85 results on '"Tilmann Bochtler"'
Search Results
2. CUP-Syndrom – die neue ESMO-Leitlinie
- Author
-
Tilmann Bochtler, Maria Pouyiourou, and Alwin Krämer
- Subjects
Oncology - Published
- 2023
3. Prognostic impact of copy number alterations and tumor mutational burden in carcinoma of unknown primary
- Author
-
Tilmann Bochtler, Timothy Wohlfromm, Thomas Hielscher, Damian Stichel, Maria Pouyiourou, Bianca Kraft, Olaf Neumann, Volker Endris, Andreas von Deimling, Albrecht Stenzinger, and Alwin Krämer
- Subjects
Chromosome Aberrations ,Chromothripsis ,Cancer Research ,DNA Copy Number Variations ,Carcinoma ,Mutation ,Biomarkers, Tumor ,Genetics ,Humans ,Neoplasms, Unknown Primary ,Microsatellite Instability ,Prognosis - Abstract
Chromosomal aberrations are known to drive metastatic spread, but their profile is still elusive in carcinoma of unknown primary (CUP). Therefore, the aim of this study was to characterize the chromosomal aberration pattern in CUP depending on histological and clinical features and to assess its prognostic impact together with chromothripsis, tumor mutational burden (TMB), microsatellite instability (MSI), and mutational profiles as potential prognostic biomarkers.Chromosomal aberrations and chromothripsis were detected by methylation-based copy number variation (CNV) analysis, whereas TMB and MSI were calculated based on large next-generation sequencing (NGS) panels. Putative primaries were assigned by consensus between two independent oncologists.CNV losses varied depending on putative primaries and were more abundant in patients harboring TP53 mutations and/or deletions 17p. CNV loss was prognostically adverse in localized CUP treated with surgery and/or radiotherapy, but not in disseminated poor-risk CUP treated with palliative chemotherapy. CNV loss also worsened the prognosis in squamous cell CUP. Chromothripsis was detected in 18/59 (30.5%) patients without prognostic effect. TMB was highest in cases with MSI, squamous cell histology, and with lung, anal or cervical putative primaries.Overall, CNV, chromothripsis, TMB, and MSI profiles in CUP are reminiscent of biological characteristics known from other cancer entities without a unifying CUP-specific signature. Markedly, high-level CNV loss is an adverse predictive biomarker in localized but not disseminated chemotherapy-treated CUP. This implies that chromosomal losses drive CUP progression, but also increase susceptibility to chemotherapy, with both effects apparently leveling out in disseminated CUP.
- Published
- 2022
4. Targeting rare and non-canonical driver variants in NSCLC – An uncharted clinical field
- Author
-
Eugen Rempel, Peter Horak, Roland Penzel, Anna-Lena Volckmar, Claus Peter Heußel, Michael Allgäuer, Michael Thomas, Regine Brandt, Huriye Seker-Cin, Mark Kriegsmann, Volker Endris, Jan Budczies, Olaf Neumann, Jonas Leichsenring, Tilman Brummer, Felix J.F. Herth, Petros Christopoulos, M. Faehling, Jürgen Fischer, Daniel Kazdal, Albrecht Stenzinger, Martina Kirchner, Julia Glade, Tilmann Bochtler, Hauke Winter, Peter Schirmacher, Stefan Fröhling, and Hannah Goldschmid
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,Thoracic Oncology ,Internal medicine ,ROS1 ,medicine ,Humans ,Precision Medicine ,Lung cancer ,business.industry ,High-Throughput Nucleotide Sequencing ,Protein-Tyrosine Kinases ,medicine.disease ,Clinical trial ,030104 developmental biology ,Non canonical ,Precision oncology ,030220 oncology & carcinogenesis ,Mutation ,Molecular targets ,business ,Systematic search - Abstract
Objectives Implementation of tyrosine kinase inhibitors (TKI) and other targeted therapies was a main advance in thoracic oncology with survival gains ranging from several months to years for non-small-cell lung cancer (NSCLC) patients. High-throughput comprehensive molecular profiling is of key importance to identify patients that can potentially benefit from these novel treatments. Material and Methods Next-generation sequencing (NGS) was performed on 4500 consecutive formalin-fixed, paraffin-embedded specimens of advanced NSCLC (n = 4172 patients) after automated extraction of DNA and RNA for parallel detection of mutations and gene fusions, respectively. Results and Conclusion Besides the 24.9 % (n = 1040) of cases eligible for approved targeted therapies based on the presence of canonical alterations in EGFR exons 18–21, BRAF, ROS1, ALK, NTRK, and RET, an additional n = 1260 patients (30.2 %) displayed rare or non-canonical mutations in EGFR (n = 748), BRAF (n = 135), ERBB2 (n = 30), KIT (n = 32), PIK3CA (n = 221), and CTNNB1 (n = 94), for which targeted therapies could also be potentially effective. A systematic literature search in conjunction with in silico evaluation identified n = 232 (5.5 %) patients, for which a trial of targeted treatment would be warranted according to available evidence (NCT level 1, i.e. published data showing efficacy in the same tumor entity). In conclusion, a sizeable fraction of NSCLC patients harbors rare or non-canonical alterations that may be associated with clinical benefit from currently available targeted drugs. Systematic identification and individualized management of these cases can expand applicability of precision oncology in NSCLC and extend clinical gain from established molecular targets. These results can also inform clinical trials.
- Published
- 2021
5. Risk stratification of EGFR+ lung cancer diagnosed with panel-based next-generation sequencing
- Author
-
Lukas C. Heukamp, Mark Kriegsmann, J. Roeper, Sonja Loges, L.M. Brückner, N. Magios, M. Faehling, Martin E. Eichhorn, Daniel Kazdal, Petros Christopoulos, A. Stenzinger, Katharina Kriegsmann, Martin Wermke, Martina Kirchner, Michael Meister, Marc A. Schneider, Frank Griesinger, Fjf Herth, Volker Endris, Thomas Muley, Roland Penzel, Tilmann Bochtler, Melanie Janning, Michael Thomas, Helge Bischoff, R. El Shafie, Farastuk Bozorgmehr, Felix C. Saalfeld, J.R. Fischer, C.P. Heussel, Peter Schirmacher, and Anna-Lena Volckmar
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.drug_class ,Hazard ratio ,medicine.disease ,Tyrosine-kinase inhibitor ,DNA sequencing ,respiratory tract diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Risk stratification ,Cohort ,medicine ,Overall survival ,Lung cancer ,business ,Risk assessment - Abstract
Objective Panel-based next-generation sequencing (NGS) is increasingly used for the diagnosis of EGFR-mutated non-small-cell lung cancer (NSCLC) and could improve risk assessment in combination with clinical parameters. Materials and methods To this end, we retrospectively analyzed the outcome of 400 tyrosine kinase inhibitor (TKI)-treated EGFR+ NSCLC patients with validation of results in an independent cohort (n = 130). Results EGFR alterations other than exon 19 deletions (non-del19), TP53 co-mutations, and brain metastases at baseline showed independent associations of similar strengths with progression-free (PFS hazard ratios [HR] 2.1–2.3) and overall survival (OS HR 1.7–2.2), in combination defining patient subgroups with distinct outcome ( E GFR+ N SCLC risk S core, "ENS", p Conclusions EGFR variant, TP53 status and brain metastases predict TKI efficacy and survival in EGFR+ NSCLC irrespective of other currently available parameters ("ENS"). Together, they constitute a practical and reproducible approach for risk stratification of newly diagnosed metastatic EGFR+ NSCLC.
- Published
- 2020
6. Das CUP-Syndrom - Stand 2020
- Author
-
Maria Pouyiourou, Laura Claßen, Göran R. Boeckel, Tilmann Bochtler, and Alwin Krämer
- Subjects
Gynecology ,medicine.medical_specialty ,business.industry ,medicine ,General Earth and Planetary Sciences ,business ,General Environmental Science - Published
- 2020
7. Diagnostik und Therapie von Krebserkrankungen mit unbekanntem Primärtumor (CUP-Syndrom)
- Author
-
L. Claßen, M. Pouyiourou, Tilmann Bochtler, G. R. Boeckel, and Alwin Krämer
- Subjects
Gynecology ,03 medical and health sciences ,medicine.medical_specialty ,0302 clinical medicine ,Oncology ,business.industry ,030220 oncology & carcinogenesis ,medicine ,business ,030215 immunology - Abstract
Bei Krebserkrankungen mit unbekanntem Primartumor (CUP, „carcinoma of unknown primary site“) handelt es sich um ein heterogenes onkologisches Krankheitsbild, bei dem nach Abschluss einer umfangreichen Primardiagnostik kein Ursprungstumor identifiziert werden kann, jedoch histologisch und/oder zytologisch gesicherte Metastasen vorliegen. Diagnostische Bemuhungen richten sich v. a. auf die Identifikation prognostisch gunstiger Subgruppen sowie die Bestimmung des Disseminationsgrads. Sie umfassen neben klinischen und radiologischen Untersuchungen insbesondere eine ausfuhrliche Histologie und Immunhistochemie. Molekulargenetische Methoden konnen helfen, einen wahrscheinlichen Primartumor sowie Mutationen fur den Einsatz zielgerichteter Therapien zu identifizieren. Sie finden derzeit v. a. in klinischen Studien Anwendung, sind aber bisher nicht als Routinediagnostik implementiert. Wahrend prognostisch gunstige Subgruppen eine spezifische Therapie in Anlehnung an den klinisch-pathologisch wahrscheinlichen Primartumor erhalten, ist eine empirische, platinbasierte Kombinationschemotherapie derzeit therapeutischer Standard zur Behandlung des prognostisch ungunstigen CUP-Syndroms. Die Prognose ist dabei mit einem medianen Uberleben von etwa einem Jahr schlecht. Der Einsatz von zielgerichteten, mutationsspezifischen Therapien und Immuncheckpointinhibitoren wird in aktuellen Studien untersucht.
- Published
- 2020
8. Local ablative treatment with surgery and/or radiotherapy in single-site and oligometastatic carcinoma of unknown primary
- Author
-
Olaf Neumann, Thomas Hielscher, Bianca Kraft, Timothy Wohlfromm, Volker Endris, Albrecht Stenzinger, Alwin Krämer, Andreas von Deimling, Stefan Delorme, Maria Pouyiourou, Tilmann Bochtler, and Damian Stichel
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,DNA Copy Number Variations ,medicine.medical_treatment ,Single site ,Ablative case ,Carcinoma ,medicine ,Humans ,Neoplasm Metastasis ,Surgical treatment ,Aged ,Retrospective Studies ,Copy number loss ,business.industry ,Retrospective cohort study ,Middle Aged ,medicine.disease ,Genes, p53 ,Prognosis ,Combined Modality Therapy ,Surgery ,Radiation therapy ,Oncology ,Mutation ,Unknown primary ,Neoplasms, Unknown Primary ,Female ,business - Abstract
Background Single-site carcinoma of unknown primary (CUP) is recognised as a distinct favourable subtype in the European Society of Medical Oncology (ESMO) classification. There is broad consensus that these patients are candidates for local ablative treatment strategies with surgery and/or radiotherapy, but data on their outcomes are scarce. Patients and methods In this study, we have addressed the prospects of cure and prognostic factors in a retrospective cohort of 63 patients who were eligible for local treatment at our centre. Results Median event-free (EFS) and overall survival (OS) were 15.6 months and 52.5 months, respectively. Of 61 patients who received local treatment, 20 (32.8%) remained event-free over a median follow-up of 28 months. Baseline clinical parameters including affected organ, number, volume and histology of metastases had no significant impact on prognosis, whereas deleterious TP53 mutations and DNA copy number loss emerged as independent adverse risk factors with respect to EFS. Surgical treatment was associated with improved OS as compared to radiation-based therapy. Conclusion Our study advocates to pursue localised treatment with surgery and/or radiotherapy whenever feasible and implies that genetic parameters might additionally determine the clinical course of single-site CUP patients.
- Published
- 2021
9. TP53 deficiency permits chromosome abnormalities and karyotype heterogeneity in acute myeloid leukemia
- Author
-
Anna Cazzola, Christin Schlegel, Anna Jauch, Tilmann Bochtler, Ilka Jansen, and Alwin Krämer
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,endocrine system diseases ,Aneuploidy ,Myeloid leukemia ,Chromosome ,Karyotype ,Hematology ,Biology ,medicine.disease ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,stomatognathic system ,Oncology ,030220 oncology & carcinogenesis ,Chromosome instability ,Complex Karyotype ,Cancer research ,medicine ,neoplasms - Abstract
Abnormal karyotypes are common in cancer cells and frequently observed in acute myeloid leukemia (AML), in which complex karyotype aberrations are associated with poor prognosis. How exactly abnormal karyotypes arise and are propagated in AML is unclear. TP53 mutations and deletions are frequent in complex karyotype AML, suggesting a role of TP53 alterations in the development of chromosome abnormalities. Here, we generated isogenic TP53-knockout versions of the euploid AML cell line EEB to investigate the impact of TP53 on karyotype stability. We show that chromosome abnormalities spontaneously arise in TP53-deficient cells. Numerical aneuploidy could, to some extent, be propagated in a TP53-proficient setting, indicating that it does not necessarily trigger TP53 activation. In contrast, tolerance to structural chromosome aberrations was almost entirely restricted to TP53-knockout clones, all of which were able to continue proliferation in the presence of damaged DNA. Mechanistically, as a source of chromosome aberrations, limited numerical but not structural chromosomal instability was tolerated by TP53-wildtype cells. In contrast, structural instability was found only in TP53-knockout cells. Together, in myeloid cells TP53 loss allows for the development of complex karyotype aberrations and karyotype heterogeneity by perpetuation of chromosome segregation errors.
- Published
- 2019
10. Comparative genetic profiling aids diagnosis and clinical decision making in challenging cases of CUP syndrome
- Author
-
Olaf Neumann, Alwin Krämer, Tilmann Bochtler, Anna Reiling, Michael Allgäuer, Albrecht Stenzinger, Peter Schirmacher, Martina Kirchner, Jonas Leichsenring, Volker Endris, and Anna-Lena Volckmar
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Clinical Decision-Making ,Ataxia Telangiectasia Mutated Proteins ,Malignancy ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Formaldehyde ,Internal medicine ,medicine ,Humans ,Aged ,BRCA2 Protein ,BAP1 ,Paraffin Embedding ,Tissue Embedding ,BRCA1 Protein ,business.industry ,Tumor Suppressor Proteins ,High-Throughput Nucleotide Sequencing ,Cancer ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Primary tumor ,Clone Cells ,DNA profiling ,030220 oncology & carcinogenesis ,Mutation ,Neoplasms, Unknown Primary ,FLAG (chemotherapy) ,Female ,sense organs ,business ,Ubiquitin Thiolesterase - Abstract
Cancer of unknown primary (CUP) denotes cancer cases where metastatic spread is histologically confirmed, but no respective primary tumor can be identified. The challenging diagnosis of CUP is further complicated in cases with previously identified malignancies or with dubious clonal relationship between metastatic sites due to ambiguous histology. Our study aims at elucidating clonal relationships by comparing the respective mutational spectra. Targeted next-generation sequencing (NGS) employing formalin-fixed and paraffin-embedded (FFPE) tumor tissue was performed on 174 consecutive CUP patients. Among these, 43/174 (24.7%) patients had a documented prior malignancy. Data on pairwise targeted NGS testing to address clonal relationships between the previous malignancy and the presumed CUP (n = 11) or between different CUP metastatic sites (n = 7) was available in 18 patients. NGS could clarify clonal relationships in 16/18 cases. Among the 11 CUP patients with antecedent malignancies, four cases were clonally independent of the previous malignancy but harbored deleterious germline mutations in BRCA/BAP1/ATM genes. Seven CUP cases were clonally related to the antecedent malignancy, changing the CUP diagnosis to relapse of the prior malignancy. In the seven CUP cases, with doubtfully related metastatic sites, NGS confirmed clonal relationship in five cases and was inconclusive in two. In conclusion, NGS proved an efficient tool to elucidate clonal relationships in clinically challenging CUP cases. Our study cautions against a premature diagnosis of CUP. Relapses of antecedent malignancies should be carefully considered. CUPs clonally independent from the antecedent malignancy should raise a red flag of a potential cancer-predisposing germline mutation.
- Published
- 2019
11. Performance analysis of AL amyloidosis cardiac biomarker staging systems with special focus on renal failure and atrial arrhythmia
- Author
-
Hugo A. Katus, Ute Hegenbart, Carsten Müller-Tidow, Christoph Kimmich, Kaya Veelken, Tobias Dittrich, Stefan Schönland, Axel Benner, Arnt V. Kristen, Fabian aus dem Siepen, and Tilmann Bochtler
- Subjects
medicine.medical_specialty ,Renal function ,Context (language use) ,Article ,Plasma Cell Disorders ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Atrial Fibrillation ,medicine ,AL amyloidosis ,Humans ,Immunoglobulin Light-chain Amyloidosis ,Renal Insufficiency ,Univariate analysis ,business.industry ,Amyloidosis ,Atrial fibrillation ,Retrospective cohort study ,Hematology ,medicine.disease ,Cardiology ,Biomarker (medicine) ,Immunoglobulin Light Chains ,business ,Biomarkers ,030215 immunology - Abstract
Systemic light chain amyloidosis is a rare and life-threatening disorder, for which accurate risk stratification is crucial. Current cardiac staging systems (MAYO2004, MAYO3b, and MAYO2012) are mainly based on biomarkers, which have uncertain reliability in the context of atrial fibrillation, arrhythmia or pacemaker stimulation as well as renal insufficiency. We compared the performance of the established staging systems with particular regard to these comorbidities in 1,224 patients with systemic light chain amyloidosis diagnosed at our center from July 2002 until March 2017. We first characterized the subsets with an estimated glomerular filtration rate
- Published
- 2019
12. Micronucleus formation in human cancer cells is biased by chromosome size
- Author
-
Mutlu Kartal-Kaess, Thomas Hielscher, Marco R. Cosenza, Martin Granzow, Alwin Krämer, Anna Jauch, Tilmann Bochtler, and Christel Herold-Mende
- Subjects
Cancer Research ,medicine.diagnostic_test ,Cell division ,Brain Neoplasms ,Chromosome ,Biology ,Molecular biology ,Chromosome segregation ,03 medical and health sciences ,0302 clinical medicine ,Chromosome Segregation ,030220 oncology & carcinogenesis ,Chromosome instability ,Micronucleus test ,Genetics ,medicine ,Chromosomes, Human ,Humans ,Glioblastoma ,Micronucleus ,Mitosis ,Cells, Cultured ,Micronuclei, Chromosome-Defective ,Fluorescence in situ hybridization - Abstract
Chromosomal instability is one of the hallmarks of cancer and caused by chromosome missegregation during mitosis, a process frequently associated with micronucleus formation. Micronuclei are formed when chromosomes fail to join a daughter nucleus during cell division and are surrounded by their own nuclear membrane. Although it has been commonly assumed that the gain or loss of specific chromosomes is random during compromised cell division, recent data suggest that the size of chromosomes can impact on chromosome segregation fidelity. To test whether chromosome missegregation rates scale with chromosome size in primary human cancer cells, we assessed chromosome sequestration into micronuclei in patient-derived primary NCH149 glioblastoma cells, which display high-level numerical chromosome instability (CIN), pronounced spontaneous micronucleus formation but virtually no structural CIN. The cells were analyzed by interphase fluorescence in situ hybridization using chromosome-specific painting probes for all chromosomes. Overall, 33% of early passage NCH149 cells harbored micronuclei. Entrapment within a micronucleus clearly correlated with chromosome size with larger chromosomes being significantly more frequently missegregated into micronuclei than smaller chromosomes in primary glioblastoma cells. These findings extend the concept that chromosome size determines segregation fidelity by implying that size-specific micronucleus entrapment occurs in primary human cancer cells as well.
- Published
- 2019
13. Cancer-of-Unknown-Primary-Origin: A SEER–Medicare Study of Patterns of Care and Outcomes among Elderly Patients in Clinical Practice
- Author
-
Linda, Mileshkin, Tilmann, Bochtler, Gemma, Gatta, Razelle, Kurzrock, Andreas, Beringer, Mathis, Müller-Ohldach, Andy, Surinach, Camille, Perret, Marlene, Thomas, Adam, Gondos, and Alwin, Krämer
- Subjects
Cancer Research ,Oncology ,diagnostic tests and procedures ,drug therapy ,Medicare Part A ,Medicare Part B ,neoplasms ,registries ,SEER program ,survival analysis ,unknown primary - Abstract
Knowledge of contemporary patterns of cancer-of-unknown-primary-origin (CUP) diagnostic work-up, treatment, and outcomes in routine healthcare is limited. Thus, we examined data from elderly patients diagnosed with CUP in real-world US clinical practice. From the Surveillance, Epidemiology, and End Results–Medicare-linked database, we included patients ≥ 66 years old with CUP diagnosed between 1 January 2013 and 31 December 2015. We analyzed baseline demographics, clinical characteristics, methods of diagnostic work-up (biopsy, immunohistochemistry, imaging), treatment-related factors, and survival. CUP diagnosis was histologically confirmed in 2813/4562 patients (61.7%). Overall, 621/4562 (13.6%) patients received anticancer pharmacotherapy; among these, 97.3% had a histologically confirmed tumor and 83.1% received all three procedures. Among those with a histologically confirmed tumor, increasing age, increasing comorbidity score, not receiving all three diagnostic measures, and having a not-further specified histologic finding of only ‘malignant neoplasm’ were all negatively associated with receipt of anticancer pharmacotherapy. Median overall survival was 1.2 months for all patients. Median time between CUP diagnosis and treatment initiation was 41 days. Limited diagnostic work-up was common and most patients did not receive anticancer pharmacotherapy. The poor outcomes highlight a substantial unmet need for further research into improving diagnostic work-up and treatment effectiveness in CUP.
- Published
- 2022
14. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial
- Author
-
Christian Thiede, C. Röllig, Christine Borkmann, Claudia D. Baldus, Ulrich Dührsen, Gerhard Ehninger, Marika Mende, Gesine Bug, Lars Fransecky, Matthias Stelljes, Richard Noppeney, Antje Schubert, Johannes Schetelig, Katharina Götze, Dominik Wolf, A.S. Kubasch, Friedrich Stölzel, Malte von Bonin, Mathias Hänel, Hubert Serve, Michael Kramer, Martin Bornhäuser, Alwin Krämer, Regina Herbst, Uta Oelschlägel, Uwe Platzbecker, Jan Moritz Middeke, Katja Sockel, Christoph Groth, Tilmann Bochtler, and Anke Mütherig
- Subjects
Male ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Neoplasm, Residual ,Time Factors ,Azacitidine ,Medizin ,Relapse prevention ,Risk Assessment ,Drug Administration Schedule ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Risk Factors ,Germany ,hemic and lymphatic diseases ,Internal medicine ,Biomarkers, Tumor ,medicine ,Clinical endpoint ,Humans ,Prospective Studies ,Progression-free survival ,Prospective cohort study ,Aged ,business.industry ,Myelodysplastic syndromes ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Transplantation ,Clinical trial ,Leukemia, Myeloid, Acute ,Oncology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Female ,business ,Nucleophosmin ,030215 immunology ,medicine.drug - Abstract
Summary Background Monitoring of measurable residual disease (MRD) in patients with advanced myelodysplastic syndromes (MDS) or acute myeloid leukaemia (AML) who achieve a morphological complete remission can predict haematological relapse. In this prospective study, we aimed to determine whether MRD-guided pre-emptive treatment with azacitidine could prevent relapse in these patients. Methods The relapse prevention with azacitidine (RELAZA2) study is an open-label, multicentre, phase 2 trial done at nine university health centres in Germany. Patients aged 18 years or older with advanced MDS or AML, who had achieved a complete remission after conventional chemotherapy or allogeneic haemopoietic stem-cell transplantation, were prospectively screened for MRD during 24 months from baseline by either quantitative PCR for mutant NPM1, leukaemia-specific fusion genes (DEK–NUP214, RUNX1–RUNX1T1, CBFb–MYH11), or analysis of donor-chimaerism in flow cytometry-sorted CD34-positive cells in patients who received allogeneic haemopoietic stem-cell transplantation. MRD-positive patients in confirmed complete remission received azacitidine 75 mg/m2 per day subcutaneously on days 1–7 of a 29-day cycle for 24 cycles. After six cycles, MRD status was reassessed and patients with major responses (MRD negativity) were eligible for a treatment de-escalation. The primary endpoint was the proportion of patients who were relapse-free and alive 6 months after the start of pre-emptive treatment. Analyses were done per protocol. This trial is registered with ClincialTrials.gov , number NCT01462578 , and finished recruitment on Aug 21, 2018. Findings Between Oct 10, 2011, and Aug 20, 2015, we screened 198 patients with advanced MDS (n=26) or AML (n=172), of whom 60 (30%) developed MRD during the 24-month screening period and 53 (88%) were eligible to start study treatment. 6 months after initiation of azacitidine, 31 (58%, 95% CI 44–72) of 53 patients were relapse-free and alive (p Interpretation Pre-emptive therapy with azacitidine can prevent or substantially delay haematological relapse in MRD-positive patients with MDS or AML who are at high risk of relapse. Our study also suggests that continuous MRD negativity during regular MRD monitoring might be prognostic for patient outcomes. Funding Celgene Pharma, Jose Carreras Leukaemia Foundation, National Center for Tumor Diseases (NCT), and German Cancer Consortium (DKTK) Foundation.
- Published
- 2018
15. 1804P Baseline mutational profiles of patients (pts) with carcinoma-of-unknown-primary-origin (CUP) enrolled onto CUPISCO
- Author
-
Sophie Golding, Holger Moch, Christoph B. Westphalen, Ethan Sokol, Andreas Beringer, Ferran Losa, E. Hoglander, Chantal Pauli, N. Chalabi, Alwin Krämer, G. Duran-Pacheco, Jeffrey S. Ross, Linda Mileshkin, A. Karapetyan, Natalie Cook, Tilmann Bochtler, and Richard W. Tothill
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Carcinoma ,medicine ,Unknown primary ,Hematology ,medicine.disease ,business ,Baseline (configuration management) - Published
- 2021
16. Cryostorage to What End? - Autologous Stem Cell Products in Burkitt Lymphoma, Acute Lymphoblastic Leukemia, Acute Myeloid Leukemia, and Myeloproliferative Neoplasm Patients
- Author
-
Patrick Wuchter, Thomas Bruckner, Harald Klüter, Sandra Sauer, Stefan Klein, Katharina Kriegsmann, Anita Schmitt, Carsten Müller-Tidow, Mark Kriegsmann, Petra Pavel, and Tilmann Bochtler
- Subjects
Chemotherapy ,Acute leukemia ,medicine.medical_specialty ,Allogeneic transplantation ,Palliative care ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Hematology ,medicine.disease ,Lymphoma ,Surgery ,Transplantation ,medicine ,Immunology and Allergy ,business ,Myeloproliferative neoplasm ,Research Article - Abstract
Introduction: Recently, we identified a huge discrepancy between the collection practice and the actual utilization of cryopreserved peripheral blood stem cells (PBSCs) for high-dose chemotherapy (HDCT) and autologous blood stem cell transplantation (ABSCT). Specifically, patients with Burkitt lymphoma, acute leukemia, and myeloproliferative neoplasms (MPN) were frequently not referred for ABSCT after successful PBSC collection. Objective: The aim of this study was to identify variables that are associated with the non-utilization of PBSC grafts. Methods: We retrospectively analyzed the collection, storage, and disposal of PBSC grafts in Burkitt lymphoma (n = 18), acute lymphoblastic leukemia (ALL, n = 22), MPN (n = 18), and acute myeloid leukemia (AML, n = 71) patients. Patients who underwent autologous PBSC collection at 2 collection and transplantation centers between 2001 and 2012 were included and followed up until 2016. Results: None of the Burkitt lymphoma patients were referred for ABSCT. Only in 1 (6%) patient, the graft was discarded after the patient’s death. In all other patients (n = 17, 94%), the grafts were stored independently of the patient’s status (death, n = 4, 22%; no follow-up, n = 6, 33%; no indication for ABSCT given, n = 7, 39%). In ALL patients, 4 (18%) patients underwent ABSCT after a median follow-up of 74 (1–182) months. In the remaining patients, PBSC grafts were either discarded (8 patients, 36%) or stored until the reference date (10 patients, 45%). Seven of 18 MPN patients (39%) underwent ABSCT. ABSCT was performed in 24 (34%) AML patients. In 20 (28%) patients who were not referred to ABSCT, an allogeneic transplantation (TPL) was performed. Fifteen (21%) patients received palliative care or deceased, and their grafts were discarded in all but 1 patient. Additional grafts were discarded in 21 (31%) patients and stored in 9 (13%) patients who underwent ABSCT or allogeneic TPL (n = 44). Conclusions: As the role and efficacy of autologous HDCT/ABSCT are not established in the analyzed entities, the indication for PBSC collection should be reanalyzed in regular intervals. Moreover, PBSC grafts from patients who have deceased, have insufficient grafts, or have already undergone an allogeneic TPL should be considered for disposal or (if applicable) for research use, to economize storage costs on a rational basis.
- Published
- 2020
17. Clinical and molecular profile of de novo vs. secondary EGFR mutated metastatic non-small-cell lung cancer
- Author
-
M. Faehling, N. Magios, J. Kuon, Albrecht Stenzinger, Daniel Kazdal, Fjf Herth, Volker Endris, Michael Meister, Thomas Muley, Michael Thomas, Stefan Rieken, Helge Bischoff, Tilmann Bochtler, Harland S. Winter, Martina Kirchner, Anna-Lena Volckmar, Farastuk Bozorgmehr, Petros Christopoulos, C.P. Heussel, and J. Budczies
- Subjects
business.industry ,Cancer research ,Medicine ,Molecular Profile ,Non small cell ,business ,Lung cancer ,medicine.disease - Published
- 2020
18. Treatment of AL amyloidosis with bendamustine: a study of 122 patients
- Author
-
Christoph Kimmich, Ute Hegenbart, Andrea Foli, Giovanni Palladini, Tilmann Bochtler, Giampaolo Merlini, Tobias Dittrich, Paolo Milani, Stefan Schönland, Carsten Müller-Tidow, and Marco Basset
- Subjects
Male ,Bendamustine ,medicine.medical_treatment ,Immunology ,Treatment outcome ,Clone (cell biology) ,Immunoglobulin light chain ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,medicine ,AL amyloidosis ,Bendamustine Hydrochloride ,Humans ,Immunoglobulin Light-chain Amyloidosis ,Prospective Studies ,Antineoplastic Agents, Alkylating ,Aged ,Chemotherapy ,business.industry ,Amyloidosis ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Treatment Outcome ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,030215 immunology ,medicine.drug - Abstract
TO THE EDITOR: Chemotherapy for light chain (AL) amyloidosis is based on combinations developed for multiple myeloma.[1][1] A better understanding of susceptibility of the AL underlying clone to specific types of treatments[2][2] and the ability to identify cytogenetic patterns with different
- Published
- 2018
19. Phase I dose-escalation trial investigating volasertib as monotherapy or in combination with cytarabine in patients with relapsed/refractory acute myeloid leukaemia
- Author
-
Tilmann Bochtler, Tillmann Taube, Oliver G. Ottmann, Florian Voss, Alwin Krämer, Pilar Garin-Chesa, Hartmut Döhner, Dan Liu, Utz Krug, Gesine Bug, Carsten Müller-Tidow, Richard F. Schlenk, and Michael Lübbert
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Volasertib ,Hematology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Internal medicine ,Relapsed refractory ,medicine ,Dose escalation ,Cytarabine ,In patient ,Myeloid leukaemia ,business ,medicine.drug - Published
- 2018
20. Integrated Histogenetic Analysis Reveals BAP1 -Mutated Epithelioid Mesothelioma in a Patient With Cancer of Unknown Primary
- Author
-
Peter Schirmacher, Fabian Stögbauer, Benjamin Goeppert, Volker Endris, Jonas Leichsenring, Michal R. Schweiger, Sebastian Klein, Anna Reiling, Albrecht Stenzinger, Tilmann Bochtler, and Alwin Krämer
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,BAP1 ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,medicine.disease ,Molecular diagnostics ,Germline ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Missense mutation ,Lymphadenectomy ,Mesothelioma ,business ,Genetic testing - Abstract
This case report presents a male patient with epithelioid mesothelioma that was initially misdiagnosed as cancer of unknown primary (CUP) and correctly identified using molecular panel sequencing. The patient had a prior history of colon and breast cancer. To assess the enlarged mediastinal lymph nodes, retrosternal lymphadenectomy was performed in 2016. The lymph nodes were histologically deemed unrelated to the known breast cancer by the reference pathologist, thus leading to the diagnosis of a CUP syndrome. When the patient presented to our center, targeted deep sequencing of both breast cancer and presumed CUP was performed to address the clonal relationship between both malignancies. A missense mutation in BAP1 was revealed in both samples, with coverage data indicating a germline event. The patient was subsequently counseled by a human geneticist and underwent genetic testing, which confirmed the germline nature of this mutation. Collectively, these data led to the diagnosis of BAP1 (BRCA1-associated protein-1) tumor predisposition syndrome (TPDS). With the knowledge of an underlying BAP1 mutation and its known frequent association with epithelioid mesothelioma, the histology was reassessed and the diagnosis was revised to epithelioid mesothelioma. At this point, peritoneal involvement of mesothelioma could be diagnosed and histologically confirmed. This case illustrates the potential of integrated histopathologic and molecular diagnostics in helping to decipher CUP syndromes and establish the correct diagnosis. Additionally, this case highlights typical features of BAP1 TPDS with its general susceptibility to cancers, with pleural and peritoneal mesotheliomas as most prevalent clinical entities and the typically more benign course of these epithelioid mesotheliomas compared with BAP1-unrelated cases of mesotheliomas.
- Published
- 2018
21. Diagnosis and management of metastatic neoplasms with unknown primary
- Author
-
Tilmann Bochtler, Harald Löffler, and Alwin Krämer
- Subjects
Male ,0301 basic medicine ,Oncology ,Pathology ,medicine.medical_specialty ,Axillary lymph nodes ,Biopsy ,medicine.medical_treatment ,Clinical Decision-Making ,Pathology and Forensic Medicine ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Predictive Value of Tests ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,business.industry ,Head and neck cancer ,Bone metastasis ,medicine.disease ,Immunohistochemistry ,Primary tumor ,Radiation therapy ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Molecular Diagnostic Techniques ,Cervical lymph nodes ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Mutation ,Neoplasms, Unknown Primary ,Adenocarcinoma ,Female ,business - Abstract
In cancer of unknown primary (CUP), metastases are clinically and histologically confirmed, but the primary tumor site remains elusive after extensive work-up. CUPs make up for 2-3% of all epithelial malignancies. The two prevailing histologies are adenocarcinomas and undifferentiated carcinomas, whereas squamous cell carcinomas, neuroendocrine carcinomas and rare histologies account for the remaining 10%. The diagnostic work-up in CUP relies strongly on a detailed immunohistological (IHC) analysis in order to characterize the tumor type, nowadays aided by molecular techniques. Diagnostics also include a thorough clinical examination, a basic lab draw with the most relevant tumor markers, and cross sectional imaging. Additional PET-CT is recommended in cervical lymph nodes suggestive of head and neck cancer and in limited metastases potentially treatable in curative intent. As for treatment, it is paramount to identify patients who fall into one of the six well defined "favorable" subset categories, namely extragonadal germ cell tumors, adenocarcinoma with isolated unilateral axillary lymph nodes in female patients, squamous cell carcinoma with neck lymph nodes, squamous cell carcinoma with inguinal lymph nodes, serous papillary peritoneal carcinomatosis in females and blastic bone metastasis in males with elevated PSA. These subsets are distinct both regarding the required treatment and the comparably favorable prognosis. Within the remaining "unfavorable" group, patients of colon and renal cancer type should be identified based on IHC and clinical picture, since the prognosis of these patients seems to improve with the use of therapy tailored to the presumed primary as well. For the few patients with limited metastases it should be assessed whether they are candidates for surgery, radiotherapy or surgery followed by irradiation in curative intent. The remaining majority of patients are treated with empiric palliative chemotherapy, typically a platinum - paclitaxel combination, though the level of evidence for this therapy recommendation is low. Gemcitabine alone or in combination can be used as an alternative. Decoding of the molecular profiles in CUP offers the prospect of targeted therapy with novel agents. However, there appears to be no uniform molecular pattern for CUP, and the observed molecular diversity thus poses a challenge to respective clinical trials.
- Published
- 2018
22. Prognostic significance of cytogenetic heterogeneity in patients with newly diagnosed multiple myeloma
- Author
-
Dirk Hose, Thomas Hielscher, Hans Salwender, Marc S. Raab, Uta Bertsch, Ingo G.H. Schmidt-Wolf, Kai Neben, Maximilian Merz, Katja Weisel, Mathias Haenel, Christof Scheid, Hartmut Goldschmidt, Hans-Walter Lindemann, Igor Wolfgang Blau, Tilmann Bochtler, Anna Jauch, Stefan Schönland, Jens Hillengass, Anja Seckinger, Hematology, and Basic (bio-) Medical Sciences
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Vincristine ,Adolescent ,Clone (cell biology) ,Transplantation, Autologous ,Bortezomib ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Autologous transplantation ,Genetics(clinical) ,Multiple Myeloma/diagnosis ,In Situ Hybridization, Fluorescence ,Multiple myeloma ,Dexamethasone ,Netherlands ,Chromosome Aberrations ,Lymphoid Neoplasia ,medicine.diagnostic_test ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Transplantation ,030220 oncology & carcinogenesis ,oncology ,young adult ,Bortezomib/therapeutic use ,Antineoplastic Combined Chemotherapy Protocols/therapeutic use ,Multiple Myeloma ,business ,030215 immunology ,medicine.drug ,Fluorescence in situ hybridization - Abstract
We investigated subclonal cytogenetic aberrations (CA) detected by interphase fluorescence in situ hybridization (iFISH) in patients with newly diagnosed multiple myeloma (MM) enrolled in the Haemato Oncology Foundation for Adults in the Netherlands (HOVON)-65/German-Speaking MM Group (GMMG)-HD4 phase 3 trial. Patients were either treated with 3 cycles of vincristine, Adriamycin, and dexamethasone or bortezomib, Adriamycin, and dexamethasone and then thalidomide or bortezomib maintenance after tandem autologous transplantation. Subclones were defined either by presence of different copy numbers of the same chromosome loci and/or CA present in at least 30% less and maximally 2/3 of cells compared with the main clone CA. Patients with subclones harbored more frequently high risk (31.0%) or hyperdiploid main clone aberrations (24.8%) than patients with t(11;14) in the main clone (10.1%). Gains and deletions of c-MYC were the only CA that occurred more frequently as subclone (8.1%/20.5%) than main clone (6.2%/3.9%, respectively). Treatment with bortezomib completely overcame the negative prognosis of high-risk CA in patients without subclones, but not in patients with additional subclonal CA. High-risk patients treated without bortezomib showed dismal outcome whether subclones were present or not. Cytogenetic heterogeneity defined by subclonal CA is of major prognostic significance in newly diagnosed MM patients treated with bortezomib within the HOVON-65/GMMG-HD4 trial.
- Published
- 2017
23. Mesenchymal stromal cells contribute to quiescence of therapy-resistant leukemic cells in acute myeloid leukemia
- Author
-
Linda Manta, Christoph Lutz, Haiju He, Patrick Wuchter, Wenwen Wang, Volker Eckstein, Tilmann Bochtler, and Anthony D. Ho
- Subjects
Adult ,Male ,0301 basic medicine ,Stromal cell ,Adolescent ,medicine.medical_treatment ,CD34 ,Apoptosis ,Cell Communication ,CD38 ,Resting Phase, Cell Cycle ,Young Adult ,03 medical and health sciences ,Immunophenotyping ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,Medicine ,Treatment Failure ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,Mesenchymal stem cell ,Myeloid leukemia ,Mesenchymal Stem Cells ,Hematology ,General Medicine ,Middle Aged ,Cell cycle ,Prognosis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,Immunology ,Neoplastic Stem Cells ,Female ,business - Abstract
Objective Persistence of leukemic cells after induction therapy has been shown to correlate with poor survival in acute myeloid leukemia (AML). In this study we tested if human mesenchymal stromal cells (hMSC) have protective effects on leukemic cells undergoing chemotherapy. Methods Persistent disease was used as marker to identify cases with therapy resistant leukemic cells in 95 AML patients. Immunophenotyping, cell cycle and apoptosis assays were assessed by flow-cytometry. AML co-culture studies were performed with hMSC of healthy donors. Results Samples from patients with persistent disease had increased fractions of CD34+CD38- and quiescent leukemic cells. Comparison of sample series collected at time points of diagnosis and blast persistence showed a relative therapy-resistance of quiescent leukemic cells. Consistent with these observations, relapsed disease always displayed higher proportions of quiescent cells compared to samples of first diagnosis suggesting that quiescence is an important therapy escape mechanism of resistant cells. Co-culture studies demonstrated that hMSC protect leukemic cells from the effect of AraC treatment by enriching for quiescent cells, mimicking the effects observed in patients. This effect was even detectable when no direct stromal contact was established. Conclusions Our data suggest that hMSC contribute to quiescence and therapy resistance of persistent AML cells. This article is protected by copyright. All rights reserved.
- Published
- 2017
24. Reduced hematopoietic stem cell frequency predicts outcome in acute myeloid leukemia
- Author
-
Isabel Hoffmann, Van T. Hoang, Rachel Blume, Anna Marciniak-Czochra, Anna Jauch, Andreas Trumpp, Wenwen Wang, Christoph Lutz, Laura Poisa-Beiro, Borhan R. Saeed, Linda Manta, Tilmann Bochtler, Patrick Wuchter, Simon Raffel, Volker Eckstein, Thomas Stiehl, Anthony D. Ho, and Marieke A.G. Essers
- Subjects
Male ,Acute Myeloid Leukemia ,0301 basic medicine ,Myeloid ,medicine.medical_treatment ,Cell Count ,Hematopoietic stem cell transplantation ,Biology ,CXCR4 ,Disease-Free Survival ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,medicine ,Animals ,Humans ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Hematology ,Hematopoietic Stem Cells ,Minimal residual disease ,Survival Rate ,Leukemia, Myeloid, Acute ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Heterografts ,Female ,Stem cell - Abstract
In patients with acute myeloid leukemia and low percentages of aldehyde-dehydrogenase-positive cells, non-leukemic hematopoietic stem cells can be separated from leukemic cells. By relating hematopoietic stem cell frequencies to outcome we detected poor overall- and disease-free survival of patients with low hematopoietic stem cell frequencies. Serial analysis of matched diagnostic and follow-up samples further demonstrated that hematopoietic stem cells increased after chemotherapy in patients who achieved durable remissions. However, in patients who eventually relapsed, hematopoietic stem cell numbers decreased dramatically at the time of molecular relapse demonstrating that hematopoietic stem cell levels represent an indirect marker of minimal residual disease, which heralds leukemic relapse. Upon transplantation in immune-deficient mice cases with low percentages of hematopoietic stem cells of our cohort gave rise to leukemic or no engraftment, whereas cases with normal hematopoietic stem cell levels mostly resulted in multi-lineage engraftment. Based on our experimental data, we propose that leukemic stem cells have increased niche affinity in cases with low percentages of hematopoietic stem cells. To validate this hypothesis, we developed new mathematical models describing the dynamics of healthy and leukemic cells under different regulatory scenarios. These models suggest that the mechanism leading to decreases in hematopoietic stem cell frequencies before leukemic relapse must be based on expansion of leukemic stem cells with high niche affinity and the ability to dislodge hematopoietic stem cells. Thus, our data suggest that decreasing numbers of hematopoietic stem cells indicate leukemic stem cell persistence and the emergence of leukemic relapse.
- Published
- 2017
25. Lenalidomide/melphalan/dexamethasone in newly diagnosed patients with immunoglobulin light chain amyloidosis: results of a prospective phase 2 study with long-term follow up
- Author
-
Dirk Hose, Stefan Schönland, Christoph Kimmich, Ute Hegenbart, Hartmut Goldschmidt, Axel Benner, Anthony D. Ho, Anja Freiberger, Jörg Beimler, Tilmann Bochtler, Arnt V. Kristen, Markus Zorn, Anna Jauch, Ernst Hund, Marianne Gawlik, Natalia Becker, Hematology, and Basic (bio-) Medical Sciences
- Subjects
Male ,Melphalan ,medicine.medical_specialty ,medicine.medical_treatment ,Immunoglobulin Light-chain Amyloidosis/drug therapy ,030204 cardiovascular system & hematology ,Gastroenterology ,Article ,Plasma Cell Disorders ,Dexamethasone ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Internal Medicine ,medicine ,Humans ,Immunoglobulin Light-chain Amyloidosis ,Lenalidomide ,Aged ,Chemotherapy ,business.industry ,Amyloidosis ,Remission Induction/methods ,Remission Induction ,Hematology ,Antineoplastic Combined Chemotherapy Protocols/administration & dosage ,Middle Aged ,medicine.disease ,Survival Analysis ,Thalidomide/analogs & derivatives ,Thalidomide ,Surgery ,Regimen ,Tolerability ,030220 oncology & carcinogenesis ,oncology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Follow-Up Studies ,medicine.drug - Abstract
Chemotherapy in light chain amyloidosis aims to normalize the involved free light chain in serum, which leads to an improvement, or at least stabilization of organ function in most responding patients. We performed a prospective single center phase 2 trial with 50 untreated patients not eligible for high-dose treatment. The treatment schedule comprised 6 cycles of oral lenalidomide, melphalan and dexamethasone every 4 weeks. After 6 months, complete remission was achieved in 9 patients (18%), very good partial remission in 16 (32%) and partial response in 9 (18%). Overall, organ response was observed in 24 patients (48%). Hematologic and cardiac toxicities were predominant adverse events. Mortality at 3 months was low at 4% (n=2) despite the inclusion of 36% of patients (n=18) with cardiac stage Mayo 3. After a median follow-up of 50 months, median overall and event-free survival were 67.5 months and 25.1 months, respectively. We conclude that the treatment of lenalidomide, melphalan and dexamethasone is very effective in achieving a hematologic remission, organ response and, consecutively, a long survival in transplant ineligible patients with light chain amyloidosis. However, as toxicity and tolerability are the major problems of a 3-drug regimen, a strict surveillance program is necessary and sufficient to avoid severe toxicities. clinicaltrials.gov Identifier: 00883623 (Eudract2008-001405-41).
- Published
- 2017
26. Novel recurrent chromosomal aberrations detected in clonal plasma cells of light chain amyloidosis patients show potential adverse prognostic effect: first results from a genome-wide copy number array analysis
- Author
-
Dirk Hose, Hartmut Goldschmidt, Kari Hemminki, Anna Jauch, Katrin Hinderhofer, Martin Granzow, Ute Hegenbart, Anja Seckinger, Tilmann Bochtler, Stefan Schönland, Hematology, and Basic (bio-) Medical Sciences
- Subjects
0301 basic medicine ,Monosomy ,DNA Copy Number Variations ,Plasma Cells ,Plasma cell dyscrasia ,Chromosomal translocation ,Biology ,Amyloidosis/diagnosis ,Article ,Plasma Cell Disorders ,Translocation, Genetic ,Polyploidy ,Immunoglobulin Light-chain Amyloidosis ,03 medical and health sciences ,0302 clinical medicine ,Internal Medicine ,medicine ,AL amyloidosis ,Humans ,Genetics(clinical) ,Alleles ,In Situ Hybridization, Fluorescence ,Chromosome 13 ,Chromosome Aberrations ,medicine.diagnostic_test ,Plasma Cells/metabolism ,Amyloidosis ,Hematology ,Prognosis ,medicine.disease ,Survival Analysis ,Molecular biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,oncology ,Mutation ,Immunoglobulin Light Chains/genetics ,Immunoglobulin Light Chains ,Genome-Wide Association Study ,Fluorescence in situ hybridization - Abstract
Immunoglobulin light chain (AL) amyloidosis is a rare plasma cell dyscrasia characterized by the deposition of abnormal amyloid fibrils in multiple organs, thus impairing their function. In the largest cohort studied up to now of 118 CD138-purified plasma cell samples from previously untreated immunoglobulin light chain amyloidosis patients, we assessed in parallel copy number alterations using high-density copy number arrays and interphase fluorescence in situ hybridization (iFISH). We used fluorescence in situ hybridization probes for the IgH translocations t(11;14), t(4;14), and t(14;16) or any other IgH rearrangement as well as numerical aberrations of the chromosome loci 1q21, 8p21, 5p15/5q35, 11q22.3 or 11q23, 13q14, 15q22, 17p13, and 19q13. Recurrent gains included chromosomes 1q (36%), 9 (24%), 11q (24%), as well as 19 (15%). Recurrent losses affected chromosome 13 (29% monosomy) and partial losses of 14q (19%), 16q (14%) and 13q (12%), respectively. In 88% of patients with translocation t(11;14), the hallmark chromosomal aberration in AL amyloidosis, a concomitant gain of 11q22.3/11q23 detected by iFISH was part of the unbalanced translocation der(14)t(11;14)(q13;q32) with the breakpoint in the CCND1/MYEOV gene region. Partial loss of chromosome regions 14q and 16q were significantly associated to gain 1q. Gain 1q21 detected by iFISH almost always resulted from a gain of the long arm of chromosome 1 and not from trisomy 1, whereas deletions on chromosome 1p were rarely found. Overall and event-free survival analysis found a potential adverse prognostic effect of concomitant gain 1q and deletion 14q as well as of deletion 1p. In conclusion, in the first whole genome report of clonal plasma cells in AL amyloidosis, novel aberrations and hitherto unknown potential adverse prognostic effects were uncovered.
- Published
- 2017
27. Systemtherapie prognostisch ungünstiger CUP-Syndrome
- Author
-
Alwin Krämer and Tilmann Bochtler
- Subjects
0301 basic medicine ,Gynecology ,03 medical and health sciences ,medicine.medical_specialty ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cancer of unknown primary ,business.industry ,030220 oncology & carcinogenesis ,Medicine ,Hematology ,business - Abstract
Beim CUP-Syndrom („cancer of unknown primary“) handelt es sich um eine Tumorerkrankung, bei der eine Metastasierung klinisch und histologisch gesichert ist, aber trotz ausfuhrlicher Diagnostik kein Primartumor nachgewiesen werden kann. CUP-Syndrome machen etwa 2–3 % aller soliden Tumorerkrankungen aus. In den meisten Fallen zeigt sich histologisch ein Adenokarzinom oder ein undifferenziertes Karzinom. An einem CUP-Syndrom erkrankte Patienten, die nicht lokal mit Operation oder Strahlentherapie behandelt werden konnen und deren Erkrankung nicht einer der definierten gunstigen Untergruppen zuzuordnen ist, werden typsicherweise mit einer empirischen Chemotherapie behandelt, auch wenn die Evidenz hierfur aus klinischen Studien aufgrund der Heterogenitat und Seltenheit des CUP-Syndroms begrenzt ist. Dabei scheint die Kombination eines der Platinpraparate Carboplatin oder Cisplatin mit einem Taxan wie Paclitaxel am effektivsten zu sein. Alternativ kommt auch eine Behandlung mit einem Platinpraparat zusammen mit Gemcitabin in Betracht. Dreifachkombinationen scheinen keine relevante Verbesserung der Prognose zu erreichen. Noch ist unklar, ob das Ansprechen auf die Chemotherapie durch die Hinzunahme eines Antikorpers verbessert werden kann. Zu dieser Fragestellung werden aktuell die Ergebnisse der deutschen PACET-CUP-Studie erwartet, die randomisiert die Hinzunahme des Antikorpers Cetuximab zu einer Chemotherapie mit Carboplatin und Paclitaxel untersucht. Da die Prognose dieser Patienten trotz empirischer Chemotherapie enttauschend ist und die meisten Patienten binnen 2 Jahren versterben, werden grose Hoffnungen in neue Substanzen gesetzt, die nach einer Mutationsanalyse des Tumorgewebes zielgerichtet eingesetzt werden konnen. Ein entsprechendes Studienkonzept wird aktuell erarbeitet. Disseminierte CUP-Syndrome werden chemotherapeutisch behandelt. Dabei werden typischerweise Kombinationstherapien eines Platinpraparats mit Paclitaxel oder Gemcitabin eingesetzt. Zunehmend kommen auch zielgerichtete Substanzen auf der Grundlage von Mutationsanalysen des Tumorgewebes zum Einsatz.
- Published
- 2017
28. Impact of Genetic Abnormalities and Measurable Residual Disease Levels on Outcome in Patients with MDS/AML Pre-Emptively Treated with Azacitidine: Correlative Results of the Prospective RELAZA2 Trial
- Author
-
Marika Mende, Malte von Bonin, Antje Schubert, Anne Kubasch, Richard Noppeney, Marion Subklewe, Sabine Kayser, Friedrich Stoelzel, Michael Kramer, Gesine Bug, Johannes Schetelig, Gerhard Ehninger, Sebastian Stasik, Schumacher Martin, Christian Thiede, Jan-Henrik Mikesch, Juergen Novotny, Katharina Götze, Mathias Haenel, Anke Mütherig, Alwin Krämer, Dominik Wolf, Regina Herbst, Karsten Spiekermann, Lars Fransecky, Tilmann Bochtler, Uwe Platzbecker, Christoph Röllig, Carsten Müller-Tidow, Jan Moritz Middeke, Katja Sockel, Matthias Stelljes, Hubert Serve, Claudia D. Baldus, Ulrich Duehrsen, and Martin Bornhäuser
- Subjects
medicine.medical_specialty ,Myeloid ,business.industry ,Myelodysplastic syndromes ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,Disease ,medicine.disease ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Clinical endpoint ,Conventional chemotherapy ,In patient ,business ,medicine.drug - Abstract
Background: Monitoring of measurable residual disease (MRD) in patients (pts) with advanced myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) who achieve complete remission (CR) can predict hematological relapse. Recently published data from the first cohort of the RELAZA2-trial have shown that pre-emptive therapy with azacitidine (AZA) can prevent or substantially delay an overt relapse in MRD-positive pts with MDS or AML (Platzbecker et al. Lancet Oncol. 2018). Aims: To evaluate outcome of the entire patient cohort of the RELAZA2-trial and determine whether MRD-guided pre-emptive AZA treatment could prevent relapse in molecularly defined cohorts. Methods: Between 12/2011 and 07/2018 380 pts with advanced MDS or AML, who had achieved CR after conventional chemotherapy or allogeneic hematopoietic stem-cell transplantation (allo-HCT) were prospectively screened for MRD in monthly intervals either in bone marrow (BM) or peripheral blood (PB). A total of 94 pts (AML, n=83; MDS, n=11) became MRD positive during 24 months from baseline by either quantitative PCR (qPCR) or analysis of CD34+ donor-chimerism and entered the treatment phase. Preemptive MRD-triggered treatment consisted of AZA 75 mg/m2 per day subcutaneously on days 1-7 of a 29-day cycle for up to 24 cycles. After six cycles, MRD status was reassessed and pts with MRD negativity were eligible for treatment de-escalation. Primary endpoint was relapse-free survival (RFS) six months after start of pre-emptive treatment. For mutational analysis next generation sequencing (NGS) with a panel of 54 genes was performed (Illumina Trusight Myeloid). Results: Median age was 60 yrs (range: 22-80 yrs); 52 (55%) of the pts were female. Prior therapy consisted of chemotherapy in 42 (45%) and allo-HCT in 52 (55%) of the pts. Cytogenetics could be analyzed in 93 (99%) of the 94 pts. Risk categorization according to ELN 2017 was favorable in 30 (37%), intermediate in 31 (38%) and adverse in 21 (26%) of the AML pts, respectively. Type of MDS was advanced in all 11 pts and all were previously transplanted. Fifty-two (61%) of 85 pts with available NPM1 status were positive. NGS on 64 (68%) pts with available DNA at the time of first diagnosis revealed additional mutations in DNMT3A (n=25), TET2 (n=15), FLT3-ITD (n=12), IDH1 (n=9), FLT3-TKD (n=8), ASXL1, NRAS, TP53 (n=7, each), IDH2 (n=6), PTPN11, WT1 (n=5, each), GATA2, U2AF1 (n=4, each), CBL (n=3), CEBPA, CSFR3, CUX1, EZH2, KIT, RAD21, RUNX1, SF3B, STAG2, ZRSR2 (n=2, each), and KRAS (n=1). MRD data were correlated with outcome in 45 pts for NPM1, in 3 for RUNX1-RUNX1T1, whereas CD34-donor-chimerism was analyzed in 39 pts (missing, n=7). There was a significant faster and deeper decline of MRD in PB as compared to BM (P=0.03). The same held true with regard to the increase of donor-chimerism, which was achieved faster in PB as compared to BM (P=0.05). Secondary molecular abnormalities (MAs) had no impact on MRD response as measured by qPCR, which was also true if MAs were categorized functionally. Similarly, additional chromosomal abnormalities had no impact on MRD response in both MRD methods. However, in pts with measurement of donor-chimerism ASXL1 mutations were a negative factor for MRD response (P Conclusions: AZA as a pre-emptive therapy was effective in delaying hematological relapse of advanced MDS or AML pts, regardless of the underlying genetic signature. Based on these encouraging results, intensifying treatment with AZA in combination with pembrolizumab is currently investigated as MRD-guided treatment in NPM1 positive AML (PEMAZA; ClinicalTrials.gov Identifier: NCT03769532). Disclosures Wolf: Celgene: Honoraria, Research Funding. Bug:Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Hexal: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Eurocept: Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel; Jazz: Honoraria; Neovii: Other: Travel; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: Travel; Sanofi: Other: Travel. Götze:Celgene: Research Funding. Stelljes:Amgen: Consultancy, Speakers Bureau; Pfizer: Consultancy, Honoraria, Research Funding, Speakers Bureau. Subklewe:Celgene: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Seattle Genetics: Research Funding; Morphosys: Research Funding; Janssen: Consultancy; AMGEN: Consultancy, Honoraria, Research Funding; Roche AG: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Gilead Sciences: Consultancy, Honoraria, Research Funding. Haenel:Amgen, Novartis, Roche, Celgene, Takeda, Bayer: Honoraria. Rollig:Amgen, Astellas, BMS, Daiichi Sankyo, Janssen, Roche: Consultancy; Abbvie, Novartis, Pfizer: Consultancy, Research Funding. Müller-Tidow:Pfizer: Research Funding, Speakers Bureau; Daiichi Sankyo: Research Funding; BiolineRx: Research Funding; Janssen-Cilag GmbH: Speakers Bureau. Platzbecker:Novartis: Consultancy, Honoraria, Research Funding; Amgen: Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria; Geron: Consultancy, Honoraria. Thiede:AgenDix GmbH: Other: Co-owner and CEO. OffLabel Disclosure: Off-label: treatment with azacitidine to prevent or substantially delay an overt relapse in MRD-positive patients with MDS or AML
- Published
- 2020
29. Integrated clinicomolecular characterization identifies RAS activation and CDKN2A deletion as independent adverse prognostic factors in cancer of unknown primary
- Author
-
Lukas C. Heukamp, Albrecht Stenzinger, Olaf Neumann, Tilmann Bochtler, Volker Endris, Anna Reiling, Michael Allgäuer, Harald Löffler, Thomas Hielscher, Martina Kirchner, Anna-Lena Volckmar, Daniel Kazdal, Peter Schirmacher, Wilko Weichert, Alwin Krämer, Jonas Leichsenring, and Jan Budczies
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,DNA Copy Number Variations ,Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,Context (language use) ,Adenocarcinoma ,Malignancy ,medicine.disease_cause ,Targeted therapy ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,CDKN2A ,Internal medicine ,Medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 4 ,Copy-number variation ,Receptor, Notch1 ,Cyclin-Dependent Kinase Inhibitor p16 ,business.industry ,High-Throughput Nucleotide Sequencing ,Fibroblast growth factor receptor 4 ,medicine.disease ,CDKN2A Deletion ,Primary tumor ,Cancer of unknown primary ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Neoplasms, Unknown Primary ,Female ,KRAS ,Tumor Suppressor Protein p53 ,business - Abstract
Background: Cancer of unknown primary (CUP) denotes a malignancy with histologically confirmed metastatic spread while the primary tumor remains elusive. Here, we address prognostic and therapeutic implications of mutations and copy number variations (CNVs) detected in tumor tissue in the context of a comprehensive clinical risk assessment. Methods: Targeted panel sequencing was performed in 252 CUP patients. 71.8% of patients had unfavorable CUP according to ESMO guidelines. 74.7% were adeno- and 13.7% squamous cell carcinomas. DNA was extracted from micro-dissected formalin-fixed, paraffin-embedded tissues. For library preparation, mostly multiplex PCR-based Ion Torrent AmpliSeqTM technology with Oncomine comprehensive assays was used. Findings: Most frequent genetic alterations were mutations/deletions of TP53 (49.6%), CDKN2A (19.0%) and NOTCH1 (14.1%) as well as oncogenic activation of KRAS (23.4%), FGFR4 (14.9%) and PIK3CA (10.7%). KRAS activation was predominantly found in adenocarcinomas (P=0.01), PIK3CA activation in squamous cell carcinomas (P=0.03). Male sex, high ECOG score, unfavorable CUP, higher number of involved organs and RAS activation predicted decreased event-free and overall survival in multivariate analysis. Deletions of CDKN2A were prognostically adverse regarding overall survival. TP53 mutations did not significantly influence prognosis in the overall cohort, but worsened prognosis in otherwise favorable CUP subtypes. Although not standard in CUP, for 17/198 (8.6%) patients molecularly targeted treatment was recommended and 10 patients (5.1%) were treated accordingly. Interpretation: Besides the identification of drug targets, panel sequencing in CUP is prognostically relevant, with RAS activation and CDKN2A deletion emerging as novel independent risk factors in a comprehensive assessment with clinico-pathological data. Funding: None. Declaration of Interest: TB and AK work as study oncologists for the CUPISCO trial, which is sponsored by Roche, and have received reimbursement for study‐related travels as well as remuneration for their work as study oncologists for the benefit of their employer. VE: advisory board and lecture fees from AstraZeneca and ThermoFisher. ALV: lecture fees from AstraZeneca. PS: advisory board honoraria from Pfizer, Roche, Novartis and AstraZeneca as well as speaker's honoraria and research funding from Roche, AstraZeneca and Novartis. WW: advisory board honoraria from MSD, BMS, Roche, AZ, Novartis, Celgene, Merck, Pfizer; speaker's honoraria from MSD, BMS, Roche, AZ, Boehringer, Lilly, Amgen, Takeda, Novartis; research funding (to institution): Roche, MSD, BMS. AS: advisory board honoraria from Bayer, Novartis, BMS, AstraZeneca, ThermoFisher, Illumina; speaker's honoraria from Takeda, Roche, BMS, Illumina, AstraZeneca, Novartis, ThermoFisher, Bayer, MSD and research funding from Chugai and BMS. All remaining authors have declared no conflicts of interest. Ethical Approval: Ethics board approval from the University of Heidelberg was obtained and patients signed informed consent.
- Published
- 2019
30. RUNX1-mutated families show phenotype heterogeneity and a somatic mutation profile unique to germline predisposed AML
- Author
-
Andrew Dubowsky, Jinghua Feng, Amanda Wells, Stefan Fröhling, Meryl Altree, Andreas W. Schreiber, Sue Morgan, Lesley Rawlings, Richard J D'Andrea, Anna L. Brown, Chan-Eng Chong, Joëlle Michaud, Andrew H. Wei, Georges Natsoulis, Jeffrey Suttle, Rachel Susman, Cassandra Vakulin, Tilmann Bochtler, Uday R. Popat, Mark S. Currie, Paul Wang, Milena Babic, Ella J Wilkins, Christopher N. Hahn, Miriam Fine, Xiaochun Li, Jessica Burdett, Belinda Mercorella, Catherine Carmichael, Nigel Patton, Denae Henry, Marshall S. Horwitz, Peer Arts, Kerry Phillips, Julian Cooney, Sarah Moore, Sally Mapp, Nicola K. Poplawski, Thuong Ha, Sarah L King-Smith, Louise Jaensch, Shai Izraeli, Devendra K Hiwase, Julia Dobbins, Lucy A. Godley, Cecily Forsyth, Kenneth F. Bradstock, Carolyn M. Butcher, Helen Mar Fan, Grace McKavanagh, Hugh Y. Rienhoff, Hamish S. Scott, Mario Nicola, Elli Papaemmanuil, Ping Cannon, Ian D. Lewis, Claire C. Homan, Peter J. Brautigan, Alwin Krämer, Brown, Anna L, Arts, Peer, Babic, Milena, Dobbins, Julia, Feng, Jinghua, Ha, Thuong, Homan, Claire C, King-Smith, Sarah L, Li, Xiao-Chun, Brautigan, Peter, Butcher, Carolyn, D'Andrea, Richard J, Hahn, Christopher N, and Scott, Hamish S
- Subjects
Genetics ,Mutation ,Myeloid Neoplasia ,Somatic cell ,Genetic heterogeneity ,Platelet disorder ,Hematology ,Biology ,medicine.disease_cause ,Phenotype ,Germline ,Epigenesis, Genetic ,Pedigree ,Leukemia, Myeloid, Acute ,Germline mutation ,Germ Cells ,hemic and lymphatic diseases ,embryonic structures ,Core Binding Factor Alpha 2 Subunit ,medicine ,Humans ,Allele - Abstract
First reported in 1999, germline runt-related transcription factor 1 (RUNX1) mutations are a well-established cause of familial platelet disorder with predisposition to myeloid malignancy (FPD-MM). We present the clinical phenotypes and genetic mutations detected in 10 novel RUNX1-mutated FPD-MM families. Genomic analyses on these families detected 2 partial gene deletions, 3 novel mutations, and 5 recurrent mutations as the germline RUNX1 alterations leading to FPD-MM. Combining genomic data from the families reported herein with aggregated published data sets resulted in 130 germline RUNX1 families, which allowed us to investigate whether specific germline mutation characteristics (type, location) could explain the large phenotypic heterogeneity between patients with familial platelet disorder and different HMs. Comparing the somatic mutational signatures between the available familial (n = 35) and published sporadic (n = 137) RUNX1-mutated AML patients showed enrichment for somatic mutations affecting the second RUNX1 allele and GATA2. Conversely, we observed a decreased number of somatic mutations affecting NRAS, SRSF2, and DNMT3A and the collective genes associated with CHIP and epigenetic regulation. This is the largest aggregation and analysis of germline RUNX1 mutations performed to date, providing a unique opportunity to examine the factors underlying phenotypic differences and disease progression from FPD to MM.
- Published
- 2019
31. RNA-Based Detection of Gene Fusions in Formalin-Fixed and Paraffin-Embedded Solid Cancer Samples
- Author
-
Martina Kirchner, Olaf Neumann, Anna-Lena Volckmar, Fabian Stögbauer, Michael Allgäuer, Daniel Kazdal, Jan Budczies, Eugen Rempel, Regine Brandt, Suranand Babu Talla, Moritz von Winterfeld, Jonas Leichsenring, Tilmann Bochtler, Alwin Krämer, Christoph Springfeld, Peter Schirmacher, Roland Penzel, Volker Endris, and Albrecht Stenzinger
- Subjects
NGS ,gene fusion ,solid tumor ,carcinoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,targeted therapy ,lcsh:RC254-282 ,Article - Abstract
Oncogenic gene fusions are important drivers in many cancer types, including carcinomas, with diagnostic and therapeutic implications. Hence, sensitive and rapid methods for parallel profiling in formalin-fixed and paraffin-embedded (FFPE) specimens are needed. In this study we analyzed gene fusions in a cohort of 517 cases where standard treatment options were exhausted. To this end the Archer®, DX Solid tumor panel (AMP, 285 cases) and the Oncomine Comprehensive Assay v3 (OCA, 232 cases) were employed. Findings were validated by Sanger sequencing, fluorescence in situ hybridization (FISH) or immunohistochemistry. Both assays demonstrated minimal dropout rates (AMP: 2.4%, n = 7/292, OCA: 2.1%, n = 5/237) with turnaround times of 6&ndash, 9 working days (median, OCA and AMP, respectively). Hands-on-time for library preparation was 6 h (AMP) and 2 h (OCA). We detected n = 40 fusion-positive cases (7.7%) with TMPRSS2::ERG in prostate cancer being most prevalent (n = 9/40, 22.5%), followed by other gene fusions identified in cancers of unknown primary (n = 6/40, 15.0%), adenoid cystic carcinoma (n = 7/40, 17.5%), and pancreatic cancer (n = 7/40, 17.5%). Our results demonstrate that targeted RNA-sequencing of FFPE samples is feasible, and a well-suited approach for the detection of gene fusions in a routine clinical setting.
- Published
- 2019
32. TP53 deficiency permits chromosome abnormalities and karyotype heterogeneity in acute myeloid leukemia
- Author
-
Anna, Cazzola, Christin, Schlegel, Ilka, Jansen, Tilmann, Bochtler, Anna, Jauch, and Alwin, Krämer
- Subjects
Chromosome Aberrations ,Aneuploidy ,Prognosis ,Leukemia, Myeloid, Acute ,Cell Line, Tumor ,Karyotyping ,Mutation ,Humans ,CRISPR-Cas Systems ,Tumor Suppressor Protein p53 ,Gene Deletion ,In Situ Hybridization, Fluorescence ,Cell Proliferation ,DNA Damage - Abstract
Abnormal karyotypes are common in cancer cells and frequently observed in acute myeloid leukemia (AML), in which complex karyotype aberrations are associated with poor prognosis. How exactly abnormal karyotypes arise and are propagated in AML is unclear. TP53 mutations and deletions are frequent in complex karyotype AML, suggesting a role of TP53 alterations in the development of chromosome abnormalities. Here, we generated isogenic TP53-knockout versions of the euploid AML cell line EEB to investigate the impact of TP53 on karyotype stability. We show that chromosome abnormalities spontaneously arise in TP53-deficient cells. Numerical aneuploidy could, to some extent, be propagated in a TP53-proficient setting, indicating that it does not necessarily trigger TP53 activation. In contrast, tolerance to structural chromosome aberrations was almost entirely restricted to TP53-knockout clones, all of which were able to continue proliferation in the presence of damaged DNA. Mechanistically, as a source of chromosome aberrations, limited numerical but not structural chromosomal instability was tolerated by TP53-wildtype cells. In contrast, structural instability was found only in TP53-knockout cells. Together, in myeloid cells TP53 loss allows for the development of complex karyotype aberrations and karyotype heterogeneity by perpetuation of chromosome segregation errors.
- Published
- 2019
33. Fludarabine-treosulfan compared to thiotepa-busulfan-fludarabine or FLAMSA as conditioning regimen for patients with primary refractory or relapsed acute myeloid leukemia: a study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation (EBMT)
- Author
-
Francesco Saraceni, Nicolaus Kröger, Hélène Labussière-Wallet, Donald Bunjes, Dietrich W. Beelen, Arnon Nagler, Johanna Tischer, Bipin N. Savani, Arne Brecht, Hermann Einsele, Matthias Eder, Tilmann Bochtler, Mohamad Mohty, Myriam Labopin, and Dietger Niederwieser
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Transplantation Conditioning ,Medizin ,Gastroenterology ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Acute myeloid leukemia (AML) ,Acute leukemia ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,lcsh:Diseases of the blood and blood-forming organs ,Hematology ,Middle Aged ,Total body irradiation ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Fludarabine ,Europe ,Leukemia, Myeloid, Acute ,Fludarabine-treosulfan (FT) ,Oncology ,Thiotepa-busulfan-fludarabine (TBF) ,030220 oncology & carcinogenesis ,Female ,Vidarabine ,medicine.drug ,Adult ,medicine.medical_specialty ,Cyclophosphamide ,ThioTEPA ,Treosulfan ,lcsh:RC254-282 ,Young Adult ,Unrelated donor (UD) ,03 medical and health sciences ,Conditioning regimen ,Internal medicine ,medicine ,Humans ,ddc:610 ,Busulfan ,Molecular Biology ,Aged ,Retrospective Studies ,lcsh:RC633-647.5 ,business.industry ,Research ,Fludarabine, intermediate dose Ara-C, amsacrine, total body irradiation/busulfan, cyclophosphamide (FLAMSA) ,Active disease ,030104 developmental biology ,Allogeneic transplantation ,business ,Thiotepa ,Sibling donor (MSD) - Abstract
Background Limited data is available to guide the choice of the conditioning regimen for patients with acute myeloid leukemia (AML) undergoing transplant with persistent disease. Methods We retrospectively compared outcome of fludarabine-treosulfan (FT), thiotepa-busulfan-fludarabine (TBF), and sequential fludarabine, intermediate dose Ara-C, amsacrine, total body irradiation/busulfan, cyclophosphamide (FLAMSA) conditioning in patients with refractory or relapsed AML. Results Complete remission rates at day 100 were 92%, 80%, and 88% for FT, TBF, and FLAMSA, respectively (p = 0.13). Non-relapse mortality, incidence of relapse, acute (a) and chronic (c) graft-versus-host disease (GVHD) rates did not differ between the three groups. Overall survival at 2 years was 37% for FT, 24% for TBF, and 34% for FLAMSA (p = 0.10). Independent prognostic factors for survival were Karnofsky performance score and patient CMV serology (p = 0.01; p = 0.02), while survival was not affected by age at transplant. The use of anti-thymocyte globulin (ATG) was associated with reduced risk of grade III–IV aGVHD (p = 0.02) and cGVHD (p = 0.006), with no influence on relapse. Conclusions In conclusion, FT, TBF, and FLAMSA regimens provided similar outcome in patients undergoing transplant with active AML. Survival was determined by patient characteristics as Karnofsky performance score and CMV serology, however was not affected by age at transplant. ATG appears able to reduce the incidence of acute and chronic GVHD without influencing relapse risk. Electronic supplementary material The online version of this article (10.1186/s13045-019-0727-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
34. In Reply
- Author
-
Tilmann Bochtler
- Subjects
General Medicine - Published
- 2019
35. 519P Patterns of care and outcomes in carcinoma of unknown primary: A SEER-Medicare study
- Author
-
Marlene Thomas, Tilmann Bochtler, M. Mueller-Ohldach, Linda Mileshkin, Andreas Beringer, A. Surinach, Alwin Krämer, A. Gondos, and C. Perret
- Subjects
Patterns of care ,Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Unknown primary ,medicine ,Carcinoma ,Hematology ,Seer medicare ,business ,medicine.disease - Published
- 2020
36. Enasidenib
- Author
-
Alwin, Krämer and Tilmann, Bochtler
- Subjects
Leukemia, Myeloid, Acute ,Mice ,Triazines ,Mutation ,Aminopyridines ,Animals ,Humans ,Antineoplastic Agents ,Isocitrate Dehydrogenase - Abstract
Enasidenib is an orally available, selective, potent, small molecule inhibitor of mutant isocitrate dehydrogenase 2 (IDH2). Neomorphic mutations in IDH2 are frequently found in both hematologic malignancies and solid tumors and lead to the production of the oncometabolite (R)-2-hydroxyglutarate. Increased levels of (R)-2-hydroxyglutarate cause histone and DNA hypermethylation associated with blocked differentiation and tumorigenesis. In PDX mice transplanted with human IDH2-mutant acute myeloid leukemia cells, enasidenib treatment led to normalization of (R)-2-hydroxyglutarate serum levels, differentiation of leukemic blasts and increased survival. Early clinical data in patients with relapsed/refractory IDH2-mutant acute myeloid leukemia show that enasidenib is well tolerated and induces durable complete remissions as a single agent in about 20% of cases. One notable drug-related adverse effect is differentiation syndrome. On the basis of these results the compound has recently been approved for the treatment of relapsed/refractory IDH2-mutant acute myeloid leukemia in the USA. Although no data are available yet, clinical trials on the treatment of patients with several types of IDH2-mutant solid tumors including gliomas, chondrosarcomas and cholangiocarcinomas are currently being performed.
- Published
- 2018
37. Cytogenetic intraclonal heterogeneity of plasma cell dyscrasia in AL amyloidosis as compared with multiple myeloma
- Author
-
Maximilian Merz, Christoph Kimmich, Korbinian Hoffmann, Anna Jauch, Hartmut Goldschmidt, Marc S. Raab, Stefan Schönland, Martin Granzow, Jens Hillengass, Alwin Krämer, Anja Seckinger, Dirk Hose, Tobias Dittrich, Carsten Müller-Tidow, Thomas Hielscher, Ute Hegenbart, Tilmann Bochtler, Hematology, and Basic (bio-) Medical Sciences
- Subjects
0301 basic medicine ,Adult ,Male ,Clone (cell biology) ,Plasma cell dyscrasia ,Paraproteinemias ,Multiple Myeloma/genetics ,Biology ,Cytogenetics/methods ,Immunoglobulin light chain ,03 medical and health sciences ,Cytogenetics ,0302 clinical medicine ,medicine ,AL amyloidosis ,Humans ,Immunoglobulin Light-chain Amyloidosis ,Multiple myeloma ,Aged ,Aged, 80 and over ,Paraproteinemias/genetics ,Lymphoid Neoplasia ,medicine.diagnostic_test ,hematology ,Middle Aged ,medicine.disease ,Molecular biology ,030104 developmental biology ,Immunoglobulin Light-chain Amyloidosis/genetics ,030220 oncology & carcinogenesis ,Female ,Hyperdiploidy ,Multiple Myeloma ,Monoclonal gammopathy of undetermined significance ,Fluorescence in situ hybridization - Abstract
Analysis of intraclonal heterogeneity has yielded insights into the clonal evolution of hematologic malignancies. We compared the clonal and subclonal compositions of the underlying plasma cell dyscrasia in 544 systemic light chain amyloidosis (PC-AL) patients with 519 patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), or symptomatic MM; ie, PC–non-AL patients). Using interphase fluorescence in situ hybridization, subclones were stringently defined as clone size below two thirds of the largest clone and an absolute difference of ≥30%. Subclones were found less frequently in the PC-AL group, at 199 (36.6%) of 544 as compared with 267 (51.4%) of 519 in the PC–non-AL group (P < .001), and were not associated with the stage of plasma cell dyscrasia in either entity. In both groups, translocation t(11;14), other immunoglobulin heavy chain translocations, and hyperdiploidy were typically found as main clones, whereas gain of 1q21 and deletions of 8p21, 13q14, and 17p13 were frequently found as subclones. There were no shifts in the subclone/main clone ratio depending on the MGUS, SMM, or MM stage of plasma cell dyscrasia. In multivariate analysis, t(11;14) was associated with lower rates of subclone formation and hyperdiploidy with higher rates. PC-AL itself lost statistical significance, demonstrating that the lower subclone frequency in AL is a reflection of its exceptionally high t(11;14) frequency. In summary, the subclone patterns in PC-AL and PC–non-AL are closely related, implying that subclone formation depends on the main cytogenetic categories and is independent of disease entity and stage.
- Published
- 2018
38. Enasidenib
- Author
-
Alwin Krämer and Tilmann Bochtler
- Published
- 2018
39. A challenging task – Identifying carcinoma of unknown primary (CUP) patients according to ESMO guidelines: The CUPISCO trial experience
- Author
-
Georgios Pentheroudakis, Stefan Foser, George Zarkavelis, Tilmann Bochtler, Andreas Beringer, Mustafa Ozguroglu, Chantal Pauli, Ferran Losa, M. Mueller-Ohldach, Alwin Krämer, J.S. Ross, J. Scarato, Holger Moch, Giulia Baciarello, Linda Mileshkin, and S Songül Yalçin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Conflict of interest ,Diagnostic algorithms ,Hematology ,Medical writing ,Clinical trial ,03 medical and health sciences ,Task (computing) ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Honorarium ,Unknown primary ,Pathology laboratory ,Medicine ,business - Abstract
Background The CUPISCO trial (NCT03498521) is an ongoing, phase II, randomised, multicentre study comparing molecularly-guided therapy with standard platinum-based chemotherapy in newly diagnosed poor-risk CUP patients. Methods Eligible patients have poor-risk adeno- or undifferentiated CUP as defined by ESMO 2015 guidelines and tissue for molecular sequencing. Local sites initiate the screening process with potentially eligible patients. Patients then undergo central Eligibility Review (ER), a cooperative effort between a central pathology laboratory, external referent oncologists and each site’s investigator and pathology laboratory to confirm the diagnosis. Patients with favourable prognostic subsets or with a strong suspicion of an existing primary site of origin based on immunohistochemistry (IHC) signature and clinical picture are excluded. Results As of 19 March 2019, 157 patients had been screened, of whom 91 (58%) failed screening. Three patients were successfully re-screened. Of the 88 patients who permanently failed screening, 23 were due to technical reasons (e.g. insufficient quality/quantity of tissue for sequencing), 20 for failure to meet inclusion/exclusion criteria not directly related to CUP diagnosis, and 14 for other reasons (e.g. declining health status). A set of 31 patients were not enrolled because the CUP diagnosis could not be confirmed at the IHC level, 19 of those after ER review. Central IHC review results included pathological signatures more typical of specific primary tumours (e.g. prostate cancer or melanoma), or marker combinations typically positive in favourable CUP subsets or rare tumour entities. Conclusions Experience with the CUPISCO study has highlighted challenges with standardised screening and diagnostic processes in an international clinical trial and the difficulties inherent in accurate diagnosis of poor-risk CUP. Confirming a CUP diagnosis for a clinical trial with multiple review checkpoints can result in many reasons for screen failures. By sharing this experience, we aim to foster understanding and to improve diagnostic algorithms for CUP. Clinical trial identification NCT03498521. Editorial acknowledgement Medical writing assistance was provided by Ian Leighton, PhD, Nspm Ltd, Meggen, Switzerland, and supported by F. Hoffmann-La Roche Ltd, Basel, Switzerland. Legal entity responsible for the study F. Hoffmann-La Roche Ltd. Funding F. Hoffmann-La Roche Ltd. Disclosure C. Pauli: Honoraria (self), Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche. T. Bochtler: Honoraria (self), Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche. L. Mileshkin: Travel / Accommodation / Expenses: Roche; Travel / Accommodation / Expenses: Beigene. G. Baciarello: Advisory / Consultancy, Travel / Accommodation / Expenses: Amgen; Advisory / Consultancy, Travel / Accommodation / Expenses: Janssen Oncology; Advisory / Consultancy, Travel / Accommodation / Expenses: Sanofi; Advisory / Consultancy, Travel / Accommodation / Expenses: Astellas-Pharma; Advisory / Consultancy: Roche; Travel / Accommodation / Expenses: AstraZeneca; Travel / Accommodation / Expenses: Ipsen. F. Losa: Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche; Advisory / Consultancy, Research grant / Funding (institution): Amgen; Advisory / Consultancy, Research grant / Funding (institution), Travel / Accommodation / Expenses: Merck; Advisory / Consultancy, Speaker Bureau / Expert testimony: Sanofi; Advisory / Consultancy: Servier. J.S. Ross: Shareholder / Stockholder / Stock options, Full / Part-time employment: Foundation Medicine Inc. S. Yalcin: Honoraria (self): Roche; Honoraria (self): Sanofi; Honoraria (self): Amgen; Honoraria (self): Novartis; Honoraria (self): Lilly; Honoraria (self): MSD; Honoraria (self): Merck Serono. A. Beringer: Full / Part-time employment: F. Hoffmann-La Roche Ltd.. S. Foser: Full / Part-time employment: F. Hoffmann-La Roche Ltd. J. Scarato: Full / Part-time employment: F. Hoffmann-La Roche Ltd.. M. Mueller-Ohldach: Full / Part-time employment: Hoffmann-La Roche Ltd. H. Moch: Honoraria (self), Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche. A. Kramer: Honoraria (self), Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche. All other authors have declared no conflicts of interest.
- Published
- 2019
40. Real-world implementation of sequential targeted therapies for EGFR-mutated NSCLC
- Author
-
Harland S. Winter, Michael Thomas, Anna-Lena Volckmar, T Muley, Helge Bischoff, Petros Christopoulos, M. Faehling, Tilmann Bochtler, Daniel Kazdal, Martina Kirchner, C.P. Heussel, Michael Meister, Juergen R. Fischer, Fjf Herth, Volker Endris, A. Stenzinger, N. Magios, Farastuk Bozorgmehr, J. Kuon, and Stefan Rieken
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Afatinib ,Clinical course ,Stock options ,Hematology ,University hospital ,Sequential treatment ,03 medical and health sciences ,Egfr tki ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,medicine ,Overall survival ,Osimertinib ,business ,medicine.drug - Abstract
Background Osimertinib is the preferable therapeutic option for many epidermal growth factor receptor (EGFR)+ non-small cell lung cancer (NSCLC) patients failing other tyrosine kinase inhibitors (TKI), but implementation of EGFR TKI sequencing is often problematic. Methods We retrospectively studied the clinical course of EGFR+ NSCLC patients that received first-/second-generation TKI at our institutions and had their last follow-up after osimertinib approval (02/2016). Results A total of n = 283 EGFR+ NSCLC patients received erlotinib (45%), gefitinib (19%) and/or afatinib (36%) in the 1st-4th treatment lines with a median age of 66 years, a median ECOG performance status of 0 (137/266 patients with available data) and a predominance of female (183/283=65%) never-/light-smokers (177/283=63%). Median overall survival (OS) from treatment start was 32.7 months (95% confidence interval [CI] 28.1 – 37.3) with 2.2 treatment lines on average (standard deviation 1.4). EGFR T790M testing was performed for 139/203 (68%) patients after TKI failure, with a positive result in 77/139 (55%) and subsequent treatment with osimertinib in 50/77 (65%). Overall, 50/203 (25%) of patients received osimertinib, with a median OS of 44.9 (27.9 – 62.1) months, significantly longer than the 30.4 (20.6 – 40.3) months for patients with alternative or no subsequent therapies (logrank p = 0.053, Breslow p = 0.002). Among the 134 deceased patients with complete follow-up, 84 (63%) received additional systemic treatment (37% chemotherapy, 16% osimertinib, 8% only alternative EGFR inhibitors, 2% only immunotherapy), while 50/134 (37%) died without next-line therapy. For patients that subsequently received chemotherapy, median time to start of chemotherapy was 11.6 (8.9 – 14.3) months. Conclusions Sequential treatment with osimertinib after first- or second-generation EGFR inhibitors significantly prolongs OS, but in the real-world setting a considerable fraction of patients will not be able to benefit from that. Main obstacles in our cohort were lack of EGFR T790M testing (32% of total cases), T790M-negative progression (45% of tested cases), and rapid clinical deterioration without the chance of next-line therapy (about one-third of patients). Legal entity responsible for the study Thoraxklinik at Heidelberg University Hospital. Funding Thoraxklinik at Heidelberg University Hospital AstraZeneca. Disclosure P. Christopoulos: Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): Novartis; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): Roche; Advisory / Consultancy: Chugai; Research grant / Funding (institution): AstraZeneca; Advisory / Consultancy: Boehringer; Research grant / Funding (institution): Takeda. F. Bozorgmehr: Honoraria (self), Research grant / Funding (institution), Travel / Accommodation / Expenses: BMS; Honoraria (self): MSD. J.B. Kuon: Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): Cellgene. V. Endris: Honoraria (self), Advisory / Consultancy: ThermoFisher; Honoraria (self), Advisory / Consultancy: AstraZeneca. T. Bochtler: Honoraria (institution), Research grant / Funding (institution): Roche. F.J.F. Herth: Honoraria (self), Research grant / Funding (institution): Lilly; Honoraria (self), Research grant / Funding (institution): Roche; Honoraria (self), Research grant / Funding (institution): AstraZeneca; Honoraria (self), Research grant / Funding (institution): Novartis; Honoraria (self), Research grant / Funding (institution): Boehringer; Honoraria (self), Research grant / Funding (institution): Chiesi; Honoraria (self), Research grant / Funding (institution): Teva; Honoraria (self): Pulmonx BTG; Honoraria (self): Olympus. C. Heussel: Honoraria (self), Honoraria (institution): Novartis, Basilea, Bayer, Grifols, Boehringer, Pierre Fabre, Covidien, Siemens, Chiesi, Intermune, MEDA Pharma, Bracco, Pfizer, MSD, Roche, Lilly, AstraZeneca, Schering-Plough, Essex, Gilead, MeVis, Fresenius, Astellas; Shareholder / Stockholder / Stock options: GSK. T. Muley: Honoraria (self), Research grant / Funding (self), Research grant / Funding (institution): Roche. J.R. Fischer: Advisory / Consultancy: Boehringer, Roche, Celgene and AstraZeneca. A. Stenzinger: Honoraria (self), Advisory / Consultancy: Novartis, AstraZeneca, ThermoFisher, BMS; Honoraria (self): BMS, Illumina, AstraZeneca, Novartis, ThermoFisher, MSD, Roche; Research grant / Funding (institution): Chugai; Honoraria (self): Illumina, AstraZeneca, Novartis, ThermoFisher . M. Thomas: Honoraria (self), Advisory / Consultancy: Novartis, Lilly, BMS, MSD, Roche, Celgene, Takeda, AbbVie, Boehringer, Lilly, MSD, Takeda; Research grant / Funding (institution): AstraZeneca, BMS, Celgene, Novartis, Roche, Takeda. All other authors have declared no conflicts of interest.
- Published
- 2019
41. Pretreatment d-2-hydroxyglutarate serum levels negatively impact on outcome in IDH1-mutated acute myeloid leukemia
- Author
-
Saadati M, Tilmann Bochtler, Anthony D. Ho, Alwin Krämer, Jörg Balss, von Deimling A, G. Ehninger, Axel Benner, Jürgen G. Okun, Christoph E. Heilig, Markus Schaich, Christian Thiede, and Stefan Pusch
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,IDH1 ,Adolescent ,Biology ,IDH2 ,Glutarates ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Survival rate ,Neoplasm Staging ,Proportional hazards model ,Myeloid leukemia ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Isocitrate Dehydrogenase ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,Isocitrate dehydrogenase ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Mutation ,Immunology ,Female ,Follow-Up Studies - Abstract
Mutations in isocitrate dehydrogenases (IDHs) 1 and 2 frequently occur in acute myeloid leukemia (AML) and result in the production of the oncometabolite d-2-hydroxyglutarate (D2HG). D2HG has been shown to promote leukemogenesis even in the absence of mutated IDH, but the prognostic significance of pretreatment serum D2HG levels in patients with IDH-mutated AML is unclear. We measured D2HG serum levels in 84 patients with IDH-mutated AML treated in the prospective, randomized multicenter AML2003 trial of the German Study Alliance Leukemia. Multivariate Cox regression showed D2HG levels to negatively impact on event-free survival (EFS) as a continuous variable in the entire IDH(mut) cohort (P=0.04), with no effect on overall survival (OS). In a subgroup analysis, the negative impact of D2HG on EFS was found to be restricted to patients with mutations in IDH1 (P=0.003), adjusted for age, leukocyte count, serum lactate dehydrogenase and European LeukemiaNet risk score. We thus conclude that pretreatment D2HG serum levels may yield prognostic information in patients with IDH1-mutated, but not in IDH2-mutated AML, possibly due to different subcellular localizations of IDH1 and IDH2.
- Published
- 2015
42. Translocation t(11;14) Is Associated With Adverse Outcome in Patients With Newly Diagnosed AL Amyloidosis When Treated With Bortezomib-Based Regimens
- Author
-
Christoph Kimmich, Martin Granzow, Tilmann Bochtler, Hartmut Goldschmidt, U Hegenbart, Christina Kunz, Anna Jauch, Axel Benner, Stefan Schönland, Anja Seckinger, Dirk Hose, Anthony D. Ho, Hematology, and Basic (bio-) Medical Sciences
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Chromosomal translocation ,Gastroenterology ,Dexamethasone ,Translocation, Genetic ,Internal medicine ,medicine ,AL amyloidosis ,Humans ,Boronic Acids/therapeutic use ,In patient ,Cyclophosphamide ,Amyloidosis/drug therapy ,In Situ Hybridization, Fluorescence ,Aged ,Retrospective Studies ,Chemotherapy ,medicine.diagnostic_test ,hematology ,Bortezomib ,business.industry ,Proportional hazards model ,Chromosomes, Human, Pair 11 ,bortezomib ,Retrospective cohort study ,Amyloidosis ,Middle Aged ,Prognosis ,medicine.disease ,Boronic Acids ,Dexamethasone/therapeutic use ,Surgery ,Pyrazines/therapeutic use ,Treatment Outcome ,Oncology ,Cyclophosphamide/therapeutic use ,Pyrazines ,Immunoglobulin Light Chains ,Female ,Chromosomes, Human, Pair 4 ,business ,Immunoglobulin Light Chains/immunology ,medicine.drug ,Fluorescence in situ hybridization - Abstract
Purpose Bortezomib has become a cornerstone in the treatment of AL amyloidosis. In this study, we addressed the prognostic impact of cytogenetic aberrations for bortezomib-treated patients. Patients and Methods We analyzed a consecutive series of 101 patients with AL amyloidosis treated with bortezomib-dexamethasone as first-line treatment by interphase fluorescence in situ hybridization (iFISH). Patients were ineligible for high-dose chemotherapy, which would put them at risk for cardiac or renal failure, and thus represented a poor-risk group. Results Presence of t(11;14), versus its absence, was associated with inferior hematologic event-free survival (median, 3.4 v 8.8 months, respectively; P = .002), overall survival (median, 8.7 v 40.7 months, respectively; P = .05), and remission rate (≥ very good partial remission; 23% v 47%, respectively; P = .02). In multivariable Cox regression models incorporating established hematologic and clinical risk factors, t(11;14) was an independent adverse prognostic marker for hematologic event-free survival (hazard ratio, 2.94; 95% CI, 1.37 to 6.25; P = .006) and overall survival (hazard ratio, 3.13; 95% CI, 1.16 to 8.33; P = .03), but not for remission (≥ very good partial remission). Markedly, the multiple myeloma high-risk iFISH aberrations t(4;14), t(14;16), del(17p), and gain of 1q21 conferred no adverse prognosis in this bortezomib-dexamethasone–treated group. After backward variable selection, the final multivariable model was validated in a consecutive series of 32 patients treated with bortezomib, dexamethasone, and cyclophosphamide. Conclusion iFISH results are important independent prognostic factors in AL amyloidosis. In contrast to our recently published results with melphalan and dexamethasone standard therapy, bortezomib is less beneficial to patients harboring t(11;14), whereas it effectively alleviates the poor prognosis inherent to high-risk aberrations. Given the discrepant response to different treatment modalities, iFISH may help to guide therapeutic choices in these poor-risk patients requiring rapid hematologic response.
- Published
- 2015
43. Role of chromosomal aberrations in clonal diversity and progression of acute myeloid leukemia
- Author
-
Tilmann Bochtler, Stefan Fröhling, and Alwin Krämer
- Subjects
Chromosome Aberrations ,Genetics ,Cancer Research ,Genetic Variation ,Myeloid leukemia ,Hematology ,Gene mutation ,Biology ,Prognosis ,medicine.disease ,Somatic evolution in cancer ,Chromosome aberration ,Clone Cells ,Leukemia, Myeloid, Acute ,Leukemia ,Oncology ,Fanconi anemia ,hemic and lymphatic diseases ,medicine ,Chromosome abnormality ,Humans ,Bloom syndrome - Abstract
Genetic abnormalities are a hallmark of cancer. Hereby, cytogenetic aberrations and small-scale abnormalities, such as single-nucleotide variations and insertion/deletion mutations, have emerged as two alternative modes of genetic diversification. Both mechanisms are at work in acute myeloid leukemia (AML), in which conventional karyotyping and molecular studies demonstrate that gene mutations occur predominantly in cytogenetically normal AML, whereas chromosomal changes are a driving force of development and progression of disease in aberrant karyotype AML. All steps of disease evolution in AML, ranging from the transformation of preleukemic clones into overt leukemia to the expansion and recurrence of malignant clones, are paralleled by clonal evolution at either the gene mutation or chromosome aberration level. Preleukemic conditions, such as Fanconi anemia and Bloom syndrome, demonstrate that the acquisition of chromosomal aberrations can contribute to leukemic transformation. Similar to what has been shown at the mutational level, expansion and recurrence of AML clones goes along with increasing genetic diversification. Hereby, cytogenetically more evolved subclones are at a proliferative advantage and outgrow ancestor clones or have evolved toward a more aggressive behavior with additional newly acquired aberrations as compared with the initial leukemic clone, respectively.
- Published
- 2015
44. Integrated Histogenetic Analysis Reveals
- Author
-
Tilmann, Bochtler, Volker, Endris, Anna, Reiling, Jonas, Leichsenring, Michal R, Schweiger, Sebastian, Klein, Fabian, Stögbauer, Benjamin, Goeppert, Peter, Schirmacher, Alwin, Krämer, and Albrecht, Stenzinger
- Subjects
Male ,Mesothelioma ,Lung Neoplasms ,Tumor Suppressor Proteins ,Mesothelioma, Malignant ,Mutation, Missense ,Neoplastic Syndromes, Hereditary ,Mutation ,Biomarkers, Tumor ,Humans ,Neoplasms, Unknown Primary ,Genetic Predisposition to Disease ,Ubiquitin Thiolesterase ,Aged - Abstract
This case report presents a male patient with epithelioid mesothelioma that was initially misdiagnosed as cancer of unknown primary (CUP) and correctly identified using molecular panel sequencing. The patient had a prior history of colon and breast cancer. To assess the enlarged mediastinal lymph nodes, retrosternal lymphadenectomy was performed in 2016. The lymph nodes were histologically deemed unrelated to the known breast cancer by the reference pathologist, thus leading to the diagnosis of a CUP syndrome. When the patient presented to our center, targeted deep sequencing of both breast cancer and presumed CUP was performed to address the clonal relationship between both malignancies. A missense mutation in
- Published
- 2017
45. AL amyloidosis patients with low amyloidogenic free light chain levels at first diagnosis have an excellent prognosis
- Author
-
Hartmut Goldschmidt, Anna Jauch, Ute Hegenbart, Anthony D. Ho, Christoph Kimmich, Natalia Becker, Stefan Schönland, Tilmann Bochtler, and Tobias Dittrich
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Amyloid ,Immunology ,Plasma Cells ,Bone Marrow Cells ,Immunoglobulin light chain ,Biochemistry ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,AL amyloidosis ,Medicine ,Humans ,Survival rate ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,Proteinuria ,business.industry ,Amyloidosis ,Plasmacytosis ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Free Light Chain ,Survival Rate ,030220 oncology & carcinogenesis ,Female ,Immunoglobulin Light Chains ,medicine.symptom ,business ,030215 immunology - Abstract
The difference between involved minus uninvolved serum free light chains (dFLC) has been established as an invaluable hematologic parameter in systemic amyloid light chain (AL) amyloidosis. However, patients with an initial dFLC level50 mg/L are currently deemed not evaluable for response to therapy. Therefore, we aimed to characterize this subgroup of patients and to define novel hematologic response parameters. We retrospectively analyzed 783 AL patients newly diagnosed at our center between 2002 and 2016. Patients with a dFLC level50 mg/L showed smaller bone marrow plasmacytosis compared to patients with a dFLC level ≥50 mg/L (7% vs 10%
- Published
- 2017
46. Azacitidine and low-dose cytarabine in palliative patients with acute myeloid leukemia and high bone marrow blast counts-a retrospective single-center experience
- Author
-
Tilmann Bochtler, Thomas Luft, Peter Dreger, Tilman Schöning, Anthony D. Ho, Sascha Dietrich, Alwin Krämer, and Aleksandar Radujkovic
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neutropenia ,Azacitidine ,Hemorrhage ,Single Center ,Gastroenterology ,Drug Administration Schedule ,Bone Marrow ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Palliative Care ,Cytarabine ,Myeloid leukemia ,Retrospective cohort study ,Pneumonia ,Hematology ,General Medicine ,Middle Aged ,medicine.disease ,Survival Analysis ,Surgery ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Karyotyping ,Toxicity ,Female ,Bone marrow ,business ,Febrile neutropenia ,medicine.drug - Abstract
We retrospectively analyzed and compared the efficacy and toxicity of azacitidine (AZA) and low-dose cytarabine (LD-Ara-C) in 65 palliative patients with acute myeloid leukemia (AML) showing high bone marrow blast counts (≥30%) before start of treatment. Twenty-seven and 38 patients received AZA and LD-Ara-C, respectively. The median patient age was 71 yr. Patient and disease characteristics did not differ between the treatment groups, except for BM blast counts, and peripheral leukocyte and blast counts which were significantly higher in the LD-Ara-C group. AZA and LD-Ara-C were first-line treatment in 12 (44%) and 17 patients (45%), respectively. Response and hematologic improvement rates were low and similar in both treatment groups. In both treatment groups, most common non-hematologic toxicities included febrile neutropenia, pneumonia, and bleedings without significant differences regarding frequencies. Estimated 1-yr survival rates were 15% (95% CI 8-22) and 13% (95% CI 7-19) in the AZA and LD-Ara-C groups, respectively, without statistically significant difference. In multivariate analysis (n = 65), previous treatment (HR 2.27, 95% CI 1.00-5.22, P = 0.05) and adverse cytogenetics (HR 2.50, 95% CI 1.20-5.22, P = 0.02) were independent predictors of poor survival. In our center and within the limitations of a retrospective study, both treatment regimens showed similar but limited efficacy in palliative patients with AML and high BM blast counts.
- Published
- 2014
47. Azacitidine for Pre-Emptive Treatment of Measurable-Residual Disease in MDS/AML Patients at High Risk of Hematological Relapse: Results of the Second Cohort of the RELAZA2 Trial
- Author
-
Carsten Müller-Tidow, Christoph Röllig, Juergen Novotny, Hubert Serve, Claudia D. Baldus, Karsten Spiekermann, Richard Noppeney, Matthias Stelljes, Uwe Platzbecker, Marika Mende, Ulrich Dührsen, Jan Moritz Middeke, Katja Sockel, Christian Thiede, Schumacher Martin, Katharina Götze, Michael Kramer, Gerhard Ehninger, Mathias Hänel, Anne Sophie Kubasch, Johannes Schetelig, Antje Schubert, Christoph Groth, Lars R. Fransecky, Martin Bornhäuser, Marion Subklewe, Alwin Krämer, Anke Mütherig, Regina Herbst, Tilmann Bochtler, Gesine Bug, and Dominik Wolf
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Azacitidine ,Medizin ,Cell Biology ,Hematology ,Disease ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Pre emptive treatment ,Recurrence risk ,Leukemia ,Internal medicine ,Cohort ,medicine ,business ,medicine.drug - Abstract
Background: Measurable residual disease (MRD) can identify patients (pts) with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) in complete hematological remission (CR) at high risk of relapse even after allogeneic hematopoietic stem cell transplantation (HSCT). We have recently shown in 53 pts treated within the first cohort of the RELAZA2 trial that pre-emptive therapy with azacitidine (AZA) at the time of MRD-positivity (MRDpos) can successfully prevent imminent hematological relapse (Platzbecker et al. Lancet Oncol. 2018). We now report on the results of the second cohort of 41 pts undergoing MRD-guided treatment in the RELAZA2 trial (ClinicalTrials.gov NCT01462578) by the Study Alliance Leukemia (SAL). Methods: Between 2015 and 2018, 166 MDS/AML pts were screened and centrally monitored for MRD in bone marrow or peripheral blood at monthly intervals for a period of 2 years prospectively in 9 centers in Germany. Of these 166, 41 pts with either advanced MDS (n=6) or AML (n=35) in CR after either conventional chemotherapy only (n=13) or consecutive allogeneic HSCT (n=28) developed MRD above a threshold defining imminent hematological relapse. Still being in morphological CR, these pts pre-emptively received 6 cycles of AZA (75mg/m2, s.c. days 1-7), which was followed by a risk-adapted AZA-maintenance therapy based on MRD-response for up to 18 additional months. Pts developing a hematological relapse went off study. MRD was detected by either the quantification of NPM1 mutation level (n=19), leukemia-specific fusion genes DEK-NUP214 (n=1) or RUNX1/RUNX1T1 (n=2) or a sensitive donor chimerism analysis of sorted CD34(+)/CD117(+) peripheral blood cells (n=28) in pts undergoing allogeneic HSCT. Here, we report the analysis of the primary endpoint of the 41 pts in the second cohort as well as the data for the entire 94 pts who entered the treatment phase of the RELAZA-2 study. Results: At a median of 110 days (range 28-476) after start of screening, 41 (25%) out of 166 prospectively screened pts became MRDpos as defined by either a decrease of CD34(+)/CD117(+) donor chimerism to 1% (NPM1 n=18) while being still in hematological CR. All of these MRDpos pts started AZA-based pre-emptive treatment to prevent imminent hematological relapse. Six months after start of MRD-guided therapy, 25 out of 41 pts were still in CR (61%, 95%-CI 45-76%, p Conclusion: These multicenter prospective data provide further strong evidence that continuous MRD monitoring is feasible and can identify MDS/AML pts at high risk of hematological relapse. Pre-emptive MRD-guided therapy with AZA is an effective treatment to prevent or at least substantially delay hematologic relapse in these pts. Disclosures Platzbecker: Novartis: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria. Wolf:Celgene: Honoraria, Research Funding; Abbvie: Honoraria. Krämer:Daiichi-Sankyo: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Bayer: Research Funding. Bug:Gilead Sciences: Membership on an entity's Board of Directors or advisory committees, Other: Travel grants; Jazz Pharmaceuticals: Honoraria; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel grants; Hexal: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; Sanofi: Other: travel grants; Celgene Neovii: Other: travel grant. Götze:AbbVie: Membership on an entity's Board of Directors or advisory committees. Stelljes:Novartis: Honoraria; Amgen: Honoraria; Jazz Pharmaceuticals: Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; MDS: Consultancy. Subklewe:AMGEN: Consultancy, Honoraria, Research Funding; Gilead: Consultancy, Honoraria, Research Funding; Miltenyi: Research Funding; Oxford Biotherapeutics: Research Funding; Janssen: Consultancy; Roche: Consultancy, Research Funding; Pfizer: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Morphosys: Research Funding. Hänel:Celgene: Other: advisory board; Novartis: Honoraria; Takeda: Other: advisory board; Amgen: Honoraria; Roche: Honoraria. Dührsen:Gilead: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Alexion: Honoraria; Takeda: Consultancy, Honoraria; Teva: Honoraria; Celgene: Research Funding; Roche: Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; Janssen: Honoraria; Amgen: Consultancy, Honoraria, Research Funding; CPT: Consultancy, Honoraria. Müller-Tidow:MSD: Membership on an entity's Board of Directors or advisory committees. Thiede:Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; AgenDix GmbH: Employment, Equity Ownership.
- Published
- 2019
48. RAS activation and CDKN2A deletion to predict prognosis in cancer of unknown primary
- Author
-
Peter Schirmacher, Michael Allgäuer, Tilmann Bochtler, Olaf Neumann, Thomas Hielscher, Alwin Kraemer, Anna Reiling, Lukas C. Heukamp, Anna-Lena Volckmar, J. Budczies, Jonas Leichsenring, Martina Kirchner, Albrecht Stenzinger, and Volker Endris
- Subjects
Cancer Research ,Oncology ,Cancer of unknown primary ,business.industry ,Cancer research ,Medicine ,business ,Malignancy ,medicine.disease ,Primary tumor ,CDKN2A Deletion - Abstract
e13026 Background: Cancer of unknown primary (CUP) denotes a malignancy with histologically confirmed metastatic spread while the primary tumor remains elusive. Given the typically dismal response to empiric chemotherapy, the deciphering of molecular profiles holds a particular promise for targeted therapy. Here, we address the prognostic implications of mutations and copy number variations (CNVs) detected in tumor tissue in the context of a comprehensive clinical risk assessment. Methods: We performed targeted panel sequencing in a series of 252 CUP patients. Median age was 60.3 years and 59.1% were female. 71.2% of patients had unfavorable CUP. Lymph nodes, bone marrow, lung, peritoneum and pleura were the most frequent metastatic sites. Among histologic subtypes, adeno- (74.7%) and squamous cell carcinomas (13.7%) prevailed. DNA was extracted from micro-dissected formalin-fixed, paraffin-embedded tissue samples. For library preparation mostly multiplex PCR-based Ion Torrent AmpliSeq technology (Life Technologies) with Oncomine comprehensive assays v1 and v3 was used. Results: The prevailing genetic alterations were deletions in tumor suppressor genes TP53 (49.6%), CDKN2A (19.0%) and NOTCH1 (14.1%) as well as oncogenic activation of KRAS (23.4%), FGFR4 (14.9%) and PIK3CA (10.7%). KRAS activation was predominantly found in adenocarcinomas (p = 0.01) and PIK3CA activation in squamous cell carcinomas (p = 0.03). Regarding prognosis, male sex, high ECOG score, unfavorable CUP subtype, a higher number of involved organs as well as RAS activation predicted decreased event-free and overall survival in multivariate analysis. Deletions in CDKN2A were a poor prognostic factor regarding overall survival. Markedly, TP53 mutations did not significantly influence prognosis. Since July 2015, 17/198 (8.6%) patients received a recommendation for molecularly guided therapy. However, due to lack of coverage by insurers or rapid clinical deterioration only 7 of these patients (3.5%) actually received targeted treatment. Conclusions: Besides offering targets for therapy, targeted panel sequencing in CUP is prognostically relevant, with RAS activation and CDKNA2 deletion emerging as independent risk factors in a comprehensive assessment with clinical risk factors.
- Published
- 2019
49. Clonal Heterogeneity As Detected by Metaphase Karyotyping Is an Indicator of Poor Prognosis in Acute Myeloid Leukemia
- Author
-
Anna Jauch, Christoph E. Heilig, Markus Schaich, Anthony D. Ho, Axel Benner, Alwin Krämer, Gerhard Ehninger, Johannes W.G. Janssen, Brigitte Mohr, Christina Kunz, Michael Kramer, Tilmann Bochtler, Friedrich Stölzel, and Martin Bornhäuser
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Myeloid ,Adolescent ,Somatic evolution in cancer ,Young Adult ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Multicenter Studies as Topic ,Metaphase ,Randomized Controlled Trials as Topic ,Chromosome Aberrations ,business.industry ,Cytogenetics ,Myeloid leukemia ,Karyotype ,Middle Aged ,Prognosis ,medicine.disease ,Transplantation ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,Karyotyping ,Population study ,Female ,business - Abstract
Purpose In acute myeloid leukemia (AML), studies based on whole-genome sequencing have shown genomic diversity within leukemic clones. The aim of this study was to address clonal heterogeneity in AML based on metaphase cytogenetics. Patients and Methods This analysis included all patients enrolled onto two consecutive, prospective, randomized multicenter trials of the Study Alliance Leukemia. Patients were newly diagnosed with non-M3 AML and were fit for intensive chemotherapy. Results Cytogenetic subclones were detected in 418 (15.8%) of 2,639 patients from the whole study population and in 418 (32.8%) of 1,274 patients with aberrant karyotypes. Among those, 252 karyotypes (60.3%) displayed a defined number of distinct subclones, and 166 (39.7%) were classified as composite karyotypes. Subclone formation was particularly frequent in the cytogenetically adverse group, with subclone formation in 69.0%, 67.1%, and 64.8% of patients with complex aberrant, monosomal, and abnl(17p) karyotypes (P < .001 each). Two-subclone patterns typically followed a mother-daughter evolution, whereas for ≥ three subclones, a branched pattern prevailed. In non–core binding factor AML, subclone formation was associated with inferior event-free and overall survival and was confirmed as an independent predictor of poor prognosis in multivariate analysis. Subgroup analysis showed that subclone formation adds prognostic information particularly in the cytogenetic adverse-risk group. Allogeneic stem-cell transplantation improved the prognosis of patients with subclone karyotypes as shown in landmark analyses. Conclusion Cytogenetic subclones are frequent in AML and permit tracing of clonal evolution and architecture. They bear prognostic significance with clonal heterogeneity as an independent adverse prognostic marker in cytogenetically adverse-risk AML.
- Published
- 2013
50. Marker chromosomes can arise from chromothripsis and predict adverse prognosis in acute myeloid leukemia
- Author
-
Albrecht Stenzinger, Friedrich Stölzel, Tilmann Bochtler, Mutlu Kartal-Kaess, Christina Kunz, Brigitte Mohr, Gerhard Ehninger, Alwin Krämer, Christian Thiede, Christoph E. Heilig, Martin Bornhäuser, Axel Benner, Anna Jauch, Martina Kirchner, Michael Kramer, Katrin Hinderhofer, Martin Granzow, Volker Endris, and Anthony D. Ho
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Immunology ,Abnormal Karyotype ,Kaplan-Meier Estimate ,Biology ,Bioinformatics ,Biochemistry ,Disease-Free Survival ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Metaphase ,In Situ Hybridization, Fluorescence ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,Chromothripsis ,Comparative Genomic Hybridization ,Cytogenetics ,Chromosome ,Myeloid leukemia ,Karyotype ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,030220 oncology & carcinogenesis ,Karyotyping ,Female ,Comparative genomic hybridization - Abstract
Metaphase karyotyping is an established diagnostic standard in acute myeloid leukemia (AML) for risk stratification. One of the cytogenetic findings in AML is structurally highly abnormal marker chromosomes. In this study, we have assessed frequency, cytogenetic characteristics, prognostic impact, and underlying biological origin of marker chromosomes. Given their inherent gross structural chromosomal damage, we speculated that they may arise from chromothripsis, a recently described phenomenon of chromosome fragmentation in a single catastrophic event. In 2 large consecutive prospective, randomized, multicenter, intensive chemotherapy trials (AML96, AML2003) from the Study Alliance Leukemia, marker chromosomes were detectable in 165/1026 (16.1%) of aberrant non-core-binding-factor (CBF) karyotype patients. Adverse-risk karyotypes displayed a higher frequency of marker chromosomes (26.5% in adverse-risk, 40.3% in complex aberrant, and 41.2% in abnormality(17p) karyotypes, P < .0001 each). Marker chromosomes were associated with a poorer prognosis compared with other non-CBF aberrant karyotypes and led to lower remission rates (complete remission + complete remission with incomplete recovery), inferior event-free survival as well as overall survival in both trials. In multivariate analysis, marker chromosomes independently predicted poor prognosis in the AML96 trial ≤60 years. As detected by array comparative genomic hybridization, about one-third of marker chromosomes (18/49) had arisen from chromothripsis, whereas this phenomenon was virtually undetectable in a control group of marker chromosome-negative complex aberrant karyotypes (1/34). The chromothripsis-positive cases were characterized by a particularly high degree of karyotype complexity, TP53 mutations, and dismal prognosis. In conclusion, marker chromosomes are indicative of chromothripsis and associated with poor prognosis per se and not merely by association with other adverse cytogenetic features.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.