51cr release
Recently Published Documents


TOTAL DOCUMENTS

131
(FIVE YEARS 2)

H-INDEX

26
(FIVE YEARS 0)

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5305-5305
Author(s):  
Judith Petersen ◽  
Thies Rösner ◽  
Max Kölling ◽  
Katja Klausz ◽  
Christian Kellner ◽  
...  

Background:Apolizumab (APO) is a humanized IgG1 antibody targeting a HLA-DRβ chain specific epitope, which is expressed by approx. 50 % of B cell lymphomas. The antibody's clinical development was hampered by toxicity, potentially mediated by its strong CDC activity. However, previous studies demonstrated that APO efficiently triggered tumor cell killing by myeloid cells, especially by granulocytes. Myeloid effector cell engagement is strongly regulated by CD47-SIRPα interactions, and blockade of this axis has been demonstrated to improve the efficacy of therapeutic antibodies. Hence, our goal was to characterize the influence of CD47-SIRPα blockade on myeloid cell engagement by APO. Methods: Expression of the APO epitope, CD20 and CD47 on different B lymphoma cell lines was measured by flow cytometry. The capacity of APO and Rituximab (RTX) to induce complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) by mononuclear (MNC) or polymorphonuclear (PMN) effector cells was analysed by 51Cr release assays. Antibody-dependent cellular phagocytosis (ADCP) by monocyte-derived macrophages was analysed by fluorescence microscopy. PMN-mediated ADCC and macrophage-mediated ADCP experiments were performed in the presence or absence of the CD47 blocking antibody 5F9-IgG2σ to analyse the impact of CD47-SIRPα blockade on APO-mediated lymphoma cell killing by myeloid cells. Results: As anticipated we observed binding of APO on approx. 50% of the tested B lymphoma cell lines, including Raji (Burkitt's lymphoma), MINO (mantle cell lymphoma) and Carnaval (double hit DLBCL). Focusing on these cell lines, APO's CDC activity was more potent (Raji) or similar (Mino) compared to RTX. Furthermore, APO significantly induced tumor cell lysis by PMN or MNC in 51Cr release assays and achieved higher lysis rates in comparison to RTX. Blocking CD47-SIRPα interactions by 5F9-IgG2σ resulted in significantly increased APO-induced ADCC by PMN against all tested B cell lymphoma lines (Mino: 28.8 ± 10.2 % vs. 41.8 ± 13.3 %; Carnaval: 37.9 ± 17.9 % vs. 55.6 ± 17.6 %; Raji: 23.6 ± 11.7 % vs. 52.4 ± 19.4 % for APO alone vs. APO + 5F9-IgG2σ, respectively, p < 0.05 by ANOVA). However, no ADCC by PMN and RTX was observed - even in the presence of CD47-SIRPα blockade. Moreover, APO mediated similar or even stronger ADCP by macrophages compared to RTX, which could be further enhanced by 5F9-IgG2σ (Figure 1). Conclusions: APO's potent myeloid cell activation could be further enhanced by disruption of CD47-SIRPα interactions. Our observations suggest to generate isotype variants of APO with lower CDC but preserved or enhanced ADCC activity. Subsequently, a combination of APO and CD47-SIRPα blocking strategies may improve B cell lymphoma immunotherapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2046-2046 ◽  
Author(s):  
Robyn A. A. Oldham ◽  
Tania Felizardo ◽  
Nathaniel Zhu ◽  
Daniel H. Fowler ◽  
Jeffrey A. Medin

Abstract Introduction: Despite some striking clinical success thus far, chimeric antigen receptor (CAR) engineered cells have the potential to cause severe side effects. Neurotoxicity and cytokine release syndrome (CRS), the latter characterized by increased levels of cytokines such as IL-6, IFN-γ, and MCP-1, are common adverse events associated with CAR therapy. Lymphodepleting preconditioning regimens are associated with improved clinical responses to CAR therapy, yet lymphodepletion has also been identified as a risk factor for CRS. Understanding and management of these toxicities has improved significantly, however these conditions are challenging to treat and can be life-threatening. The ability to limit or prevent initiation of CRS would greatly improve the safety of CAR therapy. Previous clinical trials have shown that T-Rapa cells (patient T cells that have been grown exvivo in rapamycin) can be successfully infused back into autologous recipients after a low-dose conditioning regimen. After infusion, these T-Rapa cells have potent effector functions and demonstrate long-term persistence. Here we determine that T-Rapa cells, engineered by lentivirus-mediated gene transfer to express an anti-CD19 CAR, are just as effective at killing tumor cells as similarly-engineered pan T cells but produce dramatically less IFN-γ, for example. Methods: Human CD3+ cells were treated with rapamycin in the presence of IFN-α and IL-2 to produce T-Rapa cells with a Th1/Tc1 phenotype. An anti-CD19-41BB-CD3ζ CAR construct was subcloned into a lentiviral vector backbone containing an IRES-eGFP element. Vector was prepared and used to transduce T or T-Rapa cells. Transgene expression was assessed by FACS for eGFP and Protein L staining for the CAR. CAR-T cells were then expanded using CD3/CD28 beads in the presence of IL-2. The expanded cells were used in assays including FACS assessment of T cell phenotype, co-culture assays, and 51Cr release assays in comparison with non-rapamycin treated CAR T cells and non-transduced controls. Results: Following transduction and expansion, similar eGFP and CAR expression levels were found in T and T-Rapa cells transduced at the same MOI. CAR-T and CAR-T-Rapa cells developed from multiple independent T cell donors exhibited similar phenotypes at days 5 and 14 post-thaw, as determined by analyses of T-cell subset and exhaustion markers including CD45RO, CD127, CCR7, CD95, CD25, CXCR3, CTLA-4, PD-1, LAG-3, and TIM-3. Both CAR-T and CAR-T-Rapa cells exhibited comparable levels of cytotoxicity against CD19+ Raji, SUP-B15 and RS4;11 cancer cell lines after coculture for 4 hours in a 51Cr release assay. Further, both T and T-Rapa CAR cells produced similar amounts of IL-2 following a 24-hour coculture with CD19+ Raji, SUP-B15 and RS4;11 cancer cell lines, as measured by ELISA. Interestingly, CAR-T-Rapa cells produced significantly less IFN- γ that CAR-T cells after 24 hours of coculture with CD19+ tumor cells. This observation was consistent for CAR-T and CAR-T-Rapa cells assessed at both day 5 and day 14 post-thaw. Conclusions: T-Rapa cells can be successfully transduced with a CAR vector, and show comparable T cell subset, exhaustion phenotype, and cytotoxicity to CAR-T cells that have not been treated with rapamycin. In spite of these similarities, when challenged with CD19+ tumor cells, CAR-T-Rapa cells produced less IFN-γ than CAR-T cells. Decreased production of IFN- γ may reduce the risk and severity of CRS, improving the safety of CAR therapy. Additional cytokine production studies, as well as in vivo studies, are underway to further characterize T-Rapa cells as a novel CAR effector cell type. Disclosures Felizardo: Rapa Therapeutics: Employment, Patents & Royalties. Zhu:Rapa Therapeutics: Employment, Patents & Royalties. Fowler:Rapa Therapeutics: Employment, Equity Ownership, Patents & Royalties. Medin:Rapa Therapeutics: Membership on an entity's Board of Directors or advisory committees, Patents & Royalties.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3367-3367
Author(s):  
Kotaro Miyao ◽  
Seitaro Terakura ◽  
Keisuke Watanabe ◽  
Reona Sakemura ◽  
Jakrawadee Julamanee ◽  
...  

Abstract Introduction: Endogenous or gene-modified T cells with T cell receptors (TCRs) are major designs of cytotoxic T lymphocytes (CTL) therapies. However, gene-modified TCR-CTL therapies targeting on malignant tumors have been generally unsuccessful in previous clinical trials. The major reasons were poor expansion and short persistence of CTL. To overcome these problems, TCR-affinity have been genetically enhanced, but often resulted in life-threatening toxicities. We aimed to modify TCR-CTL in order to improve the function by enhancing intracellular signaling after TCR ligation without affinity modulation. Method: We generated three artificial T cell activating adapter molecules (ATAM), which are CD3z, CD3z/CD28, and CD3z/4-1BB. CD28 or 4-1BB intracellular domain was inserted in the middle of CD3z intracellular domain, so that they can assemble with internal TCR complex when stimulated with corresponding antigen. ATAM-T2A-tEGFR was packaged into retrovirus vector to transduce the T cells. In order to evaluate the functional alteration, we established ATAM-CMV CTL model. We stimulated T cells with CMV pp65 peptide to induce endogenous CMV CTL. On day 4 after stimulation, ATAMs were retrovirally transduced into CMV CTL. After a selection with anti-EGFR mAb, we achieved more than 90% purity of ATAM positive CMV CTL and used in 51Cr release assay, cytokine release assay, and the growth assay. In the growth assay, we stimulated the ATAM CMV-CTL weekly and assayed the viable cell number, tetramer positivity, phenotype, and apoptosis. Regarding these assay, we used HLA transduced K562 (HLA-K562) as APC. Results: First, we confirmed the expression of each ATAM on CD8+ T cells, which showed similar expression among each ATAM. To determine the cytotoxicity of CMV CTL according to each ATAM or control tEGFR transduction, we performed 51Cr release assay against HLA-K562 pulsed with or without CMV peptide. In the presence of CMV peptide, each ATAM-CMV CTL showed similar cytolytic activities to control CMV CTL. [specific lysis (%): tEGFR 32.0+-6.0, CD3z 32.4+-2.6, CD3z/CD28 31.1+-8.8, and CD3z/4-1BB 37.4+-1.5: at an E:T ratio = 3:1, respectively}. In the intracellular cytokine assay against HLA-K562 pulsed with CMV peptide, similar proportions of responder were positive for IFN-g and IL-2 in control and ATAM-CMV CTL. [IFN-g (%): tEGFR, 67.3+-9.4, CD3z, 69.0+-7.4, CD3z/CD28, 66.5+-8.0, and CD3z/4-1BB, 69.6+-7.9; IL-2 (%): tEGFR, 17.5+-5.0, CD3z, 17.7+-4.3, CD3z/CD28, 15.9+-4.3, and CD3z/4-1BB, 18.9+-4.8, respectively]. The cytotoxicity and cytokine secretion upon irrelevant stimulation were completely negative among each ATAM-CMV CTL. Although the surface phenotype was not changed in control CMV-CTL, that of ATAMs-CMV CTL was significantly expanded after antigen stimulation [central memory (CD45RA-CD62L+) (%): tEGFR, 6.0+-2.9, CD3z, 26.8+-11.1, CD3z/CD28, 19.4+-9.5, and CD3z/4-1BB, 35.8+-26.8: at 7 days after stimulation]. Regarding the cell growth, CD3z/CD28 and CD3z/4-1BB showed greater CTL expansion upon weekly antigen stimulation. The fold expansion of tEGFR, CD3z, CD3z/CD28, and CD3z/4-1BB-CMV CTL were 4.9+-0.7, 8.2+-2.4, 12.3+-1.1, and 13.0+-1.6 at day7, 1.6+-0.4, 8.0+-2.5, 18.6+-3.9, and 21.3+-7.7 at day14, and 0.4+-0.2, 1.4+-0.7, 5.1+-3.0, and 30.4+-22.9 at day21, respectively (Fig). Comparing with control, CD3z/4-1BB-CMV CTL showed continuous greater expansion after several stimulations (p˂0.01: during day0 to day7, p˂0.05: during day7 to day14). To examine the reason for the growth advantage, we investigated the apoptosis after the stimulation with DAPI/Annexin V staining. It revealed that the live cells after specific stimulation elevated in ATAMs-CMV CTL compared with control CMV CTL. Conclusion: We generated three ATAMs and successfully transduced into CMV CTL. Particularly, the whole cell growth and continuance of growth after weekly stimulation were significantly improved in CD3z/4-1BB CTL. This greater cell expansion may improve in vivo persistence and further effectiveness of TCR-CTL therapy. ATAM-CMV CTL did not recognize any APC without corresponding peptide, indicating that ATAM transduction could improve the CTL effectiveness without altering CTL specificity. Thus we believe that we could potentially apply ATAM transduction to other TCR-CTLs targeting malignant tumors safely. Figure Figure. Disclosures Kiyoi: Alexion Pharmaceuticals: Research Funding; Novartis Pharma K.K.: Research Funding; Mochida Pharmaceutical Co., Ltd.: Research Funding; Toyama Chemikal Co.,Ltd.: Research Funding; Sumitomo Dainippon Pharma Co., Ltd.: Research Funding; AlexionpharmaLLC.: Research Funding; JCR Pharmaceutlcals Co.,Ltd.: Research Funding; Nippon Boehringer Ingelheim Co., Ltd.: Research Funding; MSD K.K.: Research Funding; Takeda Pharmaceutical Co., Ltd.: Research Funding; Phizer Japan Inc.: Research Funding; Yakult Honsha Co.,Ltd.: Research Funding; Eisai Co., Ltd.: Research Funding; Astellas Pharma Inc.: Consultancy, Research Funding; Nippon Shinyaku Co., Ltd.: Research Funding; Fujifilm Corporation: Patents & Royalties, Research Funding; Zenyaku Kogyo Co.LTD.: Research Funding; Kyowa-Hakko Kirin Co.LTD.: Research Funding; Chugai Pharmaceutical Co. LTD.: Research Funding; Celgene Corporation: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5193-5193
Author(s):  
Beatrice Clemenceau ◽  
Thierry Guillaume ◽  
Nelly Robillard ◽  
Regine Vivien ◽  
Pierre Peterlin ◽  
...  

Abstract Introduction: Prognosis of acute lymphoblastic leukemia (ALL) in adults remains dismal. Recent advances have consisted of targeted therapies with monoclonal antibodies and chimeric antigen receptor (CAR) T lymphocytes (anti-CD19 CAR T cells), showing very encouraging results. However, relapse still occur after this new therapeutics and increasing the armamentarium for ALL is still required. Her2 is expressed in above one third of adult B-ALL (Chevallier,Haematologica 2009). The anti-HER2 antibodytrastuzumab has been tested prospectively in such patients, showing a low efficacy (Chevallier, Blood 2012).The present work was designed to compare, in vitro, thelysis sensibility of B-ALL HER2+ mediated by two mechanisms of HER2 recognition based on antibody (Ab) specificity: firstly, when the anti-HER2 monoclonalAbtrastuzumab bridges target cells and cytotoxic lymphocytes armed with a Fc receptor (antibodydependant cellular cytotoxicity, ADCC) andsecondly, when HER2-positive target cells are directly recognized by cytotoxic lymphocytes armed with a chimeric antigen receptor (anti-HER2 CAR). Methods: Frozen samples from 7 HER2+ B-ALL adults were available, including onesample expressing also CD20. Characteristics of Patients and controls are presented in Table 1. Three human breast-cancer cell lines (BCCL) expressing different validated (HercepTest, Dako) levels of HER2 (BT474 (HER2 3+), MCF-7 (HER2 0-/1+) and MDA-MB-231 (HER2 0-/1+) were used as positive control. Leukemic HER2-negative B-ALL cells from two patients were used as negative control, including one patient with CD20+ expression. The level of HER2 expression by HER2+ B-ALL was determined by fluorescence-activated cell sorter (FACS) and compared to the HER2 expressing BCCL.Cytotoxic activity was assessed at a 30-to-1 effector-to-target ratio in a 4 hours 51Cr-release assay.We used the same cytotoxic effector lymphocytes (the human NK cell line NK-92), armed with eitheraFcγRIIIa/FcεRIγ receptor (referred to as NK-92CD16) or with an anti-HER2/FcεRIγ CAR receptor (referred to as NK-92CAR) as previously described (Clémenceau, JImmunol Research 2015). For ADCC/CAR assays, target cells were pre-incubated withtrastuzumab and /or rituximab at 10µg/ml each. The study was approved by our local review board and twoalivepatients gave informed consent. Results: The level of HER2 expression by HER2+ B-ALL (n=7) was highly variable with a mean relative fluorescence intensity ratio (RFI) of 13 (range 5 to 30). According to the HercepTestranking used for BCCL lines (see above) all B-ALL would be classified as HER2 0-/1+. In vitro HER2+ BCCL were efficiently lysed by anti-HER2 NK-92CAR in this 4 hours 51Cr-release assay and this direct pathway of killing by CAR was always more efficient than the indirect pathway mediated by ADCC. In addition, thelysis level was correlated to HER2 level expression. Trastuzumabalone has no effect on the spontaneous 51Cr-release of B-ALL cells (not shown). Anti-HER2 CAR mediatedlysis was observed for 5 out of 6 HER2+ B-ALL cells (83%, 1 case (#5) non interpretable). Although lysis levels were low, they werespecificsince the CAR mediatedlysis was inhibited when the B-ALL were pre-incubated with 10 µg/mltrastuzumab (not shown). In contrast, for these 5 HER2+ B-ALL, no anti-HER2 ADCClysis was observed. These results confirmed that in these experimental conditions, HER2 targeting by CAR is more efficient than by ADCC. For the two HER2- B-ALL, no ADCC or CARlysis were observed. Finally, for the two B-ALL expressing CD20, anti-CD20 ADCClysiscan be observed demonstrating that B-ALL cells are not intrinsically resistant to ADCCcytoxicity. Regarding the unique case of B-ALL expressing both CD20/HER2 antigens, only anti-CD20 ADCClysiswas observed. Results are presented in Table 2. Conclusion: Together, these preliminary results suggest that for the CD20-negative and HER2-positive B-ALLs, one cannot rely on ADCC as a mechanism of target celllysis, while the interest of an anti-HER2-CAR approach deserve further studies to better correlate the level of HER2 expression and the sensitivity tolysisby HER2-specific CAR. Disclosures No relevant conflicts of interest to declare.


BIO-PROTOCOL ◽  
2016 ◽  
Vol 6 (16) ◽  
Author(s):  
Petra Baumgaertner ◽  
Daniel Speiser ◽  
Pedro Romero ◽  
Nathalie Rufer ◽  
Michael Hebeisen

Sign in / Sign up

Export Citation Format

Share Document