Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Regen Ther ; 27: 234-243, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38586872

ABSTRACT

Introduction: Quality and safety of a cell product, essential to guarantee the health of patients, depends on many factors including an appropriate environmental monitoring of the manufacturing rooms. Nonetheless, the maintenance of a controlled environment is requested to minimize the risk of contamination. Thus, a timely detection of changes in microbiological trends is important to adopt promptly effective measures against resistant strains that, in turn, may invalidate not only the sanitization procedures but also the safety of the cell product. Methods: We analyzed microbes found in our cell processing clean room over the last 5 years. We used 10.147 plates for air sampler, passive air monitoring and for checking instruments and operators of the production unit. Results: From these plates, 747 colonies were subjected to identification by the MALDI-TOF Vitek® MS system and the large majority of them was gram positive (97.8%) as witnessed by the finding that the most represented genera harvested from the classified areas were Staphylococcus (65%), Micrococcus (13%), Kocuria (8%) and Bacillus (5%). We never detected fungi. Most microbes found in the operators (both from class A and B) were collected from forearms and resulted of the Staphylococcus genus. Conclusions: The observed microbial contamination is to be attributed to the personnel and no substantial microbial pitfalls in our Cell Factory has been detected.

2.
Curr Res Transl Med ; 72(4): 103445, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38493557

ABSTRACT

The use of γδ T lymphocytes as advanced therapeutic medicinal product has attracted much interest in the last years. Indeed, such cells are an ideal tool for the reconstitution of the immune system in patients receiving hematopoietic stem cell transplantation, due to their MHC-independent anti-tumor and anti-viral activities. We have here setup a protocol for the production of pure and functional γδ T lymphocytes, expanded from healthy donors' mononuclear cells, and validated the analytical methods to identify them and to analyze their potency. Next, we performed stability studies to ensure that the cell product (γδ T cells) can be used after freezing and thawing. Notably, such protocol can be promptly translated to GMP-facility, since it has been designed using only clinical grade reagents.

3.
Int J Mol Sci ; 23(6)2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35328349

ABSTRACT

HLA-G is an HLA-class Ib molecule that is involved in the establishment of tolerance at the maternal/fetal interface during pregnancy. The expression of HLA-G is highly restricted in adults, but the de novo expression of this molecule may be observed in different hematological and solid tumors and is related to cancer progression. Indeed, tumor cells expressing high levels of HLA-G are able to suppress anti-tumor responses, thus escaping from the control of the immune system. HLA-G has been proposed as an immune checkpoint (IC) molecule due to its crucial role in tumor progression, immune escape, and metastatic spread. We here review data available in the literature in which the interaction between HLA-G and other IC molecules is reported, in particular PD-1, CTLA-4, and TIM-3, but also IDO and TIGIT. Clinical trials using monoclonal antibodies against HLA-G and other IC are currently ongoing with cancer patients where antibodies and inhibitors of PD-1 and CTLA-4 showed encouraging results. With this background, we may envisage that combined therapies using antibodies targeting HLA-G and another IC may be successful for clinical purposes. Indeed, such immunotherapeutic protocols may achieve a better rescue of effective anti-tumor immune response, thus improving the clinical outcome of patients.


Subject(s)
HLA-G Antigens , Neoplasms , Adult , CTLA-4 Antigen , HLA-G Antigens/metabolism , Humans , Immune Checkpoint Proteins , Immunotherapy/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor
4.
Int J Mol Sci ; 22(8)2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33921337

ABSTRACT

The long-underestimated role of extracellular vesicles in cancer is now reconsidered worldwide by basic and clinical scientists, who recently highlighted novel and crucial activities of these moieties. Extracellular vesicles are now considered as king transporters of specific cargoes, including molecular components of parent cells, thus mediating a wide variety of cellular activities both in normal and neoplastic tissues. Here, we discuss the multifunctional activities and underlying mechanisms of extracellular vesicles in neuroblastoma, the most frequent common extra-cranial tumor in childhood. The ability of extracellular vesicles to cross-talk with different cells in the tumor microenvironment and to modulate an anti-tumor immune response, tumorigenesis, tumor growth, metastasis and drug resistance will be pinpointed in detail. The results obtained on the role of extracellular vesicles may represent a panel of suggestions potentially useful in practice, due to their involvement in the response to chemotherapy, and, moreover, their ability to predict resistance to standard therapies-all issues of clinical relevance.


Subject(s)
Extracellular Vesicles/genetics , Neuroblastoma/drug therapy , Tumor Microenvironment/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Child , Drug Resistance, Neoplasm/genetics , Extracellular Vesicles/drug effects , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Tumor Microenvironment/genetics
5.
Cells ; 10(4)2021 04 17.
Article in English | MEDLINE | ID: mdl-33920505

ABSTRACT

The tumor microenvironment (TME) represents a complex network between tumor cells and a variety of components including immune, stromal and vascular endothelial cells as well as the extracellular matrix. A wide panel of signals and interactions here take place, resulting in a bi-directional modulation of cellular functions. Many stimuli, on one hand, induce tumor growth and the spread of metastatic cells and, on the other hand, contribute to the establishment of an immunosuppressive environment. The latter feature is achieved by soothing immune effector cells, mainly cytotoxic T lymphocytes and B and NK cells, and/or through expansion of regulatory cell populations, including regulatory T and B cells, tumor-associated macrophages and myeloid-derived suppressor cells. In this context, immune checkpoints (IC) are key players in the control of T cell activation and anti-cancer activities, leading to the inhibition of tumor cell lysis and of pro-inflammatory cytokine production. Thus, these pathways represent promising targets for the development of effective and innovative therapies both in adults and children. Here, we address the role of different cell populations homing the TME and of well-known and recently characterized IC in the context of pediatric solid tumors. We also discuss preclinical and clinical data available using IC inhibitors alone, in combination with each other or administered with standard therapies.


Subject(s)
Immune Checkpoint Proteins/metabolism , Molecular Targeted Therapy , Neoplasms/metabolism , Child , Humans , Immunotherapy , Models, Biological , Tumor Microenvironment/immunology
6.
Vaccines (Basel) ; 9(1)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33450862

ABSTRACT

Neuroblastoma is the most common extracranial pediatric solid tumor with a heterogeneous clinical course, ranging from spontaneous regression to metastatic disease and death, irrespective of intensive chemotherapeutic regimen. On the basis of several parameters, children affected by neuroblastoma are stratified into low, intermediate and high risk. At present, more than 50% of high-risk patients with metastatic spread display an overall poor long-term outcome also complicated by devastating long-term morbidities. Thus, novel and more effective therapies are desperately needed to improve lifespan of high-risk patients. In this regard, adoptive cell therapy holds great promise and several clinical trials are ongoing, demonstrating safety and tolerability, with no toxicities. Starting from the immunological and clinical features of neuroblastoma, we here discuss the immunotherapeutic approaches currently adopted for high-risk patients and different innovative therapeutic strategies currently under investigation. The latter are based on the infusion of natural killer (NK) cells, as support of consolidation therapy in addition to standard treatments, or chimeric antigen receptor (CAR) T cells directed against neuroblastoma associated antigens (e.g., disialoganglioside GD2). Finally, future perspectives of adoptive cell therapies represented by γδ T lymphocyes and CAR NK cells are envisaged.

7.
J Hematol Oncol ; 13(1): 89, 2020 07 11.
Article in English | MEDLINE | ID: mdl-32653014

ABSTRACT

BACKGROUND: The oncolytic viruses have shown promising results for the treatment of multiple myeloma. However, the use of human viruses is limited by the patients' antiviral immune response. In this study, we investigated an alternative oncolytic strategy using non-human pathogen viruses as the bovine viral diarrhea virus (BVDV) that were able to interact with CD46. METHODS: We treated several human myeloma cell lines and non-myeloma cell lines with BVDV to evaluate the expression of CD46 and to study the effect on cell viability by flow cytometry. The possible synergistic effect of bortezomib in combination with BVDV was also tested. Moreover, we infected the bone marrow mononuclear cells obtained from myeloma patients and we checked the BVDV effect on different cell populations, defined by CD138, CD14, CD3, CD19, and CD56 expression evaluated by flow cytometry. Finally, the in vivo BVDV effect was tested in NOD-SCID mice injected subcutaneously with myeloma cell lines. RESULTS: Human myeloma cells were selectively sensitive to BVDV treatment with an increase of cell death and, consequently, of apoptotic markers. Consistently, bone marrow mononuclear cells isolated from myeloma patients treated with BVDV, showed a significant selective decrease of the percentage of viable CD138+ cells. Interestingly, bortezomib pre-treatment significantly increased the cytotoxic effect of BVDV in myeloma cell lines with a synergistic effect. Finally, the in vitro data were confirmed in an in vivo myeloma mouse model showing that BVDV treatment significantly reduced the tumoral burden compared to the vehicle. CONCLUSIONS: Overall, our data indicate, for the first time, a direct oncolytic effect of the BVDV in human myeloma cells suggesting its possible use as novel alternative anti-myeloma virotherapy strategy.


Subject(s)
Diarrhea Viruses, Bovine Viral , Multiple Myeloma/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Aged , Aged, 80 and over , Animals , Antigens, CD/analysis , Apoptosis , Bone Marrow Cells/chemistry , Bone Marrow Cells/drug effects , Bone Marrow Cells/virology , Bortezomib/pharmacology , Cell Line, Tumor , Cytopathogenic Effect, Viral , Diarrhea Viruses, Bovine Viral/physiology , Female , Herpesvirus 4, Bovine , Humans , Male , Membrane Cofactor Protein/biosynthesis , Membrane Cofactor Protein/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Multiple Myeloma/pathology , Oncolytic Viruses/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Specific Pathogen-Free Organisms
8.
Cells ; 9(8)2020 07 22.
Article in English | MEDLINE | ID: mdl-32707982

ABSTRACT

Most studies on genetic engineering technologies for cancer immunotherapy based on allogeneic donors have focused on adaptive immunity. However, the main limitation of such approaches is that they can lead to severe graft-versus-host disease (GvHD). An alternative approach would bolster innate immunity by relying on the natural tropism of some subsets of the innate immune system, such as γδ T and natural killer (NK) cells, for the tumor microenvironment and their ability to kill in a major histocompatibility complex (MHC)-independent manner. γδ T and NK cells have the unique ability to bridge innate and adaptive immunity while responding to a broad range of tumors. Considering these properties, γδ T and NK cells represent ideal sources for developing allogeneic cell therapies. Recently, significant efforts have been made to exploit the intrinsic anti-tumor capacity of these cells for treating hematologic and solid malignancies using genetic engineering approaches such as chimeric antigen receptor (CAR) and T cell receptor (TCR). Here, we review over 30 studies on these two approaches that use γδ T and NK cells in adoptive cell therapy (ACT) for treating cancer. Based on those studies, we propose several promising strategies to optimize the clinical translation of these approaches.


Subject(s)
Adaptive Immunity , Immunity, Innate , Immunotherapy, Adoptive/methods , Intraepithelial Lymphocytes/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Engineering/methods , Genetic Engineering/methods , Graft vs Host Disease/immunology , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology
9.
Front Immunol ; 11: 699, 2020.
Article in English | MEDLINE | ID: mdl-32477328

ABSTRACT

TcRαß/CD19-cell depleted HLA-haploidentical hematopoietic stem cell transplantation (haplo-HSCT) represents a promising new platform for children affected by acute leukemia in need of an allograft and lacking a matched donor, disease recurrence being the main cause of treatment failure. The use of zoledronic acid to enhance TcRγδ+ lymphocyte function after TcRαß/CD19-cell depleted haplo-HSCT was tested in an open-label, feasibility, proof-of-principle study. Forty-six children affected by high-risk acute leukemia underwent haplo-HSCT after removal of TcRαß+ and CD19+ B lymphocytes. No post-transplant pharmacological graft-versus-host disease (GvHD) prophylaxis was given. Zoledronic acid was administered monthly at a dose of 0.05 mg/kg/dose (maximum dose 4 mg), starting from day +20 after transplantation. A total of 139 infusions were administered, with a mean of 3 infusions per patient. No severe adverse event was observed. Common side effects were represented by asymptomatic hypocalcemia and acute phase reactions (including fever, chills, malaise, and/or arthralgia) within 24-48 h from zoledronic acid infusion. The cumulative incidence of acute and chronic GvHD was 17.3% (all grade I-II) and 4.8% (all limited), respectively. Patients given 3 or more infusions of zoledronic acid had a lower incidence of both acute GvHD (8.8 vs. 41.6%, p = 0.015) and chronic GvHD (0 vs. 22.2%, p = 0.006). Transplant-related mortality (TRM) and relapse incidence at 3 years were 4.3 and 30.4%, respectively. Patients receiving repeated infusions of zoledronic acid had a lower TRM as compared to those receiving 1 or 2 administration of the drug (0 vs. 16.7%, p = 0.01). Five-year overall survival (OS) and disease-free survival (DFS) for the whole cohort were 67.2 and 65.2%, respectively, with a trend toward a better OS for patients receiving 3 or more infusions (73.1 vs. 50.0%, p = 0.05). The probability of GvHD/relapse-free survival was significantly worse in patients receiving 1-2 infusions of zoledonic acid than in those given ≥3 infusions (33.3 vs. 70.6%, respectively, p = 0.006). Multivariable analysis showed an independent positive effect on outcome given by repeated infusions of zoledronic acid (HR 0.27, p = 0.03). These data indicate that the use of zoledronic acid after TcRαß/CD19-cell depleted haploHSCT is safe and may result in a lower incidence of acute GvHD, chronic GvHD, and TRM.


Subject(s)
Antigens, CD19/immunology , Hematopoietic Stem Cell Transplantation/methods , Immunologic Factors/administration & dosage , Leukemia, Myeloid, Acute/therapy , Lymphocyte Depletion/methods , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Transplantation Conditioning/methods , Transplantation, Haploidentical/methods , Zoledronic Acid/administration & dosage , Adolescent , B-Lymphocytes/immunology , Child , Child, Preschool , Cohort Studies , Disease-Free Survival , Feasibility Studies , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , Immunologic Factors/adverse effects , Infant , Male , T-Lymphocytes/immunology , Transplantation, Homologous/methods , Young Adult , Zoledronic Acid/adverse effects
10.
Cells ; 9(5)2020 05 24.
Article in English | MEDLINE | ID: mdl-32456316

ABSTRACT

γδ T cells have recently gained considerable attention as an attractive tool for cancer adoptive immunotherapy due to their potent anti-tumor activity and unique role in immunosurveillance. The remarkable success of engineered T cells for the treatment of hematological malignancies has revolutionized the field of adoptive cell immunotherapy. Accordingly, major efforts are underway to translate this exciting technology to the treatment of solid tumors and the development of allogeneic therapies. The unique features of γδ T cells, including their major histocompatibility complex (MHC)-independent anti-cancer activity, tissue tropism, and multivalent response against a broad spectrum of the tumors, render them ideal for designing universal 'third-party' cell products, with the potential to overcome the challenges of allogeneic cell therapy. In this review, we describe the crucial role of γδ T cells in anti-tumor immunosurveillance and we summarize the different approaches used for the ex vivo and in vivo expansion of γδ T cells suitable for the development of novel strategies for cancer therapy. We further discuss the different transduction strategies aiming at redirecting or improving the function of γδ T cells, as well as, the considerations for the clinical applications.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Cell Proliferation , Clinical Trials as Topic , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/cytology
11.
Cells ; 8(12)2019 11 27.
Article in English | MEDLINE | ID: mdl-31783629

ABSTRACT

CD38 is a multifunctional cell surface protein endowed with receptor/enzymatic functions. The protein is generally expressed at low/intermediate levels on hematological tissues and some solid tumors, scoring the highest levels on plasma cells (PC) and PC-derived neoplasia. CD38 was originally described as a receptor expressed by activated cells, mainly T lymphocytes, wherein it also regulates cell adhesion and cooperates in signal transduction mediated by major receptor complexes. Furthermore, CD38 metabolizes extracellular NAD+, generating ADPR and cyclic ADPR. This ecto-enzyme controls extra-cellular nucleotide homeostasis and intra-cellular calcium fluxes, stressing its relevance in multiple physiopathological conditions (infection, tumorigenesis and aging). In clinics, CD38 was adopted as a cell activation marker and in the diagnostic/staging of leukemias. Quantitative surface CD38 expression by multiple myeloma (MM) cells was the basic criterion used for therapeutic application of anti-CD38 monoclonal antibodies (mAbs). Anti-CD38 mAbs-mediated PC depletion in autoimmunity and organ transplants is currently under investigation. This review analyzes different aspects of CD38's role in regulatory cell populations and how these effects are obtained. Characterizing CD38 functional properties may widen the extension of therapeutic applications for anti-CD38 mAbs. The availability of therapeutic mAbs with different effects on CD38 enzymatic functions may be rapidly translated to immunotherapeutic strategies of cell immune defense.


Subject(s)
ADP-ribosyl Cyclase 1 , B-Lymphocytes, Regulatory/immunology , Extracellular Vesicles/immunology , Membrane Glycoproteins , T-Lymphocytes, Regulatory/immunology , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/physiology , Aging/immunology , Animals , Antibodies, Monoclonal/therapeutic use , B-Lymphocytes, Regulatory/cytology , B-Lymphocytes, Regulatory/pathology , Cell Line , Humans , Infections/drug therapy , Infections/immunology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/physiology , Mice , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/pathology
12.
Sci Transl Med ; 10(428)2018 02 14.
Article in English | MEDLINE | ID: mdl-29444977

ABSTRACT

Although the progression of chronic lymphocytic leukemia (CLL) requires the cooperation of the microenvironment, the exact cellular and molecular mechanisms involved are still unclear. We investigated the interleukin (IL)-23 receptor (IL-23R)/IL-23 axis and found that circulating cells from early-stage CLL patients with shorter time-to-treatment, but not of those with a more benign course, expressed a defective form of the IL-23R complex lacking the IL-12Rß1 chain. However, cells from both patient groups expressed the complete IL-23R complex in tissue infiltrates and could be induced to express the IL-12Rß1 chain when cocultured with activated T cells or CD40L+ cells. CLL cells activated in vitro in this context produced IL-23, a finding that, together with the presence of IL-23 in CLL lymphoid tissues, suggests the existence of an autocrine/paracrine loop inducing CLL cell proliferation. Interference with the IL-23R/IL-23 axis using an anti-IL-23p19 antibody proved effective in controlling disease onset and expansion in xenografted mice, suggesting potential therapeutic strategies.


Subject(s)
Interleukin-23/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptors, Interleukin/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Antibodies, Neutralizing/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lymph Nodes/metabolism , Mice , Neoplasm Staging , Risk Factors , Stromal Cells/metabolism , Up-Regulation
13.
Cell Oncol (Dordr) ; 40(5): 483-496, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28721629

ABSTRACT

PURPOSE: Multiple myeloma (MM) is a hematologic malignancy characterized by a clonal expansion of plasma cells (PCs) in the bone marrow (BM). Since MM has so far remained incurable, further insights into its pathogenesis and the concomitant identification of new therapeutic targets are urgently needed. The tyrosine kinase receptor EphA3 is known to be involved in various cellular processes including cell viability, cell movement and cell-cell interactions. Recently, EphA3 has emerged as a potential therapeutic target in several hematologic and solid tumors. Here, we aimed to uncover the role of EphA3 in MM. METHODS: EphA3 mRNA and protein expression in primary MM bone marrow plasma cells (BMPCs), in MM-derived cell lines and in healthy controls (HCs) was assessed using qRT-PCR, Western blotting and flow cytometry. The effects of siRNA-mediated EphA3 silencing and anti EphA3 antibody (EphA3mAb) treatment on MM PC trafficking and viability were evaluated using in vitro assays. The effects of EphA3mAb treatment were also assessed in two MM-derived mouse xenograft models. RESULTS: We found that EphA3 was overexpressed in primary MM BMPCs and MM-derived cell lines compared to HCs. We also found that siRNA-mediated EphA3 silencing and EphA3mAb treatment significantly inhibited the ability of MM PCs to adhere to fibronectin and stromal cells and to invade in vitro, without affecting cell proliferation and viability. Gene expression profiling showed that EphA3 silencing resulted in expression modulation of several molecules that regulate adhesion, migration and invasion processes. Importantly, we found that EphA3mAb treatment significantly inhibited in vivo tumor growth and angiogenesis in two MM-derived mouse xenograft models. CONCLUSIONS: Our findings suggest that EphA3 plays an important role in the pathogenesis of MM and provide support for the notion that its targeting may represent a novel therapeutic opportunity for MM.


Subject(s)
Cell Movement/genetics , Multiple Myeloma/genetics , Neovascularization, Pathologic/genetics , Receptor, EphA3/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Adhesion/genetics , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neovascularization, Pathologic/metabolism , RNA Interference , Receptor, EphA3/immunology , Receptor, EphA3/metabolism , Xenograft Model Antitumor Assays
14.
Cancer Res ; 76(21): 6218-6229, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27550449

ABSTRACT

The inflammatory tissue microenvironment that promotes the development of breast cancer is not fully understood. Here we report a role for elevated IL30 in supporting the breast cancer cell viability and invasive migration. IL30 was absent in normal mammary ducts, ductules, and acini of histologically normal breast and scanty in the few stromal infiltrating leukocytes. In contrast, IL30 was expressed frequently in breast cancer specimens where it was associated with triple-negative and HER2+ molecular subtypes. In stromal leukocytes found in primary tumors or tumor-draining lymph nodes, which included mainly CD14+ monocytes, CD68+ macrophages, and CD33+/CD11b+ myeloid cells, IL30 levels increased with disease stage and correlated with recurrence. A negative correlation was determined between IL30 expression by nodal stromal leukocytes and overall survival. In vitro studies showed that human recombinant IL30 upregulated expression of a pro-oncogenic program, including especially IL6 in both triple-negative and HER2+ breast cancer cells. In triple-negative breast cancer cells, IL30 boosted a broader program of proliferation, invasive migration, and an inflammatory milieu associated with KISS1-dependent metastasis. Silencing of STAT1/STAT3 signaling hindered the regulation of the primary growth and progression factors in breast cancer cells. IL30 administration in vivo fostered the growth of triple-negative breast cancer by promoting proliferation and vascular dissemination of cancer cells and the accumulation of intratumoral CD11b+/Gr1+ myeloid cell infiltrates. Overall, our results show how IL30 regulates breast cancer cell viability, migration, and gene expression to promote breast cancer growth and progression and its impact on patient outcome. Cancer Res; 76(21); 6218-29. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Interleukins/physiology , Animals , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Humans , Interleukins/analysis , Mice , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Neoplasm Staging , Receptor, ErbB-2/analysis , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Triple Negative Breast Neoplasms/pathology
15.
Blood ; 128(5): 667-79, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27268090

ABSTRACT

The importance of glutamine (Gln) metabolism in multiple myeloma (MM) cells and its potential role as a therapeutic target are still unknown, although it has been reported that human myeloma cell lines (HMCLs) are highly sensitive to Gln depletion. In this study, we found that both HMCLs and primary bone marrow (BM) CD138(+) cells produced large amounts of ammonium in the presence of Gln. MM patients have lower BM plasma Gln with higher ammonium and glutamate than patients with indolent monoclonal gammopathies. Interestingly, HMCLs expressed glutaminase (GLS1) and were sensitive to its inhibition, whereas they exhibited negligible expression of glutamine synthetase (GS). High GLS1 and low GS expression were also observed in primary CD138(+) cells. Gln-free incubation or treatment with the glutaminolytic enzyme l-asparaginase depleted the cell contents of Gln, glutamate, and the anaplerotic substrate 2-oxoglutarate, inhibiting MM cell growth. Consistent with the dependence of MM cells on extracellular Gln, a gene expression profile analysis, on both proprietary and published datasets, showed an increased expression of the Gln transporters SNAT1, ASCT2, and LAT1 by CD138(+) cells across the progression of monoclonal gammopathies. Among these transporters, only ASCT2 inhibition in HMCLs caused a marked decrease in Gln uptake and a significant fall in cell growth. Consistently, stable ASCT2 downregulation by a lentiviral approach inhibited HMCL growth in vitro and in a murine model. In conclusion, MM cells strictly depend on extracellular Gln and show features of Gln addiction. Therefore, the inhibition of Gln uptake is a new attractive therapeutic strategy for MM.


Subject(s)
Glutamine/metabolism , Molecular Targeted Therapy , Multiple Myeloma/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Transport System ASC/metabolism , Ammonium Compounds/metabolism , Animals , Asparaginase/metabolism , Biological Transport , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred NOD , Mice, SCID , Middle Aged , Minor Histocompatibility Antigens/metabolism , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Syndecan-1/metabolism
16.
Immunobiology ; 221(2): 291-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26547104

ABSTRACT

In a previous study, lack of IL-12 signaling in il12rb2 knock-out mice was found to predispose to lung adenocarcinoma (LAC). We asked whether specific polymorphisms of the human IL12RB2 gene may confer susceptibility to LAC. We studied IL12RB2 single nucleotide polymorphisms (SNPs) spanning from the promoter to the first untranslated exon of the gene. Genotypes of 49 individuals with LAC were compared with those of 93 healthy subjects. Two allele variants were found to be associated with increased susceptibility to LAC. One haplotype (hap), hap18, was more frequent in patients (18%) versus controls (6%) and significantly associated with increased probability of disease occurrence. Furthermore, IL-12 driven STAT4 phosphorylation in T cell blasts from healthy individuals was found to correlate with both single allele variants and haplotypes. In conclusion, genetically determined low signaling activity of IL-12R predisposes to the development of LAC.


Subject(s)
Adenocarcinoma/genetics , Genetic Predisposition to Disease , Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Receptors, Interleukin-12/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Alleles , Animals , Case-Control Studies , Exons , Female , Gene Expression Regulation , Gene Frequency , Haplotypes , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , Phosphorylation , Promoter Regions, Genetic , Receptors, Interleukin-12/immunology , Risk , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Untranslated Regions
17.
Oncotarget ; 7(11): 12372-85, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26540632

ABSTRACT

Critical issues in prostate cancer (PC) are a. identification of molecular drivers of the highly aggressive neuroendocrine differentiation (NED) in adenocarcinoma, and b. early assessment of disease progression. The SRY (sex determining region Y)-box 2 gene, SOX2, is an essential embryonic stem cell gene involved in prostate tumorigenesis. Here we assessed its implications in NED and progression of PC and its diagnostic and prognostic value. Laser microdissection, qRT-PCR, quantitative Methylation-Specific PCR and immunohistochemistry were used to analyze SOX2 gene expression and regulation in 206 PC samples. Results were examined according to the patient's clinical pathological profile and follow-ups. Functional studies were performed using PC cells transfected to overexpress or silence SOX2. SOX2 was consistently downregulated in PC, except in cell clusters lying within lymph node (LN)-positive PC. Multivariate analysis revealed that SOX2 mRNA expression in the primary tumor was significantly associated with LN metastasis. When SOX2 mRNA levels were ≥1.00, relative to (XpressRef) Universal Total RNA, adjusted Odds Ratio was 24.4 (95% CI: 7.54-79.0), sensitivity 0.81 (95% CI: 0.61-0.93) and specificity 0.87 (95% CI: 0.81-0.91). Patients experiencing biochemical recurrence had high median levels of SOX2 mRNA. In both PC and LN metastasis, SOX2 and NED marker, Chromogranin-A, were primarily co-expressed. In PC cells, NED genes were upregulated by SOX2 overexpression and downregulated by its silencing, which also abolished SNAI2/Slug dependent NED. Moreover, SOX2 upregulated neural CAMs, neurotrophins/neurotrophin receptors, pluripotency and epithelial-mesenchymal transition transcription factors, growth, angiogenic and lymphangiogenic factors, and promoted PC cell invasiveness and motility. This study discloses novel SOX2 target genes driving NED and spread of PC and proposes SOX2 as a functional biomarker of LN metastasization for PC.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , SOXB1 Transcription Factors/genetics , Aged , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Progression , Down-Regulation , Humans , Lymphatic Metastasis , Male , Middle Aged , Transfection
18.
Haematologica ; 101(3): e107-10, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26659917
19.
Oncotarget ; 6(41): 43267-80, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26657115

ABSTRACT

IL-27 is a member of the IL-12 family that is produced by macrophages and dendritic cells. IL-27 inhibits the growth and invasiveness of different cancers and therefore represents a potential anti-tumor agent. By contrast, it may exert immune-regulatory properties in different biological systems. We reported that IL-27 induces the expression of the IL-18 inhibitor IL-18BP, in human Epithelial Ovarian Cancer (EOC) cells, thus potentially limiting the immune response. Here, we tested whether IL-27 may modulate other immune-regulatory molecules involved in EOC progression, including Indoleamine 2,3-dioxygenase (IDO) and Programmed Death-Ligand (PD-L)1. IDO and PD-L1 were not constitutively expressed by EOC cells in vitro, but IL-27 increased their expression through STAT1 and STAT3 tyrosine phosphorylation. Differently, cells isolated from EOC ascites showed constitutive activation of STAT1 and STAT3 and IDO expression. These findings, together with the expression of IL-27 in scattered leukocytes in EOC ascites and tissues, suggest a potential role of IL-27 in immune-regulatory networks of EOC. In addition, IL-27 induced IDO or PD-L1 expression in monocytes and in human PC3 prostate and A549 lung cancer cells. A current paradigm in tumor immunology is that tumor cells may escape from immune control due to "adaptive resistance" mediated by T cell-secreted IFN-γ, which induces PD-L1 and IDO expression in tumor cells. Our present data indicate that also IL-27 has similar activities and suggest that the therapeutic use of IL-27 as anti-cancer agent may have dual effects, in some tumors.


Subject(s)
B7-H1 Antigen/metabolism , Gene Expression Regulation, Neoplastic/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Blotting, Western , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , Polymerase Chain Reaction , RNA, Small Interfering , Transfection
20.
Oncotarget ; 6(19): 17121-34, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25686823

ABSTRACT

Prostate Cancer (PCa)-related deaths are mostly due to metastasization of poorly differentiated adenocarcinomas often endowed with neuroendocrine differentiation (NED) areas.The SNAI2/Slug gene is a major regulator of cell migration and tumor metastasization. We here assessed its biological significance in NED, and metastatic potential of PCa.SNAI2 expression was down-regulated in most PCa epithelia, in association with gene promoter methylation, except for cell clusters forming: a. the expansion/invasion front of high-grade PCa, b. NED areas, or c. lymph node metastasis.Knockdown of SNAI2 in PC3 cells down-regulated the expression of neural-tissue-associated adhesion molecules, Neural-Cadherin, Neural-Cadherin-2, Neuronal-Cell-Adhesion-Molecule, and of the NED marker Neuron-Specific Enolase, whereas it abolished Chromogranin-A expression. The metastasis-suppressor genes, Nm23-H1 and KISS1, were up-regulated, while the pluripotency genes SOX2, NOTCH1, CD44v6, WWTR1/TAZ and YAP1 were dramatically down-regulated. Over-expression of SNAI2 in DU145 cells substantiated its ability to regulate metastasis-suppressor, NED and pluripotency genes. In PCa and lymph node metastasis, expression of SOX2 and NOTCH1 was highly related to that of SNAI2.In conclusion, I. SNAI2 silencing in PCa may turn-off the expression of NED markers and pluripotency genes, while turning-on that of specific metastasis-suppressors, II. SNAI2 expression in selected PCa cells, by regulating their self-renewal, NED and metastatic potential, endows them with highly malignant properties. SNAI2 may thus constitute a key target for modern approaches to PCa progression.


Subject(s)
Adenocarcinoma/pathology , Cell Differentiation , Gene Expression Regulation, Neoplastic/genetics , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Adenocarcinoma/secondary , Aged , Blotting, Western , Cell Differentiation/genetics , Cell Line, Tumor , DNA Methylation/genetics , Down-Regulation , Gene Silencing , Humans , Immunohistochemistry , Laser Capture Microdissection , Male , Middle Aged , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...