Christian H Geisler
Copenhagen University Hospital
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Christian H Geisler.
Lancet Oncology | 2015
Mohammed Farooqui; Janet Valdez; Sabrina Martyr; Georg Aue; Nakhle S. Saba; Carsten U. Niemann; Sarah E. M. Herman; Xin Tian; Gerald E. Marti; Susan Soto; Thomas Hughes; Jade Jones; Andrew Lipsky; Stefania Pittaluga; Maryalice Stetler-Stevenson; Constance Yuan; Yuh Shan Lee; Lone Bredo Pedersen; Christian H Geisler; Katherine R. Calvo; Diane C. Arthur; Irina Maric; Richard Childs; Neal S. Young; Adrian Wiestner
BACKGROUND Patients with chronic lymphocytic leukaemia (CLL) with TP53 aberrations respond poorly to first-line chemoimmunotherapy, resulting in early relapse and short survival. We investigated the safety and activity of ibrutinib in previously untreated and relapsed or refractory CLL with TP53 aberrations. METHODS In this investigator-initiated, single-arm phase 2 study, we enrolled eligible adult patients with active CLL with TP53 aberrations at the National Institutes of Health Clinical Center (Bethesda, MD, USA). Patients received 28-day cycles of ibrutinib 420 mg orally once daily until disease progression or the occurrence of limiting toxicities. The primary endpoint was overall response to treatment at 24 weeks in all evaluable patients. This study is registered with ClinicalTrials.gov, number NCT01500733, and is fully enrolled. FINDINGS Between Dec 22, 2011, and Jan 2, 2014, we enrolled 51 patients; 47 had CLL with deletion 17p13.1 and four carried a TP53 mutation in the absence of deletion 17p13.1. All patients had active disease requiring therapy. 35 enrolled patients had previously untreated CLL and 16 had relapsed or refractory disease. Median follow-up was 24 months (IQR 12.9-27.0). 33 previously untreated patients and 15 patients with relapsed or refractory CLL were evaluable for response at 24 weeks. 32 (97%; 95% CI 86-100) of 33 previously untreated patients achieved an objective response, including partial response in 18 patients (55%) and partial response with lymphocytosis in 14 (42%). One patient had progressive disease at 0.4 months. 12 (80%; 95% CI 52-96) of the 15 patients with relapsed or refractory CLL had an objective response: six (40%) achieved a partial response and six (40%) a partial response with lymphocytosis; the remaining three (20%) patients had stable disease. Grade 3 or worse treatment-related adverse events were neutropenia in 12 (24%) patients (grade 4 in one [2%] patient), anaemia in seven (14%) patients, and thrombocytopenia in five (10%) patients (grade 4 in one [2%] patient). Grade 3 pneumonia occurred in three (6%) patients, and grade 3 rash in one (2%) patient. INTERPRETATION The activity and safety profile of single-agent ibrutinib in CLL with TP53 aberrations is encouraging and supports its consideration as a novel treatment option for patients with this high-risk disease in both first-line and second-line settings. FUNDING Intramural Research Program of the National Heart, Lung, and Blood Institute and the National Cancer Institute, Danish Cancer Society, Novo Nordisk Foundation, National Institutes of Health Medical Research Scholars Program, and Pharmacyclics Inc.
European Journal of Cancer | 2002
Niels S. Andersen; Morten Krogh Jensen; P de Nully Brown; Christian H Geisler
This study presents the first large clinical analysis of 105 unselected mantle cell lymphoma (MCL) patients diagnosed from 1992 to 2000 in a well-defined Danish population. The annual incidences were 0.7/100000 for men and 0.2/100000 for women, with no significant change during the study period. Of 97 evaluable cases, 43% achieved a complete response (CR) after initial therapy. The median disease-free (DFS) and overall survival (OS) rates were 15 and 30 months, respectively. In multivariate analysis, splenomegaly (P=0.002), anaemia (P=0.0001) and age (P=0.002), but not the international prognostic index (IPI) and the Ann Arbor staging system, had an independent impact on survival. Moreover, in a sub-analysis of 45 younger MCL patients (<65 years), a trend towards an OS plateau of 58% was observed in cases without splenomegaly and anaemia (n=29). Thus, in contrast to previously suggested prognostic factors, these variables may prove useful for clinical decisions in a significant subset of MCL patients.
Leukemia | 2015
G.D. te Raa; Ingrid A. M. Derks; Veronika Navrkalová; Anna Skowronska; Perry D. Moerland; J van Laar; Ceri Oldreive; H. Monsuur; Martin Trbušek; Jitka Malčíková; M. Lodén; Christian H Geisler; Jennifer Hüllein; Alexander Jethwa; T. Zenz; Šárka Pospíšilová; Tatjana Stankovic; M. H. J. Van Oers; Arnon P. Kater; E Eldering
Mutations or deletions in TP53 or ATM are well-known determinants of poor prognosis in chronic lymphocytic leukemia (CLL), but only account for approximately 40% of chemo-resistant patients. Genome-wide sequencing has uncovered novel mutations in the splicing factor sf3b1, that were in part associated with ATM aberrations, suggesting functional synergy. We first performed detailed genetic analyses in a CLL cohort (n=110) containing ATM, SF3B1 and TP53 gene defects. Next, we applied a newly developed multiplex assay for p53/ATM target gene induction and measured apoptotic responses to DNA damage. Interestingly, SF3B1 mutated samples without concurrent ATM and TP53 aberrations (sole SF3B1) displayed partially defective ATM/p53 transcriptional and apoptotic responses to various DNA-damaging regimens. In contrast, NOTCH1 or K/N-RAS mutated CLL displayed normal responses in p53/ATM target gene induction and apoptosis. In sole SF3B1 mutated cases, ATM kinase function remained intact, and γH2AX formation, a marker for DNA damage, was increased at baseline and upon irradiation. Our data demonstrate that single mutations in sf3b1 are associated with increased DNA damage and/or an aberrant response to DNA damage. Together, our observations may offer an explanation for the poor prognosis associated with SF3B1 mutations.
Biology of Blood and Marrow Transplantation | 2017
Arne Kolstad; Lone Bredo Pedersen; Christian Winther Eskelund; Simon Husby; Kirsten Grønbæk; Mats Jerkeman; Anna Laurell; Riikka Räty; Erkki Elonen; Niels S. Andersen; Peter de Nully Brown; Eva Kimby; Hans Bentzen; Christer Sundström; Mats Ehinger; Marja-Liisa Karjalainen-Lindsberg; Jan Delabie; Elisabeth Ralfkiaer; Unn-Merete Fagerli; Herman Nilsson-Ehle; Grete F. Lauritzsen; Outi Kuittinen; Carsten U. Niemann; Christian H Geisler
The main objectives of the present study were to monitor minimal residual disease (MRD) in the bone marrow of patients with mantle cell lymphoma (MCL) to predict clinical relapse and guide preemptive treatment with rituximab. Among the patients enrolled in 2 prospective trials by the Nordic Lymphoma Group, 183 who had completed autologous stem cell transplantation (ASCT) and in whom an MRD marker had been obtained were included in our analysis. Fresh samples of bone marrow were analyzed for MRD by a combined standard nested and quantitative real-time PCR assay for Bcl-1/immunoglobulin heavy chain gene (IgH) and clonal IgH rearrangements. Significantly shorter progression-free survival (PFS) and overall survival (OS) was demonstrated for patients who were MRD positive pre-ASCT (54 patients) or in the first analysis post-ASCT (23 patients). The median PFS was only 20 months in those who were MRD-positive in the first sample post-ASCT, compared with 142 months in the MRD-negative group (P < .0001). OS was 75% at 10 years and median not reached in the MRD-negative group, compared with only 35 months in the MRD-positive group (P < .0001). Of the 86 patients (47%) who remained in continuous molecular remission, 73% were still in clinical remission after 10 years. For all patients, the median time from ASCT to first molecular relapse was 55 months, with a continuous occurrence of late molecular relapses. Fifty-eight patients who experienced MRD relapse received rituximab as preemptive treatment on 1 or more occasions, and in this group, the median time from first molecular relapse to clinical relapse was 55 months. In most cases, rituximab converted patients to MRD negativity (87%), but many patients became MRD-positive again later during follow-up (69%). By multivariate analysis, high-risk Mantle Cell Lymphoma International Prognostic Index score and positive MRD status pre-ASCT predicted early molecular relapse. In conclusion, preemptive rituximab treatment converts patients to MRD negativity and likely postpones clinical relapse. Molecular monitoring offers an opportunity to select some patients for therapeutic intervention and to avoid unnecessary treatment in others. MRD-positive patients in the first analysis post-ASCT have a dismal prognosis and thus are in need of novel strategies.
Blood | 2018
Inhye E. Ahn; Mohammed Farooqui; Xin Tian; Janet Valdez; Clare Sun; Susan Soto; Jennifer Lotter; Stephanie Housel; Maryalice Stetler-Stevenson; Constance Yuan; Irina Maric; Katherine R. Calvo; Pia Nierman; Thomas Hughes; Nakhle S. Saba; Gerald E. Marti; Stefania Pittaluga; Sarah E. M. Herman; Carsten U. Niemann; Lone Bredo Pedersen; Christian H Geisler; Richard Childs; Georg Aue; Adrian Wiestner
The safety and efficacy of ibrutinib (420 mg) in chronic lymphocytic leukemia (CLL) were evaluated in a phase 2 study; 51 patients had TP53 aberration (TP53 cohort) and 35 were enrolled because of age 65 years or older (elderly cohort). Both cohorts included patients with treatment-naive (TN) and relapsed/refractory (RR) CLL. With the median follow-up of 4.8 years, 49 (57.0%) of 86 patients remain on study. Treatment was discontinued for progressive disease in 20 (23.3%) patients and for adverse events in 5 (5.8%). Atrial fibrillation occurred in 18 (20.9%) patients for a rate of 6.4 per 100 patient-years. No serious bleeding occurred. The overall response rate at 6 months, the primary study endpoint, was 95.8% for the TP53 cohort (95% confidence interval, 85.7%-99.5%) and 93.9% for the elderly cohort (95% confidence interval, 79.8%-99.3%). Depth of response improved with time: at best response, 14 (29.2%) of 48 patients in the TP53 cohort and 9 (27.3%) of 33 in the elderly cohort achieved a complete response. Median minimal residual disease (MRD) in peripheral blood was 3.8 × 10-2 at 4 years, with MRD-negative (<10-4) remissions in 5 (10.2%) patients. In the TP53 cohort, the estimated 5-year progression-free survival (PFS) was 74.4% in TN-CLL compared with 19.4% in RR-CLL (P = .0002), and overall survival (OS) was 85.3% vs 53.7%, respectively (P = .023). In the elderly cohort, the estimated 5-year PFS and OS in RR-CLL were 64.8% and 71.6%, respectively, and no event occurred in TN-CLL. Long-term administration of ibrutinib was well tolerated and provided durable disease control for most patients. This trial was registered at www.clinicaltrials.gov as #NCT01500733.
Blood Cancer Journal | 2016
C da Cunha-Bang; Jacob Simonsen; Klaus Rostgaard; Christian H Geisler; Henrik Hjalgrim; Carsten U. Niemann
The treatment of chronic lymphocytic leukemia (CLL) is in rapid transition, and during recent decades both combination chemotherapy and immunotherapy have been introduced. To evaluate the effects of this development, we identified all CLL patients registered in the nation-wide Danish Cancer Register between 1978 and 2013. We identified 10 455 CLL patients and 508 995 CLL-free control persons from the general population. Compared with the latter, the relative mortality rate between CLL patients and their controls decreased from 3.4 (95% CI 3.2–3.6) to 1.9 (95% CI 1.7–2.1) for patients diagnosed in 1978–1984 and 2006–2013, respectively. The improved survival corresponded to a decreasing risk of death from malignant hematological diseases, whereas the risk of death from infections was stable during the study period. These population-based data substantiate the improved survival for patients treated with chemo-immunotherapy demonstrated in clinical studies.
Hematological Oncology | 2017
Catja Freiburghaus; V. Kuci Emruli; Angelica Johansson; Roger Olsson; Fredrik Ek; Christian H Geisler; Mats Jerkeman; Sara Ek
The NFκB pathway plays a crucial role in cancer cell survival, proliferation and acquisition of resistance to therapy. Mucosa‐associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is an intermediary protein paracaspase important in the classical activation of NFκB. Previous studies have postulated an important role for NFκB in facilitating BCR signaling in CLL cells. Mi‐2 is a novel irreversible small molecule inhibitor of MALT1.Herein, we studied the effect of MALT‐1 inhibition, using Mi‐2, on the survival of CLL cells. Mi‐2 rapidly reduced CLL cell viability in a dose‐dependent manner. This effect was associated with the intrinsic apoptotic pathway activation, a robust increase in cleaved PARP and accompanied by a prominent decrease in Mcl‐1 and BCL2 levels. The pro‐apoptotic effect of Mi‐2 was observed in CLL cells of both low and high‐risk prognostic features. Accordingly pre‐treatment with Mi‐2 resulted in inhibition of IκB phosphorylation after BCR activation or even reduction below its basal level.Taken together, our results provide proof of the concept that MALT1 inhibition by the small molecule Mi‐2 can efficiently induce apoptosis in CLL cells. In light of the importance of the BCR and NFκB pathways in the pathogenesis of CLL, MALT1 is a potential therapeutic target in this disease. This is particularly important in the era of novel agents used to treat CLL, when resistance to these drugs emerges over time.
Blood | 2016
Mats Jerkeman; Martin Hutchings; Riikka Räty; Karin Fahl Wader; Anna Laurell; Hanne Kuitunen; Helle Toldbod; Lone Bredo Pedersen; Christian Winther Eskelund; Kirsten Grønbæk; Carsten U. Niemann; Christian H Geisler; Arne Kolstad
Blood | 2012
Kolstad Arne; Anna Laurell; Mats Jerkeman; Kirsten Grønbæk; Erkki Elonen; Riikka Räty; Lone Bredo Pedersen; Annika Loft; Trond Velde Bogsrud; Marie Nordström; Per Boye Hansen; Unn-Merete Fagerli; Herman Nilsson-Ehle; Grete F. Lauritzsen; Anne Kristine Lehmann; Christer Sundström; Marja-Liisa Karjalainen-Lindsberg; Elisabeth Ralfkiaer; Mats Ehinger; Jan Delabie; Hans Bentzen; Jukka Schildt; Kamelia Kostova-Aherdan; Henrik Frederiksen; Peter de Nully Brown; Christian H Geisler
Blood | 2016
Christian Winther Eskelund; Jakob Werner Hansen; Maj Westman; Catja Freiburghaus; Sara Ek; Lone Bredo Pedersen; Mette K. Andersen; Riikka Räty; Arne Kolstad; Mats Jerkeman; Peter de Nully Brown; Christian H Geisler; Kirsten Groenbaek