9 results on '"Chandra Tucker"'
Search Results
2. The pediatric preclinical testing program: Description of models and early testing results
- Author
-
Richard Gorlick, Sherry S Ansher, Claire Cole, Stephen T. Keir, Javed Khan, Christopher L. Morton, John M. Maris, Chandra Tucker, Peter J. Houghton, Charles M. Stopford, E. Anders Kolb, Hernan Carol, Mimi Tajbakhsh, Joshua Courtright, Malcolm A. Smith, Jianrong Wu, Tiebin Liu, Joseph Zeidner, C. Patrick Reynolds, Henry S. Friedman, Catherine A. Billups, Jian Zhang, Edward Favours, Wendong Zhang, Richard B. Lock, and Debbie Payne
- Subjects
Ependymoma ,Oncology ,medicine.medical_specialty ,Vincristine ,Pathology ,Mice, Nude ,Mice, SCID ,Disease-Free Survival ,Drug Administration Schedule ,Mice ,Mice, Inbred NOD ,Neoplasms ,Internal medicine ,Neuroblastoma ,Animals ,Humans ,Medicine ,Rhabdomyosarcoma ,Cyclophosphamide ,Cell Proliferation ,Medulloblastoma ,Mice, Inbred BALB C ,business.industry ,Cancer ,Wilms' tumor ,Hematology ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Treatment Outcome ,Pediatrics, Perinatology and Child Health ,Female ,Sarcoma ,business ,Injections, Intraperitoneal ,medicine.drug - Abstract
Background The Pediatric Preclinical Testing Program (PPTP) is an initiative supported by the National Cancer Institute (NCI) to identify novel therapeutic agents that may have significant activity against childhood cancers. The PPTP has established panels of childhood cancer xenografts and cell lines to be used for in vivo and in vitro testing. These include panels for Wilms tumor, sarcomas (rhabdomyosarcoma, Ewing sarcoma, and osteosarcoma), neuroblastoma, brain tumors (glioblastoma, ependymoma, and medulloblastoma), rhabdoid tumors (CNS and renal), and acute lymphoblastic leukemia (ALL). Here, we describe the characteristics of the in vivo tumor panels and report results for the in vivo evaluation of two standard agents, vincristine and cyclophosphamide. Procedures Solid tumors were grown subcutaneously in immune-deficient mice and tumor dimensions were measured weekly. ALL xenografts were inoculated intravenously and human CD45-positive cells were enumerated weekly. Results Vincristine-induced objective responses in 6 of 24 (25%) and cyclophosphamide-induced objective responses in 18 of 28 (64%) solid tumor models. Comparable assessments of high levels of activity for these two agents were obtained using a tumor growth delay (TGD) measure. Both agents induced regressions in each of the ALL models evaluated. Conclusions We have established 51 solid tumor and 10 ALL in vivo models. The models identify vincristine and cyclophosphamide as having broad-spectrum activity. The PPTP tumor panels appear to generally recapitulate the activity of these agents against specific childhood cancers and to have the potential for identifying novel agents having significant clinical activity.
- Published
- 2006
3. In vivo Evaluation of Ixabepilone (BMS247550), A Novel Epothilone B Derivative, against Pediatric Cancer Models
- Author
-
Jennifer K. Peterson, Francis Y.F. Lee, Pamela J. Cheshire, Jeremy Creech, Richard Smykla, Chandra Tucker, Catherine A. Billups, Edward Favours, and Peter J. Houghton
- Subjects
Cancer Research ,Time Factors ,Maximum Tolerated Dose ,Paclitaxel ,Mitosis ,Phases of clinical research ,Mice, SCID ,Pharmacology ,Epothilone ,Microtubules ,Wilms Tumor ,Mice ,Neuroblastoma ,chemistry.chemical_compound ,Pharmacokinetics ,Cell Line, Tumor ,Rhabdomyosarcoma ,medicine ,Animals ,Humans ,Vinca Alkaloids ,Clinical Trials as Topic ,Osteosarcoma ,Dose-Response Relationship, Drug ,Brain Neoplasms ,business.industry ,Ixabepilone ,Area under the curve ,medicine.disease ,Pediatric cancer ,Disease Models, Animal ,Treatment Outcome ,Models, Chemical ,Oncology ,chemistry ,Epothilones ,Area Under Curve ,Female ,business ,Neoplasm Transplantation ,medicine.drug - Abstract
Purpose: Vinca alkaloids, agents that cause depolymerization of microtubules, are highly active in treatment of many pediatric cancers. In contrast, taxanes, agents that stabilize microtubules, are far less effective against the same cancer types. The purpose of the current study was to evaluate the antitumor activity of ixabepilone, an epothilone B derivative representing a new class of microtubule-stabilizing antimitotic agent in a wide variety of pediatric solid tumor models. Experimental Design: Ixabepilone was administered i.v. every 4 days for three doses to scid mice bearing s.c. human rhabdomyosarcoma (three lines), neuroblastoma (four), Wilms' tumors (six), osteosarcoma (four), or brain tumors (seven). Tumor diameters were measured weekly, and tumor growth or regressions were determined. Pharmacokinetic studies were done following a single administration of drug at the maximum tolerated dose (MTD) level (10 mg/kg). Results: At the MTD (10 mg/kg), ixabepilone induced objective responses (all tumors in a group achieved ≥50% volume regression) in three of three rhabdomyosarcoma lines, three of five neuroblastomas, six of seven Wilms' tumor models, two of six osteosarcoma, and four of eight brain tumor models. However, the dose-response curve was steep with only 2 of 19 tumors models regressing (≥50%) at 4.4 mg/kg. In comparison, paclitaxel administered at the MTD on the same schedule failed to induce objective regressions of three tumor lines that were highly sensitive to treatment with ixabepilone. Pharmacokinetics following single i.v. administration of ixabepilone at its MTD (10 mg/kg) were biexponential with Cmax of 12.5 μmol/L, elimination half-life of 19.2 hours, and total area under the curve of 5.8 μmol/L-h. The achieved drug exposure of ixabepilone at this efficacious MTD dose level in mice is similar to those achieved in patients given the recommended phase II dose of 40 mg/m2 by either 1- or 3-hour infusion every 3 weeks, a regimen that has shown significant anticancer activity in phase II clinical trials in adult patients. Conclusions: Administered at doses ranging from 66% to 100% of its MTD in mice, the epothilone B derivative ixabepilone shows broad spectrum activity against a panel of pediatric tumor xenograft models. Pharmacokinetic analysis indicates that the systemic ixabepilone exposure achieved in mice at its MTD is similar to that achieved in patients at the recommended phase II dose of 40 mg/m2 administered every 3 weeks. Importantly, the present results showed a clear distinction in sensitivity of pediatric solid tumors to this epothilone derivative compared with paclitaxel.
- Published
- 2005
4. Manipulating cellular processes using optical control of protein–protein interactions
- Author
-
Chandra Tucker and Thomas Knopfel
- Subjects
Cryptochrome ,Optical control ,Light control ,business.industry ,Target protein ,Computational biology ,Anion Pumps ,Optogenetics ,Biology ,Modular design ,business ,Protein–protein interaction ,Cell biology - Abstract
Tools for optical control of proteins offer an unprecedented level of spatiotemporal control over biological processes, adding a new layer of experimental opportunity. While use of light-activated cation channels and anion pumps has already revolutionized neurobiology, an emerging class of more general optogenetic tools may have similar transformative effects. These tools consist of light-dependent protein interaction modules that allow control of target protein interactions and localization with light. Such tools are modular and can be applied to regulate a wide variety of biological activities. This chapter reviews the different properties of light-induced dimerization systems, based on plant phytochromes, cryptochromes, and light-oxygen-voltage domain proteins, exploring advantages and limitations of the different systems and practical considerations related to their use. Potential applications of these tools within the neurobiology field, including light control of various signaling pathways, neuronal activity, and DNA recombination and transcription, are discussed.
- Published
- 2012
5. Evaluation of cytarabine against Ewing sarcoma xenografts by the pediatric preclinical testing program
- Author
-
C. Patrick Reynolds, Malcolm A. Smith, Edward Favours, Debbie Payne-Turner, Min H. Kang, Peter J. Houghton, Catherine A. Billups, Chandra Tucker, and Christopher L. Morton
- Subjects
Antimetabolites, Antineoplastic ,Oncogene Proteins, Fusion ,Bone Neoplasms ,Mice, SCID ,Sarcoma, Ewing ,Translocation, Genetic ,Article ,Mice ,Neuroblastoma ,In vivo ,Cell Line, Tumor ,Rhabdomyosarcoma ,Medicine ,Animals ,Humans ,Child ,Oncogene ,business.industry ,Proto-Oncogene Protein c-fli-1 ,Cytarabine ,Hematology ,medicine.disease ,Pediatric cancer ,Xenograft Model Antitumor Assays ,Oncology ,Cell culture ,Pediatrics, Perinatology and Child Health ,Cancer research ,Female ,Sarcoma ,RNA-Binding Protein EWS ,business ,medicine.drug - Abstract
Treatment with the nucleoside analog cytarabine has been shown to mimic changes in gene expression associated with downregulation of the EWS-FLI1 oncogene in Ewing sarcoma cell lines, selectively inhibit their growth in vitro, and cause tumor regression in athymic nude mice. For this report cytarabine was studied in vitro against a panel of 23 pediatric cancer cell lines and in vivo against 6 Ewing sarcoma xenografts. Acute lymphoblastic leukemia cell lines were the most sensitive to cytarabine in vitro (median IC(50) 9 nM), while Ewing sarcoma cell lines showed intermediate sensitivity (median IC(50) 232 nM). Cytarabine at a dose of 150 mg/kg administered daily 5× failed to significantly inhibit growth of five xenograft models, but reduced growth rate of the A673 xenograft by 50%. Cytarabine shows no differential in vitro activity against Ewing sarcoma cell lines and is ineffective in vivo against Ewing sarcoma xenografts at the dose and schedule studied.
- Published
- 2010
6. Evaluation of ABT-751 against childhood cancer models in vivo
- Author
-
Kimberly S Mercer, Claire R. Boltz, Chandra Tucker, Catherine A. Billups, Christopher L. Morton, Edward Favours, Peter J. Houghton, and Jeri Carol Crumpton
- Subjects
Childhood cancer ,Mice, SCID ,Pharmacology ,Antimitotic Agents ,Pediatrics ,Mice ,In vivo ,Colchicine binding ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Pharmacology (medical) ,Child ,Sulfonamides ,biology ,business.industry ,Pediatric Tumor ,Phase i trials ,Neoplasms, Experimental ,Xenograft Model Antitumor Assays ,Tubulin ,Oncology ,Cell culture ,biology.protein ,Antimitotic Agent ,Female ,business - Abstract
ABT-751 is a novel antimitotic agent that binds tubulin at the colchicine binding site. ABT-751 is undergoing Phase I trials in children, but has not been evaluated against a range of pediatric tumor models in vivo.ABT-751 was evaluated against 27 subcutaneously implanted xenograft models of childhood cancer including neuroblastoma [4], osteosarcoma [4], Ewing sarcoma [2] rhabdomyosarcoma [8], medulloblastoma [1] and eight kidney cancer lines (six Wilms tumors, two rhabdoid). ABT-751 was administered at 100 mg/kg P.O. on a schedule of 5 days on, 5 days off, 5 days on, repeating the cycle at 21 days. Tumor diameters were measured at 7 day intervals for a period of 12 weeks. Three measures of antitumor activity were used: (1) clinical response criteria [e.g., partial response (PR), complete response (CR), etc.]; (2) treated to control (T/C) tumor volume at day 21; and (3) a time to event measure based on the median event free survival (EFS) of treated and control lines.ABT-751 induced regression in 4 of 25 models (16%) including models of neuroblastoma that are refractory to vincristine and paclitaxel. Other regressions occurred in rhabdomyosarcoma and Wilms tumor models. ABT-751 significantly increased event free survival (EFS2.0) in eight models (33%) in addition to those with objective responses.ABT-751 demonstrated intermediate activity against this tumor panel. Neuroblastoma models appear somewhat more sensitive to this agent, with objective regressions also in rhabdomyosarcoma and Wilms tumor. ABT-751 was also active in several tumor lines intrinsically refractory to vincristine or paclitaxel.
- Published
- 2006
7. Evaluation of the antitumor efficacy, pharmacokinetics, and pharmacodynamics of the histone deacetylase inhibitor depsipeptide in childhood cancer models in vivo
- Author
-
Maryam Fouladi, Jeremy Creech, Tiebin Liu, Burgess B. Freeman, Claire L. Graham, Edward Favours, Chandra Tucker, Clinton F. Stewart, Peter J. Houghton, and Catherine A. Billups
- Subjects
Cancer Research ,medicine.drug_class ,Blotting, Western ,Mice, SCID ,Histone Deacetylases ,Histones ,Mice ,In vivo ,Cell Line, Tumor ,Depsipeptides ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Child ,Histone Acetyltransferases ,Depsipeptide ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Histone deacetylase inhibitor ,Cancer ,Acetylation ,Neoplasms, Experimental ,medicine.disease ,Disease Models, Animal ,Histone ,Oncology ,Primitive neuroectodermal tumor ,biology.protein ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Neoplasm Transplantation - Abstract
Purpose: Histone acetyltransferases and histone deacetylases (HDAC) control the acetylation state of histones and other proteins regulating transcription and protein function. Several structurally diverse HDAC inhibitors have been developed as cancer therapeutic agents and in vitro have been shown to cause differentiation, cell cycle arrest, or apoptosis. Here, we have evaluated depsipeptide, a natural tetrapeptide HDAC inhibitor, against a panel of pediatric solid tumor models in vivo and evaluated pharmacokinetic and pharmacodynamic variables with tumor sensitivity. Experimental Design: Depsipeptide was administered at the maximum tolerated dose (4.4 mg/kg administered every 7 days × 3 i.v. repeated q21d for a total of two cycles) to scid mice bearing 39 independently derived childhood tumors (9 brain tumors, 11 kidney cancers, 9 rhabdomyosarcomas, 3 neuroblastomas, and 7 osteosarcomas). Pharmacokinetic variables were determined, as were changes in histone and p53 acetylation, induction of p53 and p53 genotype, and alterations in Akt phosphorylation. Results: Of 39 tumors evaluated, three showed objective tumor regressions [two brain tumors (primitive neuroectodermal tumor and atypical teratoid malignant rhabdoid tumor) and one Wilms' tumor]. Depsipeptide inhibited growth of many tumor lines but achieved stable disease ( Conclusions: Our results show that depsipeptide inhibits its target in vivo causing increased histone acetylation; however, this does not correlate with drug sensitivity. The relatively low objective response rate [3 of 39 (8%) tumor lines showing greater than or equal to partial response and 4 (10%) stable disease] administered at dose levels that give clinically relevant drug exposures suggests that as a single agent depsipeptide may have limited clinical utility against pediatric solid tumors in a first-line setting.
- Published
- 2006
8. Evaluation of ABT-751 against childhood cancer models in vivo.
- Author
-
Christopher Morton, Edward Favours, Kimberly Mercer, Claire Boltz, Jeri Crumpton, Chandra Tucker, Catherine Billups, and Peter Houghton
- Subjects
ANTIMITOTIC agents ,CHILDHOOD cancer ,CLINICAL trials ,REGRESSION analysis - Abstract
Summary Objective ABT-751 is a novel antimitotic agent that binds tubulin at the colchicine binding site. ABT-751 is undergoing Phase I trials in children, but has not been evaluated against a range of pediatric tumor models in vivo. Materials and methods ABT-751 was evaluated against 27 subcutaneously implanted xenograft models of childhood cancer including neuroblastoma [4], osteosarcoma [4], Ewing sarcoma [2] rhabdomyosarcoma [8], medulloblastoma [1] and eight kidney cancer lines (six Wilms tumors, two rhabdoid). ABT-751 was administered at 100 mg/kg P.O. on a schedule of 5 days on, 5 days off, 5 days on, repeating the cycle at 21 days. Tumor diameters were measured at 7 day intervals for a period of 12 weeks. Three measures of antitumor activity were used: (1) clinical response criteria [e.g., partial response (PR), complete response (CR), etc.]; (2) treated to control (T/C) tumor volume at day 21; and (3) a time to event measure based on the median event free survival (EFS) of treated and control lines. Results ABT-751 induced regression in 4 of 25 models (16%) including models of neuroblastoma that are refractory to vincristine and paclitaxel. Other regressions occurred in rhabdomyosarcoma and Wilms tumor models. ABT-751 significantly increased event free survival (EFS > 2.0) in eight models (33%) in addition to those with objective responses. Conclusions ABT-751 demonstrated intermediate activity against this tumor panel. Neuroblastoma models appear somewhat more sensitive to this agent, with objective regressions also in rhabdomyosarcoma and Wilms tumor. ABT-751 was also active in several tumor lines intrinsically refractory to vincristine or paclitaxel. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
9. Optical control of protein-protein interactions to modulate cellular function
- Author
-
Taslimi, A., Pathak, G., Vrana, J. D., and Chandra Tucker
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.