CD7-restricted activation of Fas-mediated apoptosis: a novel therapeutic approach for acute T-cell leukemia by Edwin Bremer, Bram ten Cate, Douwe F. Samplonius,

Slides:



Advertisements
Similar presentations
RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: role in chemotherapy sensitivity in acute megakaryocytic leukemia by Holly Edwards, Chengzhi Xie,
Advertisements

Combination therapy for adult T-cell leukemia–xenografted mice: flavopiridol and anti-CD25 monoclonal antibody by Meili Zhang, Zhuo Zhang, Carolyn K. Goldman,
Dasatinib suppresses in vitro natural killer cell cytotoxicity
IFNα-stimulated neutrophils and monocytes release a soluble form of TNF-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand) displaying apoptotic activity.
Human Immunodeficiency Virus Type 1 Protease Inhibitor Modulates Activation of Peripheral Blood CD4+ T Cells and Decreases Their Susceptibility to Apoptosis.
Rabbit Anti T-Lymphocyte Globulin Induces Apoptosis in Peripheral Blood Mononuclear Cell Compartments and Leukemia Cells, While Hematopoetic Stem Cells.
by JoAnn Castelli, Elaine K
Antiangiogenic antithrombin down-regulates the expression of the proangiogenic heparan sulfate proteoglycan, perlecan, in endothelial cells by Weiqing.
Characterization of a humanized IgG4 anti-HLA-DR monoclonal antibody that lacks effector cell functions but retains direct antilymphoma activity and increases.
Expression of CD10 by Human T Cells That Undergo Apoptosis Both In Vitro and In Vivo by Giovanna Cutrona, Nicolò Leanza, Massimo Ulivi, Giovanni Melioli,
The supernatant of apoptotic cells causes transcriptional activation of hypoxia-inducible factor–1α in macrophages via sphingosine-1-phosphate and transforming.
The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages by Barbara Scheuerer, Martin Ernst,
A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils by Barbara Geering, Ursina Gurzeler, Elena Federzoni, Thomas Kaufmann,
by Feng-Ting Liu, Samir G. Agrawal, Zanyar Movasaghi, Peter B
by Kumudha Balakrishnan, William G. Wierda, Michael J
Megakaryocyte Growth and Development Factor-Induced Proliferation and Differentiation Are Regulated by the Mitogen-Activated Protein Kinase Pathway in.
by Christina K. Ullrich, Jerome E. Groopman, and Ramesh K. Ganju
Aurora kinase inhibitory VX-680 increases Bax/Bcl-2 ratio and induces apoptosis in Aurora-A-high acute myeloid leukemia by Xue-Fei Huang, Shao-Kai Luo,
by Zhengyan Wang, Tina M. Leisner, and Leslie V. Parise
Activity of vincristine, L-ASP, and dexamethasone against acute lymphoblastic leukemia is enhanced by the BH3-mimetic ABT-737 in vitro and in vivo by Min.
Cord Blood–Derived and Peripheral Blood–Derived Cytokine-Induced Killer Cells Are Sensitive to Fas-Mediated Apoptosis  Ludovic Durrieu, Mame Massar Dieng,
In the absence of IGF-1 signaling, IFN-γ suppresses human malignant T-cell growth by Laura Conti, Gabriella Regis, Angela Longo, Paola Bernabei, Roberto.
Caspases Mediate Tumor Necrosis Factor-–Induced Neutrophil Apoptosis and Downregulation of Reactive Oxygen Production by Kouhei Yamashita, Atsushi Takahashi,
Activation of ATF4 mediates unwanted Mcl-1 accumulation by proteasome inhibition by Jinsong Hu, Nana Dang, Eline Menu, Elke De Bryune, Dehui Xu, Ben Van.
CD6 Ligation Modulates the Bcl-2/Bax Ratio and Protects Chronic Lymphocytic Leukemia B Cells From Apoptosis Induced by Anti-IgM by Lyda M. Osorio, Angelina.
by Norman Nausch, Ioanna E
by Jianwei Zhang, and Keith R. McCrae
Ex vivo induction of multiple myeloma–specific cytotoxic T lymphocytes
Lipid raft adhesion receptors and Syk regulate selectin-dependent rolling under flow conditions by Claire Abbal, Martine Lambelet, Debora Bertaggia, Carole.
The heat shock protein Gp96 binds to human neutrophils and monocytes and stimulates effector functions by Markus P. Radsak, Norbert Hilf, Harpreet Singh-Jasuja,
Histone deacetylase inhibitors: a new class of immunosuppressors targeting a novel signal pathway essential for CD154 expression by Søren Skov, Klaus Rieneck,
Quinine-induced thrombocytopenia: drug-dependent GPIb/IX antibodies inhibit megakaryocyte and proplatelet production in vitro by José Perdomo, Feng Yan,
Chronic neutropenia mediated by Fas ligand
Nongenotoxic activation of the p53 pathway as a therapeutic strategy for multiple myeloma by Thorsten Stühmer, Manik Chatterjee, Martin Hildebrandt, Pia.
Cathepsin-B-dependent apoptosis triggered by antithymocyte globulins: a novel mechanism of T-cell depletion by Marie-Cécile Michallet, Frederic Saltel,
by Jun Yuan, David B. Lovejoy, and Des R. Richardson
by Asim Khwaja, and Louise Tatton
by Bindu Varghese, Adam Widman, James Do, Behnaz Taidi, Debra K
CTLA-4 is not restricted to the lymphoid cell lineage and can function as a target molecule for apoptosis induction of leukemic cells by Maria Pia Pistillo,
Melanoma-Directed Activation of Apoptosis Using a Bispecific Antibody Directed at MCSP and TRAIL Receptor-2/Death Receptor-5  Yuan He, Djoke Hendriks,
The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia by Christine Le Roy, Pierre-Antoine Deglesne, Nathalie.
by Eleanor J. Molloy, Amanda J. O'Neill, Julie J
The BH3-mimetic GX synergizes with bortezomib in mantle cell lymphoma by enhancing Noxa-mediated activation of Bak by Patricia Pérez-Galán, Gaël.
Resistance to Activation-Induced Cell Death and Bystander Cytotoxicity Via the Fas/Fas Ligand Pathway Are Implicated in the Pathogenesis of Cutaneous.
Small-molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukemia cells by Bing Z. Carter, Marcela Gronda,
Fas ligand+ fallopian tube epithelium induces apoptosis in both Fas receptor+ T lymphocytes and endometrial cells  Sebastian E. Illanes, M.D., Kevin Maisey,
Activated monocytes in sickle cell disease: potential role in the activation of vascular endothelium and vaso-occlusion by John D. Belcher, Paul H. Marker,
Bromohydrin pyrophosphate enhances antibody-dependent cell-mediated cytotoxicity induced by therapeutic antibodies by Julie Gertner-Dardenne, Cecile Bonnafous,
Cytokine-Induced CEACAM1 Expression on Keratinocytes Is Characteristic for Psoriatic Skin and Contributes to a Prolonged Lifespan of Neutrophils  Massilva.
by Kamira Maharaj, John J
by Silvia Mele, Stephen Devereux, Andrea G
PPARδ Is a Type 1 IFN Target Gene and Inhibits Apoptosis in T Cells
AML cells display differential sensitivity to inhibition of IKBKE and TBK1. AML cells display differential sensitivity to inhibition of IKBKE and TBK1.
Valerie R. Wiersma, Marco de Bruyn, Robert J
MK-8628 augments T lymphocyte apoptosis.
ABT-199 efficiently kills primary AML myeloblasts as a single agent.
Volume 61, Issue 2, Pages (February 2002)
by Martin Felices, Behiye Kodal, Peter Hinderlie, Michael F
DHA induces apoptosis independently from caspase-8 and FADD in Jurkat T-lymphoma cells. DHA induces apoptosis independently from caspase-8 and FADD in.
Volume 21, Issue 4, Pages (April 2013)
by Babs O. Fabriek, Robin van Bruggen, Dong Mei Deng, Antoon J. M
Volume 16, Issue 12, Pages (December 2008)
by Fabian C. Verbij, Nicoletta Sorvillo, Paul H. P
Bone marrow stroma partially protects multiple myeloma and AML cell lines from tipifarnib- and bortezomib-induced cell death. 8226/S myeloma cells were.
A24 induces apoptosis of MCL cells.
by Dana S. Levy, Jason A. Kahana, and Rakesh Kumar
Induction of apoptosis by statins in NB4 cells and NB4 variant cell lines. Induction of apoptosis by statins in NB4 cells and NB4 variant cell lines. A,
B-cell development in young Pax5+/− mice.
Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans by Asako Sawano,
WEE1 inhibition followed by TRAIL treatment results in apoptotic cell death in MB231 cells. WEE1 inhibition followed by TRAIL treatment results in apoptotic.
Presentation transcript:

CD7-restricted activation of Fas-mediated apoptosis: a novel therapeutic approach for acute T-cell leukemia by Edwin Bremer, Bram ten Cate, Douwe F. Samplonius, Lou F. M. H. de Leij, and Wijnand Helfrich Blood Volume 107(7):2863-2870 April 1, 2006 ©2006 by American Society of Hematology

Solution behavior and stability of scFvCD7:sFasL. Solution behavior and stability of scFvCD7:sFasL. (A) Culture medium derived from CHO-K1 production cells containing scFvCD7:sFasL was subjected to SE-FPLC using a calibrated HiLoad 16/60 FPLC column. The apoptotic activity of each individual fraction was assessed using the FasL-sensitive CD7-positive MOLT16 cells in the presence or absence of CD7-blocking mAb TH69. The horizontal bar in the graph indicates the fractions pooled for further testing. (B) scFvCD7:sFasL was stored for various lengths of time, up to 9 days, at 37°C in the presence of 15% serum. At serial time points the remaining apoptotic activity of the stored scFvCD7:sFasL was assessed by treatment of FasL-sensitive CD7-positive CEM cells for 16 hours. Apoptotic activity was assessed by ΔΨ. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

CD7-restricted binding and apoptosis induction by scFvCD7:sFasL. CD7-restricted binding and apoptosis induction by scFvCD7:sFasL. (A) CD7-restricted binding of scFvCD7:sFasL was assessed using CD7-positive CEM cells. Cells were incubated with scFvCD7:sFasL (solid line) or with unconditioned medium (filled area). Specific binding was demonstrated by preincubating CEM cells with mAb TH69 followed by incubation with scFvCD7:sFasL (dashed line). Binding of scFvCD7:sFasL was determined by flow cytometry using PE-conjugated anti-FasL mAb. (B) CD7-restricted induction of apoptosis by scFvCD7:sFasL was assessed using CD7-positive cells (MOLT-16, CEM, Jurkat, and HUT-78) and CD7-negative Raji cells. All cell types were treated for 16 hours with increasing concentrations of scFvCD7:sFasL, after which apoptosis was assessed by ΔΨ. Additionally, Raji cells were treated with increasing concentrations of cross-linked sFasL (cr-sFasL). Indicated values are mean + SEM of 3 independent experiments. (C) Jurkat cells were treated for 16 hours with scFvCD7:sFasL (20 ng/mL) in the presence or absence of MAb TH69 or FasL-neutralizing MAb Alf2.1. Apoptosis was assessed by staining for apoptotic DNA-fragmentation using mAb F7-26. Horizontal bars indicate the percentage of apoptosis. (D) To compare the specific apoptotic activity of scFvCD7:sFasL with that of the related TRAIL fusion protein scFvCD7: sTRAIL, Jurkat cells were treated with equimolar concentrations of scFvCD7:sFasL and scFvCD7:sTRAIL for 16h. After treatment, apoptosis was assessed by ΔΨ. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

Potent induction of apoptosis in CD7-negative bystander tumor cells. Potent induction of apoptosis in CD7-negative bystander tumor cells. Mixed cultures of Ramos.CD7 target cells and Raji bystander cells (ratio 1:1) were treated for 16 hours with scFvCD7:sFasL (100 ng/mL) in the presence or absence of mAb TH69 or mAb Alf2.1. The differential fluorescent labeling of target and bystander cell populations was used to separately evaluate apoptosis induction by ΔΨ in Ramos.CD7 target cells (A) and Raji bystander cells (B). Indicated values are mean + SEM of 3 independent experiments. Statistical analysis was performed using 2-tailed Student t test. *P < .05; **P < .01. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

Treatment of normal human leukocytes, activated T cells, and HUVECs with scFvCD7:sFasL. Treatment of normal human leukocytes, activated T cells, and HUVECs with scFvCD7:sFasL. (A) Resting PBLs were subjected to treatment with scFvCD7:sFasL (325 ng/mL), or cross-linked sFasL (100 ng/mL) for up to 7 days, after which experimental apoptosis was assessed by annexin V/PI staining. Indicated values are representatives of 3 independent experiments. (B) Isolated PBLs were mixed at a ratio of 1:10 with DiI-labeled Jurkat cells. Mixed cultures were treated for 24 hours with scFvCD7:sFasL (100 ng/mL) or secondarily cross-linked sFasL (100 ng/mL). Differential fluorescent labeling of Jurkat target cells and PBLs was used to separately evaluate apoptosis induction by annexinV staining. Indicated values are representatives of 3 independent experiments. (C) Activated T cells were subjected to treatment with scFvCD7:sFasL (325 ng/mL) for up to 7 days, after which apoptosis was assessed by annexin V/PI staining. Indicated values are representatives of 3 independent experiments. (D) Resting HUVECs were treated for 24 hours with scFvCD7:sFasL (100 ng/mL), secondarily cross-linked sFasL (100 ng/mL), or actinomycin D (2 μg/mL). Apoptosis was assessed by ΔΨ. (E) Resting HUVECs were mixed with fluorescently labeled Jurkat cells (ratio 1:1) and treated with scFvCD7:sFasL (100 ng/mL) or actinomycin D (2 μg/ml) for 24 hours. Differential fluorescent labeling of Jurkat target cells and HUVEC bystander cells was used to separately evaluate apoptosis by ΔΨ. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

Additive tumoricidal effect of scFvCD7:sFasL with several classes of antileukemia agents. Additive tumoricidal effect of scFvCD7:sFasL with several classes of antileukemia agents. Jurkat cells were treated for 16 hours with scFvCD7:sFasL (5 ng/mL) alone or in combination with vincristin (0.1 ng/mL) (A), farnesyl transferase inhibitor L-744832 (25 μM) (B), and the proteasome inhibitor bortezomib (10 nM) (C). Apoptosis induction was assessed by ΔΨ. The CI was calculated as described in “Materials and methods” and used to determine the cooperative effect of combination treatment. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

Cotreatment of PBLs, activated T cells, or HUVECs with scFvCD7:sFasL and several classes of antileukemia agents. Cotreatment of PBLs, activated T cells, or HUVECs with scFvCD7:sFasL and several classes of antileukemia agents. PBLs (A), activated T cells (B), and HUVECs (C) were treated with scFvCD7:sFasL (40 ng/mL) combined with vincristine (0.5 ng/mL), L-744832 (25 μM), or bortezomib (40 nM). Apoptosis was assessed by annexin V/PI staining. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology

Treatment of T-ALL, PTCL, and CD7-positive AML patient-derived blood samples in vitro. Treatment of T-ALL, PTCL, and CD7-positive AML patient-derived blood samples in vitro. (A) Blood cells derived from 4 patients with T-ALL, 1 patient with PTCL, and 1 patient with CD7-positive AML were subjected to treatment with scFvCD7:sFasL (150 ng/mL) and analyzed for apoptosis induction using annexin V/PI staining. (B) Primary CD7-positive AML cells were subjected to treatment with scFvCD7:sFasL alone or in the presence of mAb TH69 for 16 hours, after which apoptosis was assessed by annexin V/PI staining. Statistical analysis was performed using 2-tailed Student t test. *P < .05. (C) Primary T-ALL cells were subjected to scFvCD7:sFasL (150 ng/mL) and then assessed for the presence of active caspase-3 using fluorescent microscopy as described in “Fluorescence microscopy of activated caspase-3.” (D) Primary CD7-positive AML cells were subjected to single-agent treatment with scFvCD7:sFasL (100 ng/mL), vincristine (10 ng/mL), amsacrine (1 μM), or to combination treatment. Apoptosis was assessed by annexin V/PI staining. The CI was used to determine the cooperative effect of combination treatment. Edwin Bremer et al. Blood 2006;107:2863-2870 ©2006 by American Society of Hematology