Network


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

Hotspot


Dive into the research topics where Jana Hof is active.

Publication


Featured researches published by Jana Hof.


Nature Medicine | 2013

Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL

Gannie Tzoneva; Arianne Perez-Garcia; Zachary Carpenter; Hossein Khiabanian; Valeria Tosello; Maddalena Allegretta; Elisabeth Paietta; Janis Racevskis; Jacob M. Rowe; Martin S. Tallman; Maddalena Paganin; Giuseppe Basso; Jana Hof; Renate Kirschner-Schwabe; Teresa Palomero; Raul Rabadan; Adolfo A. Ferrando

Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Despite intensive chemotherapy, 20% of pediatric patients and over 50% of adult patients with ALL do not achieve a complete remission or relapse after intensified chemotherapy, making disease relapse and resistance to therapy the most substantial challenge in the treatment of this disease. Using whole-exome sequencing, we identify mutations in the cytosolic 5′-nucleotidase II gene (NT5C2), which encodes a 5′-nucleotidase enzyme that is responsible for the inactivation of nucleoside-analog chemotherapy drugs, in 20/103 (19%) relapse T cell ALLs and 1/35 (3%) relapse B-precursor ALLs. NT5C2 mutant proteins show increased nucleotidase activity in vitro and conferred resistance to chemotherapy with 6-mercaptopurine and 6-thioguanine when expressed in ALL lymphoblasts. These results support a prominent role for activating mutations in NT5C2 and increased nucleoside-analog metabolism in disease progression and chemotherapy resistance in ALL.


Journal of Clinical Oncology | 2011

Mutations and Deletions of the TP53 Gene Predict Nonresponse to Treatment and Poor Outcome in First Relapse of Childhood Acute Lymphoblastic Leukemia

Jana Hof; Stefanie Krentz; Claudia van Schewick; Shabnam Shalapour; Peter Rhein; Leonid Karawajew; Wolf-Dieter Ludwig; Karl Seeger; Günter Henze; Arend von Stackelberg; Christian Hagemeier; Cornelia Eckert; Renate Kirschner-Schwabe

PURPOSE In the clinical management of children with relapsed acute lymphoblastic leukemia (ALL), treatment resistance remains a major challenge. Alterations of the TP53 gene are frequently associated with resistance to chemotherapy, but their significance in relapsed childhood ALL has remained controversial because of small studies. PATIENTS AND METHODS Therefore, we systematically studied 265 first-relapse patients enrolled in the German Acute Lymphoblastic Leukemia Relapse Berlin-Frankfurt-Mü nster 2002 (ALL-REZ BFM 2002) trial for sequence and copy number alterations of the TP53 gene by using direct sequencing and multiplex ligation-dependent probe amplification. RESULTS We observed copy number and sequence alterations of TP53 in 12.4% (27 of 218) of patients with B-cell precursor ALL and 6.4% (three of 47) of patients with T-cell ALL relapse. Backtracking to initial ALL in 23 matched samples revealed that 54% of all TP53 alterations were gained at relapse. Within B-cell precursor ALL, TP53 alterations were consistently associated with nonresponse to chemotherapy (P < .001) and poor event-free survival (P < .001) and overall survival rates (P = .002). TP53 alterations also had a significant impact on survival within intermediate-risk (S2) and high-risk (S3/S4) relapse patients (P = .007 and P = .019, respectively). This prognostic significance of TP53 alterations was confirmed in multivariate analysis. Besides their clinical impact, TP53 alterations were associated with a higher fraction of leukemic cells in S/G(2)-M phase of the cell cycle at relapse diagnosis. CONCLUSION Alterations of the TP53 gene are of particular importance in the relapse stage of childhood ALL, in which they independently predict high risk of treatment failure in a significant number of patients. Therefore, they will aid in future risk assessment of children with ALL relapse.


Blood | 2014

Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition

Julie Irving; Elizabeth Matheson; Lynne Minto; Helen Blair; Marian Case; Christina Halsey; Isabella Swidenbank; Frida Ponthan; Renate Kirschner-Schwabe; Stefanie Groeneveld-Krentz; Jana Hof; James M. Allan; Christine J. Harrison; Josef Vormoor; Arend von Stackelberg; Cornelia Eckert

For most children who relapse with acute lymphoblastic leukemia (ALL), the prognosis is poor, and there is a need for novel therapies to improve outcome. We screened samples from children with B-lineage ALL entered into the ALL-REZ BFM 2002 clinical trial (www.clinicaltrials.gov, #NCT00114348) for somatic mutations activating the Ras pathway (KRAS, NRAS, FLT3, and PTPN11) and showed mutation to be highly prevalent (76 from 206). Clinically, they were associated with high-risk features including early relapse, central nervous system (CNS) involvement, and specifically for NRAS/KRAS mutations, chemoresistance. KRAS mutations were associated with a reduced overall survival. Mutation screening of the matched diagnostic samples found many to be wild type (WT); however, by using more sensitive allelic-specific assays, low-level mutated subpopulations were found in many cases, suggesting that they survived up-front therapy and subsequently emerged at relapse. Preclinical evaluation of the mitogen-activated protein kinase kinase 1/2 inhibitor selumetinib (AZD6244, ARRY-142886) showed significant differential sensitivity in Ras pathway-mutated ALL compared with WT cells both in vitro and in an orthotopic xenograft model engrafted with primary ALL; in the latter, reduced RAS-mutated CNS leukemia. Given these data, clinical evaluation of selumetinib may be warranted for Ras pathway-mutated relapsed ALL.


Leukemia | 2013

Prognostic value of genetic alterations in children with first bone marrow relapse of childhood B-cell precursor acute lymphoblastic leukemia.

S Krentz; Jana Hof; A. Mendioroz; R. Vaggopoulou; Petra Dörge; Claudio Lottaz; Julia C. Engelmann; T. W. L. Groeneveld; K Seeger; Christian Hagemeier; Günter Henze; C Eckert; A von Stackelberg; Renate Kirschner-Schwabe

Despite risk-adapted treatment, survival of children with relapse of acute lymphoblastic leukemia (ALL) remains poor compared with that of patients with initial diagnosis of ALL. Leukemia-associated genetic alterations may provide novel prognostic factors to refine present relapse treatment strategies. Therefore, we investigated the clinical relevance of 13 recurrent genetic alterations in 204 children treated uniformly for relapsed B-cell precursor ALL according to the ALL-REZ BFM 2002 protocol. The most common alterations were deletions of CDKN2A/2B, IKZF1, PAX5, ETV6, fusion of ETV6-RUNX1 and deletions and/or mutations of TP53. Multivariate analysis identified IKZF1 deletion and TP53 alteration as independent predictors of inferior outcome (P=0.002 and P=0.001). Next, we investigated how both alterations can improve the established risk stratification in relapsed ALL. Intermediate-risk relapse patients with low minimal residual disease are currently considered to have a good prognosis. In this group, deletion of IKZF1 and alteration of TP53 identify patients with significantly inferior outcome (P<0.001). In high-risk relapse patients, deletion of IKZF1 is strongly predictive of a second relapse after stem cell transplantation (P<0.001). We conclude that IKZF1 and TP53 represent relevant prognostic factors that should be considered in future risk assessment of children with relapsed ALL to indicate treatment intensification or intervention.


Haematologica | 2014

The activating STAT5B N642H mutation is a common abnormality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse

Obul R. Bandapalli; Stephanie Schuessele; Joachim B. Kunz; Tobias Rausch; Adrian M. Stütz; Noa Tal; Ifat Geron; Nava Gershman; Shai Izraeli; Juliane Eilers; Nina Vaezipour; Renate Kirschner-Schwabe; Jana Hof; Arend von Stackelberg; Martin Schrappe; Martin Stanulla; Martin Zimmermann; Rolf Koehler; Smadar Avigad; Rupert Handgretinger; Viktoras Frismantas; Jean Pierre Bourquin; Beat C. Bornhauser; Jan O. Korbel; Martina U. Muckenthaler; Andreas E. Kulozik

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes that accounts for approximately 15% of pediatric acute lymphoblastic leukemias. A variety of genetic events affecting cellular processes such as the cell cycle, differentiation and survival have been identified in


Haematologica | 2015

Pediatric T-cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation

Joachim B. Kunz; Tobias Rausch; Obul R. Bandapalli; Juliane Eilers; Paulina Pechanska; Stephanie Schuessele; Yassen Assenov; Adrian M. Stütz; Renate Kirschner-Schwabe; Jana Hof; Cornelia Eckert; Arend von Stackelberg; Martin Schrappe; Martin Stanulla; Rolf Koehler; Smadar Avigad; Sarah Elitzur; Rupert Handgretinger; Vladimir Benes; Joachim Weischenfeldt; Jan O. Korbel; Martina U. Muckenthaler; Andreas E. Kulozik

Relapsed precursor T-cell acute lymphoblastic leukemia is characterized by resistance against chemotherapy and is frequently fatal. We aimed at understanding the molecular mechanisms resulting in relapse of T-cell acute lymphoblastic leukemia and analyzed 13 patients at first diagnosis, remission and relapse by whole exome sequencing, targeted ultra-deep sequencing, multiplex ligation dependent probe amplification and DNA methylation array. Compared to primary T-cell acute lymphoblastic leukemia, in relapse the number of single nucleotide variants and small insertions and deletions approximately doubled from 11.5 to 26. Targeted ultra-deep sequencing sensitively detected subclones that were selected for in relapse. The mutational pattern defined two types of relapses. While both are characterized by selection of subclones and acquisition of novel mutations, ‘type 1’ relapse derives from the primary leukemia whereas ‘type 2’ relapse originates from a common pre-leukemic ancestor. Relapse-specific changes included activation of the nucleotidase NT5C2 resulting in resistance to chemotherapy and mutations of epigenetic modulators, exemplified by SUZ12, WHSC1 and SMARCA4. While mutations present in primary leukemia and in relapse were enriched for known drivers of leukemia, relapse-specific changes revealed an association with general cancer-promoting mechanisms. This study thus identifies mechanisms that drive progression of pediatric T-cell acute lymphoblastic leukemia to relapse and may explain the characteristic treatment resistance of this condition.


Haematologica | 2011

High VLA-4 expression is associated with adverse outcome and distinct gene expression changes in childhood B-cell precursor acute lymphoblastic leukemia at first relapse

Shabnam Shalapour; Jana Hof; Renate Kirschner-Schwabe; Lorenz Bastian; Cornelia Eckert; Javier Prada; Günter Henze; Arend von Stackelberg; Karl Seeger

Background Resistance to therapy and subsequent relapse remain major challenges in the clinical management of relapsed childhood acute lymphoblastic leukemia. As the bone marrow environment plays an important role in survival and chemotherapy resistance of leukemia cells by activating different signaling pathways, such as the VLA-4 and PI3K/Akt pathways, we studied the prognostic and biological impact of VLA-4 expression in leukemia cells from children with relapsed B-cell precursor acute lymphoblastic leukemia and its influence on the sensitivity of the leukemia cells to drugs. Design and Methods VLA-4 expression was quantified by real-time polymerase chain reaction in leukemia cells from 56 patients with relapsed acute lymphoblastic leukemia enrolled in the ALL-REZ BFM 2002 trial of the Berlin-Frankfurt-Münster study group. Gene expression changes related to VLA-4 expression were investigated by microarray-based mRNA profiling. The effect of VLA-4 signaling on proliferation and drug resistance was studied in co-cultures of leukemia and stromal cells. Results High expression of VLA-4 at first relapse was associated with adverse prognostic factors, poor molecular response to therapy and significantly worse probabilities of event-free and overall survival. VLA-4 expression was an independent prognostic parameter. Comparing gene expression profiles of leukemia cells with high versus low VLA-4 expression, we identified 27 differentially expressed genes primarily involved in the PI3K/Akt, ephrin and Rho GTPase pathways. Blocking of VLA-4 signaling in combination with cytarabine treatment abolished the growth supportive effect of stromal cells. Conclusions Our results show that high VLA-4 expression is a marker of poor prognosis and a potential therapeutic target in children with relapsed acute lymphoblastic leukemia and confirm that cellular interactions and biological effects related to VLA-4 play a decisive role in the survival of leukemia cells and response to therapy.


Blood Cancer Journal | 2017

Identification of a genetically defined ultra-high-risk group in relapsed pediatric T-lymphoblastic leukemia

Paulina Richter-Pechanska; Joachim B. Kunz; Jana Hof; Martin Zimmermann; Tobias Rausch; Obul R. Bandapalli; E Orlova; G Scapinello; J C Sagi; Martin Stanulla; Martin Schrappe; Gunnar Cario; Renate Kirschner-Schwabe; Cornelia Eckert; Vladimir Benes; Jan O. Korbel; Martina U. Muckenthaler; Andreas E. Kulozik

In the search for genes that define critical steps of relapse in pediatric T-cell acute lymphoblastic leukemia (T-ALL) and can serve as prognostic markers, we performed targeted sequencing of 313 leukemia-related genes in 214 patients: 67 samples collected at the time of relapse and 147 at initial diagnosis. As relapse-specific genetic events, we identified activating mutations in NT5C2 (P=0.0001, Fishers exact test), inactivation of TP53 (P=0.0007, Fishers exact test) and duplication of chr17:q11.2-24.3 (P=0.0068, Fishers exact test) in 32/67 of T-ALL relapse samples. Alterations of TP53 were frequently homozygous events, which significantly correlated with higher rates of copy number alterations in other genes compared with wild-type TP53 (P=0.0004, Mann–Whitneys test). We subsequently focused on mutations with prognostic impact and identified genes governing DNA integrity (TP53, n=8; USP7, n=4; MSH6, n=4), having key roles in the RAS signaling pathway (KRAS, NRAS, n=8), as well as IL7R (n=4) and CNOT3 (n=4) to be exclusively mutated in fatal relapses. These markers recognize 24/49 patients with a second event. In 17 of these patients with mostly refractory relapse and dire need for efficient treatment, we identified candidate targets for personalized therapy with p53 reactivating compounds, MEK inhibitors or JAK/STAT-inhibitors that may be incorporated in future treatment strategies.


Haematologica | 2017

NOTCH1 mutation, TP53 alteration and myeloid antigen expression predict outcome heterogeneity in children with first relapse of T-cell acute lymphoblastic leukemia

Jana Hof; Corinne Kox; Stefanie Groeneveld-Krentz; Obul R. Bandapalli; Leonid Karawajew; Katharina Schedel; Joachim B. Kunz; Cornelia Eckert; Wolf-Dieter Ludwig; Richard Ratei; Peter Rhein; Günter Henze; Martina U. Muckenthaler; Andreas E. Kulozik; Arend von Stackelberg; Renate Kirschner-Schwabe

Relapse of T-cell acute lymphoblastic leukemia (T-ALL) has a dismal prognosis, with only 20% of afflicted children surviving.[1][1] Children with relapsed T-ALL are commonly treated within high-risk arms of second-line treatment protocols that include mandatory hematopoietic stem cell


Blood | 2014

Clinical Significance of NT5C2 Mutations in Children with First Relapse of B-Cell Precursor Acute Lymphoblastic Leukemia

Jana Hof; A Szymansky; Arend von Stackelberg; Cornelia Eckert; Renate Kirschner-Schwabe

Collaboration


Dive into the Jana Hof's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge