Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Bus Contin Emer Plan ; 15(1): 17-29, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-34465407

ABSTRACT

Exercise GTA Unified was a functional, multi-agency, cross-jurisdictional, health-sector focused mass casualty preparedness exercise conducted in the Greater Toronto Area (GTA) on 28th November, 2019. With over 1,000 unique paper-based and electronic injects and 34 participating agencies, including 22 separate hospital sites, Exercise GTA Unified is likely the largest health-sector focused mass casualty preparedness exercise ever conducted in Canada. The exercise design approach supported a successful, objective-based functional exercise, with elements of marked realism for participants. The exercise offered a unique opportunity to collect data for future analysis and the insights gained will have a transformative impact on interagency engagement and cooperation for emergency response planning. Furthermore, the approach adopted for the exercise is affordable, reproducible, scalable and transferrable to sectors beyond the health system. This paper provides a detailed review of the key planning and design components adopted in the development and implementation of the exercise, as well as practical insights for the design and conduct of multi-agency, cross-jurisdictional functional exercises.


Subject(s)
Disaster Planning , Mass Casualty Incidents , Canada , Emergencies , Emergency Service, Hospital , Humans
2.
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
3.
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
4.
J Bus Contin Emer Plan ; 13(1): 6-21, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-31462359

ABSTRACT

This case study describes a response to a real mass-casualty incident from the perspective of Sunnybrook Health Sciences Centre, a major trauma centre in Toronto, on a day when hospital occupancy levels were at a peak. To help evaluate the lessons learned from the incident response, the article also outlines the hospital's preparedness planning and the readiness assessment it conducted six months prior to the incident. Lessons learned from the readiness assessment are closely linked to those of the real incident, highlighting the importance of readiness assessments and that routine exercises are effective methods to identify opportunities for improvement and enhance preparedness. The mitigation of issues related to interoperability during an emergency requires investment in education, planning and preparedness, as well as routine training and exercises specific to mass-casualty incidents, with the inclusion of impacted departments and services across the hospital setting. Additionally, the inclusion of surge and overcapacity management principles within hospital emergency plans would benefit from further case studies as a method to maintain continuity of operations during these events.


Subject(s)
Disaster Planning , Mass Casualty Incidents , Emergency Service, Hospital
5.
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
6.
Dev Biol ; 407(2): 224-31, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26434917

ABSTRACT

The IMD pathway induces the innate immune response to infection by gram-negative bacteria. We demonstrate strong female-to-male sex transformations in double mutants of the IMD pathway in combination with Doa alleles. Doa encodes a protein kinase playing a central role in somatic sex determination through its regulation of alternative splicing of dsx transcripts. Transcripts encoding two specific Doa isoforms are reduced in Rel null mutant females, supporting our genetic observations. A role for the IMD pathway in somatic sex determination is further supported by the induction of female-to-male sex transformations by Dredd mutations in sensitized genetic backgrounds. In contrast, mutations in either dorsal or Dif, the two other NF-κB paralogues of Drosophila, display no effects on sex determination, demonstrating the specificity of IMD signaling. Our results reveal a novel role for the innate immune IMD signaling pathway in the regulation of somatic sex determination in addition to its role in response to microbial infection, demonstrating its effects on alternative splicing through induction of a crucial protein kinase.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Genetic Loci , Immunity, Innate/genetics , Protein Serine-Threonine Kinases/genetics , Sex Determination Processes/genetics , Alleles , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/ultrastructure , Epistasis, Genetic , Female , Gene Expression Regulation , Genes, Insect , Heterozygote , Male , Mutation/genetics , NF-kappa B/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/genetics , Transcription, Genetic
7.
Blood ; 125(15): 2349-58, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25612623

ABSTRACT

We prospectively assessed functional and phenotypic characteristics of γδ T lymphocytes up to 7 months after HLA-haploidentical hematopoietic stem cell transplantation (haplo-HSCT) depleted of αß(+) T cells and CD19(+) B cells in 27 children with either malignant or nonmalignant disorders. We demonstrate that (1) γδ T cells are the predominant T-cell population in patients during the first weeks after transplantation, being mainly, albeit not only, derived from cells infused with the graft and expanding in vivo; (2) central-memory cells predominated very early posttransplantation for both Vδ1 and Vδ2 subsets; (3) Vδ1 cells are specifically expanded in patients experiencing cytomegalovirus reactivation and are more cytotoxic compared with those of children who did not experience reactivation; (4) these subsets display a cytotoxic phenotype and degranulate when challenged with primary acute myeloid and lymphoid leukemia blasts; and (5) Vδ2 cells are expanded in vitro after exposure to zoledronic acid (ZOL) and efficiently lyse primary lymphoid and myeloid blasts. This is the first detailed characterization of γδ T cells emerging in peripheral blood of children after CD19(+) B-cell and αß(+) T-cell-depleted haplo-HSCT. Our results can be instrumental to the development of clinical trials using ZOL for improving γδ T-cell killing capacity against leukemia cells. This trial was registered at www.clinicaltrials.gov as #NCT01810120.


Subject(s)
Antigens, CD19/analysis , B-Lymphocytes/cytology , Hematopoietic Stem Cell Transplantation , Leukemia/therapy , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocytes/transplantation , Adolescent , Cell Degranulation , Cells, Cultured , Child , Child, Preschool , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Male , T-Lymphocytes/cytology
8.
Oncotarget ; 5(21): 10368-81, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25362644

ABSTRACT

We have previously demonstrated that Tenascin-C (TNC)(+) human neuroblastoma (NB) cells transdifferentiate into tumor-derived endothelial cells (TDEC), which have been detected both in primary tumors and in tumors formed by human NB cell lines in immunodeficient mice. TDEC are genetically unstable and may favor tumor progression, suggesting that their elimination could reduce tumor growth and dissemination. So far, TDEC have never been targeted by antibody-mediated immunotherapy in any of the tumor models investigated. To address this issue, immunodeficient mice carrying orthotopic NB formed by the HTLA-230 human cell line were treated with TDEC-targeting cytotoxic human (h)CD31, that spares host-derived endothelial cells, or isotype-matched mAbs. hCD31 mAb treatment did not affect survival of NB-bearing mice, but increased significantly hypoxia in tumor microenvironment, where apoptotic and proliferating TDEC coexisted, indicating the occurrence of vascular remodeling. Tumor cells from hCD31 mAb treated mice showed i) up-regulation of epithelial-mesenchymal transition (EMT)-related and vascular mimicry (VM)-related gene expression, ii) expression of endothelial (i.e. CD31 and VE-cadherin) and EMT-associated (i.e. Twist-1, N-cadherin and TNC) immunophenotypic markers, and iii) up-regulation of high mobility group box-1 (HMGB-1) expression. In vitro experiments with two NB cell lines showed that hypoxia was the common driver of all the above phenomena and that human recombinant HMGB-1 amplified EMT and TDEC trans-differentiation. In conclusion, TDEC targeting with hCD31 mAb increases tumor hypoxia, setting the stage for the occurrence of EMT and of new waves of TDEC trans-differentiation. These adaptive responses to the changes induced by immunotherapy in the tumor microenvironment allow tumor cells to escape from the effects of hCD31 mAb.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Endothelial Cells/metabolism , Immunotherapy/methods , Neuroblastoma/therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Transdifferentiation , Epithelial-Mesenchymal Transition/drug effects , Female , HMGB1 Protein/metabolism , Humans , Hypoxia/chemically induced , Immunotherapy/adverse effects , Mice, Nude , Neuroblastoma/immunology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Tenascin/metabolism , Treatment Failure , Tumor Escape , Tumor Microenvironment , Vascular Remodeling , Xenograft Model Antitumor Assays
9.
Immunotherapy ; 4(7): 668, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23019744
10.
12.
J Leukoc Biol ; 92(3): 567-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22591693

ABSTRACT

AML is a hematologic malignancy that represents 15-20% of all childhood acute leukemias and is responsible for more than one-half of pediatric leukemic deaths. The bulk tumor is continuously regenerated and sustained by rare leukemic ICs that proliferate slowly, thus resulting refractory to chemotherapeutic agents targeting highly proliferating cells within the tumor. Therefore, a complete eradication of the bulk tumor may depend on efficacy of therapies that target IC. In spite of the improvements in the treatment of AML, the difficulty to eradicate completely the disease incites research for innovative therapeutic approaches. In this regard, the role of cytokines in the treatment of AML has been investigated for many years, and some of them have been tested in clinical trials as a result of their immunomodulatory properties. Furthermore, recent preclinical studies highlighted the ability of the IL-12 superfamily cytokines as potent antileukemic agents that act directly on tumor cells and on leukemic IC, thus opening new perspectives for leukemic patient treatment. Here, we review the current knowledge about the antileukemic effects of cytokines, documented in preclinical and clinical studies, discussing their potential clinical application.


Subject(s)
Cytokines/pharmacology , Immunologic Factors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cytokines/immunology , Humans , Immunologic Factors/immunology , Leukemia, Myeloid, Acute/immunology , Neoplastic Stem Cells/immunology
13.
Clin Cancer Res ; 18(6): 1630-40, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22383738

ABSTRACT

PURPOSE: Acute myeloid leukemia (AML) accounts for more than half of fatal cases in all pediatric leukemia patients; this observation highlights the need of more effective therapies. Thus, we investigated whether interleukin (IL)-27, an immunomodulatory cytokine, functions as an antitumor agent against pediatric AML cells. EXPERIMENTAL DESIGN: Expression of WSX-1 and gp130 on AML cells from 16 pediatric patients was studied by flow cytometry. Modulation of leukemia cell proliferation or apoptosis upon IL-27 treatment in vitro was tested by bromodeoxyuridine/propidium iodide (PI) and Ki67, or Annexin V/PI staining and flow cytometric analysis. The angiogenic potential of AML cells treated or not with IL-27 was studied by chorioallantoic membrane assay and PCR array. In vivo studies were carried out using nonobese diabetic/severe combined immunodeficient (NOD/SCID)/Il2rg(-/-) mice injected intravenously with five pediatric AML cell samples. Leukemic cells engrafted in PBS and IL-27-treated animals were studied by immunohistochemical/morphologic analysis and by PCR array for expression angiogenic/dissemination-related genes. RESULTS: We provided the first demonstration that (i) AML cells injected into NOD/SCID/Il2rg(-/-) mice gave rise to leukemia dissemination that was severely hampered by IL-27, (ii) compared with controls, leukemia cells harvested from IL-27-treated mice showed significant reduction of their angiogenic and spreading related genes, and (iii) similarly to what was observed in vivo, IL-27 reduced in vitro AML cell proliferation and modulated the expression of different genes involved in the angiogenic/spreading process. CONCLUSION: These results provide an experimental rationale for the development of future clinical trials aimed at evaluating the toxicity and efficacy of IL-27.


Subject(s)
Antineoplastic Agents/therapeutic use , Interleukin-17/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Proliferation/drug effects , Child , Child, Preschool , Female , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/genetics , Xenograft Model Antitumor Assays , Young Adult
14.
Crit Rev Oncol Hematol ; 83(3): 310-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22177566

ABSTRACT

B acute lymphoblastic leukemia (ALL) is the most common pediatric hematologic malignancy. Although patient cure has reached an excellent rate, a minority of cases relapse and need novel therapies. IL-12, IL-23 and IL-27 belong to the IL-12 superfamily and exert immunological and anti-tumor functions. The latter can be mediated by activation of immune responses or by the direct activity on cancer cells. Recently, the role of IL-12, IL-23 and IL-27 in the control of pediatric B-ALL has been unveiled. Here, we discuss in a translational perspective the role of IL-12 family cytokines in pediatric B-ALL, highlighting similarities and differences in their mechanisms of action.


Subject(s)
Antineoplastic Agents/therapeutic use , Interleukin-12/therapeutic use , Interleukin-23/therapeutic use , Interleukins/therapeutic use , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Humans , Interleukin-12/immunology , Interleukin-23/immunology , Interleukins/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology
15.
Cytokine Growth Factor Rev ; 22(3): 149-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21764355

ABSTRACT

B-acute lymphoblastic leukemia (ALL) is the most common hematologic tumor of pediatric age. Although patient survival has been improved, some cases still relapse and need alternative therapies. In this context, the role of microRNA in cancer is actually matter of investigation due to their regulatory function implicated in human tumorigenesis since the main target mRNA transcripts are involved in proliferation, apoptosis and differentiation. On the other hand, cytokines are actually used as adjuvants in B-ALL therapy, and recent studies reported that they may also function by acting directly against leukemic cells. Here we review the current knowledge about the role of miRNA and cytokines in B-ALL, highlighting the link between cytokine activity and miRNA expression. The translational relevance of these issues will be also discussed.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation, Leukemic , MicroRNAs/biosynthesis , Neoplasm Proteins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Neoplasm/biosynthesis , Adolescent , Child , Child, Preschool , Cytokines/immunology , Humans , Infant , MicroRNAs/immunology , Neoplasm Proteins/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , RNA, Messenger/immunology , RNA, Messenger/metabolism , RNA, Neoplasm/immunology
16.
Cell Res ; 21(10): 1470-86, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21403679

ABSTRACT

Neuroblastoma (NB)-associated endothelial microvessels (EMs) may be lined by tumor-derived endothelial cells (TECs), that are genetically unstable and chemoresistant. Here we have addressed the identification of TEC progenitors in NB by focusing on Octamer-binding transcription factor 4 (Oct-4) as a putative marker. Oct-4(+) cells were detected in primary NB samples (n = 23), metastatic bone marrow aspirates (n = 10), NB cell lines (n = 4), and orthotopic tumors (n = 10) formed by the HTLA-230 NB cell line in immunodeficient mice. Most Oct-4(+) cells showed a perivascular distribution, with 5% of them homing in perinecrotic areas. All Oct-4(+) cells were tumor-derived since they shared amplification of MYCN oncogene with malignant cells. Perivascular Oct-4(+) cells expressed stem cell-related, neural progenitor-related and NB-related markers, including surface Tenascin C (TNC), that was absent from perinecrotic Oct-4(+) cells and bulk tumor cells. TNC(+) but not TNC(-) HTLA-230 cells differentiated in vitro into endothelial-like cells expressing vascular-endothelial-cadherin, prostate-specific membrane antigen and CD31 upon culture in medium containing vascular endothelial growth factor (VEGF). TNC(+) but not TNC(-) HTLA-230 cells formed neurospheres when cultured in serum-free medium. Both cell fractions were tumorigenic, but only tumors formed by TNC(+) cells contained EMs lined by TECs. In conclusion, we have identified in NB tumors two putative niches containing Oct-4(+) tumor cells. Oct-4(+)/TNC(+) perivascular NB cells displayed a high degree of plasticity and served as progenitors of TECs. Therapeutic targeting of Oct4(+)/TNC(+) progenitors may counteract the contribution of NB-derived ECs to tumor relapse and chemoresistance.


Subject(s)
Cell Differentiation , Endothelial Cells/metabolism , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Neuroblastoma/metabolism , Octamer Transcription Factor-3/metabolism , Tenascin/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Child , Child, Preschool , Drug Resistance, Neoplasm/genetics , Endothelial Cells/pathology , Female , Gene Amplification , Humans , Infant , Male , Mice , Mice, Nude , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Tenascin/genetics , Transplantation, Heterologous
17.
J Leukoc Biol ; 89(5): 729-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21330353

ABSTRACT

This study tested the hypothesis that IL-27 and IL-23, two heterodimeric cytokines involved in physiological immune responses and immunological disorders, may function on human PC and plasmablasts. It was reported that IL-27 acts on human mature B cells, but the role of IL-27 and IL-23 in human PC remains to be established. Thus, we have asked whether these cytokines may modulate human PC functions using human PPC generated in vitro, PC isolated from tonsils, and BM. Here, we show for the first time that PC and PPC express complete IL-27R and that IL-27 exerts chemotactic properties on these cells and modulates different chemokines/chemokine receptors and secretion of IgM and IgG. Furthermore, we demonstrated that PC and PPC express both chains of IL-23R and IL-23 and exerted similar activity to IL-27 in terms of Ig production, while not inducing PC chemotaxis. These results may provide novel insight into the role of IL-27 and IL-23 in human B cell immune responses.


Subject(s)
Cell Movement , Interleukin-17/metabolism , Interleukin-23/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , B-Lymphocytes/metabolism , Chemokines/metabolism , Chemotaxis , Humans , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Receptors, Chemokine/metabolism , Signal Transduction
18.
Immunol Lett ; 133(2): 99-105, 2010 Oct 30.
Article in English | MEDLINE | ID: mdl-20705102

ABSTRACT

Acute myeloid leukemia is a haematopoietic malignancy originating from the transformation of myeloid progenitors that proliferate and accumulate in the bone marrow. In AML patients the survival rate at 5 years is 40-50% highlighting the need for novel therapies. In this study we have asked whether IL-12, an immuno-modulatory cytokine with anti-tumor activity, may inhibit directly AML cell growth. We show that the human AML cell lines U937, K562 and THP-1 expressed both chains of the IL-12 receptor (R), i.e. IL-12Rß1 and IL-12Rß2. IL-12 inhibited the angiogenic potential of AML cells in vitro, but did not affect their survival or proliferation. In vivo experiments were performed using SCID-NOD mice injected intraperitoneally (i.p.) with the human U937 AML cell line and subsequently treated with human recombinant IL-12 or PBS i.p. Histological, immunohistochemical and flow cytometric analyses on explanted tumors revealed that IL-12 reduced new vessel formation, induced apoptosis and inhibited tumor cell proliferation. Studies on a panel of angiogenesis related genes in explanted tumors using PCR arrays showed significantly down-regulated expression of numerous pro-angiogenic genes including VEGF-C, IL-6, IL-8, CXCL1, CXCL6 and alanyl aminopeptidase in IL-12 vs PBS treated mice. This study shows for the first time that IL-12 targets directly AML cell growth and paves the way to further investigation of IL-12 as potential drug for AML treatment.


Subject(s)
Cell Proliferation/drug effects , Growth Inhibitors/administration & dosage , Interleukin-12/administration & dosage , Leukemia, Myeloid, Acute/immunology , Receptors, Interleukin-12/metabolism , Animals , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Growth Inhibitors/pharmacology , Humans , Interleukin-12/pharmacology , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , U937 Cells
19.
Blood ; 116(19): 3887-98, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20671120

ABSTRACT

Interleukin (IL)-23 is a proinflammatory cytokine belonging to the IL-12 superfamily. The antitumor activity of IL-23 is controversial, and it is unknown whether or not the cytokine can act directly on tumor cells. The aim of this study was to investigate the potential direct antitumor activity of IL-23 in pediatric B-acute lymphoblastic leukemia (B-ALL) cells and to unravel the molecular mechanisms involved. Here, we show, for the first time, that IL-23R is up-regulated in primary B-ALL cells, compared with normal early B lymphocytes, and that IL-23 dampens directly tumor growth in vitro and in vivo through the inhibition of tumor cell proliferation and induction of apoptosis. The latter finding is related to IL-23-induced up-regulation of miR15a expression and the consequent down-regulation of BCL-2 protein expression in pediatric B-ALL cells. This study demonstrates that IL-23 possesses antileukemic activity and unravels the underlying mechanisms. Thus, IL-23 may be a candidate novel drug for the treatment of B-ALL patients unresponsive to current therapeutic standards.


Subject(s)
Interleukin-23 Subunit p19/immunology , Interleukin-23 Subunit p19/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Child, Preschool , Female , Gene Expression/drug effects , Genes, bcl-2/drug effects , Humans , In Vitro Techniques , Infant , Interleukin-23 Subunit p19/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Signal Transduction/immunology
20.
Clin Cancer Res ; 16(16): 4188-97, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20587591

ABSTRACT

PURPOSE: Multiple myeloma (MM) derives from plasmablast/plasma cells that accumulate in the bone marrow. Different microenvironmental factors may promote metastatic dissemination especially to the skeleton, causing bone destruction. The balance between osteoclast and osteoblast activity represents a critical issue in bone remodeling. Thus, we investigated whether interluekin-27 (IL-27) may function as an antitumor agent by acting directly on MM cells and/or on osteoclasts/osteoblasts. EXPERIMENTAL DESIGN: The IL-27 direct antitumor activity on MM cells was investigated in terms of angiogenesis, proliferation, apoptosis, and chemotaxis. The IL-27 activity on osteoclast/osteoblast differentiation and function was also tested. In vivo studies were done using severe combined immunodeficient/nonobese diabetic mice injected with MM cell lines. Tumors from IL-27- and PBS-treated mice were analyzed by immunohistochemistry and PCR array. RESULTS: We showed that IL-27 (a) strongly inhibited tumor growth of primary MM cells and MM cell lines through inhibition of angiogenesis, (b) inhibited osteoclast differentiation and activity and induced osteoblast proliferation, and (c) damped in vivo tumorigenicity of human MM cell lines through inhibition of angiogenesis. CONCLUSIONS: These findings show that IL-27 may represent a novel therapeutic agent capable of inhibiting directly MM cell growth as well as osteoclast differentiation and activity.


Subject(s)
Antineoplastic Agents/therapeutic use , Interleukins/therapeutic use , Multiple Myeloma/drug therapy , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Separation , Chemotaxis, Leukocyte/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Multiple Myeloma/pathology , Neoplasm Staging , Neovascularization, Pathologic/drug therapy , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...