Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Contemp Clin Trials Commun ; 29: 100992, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36105266

ABSTRACT

Regulation (EU) No 536/2014 (Clinical Trial Regulation, CTR) offers two precious tools to academic clinical research in Italy: - The right to transfer not-for-profit clinical trials data and results for registration purposes, and co-sponsorship. - The right to transfer data reduces the time needed to make innovative therapeutical agents and therapies accessible to the patient. Co-sponsorship, on the other hand, allows the establishment of a partnership between entities with different missions, ideals and attitudes, sharing - nevertheless - the same ultimate goal: meeting the patient's medical needs. Co-sponsorship facilitates collaboration among experts, which allows knowledge sharing, thus guaranteeing, to each contributor, recognition for their own contributions to a complex activity such as a clinical trial. However, the above-mentioned Regulation poses important challenges, especially in terms of infrastructural efficiency, which is demanding, especially for those entities suffering organizational inadequacies: unfortunately, inefficiency is sometimes a structural problem in the academic clinical environment. This publication focuses on the specific innovative aspects introduced by CTR. It also highlights the possible difficulties to be addressed by their implementation.

3.
Nat Commun ; 12(1): 4844, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381053

ABSTRACT

Acute leukemia relapsing after chemotherapy plus allogeneic hematopoietic stem cell transplantation can be treated with donor-derived T cells, but this is hampered by the need for donor/recipient MHC-matching and often results in graft-versus-host disease, prompting the search for new donor-unrestricted strategies targeting malignant cells. Leukemia blasts express CD1c antigen-presenting molecules, which are identical in all individuals and expressed only by mature leukocytes, and are recognized by T cell clones specific for the CD1c-restricted leukemia-associated methyl-lysophosphatidic acid (mLPA) lipid antigen. Here, we show that human T cells engineered to express an mLPA-specific TCR, target diverse CD1c-expressing leukemia blasts in vitro and significantly delay the progression of three models of leukemia xenograft in NSG mice, an effect that is boosted by mLPA-cellular immunization. These results highlight a strategy to redirect T cells against leukemia via transfer of a lipid-specific TCR that could be used across MHC barriers with reduced risk of graft-versus-host disease.


Subject(s)
Antigens, CD1/immunology , Glycoproteins/immunology , Leukemia/immunology , Lysophospholipids/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tissue Donors , Animals , Antigen Presentation , Antigens, CD1/metabolism , Glycoproteins/metabolism , Humans , Immunotherapy, Adoptive , Leukemia/metabolism , Leukemia/therapy , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
4.
N Engl J Med ; 383(17): 1613-1623, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33085860

ABSTRACT

BACKGROUND: Outcomes in patients with Philadelphia chromosome (Ph)-positive acute lymphoblastic leukemia (ALL) have improved with the use of tyrosine kinase inhibitors. Molecular remission is a primary goal of treatment. METHODS: We conducted a phase 2 single-group trial of first-line therapy in adults with newly diagnosed Ph-positive ALL (with no upper age limit). Dasatinib plus glucocorticoids were administered, followed by two cycles of blinatumomab. The primary end point was a sustained molecular response in the bone marrow after this treatment. RESULTS: Of the 63 patients (median age, 54 years; range, 24 to 82) who were enrolled, a complete remission was observed in 98%. At the end of dasatinib induction therapy (day 85), 29% of the patients had a molecular response, and this percentage increased to 60% after two cycles of blinatumomab; the percentage of patients with a molecular response increased further after additional blinatumomab cycles. At a median follow-up of 18 months, overall survival was 95% and disease-free survival was 88%; disease-free survival was lower among patients who had an IKZF1 deletion plus additional genetic aberrations (CDKN2A or CDKN2B, PAX5, or both [i.e., IKZF1 plus]). ABL1 mutations were detected in 6 patients who had increased minimal residual disease during induction therapy, and all these mutations were cleared by blinatumomab. Six relapses occurred. Overall, 21 adverse events of grade 3 or higher were recorded. A total of 24 patients received a stem-cell allograft, and 1 death was related to transplantation (4%). CONCLUSIONS: A chemotherapy-free induction and consolidation first-line treatment with dasatinib and blinatumomab that was based on a targeted and immunotherapeutic strategy was associated with high incidences of molecular response and survival and few toxic effects of grade 3 or higher in adults with Ph-positive ALL. (Funded by Associazione Italiana per la Ricerca sul Cancro and others; GIMEMA LAL2116 D-ALBA EudraCT number, 2016-001083-11; ClinicalTrials.gov number, NCT02744768.).


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dasatinib/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adult , Aged , Aged, 80 and over , Allografts , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Consolidation Chemotherapy , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Induction Chemotherapy , Male , Middle Aged , Mutation , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Remission Induction , Survival Analysis , Treatment Outcome
5.
J Interferon Cytokine Res ; 36(7): 462-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27379868

ABSTRACT

The effective deployment of immune responses depends on the activation of well-defined signaling pathways that interact with cell-intrinsic epigenetic features to control the activation of stimulus-specific and kinetically accurate gene expression programs. The interferon regulatory factors (IRFs) are critical regulators of both development and activation of distinct cells of the immune system. The influence of the IRF family transcription factors (TFs) on immune responses is mainly linked to the control of the type I interferon (IFN) system, which must be not only rapidly activated to mount an efficient host response, but also tightly regulated to avoid detrimental effects. In this review we discuss the different regulatory layers controlling IRF activity and specifically IRF-mediated IFN responses. We also highlight how the interplay between the DNA-binding properties of the IRF family TFs and the epigenetic landscape controls cell- and context-specific responses.


Subject(s)
Gene Expression Regulation , Genomics , Interferon Regulatory Factors/metabolism , Transcription, Genetic , Animals , Chromatin Assembly and Disassembly , Genomics/methods , Humans , Protein Binding , Signal Transduction
6.
Genes Dev ; 29(4): 394-408, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25637355

ABSTRACT

The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8-PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF-AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome. While constitutively expressed IRF8-dependent genes contained only sites mediating basal IRF8/PU.1 recruitment, inducible IRF8-dependent genes contained variable combinations of constitutive and inducible sites. Overall, these data show at the genome scale how the same TF can be linked to constitutive and inducible gene regulation via distinct combinations of alternative DNA-binding sites.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Macrophages/metabolism , Animals , Cells, Cultured , Interferon Regulatory Factors/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Protein Binding
7.
EMBO J ; 32(6): 816-28, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23422957

ABSTRACT

Dendritic cells (DC) are required for priming antigen-specific T cells and acquired immunity to many important human pathogens, including Mycobacteriuim tuberculosis (TB) and influenza. However, inappropriate priming of auto-reactive T cells is linked with autoimmune disease. Understanding the molecular mechanisms that regulate the priming and activation of naïve T cells is critical for development of new improved vaccines and understanding the pathogenesis of autoimmune diseases. The serine/threonine kinase IKKα (CHUK) has previously been shown to have anti-inflammatory activity and inhibit innate immunity. Here, we show that IKKα is required in DC for priming antigen-specific T cells and acquired immunity to the human pathogen Listeria monocytogenes. We describe a new role for IKKα in regulation of IRF3 activity and the functional maturation of DC. This presents a unique role for IKKα in dampening inflammation while simultaneously promoting adaptive immunity that could have important implications for the development of new vaccine adjuvants and treatment of autoimmune diseases.


Subject(s)
Adaptive Immunity/genetics , Cell Differentiation/genetics , Dendritic Cells/physiology , I-kappa B Kinase/physiology , Infections/immunology , Adoptive Transfer/methods , Animals , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Infections/genetics , Inflammation/genetics , Inflammation/immunology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
8.
Clin Cancer Res ; 16(3): 784-9, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103670

ABSTRACT

Tumor-associated macrophages (TAM) have been linked with the progression of cancer by favoring tumor angiogenesis, growth, and metastasis. The precise mechanisms that maintain the protumor phenotype of TAM are poorly understood, but recent research has highlighted a number of signaling pathways that are important in TAM phenotype and function. Nuclear factor-kappaB (NF-kappaB) is considered the master regulator of inflammatory and immune responses. Recently several genetic studies have indicated NF-kappaB is an important pathway in TAM for the integration of signals from the tumor microenvironment that promote carcinogenesis. This review will focus on the role of NF-kappaB in TAM and the potential of targeting this pathway as a novel therapeutic strategy against cancer.


Subject(s)
Macrophages/immunology , NF-kappa B/physiology , Neoplasms/immunology , Cell Line, Tumor , Disease Progression , Drug Delivery Systems , Gene Expression Regulation , Humans , Inflammation/etiology , NF-kappa B/antagonists & inhibitors , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction
9.
J Transl Med ; 7: 5, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19144161

ABSTRACT

BACKGROUND: Angiostatin, an endogenous angiogenesis inhibitor, is a fragment of plasminogen. Its anti-angiogenic activity was discovered with functional assays in vivo, however, its direct action on endothelial cells is moderate and identification of definitive mechanisms of action has been elusive to date. We had previously demonstrated that innate immune cells are key targets of angiostatin, however the pathway involved in this immune-related angiogenesis inhibition was not known. Here we present evidence that IL-12, a principal TH1 cytokine with potent anti-angiogenic activity, is the mediator of angiostatin's activity. METHODS: Function blocking antibodies and gene-targeted animals were employed or in vivo studies using the subcutaneous matrigel model of angiogenesis. Quantitative real-time PCR were used to assess modulation of cytokine production in vitro. RESULTS: Angiostatin inhibts angiogenesis induced by VEGF-TNFalpha or supernatants of Kaposi's Sarcoma cells (a highly angiogenic and inflammation-associated tumor). We found that function-blocking antibodies to IL-12 reverted angiostatin induced angiogenesis inhibition. The use of KO animal models revealed that angiostatin is unable to exert angiogenesis inhibition in mice with gene-targeted deletions of either the IL-12 specific receptor subunit IL-12Rbeta2 or the IL-12 p40 subunit. Angiostatin induces IL-12 mRNA synthesis by human macrophages in vitro, suggesting that these innate immunity cells produce IL-12 upon angiostatin stimulation and could be a major cellular mediator. CONCLUSION: Our data demonstrate that an endogenous angiogenesis inhibitor such as angiostatin act on innate immune cells as key targets in inflammatory angiogenesis. Angiostatin proves to be anti-angiogenic as an immune modulator rather than a direct anti-vascular agent. This article is dedicated to the memory of Prof Judah Folkman for his leadership and for encouragement of these studies.


Subject(s)
Angiogenesis Inhibitors/immunology , Angiostatins/immunology , Immune System/immunology , Immunity, Innate/immunology , Interleukin-12/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Chemokine CCL2/immunology , Humans , Interleukin-8/immunology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , Neovascularization, Pathologic , Tumor Necrosis Factor-alpha/immunology , Vascular Endothelial Growth Factor A/immunology
10.
Blood ; 112(9): 3723-34, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18694997

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that patrol tissues to sense danger signals and activate specific immune responses. In addition, they also play a role in inflammation and tissue repair. Here, we show that oxygen availability is necessary to promote full monocyte-derived DC differentiation and maturation. Low oxygen tension (hypoxia) inhibits expression of several differentiation and maturation markers (CD1a, CD40, CD80, CD83, CD86, and MHC class II molecules) in response to lipopolysaccharide (LPS), as well as their stimulatory capacity for T-cell functions. These events are paralleled by impaired up-regulation of the chemokine receptor CCR7, an otherwise necessary event for the homing of mature DCs to lymph nodes. In contrast, hypoxia strongly up-regulates production of proinflammatory cytokines, particularly TNFalpha and IL-1beta, as well as the inflammatory chemokine receptor CCR5. Subcutaneous injection of hypoxic DCs into the footpads of mice results in defective DC homing to draining lymph nodes, but enhanced leukocyte recruitment at the site of injection. Thus, hypoxia uncouples the promotion of inflammatory and tissue repair from sentinel functions in DCs, which we suggest is a safeguard mechanism against immune reactivity to damaged tissues.


Subject(s)
Cell Hypoxia/immunology , Cell Hypoxia/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chemokines/metabolism , Chemotaxis , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
11.
Semin Cancer Biol ; 18(5): 349-55, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18467122

ABSTRACT

Macrophages are a fundamental part of the innate defense mechanisms, which can promote specific immunity by inducing T cell recruitment and activation. Despite this, their presence within the tumour microenvironment has been associated with enhanced tumour progression and shown to promote cancer cell growth and spread, angiogenesis and immunosuppression. This paradoxical role of macrophages in cancer finds an explanation in their functional plasticity, that may result in the polarized expression of either pro- or anti-tumoural functions. Key players in the setting of their phenotype are the microenvironmental signals to which macrophages are exposed, which selectively tune their functions within a functional spectrum encompassing the M1 and M2 extremes. Here, we discuss recent findings suggesting that targeting tumour-associated macrophages (TAMs) polarization may represent a novel therapeutic strategy against cancer.


Subject(s)
Macrophages/physiology , Neoplasms/immunology , Neoplasms/physiopathology , Animals , Cytokines/immunology , Cytokines/metabolism , Disease Progression , Humans , Immunity, Active , Inflammation/immunology , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Macrophage Activation , Macrophages/immunology , Monocytes/immunology , Monocytes/metabolism
12.
Expert Opin Ther Targets ; 11(9): 1219-29, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17845147

ABSTRACT

Clinical and experimental evidence have highlighted that a major leukocyte population present in tumours, the so-called tumour-associated macrophages (TAM), is the principal component of the leukocyte infiltrate supporting tumour growth. Over the years the mechanisms supporting the protumoural functions of TAM have become increasingly clear and in several experimental tumour models, the activation of an inflammatory response (most frequently mediated by macrophages) has been shown to play an essential role for full neoplastic transformation and progression. This evidence strongly supports the idea that TAM are central orchestrators of the inflammatory networks expressed in the tumour microenvironment, and suggest these cells as possible targets of anticancer therapies.


Subject(s)
Macrophages/immunology , Neoplasms/immunology , Animals , Antineoplastic Agents/therapeutic use , Humans , Hypoxia-Inducible Factor 1/metabolism , Macrophages/drug effects , NF-kappa B/immunology , Neoplasms/drug therapy , Neoplasms/metabolism
13.
Adv Exp Med Biol ; 604: 67-86, 2007.
Article in English | MEDLINE | ID: mdl-17695721

ABSTRACT

Recent years have seen a renaissance of the inflammation-cancer connection stemming from different lines of work and leading to a generally accepted paradigm (Balkwill and Mantovani 2001; Mantovani et al. 2002; Coussens and Werb 2002; Balkwill et al. 2005). An inflammatory component is present in the microenvironment of most neoplastic tissues, including those not causally related to an obvious inflammatory process Cancer-associated inflammation includes: the infiltration of white blood cells, prominently phagocytic cells called macrophages (TAM) (Paik et al. 2004); the presence of polipeptide messengers of inflammation (cytokines such as tumor necrosis factor (TNF) or interleukin-1 (IL-1), chemokines such as CCL2); the occurrence of tissue remodelling and angiogenesis. Chemokines have emerged as a key component of the tumor microenvironment which shape leukocyte recruitment and function (Pollard 2004). Strong direct evidence suggests that cancer associated inflammation promotes tumor growth and progression. Therapeutic targeting of cancer promoting inflammatory reactions is in its infancy, and its development is crucially dependent on defining the underlying cellular and molecular mechanisms in relevant systems.


Subject(s)
Macrophages/metabolism , Neoplasms/metabolism , Animals , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Progression , Humans , Inflammation , Interleukin-1/metabolism , Macrophage Activation , Models, Biological , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...