1
|
Muller AJ, Mondal A, Dey S, Prendergast GC. IDO1 and inflammatory neovascularization: bringing new blood to tumor-promoting inflammation. Front Oncol 2023; 13:1165298. [PMID: 37182174 PMCID: PMC10172587 DOI: 10.3389/fonc.2023.1165298] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
In parallel with the genetic and epigenetic changes that accumulate in tumor cells, chronic tumor-promoting inflammation establishes a local microenvironment that fosters the development of malignancy. While knowledge of the specific factors that distinguish tumor-promoting from non-tumor-promoting inflammation remains inchoate, nevertheless, as highlighted in this series on the 'Hallmarks of Cancer', it is clear that tumor-promoting inflammation is essential to neoplasia and metastatic progression making identification of specific factors critical. Studies of immunometabolism and inflamometabolism have revealed a role for the tryptophan catabolizing enzyme IDO1 as a core element in tumor-promoting inflammation. At one level, IDO1 expression promotes immune tolerance to tumor antigens, thereby helping tumors evade adaptive immune control. Additionally, recent findings indicate that IDO1 also promotes tumor neovascularization by subverting local innate immunity. This newly recognized function for IDO1 is mediated by a unique myeloid cell population termed IDVCs (IDO1-dependent vascularizing cells). Initially identified in metastatic lesions, IDVCs may exert broader effects on pathologic neovascularization in various disease settings. Mechanistically, induction of IDO1 expression in IDVCs by the inflammatory cytokine IFNγ blocks the antagonistic effect of IFNγ on neovascularization by stimulating the expression of IL6, a powerful pro-angiogenic cytokine. By contributing to vascular access, this newly ascribed function for IDO1 aligns with its involvement in other cancer hallmark functionalities, (tumor-promoting inflammation, immune escape, altered cellular metabolism, metastasis), which may stem from an underlying role in normal physiological functions such as wound healing and pregnancy. Understanding the nuances of how IDO1 involvement in these cancer hallmark functionalities varies between different tumor settings will be crucial to the future development of successful IDO1-directed therapies.
Collapse
Affiliation(s)
- Alexander J. Muller
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Alexander J. Muller,
| | - Arpita Mondal
- Arbutus Biopharma, Inc., Warminster, PA, United States
| | - Souvik Dey
- Wuxi Advanced Therapeutics, Inc., Philadelphia, PA, United States
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Brochez L, Kruse V, Schadendorf D, Muller AJ, Prendergast GC. Editorial: Targeting Indoleamine 2,3-dioxygenases and Tryptophan Dioxygenase for Cancer Immunotherapy. Front Immunol 2021; 12:789473. [PMID: 34938297 PMCID: PMC8686161 DOI: 10.3389/fimmu.2021.789473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lieve Brochez
- Department of Dermatology, University Hospital Ghent and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Vibeke Kruse
- Department of Medical Oncology, University Hospital Ghent and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dirk Schadendorf
- Department of Dermatology and Comprehensive Cancer Center, University Hospital Essen, Essen, Germany
| | | | | |
Collapse
|
3
|
Zhang X, Liu X, Zhou W, Du Q, Yang M, Ding Y, Hu R. Blockade of IDO-Kynurenine-AhR Axis Ameliorated Colitis-Associated Colon Cancer via Inhibiting Immune Tolerance. Cell Mol Gastroenterol Hepatol 2021; 12:1179-1199. [PMID: 34087454 PMCID: PMC8445903 DOI: 10.1016/j.jcmgh.2021.05.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Chronic inflammation in colon section is associated with an increased risk of colorectal cancer (CRC). Proinflammatory cytokines were produced in a tumor microenvironment and correlated with poor clinical outcome. Tumor-infiltrating T cells were reported to be greatly involved in the development of colon cancer. In this study, we demonstrated that kynurenine (Kyn), a metabolite catalyzed by indoleamine 2,3-dioxygenase (IDO), was required for IDO-mediated T cell function, and adaptive immunity indeed played a critical role in CRC. METHODS Supernatant of colon cancer cells was used to culture activated T cells and mice spleen lymphocytes, and the IDO1-Kyn-aryl hydrocarbon (AhR) receptor axis was determined in vitro. In vivo, an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced CRC model was established in IDO-/-, Rag1-/-, and wild-type mice, and tumor-associated T lymphocyte infiltration and Kyn/AhR signaling pathway changes were measured in each group. RESULTS Kyn promoted AhR nuclear translocation increased the transcription of Foxp3, a marker of regulatory T cells (Tregs), through improving the interaction between AhR and Foxp3 promoter. Additionally, compared WT mice, IDO-/- mice treated with AOM/DSS exhibited fewer and smaller tumor burdens in the colon, with less Treg and more CD8+ T cells infiltration, while Kyn administration abolished this regulation. Rag1-/- mice were more sensitive to AOM/DSS-induced colitis-associated colon cancer (CRC) compared with the wild-type mice, suggesting that T cell-mediated adaptive immunity indeed played a critical role in CRC. CONCLUSIONS We demonstrated that inhibition of IDO diminished Kyn/AhR-mediated Treg differentiation and could be an effective strategy for the prevention and treatment of inflammation-related colon cancer.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China; State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiuting Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China; Department of Children Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Mengdi Yang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
4
|
Prendergast GC, Metz R. A perspective on new immune adjuvant principles: Reprogramming inflammatory states to permit clearance of cancer cells and other age-associated cellular pathologies. Oncoimmunology 2021; 1:924-929. [PMID: 23162760 PMCID: PMC3489748 DOI: 10.4161/onci.21358] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aging entails the accumulation of neoantigens comprised of aggregated, oxidized, mutated and misfolded biomolecules, including advanced-glycation end projects (AGEs). There is evidence that the immune system can recognize and clear cells fouled by these molecular debris, which contribute to the emergence of cancer and other major age-associated diseases such as atherogenic and neurodegenerative disorders. However, this process may become increasingly inefficient with aging, perhaps in part because of an insufficiency of adjuvant signals normally associated with infection that can program productive inflammatory states and properly orient the immune system toward regenerative healing. Here we propose conceptual foundations for exploring a small set of infection-associated molecules as potential immune adjuvants to reprogram non-productive inflammatory states in aging tissues, and to improve the clearance of cellular pathologies that engender age-associated disease. The proposed adjuvant classes include a subset of D-amino acids used by bacteria to disrupt biofilms; nucleoside derivatives of N6-methyladenine, which functions at the core of bacterial dam restriction systems; and derivatives of the galactosyl trisaccharide α-Gal, which invokes the hyperacute response in primates. These foreign amino acids, nucleosides and sugar molecules are generally rare or absent in humans, except in association with infections by bacteria, protists or nematodes. A rationale for exploration of these candidate adjuvant principles and their chemical derivatives is discussed in terms of their use in generalized strategies to improve the prevention or treatment of cancer and other age-associated diseases, as negative modifiers of aging.
Collapse
|
5
|
Metz R, Rust S, Duhadaway JB, Mautino MR, Munn DH, Vahanian NN, Link CJ, Prendergast GC. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: A novel IDO effector pathway targeted by D-1-methyl-tryptophan. Oncoimmunology 2021; 1:1460-1468. [PMID: 23264892 PMCID: PMC3525601 DOI: 10.4161/onci.21716] [Citation(s) in RCA: 308] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tryptophan catabolism by indoleamine 2,3-dioxygenase (IDO) alters inflammation and favors T-cell tolerance in cancer, but the underlying molecular mechanisms remain poorly understood. The integrated stress response kinase GCN2, a sensor of uncharged tRNA that is activated by amino acid deprivation, is recognized as an important effector of the IDO pathway. However, in a mouse model of inflammatory carcinogenesis, ablation of Gcn2 did not promote resistance against tumor development like the absence of IDO does, implying the existence of additional cancer-relevant pathways that operate downstream of IDO. Addressing this gap in knowledge, we report that the IDO-mediated catabolism of tryptophan also inhibits the immunoregulatory kinases mTOR and PKC-Θ, along with the induction of autophagy. These effects were relieved specifically by tryptophan but also by the experimental agent 1-methyl-D-tryptophan (D-1MT, also known as NLG8189), the latter of which reversed the inhibitory signals generated by IDO with higher potency. Taken together, our results implicate mTOR and PKC-Θ in IDO-mediated immunosuppressive signaling, and they provide timely insights into the unique mechanism of action of D-1MT as compared with traditional biochemical inhibitors of IDO. These findings are important translationally, because they suggest broader clinical uses for D-1MT against cancers that overexpress any tryptophan catabolic enzyme (IDO, IDO2 or TDO). Moreover, they define mTOR and PKC-Θ as candidate pharmacodynamic markers for D-1MT responses in patients recruited to ongoing phase IB/II cancer trials, addressing a current clinical need.
Collapse
|
6
|
Engin AB, Engin A. Indoleamine 2,3-Dioxygenase Activity-Induced Acceleration of Tumor Growth, and Protein Kinases-Related Novel Therapeutics Regimens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:339-356. [PMID: 33539022 DOI: 10.1007/978-3-030-49844-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is overexpressed in response to interferon-gamma (IFN-γ). IDO-mediated degradation of tryptophan (Trp) along the kynurenine (Kyn) pathway by immune cells is associated with the anti-microbial, and anti-tumor defense mechanisms. In contrast, IDO is constitutively expressed by various tumors and creates an immunosuppressive microenvironment around the tumor tissue both by depletion of the essential amino acid Trp and by formation of Kyn, which is immunosuppressive metabolite of Trp. IDO may activate its own expression in human cancer cells via an autocrine aryl hydrocarbon receptor (AhR)- interleukin 6 (IL-6)-signal transducer and activator of transcription 3 (STAT3) signaling loop. Although IDO is not a unique marker, in many clinical trials serum IDO activity is suggested to be an important parameter in the pathogenesis of cancer development and growth. Measuring IDO activity in serum seems to be an indicator of cancer growth rate, however, it is controversial whether this approach can be used as a reliable guide in cancer patients treated with IDO inhibitors. Thus, IDO immunostaining is strongly recommended for the identification of higher IDO producing tumors, and IDO inhibitors should be included in post-operative complementary therapy in IDO positive cancer cases only. Novel therapies that target the IDO pathway cover checkpoint protein kinases related combination regimens. Currently, multi-modal therapies combining IDO inhibitors and checkpoint kinase blockers in addition to T regulatory (Treg) cell-modifying treatments seem promising.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
7
|
Merlo LMF, DuHadaway JB, Montgomery JD, Peng WD, Murray PJ, Prendergast GC, Caton AJ, Muller AJ, Mandik-Nayak L. Differential Roles of IDO1 and IDO2 in T and B Cell Inflammatory Immune Responses. Front Immunol 2020; 11:1861. [PMID: 32973768 PMCID: PMC7461966 DOI: 10.3389/fimmu.2020.01861] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Indoleamine-2,3-dioxygenase (IDO)1 and IDO2 are two closely related tryptophan catabolizing enzymes encoded by linked genes. The IDO pathway is also immunomodulatory, with IDO1 well-characterized as a mediator of tumor immune evasion. Due to its homology with IDO1, IDO2 has been proposed to have a similar immunoregulatory function. Indeed, IDO2, like IDO1, is necessary for the differentiation of regulatory T cells in vitro. However, compared to IDO1, in vivo studies demonstrated a contrasting role for IDO2, with experiments in preclinical models of autoimmune arthritis establishing a proinflammatory role for IDO2 in mediating B and T cell activation driving autoimmune disease. Given their potentially opposing roles in inflammatory responses, interpretation of results obtained using IDO1 or IDO2 single knockout mice could be complicated by the expression of the other enzyme. Here we use IDO1 and IDO2 single and double knockout (dko) mice to define the differential roles of IDO1 and IDO2 in B cell-mediated immune responses. Autoreactive T and B cell responses and severity of joint inflammation were decreased in IDO2 ko, but not IDO1 ko arthritic mice. Dko mice had a reduction in the number of autoantibody secreting cells and severity of arthritis: however, percentages of differentiated T cells and their associated cytokines were not reduced compared to IDO1 ko or wild-type mice. These data suggest that autoreactive B cell responses are mediated by IDO2, while autoreactive T cell responses are indirectly affected by IDO1 expression in the IDO2 ko mice. IDO2 also influenced antibody responses in models of influenza infection and immunization with T cell-independent type II antigens. Taken together, these studies provide evidence for the contrasting roles IDO1 and IDO2 play in immune responses, with IDO1 mediating T cell suppressive effects and IDO2 working directly in B cells as a proinflammatory mediator of B cell responses.
Collapse
Affiliation(s)
- Lauren M F Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - James B DuHadaway
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | | | - Wei-Dan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Peter J Murray
- Immunoregulation Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States.,Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | | | | | | |
Collapse
|
8
|
Jonescheit H, Oberg HH, Gonnermann D, Hermes M, Sulaj V, Peters C, Kabelitz D, Wesch D. Influence of Indoleamine-2,3-Dioxygenase and Its Metabolite Kynurenine on γδ T Cell Cytotoxicity against Ductal Pancreatic Adenocarcinoma Cells. Cells 2020; 9:E1140. [PMID: 32384638 PMCID: PMC7290398 DOI: 10.3390/cells9051140] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a malignant gastrointestinal disease. The enzyme indoleamine-2,3-dioxgenase (IDO) is often overexpressed in PDAC and its downstream metabolite kynurenine has been reported to inhibit T cell activation and proliferation. Since γδ T cells are of high interest for T cell-based immunotherapy against PDAC, we studied the impact of IDO and kynurenine on γδ T cell cytotoxicity against PDAC cells. METHODS IDO expression was determined in PDAC cells by flow cytometry and Western blot analysis. PDAC cells were cocultured with γδ T cells in medium or were stimulated with phosphorylated antigens or bispecific antibody in the presence or absence of IDO inhibitors. Additionally, γδ T cells were treated with recombinant kynurenine. Read-out assays included degranulation, cytotoxicity and cytokine measurement as well as cell cycle analysis. RESULTS Since IDO overexpression was variable in PDAC, IDO inhibitors improved γδ T cell cytotoxicity only against some but not all PDAC cells. γδ T cell degranulation and cytotoxicity were significantly decreased after their treatment with recombinant kynurenine. CONCLUSIONS Bispecific antibody drastically enhanced γδ T cell cytotoxicity against all PDAC cells, which can be further enhanced by IDO inhibitors against several PDAC cells, suggesting a striking heterogeneity in PDAC escape mechanisms towards γδ T cell-mediated anti-tumor response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Daniela Wesch
- Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, D-24105 Kiel, Germany; (H.J.); (H.-H.O.); (D.G.); (M.H.); (V.S.); (C.P.); (D.K.)
| |
Collapse
|
9
|
Zhao Y, Wei L, Liu J, Li F. Chemoresistance was correlated with elevated expression and activity of indoleamine 2,3-dioxygenase in breast cancer. Cancer Chemother Pharmacol 2019; 85:77-93. [PMID: 31844921 DOI: 10.1007/s00280-019-04009-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) catalyses degradation of the essential amino acid tryptophan leading to the production of immunosuppressive kynurenine and tryptophan exhausting. IDO expression and activity contribute to aggressive tumor growth, inferior therapeutic gain and poor prognosis. The aim of this study was to explore the association between chemoresistance and IDO expression, activity in breast cancer METHODS: Immunohistochemistry was applied for evaluating IDO expression in biopsy tissues. Serum IDO activity was examined via High-performance liquid chromatography (HPLC). Western blots (WB), HPLC and Real-time PCR (RT-PCR) were used to analyze IDO protein, IDO enzyme activity and IDO gene expression in original and paclitaxel-resistant cells respectively. Logistic regression and survival analysis were applied to explore the association between chemoresistance and IDO expression, activity in breast cancer. RESULTS IDO expression in tumor tissues was associated with serum IDO activity (P = 0.004). Both IDO expression in tumor and serum activity were associated with clinical tumor stage, node stage and estrogen receptor (ER) status (all P < 0.05); clinical response and pathologic complete response (pCR) to NAC were both related to IDO expression and activity prior NAC (all P < 0.05). Multivariate analysis showed IDO activity before NAC was the only independent factor affected pCR (P = 0.032). ROC curves showed that the IDO expression and activity had discriminative ability for predicting the clinical response and pCR. In the prognostic analysis, patients with high IDO expression had significantly impaired overall survival (5 year survival rate: 53.57% vs 80.00%) and progression-free survival (5 year survival rate: 46.43% vs 72.00%, P = 0.031 and P = 0.046). In vitro, significantly increased IDO protein, IDO mRNA expression and IDO enzyme activity in paclitaxel-resistant cells were demonstrated in comparing of sensitive cells. CONCLUSION IDO expression and activity associated with advanced breast cancer, poor response to neoadjuvant chemotherapy and prognosis. IDO expression and activity were significantly increased in paclitaxel-resistant breast cancer cells.
Collapse
Affiliation(s)
- Yang Zhao
- The Second Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Breast Surgery, Cangzhou People's Hospital, Cangzhou, 061000, Hebei, China
| | - Lijuan Wei
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China
| | - Juntian Liu
- The Second Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China
| | - Fangxuan Li
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.
| |
Collapse
|
10
|
Xu X, Ren J, Ma Y, Liu H, Rong Q, Feng Y, Wang Y, Cheng Y, Ge R, Li Z, Bian J. Discovery of cyanopyridine scaffold as novel indoleamine-2,3-dioxygenase 1 (IDO1) inhibitors through virtual screening and preliminary hit optimisation. J Enzyme Inhib Med Chem 2019; 34:250-263. [PMID: 30734612 PMCID: PMC6327983 DOI: 10.1080/14756366.2018.1480614] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
With the aim of discovering novel IDO1 inhibitors, a combined similarity search and molecular docking approach was employed to the discovery of 32 hit compounds. Testing the screened hit compounds has led to several novel submicromolar inhibitors. Especially for compounds LVS-019 with cyanopyridine scaffold, showed good IDO1 inhibitory activity. To discover more compounds with similar structures to LVS-019, a shape-based model was then generated on the basis of it and the second-round virtual screening was carried out leading to 23 derivatives. Molecular docking studies suggested a possible binding mode of LVS-019, which provides a good starting point for the development of cyanopyridine scaffold compounds as potent IDO1 inhibitor. To improve potency of these hits, we further designed and synthesised another 14 derivatives of LVS-019. Among these compounds, LBJ-10 showed improved potency compared to the hits and displayed comparable potency to the control GDC-0919 analogue. LBJ-10 can serve as ideal leads for further modifications as IDO1 inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Xi Xu
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Jie Ren
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yinghe Ma
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Hongting Liu
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Quanjin Rong
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yifan Feng
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yameng Wang
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yu Cheng
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ruijia Ge
- c The Madeira School , McLean , VA , USA
| | - Zhiyu Li
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Jinlei Bian
- a State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
11
|
Domblides C, Lartigue L, Faustin B. Control of the Antitumor Immune Response by Cancer Metabolism. Cells 2019; 8:cells8020104. [PMID: 30708988 PMCID: PMC6406288 DOI: 10.3390/cells8020104] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
The metabolic reprogramming of tumor cells and immune escape are two major hallmarks of cancer cells. The metabolic changes that occur during tumorigenesis, enabling survival and proliferation, are described for both solid and hematological malignancies. Concurrently, tumor cells have deployed mechanisms to escape immune cell recognition and destruction. Additionally, therapeutic blocking of tumor-mediated immunosuppression has proven to have an unprecedented positive impact in clinical oncology. Increased evidence suggests that cancer metabolism not only plays a crucial role in cancer signaling for sustaining tumorigenesis and survival, but also has wider implications in the regulation of antitumor immune signaling through both the release of signaling molecules and the expression of immune membrane ligands. Here, we review these molecular events to highlight the contribution of cancer cell metabolic reprogramming on the shaping of the antitumor immune response.
Collapse
Affiliation(s)
- Charlotte Domblides
- Bordeaux University, CNRS, UMR 5164, ImmunoConcEpT, 33000 Bordeaux, France.
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, 33000 Bordeaux, France.
| | - Lydia Lartigue
- Curematch, Inc., 6440 Lusk Bvld, San Diego, CA 92121, USA.
| | - Benjamin Faustin
- Bordeaux University, CNRS, UMR 5164, ImmunoConcEpT, 33000 Bordeaux, France.
- Cellomet, CGFB, 146 Rue léo Saignat, F-33000 Bordeaux, France.
| |
Collapse
|
12
|
Fox E, Oliver T, Rowe M, Thomas S, Zakharia Y, Gilman PB, Muller AJ, Prendergast GC. Indoximod: An Immunometabolic Adjuvant That Empowers T Cell Activity in Cancer. Front Oncol 2018; 8:370. [PMID: 30254983 PMCID: PMC6141803 DOI: 10.3389/fonc.2018.00370] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/21/2018] [Indexed: 11/20/2022] Open
Abstract
Exploding interest in immunometabolism as a source of new cancer therapeutics has been driven in large part by studies of tryptophan catabolism mediated by IDO/TDO enzymes. A chief focus in the field is IDO1, a pro-inflammatory modifier that is widely overexpressed in cancers where it blunts immunosurveillance and enables neovascularization and metastasis. The simple racemic compound 1-methyl-D,L-tryptophan (1MT) is an extensively used probe of IDO/TDO pathways that exerts a variety of complex inhibitory effects. The L isomer of 1MT is a weak substrate for IDO1 and is ascribed the weak inhibitory activity of the racemate on the enzyme. In contrast, the D isomer neither binds nor inhibits the purified IDO1 enzyme. However, clinical development focused on D-1MT (now termed indoximod) due to preclinical cues of its greater anticancer activity and its distinct mechanisms of action. In contrast to direct enzymatic inhibitors of IDO1, indoximod acts downstream of IDO1 to stimulate mTORC1, a convergent effector signaling molecule for all IDO/TDO enzymes, thus possibly lowering risks of drug resistance by IDO1 bypass. In this review, we survey the unique biological and mechanistic features of indoximod as an IDO/TDO pathway inhibitor, including recent clinical findings of its ability to safely enhance various types of cancer therapy, including chemotherapy, chemo-radiotherapy, vaccines, and immune checkpoint therapy. We also review the potential advantages indoximod offers compared to selective IDO1-specific blockade, which preclinical studies and the clinical study ECHO-301 suggest may be bypassed readily by tumors. Indoximod lies at a leading edge of broad-spectrum immunometabolic agents that may act to improve responses to many anticancer modalities, in a manner analogous to vaccine adjuvants that act to boost immunity in settings of infectious disease.
Collapse
Affiliation(s)
- Eric Fox
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Thomas Oliver
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Melissa Rowe
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Yousef Zakharia
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Paul B. Gilman
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Alexander J. Muller
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Brown ZJ, Yu SJ, Heinrich B, Ma C, Fu Q, Sandhu M, Agdashian D, Zhang Q, Korangy F, Greten TF. Indoleamine 2,3-dioxygenase provides adaptive resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunol Immunother 2018; 67:1305-1315. [PMID: 29959458 DOI: 10.1007/s00262-018-2190-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. Immune checkpoint blockade with anti-CTLA-4 and anti-PD-1 antibodies has shown promising results in the treatment of patients with advanced HCC. The anti-PD-1 antibody, nivolumab, is now approved for patients who have had progressive disease on the current standard of care. However, a subset of patients with advanced HCC treated with immune checkpoint inhibitors failed to respond to therapy. Here, we provide evidence of adaptive resistance to immune checkpoint inhibitors through upregulation of indoleamine 2,3-dioxygenase (IDO) in HCC. Anti-CTLA-4 treatment promoted an induction of IDO1 in resistant HCC tumors but not in tumors sensitive to immune checkpoint blockade. Using both subcutaneous and hepatic orthotopic models, we found that the addition of an IDO inhibitor increases the efficacy of treatment in HCC resistant tumors with high IDO induction. Furthermore, in vivo neutralizing studies demonstrated that the IDO induction by immune checkpoint blockade was dependent on IFN-γ. Similar findings were observed with anti-PD-1 therapy. These results provide evidence that IDO may play a role in adaptive resistance to immune checkpoint inhibitors in patients with HCC. Therefore, inhibiting IDO in combination with immune checkpoint inhibitors may add therapeutic benefit in tumors which overexpress IDO and should be considered for clinical evaluation in HCC.
Collapse
Affiliation(s)
- Zachary J Brown
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Su Jong Yu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Bernd Heinrich
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Chi Ma
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Qiong Fu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Milan Sandhu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - David Agdashian
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Qianfei Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Firouzeh Korangy
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA
| | - Tim F Greten
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD, 20892, USA. .,National Cancer Institute, Center for Cancer Research, Liver Cancer Program, Bethesda, USA.
| |
Collapse
|
14
|
Counihan JL, Grossman EA, Nomura DK. Cancer Metabolism: Current Understanding and Therapies. Chem Rev 2018; 118:6893-6923. [DOI: 10.1021/acs.chemrev.7b00775] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jessica L. Counihan
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Elizabeth A. Grossman
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Daniel K. Nomura
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
15
|
Rosenberg AJ, Wainwright DA, Rademaker A, Galvez C, Genet M, Zhai L, Lauing KL, Mulcahy MF, Hayes JP, Odell DD, Horbinski C, Komanduri S, Tetreault MP, Kim KYA, Villaflor VM. Indoleamine 2,3-dioxygenase 1 and overall survival of patients diagnosed with esophageal cancer. Oncotarget 2018; 9:23482-23493. [PMID: 29805749 PMCID: PMC5955099 DOI: 10.18632/oncotarget.25235] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase 1 (IDO1) is an enzyme with immunomodulatory properties that has emerged as a potential immunotherapeutic target in human cancer. However, the role, expression pattern, and relevance of IDO1 in esophageal cancer (EC) are poorly understood. Here, we utilize gene expression analysis of the cancer genome atlas (TCGA) and immunohistochemistry (IHC) to better understand the role and prognostic significance of IDO1 in EC. RESULTS High IDO1 mRNA levels were associated with worse overall survival (OS) in both esophageal squamous cell carcinoma (SCC) (P = 0.02) and adenocarcinoma (AC) (P = 0.036). High co-expression of IDO1 and programmed death ligand 1 (PD-L1) was associated with worse OS in SCC (P = 0.0031) and AC (P = 0.0186). IHC for IDO1 in SCC showed a significant correlation with PD-L1 (P < 0.0001) and CD3ε (P < 0.0001). CONCLUSIONS EC with high IDO1 and PD-L1 expression is significantly correlated with decreased patient survival, and may correlate with increased T-cells. These data suggest that simultaneous inhibition of IDO1 and PD-(L)1 may overcome important barriers to T-cell mediated immune rejection of EC. MATERIALS AND METHODS mRNA expression data from TCGA (SCC N = 87; AC N = 97). IHC in a second cohort of EC (N = 93) were stained for IDO1, PD-L1, and CD3ε, followed by light microscopic analysis.
Collapse
Affiliation(s)
- Ari J. Rosenberg
- Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
- Division of Hematology and Oncology, Northwestern University, Chicago, 60611 IL, USA
| | - Derek A. Wainwright
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Alfred Rademaker
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Preventive Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Carlos Galvez
- Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Matthew Genet
- Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Lijie Zhai
- Department of Neurological Surgery, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Kristen L. Lauing
- Department of Neurological Surgery, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Mary F. Mulcahy
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
- Division of Hematology and Oncology, Northwestern University, Chicago, 60611 IL, USA
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, 60611 IL, USA
| | - John P. Hayes
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Radiation Oncology, Northwestern University, Chicago, 60611 IL, USA
| | - David D. Odell
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Thoracic Surgery, Northwestern University, Chicago, 60611 IL, USA
| | - Craig Horbinski
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Pathology, Northwestern University, Chicago, 60611 IL, USA
| | - Srinadh Komanduri
- Department of Gastroenterology, Northwestern University, Chicago, 60611 IL, USA
| | | | - Kwang-Youn A. Kim
- Department of Preventive Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
| | - Victoria M. Villaflor
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611 IL, USA
- Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, 60611 IL, USA
- Division of Hematology and Oncology, Northwestern University, Chicago, 60611 IL, USA
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, 60611 IL, USA
| |
Collapse
|
16
|
Schafer CC, Wang Y, Hough KP, Sawant A, Grant SC, Thannickal VJ, Zmijewski J, Ponnazhagan S, Deshane JS. Indoleamine 2,3-dioxygenase regulates anti-tumor immunity in lung cancer by metabolic reprogramming of immune cells in the tumor microenvironment. Oncotarget 2018; 7:75407-75424. [PMID: 27705910 PMCID: PMC5340181 DOI: 10.18632/oncotarget.12249] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) has been implicated in immune evasion by tumors. Upregulation of this tryptophan (Trp)-catabolizing enzyme, in tumor cells and myeloid-derived suppressor cells (MDSCs) within the tumor microenvironment (TME), leads to Trp depletion that impairs cytotoxic T cell responses and survival; however, exact mechanisms remain incompletely understood. We previously reported that a combination therapy of gemcitabine and a superoxide dismutase mimetic promotes anti-tumor immunity in a mouse model of lung cancer by inhibiting MDSCs, enhancing polyfunctional response of CD8+ memory T cells, and extending survival. Here, we show that combination therapy targets IDO signaling, specifically in MDSCs, tumor cells, and CD8+ T cells infiltrating the TME. Deficiency of IDO caused significant reduction in tumor burden, tumor-infiltrating MDSCs, GM-CSF, MDSC survival and infiltration of programmed death receptor-1 (PD-1)-expressing CD8+ T cells compared to controls. IDO−/− MDSCs downregulated nutrient-sensing AMP-activated protein kinase (AMPK) activity, but IDO−/− CD8+ T cells showed AMPK activation associated with enhanced effector function. Our studies provide proof-of-concept for the efficacy of this combination therapy in inhibiting IDO and T cell exhaustion in a syngeneic model of lung cancer and provide mechanistic insights for IDO-dependent metabolic reprogramming of MDSCs that reduces T cell exhaustion and regulates anti-tumor immunity.
Collapse
Affiliation(s)
- Cara C Schafer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yong Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth P Hough
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anandi Sawant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefan C Grant
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor J Thannickal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jaroslaw Zmijewski
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Jessy S Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
17
|
Zhao Q, Wang PP, Huang ZL, Peng L, Lin C, Gao Z, Su S. Tumoral indoleamine 2, 3-dioxygenase 1 is regulated by monocytes and T lymphocytes collaboration in hepatocellular carcinoma. Oncotarget 2018; 7:14781-90. [PMID: 26895379 PMCID: PMC4924751 DOI: 10.18632/oncotarget.7438] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 01/30/2016] [Indexed: 12/12/2022] Open
Abstract
Indoleamine 2, 3-Dioxygenase 1 (IDO1) in cancer cells plays a critical role in tumor immunosuppression. However, the precise mechanisms regulating tumoral IDO1 expression in tumor milieus remain unclear. Here, we reported that IDO1 expression in tumor cells of hepatocelluar carcinomas (HCC), displayed a discrete rather than uniform pattern. In vitro culture, human hepatoma cell lines did not constitutively express IDO1. Interestingly, co-culture with peripheral blood mononuclear cells (PBMC) significantly induced and maintained IDO1 expression in these tumor cells, predominantly through IFN-γ. Mechanistically, we showed that IDO1 expression in tumor cells was only induced when co-cultured with both T lymphocytes and monocytes. Moreover, the cooperation between T lymphocytes and monocytes played an indispensable role on the tumoral IDO1 expression in immunocompromised mice. Taken together, our data supported the notion that IDO1 expression in tumor cells might serve as a counter-regulatory mechanism regulated by immune system, and provided new insights into the collaborative action of different inflammatory cells in tumor immunosuppression.
Collapse
Affiliation(s)
- Qiyi Zhao
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Gene Engineering of The Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Pei-Pei Wang
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhan-Lian Huang
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoshuang Lin
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhiliang Gao
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Liver Disease, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
18
|
Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ. Discovery of IDO1 Inhibitors: From Bench to Bedside. Cancer Res 2018; 77:6795-6811. [PMID: 29247038 DOI: 10.1158/0008-5472.can-17-2285] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/23/2017] [Accepted: 11/01/2017] [Indexed: 01/11/2023]
Abstract
Small-molecule inhibitors of indoleamine 2,3-dioxygenase-1 (IDO1) are emerging at the vanguard of experimental agents in oncology. Here, pioneers of this new drug class provide a bench-to-bedside review on preclinical validation of IDO1 as a cancer therapeutic target and on the discovery and development of a set of mechanistically distinct compounds, indoximod, epacadostat, and navoximod, that were first to be evaluated as IDO inhibitors in clinical trials. As immunometabolic adjuvants to widen therapeutic windows, IDO inhibitors may leverage not only immuno-oncology modalities but also chemotherapy and radiotherapy as standards of care in the oncology clinic. Cancer Res; 77(24); 6795-811. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - James B DuHadaway
- Lankenau Institute for Medical Research (LIMR), Wynnewood, Pennsylvania
| | | |
Collapse
|
19
|
Inflammatory Reprogramming with IDO1 Inhibitors: Turning Immunologically Unresponsive 'Cold' Tumors 'Hot'. Trends Cancer 2017; 4:38-58. [PMID: 29413421 DOI: 10.1016/j.trecan.2017.11.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 01/24/2023]
Abstract
We discuss how small-molecule inhibitors of the tryptophan (Trp) catabolic enzyme indoleamine 2,3-dioxygenase (IDO) represent a vanguard of new immunometabolic adjuvants to safely enhance the efficacy of cancer immunotherapy, radiotherapy, or 'immunogenic' chemotherapy by leveraging responses to tumor neoantigens. IDO inhibitors re-program inflammatory processes to help clear tumors by blunting tumor neovascularization and restoring immunosurveillance. Studies of regulatory and effector pathways illuminate IDO as an inflammatory modifier. Recent work suggests that coordinate targeting of the Trp catabolic enzymes tryptophan 2,3-dioxygenase (TDO) and IDO2 may also safely broaden efficacy. Understanding IDO inhibitors as adjuvants to turn immunologically 'cold' tumors 'hot' can seed new concepts in how to improve the efficacy of cancer therapy while limiting collateral damage.
Collapse
|
20
|
Tajmul M, Parween F, Singh L, Mathur SR, Sharma JB, Kumar S, Sharma DN, Yadav S. Identification and validation of salivary proteomic signatures for non-invasive detection of ovarian cancer. Int J Biol Macromol 2017; 108:503-514. [PMID: 29222021 DOI: 10.1016/j.ijbiomac.2017.12.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/20/2017] [Accepted: 12/04/2017] [Indexed: 12/31/2022]
Abstract
Ovarian cancer (OC) is one of the most lethal cancers among all gynecological malignancies. An effective and non-invasive screening approach is needed urgently to reduce high mortality rate. The purpose of this study was to identify the salivary protein signatures (SPS) for non-invasive detection of ovarian cancer. Differentially expressed SPS were identified by fluorescence-based 2D-DIGE coupled with MALDI/TOF-MS. The expression levels of three differential proteins (Lipocalin-2, indoleamine-2, 3-dioxygenase1 (IDO1) and S100A8) were validated using western blotting and ELISA. Immunohistochemistry and qRT-PCR were performed in an independent cohort of ovarian tumor tissues. 25 over expressed and 19 under expressed (p<0.05) proteins between healthy controls and cancer patients were identified. Lipocalin-2, IDO1 and S100A8 were selected for initial verification and successfully verified by immunoassay. Diagnostic potential of the candidate biomarkers was evaluated by ROC analysis. The selected biomarkers were further validated by immunohistochemistry in an independent cohort of ovarian tissues. The global expression of selected targets was also analyzed by microarray and validated using qRT-PCR to strengthen our hypothesis. Tumor secreted proteins identified by 'dual-omics' strategy, whose concentration are significantly high in ovarian cancer patients have obvious potential to be used as screening biomarker after large scale validation.
Collapse
Affiliation(s)
- Md Tajmul
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Farhat Parween
- Hybridoma Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Lata Singh
- Department of Ocular Pathology, Dr. R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - J B Sharma
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sunesh Kumar
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - D N Sharma
- Department of Radiotherapy, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Savita Yadav
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
21
|
Li F, Wei L, Li S, Liu J. Indoleamine-2,3-dioxygenase and Interleukin-6 associated with tumor response to neoadjuvant chemotherapy in breast cancer. Oncotarget 2017; 8:107844-107858. [PMID: 29296206 PMCID: PMC5746108 DOI: 10.18632/oncotarget.22253] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Purpose Indoleamine-2,3-dioxygenase (IDO) and Interleukin-6 (IL-6) contribute to poor therapeutic effects, tumor relapse and aggressive tumor growth. IDO and IL-6 incorporate a positive feedback signal loop to maintain IDO and IL-6 constitutive expression and facilitate tumor progression. Results IDO expression was associated with IL-6 expression and plasma IL-6 level (P<0.05). Concentrating on clinicopathological features prior neoadjuvant chemotherapy, both IDO expression and plasma IL-6 level were associated with clinical T stage and N stage (P<0.05). IL-6 expression was associated with clinical T stage (P=0.016). The co-expression of IDO/IL-6 was correlated with clinical T, N stage and estrogen receptor (ER) status (P<0.05). IDO, IL-6 expression, clinical T stage, pathological T stage, ER status and Luminal type were correlated with clinical response to neoadjuvant chemotherapy (P<0.05). Multivariate analysis showed that IDO expression were correlated with clinical response to neoadjuvant chemotherapy (P=0.034). IL-6 expression and pathological T stage were correlated with pCR (P<0.05). In the multivariate analysis, postoperative pathological T stage associated with pCR (P=0.041). In the prognostic analysis, only clinical T stage was significant correlated with overall survival (P=0.003). Materials and Methods 46 breast cancer patients received neoadjuvant chemotherapy enrolled in this study. Immunohistochemistry was applied for evaluating IDO and IL-6 expression in biopsy tissues prior neoadjuvant chemotherapy. Immunofluorescence was applied to observe the co-localization of IDO and IL-6. Serum IL-6 level was examined via ELISA. The associations between IDO, IL-6, Serum IL-6 level and clinicopathological features, response to neoadjuvant chemotherapy were analyzed. Conclusion IDO and IL-6 expression associated with advanced breast cancer and poor response to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Fangxuan Li
- Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Lijuan Wei
- Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Shixia Li
- Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Juntian Liu
- Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
22
|
Zhai L, Ladomersky E, Lauing KL, Wu M, Genet M, Gritsina G, Győrffy B, Brastianos PK, Binder DC, Sosman JA, Giles FJ, James CD, Horbinski C, Stupp R, Wainwright DA. Infiltrating T Cells Increase IDO1 Expression in Glioblastoma and Contribute to Decreased Patient Survival. Clin Cancer Res 2017; 23:6650-6660. [PMID: 28751450 DOI: 10.1158/1078-0432.ccr-17-0120] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/28/2017] [Accepted: 07/24/2017] [Indexed: 12/15/2022]
Abstract
Purpose: Indoleamine 2,3 dioxygenase 1 (IDO1) mediates potent immunosuppression in multiple preclinical models of cancer. However, the basis for elevated IDO1 expression in human cancer, including the most common primary malignant brain tumor in adults, glioblastoma (GBM), is poorly understood. The major objective of this study is to address this gap in our understanding of how IDO1 expression contributes to the biology of GBM, and whether its level of expression is a determinant of GBM patient outcome.Experimental Design: Patient-resected GBM, The Cancer Genome Atlas, human T-cell:GBM cocultures, as well as nu/nu, NOD-scid, and humanized (NSG-SGM3-BLT) mice-engrafted human GBM form the basis of our investigation.Results:In situ hybridization for IDO1 revealed transcript expression throughout patient-resected GBM, whereas immunohistochemical IDO1 positivity was highly variable. Multivariate statistical analysis revealed that higher levels of IDO1 transcript predict a poor patient prognosis (P = 0.0076). GBM IDO1 mRNA levels positively correlated with increased gene expression for markers of cytolytic and regulatory T cells, in addition to decreased patient survival. Humanized mice intracranially engrafted human GBM revealed an IFNγ-associated T-cell-mediated increase of intratumoral IDO1Conclusions: Our data demonstrate that high intratumoral IDO1 mRNA levels correlate with a poor GBM patient prognosis. It also confirms the positive correlation between increased GBM IDO1 levels and human-infiltrating T cells. Collectively, this study suggests that future efforts aimed at increasing T-cell-mediated effects against GBM should consider combinatorial approaches that coinhibit potential T-cell-mediated IDO1 enhancement during therapy. Clin Cancer Res; 23(21); 6650-60. ©2017 AACR.
Collapse
Affiliation(s)
- Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Matthew Genet
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Galina Gritsina
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary.,2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Priscilla K Brastianos
- Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Divisions of Hematology/Oncology and Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - David C Binder
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jeffrey A Sosman
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Francis J Giles
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Charles D James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois. .,Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
23
|
Yue EW, Sparks R, Polam P, Modi D, Douty B, Wayland B, Glass B, Takvorian A, Glenn J, Zhu W, Bower M, Liu X, Leffet L, Wang Q, Bowman KJ, Hansbury MJ, Wei M, Li Y, Wynn R, Burn TC, Koblish HK, Fridman JS, Emm T, Scherle PA, Metcalf B, Combs AP. INCB24360 (Epacadostat), a Highly Potent and Selective Indoleamine-2,3-dioxygenase 1 (IDO1) Inhibitor for Immuno-oncology. ACS Med Chem Lett 2017; 8:486-491. [PMID: 28523098 PMCID: PMC5430407 DOI: 10.1021/acsmedchemlett.6b00391] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 02/24/2017] [Indexed: 01/25/2023] Open
Abstract
A data-centric medicinal chemistry approach led to the invention of a potent and selective IDO1 inhibitor 4f, INCB24360 (epacadostat). The molecular structure of INCB24360 contains several previously unknown or underutilized functional groups in drug substances, including a hydroxyamidine, furazan, bromide, and sulfamide. These moieties taken together in a single structure afford a compound that falls outside of "drug-like" space. Nevertheless, the in vitro ADME data is consistent with the good cell permeability and oral bioavailability observed in all species (rat, dog, monkey) tested. The extensive intramolecular hydrogen bonding observed in the small molecule crystal structure of 4f is believed to significantly contribute to the observed permeability and PK. Epacadostat in combination with anti-PD1 mAb pembrolizumab is currently being studied in a phase 3 clinical trial in patients with unresectable or metastatic melanoma.
Collapse
Affiliation(s)
- Eddy W. Yue
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Richard Sparks
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Padmaja Polam
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Dilip Modi
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Brent Douty
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Brian Wayland
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Brian Glass
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Amy Takvorian
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Joseph Glenn
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Wenyu Zhu
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Michael Bower
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Xiangdong Liu
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Lynn Leffet
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Qian Wang
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Kevin J. Bowman
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Michael J. Hansbury
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Min Wei
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Yanlong Li
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Richard Wynn
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Timothy C. Burn
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Holly K. Koblish
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Jordan S. Fridman
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Tom Emm
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Peggy A. Scherle
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Brian Metcalf
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Andrew P. Combs
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| |
Collapse
|
24
|
Thomas S, Izard J, Walsh E, Batich K, Chongsathidkiet P, Clarke G, Sela DA, Muller AJ, Mullin JM, Albert K, Gilligan JP, DiGuilio K, Dilbarova R, Alexander W, Prendergast GC. The Host Microbiome Regulates and Maintains Human Health: A Primer and Perspective for Non-Microbiologists. Cancer Res 2017; 77:1783-1812. [PMID: 28292977 PMCID: PMC5392374 DOI: 10.1158/0008-5472.can-16-2929] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Abstract
Humans consider themselves discrete autonomous organisms, but recent research is rapidly strengthening the appreciation that associated microorganisms make essential contributions to human health and well being. Each person is inhabited and also surrounded by his/her own signature microbial cloud. A low diversity of microorganisms is associated with a plethora of diseases, including allergy, diabetes, obesity, arthritis, inflammatory bowel diseases, and even neuropsychiatric disorders. Thus, an interaction of microorganisms with the host immune system is required for a healthy body. Exposure to microorganisms from the moment we are born and appropriate microbiome assembly during childhood are essential for establishing an active immune system necessary to prevent disease later in life. Exposure to microorganisms educates the immune system, induces adaptive immunity, and initiates memory B and T cells that are essential to combat various pathogens. The correct microbial-based education of immune cells may be critical in preventing the development of autoimmune diseases and cancer. This review provides a broad overview of the importance of the host microbiome and accumulating knowledge of how it regulates and maintains a healthy human system. Cancer Res; 77(8); 1783-812. ©2017 AACR.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.
| | - Jacques Izard
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Emily Walsh
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Kristen Batich
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Surgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Pakawat Chongsathidkiet
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Surgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, APC Microbiome Institute University College Cork, Cork, Ireland
| | - David A Sela
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts
- Center for Microbiome Research, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - James M Mullin
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Korin Albert
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
| | - John P Gilligan
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | - Rima Dilbarova
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Walker Alexander
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | |
Collapse
|
25
|
Li F, Zhang R, Li S, Liu J. IDO1: An important immunotherapy target in cancer treatment. Int Immunopharmacol 2017; 47:70-77. [PMID: 28365507 DOI: 10.1016/j.intimp.2017.03.024] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 01/01/2023]
Abstract
Indoleamine 2,3-dioxigenase 1 (IDO1) acts in pathogenic inflammatory processes and engender immune tolerance to tumor antigens. IDO1 can decrease the tryptophan and produce a series of toxic kynurenine metabolites to promote the immune toleration via GCN2 pathway, mTOR pathway, toxic effect of kynurenine and favoring differentiation of Tregs. IDO1 can be induced in most human cells, especially APCs and cancer cells through canonical and non-canonical NF-κB and Jak/STAT pathways, as well as PKC and TGF-β signaling pathways. A series of human cancers over-express IDO1 in a constitutive way. Thus, IDO1 is likely to be an attractive target for developing inhibitors of tumor treatments. Many preclinical and clinical trials have been underway and suggest that IDO1 inhibitor maybe an effective tool against a wide range of cancers. However, the IDO1 inhibitor alone had been verified that to be disappointment in achieving effective antitumor efficacy. Concentrating on its molecular mechanism in immune toleration and complex environments of cancer, IDO1 inhibitor could cooperate with chemotherapies and other immune target inhibitors to lessen the tumor.
Collapse
Affiliation(s)
- Fangxuan Li
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, China
| | - Rupeng Zhang
- Department of Gastric Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, China.
| | - Shixia Li
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, China
| | - Juntian Liu
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, China.
| |
Collapse
|
26
|
Qian S, He T, Wang W, He Y, Zhang M, Yang L, Li G, Wang Z. Discovery and preliminary structure–activity relationship of 1H-indazoles with promising indoleamine-2,3-dioxygenase 1 (IDO1) inhibition properties. Bioorg Med Chem 2016; 24:6194-6205. [DOI: 10.1016/j.bmc.2016.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 11/25/2022]
|
27
|
Kranz LM, Birtel M, Hilscher L, Grunwitz C, Petschenka J, Vascotto F, Vormehr M, Voss RH, Kreiter S, Diken M. CIMT 2016: Mechanisms of efficacy in cancer immunotherapy - Report on the 14th Annual Meeting of the Association for Cancer Immunotherapy May 10-12 2016, Mainz, Germany. Hum Vaccin Immunother 2016; 12:2805-2812. [PMID: 27435168 PMCID: PMC5137546 DOI: 10.1080/21645515.2016.1206677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Lena M Kranz
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany.,b Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| | - Matthias Birtel
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany.,b Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| | - Lina Hilscher
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | - Christian Grunwitz
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany.,c BioNTech RNA Pharmaceuticals GmbH , Mainz , Germany
| | - Jutta Petschenka
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | - Fulvia Vascotto
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | - Mathias Vormehr
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany.,c BioNTech RNA Pharmaceuticals GmbH , Mainz , Germany
| | - Ralf-Holger Voss
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | - Sebastian Kreiter
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | - Mustafa Diken
- a TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| |
Collapse
|
28
|
Qian S, Zhang M, Chen Q, He Y, Wang W, Wang Z. IDO as a drug target for cancer immunotherapy: recent developments in IDO inhibitors discovery. RSC Adv 2016. [DOI: 10.1039/c5ra25046c] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This review highlights the recent advances in research related to the role of IDO in immune escape in cancer and novel small-molecule IDO inhibitors with an emphasis on their chemical structures and modes of action.
Collapse
Affiliation(s)
- Shan Qian
- Department of Pharmaceutical Engineering
- Xihua University
- Chengdu 610039
- P. R. China
| | - Man Zhang
- Department of Pharmaceutical Engineering
- Xihua University
- Chengdu 610039
- P. R. China
| | - Quanlong Chen
- Department of Pharmaceutical Engineering
- Xihua University
- Chengdu 610039
- P. R. China
| | - Yanying He
- Department of Pharmaceutical Engineering
- Xihua University
- Chengdu 610039
- P. R. China
| | - Wei Wang
- Department of Pharmaceutical Engineering
- Xihua University
- Chengdu 610039
- P. R. China
| | - Zhouyu Wang
- Department of Chemistry
- Xihua University
- Chengdu 610039
- P. R. China
| |
Collapse
|
29
|
Zhai L, Spranger S, Binder DC, Gritsina G, Lauing KL, Giles FJ, Wainwright DA. Molecular Pathways: Targeting IDO1 and Other Tryptophan Dioxygenases for Cancer Immunotherapy. Clin Cancer Res 2015; 21:5427-33. [PMID: 26519060 PMCID: PMC4681601 DOI: 10.1158/1078-0432.ccr-15-0420] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/15/2015] [Indexed: 01/12/2023]
Abstract
Indoleamine 2, 3-dioxygenase 1 (IDO1), IDO2, and tryptophan 2, 3-dioxygenase (TDO) comprise a family of enzymes that catalyze the first- and rate-limiting step associated with the catabolic conversion of tryptophan (Trp) into kynurenine (Kyn). Through subsequent enzymatic and spontaneous reactions, Kyn is further converted into the energetic substrates, NAD(+) and ATP, to fuel cellular metabolic functions. Coincidently, the depletion of Trp and accumulation of Kyn has been demonstrated to induce effector T-cell apoptosis/dysfunction and immunosuppressive regulatory T-cell induction, respectively. Similar to other immune checkpoints, IDO1 and TDO are suggested to be important targets for immunotherapeutic intervention. This is represented by the recent growth of efforts to inhibit the Trp-to-Kyn pathway as a means to control immunosuppression. Inhibitors currently in clinical trials, INCB024360, GDC-0919, indoximod, and an IDO1 peptide-based vaccine, are being evaluated for their efficacy against a wide range of cancers including melanoma, glioblastoma, non-small cell lung, pancreatic, and/or breast cancer, as well as metastatic disease. Despite the rapid development of potent clinical grade inhibitors, strategic questions remain. Here, we review the state of the literature with respect to current therapeutic inhibitors of tryptophan catabolism, evaluation of those efforts preclinically and clinically, compensatory changes that occur with therapeutic targeting, as well as newly recognized signaling features that raise critical questions to the field. Given the rapidly evolving interest in determining how IDO1/TDO, and to an unknown extent, IDO2, can be targeted for increasing cancer immunotherapeutic efficacy, we present a brief but comprehensive analysis that addresses critical questions, while highlighting the mechanics that remain to be explored.
Collapse
Affiliation(s)
- Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stefani Spranger
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - David C Binder
- Department of Pathology, The University of Chicago, Chicago, Illinois. Committee on Cancer Biology, The University of Chicago, Chicago, Illinois
| | - Galina Gritsina
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Francis J Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, Chicago, Illinois. Division of Hematology and Oncology, Northwestern University, Chicago, Illinois. Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
30
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
31
|
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 2015; 4:e981457. [PMID: 25949879 DOI: 10.4161/2162402x.2014.981457] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/23/2014] [Indexed: 01/13/2023] Open
Abstract
The expression of the immunomodulating enzyme indoleamine-2,3-dioxygenase (IDO) suppresses T-lymphocyte function, thus correlating with poor survival in a variety of cancer patients. IDO degrades the essential amino acid tryptophan leading to immunosuppressive kynurenines production. In the present study, concentrations of tryptophan, 3-hydroxykynurenine, and kynurenine were measured in pre-treatment serum samples of 251 cervical cancer patients by a mass-spectrometric method (XLC-MS/MS) and IDO activity determined by the kynurenine/tryptophan (Kyn/Trp) ratio. A low concentration of tryptophan was found to be significantly associated with tumors greater than 4 cm and lymph node metastatic spread. Furthermore, significant positive correlations were found between high concentrations of the tryptophan metabolites kynurenine and 3-hydroxykynurenine and advanced disease stage (FIGO >IIA) and lymph node metastases. High levels of kynurenine were further associated with parametrial invasion and tumor size. A high Kyn/Trp ratio was related to lymph node metastasis, FIGO stage, tumor size, parametrial invasion and poor disease-specific survival. These results suggest that IDO activation is linked to poor clinicopathological parameters and worse survival in cervical cancer, warranting the use of IDO inhibitors in future clinical trials.
Collapse
Key Words
- FIGO, International Federation of Gynaecologists and Obstetricians
- Gy, Gray
- HPV, human papillomavirus
- IDO
- IDO, indoleamine-2,3-dioxygenase
- IFNγ, interferon γ
- Kyn/Trp ratio
- Kyn/Trp ratio, kynurenine/tryptophan ratio
- M0, no metastasis
- NK, natural killer
- SCC, squamous cell carcinoma
- TDO. tryptophan-2,3-dioxygenase
- TLR: toll-like receptor
- Tregs, regulatory T cells
- XLC-MS/MS- extraction: liquid chromatographic tandem mass spectrometry
- cervical cancer
- kynurenine
- tryptophan
Collapse
Affiliation(s)
- Debbie M Ferns
- Centre for Gynaecological Oncology Amsterdam; Free University Medical Centre ; Amsterdam, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine; University Medical Centre Groningen ; The Netherlands
| | - Marrije R Buist
- Centre for Gynaecological Oncology Amsterdam; Academic Medical Centre ; Amsterdam, The Netherlands
| | - Hans W Nijman
- Department of Gynaecological Oncology; University Medical Centre Groningen ; The Netherlands
| | - Gemma G Kenter
- Centre for Gynaecological Oncology Amsterdam; Free University Medical Centre ; Amsterdam, The Netherlands ; Centre for Gynaecological Oncology Amsterdam; Academic Medical Centre ; Amsterdam, The Netherlands
| | - Ekaterina S Jordanova
- Centre for Gynaecological Oncology Amsterdam; Free University Medical Centre ; Amsterdam, The Netherlands
| |
Collapse
|
32
|
Larsen SK, Ahmad SM, Idorn M, Met Ö, Martinenaite E, Svane IM, Straten PT, Andersen MH. Spontaneous presence of FOXO3-specific T cells in cancer patients. Oncoimmunology 2014; 3:e953411. [PMID: 25960934 DOI: 10.4161/21624011.2014.953411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/02/2014] [Indexed: 02/07/2023] Open
Abstract
In the present study, we describe forkhead box O3 (FOXO3)-specific, cytotoxic CD8+ T cells existent among peripheral-blood mononuclear cells (PBMCs) of cancer patients. FOXO3 immunogenicity appears specific, as we did not detect reactivity toward FOXO3 among T cells in healthy individuals. FOXO3 may naturally serve as a target antigen for tumor-reactive T cells as it is frequently over-expressed in cancer cells. In addition, expression of FOXO3 plays a critical role in immunosuppression mediated by tumor-associated dendritic cells (TADCs). Indeed, FOXO3-specific cytotoxic T lymphocytes (CTLs) were able to specifically recognize and kill both FOXO3-expressing cancer cells as well as dendritic cells. Thus, FOXO3 was processed and presented by HLA-A2 on the cell surface of both immune cells and cancer cells. As FOXO3 programs TADCs to become tolerogenic, FOXO3 signaling thereby comprises a significant immunosuppressive mechanism, such that FOXO3 targeting by means of specific T cells is an attractive clinical therapy to boost anticancer immunity. In addition, the natural occurrence of FOXO3-specific CTLs in the periphery suggests that these T cells hold a function in the complex network of immune regulation in cancer patients.
Collapse
Key Words
- APC, antigen presenting cell
- CTL
- CTL, cytotoxic T lymphocyte
- CTLA4, cytotoxic T-lymphocyte associated protein 4
- DC, dendritic cell
- FOXO3
- FOXO3, forkhead box O3
- IDO, indoleamine-2,3-dioxygenase
- PBMC, peripheral blood mononuclear cell
- TADC, tumor-associated DCs
- TGFβ, tumor growth factor β
- TNFα, tumor necrosis factor α
- Tregs, regulatory T cell
- antigens
- immune regulation
- tumor-associated dendritic cells
Collapse
Affiliation(s)
- Stine Kiaer Larsen
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark ; These authors contributed equally to this work
| | - Shamaila Munir Ahmad
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark ; These authors contributed equally to this work
| | - Manja Idorn
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Özcan Met
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Evelina Martinenaite
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Per Thor Straten
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| |
Collapse
|
33
|
Watcharanurak K, Zang L, Nishikawa M, Yoshinaga K, Yamamoto Y, Takahashi Y, Ando M, Saito K, Watanabe Y, Takakura Y. Effects of upregulated indoleamine 2, 3-dioxygenase 1 by interferon γ gene transfer on interferon γ-mediated antitumor activity. Gene Ther 2014; 21:794-801. [PMID: 24919418 DOI: 10.1038/gt.2014.54] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 02/07/2023]
Abstract
Interferon γ (IFN-γ), an anticancer agent, is a strong inducer of indoleamine 2,3-dioxygenase 1 (IDO1), which is a tryptophan-metabolizing enzyme involved in the induction of tumor immune tolerance. In this study, we investigated the IDO1 expression in organs after IFN-γ gene transfer to mice. IFN-γ gene transfer greatly increased the mRNA expression of IDO1 in many tissues with the highest in the liver. This upregulation was associated with reduced L-tryptophan levels and increased L-kynurenine levels in serum, indicating that IFN-γ gene transfer increased the IDO activity. Then, Lewis lung carcinoma (LLC) tumor-bearing wild-type and IDO1-knockout (IDO1 KO) mice were used to investigate the effects of IDO1 on the antitumor activity of IFN-γ. IFN-γ gene transfer significantly retarded the tumor growth in both strains without any significant difference in tumor size between the two groups. By contrast, the IDO1 activity was increased only in the wild-type mice by IFN-γ gene transfer, suggesting that cells other than LLC cells, such as tumor stromal cells, are the major contributors of IDO1 expression in LLC tumor. Taken together, these results imply that IFN-γ gene transfer mediated IDO1 upregulation in cells other than LLC cells has hardly any effect on the antitumor activity of IFN-γ.
Collapse
Affiliation(s)
- K Watcharanurak
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - L Zang
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - M Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - K Yoshinaga
- Department of Human Health Science, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Y Yamamoto
- Department of Human Health Science, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Y Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - M Ando
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - K Saito
- Department of Human Health Science, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Y Watanabe
- Department of Molecular Microbiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Y Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Prendergast GC, Smith C, Thomas S, Mandik-Nayak L, Laury-Kleintop L, Metz R, Muller AJ. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother 2014; 63:721-35. [PMID: 24711084 DOI: 10.1007/s00262-014-1549-4] [Citation(s) in RCA: 393] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/26/2014] [Indexed: 12/15/2022]
Abstract
Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-β signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.
Collapse
Affiliation(s)
- George C Prendergast
- Lankenau Institute for Medical Research (LIMR), 100 Lancaster Avenue, Wynnewood, PA, 19096, USA,
| | | | | | | | | | | | | |
Collapse
|
35
|
Becker JC, thor Straten P, Andersen MH. Self-reactive T cells: suppressing the suppressors. Cancer Immunol Immunother 2014; 63:313-9. [PMID: 24368340 PMCID: PMC11029163 DOI: 10.1007/s00262-013-1512-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/11/2013] [Indexed: 01/22/2023]
Abstract
The immune system is a tightly regulated and complex system. An important part of this immune regulation is the assurance of tolerance toward self-antigens to maintain immune homeostasis. However, in recent years, antigen-specific cellular immune responses toward several normal self-proteins expressed in regulatory immune cells have been reported, especially in patients with cancer. The seemingly lack of tolerance toward such proteins is interesting, as it suggests a regulatory function of self-reactive T (srT) cells, which may be important for the fine tuning of the immune system. In particular, surprising has been the description of cytotoxic srT cells that are able to eliminate normal regulatory immune cells. Such srT cells may be important as effector cells that suppress regulatory suppressor cells. The current knowledge of the nature and function of srT cells is still limited. Still, the therapeutic targeting of srT cells offers a novel approach to harness immune-regulatory networks in cancer.
Collapse
Affiliation(s)
- Jürgen C. Becker
- Department of General Dermatology, Medical University of Graz, Graz, Austria
| | - Per thor Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| |
Collapse
|
36
|
Stonik VA, Fedorov SN. Marine low molecular weight natural products as potential cancer preventive compounds. Mar Drugs 2014; 12:636-71. [PMID: 24473167 PMCID: PMC3944507 DOI: 10.3390/md12020636] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 12/17/2022] Open
Abstract
Due to taxonomic positions and special living environments, marine organisms produce secondary metabolites that possess unique structures and biological activities. This review is devoted to recently isolated and/or earlier described marine compounds with potential or established cancer preventive activities, their biological sources, molecular mechanisms of their action, and their associations with human health and nutrition. The review covers literature published in 2003–2013 years and focuses on findings of the last 2 years.
Collapse
Affiliation(s)
- Valentin A Stonik
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, Vladivostok 690950, Russia.
| | - Sergey N Fedorov
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, Vladivostok 690950, Russia.
| |
Collapse
|
37
|
Metz R, Smith C, DuHadaway JB, Chandler P, Baban B, Merlo LMF, Pigott E, Keough MP, Rust S, Mellor AL, Mandik-Nayak L, Muller AJ, Prendergast GC. IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation. Int Immunol 2014; 26:357-67. [PMID: 24402311 DOI: 10.1093/intimm/dxt073] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IDO2 is implicated in tryptophan catabolism and immunity but its physiological functions are not well established. Here we report the characterization of mice genetically deficient in IDO2, which develop normally but exhibit defects in IDO-mediated T-cell regulation and inflammatory responses. Construction of this strain was prompted in part by our discovery that IDO2 function is attenuated in macrophages from Ido1 (-/-) mice due to altered message splicing, generating a functional mosaic with implications for interpreting findings in Ido1 (-/-) mice. No apparent defects were observed in Ido2 (-/-) mice in embryonic development or hematopoietic differentiation, with wild-type profiles documented for kynurenine in blood serum and for immune cells in spleen, lymph nodes, peritoneum, thymus and bone marrow of naive mice. In contrast, upon immune stimulation we determined that IDO1-dependent T regulatory cell generation was defective in Ido2 (-/-) mice, supporting Ido1-Ido2 genetic interaction and establishing a functional role for Ido2 in immune modulation. Pathophysiologically, both Ido1 (-/-) and Ido2 (-/-) mice displayed reduced skin contact hypersensitivity responses, but mechanistic distinctions were apparent, with only Ido2 deficiency associated with a suppression of immune regulatory cytokines that included GM-CSF, G-CSF, IFN-γ, TNF-α, IL-6 and MCP-1/CCL2. Different contributions to inflammation were likewise indicated by the finding that Ido2 (-/-) mice did not phenocopy Ido1 (-/-) mice in the reduced susceptibility of the latter to inflammatory skin cancer. Taken together, our results offer an initial glimpse into immune modulation by IDO2, revealing its genetic interaction with IDO1 and distinguishing its non-redundant contributions to inflammation.
Collapse
Affiliation(s)
- Richard Metz
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Courtney Smith
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | | | - Phillip Chandler
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Babak Baban
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Lauren M F Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Elizabeth Pigott
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Martin P Keough
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Sonja Rust
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Andrew L Mellor
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Laura Mandik-Nayak
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexander J Muller
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA Department of Pathology, Anatomy and Cell Biology, Jefferson Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
38
|
Diken M, Attig S, Grunwitz C, Kranz L, Simon P, van de Roemer N, Vascotto F, Kreiter S. CIMT 2013: advancing targeted therapies--report on the 11th Annual Meeting of the Association for Cancer Immunotherapy, May 14-16 2013, Mainz, Germany. Hum Vaccin Immunother 2013; 9:2025-32. [PMID: 23877042 PMCID: PMC3906376 DOI: 10.4161/hv.25768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The 11th Annual Meeting of Association for Cancer Immunotherapy (CIMT) welcomed more than 700 scientists around the world to Mainz, Germany and continued to be the largest immunotherapy meeting in Europe. Renowned speakers from various fields of cancer immunotherapy gave lectures under CIMT2013’s tag: “Advancing targeted therapies” the highlights of which are summarized in this meeting report.
Collapse
Affiliation(s)
- Mustafa Diken
- TRON-Translational Oncology at the University Medical Center of Johannes Gutenberg University; Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu XQ, Lu K, Feng LL, Ding M, Gao JM, Ge XL, Wang X. Up-regulated expression of indoleamine 2,3-dioxygenase 1 in non-Hodgkin lymphoma correlates with increased regulatory T-cell infiltration. Leuk Lymphoma 2013; 55:405-14. [PMID: 23682557 DOI: 10.3109/10428194.2013.804917] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1), which is a key enzyme in tryptophan metabolism expressed in some subsets of normal and neoplastic cells, participates in tumor-induced tolerance. However, the mechanisms involved are not clearly understood. A hypothesis suggests that IDO1 may be involved in proliferation and conversion of regulatory T cells (Tregs). In this study, we evaluated the levels of IDO1 and forkhead box P3 (FoxP3) in non-Hodgkin lymphoma (NHL) tissues and performed ex vivo experiments to investigate the role of IDO1 on T-cell tolerance in NHL. The results showed that expressions of IDO1 mRNA and protein were coincidentally higher in NHL tissues than in reactive hyperplasia of lymph node tissues. Up-regulation of IDO1 was correlated with later clinical phases, larger tumors and higher serum lactate dehydrogenase (LDH), and indicated a worse prognosis. FoxP3 mRNA and protein levels were markedly increased alongside elevated IDO1 levels. Co-culture of murine CD4 + CD25- T cells with A20 cells could initiate the conversion of CD4 + CD25+ T cells, which showed a suppressive function in the mixed lymphocyte reaction. Moreover, the potent inhibitor of IDO1, 1-methyl-l-tryptophan, attenuated the conversion of CD4 + CD25- T cells into CD4 + CD25+ FoxP3 + T cells. The results suggested that up-regulation of IDO1 in NHL tissues could induce local immune tolerance by favoring development and infiltration of FoxP3 + Tregs through the conversion of CD4 + CD25- T cells into CD4 + CD25+ FoxP3 + T cells in the tumor microenvironment. This could be a novel mechanism of NHL escape from immune control.
Collapse
Affiliation(s)
- Xiao-Qian Liu
- Department of Hematology, Provincial Hospital Affiliated to Shandong University , Jinan, Shandong , P. R. China
| | | | | | | | | | | | | |
Collapse
|
40
|
Sprung CN, Yang Y, Forrester HB, Li J, Zaitseva M, Cann L, Restall T, Anderson RL, Crosbie JC, Rogers PAW. Genome-wide transcription responses to synchrotron microbeam radiotherapy. Radiat Res 2012; 178:249-59. [PMID: 22974124 DOI: 10.1667/rr2885.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The majority of cancer patients achieve benefit from radiotherapy. A significant limitation of radiotherapy is its relatively low therapeutic index, defined as the maximum radiation dose that causes acceptable normal tissue damage to the minimum dose required to achieve tumor control. Recently, a new radiotherapy modality using synchrotron-generated X-ray microbeam radiotherapy has been demonstrated in animal models to ablate tumors with concurrent sparing of normal tissue. Very little work has been undertaken into the cellular and molecular mechanisms that differentiate microbeam radiotherapy from broad beam. The purpose of this study was to investigate and compare the whole genome transcriptional response of in vivo microbeam radiotherapy versus broad beam irradiated tumors. We hypothesized that gene expression changes after microbeam radiotherapy are different from those seen after broad beam. We found that in EMT6.5 tumors at 4-48 h postirradiation, microbeam radiotherapy differentially regulates a number of genes, including major histocompatibility complex (MHC) class II antigen gene family members, and other immunity-related genes including Ciita, Ifng, Cxcl1, Cxcl9, Indo and Ubd when compared to broad beam. Our findings demonstrate molecular differences in the tumor response to microbeam versus broad beam irradiation and these differences provide insight into the underlying mechanisms of microbeam radiotherapy and broad beam.
Collapse
Affiliation(s)
- Carl N Sprung
- Centre for Innate Immunity and Infectious Disease, Monash Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Smith C, Chang MY, Parker K, Beury D, DuHadaway JB, Flick HE, Boulden J, Sutanto-Ward E, Soler AP, Laury-Kleintop LD, Mandik-Nayak L, Metz R, Ostrand-Rosenberg S, Prendergast GC, Muller AJ. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov 2012; 2:722-35. [PMID: 22822050 PMCID: PMC3677576 DOI: 10.1158/2159-8290.cd-12-0014] [Citation(s) in RCA: 261] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
UNLABELLED Indoleamine 2,3-dioxygenase (IDO) enzyme inhibitors have entered clinical trials for cancer treatment based on preclinical studies, indicating that they can defeat immune escape and broadly enhance other therapeutic modalities. However, clear genetic evidence of the impact of IDO on tumorigenesis in physiologic models of primary or metastatic disease is lacking. Investigating the impact of Ido1 gene disruption in mouse models of oncogenic KRAS-induced lung carcinoma and breast carcinoma-derived pulmonary metastasis, we have found that IDO deficiency resulted in reduced lung tumor burden and improved survival in both models. Micro-computed tomographic (CT) imaging further revealed that the density of the underlying pulmonary blood vessels was significantly reduced in Ido1-nullizygous mice. During lung tumor and metastasis outgrowth, interleukin (IL)-6 induction was greatly attenuated in conjunction with the loss of IDO. Biologically, this resulted in a consequential impairment of protumorigenic myeloid-derived suppressor cells (MDSC), as restoration of IL-6 recovered both MDSC suppressor function and metastasis susceptibility in Ido1-nullizygous mice. Together, our findings define IDO as a prototypical integrative modifier that bridges inflammation, vascularization, and immune escape to license primary and metastatic tumor outgrowth. SIGNIFICANCE This study provides preclinical, genetic proof-of-concept that the immunoregulatory enzyme IDO contributes to autochthonous carcinoma progression and to the creation of a metastatic niche. IDO deficiency in vivo negatively impacted both vascularization and IL-6–dependent, MDSC-driven immune escape, establishing IDO as an overarching factor directing the establishment of a protumorigenic environment.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/enzymology
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Adenocarcinoma of Lung
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Disease Progression
- Genes, ras
- HL-60 Cells
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Inflammation/drug therapy
- Inflammation/enzymology
- Interleukin-6/biosynthesis
- Kaplan-Meier Estimate
- Lung Neoplasms/blood supply
- Lung Neoplasms/enzymology
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Mammary Neoplasms, Experimental/enzymology
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Metastasis
- Neovascularization, Pathologic/enzymology
- Survival Analysis
- U937 Cells
Collapse
Affiliation(s)
- Courtney Smith
- Lankenau Institute for Medical Research, Wynnewood PA USA
| | | | - Katherine Parker
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore MD USA
| | - Daniel Beury
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore MD USA
| | - James B. DuHadaway
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore MD USA
| | - Hollie E. Flick
- Lankenau Institute for Medical Research, Wynnewood PA USA
- Department of Biochemistry, Drexel University College of Medicine, Philadelphia PA USA
| | | | | | - Alejandro Peralta Soler
- Lankenau Institute for Medical Research, Wynnewood PA USA
- Richfield Laboratory of Dermatopathology, Cincinnati OH USA
| | | | | | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood PA USA
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia PA USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA USA
| | - Alexander J. Muller
- Lankenau Institute for Medical Research, Wynnewood PA USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA USA
| |
Collapse
|
42
|
Chang MY, Smith C, DuHadaway JB, Pyle JR, Boulden J, Soler AP, Muller AJ, Laury-Kleintop LD, Prendergast GC. Cardiac and gastrointestinal liabilities caused by deficiency in the immune modulatory enzyme indoleamine 2,3-dioxygenase. Cancer Biol Ther 2011; 12:1050-8. [PMID: 22157149 DOI: 10.4161/cbt.12.12.18142] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) modifies adaptive immunity, in part by determining the character of inflammatory responses in the tissue microenvironment. Small molecule inhibitors of IDO are being developed to treat cancer, chronic infections and other diseases, so the systemic effects of IDO disruption on inflammatory phenomena may influence the design and conduct of early phase clinical investigations of this new class of therapeutic agents. Here, we report cardiac and gastrointestinal phenotypes observed in IDO deficient mice that warrant consideration in planned assessments of the safety risks involved in clinical development of IDO inhibitors. Calcification of the cardiac endometrium proximal to the right ventricle was a sexually dimorphic strain-specific phenotype with ~30% penetrance in BALB/c mice lacking IDO. Administration of complete Freund's adjuvant containing Toll-like receptor ligands known to induce IDO caused acute pancreatitis in IDO deficient mice, with implications for the design of planned combination studies of IDO inhibitors with cancer vaccines. In an established model of hyperlipidemia, IDO deficiency caused a dramatic elevation in levels of serum triglycerides. In the large intestine, IDO loss only slightly increased sensitivity to induction of acute colitis, but it markedly elevated tumor incidence, multiplicity and staging during inflammatory colon carcinogenesis. Together, our findings suggest potential cardiac and gastrointestinal risks of IDO inhibitors that should be monitored in patients as this new class of drugs enter early clinical development.
Collapse
|
43
|
Involvement of indoleamine 2,3-dioxygenase in impairing tumor-infiltrating CD8 T-cell functions in esophageal squamous cell carcinoma. Clin Dev Immunol 2011; 2011:384726. [PMID: 22013481 PMCID: PMC3195535 DOI: 10.1155/2011/384726] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 11/30/2022]
Abstract
The indoleamine 2,3-dioxygenase-(IDO-) mediated microenvironment plays an important role in tumor immune escape. However, the inhibitory effects of IDO on the CD8+ tumour-infiltrating lymphocytes (CD8+ TILs) in esophageal squamous cell carcinoma (ESCC) have not been clarified yet. Here, we found that the level of IDO expression in ESCC tumor specimens correlated with a reduction in the number of CD8+ TILs. Patients with high IDO expression and a low number of CD8+ TILs had significantly impaired overall survival time. IDO expression and functional enzyme activity in ESCC cell lines could be induced by IFNγ. When exposed to the milieu generated by IDO-expressing Eca109 cells, the CD8+ TILs were suppressed in proliferation, and their cytolytic functions against target tumor cells were lost. These results suggested that impairing CD8+ TIL functions by IDO expressed in ESCC possibly contributed to the finding that patients with higher IDO expression have more aggressive disease progression and shorter overall survival time.
Collapse
|
44
|
von Bubnoff D, Wilms H, Scheler M, Brenk M, Koch S, Bieber T. Human myeloid dendritic cells are refractory to tryptophan metabolites. Hum Immunol 2011; 72:791-7. [DOI: 10.1016/j.humimm.2011.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 05/24/2011] [Accepted: 05/25/2011] [Indexed: 12/18/2022]
|