Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 25(17): 5231-5241, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31171545

ABSTRACT

PURPOSE: This study was aimed at evaluating the feasibility, safety, immunologic and clinical responses in patients with follicular lymphoma treated with monocyte-derived dendritic cells generated in the presence of IFNα and GM-CSF (IFN-DC) in combination with low doses of rituximab. PATIENTS AND METHODS: Firstly, we analyzed in vitro and in vivo the immunologic properties of IFN-DC against follicular lymphoma. Thus, we performed a phase I trial in 8 patients with refractory and relapsed follicular lymphoma based on sequential intranodal injections of low-dose of rituximab and unloaded IFN-DC and report the safety, clinical, and immunologic results of the enrolled patients. RESULTS: Preclinical studies indicated that IFN-DC can synergize with rituximab leading to increased cytotoxicity and T-cell tumor infiltration. The clinical evaluation showed that the combined treatment was totally safe. The overall response rate was 50%, PET-negative complete response rate 37%, and remission is still ongoing in 2/4 of responding patients (median follow-up 26 months, range 11-47). Notably, following the combined therapy all patients showed induction/enhancement of T-cell responses by CD107 degranulation or IFNγ ELISPOT assay against patient-specific tumor IGHV sequences. CONCLUSIONS: These results represent the proof-of-principle on the effectiveness of unloaded IFN-DC in inducing durable clinical responses and promoting induction of tumor-specific peripheral T cells, thus suggesting the occurrence of an effective endogenous antitumor vaccination. The overall findings indicate that some unique properties of IFN-DC can be successfully exploited to induce/enhance antitumor responses, thus representing a valuable antitumor strategy for novel and more effective combination therapies in patients with cancer.


Subject(s)
Dendritic Cells/transplantation , Immunotherapy, Adoptive/methods , Lymphoma, Follicular/therapy , Neoplasm Recurrence, Local/therapy , Rituximab/administration & dosage , Adult , Aged , Animals , Antineoplastic Agents, Immunological/administration & dosage , Combined Modality Therapy , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Resistance, Neoplasm , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Injections, Intralymphatic , Interferon-alpha/pharmacology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Remission Induction , Salvage Therapy , Xenograft Model Antitumor Assays
2.
J Transl Med ; 13: 139, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25933939

ABSTRACT

BACKGROUND: Advanced melanoma patients have an extremely poor long term prognosis and are in strong need of new therapies. The recently developed targeted therapies have resulted in a marked antitumor effect, but most responses are partial and some degree of toxicity remain the major concerns. Dendritic cells play a key role in the activation of the immune system and have been typically used as ex vivo antigen-loaded cell drugs for cancer immunotherapy. Another approach consists in intratumoral injection of unloaded DCs that can exploit the uptake of a wider array of tumor-specific and individual unique antigens. However, intratumoral immunization requires DCs endowed at the same time with properties typically belonging to both immature and mature DCs (i.e. antigen uptake and T cell priming). DCs generated in presence of interferon-alpha (IFN-DCs), due to their features of partially mature DCs, capable of efficiently up-taking, processing and cross-presenting antigens to T cells, could successfully carry out this task. Combining intratumoral immunization with tumor-destructing therapies can induce antigen release in situ, facilitating the injected DCs in triggering an antitumor immune response. METHODS: We tested in a phase I clinical study in advanced melanoma a chemo-immunotherapy approach based on unloaded IFN-DCs injected intratumorally one day after administration of dacarbazine. Primary endpoint of the study was treatment safety and tolerability. Secondary endpoints were immune and clinical responses of patients. RESULTS: Six patients were enrolled, and only three completed the treatment. The chemo-immunotherapy was well tolerated with no major side effects. Three patients showed temporary disease stabilization and two of them showed induction of T cells specific for tyrosinase, NY-ESO-1 and gp100. Of interest, one patient showing a remarkable long-term disease stabilization kept showing presence of tyrosinase specific T cells in PBMC and high infiltration of memory T cells in the tumor lesion at 21 months. CONCLUSION: We tested a chemo-immunotherapeutic approach based on IFN-DCs injected intratumorally one day after DTIC in advanced melanoma. The treatment was well tolerated, and clinical and immunological responses, including development of vitiligo, were observed, therefore warranting additional clinical studies aimed at evaluating efficacy of this approach. TRIAL REGISTRATION: Trial Registration Number not publicly available due to EudraCT regulations: https://www.clinicaltrialsregister.eu/doc/EU_CTR_FAQ.pdf.


Subject(s)
Dacarbazine/chemistry , Dendritic Cells/cytology , Drug Therapy/methods , Immunotherapy/methods , Injections, Intralesional , Interferon-alpha/metabolism , Melanoma/therapy , Adult , Aged , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Combined Modality Therapy/methods , Female , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/cytology , Male , Membrane Proteins/metabolism , Microscopy, Confocal , Middle Aged , Monocytes/metabolism , Monophenol Monooxygenase/metabolism , Vitiligo/chemically induced , gp100 Melanoma Antigen/metabolism
3.
Blood Transfus ; 13(3): 464-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25761309

ABSTRACT

BACKGROUND: Great interest has been raised recently by the design of new adoptive immunotherapeutic strategies based on the in vivo infusion of ex vivo-expanded and activated natural killer (NK) cells. The development of good manufacturing practice (GMP) methods for the efficient production of fully functional NK cells is mandatory for clinical application. MATERIALS AND METHODS: Peripheral blood mononuclear cells were obtained by leukapheresis and processed in the GMP facility. For NK-cell enrichment, a two-step immunomagnetic procedure consisting of CD3(+) T-cell depletion followed by CD56(+) cell positive selection was used. Isolated NK cells were suspended in serum-free medium containing autologous plasma, interleukin (IL)-2 and IL-15 in the presence of irradiated autologous feeder cells and cultured for 14 days at 37 °C. IL-2 and IL-15 were also added during the last 24 hours of culture. Expanded cells underwent full quality control testing for cytogenetic characteristics, viability, sterility, phenotype and endotoxin status; functional tests, such as degranulation assays and cytotoxicity, were performed on expanded NK cells before cryopreservation and after thawing. RESULTS: NK-cell populations expanded on average 15.7±4.7 fold by day 14, with a viability of 96% ±0.5. At the end of the incubation period, 97% ±1.1 of the expanded population was CD56(+) NK cells; these effector cells showed significant up-regulation of the activating receptors NKG2D and DNAM-1. Functional tests demonstrated that expanded NK cells are fully functional with no difference whether tested before cryopreservation or after thawing. DISCUSSION: These data provide the basis for developing new NK-cell-based immunotherapeutic strategies for the treatment of patients with cancer.


Subject(s)
Cell Culture Techniques/methods , Killer Cells, Natural/cytology , Cell Culture Techniques/instrumentation , Cryopreservation/methods , Female , Humans , Killer Cells, Natural/transplantation , Leukapheresis/methods , Male
4.
Immunobiology ; 216(5): 537-47, 2011 May.
Article in English | MEDLINE | ID: mdl-21093097

ABSTRACT

We have investigated the molecular mechanisms underlying the peculiar cross-presentation efficiency of human dendritic cells (DCs) differentiated from monocytes in the presence of Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and Interferon (IFN)-α (IFN-DCs). To this end, we evaluated the capability of IFN-DCs to present and cross-present epitopes derived from Epstein-Barr Virus (EBV) or human melanoma-associated antigens after exposure to cell lysates or apoptotic cells. In an autologous setting, IFN-DCs loaded with Lymphoblastoid Cell Lines (LCL) lysates or apoptotic LCL were highly efficient in expanding, respectively, EBV-specific class II- or class I-restricted memory T cell responses. Of note, IFN-DCs loaded with apoptotic LCL were more potent than fully mature DCs in triggering the cytotoxicity of CD8(+) T lymphocytes recognizing a subdominant HLA-A*0201-restricted epitope derived from EBV latent membrane protein 2 (LMP2). In addition, IFN-DCs loaded with apoptotic human melanoma cells were highly efficient in cross-presenting the MART-1(27-35) epitope to a specific CD8(+) cytotoxic T cell clone, and this functional activity was proteasome-dependent. These IFN-DC properties were associated with an enhanced expression of all the immunoproteasome subunits as well as of TAP-1, TAP-2 and tapasin, and, interestingly, to a higher proteasome proteolytic activity as compared to immature or mature DCs. Altogether, these results highlight new mechanisms by which IFN-α influences antigen processing and cross-presentation ability of monocyte-derived DCs, with potentially important implications for the development of DC-based therapeutic vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Immunotherapy, Adoptive , Interferon-alpha/metabolism , Proteasome Endopeptidase Complex/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Cross-Priming , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/immunology , Epitopes/metabolism , Epitopes, T-Lymphocyte/metabolism , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Immunologic Memory , Interferon-alpha/immunology , Lymphocyte Activation , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/immunology , Protein Binding , Viral Matrix Proteins/metabolism
5.
AIDS ; 18(9): 1271-80, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15362659

ABSTRACT

OBJECTIVES: Herpes simplex virus (HSV) infections have been associated with reactivation of HIV-1 replication and increases of HIV-1-load in plasma of co-infected individuals. The present authors have previously reported that in epithelial cells HSV-1 induces the IkappaB-kinase (IKK) causing persistent activation of NF-kappaB, a critical regulator of HIV-1 replication. The present study was performed to investigate whether HSV-1-infection could induce IKK-mediated NF-kappaB activation and enhance HIV-1 expression in human T cells, and to analyze the effect of the IKK-inhibitor prostaglandin A1 (PGA1) and other prostanoids on the NF-kappaB-mediated HSV-HIV interaction. DESIGN AND METHODS: Induction of IKK and NF-kappaB activity was determined in lymphoblastoid Jurkat cells and HIV-1 chronically-infected H9 and ACH-2 cells by kinase assay and electrophoretic mobility shift assay, respectively. The effect of HSV-1 and different prostanoids on HIV-1 expression and replication was determined in Jurkat cells transfected with HIV-1-LTR-driven reporter genes, and in H9 and ACH-2 cells by p24-antigen level evaluation. The role of NF-kappaB in HSV-1-induced HIV-1 expression was investigated by using the IkappaBalpha dominant-negative IkappaBalpha-AA in co-transfection experiments. RESULTS: In human T lymphoblastoid cells HSV-1 potently induces IKK activity, causing a persistent induction of NF-kappaB. HSV-1-induced IKK and NF-kappaB function results in transactivation of HIV-1-LTR-regulated genes and induction of HIV-1 replication in chronically-infected T cells. The cyclopentenone PGA1 inhibits HSV-1-induced IKK and NF-kappaB activities, blocking HIV-1-LTR-driven expression and preventing HSV-1-induced HIV-1 replication in co-infected cells. CONCLUSIONS: The results indicate that IKK is a key factor in triggering HSV-1-induced HIV-1 transcription in chronically-infected cells and identify cyclopentenone prostanoids as potent inhibitors of HSV-1-induced HIV-1 reactivation.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Prostaglandins A/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Electrophoretic Mobility Shift Assay , HIV Core Protein p24/metabolism , HIV Infections/metabolism , Herpes Simplex/metabolism , Humans , I-kappa B Kinase , Jurkat Cells , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/enzymology , Transcription, Genetic , Vero Cells , Viral Load , Virus Activation/drug effects
6.
Blood ; 103(3): 980-7, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14525781

ABSTRACT

Chronic myelogenous leukemia (CML) is a malignant myeloproliferative disease arising from the clonal expansion of a stem cell expressing the bcr/abl oncogene. CML patients frequently respond to treatment with interferon-alpha (IFN-alpha), even though the mechanisms of the response remain unclear. In the present study, we evaluated the role of IFN-alpha in differentiation and activity of monocyte-derived dendritic cells (DCs) from CML patients as well as in modulation of the cell response to lipopolysaccharide (LPS). Treatment of CML monocytes with IFN-alpha and granulocyte-macrophage colony-stimulating factor (GM-CSF) resulted in the rapid generation of activated DCs (CML-IFN-DCs) expressing interleukin-15 (IL-15) and the antiapoptotic bcl-2 gene. These cells were fully competent to induce IFN-gamma production by cocultured autologous T lymphocytes and expansion of CD8(+) T cells. LPS treatment of CML-IFN-DCs, but not of immature DCs generated in the presence of IL-4/GM-CSF, induced the generation of CD8(+) T cells reactive against autologous leukemic CD34(+) cells. Altogether, these results suggest that (1) the generation of highly active monocyte-derived DCs could be important for the induction of an antitumor response in IFN-treated CML patients and (2) IFN-alpha can represent a valuable cytokine for the rapid generation of active monocyte-derived DCs to be utilized for vaccination strategies of CML patients.


Subject(s)
Dendritic Cells/drug effects , Interferon Type I/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Monocytes/drug effects , Adult , Aged , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Chemokine CXCL10 , Chemokines, CXC/genetics , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon-gamma/biosynthesis , Interleukin-15/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...