Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 21, No. 1 ( 2021-12)
    Abstract: Increased bone turnover is frequently observed in advanced cancer and predominantly related to bone metastases or therapy. Cachexia represents an important cause of morbidity and mortality in cancer patients. Key features are weight loss, muscle wasting and chronic inflammation, which induce profound metabolic changes in several organs, including the bone. However, whether cachexia contributes to abnormal bone metabolism in cancer patients is unknown. Aim of the present study was to determine the potential correlation of bone turnover markers with body composition and laboratory parameters in treatment-naïve cancer patients. Methods In this cross-sectional study we measured the levels of carboxy terminal telopeptide of collagen (CTX), an indicator of bone resorption, as well as osteocalcin (Ocn) and procollagen type I N-terminal propeptide (PINP), indicators of bone formation, in 52 cancer patients and correlated with body composition and laboratory parameters. Univariate and multivariate logistic analysis were performed to identify determinants of negative bone remodeling balance, estimated by CTX/Ocn and CTX/PINP ratio. Results Based on weight loss, body mass index and muscle mass, patients were divided into a cachectic (59.6%) and a control (40.4%) group. After correcting for the presence of bone metastases, our results showed a significant upregulation of CTX in cachectic patients compared to non-cachectic cancer patients (median 0.38 vs 0.27 ng/mL, p   〈  0.05), with no difference in Ocn and PINP levels (mean 14 vs. 16 ng/ml, p  = 0.2 and median 32 vs. 26 μg/L, p  = 0.5, respectively). In addition, the CTX/Ocn and the CTX/PINP ratio were indicative of bone resorption in 68% and 60% of cachexia patients, respectively (vs. 20% and 31% in the control group, p  = 0.002 and p  = 0.06). The main determinants of the unbalanced bone turnover were hypoalbuminemia for the CTX/Ocn ratio (OR 19.8, p  〈  0.01) and high CRP for the CTX/PINP ratio (OR 5.3, p  〈  0.01) in the multivariate regression analysis. Conclusions CTX is substantially higher in cachectic patients compared to non-cachectic oncological patients and hypoalbuminemia as well as elevated CRP concentrations are independent predictors of a negative bone remodeling balance in cancer patients. These results strongly indicate that cachexia correlates with exacerbated bone turnover in cancer.
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2041352-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancers, MDPI AG, Vol. 13, No. 17 ( 2021-08-26), p. 4312-
    Abstract: Vaccination is the primary public health strategy to cope with the COVID-19 pandemic. Although solid tumor and hematologic patients are at higher risk of serious COVID-19-related complications, data on immune responses to COVID-19 vaccines in this patient cohort are particularly scarce. The present study, therefore, aimed at the standardized determination of anti-SARS-CoV-2 spike protein antibody titers among non-vaccinated versus vaccinated solid tumor and hematologic patients who are under clinical observation or under treatment at the University Hospital Krems. Standardized anti-SARS-CoV-2 S antibody titers of a total of 441 patients were retrospectively analyzed. Our results show that antibody titers against the SARS-CoV-2 spike protein are significantly higher in solid tumor versus hematologic patients. While SARS-CoV-2 antibody titers were equal among sexes, an age-dependent decrease was observed. Of note, our studies additionally show that complete vaccination represents a valuable predictor for high anti-SARS-CoV-2 antibody responses in solid tumor and hematologic patients. In summary, to date, this is one of the largest studies to comprehensively evaluate the impact of various COVID-19 vaccines on anti-SARS-CoV-2 S antibody production in solid tumor and hematologic patients. Our findings aim to support future vaccination strategies in these highly vulnerable patients, including vaccination booster programs and alternative protective approaches.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 2354-2354
    Abstract: Angiogenesis or formation of new blood vessels from existing blood vessels, in contrast to vasculogenesis or de novo formation of new vessels, plays an important role in the progression and spread of most cancers. Multiple myeloma (MM) is characterized by increased microvessel density (MVD), a quantitative estimate of angiogenesis, which correlates with stage of disease. MVD increases with progression from MGUS to smoldering MM to newly diagnosed MM and relapsed MM. It is a powerful prognostic factor, predicting for overall survival. To further elucidate the biological basis for the prognostic value of increased angiogenesis in MM, we studied the interactions of MM cells with endothelial cells using HUVECS as a model system. Co-culture of MM cells (MM1.S, OPM2, U266) with HUVECS induced tumor cell proliferation. Enhanced tumor cell proliferation correlated with the number of HUVECs and was greater than that triggered by co-culture with patient bone marrow stromal cells. When HUVECs were fixed prior to co-culture there was a significant decrease in the tumor cell proliferation. Addition of HUVEC conditioned media to the MM cell lines also induced proliferation. Importantly, HUVECS protected against anti-MM agents including conventional agents (Dexamethasone, Doxorubicin, Melphalan) and novel drugs (Revlimid™). The protective effect afforded by co-culture was lost on HUVEC fixation. Intracellular signaling events following MM cell-endothelial cell contact were studied to understand the mechanisms of the proliferative and protective effects. Western blotting demonstrated activation of the JAK/STAT, PI3K/Akt and the MAPK pathways, mediating proliferation and survival. Ongoing studies focused on understanding cytokine as well as adhesion-mediated interactions between the endothelial cells and the MM cells will identify targets for new therapeutic approaches in MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    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. 104, No. 11 ( 2004-11-16), p. 631-631
    Abstract: Vascular endothelial growth factor (VEGF) induces proliferation of MM cells and induces interleukin-6 (IL-6) secretion in a paracrine loop involving MM cells and bone marrow stromal cells. In turn, IL-6 triggers multiple myeloma (MM) cell proliferation and also protects against apoptosis by upregulating Myeloid-cell-leukemia 1 (Mcl-1), a critical survival protein in MM cells. The goal of our study was to investigate the role of Mcl-1 in VEGF induced-proliferation and protection against apoptosis. Using two murine embryonic fibroblast cell lines as a model (a Mcl-1 deleted cell line and its wild type: Mcl-1Δ/null and Mcl-1wt/wt MEFs, respectively), we here demonstrate that deletion of Mcl-1 reduces fetal bovine serum (FBS), VEGF, and IL-6 induced-proliferation. In addition, we demonstrate that the percentage of cells in S phase is lower in Mcl-1Δ/null compared to Mcl-1wt/wt MEFs (21% (+/−1) versus 30% (+/− 3), respectively). Taken together, these results demonstrate that Mcl-1 is required to mediate VEGF, Il-6 and FBS-induced-proliferation and cell cycle progression. To highlight the key anti-apoptotic role of Mcl-1 in MM cells, humans MM1s cells were transfected with Mcl-1 siRNA. Specific inhibition of Mcl-1 was associated with decreased proliferation (42% and 61% decreases at 24 and 48 h, respectively) and induction of apoptosis (subG1 peak: 22% and 41% in Mcl-1 siRNA transfected cells versus 15% and 15 % in non-transfected cells at 24 and 48 h, respectively), confirming that Mcl-1 is critical for both proliferation and protection against apoptosis in MM cells. In 3 human MM cell lines (MM1s, U266 and MM1R) and MM patient cells we next showed that Mcl-1 protein expression, but not other bcl-2 family members, is upregulated by VEGF in a time and dose manner; and conversely that the pan-VEGF inhibitor GW654652, blocks VEGF induced-upregulation of Mcl-1. Furthermore using flow cytometry with a double staining (CD38-FITC and Apo 2.7-PE), we demonstrate that VEGF protects MM patient cells from FBS-starvation-induced-apoptosis: the percentage of apoptotic MM patient cells (CD38++ and Apo 2.7+) in non starved medium (RPMI 1640 supplemented with 10% FBS) was 15% versus 93% in starved medium (RPMI 1640 supplemented with FBS 2%), and 48% in starved medium supplemented with 25ng/ml VEGF. In conclusion, our study demonstrates that VEGF protects MM cells against apoptosis, and that VEGF-induced MM cell proliferation and survival is mediated via Mcl-1. these studies provide the preclinical framework for novel therapeutics targeting both Mcl-1 and/or VEGF 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: 2004
    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. 104, No. 13 ( 2004-12-15), p. 4188-4193
    Abstract: Previous studies have demonstrated the in vitro and in vivo activity of CC-5013 (Revlimid), an immunomodulatory analog (IMiD) of thalidomide, in multiple myeloma (MM). In the present study, we have examined the anti-MM activity of rapamycin (Rapamune), a specific mTOR inhibitor, combined with CC-5013. Based on the Chou-Talalay method, combination indices of less than 1 were obtained for all dose ranges of CC-5013 when combined with rapamycin, suggesting strong synergism. Importantly, this combination was able to overcome drug resistance when tested against MM cell lines resistant to conventional chemotherapy. Moreover, the combination, but not rapamycin alone, was able to overcome the growth advantage conferred on MM cells by interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), or adherence to bone marrow stromal cells (BMSCs). Combining rapamycin and CC-5013 induced apoptosis of MM cells. Differential signaling cascades, including the mitogen-activated protein kinase (MAPK) and the phosphatidylinositol 3′-kinase/Akt kinase (PI3K/Akt) pathways, were targeted by these drugs individually and in combination, suggesting the molecular mechanism by which they interfere with MM growth and survival. These studies, therefore, provide the framework for clinical evaluation of mTOR inhibitors combined with IMiDs 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: 2004
    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. 110, No. 5 ( 2007-09-01), p. 1656-1663
    Abstract: Activation of the extracellular signal-regulated kinase1/2 (ERK1/2) signaling cascade mediates human multiple myeloma (MM) growth and survival triggered by cytokines and adhesion to bone marrow stromal cells (BMSCs). Here, we examined the effect of AZD6244 (ARRY-142886), a novel and specific MEK1/2 inhibitor, on human MM cell growth in the bone marrow (BM) milieu. AZD6244 blocks constitutive and cytokine-stimulated ERK1/2 phosphorylation and inhibits proliferation and survival of human MM cell lines and patient MM cells, regardless of sensitivity to conventional chemotherapy. Importantly, AZD6244 (200 nM) induces apoptosis in patient MM cells, even in the presence of exogenous interleukin-6 or BMSCs associated with triggering of caspase 3 activity. AZD6244 sensitizes MM cells to both conventional (dexamethasone) and novel (perifosine, lenalidomide, and bortezomib) therapies. AZD6244 down-regulates the expression/secretion of osteoclast (OC)–activating factors from MM cells and inhibits in vitro differentiation of MM patient PBMCs to OCs induced by ligand for receptor activator of NF-κB (RANKL) and macrophage-colony stimulating factor (M-CSF). Finally, AZD6244 inhibits tumor growth and prolongs survival in vivo in a human plasmacytoma xenograft model. Taken together, these results show that AZD6244 targets both MM cells and OCs in the BM microenvironment, providing the preclinical framework for clinical trials 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: 2007
    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. 1636-1636
    Abstract: Osteolytic lesions are a hallmark of myeloma bone disease and other metastatic cancers, resulting in significant morbidity. In order to successfully target skeletal disease, it is critical to identify the mediators of osteoclastogenesis. Osteoclastogenesis is a multistep process regulated by receptor activator of nuclear factor κ B ligand (RANKL) and monocyte colony stimulating factor (mCSF). These cytokines recruit monocyte precursors, stimulate their fusion into multinucleated cells, and finally induce osteoclast (OC) formation and activation. Recent data suggest that several chemokines in combination with RANKL and/or vitamin D3 (Oba et al., Exp. Hematology 2005) modulate osteoclastogenesis. Moreover, the autocrine secretion of RANTES and MCP-1 mediate monocyte multinucleation (Kim et al., J. Biol. Chem. 2005), the initial step towards osteoclastogenesis. Since CCR1 is one of the main chemokine receptors expressed on monocytes and OC, we here studied the effects of CCR1 inhibition on osteoclastogenesis. MLN3897 is a small molecule, specific antagonist of the chemokine receptor CCR1. In order to analyze the effects of MLN3897 on osteoclastogenesis, we generated OC from peripheral blood mononuclear cells (PBMC) from healthy donors by stimulation with RANKL and M-CSF (50 ng/ml) for three weeks, in the absence or presence of MLN3897. Mature OC were multinucleated TRAP+ cells, and their functional activity was confirmed by a pit formation and a collagen release ELISA assay. Our data demonstrates that MLN3897 inhibits osteoclastogenesis in a dose and time-dependent fashion. MLN3897 at 10 nM concentration decreases OC number to 40% (range 20 to 70%) compared to untreated controls (p 〈 0.05). Time course experiments demonstrate that MLN3897 inhibites OC formation if added at the beginning of culture, whereas no inhibition is noted after 7 or 14 days. The reduced OC number is associated with decreased OC activity as demonstrated by a decrease in collagen fragments (control 82 +/−8 vs treated 25+/− 19 nM). Because the effects of MLN3897 are noted at early time points, we hypothesized that MLN3897 interfers with the monocyte multinucleation process. Our data demonstrates that MLN3897 induces a 60% reduction in the multinucleated cell number at one week (control 61+/−14 vs treated 35+/−9), associated with decreased nuclei per cell at two weeks. This correlates with an early induction of CCR1 expression on monocytes by RANKL and mCSF. Flow cytometry confirms an increment of double-stained CD14/CCR1 monocyte population (from 20% to 50%) associated with increased ERK phosphorylation at 36 hours of stimulation, which is completely abrogated by MLN3897. Taken together, these data therefore suggest that CCR1 inhibition by MLN3897 blocks ERK pathway activation and impairs the monocyte multinucleation process, an early step in osteoclastogenesis. These studies delineate a novel mechanism of action of MLN3897 on osteoclastogenesis, and provide the rationale for its clinical evaluation to treat osteolytic bone disease.
    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. 128, No. 22 ( 2016-12-02), p. 2091-2091
    Abstract: Angiogenesis significantly influences disease progression in multiple myeloma (MM) patients and is correlated with adverse prognosis. The increase in vascular density within the bone marrow (BM) microenvironment is triggered by oncogene-mediated expression and secretion of pro-angiogenic growth factors and cytokines, most prominently including VEGF. Members of the AP-1 family of transcription factors (TFs) have emerged as actively pursued therapeutic targets over the past years. Our previous studies demonstrated a critical role for the AP-1 family member JunB in MM cell proliferation, survival and drug resistance. Whether JunB also contributes to MM BM angiogenesis is currently unknown. We first sought to identify correlative expression patterns of JUNB and angiogenic factors using the Oncomine software. Indeed, similar to JUNB our data identified significant induction of VEGF, VEGFB, IGF-1 and PlGF, progressing from normal plasma cells to cells from patients with monoclonal gammopathy of undetermined significance (MGUS) and MM in two independent gene expression profiling data sets. In contrast, no correlation was observed between expression of JUNB and ANGPT1, ANGPT2, SDF-1 and FGF4. Besides BM stromal cells (BMSCs) also osteoblasts (OBs) upregulate JunB protein levels in MM: stromal cell co-cultures. This effect is, at least in part, mediated by the humoral milieu, IL-6 in particular. Whether BMSC- and OB- mediated production and secretion of angiogenic factors is mediated via JunB in MM cells was investigated next. Our data show that doxycyclin- induced inhibition of BMSC: OB- mediated JunB upregulation in TetR-shJUNB/ MM.1S cells abrogated production and secretion of VEGF, VEGFB, IGF-1 and PlGF as evidenced by qPCR and ELISA assay. Similar effects were observed in MM: stromal cell co-cultures using the IL-6R inhibitor tocilizumab. Consequently, supernatant of both doxycycline- treated BMSC: Tet-shJUNB/ MM.1S and OB: Tet-shJUNB/ MM.1S co-cultures as well as tocilizumab significantly inhibited angiogenesis, as evidenced by matrigel- based tube formation as well as wound healing assays. Conversely, tamoxifen- induced JunB activity in JunB-ER/MM cells triggered the expression and secretion of angiogenic factors and angiogenesis. Importantly, the functional role of JunB on BM angiogenesis was also verified in vivo using a MM xenograft mouse model. To this purpose, immunodeficient NSG mice were inoculated subcutaneously with Tet-SCR/ MM.1S or Tet-shJUNB/ MM.1S together with BMSCs into the left and right flanks of mice, respectively, and fed with doxycycline in their drinking water for 5 weeks. Treatment with doxycycline inhibited JunB protein levels in Tet-shJUNB/ MM.1S, but not in Tet-SCR/ MM.1S and induced a significant reduction in growth and angiogenesis in tumors formed by Tet-shJUNB/ MM.1S versus control cells, as evidenced by Ki-67 and anti-CD31 staining. In summary, our findings demonstrate for the first time a role for JunB in MM bone marrow angiogenesis, thereby strongly supporting that this TF is a promising new therapeutic target in MM. Disclosures Hose: Takeda: Other: Travel grant; Sanofi: Research Funding; EngMab: Research Funding. Goldschmidt:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium: Membership on an entity's Board of Directors or advisory committees, Research Funding; Chugai: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees. Podar:Novartis: Research Funding; Eutropics: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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. 3426-3426
    Abstract: Histone deacetylase 6 (HDAC6) has an essential role to recruit ubiquitinated proteins to transport to aggresomes, which ultimately induces lysosomal protein degradation. We have shown that inhibition of proteasomes with bortezomib and of aggresomes with HDAC6 inhibitor Tubacin demonstrated significant cytotoxicity in MM cell lines and MM patient tumor cells in vitro (Hideshima T et al., PNAS2005, 102: 8597–8572). In this study, we further examined the biologic significance of HDAC6 inhibition by Tubacin in MM cells. We found that HDAC6 is constitutively associated with heat shock protein (Hsp) 90 in MM cell lines which is enhanced by Tubacin, as evidenced by co-immunoprecipitation. Since Akt and STAT3 have been shown to play important role in proliferation, anti-apoptosis, and drug resistance in MM cells; and all are client proteins of Hsp90, we next further examined whether inhibition of HDAC6 could modulate activities of these proteins via Hsp90. Importantly, Tubacin enhanced phosphorylation of Akt, associated with augmentation of Hsp90 acetylation. Hsp90 inhibitor 17-AAG downregulated Akt phosphorylation associated with enhanced interaction of Hsp90 with Akt, which was partially blocked by Tubacin. On the other hand, 17-AAG did not enhance acetylation of α-tubulin or ubiquitination of proteins, suggesting that Hsp90 does not affect HDAC6 function. Furthermore, we found that STAT3 is also constitutively associated with Hsp90. Importantly, both Tubacin and 17-AAG inhibit phosphorylation of STAT3 in a dose- and time-dependent fashion in MM cells. Taken together, our data indicate that HDAC6 has an important role not only in aggresomal protein degradation, but also in MM cell pathogenesis by modulating Akt and STAT3 signaling cascades via Hsp90 acetylation in MM cells.
    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