Network


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

Hotspot


Dive into the research topics where Ellen Shrader is active.

Publication


Featured researches published by Ellen Shrader.


Clinical Cancer Research | 2011

Phase I Trial of Bortezomib (PS-341; NSC 681239) and Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory B-cell Neoplasms

Beata Holkova; E. Brent Perkins; Viswanathan Ramakrishnan; Mary Beth Tombes; Ellen Shrader; Neha Talreja; Martha D. Wellons; Kevin T. Hogan; G. David Roodman; Domenico Coppola; Loveleen Kang; Jana L. Dawson; Robert K. Stuart; Cody J. Peer; William D. Figg; Sarah Kolla; Austin Doyle; John J. Wright; Daniel M. Sullivan; John D. Roberts; Steven Grant

Purpose: A phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of bortezomib and alvocidib in patients with B-cell malignancies (multiple myeloma, indolent lymphoma, and mantle cell lymphoma). Experimental Design: Patients received bortezomib by intravenous push on days 1, 4, 8, and 11. Patients also received alvocidib on days 1 and 8 by 30-minute bolus infusion followed by a 4-hour continuous infusion. Treatment was on a 21-day cycle, with indefinite continuation for patients experiencing responses or stable disease. Dose escalation employed a standard 3 + 3 design until the MTD was identified on the basis of DLTs. Pharmacokinetic studies and pharmacodynamic studies were conducted. Results: Sixteen patients were treated. The MTD was established as 1.3 mg/m2 for bortezomib and 30 mg/m2 for alvocidib (both the 30-minute bolus and 4-hour infusions). Common hematologic toxicities included leukopenia, lymphopenia, neutropenia, and thrombocytopenia. Common nonhematologic toxicities included fatigue and febrile neutropenia. DLTs included fatigue, febrile neutropenia, and elevated aspartate aminotransferase (AST) levels. Two complete responses (CR; 12%) and five partial responses (PR; 31%) were observed at the MTD (overall response rate = 44%). Pharmacokinetic results were typical for alvocidib and pharmacodynamic studies yielded variable results. Conclusions: The combination of bortezomib and alvocidib is tolerable and an MTD has been established for the tested schedule. The regimen appears active in patients with relapsed and/or refractory multiple myeloma or non–Hodgkins lymphoma, justifying phase II studies to determine the activity of this regimen more definitively. Clin Cancer Res; 17(10); 3388–97. ©2011 AACR.


Clinical Cancer Research | 2013

A Phase I Trial of Vorinostat and Alvocidib in Patients with Relapsed, Refractory, or Poor Prognosis Acute Leukemia, or Refractory Anemia with Excess Blasts-2

Beata Holkova; Jeffrey G. Supko; Joel M. Reid; Geoffrey I. Shapiro; Edward B. Perkins; Viswanathan Ramakrishnan; Mary Beth Tombes; Connie Honeycutt; Renee M. McGovern; Maciej Kmieciak; Ellen Shrader; Martha D. Wellons; Heidi Sankala; Austin Doyle; John W. Wright; John D. Roberts; Steven Grant

Purpose: This phase I study was conducted to identify the maximum-tolerated dose (MTD) of alvocidib when combined with vorinostat in patients with relapsed, refractory, or poor prognosis acute leukemia, or refractory anemia with excess blasts-2. Secondary objectives included investigating the pharmacokinetic and pharmacodynamic effects of the combination. Experimental Design: Patients received vorinostat (200 mg orally, three times a day, for 14 days) on a 21-day cycle, combined with 2 different alvocidib administration schedules: a 1-hour intravenous infusion, daily × 5; or a 30-minute loading infusion followed by a 4-hour maintenance infusion, weekly × 2. The alvocidib dose was escalated using a standard 3+3 design. Results: Twenty-eight patients were enrolled and treated. The alvocidib MTD was 20 mg/m2 (30-minute loading infusion) followed by 20 mg/m2 (4-hour maintenance infusion) on days one and eight, in combination with vorinostat. The most frequently encountered toxicities were cytopenias, fatigue, hyperglycemia, hypokalemia, hypophosphatemia, and QT prolongation. Dose-limiting toxicities (DLT) were cardiac arrhythmia-atrial fibrillation and QT prolongation. No objective responses were achieved although 13 of 26 evaluable patients exhibited stable disease. Alvocidib seemed to alter vorinostat pharmacokinetics, whereas alvocidib pharmacokinetics were unaffected by vorinostat. Ex vivo exposure of leukemia cells to plasma obtained from patients after alvocidib treatment blocked vorinostat-mediated p21CIP1 induction and downregulated Mcl-1 and p-RNA Pol II for some specimens, although parallel in vivo bone marrow responses were infrequent. Conclusions: Alvocidib combined with vorinostat is well tolerated. Although disease stabilization occurred in some heavily pretreated patients, objective responses were not obtained with these schedules. Clin Cancer Res; 19(7); 1873–83. ©2013 AACR.


Clinical Cancer Research | 2014

Phase I Trial of Bortezomib (PS-341; NSC 681239) and “Nonhybrid” (Bolus) Infusion Schedule of Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory Indolent B-cell Neoplasms

Beata Holkova; Maciej Kmieciak; E. Brent Perkins; Prithviraj Bose; Rachid Baz; G. David Roodman; Robert K. Stuart; Viswanathan Ramakrishnan; Wen Wan; Cody J. Peer; Jana L. Dawson; Loveleen Kang; Connie Honeycutt; Mary Beth Tombes; Ellen Shrader; Caryn Weir-Wiggins; Martha D. Wellons; Heidi Sankala; Kevin T. Hogan; A. Dimitrios Colevas; L. Austin Doyle; William D. Figg; Domenico Coppola; John D. Roberts; Daniel M. Sullivan; Steven Grant

Purpose: This phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of bortezomib and alvocidib in patients with B-cell malignancies (multiple myeloma, indolent lymphoma, Waldenstrom macroglobulinemia, and mantle cell lymphoma). Experimental Design: Patients received bortezomib (intravenous push), followed by alvocidib (1-hour infusion), on days 1, 4, 8, and 11 of a 21-day treatment cycle. Patients experiencing responses or stable disease continued on treatment at the investigators discretion. A standard 3+3 dose-escalation design was used to identify the MTD based on DLTs, and pharmacokinetic and pharmacodynamic studies were conducted. Results: A total of 44 patients were enrolled, with 39 patients assessed for response. The MTD was established as 1.3 mg/m2 for bortezomib and 40 mg/m2 for alvocidib. The most common hematologic toxicities included leukopenia, lymphopenia, neutropenia, and thrombocytopenia. The most common nonhematologic toxicities included diarrhea, fatigue, and sensory neuropathy. Three complete remissions (8%) and 10 partial remissions (26%) were observed for a total response rate of 33%. Pharmacokinetic findings with the current dosing regimen were consistent with the comparable literature and the hybrid dosing regimen. Pharmacodynamic study results did not correlate with clinical responses. Conclusions: The combination of bortezomib and alvocidib is tolerable, and an MTD has been established for this schedule. The regimen appears to be efficacious in patients with relapsed/refractory multiple myeloma or indolent non-Hodgkin lymphoma. As the nonhybrid regimen is less cumbersome than the previous hybrid dosing schedule regimen, the current schedule is recommended for successor studies. Clin Cancer Res; 20(22); 5652–62. ©2014 AACR.


Clinical Cancer Research | 2016

A Phase II Trial of AZD6244 (Selumetinib, ARRY-142886), an Oral MEK1/2 Inhibitor, in Relapsed/Refractory Multiple Myeloma.

Beata Holkova; Adriana Zingone; Maciej Kmieciak; Prithviraj Bose; Ashraf Badros; Peter M. Voorhees; Rachid Baz; Neha Korde; Hui-Yi Lin; Jin-Qiu Chen; Michelle A. Herrmann; Liqiang Xi; Mark Raffeld; Xiuhua Zhao; Wen Wan; Mary Beth Tombes; Ellen Shrader; Caryn Weir-Wiggins; Heidi Sankala; Kevin T. Hogan; Austin Doyle; Christina M. Annunziata; Martha D. Wellons; John D. Roberts; Daniel M. Sullivan; Ola Landgren; Steven Grant

Purpose: AZD6244 is a MEK1/2 inhibitor with significant preclinical activity in multiple myeloma cells. This phase II study used a two-stage Simon design to determine the AZD6244 response rate in patients with relapsed or refractory multiple myeloma. Experimental Design: AZD6244 (75 mg) was administered orally, twice a day, continuously for 28-day cycles. Response was evaluated after three cycles. Results: Thirty-six patients received therapy. The median age was 65 years (range: 43–81) and the median number of prior therapies was 5 (range: 2–11). The most common grade 3 and 4 toxicities included anemia, neutropenia, thrombocytopenia, diarrhea, and fatigue. Three deaths occurred possibly related to AZD6244 (2 due to sepsis, 1 due to acute kidney injury). After AZD6244 discontinuation, three additional deaths occurred due to disease progression. The response rate (CR + PR) was 5.6% with a mean duration of response of 4.95 months and median progression-free survival time of 3.52 months. One patient had a very good partial response (VGPR), 1 patient had a partial response, 17 patients had stable disease, 13 patients had progressive disease, and 4 patients could not be assessed for response. Pharmacodynamic studies revealed variable effects on bone marrow CD138+ cell MEK1/2 and ERK1/2 phosphorylation. The best clinical response, a prolonged VGPR, occurred in a patient with an MMSET translocation. Conclusions: Single-agent AZD6244 was tolerable and had minimal activity in this heavily pretreated population. Clin Cancer Res; 22(5); 1067–75. ©2015 AACR.


Leukemia & Lymphoma | 2016

Phase 1 trial of carfilzomib (PR-171) in combination with vorinostat (SAHA) in patients with relapsed or refractory B-cell lymphomas

Beata Holkova; Maciej Kmieciak; Prithviraj Bose; Victor Yazbeck; Paul M. Barr; Mary Beth Tombes; Ellen Shrader; Caryn Weir-Wiggins; April D. Rollins; Erin Cebula; Emily Pierce; Megan M. Herr; Heidi Sankala; Kevin T. Hogan; Wen Wan; Changyong Feng; Derick R. Peterson; Richard I. Fisher; Steven Grant; Jonathan W. Friedberg

Abstract A phase 1 study with carfilzomib and vorinostat was conducted in 20 B-cell lymphoma patients. Vorinostat was given orally twice daily on days 1, 2, 3, 8, 9, 10, 15, 16, and 17 followed by carfilzomib (given as a 30-min infusion) on days 1, 2, 8, 9, 15, and 16. A treatment cycle was 28 days. Dose escalation initially followed a standard 3 + 3 design, but adapted a more conservative accrual rule following dose de-escalation. The maximum tolerated dose was 20 mg/m2 carfilzomib and 100 mg vorinostat (twice daily). The dose-limiting toxicities were grade 3 pneumonitis, hyponatremia, and febrile neutropenia. One patient had a partial response and two patients had stable disease. Correlative studies showed a decrease in NF-κB activation and an increase in Bim levels in some patients, but these changes did not correlate with clinical response.


Leukemia & Lymphoma | 2017

A phase 1 study of bortezomib and romidepsin in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, indolent B-cell lymphoma, peripheral T-cell lymphoma, or cutaneous T-cell lymphoma

Beata Holkova; Victor Yazbeck; Maciej Kmieciak; Prithviraj Bose; Shuo Ma; Amy S. Kimball; Mary Beth Tombes; Ellen Shrader; Wen Wan; Caryn Weir-Wiggins; Amanda Singh; Kevin T. Hogan; Sarah Conine; Heidi Sankala; John D. Roberts; Thomas C. Shea; Steven Grant

Abstract A phase 1 study was conducted to determine the dose-limiting toxicities and maximum-tolerated dose (MTD) for bortezomib followed by romidepsin on days 1, 8, and 15 in patients with relapsed/refractory CLL/SLL or B- or T-cell lymphoma. Eighteen treated patients were evaluable for response. The MTD was 1.3 mg/m2 bortezomib and 10 mg/m2 romidepsin; median treatment duration was 3 cycles at this dose. The dose-limiting toxicities were grade 3 fatigue, vomiting, and chills. Two patients had partial responses, one lasting >2 years, 8 had stable disease, and 8 had progressive disease. The median duration of stable disease was 3.5 cycles. Correlative studies examining expression of NF-кB, XIAP, Bcl-xL, and Bim yielded variable results. The safety profile was consistent with that reported for single-agent bortezomib and romidepsin. This regimen has modest activity in heavily pretreated patients with relapsed/refractory CLL or B- or T-cell lymphoma. NCT00963274.


Oncotarget | 2016

Phase I study of pemetrexed with sorafenib in advanced solid tumors

Andrew Poklepovic; Sarah Gordon; Danielle Shafer; John D. Roberts; Prithviraj Bose; Charles E. Geyer; William P. McGuire; Mary Beth Tombes; Ellen Shrader; Katie Strickler; Maria A. Quigley; Wen Wan; Maciej Kmieciak; H. Davis Massey; Laurence Booth; Richard G. Moran; Paul Dent

Purpose To determine if combination treatment with pemetrexed and sorafenib is safe and tolerable in patients with advanced solid tumors. Results Thirty-seven patients were enrolled and 36 patients were treated (24 in cohort A; 12 in cohort B). The cohort A dose schedule resulted in problematic cumulative toxicity, while the cohort B dose schedule was found to be more tolerable. The maximum tolerated dose (MTD) was pemetrexed 750 mg/m2 every 14 days with oral sorafenib 400 mg given twice daily on days 1–5. Because dosing delays and modifications were associated with the MTD, the recommended phase II dose was declared to be pemetrexed 500 mg/m2 every 14 days with oral sorafenib 400 mg given twice daily on days 1–5. Thirty-three patients were evaluated for antitumor activity. One complete response and 4 partial responses were observed (15% overall response rate). Stable disease was seen in 15 patients (45%). Four patients had a continued response at 6 months, including 2 of 5 patients with triple-negative breast cancer. Experimental Design A phase I trial employing a standard 3 + 3 design was conducted in patients with advanced solid tumors. Cohort A involved a novel dose escalation schema exploring doses of pemetrexed every 14 days with continuous sorafenib. Cohort B involved a modified schedule of sorafenib dosing on days 1–5 of each 14-day pemetrexed cycle. Radiographic assessments were conducted every 8 weeks. Conclusions Pemetrexed and intermittent sorafenib therapy is a safe and tolerable combination for patients, with promising activity seen in patients with breast cancer.


Clinical Lymphoma, Myeloma & Leukemia | 2018

A Phase II Trial of Bortezomib and Vorinostat in Mantle Cell Lymphoma and Diffuse Large B-cell Lymphoma

Victor Yazbeck; Danielle Shafer; Edward B. Perkins; Domenico Coppola; Lubomir Sokol; Kristy L. Richards; Thomas B. Shea; Jia Ruan; Samir Parekh; Roger Strair; Christopher R. Flowers; David Morgan; Maciej Kmieciak; Prithviraj Bose; Amy Kimball; Ashraf Badros; Rachid Baz; Hui-Yi Lin; Xiuhua Zhao; Richard R. Reich; Mary Beth Tombes; Ellen Shrader; Heidi Sankala; John D. Roberts; Daniel C. Sullivan; Steven Grant; Beata Holkova

Micro‐Abstract Patients with relapsed/refractory non‐Hodgkin lymphoma remain a population with an unmet medical need. Histone deacetylase inhibitors and proteasome inhibitors have shown synergistic interactions preclinically in several B‐cell malignancies. The present phase II trial examined the combination of the proteasome inhibitor bortezomib and the histone deacetylase inhibitor vorinostat in patients with relapsed/refractory diffuse large B‐cell lymphoma or mantle cell lymphoma. The results of the present trial revealed a modest overall response rate. Background The proteasome inhibitor bortezomib has demonstrated marked preclinical activity when combined with the histone deacetylase inhibitor vorinostat in leukemia, multiple myeloma, and mantle cell lymphoma (MCL) cells. The present study evaluated the efficacy and safety of the combination in patients with relapsed or refractory MCL and diffuse large B‐cell lymphoma (DLBCL). Patients and Methods The present multicenter, nonrandomized phase II trial used a Simon 2‐stage design with 3 cohorts: cohort A, MCL with no previous bortezomib (including untreated MCL); cohort B, MCL with previous bortezomib; and cohort C, relapsed or refractory DLBCL with no previous bortezomib. Vorinostat (400 mg) was administered orally on days 1 to 5 and 8 to 12 before bortezomib (1.3 mg/m2), which was administered intravenously on days 1, 4, 8, and 11 of each 21‐day cycle. Results For the 65 treated patients (22 in cohort A, 4 in cohort B, and 39 in cohort C), the overall response rate was 31.8%, 0%, and 7.7%, respectively. The median progression‐free survival was 7.6 months for cohort A and 1.8 months for cohort C. In cohort A, 7 patients had a partial response (PRs), 5 had stable disease (SD), 7 had progressive disease (PD), 1 was not assessed, and 2 were not evaluable. In cohort B, 2 had SD and 2 had PD. In cohort C, 3 had a PR, 8 had SD, 23 had PD, and 5 were not assessed. Baseline NF‐&kgr;B activation, measured as nuclear RelA by immunohistochemistry, did not correlate with clinical response. Conclusion The combination of bortezomib and vorinostat is safe and has modest activity in MCL and limited activity in DLBCL.


Blood | 2011

A Phase II Trial of Bortezomib and Vorinostat in Mantle Cell Lymphoma and Diffuse Large B-Cell Lymphoma

Beata Holkova; Edward B. Perkins; Lubomir Sokol; Kristy L. Richards; Samir Parekh; Rebecca Elstrom; Ashraf Badros; Igor Espinoza-Delgado; Michael J. Schell; Amy Kimball; Mary Beth Tombes; Ellen Shrader; Heidi Sankala; Domenico Coppola; Maciej Kmieciak; Daniel M. Sullivan; John D. Roberts; Steven Grant


Blood | 2011

Phase I Trial of Belinostat and Bortezomib in Patients with Relapsed or Refractory Acute Leukemia, Myelodysplastic Syndrome, or Chronic Myelogenous Leukemia in Blast Crisis - One Year Update

Beata Holkova; Prithviraj Bose; Mary Beth Tombes; Ellen Shrader; Wen Wan; Caryn Weir-Wiggins; Elizabeth Stoddert; Heidi Sankala; Maciej Kmieciak; John D. Roberts; Guillermo Garcia-Manero; Steven Grant

Collaboration


Dive into the Ellen Shrader's collaboration.

Top Co-Authors

Avatar

Mary Beth Tombes

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Maciej Kmieciak

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Steven Grant

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Beata Holkova

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Heidi Sankala

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

John D. Roberts

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Prithviraj Bose

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Wen Wan

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Caryn Weir-Wiggins

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Sullivan

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge