Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Immunol ; 187(8): 3987-96, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21908738

ABSTRACT

The graft-versus-myeloma (GVM) effect represents a powerful form of immune attack exerted by alloreactive T cells against multiple myeloma cells, which leads to clinical responses in multiple myeloma transplant recipients. Whether myeloma cells are themselves able to induce alloreactive T cells capable of the GVM effect is not defined. Using adoptive transfer of T naive cells into myeloma-bearing mice (established by transplantation of human RPMI8226-TGL myeloma cells into CD122(+) cell-depleted NOD/SCID hosts), we found that myeloma cells induced alloreactive T cells that suppressed myeloma growth and prolonged survival of T cell recipients. Myeloma-induced alloreactive T cells arising in the myeloma-infiltrated bones exerted cytotoxic activity against resident myeloma cells, but limited activity against control myeloma cells obtained from myeloma-bearing mice that did not receive T naive cells. These myeloma-induced alloreactive T cells were derived through multiple CD8(+) T cell divisions and enriched in double-positive (DP) T cells coexpressing the CD8αα and CD4 coreceptors. MHC class I expression on myeloma cells and contact with T cells were required for CD8(+) T cell divisions and DP-T cell development. DP-T cells present in myeloma-infiltrated bones contained a higher proportion of cells expressing cytotoxic mediators IFN-γ and/or perforin compared with single-positive CD8(+) T cells, acquired the capacity to degranulate as measured by CD107 expression, and contributed to an elevated perforin level seen in the myeloma-infiltrated bones. These observations suggest that myeloma-induced alloreactive T cells arising in myeloma-infiltrated bones are enriched with DP-T cells equipped with cytotoxic effector functions that are likely to be involved in the GVM effect.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Tumor Effect/immunology , Multiple Myeloma/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Cell Separation , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Homologous
2.
Cancer Cell ; 8(5): 407-19, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16286248

ABSTRACT

Bortezomib therapy has proven successful for the treatment of relapsed and/or refractory multiple myeloma (MM); however, prolonged treatment is associated with toxicity and development of drug resistance. Here, we show that the novel proteasome inhibitor NPI-0052 induces apoptosis in MM cells resistant to conventional and Bortezomib therapies. NPI-0052 is distinct from Bortezomib in its chemical structure, effects on proteasome activities, mechanisms of action, and toxicity profile against normal cells. Moreover, NPI-0052 is orally bioactive. In animal tumor model studies, NPI-0052 is well tolerated and prolongs survival, with significantly reduced tumor recurrence. Combining NPI-0052 and Bortezomib induces synergistic anti-MM activity. Our study therefore provides the rationale for clinical protocols evaluating NPI-0052, alone and together with Bortezomib, to improve patient outcome in MM.


Subject(s)
Boronic Acids/pharmacology , Lactones/pharmacology , Multiple Myeloma/drug therapy , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrroles/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Caspases/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Genes, bcl-2 , Humans , Lactones/administration & dosage , Lactones/chemistry , Lymphocytes/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Plasmacytoma/drug therapy , Proteasome Endopeptidase Complex/pharmacology , Pyrazines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/chemistry , Tumor Cells, Cultured
3.
Mol Cancer Ther ; 8(1): 26-35, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139110

ABSTRACT

Protein tyrosine kinases of the Janus kinase (JAK) family are associated with many cytokine receptors, which, on ligand binding, regulate important cellular functions such as proliferation, survival, and differentiation. In multiple myeloma, JAKs may be persistently activated due to a constant stimulation by interleukin (IL)-6, which is produced in the bone marrow environment. INCB20 is a synthetic molecule that potently inhibits all members of the JAK family with a 100- to 1,000-fold selectivity for JAKs over >70 other kinases. Treatment of multiple myeloma cell lines and patient tumor cells with INCB20 resulted in a significant and dose-dependent inhibition of spontaneous as well as IL-6-induced cell growth. Importantly, multiple myeloma cell growth was inhibited in the presence of bone marrow stromal cells. The IL-6 dependent cell line INA-6 was particularly sensitive to the drug (IC50<1 micromol/L). Growth suppression of INA-6 correlated with an increase in the percentage of apoptotic cells and inhibition of signal transducer and activator of transcription 3 phosphorylation. INCB20 also abrogated the protective effect of IL-6 against dexamethasone by blocking phosphorylation of SHP-2 and AKT. In contrast, AKT phosphorylation induced by insulin-like growth factor-I remained unchanged, showing selectivity of the compound. In a s.c. severe combined immunodeficient mouse model with INA-6, INCB20 significantly delayed INA-6 tumor growth. Our studies show that disruption of JAKs and downstream signaling pathways may both inhibit multiple myeloma cell growth and survival and overcome cytokine-mediated drug resistance, thereby providing the preclinical rationale for the use of JAK inhibitors as a novel therapeutic approach in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Janus Kinases/antagonists & inhibitors , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Dexamethasone/pharmacology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Multiple Myeloma/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Stromal Cells/drug effects , Stromal Cells/enzymology , Substrate Specificity , Survival Rate , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 14(21): 6955-62, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980990

ABSTRACT

PURPOSE: CD1d-restricted invariant natural killer T (iNKT) cells are important immunoregulatory cells in antitumor immune responses. However, the quantitative and qualitative defects of iNKT cells in advanced multiple myeloma hamper their antitumor effects. Therefore, the development of functional iNKT cells may provide a novel strategy for the immunotherapy in multiple myeloma. EXPERIMENTAL DESIGN: We activated and expanded iNKT cells from multiple myeloma patients with alpha-galactosylceramide (alpha-GalCer)-pulsed dendritic cells, characterized their antitumor effects by the cytokine production profile and cytotoxicity against multiple myeloma cells, and explored the effects of immunomodulatory drug lenalidomide on these iNKT cells. We also investigated the expression of CD1d by primary multiple myeloma cells and its function to activate iNKT cells. RESULTS: We established highly purified functional iNKT cell lines from newly diagnosed and advanced multiple myeloma patients. These CD1d-restricted iNKT cell lines produced high level of antitumor Th1 cytokine in response to alpha-GalCer-pulsed primary multiple myeloma cells, CD1d-transfected MM1S cell line, and dendritic cells. Moreover, iNKT cell lines displayed strong cytotoxicity against alpha-GalCer-pulsed primary multiple myeloma cells. Importantly, lenalidomide further augmented the Th1 polarization by iNKT cell lines via increased Th1 cytokine production and reduced Th2 cytokine production. We also showed that CD1d was expressed in primary multiple myeloma cells at mRNA and protein levels from the majority of multiple myeloma patients, but not in normal plasma cells and multiple myeloma cell lines, and CD1d(+) primary multiple myeloma cells presented antigens to activate iNKT cell lines. CONCLUSIONS: Taken together, our results provide the preclinical evidence for the iNKT cell-mediated immunotherapy and a rationale for their use in combination with lenalidomide in multiple myeloma treatment.


Subject(s)
Cell Line , Killer Cells, Natural , Multiple Myeloma/immunology , Thalidomide/analogs & derivatives , Antigens, CD1/immunology , Cytotoxicity, Immunologic , Humans , Immunotherapy/methods , Lenalidomide , Thalidomide/pharmacology
7.
Cancer Res ; 66(13): 6675-82, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16818641

ABSTRACT

Recent studies have underscored the role of B-cell-activating factor (BAFF), a member of the tumor necrosis factor superfamily, in promoting the survival of malignant B cells, including human multiple myeloma. We here characterized the functional significance of BAFF in the interaction between multiple myeloma and bone marrow stromal cells (BMSC) and further defined the molecular mechanisms regulating these processes. BAFF is detected on BMSCs derived from multiple myeloma patients as evidenced by flow cytometry. BAFF secretion is 3- to 10-fold higher in BMSCs than in multiple myeloma cells, and tumor cell adhesion to BMSCs augments BAFF secretion by 2- to 5-fold, confirmed by both ELISA and immunoblotting. Adhesion of MM1S and MCCAR multiple myeloma cell lines to KM104 BMSC line transfected with a luciferase reporter vector carrying the BAFF gene promoter (BAFF-LUC) significantly enhanced luciferase activity, suggesting that nuclear factor-kappaB (NF-kappaB) activation induced by multiple myeloma adhesion to BMSCs mediates BAFF up-regulation. Moreover, BAFF (0-100 ng/mL) increases adhesion of multiple myeloma lines to BMSCs in a dose-dependent manner; conversely, transmembrane activator and calcium modulator and cyclophylin ligand interactor-Ig or B-cell maturation antigen/Fc blocked BAFF stimulation. Using adenoviruses expressing dominant-negative and constitutively expressed AKT as well as NF-kappaB inhibitors, we further showed that BAFF-induced multiple myeloma cell adhesion is primarily mediated via activation of AKT and NF-kappaB signaling. Importantly, BAFF similarly increased adhesion of CD138-expressing patient multiple myeloma cells to BMSCs. These studies establish a role for BAFF in localization and survival of multiple myeloma cells in the bone marrow microenvironment and strongly support novel therapeutics, targeting the interaction between BAFF and its receptors in human multiple myeloma.


Subject(s)
Bone Marrow Neoplasms/pathology , Membrane Proteins/physiology , Multiple Myeloma/pathology , Tumor Necrosis Factor-alpha/physiology , B-Cell Activating Factor , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Neoplasms/metabolism , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-4/biosynthesis , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
Exp Hematol ; 35(7): 1038-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17588472

ABSTRACT

OBJECTIVES: Alkyl-lysophospholipids are a novel class of antitumor agents. Perifosine is a novel alkyl-lysophospholipid that can induce apoptosis in multiple myeloma (MM) tumor cells, both in vitro and in vivo. We investigated the effects of perifosine on the peripheral blood, bone marrow, and spleen of mice inoculated with subcutaneous plasmacytomas. METHODS: Immunocompromised mice were inoculated with myeloma cell lines and treated with oral perifosine in either a daily or weekly schedule, or with vehicle only. When plasmacytomas reached 2 cm, mice were sacrificed. Terminal blood was analyzed with a Coulter counter, and counts were confirmed by light microscopy. Marrow and spleen were also analyzed by light microscopy. RESULTS: In control mice, mean hemoglobin was 12 g/dL, white blood cell (WBC) count 7 x 10(9)/L, and mean platelet count was 292 x 10(9)/L. In contrast, the respective values for mice treated with perifosine weekly were 11 g/dL, 9 x 10(9)/L, and 944 x 10(9)/L; and for mice treated with perifosine daily were 10 g/dL, 11 x 10(9)/L, and 752 x 10(9)/L. The increase in WBCs was due, predominantly, to a neutrophilia. Compared to control mice, perifosine treatment induced marrow hypercellularity and splenic white pulp expansion. CONCLUSIONS: These findings have clinical relevance because myeloid suppression is a dose-limiting toxicity of many cytotoxic agents, and myeloid hyperplasia is usually only observed in the setting of growth factor stimulation. Coupled with its remarkable in vitro MM cytotoxicity, these results strongly support the use of perifosine in clinical trials for patients with MM.


Subject(s)
Multiple Myeloma/pathology , Myeloid Cells/drug effects , Phosphorylcholine/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Line , Female , Hematopoiesis/drug effects , Humans , Hyperplasia , Leukocytosis/chemically induced , Mice , Multiple Myeloma/drug therapy , Myeloid Cells/pathology , Phosphorylcholine/pharmacology , Thrombocytosis/chemically induced
9.
Mol Cancer Ther ; 6(6): 1718-27, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17575103

ABSTRACT

In this study, we investigated the cytotoxicity of 5-azacytidine, a DNA methyltransferase inhibitor, against multiple myeloma (MM) cells, and characterized DNA damage-related mechanisms of cell death. 5-Azacytidine showed significant cytotoxicity against both conventional therapy-sensitive and therapy-resistant MM cell lines, as well as multidrug-resistant patient-derived MM cells, with IC(50) of approximately 0.8-3 micromol/L. Conversely, 5-azacytidine was not cytotoxic to peripheral blood mononuclear cells or patient-derived bone marrow stromal cells (BMSC) at these doses. Importantly, 5-azacytidine overcame the survival and growth advantages conferred by exogenous interleukin-6 (IL-6), insulin-like growth factor-I (IGF-I), or by adherence of MM cells to BMSCs. 5-Azacytidine treatment induced DNA double-strand break (DSB) responses, as evidenced by H2AX, Chk2, and p53 phosphorylations, and apoptosis of MM cells. 5-Azacytidine-induced apoptosis was both caspase dependent and independent, with caspase 8 and caspase 9 cleavage; Mcl-1 cleavage; Bax, Puma, and Noxa up-regulation; as well as release of AIF and EndoG from the mitochondria. Finally, we show that 5-azacytidine-induced DNA DSB responses were mediated predominantly by ATR, and that doxorubicin, as well as bortezomib, synergistically enhanced 5-azacytidine-induced MM cell death. Taken together, these data provide the preclinical rationale for the clinical evaluation of 5-azacytidine, alone and in combination with doxorubicin and bortezomib, to improve patient outcome in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azacitidine/pharmacology , Boronic Acids/pharmacology , DNA Damage , DNA, Neoplasm/drug effects , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Multiple Myeloma/pathology , Pyrazines/pharmacology , Bortezomib , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans
10.
Cancer Res ; 65(13): 5898-906, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994968

ABSTRACT

Monoclonal antibodies (mAb) directed against lineage-specific B-cell antigens have provided clinical benefit for patients with hematologic malignancies, but to date no antibody-mediated immunotherapy is available for multiple myeloma. In the present study, we assessed the efficacy of a fully human anti-CD40 mAb CHIR-12.12 against human multiple myeloma cells. CHIR-12.12, generated in XenoMouse mice, binds to CD138-expressing multiple myeloma lines and freshly purified CD138-expressing cells from >80% multiple myeloma patients, as assessed by flow cytometry. Importantly, CHIR-12.12 abrogates CD40L-induced growth and survival of CD40-expressing patient multiple myeloma cells in the presence or absence of bone marrow stromal cells (BMSC), without altering constitutive multiple myeloma cell proliferation. Immunoblotting analysis specifically showed that PI3-K/AKT, nuclear factor-kappaB (NF-kappaB), and extracellular signal-regulated kinase activation induced by CD40L (5 mug/mL) was inhibited by CHIR-12.12 (5 mug/mL). Because CD40 activation induces multiple myeloma cell adhesion to both fibronectin and BMSCs, we next determined whether CHIR-12.12 inhibits this process. CHIR-12.12 decreased CD40L-induced multiple myeloma cell adhesion to fibronectin and BMSCs, whereas control human IgG1 did not. Adhesion of multiple myeloma cells to BMSCs induces interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) secretion, and treatment of multiple myeloma cells with CD40L further enhanced adhesion-induced cytokine secretion; conversely, CHIR-12.12 blocks CD40L-enhanced IL-6 and VEGF secretion in cocultures of multiple myeloma cells with BMSCs. Finally, CHIR-12.12 triggered lysis of multiple myeloma cells via antibody-dependent cellular cytotoxicity (ADCC) but did not induce ADCC against CD40-negative multiple myeloma cells, confirming specificity against CD40-expressing multiple myeloma cells. These results provide the preclinical rationale for clinical trials of CHIR-12.12 to improve patient outcome in multiple myeloma.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , Immunization, Passive/methods , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Antibody-Dependent Cell Cytotoxicity , Antigen-Antibody Reactions , CD40 Antigens/biosynthesis , CD40 Ligand/immunology , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Humans , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Multiple Myeloma/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteoglycans/immunology , Proteoglycans/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Syndecan-1 , Syndecans , Vascular Endothelial Growth Factor A/metabolism
11.
Cancer Res ; 65(24): 11712-20, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357183

ABSTRACT

SGN-40, a humanized immoglobulin G1 (IgG1) anti-CD40 monoclonal antibody, mediates cytotoxicity against human multiple myeloma (MM) cells via suppression of interleukin (IL)-6-induced proliferative and antiapoptotic effects as well as antibody-dependent cell-mediated cytotoxicity (ADCC). Here, we studied the clinical significance of an immunomodulatory drug lenalidomide on SGN-40-induced cytotoxicity against CD138(+)CD40(+) MM lines and patient MM cells. Pretreatment with lenalidomide sensitized MM cells to SGN-40-induced cell death. Combined lenalidomide and SGN-40 significantly induced MM apoptosis, evidenced by enhanced cleavage of caspase-3/8/poly(ADP-ribose)polymerase and increased sub-G(0) cells, compared with either single agent at the same doses. Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Importantly, pretreatment with lenalidomide or lenalidomide and SGN-40 markedly enhanced NK-cell-mediated lysis of autologous patient MM cells triggered by SGN-40. Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. In addition, lenalidomide induced the CD56(dim) NK subset, which are more potent mediators of ADCC against target MM cells than the CD56(bright) NK subset. Finally, pretreatment of both effector and target MM cells with lenalidomide markedly enhanced SGN-40-mediated ADCC against CD40-expressing MM cells. These studies, therefore, show that the addition of lenalidomide to SGN-40 enhances cytotoxicity against MM cells, providing the framework for combined lenalidomide and SGN-40 in a new treatment paradigm to both target MM cells directly and induce immune effectors against MM.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , CD40 Antigens/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Thalidomide/analogs & derivatives , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Humans , Immunization, Passive , Interleukin-6/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Multiple Myeloma/metabolism , Thalidomide/therapeutic use
12.
Cancer Res ; 65(16): 7478-84, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103102

ABSTRACT

The novel immunomodulator FTY720 down-modulates sphingosine-1-phosphate receptor 1 on lymphocytes at low nanomolar concentrations, thereby inhibiting sphingosine-1-phosphate receptor 1-dependent egress of lymphocytes from lymph nodes into efferent lymphatics and blood. At high micromolar concentration, FTY720 has been shown to induce growth inhibition and/or apoptosis in human cancer cells in vitro. In this study, we investigated the biological effects of FTY720 on multiple myeloma cells. We found that FTY720 induces potent cytotoxicity against drug-sensitive and drug-resistant multiple myeloma cell lines as well as freshly isolated tumor cells from multiple myeloma patients who do not respond to conventional agents. FTY720 triggers activation of caspase-8, -9, and -3, followed by poly(ADP-ribose) polymerase cleavage. Interestingly, FTY720 induces alterations in mitochondrial membrane potential (DeltaPsim) and Bax cleavage, followed by translocation of cytochrome c and Smac/Diablo from mitochondria to the cytosol. In combination treatment studies, both dexamethasone and anti-Fas antibodies augment anti-multiple myeloma activity induced by FTY720. Neither interleukin-6 nor insulin-like growth factor-I, which both induce multiple myeloma cell growth and abrogate dexamethasone-induced apoptosis, protect against FTY720-induced growth inhibition. Importantly, growth of multiple myeloma cells adherent to bone marrow stromal cells is also significantly inhibited by FTY720. Finally, it down-regulates interleukin-6-induced phosphorylation of Akt, signal transducers and activators of transcription 3, and p42/44 mitogen-activated protein kinase; insulin-like growth factor-I-triggered Akt phosphorylation; and tumor necrosis factor alpha-induced IkappaBalpha and nuclear factor-kappaB p65 phosphorylation. These results suggest that FTY720 overcomes drug resistance in multiple myeloma cells and provide the rationale for its clinical evaluation to improve patient outcome in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Multiple Myeloma/drug therapy , Propylene Glycols/pharmacology , Apoptosis/physiology , Bone Marrow Cells/cytology , Caspases/metabolism , Cell Growth Processes/physiology , Coculture Techniques , Drug Resistance, Neoplasm , Fingolimod Hydrochloride , Immunosuppressive Agents/pharmacology , Insulin-Like Growth Factor I/pharmacology , Interleukin-6/pharmacology , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Stromal Cells/cytology , bcl-2-Associated X Protein
13.
Clin Cancer Res ; 11(11): 4251-8, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930364

ABSTRACT

Interleukin-6 (IL-6) protects multiple myeloma cells against apoptosis induced by glucocorticoids. Here, we investigated whether inhibition of the IL-6 signaling pathway by the IL-6 receptor superantagonist Sant7 enhances the in vivo antitumor effects of dexamethasone on the IL-6-dependent multiple myeloma cell line INA-6. For this purpose, we used a novel murine model of human multiple myeloma in which IL-6-dependent INA-6 multiple myeloma cells were directly injected into human bone marrow implants in severe combined immunodeficient (SCID) mice (SCID-hu). The effect of in vivo drug treatments on multiple myeloma cell growth was monitored by serial determinations of serum levels of soluble IL-6 receptor (shuIL-6R), which is released by INA-6 cells and served as a marker of tumor growth. In SCID-hu mice engrafted with INA-6 cells, treatment with either Sant7 or dexamethasone alone did not induce significant reduction in serum shuIL-6R levels. In contrast, the combination of Sant7 with dexamethasone resulted in a synergistic reduction in serum shuIL-6R levels after 6 consecutive days of treatment. Gene expression profiling of INA-6 cells showed down-regulation of proliferation/maintenance and cell cycle control genes, as well as up-regulation of apoptotic genes in multiple myeloma cells triggered by Sant7 and dexamethasone combination. In vitro colony assays showed inhibition of myeloid and erythroid colonies from normal human CD34(+) progenitors in response to dexamethasone, whereas Sant7 neither inhibited colony growth nor potentiated the inhibitory effect of dexamethasone. Taken together, these results indicate that inhibition of IL-6 signaling by Sant7 significantly potentiates the therapeutic action of dexamethasone against multiple myeloma cells, providing the preclinical rationale for clinical trials of Sant7 in combination with dexamethasone to improve patient outcome in multiple myeloma.


Subject(s)
Dexamethasone/pharmacology , Interleukin-6/analogs & derivatives , Multiple Myeloma/drug therapy , Receptors, Interleukin-6/antagonists & inhibitors , Animals , Antigens, CD34/analysis , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/genetics , Cell Line, Tumor , Cluster Analysis , Colony-Forming Units Assay , Dexamethasone/therapeutic use , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Interleukin-6/pharmacology , Interleukin-6/therapeutic use , Male , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Receptors, Interleukin-6/metabolism
14.
Cancer Res ; 64(23): 8746-53, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15574786

ABSTRACT

Topoisomerase I inhibitors are effective anticancer therapies and have shown activity in hematologic malignancies. Here we show for the first time that SN38, the potent active metabolite of irinotecan, induces c-Jun NH(2)-terminal kinase activation, Fas up-regulation, and caspase 8-mediated apoptosis in multiple myeloma (MM) cells. Proteasomal degradation of nuclear topoisomerase I has been proposed as a resistance mechanism in solid malignancies. SN38-induced proteasomal degradation of topoisomerase I was observed during SN38-mediated cytotoxicity against MM.1S myeloma cell line but occurred after c-Jun NH(2)-terminal kinase activation, Fas up-regulation, and poly(ADP-ribose) polymerase cleavage and failed to protect cells from apoptosis. Differential toxicity was observed against MM cells versus bone marrow stromal cells, and SN38 inhibited adhesion-induced up-regulation of MM cell proliferation when MM cells adhere to bone marrow stromal cells. In addition, SN38 directly inhibited constitutive and inducible interleukin 6 and vascular endothelial growth factor secretion by bone marrow stromal cells. Synergy was observed when SN38 was used in combination with doxorubicin, bortezomib, as well as poly(ADP-ribose) polymerase inhibitor NU1025 and Fas-activator CH11. These findings have clinical significance, because identification of downstream apoptotic signaling after topoisomerase I inhibition will both elucidate mechanisms of resistance and optimize future combination chemotherapy against MM.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , DNA Topoisomerases, Type I/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex/metabolism , fas Receptor/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Boronic Acids/administration & dosage , Boronic Acids/pharmacology , Bortezomib , Cell Communication/drug effects , Cell Line, Tumor , DNA, Neoplasm/biosynthesis , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Enzyme Activation , Humans , Irinotecan , Lymphoma/drug therapy , Lymphoma/enzymology , Lymphoma/genetics , Lymphoma/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Pyrazines/administration & dosage , Pyrazines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Stromal Cells/drug effects , Stromal Cells/pathology , Up-Regulation/drug effects , fas Receptor/genetics , fas Receptor/immunology
15.
Cancer Res ; 64(8): 2846-52, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15087402

ABSTRACT

CD40 is expressed on B-cell malignancies, including human multiple myeloma (MM) and a variety of carcinomas. We examined the potential therapeutic utility of SGN-40, the humanized anti-CD40 monoclonal antibody, for treating human MM using MM cell lines and patient MM cells (CD138(++), CD40(+)). SGN-40 (0.01-100 micro g/ml) induces modest cytotoxicity in MM cell lines and patient MM cells. In the presence of de novo protein synthesis inhibitor cycloheximide, SGN-40 significantly induced apoptosis in Dexamethasone (Dex)-sensitive MM.1S and Dex-resistant MM.1R cells and in patient MM cells. SGN-40-mediated cytotoxicity is associated with up-regulation of cytotoxic ligands of the tumor necrosis factor family (Fas/FasL, tumor necrosis factor-related apoptosis-inducing ligand, and tumor necrosis factor alpha). SGN-40 treatment also induces a down-regulation of CD40 dependent on an endocytic pathway. Consequently, pretreatment of MM cells with SGN-40 blocked sCD40L-mediated phosphatidylinositol 3'-kinase/AKT and nuclear factor kappaB activation. Importantly, pretreatment of MM.1S and MM.1R cells with SGN-40 inhibited proliferation triggered by interleukin 6 (IL-6) but not by insulin-like growth factor-I. In addition, SGN-40 pretreatment of MM.1S cells blocked the ability of IL-6 to protect against Dex-induced inhibition of DNA synthesis. This was associated with a 2-4-fold reduction of IL-6 receptor at protein and mRNA levels in SGN-40-treated MM.1S cells and patient MM cells. Taken together, these results provide the preclinical rationale for the evaluation of SGN-40 as a potential new therapy to improve patient outcome in MM.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Protein Serine-Threonine Kinases , Antibodies, Monoclonal/immunology , CD40 Antigens/biosynthesis , CD40 Ligand/immunology , CD40 Ligand/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , I-kappa B Proteins/metabolism , Immunization, Passive/methods , Insulin-Like Growth Factor I/pharmacology , Interleukin-6/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/genetics
16.
Cancer Res ; 63(18): 5850-8, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522909

ABSTRACT

Insulin-like growth factor-1 (IGF-I) is a growth and survival factor in human multiple myeloma (MM) cells. Here we examine the effect of IGF-I on MM cell adhesion and migration, and define the role of beta1 integrin in these processes. IGF-I increases adhesion of MM.1S and OPM6 MM cells to fibronectin (FN) in a time- and dose-dependent manner, as a consequence of IGF-IR activation. Conversely, blocking anti-beta1 integrin monoclonal antibody, RGD peptide, and cytochalasin D inhibit IGF-I-induced cell adhesion to FN. IGF-I rapidly and transiently induces association of IGF-IR and beta1 integrin, with phosphorylation of IGF-IR, IRS-1, and p85(PI3-K). IGF-I also triggers phosphorylation of AKT and ERK significantly. Both IGF-IR and beta1 integrin colocalize to lipid rafts on the plasma membrane after IGF-I stimulation. In addition, IGF-I triggers polymerization of F-actin, induces phosphorylation of p125(FAK) and paxillin, and enhances beta1 integrin interaction with these focal adhesion proteins. Importantly, using pharmacological inhibitors of phosphatidylinositol 3'-kinase (PI3-K) (LY294002 and wortmannin) and extracellular signal-regulated kinase (PD98059), we demonstrate that IGF-I-induced MM cell adhesion to FN is achieved only when PI3-K/AKT is activated. IGF-I induces a 1.7-2.2 (MM.1S) and 2-2.5-fold (OPM6) increase in migration, whereas blocking anti-IGF-I and anti-beta1 integrin monoclonal antibodies, PI3-K inhibitors, as well as cytochalasin D abrogate IGF-I-induced MM cell transmigration. Finally, IGF-I induces adhesion of CD138+ patient MM cells. Therefore, these studies suggest a role for IGF-I in trafficking and localization of MM cells in the bone marrow microenvironment. Moreover, they define the functional association of IGF-IR and beta1 integrin in mediating MM cell homing, providing the preclinical rationale for novel treatment strategies targeting IGF-I/IGF-IR in MM.


Subject(s)
Insulin-Like Growth Factor I/physiology , Integrin beta1/physiology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Enzyme Activation , Fibronectins/metabolism , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Integrin beta1/metabolism , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Oligopeptides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteoglycans/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor Cross-Talk/physiology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/physiology , Signal Transduction/physiology , Syndecan-1 , Syndecans
17.
Cancer Res ; 62(17): 4996-5000, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12208752

ABSTRACT

The proteasome is a ubiquitous and essential intracellular enzyme that degrades many proteins regulating cell cycle, apoptosis, transcription, cell adhesion,angiogenesis, and antigen presentation. We have shown recently that the proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human myeloma cells in vitro. In this study, we examined the efficacy, toxicity, and in vivo mechanism of action of PS-341 using a human plasmacytoma xenograft mouse model. One hundred immunodeficient (beige-nude-xid) mice were used in two independent experiments. The mice were injected s.c. with 3 x 10(7) RPMI-8226 myeloma cells. When tumors became measurable (9.2 days; range, 6-13 days after tumor injection), mice were assigned to treatment groups receiving PS-341 0.05 mg/kg (n = 13), 0.1 mg/kg (n = 15), 0.5 mg/kg (n = 14), or 1.0 mg/kg (n = 14) twice weekly via tail vein, or to control groups (n = 13) receiving the vehicle only. Significant inhibition of tumor growth, even with some complete tumor regression, was observed in PS-341-treated mice. The median overall survival was also significantly prolonged compared with controls (30 and 34 days for high dose-treated mice versus 14 days for controls; P < 0.0001). PS-341 was well tolerated up to 0.5 mg/kg, but some mice treated at 1.0 mg/kg became moribund and lost weight. Analysis of tumors harvested from treated animals showed that PS-341 induced apoptosis and decreased angiogenesis in vivo. These studies therefore demonstrate that PS-341 has significant in vivo antimyeloma activity at doses that are well tolerated in a murine model, confirming our in vitro data and further supporting the early clinical promise of PS-341 to overcome drug resistance and improve patient outcome.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Boronic Acids/adverse effects , Bortezomib , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice , Multiple Myeloma/blood supply , Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Protease Inhibitors/adverse effects , Pyrazines/adverse effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Exp Hematol ; 30(7): 711-20, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12135668

ABSTRACT

OBJECTIVE: To evaluate the anti-tumor potential of beta-lapachone in multiple myeloma (MM) cell lines (U266, RPMI8226, and MM.1S); MM cell lines resistant to dexamethasone (MM.1R), melphalan (RPMI8226/LR5), doxorubicin (RPMI8226/DOX40), and mitoxantrone (RPMI8226/ MR20); and MM cells from patients (MM1-MM4). MATERIALS AND METHODS: Cytotoxicity of beta-lapachone was assessed by MTT and [3H]-thymidine uptake assays. Apoptosis was analyzed using propidium iodide staining, DNA fragmentation, TUNEL assay, caspase-9 colorimetric assay, and immunoblotting for caspase-3, poly (ADP-ribose) polymerase (PARP), and caspase-8 cleavage products. Paracrine growth of MM cells was assessed by [3H]-thymidine uptake in cultures of bone marrow stromal cells (BMSCs) and MM cells. Interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) secretion in the culture supernatants was measured by specific enzyme-linked immunosorbent assays (ELISAs). RESULTS: beta-lapachone showed significant cytotoxicity in MM cells (IC(50): 4-8 microM). In contrast, normal peripheral blood mononuclear cells (PBMCs) and BMSCs from MM patients were relatively resistant (IC(50): 8-16 microM). IL-6 did not protect against beta-lapachone-induced apoptosis in MM.1S cells, and dexamethasone showed additive cytotoxicity. beta-lapachone also decreased binding of MM.1S cells to BMSCs; abrogated IL-6 and VEGF secretion triggered by adhesion of BMSCs to MM.1S cells; reduced proliferation of MM.1S cells adherent to BMSCs; and decreased intracellular adhesion molecule-1 (ICAM-1) expression on MM.1S cells. Furthermore, beta-lapachone induced typical PARP cleavage, increased caspase-9 proteolytic activity, and activation of caspase-3, without activation of caspase-8 in U266 cells. CONCLUSION: These studies provide a framework for clinical evaluation of beta-lapachone to improve the outcome for patients with MM.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Multiple Myeloma/pathology , Naphthoquinones/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Caspases/metabolism , Cell Adhesion/drug effects , Cell Division/drug effects , Dexamethasone/pharmacology , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Enzyme Precursors/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/pharmacology , Leukocytes, Mononuclear/drug effects , Melphalan/pharmacology , Mitoxantrone/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Paracrine Communication , Poly(ADP-ribose) Polymerases/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Tumor Cells, Cultured/drug effects
19.
Hematol J ; 5(1): 9-23, 2004.
Article in English | MEDLINE | ID: mdl-14745425

ABSTRACT

Multiple myeloma (MM) is an incurable hematological malignancy with an average survival of 3 years with conventional therapy. Allogeneic hematopoietic cell transplantation (allo-HCT) may cure some patients, but has been associated with a very high transplantation-related mortality (TRM) of over 40%.(1) In contrast to allo-HCT, autologous hematopoietic cell transplantation (AHCT) has been much safer, with a TRM <3% in the 1990s. Therefore, in the last 15 years AHCT has become a common procedure for MM patients. The widespread use of AHCT has been associated with a median survival of 55-72 months,(2,3,4,5,6) and two large randomized trials have shown that AHCT is superior to conventional chemotherapy for the treatment of MM.(3,7) Approaches to improve the outcome of stem cell transplantation for MM patients include consideration of patient status, efficacy and toxicity of induction therapy, source of hematopoietic rescue, conditioning regimens, and maintenance therapy. Recent attempts to improve outcome include tandem AHCT, AHCT followed by RIC (reduced intensity conditioning) allo-HCT, and allo-HCT with T-cell depletion and subsequent donor-lymphocyte infusions (DLI), while novel therapies and improved supportive care may improve the overall survival (OS) of all MM patients with or without transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Kidney Diseases/chemically induced , Kidney Diseases/etiology , Multiple Myeloma/complications , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome
20.
J Hematol Oncol ; 6: 24, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23547827

ABSTRACT

BACKGROUND: Inosine triphosphate pyrophosphohydrolase (ITPase) is a 'house-cleaning' enzyme that degrades non-canonical ('rogue') nucleotides. Complete deficiency is fatal in knockout mice, but a mutant polymorphism resulting in low enzyme activity with an accumulation of ITP and other non-canonical nucleotides, appears benign in humans. We hypothesised that reduced ITPase activity may cause acquired mitochondrial DNA (mtDNA) defects. Furthermore, we investigated whether accumulating mtDNA defects may then be a risk factor for cell transformation, in adult haematological malignancy (AHM). METHODS: DNA was extracted from peripheral blood and bone marrow samples. Microarray-based sequencing of mtDNA was performed on 13 AHM patients confirmed as carrying the ITPA 94C>A mutation causing low ITPase activity, and 4 AHM patients with wildtype ITPA. The frequencies of ITPA 94C>A and IVS2+21A>C polymorphisms were studied from 85 available AHM patients. RESULTS: ITPA 94C>A was associated with a significant increase in total heteroplasmic/homoplasmic mtDNA mutations (p<0.009) compared with wildtype ITPA, following exclusion of haplogroup variants. This suggested that low ITPase activity may induce mitochondrial abnormalities. Compared to the normal population, frequencies for the 94C>A and IVS2+21A>C mutant alleles among the AHM patients were higher for myelodyplastic syndrome (MDS) - but below significance; were approximately equivalent for chronic lymphoblastic leukemia; and were lower for acute myeloid leukemia. CONCLUSIONS: This study invokes a new paradigm for the evolution of MDS, where nucleotide imbalances produced by defects in 'house-cleaning' genes may induce mitochondrial dysfunction, compromising cell integrity. It supports recent studies which point towards an important role for ITPase in cellular surveillance of rogue nucleotides.


Subject(s)
DNA, Mitochondrial/genetics , Hematologic Neoplasms/enzymology , Hematologic Neoplasms/genetics , Pyrophosphatases/genetics , Adult , Aged , Aged, 80 and over , Animals , Humans , Mice , Mice, Knockout , Microarray Analysis , Middle Aged , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL