Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Uruguay Oncology Collection
Country/Region as subject
Publication year range
1.
Kidney Int ; 103(3): 627-637, 2023 03.
Article in English | MEDLINE | ID: mdl-36306921

ABSTRACT

Kidney transplant survival is shortened by chronic rejection and side effects of standard immunosuppressive drugs. Cell-based immunotherapy with tolerogenic dendritic cells has long been recognized as a promising approach to reduce general immunosuppression. Published trials report the safety and the absence of therapy-related adverse reactions in patients treated with tolerogenic dendritic cells suffering from several inflammatory diseases. Here, we present the first phase I clinical trial results using human autologous tolerogenic dendritic cells (ATDC) in kidney transplantation. Eight patients received ATDC the day before transplantation in conjunction with standard steroids, mycophenolate mofetil and tacrolimus immunosuppression with an option to taper mycophenolate mofetil. ATDC preparations were manufactured in a Good Manufacturing Practice-compliant facility and fulfilled cell count, viability, purity and identity criteria for release. A control group of nine patients received the same standard immunosuppression, except basiliximab induction replaced ATDC therapy and mycophenolate tapering was not allowed. During the three-year follow-up, no deaths occurred and there was 100% graft survival. No significant increase of adverse events was associated with ATDC infusion. Episodes of rejection were observed in two patients from the ATDC group and one patient from the control group. However, all rejections were successfully treated by glucocorticoids. Mycophenolate was successfully reduced/stopped in five patients from the ATDC group, allowing tacrolimus monotherapy for two of them. Regarding immune monitoring, reduced CD8 T cell activation markers and increased Foxp3 expression were observed in the ATDC group. Thus, our results demonstrate ATDC administration safety in kidney-transplant recipients.


Subject(s)
Kidney Transplantation , Tacrolimus , Humans , Tacrolimus/therapeutic use , Mycophenolic Acid/therapeutic use , Kidney Transplantation/adverse effects , Transplant Recipients , Immunosuppressive Agents/therapeutic use , Immunosuppression Therapy/methods , Dendritic Cells , Graft Rejection , Graft Survival
2.
J Immunol ; 207(2): 421-435, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34233909

ABSTRACT

Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Membrane Proteins/immunology , Animals , Endosomes/immunology , Female , Genes, MHC Class II/immunology , Golgi Apparatus/immunology , Immunity, Innate/immunology , Intestinal Mucosa/immunology , Ion Channels/immunology , Lymphocytes/immunology , Lysosomes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Tretinoin/immunology
3.
Cancer Immunol Immunother ; 68(4): 661-672, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30357490

ABSTRACT

Myeloid cells play a pivotal role in regulating innate and adaptive immune responses. In inflammation, autoimmunity, and after transplantation, myeloid cells have contrasting roles: on the one hand they initiate the immune response, promoting activation and expansion of effector T-cells, and on the other, they counter-regulate inflammation, maintain tissue homeostasis, and promote tolerance. The latter activities are mediated by several myeloid cells including polymorphonuclear neutrophils, macrophages, myeloid-derived suppressor cells, and dendritic cells. Since these cells have been associated with immune suppression and tolerance, they will be further referred to as myeloid regulatory cells (MRCs). In recent years, MRCs have emerged as a therapeutic target or have been regarded as a potential cellular therapeutic product for tolerance induction. However, several open questions must be addressed to enable the therapeutic application of MRCs including: how do they function at the site of inflammation, how to best target these cells to modulate their activities, and how to isolate or to generate pure populations for adoptive cell therapies. In this review, we will give an overview of the current knowledge on MRCs in inflammation, autoimmunity, and transplantation. We will discuss current strategies to target MRCs and to exploit their tolerogenic potential as a cell-based therapy.


Subject(s)
Autoimmunity , Homeostasis , Immune Tolerance , Inflammation/etiology , Inflammation/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , Biomarkers , Disease Susceptibility , Humans , Immunomodulation , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Organ Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation, Homologous
4.
Transpl Int ; 30(8): 754-764, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27864897

ABSTRACT

Although the occurrence of acute rejection was significantly reduced and the allograft survival at 1 year was massively improved by the development of pharmacological immunosuppressive drugs, little progress has been made regarding long-term graft survival. Cell therapy appears to be an innovative and promising strategy to minimize the use of immunosuppression in transplantation and consequently increases long-term graft survival. The strength of cell therapy is that it will induce graft-specific tolerance and not a general immunosuppression of the patients. Several candidates, such as tolerogenic dendritic cells, have been gaining interest as an efficient means of promoting antigen-specific tolerance over recent years. Studies performed in rodent models have demonstrated the feasibility and efficacy of tolerogenic dendritic cells for the induction of tolerance in transplantation. In parallel, protocols to generate human tolerogenic dendritic cells in vitro have been defined, and some phase I clinical trials in autoimmune diseases have been recently performed to evaluate the safety of tolerogenic dendritic cell therapy. In this review, we will focus on the potential therapeutic interest of these cells in transplantation as well as their generation and characterization in humans. Finally, we will describe our current clinical trial using autologous tolerogenic dendritic cells in transplantation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Animals , Cell Transplantation/methods , Cell- and Tissue-Based Therapy/methods , Clinical Trials as Topic , Dendritic Cells/drug effects , Graft Survival/immunology , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , Kidney Transplantation/methods , Models, Animal , Organ Transplantation , Transplantation Immunology , Transplantation Tolerance
5.
J Immunol ; 195(10): 5035-44, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26432892

ABSTRACT

Emerging knowledge regarding B cells in organ transplantation has demonstrated that these cells can no longer be taken as mere generators of deleterious Abs but can also act as beneficial players. We previously demonstrated in a rat model of cardiac allograft tolerance induced by short-term immunosuppression an accumulation in the blood of B cells overexpressing inhibitory molecules, a phenotype also observed in the blood of patients that spontaneously develop graft tolerance. In this study, we demonstrated the presence in the spleen of regulatory B cells enriched in the CD24(int)CD38(+)CD27(+)IgD(-)IgM(+/low) subpopulation, which are able to transfer donor-specific tolerance via IL-10 and TGF-ß1-dependent mechanisms and to suppress in vitro TNF-α secretion. Following anti-CD40 stimulation, IgD(-)IgM(+/low) B cells were blocked in their plasma cell differentiation pathway, maintained high expression of the inhibitory molecules CD23 and Bank1, and upregulated Granzyme B and Irf4, two molecules described as highly expressed by regulatory B cells. Interestingly, these B cells recognized specifically a dominant donor Ag, suggesting restricted specificity that could lead to a particular B cell response. Regulatory B cells were not required for induction of tolerance and appeared following Foxp3(+)CD4(+)CD25(+) regulatory T cells, suggesting cooperation with regulatory T cells for their expansion. Nevertheless, following transfer to new recipients, these B cells migrated to the allograft, kept their regulatory profile, and promoted local accumulation of Foxp3(+)CD4(+)CD25(+) regulatory T cells. Mechanisms of regulatory B cells and their cell therapy potential are important to decipher in experimental models to pave the way for future developments in the clinic.


Subject(s)
B-Lymphocytes, Regulatory/immunology , CD40 Antigens/immunology , Granzymes/immunology , Heart Transplantation , Plasma Cells/immunology , Signal Transduction/immunology , Transplantation Tolerance , Allografts , Animals , Antigens, CD/immunology , Cytokines/immunology , Isoantigens/immunology , Male , Rats , T-Lymphocytes, Regulatory/immunology
6.
J Pediatr Gastroenterol Nutr ; 65(3): e53-e59, 2017 09.
Article in English | MEDLINE | ID: mdl-28319600

ABSTRACT

As pediatric liver transplantation comes of age, experts gathered to discuss current paradigms and define gaps in knowledge warranting research to further improve patient and graft outcomes. Identified areas ripe for collaborative research include understanding the molecular and cellular mechanisms of tolerance and the role of donor-specific antibodies, considering ways to expand donor pool, minimizing long-term side effects of immunosuppression, and fine-tuning surgical techniques to minimize biliary and vascular complications.


Subject(s)
Liver Transplantation , Child , Drug Administration Schedule , Graft Rejection/diagnosis , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Humans , Immunosuppressive Agents/therapeutic use , Outcome Assessment, Health Care , Pediatrics , Postoperative Complications/diagnosis , Postoperative Complications/therapy , Quality of Life , Tissue and Organ Procurement/methods
7.
J Immunol ; 193(9): 4696-703, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25252962

ABSTRACT

Cell therapy and the use of mAbs that interfere with T cell effector functions constitute promising approaches for the control of allograft rejection. In the current study, we investigated a novel approach combining administration of autologous tolerogenic dendritic cells with short-term treatment with CD3-specific Abs. Permanent acceptance of pancreatic islet allografts was achieved in mice treated with the combination therapy the day before transplantation but not in recipients treated with either therapy alone. The combination treatment induced a marked decrease in T cells infiltrating the allografts and a sustained reduction of antidonor responses. Importantly, CD4(+)Foxp3(+) regulatory T cells appeared to play a crucial role in the long-term graft acceptance. Their frequency increased significantly in the spleen, draining lymph nodes, and transplanted islets and remained elevated over the long term; they exhibited increased donor-specific suppressive functions; and their removal at the time of transplantation abrogated the therapeutic effect of the combined therapy. These results support the therapeutic potential of protocols combining autologous dendritic cells and low-dose CD3 Abs, both currently in clinical development, and that act in synergy to control allogeneic immune responses and favor graft survival in a full-mismatch situation.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD3 Complex/metabolism , Dendritic Cells/immunology , Dendritic Cells/transplantation , Islets of Langerhans Transplantation , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , Epitopes/immunology , Graft Survival/drug effects , Graft Survival/immunology , Immunomodulation/drug effects , Immunomodulation/immunology , Islets of Langerhans Transplantation/methods , Mice , Models, Animal , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Transplantation, Autologous
8.
Eur J Immunol ; 42(11): 2881-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22865279

ABSTRACT

Galectin-1 (Gal-1) is a member of a family of endogenous ß-galactose-binding proteins with a role in preventing autoimmune diseases and chronic inflammation. In this study, the involvement of Gal-1 in graft rejection was investigated by using Gal-1-deficient mice (Gal-1⁻/⁻). We demonstrate that in the absence of Gal-1, skin grafts are rejected earlier compared with those of WT mice, and that this is due to the role played by CD8⁺ T cells in graft rejection. The difference in graft survival observed between Gal-1⁻/⁻ and WT mice was explained by both an increase in the percentage of antigen-specific CD8+ T cells and by preferential secretion of IFN-γ and IL-17 by CD8⁺ T cells in Gal-1⁻/⁻ mice compared with WT mice. This study suggests that endogenous expression of Gal-1 contributes to graft survival. The results obtained from the use of mice deficient in Gal-1 also confirm a key role for CD8⁺ T cells in graft rejection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Galectin 1/immunology , Graft Rejection/immunology , Skin Transplantation/immunology , Animals , Female , Flow Cytometry , Interferon-gamma/immunology , Interleukin-17/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Spleen/immunology
9.
J Immunol ; 185(2): 823-33, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543104

ABSTRACT

Despite accumulating evidence for the importance of allospecific CD8(+) regulatory T cells (Tregs) in tolerant rodents and free immunosuppression transplant recipients, mechanisms underlying CD8(+) Treg-mediated tolerance remain unclear. By using a model of transplantation tolerance mediated by CD8(+) Tregs following CD40Ig treatment in rats, in this study, we show that the accumulation of tolerogenic CD8(+) Tregs and plasmacytoid dendritic cells (pDCs) in allograft and spleen but not lymph nodes was associated with tolerance induction in vascularized allograft recipients. pDCs preferentially induced tolerogenic CD8(+) Tregs to suppress CD4(+) effector cells responses to first-donor Ags in vitro. When tolerogenic CD8(+) Tregs were not in contact with CD4(+) effector cells, suppression was mediated by IDO. Contact with CD4(+) effector cells resulted in alternative suppressive mechanisms implicating IFN-gamma and fibroleukin-2. In vivo, both IDO and IFN-gamma were involved in tolerance induction, suggesting that contact with CD4(+) effector cells is crucial to modulate CD8(+) Tregs function in vivo. In conclusion, CD8(+) Tregs and pDCs interactions were necessary for suppression of CD4(+) T cells and involved different mechanisms modulated by the presence of cell contact between CD8(+) Tregs, pDCs, and CD4(+) effector cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Heart Transplantation/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Adenoviridae/genetics , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/immunology , Flow Cytometry , Genetic Vectors/genetics , Heart Transplantation/methods , Male , Models, Animal , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , Transduction, Genetic , Transplantation, Homologous
10.
MAbs ; 14(1): 2095949, 2022.
Article in English | MEDLINE | ID: mdl-35867844

ABSTRACT

Cluster of differentiation 38 (CD38) is an ecto-enzyme expressed primarily on immune cells that metabolize nicotinamide adenine dinucleotide (NAD+) to adenosine diphosphate ribose or cyclic ADP-ribose and nicotinamide. Other substrates of CD38 include nicotinamide adenine dinucleotide phosphate and nicotinamide mononucleotide, a critical NAD+ precursor in the salvage pathway. NAD+ is an important coenzyme involved in several metabolic pathways and is a required cofactor for the function of sirtuins (SIRTs) and poly (adenosine diphosphate-ribose) polymerases. Declines in NAD+ levels are associated with metabolic and inflammatory diseases, aging, and neurodegenerative disorders. To inhibit CD38 enzyme activity and boost NAD+ levels, we developed TNB-738, an anti-CD38 biparatopic antibody that pairs two non-competing heavy chain-only antibodies in a bispecific format. By simultaneously binding two distinct epitopes on CD38, TNB-738 potently inhibited its enzymatic activity, which in turn boosted intracellular NAD+ levels and SIRT activities. Due to its silenced IgG4 Fc, TNB-738 did not deplete CD38-expressing cells, in contrast to the clinically available anti-CD38 antibodies, daratumumab, and isatuximab. TNB-738 offers numerous advantages compared to other NAD-boosting therapeutics, including small molecules, and supplements, due to its long half-life, specificity, safety profile, and activity. Overall, TNB-738 represents a novel treatment with broad therapeutic potential for metabolic and inflammatory diseases associated with NAD+ deficiencies.Abbreviations: 7-AAD: 7-aminoactinomycin D; ADCC: antibody dependent cell-mediated cytotoxicity; ADCP: antibody dependent cell-mediated phagocytosis; ADPR: adenosine diphosphate ribose; APC: allophycocyanin; cADPR: cyclic ADP-ribose; cDNA: complementary DNA; BSA: bovine serum albumin; CD38: cluster of differentiation 38; CDC: complement dependent cytotoxicity; CFA: Freund's complete adjuvant; CHO: Chinese hamster ovary; CCP4: collaborative computational project, number 4; COOT: crystallographic object-oriented toolkit; DAPI: 4',6-diamidino-2-phenylindole; DNA: deoxyribonucleic acid; DSC: differential scanning calorimetry; 3D: three dimensional; εNAD+: nicotinamide 1,N6-ethenoadenine dinucleotide; ECD: extracellular domain; EGF: epidermal growth factor; FACS: fluorescence activated cell sorting; FcγR: Fc gamma receptors; FITC: fluorescein isothiocyanate; HEK: human embryonic kidney; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IgG: immunoglobulin; IFA: incomplete Freund's adjuvant; IFNγ: Interferon gamma; KB: kinetic buffer; kDa: kilodalton; KEGG: kyoto encyclopedia of genes and genomes; LDH: lactate dehydrogenase; M: molar; mM: millimolar; MFI: mean fluorescent intensity; NA: nicotinic acid; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; NAM: nicotinamide; NGS: next-generation sequencing; NHS/EDC: N-Hydroxysuccinimide/ ethyl (dimethylamino propyl) carbodiimide; Ni-NTA: nickel-nitrilotriacetic acid; nL: nanoliter; NK: natural killer; NMN: nicotinamide mononucleotide; OD: optical density; PARP: poly (adenosine diphosphate-ribose) polymerase; PBS: phosphate-buffered saline; PBMC: peripheral blood mononuclear cell; PDB: protein data bank; PE: phycoerythrin; PISA: protein interfaces, surfaces, and assemblies: PK: pharmacokinetics; mol: picomolar; RNA: ribonucleic acid; RLU: relative luminescence units; rpm: rotations per minute; RU: resonance unit; SEC: size exclusion chromatography; SEM: standard error of the mean; SIRT: sirtuins; SPR: surface plasmon resonance; µg: microgram; µM: micromolar; µL: microliter.


Subject(s)
NAD , Sirtuins , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic ADP-Ribose , Humans , Immunoglobulin G , Leukocytes, Mononuclear/metabolism , NAD/chemistry , NAD/metabolism , NADP , Niacinamide , Nicotinamide Mononucleotide , Ribose
11.
Sci Adv ; 8(38): eabn6545, 2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36129987

ABSTRACT

Severe COVID-19 is associated with hyperinflammation and weak T cell responses against SARS-CoV-2. However, the links between those processes remain partially characterized. Moreover, whether and how therapeutically manipulating T cells may benefit patients are unknown. Our genetic and pharmacological evidence demonstrates that the ion channel TMEM176B inhibited inflammasome activation triggered by SARS-CoV-2 and SARS-CoV-2-related murine ß-coronavirus. Tmem176b-/- mice infected with murine ß-coronavirus developed inflammasome-dependent T cell dysfunction and critical disease, which was controlled by modulating dysfunctional T cells with PD-1 blockers. In critical COVID-19, inflammasome activation correlated with dysfunctional T cells and low monocytic TMEM176B expression, whereas PD-L1 blockade rescued T cell functionality. Here, we mechanistically link T cell dysfunction and inflammation, supporting a cancer immunotherapy to reinforce T cell immunity in critical ß-coronavirus disease.

12.
J Immunol ; 183(5): 3099-108, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667084

ABSTRACT

C-type lectin receptors have recently been described as playing crucial roles in immunity and homeostasis since these proteins are able to recognize pathogens as well as self-Ags. We identified the C-type lectin-like receptor-1, CLEC-1, as being overexpressed in a model of rat allograft tolerance. We previously described in this model the expression of numerous cytoprotective molecules by graft endothelial cells and their interplay with regulatory CD4(+)CD25(+) T cells. In this study, we demonstrate that CLEC-1 is expressed by myeloid cells and specifically by endothelial cells in tolerated allografts and that CLEC-1 expression can be induced in endothelial cells by alloantigen-specific regulatory CD4(+)CD25(+) T cells. Analysis of CLEC-1 expression in naive rats demonstrates that CLEC-1 is highly expressed by myeloid cells and at a lower level by endothelial cells, and that its expression is down-regulated by inflammatory stimuli but increased by the immunoregulators IL-10 or TGFbeta. Interestingly, we demonstrate in vitro that inhibition of CLEC-1 expression in rat dendritic cells increases the subsequent differentiation of allogeneic Th17 T cells and decreases the regulatory Foxp3(+) T cell pool. Additionally, in chronically rejected allograft, the decreased expression of CLEC-1 is associated with a higher production of IL-17. Taken together, our data suggest that CLEC-1, expressed by myeloid cells and endothelial cells, is enhanced by regulatory mediators and moderates Th17 differentiation. Therefore, CLEC-1 may represent a new therapeutic agent to modulate the immune response in transplantation, autoimmunity, or cancer settings.


Subject(s)
Endothelial Cells/immunology , Endothelial Cells/metabolism , Lectins, C-Type/biosynthesis , Lymphocyte Activation/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Endothelial Cells/pathology , Gene Expression Regulation/immunology , Graft Survival/genetics , Graft Survival/immunology , Heart Transplantation/immunology , Heart Transplantation/pathology , Immune Tolerance/genetics , Inflammation Mediators/physiology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/genetics , Lectins, C-Type/physiology , Lymphocyte Activation/genetics , Molecular Sequence Data , Rats , Rats, Inbred Lew , T-Lymphocyte Subsets/metabolism
13.
Transplantation ; 105(4): 832-841, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32433241

ABSTRACT

BACKGROUND: Kidney transplantation is the therapeutic of choice for patients with kidney failure. While immunosuppressive drugs can control graft rejection, their use is associated with increased infections and cancer, and they do not effectively control chronic graft rejection. Cell therapy is an attractive strategy to minimize the use of pharmacological drugs. METHODS: We recently developed a protocol to generate human monocyte-derived autologous tolerogenic dendritic cells (ATDCs) from healthy volunteers. Herein, we transferred the ATDC manufacturing protocol to a Good Manufacturing Practice (GMP)-compliant facility. Furthermore, we compared the phenotype and in vitro functions of ATDCs generated from patients with end-stage renal disease to those generated from healthy volunteers. RESULTS: We describe the critical steps for GMP-compliant production of ATDCs and define the quality criteria required to allow release of the cell products. Furthermore, we showed that ATDCs generated from healthy volunteers and patients with kidney failure display the same tolerogenic profile based on their phenotype, resistance to maturation, and ability to modulate T-cell responses. CONCLUSIONS: Together, these results allowed us to define the production process and the quality criteria for the release of ATDCs before their administration in patients receiving a kidney transplant.


Subject(s)
Dendritic Cells/immunology , Kidney Failure, Chronic/immunology , Self Tolerance , Case-Control Studies , Cell Proliferation , Cell Separation , Cell Transplantation , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/surgery , Phenotype , Time Factors , Transplantation Tolerance , Transplantation, Autologous
14.
Sci Rep ; 11(1): 10592, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011961

ABSTRACT

The use of recombinant interleukin-2 (IL-2) as a therapeutic protein has been limited by significant toxicities despite its demonstrated ability to induce durable tumor-regression in cancer patients. The adverse events and limited efficacy of IL-2 treatment are due to the preferential binding of IL-2 to cells that express the high-affinity, trimeric receptor, IL-2Rαßγ such as endothelial cells and T-regulatory cells, respectively. Here, we describe a novel bispecific heavy-chain only antibody which binds to and activates signaling through the heterodimeric IL-2Rßγ receptor complex that is expressed on resting T-cells and NK cells. By avoiding binding to IL-2Rα, this molecule circumvents the preferential T-reg activation of native IL-2, while maintaining the robust stimulatory effects on T-cells and NK-cells in vitro. In vivo studies in both mice and cynomolgus monkeys confirm the molecule's in vivo biological activity, extended pharmacodynamics due to the Fc portion of the molecule, and enhanced safety profile. Together, these results demonstrate that the bispecific antibody is a safe and effective IL-2R agonist that harnesses the benefits of the IL-2 signaling pathway as a potential anti-cancer therapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Interleukin Receptor Common gamma Subunit/agonists , Interleukin-2 Receptor beta Subunit/agonists , Lymphocytes/drug effects , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2 Receptor beta Subunit/immunology , Macaca fascicularis , Male , Mice, Inbred BALB C
15.
J Clin Invest ; 117(4): 1096-106, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404623

ABSTRACT

Treatment with CD40Ig results in indefinite allograft survival in a complete MHC-mismatched heart allograft model in the rat. Here we show that serial second, third, and fourth adoptive transfers of total splenocytes from CD40Ig-treated recipients into secondary recipients led to indefinite donor-specific allograft acceptance. Purification of splenocyte subpopulations from CD40Ig-treated recipients demonstrated that only the adoptively transferred CD8(+)CD45RC(low) subset resulted in donor-specific long-term survival, whereas CD8(+)CD45RC(low) T cells from naive animals did not. Accepted grafts displayed increased indoleamine 2,3-dioxygenase (IDO) expression restricted in the graft to ECs. Coculture of donor ECs with CD8(+)CD45RC(low) T cells purified from CD40Ig-treated animals resulted in donor-specific IDO expression dependent on IFN-gamma. Neutralization of IFN-gamma or IDO triggered acute allograft rejection in both CD40Ig-treated and adoptively transferred recipients. This study demonstrates for what we believe to be the first time that interference in CD40-CD40 ligand (CD40-CD40L) interactions induces allospecific CD8(+) Tregs that maintain allograft survival. CD8(+)CD45RC(low) T cells act through IFN-gamma production, which in turn induces IDO expression by graft ECs. Thus, donor alloantigen-specific CD8(+) Tregs may promote local graft immune privilege through IDO expression.


Subject(s)
Graft Survival/physiology , Heart Transplantation/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/physiology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Graft Survival/drug effects , Graft Survival/immunology , Heart Transplantation/immunology , Immune Tolerance/drug effects , Rats , Rats, Inbred Lew , Transplantation, Homologous/immunology
16.
Cancer Cell ; 35(5): 767-781.e6, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31085177

ABSTRACT

Although immune checkpoint blockers have yielded significant clinical benefits in patients with different malignancies, the efficacy of these therapies is still limited. Here, we show that disruption of transmembrane protein 176B (TMEM176B) contributes to CD8+ T cell-mediated tumor growth inhibition by unleashing inflammasome activation. Lack of Tmem176b enhances the antitumor activity of anti-CTLA-4 antibodies through mechanisms involving caspase-1/IL-1ß activation. Accordingly, patients responding to checkpoint blockade therapies display an activated inflammasome signature. Finally, we identify BayK8644 as a potent TMEM176B inhibitor that promotes CD8+ T cell-mediated tumor control and reinforces the antitumor activity of both anti-CTLA-4 and anti-PD-1 antibodies. Thus, pharmacologic de-repression of the inflammasome by targeting TMEM176B may enhance the therapeutic efficacy of immune checkpoint blockers.


Subject(s)
Antineoplastic Agents/pharmacology , Inflammasomes/drug effects , Inflammasomes/immunology , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CHO Cells , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetulus , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/metabolism , Xenopus laevis/metabolism
17.
Cell Metab ; 30(6): 1075-1090.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801055

ABSTRACT

Cell therapy is a promising strategy for treating patients suffering from autoimmune or inflammatory diseases or receiving a transplant. Based on our preclinical studies, we have generated human autologous tolerogenic dendritic cells (ATDCs), which are being tested in a first-in-man clinical trial in kidney transplant recipients. Here, we report that ATDCs represent a unique subset of monocyte-derived cells based on phenotypic, transcriptomic, and metabolic analyses. ATDCs are characterized by their suppression of T cell proliferation and their expansion of Tregs through secreted factors. ATDCs produce high levels of lactate that shape T cell responses toward tolerance. Indeed, T cells take up ATDC-secreted lactate, leading to a decrease of their glycolysis. In vivo, ATDCs promote elevated levels of circulating lactate and delay graft-versus-host disease by reducing T cell proliferative capacity. The suppression of T cell immunity through lactate production by ATDCs is a novel mechanism that distinguishes ATDCs from other cell-based immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance , Immunosuppression Therapy , Lactic Acid/biosynthesis , Animals , Autoimmune Diseases/therapy , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Dendritic Cells/metabolism , Female , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/immunology
18.
Transplantation ; 85(9): 1351-6, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18475195

ABSTRACT

Regulatory T cells (Treg) have been identified as playing a pivotal role in the control of tolerance and in the suppression of pathologic immune responses in autoimmune diseases, transplantation, and graft-versus-host disease. Treg expanded ex vivo by dendritic cells could be potential reagents to promote antigen-specific tolerance in vivo. However, in vivo studies have been carried out mostly in rodents and will need validation in primates before clinical application. We characterized macaque dendritic cell derived either from bone marrow with and without prior CD34+ cell selection (BMDC), or from CD14+ peripheral blood mononuclear cells (Mo-DC). We demonstrate that with a semi-mature phenotype, BMDC are superior to Mo-DC in their capacity to expand freshly isolated allogeneic macaque CD4+ CD25+ CD127- Foxp3+ Treg in vitro in the presence of interleukin-2. Moreover, the expanded Treg maintain their phenotype and suppressive activity. These data provide a step toward the use of macaque dendritic cell to expand Treg for future preclinical testing.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Monocytes/cytology , Monocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD34/immunology , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/immunology , Lipopolysaccharide Receptors/immunology , Macaca fascicularis , Models, Animal
19.
Front Immunol ; 9: 274, 2018.
Article in English | MEDLINE | ID: mdl-29520275

ABSTRACT

Over the past century, solid organ transplantation has been improved both at a surgical and postoperative level. However, despite the improvement in efficiency, safety, and survival, we are still far from obtaining full acceptance of all kinds of allograft in the absence of concomitant treatments. Today, transplanted patients are treated with immunosuppressive drugs (IS) to minimize immunological response in order to prevent graft rejection. Nevertheless, the lack of specificity of IS leads to an increase in the risk of cancer and infections. At this point, cell therapies have been shown as a novel promising resource to minimize the use of IS in transplantation. The main strength of cell therapy is the opportunity to generate allograft-specific tolerance, promoting in this way long-term allograft survival. Among several other regulatory cell types, tolerogenic monocyte-derived dendritic cells (Tol-MoDCs) appear to be an interesting candidate for cell therapy due to their ability to perform specific antigen presentation and to polarize immune response to immunotolerance. In this review, we describe the characteristics and the mechanisms of action of both human Tol-MoDCs and rodent tolerogenic bone marrow-derived DCs (Tol-BMDCs). Furthermore, studies performed in transplantation models in rodents and non-human primates corroborate the potential of Tol-BMDCs for immunoregulation. In consequence, Tol-MoDCs have been recently evaluated in sundry clinical trials in autoimmune diseases and shown to be safe. In addition to autoimmune diseases clinical trials, Tol-MoDC is currently used in the first phase I/II clinical trials in transplantation. Translation of Tol-MoDCs to clinical application in transplantation will also be discussed in this review.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Dendritic Cells/immunology , Graft Rejection/immunology , Organ Transplantation , Animals , Bone Marrow/immunology , Clinical Trials as Topic , Dendritic Cells/transplantation , Graft Rejection/prevention & control , Graft Survival , Humans , Immune Tolerance , Immunomodulation , Mice , Primates , Rats
20.
Blood Adv ; 1(9): 557-568, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-29296975

ABSTRACT

Dendritic cells (DCs) represent essential antigen-presenting cells that are critical for linking innate and adaptive immunity, and influencing T-cell responses. Among pattern recognition receptors, DCs express C-type lectin receptors triggered by both exogenous and endogenous ligands, therefore dictating pathogen response, and also shaping T-cell immunity. We previously described in rat, the expression of the orphan C-type lectin-like receptor-1 (CLEC-1) by DCs and demonstrated in vitro its inhibitory role in downstream T helper 17 (Th17) activation. In this study, we examined the expression and functionality of CLEC-1 in human DCs, and show a cell-surface expression on the CD16- subpopulation of blood DCs and on monocyte-derived DCs (moDCs). CLEC-1 expression on moDCs is downregulated by inflammatory stimuli and enhanced by transforming growth factor ß. Moreover, we demonstrate that CLEC-1 is a functional receptor on human moDCs and that although not modulating the spleen tyrosine kinase-dependent canonical nuclear factor-κB pathway, represses subsequent Th17 responses. Interestingly, a decreased expression of CLEC1A in human lung transplants is predictive of the development of chronic rejection and is associated with a higher level of interleukin 17A (IL17A). Importantly, using CLEC-1-deficient rats, we showed that disruption of CLEC-1 signaling led to an enhanced Il12p40 subunit expression in DCs, and to an exacerbation of downstream in vitro and in vivo CD4+ Th1 and Th17 responses. Collectively, our results establish a role for CLEC-1 as an inhibitory receptor in DCs able to dampen activation and downstream effector Th responses. As a cell-surface receptor, CLEC-1 may represent a useful therapeutic target for modulating T-cell immune responses in a clinical setting.

SELECTION OF CITATIONS
SEARCH DETAIL