Network


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

Hotspot


Dive into the research topics where Peter Gimsing is active.

Publication


Featured researches published by Peter Gimsing.


The New England Journal of Medicine | 2015

Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma.

Abstr Act; Henk M. Lokhorst; Torben Plesner; Hareth Nahi; Peter Gimsing; Markus Hansson; Ulrik V. Lassen; Jakub Krejcik; A Palumbo; Tahamtan Ahmadi; I. Khan; Jianping Wang; N. Losic; Steen Lisby; Linda Basse; Nikolai C. Brun

BACKGROUND Multiple myeloma cells uniformly overexpress CD38. We studied daratumumab, a CD38-targeting, human IgG1κ monoclonal antibody, in a phase 1-2 trial involving patients with relapsed myeloma or relapsed myeloma that was refractory to two or more prior lines of therapy. METHODS In part 1, the dose-escalation phase, we administered daratumumab at doses of 0.005 to 24 mg per kilogram of body weight. In part 2, the dose-expansion phase, 30 patients received 8 mg per kilogram of daratumumab and 42 received 16 mg per kilogram, administered once weekly (8 doses), twice monthly (8 doses), and monthly for up to 24 months. End points included safety, efficacy, and pharmacokinetics. RESULTS No maximum tolerated dose was identified in part 1. In part 2, the median time since diagnosis was 5.7 years. Patients had received a median of four prior treatments; 79% of the patients had disease that was refractory to the last therapy received (64% had disease refractory to proteasome inhibitors and immunomodulatory drugs and 64% had disease refractory to bortezomib and lenalidomide), and 76% had received autologous stem-cell transplants. Infusion-related reactions in part 2 were mild (71% of patients had an event of any grade, and 1% had an event of grade 3), with no dose-dependent adverse events. The most common adverse events of grade 3 or 4 (in ≥ 5% of patients) were pneumonia and thrombocytopenia. The overall response rate was 36% in the cohort that received 16 mg per kilogram (15 patients had a partial response or better, including 2 with a complete response and 2 with a very good partial response) and 10% in the cohort that received 8 mg per kilogram (3 had a partial response). In the cohort that received 16 mg per kilogram, the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 8.1), and 65% (95% CI, 28 to 86) of the patients who had a response did not have progression at 12 months. CONCLUSIONS Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma. (Funded by Janssen Research and Development and Genmab; ClinicalTrials.gov number, NCT00574288.).


The New England Journal of Medicine | 2016

Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma

Philippe Moreau; Tamas Masszi; Norbert Grzasko; Nizar J. Bahlis; Markus Hansson; Luděk Pour; Irwindeep Sandhu; Peter Ganly; Bartrum Baker; Sharon Jackson; Anne-Marie Stoppa; David R Simpson; Peter Gimsing; A Palumbo; L. Garderet; Michele Cavo; Shaji Kumar; Cyrille Touzeau; Francis Buadi; Jacob P. Laubach; Deborah Berg; Jianchang Lin; A. Di Bacco; Ai-Min Hui; H van de Velde; Paul G. Richardson; Eric Kupperman; Allison Berger; Larry Dick; Mark Williamson

BACKGROUND Ixazomib is an oral proteasome inhibitor that is currently being studied for the treatment of multiple myeloma. METHODS In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned 722 patients who had relapsed, refractory, or relapsed and refractory multiple myeloma to receive ixazomib plus lenalidomide-dexamethasone (ixazomib group) or placebo plus lenalidomide-dexamethasone (placebo group). The primary end point was progression-free survival. RESULTS Progression-free survival was significantly longer in the ixazomib group than in the placebo group at a median follow-up of 14.7 months (median progression-free survival, 20.6 months vs. 14.7 months; hazard ratio for disease progression or death in the ixazomib group, 0.74; P=0.01); a benefit with respect to progression-free survival was observed with the ixazomib regimen, as compared with the placebo regimen, in all prespecified patient subgroups, including in patients with high-risk cytogenetic abnormalities. The overall rates of response were 78% in the ixazomib group and 72% in the placebo group, and the corresponding rates of complete response plus very good partial response were 48% and 39%. The median time to response was 1.1 months in the ixazomib group and 1.9 months in the placebo group, and the corresponding median duration of response was 20.5 months and 15.0 months. At a median follow-up of approximately 23 months, the median overall survival has not been reached in either study group, and follow-up is ongoing. The rates of serious adverse events were similar in the two study groups (47% in the ixazomib group and 49% in the placebo group), as were the rates of death during the study period (4% and 6%, respectively); adverse events of at least grade 3 severity occurred in 74% and 69% of the patients, respectively. Thrombocytopenia of grade 3 and grade 4 severity occurred more frequently in the ixazomib group (12% and 7% of the patients, respectively) than in the placebo group (5% and 4% of the patients, respectively). Rash occurred more frequently in the ixazomib group than in the placebo group (36% vs. 23% of the patients), as did gastrointestinal adverse events, which were predominantly low grade. The incidence of peripheral neuropathy was 27% in the ixazomib group and 22% in the placebo group (grade 3 events occurred in 2% of the patients in each study group). Patient-reported quality of life was similar in the two study groups. CONCLUSIONS The addition of ixazomib to a regimen of lenalidomide and dexamethasone was associated with significantly longer progression-free survival; the additional toxic effects with this all-oral regimen were limited. (Funded by Millennium Pharmaceuticals; TOURMALINE-MM1 ClinicalTrials.gov number, NCT01564537.).


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.


Blood | 2011

Thalidomide for previously untreated elderly patients with multiple myeloma: meta-analysis of 1685 individual patient data from 6 randomized clinical trials

Peter Fayers; Antonio Palumbo; Cyrille Hulin; Anders Waage; Pierre W. Wijermans; Meral Beksac; Sara Bringhen; Jean Yves Mary; Peter Gimsing; Fabian Termorshuizen; Rauf Haznedar; Tommaso Caravita; Philippe Moreau; Ingemar Turesson; Pellegrino Musto; Lotfi Benboubker; Martijn R. Schaafsma; Pieter Sonneveld; Thierry Facon

The role of thalidomide for previously untreated elderly patients with multiple myeloma remains unclear. Six randomized controlled trials, launched in or after 2000, compared melphalan and prednisone alone (MP) and with thalidomide (MPT). The effect on overall survival (OS) varied across trials. We carried out a meta-analysis of the 1685 individual patients in these trials. The primary endpoint was OS, and progression-free survival (PFS) and 1-year response rates were secondary endpoints. There was a highly significant benefit to OS from adding thalidomide to MP (hazard ratio = 0.83; 95% confidence interval 0.73-0.94, P = .004), representing increased median OS time of 6.6 months, from 32.7 months (MP) to 39.3 months (MPT). The thalidomide regimen was also associated with superior PFS (hazard ratio = 0.68, 95% confidence interval 0.61-0.76, P < .0001) and better 1-year response rates (partial response or better was 59% on MPT and 37% on MP). Although the trials differed in terms of patient baseline characteristics and thalidomide regimens, there was no evidence that treatment affected OS differently according to levels of the prognostic factors. We conclude that thalidomide added to MP improves OS and PFS in previously untreated elderly patients with multiple myeloma, extending the median survival time by on average 20%.


Blood | 2010

Melphalan and prednisone plus thalidomide or placebo in elderly patients with multiple myeloma

Peter Gimsing; Peter Fayers; Niels Abildgaard; Lucia Ahlberg; Bo Björkstrand; Kristina Carlson; Inger Marie S. Dahl; Karin Forsberg; Nina Gulbrandsen; Einar Haukås; Øyvind Hjertner; Martin Hjorth; Torbjörn Karlsson; Lene Meldgaard Knudsen; Johan Lanng Nielsen; Olle Linder; Ulf-Henrik Mellqvist; Ingerid Nesthus; Jürgen Rolke; Maria Strandberg; Jon Hjalmar Sørbø; Finn Wisløff; Gunnar Juliusson; Ingemar Turesson

In this double-blind, placebo-controlled study, 363 patients with untreated multiple myeloma were randomized to receive either melphalan-prednisone and thalidomide (MPT) or melphalan-prednisone and placebo (MP). The dose of melphalan was 0.25 mg/kg and prednisone was 100 mg given daily for 4 days every 6 weeks until plateau phase. The dose of thalidomide/placebo was escalated to 400 mg daily until plateau phase and thereafter reduced to 200 mg daily until progression. A total of 357 patients were analyzed. Partial response was 34% and 33%, and very good partial response or better was 23% and 7% in the MPT and MP arms, respectively (P < .001). There was no significant difference in progression-free or overall survival, with median survival being 29 months in the MPT arm and 32 months in the MP arm. Most quality of life outcomes improved equally in both arms, apart from constipation, which was markedly increased in the MPT arm. Constipation, neuropathy, nonneuropathy neurologic toxicity, and skin reactions were significantly more frequent in the MPT arm. The number of thromboembolic events was equal in the 2 treatment arms. In conclusion, MPT had a significant antimyeloma effect, but this did not translate into improved survival. This trial was registered at www.clinicaltrials.gov as #NCT00218855.


European Journal of Haematology | 2008

A phase I clinical trial of the histone deacetylase inhibitor belinostat in patients with advanced hematological neoplasia

Peter Gimsing; Mads Hansen; Lene Meldgaard Knudsen; P. Knoblauch; Ib Jarle Christensen; Chean Eng Ooi; Peter Buhl-Jensen

Purpose:  To determine the safety, dose‐limiting toxicity and maximum tolerated dose (MTD) of the novel hydroxamate histone deacetylase inhibitor belinostat (PXD101) in patients with advanced hematological neoplasms.


Scandinavian Journal of Immunology | 2010

Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR⁻/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma.

Marie Klinge Brimnes; Annette Juul Vangsted; Lene Meldgaard Knudsen; Peter Gimsing; Anne O. Gang; Hans Erik Johnsen; Inge Marie Svane

Patients with multiple myeloma (MM) suffer from a general impaired immunity comprising deficiencies in humoral responses, T‐cell responses as well as dendritic cell (DC) function. Thus, to achieve control of tumour growth through immune therapy constitutes a challenge. Careful evaluation of the immune status in patients with MM seems crucial prior to active immune therapy. We evaluated the proportion of both, DC, Treg cells and myeloid‐derived suppressor cells (MDSC) in peripheral blood from patients with MM at diagnosis and in remission as well as patients with monoclonal gammopathy of undetermined significance (MGUS). We found that the proportion of both myeloid (m) DC and plasmacytoid (p) DC in patients at diagnosis was lowered compared to control donors, while only the proportion of pDC in patients in remission and with MGUS was significantly lower than in controls. The proportion of CD4+FOXP3+ Treg cells was increased in patients at diagnosis and not in patients in remission or with MGUS. Also, Treg cells from patients with MM were functionally intact as they were able to inhibit proliferation of both CD4 and CD8 T cells. Finally, we observed an increase in the proportion of CD14+HLA‐DR−/low MDSC in patients with MM at diagnosis, illustrating that this cell fraction is also distorted in patients with MM. Taken together, our results illustrate that, both mDC, pDC, Treg cells and MDSC are affected in patients with MM underlining the fact that the immune system is dysregulated as a consequence of the disease.


British Journal of Haematology | 1998

Failure of oral pamidronate to reduce skeletal morbidity in multiple myeloma: a double‐blind placebo‐controlled trial

Hans Brincker; Jan Westin; Niels Abildgaard; Peter Gimsing; Ingemar Turesson; Michael Hedenus; John M. Ford; Albert Kandra

In order to study whether oral bisphosphonate therapy might prevent or reduce skeletal‐related morbidity in patients with newly diagnosed multiple myeloma who required chemotherapy, 300 patients were included in a randomized multi‐centre trial. Patients were given oral pamidronate at a dose of 300 mg daily, or placebo, in addition to conventional intermittent melphalan/prednisolone (and in some cases alpha‐interferon) treatment. With a median treatment duration of about 550 d, no statisticallly significant reduction in skeletal‐related morbidity (defined as bone fracture, related surgery, vertebral collapse, or increase in number and/or size of bone lesions) could be demonstrated. Pamidronate treatment also did not have any influence on patient survival or on the frequency of hypercalcaemia. However, in patients treated with pamidronate there were fewer episodes of severe pain (P = 0.02) and a decreased reduction of body height of 1.5 cm (P = 0.02). The overall negative result of the study is attributed to the very low absorption of orally administered bisphosphonates in general.


European Journal of Haematology | 2005

Autologous stem cell transplantation in multiple myeloma: outcome in patients with renal failure.

Lene Meldgaard Knudsen; Bendt Nielsen; Peter Gimsing; Christian H. Geisler

Abstract:  The impact of renal failure on prognosis of multiple myeloma patients treated with high‐dose chemotherapy and stem cell support is incompletely studied. A total of 137 patients received high‐dose chemotherapy with autologous transplantation at our centre. The patient population was divided into three groups based on their estimated creatinine clearance (Ccr); renal failure defined as Ccr < 60 mL/min: Group A: normal renal function both at diagnosis and at transplant (n = 78), Group B: renal failure at diagnosis but normal renal function at transplant (n = 30), Group C: renal failure both at diagnosis and at transplant (n = 29). There were no differences in the number of stem cells harvested, time to engraftment or response to transplantation between the groups. Ten of the patients in Group C had a normalisation of renal function post‐transplant. Significantly longer hospitalisation, increased use of blood products and increased number of infections were seen in Group C compared to Groups A and B. The transplant‐related mortality was 17% in Group C compared to 0% and 1% in Groups B and A respectively. Eight patients were on dialysis during transplant and four of these died within the first 100 d post‐transplant. Disease response was similar in the three groups. Overall survival was significantly longer in Group A than in Groups B and C. High‐dose chemotherapy with autologous transplantation is feasible in MM with renal failure. Whereas patients with moderate renal insufficiency seem to benefit from this treatment, patients in need for dialysis at the time of transplant must be carefully evaluated before proceeding to high‐dose chemotherapy.


Blood | 2013

Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial

Ulf-Henrik Mellqvist; Peter Gimsing; Øyvind Hjertner; Stig Lenhoff; Edward Laane; Kari Remes; Hlif Steingrimsdottir; Niels Abildgaard; Lucia Ahlberg; Cecilie Blimark; Inger Marie S. Dahl; Karin Forsberg; Tobias Gedde-Dahl; Henrik Gregersen; Astrid Gruber; Nina Guldbrandsen; Einar Haukås; Kristina Carlson; Ann Kristin Kvam; Hareth Nahi; Roald Lindås; Niels Frost Andersen; Ingemar Turesson; Anders Waage; Jan Westin

The Nordic Myeloma Study Group conducted an open randomized trial to compare bortezomib as consolidation therapy given after high-dose therapy and autologous stem cell transplantation (ASCT) with no consolidation in bortezomib-naive patients with newly diagnosed multiple myeloma. Overall, 370 patients were centrally randomly assigned 3 months after ASCT to receive 20 doses of bortezomib given during 21 weeks or no consolidation. The hypothesis was that consolidation therapy would prolong progression-free survival (PFS). The PFS after randomization was 27 months for the bortezomib group compared with 20 months for the control group (P = .05). Fifty-one of 90 patients in the treatment group compared with 32 of 90 controls improved their response after randomization (P = .007). No difference in overall survival was seen. Fatigue was reported more commonly by the bortezomib-treated patients in self-reported quality-of-life (QOL) questionnaires, whereas no other major differences in QOL were recorded between the groups. Consolidation therapy seemed to be beneficial for patients not achieving at least a very good partial response (VGPR) but not for patients in the ≥ VGPR category at randomization. Consolidation with bortezomib after ASCT in bortezomib-naive patients improves PFS without interfering with QOL. This trial was registered at www.clinicaltrials.gov as #NCT00417911.

Collaboration


Dive into the Peter Gimsing's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Niels Abildgaard

Aarhus University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Torben Plesner

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders Waage

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Erik Hippe

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge