Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 44
1.
Cancers (Basel) ; 15(23)2023 Nov 28.
Article En | MEDLINE | ID: mdl-38067325

Several types of cancer spread through the lymphatic system via the sentinel lymph nodes (LNs). Such LN-draining primary tumors, modified by tumor factors, lead to the formation of a metastatic niche associated with an increased number of Foxp3+ regulatory T cells (Tregs). These cells are expected to contribute to the elaboration of an immune-suppressive environment. Activated Tregs express glycoprotein A repetitions predominant (GARP), which binds and presents latent transforming growth factor beta 1 (TGF-ß1) at their surface. GARP is also expressed by other non-immune cell types poorly described in LNs. Here, we mapped GARP expression in non-immune cells in human and mouse metastatic LNs. The mining of available (human and murine) scRNA-Seq datasets revealed GARP expression by blood (BEC)/lymphatic (LEC) endothelial, fibroblastic, and perivascular cells. Consistently, through immunostaining and in situ RNA hybridization approaches, GARP was detected in and around blood and lymphatic vessels, in (αSMA+) fibroblasts, and in perivascular cells associated with an abundant matrix. Strikingly, GARP was detected in LECs forming the subcapsular sinus and high endothelial venules (HEVs), two vascular structures localized at the interface between LNs and the afferent lymphatic and blood vessels. Altogether, we here provide the first distribution maps for GARP in human and murine LNs.

2.
Blood ; 141(5): 490-502, 2023 02 02.
Article En | MEDLINE | ID: mdl-36322928

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by the clonal expansion of myeloid cells, notably megakaryocytes (MKs), and an aberrant cytokine production leading to bone marrow (BM) fibrosis and insufficiency. Current treatment options are limited. TGF-ß1, a profibrotic and immunosuppressive cytokine, is involved in PMF pathogenesis. While all cell types secrete inactive, latent TGF-ß1, only a few activate the cytokine via cell type-specific mechanisms. The cellular source of the active TGF-ß1 implicated in PMF is not known. Transmembrane protein GARP binds and activates latent TGF-ß1 on the surface of regulatory T lymphocytes (Tregs) and MKs or platelets. Here, we found an increased expression of GARP in the BM and spleen of mice with PMF and tested the therapeutic potential of a monoclonal antibody (mAb) that blocks TGF-ß1 activation by GARP-expressing cells. GARP:TGF-ß1 blockade reduced not only fibrosis but also the clonal expansion of transformed cells. Using mice carrying a genetic deletion of Garp in either Tregs or MKs, we found that the therapeutic effects of GARP:TGF-ß1 blockade in PMF imply targeting GARP on Tregs. These therapeutic effects, accompanied by increased IFN-γ signals in the spleen, were lost upon CD8 T-cell depletion. Our results suggest that the selective blockade of TGF-ß1 activation by GARP-expressing Tregs increases a CD8 T-cell-mediated immune reaction that limits transformed cell expansion, providing a novel approach that could be tested to treat patients with myeloproliferative neoplasms.


Primary Myelofibrosis , Transforming Growth Factor beta1 , Mice , Animals , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/metabolism , Cytokines/metabolism , Fibrosis , T-Lymphocytes, Regulatory
3.
Front Immunol ; 12: 704050, 2021.
Article En | MEDLINE | ID: mdl-34386010

When combined with anti-PD-1, monoclonal antibodies (mAbs) against GARP:TGF-ß1 complexes induced more frequent immune-mediated rejections of CT26 and MC38 murine tumors than anti-PD-1 alone. In both types of tumors, the activity of anti-GARP:TGF-ß1 mAbs resulted from blocking active TGF-ß1 production and immunosuppression by GARP-expressing regulatory T cells. In CT26 tumors, combined GARP:TGF-ß1/PD-1 blockade did not augment the infiltration of T cells, but did increase the effector functions of already present anti-tumor T cells. Here we show that, in contrast, in MC38, combined GARP:TGF-ß1/PD-1 blockade increased infiltration of T cells, as a result of increased extravasation of T cells from blood vessels. Unexpectedly, combined GARP:TGF-ß1/PD-1 blockade also increased the density of GARP+ blood vessels covered by pericytes in MC38, but not in CT26 tumors. This appears to occur because anti-GARP:TGF-ß1, by blocking TGF-ß1 signals, favors the proliferation of and expression of adhesion molecules such as E-selectin by blood endothelial cells. The resulting densification of intratumoral blood vasculature probably contributes to increased T cell infiltration and to the therapeutic efficacy of GARP:TGF-ß1/PD-1 blockade in MC38. We conclude from these distinct observations in MC38 and CT26, that the combined blockades of GARP:TGF-ß1 and PD-1 can exert anti-tumor activity via multiple mechanisms, including the densification and normalization of intratumoral blood vasculature, the increase of T cell infiltration into the tumor and the increase of the effector functions of intratumoral tumor-specific T cells. This may prove important for the selection of cancer patients who could benefit from combined GARP:TGF-ß1/PD-1 blockade in the clinics.


Antineoplastic Agents, Immunological , Blood Vessels/immunology , Membrane Proteins , Neoplasms, Experimental , Neovascularization, Pathologic , Pericytes/immunology , Programmed Cell Death 1 Receptor , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1 , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/immunology
4.
Nat Commun ; 11(1): 4545, 2020 09 11.
Article En | MEDLINE | ID: mdl-32917858

TGF-ß1, ß2 and ß3 bind a common receptor to exert vastly diverse effects in cancer, supporting either tumor progression by favoring metastases and inhibiting anti-tumor immunity, or tumor suppression by inhibiting malignant cell proliferation. Global TGF-ß inhibition thus bears the risk of undesired tumor-promoting effects. We show that selective blockade of TGF-ß1 production by Tregs with antibodies against GARP:TGF-ß1 complexes induces regressions of mouse tumors otherwise resistant to anti-PD-1 immunotherapy. Effects of combined GARP:TGF-ß1/PD-1 blockade are immune-mediated, do not require FcγR-dependent functions and increase effector functions of anti-tumor CD8+ T cells without augmenting immune cell infiltration or depleting Tregs within tumors. We find GARP-expressing Tregs and evidence that they produce TGF-ß1 in one third of human melanoma metastases. Our results suggest that anti-GARP:TGF-ß1 mAbs, by selectively blocking a single TGF-ß isoform emanating from a restricted cellular source exerting tumor-promoting activity, may overcome resistance to PD-1/PD-L1 blockade in patients with cancer.


Antineoplastic Agents, Immunological/pharmacology , Drug Resistance, Neoplasm/drug effects , Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/immunology , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism
5.
Nat Commun ; 11(1): 2408, 2020 05 15.
Article En | MEDLINE | ID: mdl-32415113

Uveal melanoma (UM) is the most common intraocular tumour in adults and despite surgical or radiation treatment of primary tumours, ~50% of patients progress to metastatic disease. Therapeutic options for metastatic UM are limited, with clinical trials having little impact. Here we perform whole-genome sequencing (WGS) of 103 UM from all sites of the uveal tract (choroid, ciliary body, iris). While most UM have low tumour mutation burden (TMB), two subsets with high TMB are seen; one driven by germline MBD4 mutation, and another by ultraviolet radiation (UVR) exposure, which is restricted to iris UM. All but one tumour have a known UM driver gene mutation (GNAQ, GNA11, BAP1, PLCB4, CYSLTR2, SF3B1, EIF1AX). We identify three other significantly mutated genes (TP53, RPL5 and CENPE).


Iris Neoplasms/genetics , Iris Neoplasms/pathology , Melanoma/genetics , Melanoma/pathology , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Cell Line, Tumor , Chromosome Aberrations , Computational Biology , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Gene Dosage , Genome, Human , Genomics , Humans , Kaplan-Meier Estimate , Markov Chains , Melanocytes/metabolism , Mutation , Phenotype , Prognosis , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
6.
Cancer Immunol Res ; 8(1): 19-31, 2020 01.
Article En | MEDLINE | ID: mdl-31806639

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 PDGFRß 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.


Antibodies, Monoclonal/immunology , Biomarkers, Tumor/metabolism , Lung Diseases/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Pericytes/pathology , Tryptophan Oxygenase/metabolism , Animals , Antibodies, Monoclonal/isolation & purification , Antibody Formation , Cell Line, Tumor , Humans , Lung Diseases/immunology , Lung Diseases/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Grading , Neoplasms/immunology , Neoplasms/pathology , Pericytes/metabolism , Tryptophan/metabolism , Tryptophan Oxygenase/immunology
7.
Leuk Lymphoma ; 60(4): 1043-1052, 2019 04.
Article En | MEDLINE | ID: mdl-30277098

High-grade B-cell lymphomas with MYC and BCL2 or BCL6 rearrangements are highly aggressive B-cell lymphomas called double-hit lymphomas (HGBL-DH). They are particularly refractory to standard treatments and carry a poor prognosis. Fragments of resected tumoral lymph nodes from two HGBL-DH patients were put in culture. Continuously proliferating cells were characterized and compared with the original tumors. In both cases, the proliferating cells and the tumor displayed MYC and BCL2 rearrangements. Both cell lines (called LB5848-LYMP and LB5871-LYMP) presented a high proliferation rate and were maintained in culture for more than one year. Upon injection in immunodeficient mice, LB5848-LYMP gave rise to lymphoid tumors. In vitro treatment of these cell lines with a BCL2-inhibitory drug (ABT-199) selectively stopped their proliferation. These new cell lines represent valuable tools for studying HGBL-DH and for the in vitro testing of candidate therapies targeting HGBL-DH. LB5848-LYMP is also suitable for similar experiments in vivo.


Biomarkers, Tumor , Gene Rearrangement , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Aged , Animals , Apoptosis/genetics , Biopsy , Cell Line , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Genetic Predisposition to Disease , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Karyotyping , Lymphoma, B-Cell/pathology , Male , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Xenograft Model Antitumor Assays
8.
Nat Commun ; 8(1): 1404, 2017 11 10.
Article En | MEDLINE | ID: mdl-29123081

Despite impressive clinical success, cancer immunotherapy based on immune checkpoint blockade remains ineffective in many patients due to tumoral resistance. Here we use the autochthonous TiRP melanoma model, which recapitulates the tumoral resistance signature observed in human melanomas. TiRP tumors resist immunotherapy based on checkpoint blockade, cancer vaccines or adoptive T-cell therapy. TiRP tumors recruit and activate tumor-specific CD8+ T cells, but these cells then undergo apoptosis. This does not occur with isogenic transplanted tumors, which are rejected after adoptive T-cell therapy. Apoptosis of tumor-infiltrating lymphocytes can be prevented by interrupting the Fas/Fas-ligand axis, and is triggered by polymorphonuclear-myeloid-derived suppressor cells, which express high levels of Fas-ligand and are enriched in TiRP tumors. Blocking Fas-ligand increases the anti-tumor efficacy of adoptive T-cell therapy in TiRP tumors, and increases the efficacy of checkpoint blockade in transplanted tumors. Therefore, tumor-infiltrating lymphocytes apoptosis is a relevant mechanism of immunotherapy resistance, which could be blocked by interfering with the Fas/Fas-ligand pathway.


Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Apoptosis/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Humans , Immunotherapy/methods , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Tumor Microenvironment/immunology , fas Receptor/antagonists & inhibitors , fas Receptor/immunology
9.
Cell Rep ; 19(12): 2529-2543, 2017 06 20.
Article En | MEDLINE | ID: mdl-28636941

Unlimited replicative potential is one of the hallmarks of cancer cells. In melanoma, hTERT (telomerase reverse transcriptase) is frequently overexpressed because of activating mutations in its promoter, suggesting that telomerase is necessary for melanoma development. We observed, however, that a subset of melanoma metastases and derived cell lines had no telomere maintenance mechanism. Early passages of the latter displayed long telomeres that progressively shortened and fused before cell death. We propose that, during melanoma formation, oncogenic mutations occur in precursor melanocytes with long telomeres, providing cells with sufficient replicative potential, thereby bypassing the need to re-activate telomerase. Our data further support the emerging idea that long telomeres promote melanoma formation. These observations are important when considering anticancer therapies targeting telomerase.


Melanoma/secondary , Skin Neoplasms/pathology , Telomere Homeostasis , Telomere/metabolism , Adult , Aged , Animals , Cell Line, Tumor , Female , Humans , Lymphatic Metastasis , Male , Melanoma/genetics , Melanoma/metabolism , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Telomerase/metabolism , Young Adult
10.
J Transl Med ; 14(1): 232, 2016 08 02.
Article En | MEDLINE | ID: mdl-27484791

BACKGROUND: Ipilimumab (Ipi) improves the survival of advanced melanoma patients with an incremental long-term benefit in 10-15 % of patients. A tumor signature that correlates with this survival benefit could help optimizing individualized treatment strategies. METHODS: Freshly frozen melanoma metastases were collected from patients treated with either Ipi alone (n: 7) or Ipi combined with a dendritic cell vaccine (TriMixDC-MEL) (n: 11). Samples were profiled by immunohistochemistry (IHC), whole transcriptome (RNA-seq) and methyl-DNA sequencing (MBD-seq). RESULTS: Patients were divided in two groups according to clinical evolution: durable benefit (DB; 5 patients) and no clinical benefit (NB; 13 patients). 20 metastases were profiled by IHC and 12 were profiled by RNA- and MBD-seq. 325 genes were identified as differentially expressed between DB and NB. Many of these genes reflected a humoral and cellular immune response. MBD-seq revealed differences between DB and NB patients in the methylation of genes linked to nervous system development and neuron differentiation. DB tumors were more infiltrated by CD8(+) and PD-L1(+) cells than NB tumors. B cells (CD20(+)) and macrophages (CD163(+)) co-localized with T cells. Focal loss of HLA class I and TAP-1 expression was observed in several NB samples. CONCLUSION: Combined analyses of melanoma metastases with IHC, gene expression and methylation profiling can potentially identify durable responders to Ipi-based immunotherapy.


Antibodies, Monoclonal/therapeutic use , Epigenesis, Genetic , Immunotherapy , Melanoma/genetics , Melanoma/therapy , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Tumor Microenvironment/immunology , Adult , Aged , Antibodies, Monoclonal/pharmacology , DNA Methylation/drug effects , DNA Methylation/genetics , Demography , Epigenesis, Genetic/drug effects , Female , Frozen Sections , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Ipilimumab , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Paraffin Embedding , Remission Induction , Skin Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
11.
J Clin Oncol ; 34(12): 1330-8, 2016 Apr 20.
Article En | MEDLINE | ID: mdl-26926680

PURPOSE: Autologous monocyte-derived dendritic cells (DCs) electroporated with synthetic mRNA (TriMixDC-MEL) are immunogenic and have antitumor activity as a monotherapy in patients with pretreated advanced melanoma. Ipilimumab, an immunoglobulin G1 monoclonal antibody directed against the cytotoxic T-lymphocyte-associated protein 4 receptor that counteracts physiologic suppression of T-cell function, improves the overall survival of patients with advanced melanoma. This phase II study investigated the combination of TriMixDC-MEL and ipilimumab in patients with pretreated advanced melanoma. PATIENTS AND METHODS: Thirty-nine patients were treated with TriMixDC-MEL (4 × 10(6) cells administered intradermally and 20 × 10(6) cells administered intravenously) plus ipilimumab (10 mg/kg every 3 weeks for a total of four administrations, followed by maintenance therapy every 12 weeks in patients who remained progression free). Six-month disease control rate according to the immune-related response criteria served as the primary end point. RESULTS: The 6-month disease control rate was 51% (95% CI, 36% to 67%), and the overall tumor response rate was 38% (including eight complete and seven partial responses). Seven complete responses and one partial tumor response are ongoing after a median follow-up time of 36 months (range, 22 to 43 months). The most common treatment-related adverse events (all grades) consisted of local DC injection site skin reactions (100%), transient post-DC infusion chills (38%) and flu-like symptoms (84%), dermatitis (64%), hepatitis (13%), hypophysitis (15%), and diarrhea/colitis (15%). Grade 3 or 4 immune-related adverse events occurred in 36% of patients. There was no grade 5 adverse event. CONCLUSION: The combination of TriMixDC-MEL and ipilimumab is tolerable and results in an encouraging rate of highly durable tumor responses in patients with pretreated advanced melanoma.


Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Dendritic Cells/transplantation , Electroporation , Genetic Therapy/methods , Melanoma/therapy , RNA, Messenger/genetics , Skin Neoplasms/therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cells, Cultured , Chemotherapy, Adjuvant , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Progression , Disease-Free Survival , Drug Administration Schedule , Female , Genetic Therapy/adverse effects , Genetic Therapy/mortality , Humans , Ipilimumab , Kaplan-Meier Estimate , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/mortality , Middle Aged , Proportional Hazards Models , RNA, Messenger/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Time Factors , Transplantation, Autologous , Treatment Outcome , Young Adult
12.
Mod Pathol ; 29(4): 347-58, 2016 Apr.
Article En | MEDLINE | ID: mdl-26867783

Melanoma is not only one of the most immunogenic cancers but also one of the most effective cancers at subverting host immunity. The role of T lymphocytes in tumor immunity has been extensively studied in melanoma, whereas less is known about the importance of B lymphocytes. The effects of plasma cells (PCs), in particular, are still obscure. The aim of this study was to characterize pathological features and clinical outcome of primary cutaneous melanomas associated with PCs. Moreover, we investigated the origins of the melanoma-associated PCs. Finally, we studied the outcome of patients with primary melanomas with PCs. We reviewed 710 melanomas to correlate the presence of PCs with histological prognostic markers. Immunohistochemistry for CD138 and heavy and light chains was performed in primary melanomas (PM) and in loco-regional lymph nodes (LN), both metastatic and not metastatic. In three PM and nine LN with frozen material, VDJ-rearrangement was analyzed by Gene Scan Analysis. Survival analysis was performed on a group of 85 primary melanomas >2 mm in thickness. Forty-one cases (3.7%) showed clusters/sheets of PCs. PC-rich melanomas occurred at an older age and were thicker, more often ulcerated and more mitotically active (P<0.05). PCs were polyclonal and often expressed IgA in addition to IgG. In LN, clusters/sheets of IgA+ PCs were found both in the sinuses and subcapsular areas. Analysis of VDJ-rearrangements showed the IgA to be oligoclonal. Melanomas with clusters/sheets of PCs had a significantly worse survival compared with melanomas without PCs while, interestingly, melanomas with sparse PCs were associated with a better clinical outcome (P=0.002). In conclusion, melanomas with sheets/clusters of PCs are associated with worse prognosis. IgG and IgA are the isotypes predominantly produced by these PCs. IgA oligoclonality suggests an antigen-driven response that facilitates melanoma progression by a hitherto unknown mechanism.


Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Plasma Cells/immunology , Skin Neoplasms/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Immunoglobulin A , Immunohistochemistry , Kaplan-Meier Estimate , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Young Adult
13.
Cancer Immunol Res ; 3(9): 978-85, 2015 Sep.
Article En | MEDLINE | ID: mdl-26269528

Cancer patients mount T-lymphocyte responses against antigens expressed selectively by their malignancy, but these responses often fail to control their disease, because tumors select mechanisms that allow them to resist immune destruction. Among the numerous resistance mechanisms that have been proposed, metabolic inhibition of T cells by tryptophan catabolism deserves particular attention, because of the frequent expression of tryptophan-degrading enzymes in human tumors, and because in vitro and in vivo studies have shown that their enzymatic activity can be readily blocked by pharmacologic inhibitors, thereby restoring T-cell-mediated tumor cell killing and paving the way to targeted therapeutic intervention. In view of recent observations, and taking into account the differences between human and mouse data that differ in several aspects, in this Cancer Immunology at the Crossroads article, we discuss the role of the three enzymes that have been proposed to control tryptophan catabolism in tumoral immune resistance: indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO), and indoleamine 2,3-dioxygenase 2 (IDO2).


Neoplasms/immunology , Tryptophan/metabolism , Tumor Escape/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/enzymology , Tryptophan Oxygenase/physiology
14.
Oncoimmunology ; 4(5): e1003012, 2015 May.
Article En | MEDLINE | ID: mdl-26155395

Tryptophan catabolism by indoleamine 2,3-dioxygenase (IDO1) is a physiological immunoregulatory mechanism often hijacked by tumors. Our recent extensive study of IDO1 protein expression in human tissues showed expression in mature dendritic cells and in pulmonary and placental endothelial cells. IDO1 was also expressed in 56% of tumors, either by tumoral, stromal, or endothelial cells. These results and reagent will guide the clinical development of IDO1 inhibitors for cancer therapy.

15.
Front Immunol ; 6: 34, 2015.
Article En | MEDLINE | ID: mdl-25691885

Tryptophan is required for T lymphocyte effector functions. Its degradation is one of the mechanisms selected by tumors to resist immune destruction. Two enzymes, tryptophan-2,3-dioxygenase and indoleamine 2,3-dioxygenase 1, control tryptophan degradation through the kynurenine pathway. A third protein, indoleamine 2,3-dioxygenase 2, was identified more recently. All three enzymes were reported to be expressed in tumors, and are candidate targets for pharmacological inhibition aimed at restoring effective anti-tumoral immunity. In this review, we compare these three enzymes in terms of structure, activity, regulation, and expression in healthy and cancerous tissues, in order to appreciate their relevance to tumoral immune resistance.

16.
Cancer Immunol Res ; 3(2): 161-72, 2015 Feb.
Article En | MEDLINE | ID: mdl-25271151

Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 (IDO1) plays a key role in tumoral resistance to immune rejection. In humans, constitutive expression of IDO1 has been observed in several tumor types. However, a comprehensive analysis of its expression in normal and tumor tissues is still required to anticipate the risks and potential benefits of IDO1 inhibitors. Using a newly validated monoclonal antibody to human IDO1, we performed an extensive immunohistochemical analysis of IDO1 expression in normal and tumor tissues. In normal tissues, IDO1 was expressed by endothelial cells in the placenta and lung and by epithelial cells in the female genital tract. In lymphoid tissues, IDO1 was expressed in mature dendritic cells with a phenotype (CD83(+), DC-LAMP(+), langerin(-), CD123(-), CD163(-)) distinct from plasmacytoid dendritic cells. Importantly, IDO1-expressing dendritic cells were not enriched in tumor-draining lymph nodes, in contrast with previously reported findings. IDO1-expressing cells were observed in a large fraction (505/866, 58%) of human tumors. They comprised tumor cells, endothelial cells, and stromal cells in proportions that varied depending on the tumor type. Tumors showing the highest proportions of IDO1-immunolabeled samples were carcinomas of the endometrium and cervix, followed by kidney, lung, and colon. This hierarchy of IDO1 expression was confirmed by gene expression data mined from The Cancer Genome Atlas database. Expression of IDO1 may be used to select tumors likely to benefit from targeted therapy with IDO1 inhibitors.


Biomarkers, Tumor/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Neoplasm Proteins/metabolism , Neoplasms/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/immunology , Genitalia, Female/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lung/immunology , Lymph Nodes/immunology , Lymphatic Metastasis , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Placenta/immunology , Pregnancy , RNA, Messenger/genetics
17.
Epigenetics ; 9(8): 1163-71, 2014 Aug.
Article En | MEDLINE | ID: mdl-25089631

Genome hypomethylation is a common epigenetic alteration in human tumors, where it often leads to aberrant activation of a group of germline-specific genes, commonly referred to as "cancer-germline" genes. The cellular functions and tumor promoting potential of these genes remain, however, largely uncertain. Here, we report identification of a novel cancer-germline transcript (CT-GABRA3) displaying DNA hypomethylation-dependent activation in various tumors, including melanoma and lung carcinoma. Importantly, CT-GABRA3 harbors a microRNA (miR-105), which has recently been identified as a promoter of cancer metastasis by its ability to weaken vascular endothelial barriers following exosomal secretion. CT-GABRA3 also carries a microRNA (miR-767) with predicted target sites in TET1 and TET3, two members of the ten-eleven-translocation family of tumor suppressor genes, which are involved in the conversion of 5-methylcytosines to 5-hydroxymethylcytosines (5hmC) in DNA. Decreased TET activity is a hallmark of cancer; here, we provide evidence that aberrant activation of miR-767 contributes to this phenomenon. We demonstrate that miR-767 represses TET1/3 mRNA and protein expression and regulates genomic 5hmC levels. Additionally, we show that high CT-GABRA3 transcription correlates with reduced TET1 mRNA levels in vivo in lung tumors. Together, our study identified a cancer-germline gene that produces microRNAs with oncogenic potential. Moreover, our data indicate that DNA hypomethylation in tumors can contribute to reduced 5hmC levels via activation of a TET-targeting microRNA.


DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , MicroRNAs/metabolism , Neoplasms/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Humans , Mixed Function Oxygenases , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
18.
Proc Natl Acad Sci U S A ; 111(9): 3502-7, 2014 Mar 04.
Article En | MEDLINE | ID: mdl-24550491

Female mice of inbred strain CBA do not reject syngeneic male skin grafts even though they mount a T-cell response against the male-specific HY antigen. We show that local immunostimulation performed by injecting cytokines and Toll-like receptor ligands in close vicinity to the graft causes rejection. We feel that this approach should be tested in tumor-bearing human patients in combination with antitumor vaccination. Relief of intratumor immunosuppression may increase considerably the fraction of patients who respond to vaccination directed against tumor antigens recognized by T cells.


Disease Models, Animal , Graft Rejection/chemically induced , Immune Tolerance/immunology , Immunization/methods , Neoplasms/therapy , Adoptive Transfer , Animals , Cytokines/adverse effects , Cytokines/immunology , DNA Primers/genetics , Female , Immunization/adverse effects , Male , Mice , Mice, Inbred CBA , Neoplasms/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Skin Transplantation/methods
19.
Cancer Discov ; 4(1): 80-93, 2014 Jan.
Article En | MEDLINE | ID: mdl-24265155

BRAF inhibitors elicit rapid antitumor responses in the majority of patients with BRAF(V600)-mutant melanoma, but acquired drug resistance is almost universal. We sought to identify the core resistance pathways and the extent of tumor heterogeneity during disease progression. We show that mitogen-activated protein kinase reactivation mechanisms were detected among 70% of disease-progressive tissues, with RAS mutations, mutant BRAF amplification, and alternative splicing being most common. We also detected PI3K-PTEN-AKT-upregulating genetic alterations among 22% of progressive melanomas. Distinct molecular lesions in both core drug escape pathways were commonly detected concurrently in the same tumor or among multiple tumors from the same patient. Beyond harboring extensively heterogeneous resistance mechanisms, melanoma regrowth emerging from BRAF inhibitor selection displayed branched evolution marked by altered mutational spectra/signatures and increased fitness. Thus, melanoma genomic heterogeneity contributes significantly to BRAF inhibitor treatment failure, implying upfront, cotargeting of two core pathways as an essential strategy for durable responses.


Drug Resistance, Neoplasm/physiology , Melanoma/genetics , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Clonal Evolution , Female , Humans , Imidazoles/therapeutic use , Indoles/therapeutic use , Male , Melanoma/drug therapy , Melanoma/metabolism , Middle Aged , Oximes/therapeutic use , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Sulfonamides/therapeutic use , Vemurafenib , ras Proteins/genetics
20.
Ann N Y Acad Sci ; 1284: 62-5, 2013 May.
Article En | MEDLINE | ID: mdl-23651195

A large body of evidence supports the idea that the tumor environment is immunosuppressive, notably for T lymphocytes. Yet, in some tumor types, the presence of tumor-infiltrating T cells has favorable prognostic value. In order to better understand the functional value of T cells in human tumors, we focused on cutaneous metastases of melanoma and observed that some of them host ectopic lymphoid structures in which B cell, and possibly T cell, responses take place. This observation is discussed in the context of current views on immunosuppression in tumors.


Adaptive Immunity , Melanoma/immunology , Melanoma/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Animals , Humans , Immunity, Humoral , Immunoglobulin A/immunology , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasm Metastasis , Prognosis
...