Network


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

Hotspot


Dive into the research topics where Beata Holkova is active.

Publication


Featured researches published by Beata Holkova.


Clinical Cancer Research | 2005

Immunomodulatory Drug CC-5013 or CC-4047 and Rituximab Enhance Antitumor Activity in a Severe Combined Immunodeficient Mouse Lymphoma Model

Francisco J. Hernandez-Ilizaliturri; Nishitha Reddy; Beata Holkova; Edris Ottman; Myron S. Czuczman

New thalidomide derivatives CC-5013 and CC-4047 (immunomodulatory drugs, IMiD) are up to 10,000 times more potent than Thalidomide. The biological effects of IMiDs are presumed to be mediated by (a) activation of some components of the innate [natural killer (NK) cells] or adoptive immune system (T cells), (b) modification of cytokine microenvironment in the tumor bed, or by (c) inhibition of angiogenesis. In this article, we tested an innovative combination strategy involving rituximab and IMiDs in aggressive lymphoma cell lines and human lymphoma xenografts. Treatment of non-Hodgkins lymphoma cells with CC-5013 resulted in a 40% to 70% growth inhibition when compared with controls (P < 0.05). Exposure of lymphoma cells to CC-4047 resulted in a lesser degree of growth inhibition. Induction of apoptosis was shown in 10% to 26% of lymphoma cells 24 hours following exposure to either IMiD. In vivo studies in severe combined immunodeficient mice showed synergistic activity between CC-4047 (and to a lesser degree, CC-5013) plus rituximab. Animals treated with the CC-4047/rituximab combination had a median survival of 74 days (P = 0.0012) compared with 58 days (P = 0.167) in CC-5013/rituximab-treated animals compared with 45 days in rituximab monotherapy–treated animals. The synergistic effect between IMiDs and rituximab in our mouse model was attributed to NK cell expansion. The enhancement of rituximab activity by IMiDs was abrogated by in vivo depletion of NK cells. Augmenting NK cell function by CC-4047 or CC-5013 exposure may increase the antitumor effects of rituximab against B-cell lymphomas and warrants further exploration in the context of a clinical trial.


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.


Expert Opinion on Pharmacotherapy | 2014

Bortezomib for the treatment of non-Hodgkin's lymphoma

Prithviraj Bose; Michael Batalo; Beata Holkova; Steven Grant

Introduction: Bortezomib, the first proteasome inhibitor (PI) to be evaluated in humans, is approved in the USA and Europe for the treatment of patients with multiple myeloma, and in the USA for patients with relapsed mantle cell lymphoma (MCL). Areas covered: This review examines the role of bortezomib in the therapy of non-Hodgkin’s lymphoma (NHL). Bortezomib may be particularly effective against the NF-κB-dependent activated B-cell subtype of diffuse large B-cell lymphoma. The combination of bortezomib with rituximab and dexamethasone represents a standard approach for the treatment of Waldenström’s macroglobulinemia, and that with bendamustine and rituximab has demonstrated excellent efficacy in follicular lymphoma. Combinations with other novel agents, such as inhibitors of cyclin-dependent kinases or histone deacetylases, also hold substantial promise in NHL. Unmet needs in NHL, competitor compounds, chemistry, pharmacokinetics, pharmacodynamics and safety and tolerability of bortezomib are also discussed. Expert opinion: The success of bortezomib in MCL has validated the proteasome as a therapeutic target in NHL. Rational combinations, for example, with Bruton’s tyrosine kinase inhibitors or BH3-mimetics, may hold the key to optimizing the therapeutic potential of PIs in NHL. Future trials are likely to involve newer agents with improved pharmacodynamic (e.g., carfilzomib, marizomib) or pharmacokinetic (e.g., ixazomib, oprozomib) properties.


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.


Best Practice & Research Clinical Haematology | 2012

Proteasome inhibitors in mantle cell lymphoma

Beata Holkova; Steven Grant

Mantle cell lymphoma (MCL) represents a subtype of non-Hodgkins lymphoma (NHL) which has a relatively poor prognosis compared to other forms of NHL. Despite multiple options for cytotoxic chemotherapy, attempts to prolong the survival of patients with this disease have not yet met with success. Consequently, the development of targeted approaches to therapy which minimize toxicities has potentially important implications for MCL. Proteasome inhibitors preferentially kill transformed cells through diverse mechanisms. The proteasome inhibitor bortezomib was initially approved for patients with relapsed or refractory multiple myeloma and now has been approved for relapsed or refractory MCL. The introduction of newer proteasome inhibitors with activity in bortezomib-resistant disease and reduced toxicity profiles may yield further benefits. Multiple ongoing studies are building on the known efficacy of proteasome inhibitors in MCL by evaluating combination regimens involving either cytotoxic or targeted therapies, with the ultimate goal of prolonging survival in this patient population.


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 Colorectal Cancer | 2015

A Phase II Study of Sorafenib Combined With Cetuximab in EGFR-Expressing, KRAS-Mutated Metastatic Colorectal Cancer

Khanh Do; Liang Cao; Zhigang Kang; Baris Turkbey; Maria Liza Lindenberg; Erin Larkins; Beata Holkova; Seth M. Steinberg; Mark Raffeld; Cody J. Peer; William D. Figg; Michelle Eugeni; Paula Jacobs; Peter L. Choyke; John J. Wright; James H. Doroshow; Shivaani Kummar

BACKGROUND Mutations in the KRAS gene predict for resistance to anti-epidermal growth factor receptor (EGFR) therapies, including cetuximab. Upregulation of vascular endothelial growth factor (VEGF)-A has been implicated in resistance to anti-EGFR treatment. Abrogation of the VEGF and RAS/RAF/MEK/ERK pathways has the potential to restore cetuximab sensitivity. PATIENTS AND METHODS Adult patients with histologically documented, measurable, EGFR-expressing, KRAS-mutated metastatic colorectal cancer (mCRC) that had progressed after 5-fluorouracil-based regimens were treated with sorafenib 400 mg orally twice daily and intravenous cetuximab weekly in 28-day cycles. The primary endpoint was the response rate (complete response, partial response, and stable disease at 4 cycles). The secondary endpoints included plasma biomarker analysis of angiogenic cytokines and correlative imaging studies with dynamic contrast-enhanced magnetic resonance imaging and zirconium 89-panitumumab. RESULTS Of the 30 patients enrolled, 26 were evaluable for response. Of the 26 patients evaluated, 4 had stable disease at 4 cycles and 1 had stable disease at 8 cycles. The median progression-free survival was 1.84 months. The common toxicities were rash, diarrhea, and liver enzyme elevations. Of the angiogenic cytokines evaluated, only the placental growth factor increased significantly with treatment (P < .0001). No pharmacodynamic parameters were associated with the treatment response. CONCLUSION We report the results of a trial that combined cetuximab and sorafenib for the treatment of KRAS-mutated mCRC, with correlative imaging studies and pharmacodynamic angiogenic cytokine profiling as downstream markers of EGFR and VEGF receptor (VEGFR) signaling. No objective responses were observed. Additional development of biomarkers for patient selection is needed to evaluate combined EGFR and VEGFR blockade as a therapeutic option in KRAS-mutated CRC.


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.

Collaboration


Dive into the Beata Holkova's collaboration.

Top Co-Authors

Avatar

Steven Grant

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Maciej Kmieciak

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Ellen Shrader

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Mary Beth Tombes

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Prithviraj Bose

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

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