Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zev A. Binder is active.

Publication


Featured researches published by Zev A. Binder.


Journal of Neuro-oncology | 2017

SHP2 regulates proliferation and tumorigenicity of glioma stem cells

Laura Roccograndi; Zev A. Binder; Logan Zhang; Nicola Aceto; Zhuo Zhang; Mohamed Bentires-Alj; Ichiro Nakano; Nadia Dahmane; Donald M. O’Rourke

SHP2 is a cytoplasmic protein tyrosine phosphatase (PTPase) involved in multiple signaling pathways and was the first identified proto-oncogene PTPase. Previous work in glioblastoma (GBM) has demonstrated the role of SHP2 PTPase activity in modulating the oncogenic phenotype of adherent GBM cell lines. Mutations in PTPN11, the gene encoding SHP2, have been identified with increasing frequency in GBM. Given the importance of SHP2 in developing neural stem cells, and the importance of glioma stem cells (GSCs) in GBM oncogenesis, we explored the functional role of SHP2 in GSCs. Using paired differentiated and stem cell primary cultures, we investigated the association of SHP2 expression with the tumor stem cell compartment. Proliferation and soft agar assays were used to demonstrate the functional contribution of SHP2 to cell growth and transformation. SHP2 expression correlated with SOX2 expression in GSC lines and was decreased in differentiated cells. Forced differentiation of GSCs by removal of growth factors, as confirmed by loss of SOX2 expression, also resulted in decreased SHP2 expression. Lentiviral-mediated knockdown of SHP2 inhibited proliferation. Finally, growth in soft-agar was similarly inhibited by loss of SHP2 expression. Our results show that SHP2 function is required for cell growth and transformation of the GSC compartment in GBM.


Scientific Reports | 2018

Radiomic MRI signature reveals three distinct subtypes of glioblastoma with different clinical and molecular characteristics, offering prognostic value beyond IDH1

Saima Rathore; Hamed Akbari; Martin Rozycki; Kalil G. Abdullah; MacLean P. Nasrallah; Zev A. Binder; Ramana V. Davuluri; Robert A. Lustig; Nadia Dahmane; Michel Bilello; Donald M. O’Rourke; Christos Davatzikos

The remarkable heterogeneity of glioblastoma, across patients and over time, is one of the main challenges in precision diagnostics and treatment planning. Non-invasive in vivo characterization of this heterogeneity using imaging could assist in understanding disease subtypes, as well as in risk-stratification and treatment planning of glioblastoma. The current study leveraged advanced imaging analytics and radiomic approaches applied to multi-parametric MRI of de novo glioblastoma patients (nu2009=u2009208 discovery, nu2009=u200953 replication), and discovered three distinct and reproducible imaging subtypes of glioblastoma, with differential clinical outcome and underlying molecular characteristics, including isocitrate dehydrogenase-1 (IDH1), O6-methylguanine–DNA methyltransferase, epidermal growth factor receptor variant III (EGFRvIII), and transcriptomic subtype composition. The subtypes provided risk-stratification substantially beyond that provided by WHO classifications. Within IDH1-wildtype tumors, our subtypes revealed different survival (pu2009<u20090.001), thereby highlighting the synergistic consideration of molecular and imaging measures for prognostication. Moreover, the imaging characteristics suggest that subtype-specific treatment of peritumoral infiltrated brain tissue might be more effective than current uniform standard-of-care. Finally, our analysis found subtype-specific radiogenomic signatures of EGFRvIII-mutated tumors. The identified subtypes and their clinical and molecular correlates provide an in vivo portrait of phenotypic heterogeneity in glioblastoma, which points to the need for precision diagnostics and personalized treatment.


SLAS TECHNOLOGY: Translating Life Sciences Innovation | 2018

Mutation Profiles in Glioblastoma 3D Oncospheres Modulate Drug Efficacy

Kelli Wilson; Lesley Mathews-Griner; Tara Williamson; Rajarshi Guha; Lu Chen; Paul Shinn; Crystal McKnight; Sam Michael; Carleen Klumpp-Thomas; Zev A. Binder; Marc Ferrer; Gary L. Gallia; Craig J. Thomas; Gregory J. Riggins

Glioblastoma (GBM) is a lethal brain cancer with a median survival time of approximately 15 months following treatment. Common in vitro GBM models for drug screening are adherent and do not recapitulate the features of human GBM in vivo. Here we report the genomic characterization of nine patient-derived, spheroid GBM cell lines that recapitulate human GBM characteristics in orthotopic xenograft models. Genomic sequencing revealed that the spheroid lines contain alterations in GBM driver genes such as PTEN, CDKN2A, and NF1. Two spheroid cell lines, JHH-136 and JHH-520, were utilized in a high-throughput drug screen for cell viability using a 1912-member compound library. Drug mechanisms that were cytotoxic in both cell lines were Hsp90 and proteasome inhibitors. JHH-136 was uniquely sensitive to topoisomerase 1 inhibitors, while JHH-520 was uniquely sensitive to Mek inhibitors. Drug combination screening revealed that PI3 kinase inhibitors combined with Mek or proteasome inhibitors were synergistic. However, animal studies to test these drug combinations in vivo revealed that Mek inhibition alone was superior to the combination treatments. These data show that these GBM spheroid lines are amenable to high-throughput drug screening and that this dataset may deliver promising therapeutic leads for future GBM preclinical studies.


Molecular Therapy - Oncolytics | 2018

Checkpoint Blockade Reverses Anergy in IL-13Rα2 Humanized scFv-Based CAR T Cells to Treat Murine and Canine Gliomas

Yibo Yin; Alina C. Boesteanu; Zev A. Binder; Chong Xu; Reiss A. Reid; Jesse L. Rodriguez; Danielle R. Cook; Radhika Thokala; Kristin Blouch; Bevin McGettigan-Croce; Logan Zhang; Christoph Konradt; Alexandria P. Cogdill; M. Kazim Panjwani; Shuguang Jiang; Denis Migliorini; Nadia Dahmane; Avery D. Posey; Carl H. June; Nicola J. Mason; Zhiguo Lin; Donald M. O’Rourke; Laura A. Johnson

We generated two humanized interleukin-13 receptor α2 (IL-13Rα2) chimeric antigen receptors (CARs), Hu07BBz and Hu08BBz, that recognized human IL-13Rα2, but not IL-13Rα1. Hu08BBz also recognized canine IL-13Rα2. Both of these CAR T cell constructs demonstrated superior tumor inhibitory effects in a subcutaneous xenograft model of human glioma compared with a humanized EGFRvIII CAR T construct used in a recent phase 1 clinical trial (ClinicalTrials.gov: NCT02209376). The Hu08BBz demonstrated a 75% reduction in orthotopic tumor growth using low-dose CAR T cell infusion. Using combination therapy with immune checkpoint blockade, humanized IL-13Rα2 CAR T cells performed significantly better when combined with CTLA-4 blockade, and humanized EGFRvIII CAR T cells’ efficacy was improved by PD-1 and TIM-3 blockade in the same mouse model, which was correlated with the levels of checkpoint molecule expression in co-cultures with the same tumor in vitro. Humanized IL-13Rα2 CAR T cells also demonstrated benefit from a self-secreted anti-CTLA-4 minibody in the same mouse model. In addition to a canine glioma cell line (J3T), canine osteosarcoma lung cancer and leukemia cell lines also express IL-13Rα2 and were recognized by Hu08BBz. Canine IL-13Rα2 CAR T cell was also generated and tested in vitro by co-culture with canine tumor cells and in vivo in an orthotopic model of canine glioma. Based on these results, we are designing a pre-clinical trial to evaluate the safety of canine IL-13Rα2 CAR T cells in dog with spontaneous IL-13Rα2-positive glioma, which will help to inform a human clinical trial design for glioblastoma using humanized scFv-based IL-13Rα2 targeting CAR T cells.


Cancer Cell | 2018

Epidermal Growth Factor Receptor Extracellular Domain Mutations in Glioblastoma Present Opportunities for Clinical Imaging and Therapeutic Development

Zev A. Binder; Amy Haseley Thorne; Spyridon Bakas; E. Paul Wileyto; Michel Bilello; Hamed Akbari; Saima Rathore; Sung Min Ha; Logan Zhang; Cole J. Ferguson; Sonika Dahiya; Wenya Linda Bi; David A. Reardon; Ahmed Idbaih; Joerg Felsberg; Bettina Hentschel; Michael Weller; Stephen J. Bagley; Jennifer J.D. Morrissette; MacLean P. Nasrallah; Jianhui Ma; Ciro Zanca; Andrew M. Scott; Laura Orellana; Christos Davatzikos; Frank Furnari; Donald M. O'Rourke

We explored the clinical and pathological impact of epidermal growth factor receptor (EGFR) extracellular domain missense mutations. Retrospective assessment of 260 de novo glioblastoma patients revealed a significant reduction in overall survival of patients having tumors with EGFR mutations at alanine 289 (EGFRA289D/T/V). Quantitative multi-parametric magnetic resonance imaging analyses indicated increased tumor invasion for EGFRA289D/T/V mutants, corroborated in mice bearing intracranial tumors expressing EGFRA289V and dependent on ERK-mediated expression of matrix metalloproteinase-1. EGFRA289V tumor growth was attenuated with an antibody against a cryptic epitope, based on in silico simulation. The findings of this study indicate a highly invasive phenotype associated with the EGFRA289V mutation in glioblastoma, postulating EGFRA289V as a molecular marker for responsiveness to therapy with EGFR-targeting antibodies.


Neuro-oncology | 2016

MPTH-02. EXTRACELLULAR EGFR289 ACTIVATING MUTATIONS CONFER POORER SURVIVAL AND SUGGEST ENHANCED MOTILITY IN PRIMARY GBMs

Zev A. Binder; Spyridon Bakas; E. Paul Wileyto; Hamed Akbari; Saima Rathore; Martin Rozycki; Jennifer J.D. Morrissette; Maria Martinez-Lage; Nadia Dahmane; Christos Davatzikos; Donald M. O’Rourke


Neuro-oncology | 2016

NIMG-11. HIGHLY-EXPRESSED WILD-TYPE EGFR AND EGFRvIII MUTANT GLIOBLASTOMAS HAVE SIMILAR MRI SIGNATURE, CONSISTENT WITH DEEP PERITUMORAL INFILTRATION

Spyridon Bakas; Zev A. Binder; Hamed Akbari; Maria Martinez-Lage; Martin Rozycki; Jennifer J.D. Morrissette; Nadia Dahmane; Donald M. O’Rourke; Christos Davatzikos


Neuro-oncology | 2017

NIMG-07. UNIFYING MAGNETIC RESONANCE IMAGING SIGNATURE OF EGFR PATHWAY ACTIVATION IN GLIOBLASTOMA CONSISTENT WITH UNIFORMLY AGGRESSIVELY INFILTRATION

Spyridon Bakas; Zev A. Binder; Jennifer J.D. Morrissette; Hamed Akbari; Donald M. O’Rourke; Christos Davatzikos


Neuro-oncology | 2017

NIMG-59. RADIOLOGIC SUBTYPES OF GLIOBLASTOMA CALCULATED VIA MULTI-PARAMETRIC IMAGING SIGNATURES REVEAL COMPLEMENTARY INFORMATION TO CURRENT WHO CLASSIFICATION

Saima Rathore; Hamed Akbari; Martin Rozycki; Kalil Abdullah; MacLean P. Nasrallah; Zev A. Binder; Robert H. Lustig; Nadia Dahmane; Michel Bilello; Donald M. O’Rourke; Christos Davatzikos


Cancer Research | 2018

Abstract LB-340: Combinatorial platform for CART cell therapy for glioblastoma

Zev A. Binder; Yibo Yin; Radhika Thokala; Donald M. O'Rourke

Collaboration


Dive into the Zev A. Binder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hamed Akbari

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saima Rathore

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Spyridon Bakas

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Rozycki

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge