Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
PLoS One ; 16(12): e0261987, 2021.
Article in English | MEDLINE | ID: mdl-34972158

ABSTRACT

Nanoparticles made of the coat protein of papaya mosaic virus (PapMV) and a single-strand RNA were previously shown to be an efficient antigen presentation system for the trigger of cellular immunity. Engineering of PapMV nano with a cytotoxic T lymphocyte epitope was previously shown activating specific T lymphocytes through a proteasome-independent major histocompatibility complex class I (MHC-I) cross-presentation. In this study, we provide new insights into the mechanism of the MHC-I cross-presentation mediated by PapMV nanoparticles. We demonstrate that PapMV nanoparticles do not require the transporter associated with antigen presentation (TAP), but rather depend on lysosome acidification and cathepsin S protease activity for presentation of the T cell epitope. We have also linked the induction of autophagy with this vacuolar MHC-I cross-presentation process. Interestingly, autophagy is induced in antigen-presenting cells after PapMV nanoparticles exposure and inhibition of autophagy reduce MHC-I cross-presentation. This study demonstrates that autophagy is associated with TAP- and proteasome-independent MHC-I cross-presentation. A deeper understanding of the autophagy-dependent MHC-I cross-presentation will be useful in designing vaccination platforms that aim to trigger an efficient cytotoxic T lymphocyte response.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Autophagy , Cross-Priming/immunology , Histocompatibility Antigens Class I/immunology , Cathepsins/chemistry , Chloroquine/chemistry , Epitopes/chemistry , Epitopes, T-Lymphocyte/chemistry , Humans , Lysosomes/chemistry , Microscopy, Confocal , Nanoparticles/chemistry , Potexvirus , Protein Engineering , RNA/chemistry
2.
Cytometry B Clin Cytom ; 100(1): 79-91, 2021 01.
Article in English | MEDLINE | ID: mdl-33373096

ABSTRACT

Chimeric Antigen Receptor (CAR) T cells are recognized as efficacious therapies with demonstrated ability to produce durable responses in blood cancer patients. Regulatory approvals and acceptance of these unique therapies by patients and reimbursement agencies have led to a significant increase in the number of next generation CAR T clinical trials. Flow cytometry is a powerful tool for comprehensive profiling of individual CAR T cells at multiple stages of clinical development, from product characterization during manufacturing to longitudinal evaluation of the infused product in patients. There are unique challenges with regard to the development and validation of flow cytometric methods for CAR T cells; moreover, the assay requirements for manufacturing and clinical monitoring differ. Based on the collective experience of the authors, this recommendation paper aims to review these challenges and present approaches to address them. The discussion focuses on describing key considerations for the design, optimization, validation and implementation of flow cytometric methods during the clinical development of CAR T cell therapies.


Subject(s)
Flow Cytometry , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/analysis , T-Lymphocytes/cytology , Humans , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
3.
Nat Commun ; 11(1): 219, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924795

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has produced remarkable anti-tumor responses in patients with B-cell malignancies. However, clonal kinetics and transcriptional programs that regulate the fate of CAR-T cells after infusion remain poorly understood. Here we perform TCRB sequencing, integration site analysis, and single-cell RNA sequencing (scRNA-seq) to profile CD8+ CAR-T cells from infusion products (IPs) and blood of patients undergoing CD19 CAR-T immunotherapy. TCRB sequencing shows that clonal diversity of CAR-T cells is highest in the IPs and declines following infusion. We observe clones that display distinct patterns of clonal kinetics, making variable contributions to the CAR-T cell pool after infusion. Although integration site does not appear to be a key driver of clonal kinetics, scRNA-seq demonstrates that clones that expand after infusion mainly originate from infused clusters with higher expression of cytotoxicity and proliferation genes. Thus, we uncover transcriptional programs associated with CAR-T cell behavior after infusion.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Immunotherapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated/immunology , Humans , Kinetics , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, RNA , T-Lymphocytes, Cytotoxic/immunology , Transcriptome
4.
Biol Blood Marrow Transplant ; 24(2): 242-251, 2018 02.
Article in English | MEDLINE | ID: mdl-29024803

ABSTRACT

Mucosal-associated invariant T (MAIT) cells express a semi-invariant Vα7.2+ T cell receptor (TCR) that recognizes ligands from distinct bacterial and fungal species. In neonates, MAIT cells proliferate coincident with gastrointestinal (GI) bacterial colonization. In contrast, under noninflammatory conditions adult MAIT cells remain quiescent because of acquired regulation of TCR signaling. Effects of inflammation and the altered GI microbiota after allogeneic hematopoietic cell transplantation (HCT) on MAIT cell reconstitution have not been described. We conducted an observational study of MAIT cell reconstitution in myeloablative (n = 41) and nonmyeloablative (n = 66) allogeneic HCT recipients and found that despite a rapid and early increase to a plateau at day 30 after HCT, MAIT cell numbers failed to normalize for at least 1 year. Cord blood transplant recipients and those who received post-HCT cyclophosphamide for graft versus host disease (GVHD) prophylaxis had profoundly impaired MAIT cell reconstitution. Sharing of TCRß gene sequences between MAIT cells isolated from HCT grafts and blood of recipients after HCT showed early MAIT cell reconstitution was due at least in part to proliferation of MAIT cells transferred in the HCT graft. Inflammatory cytokines were required for TCR-dependent MAIT cell proliferation, suggesting that bacterial Vα7.2+ TCR ligands might promote MAIT cell reconstitution after HCT. Robust MAIT cell reconstitution was associated with an increased GI abundance of Blautia spp. MAIT cells suppressed proliferation of conventional T cells consistent with a possible regulatory role. Our data identify modifiable factors impacting MAIT cell reconstitution that could influence the risk of GVHD after HCT.


Subject(s)
Allografts/cytology , Mucosal-Associated Invariant T Cells/cytology , Hematopoietic Stem Cell Transplantation , Humans , Kinetics , Peripheral Blood Stem Cell Transplantation , Prospective Studies , Receptors, Antigen, T-Cell , Tissue Donors
5.
Cancer Discov ; 7(12): 1404-1419, 2017 12.
Article in English | MEDLINE | ID: mdl-29025771

ABSTRACT

Lymphodepletion chemotherapy followed by infusion of CD19-targeted chimeric antigen receptor-modified T (CAR-T) cells can be complicated by neurologic adverse events (AE) in patients with refractory B-cell malignancies. In 133 adults treated with CD19 CAR-T cells, we found that acute lymphoblastic leukemia, high CD19+ cells in bone marrow, high CAR-T cell dose, cytokine release syndrome, and preexisting neurologic comorbidities were associated with increased risk of neurologic AEs. Patients with severe neurotoxicity demonstrated evidence of endothelial activation, including disseminated intravascular coagulation, capillary leak, and increased blood-brain barrier (BBB) permeability. The permeable BBB failed to protect the cerebrospinal fluid from high concentrations of systemic cytokines, including IFNγ, which induced brain vascular pericyte stress and their secretion of endothelium-activating cytokines. Endothelial activation and multifocal vascular disruption were found in the brain of a patient with fatal neurotoxicity. Biomarkers of endothelial activation were higher before treatment in patients who subsequently developed grade ≥4 neurotoxicity.Significance: We provide a detailed clinical, radiologic, and pathologic characterization of neurotoxicity after CD19 CAR-T cells, and identify risk factors for neurotoxicity. We show endothelial dysfunction and increased BBB permeability in neurotoxicity and find that patients with evidence of endothelial activation before lymphodepletion may be at increased risk of neurotoxicity. Cancer Discov; 7(12); 1404-19. ©2017 AACR.See related commentary by Mackall and Miklos, p. 1371This article is highlighted in the In This Issue feature, p. 1355.


Subject(s)
Antigens, CD19/immunology , Blood-Brain Barrier/metabolism , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/metabolism , Humans , Treatment Outcome
6.
Blood ; 130(21): 2295-2306, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28924019

ABSTRACT

Lymphodepletion chemotherapy followed by infusion of CD19-specific chimeric antigen receptor-modified (CAR) T cells has produced impressive antitumor responses in patients with refractory CD19+ B-cell malignancies but is often associated with cytokine release syndrome (CRS). Our understanding of CRS continues to evolve, and identification of the kinetics of CRS and predictive clinical and laboratory biomarkers of severity are needed to evaluate strategies to mitigate toxicity. We report the clinical presentation of and identify biomarkers of severe CRS in 133 adult patients who received CD19 CAR T cells. CRS developed in 70% of patients, including 62.5% with grade 1 to 3 CRS (grade 1, 26%; grade 2, 32%; grade 3, 4.5%), 3.8% with grade 4, and 3.8% with grade 5. A majority of cases of grade ≥4 CRS occurred during CAR T-cell dose finding. Multivariable analysis of baseline characteristics identified high marrow tumor burden, lymphodepletion using cyclophosphamide and fludarabine, higher CAR T-cell dose, thrombocytopenia before lymphodepletion, and manufacturing of CAR T cells without selection of CD8+ central memory T cells as independent predictors of CRS. Severe CRS was characterized by hemodynamic instability, capillary leak, and consumptive coagulopathy. Angiopoietin-2 and von Willebrand factor, which are biomarkers of endothelial activation, were increased during severe CRS and also before lymphodepletion in patients who subsequently developed CRS. We describe a classification-tree algorithm to guide studies of early intervention after CAR T-cell infusion for patients at high risk of severe CRS. These data provide a framework for early intervention studies to facilitate safer application of effective CD19 CAR T-cell therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Cytokines/metabolism , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Adult , Aged , Blood Coagulation Disorders/complications , Blood Coagulation Disorders/physiopathology , Blood Coagulation Disorders/therapy , Endothelial Cells/pathology , Female , Fever/etiology , Hematopoiesis , Hemodynamics , Humans , Kinetics , Lymphocyte Count , Male , Middle Aged , Multivariate Analysis , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/therapy , Risk Factors , Syndrome , Treatment Outcome , Young Adult
7.
J Clin Oncol ; 35(26): 3010-3020, 2017 Sep 10.
Article in English | MEDLINE | ID: mdl-28715249

ABSTRACT

Purpose We evaluated the safety and feasibility of anti-CD19 chimeric antigen receptor-modified T (CAR-T) cell therapy in patients with chronic lymphocytic leukemia (CLL) who had previously received ibrutinib. Methods Twenty-four patients with CLL received lymphodepleting chemotherapy and anti-CD19 CAR-T cells at one of three dose levels (2 × 105, 2 × 106, or 2 × 107 CAR-T cells/kg). Nineteen patients experienced disease progression while receiving ibrutinib, three were ibrutinib intolerant, and two did not experience progression while receiving ibrutinib. Six patients were venetoclax refractory, and 23 had a complex karyotype and/or 17p deletion. Results Four weeks after CAR-T cell infusion, the overall response rate (complete response [CR] and/or partial response [PR]) by International Workshop on Chronic Lymphocytic Leukemia (IWCLL) criteria was 71% (17 of 24). Twenty patients (83%) developed cytokine release syndrome, and eight (33%) developed neurotoxicity, which was reversible in all but one patient with a fatal outcome. Twenty of 24 patients received cyclophosphamide and fludarabine lymphodepletion and CD19 CAR-T cells at or below the maximum tolerated dose (≤ 2 × 106 CAR-T cells/kg). In 19 of these patients who were restaged, the overall response rate by IWCLL imaging criteria 4 weeks after infusion was 74% (CR, 4/19, 21%; PR, 10/19, 53%), and 15/17 patients (88%) with marrow disease before CAR-T cells had no disease by flow cytometry after CAR-T cells. Twelve of these patients underwent deep IGH sequencing, and seven (58%) had no malignant IGH sequences detected in marrow. Absence of the malignant IGH clone in marrow of patients with CLL who responded by IWCLL criteria was associated with 100% progression-free survival and overall survival (median 6.6 months follow-up) after CAR-T cell immunotherapy. The progression-free survival was similar in patients with lymph node PR or CR by IWCLL criteria. Conclusion CD19 CAR-T cells are highly effective in high-risk patients with CLL after they experience treatment failure with ibrutinib therapy.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Adenine/analogs & derivatives , Adult , Aged , Antigens, CD19/immunology , Disease-Free Survival , Humans , Immunotherapy, Adoptive/adverse effects , Leukapheresis/methods , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Depletion/methods , Middle Aged , Piperidines , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Remission Induction
8.
Nat Biotechnol ; 35(8): 765-772, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28504668

ABSTRACT

Polymorphisms in the human leukocyte antigen (HLA) class I genes can cause the rejection of pluripotent stem cell (PSC)-derived products in allogeneic recipients. Disruption of the Beta-2 Microglobulin (B2M) gene eliminates surface expression of all class I molecules, but leaves the cells vulnerable to lysis by natural killer (NK) cells. Here we show that this 'missing-self' response can be prevented by forced expression of minimally polymorphic HLA-E molecules. We use adeno-associated virus (AAV)-mediated gene editing to knock in HLA-E genes at the B2M locus in human PSCs in a manner that confers inducible, regulated, surface expression of HLA-E single-chain dimers (fused to B2M) or trimers (fused to B2M and a peptide antigen), without surface expression of HLA-A, B or C. These HLA-engineered PSCs and their differentiated derivatives are not recognized as allogeneic by CD8+ T cells, do not bind anti-HLA antibodies and are resistant to NK-mediated lysis. Our approach provides a potential source of universal donor cells for applications where the differentiated derivatives lack HLA class II expression.


Subject(s)
HLA Antigens/immunology , Killer Cells, Natural/immunology , Pluripotent Stem Cells/immunology , Transplants/immunology , Animals , Female , Graft Rejection/immunology , HLA Antigens/chemistry , HLA Antigens/genetics , Humans , Mice , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/cytology , Transplants/chemistry , Transplants/cytology
9.
Sci Transl Med ; 8(355): 355ra116, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27605551

ABSTRACT

CD19-specific chimeric antigen receptor (CAR)-modified T cells have antitumor activity in B cell malignancies, but factors that affect toxicity and efficacy have been difficult to define because of differences in lymphodepletion and heterogeneity of CAR-T cells administered to individual patients. We conducted a clinical trial in which CD19 CAR-T cells were manufactured from defined T cell subsets and administered in a 1:1 CD4(+)/CD8(+) ratio of CAR-T cells to 32 adults with relapsed and/or refractory B cell non-Hodgkin's lymphoma after cyclophosphamide (Cy)-based lymphodepletion chemotherapy with or without fludarabine (Flu). Patients who received Cy/Flu lymphodepletion had increased CAR-T cell expansion and persistence, and higher response rates [50% complete remission (CR), 72% overall response rate (ORR)] than patients who received Cy-based lymphodepletion without Flu (8% CR, 50% ORR). The CR rate in patients treated with Cy/Flu at the maximally tolerated dose was 64% (82% ORR; n = 11). Cy/Flu minimized the effects of an immune response to the murine single-chain variable fragment component of the CAR, which limited CAR-T cell expansion and clinical efficacy in patients who received Cy-based lymphodepletion without Flu. Severe cytokine release syndrome (sCRS) and grade ≥3 neurotoxicity were observed in 13 and 28% of all patients, respectively. Serum biomarkers, one day after CAR-T cell infusion, correlated with subsequent sCRS and neurotoxicity. Immunotherapy with CD19 CAR-T cells in a defined CD4(+)/CD8(+) ratio allowed identification of correlative factors for CAR-T cell expansion, persistence, and toxicity, and facilitated optimization of lymphodepletion that improved disease response and overall and progression-free survival.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Receptors, Antigen, T-Cell/immunology , Adult , Aged , Biomarkers, Tumor/metabolism , Cell Proliferation , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Female , Humans , Immunotherapy/adverse effects , Lymphocyte Depletion , Lymphocyte Subsets/immunology , Lymphoma, Non-Hodgkin/drug therapy , Male , Middle Aged , Syndrome , Transgenes , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Vidarabine/therapeutic use , Young Adult
10.
J Clin Invest ; 126(6): 2123-38, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27111235

ABSTRACT

BACKGROUND: T cells that have been modified to express a CD19-specific chimeric antigen receptor (CAR) have antitumor activity in B cell malignancies; however, identification of the factors that determine toxicity and efficacy of these T cells has been challenging in prior studies in which phenotypically heterogeneous CAR-T cell products were prepared from unselected T cells. METHODS: We conducted a clinical trial to evaluate CD19 CAR-T cells that were manufactured from defined CD4+ and CD8+ T cell subsets and administered in a defined CD4+:CD8+ composition to adults with B cell acute lymphoblastic leukemia after lymphodepletion chemotherapy. RESULTS: The defined composition product was remarkably potent, as 27 of 29 patients (93%) achieved BM remission, as determined by flow cytometry. We established that high CAR-T cell doses and tumor burden increase the risks of severe cytokine release syndrome and neurotoxicity. Moreover, we identified serum biomarkers that allow testing of early intervention strategies in patients at the highest risk of toxicity. Risk-stratified CAR-T cell dosing based on BM disease burden decreased toxicity. CD8+ T cell-mediated anti-CAR transgene product immune responses developed after CAR-T cell infusion in some patients, limited CAR-T cell persistence, and increased relapse risk. Addition of fludarabine to the lymphodepletion regimen improved CAR-T cell persistence and disease-free survival. CONCLUSION: Immunotherapy with a CAR-T cell product of defined composition enabled identification of factors that correlated with CAR-T cell expansion, persistence, and toxicity and facilitated design of lymphodepletion and CAR-T cell dosing strategies that mitigated toxicity and improved disease-free survival. TRIAL REGISTRATION: ClinicalTrials.gov NCT01865617. FUNDING: R01-CA136551; Life Science Development Fund; Juno Therapeutics; Bezos Family Foundation.


Subject(s)
Immunotherapy, Adoptive/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Adult , Aged , CD4-CD8 Ratio , Disease-Free Survival , Humans , Immunotherapy, Adoptive/adverse effects , Lymphocyte Depletion/methods , Middle Aged , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , T-Lymphocyte Subsets/transplantation , Tumor Burden/immunology , Young Adult
11.
Blood ; 127(20): 2406-10, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26907630

ABSTRACT

Administration of lymphodepletion chemotherapy followed by CD19-specific chimeric antigen receptor (CAR)-modified T cells is a remarkably effective approach to treating patients with relapsed and refractory CD19(+) B-cell malignancies. We treated 7 patients with B-cell acute lymphoblastic leukemia (B-ALL) harboring rearrangement of the mixed lineage leukemia (MLL) gene with CD19 CAR-T cells. All patients achieved complete remission (CR) in the bone marrow by flow cytometry after CD19 CAR-T-cell therapy; however, within 1 month of CAR-T-cell infusion, 2 of the patients developed acute myeloid leukemia (AML) that was clonally related to their B-ALL, a novel mechanism of CD19-negative immune escape. These reports have implications for the management of patients with relapsed and refractory MLL-B-ALL who receive CD19 CAR-T-cell therapy.


Subject(s)
Antigens, CD19/genetics , Histone-Lysine N-Methyltransferase/genetics , Immunotherapy, Adoptive , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Tumor Escape , Antigens, CD19/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Chromosomes, Human, Pair 11/genetics , Clone Cells , Combined Modality Therapy , Female , Humans , Immunophenotyping , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Lymphocyte Depletion , Middle Aged , Neoplastic Stem Cells , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Remission Induction , Salvage Therapy , Translocation, Genetic
12.
PLoS One ; 9(6): e99211, 2014.
Article in English | MEDLINE | ID: mdl-24911872

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is found in multiple malignancies and exerts immunosuppressive effects that are central in protecting tumors from host T lymphocyte rejection. IDO is an enzyme involved in the catabolism of tryptophan resulting in inhibition of T lymphocyte function. While inhibition of IDO enzymatic activity results in tumor rejection, it is still unknown how we can directly target IDO expression within tumors using drugs. We have chosen to interfere with IDO expression by targeting the key-signaling event signal transducer and activator of transcription 1 (STAT1). We evaluated the efficacy of fludarabine, previously described to inhibit STAT1 phosphorylation. Interestingly, fludarabine was efficient in suppressing protein expression and consequently IDO activity in two different cell lines derived from breast cancer and melanoma when IDO was activated with interferon-gamma (IFN-γ) or supernatants prepared from activated T lymphocytes. However, fludarabine had no inhibitory effect on STAT1 phosphorylation. Other IFN-γ-responsive genes were only marginally inhibited by fludarabine. The level of IDO transcript was unaffected by this inhibitor, suggesting the involvement of post-transcriptional control. Strikingly, we have found that the inhibition of proteasome partially protected IDO from fludarabine-induced degradation, indicating that fludarabine induces IDO degradation through a proteasome-dependent pathway. Currently used in the clinic to treat some malignancies, fludarabine has the potential for use in the treatment of human tumors through induction of IDO degradation and consequently, for the promotion of T cell-mediated anti-tumor response.


Subject(s)
Antineoplastic Agents/toxicity , Down-Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Proteasome Endopeptidase Complex/metabolism , Vidarabine/analogs & derivatives , B7-H1 Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/pharmacology , Phosphorylation/drug effects , Protein Stability , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Vidarabine/toxicity
13.
Proteomics ; 13(7): 1108-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436686

ABSTRACT

The stress status of the apoptotic cell can promote phenotypic changes that have important consequences on the immunogenicity of the dying cell. Autophagy is one of the biological processes activated in response to a stressful condition. It is an important mediator of intercellular communications, both by regulating the unconventional secretion of molecules, including interleukin 1ß, and by regulating the extracellular release of ATP from early stage apoptotic cells. Additionally, autophagic components can be released in a caspase-dependent manner by serum-starved human endothelial cells that have engaged apoptotic and autophagic processes. The nature and the components of the extracellular vesicles released by dying autophagic cells are not known. In this study, we have identified extracellular membrane vesicles that are released by human endothelial cells undergoing apoptosis and autophagy, and characterized their biochemical, ultrastructural, morphological properties as well as their proteome. These extracellular vesicles differ from classical apoptotic bodies because they do not contain nucleus components and are released independently of Rho-associated, coiled-coil containing protein kinase 1 activation. Instead, they are enriched with autophagosomes and mitochondria and convey various danger signals, including ATP, suggesting that they could be involved in the modulation of innate immunity.


Subject(s)
Autophagy , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Culture Media, Serum-Free/pharmacology , Enzyme Activation/drug effects , Exosomes/ultrastructure , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Necrosis , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Proteomics , rho-Associated Kinases/metabolism
14.
Clin Cancer Res ; 17(22): 6985-91, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22068654

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-catabolizing enzyme with immune-regulating activities in many contexts, such as fetal protection, allograft protection, and cancer progression. Clinical trials are currently evaluating IDO inhibition with 1-methyltryptophan in cancer immunotherapy. However, the exact role of tryptophan catabolism by IDO in human cancers remains poorly understood. Here, we review several studies that correlate IDO expression in human cancer samples and tumor-draining lymph nodes, with relevant clinical or immunologic parameters. IDO expression in various histologic cancer types seems to decrease tumor infiltration of immune cells and to increase the proportion of regulatory T lymphocytes in the infiltrate. The impact of IDO on different immune cell infiltration leads to the conclusion that IDO negatively regulates the recruitment of antitumor immune cells. In addition, increased IDO expression correlates with diverse tumor progression parameters and shorter patient survival. In summary, in the vast majority of the reported studies, IDO expression is correlated with a less favorable prognosis. As we may see results from the first clinical trials with 1-methyltryptophan in years to come, this review brings together IDO studies from human studies and aims to help appreciate outcomes from current and future trials. Consequently, IDO inhibition seems a promising approach for cancer immunotherapy.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Neoplasms/enzymology , Tumor Escape , Disease Progression , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Prognosis , T-Lymphocytes, Regulatory/immunology
15.
Mol Immunol ; 49(1-2): 253-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21937116

ABSTRACT

The immune system must be under tight control to avoid undesired responses. The enzyme indoleamine 2,3-dioxygenase (IDO) can exert necessary regulating effects by catabolizing tryptophan, leading to the suppression of immune responses in different settings, such as pregnancy and tumor growth. IDO's immuno-suppressive actions are mediated by tryptophan starvation and the accumulation of toxic tryptophan metabolites, resulting in T cell anergy, inhibition of clonal expansion or apoptosis. IDO activity in human macrophages and dendritic cells has been observed after interaction with T lymphocytes, and is triggered by interferon-gamma (IFN-γ) as well as CD40-ligand (CD40L). However, it is unclear whether IDO activity is present in B lymphocytes, which have been identified as having suppressive properties involved in anti-tumor immunity inhibition. In this study, we investigated whether IDO expression is induced in human B cells after exposure to T lymphocyte stimuli and TLR ligands. We report IDO1 and IDO2 mRNA up-regulation by exogenous stimulation with CD40L and IFN-γ. IDO is also upregulated by imiquimod, a TLR 7/8 agonist. In addition, IDO protein is detected after treatment with these exogenous factors or with supernatant from activated CD4(+) T cells. We, however, report weak or absent enzymatic activity from these IDO-expressing cells, as assessed by tryptophan consumption. We conclude that IDO may not be a counter-regulatory mechanism utilized by B lymphocytes to down-regulate immune responses, although its expression is inducible.


Subject(s)
B-Lymphocytes/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism , B-Lymphocytes/immunology , Blotting, Western , Cells, Cultured , Chromatography, High Pressure Liquid , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lymphocyte Activation/immunology , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Toll-Like Receptors/immunology
16.
Vaccine ; 28(34): 5617-26, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20600515

ABSTRACT

Chimeric VLPs made of papaya mosaic virus (PapMV) trigger a CTL response through antigenic presentation of epitopes on MHC class I. Here, a chimeric VLP composed of malva mosaic virus (MaMV) was shown to share similar properties. We demonstrated the capacity of both VLPs to enter human APCs. The chimeric constructions were cross-presented in CD40-activated B lymphocytes leading to in vitro expansion of antigen-specific T lymphocytes. We showed that high concentrations of chimeric MaMV induced cell death, suggesting that some modifications can trigger collateral effects in vitro. Results suggest that potexvirus VLPs are an attractive vaccine platform for inducing a CTL response.


Subject(s)
Antigen Presentation , Cross-Priming , Epitopes/immunology , Potexvirus/immunology , Amino Acid Sequence , B-Lymphocytes/immunology , CD40 Antigens/immunology , Capsid Proteins/immunology , Cell Proliferation , Cloning, Molecular , Humans , Molecular Sequence Data , Protein Engineering , T-Lymphocytes, Cytotoxic/immunology
17.
J Immunol ; 183(12): 7752-60, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933867

ABSTRACT

Previous cancer vaccination approaches have shown some efficiency in generating measurable immune responses, but they have rarely led to tumor regression. It is therefore possible that tumors emerge with the capacity to down-regulate immune counterparts, through the local production of immunosuppressive molecules, such as IDO. Although it is known that IDO exerts suppressive effects on T cell functions, the mechanisms of IDO regulation in tumor cells remain to be characterized. Here, we demonstrate that activated T cells can induce functional IDO expression in breast and kidney tumor cell lines, and that this is partly attributable to IFN-gamma. Moreover, we found that IL-13, a Th2 cytokine, has a negative modulatory effect on IDO expression. Furthermore, we report IDO expression in the majority of breast and kidney carcinoma samples, with infiltration of activated Th1-polarized T cells in human tumors. These findings demonstrate complex control of immune activity within tumors. Future immune therapeutic interventions should thus include strategies to counteract these negative mechanisms.


Subject(s)
Breast Neoplasms/immunology , Carcinoma, Renal Cell/immunology , Gene Expression Regulation, Enzymologic/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Lymphocyte Activation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/immunology , Coculture Techniques , Humans , Immunophenotyping , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/metabolism , Interferon-gamma/physiology , Lymphocyte Activation/genetics , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Th1 Cells/enzymology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/enzymology , Th2 Cells/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...