Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Cancer J ; 5: e328, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26230952

ABSTRACT

The biological role of monocytes and macrophages in B-cell non-Hodgkin lymphoma (NHL) is not fully understood. We have previously reported that monocytes from patients with B-cell NHL have an immunosuppressive CD14(+)HLA-DR(low/-) phenotype that correlates with a poor prognosis. However, the underlying mechanism by which CD14(+)HLA-DR(low/-) monocytes develop in lymphoma is unknown. In the present study, we found that interleukin (IL)-10, which is increased in the serum of patients with B-cell NHL, induced the development of the CD4(+)HLA-DR(low/-) population. Using peripheral blood samples from patients with B-cell NHL, we found that absolute numbers of CD14(+) monocytic cells with an HLA-DR(low/-) phenotype were higher than healthy controls and correlated with a higher International Prognostic Index score. IL-10 serum levels were elevated in lymphoma patients compared with controls and were associated with increased peripheral monocyte counts. Treatment of monocytes with IL-10 in vitro significantly decreased HLA-DR expression and resulted in the expansion of CD14(+)HLA-DR(low/-) population. We found that lymphoma B cells produce IL-10 and supernatants from cultured lymphoma cells increased the CD14(+)HLA-DR(low/-) population. Furthermore, we found that IL-10-induced CD14(+)HLA-DR(low/-) monocytes inhibited the activation and proliferation of T cells. Taken together, these results suggest that elevated IL-10 serum levels contribute to increased numbers of immunosuppressive CD14(+)HLA-DR(low/-) monocytes in B-cell NHL.


Subject(s)
Interleukin-10/physiology , Lymphoma, Non-Hodgkin/immunology , Monocytes/metabolism , B-Lymphocytes/metabolism , Case-Control Studies , Cell Proliferation , Cells, Cultured , HLA-DR Antigens/metabolism , Humans , Immune Tolerance , Lipopolysaccharide Receptors/metabolism , Lymphocyte Activation , Lymphoma, Non-Hodgkin/blood , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Blood Cancer J ; 5: e281, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25700246

ABSTRACT

To determine the biological and clinical relevance of programmed death 1 (PD-1) in follicular lymphoma (FL), we characterized PD-1(+) T-cell subsets and assessed their biological function as well as potential clinical impact. We found that PD-1 is expressed on intratumoral CD4(+) T cells with both bright and dim intensity, representing two different sub-populations of cells. By immunohistochemistry, we found that CD4(+)PD-1(high) T cells predominantly reside in the lymph node follicles, while PD-1(low) T cells are mainly located in an interfollicular pattern. Intratumoral CD4(+)PD-1(high) T cells have a TFH cell phenotype, express CXCR5, secrete IL-21 and are BCL-6 positive with no TIM-3 expression. In contrast, CD4(+)PD-1(low) T cells have an exhausted phenotype, express TIM-3 and do not express BCL-6 and CXCR5. Functionally, CD4(+)PD-1(high) T cells actively supported B-cell growth, while CD4(+)PD-1(low) T cells displayed a reduced cytokine production and cell-signal transduction. Clinically, we observed that the numbers of CD4(+) or CD8(+)PD-1(low) T cells significantly correlate with a reduced overall survival in FL patients (P=0.007 and 0.04 respectively; n=32). In contrast, the number of CD4(+)PD-1(high) T cells was not associated with patient outcome. Taken together, these results indicated that PD-1 expression defines two sub-populations with distinct functions that differentially impact patient outcome in FL.


Subject(s)
Lymphoma, Follicular/genetics , Programmed Cell Death 1 Receptor/biosynthesis , Survival Analysis , T-Lymphocyte Subsets/pathology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphoma, Follicular/epidemiology , Lymphoma, Follicular/pathology , Male , Middle Aged , Neoplasm Staging , Programmed Cell Death 1 Receptor/genetics , T-Lymphocyte Subsets/metabolism
3.
Leukemia ; 28(9): 1872-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24569779

ABSTRACT

Transforming growth factor beta (TGF-ß) has an important role in mediating T-cell suppression in B-cell non-Hodgkin lymphoma (NHL). However, the underlying mechanism responsible for TGF-ß-mediated inhibition of effector memory T (Tm) cells is largely unknown. As reported here, we show that exhaustion is a major mechanism by which TGF-ß inhibits Tm cells, and TGF-ß mediated exhaustion is associated with upregulation of CD70. We found that TGF-ß upregulates CD70 expression on effector Tm cells while it preferentially induces Foxp3 expression in naive T cells. CD70 induction by TGF-ß is Smad3-dependent and involves IL-2/Stat5 signaling. CD70+ T cells account for TGF-ß-induced exhaustion of effector Tm cells. Both TGF-ß-induced and preexisting intratumoral CD70+ effector Tm cells from B-cell NHL have an exhausted phenotype and express higher levels of PD-1 and TIM-3 compared with CD70- T cells. Signaling transduction, proliferation and cytokine production are profoundly decreased in these cells, and they are highly susceptible to apoptosis. Clinically, intratumoral CD70-expressing T cells are prevalent in follicular B-cell lymphoma (FL) biopsy specimens, and increased numbers of intratumoral CD70+ T cells correlate with an inferior patient outcome. These findings confirm TGF-ß-mediated effector Tm cell exhaustion as an important mechanism of immune suppression in B-cell NHL.


Subject(s)
CD27 Ligand/physiology , Immunologic Memory , Lymphoma, B-Cell/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/pharmacology , Apoptosis , CD27 Ligand/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatitis A Virus Cellular Receptor 2 , Humans , Interleukin-2/physiology , Membrane Proteins/analysis , Programmed Cell Death 1 Receptor/analysis , STAT5 Transcription Factor/physiology , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology
4.
Leukemia ; 26(5): 1046-52, 2012 May.
Article in English | MEDLINE | ID: mdl-22015775

ABSTRACT

Follicular lymphoma exhibits intratumoral infiltration by non-malignant T lymphocytes, including CD4+CD25+ regulatory T (T(reg)) cells. We combined denileukin diftitox with rituximab in previously untreated, advanced-stage follicular lymphoma patients anticipating that denileukin diftitox would deplete CD25+ T(reg) cells while rituximab would deplete malignant B cells. Patients received rituximab 375 mg/m(2) weekly for 4 weeks and denileukin diftitox 18 mcg/kg/day for 5 days every 3 weeks for 4 cycles; neither agent was given as maintenance therapy. Between August 2008 and March 2010, 24 patients were enrolled. One patient died before treatment was given and was not included in the analysis. Eleven of 23 patients (48%; 95% confidence interval (CI): 27-69%) responded; 2 (9%) had complete responses and 9 (39%) had partial responses. The progression-free rate at 2 years was 55% (95%CI: 37-82%). Thirteen patients (57%) experienced grade ≥3 adverse events and one patient (4%) died. In correlative studies, soluble CD25 and the number of CD25+ T cells decreased after treatment; however, there was a compensatory increase in IL-15 and IP-10. We conclude that although the addition of denileukin diftitox to rituximab decreased the number of CD25+ T cells, denileukin diftitox contributed to the toxicity of the combination without an improvement in response rate or time to progression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, B-Cell/drug therapy , Lymphoma, Follicular/drug therapy , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Diphtheria Toxin/administration & dosage , Female , Humans , Interleukin-2/administration & dosage , Interleukin-2 Receptor alpha Subunit/immunology , Lymphoma, B-Cell/immunology , Lymphoma, Follicular/immunology , Male , Middle Aged , Recombinant Fusion Proteins/administration & dosage , Rituximab , T-Lymphocytes/immunology
5.
Blood Cancer J ; 1(6): e24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22829168

ABSTRACT

MicroRNAs (miRNAs) are involved in the regulation of many cellular processes including hematopoiesis, with the aberrant expression of differentiation-stage specific miRNA associated with lymphomagenesis. miRNA profiling has been essential for understanding the underlying biology of many hematological malignancies; however the miRNA signature of the diverse tumor clone associated with Waldenstrom's macroglobulinemia (WM), consisting of B lymphocytes, plasmacytes and lymphoplasmacytic cells, has not been characterized. We have investigated the expression of over 13 000 known and candidate miRNAs in both CD19(+) and CD138(+) WM tumor cells, as well as in their malignant and non-malignant counterparts. Although neither CD19(+) nor CD138(+) WM cells were defined by a distinct miRNA profile, the combination of all WM cells revealed a unique miRNA transcriptome characterized by the dysregulation of many miRNAs previously identified as crucial for normal B-cell lineage differentiation. Specifically, miRNA-9(*)/152/182 were underexpressed in WM, whereas the expression of miRNA-21/125b/181a/193b/223/363 were notably increased (analysis of variance; P<0.0001). Future studies focusing on the effects of these dysregulated miRNAs will provide further insight into the mechanisms responsible for the pathogenesis of WM.

6.
Leukemia ; 22(12): 2240-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18830264

ABSTRACT

Multiple myeloma (MM) is a progressive disease that results from dysregulated proliferation of plasma cells. Although, causative factors such as genetic events and altered expression of anti-apoptotic factors have been described in a number of patients, the mechanistic details that drive myeloma development and continued growth of malignant cells remain largely undefined. Numerous growth factors, including interleukin (IL)-6, Insulin-like growth factor-1 and IL-10 have been shown to promote growth of MM cells suggesting a significant role for cytokines in this disease. Interferon (IFN)-lambda1 is a new member of the Class II cytokine family that, similar to IFN-alpha, has been shown to mediate viral immunity. In light of data supporting a role for cytokines in myeloma, we investigated the significance of IFN-lambda1 on myeloma cell biology. Our studies show for the first time that myeloma cells bind to soluble IFN-lambda1, and that IFN-lambda1 induces myeloma cell growth and protects against dexamethasone-induced cell death. Our data also show that IFN-lambda1 induces phosphorylation of STAT1, STAT3 and Erk. Taken together, our results suggest that IFN-lambda1 may regulate myeloma cell biology and could prove to be therapeutically important.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Interleukins/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , B-Lymphocytes/immunology , Cell Death/drug effects , Cell Death/immunology , Cell Division/drug effects , Cell Division/immunology , Cell Line, Tumor , Dexamethasone/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucocorticoids/pharmacology , Humans , Interferons , Interleukins/genetics , Interleukins/immunology , Multiple Myeloma/immunology , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured
7.
Leukemia ; 18(3): 616-23, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14749704

ABSTRACT

Survivin is a member of the inhibitor of apoptosis protein (IAP) family and functions both as an apoptosis inhibitor and a regulator of cell division. Survivin overexpression is common in many human tumors and correlates with survival in large cell non-Hodgkin's lymphoma. To evaluate this molecule as a potential therapeutic target in large-cell lymphoma, we evaluated the effect of survivin inhibition both in vitro and in vivo. Using an antisense oligonucleotide (ASO) approach, cell growth was significantly inhibited in the DoHH2, RL and HT lymphoma cell lines. In a lymphoma xenograft model, the development of tumors as well as the growth of established tumors was inhibited in the survivin ASO-treated mice compared to controls. To assess the efficacy of the survivin ASO in combination with other biological agents, we combined the survivin ASO with an anti-CD20 monoclonal antibody, rituximab. The effect of survivin ASO and rituximab in combination was additive in vitro. In vivo, however, suppression of tumor growth with the combination was not significantly superior to controls. We conclude that inhibition of survivin expression is an attractive therapeutic strategy in aggressive non-Hodgkin's lymphomas, and that combining survivin ASO with rituximab may enhance the efficacy of this approach.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, Non-Hodgkin/pathology , Microtubule-Associated Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Division/drug effects , Humans , Inhibitor of Apoptosis Proteins , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/prevention & control , Mice , Mice, SCID , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins , Oligonucleotides, Antisense/pharmacology , Survivin , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...