Network


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

Hotspot


Dive into the research topics where Christine von Neuhoff is active.

Publication


Featured researches published by Christine von Neuhoff.


Journal of Clinical Oncology | 2010

Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98.

Christine von Neuhoff; Dirk Reinhardt; Annette Sander; Martin Zimmermann; Jutta Bradtke; David R. Betts; Zuzana Zemanova; Jan Stary; Jean-Pierre Bourquin; Oskar A. Haas; Michael Dworzak; Ursula Creutzig

PURPOSE Because cytogenetic data are essential for risk stratification of childhood acute myeloid leukemia (AML), the impact of chromosomal aberrations is crucial. PATIENTS AND METHODS Data of a large group of patients younger than 18 years treated according to study AML-Berlin-Frankfurt-Münster (BFM) 98 (n = 454), including their cytogenetics, were analyzed. RESULTS The favorable outcome in the subgroups of patients with t(8;21), inv(16), and t(15;17), with an overall survival of 91% (SE, 4%), 92% (SE, 6%), and 87% (SE, 5%), respectively, was confirmed. Within this group, the 5-year probability of event-free survival (pEFS) of all 17 children with t(8;21) and additional aberrations apart from del(9q) or -X/-Y was 100%. As expected, the cytogenetic finding of a complex karyotype (n = 35; pEFS, 33%; SE, 8%) or a monosomy 7 (n = 12; pEFS, 17%; SE, 11%) was associated with a poor outcome. Compared with remaining patients with cytogenetic data (pEFS, 48%; SE, 2%), prognosis in patients with an MLL rearrangement (n = 91) was inferior (pEFS, 34%; SE, 5%; P = .0005). Particularly, children with t(9;11) and additional aberrations (n = 13; pEFS, 31%; SE, 14%) and MLL rearrangements other than t(9;11) and t(11;19) (n = 41; pEFS, 24%; SE, 7%) had an unfavorable outcome. Nine patients with aberrations in 12p showed an adverse prognosis (pEFS, 11%; SE, 10%). The outcome of patients with aberrations of chromosome 5 (n = 13) was better than expected (pEFS, 50%; SE, 13%). CONCLUSION Because the prognostic value of rare recurrent chromosomal aberrations still has to be elucidated, these data will contribute to future risk stratification for the treatment of pediatric AML.


Blood | 2013

Randomized trial comparing liposomal daunorubicin with idarubicin as induction for pediatric acute myeloid leukemia: results from Study AML-BFM 2004

Ursula Creutzig; Martin Zimmermann; Jean-Pierre Bourquin; Michael Dworzak; Gudrun Fleischhack; Norbert Graf; Thomas Klingebiel; Bernhard Kremens; Thomas Lehrnbecher; Christine von Neuhoff; J. Ritter; Annette Sander; André Schrauder; Arend von Stackelberg; Jan Starý; Dirk Reinhardt

Outcomes of patients with acute myeloid leukemia (AML) improve significantly by intensification of induction. To further intensify anthracycline dosage without increasing cardiotoxicity, we compared potentially less cardiotoxic liposomal daunorubicin (L-DNR) to idarubicin at a higher-than-equivalent dose (80 vs 12 mg/m(2) per day for 3 days) during induction. In the multicenter therapy-optimization trial AML-BFM 2004, 521 of 611 pediatric patients (85%) were randomly assigned to L-DNR or idarubicin induction. Five-year results in both treatment arms were similar (overall survival 76% ± 3% [L-DNR] vs 75% ± 3% [idarubicin], Plogrank = .65; event-free survival [EFS] 59% ± 3% vs 53% ± 3%, Plogrank = .25; cumulative incidence of relapse 29% ± 3% vs 31% ± 3%, P(Gray) = .75), as were EFS results for standard (72% ± 5% vs 68% ± 5%, Plogrank = .47) and high-risk (51% ± 4% vs 46% ± 4%, Plogrank = .45) patients. L-DNR resulted in significantly better probability of EFS in patients with t(8;21). Overall, treatment-related mortality was lower with L-DNR than idarubicin (2/257 vs 10/264 patients, P = .04). Grade 3/4 cardiotoxicity was rare after induction (4 L-DNR vs 5 idarubicin). Only 1 L-DNR and 3 idarubicin patients presented with subclinical or mild cardiomyopathy during follow-up. In conclusion, at the given dose, L-DNR has overall antileukemic activity comparable to idarubicin, promises to be more active in subgroups, and causes less treatment-related mortality. This trial was registered at www.clinicaltrials.gov as #NCT00111345.


Blood | 2011

Analysis of GATA1 mutations in Down syndrome transient myeloproliferative disorder and myeloid leukemia

Kate A. Alford; Katarina Reinhardt; Catherine Garnett; Alice Norton; Katarina Böhmer; Christine von Neuhoff; Alexandra Kolenova; Emanuele Marchi; Jan-Henning Klusmann; Irene Roberts; Henrik Hasle; Dirk Reinhardt; Paresh Vyas

Children with Down syndrome (DS) up to the age of 4 years are at a 150-fold excess risk of developing myeloid leukemia (ML-DS). Approximately 4%-5% of newborns with DS develop transient myeloproliferative disorder (TMD). Blast cell structure and immunophenotype are similar in TMD and ML-DS. A mutation in the hematopoietic transcription factor GATA1 is present in almost all cases. Here, we show that simple techniques detect GATA1 mutations in the largest series of TMD (n = 134; 88%) and ML-DS (n = 103; 85%) cases tested. Furthermore, no significant difference in the mutational spectrum between the 2 disorders was seen. Thus, the type of GATA1 sequence mutation is not a reliable tool and is not prognostic of which patients with TMD are probable to develop ML-DS.


Pediatric Blood & Cancer | 2011

CNS Irradiation in Pediatric Acute Myleoid Leukemia: Equal Results by 12 or 18 Gy in Studies AML-BFM98 and 2004

Ursula Creutzig; Martin Zimmermann; Jean-Pierre Bourquin; Michael Dworzak; Gudrun Fleischhack; Christine von Neuhoff; Annette Sander; André Schrauder; Arend von Stackelberg; J. Ritter; Jan Starý; Dirk Reinhardt

The impact of preventive central nervous system irradiation (CNS‐RT) in childhood acute myeloid leukemia (AML) is still discussed. As results of study AML‐BFM87 revealed an increased risk for relapse when CNS‐RT was not performed, studies AML‐BFM98 and ‐2004 randomized CNS‐RT of 18 or 12 Gy in order to evaluate the efficacy of the lower dose and to reduce late effects.


Haematologica | 2014

The prognostic significance of early treatment response in pediatric relapsed acute myeloid leukemia: results of the international study Relapsed AML 2001/01

Ursula Creutzig; Martin Zimmermann; Michael Dworzak; Brenda Gibson; Rienk Tamminga; Jonas Abrahamsson; Shau-Yin Ha; Henrik Hasle; Alexey Maschan; Yves Bertrand; Guy Leverger; Christine von Neuhoff; Bassem I. Razzouk; Carmelo Rizzari; Petr Smisek; Owen P. Smith; Batia Stark; Dirk Reinhardt; G.J.L. Kaspers

The prognostic significance of early response to treatment has not been reported in relapsed pediatric acute myeloid leukemia. In order to identify an early and easily applicable prognostic factor allowing subsequent treatment modifications, we assessed leukemic blast counts in the bone marrow by morphology on days 15 and 28 after first reinduction in 338 patients of the international Relapsed-AML2001/01 trial. Both day 15 and day 28 status was classified as good (≤20% leukemic blasts) in 77% of patients. The correlation between day 15 and 28 blast percentages was significant, but not strong (Spearman correlation coefficient = 0.49, P<0.001). Survival probability decreased in a stepwise fashion along with rising blast counts at day 28. Patients with bone marrow blast counts at this time-point of ≤5%, 6–10%, 11–20% and >20% had 4-year probabilities of survival of 52%±3% versus 36%±10% versus 21%±9% versus 14%±4%, respectively, P<0.0001; this trend was not seen for day 15 results. Multivariate analysis showed that early treatment response at day 28 had the strongest prognostic significance, superseding even time to relapse (< or ≥12 months). In conclusion, an early response to treatment, measured on day 28, is a strong and independent prognostic factor potentially useful for treatment stratification in pediatric relapsed acute myeloid leukemia. This study was registered with ISRCTN code: 94206677.


Haematologica | 2014

Normal karyotype is a poor prognostic factor in myeloid leukemia of Down syndrome: A retrospective, international study

Marjolein Blink; Martin Zimmermann; Christine von Neuhoff; Dirk Reinhardt; Valerie de Haas; Henrik Hasle; Maureen M. O'Brien; Batia Stark; Julie Tandonnet; Andrea Pession; Daniel K. L. Cheuk; Kazuko Kudo; Takashi Taga; Jeffrey E. Rubnitz; Irén Haltrich; Walentyna Balwierz; Rob Pieters; Erik Forestier; Bertil Johansson; Marry M. van den Heuvel-Eibrink; C. Michel Zwaan

Myeloid leukemia of Down syndrome has a better prognosis than sporadic pediatric acute myeloid leukemia. Most cases of myeloid leukemia of Down syndrome are characterized by additional cytogenetic changes besides the constitutional trisomy 21, but their potential prognostic impact is not known. We, therefore, conducted an international retrospective study of clinical characteristics, cytogenetics, treatment, and outcome of 451 children with myeloid leukemia of Down syndrome. All karyotypes were centrally reviewed before assigning patients to subgroups. The overall 7-year event-free survival for the entire cohort was 78% (±2%), with the overall survival rate being 79% (±2%), the cumulative incidence of relapse 12% (±2%), and the cumulative incidence of toxic death 7% (±1%). Outcome estimates showed large differences across the different cytogenetic subgroups. Based on the cumulative incidence of relapse, we could risk-stratify patients into two groups: cases with a normal karyotype (n=103) with a higher cumulative incidence of relapse (21%±4%) than cases with an aberrant karyotype (n=255) with a cumulative incidence of relapse of 9% (±2%) (P=0.004). Multivariate analyses revealed that white blood cell count ≥20×109/L and age >3 years were independent predictors for poor event-free survival, while normal karyotype independently predicted inferior overall survival, event-free survival, and relapse-free survival. In conclusion, this study showed large differences in outcome within patients with myeloid leukemia of Down syndrome and identified novel prognostic groups that predicted clinical outcome and hence may be used for stratification in future treatment protocols.


Cancer | 2016

Changes in cytogenetics and molecular genetics in acute myeloid leukemia from childhood to adult age groups.

Ursula Creutzig; Martin Zimmermann; Dirk Reinhardt; Mareike Rasche; Christine von Neuhoff; Tamara Alpermann; Michael Dworzak; Karolína Perglerová; Zuzana Zemanova; Joelle Tchinda; Jutta Bradtke; Christian Thiede; Claudia Haferlach

To obtain better insight into the biology of acute myeloid leukemia (AML) in various age groups, this study focused on the genetic changes occurring during a lifetime.


Pediatric Blood & Cancer | 2017

First experience of the AML-Berlin-Frankfurt-Münster group in pediatric patients with standard-risk acute promyelocytic leukemia treated with arsenic trioxide and all-trans retinoid acid

Ursula Creutzig; Michael Dworzak; Konrad Bochennek; Jörg Faber; Christian Flotho; Norbert Graf; Udo Kontny; Claudia Rossig; Irene Schmid; Arend von Stackelberg; Jans-Enno Mueller; Christine von Neuhoff; Dirk Reinhardt; Nils von Neuhoff

Recently, studies in adults with acute promyelocytic leukemia (APL) showed high cure rates in low‐risk patients treated with all‐trans retinoid acid (ATRA) and arsenic trioxide (ATO), while toxicities were significantly reduced compared to the standard treatment with ATRA and chemotherapy. Here we report about first experience with 11 pediatric patients with low‐risk APL treated with ATRA and ATO. All patients stayed in molecular remission. All suffered from hyperleukocytosis. Two patients experienced reversible severe side effects. One suffered from osteonecroses at both femurs, seizures, as well as posterior reversible encephalopathy syndrome, the other patient had an abducens paresis.


Blood | 2017

Therapy reduction in patients with Down syndrome and myeloid leukemia: the international ML-DS 2006 trial

Madita Uffmann; Mareike Rasche; Martin Zimmermann; Christine von Neuhoff; Ursula Creutzig; Michael Dworzak; Lenie Scheffers; Henrik Hasle; C. Michel Zwaan; Dirk Reinhardt; Jan-Henning Klusmann

Children with myeloid leukemia associated with Down syndrome (ML-DS) have superior outcome compared with non-DS patients, but suffer from higher constitutional cytotoxic drug susceptibility. We analyzed the outcome of 170 pediatric patients with ML-DS enrolled in the prospective, multicenter, open-label, nonrandomized ML-DS 2006 trial by Nordic Society for Pediatric Hematology and Oncology (NOPHO), Dutch Childhood Oncology Group (DCOG), and Acute Myeloid Leukemia-Berlin-Frankfurt-Münster (AML-BFM) study group. Compared with the historical control arm (reduced-intensity protocol for ML-DS patients from the AML-BFM 98 trial), treatment intensity was reduced by lowering the cumulative dose of etoposide (950 to 450 mg/m2) and intrathecal central nervous system prophylaxis while omitting maintenance therapy. Still, 5-year overall survival (89% ± 3% vs 90% ± 4%; Plog-rank = .64), event-free survival (EFS; 87% ± 3% vs 89% ± 4%; Plog-rank = .71), and cumulative incidence of relapse/nonresponse (CIR/NR; 6% ± 3% vs 6% ± 2%; PGray = .03) did not significantly differ between the ML-DS 2006 trial and the historical control arm. Poor early treatment response (5-year EFS, 58% ± 16% vs 88% ± 3%; Plog rank = .0008) and gain of chromosome 8 (CIR/NR, 16% ± 7% vs 3% ± 2%, PGray = .02; 5-year EFS, 73% ± 8% vs 91% ± 4%, Plog rank = .018) were identified as independent prognostic factors predicting a worse EFS. Five of 7 relapsed patients (71%) with cytogenetic data had trisomy 8. Our study reveals prognostic markers for children with ML-DS and illustrates that reducing therapy did not impair excellent outcome. The trial was registered at EudraCT as #2007-006219-2.


Blood Advances | 2018

Low-dose cytarabine to prevent myeloid leukemia in children with Down syndrome: TMD Prevention 2007 study

Marius Flasinski; Kira Scheibke; Martin Zimmermann; Ursula Creutzig; Katarina Reinhardt; Femke Verwer; Valerie de Haas; V H J van der Velden; Christine von Neuhoff; C. Michel Zwaan; Dirk Reinhardt; Jan-Henning Klusmann

Approximately 5% to 10% of children with Down syndrome (DS) are diagnosed with transient myeloproliferative disorder (TMD). Approximately 20% of these patients die within 6 months (early death), and another 20% to 30% progress to myeloid leukemia (ML-DS) within their first 4 years of life. The aim of the multicenter, nonrandomized, historically controlled TMD Prevention 2007 trial was to evaluate the impact of low-dose cytarabine treatment on survival and prevention of ML-DS in patients with TMD. Patients received cytarabine (1.5 mg/kg for 7 days) in case of TMD-related symptoms at diagnosis (high white blood cell count, ascites, liver dysfunction, hydrops fetalis) or detection of minimal residual disease (MRD) 8 weeks after diagnosis. The 5-year probability of event-free and overall survival of 102 enrolled TMD patients was 72 ± 5% and 91 ± 3%, respectively. In patients eligible for treatment because of symptoms (n = 43), we observed a significantly lower cumulative incidence (CI) of early death as compared with symptomatic patients in the historical control (n = 45) (12 ± 5% vs 33 ± 7%, PGray = .02). None of the asymptomatic patients in the current study suffered early death. However, the treatment of symptomatic or MRD-positive patients did not result in a significantly lower CI of ML-DS (25 ± 7% [treated] vs 14 ± 7% [untreated], PGray = .34 [per protocol analysis]; historical control: 22 ± 4%, PGray = .55). Thus, low-dose cytarabine treatment helped to reduce TMD-related mortality when compared with the historical control but was insufficient to prevent progression to ML-DS. This trial was registered at EudraCT as #2006-002962-20.

Collaboration


Dive into the Christine von Neuhoff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Zimmermann

Leibniz University of Hanover

View shared research outputs
Top Co-Authors

Avatar

Michael Dworzak

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ursula Creutzig

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ursula Creutzig

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Michel Zwaan

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge