Network


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

Hotspot


Dive into the research topics where Hans Erik Johnsen is active.

Publication


Featured researches published by Hans Erik Johnsen.


Lancet Oncology | 2014

International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma

S. Vincent Rajkumar; Meletios A. Dimopoulos; Antonio Palumbo; Joan Bladé; Giampaolo Merlini; Maria Victoria Mateos; Shaji Kumar; Jens Hillengass; Efstathios Kastritis; Paul G. Richardson; Ola Landgren; Bruno Paiva; Angela Dispenzieri; Brendan M. Weiss; Xavier Leleu; Sonja Zweegman; Sagar Lonial; Laura Rosiñol; Elena Zamagni; Sundar Jagannath; Orhan Sezer; Sigurdur Y. Kristinsson; Jo Caers; Saad Z Usmani; Juan José Lahuerta; Hans Erik Johnsen; Meral Beksac; Michele Cavo; Hartmut Goldschmidt; Evangelos Terpos

This International Myeloma Working Group consensus updates the disease definition of multiple myeloma to include validated biomarkers in addition to existing requirements of attributable CRAB features (hypercalcaemia, renal failure, anaemia, and bone lesions). These changes are based on the identification of biomarkers associated with near inevitable development of CRAB features in patients who would otherwise be regarded as having smouldering multiple myeloma. A delay in application of the label of multiple myeloma and postponement of therapy could be detrimental to these patients. In addition to this change, we clarify and update the underlying laboratory and radiographic variables that fulfil the criteria for the presence of myeloma-defining CRAB features, and the histological and monoclonal protein requirements for the disease diagnosis. Finally, we provide specific metrics that new biomarkers should meet for inclusion in the disease definition. The International Myeloma Working Group recommends the implementation of these criteria in routine practice and in future clinical trials, and recommends that future studies analyse any differences in outcome that might occur as a result of the new disease definition.


Journal of Clinical Oncology | 2003

High-Dose Therapy Improves Progression-Free Survival and Survival in Relapsed Follicular Non-Hodgkin's Lymphoma: Results From the Randomized European CUP Trial

Harry C. Schouten; Wendi Qian; Stein Kvaloy; Adolfo Porcellini; Hans Hagberg; Hans Erik Johnsen; Jeanette K. Doorduijn; Matthew R. Sydes; Gunnar Kvalheim

PURPOSE To determine, in a randomized clinical trial, whether high-dose therapy (HDT) followed by autologous stem-cell transplantation is more effective than standard treatment with regard to progression-free survival (PFS) and overall survival (OS) in patients with relapsed follicular non-Hodgkins lymphoma; and to assess the additional value of B-cell purging of the stem-cell graft with regards to PFS and OS. PATIENTS AND METHODS Patients received three cycles of chemotherapy. Responding patients with limited bone marrow infiltration were eligible for random assignment to three further cycles of chemotherapy (C), unpurged HDT (U), or purged HDT (P). RESULTS Between August 1993 and April 1997, 140 patients were registered from 36 centers internationally, and 89 were randomly assigned. Reasons for not randomizing included patient refusal, early progression, or death on induction therapy. With a 69-month median follow-up, the log-rank P value for PFS and OS were.0037 and.079, respectively. For PFS, the hazard ratios (95% CIs) for U versus C, P versus C, and P versus U were 0.33 (0.16 to 0.70), 0.38 (0.19 to 0.79), and 1.02 (0.51 to 2.05), respectively. The hazard ratio (95% CI) for C versus U + P was 0.30 (0.15 to 0.61). Hazard ratios (95% CIs) for OS were 0.43 (0.18 to 1.06), 0.43 (0.18 to 1.02), and 0.72 (0.32 to 1.63). For C versus U + P, the hazard ratio (95% CI) was 0.40 (0.18 to 0.89). Kaplan-Meier estimates (95% CIs) of 2-year PFS for C, U, and P were 26% (8% to 44%), 58% (37% to 79%), and 55% (34% to 75%), respectively. OS at 4 years for C, U, and P are 46% (25% to 67%), 71% (52% to 91%), and 77% (60% to 95%) respectively. CONCLUSION HDT significantly improves PFS and OS. There is no clear evidence of benefit through purging.


Circulation | 2006

Stem Cell Mobilization Induced by Subcutaneous Granulocyte-Colony Stimulating Factor to Improve Cardiac Regeneration After Acute ST-Elevation Myocardial Infarction Result of the Double-Blind, Randomized, Placebo-Controlled Stem Cells in Myocardial Infarction (STEMMI) Trial

Rasmus Sejersten Ripa; Erik Jørgensen; Yongzhong Wang; Jens Jakob Thune; Jens C. Nilsson; Lars Søndergaard; Hans Erik Johnsen; Lars Køber; Peer Grande; Jens Kastrup

Background— Phase 1 clinical trials of granulocyte-colony stimulating factor (G-CSF) treatment after myocardial infarction have indicated that G-CSF treatment is safe and may improve left ventricular function. This randomized, double-blind, placebo-controlled trial aimed to assess the efficacy of subcutaneous G-CSF injections on left ventricular function in patients with ST-elevation myocardial infarction. Methods and Results— Seventy-eight patients (62 men; average age, 56 years) with ST-elevation myocardial infarction were included after successful primary percutaneous coronary stent intervention <12 hours after symptom onset. Patients were randomized to double-blind treatment with G-CSF (10 &mgr;g/kg of body weight) or placebo for 6 days. The primary end point was change in systolic wall thickening from baseline to 6 months determined by cardiac magnetic resonance imaging (MRI). An independent core laboratory analyzed all MRI examinations. Systolic wall thickening improved 17% in the infarct area in the G-CSF group and 17% in the placebo group (P=1.0). Comparable results were found in infarct border and noninfarcted myocardium. Left ventricular ejection fraction improved similarly in the 2 groups measured by both MRI (8.5 versus 8.0; P=0.9) and echocardiography (5.7 versus 3.7; P=0.7). The risk of severe clinical adverse events was not increased by G-CSF. In addition, in-stent late lumen loss and target vessel revascularization rate in the follow-up period were similar in the 2 groups. Conclusions— Bone marrow stem cell mobilization with subcutaneous G-CSF is safe but did not lead to further improvement in ventricular function after acute myocardial infarction compared with the recovery observed in the placebo group.


Nature Reviews Cancer | 2012

Cancer stem cell definitions and terminology: the devil is in the details

Peter Valent; Dominique Bonnet; Ruggero De Maria; Tsvee Lapidot; Mhairi Copland; Junia V. Melo; Christine Chomienne; Fumihiko Ishikawa; Jan Jacob Schuringa; Giorgio Stassi; Brian J. P. Huntly; Harald Herrmann; Jean Soulier; Alexander Roesch; G.J. Schuurhuis; Stefan Wöhrer; Michel Arock; Johannes Zuber; Sabine Cerny-Reiterer; Hans Erik Johnsen; Michael Andreeff; Connie J. Eaves

The cancer stem cell (CSC) concept has important therapeutic implications, but its investigation has been hampered both by a lack of consistency in the terms used for these cells and by how they are defined. Evidence of their heterogeneous origins, frequencies and their genomic, as well as their phenotypic and functional, properties has added to the confusion and has fuelled new ideas and controversies. Participants in The Year 2011 Working Conference on CSCs met to review these issues and to propose a conceptual and practical framework for CSC terminology. More precise reporting of the parameters that are used to identify CSCs and to attribute responses to them is also recommended as key to accelerating an understanding of their biology and developing more effective methods for their eradication in patients.


Haematologica | 2008

Report of the European Myeloma Network on multiparametric flow cytometry in multiple myeloma and related disorders

Andy C. Rawstron; Alberto Orfao; Meral Beksac; Ludmila Bezdickova; Rik A. Brooimans; Horia Bumbea; Klara Dalva; Gwenny M. Fuhler; Jan W. Gratama; Dirk Hose; Lucie Kovarova; Michael Lioznov; Gema Mateo; Ricardo Morilla; Anne K. Mylin; Paola Omedè; Catherine Pellat-Deceunynck; Martin Perez Andres; Maria Teresa Petrucci; Marina Ruggeri; Grzegorz Rymkiewicz; Alexander Schmitz; Martin Schreder; Carine Seynaeve; Martin Spacek; Ruth de Tute; Els Van Valckenborgh; Nicola J. Weston-Bell; Roger G. Owen; Jesús F. San Miguel

In multiple myeloma, the use of multiparametric flow cytometry in many laboratories is currently restricted to clinical research studies and the differential diagnosis of unusual cases. This article report the indications of the European Myeloma Network for flow cytometry in patients with monoclonal gammopathies, and the technical recommendations for the analysis of plasma cells. The European Myeloma Network (EMN) organized two flow cytometry workshops. The first aimed to identify specific indications for flow cytometry in patients with monoclonal gammopathies, and consensus technical approaches through a questionnaire-based review of current practice in participating laboratories. The second aimed to resolve outstanding technical issues and develop a consensus approach to analysis of plasma cells. The primary clinical applications identified were: differential diagnosis of neoplastic plasma cell disorders from reactive plasmacytosis; identifying risk of progression in patients with MGUS and detecting minimal residual disease. A range of technical recommendations were identified, including: 1) CD38, CD138 and CD45 should all be included in at least one tube for plasma cell identification and enumeration. The primary gate should be based on CD38 vs. CD138 expression; 2) after treatment, clonality assessment is only likely to be informative when combined with immunophenotype to detect abnormal cells. Flow cytometry is suitable for demonstrating a stringent complete remission; 3) for detection of abnormal plasma cells, a minimal panel should include CD19 and CD56. A preferred panel would also include CD20, CD117, CD28 and CD27; 4) discrepancies between the percentage of plasma cells detected by flow cytometry and morphology are primarily related to sample quality and it is, therefore, important to determine that marrow elements are present in follow-up samples, particularly normal plasma cells in MRD negative cases.


Lancet Oncology | 2016

International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma

Shaji Kumar; Bruno Paiva; Kenneth C. Anderson; Brian G. M. Durie; Ola Landgren; Philippe Moreau; Nikhil C. Munshi; Sagar Lonial; Joan Bladé; Maria-Victoria Mateos; Meletios A. Dimopoulos; Efstathios Kastritis; Mario Boccadoro; Robert Z. Orlowski; Hartmut Goldschmidt; Andrew Spencer; Jian Hou; Wee Joo Chng; Saad Z Usmani; Elena Zamagni; Kazuyuki Shimizu; Sundar Jagannath; Hans Erik Johnsen; Evangelos Terpos; Anthony Reiman; Robert A. Kyle; Pieter Sonneveld; Paul G. Richardson; Philip L. McCarthy; Heinz Ludwig

Treatment of multiple myeloma has substantially changed over the past decade with the introduction of several classes of new effective drugs that have greatly improved the rates and depth of response. Response criteria in multiple myeloma were developed to use serum and urine assessment of monoclonal proteins and bone marrow assessment (which is relatively insensitive). Given the high rates of complete response seen in patients with multiple myeloma with new treatment approaches, new response categories need to be defined that can identify responses that are deeper than those conventionally defined as complete response. Recent attempts have focused on the identification of residual tumour cells in the bone marrow using flow cytometry or gene sequencing. Furthermore, sensitive imaging techniques can be used to detect the presence of residual disease outside of the bone marrow. Combining these new methods, the International Myeloma Working Group has defined new response categories of minimal residual disease negativity, with or without imaging-based absence of extramedullary disease, to allow uniform reporting within and outside clinical trials. In this Review, we clarify several aspects of disease response assessment, along with endpoints for clinical trials, and highlight future directions for disease response assessments.


Blood | 2011

International myeloma working group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation

Michele Cavo; S. Vincent Rajkumar; Antonio Palumbo; Philippe Moreau; Robert Z. Orlowski; Joan Bladé; Orhan Sezer; Heinz Ludwig; Meletios A. Dimopoulos; Michel Attal; Pieter Sonneveld; Mario Boccadoro; Kenneth C. Anderson; Paul G. Richardson; William Bensinger; Hans Erik Johnsen; Nicolaus Kroeger; Gösta Gahrton; P. Leif Bergsagel; David H. Vesole; Hermann Einsele; Sundar Jagannath; Ruben Niesvizky; Brian G. M. Durie; Jesús F. San Miguel; Sagar Lonial

The role of high-dose therapy followed by autologous stem cell transplantation (ASCT) in the treatment of multiple myeloma (MM) continues to evolve in the novel agent era. The choice of induction therapy has moved from conventional chemotherapy to newer regimens incorporating the immunomodulatory derivatives thalidomide or lenalidomide and the proteasome inhibitor bortezomib. These drugs combine well with traditional therapies and with one another to form various doublet, triplet, and quadruplet regimens. Up-front use of these induction treatments, in particular 3-drug combinations, has affected unprecedented rates of complete response that rival those previously seen with conventional chemotherapy and subsequent ASCT. Autotransplantation applied after novel-agent-based induction regimens provides further improvement in the depth of response, a gain that translates into extended progression-free survival and, potentially, overall survival. High activity shown by immunomodulatory derivatives and bortezomib before ASCT has recently led to their use as consolidation and maintenance therapies after autotransplantation. Novel agents and ASCT are complementary treatment strategies for MM. This article reviews the current literature and provides important perspectives and guidance on the major issues surrounding the optimal current management of younger, transplantation-eligible MM patients.


Blood | 2011

Personalized therapy in multiple myeloma according to patient age and vulnerability: a report of the European Myeloma Network (EMN)

A. Palumbo; Sara Bringhen; Heinz Ludwig; Meletios A. Dimopoulos; Joan Bladé; M.V. Mateos; Laura Rosiñol; Mario Boccadoro; Michele Cavo; Henk M. Lokhorst; Sonja Zweegman; Evangelos Terpos; Faith E. Davies; Christoph Driessen; Peter Gimsing; Martin Gramatzki; Roman Hájek; Hans Erik Johnsen; F. Leal da Costa; Orhan Sezer; Andrew Spencer; Meral Beksac; Gareth J. Morgan; Hermann Einsele; J. F. San Miguel; Pieter Sonneveld

Most patients with newly diagnosed multiple myeloma (MM) are aged > 65 years with 30% aged > 75 years. Many elderly patients are also vulnerable because of comorbidities that complicate the management of MM. The prevalence of MM is expected to rise over time because of an aging population. Most elderly patients with MM are ineligible for autologous transplantation, and the standard treatment has, until recently, been melphalan plus prednisone. The introduction of novel agents, such as thalidomide, bortezomib, and lenalidomide, has improved outcomes; however, elderly patients with MM are more susceptible to side effects and are often unable to tolerate full drug doses. For these patients, lower-dose-intensity regimens improve the safety profile and thus optimize treatment outcome. Further research into the best treatment strategies for vulnerable elderly patients is urgently needed. Appropriate screening for vulnerability and an assessment of cardiac, pulmonary, renal, hepatic, and neurologic functions, as well as age > 75 years, at the start of therapy allows treatment strategies to be individualized and drug doses to be tailored to improve tolerability and optimize efficacy. Similarly, occurrence of serious nonhematologic adverse events during treatment should be carefully taken into account to adjust doses and optimize outcomes.


Cytometry | 1998

Flow cytometric enumeration of CD34+ hematopoietic stem and progenitor cells.

Jan W. Gratama; Alberto Orfao; David Barnett; Bruno Brando; Andreas Huber; George Janossy; Hans Erik Johnsen; Michael Keeney; Gerald E. Marti; Frank Preijers; Gregor Rothe; Stefan Serke; D. Robert Sutherland; C. Ellen Van der Schoot; Gerd Schmitz; Stefano Papa

The need for a rapid and reliable marker for the engraftment potential of hematopoietic stem and progenitor cell (HPC) transplants has led to the development of flow cytometric assays to quantitate such cells on the basis of their expression of CD34. The variability associated with enumeration of low-frequency cells (i.e., as low as 0.1% or 5 cells/microl) is exceedingly large, but recent developments have improved the accuracy and precision of the assay. Here, we review and compare the major techniques. Based on the current state of the art, we recommend 1) bright fluorochrome conjugates of class II or III monoclonal antibodies (mAbs) that detect all glycoforms of CD34, 2) use of a vital nucleic acid dye to exclude platelets, unlysed red cells, and debris or use of 7-amino actinomycin D to exclude dead cells during data acquisition, 3) counterstaining with CD45 mAb to be included in the definition of HPC, 4) during list mode data analysis, Boolean gating to resolve the CD34+ HPCs from irrelevant cell populations on the basis of the low levels of CD45 expression and low sideward light-scatter signals of HPCs, 5) inclusion of CD34dim and CD34bright populations in the CD34+ cell count, 6) omission of the negative control staining, and 7) for apheresis products, enumeration of at least 100 CD34+ cells to ensure a 10% precision. Unresolved technical questions are 1) the replacement of conventional dual-platform by single-platform assay formats, i.e., derivation of absolute CD34+ cell counts from a single flow cytometric assessment instead of from combined flow cytometer (percent CD34+) and hematology analyzer (absolute leukocyte count) data, 2) the cross-calibration of the available single-platform assays, and 3) the optimal method for sample preparation. An important clinical question to be addressed is the definition of the precise phenotypes and required numbers of HPCs responsible for short- and long-term recovery to optimize HPC transplant strategies.


Haematologica | 2010

Circulating human B and plasma cells. Age-associated changes in counts and detailed characterization of circulating normal CD138− and CD138+ plasma cells

Anouk Caraux; Bernard Klein; Bruno Paiva; Caroline Bret; Alexander Schmitz; Gwenny M. Fuhler; Nico A. Bos; Hans Erik Johnsen; Alberto Orfao; Martin Perez-Andres

Generation of B and plasma cells involves several organs with a necessary cell trafficking between them. A detailed phenotypic characterization of four circulating B-cell subsets (immature-, naïve-, memory- B-lymphocytes and plasma cells) of 106 healthy adults was realized by multiparametric flow cytometry. We show that CD10, CD27 and CD38 is the minimal combination of subsetting markers allowing unequivocal identification of immature (CD10+CD27−CD38+, 6±6 cells/μL), naïve (CD10−CD27−CD38−, 125±90 cells/μL), memory B lymphocytes (CD10−CD27+CD38−, 58±42 cells/μL), and plasma cells (CD10−CD27++CD38++, 2.1±2.1 cells/μL) within circulating CD19+ cells. From these four subsets, only memory B lymphocytes and plasma cells decreased with age, both in relative and absolute counts. Circulating plasma cells split into CD138− (57±12%) and CD138+ (43±12%) cells, the latter displaying a more mature phenotypic profile: absence of surface immunoglobulin, lower CD45 positivity and higher amounts of cytoplasmic immunoglobulin, CD38 and CD27. Unlike B lymphocytes, both populations of plasma cells are KI-67+ and show weak CXCR4 expression.

Collaboration


Dive into the Hans Erik Johnsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge