Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Blood ; 130(2): 176-180, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28566492

ABSTRACT

Langerhans cell histiocytosis (LCH) and the non-LCH neoplasm Erdheim-Chester disease (ECD) are heterogeneous neoplastic disorders marked by infiltration of pathologic macrophage-, dendritic cell-, or monocyte-derived cells in tissues driven by recurrent mutations activating MAPK signaling. Although recent data indicate that at least a proportion of LCH and ECD patients have detectable activating kinase mutations in circulating hematopoietic cells and bone marrow-based hematopoietic progenitors, functional evidence of the cell of origin of histiocytosis from actual patient materials has long been elusive. Here, we provide evidence for mutations in MAPK signaling intermediates in CD34+ cells from patients with ECD and LCH/ECD, including detection of shared origin of LCH and acute myelomonocytic leukemia driven by TET2-mutant CD34+ cell progenitors in one patient. We also demonstrate functional self-renewal capacity for CD34+ cells to drive the development of histiocytosis in xenotransplantation assays in vivo. These data indicate that the cell of origin of at least a proportion of patients with systemic histiocytoses resides in hematopoietic progenitor cells prior to committed monocyte/macrophage or dendritic cell differentiation and provide the first example of a patient-derived xenotransplantation model for a human histiocytic neoplasm.


Subject(s)
Bone Marrow Cells/pathology , DNA-Binding Proteins/genetics , Erdheim-Chester Disease/pathology , Hematopoietic Stem Cells/pathology , Histiocytosis, Langerhans-Cell/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , Adult , Alleles , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Differentiation , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dioxygenases , Erdheim-Chester Disease/genetics , Erdheim-Chester Disease/immunology , Gene Expression , Hematopoietic Stem Cells/immunology , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/immunology , Humans , Immunophenotyping , Macrophages/immunology , Macrophages/pathology , Mice , Monocytes/immunology , Monocytes/pathology , Mutation , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins B-raf/immunology , Transplantation, Heterologous
2.
Proc Natl Acad Sci U S A ; 113(8): 2182-7, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26862175

ABSTRACT

Viral clearance requires effector T-cell egress from the draining lymph node (dLN). The mechanisms that regulate the complex process of effector T-cell egress from the dLN after infection are poorly understood. Here, we visualized endogenous pathogen-specific effector T-cell migration within, and from, the dLN. We used an inducible mouse model with a temporally disrupted sphingosine-1-phosphate receptor-1 (S1PR1) gene specifically in endogenous effector T cells. Early after infection, WT and S1PR1(-/-) effector T cells localized exclusively within the paracortex. This localization in the paracortex by CD8 T cells was followed by intranodal migration by both WT and S1PR1(-/-) T cells to positions adjacent to both cortical and medullary lymphatic sinuses where the T cells exhibited intense probing behavior. However, in contrast to WT, S1PR1(-/-) effector T cells failed to enter the sinuses. We demonstrate that, even when LN retention signals such as CC chemokine receptor 7 (CCR7) are down-regulated, T cell intrinsic S1PR1 is the master regulator of effector T-cell emigration from the dLN.


Subject(s)
Infections/immunology , Infections/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Receptors, Lysosphingolipid/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Cell Movement/immunology , Endothelial Cells/immunology , Endothelial Cells/pathology , Lymphocyte Activation , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Vesicular stomatitis Indiana virus
3.
J Immunol ; 188(3): 981-91, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22184726

ABSTRACT

By revisiting CD90, a GPI-anchored glycoprotein, we show that CD90 is expressed by a subset of CD4(+) and CD8(+) human T cells. CD4(+)CD90(+) cells share similarities with Th17 cells because they express the Th17-specific transcription factor RORC2 and produce IL-17A. CD4(+)CD90(+) cells are activated memory T cells that express the gut mucosal markers CCR6, CD161, and the α(4) and ß(7) integrins. Compared with CD90-depleted CCR6(+) memory Th17 cells, CD4(+)CD90(+) cells express higher levels of IL-22 and proinflammatory cytokines (IL-6, TNF-α and GM-CSF), but they produce lower levels of IL-21 and no IL-9. Analyses of CD8(+)CD90(+) cells reveal that they express RORC2 and are able to produce higher levels of IL-17A, IL-22, and CCL20 compared with CD90-depleted CD8(+) cells. These data show that CD90 identifies Th17 and Tc17 cells with a peculiar cytokine profile. Studies of circulating CD90(+) cells in HIV patients show that CD90(+) cells are decreased with an imbalance of the CD4(+)CD90(+)/regulatory T cell ratio in nontreated patients compared with treated patients and healthy donors. Overall, human CD90 identifies a subset of Th17 and Tc17 cells within CD4(+) and CD8(+) T cells, respectively, which are depleted during HIV infection.


Subject(s)
HIV Infections/immunology , T-Lymphocyte Subsets/virology , Th17 Cells/pathology , Thy-1 Antigens , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Chemokine CCL20 , Cytokines/analysis , Humans , Interleukins , Nuclear Receptor Subfamily 1, Group F, Member 3 , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Th17 Cells/virology , Interleukin-22
4.
Brain ; 135(Pt 2): 483-92, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22240777

ABSTRACT

γ-Sarcoglycanopathy or limb girdle muscular dystrophy type 2C is an untreatable disease caused by autosomal recessively inherited mutations of the γ-sarcoglycan gene. Nine non-ambulatory patients (two males, seven females, mean age 27 years; range 16-38 years) with del525T homozygous mutation of the γ-sarcoglycan gene and no γ-sarcoglycan immunostaining on muscle biopsy were divided into three equal groups to receive three escalating doses of an adeno-associated virus serotype 1 vector expressing the human γ-sarcoglycan gene under the control of the desmin promoter, by local injection into the extensor carpi radialis muscle. The first group received a single injection of 3 × 10(9) viral genomes in 100 µl, the second group received a single injection of 1.5 × 10(10) viral genomes in 100 µl, and the third group received three simultaneous 100-µl injections at the same site, delivering a total dose of 4.5 × 10(10) viral genomes. No serious adverse effects occurred during 6 months of follow-up. All nine patients became adeno-associated virus serotype 1 seropositive and one developed a cytotoxic response to the adeno-associated virus serotype 1 capsid. Thirty days later, immunohistochemical analysis of injected-muscle biopsy specimens showed γ-sarcoglycan expression in all three patients who received the highest dose (4.7-10.5% positively stained fibres), while real-time polymerase chain reaction detected γ-sarcoglycan messenger RNA. In one patient, γ-sarcoglycan protein was detected by western blot. For two other patients who received the low and intermediate doses, discrete levels of γ-sarcoglycan expression (<1% positively stained fibres) were also detectable. Expression of γ-sarcoglycan protein can be induced in patients with limb girdle muscular dystrophy type 2C by adeno-associated virus serotype 1 gene transfer, with no serious adverse effects.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Muscular Dystrophies, Limb-Girdle/therapy , Sarcoglycans/genetics , Adolescent , Adult , Dependovirus/genetics , Dependovirus/metabolism , Female , Follow-Up Studies , Genetic Vectors , Humans , Male , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Sarcoglycans/metabolism , Treatment Outcome
5.
Front Immunol ; 12: 626776, 2021.
Article in English | MEDLINE | ID: mdl-33763071

ABSTRACT

The presence of tertiary lymphoid structures (TLS) in the tumor microenvironment is associated with better clinical outcome in many cancers. In non-small cell lung cancer (NSCLC), we have previously showed that a high density of B cells within TLS (TLS-B cells) is positively correlated with tumor antigen-specific antibody responses and increased intratumor CD4+ T cell clonality. Here, we investigated the relationship between the presence of TLS-B cells and CD4+ T cell profile in NSCLC patients. The expression of immune-related genes and proteins on B cells and CD4+ T cells was analyzed according to their relationship to TLS-B density in a prospective cohort of 56 NSCLC patients. We observed that tumor-infiltrating T cells showed marked differences according to TLS-B cell presence, with higher percentages of naïve, central-memory, and activated CD4+ T cells and lower percentages of both immune checkpoint (ICP)-expressing CD4+ T cells and regulatory T cells (Tregs) in the TLS-Bhigh tumors. A retrospective study of 538 untreated NSCLC patients showed that high TLS-B cell density was even able to counterbalance the deleterious impact of high Treg density on patient survival, and that TLS-Bhigh Treglow patients had the best clinical outcomes. Overall, the correlation between the density of TLS-Bhigh tumors with early differentiated, activated and non-regulatory CD4+ T cell cells suggest that B cells may play a central role in determining protective T cell responses in NSCLC patients.


Subject(s)
B-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Humans , Lymphocyte Activation , Male , Middle Aged , Retrospective Studies , Transcriptome , Tumor Microenvironment/immunology
6.
Sci Rep ; 10(1): 6488, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32300208

ABSTRACT

Chronic exposure to environmental pollutants is often associated with systemic inflammation. As such, cigarette smoking contributes to inflammation and lung diseases by inducing senescence of pulmonary cells such as pneumocytes, fibroblasts, and endothelial cells. Yet, how smoking worsens evolution of chronic inflammatory disorders associated with Th17 lymphocytes, such as rheumatoid arthritis, psoriasis, Crohn's disease, and multiple sclerosis, is largely unknown. Results from human studies show an increase in inflammatory CD4+ Th17 lymphocytes at blood- and pulmonary level in smokers. The aim of the study was to evaluate the sensitivity of CD4+ Th17 lymphocytes to cigarette smoke-induced senescence. Mucosa-homing CCR6+ Th17- were compared to CCR6neg -and regulatory T peripheral lymphocytes after exposure to cigarette smoke extract (CSE). Senescence sensitivity of CSE-exposed cells was assessed by determination of various senescence biomarkers (ß-galactosidase activity, p16Ink4a- and p21 expression) and cytokines production. CCR6+ Th17 cells showed a higher sensitivity to CSE-induced senescence compared to controls, which is associated to oxidative stress and higher VEGFα secretion. Pharmacological targeting of ROS- and ERK1/2 signalling pathways prevented CSE-induced senescence of CCR6+Th17 lymphocytes as well as VEGFα secretion. Altogether, these results identify mechanisms by which pro-oxidant environmental pollutants contribute to pro-angiogenic and pathogenic CCR6+Th17 cells, therefore potential targets for therapeutic purposes.


Subject(s)
Cellular Senescence/immunology , Cigarette Smoking/immunology , Th17 Cells/immunology , Vascular Endothelial Growth Factor A/metabolism , Blood Buffy Coat/cytology , Cells, Cultured , Cellular Senescence/drug effects , Cigarette Smoking/adverse effects , Cigarette Smoking/blood , Cytokines/metabolism , Healthy Volunteers , Humans , MAP Kinase Signaling System/immunology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Primary Cell Culture , Reactive Oxygen Species/metabolism , Receptors, CCR6/metabolism , Smoke/adverse effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
7.
Int J Oncol ; 35(3): 569-81, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639177

ABSTRACT

Cytotoxic chemotherapy is ineffective in metastatic renal cancer. However, systemic administration of interleukin 2 (IL-2) or infusion of dendritic cells (DCs) loaded with tumor extracts can lead to some response rates with concomitant survival improvements. We report the results of a phase I-II pilot study combining DCs and IL-2 where six patients were included. DCs were derived from bone marrow CD34+ cells and loaded with autologous tumor extracts. CD34-DC vaccines were infused subcutaneously at day 45, 52, 59, 90 and 120 following surgery in combination with IL-2, that was subsequently administrated after the 3rd and 4th DC vaccinations. Preparation of tumor extracts and CD34-DCs were satisfactory in all patients but one. Due to rapid tumor progression, one patient was excluded before vaccination. In the 4 remaining patients, two received 3 vaccinations, while the 2 others received 5 vaccinations and the full IL-2 treatment. No adverse effect due to the vaccinations was observed. A specific immune response against autologous tumor cells was observed in the 2 patients who completed the treatment. Interestingly, these 2 patients had a more prolonged survival than the patients receiving 3 vaccinations. Importantly, a transient and massive increase of circulating natural regulatory T-cells (nTregs) was evidenced in 3 patients following IL-2 administration. Overall, the use of CD34-DC vaccines is feasible, safe and non-toxic. A specific anti-tumor immune response can be detected. However, our data highlights that IL-2 is a potent inducer of nTregs in vivo and as such may have a negative impact on cancer immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Antigens, CD34/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Carcinoma, Renal Cell/immunology , Dendritic Cells/immunology , Female , Humans , Immunophenotyping , Immunotherapy/methods , Kidney Neoplasms/immunology , Male , Middle Aged , T-Lymphocytes, Regulatory/drug effects
8.
Sci Rep ; 9(1): 12910, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501487

ABSTRACT

Currently, there is a lack of systems for studying the role of hepatitis B viral proteins, such as HBeAg and HBcAg, on liver injury. It is necessary to develop an original tool in order to clarify the role of these viral proteins in hepatic stellate cell activation, and to understand the molecular mechanisms of liver injury. HepaRG are the most reliable hepatocyte-like cells for studying liver functions or disorders. In this paper, we demonstrate that the transduction of differentiated HepaRG (dHepaRG) cells can be performed successfully using lentiviral particles. The production of a functional Green Fluorescent Protein (GFP) assessed by Fluorescence Activated Cell Sorting and fluorescence microscopy is up to 16% of GFP positive cells using a multiplicity of infection (MOI) of 2.4. We demonstrate that this technology can allow the stable expression of GFP during the long lifecycle of the cell (up to four weeks after the cell's passage). With this innovative tool, we aim to express viral proteins such as HBeAg or HBcAg in dHepaRG cells. The preliminary results of this work shows that HBeAg can be efficiently produced in dHepaRG cells and that increased MOI allows a better production of this protein. Our future objective will be to study the role of HBc and HBe proteins on the induction of hepatic fibrosis.


Subject(s)
Genetic Vectors , Hepatocytes/metabolism , Lentivirus , Transduction, Genetic , Animals , Cell Culture Techniques , Cell Line , Flow Cytometry , Gene Expression , Genes, Reporter , Genetic Engineering , Genetic Vectors/genetics , Lentivirus/genetics , Mice , Transgenes
9.
J Gene Med ; 10(8): 834-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18615770

ABSTRACT

BACKGROUND: Human CD4+CD25+FOXP3+ natural regulatory T-cells (nTreg) have a great therapeutic potential for the induction of tolerance in allo-transplanted patients or for the control of severe auto-immune diseases. However, clinical-grade production of nTreg remains difficult to achieve because of the absence of a truly specific surface marker and of their low frequency that implies a need for their ex vivo expansion. Furthermore, safety issues should be taken into consideration due to the risk of either uncontrolled nTreg-induced immunosuppression or uncontrolled proliferation of autoreactive contaminating T-cells particularly in an auto-immune context. METHODS: We compared different clinical-grade conditions for immuno-magnetic selection and ex vivo expansion of nTreg. For safety, expanded cells were genetically modified with retroviral vectors co-expressing human CD90 and HSV1 thymidine kinase. The CD90 surface marker and thymidine kinase allow for selection and elimination of transduced cells by ganciclovir, respectively. RESULTS: We showed that (i) nTreg could be enriched in a one step using CD25 microbeads, were functionally suppressive and mainly FOXP3+; (ii) using anti-CD28- and anti-CD3-coated beads, interleukin-2 and rapamycin, nTreg were expanded 150-200-fold after 3 weeks. Under these clinical-grade conditions, they remained suppressive, and no major alteration of the TCR repertoire was observed; (iii) after efficient retroviral transduction and CD90 selection, nTreg maintained their suppressive activity; (iv) transduced nTreg could be eliminated by ganciclovir upon activation. CONCLUSIONS: The efficient procedure reported here for the preparation of nTreg, whose safety has been ensured, is now applicable for further clinical trials.


Subject(s)
Genes, Transgenic, Suicide/genetics , Retroviridae/genetics , T-Lymphocytes, Regulatory/immunology , Thy-1 Antigens/genetics , Thymidine Kinase/genetics , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Death/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Forkhead Transcription Factors/metabolism , Ganciclovir/pharmacology , Genetic Vectors , Herpesvirus 1, Human/enzymology , Humans , Immunomagnetic Separation , Immunosuppressive Agents/pharmacology , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation/drug effects , Microspheres , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , Time Factors , Transduction, Genetic
10.
Mol Immunol ; 44(4): 267-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16630660

ABSTRACT

In humans, the CD4 molecule is expressed on a subset of T-cells and at various levels on myeloid and lymphoid cells. The mechanisms regulating human CD4 gene expression are yet poorly understood. We speculated that the CD4 silencer, which operates in CD8+ T-cells to repress CD4 expression, could be responsible for CD4 repression in human lymphoid non-T-cells. To test this possibility, we used lentiviral vectors carrying CD4 regulatory sequences, with or without the silencer element, to express an eGFP reporter gene. We observed that (i) in the absence of the silencer element, eGFP expression was detected in CD34+-derived B- and NK-cells that otherwise lacked endogenous CD4 mRNA, indicating active repression of the CD4 regulatory sequences and (ii) the addition of the CD4 silencer could repress eGFP expression in these same cells, as well as in human B-cells generated in vivo in NOD/SCID mice. Collectively, our results suggest that beyond its well-characterized function in T-cells, the CD4 silencer also regulates CD4 gene expression in human lymphoid non-T-cells.


Subject(s)
CD4 Antigens/genetics , Gene Expression Regulation , Lymphoid Tissue/immunology , Silencer Elements, Transcriptional , CD4 Antigens/immunology , Cell Line , Enhancer Elements, Genetic , Gene Expression Regulation/immunology , Genes, Reporter , Green Fluorescent Proteins , Humans , Promoter Regions, Genetic , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
11.
Int J Oncol ; 31(6): 1357-65, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982663

ABSTRACT

Many strategies have been proposed to circumvent cancer development or prevent its growth. One of the promising strategies is to direct the immune response toward tumour antigens. This can be achieved by loading dendritic cells, the most potent antigen presenting cells, with tumour antigens. Fusion of dendritic cells (DC) with tumour cells is an attractive way to load the DC with all tumour antigens regardless of their immunogenicity status and the fact that they have, or not, been identified. The aim of our study was to characterise the immunophenotype of fused cells, monitor the evolution of the fusion interface and the distribution of surface antigens over time and assess for their maturation status and functionality in vitro. We used polyethylene glycol to fuse DC with Her2/neu positive breast cancer cell line T-47D. We demonstrate that false positive events accounted in flow cytometry can be identified using confocal microscopy to avoid an overestimation of fusion efficiency and to distinguish clearly hybrid cells from aggregated or phagocytosed cells. We used imaging means to demonstrate the conservation of presentation molecules (MHC II, CD1a), co-stimulatory molecules (CD40, CD80, CD86), as well as tumour antigens (Her2/neu, cytokeratins) in optimised conditions. Fused cells were only recognisable for 48 h as assessed by membrane staining and membranous antigen distribution. Fusion was necessary for their maturation to be accompanied by functional activity such as secretion of cytokines and perforin. These results suggest that hybrid cells generated by the fusion of DC and tumour cells can be easily identified and characterised using imaging techniques, and that, regarding functionality and cytokine secretion, they appear to be good candidates for anti-tumour therapies namely in breast cancer.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Dendritic Cells/physiology , Hybrid Cells/physiology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Fusion , Cell Line, Tumor , Female , Flow Cytometry , HLA-DR Antigens/analysis , Humans , Interleukin-10/biosynthesis , Receptor, ErbB-2/analysis
12.
J Ocul Pharmacol Ther ; 33(4): 278-284, 2017 05.
Article in English | MEDLINE | ID: mdl-28448237

ABSTRACT

Uveitis is a sight-threatening primary intraocular inflammation of various origins in mainly young and active patients. Due to the absence of biomarkers in most of the cases, the current treatment of noninfectious entities remains nonspecific, using corticosteroids, conventional immunosuppressors, and more recently biological agents. Identification of regulatory T cells in different models of autoimmune uveitis together with the evaluation of this important subpopulation in different entities paved the way for new therapeutic strategies, in addition to exclusive pharmaceutical approaches. Upregulation of regulatory T cells induced by biological agents has been recently highlighted. Development of cell therapy in autoimmune diseases is at its stammering needing more experimental data and robust clinical trials to demonstrate safety and efficacy before larger developments. Specific or polyclonal Tregs may be used, but it is of utmost importance to determine the method of selection, the level of activation, and the route of administration. Mastering immune cell therapy remains a challenging goal in patients with autoimmune diseases, but it may significantly enlarge our therapeutic possibilities in severe and refractory situations.


Subject(s)
T-Lymphocytes, Regulatory/immunology , Uveitis/therapy , Animals , Humans , Uveitis/immunology
13.
Eur J Heart Fail ; 19(11): 1534-1541, 2017 11.
Article in English | MEDLINE | ID: mdl-28393439

ABSTRACT

AIMS: Restoration of sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2a) activity through gene transfer improved cardiac function in experimental and pilot studies in humans with heart failure. The AGENT-HF (NCT01966887) trial investigated the impact of adeno-associated virus (AAV1)/SERCA2a on ventricular remodelling using multimodality non-invasive cardiac imaging. METHODS AND RESULTS: AGENT-HF was a single centre, randomized, double-blind, placebo-controlled trial in adult patients with NYHA class III-IV ischaemic or non-ischaemic heart failure and left ventricular ejection fraction ≤35%. Eligible patients were randomized to receive a single intracoronary infusion of either 1 × 1013 DNase-resistant particles of AAV1/SERCA2a or placebo. The primary endpoint was change in left ventricular end-systolic volume (LVESV), measured by cardiac computed tomography at 6 month follow-up. We planned to include 40 patients but the trial was terminated prematurely as the sponsor suspended further enrolment following neutral results of the CUPID-2 outcome trial. At the time of termination, nine patients were randomized with five patients infused with AAV1/SERCA2a and four with placebo. At 6 months, LVESV was increased in both groups compared with baseline: median (interquartile range) in AAV1/SERCA2a vs. placebo: 13 (13;14) mL vs. 3.5 (-36;36) mL, P = 0.74, with a mean difference between groups of 11.4 mL in favour of placebo. No safety issues were noted. CONCLUSION: AGENT-HF failed to demonstrate any improvement in ventricular remodelling in response to AAV1/SERCA2a at the dose studied. However, because of premature termination, the study was underpowered to demonstrate an effect of AAV1/SERCA2a and these data should be interpreted with caution.


Subject(s)
Genetic Therapy/methods , Heart Failure, Systolic/drug therapy , Sarcoplasmic Reticulum Calcium-Transporting ATPases/administration & dosage , Ventricular Remodeling/physiology , Aged , Coronary Vessels , Double-Blind Method , Female , Heart Failure, Systolic/diagnosis , Heart Failure, Systolic/physiopathology , Humans , Infusions, Intra-Arterial , Male , Middle Aged , Treatment Outcome , Ventricular Remodeling/genetics
15.
J Control Release ; 239: 82-91, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27565211

ABSTRACT

Gene-directed enzyme pro-drug therapy (GDEPT) consists of expressing, in tumor cells, a suicide gene which converts a pro-drug into cytotoxic metabolites, in situ. In a previous work, we demonstrated that the combination of the suicide gene CYP2B6TM-RED (a fusion of a triple mutant of CYP2B6 with NADPH cytochrome P450 reductase) and cyclophosphamide (CPA) constituted a powerful treatment for solid tumors. In this work, we investigated the use of mesenchymal stem cells (MSCs) as cellular vehicles for the delivery of our suicide gene. MSCs were genetically engineered ex-vivo to stably express CYP2B6TM-RED. Ex vivo and in vivo investigations showed that MSCs expressing CYP2B6TM-RED were able 1) to bioactivate CPA and produce local cytotoxic metabolites in tumor sites and 2) to destroy neighboring tumor cells through a bystander effect. Intratumoral injections of CYP2B6TM-RED-MSCs and CPA completely eradicated tumors in 33% of mice without recurrence after 6months. Rechallenge experiments demonstrated an efficient immune response. These data suggest that MSCs expressing CYP2B6TM-RED with CPA could represent a promising treatment for solid tumors to test in future clinical trials.


Subject(s)
Genes, Transgenic, Suicide/genetics , Genetic Engineering/methods , Genetic Therapy/methods , Genetic Vectors/genetics , Mesenchymal Stem Cells/physiology , Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Neoplasms/therapy
16.
Cancer Res ; 76(22): 6483-6494, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27680685

ABSTRACT

The CCL2 chemokine receptor CCR2 drives cancer by mediating the recruitment of monocytes and myeloid-derived suppressor cells to the tumor microenvironment. In this study, we extend the significance of CCR2 in this setting by identifying a new role for it in mediating recruitment of CD4+ T regulatory cells (Treg). Following tumor initiation, an expanded population of CCR2+ Tregs required CCR2 expression to traffic between draining lymph nodes (dLN) and the tumor. This Treg subset was enriched in the fraction of tumor antigen-specific cells in the dLN, where they displayed an activated immunosuppressive phenotype. Notably, in mouse models, low-dose cyclophosphamide treatment preferentially depleted CCR2+ Treg, enhancing priming of tumor-specific CD8+ T cells. In the MMTV-PyMT transgenic mouse model of breast cancer and in oral squamous cell carcinoma patients, tumor development was associated with decreased blood frequency and inversely increased tumor frequency of CCR2+ Tregs. Our results define a novel subset of CCR2+ Treg involved in tumoral immune escape, and they offer evidence that this Treg subset may be preferentially eradicated by low-dose cyclophosphamide treatment. Cancer Res; 76(22); 6483-94. ©2016 AACR.


Subject(s)
Cyclophosphamide/therapeutic use , T-Lymphocytes, Regulatory/immunology , Animals , Biomarkers , Cell Movement , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Female , Humans , Male , Mice , Middle Aged , Receptors, CCR2/metabolism
17.
Oncoimmunology ; 5(7): e1164363, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622018

ABSTRACT

Despite current therapy, head and neck squamous cell carcinomas (HNSCCs) arising from various mucosal sites of the upper aero-digestive tract frequently relapse in a loco-regional manner and have a poor prognosis. Our objective was to validate an innovative mucosal route of vaccination using plasmo virus-like particles (pVLPs) in a pre-clinical orthotopic model of HNSCCs. For this purpose, we used pVLP-E7, that are plasmid DNA encoding retroviral virus-like particles carrying a truncated E7 oncoprotein from HPV-16 as antigen model, to vaccinate mice bearing pre-established TC-1 tumors implanted into the buccal mucosa. pVLP-E7 were combined with clinical grade TLR agonists (Imiquimod and CpG-ODN). In this pre-clinical orthotopic model, whose tumor microenvironment resembles to those of human HNSCCs, different mucosal vaccination routes were tested for their ability to elicit efficient immune and antitumoral responses. Results showed that mucosal intra-cheek (IC) vaccinations using pVLP-E7, comparatively to intradermic vaccinations (ID), gave rise to higher mobilization of mucosal (CD49a(+)) CD8(+) specific effector T cells in both tumor draining lymph nodes (TdLNs) and tumor microenvironment resulting in better antitumor effects and in a long-term protection against tumor rechallenge. In vivo CD8(+) depletion demonstrated that antitumoral effects were fully dependent upon the presence of CD8(+) T cells. Validation of IC mucosal vaccinations with pVLPs combined with adjuvants using a pre-clinical orthotopic model of HNSCC provides valuable pre-clinical data to rapidly envision the use of such therapeutic vaccines in patients with HNSCCs, inasmuch as vaccinal components and adjuvants can be easily obtained as clinical grade reagents.

18.
Cell Transplant ; 24(12): 2527-40, 2015.
Article in English | MEDLINE | ID: mdl-25198125

ABSTRACT

Natural regulatory T cells (Tregs) may have a great therapeutic potential to induce tolerance in allogeneic cells and organ transplantations. In mice, we showed that alloantigen-specific Tregs (spe-Tregs) were more efficient than polyclonal Tregs (poly-Tregs) in controlling graft-versus-host disease (GVHD). Here we describe a clinical-grade compliant method for generating human spe-Tregs. Tregs were enriched from leukapheresis products with anti-CD25 immunomagnetic beads, primed twice by allogeneic mature monocyte-derived dendritic cells (mDCs), and cultured during 3 weeks in medium containing interleukin 2 (IL-2), IL-15, and rapamycin. After 3 weeks of culture, final cell products were expanded 8.3-fold from the initial CD25(+) purifications. Immunophenotypic analyses of final cells indicate that they were composed of 88 ± 2.6% of CD4(+) T cells, all expressing Treg-specific markers (FOXP3, Helios, GARP, LAP, and CD152). Spe-Tregs were highly suppressive in vitro and also in vivo using a xeno-GVHD model established in immunodeficient mice. The specificity of their suppressive activity was demonstrated on their ability to significantly suppress the proliferation of autologous effector T cells stimulated by the same mDCs compared to third-party mDCs. Our data provide evidence that functional alloantigen Tregs can be generated under clinical-grade compliant conditions. Taking into account that 130 × 10(6) CD25(+) cells can be obtained at large scale from standard leukapheresis, our cell process may give rise to a theoretical final number of 1 × 10(9) spe-Tregs. Thus, using our strategy, we can propose to prepare spe-Tregs for clinical trials designed to control HLA-mismatched GVHD or organ transplantation rejection.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Isoantigens/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Adult , Aged , Animals , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Dendritic Cells/immunology , Female , Graft vs Host Disease/therapy , Humans , Immune Tolerance/immunology , Immunomagnetic Separation , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Leukapheresis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Organ Transplantation/methods , Sirolimus/pharmacology , Young Adult
19.
AIDS ; 17(4): 563-74, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12598777

ABSTRACT

OBJECTIVES: To study, in asymptomatic HIV-1-infected (HIV+) patients, whether peripheral blood hematopoietic progenitor/stem cells (PBPC) mobilized by granulocyte colony stimulating factor (G-CSF), can be used as a source of cells for retroviral gene therapy. DESIGN: PBPC from two groups of HIV+ patients (treated or untreated by highly active antiretroviral therapy) and from seronegative donors were mobilized with G-CSF. METHODS: PBPC collected by leukapheresis were enriched for CD34 cells, immunophenotypically and functionally characterized, cultured and infected with retroviral vectors. HIV proviral integration was studied on fresh and cultured cells. RESULTS: G-CSF moderately and transiently increased the viral load in untreated patients only, and induced in both groups of HIV+ patients mobilization of percentages and numbers of CD34 cells comparable to those of seronegative volunteers. The most immature CD34 cell subset, the clonogenic progenitor and long-term culture initiating cells were significantly decreased in leukapheresis products and CD34-enriched fractions from untreated HIV+ patients but not in those from treated HIV+ patients. Cell cycle activation and growth factor responses of CD34 cells from both groups of HIV+ patients were not different from those of the control group. Culture and retroviral infection of CD34 cells from HIV+ patients did not enhance HIV replication, and yielded transduction levels similar to those obtained using CD34 cells from seronegative donors. CONCLUSIONS: G-CSF-mobilized PBPC can be safely used for HIV retroviral gene therapy in asymptomatic treated patients while highly active antiretroviral therapy would control the G-CSF-induced increase in viral load and correct the defective hematopoiesis observed in untreated patients, without inhibiting the retroviral transduction of PBPC.


Subject(s)
Antiretroviral Therapy, Highly Active , Genetic Therapy/methods , HIV Infections/blood , HIV Infections/drug therapy , HIV-1 , Hematopoietic Stem Cell Mobilization , Peripheral Blood Stem Cell Transplantation , Antigens, CD34 , Granulocyte Colony-Stimulating Factor/therapeutic use , HIV Infections/therapy , Humans , Leukocytes/immunology , Retroviridae/genetics , Thy-1 Antigens/genetics , Transduction, Genetic/methods , Viral Load , Virus Integration
20.
Hum Gene Ther ; 15(6): 542-52, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15212713

ABSTRACT

We have previously proposed the use of HLA-specific T-cell clones transduced with a suicide gene to produce an allogeneic effect that can be controlled after allogeneic bone marrow transplantation. Procedures described so far to obtain specific T-cells transduced with a suicide gene have led to the recovery of heterogeneous polyclonal T-cells with a limited level of purity. We have therefore developed an approach to select specific T-cell clones in which the suicide transgene is inserted at a unique site of the genome, and used it to produce CD(+)-cytotoxic HLA-DP-specific T-cell clones. Immunization was performed by a one-way mixed lymphocyte culture and responder T lymphocytes were transduced at day 16, 6 days after the second stimulation. Transductions were carried out using gibbon ape leukemia virus (GALV)-pseudotyped retroviral particles harboring a bicistronic Thy-1/TK vector produced by TEFLY GA16-pKM4 clone 34 packaging cells. Three to 5 days later, CD90 immunomagnetic selection and cloning were performed on the transduced T cells. Our results demonstrate that this procedure led to the recovery of T-cell clones, the majority of which had the expected specificity and a single site of transgene insertion. Such clonotransgenic T-cell populations represent suitable tools to drive a defined alloreaction that can be controlled after bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation , Genes, Transgenic, Suicide/physiology , HLA-DP Antigens/immunology , Immunization , T-Lymphocytes/enzymology , Thymidine Kinase/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , Cells, Cultured/transplantation , Clone Cells , HLA-DP Antigens/genetics , Humans , Immunomagnetic Separation , Immunotherapy , Leukemia Virus, Gibbon Ape/genetics , Lymphocyte Activation , Retroviridae/genetics , T-Lymphocytes/immunology , Thy-1 Antigens/immunology , Thy-1 Antigens/metabolism , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL