Volume 10, Issue 4, Pages (October 2006)

Slides:



Advertisements
Similar presentations
Volume 14, Issue 4, Pages (October 2008)
Advertisements

Volume 114, Issue 5, Pages (May 1998)
Cantharidin Inhibits the Growth of Triple-Negative Breast Cancer Cells by Suppressing Autophagy and Inducing Apoptosis in Vitro and in.
Volume 18, Issue 6, Pages (December 2010)
An RNA Molecule Derived From Sendai Virus DI Particles Induces Antitumor Immunity and Cancer Cell-selective Apoptosis  Li-Wen Liu, Tomoyuki Nishikawa,
An anti-CD20–IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma by Stephen D. Gillies, Yan Lan, Steven Williams,
Volume 15, Issue 6, Pages (June 2009)
Volume 12, Issue 3, Pages (September 2007)
Cell Surface CD74–MIF Interactions Drive Melanoma Survival in Response to Interferon-γ  Keiji Tanese, Yuuri Hashimoto, Zuzana Berkova, Yuling Wang, Felipe.
Upregulation of PD-L1 by EGFR Activation Mediates the Immune Escape in EGFR- Driven NSCLC: Implication for Optional Immune Targeted Therapy for NSCLC Patients.
Characterization of a humanized IgG4 anti-HLA-DR monoclonal antibody that lacks effector cell functions but retains direct antilymphoma activity and increases.
Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor–κB and IκBα kinase in human multiple myeloma cells, leading to.
Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas by Pankaj Gupta,
Volume 5, Issue 2, Pages (August 2009)
Regulation of Mast Cell Survival by IgE
Myung Jin Son, Kevin Woolard, Do-Hyun Nam, Jeongwu Lee, Howard A. Fine 
Inhibition of glycogen synthase kinase-3 activity leads to epigenetic silencing of nuclear factor κB target genes and induction of apoptosis in chronic.
Volume 14, Issue 3, Pages (September 2008)
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Cathepsin-B-dependent apoptosis triggered by antithymocyte globulins: a novel mechanism of T-cell depletion by Marie-Cécile Michallet, Frederic Saltel,
Volume 17, Issue 2, Pages (February 2010)
KIR3DL2/CpG ODN Interaction Mediates Sézary Syndrome Malignant T Cell Apoptosis  Bouchra Ghazi, Nicolas Thonnart, Martine Bagot, Armand Bensussan, Anne.
Dienogest enhances autophagy induction in endometriotic cells by impairing activation of AKT, ERK1/2, and mTOR  JongYeob Choi, Ph.D., MinWha Jo, M.S.,
Volume 18, Issue 5, Pages (May 2010)
Volume 12, Issue 5, Pages (November 2007)
Volume 6, Issue 1, Pages (January 2014)
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Volume 25, Issue 3, Pages (March 2017)
Volume 23, Issue 1, Pages (July 2006)
Involvement of Oxidative Stress in Apoptosis Induced by a Mixture of Isothiazolinones in Normal Human Keratinocytes  Anna Ettorre, Paolo Neri, Anna Di.
Volume 18, Issue 6, Pages (December 2010)
Volume 36, Issue 1, Pages (January 2012)
Volume 73, Issue 5, Pages (March 2008)
Activation of the Innate Signaling Molecule MAVS by Bunyavirus Infection Upregulates the Adaptor Protein SARM1, Leading to Neuronal Death  Piyali Mukherjee,
Non-canonical PI3K-Cdc42-Pak-Mek-Erk Signaling Promotes Immune-Complex- Induced Apoptosis in Human Neutrophils  Julia Y. Chu, Ian Dransfield, Adriano G.
Protodynamic Intracellular Acidification by cis-Urocanic Acid Promotes Apoptosis of Melanoma Cells In Vitro and In Vivo  Jarmo K. Laihia, Janne P. Kallio,
Lamp-2a Facilitates MHC Class II Presentation of Cytoplasmic Antigens
Volume 22, Issue 3, Pages (March 2015)
Volume 22, Issue 1, Pages (July 2012)
Inhibition of KLF4 by Statins Reverses Adriamycin-Induced Metastasis and Cancer Stemness in Osteosarcoma Cells  Yangling Li, Miao Xian, Bo Yang, Meidan.
Cisplatin augments cytotoxic T-lymphocyte–mediated antitumor immunity in poorly immunogenic murine lung cancer  Robert E Merritt, MD, Ali Mahtabifard,
Volume 82, Issue 7, Pages (October 2012)
Volume 10, Issue 1, Pages (July 2006)
Regulation of Mast Cell Survival by IgE
Volume 22, Issue 3, Pages (March 2005)
Volume 6, Issue 1, Pages (January 2014)
Volume 29, Issue 6, Pages (December 2008)
B Cell Receptor and CD40 Signaling Are Rewired for Synergistic Induction of the c-Myc Transcription Factor in Germinal Center B Cells  Wei Luo, Florian.
Death Receptor-Independent Apoptosis in Malignant Melanoma Induced by the Small- Molecule Immune Response Modifier Imiquimod  Michael P. Schön, B. Gregor.
Myeloma cell–derived Runx2 promotes myeloma progression in bone
Select Cancer Testes Antigens of the MAGE-A, -B, and -C Families Are Expressed in Mast Cell Lines and Promote Cell Viability In Vitro and In Vivo  Bing.
Activation of Akt as a Mechanism for Tumor Immune Evasion
Volume 6, Issue 1, Pages (January 1997)
Thomas S. Griffith, Elizabeth L. Broghammer  Molecular Therapy 
Volume 17, Issue 2, Pages (February 2009)
Volume 12, Issue 5, Pages (November 2007)
A CTLA-4 Antagonizing DNA Aptamer with Antitumor Effect
Volume 44, Issue 4, Pages (April 2016)
Chie Kudo-Saito, Hiromi Shirako, Tadashi Takeuchi, Yutaka Kawakami 
Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis
Volume 8, Issue 4, Pages (October 2005)
Volume 22, Issue 1, Pages (July 2012)
Targeted Cleavage of Signaling Proteins by Caspase 3 Inhibits T Cell Receptor Signaling in Anergic T Cells  Irene Puga, Anjana Rao, Fernando Macian  Immunity 
A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma
Volume 18, Issue 3, Pages (September 2010)
Yann Leverrier, Anne J. Ridley  Current Biology 
Volume 14, Issue 3, Pages (September 2008)
Yoshinori Aragane, Akira Maeda, Chang-Yi Cui, Tadashi Tezuka 
Sean P. Cullen, Conor J. Kearney, Danielle M. Clancy, Seamus J. Martin 
Inhibition of NF-κB in cancer cells converts inflammation- induced tumor growth mediated by TNFα to TRAIL-mediated tumor regression  Jun-Li Luo, Shin.
Presentation transcript:

Volume 10, Issue 4, Pages 295-307 (October 2006) Targeting β2-microglobulin for induction of tumor apoptosis in human hematological malignancies  Jing Yang, Jianfei Qian, Michele Wezeman, Siqing Wang, Pei Lin, Michael Wang, Shmuel Yaccoby, Larry W. Kwak, Bart Barlogie, Qing Yi  Cancer Cell  Volume 10, Issue 4, Pages 295-307 (October 2006) DOI: 10.1016/j.ccr.2006.08.025 Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 1 In vitro tumoricidal activity of β2M-specific mAbs A: Annexin V-staining, and B, TUNEL assay for detection of apoptosis in myeloma cells in cultures for 48 hr with medium or with the addition of 100 μg/ml β2M-specific mAb B2, HLA-ABC-specific mAb LY5.1, or mouse IgG1. C: Dose-dependent (in a 48 hr culture), and D, time-dependent response of β2M-specific mAb-induced apoptosis (50 μg/ml mAbs) in myeloma cells induced by B2, LY5.1, and mouse IgG1. Results of four independent experiments using myeloma cell line RPMI-8226 are shown. Similar results were obtained with other myeloma cell lines. E: Induction of apoptosis, detected by Annexin V binding assay, in primary myeloma cells from eight patients (P1–P8) with multiple myeloma in medium or induced by 50 μg/ml B2 or D1 β2M-specific mAbs in 24 hr cultures. Results of three experiments are shown. Similar results were obtained with TUNEL assay. ∗p < 0.05; ∗∗p < 0.01. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 2 Killing of myeloma, but not normal hematopoietic cells, by β2M-specific mAbs Effect of β2M-specific mAbs on primary myeloma cells and normal osteoclasts in their cocultures. A: Viable myeloma cells; B, percentage of apoptotic myeloma cells; and C, viability of osteoclasts in the cocultures with the addition of 50 μg/ml B2 mAb or mouse IgG1 for 48 hr. Cocultures with medium alone served as control. Details are described in Experimental Procedures. Effect of β2M-specific mAbs on myeloma cell lines and normal PBMCs in their cocultures: D, Apoptotic myeloma cells, and E, apoptotic PBMCs and lymphocyte subsets in their cocultures, in which PBMCs (106/ml) were seeded on wells and myeloma cells (0.2 × 106/ml) were placed on inserts. β2M-specific mAbs (50 μg/ml) were added to the wells and incubated for 48 hr. Cells were cultured in RPMI-1640 supplemented with 10% fetal calf serum. No lymphocyte growth factors were added. After culture, cells were recovered and apoptotic cells were detected using the Annexin V binding assay. To examine apoptotic lymphocyte subsets, PE-conjugated CD3-, CD19-, and CD16-specific antibodies were used to identify the subsets, while FITC-conjugated Annexin V was used to identify apoptotic cells. Shown are the results of three experiments using D1 mAb on MM.1S. Similar results were obtained with ARP-1 and B2 mAb. F: Detection of apoptosis in lymphocytes (Lym; both resting and PHA-activated), and G, bone marrow-derived stem cells in cultures with the mAbs. Experiments were performed with 50 μg/ml B2 or D1 mAbs in 48 hr cultures. Results of four independent experiments are shown. ∗p < 0.05. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 3 Selectivity and mechanism of β2M-specific mAb-induced cell death Expression and modulation of β2M and HLA-ABC on myeloma cells by β2M-specific mAbs. A: Flow cytometry examining the levels of surface β2M and HLA-ABC molecules on myeloma cell lines (cell lines, n = 8), primary myeloma cells (MM cells, n = 6), and peripheral blood monocytes and lymphocytes from healthy donors (n = 6). B: Staining for β2M in bone marrow biopsies of patients with myeloma by immunohistochemistry, which showed that only myeloma plasma cells, but not normal hematopoietic cells, were stained by β2M-specific antibody. Scale bar, 20 μM. The insert shows a higher magnification (×1200). C: Effects of soluble β2M on β2M-specific mAb (B2 and D1)-induced apoptosis in myeloma cells, and D, effects of soluble β2M on β2M-specific mAb (B2) binding to surface β2M. In these experiments, equal amounts, unless otherwise indicated, of β2M-specific mAbs and soluble β2M were preincubated to form immune complexes, followed by the addition of myeloma cells (RPMI-8226; similar results were obtained with other cells) to the culture. Apoptosis was detected after cells were cultured 24 hr at 37°C by Annexin V binding assay, and surface staining was performed after 30 min incubation (on ice) by washing and incubating the cells with FITC-conjugated antibody to mouse IgG. Cells preincubated with B2 mAb alone (B2) served as control for surface binding of the mAb. Representative results of five independent experiments are shown. E: knockdown of surface β2M by specific siRNA abrogated β2M-specific mAb-induced apoptosis in myeloma cells. Shown are the results of two myeloma cell lines, MM.1S and ARP-1. The upper panels show the reduction of surface β2M, and middle panels show the level of HLA-DR on myeloma cells detected by flow cytometry. Numbers above the histograms represent mean fluorescence intensity. Filled blue histograms represent cells stained with control IgG, and open histograms show the levels of β2M expression on cells treated with mock siRNA (green) or transfected with control siRNA (blue) or β2M-specific siRNA (red). Assay was performed 72 hr after transfection. Representative results of three independent experiments are shown. The lower panels show percentages of apoptotic cells induced by β2M-specific mAb D1. Similar results were obtained with B2 mAb. In these experiments, cells were transfected with 400 nM siRNA, and 72 hr later, washed and incubated with 50 μg/ml β2M-specific mAbs for another 48 hr. Apoptosis was detected by Annexin V binding assay. F: Immunofluorescence staining for extracellular and intracellular localization of β2M-specific (B2) mAb/MHC class I molecules, visualized by FITC-conjugated antibody to mouse IgG1. Scale bar, 10 μM. G: Staining for surface HLA-ABC molecules on myeloma cells in cultures (at different time points) with the addition of B2 or D1 β2M-specific mAbs. Medium or mouse IgG1 served as controls. Representative results of three independent experiments are shown. ∗p < 0.05; ∗∗p < 0.01. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 4 β2M-specific mAb-induced apoptosis and signaling pathways A: Western blot analysis showing protein levels of procaspase and cleaved caspases and PARP in β2M-specific mAb (D1)-treated myeloma cells. Inhibition of caspase and PARP activation and processing (B) and apoptosis (C) is shown in β2M-specific mAb (D1)-treated myeloma cells with the addition of the pan-caspase inhibitor Z-VAD or caspase-9 inhibitor Z-LEHD. Arrows indicate cleaved (c) caspases and PARP. D: Western blot analysis showing protein levels of mitochondria-related proapoptotic and antiapoptotic proteins (cCyto C: cytosolic cytochrome c; tCyto C: total cytochrome c), and E, phosphorylated (p) and nonphosphorylated JNK, Akt, and ERK in β2M-specific mAb (D1)-treated myeloma cells. Results obtained with D1 mAb on MM.1S myeloma cells from one representative experiment of four performed are shown. Similar results are obtained with other β2M-specific mAbs on other myeloma cell lines as well as primary myeloma cells. JNK inhibitor II abrogated (F) β2M-specific mAb-induced JNK activation, and G, apoptosis in myeloma cells. Medium control contained DMSO. Results obtained with B2 or D1 mAb on MM.1S and/or ARP-1 myeloma cells from one representative experiment out of four performed are shown. Similar results are obtained with other β2M-specific mAbs on this and other myeloma cell lines. ∗p < 0.05; ∗∗p < 0.01. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 5 Involvement of lipid rafts and kinases in β2M-specific mAb-induced apoptosis in tumor cells Localization of MHC class I, Lyn, and Syk in lipid rafts (fractions 2–5) or nonraft fractions (fractions 7–9) in A, myeloma cells, or B, normal B cells after treatment with mouse IgG1 (left panels) or β2M-specific mAbs (right panels). Lipid-raft fractions were confirmed by positive staining for caveolin-1, a raft-associated protein. C: Methyl-β-cyclodextrin (MCD) inhibits β2M-specific mAb-induced apoptosis of myeloma cells. In these experiments, cells were preincubated with MCD (5 mM, titrated in preliminary experiments) for 30 min, washed, and incubated further with or without β2M-specific mAbs D1 or B2 (50 μg/ml). Apoptosis was measured at 48 hr by the Annexin V binding assay. Immunoprecipitation using antibodies against D, MHC class I; E, Lyn; or F, PLCγ2 in myeloma cells treated with IgG1 or the mAbs (D1 or B2). Blotting antibodies used were against MHC class I, Lyn, Syk, and PLCγ2 (D), MHC class I and Lyn (E), or MHC class I and PLCγ2 (F). G: Western blot showing phosphorylated Lyn (pLyn) and pPLCγ2 in myeloma cells after treatment with β2M-specific mAbs. Nonphosphorylated kinases were also shown. Results obtained with D1 mAb on MM.1S and/or ARP-1 myeloma cells from one representative experiment out of four performed are shown. Similar results are obtained with other tumor cell lines and primary myeloma cells. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 6 In vivo therapeutic effects of β2M-specific mAbs on established human myeloma and other hematological tumors in SCID or SCID-hu mouse models SCID mice were xenografted subcutaneously with ARP-1, MM.1S, CA-46, Granta 519, or KG1 cells, and tumor burdens were monitored as tumor volumes (upper panels; A for myeloma cells, and B for other tumor cells). Mice received intraperitoneal injections (every 3 days for a total of four injections) of 250 μl ascites containing about 500 μg D1 β2M-specific mAb or 500 μg mouse IgG1, or an equal volume of PBS. Survival of SCID mice is shown in lower panels; A for myeloma cells, and B for other tumor cells (survival times: β2M-specific mAb group versus PBS or IgG1 groups: p < 0.01 in all models). Results from one representative experiment with five mice per group of three performed using D1 mAb are shown. Similar results are obtained with another β2M-specific mAb (E6) on these two myeloma cell lines. In SCID-hu mice (six mice per group), primary myeloma cells from patients (n = 3) were directly injected into implanted human bones, and tumor growth was monitored as levels of circulating human M-protein or its light chain (A, far right; Primary MM). Mice received the same treatments as SCID. Note: because primary myeloma cells only grow in implanted human bone and not in murine bone marrow or other organs, these mice usually do not develop systemic symptoms and become moribund; therefore, no death data were available. ∗p < 0.05; ∗∗p < 0.01. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 7 In vivo validation of signaling and apoptosis mechanisms A: Myeloma (MM.1S)-bearing mice were treated as previously indicated, sacrificed, and had tumors removed for immunohistochemical staining for phosphorylated JNK (pJNK), cleaved caspase-9 (cCaspase-9), and apoptotic cells by in situ TUNEL assay. Shown are staining of tumors after one treatment with intraperitoneal injection of the mAbs. In some experiments, JNK inhibitor (JNK inhibitor II, 20 μM) was injected around the tumors before injecting the mAbs. Scale bar, 20 μM. B: Effect of the JNK inhibitor in abrogating the therapeutic efficacy of β2M mAbs on established myeloma (MM.1S) in SCID mice. PBS contained DMSO. It is evident that the JNK inhibitor at ≥10 μM was effective. Representative results of three experiments are shown. The same results were also obtained with other tumor cells lines in SCID mice. Error bars = SEM. Cancer Cell 2006 10, 295-307DOI: (10.1016/j.ccr.2006.08.025) Copyright © 2006 Elsevier Inc. Terms and Conditions