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. 126, No. 23 ( 2015-12-03), p. 3024-3024
    Abstract: Background: Monoclonal antibodies (mAbs) are an emerging therapeutic class for MM patients (pts). Elotuzumab, a mAb in late-phase clinical development, targets the SLAMF7 receptor expressed highly on MM cells. While its primary mechanism of action is through CD16-mediated ADCC, elotuzumab can also directly activate SLAMF7-expressing NK cells. Gaining a greater understanding of phenotypic and functional changes in NK cells over the course of the disease, and how these changes impact capacity for ADCC, may help identify profiles that can better select pts likely to benefit from elotuzumab or other mAb therapies. Methods: We prospectively performed a comprehensive flow cytometry-based analysis of lymphocyte subsets, focusing on expression of NK cell activating and inhibitory receptors, activation and maturation markers, and degranulation in 30 MM pts (12 newly-diagnosed (ND), 18 relapsed/refractory (RR)) and 19 aged-matched healthy donors (HD). Over 140 immune parameters were analyzed, with differences in expression between HD and pt subsets compared by Wilcoxon rank-sum test. We analyzed correlations between expression of certain markers with each other, and with elotuzumab-induced NK cell degranulation against MM cell targets (MM1R) in a 2-hour co-culture assay. We also compared NK cell parameters in blood and bone marrow (BM) from pts with matched samples available. Results: Within the blood, there was no difference in relative NK cell frequency between the groups, and little difference phenotypically between HD and ND pt NK cells, except for decreased DNAM1 expression in ND. In contrast, in comparison to HD, CD56dim NK cells in RR pts were less mature with a higher CD56bright to CD56dim NK cell ratio and reduced expression of the terminal differentiation/maturation markers, CD57 and KLRG1. RR pts also showed increased expression of the activation marker CD69 on all NK cells, and their CD56dim NK cells had increased levels of the natural cytotoxicity receptors, NKp30 and NKp46 and decreased expression of activating receptors DNAM1 and NKG2D. SLAMF7 expression was also increased in RR pts, but only on the CD56bright subset. Consistent changes in NK cell expression of checkpoint/co-stimulatory molecules (eg. PD-1, Tim3, LAG3, CD137) were not seen. Despite these phenotypic changes, no significant differences between groups were noted for elotuzumab-induced ADCC against MM1R targets, as measured by CD107a degranulation by CD56dim NK cells, with significant variability noted within groups. Interestingly, the expression levels of SLAMF7 on CD56dim NK cells directly correlated with CD16 levels, particularly within RR pts (Fig.), suggesting cooperative interactions between these receptors that may be beneficial in MM patients treated with elotuzumab. In addition, degranulation toward elotuzumab-treated MM1R targets was significantly associated with surface expression levels of both SLAMF7 and CD16 on the CD56dim NK cells. The status of NK cells was also compared between matching blood and BM samples from ND (n=7) and RR (n=8) pts. NK cell phenotype and degranulation in blood and BM were similar in ND pts, but in RR pts, expression of CD69 and SLAMF7 were higher on BM-derived NK cells, and CD56dim NK cells from BM demonstrated greater degranulation toward elotuzumab-treated MM1R targets. DNAM1 expression was reduced, but NKG2D, NKp30, and NKp46 were upregulated on various NK cell populations in BM from RR pts compared to peripheral blood. Conclusions: Taken together, our data indicate that NK cells in RR MM pts had increased activation, reduced maturation status, and distinct changes in activating receptor expression levels that are often further enhanced in the BM microenvironment. Furthermore, CD56dim NK cells in many RR pts had parallel increased expression levels of CD16 and SLAMF7, which correlated with enhanced degranulation toward elotuzumab-treated MM target cells. The fact that these changes are seen primarily in RR pts rather than untreated ND pts implies a significant impact of disease evolution and prior therapy on the NK cell compartment, and supports further exploration of these parameters as potential biomarkers of activity of elotuzumab and other therapeutic mAbs in myeloma. Figure 1. Figure 1. Disclosures Campbell: Bristol-Meyers Squibb: Membership on an entity's Board of Directors or advisory committees, Research Funding. Cohen:Bristol-Meyers Squibb: 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.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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: OncoImmunology, Informa UK Limited, Vol. 6, No. 9 ( 2017-09-02), p. e1339853-
    Type of Medium: Online Resource
    ISSN: 2162-402X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2017
    detail.hit.zdb_id: 2645309-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 8 ( 2017-03-30)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2017
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 2 ( 2021-02-01), p. 446-479
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has a poor 5-year survival rate and lacks effective therapeutics. Therefore, it is of paramount importance to identify new targets. Using multiplex data from patient tissue, three-dimensional coculturing in vitro assays, and orthotopic murine models, we identified Netrin G1 (NetG1) as a promoter of PDAC tumorigenesis. We found that NetG1+ cancer-associated fibroblasts (CAF) support PDAC survival, through a NetG1-mediated effect on glutamate/glutamine metabolism. Also, NetG1+ CAFs are intrinsically immunosuppressive and inhibit natural killer cell–mediated killing of tumor cells. These protumor functions are controlled by a signaling circuit downstream of NetG1, which is comprised of AKT/4E-BP1, p38/FRA1, vesicular glutamate transporter 1, and glutamine synthetase. Finally, blocking NetG1 with a neutralizing antibody stunts in vivo tumorigenesis, suggesting NetG1 as potential target in PDAC. Significance: This study demonstrates the feasibility of targeting a fibroblastic protein, NetG1, which can limit PDAC tumorigenesis in vivo by reverting the protumorigenic properties of CAFs. Moreover, inhibition of metabolic proteins in CAFs altered their immunosuppressive capacity, linking metabolism with immunomodulatory function. See related commentary by Sherman, p. 230. This article is highlighted in the In This Issue feature, p. 211
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: ACS Nano, American Chemical Society (ACS), Vol. 6, No. 6 ( 2012-06-26), p. 5143-5149
    Type of Medium: Online Resource
    ISSN: 1936-0851 , 1936-086X
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2012
    detail.hit.zdb_id: 2383064-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2998-2998
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2998-2998
    Abstract: Elotuzumab (Elo) is an IgG1 monoclonal antibody targeting SLAMF7 (CS1, CRACC, CD319) protein, which is highly and uniformly expressed on multiple myeloma cells. Improved survival has been observed in multiple myeloma patients treated with Elo in combination with immunomodulatory drugs such as dexamethasone and lenalidomide. Previous work showed that the main mechanism of Elo action in vitro is CD16-mediated antibody dependent cellular cytotoxicity (ADCC) via Fc interaction with CD16 (FcγRIIIa) receptor on NK cells. However, SLAMF7 is also expressed on NK cells where it transmits an activating signal. We hypothesized that Elo can directly activate NK cells via SLAMF7 ligation. Utilizing non-fucosylated Elo that has higher affinity to CD16 and Elo mutant that has an Fc region mutation that abrogates binding to CD16, we confirmed that Elo promotes NK cell activation and degranulation in a CD16-dependent manner. Specifically, non-fucosylated Elo had higher potency, whereas Elo mutant did not stimulate degranulation or CD69 expression. To test for co-stimulatory effects of Elo and Elo mutant we measured calcium signaling responses triggered by those antibodies in combination with antibodies to engage the activating receptors NKp46 and NKG2D. Elo or Elo mut alone had no effect on calcium signaling, but when added in combination with multimeric engagement of NKp46 and NKG2D both antibodies co-stimulated calcium signaling responses. We conclude that Elo stimulates NK cells via interaction with CD16, but direct engagement with SLAMF7 can also transduce co-activating signals to potentiate calcium signals generated through NKp46 and NKG2D in a CD16-independent manner. Our results provide evidence that direct engagement with SLAMF7 by Elo has the potential to reduce the threshold of NK cell activation triggered through other activating receptors. Citation Format: Tatiana Pazina, Ashley Mentlik James, Alexander W. MacFarlane, Natalie A. Bezman, Robert F. Graziano, Michael D. Robbins, Adam D. Cohen, Kerry S. Campbell. Elotuzumab can costimulate NK cell responses by engaging SLAMF7 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2998. doi:10.1158/1538-7445.AM2017-2998
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2038-2038
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is predicted to soon become the second killer cancer in the USA. One of the hallmarks of PDAC is desmoplasia, which consists of an expansion in activated stellate cells, known as cancer associated fibroblasts (CAFs), and remodeling of their extracellular matrix. Desmoplasia is a major culprit for the unique PDAC microenvironment, which both collapses blood vessels restricting nutrients and fosters immunosuppression. Crosstalk between CAFs, PDAC, and immune cells, within this microenvironment, is not fully understood. To study the functions of CAFs on PDAC development, we first performed a microarray comparing patient matched normal pancreatic stellate cells and CAFs cultured using our stromal 3D system. We identified proteins upregulated in CAFs that have not been previously implicated in desmoplasia. To understand the functional role of these proteins, CRISPR/Cas9 mediated deletion of target proteins was undertaken and cells were subjected to a number of co-culture assays, with PDAC cells and NK cells. Assays consisted of material transfer, cell engagement, proliferation, survival, NK cell activation and PDAC killing. Syngeneic/orthotopic in vivo models, RNAseq analyzes and quantitative multi-plex microscopy, on pathological samples, were also used. We uncovered the ectopic expressions of NetrinG1 (NG1), a neural pre-synaptic protein, in CAFs, and of NG1’s post-synaptic binding partner, NGL1, in PDAC cells. Using our 3D system, we observed that heterotypic CAF-to-PDAC interactions, via NG1/NGL1 engagement, are critical for providing survival advantages to nutrient deprived PDAC cells and for CAFs to protect PDAC from NK cell-driven elimination. Mechanistically, we uncovered that PDAC starvation is overcome by NG1 expressing CAFs that provide nutrition via material transfer. Further, knockout of NG1, in CAFs, significantly reduced the production of immunosuppressive cytokines, such as IL-6, IL-8, and TGF-β. Moreover, loss of NG1 from CAFs resulted in anti-tumor NK cell activation. Unexpectedly, we observed a link between glutamate metabolism in CAFs and the inactivation of anti-tumor NK cells, and this was dependent on CAFs’ NG1 expression. These results were confirmed in an orthotopic PDAC mouse model in which ablation of NGL1, in syngeneic mouse PDAC cells, significantly halted tumor growth. RNAseq comparing CON or NG1+ CAFs vs. knockout NG1- CAFs rendered changes in neural, metabolic and fibrous signature pathways. Translationally, we saw clear expression of stromal NG1 only in PDAC affected pancreata. Overall, this study identifies two novel targets for PDAC; a cancer that lacks effective therapies. Inhibition of the NG1/NGL1 axis, constitutes a potential future therapeutic approach, as it alters CAF mediated metabolism and reverts immunosuppression, stunting PDAC growth in vitro and in vivo. Citation Format: Ralph Francescone, Débora Barbosa Vendramini-Costa, Janusz Franco-Barraza, Jessica Wagner, Alex Muir, Linara Gabitova, Tatiana Pazina, Tiffany Luong, Neelima Shah, Dustin Rollins, Ruchi Malik, Sapna Gupta, Roshan Thapa, Diana Restifo, Allison Lau, Yan Zhou, Kathy Q. Cai, Harvey Harvey Hensley, Emmanuelle Nicolas, Warren D. Kruger, Karthik Devarajan, Siddharth Balachandran, Wafik S. El-Deiry, Matthew Vander Heiden, Kerry Campbell, Igor Astsaturov, Edna Cukierman. NG1/NGL1 engagement supports PDAC development via CAF to PDAC nutrition and CAF-regulated immunosuppression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2038.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Frontiers Media SA ; 2018
    In:  Frontiers in Immunology Vol. 9 ( 2018-11-5)
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 9 ( 2018-11-5)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2018
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 2092-2092
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2092-2092
    Abstract: Multiple myeloma (MM) is a malignancy of plasma cells characterized by a monoclonal expansion of tumor cells in bone marrow and accumulation of a monoclonal protein in blood and urine, which ultimately leads to end-stage organ damage. Despite advances in therapy, MM remains an incurable disease. Recent investigations have revealed a new therapeutic target for MM, SLAMF7 (CS1), which is expressed at high levels on MM tumor cells. SLAMF7 is also found on NK, CD8+ T, CD4+ T and activated B cells. Elotuzumab (HuLuc63) is a humanized IgG1 anti-SLAMF7 antibody, which can facilitate killing of primary MM cells by NK cells via antibody-dependent cellular cytotoxicity (ADCC) and has shown promise in treating relapsed MM. A key question in understanding elotuzumab’s mechanism of action is whether it can directly activate NK cells via SLAMF7 binding, in addition to Fc-mediated activation via binding of CD16 (FcγRIIIA). We have examined the in vitro effect of elotuzumab on NK cells in fresh human peripheral blood from healthy donors and MM patients incubated with or without MM cell lines that have varied levels of SLAMF7 surface expression. Cytotoxic degranulation of NK cells was quantified by staining for CD107a (marker of degranulation) after 2 hours, and direct effects of elotuzumab on NK cells were assessed by measuring upregulation of the CD69 activation marker after 24 hours of antibody exposure. Degranulation of fresh primary NK cells toward SLAMF7-bearing MM target cell lines was stimulated by addition of elotuzumab. Intensity of the degranulation response was dependent upon the concentration of elotuzumab and the expression level of SLAMF7 on the MM target cells. While elotuzumab alone could stimulate low-level degranulation of NK cells in fresh PBL from healthy donors, the degranulation response was stronger if SLAMF7-expressing MM target cells were added in combination with elotuzumab. Addition of F(ab’)2 elotuzumab or an Fc mutant form of elotuzumab that cannot bind CD16 did not stimulate degranulation, implying that binding of the antibody to SLAMF7 on NK cells and MM cells, without concomitant engagement of NK cell Fc receptors, is insufficient to trigger degranulation. Soluble intact elotuzumab stimulated CD69 expression and degranulation by fresh primary NK cells in the absence of MM target cells, which peaked at 1 µg/ml and only modestly increased up to 100 µg/ml elotuzumab. In the same assays, we only observed modest stimulation of CD69 expression using soluble F(ab’)2 or Fc mutant elotuzumab. In contrast, plate-bound F(ab’)2 elotuzumab stimulated robust CD69 expression, indicating that the antibody can induce direct stimulation of NK cells through SLAMF7, but strong stimulation requires aggregation of the antibody. In soluble form, a non-fucosylated version of elotuzumab with greater affinity for CD16 stimulated significantly stronger degranulation and CD69 expression than intact elotuzumab, both in the presence or absence of MM target cells, and with both high and low SLAMF7 expression on target cells. Importantly, elotuzumab was found to induce similar degranulation by NK cells from freshly isolated peripheral blood of MM patients as compared to healthy donors. Taken together, our data support the hypothesis that binding of soluble elotuzumab to SLAMF7 can weakly activate NK cells, but strong activation and degranulation require antibody aggregation or engagement of the Fc domain with CD16. Elotuzumab variants with enhanced CD16 binding, such as a non-fucosylated version, can augment ADCC against myeloma cell lines, and warrant further study as a potential strategy to improve clinical efficacy in MM patients. Disclosures Cohen: Celgene: Member, Independent Response Adjudication Committee Other; Janssen: Advisory Board, Advisory Board Other; Bristol-Myers Squibb: Advisory Board, Advisory Board Other, Research Funding; Onyx: Advisory Board, Advisory Board Other. Campbell:Bristol-Myers Squibb: Advisory Board Other, Research Funding; Janssen: Advisory Board, Advisory Board Other; Conkwest: Consultancy, Patented NK-92 cell lines, Patented NK-92 cell lines Patents & Royalties, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    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
    Online Resource
    Online Resource
    American Society of Hematology ; 2015
    In:  Blood Vol. 126, No. 23 ( 2015-12-03), p. 2964-2964
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2964-2964
    Abstract: Introduction: Elotuzumab is a humanized monoclonal antibody that binds specifically to Signaling Lymphocyte Activation Molecule Family Member 7 (SLAMF7; CS1), a glycoprotein highly expressed on the surface of multiple myeloma (MM) cells and on subsets of immune cells, including natural killer cells. SLAMF7 also exists as a soluble form (sSLAMF7) and has been detected in the serum of patients with MM at statistically significant higher levels than healthy individuals. Although the mechanism of action of elotuzumab involves the direct activation of natural killer cells and the initiation of antibody-dependent cell-mediated cytotoxicity of SLAMF7-expressing myeloma cells, its effects on sSLAMF7 have not been reported. The biological significance of sSLAMF7 has not been described in MM; however, it has been suggested that sSLAMF7 may be associated with disease stage. In order to assess the pharmacodynamic and predictive utility of this biomarker, we investigated levels of serum sSLAMF7 at baseline and on treatment (Cycle 2 Day 1 [C2D1]) across several elotuzumab clinical studies (HuLuc63-1703, CA204-009, and CA204-011) in both smoldering MM and relapsed/refractory MM. Additionally, associations between changes in sSLAMF7 and pharmacokinetic parameters were investigated. Lastly, associations between sSLAMF7 and markers of disease burden were also assessed. Methods: Two enzyme-linked immunosorbent assays were developed to detect sSLAMF7 in the presence of elotuzumab: the total assay (TA) detected all forms of sSLAMF7 (both unbound and elotuzumab-bound sSLAMF7) and the free assay (FA) detected only unbound sSLAMF7. Serum was collected from peripheral blood before initiation of treatment (C1D1) and during treatment (C2D1) and assessed for levels of sSLAMF7 by the TA and FA. Soluble component (non-cellular) from bone marrow aspirates were obtained prior to any therapy to assess the relationship between concentrations of sSLAMF7 in matched marrow and blood samples. Results: As previously reported, these data confirm sSLAMF7 expression in patients with MM. The TA demonstrated a significant dose- and time-dependent increase in sSLAMF7 following elotuzumab treatment alone or in combination with either bortezomib/dexamethasone (E-B/d) or lenalidomide/dexamethasone (E-L/d); this increase was not observed in patients treated with bortezomib/dexamethasone (B/d) only. The increase in total sSLAMF7 observed in the TA demonstrated elotuzumab pharmacodynamic specificity and likely reflected stabilization of the protein in the serum. The FA detected a significant decrease in free sSLAMF7 (at C2D1) in both elotuzumab- and non-elotuzumab-treated patients, with the greatest reduction observed in elotuzumab-treated patients. The decrease in free sSLAMF7 detected using the FA, regardless of treatment, suggest the potential use of this biomarker as a surrogate for disease burden. Additionally, the extent of the C2D1 changes in free and total sSLAMF7 following elotuzumab-containing regimens demonstrated a trend for association with increased depth of response (comparing progressive disease/stable disease/minimal response vs partial response vs very good partial response/complete response). Baseline and C2D1 levels of serum sSLAMF7 showed a trend for an inverse relationship with progression-free survival (for E-L/d) but this was not statistically significant. Baseline sSLAMF7 did demonstrate a positive relation with the percentage of bone marrow plasma cells. At C2D1, with the dose of 10 mg/kg, the mean elotuzumab concentration was 337.1 µg/mL (N=38; CV of 51.6%), which was significantly higher than the mean free sSLAMF7 concentration 0.255 ng/mL (N=37; CV of 200%). In addition, E-L/d treatment demonstrated the greatest reduction of free sSLAMF7 (more than 80% of patients treated with E-L/d reached 〉 75% of free sSLAMF7 reduction) compared with that observed for E-B/d and B/d. Conclusions: Together, these data suggest that sSLAMF7 may be a surrogate marker of disease burden in MM, and early changes in sSLAMF7 may provide an indication of likelihood of response. Additional analysis of sSLAMF7 is ongoing and future studies will help us to further understand the use of this biomarker for prognostic or predictive purposes. Study supported by: Bristol-Myers Squibb Disclosures Postelnek: Bristol-Myers Squibb: Employment. Off Label Use: Elotuzumab is an investigational agent being studied for the treatment of multiple myeloma.. Sheridan:AbbVie Biotherapeutics: Employment, Equity Ownership. Keller:AbbVie Biotherapeutics: Employment, Equity Ownership. Sheng:Bristol-Myers Squibb: Employment, Equity Ownership. Poulart:Bristol-Myers Squibb: Employment. Robbins:Bristol-Myers Squibb: Employment, Other: shareholder.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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