Deep learning model enables the discovery of a novel immunotherapeutic agent regulating the kynurenine pathway

Background: Kynurenine (Kyn) is a key inducer of an immunosuppressive tumor microenvironment (TME). Although indoleamine 2,3-dioxygenase (IDO)-selective inhibitors have been developed to suppress the Kyn pathway, the results were not satisfactory due to the presence of various opposing mechanisms. Here, we employed an orally administered novel Kyn pathway regulator to overcome the limitation of anti-tumor immune response.Methods: We identified a lead compound through the development of a deep learning model to predict compound-target interaction. Inhibitory activity of the lead compound against both IDO and tryptophan 2,3-dioxygenase (TDO) was evaluated using enzyme- and cell-based assays. CT26 colon cancer-bearing mice were treated with the lead compound and/or immune checkpoint inhibitors. Tumor samples were analyzed using histological, flow cytometry, and immune profiling assays.Results: We identified a novel core structure that inhibited both IDO and TDO. An orally available lead compound, STB-C017 (designated hereafter as STB), was then selected via validation of structural modification and in vitro assays. STB effectively inhibited the enzymatic and cellular activity of IDO and TDO in vitro. Moreover, it potently suppressed Kyn levels in both the plasma and tumor in vivo. The dose and schedule of STB administration were optimized based on the immunotherapeutic efficacy and toxicity. STB monotherapy increased the infiltration of CD8+ T cells into TME. In addition, it elicited an anti-cancer immune response with a strong activation of T cell immunity. STB reprogrammed the TME with widespread changes in immune-mediated gene signatures. Notably, STB-based combination immunotherapy elicited the most potent anti-tumor efficacy through concurrent treatment with immune checkpoint inhibitors (ɑPD-1 and/or ɑCTLA-4), leading to complete tumor regression and long-term overall survival.Conclusions: Our study demonstrated that a novel Kyn pathway regulator derived using deep learning technology can activate T cell immunity and potentiate immune checkpoint blockade by overcoming an immunosuppressive TME.

[1]  J. Fridman,et al.  Hydroxyamidine Inhibitors of Indoleamine-2,3-dioxygenase Potently Suppress Systemic Tryptophan Catabolism and the Growth of IDO-Expressing Tumors , 2010, Molecular Cancer Therapeutics.

[2]  Bao-sheng Chen,et al.  IDO1 and Kynurenine Pathway Metabolites Activate PI3K-Akt Signaling in the Neoplastic Colon Epithelium to Promote Cancer Cell Proliferation and Inhibit Apoptosis. , 2019, Cancer research.

[3]  A. von Deimling,et al.  Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, STAT3 and the AHR , 2014, Oncotarget.

[4]  Yu Seong Lee,et al.  STING signaling is a potential immunotherapeutic target in colorectal cancer , 2019, Journal of Cancer.

[5]  J. McCarter,et al.  Purification and kinetic characterization of human indoleamine 2,3-dioxygenases 1 and 2 (IDO1 and IDO2) and discovery of selective IDO1 inhibitors. , 2011, Biochimica et biophysica acta.

[6]  Joo Hoon Kim,et al.  STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade. , 2019, The Journal of clinical investigation.

[7]  Yi Fang,et al.  Tumor-Repopulating Cells Induce PD-1 Expression in CD8+ T Cells by Transferring Kynurenine and AhR Activation. , 2018, Cancer cell.

[8]  J. Wolchok,et al.  Tumor-Expressed IDO Recruits and Activates MDSCs in a Treg-Dependent Manner. , 2015, Cell reports.

[9]  Robert R Yauch,et al.  Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice , 2017, Nature Communications.

[10]  D. Han,et al.  Combination of Irreversible Electroporation and STING Agonist for Effective Cancer Immunotherapy , 2020, Cancers.

[11]  Asher Mullard IDO takes a blow , 2018, Nature Reviews Drug Discovery.

[12]  B. J. Van den Eynde,et al.  Inhibition of Tryptophan-Dioxygenase Activity Increases the Antitumor Efficacy of Immune Checkpoint Inhibitors , 2019, Cancer Immunology Research.

[13]  Qingzhu Wang,et al.  Carboxyamidotriazole combined with IDO1-Kyn-AhR pathway inhibitors profoundly enhances cancer immunotherapy , 2019, Journal of Immunotherapy for Cancer.

[14]  Y. Saga,et al.  Indoleamine 2,3-dioxygenase promotes peritoneal dissemination of ovarian cancer through inhibition of natural killercell function and angiogenesis promotion. , 2011, International journal of oncology.

[15]  Tadhg P Begley,et al.  Crystal structure and mechanism of tryptophan 2,3-dioxygenase, a heme enzyme involved in tryptophan catabolism and in quinolinate biosynthesis. , 2007, Biochemistry.

[16]  S. Gou,et al.  The introduction of immunosuppressor (TDO inhibitor) significantly improved the efficacy of irinotecan in treating hepatocellular carcinoma , 2020, Cancer Immunology, Immunotherapy.

[17]  Su-Jin Shin,et al.  High endothelial venule is a surrogate biomarker for T-cell inflamed tumor microenvironment and prognosis in gastric cancer , 2021, Journal for ImmunoTherapy of Cancer.

[18]  G. Kenter,et al.  Indoleamine-2,3-dioxygenase (IDO) metabolic activity is detrimental for cervical cancer patient survival , 2015, Oncoimmunology.

[19]  J. Wolchok,et al.  Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine , 2020, Nature Communications.

[20]  C. Uyttenhove,et al.  Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase , 2012, Proceedings of the National Academy of Sciences.

[21]  C. Drake,et al.  Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.

[22]  Fangxuan Li,et al.  Tumor-infiltrating Treg, MDSC, and IDO expression associated with outcomes of neoadjuvant chemotherapy of breast cancer , 2018, Cancer biology & therapy.

[23]  Y. Cheng,et al.  Durable Therapeutic Efficacy Utilizing Combinatorial Blockade against IDO, CTLA-4, and PD-L1 in Mice with Brain Tumors , 2014, Clinical Cancer Research.

[24]  H. Schreiber,et al.  Innate and adaptive immune cells in the tumor microenvironment , 2013, Nature Immunology.

[25]  W. Lee,et al.  Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity , 2020, Experimental & Molecular Medicine.

[26]  G. G. Galli,et al.  The landscape of cancer cell line metabolism , 2019, Nature Medicine.

[27]  M. Bower,et al.  INCB24360 (Epacadostat), a Highly Potent and Selective Indoleamine-2,3-dioxygenase 1 (IDO1) Inhibitor for Immuno-oncology , 2017, ACS medicinal chemistry letters.

[28]  M. Weller,et al.  An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor , 2011, Nature.

[29]  J. Luke,et al.  Reimagining IDO Pathway Inhibition in Cancer Immunotherapy via Downstream Focus on the Tryptophan–Kynurenine–Aryl Hydrocarbon Axis , 2018, Clinical Cancer Research.

[30]  J. Hunt,et al.  Preclinical Characterization of Linrodostat Mesylate, a Novel, Potent, and Selective Oral Indoleamine 2,3-Dioxygenase 1 Inhibitor , 2020, Molecular Cancer Therapeutics.

[31]  A. Zippelius,et al.  A highly efficient modality to block the degradation of tryptophan for cancer immunotherapy: locked nucleic acid-modified antisense oligonucleotides to inhibit human indoleamine 2,3-dioxygenase 1/tryptophan 2,3-dioxygenase expression , 2019, Cancer Immunology, Immunotherapy.

[32]  Olivier Michielin,et al.  Challenges in the Discovery of Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitors. , 2015, Journal of medicinal chemistry.

[33]  C. Junghanss,et al.  Activation of the Kynurenine Pathway in Human Malignancies Can Be Suppressed by the Cyclin-Dependent Kinase Inhibitor Dinaciclib , 2020, Frontiers in Immunology.

[34]  E. Shin,et al.  Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. , 2020, Journal of hepatology.

[35]  Masahiro Ito,et al.  Serum kynurenine levels are a novel biomarker to predict the prognosis of patients with hepatocellular carcinoma , 2020, PloS one.

[36]  Joo Hoon Kim,et al.  Tumor Microenvironment Remodeling by Intratumoral Oncolytic Vaccinia Virus Enhances the Efficacy of Immune-Checkpoint Blockade , 2018, Clinical Cancer Research.

[37]  J. Wargo,et al.  Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy , 2017, Cell.

[38]  T. Gajewski The Next Hurdle in Cancer Immunotherapy: Overcoming the Non-T-Cell-Inflamed Tumor Microenvironment. , 2015, Seminars in oncology.

[39]  Hyae-Young Kim,et al.  Sequential Treatment with an Immune Checkpoint Inhibitor Followed by a Small-Molecule Targeted Agent Increases Drug-Induced Pneumonitis , 2020, Cancer research and treatment : official journal of Korean Cancer Association.

[40]  Qian Wang,et al.  Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. , 2010, Blood.

[41]  C. Uyttenhove,et al.  Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase , 2003, Nature Medicine.

[42]  Shanshan Zhu,et al.  High Indoleamine 2,3-Dioxygenase Is Correlated With Microvessel Density and Worse Prognosis in Breast Cancer , 2018, Front. Immunol..

[43]  P. Wallace,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. , 2009, Cancer research.

[44]  H. Koblish,et al.  Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8+ T cells directly within the tumor microenvironment , 2014, Journal of Immunotherapy for Cancer.

[45]  Jedd D. Wolchok,et al.  Cancer immunotherapy using checkpoint blockade , 2018, Science.

[46]  G. Prendergast,et al.  Accumulation of an Endogenous Tryptophan-Derived Metabolite in Colorectal and Breast Cancers , 2015, PloS one.

[47]  H. Oberg,et al.  Influence of Indoleamine-2,3-Dioxygenase and Its Metabolite Kynurenine on γδ T Cell Cytotoxicity against Ductal Pancreatic Adenocarcinoma Cells , 2020, Cells.

[48]  Milton W. Taylor,et al.  Indoleamine 2,3-Dioxygenase Production by Human Dendritic Cells Results in the Inhibition of T Cell Proliferation , 2000, The Journal of Immunology.

[49]  Xiaomin Luo,et al.  Discovery of Novel Inhibitors of Indoleamine 2,3-Dioxygenase 1 Through Structure-Based Virtual Screening , 2018, Front. Pharmacol..

[50]  P. Marchetti,et al.  Can IDO activity predict primary resistance to anti-PD-1 treatment in NSCLC? , 2018, Journal of Translational Medicine.

[51]  Xiaoyue Lin,et al.  Local expression of indoleamine 2,3-dioxygenase protects engraftment of xenogeneic skin substitute. , 2006, The Journal of investigative dermatology.

[52]  James R. Anderson,et al.  Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. , 2019, The Lancet. Oncology.

[53]  Chan Kim,et al.  Combination Immunotherapy Using Oncolytic Virus for the Treatment of Advanced Solid Tumors , 2020, International journal of molecular sciences.

[54]  Yu Seong Lee,et al.  Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer , 2020, Journal for ImmunoTherapy of Cancer.

[55]  G. Prendergast,et al.  Inhibiting IDO pathways to treat cancer: lessons from the ECHO-301 trial and beyond , 2018, Seminars in Immunopathology.