Network


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

Hotspot


Dive into the research topics where Wieslaw Wiktor-Jedrzejczak is active.

Publication


Featured researches published by Wieslaw Wiktor-Jedrzejczak.


The New England Journal of Medicine | 2012

Continuous Lenalidomide Treatment for Newly Diagnosed Multiple Myeloma

Antonio Palumbo; Roman Hájek; Michel Delforge; Martin Kropff; Maria Teresa Petrucci; John Catalano; Heinz Gisslinger; Wieslaw Wiktor-Jedrzejczak; Mamia Zodelava; Katja Weisel; Nicola Cascavilla; Genadi Iosava; Michele Cavo; Janusz Kloczko; Joan Bladé; Meral Beksac; Ivan Spicka; Torben Plesner; Joergen Radke; Christian Langer; Dina Ben Yehuda; Alessandro Corso; Lindsay Herbein; Zhinuan Yu; Jay Mei; Christian Jacques; Meletios A. Dimopoulos

BACKGROUND Lenalidomide has tumoricidal and immunomodulatory activity against multiple myeloma. This double-blind, multicenter, randomized study compared melphalan-prednisone-lenalidomide induction followed by lenalidomide maintenance (MPR-R) with melphalan-prednisone-lenalidomide (MPR) or melphalan-prednisone (MP) followed by placebo in patients 65 years of age or older with newly diagnosed multiple myeloma. METHODS We randomly assigned patients who were ineligible for transplantation to receive MPR-R (nine 4-week cycles of MPR followed by lenalidomide maintenance therapy until a relapse or disease progression occurred [152 patients]) or to receive MPR (153 patients) or MP (154 patients) without maintenance therapy. The primary end point was progression-free survival. RESULTS The median follow-up period was 30 months. The median progression-free survival was significantly longer with MPR-R (31 months) than with MPR (14 months; hazard ratio, 0.49; P<0.001) or MP (13 months; hazard ratio, 0.40; P<0.001). Response rates were superior with MPR-R and MPR (77% and 68%, respectively, vs. 50% with MP; P<0.001 and P=0.002, respectively, for the comparison with MP). The progression-free survival benefit associated with MPR-R was noted in patients 65 to 75 years of age but not in those older than 75 years of age (P=0.001 for treatment-by-age interaction). After induction therapy, a landmark analysis showed a 66% reduction in the rate of progression with MPR-R (hazard ratio for the comparison with MPR, 0.34; P<0.001) that was age-independent. During induction therapy, the most frequent adverse events were hematologic; grade 4 neutropenia was reported in 35%, 32%, and 8% of the patients in the MPR-R, MPR, and MP groups, respectively. The 3-year rate of second primary tumors was 7% with MPR-R, 7% with MPR, and 3% with MP. CONCLUSIONS MPR-R significantly prolonged progression-free survival in patients with newly diagnosed multiple myeloma who were ineligible for transplantation, with the greatest benefit observed in patients 65 to 75 years of age. (Funded by Celgene; MM-015 ClinicalTrials.gov number, NCT00405756.).


Blood | 2011

Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse

Mark Levis; Farhad Ravandi; Eunice S. Wang; Maria R. Baer; Alexander E. Perl; Steven Coutre; Harry P. Erba; Robert K. Stuart; Michele Baccarani; Larry D. Cripe; Martin S. Tallman; Giovanna Meloni; Lucy A. Godley; Amelia Langston; S. Amadori; Ian D. Lewis; Arnon Nagler; Richard Stone; Karen Yee; Anjali S. Advani; Dan Douer; Wieslaw Wiktor-Jedrzejczak; Gunnar Juliusson; Mark R. Litzow; Stephen H. Petersdorf; Miguel A. Sanz; Hagop M. Kantarjian; Takashi Sato; Lothar Tremmel; Debra M. Bensen-Kennedy

In a randomized trial of therapy for FMS-like tyrosine kinase-3 (FLT3) mutant acute myeloid leukemia in first relapse, 224 patients received chemotherapy alone or followed by 80 mg of the FLT3 inhibitor lestaurtinib twice daily. Endpoints included complete remission or complete remission with incomplete platelet recovery (CR/CRp), overall survival, safety, and tolerability. Correlative studies included pharmacokinetics and analysis of in vivo FLT3 inhibition. There were 29 patients with CR/CRp in the lestaurtinib arm and 23 in the control arm (26% vs 21%; P = .35), and no difference in overall survival between the 2 arms. There was evidence of toxicity in the lestaurtinib-treated patients, particularly those with plasma levels in excess of 20 μM. In the lestaurtinib arm, FLT3 inhibition was highly correlated with remission rate, but target inhibition on day 15 was achieved in only 58% of patients receiving lestaurtinib. Given that such a small proportion of patients on this trial achieved sustained FLT3 inhibition in vivo, any conclusions regarding the efficacy of combining FLT3 inhibition with chemotherapy are limited. Overall, lestaurtinib treatment after chemotherapy did not increase response rates or prolong survival of patients with FLT3 mutant acute myeloid leukemia in first relapse. This study is registered at www.clinicaltrials.gov as #NCT00079482.


Bone Marrow Transplantation | 2014

Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice.

Tapani Ruutu; Alois Gratwohl; T.J.M. de Witte; Boris Afanasyev; J. Apperley; Andrea Bacigalupo; Francesco Dazzi; Peter Dreger; Rafael F. Duarte; J Finke; Laurent Garderet; Hildegard Greinix; Ernst Holler; N Kröger; A. Lawitschka; M. Mohty; Arnon Nagler; Jakob Passweg; Olle Ringdén; Gérard Socié; Jorge Sierra; Anna Sureda; Wieslaw Wiktor-Jedrzejczak; Alejandro Madrigal; Dietger Niederwieser

GVHD remains the major impediment to broader application of allogeneic haematopoietic SCT. It can be prevented completely, but at the expense of other complications, rejection, relapse or delayed immune reconstitution. No optimal prevention or treatment method has been defined. This is reflected by enormous heterogeneity in approaches in Europe. Retrospective comparisons between different policies, although warranted, do not give definite answers. In order to improve the present situation, an European Group for Blood and Marrow Transplantation and the European LeukemiaNet working group has developed in a Delphi-like approach recommendations for prophylaxis and treatment of GVHD in the most common allogeneic transplant setting, transplantation from an HLA-identical sibling or unrelated donor for standard risk malignant disease. The working group proposes these guidelines to be adopted as routine standard in transplantation centres and to be used as comparator in systematic studies evaluating the advantages and disadvantages of practices differing from these recommendations.


Blood | 2012

Rac2-MRC-cIII-generated ROS cause genomic instability in chronic myeloid leukemia stem cells and primitive progenitors

Margaret Nieborowska-Skorska; Piotr Kopinski; Regina Ray; Grazyna Hoser; Danielle Ngaba; Sylwia Flis; Kimberly Cramer; Mamatha M. Reddy; Mateusz Koptyra; Tyrone Penserga; Eliza Glodkowska-Mrowka; Elisabeth Bolton; Tessa L. Holyoake; Connie J. Eaves; Sabine Cerny-Reiterer; Peter Valent; Andreas Hochhaus; Timothy P. Hughes; Heiko van der Kuip; Martin Sattler; Wieslaw Wiktor-Jedrzejczak; Charles C. Richardson; Adrienne M. Dorrance; Tomasz Stoklosa; David A. Williams; Tomasz Skorski

Chronic myeloid leukemia in chronic phase (CML-CP) is induced by BCR-ABL1 oncogenic tyrosine kinase. Tyrosine kinase inhibitors eliminate the bulk of CML-CP cells, but fail to eradicate leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) displaying innate and acquired resistance, respectively. These cells may accumulate genomic instability, leading to disease relapse and/or malignant progression to a fatal blast phase. In the present study, we show that Rac2 GTPase alters mitochondrial membrane potential and electron flow through the mitochondrial respiratory chain complex III (MRC-cIII), thereby generating high levels of reactive oxygen species (ROS) in CML-CP LSCs and primitive LPCs. MRC-cIII-generated ROS promote oxidative DNA damage to trigger genomic instability, resulting in an accumulation of chromosomal aberrations and tyrosine kinase inhibitor-resistant BCR-ABL1 mutants. JAK2(V617F) and FLT3(ITD)-positive polycythemia vera cells and acute myeloid leukemia cells also produce ROS via MRC-cIII. In the present study, inhibition of Rac2 by genetic deletion or a small-molecule inhibitor and down-regulation of mitochondrial ROS by disruption of MRC-cIII, expression of mitochondria-targeted catalase, or addition of ROS-scavenging mitochondria-targeted peptide aptamer reduced genomic instability. We postulate that the Rac2-MRC-cIII pathway triggers ROS-mediated genomic instability in LSCs and primitive LPCs, which could be targeted to prevent the relapse and malignant progression of CML.


Cancer | 2014

Results of a randomized, double-blind study of romiplostim versus placebo in patients with low/intermediate-1–risk myelodysplastic syndrome and thrombocytopenia

Aristoteles Giagounidis; Ghulam J. Mufti; Pierre Fenaux; Mikkael A. Sekeres; Jeff Szer; Uwe Platzbecker; Andrea Kuendgen; Gianluca Gaidano; Wieslaw Wiktor-Jedrzejczak; Kuolung Hu; Paul Woodard; Allen S. Yang; Hagop M. Kantarjian

Thrombocytopenia in patients with myelodysplastic syndrome (MDS) is associated with shortened survival and an increased risk of evolution to acute myeloid leukemia (AML). In this study, the authors evaluated the efficacy of romiplostim in patients who had thrombocytopenia with low‐risk/intermediate‐1–risk MDS.


Bone Marrow Transplantation | 2011

Independence of exogenous insulin following immunoablation and stem cell reconstitution in newly diagnosed diabetes type I

Emilian Snarski; A Milczarczyk; T Torosian; M Paluszewska; E Urbanowska; M Król; P Boguradzki; K Jedynasty; E Franek; Wieslaw Wiktor-Jedrzejczak

Type I diabetes mellitus is a metabolic disease caused by chronic immune attack against the insulin-producing cells of the pancreas. It has recently been shown that the clinical course of this disease can be interrupted by immune ablation and PBSCT. In this report, we describe our experience with this treatment modality in a series of eight cases. Patients with newly diagnosed type I diabetes were received treatment consisting of two to three plasmaphereses, hematopoietic stem cell mobilization with CY and G-CSF, collection of at least 3 × 106 per kg of CD34+ cells, and conditioning with CY and anti-thymocyte globulin followed by stem cell infusion. All patients became independent of exogenous insulin after the transplantation. One patient resumed low-dose insulin 7 months after transplantation. Six out of eight patients were given acarbose for better glycemic control after transplantation. All patients exhibited good glycemic control: the average HbA1c concentrations were 12.3% at diagnosis, and 5.6 and 6.2% at 3 and 6 months after transplantation, respectively. We conclude that at least temporary independence of exogenous insulin can be achieved in type I diabetes patients following immunoablation and reconstitution of the immune system with autologous PBSCs.


Leukemia | 2016

Frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the European ENEST1st study

Andreas Hochhaus; Gianantonio Rosti; Nicholas C.P. Cross; Juan-Luis Steegmann; P. le Coutre; Gert J. Ossenkoppele; Ljubomir Petrov; Tamas Masszi; Andrzej Hellmann; Laimonas Griskevicius; Wieslaw Wiktor-Jedrzejczak; Delphine Rea; Daniel Coriu; Tim H. Brümmendorf; Kimmo Porkka; G. Saglio; Günther Gastl; Markus Müller; Peter Schuld; P. Di Matteo; Angela Pellegrino; Luca Dezzani; F-X Mahon; Michele Baccarani; Frank Giles

The Evaluating Nilotinib Efficacy and Safety in Clinical Trials as First-Line Treatment (ENEST1st) study included 1089 patients with newly diagnosed chronic myeloid leukemia in chronic phase. The rate of deep molecular response (MR4 (BCR-ABL1⩽0.01% on the International Scale or undetectable BCR-ABL1 with ⩾10 000 ABL1 transcripts)) at 18 months was evaluated as the primary end point, with molecular responses monitored by the European Treatment and Outcome Study network of standardized laboratories. This analysis was conducted after all patients had completed 24 months of study treatment (80.9% of patients) or discontinued early. In patients with typical BCR-ABL1 transcripts and ⩽3 months of prior imatinib therapy, 38.4% (404/1052) achieved MR4 at 18 months. Six patients (0.6%) developed accelerated or blastic phase, and 13 (1.2%) died. The safety profile of nilotinib was consistent with that of previous studies, although the frequencies of some nilotinib-associated adverse events were lower (for example, rash, 21.4%). Ischemic cardiovascular events occurred in 6.0% of patients. Routine monitoring of lipid and glucose levels was not mandated in the protocol. These results support the use of frontline nilotinib, particularly when achievement of a deep molecular response (a prerequisite for attempting treatment-free remission in clinical trials) is a treatment goal.


Journal of International Medical Research | 2010

Update on the Incidence of Metamizole Sodium-Induced Blood Dyscrasias in Poland

Grzegorz W. Basak; Joanna Drozd-Sokołowska; Wieslaw Wiktor-Jedrzejczak

Metamizole sodium (metamizole) is a popular non-opioid analgesic and a common non-prescription product in Poland. Controversy exists regarding the level of risk of agranulocytosis or aplastic anaemia associated with its use. Two previous pharmacovigilance studies conducted in Poland found the risk was low. Twenty-four of the 25 haematology centres that provide specialist care for the 30 million adults in Poland participated in this prospective 12-month study. Twenty-one cases of agranulocytosis, 48 of aplastic anaemia, 15 of neutropenia and 11 of pancytopenia were reported. Of these cases, three (two agranulocytosis; one aplastic anaemia) were judged as being possibly related to metamizole. Crude estimates of the rate of agranulocytosis and aplastic anaemia associated with metamizole were 0.16 and 0.08 cases/million person-days of use, respectively. Ongoing national safety surveillance in Poland shows that, despite the possibility of drug-induced blood dyscrasias with metamizole, the risk is very low.


Clinical Infectious Diseases | 2017

Fecal Microbiota Transplantation in Patients With Blood Disorders Inhibits Gut Colonization With Antibiotic-Resistant Bacteria: Results of a Prospective, Single-Center Study

Jaroslaw Bilinski; Pawel Grzesiowski; Nikolaj Sorensen; Krzysztof Madry; Jacek Muszyński; Katarzyna Robak; Marta Wróblewska; Tomasz Dzieciatkowski; Grażyna Dulny; Jadwiga Dwilewicz-Trojaczek; Wieslaw Wiktor-Jedrzejczak; Grzegorz W. Basak

Background Patients with blood disorders colonized with antibiotic-resistant bacteria (ARB) are prone to systemic infections that are difficult to treat. Reintroduction of commensal bacteria in a murine model of enterococcal colonization of the gut can lead to eradication of enterococci. We hypothesized that fecal microbiota transplantation (FMT) could be used to eradicate ARB in humans. Methods Participants colonized with ARB were treated with intraduodenal FMT according to a prospective protocol (NCT02461199). The primary endpoint was complete ARB decolonization at 1 month after FMT. Secondary endpoints included safety assessment and partial ARB decolonization. Microbiome sequencing was performed to investigate the influence of microbial composition of the transplanted material on the outcome of FMT. Results Twenty-five FMTs were performed in 20 participants (including 40% who had neutropenia) who were colonized by a median of 2 (range, 1-4) strains of ARB. The primary endpoint was reached in 15/25 (60%) of the FMTs and more frequently in cases in which there was no periprocedural use of antibiotics (79% vs 36%, P < .05). Among participants, 15/20 (75%) experienced complete ARB decolonization. There were no severe adverse events, and partial ARB decolonization was observed in 20/25 (80%) of the FMTs. The microbiota composition analysis revealed higher abundance of Barnesiella spp., Bacteroides, and Butyricimonas and greater bacterial richness in the fecal material, resulting in eradication of Klebsiella pneumoniae compared with nonresponders. Conclusions FMT in patients with blood disorders is safe and promotes eradication of ARB from the gastrointestinal tract. Clinical Trials Registration NCT02461199.


Leukemia | 2017

Treatment-free remission following frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the ENESTfreedom study

Andreas Hochhaus; Tamas Masszi; Frank Giles; Jerry Radich; David M. Ross; M T Gómez Casares; Andrzej Hellmann; Jesper Stentoft; Eibhlin Conneally; Valentin Garcia-Gutierrez; Norbert Gattermann; Wieslaw Wiktor-Jedrzejczak; P. le Coutre; Bruno Martino; Susanne Saussele; Hans D. Menssen; Weiping Deng; Nancy Krunic; Véronique Bédoucha; G. Saglio

The single-arm, phase 2 ENESTfreedom trial assessed the potential for treatment-free remission (TFR; i.e., the ability to maintain a molecular response after stopping therapy) following frontline nilotinib treatment. Patients with Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase with MR4.5 (BCR-ABL1⩽0.0032% on the International Scale (BCR-ABL1IS)) and ⩾2 years of frontline nilotinib therapy were enrolled. Patients with sustained deep molecular response during the 1-year nilotinib consolidation phase were eligible to stop treatment and enter the TFR phase. Patients with loss of major molecular response (MMR; BCR-ABL1IS⩽0.1%) during the TFR phase reinitiated nilotinib. In total, 215 patients entered the consolidation phase, of whom 190 entered the TFR phase. The median duration of nilotinib before stopping treatment was 43.5 months. At 48 weeks after stopping nilotinib, 98 patients (51.6%; 95% confidence interval, 44.2–58.9%) remained in MMR or better (primary end point). Of the 86 patients who restarted nilotinib in the treatment reinitiation phase after loss of MMR, 98.8% and 88.4%, respectively, regained MMR and MR4.5 by the data cutoff date. Consistent with prior reports of imatinib-treated patients, musculoskeletal pain-related events were reported in 24.7% of patients in the TFR phase (consolidation phase, 16.3%).

Collaboration


Dive into the Wieslaw Wiktor-Jedrzejczak's collaboration.

Top Co-Authors

Avatar

Grzegorz W. Basak

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Elżbieta Urbanowska

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tigran Torosian

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Emilian Snarski

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Król

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Krzysztof Warzocha

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Małgorzata Król

Medical University of Warsaw

View shared research outputs
Researchain Logo
Decentralizing Knowledge