Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Commun ; 14(1): 5529, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37684243

ABSTRACT

Immune checkpoint inhibitors cause side effects ranging from autoimmune endocrine disorders to severe cardiotoxicity. Periodic Fasting mimicking diet (FMD) cycles are emerging as promising enhancers of a wide range of cancer therapies including immunotherapy. Here, either FMD cycles alone or in combination with anti-OX40/anti-PD-L1 are much more effective than immune checkpoint inhibitors alone in delaying melanoma growth in mice. FMD cycles in combination with anti-OX40/anti-PD-L1 also show a trend for increased effects against a lung cancer model. As importantly, the cardiac fibrosis, necrosis and hypertrophy caused by immune checkpoint inhibitors are prevented/reversed by FMD treatment in both cancer models whereas immune infiltration of CD3+ and CD8+ cells in myocardial tissues and systemic and myocardial markers of oxidative stress and inflammation are reduced. These results indicate that FMD cycles in combination with immunotherapy can delay cancer growth while reducing side effects including cardiotoxicity.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Lung Neoplasms , Animals , Mice , Cardiotoxicity , Immune Checkpoint Inhibitors/adverse effects , Fasting , Diet , Immunotherapy/adverse effects , Lung Neoplasms/therapy , Myocardium
2.
Eur Rev Med Pharmacol Sci ; 27(17): 8269-8279, 2023 09.
Article in English | MEDLINE | ID: mdl-37750654

ABSTRACT

OBJECTIVE: Hemorrhoids are a common anorectal disease that causes pain, itching, and burning. The prevalence of hemorrhoids is estimated to be as high as 36% in the general population, with approximately 50% of individuals experiencing symptomatic hemorrhoids at least once in their life. Middle age, obesity, and pregnancy are risk factors. The combination of tribenoside and lidocaine (Procto-Glyvenol©, Recordati) has been used for decades to treat low-grade hemorrhoids, and its efficacy and safety are well supported by clinical experience. Tribenoside has been shown to have an anti-inflammatory effect, ameliorate the local microcirculation and vascular tone, and promote the healing of basement membrane. However, the molecular mechanism behind its wound-healing properties is still unclear. MATERIALS AND METHODS: Human dermal fibroblasts were used to test the effect of tribenoside on cell proliferation, cell migration, and production of reactive oxygen species in vitro. Full-thickness excisional wound model in rats was used to test the wound-healing properties of Procto-Glyvenol© in vivo. RESULTS: Tribenoside has been found to increase the migration rate of fibroblasts in vitro and to improve the wound healing process by promoting re-epithelialization in rats. Furthermore, novel antioxidant activity of tribenoside has been reported, which may represent a further mechanism of action in wound healing. CONCLUSIONS: Procto-Glyvenol© improves the natural healing process of wounds by stimulating cell migration and protecting against the toxic effects of reactive oxygen species. Therefore, it may represent a first-line treatment for hemorrhoids, which are a significant medical and socioeconomic problem that can deteriorate the quality of life.


Subject(s)
Hemorrhoids , Middle Aged , Female , Pregnancy , Humans , Animals , Rats , Quality of Life , Reactive Oxygen Species , Lidocaine/pharmacology , Wound Healing
3.
ESMO Open ; 8(4): 101590, 2023 08.
Article in English | MEDLINE | ID: mdl-37393630

ABSTRACT

BACKGROUND: Gene expression profiling (GEP)-based prognostic signatures are being rapidly integrated into clinical decision making for systemic management of breast cancer patients. However, GEP remains relatively underdeveloped for locoregional risk assessment. Yet, locoregional recurrence (LRR), especially early after surgery, is associated with poor survival. PATIENTS AND METHODS: GEP was carried out on two independent luminal-like breast cancer cohorts of patients developing early (≤5 years after surgery) or late (>5 years) LRR and used, by a training and testing approach, to build a gene signature able to intercept women at risk of developing early LRR. The GEP data of two in silico datasets and of a third independent cohort were used to explore its prognostic value. RESULTS: Analysis of the first two cohorts led to the identification of three genes, CSTB, CCDC91 and ITGB1, whose expression, derived by principal component analysis, generated a three-gene signature significantly associated with early LRR in both cohorts (P value <0.001 and 0.005, respectively), overcoming the discriminatory capability of age, hormone receptor status and therapy. Remarkably, the integration of the signature with these clinical variables led to an area under the curve of 0.878 [95% confidence interval (CI) 0.810-0.945]. In in silico datasets we found that the three-gene signature retained its association, showing higher values in the early relapsed patients. Moreover, in the third additional cohort, the signature significantly associated with relapse-free survival (hazard ratio 1.56, 95% CI 1.04-2.35). CONCLUSIONS: Our three-gene signature represents a new exploitable tool to aid treatment choice in patients with luminal-like breast cancer at risk of developing early recurrence.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/drug therapy , Prognosis , Transcriptome , Risk Assessment
4.
Virchows Arch ; 475(6): 799, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31664506

ABSTRACT

The first and family names of the authors were interchanged and are now presented correctly. The original article has been corrected.

6.
Virchows Arch ; 475(4): 513-518, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31388760

ABSTRACT

An accurate diagnosis of clinically distinct subgroups of aggressive mature B cell lymphomas is crucial for the choice of proper treatment. Presently, precise recognition of these disorders relies on the combination of morphological, immunophenotypical, and cytogenetic/molecular features. The diagnostic workup in such situations implies the application of costly and time-consuming analyses, which are not always required, since an intensified treatment option is reasonably reserved to fit patients. The Italian Group of Haematopathology proposes herein a practical algorithm for the diagnosis of aggressive mature B cell lymphomas based on a stepwise approach, aimed to select cases deserving molecular analysis, in order to optimize time and resources still assuring the optimal management for any patient.


Subject(s)
Algorithms , Lymphoma, B-Cell/diagnosis , Humans , Immunophenotyping/methods , In Situ Hybridization, Fluorescence/methods
7.
Cytokine Growth Factor Rev ; 44: 1-10, 2018 12.
Article in English | MEDLINE | ID: mdl-30393044

ABSTRACT

The eighth annual conference of "Innovative therapy, monoclonal antibodies, and beyond" was held in Milan on Jan. 26, 2018, and hosted by Fondazione IRCCS-Istituto Nazionale dei Tumori (Fondazione IRCCS INT). The conference was divided into two main scientific sessions, of i) pre-clinical assays and novel biotargets, and ii) clinical translation, as well as a third session of presentations from young investigators, which focused on recent achievements within Fondazione IRCCS INT on immunotherapy and targeted therapies. Presentations in the first session addressed the issue of cancer immunotherapy activity with respect to tumor heterogeneity, with key topics addressing: 1) tumor heterogeneity and targeted therapy, with the definition of the evolutionary Index as an indicator of tumor heterogeneity in both space and time; 2) the analysis of cancer evolution, with the introduction of the TRACERx Consortium-a multi-million pound UK research project focused on non-small cell lung cancer (NSCLC); 3) the use of anti-estrogen agents to boost immune recognition of breast cancer cells; and 4) the high degree of functional plasticity within the NK cell repertoire, including the expansion of adaptive NK cells following viral challenges. The second session addressed: 1) the effectiveness of radiotherapy to enhance the proportion of patients responsive to immune-checkpoint blockers (ICBs); 2) the use of MDSC scores in selecting melanoma patients with high probability to be responsive to ICBs; and 3) the relevance of the gut microbiome as a predictive factor, and the potential of its perturbation in increasing the immune response rate to ICBs. Overall, a picture emerged of tumor heterogeneity as the main limitation that impairs the effectiveness of anti-cancer therapies. Thus, the choice of a specific therapy based on reproducible and selective predictive biomarkers is an urgent unmet clinical need that should be addressed in order to increase the proportion of long-term responding patients and to improve the sustainability of novel drugs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy , Neoplasms/therapy , Animals , Gastrointestinal Microbiome , Humans , Neoplasms/immunology , Neoplasms/microbiology
8.
Ann Oncol ; 29(12): 2363-2370, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30307529

ABSTRACT

Background: Gene expression profiling (GEP) studies recognized a prognostic role for tumor microenvironment (TME) in diffuse large B-cell lymphoma (DLBCL), but the routinely adoption of prognostic stromal signatures remains limited. Patients and methods: Here, we applied the computational method CIBERSORT to generate a 1028-gene matrix incorporating signatures of 17 immune and stromal cytotypes. Then, we carried out a deconvolution on publicly available GEP data of 482 untreated DLBCLs to reveal associations between clinical outcomes and proportions of putative tumor-infiltrating cell types. Forty-five genes related to peculiar prognostic cytotypes were selected and their expression digitally quantified by NanoString technology on a validation set of 175 formalin-fixed, paraffin-embedded DLBCLs from two randomized trials. Data from an unsupervised clustering analysis were used to build a model of clustering assignment, whose prognostic value was also assessed on an independent cohort of 40 cases. All tissue samples consisted of pretreatment biopsies of advanced-stage DLBCLs treated by comparable R-CHOP/R-CHOP-like regimens. Results: In silico analysis demonstrated that higher proportion of myofibroblasts (MFs), dendritic cells, and CD4+ T cells correlated with better outcomes and the expression of genes in our panel is associated with a risk of overall and progression-free survival. In a multivariate Cox model, the microenvironment genes retained high prognostic performance independently of the cell-of-origin (COO), and integration of the two prognosticators (COO + TME) improved survival prediction in both validation set and independent cohort. Moreover, the major contribution of MF-related genes to the panel and Gene Set Enrichment Analysis suggested a strong influence of extracellular matrix determinants in DLBCL biology. Conclusions: Our study identified new prognostic categories of DLBCL, providing an easy-to-apply gene panel that powerfully predicts patients' survival. Moreover, owing to its relationship with specific stromal and immune components, the panel may acquire a predictive relevance in clinical trials exploring new drugs with known impact on TME.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/mortality , Transcriptome/genetics , Tumor Microenvironment/genetics , Adult , Aged , Algorithms , Biopsy , Cluster Analysis , Cohort Studies , Computational Biology , Datasets as Topic , Female , Gene Expression Profiling/methods , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Paraffin Embedding , Predictive Value of Tests , Prognosis , Progression-Free Survival , Randomized Controlled Trials as Topic , Reproducibility of Results , Survival Analysis , Young Adult
9.
Oncogene ; 35(23): 3016-26, 2016 06 09.
Article in English | MEDLINE | ID: mdl-26434590

ABSTRACT

Accumulating evidences have shown the association between aberrantly expressed microRNAs (miRs) and cancer, where these small regulatory RNAs appear to dictate the cell fate by regulating all the main biological processes. We demonstrated the responsibility of the circuitry connecting the oncomiR-221&222 with the tumor suppressors miR-126&126* in melanoma development and progression. According to the inverse correlation between endogenous miR-221&222 and miR-126&126*, respectively increasing or decreasing with malignancy, their enforced expression or silencing was sufficient for a reciprocal regulation. In line with the opposite roles of these miRs, protein analyses confirmed the reverse expression pattern of miR-126&126*-targeted genes that were induced by miR-221&222. Looking for a central player in this complex network, we revealed the dual regulation of AP2α, on one side directly targeted by miR-221&222 and on the other a transcriptional activator of miR-126&126*. We showed the chance of restoring miR-126&126* expression in metastatic melanoma to reduce the amount of mature intracellular heparin-binding EGF like growth factor, thus preventing promyelocytic leukemia zinc finger delocalization and maintaining its repression on miR-221&222 promoter. Thus, the low-residual quantity of these two miRs assures the release of AP2α expression, which in turn binds to and induces miR-126&126* transcription. All together these results point to an unbalanced ratio functional to melanoma malignancy between these two couples of miRs. During progression this balance gradually moves from miR-126&126* toward miR-221&222. This circuitry, besides confirming the central role of AP2α in orchestrating melanoma development and/or progression, further displays the significance of these miRs in cancer and the option of utilizing them for novel therapeutics.


Subject(s)
Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Melanoma/genetics , Melanoma/metabolism , MicroRNAs/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Disease Progression , Humans , Melanoma/pathology , MicroRNAs/genetics
10.
Leukemia ; 29(4): 958-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25283844

ABSTRACT

Interleukin (IL)-31A binds to an heterodimer composed of IL-31 receptor A (IL-31RA) and Oncostatin M Receptor (OSMR). The IL-31/IL-31R complex is involved in the pathogenesis of various skin diseases, including cutaneous T-cell lymphoma. No information is available on the relations between the IL-31/IL-31R complex and B-cell lymphoma. Here we have addressed this issue in follicular lymphoma (FL), a prototypic germinal center(GC)-derived B-cell malignancy. IL-31 enhanced primary FL cell proliferation through IL-31R-driven signal transducer and activator of transcription factor 1/3 (STAT1/3), extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt phosphorylation. In contrast, GC B cells did not signal to IL-31 in spite of IL-31R expression. GC B cells expressed predominantly the inhibitory short IL-31RA isoform, whereas FL cells expressed predominantly the long signaling isoform. Moreover, GC B cells lacked expression of other IL-31RA isoforms potentially involved in the signaling pathway. IL-31 protein expression was significantly higher in surface membrane than in cytosol of both FL and GC B cells. IL-31 was detected in plasma membrane microvesicles from both cell types but not released in soluble form in culture supernatants. IL-31 and IL-31RA expression was higher in lymph nodes from FL patients with grade IIIa compared with grade I/II, suggesting a paracrine and/or autocrine role of IL-31/IL-31RA complex in tumor progression through microvesicle shedding.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Leukemic , Germinal Center/metabolism , Interleukins/genetics , Lymphoma, Follicular/genetics , Receptors, Interleukin/genetics , B-Lymphocytes/pathology , Cell Membrane/metabolism , Cell Proliferation , Cell-Derived Microparticles/metabolism , Cytosol/metabolism , Female , Germinal Center/pathology , Humans , Interleukins/metabolism , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Grading , Phosphorylation , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
11.
Leukemia ; 29(2): 406-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24903480

ABSTRACT

The efficacy of antibody-based immunotherapy is due to the activation of apoptosis, the engagement of antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity (CDC). We developed a novel strategy to enhance CDC using bispecific antibodies (bsAbs) that neutralize the C-regulators CD55 and CD59 to enhance C-mediated functions. Two bsAbs (MB20/55 and MB20/59) were designed to recognize CD20 on one side. The other side neutralizes CD55 or CD59. Analysis of CDC revealed that bsAbs could kill 4-25 times more cells than anti-CD20 recombinant antibody in cell lines or cells isolated from patients with chronic lymphocytic leukemia. The pharmacokinetics of the bsAbs was evaluated in a human-SCID model of Burkitt lymphoma. The distribution profile of bsAbs mimics the data obtained by studying the pharmacokinetics of anti-CD20 antibodies, showing a peak in the tumor mass 3-4 days after injection. The treatment with bsAbs completely prevented the development of human/SCID lymphoma. The tumor growth was blocked by the activation of the C cascade and by the recruitment of macrophages, polymorphonuclear and natural killer cells. This strategy can easily be applied to the other anti-tumor C-fixing antibodies currently used in the clinic or tested in preclinical studies using the same vector with the appropriate modifications.


Subject(s)
Antibodies, Bispecific/chemistry , Antibodies/chemistry , Burkitt Lymphoma/immunology , Immunotherapy/methods , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/chemistry , CD55 Antigens/chemistry , CD59 Antigens/chemistry , Cell Separation , Cloning, Molecular , Complement System Proteins , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Killer Cells, Natural/cytology , Macrophages/cytology , Mice , Mice, SCID , Microscopy, Fluorescence
12.
Blood Cancer J ; 4: 259, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25382608

ABSTRACT

Peripheral T-cell lymphomas not otherwise specified (PTCLs/NOS) are rare and aggressive tumours whose molecular pathogenesis and diagnosis are still challenging. The microRNA (miRNA) profile of 23 PTCLs/NOS was generated and compared with that of normal T-lymphocytes (CD4+, CD8+, naive, activated). The differentially expressed miRNA signature was compared with the gene expression profile (GEP) of the same neoplasms. The obtained gene patterns were tested in an independent cohort of PTCLs/NOS. The miRNA profile of PTCLs/NOS then was compared with that of 10 angioimmunoblastic T-cell lymphomas (AITLs), 6 anaplastic large-cell lymphomas (ALCLs)/ALK+ and 6 ALCLs/ALK-. Differentially expressed miRNAs were validated in an independent set of 20 PTCLs/NOS, 20 AITLs, 19 ALCLs/ALK- and 15 ALCLs/ALK+. Two hundred and thirty-six miRNAs were found to differentiate PTCLs/NOS from activated T-lymphocytes. To assess which miRNAs impacted on GEP, a multistep analysis was performed, which identified all miRNAs inversely correlated to different potential target genes. One of the most discriminant miRNAs was selected and its expression was found to affect the global GEP of the tumours. Moreover, two sets of miRNAs were identified distinguishing PTCL/NOS from AITL and ALCL/ALK-, respectively. The diagnostic accuracy of this tool was very high (83.54%) and its prognostic value validated.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/metabolism , MicroRNAs/biosynthesis , RNA, Neoplasm/biosynthesis , Female , Gene Expression Profiling/methods , Humans , Lymphoma, T-Cell, Peripheral/genetics , Male , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis/methods , RNA, Neoplasm/genetics
13.
Ann Oncol ; 25(10): 2080-2086, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25015333

ABSTRACT

BACKGROUND: At diagnosis, identification of reliable biological indicators of prognosis to allow stratification of patients according to different risks is an important but still unresolved aspect in the treatment of Ewing sarcoma (EWS) patients. This study aimed to explore the role of miR-34A expression on prognosis of EWS patients. PATIENTS AND METHODS: Specimens from 109 patients with non-metastatic EWS treated at the Rizzoli Institute with neoadjuvant chemotherapy (protocols ISG/SSGIII, EW-1, EW-2, EW-REN2, EW-REN3, EW-PILOT) and 17 metastases were studied. Sixty-eight patients (62%) remained disease-free and 41 (38%) relapsed (median follow-up: 67 months, range 9-241 months). Expression of miR-34a and of some of its targets (cyclin D1, bcl-2, SIRT1 and YY1) was evaluated by qRT-PCR using TaqMan MicroRNA Assays and/or by immunohistochemistry on tissue microarrays from the same patients. RESULTS: High expression of miR-34a in localized tumors was significantly related to better event-free and overall survival (P = 0.004). Relevance of miR-34a was confirmed by using different calibrators (normal mesenchymal stem cells and different normal tissues). By multivariate Cox regression analysis, low miR-34a expression as well as nontotal necrosis and high levels of lactate dehydrogenase were all confirmed as independent risk factors associated with poor outcome. Expression of miR-34a was lower in metastases than in primary tumors. It inversely correlated with expression of cyclin D1 and Ki-67. CONCLUSIONS: By demonstrating its relationship with clinical outcome, we propose evaluation of miR-34a at diagnosis of EWS patients to allow early risk stratification. Validation of these results would nonetheless ultimately need a prospective assessment.


Subject(s)
Cyclin D1/biosynthesis , Ki-67 Antigen/biosynthesis , MicroRNAs/biosynthesis , Sarcoma, Ewing/genetics , Sarcoma, Ewing/therapy , Adult , Aged, 80 and over , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hydro-Lyases/biosynthesis , Male , MicroRNAs/genetics , Middle Aged , Neoadjuvant Therapy , Neoplasm Metastasis , Prognosis , Sarcoma, Ewing/pathology , Treatment Outcome
14.
Leukemia ; 28(8): 1687-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24480986

ABSTRACT

Peripheral T-cell lymphomas not otherwise specified (PTCL/NOS) are very aggressive tumors characterized by consistent aberrant expression of platelet-derived growth factor receptor alpha (PDGFRA). In this study, we aimed to identify the determinants of PDGFRA activity in PTCL/NOS and to elucidate the biological consequences of its activation. We observed overexpression of the PDGFRA gene by gene expression profiling in most of the tested PTCLs and confirmed the expression of PDGFRA and phospho-PDGFRA using immunohistochemistry. The integrity of the PDFGRA locus was demonstrated using several different approaches, including massive parallel sequencing and Sanger sequencing. PDGF-AA was found to be expressed and secreted by PTCL/NOS cells and to be necessary and sufficient for PDGFRA phosphorylation ex vivo by sustaining an autocrine stimulation. We documented consistently high PDGF-A expression in primary biopsies and patients' plasma and tracked PDGFRA signaling in primary tumors, achieving evidence of its activation. Indeed, we found that STAT1 and STAT5 are implicated in PDGFRA signaling transduction. Finally, we demonstrated that PDGFRA activation supported tumor cell proliferation and provided the first evidence of the anti-lymphoma activity of PDGRA inhibition in a PTCL/NOS patient. Altogether, our data demonstrated that PDGFRA activity fosters PTCL/NOS proliferation via an autocrine loop.


Subject(s)
Autocrine Communication , Lymphoma, T-Cell, Peripheral/pathology , Platelet-Derived Growth Factor/physiology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Proto-Oncogene Proteins c-akt/physiology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , STAT1 Transcription Factor , STAT5 Transcription Factor/physiology
15.
Leukemia ; 28(8): 1606-16, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24504027

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare disease of controversial origin recently recognized as a neoplasm deriving from plasmacytoid dendritic cells (pDCs). Nevertheless, it remains an orphan tumor with obscure biology and dismal prognosis. To better understand the pathobiology of BPDCN and discover new targets for effective therapies, the gene expression profile (GEP) of 25 BPDCN samples was analyzed and compared with that of pDCs, their postulated normal counterpart. Validation was performed by immunohistochemistry (IHC), whereas functional experiments were carried out ex vivo. For the first time at the molecular level, we definitely recognized the cellular derivation of BPDCN that proved to originate from the myeloid lineage and in particular, from resting pDCs. Furthermore, thanks to an integrated bioinformatic approach we discovered aberrant activation of the NF-kB pathway and suggested it as a novel therapeutic target. We tested the efficacy of anti-NF-kB-treatment on the BPDCN cell line CAL-1, and successfully demonstrated by GEP and IHC the molecular shutoff of the NF-kB pathway. In conclusion, we identified a molecular signature representative of the transcriptional abnormalities of BPDCN and developed a cellular model proposing a novel therapeutic approach in the setting of this otherwise incurable disease.


Subject(s)
Dendritic Cells/pathology , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Cell Cycle , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , NF-kappa B/physiology
16.
Am J Transplant ; 13(8): 2201-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23731345

ABSTRACT

Hemolytic uremic syndrome (HUS) is a disease of microangiopathic hemolytic anemia, thrombocytopenia and acute renal failure. About 90% of cases are secondary to infections by Escherichia coli strains producing Shiga-like toxins (STEC-HUS), while 10% are associated with mutations in genes encoding proteins of complement system (aHUS). We describe two patients with a clinical history of STEC-HUS, who developed end-stage renal disease (ESRD) soon after disease onset. They received a kidney transplant but lost the graft for HUS recurrence, a complication more commonly observed in aHUS. Before planning a second renal transplantation, the two patients underwent genetic screening for aHUS-associated mutations that revealed the presence of a heterozygous CFI mutation in patient #1 and a heterozygous MCP mutation in patient #2, and also in her mother who donated the kidney. This finding argues that the two cases originally diagnosed as STEC-HUS had indeed aHUS triggered by STEC infection on a genetic background of impaired complement regulation. Complement gene sequencing should be performed before kidney transplantation in patients who developed ESRD following STEC-HUS since they may be undiagnosed cases of aHUS, at risk of posttransplant recurrence. Furthermore, genetic analysis of donors is mandatory before living-related transplantation to exclude carriers of HUS-predisposing mutations.


Subject(s)
Complement Factor I/genetics , Escherichia coli Infections/complications , Hemolytic-Uremic Syndrome/complications , Kidney Failure, Chronic/etiology , Membrane Cofactor Protein/genetics , Mutation/genetics , Adult , Case-Control Studies , DNA Primers/chemistry , DNA Primers/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Female , Genetic Testing , Graft Rejection/diagnosis , Graft Rejection/etiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Heterozygote , Humans , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/therapy , Kidney Transplantation , Male , Middle Aged , Pedigree , Prognosis , Recurrence , Risk Factors , Shiga-Toxigenic Escherichia coli , Thrombocytopenia/complications , Thrombocytopenia/genetics , Thrombocytopenia/microbiology , Young Adult
17.
Leukemia ; 25(8): 1268-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21546901

ABSTRACT

Several chemokines/chemokine receptors such as CCR7, CXCR4 and CXCR5 attract chronic lymphocytic leukemia (CLL) cells to specific microenvironments. Here we have investigated whether the CX(3)CR1/CX(3)CL1 axis is involved in the interaction of CLL with their microenvironment. CLL cells from 52 patients expressed surface CX(3)CR1 and CX(3)CL1 and released constitutively soluble CX(3)CL1. One third of these were attracted in vitro by soluble CX(3)CL1. CX(3)CL1-induced phosphorylation of PI3K, Erk1/2, p38, Akt and Src was involved in induction of CLL chemotaxis. Leukemic B cells upregulated CXCR4 upon incubation with CX(3)CL1 and this was paralleled by increased chemotaxis to CXCL12. Akt phosphorylation was involved in CX(3)CL1-induced upregulation of CXCR4 on CLL. In proliferation centers from CLL lymph node and bone marrow, CX(3)CL1 was expressed by CLL cells whereas CX(3)CR1 was detected in CLL and stromal cells. Nurselike cells (NLCs) generated from CLL patient blood co-expressed surface CX(3)CR1 and CX(3)CL1, but did not secrete soluble CX(3)CL1. Only half of NLC cell fractions were attracted in vitro by CX(3)CL1. In conclusion, the CX(3)CR1/CX(3)CL1 system may contribute to interactions between CLL cells and tumor microenvironment by increasing CXCL12-mediated attraction of leukemic cells to NLC and promoting directly adhesion of CLL cells to NLC.


Subject(s)
Cell Communication , Chemokine CX3CL1/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptors, Chemokine/physiology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , CX3C Chemokine Receptor 1 , Chemotaxis , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymph Nodes/metabolism , Male , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
18.
Oncogene ; 30(7): 757-69, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21057534

ABSTRACT

c-Kit tyrosine kinase receptor and its ligand stem cell factor have multiple functions during development, whereas in adulthood they are mostly needed for stem cell (SC) maintenance and mast cell (MC) biology. c-Kit plays an essential tumor-cell-intrinsic role in many types of cancer, either providing the tumorigenic force when aberrantly activated or conferring stem-like features characterizing the most aggressive variants. A tumor-cell-extrinsic role occurs through c-Kit-dependent accessory cells (such as MCs) that infiltrate tumors and deeply influence their progression. c-Kit-targeted therapy with tyrosine kinase inhibitors (TKIs) may ideally work against both tumor and stromal cells. Here, we summarize the tumor-intrinsic and -extrinsic roles of c-Kit in cancer and discuss TKIs with their on- and off-targets, with a special emphasis on MCs as paradigmatic c-Kit-dependent accomplices for tumor progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Mast Cells/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Animals , Humans , Mast Cells/drug effects , Mast Cells/immunology , Mastocytosis/blood supply , Mastocytosis/drug therapy , Mastocytosis/immunology , Mice , Neoplasm Invasiveness , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Rats
19.
Curr Cancer Drug Targets ; 9(5): 617-25, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19508173

ABSTRACT

Monoclonal antibodies (mAbs), successfully adopted in the treatment of several haematological malignancies, have proved almost ineffective in multiple myeloma (MM), because of the lack of an appropriate antigen for targeting and killing MM cells. Here, we demonstrate that PSGL1, the major ligand of P-Selectin, a marker of plasmacytic differentiation expressed at high levels on normal and neoplastic plasma cells, may represent a novel target for mAb-mediated MM immunotherapy. The primary effectors of mAb-induced cell-death, complement-mediated lysis (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC), were investigated using U266B1 and LP1 cell-lines as models. Along with immunological mechanisms, the induction of apoptosis by PSGL1 cross-linking was assessed. The anti-PSGL1 murine mAb KPL1 induced death of MM cells in a dose- and time-dependent fashion and mediated a significant amount of ADCC. KPL1 alone mediated C1q deposition on target cells but proved unable to induce CDC due to inhibition of the lytic activity of complement by membrane complement regulators (mCRP) expressed on the cell surface. Consistently, CDC was induced by KPL1 upon mCRP blockage. Our results suggest a role for PSGL1 in MM humoral immunotherapy and support further in vivo studies assessing the effects of anti-PSGL1 mAbs on MM growth and interaction with the bone marrow microenvironment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Membrane Glycoproteins/immunology , Multiple Myeloma/therapy , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , Complement Activation/drug effects , Complement Activation/immunology , Cytotoxicity, Immunologic/drug effects , Drug Delivery Systems , Drug Evaluation, Preclinical , Humans , Membrane Glycoproteins/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology
20.
Int J Immunopathol Pharmacol ; 22(1): 73-83, 2009.
Article in English | MEDLINE | ID: mdl-19309554

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising natural anticancer therapeutic agent because through its death receptors, TRAIL-R1 and TRAIL-R2, it induces apoptosis in many transformed tumor cells, but not in the majority of normal cells. Hence, agonistic compounds directed against TRAIL death receptors have the potential of being excellent cancer therapeutic agents, with minimal cytotoxicity in normal tissues. Here, we report the selection and characterization of a new single-chain fragment variable (scFv) to TRAIL-R2 receptor isolated from a human phage-display library, produced as minibody (MB), and characterized for the in vitro anti-leukemic tumoricidal activity. The anti-TRAIL-R2 MB2.23 efficiently and specifically bound to membrane-associated TRAIL-R2 on different leukemic cell lines and could act as a direct agonist in vitro, initiating apoptotic signaling as well as complement-dependent cytotoxicity and antibody-dependent cell cytotoxicity, providing a rationale for further investigations of MB2.23 in anticancer therapy.


Subject(s)
Immunoglobulin Fragments/therapeutic use , Leukemia/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Animals , Antibody-Dependent Cell Cytotoxicity , Apoptosis/drug effects , CHO Cells , Cricetinae , Cricetulus , Humans , Leukemia/pathology , Peptide Library , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...