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.
Hematol Oncol ; 41(3): 578-582, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36043430

ABSTRACT

We hypothesized that ruxolitinib may inhibit the immune checkpoint protein, B7H3; and, thus, investigated its effects on this immune inhibitor using multiple myeloma (MM) cell lines, bone marrow (BM) mononuclear cells from MM patients and human MM LAGλ -1A xenografts. Ruxolitinib reduced B7H3 gene and protein expression and increased IL-2 and CD8 gene expression. These results suggest that ruxolitinib inhibition of B7H3 may restore exhausted T-cell activity in the MM BM tumor microenvironment.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/pathology , Immune Checkpoint Proteins/pharmacology , Janus Kinase 1 , Signal Transduction , Tumor Microenvironment
2.
Br J Haematol ; 192(3): 568-576, 2021 02.
Article in English | MEDLINE | ID: mdl-33341940

ABSTRACT

Multiple myeloma (MM) tumour cells evade host immunity through a variety of mechanisms, which may potentially include the programmed cell death ligand-1 (PD-L1):programmed cell death protein-1 (PD-1) axis. This interaction contributes to the immunosuppressive bone marrow (BM) microenvironment, ultimately leading to reduced effector cell function. PD-L1 is overexpressed in MMBM and is associated with the resistance to immune-based approaches for treating MM. Ruxolitinib (RUX), an inhibitor of the Janus kinase (JAK) family of protein tyrosine kinases, is approved for myeloproliferative diseases. We investigated the effects of RUX alone or in combination with anti-MM agents on the expression of PD-L1 and T-cell cytotoxicity in MM. We showed that the expression of the PD-L1 gene was markedly increased in BM mononuclear cells from patients with MM with progressive disease versus those in complete remission. Furthermore, RUX treatment resulted in a concentration-dependent reduction of PD-L1 gene expression in the MM tumour cells cultured alone or co-cultured with stromal cells compared with untreated cells. The results also demonstrated that RUX increased MM cell apoptosis in the presence of interleukin-2-stimulated T cells to a similar degree as the treatment with anti-PD-1 or anti-PD-L1 antibodies. In summary, these results indicate that RUX can block PD-L1 expression resulting in augmentation of anti-MM effects of T cells.


Subject(s)
Antineoplastic Agents/therapeutic use , B7-H1 Antigen/genetics , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Animals , Apoptosis/drug effects , Down-Regulation/drug effects , Humans , Janus Kinases/antagonists & inhibitors , Male , Mice, SCID , Multiple Myeloma/genetics , Nitriles , Pyrimidines , Tumor Microenvironment/drug effects
3.
Br J Haematol ; 188(2): 283-294, 2020 01.
Article in English | MEDLINE | ID: mdl-31423579

ABSTRACT

Monocytes polarize into pro-inflammatory macrophage-1 (M1) or alternative macrophage-2 (M2) states with distinct phenotypes and physiological functions. M2 cells promote tumour growth and metastasis whereas M1 macrophages show anti-tumour effects. We found that M2 cells were increased whereas M1 cells were decreased in bone marrow (BM) from multiple myeloma (MM) patients with progressive disease (PD) compared to those in complete remission (CR). Gene expression of Tribbles homolog 1 (TRIB1) protein kinase, an inducer of M2 polarization, was increased in BM from MM patients with PD compared to those in CR. Ruxolitinib (RUX) is an inhibitor of the Janus kinase family of protein tyrosine kinases (JAKs) and is effective for treating patients with myeloproliferative disorders. RUX markedly reduces both M2 polarization and TRIB1 gene expression in MM both in vitro and in vivo in human MM xenografts in severe combined immunodeficient mice. RUX also downregulates the expression of CXCL12, CXCR4, MUC1, and CD44 in MM cells and monocytes co-cultured with MM tumour cells; overexpression of these genes is associated with resistance of MM cells to the immunomodulatory agent lenalidomide. These results provide the rationale for evaluation of JAK inhibitors, including MM BM in combination with lenalidomide, for the treatment of MM patients.


Subject(s)
Chemokines, CXC/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinases/metabolism , Lenalidomide/pharmacology , Mucin-1/biosynthesis , Multiple Myeloma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Case-Control Studies , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Chemokines, CXC/metabolism , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, SCID , Monocytes/drug effects , Monocytes/metabolism , Mucin-1/metabolism , Multiple Myeloma/blood , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/metabolism , Signal Transduction , THP-1 Cells
4.
Leuk Res ; 81: 62-66, 2019 06.
Article in English | MEDLINE | ID: mdl-31035033

ABSTRACT

B-cell maturation antigen (BCMA), a tumor necrosis factor receptor (TNFR) family member, is selectively expressed on terminally differentiated B-lymphocytes including multiple myeloma (MM) tumor cells. We sought to determine whether circulating (c)BCMA in MM serum interferes with antiBCMA antibody binding to MM cells. An enzyme-linked immunosorbent assay (ELISA) was used to determine serum (s) BCMA levels among 379 samples from patients with relapsed/refractory MM (RRMM). Furthermore, flow cytometric and immunofluorescent studies were used to examine if concentrations of BCMA in patients' serum were high enough to interfere with the binding of anti-BCMA antibody to MM tumor cells. We have shown that BCMA is elevated in the serum from MM patients and that the median concentration of sBCMA from RRMM patients was 176 ng/mL (n = 379). Additionally, there was a consistent decrease in the binding of anti-BCMA antibody to MM tumor cells with sBCMA level ≥156 ng/mL. Together, these results demonstrate that circulating BCMA levels in most RRMM patients are high enough to interfere with anti-BCMA antibody binding to MM tumor cells and may interfere with BCMA-targeted immune-based therapies.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/metabolism , B-Cell Maturation Antigen/metabolism , Biomarkers, Tumor/metabolism , Multiple Myeloma/pathology , Antibodies, Monoclonal/immunology , Antibody Affinity , Antigen-Antibody Complex/immunology , B-Cell Maturation Antigen/immunology , Binding Sites, Antibody , Case-Control Studies , Humans , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Prognosis
5.
Ann Hematol ; 98(3): 691-703, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30635766

