, K. M., Mutkus, L., Stumpf, K., Gomez-Manzano, C. & Marini, F. C.

Tumor-associated stromal cells as key contributors to the tumor microenvironment.

Breast Cancer Res 18, 84 (2016).

4. Whiteside, T. L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 27, 5904-5912 (2008).

5. Hinshaw, D. C. & Shevde, L. A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res 79, 4557-4566 (2019).

6. Yáñez-Mó, M. et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles 4, 27066 (2015).

7. De Broe, M., Wieme, R. & Roels, F. Letter: Membrane fragments with koinozymic properties released from villous adenoma of the rectum. Lancet 2, 1214-1215 (1975).

8. Bebelman, M. P., Smit, M. J., Pegtel, D. M. & Baglio, S. R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol Ther 188, 1-11 (2018).

9. Urabe, F. et al. Extracellular vesicles as biomarkers and therapeutic targets for cancer.

Am J Physiol Cell Physiol 318, C29-C39 (2020).

10. Aheget, H. et al. Exosome: A New Player in Translational Nanomedicine. J Clin Med 9 (2020).

11. Khawar, M. B., Abbasi, M. H., Siddique, Z., Arif, A. & Sheikh, N. An Update on Novel Therapeutic Warfronts of Extracellular Vesicles (EVs) in Cancer Treatment:

Where We Are Standing Right Now and Where to Go in the Future. Oxid Med Cell Longev 2019, 9702562 (2019).

12. Willemze R, Cerroni L, Kempf W, et al. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood. 2019;133(16):1703-1714.

Blood 134, 1112 (2019).

13. Stolearenco, V. et al. Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma. Front Cell Dev Biol 8, 851 (2020).

14. Bobrowicz, M. et al. Pathogenesis and Therapy of Primary Cutaneous T-Cell Lymphoma: Collegium Internationale Allergologicum (CIA) Update 2020. Int Arch

15. Yumeen, S. & Girardi, M. Insights Into the Molecular and Cellular Underpinnings of Cutaneous T Cell Lymphoma. Yale J Biol Med 93, 111-121 (2020).

16. Keto, J., Hahtola, S., Linna, M. & Väkevä, L. Mycosis fungoides and Sézary syndrome: a population-wide study on prevalence and health care use in Finland in 1998-2016. BMC Health Serv Res 21, 166 (2021).

17. Hodak, E. et al. Juvenile mycosis fungoides: cutaneous T-cell lymphoma with frequent follicular involvement. J Am Acad Dermatol 70, 993-1001 (2014).

18. Melhoranse Gouveia, B. et al. Reflectance confocal microscopy role in mycosis fungoides follow-up. Skin Res Technol (2020).

19. Girardi, M., Heald, P. W. & Wilson, L. D. The pathogenesis of mycosis fungoides.

N Engl J Med 350, 1978-1988 (2004).

20. Talpur, R. et al. Long-term outcomes of 1,263 patients with mycosis fungoides and Sézary syndrome from 1982 to 2009. Clin Cancer Res 18, 5051-5060 (2012).

21. Oka, T. & Miyagaki, T. Novel and Future Therapeutic Drugs for Advanced Mycosis Fungoides and Sézary Syndrome. Front Med (Lausanne) 6 (2019).

22. Morris, L., Tran, J. & Duvic, M. Non-Classic Signs of Sézary Syndrome: A Review.

Am J Clin Dermatol 21, 383-391 (2020).

23. Quaglino, P. et al. Phenotypical Markers, Molecular Mutations, and Immune Microenvironment as Targets for New Treatments in Patients with Mycosis Fungoides and/or Sézary Syndrome. J Invest Dermatol (2020).

24. Cheng, J., Zhu, H. & Choi, J. K. CD30 Expression in Pediatric Neoplasms, Study of 585 Cases. Pediatr Dev Pathol 20, 191-196 (2017).

25. Brown, R. A., Fernandez-Pol, S. & Kim, J. Primary cutaneous anaplastic large cell lymphoma. J Cutan Pathol 44, 570-577 (2017).

26. Moodley, N., Nombona, P. & Mosam, A. Primary Cutaneous Anaplastic Large-Cell Lymphoma. Dermatopathology (Basel) 6, 163-169 (2019).

27. Querfeld, C. et al. Primary cutaneous and systemic anaplastic large cell lymphoma:

clinicopathologic aspects and therapeutic options. Oncology (Williston Park) 24, 574-587 (2010).

28. Martinez-Cabriales, S. A., Walsh, S., Sade, S. & Shear, N. H. Lymphomatoid papulosis: an update and review. J Eur Acad Dermatol Venereol 34, 59-73 (2020).

29. Nijsten, T., Curiel-Lewandrowski, C. & Kadin, M. E. Lymphomatoid papulosis in children: a retrospective cohort study of 35 cases. Arch Dermatol 140, 306-312 (2004).

30. Willemze, R. et al. Subcutaneous panniculitis-like T-cell lymphoma: definition, classification, and prognostic factors: an EORTC Cutaneous Lymphoma Group Study of 83 cases. Blood 111, 838-845 (2008).

31. Sugeeth, M. T., Narayanan, G., Jayasudha, A. V. & Nair, R. A. Subcutaneous panniculitis-like T-cell lymphoma. Proc (Bayl Univ Med Cent) 30, 76-77 (2017).

32. Trautinger, F. Phototherapy of mycosis fungoides. Photodermatol Photoimmunol Photomed 27, 68-74 (2011).

33. Trautinger, F. et al. European Organisation for Research and Treatment of Cancer consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome – Update 2017. European Journal of Cancer 77, 57-74 (2017).

34. Yu, X., Marshall, M. J. E., Cragg, M. S. & Crispin, M. Improving Antibody-Based Cancer Therapeutics Through Glycan Engineering. BioDrugs 31, 151-166 (2017).

35. Scarisbrick, J. J. Brentuximab vedotin therapy for CD30-positive cutaneous T-cell lymphoma: a targeted approach to management. Future Oncol 13, 2405-2411 (2017).

36. Sunshine, J. & Taube, J. M. PD-1/PD-L1 inhibitors. Curr Opin Pharmacol 23, 32-38 (2015).

37. Cetinözman, F., Jansen, P. M., Vermeer, M. H. & Willemze, R. Differential expression of programmed death-1 (PD-1) in Sézary syndrome and mycosis fungoides.

Arch Dermatol 148, 1379-1385 (2012).

38. Samimi, S. et al. Increased programmed death-1 expression on CD4+ T cells in cutaneous T-cell lymphoma: implications for immune suppression. Arch Dermatol 146, 1382-1388 (2010).

39. Khodadoust, M. S. et al. Pembrolizumab in Relapsed and Refractory Mycosis Fungoides and Sézary Syndrome: A Multicenter Phase II Study. J Clin Oncol 38, 20-28 (2020).

40. Lindahl, L. M. et al. Antibiotics inhibit tumor and disease activity in cutaneous T-cell lymphoma. Blood 134, 1072-1083 (2019).

41. Larocca, C. & Kupper, T. Mycosis Fungoides and Sézary Syndrome: An Update.

Hematol Oncol Clin North Am 33, 103-120 (2019).

42. Johnson, W. T. et al. Allogeneic hematopoietic stem cell transplantation in advanced stage mycosis fungoides and Sézary syndrome: a concise review. Chin Clin Oncol 8, 12 (2019).

43. Alberts, B. et al. Lymphocytes and the Cellular Basis of Adaptive Immunity. Molecular Biology of the Cell� 4th edition (2002).

44. Krejsgaard, T., Odum, N., Geisler, C., Wasik, M. A. & Woetmann, A. Regulatory T cells and immunodeficiency in mycosis fungoides and Sézary syndrome. Leukemia 26, 424-432 (2012).

45. Iyer, A. et al. Skin colonization by circulating neoplastic clones in cutaneous T-cell lymphoma. Blood 134, 1517-1527 (2019).

46. Iyer, A. et al. Branched evolution and genomic intratumor heterogeneity in the

47. Gaydosik, A. M. et al. Genome-wide transcriptome analysis of the STAT6-regulated genes in advanced-stage cutaneous T-cell lymphoma. Blood 136, 1748-1759 (2020).

48. Thode, C. et al. Malignant T cells secrete galectins and induce epidermal hyperproliferation and disorganized stratification in a skin model of cutaneous T-cell lymphoma. J Invest Dermatol 135, 238-246 (2015).

49. Willerslev-Olsen, A. et al. Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma. Blood 127, 1287-1296 (2016).

50. Krejsgaard, T. et al. Malignant inflammation in cutaneous T-cell lymphoma-a hostile takeover. Semin Immunopathol 39, 269-282 (2017).

51. Rassek, K. & Iżykowska, K. Single-Cell Heterogeneity of Cutaneous T-Cell Lymphomas Revealed Using RNA-Seq Technologies. Cancers (Basel) 12 (2020).

52. Borcherding, N. et al. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin Cancer Res 25, 2996-3005 (2019).

53. Ghazawi, F. M. et al. Environmental and Other Extrinsic Risk Factors Contributing to the Pathogenesis of Cutaneous T Cell Lymphoma (CTCL). Front Oncol 9, 300 (2019).

54. Ferlay, J. et al. Cancer statistics for the year 2020: an overview. Int J Cancer (2021).

55. Gupta, S. et al. Prostate Cancer: How Young is too Young? Curr Urol 9, 212-215 (2017).

56. The Molecular Taxonomy of Primary Prostate Cancer. Cell 163, 1011-1025 (2015).

57. Merriel, S. W. D., Funston, G. & Hamilton, W. Prostate Cancer in Primary Care.

Adv Ther 35, 1285-1294 (2018).

58. Wang, G., Zhao, D., Spring, D. J. & DePinho, R. A. Genetics and biology of prostate cancer. Genes Dev 32, 1105-1140 (2018).

59. Lima, A. R., Bastos, M. d. L., Carvalho, M. & Guedes de Pinho, P. Biomarker Discovery in Human Prostate Cancer: an Update in Metabolomics Studies. Transl Oncol 9, 357-370 (2016).

60. Aoun, F., Peltier, A. & van Velthoven, R. A comprehensive review of contemporary role of local treatment of the primary tumor and/or the metastases in metastatic prostate cancer. Biomed Res Int 2014, 501213 (2014).

61. Roberts, M. J., Schirra, H. J., Lavin, M. F. & Gardiner, R. A. Metabolomics: a novel approach to early and noninvasive prostate cancer detection. Korean J Urol 52, 79-89 (2011).

62. Trock, B. J. Application of metabolomics to prostate cancer. Urol Oncol 29, 572-581 (2011).

63. Bader, D. A. & McGuire, S. E. Tumour metabolism and its unique properties in prostate adenocarcinoma. Nat Rev Urol 17, 214-231 (2020).

64. DeSantis, C. E. et al. Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin 64, 252-271 (2014).

65. Araghi, M. et al. Global trends in colorectal cancer mortality: projections to the year 2035. Int J Cancer 144, 2992-3000 (2019).

66. Baran, B. et al. Difference Between Left-Sided and Right-Sided Colorectal Cancer:

A Focused Review of Literature. Gastroenterology Res 11, 264-273 (2018).

67. Paschke, S. et al. Are Colon and Rectal Cancer Two Different Tumor Entities? A Proposal to Abandon the Term Colorectal Cancer. Int J Mol Sci 19 (2018).

68. Galon, J. et al. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 232, 199-209 (2014).

69. Brenner, H., Kloor, M. & Pox, C. P. Colorectal cancer. Lancet 383, 1490-1502 (2014).

70. Hong, T. S., Clark, J. W. & Haigis, K. M. Cancers of the colon and rectum: identical or fraternal twins? Cancer Discov 2, 117-121 (2012).

71. Glebov, O. K. et al. Distinguishing right from left colon by the pattern of gene expression. Cancer Epidemiol Biomarkers Prev 12, 755-762 (2003).

72. Boland, C. R. & Goel, A. Microsatellite instability in colorectal cancer. Gastroenterology 138, 2073-2087.e3 (2010).

73. Gatalica, Z., Vranic, S., Xiu, J., Swensen, J. & Reddy, S. High microsatellite instability (MSI-H) colorectal carcinoma: a brief review of predictive biomarkers in the era of personalized medicine. Fam Cancer 15, 405-412 (2016).

74. André, T. et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N Engl J Med 383, 2207-2218 (2020).

75. Fearon, E. R. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61, 759-767 (1990).

76. Opitz, C. A. et al. The therapeutic potential of targeting tryptophan catabolism in cancer. Br J Cancer 122, 30-44 (2020).

77. Platten, M., Nollen, E. A. A., Röhrig, U. F., Fallarino, F. & Opitz, C. A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond.

Nat Rev Drug Discov 18, 379-401 (2019).

78. Fuertig, R. et al. LC-MS/MS-based quantification of kynurenine metabolites, tryptophan, monoamines and neopterin in plasma, cerebrospinal fluid and brain.

Bioanalysis 8, 1903-1917 (2016).

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

80. Zhai, L. et al. Immunosuppressive IDO in Cancer: Mechanisms of Action, Animal Models, and Targeting Strategies. Front Immunol 11, 1185 (2020).

81. Walczak, K., Wnorowski, A., Turski, W. A. & Plech, T. Kynurenic acid and cancer:

facts and controversies. Cell Mol Life Sci 77, 1531-1550 (2020).

82. Munn, D. H. et al. Prevention of allogeneic fetal rejection by tryptophan catabolism.

Science 281, 1191-1193 (1998).

83. Munn, D. H. & Mellor, A. L. IDO in the Tumor Microenvironment: Inflammation,

84. Meireson, A., Devos, M. & Brochez, L. IDO Expression in Cancer: Different Compartment, Different Functionality? Front� Immunol� 11 (2020).

85. 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).

86. 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).

87. Stevens, E. A., Mezrich, J. D. & Bradfield, C. A. The aryl hydrocarbon receptor: a perspective on potential roles in the immune system. Immunology 127, 299-311 (2009).

88. 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).

89. Hwu, P. et al. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol 164, 3596-3599 (2000).

90. Pantouris, G., Serys, M., Yuasa, H. J., Ball, H. J. & Mowat, C. G. Human indoleamine 2,3-dioxygenase-2 has substrate specificity and inhibition characteristics distinct from those of indoleamine 2,3-dioxygenase-1. Amino Acids 46, 2155-2163 (2014).

91. Taylor, M. W. & Feng, G. S. Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB J 5, 2516-2522 (1991).

92. Fujigaki, H. et al. The signal transducer and activator of transcription 1alpha and interferon regulatory factor 1 are not essential for the induction of indoleamine

92. Fujigaki, H. et al. The signal transducer and activator of transcription 1alpha and interferon regulatory factor 1 are not essential for the induction of indoleamine