Network


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

Hotspot


Dive into the research topics where Michael E. Barish is active.

Publication


Featured researches published by Michael E. Barish.


Clinical Cancer Research | 2015

Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma

Christine E. Brown; Behnam Badie; Michael E. Barish; Lihong Weng; Julie R. Ostberg; Wen-Chung Chang; Araceli Naranjo; Renate Starr; Jamie Wagner; Christine Wright; James R. Bading; Julie A. Ressler; Jana Portnow; Massimo D'Apuzzo; Stephen J. Forman; Michael C. Jensen

Purpose: A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)–engineered, autologous primary human CD8+ cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). Experimental Design: Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8+ CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 108 CAR-engineered T cells via a catheter/reservoir system. Results: We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine+ CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine+ CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine+ T-cell administration. Conclusions: These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied. Clin Cancer Res; 21(18); 4062–72. ©2015 AACR.


Clinical Cancer Research | 2012

Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells.

Christine E. Brown; Renate Starr; Brenda Aguilar; Andrew F. Shami; Catalina Martinez; Massimo D'Apuzzo; Michael E. Barish; Stephen J. Forman; Michael C. Jensen

Purpose: To evaluate IL13Rα2 as an immunotherapeutic target for eliminating glioma stem–like cancer initiating cells (GSC) of high-grade gliomas, with particular focus on the potential of genetically engineered IL13Rα2-specific primary human CD8+ CTLs (IL13-zetakine+ CTL) to target this therapeutically resistant glioma subpopulation. Experimental Design: A panel of low-passage GSC tumor sphere (TS) and serum-differentiated glioma lines were expanded from patient glioblastoma specimens. These glioblastoma lines were evaluated for expression of IL13Rα2 and for susceptibility to IL13-zetakine+ CTL-mediated killing in vitro and in vivo. Results: We observed that although glioma IL13Rα2 expression varies between patients, for IL13Rα2pos cases this antigen was detected on both GSCs and more differentiated tumor cell populations. IL13-zetakine+ CTL were capable of efficient recognition and killing of both IL13Rα2pos GSCs and IL13Rα2pos differentiated cells in vitro, as well as eliminating glioma-initiating activity in an orthotopic mouse tumor model. Furthermore, intracranial administration of IL13-zetakine+ CTL displayed robust antitumor activity against established IL13Rα2pos GSC TS-initiated orthotopic tumors in mice. Conclusions: Within IL13Rα2 expressing high-grade gliomas, this receptor is expressed by GSCs and differentiated tumor populations, rendering both targetable by IL13-zetakine+ CTLs. Thus, our results support the potential usefullness of IL13Rα2-directed immunotherapeutic approaches for eradicating therapeutically resistant GSC populations. Clin Cancer Res; 18(8); 2199–209. ©2012 AACR.


Science Translational Medicine | 2013

Neural Stem Cell–Mediated Enzyme/Prodrug Therapy for Glioma: Preclinical Studies

Karen S. Aboody; Joseph Najbauer; Marianne Z. Metz; Massimo D'Apuzzo; Margarita Gutova; Alexander J. Annala; Timothy W. Synold; Larry A. Couture; Suzette Blanchard; Rex Moats; Elizabeth Garcia; Soraya Aramburo; Valenzuela Vv; Richard T. Frank; Michael E. Barish; Christine E. Brown; Seung U. Kim; Behnam Badie; Jana Portnow

Neural stem cells home to gliomas in mice where they convert a prodrug to 5-fluorouracil, leading to tumor regression. Cellular Assassins Derived from the supporting cells of the brain, gliomas are deadly tumors that can be only temporarily held at bay, but not cured. New ways to treat these cancers are needed. To get regulatory approval to test a new stem cell–based therapy in patients, Aboody et al. performed a series of preclinical experiments in mice with artificially implanted gliomas in their brains. By mimicking closely the treatments that they hoped to perform in humans, these authors were able to show to the satisfaction of the regulatory agency that the treatment was safe and effective enough in the mice to warrant a first-in-human trial in patients. The authors used a neural stem cell line carrying a v-myc gene and a gene for cytosine deaminase. These cells exhibit tropism to human glioma cells. When injected into mice with gliomas, they migrate to the site of the tumor, even when the mice are treated with steroids or radiation, as might be the case for human patients. The cytosine deaminase in the cells provides another anticancer weapon. This enzyme converts the prodrug 5-fluorocytosine (5-FC) to the toxic 5-flurouracil (5-FU), delivering a high concentration of the therapeutic agent directly in and around the tumor and causing it to shrink significantly. Injection of excess numbers of cells or increasing the dose of 5-FU did not result in any abnormalities in the animals; in fact, by 12 weeks after injection, no cells were to be seen in the brain or elsewhere, even when a highly sensitive polymerase chain reaction method was used to look for the v-myc DNA. This targeted cell-based approach to cancer therapy that concentrates the therapeutic agent in the vicinity of the tumor is expected to reduce toxicity to other tissues. Thus, a higher local dose is possible, potentially improving efficacy against the tumor. The phase 1 trial derived from these preclinical results is ongoing; its end will allow evaluation of how well these preclinical in vivo studies set the stage for humans. High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)–expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non–tumor-bearing and orthotopic glioma–bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


The Journal of Neuroscience | 1994

Perturbation of Intracellular Calcium and Hydrogen Ion Regulation in Cultured Mouse Hippocampal Neurons by Reduction of the Sodium Ion Concentration Gradient

Ra Koch; Michael E. Barish

Na(+)-Ca2+ exchange has been identified as a mechanism for regulation of intracellular Ca ion concentration ([Ca2+]i) in neurons of invertebrates and vertebrates, but for mammalian central neurons its role in restoration of resting [Ca2+]i after transient increases induced by stimulation has been less clear. We have examined the recovery of [Ca2+]i following K+ depolarization and glutamate receptor activation of cultured mouse hippocampal neurons using the Ca(2+)- sensitive dye Fura-2. Reduction of the transmembrane Na+ gradient by removal of external Na+ slowed the recovery of neurons from imposed Ca2+ loads. We observed that [Ca2+]i regulation was disrupted more severely when N-methyl-D-glucamine (N-MG), Tris, or choline rather than Li+ replaced external Na+. Additional disruption of intracellular pH regulation by substitutes other than Li+ may account for this difference. Measurement of [Ca2+]i and [H+]i (using the H(+)-sensitive dye BCECF) during glutamate receptor activation indicated that Ca2+ influx resulted in production of intracellular H+, and that Li+ but not N-MG could prevent cytoplasmic acidification on removal of external Na+. We also observed that intracellular acidification alone was sufficient to slow recovery from Ca2+ load. We conclude, therefore, that Na(+)-Ca2+ exchange contributes to recovery of [Ca2+]i after stimulation leading to Ca2+ entry into hippocampal neurons, and that Na(+)-H+ exchange limits the acidification (and secondary increase in [Ca2+]i) that accompanies Ca2+ influx. We suggest that because both Na(+)-Ca2+ and Na(+)-H+ exchangers will be compromised during ischemia and hypoglycemia, increased intracellular H+ may synergize with cytoplasmic Ca2+ to potentiate excitotoxic neuronal death.


Journal of Clinical Investigation | 2004

Enhanced neuronal excitability in the absence of neurodegeneration induces cerebellar ataxia

Vikram G. Shakkottai; Chin Hua Chou; Salvatore Oddo; Claudia A. Sailer; Hans-Günther Knaus; George A. Gutman; Michael E. Barish; Frank M. LaFerla; K. George Chandy

Cerebellar ataxia, a devastating neurological disease, may be initiated by hyperexcitability of deep cerebellar nuclei (DCN) secondary to loss of inhibitory input from Purkinje neurons that frequently degenerate in this disease. This mechanism predicts that intrinsic DCN hyperexcitability would cause ataxia in the absence of upstream Purkinje degeneration. We report the generation of a transgenic (Tg) model that supports this mechanism of disease initiation. Small-conductance calcium-activated potassium (SK) channels, regulators of firing frequency, were silenced in the CNS of Tg mice with the dominant-inhibitory construct SK3-1B-GFP. Transgene expression was restricted to the DCN within the cerebellum and was detectable beginning on postnatal day 10, concomitant with the onset of cerebellar ataxia. Neurodegeneration was not evident up to the sixth month of age. Recordings from Tg DCN neurons revealed loss of the apamin-sensitive after-hyperpolarization current (IAHP) and increased spontaneous firing through SK channel suppression, indicative of DCN hyperexcitability. Spike duration and other electrogenic conductance were unaffected. Thus, a purely electrical alteration is sufficient to cause cerebellar ataxia, and SK openers such as the neuroprotective agent riluzole may reduce neuronal hyperexcitability and have therapeutic value. This dominant-inhibitory strategy may help define the in vivo role of SK channels in other neuronal pathways.


Stem Cells | 2008

Neural Stem Cell Targeting of Glioma Is Dependent on Phosphoinositide 3‐Kinase Signaling

Stephen E. Kendall; Joseph Najbauer; Heather F. Johnston; Marianne Z. Metz; Shan Li; Marisa Bowers; Elizabeth Garcia; Seung U. Kim; Michael E. Barish; Karen S. Aboody; Carlotta A. Glackin

The utility of neural stem cells (NSCs) has extended beyond regenerative medicine to targeted gene delivery, as NSCs possess an inherent tropism to solid tumors, including invasive gliomas. However, for optimal clinical implementation, an understanding of the molecular events that regulate NSC tumor tropism is needed to ensure their safety and to maximize therapeutic efficacy. We show that human NSC lines responded to multiple tumor‐derived growth factors and that hepatocyte growth factor (HGF) induced the strongest chemotactic response. Gliomatropism was critically dependent on c‐Met signaling, as short hairpin RNA‐mediated ablation of c‐Met significantly attenuated the response. Furthermore, inhibition of Ras‐phosphoinositide 3‐kinase (PI3K) signaling impaired the migration of human neural stem cells (hNSCs) toward HGF and other growth factors. Migration toward tumor cells is a highly regulated process, in which multiple growth factor signals converge on Ras‐PI3K, causing direct modification of the cytoskeleton. The signaling pathways that regulate hNSC migration are similar to those that promote unregulated glioma invasion, suggesting shared cellular mechanisms and responses.


Stem Cells Translational Medicine | 2013

Magnetic Resonance Imaging Tracking of Ferumoxytol-Labeled Human Neural Stem Cells: Studies Leading to Clinical Use

Margarita Gutova; Joseph A. Frank; Massimo D'Apuzzo; Vazgen Khankaldyyan; Megan Gilchrist; Alexander J. Annala; Marianne Z. Metz; Yelena Abramyants; Kelsey Herrmann; Lucy Y. Ghoda; Joseph Najbauer; Christine E. Brown; M. Suzette Blanchard; Maciej S. Lesniak; Seung U. Kim; Michael E. Barish; Karen S. Aboody; Rex Moats

Numerous stem cell‐based therapies are currently under clinical investigation, including the use of neural stem cells (NSCs) as delivery vehicles to target therapeutic agents to invasive brain tumors. The ability to monitor the time course, migration, and distribution of stem cells following transplantation into patients would provide critical information for optimizing treatment regimens. No effective cell‐tracking methodology has yet garnered clinical acceptance. A highly promising noninvasive method for monitoring NSCs and potentially other cell types in vivo involves preloading them with ultrasmall superparamagnetic iron oxide nanoparticles (USPIOs) to enable cell tracking using magnetic resonance imaging (MRI). We report here the preclinical studies that led to U.S. Food and Drug Administration approval for first‐in‐human investigational use of ferumoxytol to label NSCs prior to transplantation into brain tumor patients, followed by surveillance serial MRI. A combination of heparin, protamine sulfate, and ferumoxytol (HPF) was used to label the NSCs. HPF labeling did not affect cell viability, growth kinetics, or tumor tropism in vitro, and it enabled MRI visualization of NSC distribution within orthotopic glioma xenografts. MRI revealed dynamic in vivo NSC distribution at multiple time points following intracerebral or intravenous injection into glioma‐bearing mice that correlated with histological analysis. Preclinical safety/toxicity studies of intracerebrally administered HPF‐labeled NSCs in mice were also performed, and they showed no significant clinical or behavioral changes, no neuronal or systemic toxicities, and no abnormal accumulation of iron in the liver or spleen. These studies support the clinical use of ferumoxytol labeling of cells for post‐transplant MRI visualization and tracking.


PLOS ONE | 2013

Glioma IL13Rα2 is associated with mesenchymal signature gene expression and poor patient prognosis.

Christine E. Brown; Charles Warden; Renate Starr; Xutao Deng; Behnam Badie; Yate-Ching Yuan; Stephen J. Forman; Michael E. Barish

A major challenge for successful immunotherapy against glioma is the identification and characterization of validated targets. We have taken a bioinformatics approach towards understanding the biological context of IL-13 receptor α2 (IL13Rα2) expression in brain tumors, and its functional significance for patient survival. Querying multiple gene expression databases, we show that IL13Rα2 expression increases with glioma malignancy grade, and expression for high-grade tumors is bimodal, with approximately 58% of WHO grade IV gliomas over-expressing this receptor. By several measures, IL13Rα2 expression in patient samples and low-passage primary glioma lines most consistently correlates with the expression of signature genes defining mesenchymal subclass tumors and negatively correlates with proneural signature genes as defined by two studies. Positive associations were also noted with proliferative signature genes, whereas no consistent associations were found with either classical or neural signature genes. Probing the potential functional consequences of this mesenchymal association through IPA analysis suggests that IL13Rα2 expression is associated with activation of proinflammatory and immune pathways characteristic of mesenchymal subclass tumors. In addition, survival analyses indicate that IL13Rα2 over-expression is associated with poor patient prognosis, a single gene correlation ranking IL13Rα2 in the top ~1% of total gene expression probes with regard to survival association with WHO IV gliomas. This study better defines the functional consequences of IL13Rα2 expression by demonstrating association with mesenchymal signature gene expression and poor patient prognosis. It thus highlights the utility of IL13Rα2 as a therapeutic target, and helps define patient populations most likely to respond to immunotherapy in present and future clinical trials.


Stem Cells Translational Medicine | 2013

Neural Stem Cell-Mediated Delivery of Irinotecan-Activating Carboxylesterases to Glioma: Implications for Clinical Use

Marianne Z. Metz; Margarita Gutova; Simon F. Lacey; Yelena Abramyants; Tien Vo; Megan Gilchrist; Revathiswari Tirughana; Lucy Y. Ghoda; Michael E. Barish; Christine E. Brown; Joseph Najbauer; Philip M. Potter; Jana Portnow; Timothy W. Synold; Karen S. Aboody

CPT‐11 (irinotecan) has been investigated as a treatment for malignant brain tumors. However, limitations of CPT‐11 therapy include low levels of the drug entering brain tumor sites and systemic toxicities associated with higher doses. Neural stem cells (NSCs) offer a novel way to overcome these obstacles because of their inherent tumor tropism and ability to cross the blood‐brain barrier, which enables them to selectively target brain tumor sites. Carboxylesterases (CEs) are enzymes that can convert the prodrug CPT‐11 (irinotecan) to its active metabolite SN‐38, a potent topoisomerase I inhibitor. We have adenovirally transduced an established clonal human NSC line (HB1.F3.CD) to express a rabbit carboxylesterase (rCE) or a modified human CE (hCE1m6), which are more effective at converting CPT‐11 to SN‐38 than endogenous human CE. We hypothesized that NSC‐mediated CE/CPT‐11 therapy would allow tumor‐localized production of SN‐38 and significantly increase the therapeutic efficacy of irinotecan. Here, we report that transduced NSCs transiently expressed high levels of active CE enzymes, retained their tumor‐tropic properties, and mediated an increase in the cytotoxicity of CPT‐11 toward glioma cells. CE‐expressing NSCs (NSC.CEs), whether administered intracranially or intravenously, delivered CE to orthotopic human glioma xenografts in mice. NSC‐delivered CE catalyzed conversion of CPT‐11 to SN‐38 locally at tumor sites. These studies demonstrate the feasibility of NSC‐mediated delivery of CE to glioma and lay the foundation for translational studies of this therapeutic paradigm to improve clinical outcome and quality of life in patients with malignant brain tumors.


NeuroImage | 2007

Novel method for visualizing and modeling the spatial distribution of neural stem cells within intracranial glioma

David Lin; Joseph Najbauer; Paul M. Salvaterra; Adam N. Mamelak; Michael E. Barish; Elizabeth Garcia; Marianne Z. Metz; Stephen E. Kendall; Marisa Bowers; Babak Kateb; Seung U. Kim; Margaret L. Johnson; Karen S. Aboody

Neural stem cells (NSCs) hold great promise for glioma therapy due to their inherent tumor-tropic properties, enabling them to deliver therapeutic agents directly to invasive tumor sites. In the present study, we visualized and quantitatively analyzed the spatial distribution of tumor-tropic NSCs in a mouse model of orthotopic glioma in order to predict the therapeutic efficacy of a representative NSC-based glioma therapy. U251.eGFP human glioma was established in the brain of athymic mice, followed by stereotactic injection of CM-DiI-labeled human NSCs posterior-lateral to the tumor site. Confocal microscopy, three-dimensional modeling and mathematical algorithms were used to visualize and characterize the spatial distribution of NSCs throughout the tumor. The pattern of NSC distribution showed a gradient with higher densities toward the centroid of the tumor mass. We estimate that NSC-mediated therapy would eradicate 70-90% of the primary tumor mass and the majority of invasive tumor foci. Our method may serve as a model for optimizing the efficacy of NSC-based glioma therapy.

Collaboration


Dive into the Michael E. Barish's collaboration.

Top Co-Authors

Avatar

Christine E. Brown

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Karen S. Aboody

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Behnam Badie

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Renate Starr

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Margarita Gutova

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Massimo D'Apuzzo

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brenda Aguilar

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marianne Z. Metz

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph Najbauer

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge