Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 5150-5150
    Abstract: SGN-40, a humanized immoglobulin G1 (IgG1) anti-CD40 monoclonal antibody, mediates cytotoxicity against human multiple myeloma (MM) cells via suppression of IL-6-induced proliferative and antiapoptotic effects, as well as antibody-dependent cell-mediated cytotoxicity (ADCC). We here 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/PARP and increased subG0 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− 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 by SGN-40. Lenalidomide also upregulated CD40L on CD56+CD3− NK cells, facilitating IL-2-mediated activation of NK cells. In addition, lenalidomide induced the CD56dim NK subset, which are more potent mediators of ADCC against target MM cells than the CD56bright 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 demonstrate 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.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 5104-5104
    Abstract: Invariant NKT (iNKT) cells are important immunoregulatory cells that recognize glycolipid antigens with CD1d restriction and contribute to antitumor immune responses through the production of IFN-γ and IL-2. However, in progressive multiple myeloma (MM), the iNKT cell population is decreased along with its capacity to produce IFN-γ. Thus, a novel strategy for the immunotherapy of MM entails the enhancement of iNKT cell functions. In this study, we established iNKT cell lines from MM patients via enrichment with Vα24+ and subsequently with Vβ11+ cells, followed by several rounds of stimulation with α-GalCer-pulsed DCs. These techniques resulted in highly purified iNKT cell lines ( & gt;97%). To evaluate potential in vivo interaction between iNKT cells and myeloma cells, we evaluated the CD1d expression on primary myeloma cells as well as MM cell lines. Gene expression profiling revealed compared to normal plasma cells, majority of primary MM cells (11 out of 15) expressed higher levels of CD1d; in contrast, all 6 MM cell lines tested had no expression. Flow cytometric analysis further confirmed the expression of CD1d on primary MM cells and lack of its expression on 12 different MM cell lines. A CD1d-transfected MM1S cell line (MM1S-CD1d) was therefore established for the functional study. To determine whether CD1d-expressing primary MM cells have the antigen presenting capacity, iNKT cell lines from healthy donors (n=2) and MM patients (n=2) were cocultured with 5 cases of CD1d positive primary MM cells with or without α-GalCer. Monitored by the CD25 expression, we demonstrated primary MM cells presented α-GalCer and also endogenous antigen(s) to activate iNKT cells. We have further evaluated the functional profile of expanded iNKT cell lines from MM patients (n=5). Upon stimulation with α-GalCer-pulsed MM.1S-CD1d cells, iNKT cells produced high levels of Th1-type cytokines (IFN-γ and IL-2) compared to low level Th2-type cytokine production (IL-4). Our results thus demonstrate that iNKT cell lines from MM patients were functionally restored by expansion with α-GalCer-pulsed DCs in vitro. To further augment iNKT cells function, we evaluated effects of lenalidomide on iNKT cell lines, an immunomodulatory drug which has been demonstrated to enhance T cell costimulation and NK cell activity. Lenalidomide did not directly stimulate iNKT cells in the presence or absence of α-GalCel. Importantly, upon CD1d-restricted activation by α-GalCer-loaded MM1S-CD1d cells, lenalidomide significantly enhanced the Th1-type immune responses of iNKT cell lines from both healthy donors and MM patients. Compared to those of controls, a significant increase of IFN- γ (healthy donor, p & lt; 0.001, n=7; MM patients, p & lt;0.05, n=3) and IL-2 (MM patients, p & lt;0.0015, n=3) occurred. Meanwhile, lenalidomide had no significant effect on the production of IL-4 by iNKT cell lines (healthy donor, p & gt;0.05, n=7; MM patients, p & gt;0.05, n=3). Taken together, our results provide preclinical feasibility and support a rationale to evaluate efficacy of adoptive transfer of iNKT cells in MM. Moreover, it provides a clinical basis for use of lenalidomide to enhance iNKT cell mediated immunotherapy in myeloma.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 655-655
    Abstract: Monoclonal gammopathy of undetermined significance (MGUS) is an indolent condition that may be modulated by various factors including immunologic responses directed at the monoclonal cells. Several evidences have supported the idea that the immune system, in patients with MGUS, may play a role in controlling the progression to myeloma (MM) and the identification of antigenic targets could open the way for future immunotherapeutic approaches to delay or prevent such progression. We have screened a cDNA expression library from primary myeloma cells and used Serological Analysis of Recombinant cDNA Expression Library (SEREX) to identify antigens that are recognized by antibodies in MGUS patients and therefore may be targets of the immune system and possibly involved in the pathogenesis of this disease. We used high dilution (1:500) serum from 3 MGUS patients with stable disease for 1 to 4 years and identified a panel of 11 novel antigenic targets eliciting an immune response. Antibody response appeared to be directed against intracellular proteins involved in apoptosis (SON, Hip1), DNA and RNA binding proteins (KIAA0530, GPATC4), signal transduction regulators (AKAP11), developmental proteins (OFD1), transcriptional co-repressors (IRF2BP2), proteins of the ubiquitin-proteosome pathways (PSMC1). We have further analyzed frequency of antibody response against these antigens in additional 26 MGUS sera, 10 newly diagnosed and 10 MM patients in remission after auto-transplant and 25 normal donors. We have observed antibody response against OFD1 (20.6 %); KIAA0530 (10.3%); AKAP11 (10.3%); and GPATC4 (6.8%) in patients with MGUS. Interestingly, 1/10 patients with newly diagnosed MM (10%) and 3/10 (30%) patients in remission after auto-transplant had an antibody response against OFD1 with evidence of increase in antibody titer in one patient after transplant suggesting its importance as a target. No significant antibody responses were observed against any of these antigens in the sera of 25 health donors. We have further focused our studies on OFD1, a protein developmentally expressed in adult human organs that colocalizes with γ-tubulin in the centrosome and has LIS1 homology motifs suggesting its contribution in regulation of microtubule dynamics. We have confirmed, by western blot analysis and RT-PCR, the expression of OFD1 in MM cell lines and lack of its expression in normal cells including normal BMSC. In addition some myeloma cell lines express different spliced variant of this protein. Specific T cell responses directed at OFD1 and its role in cell signaling are under investigation. These data open the possibility to identify target antigens that are important in the disease process of MGUS and may allow us to design future vaccines and immunotherapeutic approaches targeting these antigens in MGUS as well as in MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 609-609
    Abstract: Abstract 609 HM1.24/CD317 or BST2, a cell surface protein highly expressed on malignant plasma cells, represents a potential target of immunotherapy for multiple myeloma (MM). Here we characterized XmAb®5592, a novel Fc-engineered and humanized anti-HM1.24 antibody (Ab), and studied mechanisms of its anti-MM activity. XmAb®5592, with double amino acid substitution in Fc region of the wild type IgG1, has approximately 40-fold and 10-fold increases in affinity for Fc gamma receptor III (FcγRIIIa) and (FcγRIIa), respectively, expressed on effector cells including NK cells. The Fv region of XmAb®5592 was humanized and engineered to achieve high affinity and specificity of binding to HM1.24-expressing target cells. XmAb®5592 reacts against a panel of MM cell lines (n=19) which are both sensitive or resistant to current anti-MM conventional and novel therapies. Importantly, it triggers 10-100-fold higher antibody-dependent cell-mediated cytotoxicity (ADCC) against these MM cell lines than a native/non Fc-engineered version (anti-HM1.24 IgG1) of the Ab. Specifically, the maximum specific lysis of MM1S, MM1R, and RPMI8226 target cells induced by XmAb®5592 is at a concentration of 0.001-0.01 μg/ml, whereas the IgG1 analog did not induce maximum cell lysis until 0.1 μg/ml. The maximum 100% specific lysis of INA-6 target cells occurred at 0.1 μg/ml of XmAb5592, in contrast to 60% maximum lysis induced by10 μg/ml of the IgG1 analog. Since the bone marrow (BM) microenvironment induces resistance in MM cells to conventional therapies, we next asked whether XmAb®5592 induced ADCC against MM cells even in the presence of BM stromal cells (BMSCs). Importantly, XmAb®5592 triggered significant ADCC against MM1S, MM1R, and INA-6 MM cells in the context of BMSCs. XmAb5592 also reacts against patient MM cells, and triggers robust ADCC against CD138-purified patient MM cells in assays using NK effector cells from normal donors. Furthermore, cross-linked XmAb5592 inhibited RMPI 8226 cell growth in the absence of effector cells. The in vivo efficacy of XmAb®5592 was next evaluated in murine subcutaneous (sc) xenograft murine models using RPMI 8226 cells. Administration of XmAb5592 (9mg/kg, ip, 2x/week for 4 weeks) led to a significant reduction in growth of established tumors in vivo compared to a non-engineered IgG1 anti-HM1.24 analog. At termination of the study. 7/15 mice were tumor free in the XmAb- treated group versus only 1/15 tumor free mice in the IgG1 analog treated group. An anti-HM1.24 antibody with Fc region engineered to completely ablate binding to FcγRs (knock-out) behaved equivalent to the PBS vehicle control in these studies, again underlining the significance of interaction with FcγR for anti-tumor efficacy. These results therefore suggest that XmAb5592, an anti-HM1.24 antibody engineered for improved effector function and antitumor potency in vitro and in vivo, is a promising next-generation immunotherapeutic for MM. Disclosures: Muchhal: Xencor: Employment. Horton:Xencor: Employment. Nguyen:Xencor: Employment. Karki:Xencor: Employment. Desjarlais:Xencor: Employment. Munshi:Millennium Pharmaceuticals: Honoraria, Speakers Bureau. Richardson:Keryx Biopharmaceuticals: Honoraria. Anderson:Millennium Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3485-3485
    Abstract: Osteolytic bone disease in Multiple Myeloma (MM) is caused by enhanced osteoclast (OCL) activation and inhibition of osteoblast function. The proteasome inhibitor bortezomib (PS341, Velcade) has potent anti-myeloma activity with impressive clinical responses. A recent study indicated that bortezomib has inhibitory effects on OCL (ASH 2005, Abstract #2488). Lenalidomide (CC-5013, Revlimid) is an immunomodulatory derivative of thalidomide that has shown promising anti-MM effects in patients with relapsed or refractory MM (Richardson et. al, Blood Jul 06). Significantly, a phase I clinical trial showed that lenalidomide and bortezomib could achieve responses in the majority of patients with MM, refractory to either agent alone (ASH 2005, Abstract #365). However, the effect of lenalidomide on human OCL lineage is unknown. Here we investigated the effect of lenalidomide and bortezomib on human OCL. Peripheral blood mononuclear cells (PBMC) from MM patients (n=11) and healthy donors (n=5) were stimulated with receptor activator of NFk-B ligand (RANKL) (50ng/ml) and M-CSF (25ng/ml) for two weeks to induce OCL formation, in the presence or absence of lenalidomide or bortezomib. OCL were identified by flow cytometric analysis using anti-aVb3 integrin. Lenalidomide and bortezomib inhibited OCL differentiation in a dose-dependent manner (n=13, median control: 70.9% at 0 μM; 63% at lenalidomide 2μM and 45% at 10μM; 35% at bortezomib 2nM and 11% at 5nM). TRAP staining (tartrate-resistant acid phosphatase) was performed to identify OCL and confirm OCL activity. Lenalidomide, as well as bortezomib inhibited OCL in a dose-dependent manner, as evidenced by a marked decrease in TRAP+ cells. To assess bone resorption activity, OCL were cultured with dentine discs, in the presence or absence of lenalidomide and bortezomib, followed by light microscopic analysis and additional measurement of soluble collagen I fragments from the supernatant. Both lenalidomide and bortezomib inhibited bone resorption in a dose-dependent manner. We next asked whether mature OCL were affected. OCL were induced by cytokine stimulation for 3 weeks and treated for 72h, followed by flow cytometry. Neither lenalidomide nor bortezomib altered total number of aVb3 integrin-expressing mature OCL (n=6). In addition, OCL culture supernatants were collected, and two major MM growth and survival factors produced by OCL, B-cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL), were measured by specific ELISA. Both lenalidomide and bortezomib strongly inhibited secretion of BAFF and APRIL. Finally, we determined whether lenalidomide and bortezomib inhibited expression of transcriptional factors important for OCL differentiation and survival. Cell lysates of CD14-expressing monocytic cells from normal donor PBMCs were subject to immunoblotting. Importantly, lenalidomide inhibited OCL differentiation by downregulation of PU.1 expression. These results therefore indicate, that lenalidomide and bortezomib inhibit OCL differentiation, thereby directly preventing the development of new osteolytic lesions. Moreover, BAFF and APRIL secretion by OCL is downregulated, thereby inhibiting MM cell survival in the bone marrow microenvironment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1963-1963
    Abstract: Abstract 1963 Poster Board I-986 Background: Multiple Myeloma (MM), a cancer of plasma cells is characterized by frequent chromosomal alterations. Deletion of chromosome 13, especially band 13q14, is commonly observed in early stages of MM, suggesting the presence of tumor suppressor genes within this region. When studied in the context of CLL, the miR 15a and 16-1 cluster was associated with a distinct miR signature and clinical outcome. Over-expression of miR16 caused induction of apoptosis and downregulation of the anti apoptotic gene BCL2 in a megakaryocytic leukemia cell line and induced growth arrest in MM cells. Nonetheless, being lost in CLL, MM, MCL and LPL, their functional role has not been studied using a loss-of-function approach in any of these lymphoid malignancies. Here, we describe the generation of an in vivo system for the long term, stable knockdown of miR 15a/ 16-1 expression in myeloma cells to recapitulate the conditions seen in chromosome 13q14 deleted MM. Methods: Using lentiviral vectors to stably express a competitive sponge miR16 inhibitor we set up a system to functionally validate the role of microRNA 15a/16-1 cluster. Pure populations of lentivirally transduced MM cell lines were sorted by flow cytometry using GFP marker. Decreased miRs 15a and 16 expression levels were assessed by Northern blot and R-luciferase reporter system. Cell growth rate was measured using trypan blue counting, and thymidine incorporation. Cell cycle analysis was measured by flow cytometry after staining with PI. Intracellular signal modulation was demonstrated by Western blotting. RNA from MM cell lines expressing the control sponge or sponge16 were hybridized on an Affymetrix U133A 2.0 array chip, and validated using quantitative real time PCR. Xenograft murine models were performed using the stable MM cell lines injected into 6-week old NOD.CB17-PrkdcSCID/J irradiated mice. Results: Selected stable miR knockdown MM cell lines exhibited significantly reduced expression of miRs15a/16-1 as assessd by both by mRNA levels and miR luciferase reporter assays. The knockdown cells showed a significant increase in growth rates (1.5-2 fold) compared to control cells, as measured by viable cell counts and proliferation by thymidine incorporation in vitro. Importantly, miR16 inhibition decreased animal survival in a xenograft model of MM by increasing tumor load, invasiveness and host angiogenesis. To further delineate the role of miR15a/16 in MM and to gain additional insight into the possible target genes regulated by this cluster, we performed gene expression-profiling analysis in controls and miR16 deficient MMS1 and RPMI cell lines. Since our sponge system produces downregulation of the miRs, we focused on the upregulated probes. Expression profiling analysis of miR16 deficient cells identified a surprisingly large number of downstream target-genes such as FGFR1, PI3KCa, MDM4, VEGFa, as well as secondary affected genes such as JUN and Jag1. These results were verified both at the mRNA level and the protein level, as well as in other MM cell lines. Moreover, we were able to show that these knockdown cells were partially addicted to some of these pathways using specific drug inhibitors. We further validated designated genes as direct miR16 targets by showing binding sites within the conserved 3' UTR and also within the mRNA coding region, thus indicating that the miRs may have many more possible targets than anticipated by conventional prediction methods. Conclusions: Using this loss-of-function system, which mimics the pleiotropic chronic effects of microRNA loss at the 13q chromosomal deletion, provides a valuable tool to investigate their function as tumor suppressor genes in MM pathogenesis, affecting multiple targets, and may represent a novel potential for therapeutic targeting in MM and other lymphoid malignancies. Disclosures: Munshi: Seattle Genetics, Inc.: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3591-3591
    Abstract: Osteonecrosis of the jaw (ONJ) is a recently described entity observed in patients with a history of aminobisphosphonate use. To date nearly 400 patients with ONJ have been reported in literature and this number continues to rise. However,prospective characterization of ONJ is lacking. We here characterize ONJ clinically and radiographically using multiple imaging modalities including panorex films, CAT scans, magnetic resonance imaging (MRI), FDG-PET scans, and NaF-PET bone scans in 11 patients with multiple myeloma (MM). Moreover, bone turnover and remodelling markers in these patients were analyzed prospectively in order to gain insights into the pathophysiology of ONJ. Eleven patients between the ages of 57 and 81 yrs were included. There were 8 men and 3 women, and 6 patients presented with Durie Salmon stage III disease. Patients were treated with various combinations of conventional and novel agents, including stem cell transplantation (4/11). Patients received either pamidronate (n=3), zolendronic acid (n=4), or both agents sequentially (n=4). The mean duration of bisphosphonate (BP) therapy was 38.7 months (range: 9–81 months). All patients were examined independently by 2 oral medicine physicians, and clinical data was validated on 2 separate dental visits. Six of 11 patients had mandibular lesions, 3 had lesions in the maxilla, and 2/11 patients had lesions in both the maxilla and mandible. Plain and panorex filmsdemonstrated a mottled appearance with increased radiolucency at the site of ONJ.This was associated with an increase in both glucose metabolism and mineralization at sites of ONJ, as measured with the maximum standardized uptake value (SUVmax) on FDG and NaF-PET scans, respectively. However, one patient with increased uptake on NaF-PET did not have increased glucose metabolism with FDG-PET. The target-to-background ratio of SUVmax for NaF-PET scans was significantly greater than FDG-PET suggesting that NaF-PET may have greater sensitivity than FDG-PET in confirming a diagnosis of ONJ. Several markers of bone turnover and remodelling were measured including serum calcium, vitamin D (25-OH), urinary N telopeptides, bone alkaline phosphatase (BAP), osteopontin, MIP 1a, RANK-L, osteoprotegrin, and DKK. Transcriptional profiling on peripheral blood mononuclear cells (PBMCs) using the Affymetrix U133Plus 2.0 gene chip was also performed in all 11 patients and compared with those of 10 MM patients on BP therapy without ONJ and 5 normal donors. These correlative studies will be presented and provide insights into the pathophysiology of ONJ. Importantly these studies both define clinical and radiographic features on ONJ, but also identify biomarkers to be evaluated in future prospective studies of BP therapy and ONJ.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3468-3468
    Abstract: Multiple Myeloma (MM) remains an incurable plasma cell neoplasia, despite recent additions in the therapeutic arsenal for its management. Aurora kinases play integral roles in the orchestration of chromosomes and cytoskeletal mobility during the process of cell division. Aurora kinase activity has been implicated in several tumor types, including ovarian, colon, and prostate cancers. To determine whether inhibition of Aurora kinase activity could attenuate myeloma cell survival, we performed studies of the Aurora kinase inhibitor VE465 (Vertex Pharmaceuticals / Merck & Co., Inc.). VE465 inhibits all 3 Aurora isoforms (Aur A, B and C) with approximate Ki values of 1, 26, and 8.7 nM respectively. MTT colormetric survival assays (72–96hrs exposure) showed that VE465 is active against a wide panel of human MM cell lines: 26 of 38 MM cell lines had IC50 values at or 〈 100 nM, which are significantly lower than IC50 values for normal hematopoietic cells, e.g. unstimulated or PHA-stimatuled PBMCs. Importantly, VE465 was active in vitro against MM cell lines and/or primary MM tumor cells resistant to various anti-MM therapeutics, including dexamethasone, alkylating agents, anthracyclines, the proteasome inhibitor bortezomib, and/or immunomodulatory thalidomide derivatives (IMiDs). Moreover, VE465 maintained its activity despite the presence of protective bone marrow-derived cytokines (e.g. IL-6). PI cell cycle analyses showed that VE465 causes (even within 8 hrs of treatment) caused pronounced G2 arrest, followed by significant shift of MM cells to sub-G1 gate, consistent with cell death. Immunoblotting analyses confirmed that VE465 treatment induces cleavage of PARP, as well as cleavage of caspases-8 and -9, without significant changes in the expression levels of several key molecular effectors (e.g. Mcl-1, Bax, p53, hsp70, hsp90, hs27) which have been previously implicated in the mechanism of anti-MM activity of diverse other therapeutics. Screening of VE465-based combination regimens with other anti-MM agents showed additive effects of VE465 with the histone deacetylase inhibitor Vorinostat (SAHA) (Merck & Co., Inc). Ongoing studies in our Center are addressing the identification of specific molecular markers correlating with the degree of sensitivity of MM cells to VE465. Our in vitro evidence for induction of MM cell death and therapeutic window for the anti-MM effect of VE465, its ability to overcome protective effect of BM-derived cytokines, and the clearly distinct pattern of molecular sequelae of VE465 compared to several other agents in our current anti-MM therapeutic armamentarium, all suggest that Aurora kinase inhibition represents an intriguing novel targeted treatment strategy in MM. Importantly, these studies, particularly the identification of a sizeable subset of MM cell lines with higher sensitivity to VE465 than normal cells, provide the framework for in vivo VE465 studies in progress, alone and in combination with other anti-MM agents, to inform the design of potential clinical trials of this class of agents for MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3409-3409
    Abstract: Interaction between multiple myeloma (MM) cells and the bone marrow (BM) microenvironment plays a critical role in promoting MM cell growth, survival, migration and development of drug resistance. This interaction within the bone marrow milieu is unique and its understanding is important in evaluating effects of novel agents in vitro and in vivo. We here describe a novel murine model that allows us to study the expression changes in vivo in MM cells within the human BM milieu. In this model, the green fluorescent protein (INA-6 GFP+) transduced IL-6-dependent human MM cell line, INA-6, was injected in human bone chip implanted into SCID mice. At different time points the bone chip was retrieved, cells flushed out and GFP+ MM cells were purified by CD138 MACS microbeads. Similar isolation process was used on INA-6 GFP+ cells cultured in vitro and used as control. Total RNA was isolated from these cells and gene expression profile analyzed using the HG-U133 array chip (Affymetrix) and DChip analyzer program. We have identified significant changes in expression of several genes following in vivo interaction between INA-6 and the BM microenvironment. Specifically, we observed up-regulation of genes associated with cytokines (IL-4, IL-8, IGFB 2–5) and chemokines (CCL2, 5, 6, 18, 24, CCR1, 2, 4), implicated in cell-cell signalling. Moreover genes implicated in DNA transcription (V-Fos, V-Jun, V-kit), adhesion (Integrin alpha 2b, 7, cadherin 1 and 11) and cell growth (CDC14, Cyclin G2, ADRA1A) were also up-regulated and genes involved in apoptosis and cell death (p-57, BCL2, TNF1a) were down-regulated. Using the Ingenuity Pathway Analysis the most relevant pathways modulated by the in vivo interaction between MM cells and BMSCs were IL-6, IGF1, TGF-beta and ERK/MAPK-mediated pathways as well as cell-cycle regulation and chemokine signalling. These results are consistent with previously observed in vitro cell signalling studies. Taken together these results highlight the ability of BM microenvironment to modulate the gene expression profile of the MM cells and our ability to in vivo monitor the changes. This model thus provides us with an ability to study in vivo effects of novel agents on expression profile of MM cells in BM milieu, to pre-clinically characterize their activity.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3452-3452
    Abstract: We previously identified a role of B-cell activating factor (BAFF), a member of the tumor necrosis factor superfamily, in localization and survival of MM cells in the BM microenvironment (Cancer Res2006, 66:6675–82). In the present study, we examined the potential therapeutic utility of the BAFF inhibitor, AMG523, for treating human MM using MM lines, either sensitive or resistant to conventional chemotherapy, as well as freshly isolated patient MM cells, in the presence or absence of bone marrow stromal cells (BMSCs). AMG523 induces modest cytotoxicity in MM cell lines and patient MM cells, suggesting a minor role of autocrine mechanism of BAFF for MM growth and survival. In the presence of BMSCs, AMG523 significantly decreased growth and survival in dexamethasone (Dex)-sensitive MM1S, Dex-resistant MM1R, INA6 MM cells and in patient MM cells (n=7), in a dose-dependent manner (0.1–10 μg/ml). BAFF-augmented MM adhesion to BMSCs is also blocked by AMG523 at 0.1 mg/ml in MM lines (MM1S, 28PE, INA6), as well as in freshly isolated patient MM cells (n=4). BAFF protects MM cells against dex- and lenalidomide-induced cytotoxicity; conversely, AMG523 blocks BAFF-induced protection against drug-induced apoptosis. Importantly, pretreatment of AMG523 blocks BAFF-induced activation of AKT, nuclear factor kB, and ERK in MM cells, confirming its inhibitory effect on BAFF-mediated adhesion and survival. We next asked whether AMG523 enhances Dex-, bortezomib-, Lenalidomide-induced MM cell cytotoxicity. AMG523 augments the inhibitory effect of Dex and lenalidomide in patient MM cells in the presence of BMSCs. Since osteoclasts (OCLs) secrete BAFF in the bone marrow microenvironment, we further asked whether AMG523 inhibits protection by MM-OCL interaction. OCLs were derived from peripheral blood mononuclear cells from MM patients after 2-week culture with M-CSF and RANKL, and MM cells were added in the presence or absence of AMG523. OCLs significantly increased MM cell survival, evidenced by annexin V and PI staining followed by flow cytometric analysis; conversely, AMG523 blocked MM cell survival by coculture with OCLs. Taken together, our data demonstrate that the novel therapeutic AMG523 blocks the interaction between BAFF and its receptors in human MM, thereby providing the rationale for clinical trials of AMG523 to improve patient outcome in MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages