Tryptophan catabolism is used by tumors to resist immune attack. It can be catalyzed by indoleamine 2,3-dioxygenase (IDO1) and tryptophan 2,3-dioxygenase (TDO). IDO1 is frequently expressed in tumors and has been widely studied as a potential therapeutic target to reduce resistance to cancer immunotherapy. In contrast, TDO expression in tumors is not well characterized. Several human tumor cell lines constitutively express enzymatically active TDO. In human tumor samples, TDO expression has previously been detected by transcriptomics, but the lack of validated antibodies has precluded detection of the TDO protein and identification of TDO-expressing cells. Here, we developed novel TDO-specific monoclonal antibodies and confirmed by immunohistochemistry the expression of TDO in the majority of human cancers. In all hepatocarcinomas (10/10), TDO was expressed by most tumor cells. Some glioblastomas (10/39) and kidney carcinomas (1/10) also expressed TDO in tumor cells themselves but only in focal tumor areas. In addition, all cancers tested contained foci of nontumoral TDO-expressing cells, which were identified as pericytes by their expression of PDGFRβ and their location in vascular structures. These TDO-expressing pericytes belonged to morphologically abnormal tumor vessels and were found in high-grade tumors in the vicinity of necrotic or hemorrhagic areas, which were characterized by neoangiogenesis. We observed similar TDO-expressing pericytes in inflammatory pulmonary lesions containing granulation tissue, and in chorionic villi, two tissue types that also feature neoangiogenesis. Our results confirm TDO as a relevant immunotherapeutic target in hepatocellular carcinoma and suggest a proangiogenic role of TDO in other cancer types.

See article by Schramme et al., p. 32

Tryptophan catabolism is one of the immunosuppressive mechanisms acting in the tumor microenvironment (1–3). Two distinct enzymes can convert tryptophan into kynurenine: indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO protein encoded by gene TDO2; refs. 4, 5). Although both enzymes are located in the cytosol, their activity induces tryptophan depletion and kynurenine accumulation in the extracellular space, because of the action of amino-acid transporters (6–10). The concerted effects of tryptophan depletion and kynurenine accumulation make the tumor microenvironment strongly immunosuppressive, reducing the proliferation of effector T lymphocytes and favoring the differentiation of regulatory T cells (11–14).

In steady state, the expression of IDO1 is restricted to endothelial cells in the placenta and lung, to mature dendritic cells in secondary lymphoid organs and to scattered epithelial cells in the female genital tract (15). However, in inflammatory conditions, IDO1 expression is strongly induced by interferon gamma (IFNγ; refs. 16, 17). It is therefore highly expressed in inflammatory sites, where it appears to contribute to the negative feedback of the immune response that prevents immunopathology (15). Placental IDO1 expression prevents immune rejection of the fetus by the maternal immune system (18). IDO1 is also expressed in many cancers and contributes to their resistance to immune rejection (1, 15). Inflamed tumors can express IDO1 as an adaptive resistance mechanism, in which IDO1 is induced by the IFNγ produced by tumor-infiltrating T cells (19). However, a number of human tumors express IDO1 constitutively, as an intrinsic resistance mechanism that prevents T-cell infiltration and may account for a number of cold, poorly immune cell–infiltrated, tumors (20). Such constitutive IDO1 expression, which depends on oncogenic signaling through the MAPK and/or PI3K pathways, is exerted via COX2 inducing an autocrine loop of prostaglandin secretion, and can be repressed by clinically available COX2 inhibitors, thereby promoting tumor rejection (20). IDO1 activity can also be blocked by small-molecule inhibitors, several of which are currently in clinical development (21). The most advanced IDO1 inhibitor, epacadostat, recently completed a phase III clinical trial testing the combination of epacadostat with anti–PD-1, pembrolizumab, in advanced melanoma (22). This trial showed no clinical benefit of the combination compared with anti–PD-1 alone. The potential reasons for this failure are numerous, but include a suboptimal dosing regimen and a lack of inclusion of tumors that constitutively express IDO1 in the trial. Another potential reason is the involvement of additional actors mediating tryptophan catabolism in the tumor microenvironment (23, 24).

The other enzyme able to degrade tryptophan into kynurenine is TDO, which is expressed at a high level in the liver and degrades excess dietary tryptophan to maintain stable levels of systemic tryptophan (25, 26). Expression of TDO is regulated in part by glucocorticoids (27), whereas TDO is posttranslationally stabilized by its substrate (28, 29). TDO expression is also detected in the decidualized endometrium, where its function is unknown (30–32), and in the brain where it might contribute to the synthesis of neuroactive compounds (33). TDO2 mRNA is also found in a number of human tumor samples, including hepatocarcinomas, glioblastomas, melanomas, and bladder carcinomas (12, 34). Expression of TDO in mouse tumors can prevent their immune rejection, an effect reverted by systemic treatment of mice with a small-molecule inhibitor of TDO (34, 35). These results prompted the preclinical and early clinical development of TDO inhibitors (26, 34–36). TDO is constitutively expressed in a number of human tumor cell lines, including glioblastoma, colorectal, head and neck, lung and gall bladder carcinoma cell lines (34). In these lines, TDO protein expression and activity was confirmed by enzymatic assays (34). In human tumor samples, TDO2 expression is detected by RT-qPCR (12, 34), but the lack of a reliable TDO-specific antibody prevented confirmation of protein expression as well as identification of the cell type(s) expressing TDO. These uncertainties have hampered clinical development of TDO inhibitors. In this report, using novel TDO-specific monoclonal antibodies (mAb), we confirm TDO protein expression in many human tumor samples, and identify the TDO-expressing cells in these samples.

Mouse experiments

DBA/2 mice (Harlan Laboratories), C57BL/6J Ola Hsd mice (Envigo), and C57BL/6 TDO-KO mice (provided by Dr Hiroshi Funakoshi and Dr Toshikazu Nakamura from Osaka University; ref. 25) were bred at the animal facility of the Ludwig Institute for Cancer Research, Brussels, Belgium. C57BL/6 and C57BL/6 TDO-KO mice were mated to obtain heterozygous offspring, which were intercrossed to obtain Tdo2+/−, Tdo2+/+ (WT), and Tdo2−/− (TDO-KO). Homozygous TDO-KO and WT mice littermates were selected by PCR on genomic DNA using primers: mTdo2-forward: GTA-TCT-ATG-GAG-GAC-AAT-GAA-G, mTdo2-reverse: GAT-GAA-TAG-GTG-CTC-GTC-ATG; neomycin-resistance-gene-forward: GTT-CTT-TTT-GTC-AAG-ACC-GA, neomycin-resistance-gene-reverse: TTT-CCA-CCA-TGA- TAT-TCG-GC. Mice were used at 8 to 12 weeks of age. Animal studies were conducted in accordance with national and institutional guidelines for animal care and with the approval of the Comité d'Ethique pour l'Expérimentation Animale from the Secteur des Sciences de la Santé, UCLouvain (2011/UCL/MD/015 and 2015/UCL/MD/015).

Production of TDO-specific mouse mAbs

We created a pcDNA3 plasmid construct (Thermo Fisher Scientific) encoding a fusion protein comprising human TDO at the C-terminal end of the type II transmembrane human CD134 ligand (hCD134L) protein (Supplementary Fig. S1A). We immunized DBA/2 mice with this plasmid three times at 2-week intervals by electrotransfer of 20 μg DNA into tibialis muscles (10 μg into each muscle) using the pulse generator Clinivet (IGEA). Electrotransfer comprised eight pulses of 200 V/cm amplitude lasting 20 ms followed by a pause of 480 ms. Two weeks after the last immunization (i.e., on day 42), we boosted mice by injecting in the footpad 105 living P1.HTR cells stably transfected with pCD1 hCD134L hTDO expression vector, which were created as described (ref. 37; Supplementary Fig. S1B). The boost was repeated on days 56, 70, and 115 using 105, 106, and 90 × 106 cells, respectively. On day 119, the mouse with the best TDO antibody response was sacrificed, and lymphocytes from the spleen and lymph nodes (inguinal and mesenteric) were hybridized with cells from fusion partner SP2neo using standard procedures (polyethylene glycol or electrofusion). Hybridomas were selected in HAT medium, screened for production of TDO antibodies by ELISA, and confirmed by FACS.

Cancer cell lines

P1.HTR cells were derived in our lab from the P815 mastocytoma cell line (38). HEK-293-EBNA (293E) cells were purchased from InvivoGen. 293E cells transfected with human TDO cl119 were previously created (34), and confirmed to still express enzymatically active TDO (Supplementary Fig. S2A). HEK-293T cells were a generous gift from Prof. Kris Thielemans in 2006. A172 (glioblastoma) cells were purchased from Antisense Pharma. Huh-7 (liver cancer) cells were a gift from A. Patel (Oxford). TDO-expressing LB159-CRCA cells were derived in 1991 at the Ludwig Institute for Cancer Research Brussels Branch from a colorectal carcinoma patient sample from the Cliniques universitaires Saint-Luc, Brussels (34). They were cultured with homemade ACL-4 supplement. LB159-CRCA TDO−/− cells were generated using CRISPR/Cas9 as described below. SK-CO-11 (colon cancer) and MZ-CHA-3 (gall bladder cancer) were received from collaborators Alex Knuth and Thomas Wölfel in Mainz in 1991. LB1317-SCCHN (head and neck cancer) cells were derived at the Ludwig Institute Brussels Branch from a patient sample received from Marc Hamoir and Vincent Grégoire (Cliniques universitaires Saint-Luc, Brussels) in 1996. U-87 MG (glioblastoma) cells were purchased from Cell Lines Services GmBH in 2011. All cell lines were tested for Mycoplasma in October 2019. Human cell lines were authenticated in November 2019 by short tandem repeat profiling (Promega Powerplex hs 16). To prepare formalin-fixed, paraffin-embedded (FFPE) cell pellets, cells were fixed for 20 minutes with 4% formaldehyde, washed with PBS, and resuspended in 3% low-melting point agarose-PBS. The agarose blocks were embedded in paraffin using the Vacuum Infiltration Processor (Tissue-Tek). Briefly, they were dehydrated using ethanol-water baths with increasing ethanol concentrations, incubated in xylene, and finally embedded in paraffin.

Generation of LB159-CRCA TDO−/− cells

LB159-CRCA TDO−/− cells were generated using CRISPR/Cas9 as follows. Two different pairs of guide RNA (GR1-forward: CACCGTTTAAAAAACTCCCCGTAGA, GR1-reverse: AAACTCTACGGGGAGTTTTTTAAAC; GR2-forward: CACCGGCAAAGGAGGTCTTATCTAT, GR2-reverse: AAACATAGATAAGACCTCCTTTGCC) were cloned into lentiCRISPR v2 plasmid (Addgene). Target guide sequence cloning was performed as follows: Digestion and dephosphorylation of lentiviral v2 CRISPR plasmid were performed with BsmBI (NEB, #R0580S) and SAP (NEB, #M0371S) for 30 minutes at 37°C, followed by inactivation for 15 minutes at 65°C. Gel purification of the digested plasmid was performed using QIAquick Gel Extraction (Qiagen 28706). Phosphorylation and annealing of each pair of oligonucleotides (GR1 and GR2) were performed using T4 Ligase (NEB, #M0202S) and the T4 Ligation Buffer containing ATP (NEB, #B0202S) with a cycle of 37°C for 30 minutes, 95°C for 5 minutes, and cooling to 25°C at 5°C/minute. Ligation was performed using the Quick Ligation protocol (NEB, #M2200S) according to the manufacturer's instructions. XL-1 blue competent cells (Agilent 200249) were transformed, and the plasmids were purified with NucleoSpin Plasmid (Macherey Nagel). Sequencing of selected plasmid clones was performed prior to generating the lentiviruses. Lentiviruses were produced in HEK-293T cells seeded at 2 × 106 cells/plate (standard petri dish). Twenty-four hours after plating, the cells were transfected with 5 μg/mL lentiviral plasmid and a second-generation lentivirus packaging system. Plasmids were diluted in FBS-free DMEM with Turbofect reagent (Thermo Fisher, #R0531) and incubated at room temperature for 15 to 20 minutes. The plasmid/turbofect mix was then added to the cells in a drop-wise fashion, and the plate was gently rocked to achieve even distribution of the plasmids. Six hours after transfection, the medium was exchanged with fresh medium. The supernatant of the cells was collected 24 and 48 hours after the transfection, filtered using a 0.45-μm filter and stored at −80°C. LB159-CRCA cells were infected with both lentiviruses. LB159-CRCA cells were seeded at 1 × 106 cells/well in 6-well plates with 4 mL of medium. Twenty-four hours after seeding, the medium was removed, and 1 mL of lentivirus-GR1 or -GR2 supernatant collected 48 hours after transfection was added. Cells were incubated for 4 hours prior to adding 1 mL of fresh culture medium. Cells were cultured for 72 hours prior to selection with puromycin (6 μg/mL). The knockout of TDO was validated in clone 1 by DNA sequencing and by enzymatic assays (Supplementary Fig. S2A).

Human tissues

Normal tissues and tumors were obtained from surgical resections or autopsies from 171 patients. The tissues used in this publication were collected, fixed in formaldehyde, embedded in paraffin and provided by (i) the BioLibrary of the Institut Roi Albert II of the Cliniques universitaires Saint-Luc, Brussels, Belgium, projects CDCUCLR27 and CDCUCLR16-2016; (ii) the Service d'Anatomie Pathologique of the CHU-Brugmann, Brussels, Belgium; (iii) BHUL—Université de Liège—CHU de Liège, Belgium. The study was approved by the Commission d'Ethique Biomédicale Hospitalo-Facultaire from the UCLouvain (B403201316588) and conducted under the guidelines of the Helsinki declaration. Patients gave informed consent for the use of their tissues in this study.

Immunohistochemistry and immunofluorescence multiplex stainings

Five μm-thick paraffin sections were deparaffinized in 3 baths of Histo-Clear (3 minutes each), washed in butanol for 3 minutes and progressively rehydrated in 100%, 90%, 70%, 50% ethanol and in demineralized water (3 minutes each). Antigen retrieval was performed with Tris/EDTA buffer at pH 9 in a 2100 Antigen Retriever (Aptum) using a predefined heating cycle. All the following steps were performed at room temperature. Endogenous peroxidases were blocked with Peroxidase Block (Dako) for 15 minutes and protein blocking was done for 1 hour with TBS-Tween containing 2% milk, 5% bovine serum albumin (biotin-free BSA), and 1% human IgG. The primary antibodies (Table 1) were diluted in immunohistochemistry (IHC) diluent (Enzo) and incubated for 1 hour. After washing, the secondary antibodies (Table 1) were incubated for 1 hour. For IHC, the staining was revealed with HIGHDEF DAB substrate (Enzo) or with AEC+ Substrate Chromogen (Dako) between 5 and 20 minutes, and counterstaining was performed with hematoxylin. Slides were incubated for 5 minutes in hematoxylin, washed with demineralized water, washed in tap water for 5 minutes and transferred again to demineralized water. Slides were mounted with HIGHDEF IHC mount (Enzo). For multiplex immunofluorescence (IF), the staining was revealed with the Tyramide Signal Amplification system (Table 1). Tyramide reagent was used at 1:50 in borate buffer. The antibodies were then eluted with citrate buffer at pH 6 using microwave treatment at 900 W until boiling followed by 15 minutes at 90 W. The following staining was started with protein blocking and performed as described above. After the last staining, microwave treatment was performed, the nuclei were counterstained for 5 minutes with Hoechst 33342 (Invitrogen) used at 20 μg/mL in TBS-Tween 0.1% with 10% BSA, and the slides were mounted with HIGHDEF IHC fluoromount (Enzo). The slides were digitalized using a Pannoramic 250 Flash III tissue scanner (3DHISTECH) at ×20 magnification. Multiplex-immunostained entire tumor sections were evaluated quantitatively using the image analysis tool Oncotopix version 2017.2 (Visiopharm).

Table 1.

IHC antibodies.

Primary antibodies
SpeciesAntigenCloneReferenceDilution
Rabbit CA9 Polyclonal NovusBio (#NB100-417) 1:1,000 
Rabbit CD3 SP7 Abcam (#ab16669) 1:500 
Rabbit CD8 SP16 Thermo Fisher Scientific (#MA5-14548) 1:200 
Rabbit CD34 EP373Y GeneTex (#GTX61737) 1:1,000 
Mouse CD68 PG-M1 Thermo Fisher Scientific (#MA5-12407) 1:50 
Mouse Cytokeratin AE1/AE3 Dako (#M3515) 1:50 
Rat GFAP 2.2B10 Invitrogen (#13-0300) 1:50 
Mouse HLA-DR LN-3 Novocastra (#NCL-LN3) 1:100 
Mouse IDO1 4.16H1 Theate et al. 2015 (15) 0.5 μg/mL 
Rabbit Ki67 SP6 Thermo Fisher Scientific (#RM9106) 1:200 
Rabbit PDGFRβ Y92 Abcam (#ab32570) 1:200 
Rabbit αSMA EPR5368 Merck (#MABT381) 1:2,000 
Mouse TDO III iTeos Therapeutics 0.5 μg/mL 
Mouse TDO iTeos Therapeutics 0.5 μg/mL 
Rabbit β3-tubulin TUJ1 Cell Signaling (#5666) 1:100 
Mouse IgG2a, κ isotype ctrl MOPC173 BioLegend (#400209) 0.5 μg/mL 
Secondary antibodies 
EnVision+ HRP goat anti-mouse Dako (#K4001) — 
EnVision+ HRP goat anti-rabbit Dako (#K4003) — 
ImmPRESS HRP goat anti-rat Vector (#MP-7444) — 
Tyramide reagents 
Alexa Fluor Used to stain Reference Dilution 
488 CA9, CD3, HLA-DR, PDGFRβ, αSMA Thermo Fisher Scientific (#B40953) 1:50 
555 CD8, CD34, β3-tubulin Thermo Fisher Scientific (#B40955) 1:50 
594 CD68, GFAP, Ki67, cytokeratin Thermo Fisher Scientific (#B40957) 1:50 
647 TDO Thermo Fisher Scientific (#B40958) 1:50 
Primary antibodies
SpeciesAntigenCloneReferenceDilution
Rabbit CA9 Polyclonal NovusBio (#NB100-417) 1:1,000 
Rabbit CD3 SP7 Abcam (#ab16669) 1:500 
Rabbit CD8 SP16 Thermo Fisher Scientific (#MA5-14548) 1:200 
Rabbit CD34 EP373Y GeneTex (#GTX61737) 1:1,000 
Mouse CD68 PG-M1 Thermo Fisher Scientific (#MA5-12407) 1:50 
Mouse Cytokeratin AE1/AE3 Dako (#M3515) 1:50 
Rat GFAP 2.2B10 Invitrogen (#13-0300) 1:50 
Mouse HLA-DR LN-3 Novocastra (#NCL-LN3) 1:100 
Mouse IDO1 4.16H1 Theate et al. 2015 (15) 0.5 μg/mL 
Rabbit Ki67 SP6 Thermo Fisher Scientific (#RM9106) 1:200 
Rabbit PDGFRβ Y92 Abcam (#ab32570) 1:200 
Rabbit αSMA EPR5368 Merck (#MABT381) 1:2,000 
Mouse TDO III iTeos Therapeutics 0.5 μg/mL 
Mouse TDO iTeos Therapeutics 0.5 μg/mL 
Rabbit β3-tubulin TUJ1 Cell Signaling (#5666) 1:100 
Mouse IgG2a, κ isotype ctrl MOPC173 BioLegend (#400209) 0.5 μg/mL 
Secondary antibodies 
EnVision+ HRP goat anti-mouse Dako (#K4001) — 
EnVision+ HRP goat anti-rabbit Dako (#K4003) — 
ImmPRESS HRP goat anti-rat Vector (#MP-7444) — 
Tyramide reagents 
Alexa Fluor Used to stain Reference Dilution 
488 CA9, CD3, HLA-DR, PDGFRβ, αSMA Thermo Fisher Scientific (#B40953) 1:50 
555 CD8, CD34, β3-tubulin Thermo Fisher Scientific (#B40955) 1:50 
594 CD68, GFAP, Ki67, cytokeratin Thermo Fisher Scientific (#B40957) 1:50 
647 TDO Thermo Fisher Scientific (#B40958) 1:50 

In situ hybridization

Five μm-thick paraffin sections were deparaffinized and progressively rehydrated as described for IHC and IF. Endogenous peroxidases were blocked with RNAscope H2O2 (ACD) for 10 minutes at room temperature (RT). Antigen retrieval was performed with RNAscope Target Retrieval Reagents (ACD) for 15 minutes in a steam cooker. The slides were immediately transferred to water at RT, washed with ethanol and dried for several minutes up to overnight at RT. The slides were incubated with RNAscope Protease Plus (ACD) for 30 minutes at 40°C in a Dako Hybridizer. After washing with RNAscope Wash Buffer Reagents (ACD), the slides were incubated for 2 hours at 40°C with the primary probes: RNAscope Probe Hs-TDO2 (ACD, #416471), RNAscope Negative Control Probe dapB (ACD, #310043), RNAscope Hs-POLR2A (positive control, ACD, #310451). After extensive washing, the slides underwent incubation with 6 successive amplification probes from the revelation kit RNAscope 2.5 HD Detection Reagent-BROWN (ACD, #322310): AMP1 (30 minutes, 40°C), AMP2 (15 minutes, 40°C), AMP3 (30 minutes, 40°C), AMP4 (15 minutes, 40°C), AMP5 (30 minutes, RT), AMP6 (15 minutes, RT). The staining was revealed with DAB from the same kit for 10 minutes at RT. Counterstaining was done with hematoxylin, and the slides were mounted with HIGHDEF IHC mount (Enzo).

Quantitative RT-PCR

RNA of frozen cells and tissues was extracted with NucleoSpin RNA (Macherey Nagel) according to the manufacturer's instructions. Whole tissues were beforehand crushed in the lysis buffer of the kit using a TissueLyser LT (Qiagen). RNA of FFPE tissue sections was extracted with NucleoSpin total RNA FFPE XS (Macherey Nagel) according to the manufacturer's instructions. RNA was quantified with a NanoDrop spectrophotometer and a defined amount of RNA was retrotranscribed by the RevertAid RT Kit (Thermo Fisher Scientific). TaqMan qPCR was performed with Takyon ROX Probe 2X MasterMix dTTP blue (Eurogentec) in a StepOnePlus thermal cycler (Applied Biosystems) using the following program for human IDO1, human TDO2, and murine Tdo2: 3 minutes at 95°C, then 40 cycles of 10 seconds at 95°C and 1 minute at 60°C; for human EF1 and murine β-actin: 3 minutes at 95°C, then 40 cycles of 3 seconds at 95°C and 30 seconds at 60°C. The following primers were used (F = forward, R = reverse, P = probe):

hIDO1:

F 5′-GGTCATGGAGATGTCCGTAA-3′

R 5′-ACCAATAGAGAGACCAGGAAGAA-3′

P 5′-CTGTTCCTTACTGCCAACTCTCCAAGAAACTG-3′

hTDO2:

F 5′-CATGGCTGGAAAGAACTC-3′

R 5′-CTGAAGTGCTCTGTATGAC-3′

P 5′-TTTAGAGCCACATGGATTTAACTTCTGGG-3′

hEF1 (EEF1A1):

F 5′-GCTTCACTGCTCAGGTGAT-3′

R 5′-GCCGTGTGGCAATCCAAT-3′

P 5′-AAATAAGCGCCGGCTATGCCCCTG-3′

mTdo2:

F 5′-GTATCTATGGAGGACAATGAAG-3′

R 5′-GATGAATAGGTGCTCGTCATG-3′

P 5′-CCTCCTTTGCTGGCTCTGTTTACACC-3′

mβ-actin (Actb):

F 5′-CTCTGGCTCCTAGCACCATGAAG-3′

R 5′-GCTGGAAGGTGGACAGTGAG-3′

P 5′-ATCGGTGGCTCCATCCTGGC-3′

The probes were coupled to 5′ FAM and 3′ TAMRA. Standard curves were added for each gene. Mβ-Actin or hEF1 were used for normalization.

Tryptophan and kynurenine quantification

Tryptophan and kynurenine were quantified in the cell culture supernatants by HPLC based on the retention time and the UV absorption (280 nm for tryptophan, 360 nm for kynurenine; ref. 34).

Western blot

Protocol #1

This protocol was used in Supplementary Fig. S2A. The cells were lysed in a homemade lysis buffer (0.1% SDS, 1% sodium deoxycholate, 0.5% Nonidet P40) with complete Protease Inhibitor Cocktail (Sigma-Aldrich). A defined amount of protein was heated at 70°C for 10 minutes with NuPAGE LDS Sample Buffer (Thermo Fisher Scientific) and NuPAGE Sample Reducing Agent (Thermo Fisher Scientific). Proteins (20 μg) were loaded on Bolt 4%–12% Bis-Tris Plus Gels (Thermo Fisher Scientific) and separated by gel electrophoresis using Bolt MOPS SDS Running Buffer (Thermo Fisher Scientific). Dry transfer was performed by iBlot (Thermo Fisher Scientific) using iBlot Gel Transfer Stacks, nitrocellulose, regular size (Thermo Fisher Scientific), and a transfer program of 7 minutes (1 minute at 20 V, 4 minutes at 23 V, 2 minutes at 25 V). The membranes were incubated overnight at 4°C with the primary antibodies: mouse anti-TDO clone III at 1 μg/mL, clone V at 1 μg/mL, or mouse anti-β-actin clone AC-15 at 1:10,000 (Sigma-Aldrich, #A5441). After washing in PBS Tween 20, the membranes were incubated with HRP-linked goat anti-mouse IgG (R&D Systems, #HAF007) at 1:5,000 for 1 hour at RT and washed again. HRP was revealed using SuperSignal West Pico (Thermo Fisher Scientific) and detected with Fusion Solo S (Vilber Lourmat). TDO and β-actin were stained on the same membrane, and stripping between both stainings was performed by 15-minute incubation in Restore PLUS Western Blot Stripping Buffer (Thermo Fisher Scientific).

Protocol #2

This protocol was used in Supplementary Fig. S2B and S2C. The cells and whole livers were lysed in Pierce Ripa buffer (Thermo Fisher Scientific) with Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific). The livers were crushed using a TissueLyser LT (Qiagen). A defined amount of proteins was heated at 95°C for 5 minutes with Laemmli loading buffer. Proteins (20 μg) were loaded on NuPAGE Bis-Tris 4–12% gels (Thermo Fisher Scientific) and separated by gel electrophoresis using NuPAGE MOPS SDS Running Buffer (Thermo Fisher Scientific). Dry transfer was performed like in protocol #1. After blocking, the membranes were incubated for 2 hours at RT with the primary antibodies: mouse anti-TDO clone III at 1 μg/mL, clone V at 1 μg/mL or mouse anti-vinculin clone hVIN-1 at 1:10,000 (Sigma-Aldrich, #V9131). After washing, the membranes were incubated with HRP-linked goat anti-mouse IgG (BioLegend, #405306) at 1:2,500 in blocking buffer for 1 hour at RT and washed again. HRP was revealed like in protocol #1.

Statistical analyses

Statistical analyses were performed using Prism 6 (GraphPad Software). P values of ≤ 0.05 were considered statistically significant. Correlation analysis was performed using two-tailed Spearman rank correlation. Two paired groups were compared using the two-tailed Wilcoxon matched-pairs signed rank test.

Production and validation of TDO-specific antibodies

Given the lack of fully validated antibodies recognizing TDO, we first produced new TDO-specific mAbs by immunizing mice with DNA and boosting with cells engineered to express full-length human TDO at the cell surface (37). We selected two clones (clone V, IgG2aκ, and clone III, IgG2bκ) and validated them based on the following criteria: They recognized a single band of the expected size (47 kDa) only in human cell lines that express TDO2 mRNA (Supplementary Fig. S2A and S2B). This band disappeared after CRISPR-Cas9 genetic inactivation of TDO2 (Supplementary Fig. S2A and S2B). In TDO2-negative human cells, the same band appeared upon transfection of TDO2 (Supplementary Fig. S2A). This band was absent in cells expressing IDO1 but not TDO2, indicating specificity (Supplementary Fig. S2B). Although both mAbs recognized human TDO, only mAb V recognized mouse TDO, and stained a single band in liver lysates from WT but not from TDO−/− mice (Supplementary Fig. S2C). Of note, human and murine TDO are 89% identical (protein BLAST database). IHC of fixed pellets of human cells showed a cytoplasmic staining in cell lines expressing TDO but not in TDO-negative cell lines (Supplementary Fig. S2D).

TDO was expressed in the majority of late-stage human cancers

We used the aforementioned antibodies to analyze the expression of TDO in human tumors and determine the proportion of TDO expression and the type of cells that express TDO. We stained a large number of human tumors of various histologies with both mAbs and found TDO-positive cells in most of them (Fig. 1A and Table 2). The results obtained with both TDO mAbs were identical. Importantly, in all cases we used adjacent sections to detect the presence of TDO mRNA by RT-qPCR and/or in situ hybridization (ISH) to validate the specificity of the stainings (Fig. 1B). Samples were considered positive only when they were stained by IHC with both antibodies and contained TDO2 mRNA.

Figure 1.

TDO expression in human tumors. A, Representative pictures (from 146 tumors and 24 infiltrated lymph nodes) showing TDO IHC staining with the mouse TDO mAb V on FFPE sections of different human tumors. The dotted lines delineate necrotic or hemorrhagic tumor areas. Scale bars, 50–2,000 μm. B, TDO was stained by IHC with the mouse TDO mAb V and mAb III, and by ISH with anti-hTDO2 mRNA probes on adjacent sections of an FFPE kidney carcinoma. Scale bar, 50 μm. C, Representative pictures illustrating different TDO expression levels: Level 0 is negative for TDO, level 1 contains some isolated TDO-positive cells, level 2 contains regions enriched in TDO-positive cells, and level 3 contains several regions with high amounts of TDO-positive cells. Scale bar, 50 μm. The graph and the cross-tabulation represent the numbers and percentages of tumors organized according to the level of TDO expression and the tumor grade. For all IHC stainings, negative controls were performed with a mouse IgG2a isotype control. ISH-negative controls were performed with an anti-dapB probe. All negative controls remained unstained.

Figure 1.

TDO expression in human tumors. A, Representative pictures (from 146 tumors and 24 infiltrated lymph nodes) showing TDO IHC staining with the mouse TDO mAb V on FFPE sections of different human tumors. The dotted lines delineate necrotic or hemorrhagic tumor areas. Scale bars, 50–2,000 μm. B, TDO was stained by IHC with the mouse TDO mAb V and mAb III, and by ISH with anti-hTDO2 mRNA probes on adjacent sections of an FFPE kidney carcinoma. Scale bar, 50 μm. C, Representative pictures illustrating different TDO expression levels: Level 0 is negative for TDO, level 1 contains some isolated TDO-positive cells, level 2 contains regions enriched in TDO-positive cells, and level 3 contains several regions with high amounts of TDO-positive cells. Scale bar, 50 μm. The graph and the cross-tabulation represent the numbers and percentages of tumors organized according to the level of TDO expression and the tumor grade. For all IHC stainings, negative controls were performed with a mouse IgG2a isotype control. ISH-negative controls were performed with an anti-dapB probe. All negative controls remained unstained.

Close modal
Table 2.

TDO protein expression in human tumors.

TumorsTDO+/total (%)Level 0Level 1Level 2Level 3
Hepatocarcinoma 10/10 (100%) 10 
Glioblastoma 38/39 (97%) 25 
Bladder carcinoma 12/13 (92%) 
Pancreatic carcinoma 10/11 (91%) 
Colon carcinoma 9/10 (90%) 
Stomach carcinoma 6/8 (75%) 
Endometrial carcinoma 5/7 (71%) 
Lung carcinoma 7/11 (64%) 
Melanoma 3/6 (50%) 
 Metastatic lymph nodes 8/9 (89%) 
 Nonmetastatic lymph nodes 1/9 (11%) 
Breast carcinoma 4/9 (44%) 
 Metastatic lymph nodes 12/15 (80%) 
 Nonmetastatic lymph nodes 0/6 (0%) 
Kidney carcinoma 4/10 (40%) 
Astrocytoma 0/12 (0%) 12 
Total of all tumors (without lymph nodes) 108/146 (74%)     
TumorsTDO+/total (%)Level 0Level 1Level 2Level 3
Hepatocarcinoma 10/10 (100%) 10 
Glioblastoma 38/39 (97%) 25 
Bladder carcinoma 12/13 (92%) 
Pancreatic carcinoma 10/11 (91%) 
Colon carcinoma 9/10 (90%) 
Stomach carcinoma 6/8 (75%) 
Endometrial carcinoma 5/7 (71%) 
Lung carcinoma 7/11 (64%) 
Melanoma 3/6 (50%) 
 Metastatic lymph nodes 8/9 (89%) 
 Nonmetastatic lymph nodes 1/9 (11%) 
Breast carcinoma 4/9 (44%) 
 Metastatic lymph nodes 12/15 (80%) 
 Nonmetastatic lymph nodes 0/6 (0%) 
Kidney carcinoma 4/10 (40%) 
Astrocytoma 0/12 (0%) 12 
Total of all tumors (without lymph nodes) 108/146 (74%)     

Note: The table lists all cancer types tested, the number and percentage of samples containing TDO-positive cells, as well as the level of TDO expression (illustrated in Fig. 1C). We performed the screening by staining three adjacent tissue sections with the anti-TDO mAb III and mAb V, and with an isotype control. The two following sections were used for RNA extraction and RT-qPCR analyses to confirm that tissues showing staining by IHC also contained TDO2 mRNA. When IHC staining was present, but no mRNA was detected by RT-qPCR, the mRNA of the housekeeping gene EF1 was often highly degraded. If, on the contrary, EF1 mRNA was in a normal range, we performed ISH to confirm the IHC staining. In cases with a positive RT-qPCR result but a negative IHC result, we suspected contamination of the mRNA when cutting the tissues on the microtome. To confirm the IHC result, we performed ISH on these tissues. In some cases, especially in lung and kidney samples, we detected TDO2 by ISH, showing that the positive RT-qPCR result was correct, but the expression of the TDO protein probably too low to be detected by the antibody. A tumor was considered positive when IHC staining was detected. We did not take into account tumors that were positive by RT-qPCR but negative in IHC, because we knew from in vitro experiments that cell lines displaying the same expression and staining pattern did not have any catalytic activity.

With the exception of astrocytomas, we found TDO-positive samples in all the tumor types tested. All hepatocarcinomas expressed TDO and showed cytoplasmic staining in most tumor cells (Fig. 1A). This is in line with the physiologic expression of TDO in normal hepatocytes (39). Other tumors, including glioblastomas, bladder, pancreatic and colon carcinomas, also showed a high proportion of TDO-positive samples, but here the staining was limited to some cells in focal tumor areas, usually corresponding to vascular structures. Such focal staining pattern was observed in all the TDO-positive tumors other than hepatocarcinomas (Fig. 1A; Table 2). Interestingly, although only half of the tested melanomas and breast carcinomas were TDO positive, more than 80% of metastatic lymph nodes contained TDO-expressing cells, which were absent from nonmetastatic lymph nodes (Table 2).

The vascular structures expressing TDO were often located at the tumor periphery or surrounded necrotic and hemorrhagic areas (Fig. 1A). In tumor-infiltrated lymph nodes, TDO-positive vessels were found mainly at the borders of the metastases and at the interface between tumor and lymphatic tissue. We then wondered whether the level of TDO expression correlated with clinical features, such as the age of the patients, their treatment, the tumor grade, and the presence of lymph node or distant metastases. Considering all the tumor types tested (except hepatocarcinoma), we found that the proportion of TDO-expressing cells was inversely correlated with tumor differentiation, with higher proportions in higher histologic grades (Fig. 1C). An illustrative example is glioma: whereas grade II and III astrocytomas were all TDO-negative, 97% of glioblastomas multiforme (grade IV gliomas) contained TDO-positive cells (Table 2).

TDO was expressed in human tumors by morphologically abnormal vessels

To identify the cell type expressing TDO in these vascular structures, we performed a series of costainings (Fig. 2A). We observed that all TDO-positive cells expressed the beta-type platelet-derived growth factor receptor (PDGFRβ), a marker of mesenchymal cells [fibroblasts, pericytes, vascular smooth muscle cells (vSMCs); ref. 40], whereas not all PDGFRβ-positive cells expressed TDO (Fig. 2A). Some, but not all, TDO-positive cells expressed α-smooth muscle actin (αSMA) and NG2 (neural/glial antigen 2; Fig. 2A). TDO-positive cells also surrounded CD34-positive endothelial cells, but TDO never colocalized with CD34 (Fig. 2A). We therefore concluded that TDO-expressing cells corresponded to vascular smooth muscle cells or pericytes. These two cell types, which have very similar phenotypes, can be identified according to their localization toward the basement membrane in normal vessels, but can no longer be distinguished in disorganized tumor vessels (41, 42). In addition to TDO-positive pericytes, which were identified in all tumor types including hepatocellular carcinomas (HCC), some tumors also contained TDO-expressing tumor cells, including, as mentioned above, all HCCs, but also 10 of 39 glioblastomas, in which TDO was partially coexpressed with glial fibrillary acidic protein (GFAP) and β3-tubulin (Fig. 2A), and 1 of 10 kidney carcinoma (Fig. 1B). These TDO-positive tumor cells surrounded TDO-positive vessels bordering necrotic tumor areas. In none of the tested tumors was TDO expressed by macrophages or dendritic cells (HLA-DR-positive; Fig. 2A).

Figure 2.

TDO expression by pericytes in abnormal tumor vessels. A, Costainings of TDO with different cell-type markers were performed on a certain number of TDO-positive tumors from Table 2: endometrial carcinoma (4 costainings performed/6 TDO-positive tumors), kidney carcinoma (2/4), melanoma (2/4), pancreatic carcinoma (4/9), glioblastoma (15/37), hepatocarcinoma (4/10), stomach carcinoma (2/6), bladder carcinoma (4/12), lung carcinoma (2/7), and colon carcinoma (2/9). Representative pictures of FFPE glioblastoma sections stained by IF with TDO mAb V in orange and the comarkers in green are shown. Colocalization appears in yellow. Orange arrows point to TDO-positive cells, green arrows point to cells labeled with comarkers, and yellow arrows point to cells where TDO colocalizes with one of the cell-type markers. Scale bars, 50 μm. B, Triple IF costaining of glioblastoma sections for TDO mAb V (orange), PDGFRβ (green), and CD34 (purple). Colocalization of TDO with PDGFRβ is represented in yellow. Scale bars, 20 μm. C, Representative pictures of an IF costaining for TDO mAb V (orange) and CA9 (green) on a glioblastoma section (out of 17 stained tissues). The dotted line delineates the necrotic tumor area. Scale bar, 100 μm. For all IF stainings, negative controls were performed by omitting the primary antibody and remained unstained.

Figure 2.

TDO expression by pericytes in abnormal tumor vessels. A, Costainings of TDO with different cell-type markers were performed on a certain number of TDO-positive tumors from Table 2: endometrial carcinoma (4 costainings performed/6 TDO-positive tumors), kidney carcinoma (2/4), melanoma (2/4), pancreatic carcinoma (4/9), glioblastoma (15/37), hepatocarcinoma (4/10), stomach carcinoma (2/6), bladder carcinoma (4/12), lung carcinoma (2/7), and colon carcinoma (2/9). Representative pictures of FFPE glioblastoma sections stained by IF with TDO mAb V in orange and the comarkers in green are shown. Colocalization appears in yellow. Orange arrows point to TDO-positive cells, green arrows point to cells labeled with comarkers, and yellow arrows point to cells where TDO colocalizes with one of the cell-type markers. Scale bars, 50 μm. B, Triple IF costaining of glioblastoma sections for TDO mAb V (orange), PDGFRβ (green), and CD34 (purple). Colocalization of TDO with PDGFRβ is represented in yellow. Scale bars, 20 μm. C, Representative pictures of an IF costaining for TDO mAb V (orange) and CA9 (green) on a glioblastoma section (out of 17 stained tissues). The dotted line delineates the necrotic tumor area. Scale bar, 100 μm. For all IF stainings, negative controls were performed by omitting the primary antibody and remained unstained.

Close modal

Although most tumors contained TDO-positive cells, these cells were few in numbers. In the 34 analyzed glioblastoma multiforme tumors (the tumor type with the highest amount of TDO-expressing pericytes), only 5.9% of the PDGFRβ-stained area was also positive for TDO (workflow described in Supplementary Fig. S3). We used PDGFRβ rather than αSMA or NG2 as a pericyte marker, because not all TDO-positive cells expressed αSMA or NG2 (Fig. 2A). However, PDGFRβ is not exclusively expressed by pericytes, but also by cancer-associated fibroblasts (43). Therefore, 5.9% is probably a slight underestimation of the proportion of TDO-positive pericytes.

TDO-positive tumor vessels often displayed abnormal features that were similar to the vascular anomalies previously observed in tumors (42). These features included the accumulation of little vessels surrounded by several layers of pericytes (Fig. 2B, first line), and enlarged vessels with a single, irregular layer of pericytes that did not completely cover the endothelial cells (Fig. 2B, second line). We also found normal TDO-positive capillaries (with a diameter of about 10 μm and a single pericyte layer that completely surrounded the endothelial cells), but they were much less abundant (Fig. 2B, third line). We therefore wondered whether TDO-positive vessels were well perfused with blood or whether these tumor areas were hypoxic. We found no costaining of TDO with the hypoxia marker carbonic anhydrase 9 (CA9; Fig. 2C). This result suggested that TDO-positive tumor regions were well oxygenated, but did not exclude that blood and oxygen were supplied by the neighboring TDO-negative vessels.

TDO and IDO1 did not colocalize in glioblastomas

We wondered if TDO colocalized with IDO1, the other tryptophan-metabolizing enzyme. IHC stainings of glioblastoma multiforme showed that IDO1 was expressed by very few cells, which were either tumor cells or cells with a dendritic cell–like shape (Supplementary Fig. S4A). Costainings with TDO were performed on five tumors and showed that both proteins never colocalized (Supplementary Fig. S4B).

TDO did not affect the infiltration of lymphocytes in glioblastomas

In mouse tumor models, we previously described the immunosuppressive role of TDO when expressed homogeneously by tumor cells (34). This situation was nicely reflected in human hepatocarcinomas, which expressed TDO in all tumor cells (Fig. 1A). It was unclear, however, whether the low proportion of TDO-expressing vascular cells we observed in other tumors would suffice to catabolize tryptophan in a meaningful immunosuppressive way. This was difficult to address in mouse models, in which we never observed a similar expression of TDO in tumor vessels (36). To try to address this question, we costained 29 human glioblastomas for T lymphocytes (CD3 and CD8) and TDO and used an image analysis software to quantify lymphocyte infiltration in TDO-positive versus TDO-negative tumor areas (workflow described in Supplementary Fig. S5). Glioblastomas have the advantage that they rarely expressed IDO1 (ref. 15; Supplementary Fig. S4). As expected, T-cell infiltration was low in these glioblastomas (Fig. 3A; ref. 44). Tumor regions within 60 μm of a TDO-expressing cell (whether a pericyte or a tumor cell) contained slightly more—not less—CD3-expressing lymphocytes, whether or not they also expressed CD8 (Fig. 3A; Supplementary Fig. S5). Moreover, T lymphocytes present in TDO-positive areas tended to be more proliferative, as indicated by Ki67 staining (Fig. 3B; Supplementary Fig. S5). These results suggested that the presence of TDO-positive pericytes does not prevent but might instead favor lymphocyte infiltration in glioblastoma, although this enrichment might result from the fact that the TDO-positive areas were close to vessels, whereas the TDO-negative areas we used for comparison were not, for technical reasons, selected to be close to vessels. Although our approach did not assess lymphocyte function or differentiation, these data do not support an important immunosuppressive role of TDO-expressing pericytes in the tumor microenvironment.

Figure 3.

Quantification of T lymphocytes in TDO-positive and TDO-negative glioblastoma areas. IF costainings were performed for TDO mAb V, CD3, CD8, and Ki67 on tissue sections from 29 glioblastomas multiforme. Negative controls were performed by omitting the primary antibody and remained unstained. Infiltrating lymphocytes were counted in zones 60 μm away from TDO-positive cells and compared with lymphocytes in the remaining tumor regions (the quantification workflow is illustrated in Supplementary Fig. S5). A, CD3+, CD3+CD8, or CD3+CD8+ cells were counted and expressed as the number of cells per mm2 of tissue area. B, Ki67-positive lymphocytes were counted and expressed as a percentage of the parental lymphocyte population. Mean ± SD, Wilcoxon matched-pairs signed rank test (*, P < 0.05; ****, P < 0.0001).

Figure 3.

Quantification of T lymphocytes in TDO-positive and TDO-negative glioblastoma areas. IF costainings were performed for TDO mAb V, CD3, CD8, and Ki67 on tissue sections from 29 glioblastomas multiforme. Negative controls were performed by omitting the primary antibody and remained unstained. Infiltrating lymphocytes were counted in zones 60 μm away from TDO-positive cells and compared with lymphocytes in the remaining tumor regions (the quantification workflow is illustrated in Supplementary Fig. S5). A, CD3+, CD3+CD8, or CD3+CD8+ cells were counted and expressed as the number of cells per mm2 of tissue area. B, Ki67-positive lymphocytes were counted and expressed as a percentage of the parental lymphocyte population. Mean ± SD, Wilcoxon matched-pairs signed rank test (*, P < 0.05; ****, P < 0.0001).

Close modal

TDO expression in pericytes from inflammatory pulmonary lesions

We then took advantage of our validated TDO-specific mAbs to evaluate the expression profile of TDO in normal tissues. We stained nontumoral sections from the organs harboring the tumors listed in Table 2, including 5 pancreata, 1 uterus, 3 colons, 2 stomachs, 2 lungs, 6 kidneys, 10 livers, and 10 bladders. These tissues were negative for TDO, with the exception of liver and lung. As expected, all liver samples showed TDO expression in hepatocytes (Fig. 4A).

Figure 4.

TDO expression in human normal liver, pulmonary granulomas, and placental tissues. A, Representative pictures showing TDO mAb V staining of FFPE sections of liver (10 samples), lung (13 samples), decidua (9 samples), and ectopic pregnancies (5 samples). Scale bars, 50–2,000 μm. Similar stainings were observed with both primary antibodies mAb III and V. Negative controls were performed with a mouse IgG2a isotype control and remained unstained. B, Costainings of TDO with different cell-type markers were performed on a certain number of TDO-positive samples: lung (6 costainings performed/7 TDO-positive samples), decidua (4/7), and ectopic pregnancies (2/5). Representative pictures of FFPE pulmonary granuloma and ectopic pregnancy sections stained by IF with TDO mAb V (orange), PDGFRβ (green), CD34 (purple), cytokeratin (blue), or CD68 (macrophages, blue) are shown. Colocalization of TDO with PDGFRβ is represented in yellow. Colocalization of TDO with cytokeratin is represented in clear blue. The arrows point to a pericyte that coexpresses TDO and PDGFRβ. The surrounded cell is a syncytiotrophoblast that expresses both TDO and cytokeratin. Scale bars, 20–50 μm. Negative controls were performed by omitting the primary antibody and remained unstained.

Figure 4.

TDO expression in human normal liver, pulmonary granulomas, and placental tissues. A, Representative pictures showing TDO mAb V staining of FFPE sections of liver (10 samples), lung (13 samples), decidua (9 samples), and ectopic pregnancies (5 samples). Scale bars, 50–2,000 μm. Similar stainings were observed with both primary antibodies mAb III and V. Negative controls were performed with a mouse IgG2a isotype control and remained unstained. B, Costainings of TDO with different cell-type markers were performed on a certain number of TDO-positive samples: lung (6 costainings performed/7 TDO-positive samples), decidua (4/7), and ectopic pregnancies (2/5). Representative pictures of FFPE pulmonary granuloma and ectopic pregnancy sections stained by IF with TDO mAb V (orange), PDGFRβ (green), CD34 (purple), cytokeratin (blue), or CD68 (macrophages, blue) are shown. Colocalization of TDO with PDGFRβ is represented in yellow. Colocalization of TDO with cytokeratin is represented in clear blue. The arrows point to a pericyte that coexpresses TDO and PDGFRβ. The surrounded cell is a syncytiotrophoblast that expresses both TDO and cytokeratin. Scale bars, 20–50 μm. Negative controls were performed by omitting the primary antibody and remained unstained.

Close modal

Unexpectedly, both lung samples contained TDO-positive pericytes (Fig. 4A and B). To determine whether TDO expression was linked to the presence of a tumor in the lung, we stained 11 additional nontumoral lung samples collected at a distance from the lesion in patients with or without benign or malignant tumor, as indicated in Table 3. Five of them contained TDO-expressing pericytes (Fig. 4A and B). Interestingly, all 7 samples containing TDO-positive cells also showed signs of inflammation, some of them also containing granulation tissue, which is tissue formed during wound healing and characterized by neovascularization, extracellular matrix formation, and the presence of immune cells (Table 3). In contrast, none of the six samples without inflammation contained TDO-expressing vessels (Table 3). These results indicated that TDO expression in lung pericytes was associated with inflammatory processes.

Table 3.

TDO expression in nontumoral lung pericytes.

Clinical characteristicsTDO levelInflammationGranulation tissue
Normal lung (autopsy after sudden death) No No 
Nonmalignant lung (atelectasis) No No 
Distant metastasis (colorectal adenocarcinoma) No No 
Distant metastasis (sarcoma) No No 
Distant metastasis (sarcoma) No No 
Distant metastasis (sarcoma) No No 
Normal lung (negative search for metastases) Some cells No 
Nonmalignant lung (atelectasis and cyst) Some cells No 
Malignant lung carcinoma Yes No 
Nonmalignant lung (emphysema) Yes No 
Distant metastasis (sarcoma) Yes Yes 
Malignant carcinoid tumor Yes Yes 
Malignant lung carcinoma Yes No 
Clinical characteristicsTDO levelInflammationGranulation tissue
Normal lung (autopsy after sudden death) No No 
Nonmalignant lung (atelectasis) No No 
Distant metastasis (colorectal adenocarcinoma) No No 
Distant metastasis (sarcoma) No No 
Distant metastasis (sarcoma) No No 
Distant metastasis (sarcoma) No No 
Normal lung (negative search for metastases) Some cells No 
Nonmalignant lung (atelectasis and cyst) Some cells No 
Malignant lung carcinoma Yes No 
Nonmalignant lung (emphysema) Yes No 
Distant metastasis (sarcoma) Yes Yes 
Malignant carcinoid tumor Yes Yes 
Malignant lung carcinoma Yes No 

Note: The table lists all lung samples resected at distant sites from the indicated lesion, the level of TDO expression, and the presence of inflammation or granulation tissue (illustrated in Fig. 4). The staining protocol is the same as in Table 2. We observed that TDO was exclusively expressed by pericytes.

TDO expression in decidua and placenta

Because the murine decidua is known to express TDO (30–32), we also tested human deciduas of the first trimester and ectopic pregnancies by IHC. We found TDO expression only in a few cells, which corresponded to pericytes (PDGFRβ-positive) located inside chorionic villi, and to interstitial syncytiotrophoblasts (multinuclear cells expressing cytokeratin; Fig. 4A and B). Trying to identify the mechanism responsible for TDO expression in pericytes, we treated murine mesenchymal stem cells (MSC), which are precursors of pericytes, with dexamethasone, or with different combinations of pregnancy hormones or cytokines able to induce inflammation or differentiation to fibroblasts/vSMCs. However, we could not detect any induction of TDO2 mRNA (Supplementary Table S1). As TDO is expressed in the murine decidua, the maternal part of the placenta (31, 32), we also treated decidualized and undecidualized uterine cells. Except for a small increase of TDO expression by LPS and IFNγ in decidualized cells, TDO was not induced (Supplementary Table S1).

In conclusion, we observed massive TDO expression in human hepatocarcinoma tumor cells, but a limited expression in other tumors, restricted to pericytes in tumor vessels located in angiogenic areas. Similar TDO-expressing pericytes were observed in nontumoral inflammatory lesions of the lung and in placental tissue.

So far, the evidence supporting TDO expression in human tumors is 3-fold. First, TDO2 mRNA is detectable in human tumors by RT-qPCR performed on bulk tumor RNA (34). Second, a number of human tumor lines express TDO at the mRNA and protein level and effectively degrade tryptophan into kynurenine, which is inhibited by TDO inhibitors (12, 34). Third, IHC studies of several human tumor types (colon carcinoma, breast cancer, non–small cell lung carcinoma, ovarian carcinoma, renal cell carcinoma, and brain metastases of malignant melanoma) report widespread and homogeneous expression of TDO in tumor cells (12). Based on the expression of TDO in human tumor lines and on the staining patterns reported by Opitz and colleagues (12), it is considered that TDO is expressed by tumor cells themselves in human tumors. However, no validated TDO-specific mAb was available at the time, so that the reported IHC stainings were performed with nonvalidated polyclonal antibodies, which carry the inherent risk of cross-reactivity, particularly when used for IHC, which does not control for the molecular size of the signal (15). In this study, we described and fully validated two highly specific mAbs recognizing TDO. They stained a single band of the expected molecular weight when used in western blots and stained, by both western blot and IHC, cell lines in a TDO expression–dependent manner. Using these mAbs, we confirmed TDO expression in a large number of human tumors. In HCC, TDO was highly expressed in tumor cells themselves. In most other tumors, however, we did not observe TDO expression in tumor cells, but only in scattered cells often located next to necrotic or hemorrhagic areas. The TDO specificity of this staining was further confirmed by ISH, which detected a similar expression pattern of TDO2 mRNA in adjacent sections. TDO-expressing cells belonged to vascular structures with abnormal morphology, typical of vascular anomalies previously reported in tumor vessels (42). Further stainings identified these TDO-expressing cells as pericytes/vSMCs, expressing PDGFRβ. The lack of TDO expression in tumor cells other than HCC came as a surprise, particularly given the expression of TDO in several human tumor lines of glioblastoma and carcinoma of the colon, head and neck, lung and gall bladder (12, 34). This disconnection between tumor lines and tumor samples is intriguing, even more so when considering human HCC, in which the opposite disconnection was observed: although TDO was expressed in tumor cells in HCC samples, HCC lines tested proved negative for TDO expression (34).

The function of hepatic TDO is to regulate systemic tryptophan concentrations, as illustrated by the 9-fold increased level of tryptophan in the serum of TDO-KO mice (25, 36). By analogy to the tryptophan-degrading enzyme IDO1, the proposed function of TDO in tumors is the inhibition of immune-mediated tumor rejection (12, 34). The impact of IDO1 on immune evasion is well defined via several in vivo experiments using different tumor models (1, 20, 45, 46). In humans, IDO1 is expressed by a large number of tumors, and high IDO1 expression is positively correlated with tumor progression and decreased survival (2, 21, 47). Several IDO1 inhibitors are now in clinical development (21). Similar to IDO1, TDO negatively affects T lymphocyte activity and proliferation as seen in several in vitro and in vivo experiments (12, 34, 48). TDO-positive cells block in vitro IFNγ production and proliferation of T lymphocytes (48) and TDO favors escape of murine tumors to immune rejection (12, 34). The outcome of LPS-induced endotoxemia is worsened in TDO−/− mice due to the increased secretion of proinflammatory cytokines (49). The dominant expression of TDO in human HCC confirms the potential of TDO as a therapeutic target in this cancer type. Although checkpoint inhibitors were recently approved in HCC (50, 51), their clinical efficacy remains limited and could be improved upon combination with TDO inhibitors, several of which have been developed (26, 34–36).

In tumor types other than hepatocarcinomas, including gliobastomas, melanomas, and carcinomas of the pancreas, colon, stomach, lung, endometrium, breast and kidney, the role of TDO expression in pericytes associated with abnormal tumor vessels is unclear at this stage. A local immunosuppressive function is possible and in line with previous reports indicating that tumor-derived vascular pericytes can cause T helper cell anergy (52). Pericytes derived from human glioblastomas have stem cell–like and immunosuppressive properties associated with TDO expression, albeit at a very low level (53). Because we could not find expression of TDO in murine tumor–associated pericytes, we could not experimentally test the immunosuppressive role of TDO-expressing pericytes in vivo (36). In human glioblastoma, our quantification of T cells in tumor areas containing TDO-positive cells did not show a reduced infiltration or proliferation of CD3- or CD8-positive cells. This result suggests that TDO-expressing pericytes did not negatively affect the recruitment and infiltration of T cells into the tumor, although they could still affect the functional status or anergic state of tumor-infiltrating lymphocytes. However, although present in most tumor samples, TDO-expressing pericytes comprised only about 6% of pericytes within a given sample. It was unclear whether such a low proportion of TDO-expressing cells was sufficient to induce the level of tryptophan degradation that is required to induce immunosuppression in the whole tumor mass.

We observed that TDO-positive vessels were often located around necrotic and hemorrhagic tumor areas, which were characterized by neovascularization. We also found TDO-expressing vessels in nontumoral lung granulation tissue, which also involved neovascularization, and in chorionic villi at the time of embryonic vessel formation. TDO could therefore play a role in neovascularization, by way of the vasoactive function of one of the tryptophan-derived metabolites. Kynurenine produced by endothelial cells expressing IDO1 during inflammation can induce vessel relaxation, and the severity of septic shock is directly correlated to the expression of IDO1 (54, 55). IDO1-deficient mice have a reduced density of pulmonary blood vessels (56) and show reduced neovascularization in models of oxygen-induced retinopathy and lung metastasis (57). In addition, a newly described tryptophan-derived tricyclic hydroperoxide, formed by IDO1 in the presence of H2O2, causes vessel relaxation of IFNγ-pretreated mouse abdominal aortas (58). This or a similar compound could also be produced by TDO, given the similar enzymatic mechanism of both enzymes. A link between TDO expression and neovascularization would also be in line with our observation that glioblastomas, which display much more neoangiogenesis as compared with astrocytomas, also contained much more TDO-expressing vessels.

It is unclear what the mechanism that triggers TDO expression in tumor pericytes is. Rat hepatocytes increase TDO2 expression upon treatment with glucocorticoids (27), whereas murine decidual cells do so upon exposure to estrogen and progesterone (32). However, we were unable to induce TDO2 expression by adding dexamethasone, different combinations of pregnancy hormones or cytokines or inflammatory stimuli to murine MSC or to uterine decidualized cells. As we observed TDO-positive pericytes in angiogenic tumor areas, granuloma tissues and in placenta, the induction of TDO might have resulted from a complex interplay of different physiologic and pathologic phenomena related to hypoxia and inflammation. In line with this, recent reports describe the induction of TDO in fibroblasts, which, like pericytes, are derived from MSCs: TDO was upregulated in uterine stromal fibroblasts by natural killer cells (59) and in normal lung fibroblasts by galectin-1 secreted from lung tumor cell lines (60).

In sum, our results confirm TDO expression at the protein level in a large range of human cancers. The high TDO expression by HCC cells warrants the development of TDO inhibitors in this indication. In cancers without massive TDO expression in tumor cells, TDO inhibitors may synergize with immunotherapy in a different manner, by blocking hepatic TDO and increasing systemic tryptophan levels, as we report in the accompanying manuscript (36).

B.J. Van den Eynde is a scientific advisory board member for and has ownership interest (including patents) in iTeos Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: D. Hoffmann, B.J. Van den Eynde

Development of methodology: D. Hoffmann, J.-C. Renauld, N. van Baren, B.J. Van den Eynde

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D. Hoffmann, T. Dvorakova, V. Stroobant, M. Solvay, S. Klaessens, M.-C. Letellier, N. van Baren

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D. Hoffmann, V. Stroobant, C. Bouzin, A. Daumerie, N. van Baren, J. Lelotte, E. Marbaix, B.J. Van den Eynde

Writing, review, and/or revision of the manuscript: D. Hoffmann, N. van Baren, E. Marbaix, B.J. Van den Eynde

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D. Hoffmann, T. Dvorakova, V. Stroobant, C. Bouzin, A. Daumerie, M. Solvay, S. Klaessens, M.-C. Letellier, J.-C. Renauld, N. van Baren, J. Lelotte, E. Marbaix, B.J. Van den Eynde

Study supervision: B.J. Van den Eynde

Other (contribution of material for the study): E. Marbaix

The authors thank Dr. Hiroshi Funakoshi, Dr. Toshikazu Nakamura, and Prof. Michael Platten for providing TDO-KO mice; Guy Warnier and his team for the production of TDO-KO mice; the BioLibrary of the Institut Roi Albert II of the Cliniques universitaires Saint-Luc (Brussels, Belgium), the Service d'Anatomie Pathologique of the CHU-Brugmann (Brussels, Belgium), BHUL—Université de Liège—CHU de Liège (Belgium), and Prof. Christine Galant for providing tissue samples; Vanesa Bol for the ISH protocol; Dr. Pascal Brouillard, senior manager of the Genomics Platform of UCLouvain, for genotyping of the cell lines; and Luc Pilotte and Auriane Sibille for editorial assistance. D. Hoffmann was supported by FNRS-FRIA (grant number: 1.E082.14), T. Dvorakova by FNRS-Télévie (grant number: 7.4597.18), V. Stroobant and B.J. Van den Eynde by the Ludwig Institute for Cancer Research, C. Bouzin, A. Daumerie, and J.-C. Renauld by UCLouvain, M. Solvay by FNRS-Aspirant (grant number: 1.A385.16), S. Klaessens by FNRS-FRIA (grant number: 1.E100.14), M.-C. Letellier by iTeos Therapeutics, N. van Baren by de Duve Institute, J. Lelotte by Cliniques universitaires Saint-Luc, and E. Marbaix by Cliniques universitaires Saint-Luc.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Uyttenhove
C
,
Pilotte
L
,
Theate
I
,
Stroobant
V
,
Colau
D
,
Parmentier
N
, et al
Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase
.
Nat Med
2003
;
9
:
1269
74
.
2.
van Baren
N
,
Van den Eynde
BJ
. 
Tryptophan-degrading enzymes in tumoral immune resistance
.
Front Immunol
2015
;
6
:
34
.
3.
van Baren
N
,
Van den Eynde
BJ
. 
Tumoral immune resistance mediated by enzymes that degrade tryptophan
.
Cancer Immunol Res
2015
;
3
:
978
85
.
4.
Shimizu
T
,
Nomiyama
S
,
Hirata
F
,
Hayaishi
O
. 
Indoleamine 2,3-dioxygenase. Purification and some properties
.
J Biol Chem
1978
;
253
:
4700
6
.
5.
Rafice
SA
,
Chauhan
N
,
Efimov
I
,
Basran
J
,
Raven
EL
. 
Oxidation of L-tryptophan in biology: a comparison between tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase
.
Biochem Soc Trans
2009
;
37
:
408
12
.
6.
Oxender
DL
,
Christensen
HN.
Evidence for two types of mediation of neutral and amino-acid transport in Ehrlich cells
.
Nature
1963
;
197
:
765
7
.
7.
Vumma
R
,
Johansson
J
,
Lewander
T
,
Venizelos
N
. 
Tryptophan transport in human fibroblast cells-a functional characterization
.
Int J Tryptophan Res
2011
;
4
:
19
27
.
8.
Kudo
Y
,
Boyd
CA.
Characterisation of L-tryptophan transporters in human placenta: a comparison of brush border and basal membrane vesicles
.
J Physiol
2001
;
531
:
405
516
.
9.
Kaper
T
,
Looger
LL
,
Takanaga
H
,
Platten
M
,
Steinman
L
,
Frommer
WB
. 
Nanosensor detection of an immunoregulatory tryptophan influx/kynurenine efflux cycle
.
PLoS Biol
2007
;
5
:
e257
.
10.
Speciale
C
,
Hares
K
,
Schwarcz
R
,
Brookes
N
. 
High-affinity uptake of L-kynurenine by a Na+-independent transporter of neutral amino acids in astrocytes
.
J Neurosci
1989
;
9
:
2066
72
.
11.
Munn
DH
,
Sharma
MD
,
Baban
B
,
Harding
HP
,
Zhang
Y
,
Ron
D
, et al
GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase
.
Immunity
2005
;
22
:
633
42
.
12.
Opitz
CA
,
Litzenburger
UM
,
Sahm
F
,
Ott
M
,
Tritschler
I
,
Trump
S
, et al
An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor
.
Nature
2011
;
478
:
197
203
.
13.
Terness
P
,
Bauer
TM
,
Rose
L
,
Dufter
C
,
Watzlik
A
,
Simon
H
, et al
Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites
.
J Exp Med
2002
;
196
:
447
57
.
14.
Mezrich
JD
,
Fechner
JH
,
Zhang
X
,
Johnson
BP
,
Burlingham
WJ
,
Bradfield
CA
. 
An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells
.
J Immunol
2010
;
185
:
3190
8
.
15.
Theate
I
,
van Baren
N
,
Pilotte
L
,
Moulin
P
,
Larrieu
P
,
Renauld
JC
, et al
Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues
.
Cancer Immunol Res
2015
;
3
:
161
72
.
16.
Dai
W
,
Gupta
SL.
Regulation of indoleamine 2,3-dioxygenase gene expression in human fibroblasts by interferon-gamma. Upstream control region discriminates between interferon-gamma and interferon-alpha
.
J Biol Chem
1990
;
265
:
19871
77
.
17.
Chon
SY
,
Hassanain
HH
,
Pine
R
,
Gupta
SL
. 
Involvement of two regulatory elements in interferon-gamma-regulated expression of human indoleamine 2,3-dioxygenase gene
.
J Interferon Cytokine Res
1995
;
15
:
517
26
.
18.
Munn
DH
,
Zhou
M
,
Attwood
JT
,
Bondarev
I
,
Conway
SJ
,
Marshall
B
, et al
Prevention of allogeneic fetal rejection by tryptophan catabolism
.
Science
1998
;
281
:
1191
3
.
19.
Spranger
S
,
Spaapen
RM
,
Zha
Y
,
Williams
J
,
Meng
Y
,
Ha
TT
, et al
Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells
.
Sci Transl Med
2013
;
5
:
200ra116
.
20.
Hennequart
M
,
Pilotte
L
,
Cane
S
,
Hoffmann
D
,
Stroobant
V
,
De Plaen
E
, et al
Constitutive IDO1 expression in human tumors is driven by cyclooxygenase-2 and mediates intrinsic immune resistance
.
Cancer Immunol Res
2017
;
5
:
695
709
.
21.
Brochez
L
,
Chevolet
I
,
Kruse
V
. 
The rationale of indoleamine 2,3-dioxygenase inhibition for cancer therapy
.
Eur J Cancer
2017
;
76
:
167
82
.
22.
Long
GV
,
Dummer
R
,
Hamid
O
,
Gajewski
T
,
Caglevic
C
,
Dalle
S
, et al
Epacadostat (E) plus pembrolizumab (P) versus pembrolizumab alone in patients (pts) with unresectable or metastatic melanoma: results of the phase 3 ECHO-301/KEYNOTE-252 study
.
J Clin Oncol
36
, 
2018
(suppl; abstr 108).
23.
Van den Eynde
BJ
,
van Baren
N
,
Baurain
JF
. 
Is there a clinical future for IDO1 inhibitors after the failure
of epacadostat in melanoma?
Annu Rev Cancer Biol
2020
;
4
:
241
56
.
24.
Muller
AJ
,
Manfredi
MG
,
Zakharia
Y
,
Prendergast
GC
. 
Inhibiting IDO pathways to treat cancer: lessons from the ECHO-301 trial and beyond
.
Semin Immunopathol
2019
;
41
:
41
8
.
25.
Kanai
M
,
Funakoshi
H
,
Takahashi
H
,
Hayakawa
T
,
Mizuno
S
,
Matsumoto
K
, et al
Tryptophan 2,3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice
.
Mol Brain
2009
;
2
:
8
.
26.
Salter
M
,
Hazelwood
R
,
Pogson
CI
,
Iyer
R
,
Madge
DJ
. 
The effects of a novel and selective inhibitor of tryptophan 2,3-dioxygenase on tryptophan and serotonin metabolism in the rat
.
Biochem Pharmacol
1995
;
49
:
1435
42
.
27.
Danesch
U
,
Hashimoto
S
,
Renkawitz
R
,
Schutz
G
. 
Transcriptional regulation of the tryptophan oxygenase gene in rat liver by glucocorticoids
.
J Biol Chem
1983
;
258
:
4750
3
.
28.
Salter
M
,
Pogson
CI.
The role of haem in the regulation of rat liver tryptophan metabolism
.
Biochem J
1986
;
240
:
259
63
.
29.
Lewis-Ballester
A
,
Forouhar
F
,
Kim
SM
,
Lew
S
,
Wang
Y
,
Karkashon
S
, et al
Molecular basis for catalysis and substrate-mediated cellular stabilization of human tryptophan 2,3-dioxygenase
.
Sci Rep
2016
;
6
:
35169
.
30.
Suzuki
S
,
Tone
S
,
Takikawa
O
,
Kubo
T
,
Kohno
I
,
Minatogawa
Y
. 
Expression of indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase in early concepti
.
Biochem J
2001
;
355
:
425
9
.
31.
Tatsumi
K
,
Higuchi
T
,
Fujiwara
H
,
Nakayama
T
,
Egawa
H
,
Itoh
K
, et al
Induction of tryptophan 2,3-dioxygenase in the mouse endometrium during implantation
.
Biochem Biophys Res Commun
2000
;
274
:
166
70
.
32.
Li
DD
,
Gao
YJ
,
Tian
XC
,
Yang
ZQ
,
Cao
H
,
Zhang
QL
, et al
Differential expression and regulation of Tdo2 during mouse decidualization
.
J Endocrinol
2014
;
220
:
73
83
.
33.
Yu
CP
,
Pan
ZZ
,
Luo
DY
. 
TDO as a therapeutic target in brain diseases
.
Metab Brain Dis
2016
;
31
:
737
47
.
34.
Pilotte
L
,
Larrieu
P
,
Stroobant
V
,
Colau
D
,
Dolusic
E
,
Frederick
R
, et al
Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase
.
Proc Natl Acad Sci U S A
2012
;
109
:
2497
502
.
35.
Dolusic
E
,
Larrieu
P
,
Moineaux
L
,
Stroobant
V
,
Pilotte
L
,
Colau
D
, et al
Tryptophan 2,3-dioxygenase (TDO) inhibitors. 3-(2-(pyridyl)ethenyl)indoles as potential anticancer immunomodulators
.
J Med Chem
2011
;
54
:
5320
34
.
36.
Schramme
F
,
Crosignani
S
,
Frederix
K
,
Hoffmann
D
,
Pilotte
L
,
Stroobant
V
, et al
Inhibition of tryptophan-dioxygenase activity increases the antitumor efficacy of immune checkpoint inhibitors
.
Cancer Immunol Res
2020
;
8
:
32
45
.
37.
Nizet
Y
,
Gillet
L
,
Schroeder
H
,
Lecuivre
C
,
Louahed
J
,
Renauld
JC
, et al
Antibody production by injection of living cells expressing non self antigens as cell surface type II transmembrane fusion protein
.
J Immunol Methods
2011
;
367
:
70
7
.
38.
Van Pel
A
,
De Plaen
E
,
Boon
T
. 
Selection of highly transfectable variant from mouse mastocytoma P815
.
Somat Cell Mol Genet
1985
;
11
:
467
75
.
39.
Badawy
AA.
Kynurenine pathway of tryptophan metabolism: regulatory and functional aspects
.
Int J Tryptophan Res
2017
;
10
:
1178646917691938
.
40.
Armulik
A
,
Abramsson
A
,
Betsholtz
C
. 
Endothelial/pericyte interactions
.
Circ Res
2005
;
97
:
512
23
.
41.
De Bock
K
,
De Smet
F
,
Leite De Oliveira
R
,
Anthonis
K
,
Carmeliet
P
. 
Endothelial oxygen sensors regulate tumor vessel abnormalization by instructing phalanx endothelial cells
.
J Mol Med (Berl)
2009
;
87
:
561
9
.
42.
Nagy
JA
,
Chang
SH
,
Dvorak
AM
,
Dvorak
HF
. 
Why are tumour blood vessels abnormal and why is it important to know?
Br J Cancer
2009
;
100
:
865
9
.
43.
Liu
T
,
Zhou
L
,
Li
D
,
Andl
T
,
Zhang
Y
. 
Cancer-associated fibroblasts build and secure the tumor microenvironment
.
Front Cell Dev Biol
2019
;
7
:
60
.
44.
Han
S
,
Zhang
C
,
Li
Q
,
Dong
J
,
Liu
Y
,
Huang
Y
, et al
Tumour-infiltrating CD4(+) and CD8(+) lymphocytes as predictors of clinical outcome in glioma
.
Br J Cancer
2014
;
110
:
2560
8
.
45.
Banerjee
T
,
Duhadaway
JB
,
Gaspari
P
,
Sutanto-Ward
E
,
Munn
DH
,
Mellor
AL
, et al
A key in vivo antitumor mechanism of action of natural product-based brassinins is inhibition of indoleamine 2,3-dioxygenase
.
Oncogene
2008
;
27
:
2851
7
.
46.
Muller
AJ
,
DuHadaway
JB
,
Jaller
D
,
Curtis
P
,
Metz
R
,
Prendergast
GC
. 
Immunotherapeutic suppression of indoleamine 2,3-dioxygenase and tumor growth with ethyl pyruvate
.
Cancer Res
2010
;
70
:
1845
53
.
47.
Godin-Ethier
J
,
Hanafi
LA
,
Piccirillo
CA
,
Lapointe
R
. 
Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives
.
Clin Cancer Res
2011
;
17
:
6985
91
.
48.
Schmidt
SK
,
Muller
A
,
Heseler
K
,
Woite
C
,
Spekker
K
,
MacKenzie
CR
, et al
Antimicrobial and immunoregulatory properties of human tryptophan 2,3-dioxygenase
.
Eur J Immunol
2009
;
39
:
2755
64
.
49.
Bessede
A
,
Gargaro
M
,
Pallotta
MT
,
Matino
D
,
Servillo
G
,
Brunacci
C
, et al
Aryl hydrocarbon receptor control of a disease tolerance defence pathway
.
Nature
2014
;
511
:
184
90
.
50.
Xu
F
,
Jin
T
,
Zhu
Y
,
Dai
C
. 
Immune checkpoint therapy in liver cancer
.
J Exp Clin Cancer Res
2018
;
37
:
110
.
51.
Finkelmeier
F
,
Waidmann
O
,
Trojan
J
. 
Nivolumab for the treatment of hepatocellular carcinoma
.
Expert Rev Anticancer Ther
2018
;
18
:
1169
75
.
52.
Bose
A
,
Barik
S
,
Banerjee
S
,
Ghosh
T
,
Mallick
A
,
Bhattacharyya Majumdar
S
, et al
Tumor-derived vascular pericytes anergize Th cells
.
J Immunol
2013
;
191
:
971
81
.
53.
Ochs
K
,
Sahm
F
,
Opitz
CA
,
Lanz
TV
,
Oezen
I
,
Couraud
PO
, et al
Immature mesenchymal stem cell-like pericytes as mediators of immunosuppression in human malignant glioma
.
J Neuroimmunol
2013
;
265
:
106
16
.
54.
Wang
Y
,
Liu
H
,
McKenzie
G
,
Witting
PK
,
Stasch
JP
,
Hahn
M
, et al
Kynurenine is an endothelium-derived relaxing factor produced during inflammation
.
Nat Med
2010
;
16
:
279
85
.
55.
Tattevin
P
,
Monnier
D
,
Tribut
O
,
Dulong
J
,
Bescher
N
,
Mourcin
F
, et al
Enhanced indoleamine 2,3-dioxygenase activity in patients with severe sepsis and septic shock
.
J Infect Dis
2010
;
201
:
956
66
.
56.
Smith
C
,
Chang
MY
,
Parker
KH
,
Beury
DW
,
DuHadaway
JB
,
Flick
HE
, et al
IDO is a nodal pathogenic driver of lung cancer and metastasis development
.
Cancer Discov
2012
;
2
:
722
35
.
57.
Mondal
A
,
Smith
C
,
DuHadaway
JB
,
Sutanto-Ward
E
,
Prendergast
GC
,
Bravo-Nuevo
A
, et al
IDO1 is an integral mediator of inflammatory neovascularization
.
EBioMedicine
2016
;
14
:
74
82
.
58.
Stanley
CP
,
Maghzal
GJ
,
Ayer
A
,
Talib
J
,
Giltrap
AM
,
Shengule
S
, et al
Singlet molecular oxygen regulates vascular tone and blood pressure in inflammation
.
Nature
2019
;
566
:
548
52
.
59.
Germeyer
A
,
Sharkey
AM
,
Prasadajudio
M
,
Sherwin
R
,
Moffett
A
,
Bieback
K
, et al
Paracrine effects of uterine leucocytes on gene expression of human uterine stromal fibroblasts
.
Mol Hum Reprod
2009
;
15
:
39
48
.
60.
Hsu
YL
,
Hung
JY
,
Chiang
SY
,
Jian
SF
,
Wu
CY
,
Lin
YS
, et al
Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis
.
Oncotarget
2016
;
7
:
27584
98
.