GLORIA

GEOMAR Library Ocean Research Information Access

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. 128, No. 22 ( 2016-12-02), p. 2767-2767
    Abstract: Background Targeting new antigens in chronic lymphocytic leukemia (CLL) and lymphoma may increase flexibility in the clinic and help circumvent resistance. The tetraspanin CD37 domain mediates transduction of survival and apoptotic signals (Lapalombella et al.,Cancer Cell, 2014), and has been clinically validated by recent trials of otlertuzumab (TRU-016) in CLL and Non-Hodgkin Lymphoma . Ligation of CD37 by this reagent simultaneously induced pro-apoptotic signaling and inhibited pro-survival signaling of phosphoinositide 3-kinase δ (PI3Kδ), which introduces a unique opportunity to use combination strategies employing activation of CD37 and inhibition of PI3Kδ. A new agent BI 836826 is an Fc-engineered anti-CD37 IgG1 that displays improved effector activities as well as crosslinker-independent direct cytotoxicity. We have evaluated the efficacy of BI 836826 combined with the PI3Kδ-selective inhibitor idelalisib in diffuse large B-cell lymphoma (DLBCL) cell lines and primary human CLL B-cells in the University and then by industry to validate the synergistic finding initially reported. Methods Cell viability assays usedCellTiterGlo to measure inhibition of antibody, isotype control, idelalisib or a combination of antibody and compound over 72h in culture. The cell viability of vehicle is measured at the time of dosing (T0) and after seventy-two hours (T72). A GI reading of 0% represents no growth inhibition, GI 100% represents complete growth inhibition, and a GI 200% represents complete death of all cells in the culture well. Annexin V-FITC and propidium iodide measure by flow cytometry was used to assess enhanced killing of primary CLL cells, with incubation of BI 836826 (0.1 µg/mL) and/or idelalisib (1 µM) at 37°C for 24 hours. Trastuzumab included as a non-specific IgG1 control. Data was reported as percentage of viable cells (Annexin V negative, PI negative) normalized to untreated control. Results DLBCL cell lines were variably sensitive to single agent BI 836826. In most of the cell lines tested, the cell viability was inhibited by 40%-50% with BI 836826 in the concentration range of 1-1000 ng/mL (Figure 1A). A synergistic effect was noted in several DLBCL cell lines when BI 836826 was combined with idelalisib. When the maximal effect of BI 836826 was greater than isotype control (GI% 〉 12, dotted line) and the effect of idelalisib showed a GI50 〈 1uM, 3/5 cell lines showed synergy in combination (red dot, Figure 1B). A shift in the EC50of idelalisib can be seen with the addition of increasing amounts of BI 836826 (Figure 1C). In primary CLL B-cell cultures, 1 µM idelalisib displayed weak single agent activity following 24-hour incubation. The cytotoxicity of BI 836826 at 0.1 µg/mL was more variable, although treatment of samples from most CLL patients resulted in 20-50% B-cell death. The combination of these 2 agents resulted in enhanced cytotoxic activity (Figure 2A), and this effect was not attenuated by the presence of del(17)(p13.1), as there was no significant difference in cytotoxicity against these cells compared to those with lower risk cytogenetics (Figure 2B,C). Additionally, the combination was beneficial in CLL B-cells isolated from patients who were refractory to ibrutinib (Figure 2D). Conclusions This collaborative industry and academic endeavor with cross validation of initial mechanistic studies of synergy between CD37 and idelalisib demonstrates that addition of idelalisib to BI 836826 augments cytotoxicity against DLBCL cell lines and primary human CLL B-cells in an additive-to-synergistic manner. In addition, it maintains efficacy against CLL B-cells with del(17)(p13.1) and those from ibrutinib-refractory patients. Further exploration of this therapeutic strategy in clinical trials is strongly warranted. Disclosures Jones: AbbVie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics, LLC, an AbbVie Company: 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. Awan:Innate Pharma: Research Funding; Pharmacyclics: Consultancy; Novartis Oncology: Consultancy. Grosmaire:Gilead: Employment. Jones:Gilead: Employment. DiPaolo:Gilead: Employment. Tannheimer:Gilead Sciences: Employment. Heider:4Boehringer Ingelheim RCV: Employment.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 127, No. 23 ( 2016-06-09), p. 2879-2889
    Abstract: BI 836858, an Fc-engineered anti-CD33 antibody, mediates autologous and allogeneic NK cell–mediated ADCC. Decitabine increases ligands for activating NK receptors potentiating BI 836858 activity, providing a rationale for combination therapy.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: OncoImmunology, Informa UK Limited, Vol. 5, No. 10 ( 2016-10-02), p. e1226720-
    Type of Medium: Online Resource
    ISSN: 2162-402X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2016
    detail.hit.zdb_id: 2645309-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 288, No. 37 ( 2013-09), p. 26800-26809
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4681-4681
    Abstract: Novel chronic lymphocytic leukemia (CLL) therapies target phosphoinositide 3-kinase (PI3K; idelalisib) and Bruton's tyrosine kinase (BTK; ibrutinib). Despite their success, certain high-risk groups, notably those with dysfunctional P53, demonstrate lower response rates than other CLL groups. As such, combination therapies could potentially overcome this deficiency. As CD37-mediated signaling resulted in direct cytotoxicity that was enhanced with PI3K inhibition (Lapalombella 2012), we aimed to combine BI 836826, a novel IgG1 chimerized and Fc-engineered anti-CD37 mAb, with BCR-pathway inhibitors to possibly enhance clinical efficacy in high-risk CLL patients and overcome potential inhibition of ADCC. Consistent with previous report, BI 836826 displays both ADCC and direct pro-apoptotic activity (Heider 2011). We assessed the potential effect of idelalisib and ibrutinib on CD37 expression and NK-cell function. Following 24hr incubation of primary CLL cells (n=4) with idelalisib, ibrutinib, or DMSO, there was no significant difference in surface expression of CD37 (fold change ~ 1.01; p 〉 0.16 for all comparisons). To ensure that NK-cell Fc-receptor function was unimpaired by BCR-pathway inhibitors, we conducted CD107ab degranulation assays (n=5). Data revealed that Fc-receptor engagement of BI 836826 and NK-cell lytic degranulation was not abrogated by idelalisib or ibrutinib (mean CD107ab expression: DMSO = 10.14%; idelalisib = 5.67%; ibrutinib = 2.41%). In combination with idelalisib, BI 836826 was still capable of eliciting an effective NK-cell response superior to rituximab (difference = 4.51%; p 〈 0.0001). To further qualify NK-cell function, TNFα (n = 6) or INFγ (n = 12) release by NK cells after pretreatment with idelalisib or ibrutinib was evaluated. Similarly, cytokine release was not completely inhibited by idelalisib and ibrutinib; and BI 836826 was still capable of inducing cytokine release. To confirm the potential added benefit of combination therapy, we conducted 51Cr release ADCC assays with primary CLL cells and healthy allogeneic NK-cells (Fig1) or CLL patient autologous NK-cells (Fig2). Results confirmed that neither ibrutinib nor idelalisib could ablate BI 836826-induced NK-cell ADCC. However, there was a more pronounced inhibition of NK-cell ADCC with ibrutinib. As previously demonstrated (Kohrt 2014), NK-cell ADCC mediated by rituximab was almost completely abrogated by ibrutinib, likely secondary to off-target interleukin-2-inducible T-cell kinase inhibition (Dubovsky 2013). Our preliminary data show that signaling induced by BI 836826 may demonstrate the same enhanced direct cytotoxicity with PI3K inhibition as previously reported with other CD37 mediated signaling agents (Lapalombella 2012). Evaluation of direct pro-apoptotic activity of BI 836826 (0.1ug/mL) in combination with idelalisib (1uM) on direct cell death (24hr incubation) demonstrated enhanced activity in the combination setting with similar activity in primary patient CLL cells with dysfunctional P53 (n=6; Fig3A) and functional P53 (n=4; Fig3B). In conclusion, these results demonstrate that idelalisib could potentially be used in combination with BI 836826, which has the added benefit of directly killing inhibitor-resistant P53-null primary CLL cells and lacks complete inhibitor-induced ADCC ablation. Figure 1. Allogeneic ADCC after Pretreatment of NK-cells with BTK inhibitor (Ibrutinib) or PI3K inhibitor (Idelalisib) Figure 1. Allogeneic ADCC after Pretreatment of NK-cells with BTK inhibitor (Ibrutinib) or PI3K inhibitor (Idelalisib) Figure 2. Autologous ADCC after Pretreatment of NK-cells with BTK inhibitor (Ibrutinib) or PI3K inhibitor (Idelalisib) Figure 2. Autologous ADCC after Pretreatment of NK-cells with BTK inhibitor (Ibrutinib) or PI3K inhibitor (Idelalisib) Figure 3. BI 836826 Direct Cytotoxicity in High- and Low-Risk CLL Patients Figure 3. BI 836826 Direct Cytotoxicity in High- and Low-Risk CLL Patients Disclosures Off Label Use: Off-label use of idelalisib and BI 836826 in combination for CLL patients. Jones:Pharmacyclics: Consultancy, Research Funding. Heider:Boehringer Ingelheim: Employment.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 115, No. 6 ( 2010-02-11), p. 1204-1213
    Abstract: CD19 is a B cell–specific antigen expressed on chronic lymphocytic leukemia (CLL) cells but to date has not been effectively targeted with therapeutic monoclonal antibodies. XmAb5574 is a novel engineered anti-CD19 monoclonal antibody with a modified constant fragment (Fc)–domain designed to enhance binding of FcγRIIIa. Herein, we demonstrate that XmAb5574 mediates potent antibody-dependent cellular cytotoxicity (ADCC), modest direct cytotoxicity, and antibody-dependent cellular phagocytosis but not complement-mediated cytotoxicity against CLL cells. Interestingly, XmAb5574 mediates significantly higher ADCC compared with both the humanized anti-CD19 nonengineered antibody it is derived from and also rituximab, a therapeutic antibody widely used in the treatment of CLL. The XmAb5574-dependent ADCC is mediated by natural killer (NK) cells through a granzyme B–dependent mechanism. The NK cell–mediated cytolytic and secretory function with XmAb5574 compared with the nonengineered antibody is associated with enhanced NK-cell activation, interferon production, extracellular signal-regulated kinase phosphorylation downstream of Fcγ receptor, and no increased NK-cell apoptosis. Notably, enhanced NK cell–mediated ADCC with XmAb5574 was enhanced further by lenalidomide. These findings provide strong support for further clinical development of XmAb5574 as both a monotherapy and in combination with lenalidomide for the therapy of CLL and related CD19+ B-cell malignancies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2019
    In:  The Journal of Immunology Vol. 202, No. 9 ( 2019-05-01), p. 2806-2816
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 202, No. 9 ( 2019-05-01), p. 2806-2816
    Abstract: The clinical benefit of CTLA-4 blockade on T cells is known, yet the impact of its expression on cancer cells remains unaddressed. We define an immunosuppressive role for tumor-expressed CTLA-4 using chronic lymphocytic leukemia (CLL) as a disease model. CLL cells, among other cancer cells, are CTLA-4+. Coculture with activated human T cells induced surface CTLA-4 on primary human CLL B cells. CTLA-4 on CLL-derived human cell lines decreased CD80 expression on cocultured CD80+ cells, with restoration upon CTLA-4 blockade. Coculture of CTLA-4+ CLL cells with CD80-GFP+ cell lines revealed transfer of CD80-GFP into CLL tumor cells, similar to CTLA-4+ T cells able to trans-endocytose CD80. Coculture of T cells with CTLA-4+ CLL cells decreased IL-2 production. Using a human CTLA-4 knock-in mouse lacking FcγR function, antitumor efficacy was observed by blocking murine CTLA-4 on tumor cells in isolation of the T cell effect and Fc-mediated depletion. These data implicate tumor CTLA-4 in cancer cell–mediated immunosuppression in vitro and as having a functional role in tumor cells in vivo.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2019
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Oncotarget, Impact Journals, LLC, Vol. 9, No. 11 ( 2018-02-09), p. 9706-9713
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3717-3717
    Abstract: Abstract 3717 The anti-CD20 antibody rituximab represents the first therapy to contribute to prolongation of survival in CLL yet controversy exists as to how it promotes tumor elimination. Pre-clinical studies in CLL have suggested that innate immune cells, complement, and direct antibody killing might contribute to anti-CD20 antibody efficacy. Murine models of B-cell depletion by CD20 antibodies have shown monocytes to be the most important effector cell, although significant controversy around this point exists. Given the success of rituximab, second generation anti-CD20 antibodies (Ofatumumab and GA101 are now in phase III testing in CLL with reported improved direct (GA101), complement (Ofatumumab), or NK cell effector cell killing (GA101). To date, direct comparison of these three antibodies for direct, complement, and effector cell engagement has not been performed. Additionally, the impact of afucosylation engineering of GA101 on monocyte and macrophage function has not been reported. As a Type II anti-CD20 antibody, GA101 mediated significantly increased cell death (∼25%) without Fc crosslinking when compared to Ofatumumab or rituximab (5–8%). Direct cytotoxicity assessment of 19 pts with cross-linking demonstrated GA101 (25%) to mediate significantly greater (p=0.0003) killing than rituximab (15%) but not Ofatumumab (20%). Complement mediated killing was significantly increased over media with Ofatumumab (∼30%), whereas an average of only 5–10% killing was observed with rituximab or GA101. Immobilized GA101 significantly increased NK cell activation as detected by IFNg production and CD107a induction (p=0.005) more than rituximab or Ofatumumab. In addition, GA101 mediates 2 to 3 fold greater NK cell-mediated Antibody Dependent Cellular Cytotoxicity (ADCC) compared to Ofatumumab or rituximab at higher antibody concentrations, but not at concentrations less than 0.05 mg/ml. This enhanced ADCC with GA101 is seen with normal NK cells as well as NK cells from CLL patients. Ofatumumab mediated significantly greater (p=0.0001) NK cell ADCC than rituximab. Given the importance of macrophages in depletion of anti-CD20 tumors in mouse models of lymphoma, we next focused on this. The three anti-CD20 antibodies show Antibody Dependent Cellular Phagocytosis (ADCP) capability with Monocyte Derived Macrophages (MDM) against CLL B cells, with Ofatumumab exhibiting the greatest ADCP compared to GA101 (60% vs 40%; p=0.0036). In addition, primary monocytes stimulated with immobilized GA101 show less TNFa release, when compared to rituximab or Ofatumumab. To investigate this mechanistically, monocytes were stimulated with plate bound rituximab, Ofatumumab or GA101. Compared to the non Fc engineered antibodies (rituximab and Ofatumumab) Fc engineered GA101 induces reduced pan tyrosine phosphorylation and phosphorylation of ERK. No differences in phosphorylation of FcgRIIa or FcgRIIb was observed between the engineered and non-engineered antibodies. Further mechanistic studies to elucidate these differences in monocytes based upon antibody afucosylation changes, or differential phosphorylation of FcgR are ongoing. Collectively, our data indicate that GA101 and Ofatumumab are both superior to rituximab against CLL cells and have differential properties with respect to apoptosis, CDC, and effector cell-mediated killing. GA-101 mediates the most potent NK cell mediated killing at high concentrations whereas Ofatumumab has the greatest monocyte activation and phagocytosis despite absence of any engineering. These findings have relevance in the choice of the optimal CD20 antibody for treatment of CLL and combination strategies used. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3725-3725
    Abstract: Abstract 3725 Poster Board III-661 CD19 is a lineage-specific B-cell antigen, expressed at a high density on CLL cells, that contributes to B-cell receptor signaling but to date has not been effectively targeted with therapeutic monoclonal antibodies. XmAb5574 is a novel engineered anti-CD19 monoclonal antibody with a modified Fc-domain designed to enhance binding of FcγRIIIa that is predominately expressed on Natural Killer (NK)-cells. Utilizing freshly isolated chronic lymphocytic leukemia (CLL) patient B-cells we demonstrate that XmAb5574 lacks significant internalization seen with other anti-CD19 antibodies [maximum internalization for XmAb5574 was only 27.9% at 30-minutes (95%CI 14.5%, 41.4%)], thereby enhancing its ability to induce potent antibody-dependent cellular cytotoxicity (ADCC). Annexin V/PI flow cytometry analysis revealed that XmAb5574 mediates modest direct cytotoxicity not significantly different from Rituximab (0.6% increase, 95%CI -10.5%, 11.7%, p=0.91), and no complement mediated cytotoxicity (CDC) against primary CLL B-cells. Multi-color flow cytometry and monocyte derived macrophages (MDM) were used to assess XmAb5574 antibody dependent cellular phagocytosis (ADCP) against CLL cells and revealed no significant impact of the Fc-domain modification on MDM induced ADCP against CLL cells as compared to the wild type parental anti-CD19 antibody (12.37% vs. 10.51%, p=0.58). Interestingly, utilizing NK-cells and CLL cells isolated from normal donors and CLL patients, and employing autologous and allogeneic effector-target (E:T) conditions, XmAb5574 was found to mediate significantly higher ADCC when compared to the control humanized anti-CD19 non-engineered antibody (26.9% higher at E:T 25:1, p=0.0004 for allogeneic conditions, and 23.6% higher, p=0.004 for autologous conditions). ADCC mediated by XmAb5574 was also significantly higher as compared to Rituximab (33.5% higher at E:T 25:1, p 〈 0.0001 for allogeneic conditions and 27.1% higher, p=0.004 for autologous conditions), a therapeutic antibody widely utilized in the treatment of CLL, hence confirming the functional in vitro efficacy and utility of the Fc-domain modification. By using inhibitor studies we further provide mechanistic insight into the XmAb5574–dependent ADCC mediated by NK-cells through a Granzyme B dependent mechanism. XmAb5574 also enhanced NK-cell activation as exhibited by an increased phosphorylation of Erk1/2 downstream of Fcγ receptor. The enhancement of subsequent cytolytic and secretory function was shown by the measurement CD107a up regulation on the surface of NK-cells (19.4% increase, p=0.005, as compared to wild type anti-CD19 antibody), and interferon-gamma release as measured by ELISA assays (6.4 times higher, p=0.007, as compared to wild type anti-CD19 antibody). Notably, enhanced NK-cell mediated ADCC observed with XmAb5574 against primary CLL B-cells could be augmented further by treatment with Lenalidomide (17.9% higher, p=0.04). These findings provide strong pre-clinical evidence for further clinical development of XmAb5574, both as monotherapy and in combination with Lenalidomide, for the therapy of CLL and related CD19+ B-cell malignancies. We also provide mechanistic insight into the utility and feasibility of Fc-domain engineering of specific antibodies, which will enhance their efficacy through an increased ability to recruit the innate immune system to more effectively control tumor progression. Disclosures: Desjarlais: Xencor: Employment.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...