Wiesław Wiktor Jędrzejczak
Medical University of Warsaw
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Wiesław Wiktor Jędrzejczak.
The New England Journal of Medicine | 2013
Michael C. Wang; Simon Rule; Peter Martin; Andre Goy; Rebecca Auer; Brad S. Kahl; Wojciech Jurczak; Ranjana H. Advani; Jorge Romaguera; Michael E. Williams; Jacqueline C. Barrientos; Ewa Chmielowska; John Radford; Stephan Stilgenbauer; M. Dreyling; Wiesław Wiktor Jędrzejczak; Peter E. Johnson; Stephen E. Spurgeon; Lei Li; Liang Zhang; Kate J. Newberry; Zhishuo Ou; Nancy Cheng; Bingliang Fang; Jesse McGreivy; Fong Clow; Joseph J. Buggy; Betty Y. Chang; Darrin M. Beaupre; Lori Kunkel
BACKGROUND Brutons tyrosine kinase (BTK) is a mediator of the B-cell-receptor signaling pathway implicated in the pathogenesis of B-cell cancers. In a phase 1 study, ibrutinib, a BTK inhibitor, showed antitumor activity in several types of non-Hodgkins lymphoma, including mantle-cell lymphoma. METHODS In this phase 2 study, we investigated oral ibrutinib, at a daily dose of 560 mg, in 111 patients with relapsed or refractory mantle-cell lymphoma. Patients were enrolled into two groups: those who had previously received at least 2 cycles of bortezomib therapy and those who had received less than 2 complete cycles of bortezomib or had received no prior bortezomib therapy. The primary end point was the overall response rate. Secondary end points were duration of response, progression-free survival, overall survival, and safety. RESULTS The median age was 68 years, and 86% of patients had intermediate-risk or high-risk mantle-cell lymphoma according to clinical prognostic factors. Patients had received a median of three prior therapies. The most common treatment-related adverse events were mild or moderate diarrhea, fatigue, and nausea. Grade 3 or higher hematologic events were infrequent and included neutropenia (in 16% of patients), thrombocytopenia (in 11%), and anemia (in 10%). A response rate of 68% (75 patients) was observed, with a complete response rate of 21% and a partial response rate of 47%; prior treatment with bortezomib had no effect on the response rate. With an estimated median follow-up of 15.3 months, the estimated median response duration was 17.5 months (95% confidence interval [CI], 15.8 to not reached), the estimated median progression-free survival was 13.9 months (95% CI, 7.0 to not reached), and the median overall survival was not reached. The estimated rate of overall survival was 58% at 18 months. CONCLUSIONS Ibrutinib shows durable single-agent efficacy in relapsed or refractory mantle-cell lymphoma. (Funded by Pharmacyclics and others; ClinicalTrials.gov number, NCT01236391.)
Journal of Clinical Oncology | 2008
S. Vincent Rajkumar; Laura Rosiñol; Mohamad A. Hussein; John Catalano; Wiesław Wiktor Jędrzejczak; Lela M. Lucy; Marta Olesnyckyj; Zhinuan Yu; Robert Knight; Jerome B. Zeldis; Joan Bladé
PURPOSE The long-term impact of thalidomide plus dexamethasone (thal/dex) as primary therapy for newly diagnosed multiple myeloma (MM) is unknown. The goal of this study was to compare thalidomide plus dexamethasone versus placebo plus dexamethasone (placebo/dex)as primary therapy for newly diagnosed MM. PATIENTS AND METHODS In this double-blind, placebo-controlled trial, patients with untreated symptomatic MM were randomized to thal/dex (arm A) or to placebo plus dexamethasone (dex) (arm B). Patients in arm A received oral thalidomide 50 mg daily, escalated to 100 mg on day 15, and to 200 mg from day 1 of cycle 2 (28-day cycles). Oral dex 40 mg was administered on days 1 through 4, 9 through 12, and 17 through 20 during cycles 1 through 4 and on days 1 through 4 only from cycle 5 onwards. Patients in arm B received placebo and dex, administered as in arm A. The primary end point of the study was time to progression. This study is registered at http://ClinicalTrials.gov (NCT00057564). RESULTS A total of 470 patients were enrolled (235 randomly assigned to thal/dex and 235 to placebo/dex). The overall response rate was significantly higher with thal/dex compared with placebo/dex (63% v 46%), P < .001. Time to progression (TTP) was significantly longer with thal/dex compared with placebo/dex (median, 22.6 v 6.5 months, P < .001). Grade 4 adverse events were more frequent with thal/dex than with placebo/dex (30.3% v 22.8%). CONCLUSION Thal/dex results in significantly higher response rates and significantly prolongs TTP compared with dexamethasone alone in patients with newly diagnosed MM.
Blood | 2009
Jean-Luc Harousseau; Giovanni Martinelli; Wiesław Wiktor Jędrzejczak; Joseph Brandwein; Dominique Bordessoule; Tamas Masszi; Gert J. Ossenkoppele; Julia Alexeeva; Gernot Beutel; Johan Maertens; María-Belén Vidriales; Hervé Dombret; Xavier Thomas; Alan Kenneth Burnett; Tadeusz Robak; Nuriet K. Khuageva; Anatoly Golenkov; Elena Tóthová; Lars Möllgård; Youn C. Park; Annick Bessems; Peter De Porre; Angela Howes
This phase 3, multicenter, open-label study evaluated the efficacy and safety of tipifarnib compared with best supportive care (BSC), including hydroxyurea, as first-line therapy in elderly patients (>or=70 years) with newly diagnosed, de novo, or secondary acute myeloid leukemia. A total of 457 patients were enrolled with 24% 80 years of age or older. Tipifarnib 600 mg orally twice a day was administered for the first 21 consecutive days, in 28-day cycles. The primary endpoint was overall survival. The median survival was 107 days for the tipifarnib arm and 109 days for the BSC arm. The hazard ratio (tipifarnib vs BSC) for overall survival was 1.02 (P value by stratified log-rank test, .843). The complete response rate for tipifarnib in this study (8%) was lower than that observed previously, but with a similar median duration of 8 months. The most frequent grade 3 or 4 adverse events were cytopenias in both arms, slightly more infections (39% vs 33%), and febrile neutropenia (16% vs 10%) seen in the tipifarnib arm. The results of this randomized study showed that tipifarnib treatment did not result in an increased survival compared with BSC, including hydroxyurea. This trial was registered at www.clinicaltrials.gov as #NCT00093990.
Blood | 2015
Michael L. Wang; Kristie A. Blum; Peter Martin; Andre Goy; Rebecca Auer; Brad S. Kahl; Wojciech Jurczak; Ranjana H. Advani; Jorge Romaguera; Michael E. Williams; Jacqueline C. Barrientos; Ewa Chmielowska; John Radford; Stephan Stilgenbauer; Martin Dreyling; Wiesław Wiktor Jędrzejczak; Peter Johnson; Stephen E. Spurgeon; Liang Zhang; Linda Baher; Mei Cheng; Dana Lee; Darrin M. Beaupre; Simon Rule
Ibrutinib, an oral inhibitor of Bruton tyrosine kinase, is approved for patients with mantle cell lymphoma (MCL) who have received one prior therapy. We report the updated safety and efficacy results from the multicenter, open-label phase 2 registration trial of ibrutinib (median 26.7-month follow-up). Patients (N = 111) received oral ibrutinib 560 mg once daily, and those with stable disease or better could enter a long-term extension study. The primary end point was overall response rate (ORR). The median patient age was 68 years (range, 40-84), with a median of 3 prior therapies (range, 1-5). The median treatment duration was 8.3 months; 46% of patients were treated for >12 months, and 22% were treated for ≥2 years. The ORR was 67% (23% complete response), with a median duration of response of 17.5 months. The 24-month progression-free survival and overall survival rates were 31% (95% confidence interval [CI], 22.3-40.4) and 47% (95% CI, 37.1-56.9), respectively. The most common adverse events (AEs) in >30% of patients included diarrhea (54%), fatigue (50%), nausea (33%), and dyspnea (32%). The most frequent grade ≥3 infections included pneumonia (8%), urinary tract infection (4%), and cellulitis (3%). Grade ≥3 bleeding events in ≥2% of patients were hematuria (2%) and subdural hematoma (2%). Common all-grade hematologic AEs were thrombocytopenia (22%), neutropenia (19%), and anemia (18%). The prevalence of infection, diarrhea, and bleeding was highest for the first 6 months of therapy and less thereafter. With longer follow-up, ibrutinib continues to demonstrate durable responses and favorable safety in relapsed/refractory MCL. The trial is registered to www.ClinicalTrials.gov as #NCT01236391.
Journal of Clinical Oncology | 2012
Jerzy Holowiecki; Sebastian Grosicki; Sebastian Giebel; Tadeusz Robak; Slawomira Kyrcz-Krzemien; Aleksander B. Skotnicki; Andrzej Hellmann; Kazimierz Sulek; Anna Dmoszynska; Janusz Kloczko; Wiesław Wiktor Jędrzejczak; Barbara Zdziarska; Krzysztof Warzocha; Krystyna Zawilska; Mieczysław Komarnicki; Marek Kielbinski; Beata Piatkowska-Jakubas; Agnieszka Wierzbowska; Malgorzata Wach; Olga Haus
PURPOSE The goal of this study was to evaluate whether the addition of a purine analog, cladribine or fludarabine, to the standard induction regimen affects the outcome of adult patients with acute myeloid leukemia (AML). PATIENTS AND METHODS A cohort of 652 untreated AML patients with median age 47 years (range, 17 to 60 years) were randomly assigned to receive one of three induction regimens: DA (daunorubicin plus cytarabine), DAC (DA plus cladribine), or DAF (DA plus fludarabine). Postremission treatment was the same for all arms. RESULTS Complete remission rate in the DAC arm was higher compared with the DA arm (67.5% v 56%; P = .01) as a consequence of reduced incidence of resistant disease (21% v 34%; P = .004). There was no significant difference in early outcome between the DAF and DA arms. The probability of overall survival was improved for the DAC arm (45% ± 4% at 3 years) compared with the DA arm (33% ± 4%; P = .02), and leukemia-free survival was comparable. Long-term outcome did not differ significantly for the comparison of the DAF and DA arms. A survival advantage of the DAC arm over the DA arm was observed among patients age 50 years or older (P = .005), those with initial leukocyte count above 50 × 10(9)/L (P = .03), and those with unfavorable karyotype (P = .03). DAF revealed a significant advantage over DA in patients with adverse karyotype (P = .02). CONCLUSION The addition of cladribine to the standard induction regimen is associated with increased rate of complete remission and improved survival of adult patients with AML.
British Journal of Haematology | 2008
Jerzy Holowiecki; Malgorzata Krawczyk-Kulis; Sebastian Giebel; Krystyna Jagoda; Beata Stella-Holowiecka; Beata Piatkowska-Jakubas; Monika Paluszewska; Ilona Seferynska; Krzysztof Lewandowski; Marek Kielbinski; Anna Czyż; Agnieszka Balana-Nowak; Maria Król; Aleksander B. Skotnicki; Wiesław Wiktor Jędrzejczak; Krzysztof Warzocha; Andrzej Lange; Andrzej Hellmann
The treatment of adults with Philadelphia‐negative acute lymphoblastic leukaemia (ALL) depends on the presence of risk factors including age, white blood cell count, immunophenotype and time to complete remission. In recent years, status of minimal residual disease (MRD) has been postulated as an additional risk criterion. This study prospectively evaluated the significance of MRD. Patients were treated with a uniform Polish Adult Leukemia Group (PALG) 4‐2002 protocol. MRD status was assessed after induction and consolidation by multiparametric flow cytometry. Out of 132 patients included (age, 17–60 years), 116 patients were suitable for analysis. MRD level ≥0·1% of bone marrow cells after induction was found to be a strong and independent predictor for relapse in the whole study population (P < 0·0001), as well as in the standard risk (SR, P = 0·0003) and high‐risk (P = 0·008) groups. The impact of MRD after consolidation on outcome was not significant. The combination of MRD status with conventional risk stratification system identified a subgroup of patients allocated to the SR group with MRD <0·1% after induction who had a very low risk of relapse of 9% at 3 years as opposed to 71% in the remaining subjects (P = 0·001). We conclude that MRD evaluation after induction should be considered with conventional risk criteria for treatment decisions in adult ALL.
Blood | 2016
Paul G. Richardson; Vânia Tm Hungria; Sung-Soo Yoon; Meral Beksac; Meletios A. Dimopoulos; Ashraf Elghandour; Wiesław Wiktor Jędrzejczak; Andreas Guenther; Thanyaphong Na Nakorn; Noppadol Siritanaratkul; Robert Schlossman; Jian Hou; Philippe Moreau; Sagar Lonial; Jae Hoon Lee; Hermann Einsele; Monika Sopala; Bourras-Rezki Bengoudifa; Claudia Corrado; Florence Binlich; Jesús F. San-Miguel
Panobinostat is a potent pan-deacetylase inhibitor that affects the growth and survival of multiple myeloma (MM) cells through alteration of epigenetic mechanisms and protein metabolism. Panobinostat plus bortezomib and dexamethasone (PAN-BTZ-Dex) led to a significant increase in progression-free survival (PFS) vs placebo plus bortezomib and dexamethasone (Pbo-BTZ-Dex) in patients with relapsed or relapsed and refractory MM in the phase 3 PANORAMA 1 trial. This subgroup analysis evaluated outcomes in patients in the PANORAMA 1 trial based on prior treatment: a prior immunomodulatory drug (IMiD; n = 485), prior bortezomib plus an IMiD (n = 193), and ≥2 prior regimens including bortezomib and an IMiD (n = 147). Median PFS with PAN-BTZ-Dex vs Pbo-BTZ-Dex across subgroups was as follows: prior IMiD (12.3 vs 7.4 months; hazard ratio [HR], 0.54; 95% confidence interval [CI], 0.43-0.68), prior bortezomib plus IMiD (10.6 vs 5.8 months; HR, 0.52; 95% CI, 0.36-0.76), and ≥2 prior regimens including bortezomib and an IMiD (12.5 vs 4.7 months; HR, 0.47; 95% CI, 0.31-0.72). Common grade 3/4 adverse events and laboratory abnormalities in patients who received PAN-BTZ-Dex across the prior treatment groups included thrombocytopenia, lymphopenia, neutropenia, diarrhea, and asthenia/fatigue. Incidence of on-treatment deaths among patients who received prior bortezomib and an IMiD (regardless of number of prior regimens) was similar between treatment arms. This analysis demonstrated a clear PFS benefit of 7.8 months with PAN-BTZ-Dex among patients who received ≥2 prior regimens including bortezomib and an IMiD, a population with limited treatment options and poorer prognosis. This trial was registered at www.clinicaltrials.gov as #NCT01023308.
Bone Marrow Transplantation | 2007
B Nasilowska-Adamska; Piotr Rzepecki; J Manko; Anna Czyż; Miroslaw Markiewicz; I Federowicz; A Tomaszewska; B Piatkowska-Jakubas; A Wrzesien-Kus; M Bieniaszewska; D Duda; Richard Szydlo; Kazimierz Hałaburda; A Szczepinski; Andrzej Lange; A Hellman; Tadeusz Robak; Aleksander B. Skotnicki; Wiesław Wiktor Jędrzejczak; Jan Walewski; Jerzy Holowiecki; Mieczysław Komarnicki; Anna Dmoszynska; Krzysztof Warzocha; B Marianska
In this multicenter study, we assessed the use of palifermin (recombinant human–keratinocyte growth factor 1) in the prevention of oral mucositis (OM) and acute GvHD (aGvHD) induced by a hematopoietic stem cell transplant (HSCT). Fifty-three patients with hematological diseases received three doses of palifermin (60 μg/kg once daily i.v.) pre- and post-conditioning regimens (total six doses). A retrospective control group of 53 transplant patients received no palifermin. There was a significant reduction in the incidence of OM of WHO (World Health Organization) grades 1–4 (58 vs 94%, P<0.001), 3–4 (13 vs 43%, P<0.001) and the median duration of OM (4 vs 9 days, P<0.001) in the palifermin group compared to the control group. The incidence of analgesics (32 vs 75.5%, P<0.001), opioid analgesics (24 vs 64%, P<0.001) and total parenteral nutrition (11 vs 45%, P<0.001) was also significantly reduced. The analysis of distribution of affected organs revealed that aGvHD was less prevalent in the palifermin group (P=0.036). There was no significant difference in the onset of any OM after HSCT, time to engraftment and length of hospitalization between groups. The drug was generally well tolerated and safe. Our results suggest that the use of palifermin reduces OM and probably aGvHD after HSCT, but a randomized trial is needed.
Blood | 2012
Tadeusz Robak; Jerzy Windyga; Jacek Treliński; Mario von Depka Prondzinski; Aristoteles Giagounidis; Chantal Doyen; Ann Janssens; María Teresa Álvarez-Román; Isidro Jarque; Javier Loscertales; Gloria Pérez Rus; Andrzej Hellmann; Wiesław Wiktor Jędrzejczak; Lana M. Golubovic; Dusica Celeketic; Andrei Cucuianu; Emanuil Gheorghita; Mihaela Lazaroiu; Ofer Shpilberg; Dina Attias; Elena Karyagina; Kalinina Svetlana; Kateryna Vilchevska; Nichola Cooper; Kate Talks; Mukhyaprana Prabhu; Prasad Sripada; T. P. R. Bharadwaj; Henrik Næsted; Niels Jørgen Østergaard Skartved
Rozrolimupab, a recombinant mixture of 25 fully human RhD-specific monoclonal antibodies, represents a new class of recombinant human antibody mixtures. In a phase 1 or 2 dose escalation study, RhD(+) patients (61 subjects) with primary immune thrombocytopenia received a single intravenous dose of rozrolimupab ranging from 75 to 300 μg/kg. The primary outcome was the occurrence of adverse events. The principal secondary outcome was the effect on platelet levels 7 days after the treatment. The most common adverse events were headache and pyrexia, mostly mild, and reported in 20% and 13% of the patients, respectively, without dose relationship. Rozrolimupab caused an expected transient reduction of hemoglobin concentration in the majority of the patients. At the dose of 300 μg/kg platelet responses, defined as platelet count ≥ 30 × 10(9)/L and an increase in platelet count by > 20 × 10(9)/L from baseline were observed after 72 hours and persisted for at least 7 days in 8 of 13 patients (62%). Platelet responses were observed within 24 hours in 23% of patients and lasted for a median of 14 days. Rozrolimupab was well tolerated and elicited rapid platelet responses in patients with immune thrombocytopenia and may be a useful alternative to plasma-derived products. This trial is registered at www.clinicaltrials.gov as #NCT00718692.
European Journal of Haematology | 2003
A. Wrzesien-Kus; Tadeusz Robak; Ewa Lech-Marańda; Agnieszka Wierzbowska; Anna Dmoszynska; M. Kowal; Jerzy Holowiecki; Slawomira Kyrcz-Krzemien; Sebastian Grosicki; S. Maj; Andrzej Hellmann; Aleksander B. Skotnicki; Wiesław Wiktor Jędrzejczak
Objectives: To evaluate the efficacy and toxicity of cladribine (2‐chlorodeoxyadenosine, 2‐CdA), cytarabine (Ara‐C), and granulocyte‐colony stimulating factor (G‐CSF) (CLAG) regimen in refractory acute myeloid leukemia (AML) in the multicenter phase II study.