ABSTRACT

The Janus kinase (JAK) pathway has been shown to play key roles in the growth and resistance to drugs that develop in multiple myeloma (MM) patients. The anti-MM effects of the selective JAK1 inhibitor INCB052793 (INCB) alone and in combination with anti-MM agents were evaluated in vitro and in vivo. Significant inhibition of cell viability of primary MM cells obtained fresh from MM patients, and the MM cell lines RPMI8226 and U266, was observed with single agent INCB and was enhanced in combination with other anti-MM agents including proteasome inhibitors and glucocorticosteroids. Single-agent INCB resulted in decrease in tumor growth of the MM xenograft LAGκ-1A growing in severe combined immunodeficient mice. Mice dosed with INCB (30 mg/kg) showed significant reductions in tumor volume on days 28, 35, 42, 49, 56, and 63. Similarly, INCB at 10 mg/kg showed anti-tumor effects on days 56 and 63. Tumor-bearing mice receiving combinations of INCB with carfilzomib, bortezomib, dexamethasone, or lenalidomide showed significantly smaller tumors when compared to vehicle control and mice treated with single agents. These results provide further support for the clinical evaluation of INCB052793 alone and in combination treatment for MM patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Janus Kinase 1/antagonists & inhibitors , Multiple Myeloma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bortezomib/administration & dosage , Bortezomib/pharmacology , Cell Line, Tumor , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Lenalidomide/administration & dosage , Lenalidomide/pharmacology , Male , Mice, SCID , Molecular Targeted Therapy , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Specific Pathogen-Free Organisms , Xenograft Model Antitumor Assays
6.
Expert Rev Mol Diagn ; 18(4): 319-329, 2018 04.
Article in English | MEDLINE | ID: mdl-29504446

ABSTRACT

INTRODUCTION: B-cell maturation antigen (BCMA) is a cell membrane bound tumor necrosis factor receptor family member that is expressed exclusively on late stage normal and malignant B-cells and plasma cells. Addition of two of its ligands, B-cell activating factor and a proliferation inducting ligand, to normal B-cells cause B-cell proliferation and antibody production. Serum BCMA is elevated among patients with multiple myeloma (MM) and chronic lymphocytic leukemia (CLL), and is a prognostic and monitoring tool for these patients. The first anti-BCMA antibody (Ab) was developed in 2007. Recently, biotech and pharmaceutical companies have created various forms of BCMA-directed Abs (naked Abs, Ab drug conjugates, and bispecific Abs) and cellular therapies (chimeric antigen receptor T-cells) with promising clinical results. Areas covered: This BCMA review encompasses full-text publications of original research articles and abstracts presented at hematology/oncology meetings. Expert commentary: The limited preclinical and ongoing clinical studies published to date evaluating BCMA-directed therapies have shown great promise. It has also been demonstrated that BCMA is solubilized and elevated in the blood of MM, Waldenstrom's macroglobulinemia and CLL patients, and is also responsible for the immune deficiency in MM. Reducing circulating levels may improve the efficacy of these treatments.


Subject(s)
B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/blood , Biomarkers, Tumor/blood , B-Cell Activating Factor/blood , B-Cell Activating Factor/immunology , B-Cell Maturation Antigen/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Humans , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Multiple Myeloma/therapy , Tumor Necrosis Factor Ligand Superfamily Member 13/blood , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
7.
Target Oncol ; 13(1): 39-47, 2018 02.
Article in English | MEDLINE | ID: mdl-29230672

ABSTRACT

B-cell maturation antigen (BCMA) was originally identified as a cell membrane receptor, expressed exclusively on late stage B-cells and plasma cells (PCs). Investigations of BCMA as a target for therapeutic intervention in multiple myeloma (MM) were initiated in 2007, using cSG1 as a naked antibody (Ab) as well as an Ab-drug conjugate (ADC) targeting BCMA, ultimately leading to ongoing clinical studies for previously treated MM patients. Since then, multiple companies have developed anti-BCMA-directed ADCs. Additionally, there are now three bispecific antibodies in development, which bind to both BCMA and CD3ε on T-cells. This latter binding results in T-cell recruitment and activation, causing target cell lysis. More recently, T-cells have been genetically engineered to recognize BCMA-expressing cells and, in 2013, the first report of anti-BCMA-chimeric antigen receptor T-cells showed that these killed MM cell lines and human MM xenografts in mice. BCMA is also solubilized in the blood (soluble BCMA [sBCMA]) and MM patients with progressive disease have significantly higher sBCMA levels than those responding to treatment. sBCMA circulating in the blood may limit the efficacy of these anti-BCMA-directed therapies. When sBCMA binds to B-cell activating factor (BAFF), BAFF is unable to perform its major biological function of inducing B-cell proliferation and differentiation into Ab-secreting PC. However, the use of γ-secretase inhibitors, which prevent shedding of BCMA from PCs, may improve the efficacy of these BCMA-directed therapies.


Subject(s)
B-Lymphocytes/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Humans , Multiple Myeloma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...