Network


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

Hotspot


Dive into the research topics where Darrell D. Bigner is active.

Publication


Featured researches published by Darrell D. Bigner.


Nature Genetics | 2009

Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma

Paul A. Northcott; Yukiko Nakahara; Xiaochong Wu; Lars Feuk; David W. Ellison; Sid Croul; Stephen C. Mack; Paul N. Kongkham; John Peacock; Adrian Dubuc; Young Shin Ra; Karen Zilberberg; Jessica McLeod; Stephen W. Scherer; J. Sunil Rao; Charles G. Eberhart; Wiesia Grajkowska; Yancey Gillespie; Boleslaw Lach; Richard Grundy; Ian F. Pollack; Ronald L. Hamilton; Timothy Van Meter; Carlos Gilberto Carlotti; Frederick A. Boop; Darrell D. Bigner; Richard J. Gilbertson; James T. Rutka; Michael D. Taylor

We used high-resolution SNP genotyping to identify regions of genomic gain and loss in the genomes of 212 medulloblastomas, malignant pediatric brain tumors. We found focal amplifications of 15 known oncogenes and focal deletions of 20 known tumor suppressor genes (TSG), most not previously implicated in medulloblastoma. Notably, we identified previously unknown amplifications and homozygous deletions, including recurrent, mutually exclusive, highly focal genetic events in genes targeting histone lysine methylation, particularly that of histone 3, lysine 9 (H3K9). Post-translational modification of histone proteins is critical for regulation of gene expression, can participate in determination of stem cell fates and has been implicated in carcinogenesis. Consistent with our genetic data, restoration of expression of genes controlling H3K9 methylation greatly diminishes proliferation of medulloblastoma in vitro. Copy number aberrations of genes with critical roles in writing, reading, removing and blocking the state of histone lysine methylation, particularly at H3K9, suggest that defective control of the histone code contributes to the pathogenesis of medulloblastoma.


Neuro-oncology | 2004

Phase 2 trial of BCNU plus irinotecan in adults with malignant glioma

David A. Reardon; Jennifer A. Quinn; Jeremy N. Rich; Sridharan Gururangan; James J. Vredenburgh; John H. Sampson; James M. Provenzale; Amy Walker; Michael A. Badruddoja; Sandra Tourt-Uhlig; James E. Herndon; Jeannette M. Dowell; Mary Lou Affronti; Susanne Jackson; Deborah Allen; Karen Ziegler; Steven Silverman; Cindy Bohlin; Allan H. Friedman; Darrell D. Bigner; Henry S. Friedman

In preclinical studies, BCNU, or 1,3-bis(2-chloroethyl)-1-nitrosourea, plus CPT-11 (irinotecan) exhibits schedule-dependent, synergistic activity against malignant glioma (MG). We previously established the maximum tolerated dose of CPT-11 when administered for 4 consecutive weeks in combination with BCNU administered on the first day of each 6-week cycle. We now report a phase 2 trial of BCNU plus CPT-11 for patients with MG. In the current study, BCNU (100 mg/m2) was administered on day 1 of each 6-week cycle. CPT-11 was administered on days 1, 8, 15, and 22 at 225 mg/m2 for patients receiving CYP3A1- or CYP3A4-inducing anticonvulsants and at 125 mg/m2 for those not on these medications. Newly diagnosed patients received up to 3 cycles before radiotherapy, while recurrent patients received up to 8 cycles. The primary end point of this study was radiographic response, while time to progression and overall survival were also assessed. Seventy-six patients were treated, including 37 with newly diagnosed tumors and 39 with recurrent disease. Fifty-six had glioblastoma multiforme, 18 had anaplastic astrocytoma, and 2 had anaplastic oligodendroglioma. Toxicities (grade > or =3) included infections (13%), thromboses (12%), diarrhea (10%), and neutropenia (7%). Interstitial pneumonitis developed in 4 patients. Five newly diagnosed patients (14%; 95% CI, 5%-29%) achieved a radiographic response (1 complete response and 4 partial responses). Five patients with recurrent MG also achieved a response (1 complete response and 4 partial responses; 13%; 95% CI, 4%-27%). More than 40% of both newly diagnosed and recurrent patients achieved stable disease. Median time to progression was 11.3 weeks for recurrent glioblastoma multiforme patients and 16.9 weeks for recurrent anaplastic astrocytoma/ anaplastic oligodendroglioma patients. We conclude that the activity of BCNU plus CPT-11 for patients with MG appears comparable to that of CPT-11 alone and may be more toxic.


Medical and Pediatric Oncology | 1996

Successful treatment of childhood pilocytic astrocytomas metastatic to the leptomeninges with high‐dose Cyclophosphamide

Geoffrey McCowage; Robert D. Tien; Roger E. McLendon; Gary J. Felsberg; Herbert E. Fuchs; Michael L. Graham; Joanne Kurtzberg; Albert Moghrabi; Lee Ferrell; Tracy Kerby; Margaret Duncan-Brown; Elizabeth A. Stewart; Patricia L. Robertson; O. Michael Colvin; Barry Golembe; Darrell D. Bigner; Henry S. Friedman

Leptomeningeal dissemination of childhood pilocytic astrocytoma (PA) is a rare event with little information available regarding therapy. We report here four children with disseminated PA whom we treated with high doses of cyclophosphamide with clinical benefit. The patients were aged 2.5 to 8 years. Three patients presented with PA localized in the posterior fossa, initially treated with surgical resection (n = 3) and radiotherapy (n = 1). Leptomeningeal dissemination occurred at 32, 44, and 8 months from diagnosis, respectively. The fourth patient presented with an optic pathway tumor with leptomeningeal dissemination at diagnosis. At commencement of cyclophosphamide therapy, disease was present in the subarachnoid space (intracranial, n = 2; spinal, n = 4), cerebral ventricles (n = 2), and primary site (n = 3). Histology was identical at diagnosis and recurrence in the two biopsied cases and cerebrospinal fluid was negative in all cases. Treatment was with cyclophosphamide 4-5 g/m2/cycle given every 4 weeks for a total of two cycles (n = 1) and four cycles (n = 3). One patient achieved disease stabilization (duration 27 months at the time of publication) and three patients experienced significant reductions in tumor burden. Subsequent intrathecal therapy was administered to two patients. Two patients developed disease progression at 10 and 9 months from cessation of chemotherapy. The one re-treated patient responded to further, lower dose, cyclophosphamide. This is the first report of the use of high dose cyclophosphamide for disseminated PA. The recurrence of disease in two cases with a further response to lower dose cyclophosphamide has implications for the optimal duration of therapy for these low grade, aggressive tumors.


Journal of Neuro-oncology | 2010

Convection-enhanced delivery of free gadolinium with the recombinant immunotoxin MR1-1

Dale Ding; Charles W. Kanaly; Darrell D. Bigner; Thomas J. Cummings; James E. Herndon; Ira Pastan; Raghu Raghavan; John H. Sampson

A major obstacle in glioblastoma (GBM) therapy is the restrictive nature of the blood-brain barrier (BBB). Convection-enhanced delivery (CED) is a novel method of drug administration which allows direct parenchymal infusion of therapeutics, bypassing the BBB. MR1-1 is a novel recombinant immunotoxin that targets the GBM tumor-specific antigen EGFRvIII and can be delivered via CED infusion. However, drug distribution via CED varies dramatically, which necessitates active monitoring. Gadolinium conjugated to diethylenetriamine penta-acetic acid (Gd-DTPA) is a commonly used MRI contrast agent which can be co-infused with therapies using CED and may be useful in monitoring infusion leak and early distribution. Forty immunocompetent rats were implanted with intracerebral cannulas that were connected to osmotic pumps and subsequently randomized into four groups that each received 0.2% human serum albumin (HSA) mixed with a different experimental infusion: (1) 25xa0ng/ml MR1-1; (2) 0.1xa0μmol/ml Gd-DTPA; (3) 25xa0ng/ml MR1-1 and 0.1xa0μmol/ml Gd-DTPA; (4) 250xa0ng/ml MR1-1 and 0.1xa0μmol/ml Gd-DTPA. The rats were monitored clinically for 6xa0weeks then necropsied and histologically assessed for CNS toxicity. All rats survived the entirety of the study without clinical or histological toxicity attributable to the study drugs. There was no statistically significant difference in weight change over time among groups (Pxa0>xa00.999). MR1-1 co-infused with Gd-DTPA via CED is safe in the long-term setting in a pre-clinical animal model. Our data supports the use of Gd-DTPA, as a surrogate tracer, co-infused with MR1-1 for drug distribution monitoring in patients with GBM.


Cancer Chemotherapy and Pharmacology | 2005

Signal transduction through substance P receptor in human glioblastoma cells: roles for Src and PKCδ

Keisuke Yamaguchi; Mark D. Richardson; Darrell D. Bigner; Madan M. Kwatra

Substance P receptor (SPR), a G protein-coupled receptor (GPCR), is found in human glioblastomas, and has been implicated in their growth. Consistent with a role for SPR in cell growth, activation of SPR in U373 MG human glioblastoma cells leads to the phosphorylation of mitogen-activated protein kinases [extracellular signal-regulated kinase 1 and 2 (ERK1/2)] and stimulation of cell proliferation. The purpose of the present study was to elucidate the pathway through which these actions occur. Using either the epidermal growth factor receptor (EGFR) kinase inhibitor, AG 1478, or a small-interfering RNA (siRNA) directed against human EGFR, we found that transactivation of EGFR by SPR is only marginally involved in SP-dependent ERK1/2 phosphorylation. Src, however, is shown to be a major component of SPR signaling because the Src kinase inhibitor, PP2, and a kinase-dead Src mutant both inhibit SP-dependent ERK1/2 phosphorylation. We also report that SPR stimulates the phosphorylation of protein kinase Cδ(PKCδ), and that this stimulation is blocked by PP2. SP-dependent ERK1/2 phosphorylation is also blocked by rottlerin, a PKCδ inhibitor, and the calcium scavenger, BAPTA/AM. Finally, rottlerin and PP2 were both found to inhibit the growth of several glioblastoma cell lines, underscoring the potential of these agents to block glioblastoma growth.


International Journal of Radiation Oncology Biology Physics | 2002

Vascular targeted endoradiotherapy of tumors using alpha-particle-emitting compounds: theoretical analysis☆

Gamal Akabani; Roger E. McLendon; Darrell D. Bigner; Michael R. Zalutsky

PURPOSEnTo establish the theoretical framework and study the feasibility of (211)At-labeled anti-tenascin chimeric 81C6 monoclonal antibody (mAb) as anti-vascular endoradiotherapy for the treatment of glioblastoma multiforme (GBM) tumors.nnnMETHODS AND MATERIALSnThe morphology of blood vessels from histologic images was analyzed and used along with reaction-diffusion equations to assess the activity concentration of (211)At-labeled chimeric 81C6 mAb in GBM tumor and normal-brain tissue. Alpha particle microdosimetry was then used to assess the survival probability and average absorbed dose for tumor and normal tissue endothelial cells (ECs) per unit vascular cumulated activity concentration q(source) (MBq-s g(-1)). In turn, these survival probabilities were used to assess the probability of failure Phi for a single vessel. Furthermore, using the vessel density, the specific tumor control probability per unit mass of tumor tissue (tcp) and the specific normal-tissue complication probability per unit mass of normal-brain tissue (ntcp) were estimated. The specific tumor control probability, tcp, was used to assess the overall tumor control probability (TCP) as a function of tumor mass.nnnRESULTSnThe levels of (211)At-labeled ch81C6 mAb cumulated activity concentration in GBM tumor tissue were approximately five times higher than that in normal-brain tissue. Thus, the average absorbed dose to tumor ECs was higher than that of normal tissue ECs, and the survival probability for GBM ECs was lower than for normal-brain tissue ECs. Consequently, the resulting vessel-failure probability, Phi, for GBM tumor and for normal-brain tissue differ considerably, yielding a q(source) range between 10(3) and 10(4) MBq-s g(-1).nnnCONCLUSIONSnThis theoretical analysis demonstrated that (211)At-labeled chimeric 81C6 is an effective anti-vascular therapy for the treatment of GBM tumors, yielding a tcp higher than 0.999 for vascular cumulated activity concentrations q(source) higher than 1 x 10(4) MBq-s g(-1), while yielding a low probability for normal-brain tissue damage.


Cancer | 2006

Patterns of exercise across the cancer trajectory in brain tumor patients.

Lee W. Jones; Bebe Guill; Stephen T. Keir; B S Karen Carter; Henry S. Friedman; Darrell D. Bigner; David A. Reardon

Exercise may represent a supportive intervention that may complement existing neurooncologic therapies and address a multitude of therapy‐induced debilitating side effects in patients with brain tumors. Given the limited evidence, the authors conducted a survey to examine the exercise patterns of brain tumor patients across the cancer trajectory.


International Journal of Cancer | 2010

IDH1 and IDH2 hotspot mutations are not found in canine glioma.

Zachary J. Reitman; Natasha J. Olby; Christopher L. Mariani; Rachael Thomas; Matthew Breen; Darrell D. Bigner; Roger E. McLendon; Hai Yan

Human diffuse and anaplastic astrocytomas, well-differenti-ated and anaplastic oligodendrogliomas and secondary glio-blastomas frequently (>70%) contain somatic mutations ofthe R132 codon of the cytoplasmic NADPþ-dependent iso-citrate dehydrogenase (IDH1) or the corresponding R172codon in its homolog, IDH2.


CNS Neuroscience & Therapeutics | 2014

AJAP1 is Dysregulated at an Early Stage of Gliomagenesis and Suppresses Invasion Through Cytoskeleton Reorganization

Lei Han; Kailiang Zhang; Junxia Zhang; Liang Zeng; Chunhui Di; Brian E. Fee; Miriam V. Rivas; Zhaoshi Bao; Tao Jiang; Darrell D. Bigner; Chunsheng Kang; David Cory Adamson

Down‐regulation of AJAP1 in glioblastoma multiforme (GBM) has been reported. However, the expression profiles of AJAP1 in gliomas and the underlying mechanisms of AJAP1 function on invasion are still poorly understood.


Cancer Chemotherapy and Pharmacology | 1991

Positive therapeutic interaction between thiopurines and alkylating drugs in human glioma xenografts

A. M. Wang; Gertrude B. Elion; Henry S. Friedman; W. J. Bodell; Darrell D. Bigner; Schold Sc

SummaryWe used human anaplastic glioma xenografts to evaluate the therapeutic efficacy of combinations of alkylating drugs, either 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), 1-(2-chloroethyl)-3-(2,5-dioxo-3-piperidyl)-1-nitrosourea (PCNU), or procarbazine, and thiopurines, either 6-mercaptopurine (6MP) or 6-thioguanine (6TG). Using growth delay as the endpoint in subcutaneous (s.c.) tumors and increased life span as the endpoint in intracranial (i.c.) tumors, we found that combinations of chloroethylnitrosoureas (CENUs) and thiopurines were significantly more active than either type of agent alone. In contrast, combinations of procarbazine and thiopurines were not significantly more active than procarbazine alone. The therapeutic potentiation of the CENU was greater when the latter was given on the 4th day of the thiopurine treatment cycle than when it was given on the 1st day. Characterization of the interaction between CENUs and thiopurines also revealed a supraadditive therapeutic response at higher BCNU doses in combination with 6TG. Interaction between the nitrosoureas and the thiopurines probably occurs in the guanine base of tumor DNA and has important therapeutic implications.

Collaboration


Dive into the Darrell D. Bigner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dale Ding

Barrow Neurological Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge