Network


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

Hotspot


Dive into the research topics where Philipp Erben is active.

Publication


Featured researches published by Philipp Erben.


Leukemia | 2012

Early molecular and cytogenetic response is predictive for long-term progression-free and overall survival in chronic myeloid leukemia (CML)

Benjamin Hanfstein; Markus Müller; Rüdiger Hehlmann; Philipp Erben; Michael Lauseker; A. Fabarius; S Schnittger; Claudia Haferlach; Gudrun Göhring; Ulrike Proetel; H. J. Kolb; S. W. Krause; Wolf-Karsten Hofmann; Jörg Schubert; H. Einsele; Jolanta Dengler; Matthias Hänel; C. Falge; Lothar Kanz; Andreas Neubauer; Michael Kneba; Frank Stegelmann; Michael Pfreundschuh; Cornelius F. Waller; S Branford; Timothy P. Hughes; Karsten Spiekermann; Markus Pfirrmann; Joerg Hasford; Susanne Saußele

In the face of competing first-line treatment options for CML, early prediction of prognosis on imatinib is desirable to assure favorable survival or otherwise consider the use of a second-generation tyrosine kinase inhibitor (TKI). A total of 1303 newly diagnosed imatinib-treated patients (pts) were investigated to correlate molecular and cytogenetic response at 3 and 6 months with progression-free and overall survival (PFS, OS). The persistence of BCR-ABL transcript levels >10% according to the international scale (BCR-ABLIS) at 3 months separated a high-risk group (28% of pts; 5-year OS: 87%) from a group with >1–10% BCR-ABLIS (41% of pts; 5-year OS: 94%; P=0.012) and from a group with ⩽1% BCR-ABLIS (31% of pts; 5-year OS: 97%; P=0.004). Cytogenetics identified high-risk pts by >35% Philadelphia chromosome-positive metaphases (Ph+, 27% of pts; 5-year OS: 87%) compared with ⩽35% Ph+ (73% of pts; 5-year OS: 95%; P=0.036). At 6 months, >1% BCR-ABLIS (37% of pts; 5-year OS: 89%) was associated with inferior survival compared with ⩽1% (63% of pts; 5-year OS: 97%; P<0.001) and correspondingly >0% Ph+ (34% of pts; 5-year OS: 91%) compared with 0% Ph+ (66% of pts; 5-year OS: 97%; P=0.015). Treatment optimization is recommended for pts missing these landmarks.


Journal of Clinical Oncology | 2009

Impact of Baseline BCR-ABL Mutations on Response to Nilotinib in Patients With Chronic Myeloid Leukemia in Chronic Phase

Timothy P. Hughes; Giuseppe Saglio; Susan Branford; Simona Soverini; Dong-Wook Kim; Martin C. Müller; Giovanni Martinelli; Jorge Cortes; Lan Beppu; Enrico Gottardi; Dongho Kim; Philipp Erben; Yaping Shou; Ariful Haque; Neil Gallagher; Jerald P. Radich; Andreas Hochhaus

PURPOSE Nilotinib is a second-generation tyrosine kinase inhibitor indicated for the treatment of patients with chronic myeloid leukemia (CML) in chronic phase (CP; CML-CP) and accelerated phase (AP; CML-AP) who are resistant to or intolerant of prior imatinib therapy. In this subanalysis of a phase II study of nilotinib in patients with imatinib-resistant or imatinib-intolerant CML-CP, the occurrence and impact of baseline and newly detectable BCR-ABL mutations were assessed. PATIENTS AND METHODS Baseline mutation data were assessed in 281 (88%) of 321 patients with CML-CP in the phase II nilotinib registration trial. RESULTS Among imatinib-resistant patients, the frequency of mutations at baseline was 55%. After 12 months of therapy, major cytogenetic response (MCyR) was achieved in 60%, complete cytogenetic response (CCyR) in 40%, and major molecular response (MMR) in 29% of patients without baseline mutations versus 49% (P = .145), 32% (P = .285), and 22% (P = .366), respectively, of patients with mutations. Responses in patients who harbored mutations with high in vitro sensitivity to nilotinib (50% inhibitory concentration [IC(50)] <or= 150 nM) or mutations with unknown nilotinib sensitivity were equivalent to those responses for patients without mutations (not significant). Patients with mutations that were less sensitive to nilotinib in vitro (IC(50) > 150 nM; Y253H, E255V/K, F359V/C) had less favorable responses, as 13%, 43%, and 9% of patients with each of these mutations, respectively, achieved MCyR; none achieved CCyR. CONCLUSION For most patients with imatinib resistance and with mutations, nilotinib offers a substantial probability of response. However, mutational status at baseline may influence response. Less sensitive mutations that occurred at three residues defined in this study, as well as the T315I mutation, may be associated with less favorable responses to nilotinib.


Blood | 2009

Dasatinib treatment of chronic phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations

Martin C. Müller; Jorge Cortes; Dong-Wook Kim; Brian J. Druker; Philipp Erben; Ricardo Pasquini; Susan Branford; Timothy P. Hughes; Jerald P. Radich; Lynn Ploughman; Jaydip Mukhopadhyay; Andreas Hochhaus

Dasatinib is a BCR-ABL inhibitor with 325-fold higher potency than imatinib against unmutated BCR-ABL in vitro. Imatinib failure is commonly caused by BCR-ABL mutations. Here, dasatinib efficacy was analyzed in patients recruited to phase 2/3 trials with chronic-phase chronic myeloid leukemia with or without BCR-ABL mutations after prior imatinib. Among 1043 patients, 39% had a preexisting BCR-ABL mutation, including 48% of 805 patients with imatinib resistance or suboptimal response. Sixty-threedifferent BCR-ABL mutations affecting 49 amino acids were detected at baseline, with G250, M351, M244, and F359 most frequently affected. After 2 years of follow-up, dasatinib treatment of imatinib-resistant patients with or without a mutation resulted in notable response rates (complete cytogenetic response: 43% vs 47%) and durable progression-free survival (70% vs 80%). High response rates were achieved with different mutations except T315I, including highly imatinib-resistant mutations in the P-loop region. Impaired responses were observed with some mutations with a dasatinib median inhibitory concentration (IC(50)) greater than 3nM; among patients with mutations with lower or unknown IC(50), efficacy was comparable with those with no mutation. Overall, dasatinib has durable efficacy in patients with or without BCR-ABL mutations. All trials were registered at http://www.clinicaltrials.gov as NCT00123474, NCT00101660, and NCT00103844.


Leukemia | 2009

Harmonization of molecular monitoring of CML therapy in Europe.

Markus Müller; Nicholas C.P. Cross; Philipp Erben; Thomas Schenk; Benjamin Hanfstein; Thomas Ernst; R. Hehlmann; S Branford; Giuseppe Saglio; Andreas Hochhaus

The high efficacy of the standard treatment of chronic myeloid leukemia (CML) with imatinib has prompted the need for accurate methods to monitor response at levels below the landmark of complete cytogenetic remission. Quantification of BCR–ABL transcripts has proven to be the most sensitive method available, and has shown prognostic impact with regard to progression-free survival. Until recently, variations in methods used to quantify BCR–ABL made it difficult to compare results between laboratories. An international program is now underway to harmonize the reporting of results according to an international scale (IS). In this review, we consider the background to the IS and the progress that has been made to date, with a particular focus on ongoing harmonization efforts in Europe. We provide recommendations for the propagation of the IS by national or regional laboratory networks.


Journal of Clinical Oncology | 2009

Dasatinib in the treatment of chronic myeloid leukemia in accelerated phase after imatinib failure: the START a trial.

Jane F. Apperley; Jorge Cortes; Dong-Wook Kim; Lydia Roy; Gail J. Roboz; Gianantonio Rosti; Eduardo Bullorsky; Elisabetta Abruzzese; Andreas Hochhaus; Dominik Heim; Carmino Antonio de Souza; Richard A. Larson; Jeffrey H. Lipton; H. Jean Khoury; Hyeoung Joon Kim; Christian Sillaber; Timothy P. Hughes; Philipp Erben; Jan Van Tornout; Richard Stone

PURPOSE Patients with chronic myelogenous leukemia in accelerated phase (CML-AP) that is resistant or intolerant to imatinib have limited therapeutic options. Dasatinib, a potent inhibitor of BCR-ABL and SRC-family kinases, has efficacy in patients with CML-AP who have experienced treatment failure with imatinib. We now report follow-up data from the full patient cohort of 174 patients enrolled onto a phase II trial to provide a more complete assessment of the efficacy and safety of dasatinib in this population. PATIENTS AND METHODS Patients with imatinib-resistant (n = 161) or -intolerant (n = 13) CML-AP received dasatinib 70 mg orally twice daily. Results At a median follow-up of 14.1 months (treatment duration, 0.1 to 21.7 months), major and complete hematologic responses were attained by 64% and 45% of patients, respectively, and major and complete cytogenetic responses were achieved in 39% and 32% of patients, respectively. Responses were achieved irrespective of imatinib status (resistant or intolerant), prior stem-cell transplantation, or the presence of prior BCR-ABL mutation. The 12-month progression-free survival and overall survival rates were 66% and 82%, respectively. Dasatinib was generally well tolerated; the most frequent nonhematologic severe treatment-related adverse event was diarrhea (52%; grade 3 to 4, 8%). Cytopenias were common, including grade 3 to 4 neutropenia (76%) and thrombocytopenia (82%). Pleural effusion occurred in 27% of patients (grade 3 to 4, 5%). CONCLUSION Dasatinib is effective in patients with CML-AP after imatinib treatment failure.


Journal of Clinical Oncology | 2010

Sustained Molecular Response With Interferon Alfa Maintenance After Induction Therapy With Imatinib Plus Interferon Alfa in Patients With Chronic Myeloid Leukemia

Andreas Burchert; Martin C. Müller; Philippe Kostrewa; Philipp Erben; Tilman Bostel; Simone Liebler; Rüdiger Hehlmann; Andreas Neubauer; Andreas Hochhaus

PURPOSE Imatinib induces sustained remissions in patients with chronic myelogenous leukemia (CML), but fails to eradicate CML stem cells. This is of major concern regarding the issues of cure, long-term imatinib tolerability, and imatinib resistance. We therefore asked whether interferon alfa-2a (IFN) alone could maintain molecular remissions achieved by a prior combination therapy with imatinib and IFN. PATIENTS AND METHODS Imatinib therapy was stopped in 20 patients who had concomitantly been pretreated with imatinib and IFN for a median of 2.4 years (range, 0.2 to 4.8 years) and 2.5 years (range, 0.2 to 4.9 years), respectively. After imatinib discontinuation, remission status was monitored monthly by quantitative analysis of the peripheral-blood BCR-ABL mRNA levels using real-time polymerase chain reaction. Proteinase-3 expression and proteinase-3-specific cytotoxic T cells (CTLs) were longitudinally measured to assess putative markers of IFN response. RESULTS With a median time of 2.4 years after imatinib withdrawal (range, 0.5 to 4.0 years), 15 (75%) of 20 patients remained in remission. The number of patients in complete molecular remission increased under IFN from two patients at baseline to five patients after 2 years. Relapses occurred in five patients within 0.4 years (range, 0.2 to 0.8 years), but patients underwent rescue treatment with imatinib, re-establishing molecular remission. IFN therapy was associated with an increase in the expression of leukemia-associated antigen proteinase 3 and induction of proteinase-3-specific CTLs. CONCLUSION Treatment with IFN enables discontinuation of imatinib in most patients after prior imatinib/IFN combination therapy and may result in improved molecular response. Induction of a proteinase-3-specific CTL response by IFN may contribute to this effect.


Leukemia | 2007

Recurrent finding of the FIP1L1-PDGFRA fusion gene in eosinophilia-associated acute myeloid leukemia and lymphoblastic T-cell lymphoma

Georgia Metzgeroth; Christoph Walz; Joannah Score; Reiner Siebert; Susanne Schnittger; Claudia Haferlach; Henning D. Popp; T Haferlach; Philipp Erben; Jürgen Mix; Markus Müller; H Beneke; Lothar Müller; F del Valle; Walter E. Aulitzky; G Wittkowsky; Norbert Schmitz; C Schulte; K Müller-Hermelink; E Hodges; Sean Whittaker; F Diecker; H Döhner; Peter Schuld; R. Hehlmann; Andreas Hochhaus; Nicholas C.P. Cross; Andreas Reiter

The FIP1L1-PDGFRA fusion gene has been described in patients with eosinophilia-associated myeloproliferative disorders (Eos-MPD). Here, we report on seven FIP1L1-PDGFRA-positive patients who presented with acute myeloid leukemia (AML, n=5) or lymphoblastic T-cell non-Hodgkin-lymphoma (n=2) in conjunction with AML or Eos-MPD. All patients were male, the median age was 58 years (range, 40–66). AML patients were negative for common mutations of FLT3, NRAS, NPM1, KIT, MLL and JAK2; one patient revealed a splice mutation of RUNX1 exon 7. Patients were treated with imatinib (100 mg, n=5; 400 mg, n=2) either as monotherapy (n=2), as maintenance treatment after intensive chemotherapy (n=3) or in overt relapse 43 and 72 months, respectively, after primary diagnosis and treatment of FIP1L1-PDGFRA-positive disease (n=2). All patients are alive, disease-free and in complete hematologic and complete molecular remission after a median time of 20 months (range, 9–36) on imatinib. The median time to achievement of complete molecular remission was 6 months (range, 1–14). We conclude that all eosinophilia-associated hematological malignancies should be screened for the presence of the FIP1L1-PDGFRA fusion gene as they are excellent candidates for treatment with tyrosine kinase inhibitors even if they present with an aggressive phenotype such as AML.


Blood | 2013

Comprehensive mutational profiling in advanced systemic mastocytosis

Juliana Schwaab; Susanne Schnittger; Karl Sotlar; Christoph Walz; Alice Fabarius; Markus Pfirrmann; Alexander Kohlmann; Vera Grossmann; Manja Meggendorfer; Hans-Peter Horny; Peter Valent; Mohamad Jawhar; Martina Teichmann; Georgia Metzgeroth; Philipp Erben; Thomas Ernst; Andreas Hochhaus; Torsten Haferlach; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Andreas Reiter

To explore mechanisms contributing to the clinical heterogeneity of systemic mastocytosis (SM) and to suboptimal responses to diverse therapies, we analyzed 39 KIT D816V mutated patients with indolent SM (n = 10), smoldering SM (n = 2), SM with associated clonal hematologic nonmast cell lineage disorder (SM-AHNMD, n = 5), and aggressive SM (n = 15) or mast cell leukemia (n = 7) with (n = 18) or without (n = 4) AHNMD for additional molecular aberrations. We applied next-generation sequencing to investigate ASXL1, CBL, IDH1/2, JAK2, KRAS, MLL-PTD, NPM1, NRAS, TP53, SRSF2, SF3B1, SETBP1, U2AF1 at mutational hotspot regions, and analyzed complete coding regions of EZH2, ETV6, RUNX1, and TET2. We identified additional molecular aberrations in 24/27 (89%) patients with advanced SM (SM-AHNMD, 5/5; aggressive SM/mast cell leukemia, 19/22) whereas only 3/12 (25%) indolent SM/smoldering SM patients carried one additional mutation each (U2AF1, SETBP1, CBL) (P < .001). Most frequently affected genes were TET2, SRSF2, ASXL1, CBL, and RUNX1. In advanced SM, 21/27 patients (78%) carried ≥3 mutations, and 11/27 patients (41%) exhibited ≥5 mutations. Overall survival was significantly shorter in patients with additional aberrations as compared to those with KIT D816V only (P = .019). We conclude that biology and prognosis in SM are related to the pattern of mutated genes that are acquired during disease evolution.


Haematologica | 2008

Dynamics of BCR-ABL mutated clones prior to hematologic or cytogenetic resistance to imatinib.

Thomas Ernst; Philipp Erben; Martin C. Müller; Peter Paschka; Thomas Schenk; Jana Hoffmann; Sebastian Kreil; Paul La Rosée; Rüdiger Hehlmann; Andreas Hochhaus

Mutations of the BCR-ABL tyrosine domain represent a major cause of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia. The appearance of BCR-ABL mutations during imatinib therapy likely indicates imminent relapse, and their early detection might allow reconsideration of the therapeutic strategy. Background Mutations of the BCR-ABL tyrosine kinase domain constitute a major cause of resistance to tyrosine kinase inhibitors in patients with chronic myeloid leukemia. We sought to improve the diagnostic armamentarium by screening and to analyze the dynamics of mutated clones in chronic myeloid leukemia patients who experienced hematologic or cytogenetic relapse. Design and Methods Ninety-five patients who relapsed during imatinib therapy were screened for BCR-ABL kinase domain mutations using sensitive denaturing high-performance liquid chromatography (D-HPLC) and direct sequencing. To investigate the dynamics of mutated clones D-HPLC was applied to 453 cDNA samples tracking back from relapse towards the start of imatinib therapy. Results Twenty-two different point mutations affecting 18 amino acids were detectable in 46/79 (58%) and in 7/16 patients (44%) with hematologic or cytogenetic relapse, respectively. A deletion of 81 nucleotides (del248-274) of ABL exon 4 was observed in two patients. Three patients had exclusively single nucleotide polymorphisms (K247R, T315T, E499E, n=1 each) within the BCR-ABL kinase domain. In patients harboring mutations, hematologic relapse occurred after a median of 12.9 months (range, 0.9–44.2), and BCR-ABL mutations first became detectable at a median of 5.8 months (range, 0–30.5) after starting imatinib therapy (p<0.0001). Nine patients showed evidence of BCR-ABL mutations prior to imatinib therapy (T315I, n=4; M351T, n=3; M244V and Y253H, n=1 each). Conclusions We conclude that: (i) D-HPLC is a sensitive method for screening for BCR-ABL mutations before and during therapy with tyrosine kinase inhibitors; (ii) the occurrence of BCR-ABL mutations during imatinib therapy is predictive of relapse; (iii) mutations may be detectable several months before relapse, and (iv) the sensitive detection of small numbers of mutated clones could provide clinical benefit by triggering early therapeutic interventions.


British Journal of Haematology | 2008

Safety and efficacy of imatinib in chronic eosinophilic leukaemia and hypereosinophilic syndrome – a phase-II study

Georgia Metzgeroth; Christoph Walz; Philipp Erben; Helena Popp; Annette Schmitt-Graeff; Claudia Haferlach; Alice Fabarius; Susanne Schnittger; David Grimwade; Nicholas C.P. Cross; Riidiger Hehlmann; Andreas Hochhaus; Andreas Reiter

This study evaluated the efficacy and safety of imatinib in chronic eosinophilic leukaemia (CEL, n = 23) and hypereosinophilic syndrome (HES, n = 13). In CEL with FIP1L1‐PDGFRA (n = 16) or various PDGFRB fusion genes (n = 5), complete haematological remission (CHR) was achieved in 95% (20/21) after 3 months. Complete molecular remission (CMR) was seen in 75% (12/16) of cases with FIP1L1‐PDGFRA positive CEL by 6 months, and in 87% (13/15) after 12 months. CMR was achieved in three of five PDGFRB fusion positive patients after 3, 9 and 18 months respectively. All patients are currently on imatinib (100 mg; n = 13, 400 mg; n = 8) and no molecular relapse has yet been observed (median 26·7 months; range, 6·9–39·9). Imatinib was less effective in HES and CEL without known molecular aberration (n = 15); CHR was observed in 40% (6/15) of patients, two patients relapsed after 4·8 and 24·5 months. Three patients died due to imatinib‐resistant progressive CEL (n = 2) or myocardial infarction (n = 1) unrelated to study treatment. Overall, imatinib was well tolerated with a low incidence of grade III/IV toxicities. These data confirmed the long‐term efficacy of imatinib for PDGFR‐rearranged CEL patients, and also showed that a minority of HES cases without known molecular aberrations may benefit from imatinib.

Collaboration


Dive into the Philipp Erben'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
Top Co-Authors

Avatar

Thomas Ernst

University of Southampton

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge