EPHB4 is a therapeutic target in AML and promotes leukemia cell survival via AKT by Akil A. Merchant, Aparna Jorapur, Amy McManus, Ren Liu, Valery Krasnoperov,

Slides:



Advertisements
Similar presentations
An aptamer-based targeted delivery of miR-26a protects mice against chemotherapy toxicity while suppressing tumor growth by Toshihiko Tanno, Peng Zhang,
Advertisements

High TCL1 levels are a marker of B-cell receptor pathway responsiveness and adverse outcome in chronic lymphocytic leukemia by Marco Herling, Kaushali.
Combination therapy for adult T-cell leukemia–xenografted mice: flavopiridol and anti-CD25 monoclonal antibody by Meili Zhang, Zhuo Zhang, Carolyn K. Goldman,
High-Risk Acute Lymphoblastic Leukemia Cells with bcr-abl and Ink4a/Arf Mutations Retain Susceptibility to Alloreactive T Cells  Faith M. Young, Andrew.
Protein kinase B (PKB/c-akt) regulates homing of hematopoietic progenitors through modulation of their adhesive and migratory properties by Miranda Buitenhuis,
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 Lianne van de Laar, Aniek van den Bosch, André Boonstra, Rekha S
Characterization of a humanized IgG4 anti-HLA-DR monoclonal antibody that lacks effector cell functions but retains direct antilymphoma activity and increases.
The TCL1 oncoprotein inhibits activation-induced cell death by impairing PKCθ and ERK pathways by Gilles Despouy, Marjorie Joiner, Emilie Le Toriellec,
Rapid and selective death of leukemia stem and progenitor cells induced by the compound 4-benzyl, 2-methyl, 1,2,4-thiadiazolidine, 3,5 dione (TDZD-8)‏
Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment by Francis Mussai, Carmela De Santo, Issa Abu-Dayyeh, Sarah Booth,
by Rafijul Bari, Teresa Bell, Wai-Hang Leung, Queenie P
Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas by Pankaj Gupta,
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 Daniel Sasca, Patricia S
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.
Sustained signaling through the B-cell receptor induces Mcl-1 and promotes survival of chronic lymphocytic leukemia B cells by Aleksandar Petlickovski,
Suppression of Fas-FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo by Ying Liu, Ramona.
Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy by Mark P. Chao, Chad Tang, Russell K. Pachynski,
AKR1C3 is a biomarker of sensitivity to PR-104 in preclinical models of T-cell acute lymphoblastic leukemia by Donya Moradi Manesh, Jad El-Hoss, Kathryn.
Improving T-cell expansion and function for adoptive T-cell therapy using ex vivo treatment with PI3Kδ inhibitors and VIP antagonists by Christopher T.
Preactivation with IL-12, IL-15, and IL-18 Induces CD25 and a Functional High-Affinity IL-2 Receptor on Human Cytokine-Induced Memory-like Natural Killer.
by Takahiro Kamiya, Desmond Wong, Yi Tian Png, and Dario Campana
Attenuation of AML1-ETO cellular dysregulation correlates with increased leukemogenic potential by Russell C. DeKelver, Ming Yan, Eun-Young Ahn, Wei-Jong.
TLR5 signaling in murine bone marrow induces hematopoietic progenitor cell proliferation and aids survival from radiation by Benyue Zhang, Damilola Oyewole-Said,
A potent tetravalent T-cell–engaging bispecific antibody against CD33 in acute myeloid leukemia by Sayed Shahabuddin Hoseini, Hongfen Guo, Zhihao Wu, Miho.
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,
Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15 by Lazar.
Generation of Highly Cytotoxic Natural Killer Cells for Treatment of Acute Myelogenous Leukemia Using a Feeder-Free, Particle-Based Approach  Jeremiah.
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,
C-FLIP confers resistance to FAS-mediated apoptosis in anaplastic large-cell lymphoma by Mauricio P. Oyarzo, L. Jeffrey Medeiros, Coralyn Atwell, Marianna.
Stimulation of the B-cell receptor activates the JAK2/STAT3 signaling pathway in chronic lymphocytic leukemia cells by Uri Rozovski, Ji Yuan Wu, David.
Selinexor is effective in acquired resistance to ibrutinib and synergizes with ibrutinib in chronic lymphocytic leukemia by Zachary A. Hing, Rose Mantel,
Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival by Kyu-Tae Kim, Kristin Baird, Joon-Young Ahn, Paul.
Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia by Parvathi Ranganathan, Xueyan Yu, Caroline Na, Ramasamy Santhanam, Sharon Shacham,
Selective Purging of Human Multiple Myeloma Cells from Autologous Stem Cell Transplantation Grafts using Oncolytic Myxoma Virus  Eric Bartee, Winnie M.
An aptamer-based targeted delivery of miR-26a protects mice against chemotherapy toxicity while suppressing tumor growth by Toshihiko Tanno, Peng Zhang,
Crosstalk between ROR1 and BCR pathways defines novel treatment strategies in mantle cell lymphoma by Hanna Karvonen, David Chiron, Wilhelmiina Niininen,
T Cell and B Cell Immunity can be Reconstituted with Mismatched Hematopoietic Stem Cell Transplantation Without Alkylator Therapy in Artemis-Deficient.
by Kamira Maharaj, John J
Volume 24, Issue 9, Pages (September 2016)
Imetelstat, a telomerase inhibitor, is capable of depleting myelofibrosis stem and progenitor cells by Xiaoli Wang, Cing Siang Hu, Bruce Petersen, Jiajing.
The kinases IKBKE and TBK1 regulate MYC-dependent survival pathways through YB-1 in AML and are targets for therapy by Suhu Liu, Anna E. Marneth, Gabriela.
by Kalpana Parvathaneni, and David W. Scott
Multiple distinct molecular mechanisms influence sensitivity and resistance to MDM2 inhibitors in adult acute myelogenous leukemia by Jianting Long, Brian.
Volume 25, Issue 9, Pages (September 2017)
Agonistic targeting of TLR1/TLR2 induces p38 MAPK-dependent apoptosis and NFκB-dependent differentiation of AML cells by Mia Eriksson, Pablo Peña-Martínez,
MLL-AF9 leukemias are sensitive to PARP1 inhibitors combined with cytotoxic drugs by Silvia Maifrede, Esteban Martinez, Margaret Nieborowska-Skorska, Daniela.
Myeloma cell–derived Runx2 promotes myeloma progression in bone
T exosomes bind MAdCAM-1 via RA-increased integrin α4β7.
Volume 38, Issue 3, Pages (March 2013)
Paradoxical enhancement of leukemogenesis in acute myeloid leukemia with moderately attenuated RUNX1 expressions by Ken Morita, Shintaro Maeda, Kensho.
Volume 8, Issue 4, Pages (October 2005)
by Yue Wei, Hong Zheng, Naran Bao, Shan Jiang, Carlos E
Targeting ubiquitin-activating enzyme induces ER stress–mediated apoptosis in B-cell lymphoma cells by Scott Best, Taylor Hashiguchi, Adam Kittai, Nur.
A phase 1 trial evaluating thioridazine in combination with cytarabine in patients with acute myeloid leukemia by Lili Aslostovar, Allison L. Boyd, Mohammed.
BRAFV600E accelerates disease progression and enhances immune suppression in a mouse model of B-cell leukemia by Yo-Ting Tsai, Aparna Lakshmanan, Amy Lehman,
AML cells display differential sensitivity to inhibition of IKBKE and TBK1. AML cells display differential sensitivity to inhibition of IKBKE and TBK1.
Complement C5 but not C3 is expendable for tissue factor activation by cofactor-independent antiphospholipid antibodies by Nadine Müller-Calleja, Svenja.
Defective RAB1B-related megakaryocytic ER-to-Golgi transport in RUNX1 haplodeficiency: impact on von Willebrand factor by Gauthami Jalagadugula, Lawrence.
by Pamela J. Sung, Mayumi Sugita, Holly Koblish, Alexander E
Volume 21, Issue 4, Pages (April 2013)
The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells.
by Samuel J. Taylor, Johanna M. Duyvestyn, Samantha A. Dagger, Emma J
by Fabian C. Verbij, Nicoletta Sorvillo, Paul H. P
Volume 25, Issue 9, Pages (September 2017)
SY-1425 induces maturation in RARA-high AML
B-cell development in young Pax5+/− mice.
Presentation transcript:

EPHB4 is a therapeutic target in AML and promotes leukemia cell survival via AKT by Akil A. Merchant, Aparna Jorapur, Amy McManus, Ren Liu, Valery Krasnoperov, Parvesh Chaudhry, Mohan Singh, Lisa Harton, Mary Agajanian, Miriam Kim, Timothy J. Triche, Brian J. Druker, Jeffrey W. Tyner, and Parkash S. Gill BloodAdv Volume 1(20):1635-1644 September 12, 2017 © 2017 by The American Society of Hematology

Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology

EPHB4 is expressed in AML EPHB4 is expressed in AML. (A) Representative plot of 1 primary AML sample that screened positive for sensitivity to RNAi against EPHB4. EPHB4 is expressed in AML. (A) Representative plot of 1 primary AML sample that screened positive for sensitivity to RNAi against EPHB4. This particular sample had a validated mutation in NRAS, which serves as a positive control, because AML cells demonstrate similar sensitivity to RNAi against NRAS as they do against EPHB4. *P < .05. (B) Cell surface EPHB4 expression in myeloid leukemia cell lines as measured by flow cytometry. The IgG isotype used to determine negative population is in gray. (C) Cell surface EPHB4 expression on primary AML blasts representing high (AML5), moderate (AML11), and no (AML16) expression as measured by flow cytometry. (D) Summary of EPHB4 expression on primary AML (n = 25) and normal, human CD34+ bone marrow cells obtained from healthy donors. Expression was measured by flow cytometry, and MFI was calculated and divided by the MFI of negative control. Results are plotted on log scale, therefore the expression ratio of 1 corresponds to no expression. The bar represents average MFI. P = .0012 Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology

EPHB4 in AML can be targeted with a monoclonal antibody (MAb131). EPHB4 in AML can be targeted with a monoclonal antibody (MAb131). (A) EPHB4+ leukemia cells (K562 and Molm14) and EPHB4– (KG1) cells treated with 1 dose of MAb131 at time 0 and cultured under normal conditions. Live cells were counted by trypan blue exclusion at specified time points. MAb131 treatment reduced cell growth in EPHB4+ cells only. The average counts from 3 separate experiments are displayed, and error bars represent standard error of the mean. P values were calculated at 72 hours by pairwise comparison between control IgG-treated and MAb131-treated cells. *P < .01. (B) MAb131 induces apoptosis in EPHB4+ leukemia cells. K562 cells were treated with MAb131 (1, 10, and 100 μg/mL) and assayed for apoptosis by Annexin V/PI staining. The percentage of cells live (double negative), early apoptotic (Annexin V+/PI+), and late apoptotic/dead (Annexin V+/PI+) are noted. (C) K562 cells were treated with MAb131 and harvested for protein analysis by immunoblot (top) or by flow cytometry (bottom). MAb131 treatment causes loss of cell surface EPHB4 and degradation of total EPHB4 in cells. (D) K562 cells were treated with Alexa 488–labeled MAb131 and incubated at 37°C and 4°C for 1 hour, followed by imaging (40× objective). Incubation at 4°C (top) prevents the internalization and degradation of EPHB4, as evidenced by the bright cell surface staining, whereas incubation at 37°C (bottom) leads to internalization of labeled antibody and decreased overall staining. (E) EPHB4+ primary AML blasts (n = 3) were treated with MAb131. Live cells were counted on a hemacytometer by trypan blue exclusion at 24 and 48 hours and normalized to untreated controls. Treatment with MAb131 lead to increased cell death in primary AML samples. (F) Primary AML samples were treated with MAb131 and show downregulation of cell surface EPHB4 as measured by flow cytometry. (G) K562 cells were treated with increasing concentrations of soluble EPHB4, which blocks the cytotoxic effect of MAb131, confirming that MAb131 effects are due to EPHB4 binding and not off-target effects. Cells were treated and live cells counted by trypan blue exclusion at 72 hours. The average counts from 3 separate experiments are displayed, and error bars represent the standard error of the mean. P values for selected pairwise comparison are noted on the chart. Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology

EPHB4 promotes AML survival through AKT signaling. EPHB4 promotes AML survival through AKT signaling. (A) K562 cells were transduced with siRNA against EPHB4 and analyzed for EPHB4, pAKT, and total AKT at 48 hours. pAKT levels were reduced in EPHB4 knockdown cells as measured by immunoblotting, whereas total AKT levels were unchanged. (B) K562 cells were co-transduced with siRNA against EPHB4 and/or the constitutively active form of AKT (myr-AKT). Expression of myr-AKT restores cell growth and survival after EPHB4 knockdown. The average counts from 3 separate experiments are displayed, and error bars represent the standard error of the mean. (C) Levels of AKT activation in cells treated in Figure 3B as measured by phosphor-flow for pAKT. (D) K562 cells were treated with MAb131, and pAKT was measured by flow cytometry at various time points. At 30 minutes after treatment, a transient increase in pAKT was observed due to EPHB4 activation by MAb131, followed by a decrease in pAKT as EPHB4 activity was lost. (E) Primary AML blasts (unstimulated) treated with MAb131 show decreased pAKT after treatment with MAb131. Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology

MAb131 demonstrates in vivo activity against primary AML MAb131 demonstrates in vivo activity against primary AML. (A) Immunodeficient (NSG) mice were injected with 1 × 106 EPHB4+ primary AML cells (AML18) to create orthotopic xenografts. MAb131 demonstrates in vivo activity against primary AML. (A) Immunodeficient (NSG) mice were injected with 1 × 106 EPHB4+ primary AML cells (AML18) to create orthotopic xenografts. AML18 was characterized by normal cytogenetics, CEBPA mutant, and NPM1 mutation; 5 to 6 NSG mice per treatment group. Two weeks after injection, treatment mice were treated with PBS (untreated), AraC (50 mg/kg IP weekly), MAb131 10 mg/kg 3 times per week, or the combination. Treatments continued until all control mice were killed for evidence of disease. Remaining mice were observed for survival. Engraftment was confirmed at necropsy. (B) The table predicts median survival, range, and adjusted P values, after Bonferroni correction, for each survival curve. Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology

EPHB4 expression is driven by mutations found in AML EPHB4 expression is driven by mutations found in AML. (A) Treatment with inhibitor of BCR-ABL (imatinib) led to a decrease of EPHB4 expression on the surface of bcr-ABL–containing K562 cells lines, with no change in ML-2 cells, which had wild-type (wt) ABL. (B) Treatment with the specific FLT3 inhibitor, quizartinib led to a decrease of EPHB4 expression in FLT3-ITD–containing MV-4-11 and MOLM-14 cells, but no change in FLT3-wt cells. EPHB4 expression is driven by mutations found in AML. (A) Treatment with inhibitor of BCR-ABL (imatinib) led to a decrease of EPHB4 expression on the surface of bcr-ABL–containing K562 cells lines, with no change in ML-2 cells, which had wild-type (wt) ABL. (B) Treatment with the specific FLT3 inhibitor, quizartinib led to a decrease of EPHB4 expression in FLT3-ITD–containing MV-4-11 and MOLM-14 cells, but no change in FLT3-wt cells. (C) Treatment with the JAK inhibitor ruxolitinib resulted in a decrease of EPHB4 expression on JAK2V617F-containing HEL cells, with no change in JAK2-wt controls. Akil A. Merchant et al. Blood Adv 2017;1:1635-1644 © 2017 by The American Society of Hematology