Network


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

Hotspot


Dive into the research topics where Anders Waage is active.

Publication


Featured researches published by Anders Waage.


The Lancet | 1987

ASSOCIATION BETWEEN TUMOUR NECROSIS FACTOR IN SERUM AND FATAL OUTCOME IN PATIENTS WITH MENINGOCOCCAL DISEASE

Anders Waage; Alfred Halstensen; Terje Espevik

Serum samples taken on admission from 79 patients with meningococcal meningitis, septicaemia, or both, were examined in a highly sensitive bioassay for tumour necrosis factor (TNF). TNF was detected in samples from 10 of 11 patients who died but from only 8 of 68 survivors. All 5 patients with serum TNF levels over 440 units/ml (corresponding to 0.1 ng/ml recombinant TNF) died.


Leukemia | 2008

Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma

A. Palumbo; S V Rajkumar; M. A. Dimopoulos; Paul G. Richardson; J. F. San Miguel; Bart Barlogie; Jean Luc Harousseau; Jeffrey A. Zonder; Michele Cavo; Maurizio Zangari; Michel Attal; Andrew R. Belch; S. Knop; Douglas E. Joshua; Orhan Sezer; H. Ludwig; David H. Vesole; J. Bladé; Robert A. Kyle; Jan Westin; Donna M. Weber; Sara Bringhen; Ruben Niesvizky; Anders Waage; M. von Lilienfeld-Toal; Sagar Lonial; Gareth J. Morgan; Robert Z. Orlowski; Kazuyuki Shimizu; Kenneth C. Anderson

The incidence of venous thromboembolism (VTE) is more than 1‰ annually in the general population and increases further in cancer patients. The risk of VTE is higher in multiple myeloma (MM) patients who receive thalidomide or lenalidomide, especially in combination with dexamethasone or chemotherapy. Various VTE prophylaxis strategies, such as low-molecular-weight heparin (LMWH), warfarin or aspirin, have been investigated in small, uncontrolled clinical studies. This manuscript summarizes the available evidence and recommends a prophylaxis strategy according to a risk-assessment model. Individual risk factors for thrombosis associated with thalidomide/lenalidomide-based therapy include age, history of VTE, central venous catheter, comorbidities (infections, diabetes, cardiac disease), immobilization, surgery and inherited thrombophilia. Myeloma-related risk factors include diagnosis and hyperviscosity. VTE is very high in patients who receive high-dose dexamethasone, doxorubicin or multiagent chemotherapy in combination with thalidomide or lenalidomide, but not with bortezomib. The panel recommends aspirin for patients with ⩽1 risk factor for VTE. LMWH (equivalent to enoxaparin 40 mg per day) is recommended for those with two or more individual/myeloma-related risk factors. LMWH is also recommended for all patients receiving concurrent high-dose dexamethasone or doxorubicin. Full-dose warfarin targeting a therapeutic INR of 2–3 is an alternative to LMWH, although there are limited data in the literature with this strategy. In the absence of clear data from randomized studies as a foundation for recommendations, many of the following proposed strategies are the results of common sense or derive from the extrapolation of data from many studies not specifically designed to answer these questions. Further investigation is needed to define the best VTE prophylaxis.


Nature Communications | 2015

Variants in ELL2 influencing immunoglobulin levels associate with multiple myeloma

Bhairavi Swaminathan; Guðmar Thorleifsson; Magnus Jöud; Mina Ali; Ellinor Johnsson; Ram Ajore; Patrick Sulem; Britt-Marie Halvarsson; Guðmundur Eyjolfsson; Vilhelmína Haraldsdóttir; Christina M. Hultman; Erik Ingelsson; Sigurður Yngvi Kristinsson; Anna K. Kähler; Stig Lenhoff; Gisli Masson; Ulf-Henrik Mellqvist; Robert Månsson; Sven Nelander; Isleifur Olafsson; Olof Sigurðardottir; Hlif Steingrimsdottir; Annette Juul Vangsted; Ulla Vogel; Anders Waage; Hareth Nahi; Daniel F. Gudbjartsson; Thorunn Rafnar; Ingemar Turesson; Urban Gullberg

Multiple myeloma (MM) is characterized by an uninhibited, clonal growth of plasma cells. While first-degree relatives of patients with MM show an increased risk of MM, the genetic basis of inherited MM susceptibility is incompletely understood. Here we report a genome-wide association study in the Nordic region identifying a novel MM risk locus at ELL2 (rs56219066T; odds ratio (OR)=1.25; P=9.6 × 10−10). This gene encodes a stoichiometrically limiting component of the super-elongation complex that drives secretory-specific immunoglobulin mRNA production and transcriptional regulation in plasma cells. We find that the MM risk allele harbours a Thr298Ala missense variant in an ELL2 domain required for transcription elongation. Consistent with a hypomorphic effect, we find that the MM risk allele also associates with reduced levels of immunoglobulin A (IgA) and G (IgG) in healthy subjects (P=8.6 × 10−9 and P=6.4 × 10−3, respectively) and, potentially, with an increased risk of bacterial meningitis (OR=1.30; P=0.0024).


American Journal of Obstetrics and Gynecology | 1993

Tumor necrosis factor, interleukin-1, and interleukin-6 in normal human pregnancy

Sissel Linda Opsjøn; Neville C. Wathen; Solveig Tingulstad; Gro Wiedswang; Anders Sundan; Anders Waage; Rigmor Austgulen

OBJECTIVE Our purpose was to investigate the cytokines, tumor necrosis factor, interleukin-1, and interleukin-6 in normal human pregnancy and labor. STUDY DESIGN Bioassays were used to measure these factors in extraembryonic coelomic fluid, amniotic fluid, placenta, and maternal and cord serum. RESULTS Little or no tumor necrosis factor, interleukin-1, or interleukin-6 was found in coelomic fluid or amniotic fluid in the first trimester. Interleukin-6 appeared in second-trimester amniotic fluid. At term tumor necrosis factor was present (median 17 pg/ml) and increased with the onset of labor (median 58 pg/ml), as did interleukin-1 (median 188 to 680 pg/ml) and interleukin-6 (median 399 to 4800 pg/ml). Maternal serum interleukin-6 increased during pregnancy with a further increment with the onset of labor. Cord interleukin-6 also increased with labor but at a lower level. CONCLUSION The cytokines tumor necrosis factor, interleukin-1, and interleukin-6 may play a role in the onset of normal labor.


Blood | 2008

Thalidomide for treatment of multiple myeloma: 10 years later

Antonio Palumbo; Thierry Facon; Pieter Sonneveld; Joan Bladé; Massimo Offidani; Philippe Moreau; Anders Waage; Andrew Spencer; Heinz Ludwig; Mario Boccadoro; Jean-Luc Harousseau

Thalidomide, bortezomib, and lenalidomide have recently changed the treatment paradigm of myeloma. In young, newly diagnosed patients, the combination of thalidomide and dexamethasone has been widely used as induction treatment before autologous stem cell transplantation (ASCT). In 2 randomized studies, consolidation or maintenance with low-dose thalidomide has extended both progression-free and overall survival in patients who underwent ASCT at diagnosis. In elderly, newly diagnosed patients, 3 independent randomized studies have reported that the oral combination of melphalan and prednisone plus thalidomide (MPT) is better than the standard melphalan and prednisone (MP). These studies have shown better progression-free survival, and 2 have shown improved overall survival for patients assigned to MPT. In refractory-relapsed disease, combinations including thalidomide with dexamethasone, melphalan, doxorubicin, or cyclophosphamide have been extensively investigated. The risks of side effects are greater when thalidomide is used in combination with other drugs. Thromboembolism and peripheral neuropathy are the major concern. The introduction of anticoagulant prophylaxis has reduced the rate of thromboembolism to less than 10%. Immediate thalidomide dose reduction or discontinuation when paresthesia is complicated by pain or motor deficit has decreased the severity of neuropathy. Future studies will define the most effective or the best sequence of combinations which could improve life expectancy.


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%.


Annals of Oncology | 2009

The use of bisphosphonates in multiple myeloma: recommendations of an expert panel on behalf of the European Myeloma Network

Evangelos Terpos; Orhan Sezer; Peter I. Croucher; Ramón García-Sanz; Mario Boccadoro; J. F. San Miguel; J. Ashcroft; J. Bladé; Michele Cavo; Michel Delforge; M. A. Dimopoulos; Thierry Facon; M Macro; Anders Waage; Pieter Sonneveld

BACKGROUND Bisphosphonates (BPs) prevent, reduce, and delay multiple myeloma (MM)-related skeletal complications. Intravenous pamidronate and zoledronic acid, and oral clodronate are used for the management of MM bone disease. The purpose of this paper is to review the current evidence for the use of BPs in MM and provide European Union-specific recommendations to support the clinical practice of treating myeloma bone disease. DESIGN AND METHODS An interdisciplinary, expert panel of specialists on MM and myeloma-related bone disease convened for a face-to-face meeting to review and assess the evidence and develop the recommendations. The panel reviewed and graded the evidence available from randomized clinical trials, clinical practice guidelines, and the body of published literature. Where published data were weak or unavailable, the panel used their own clinical experience to put forward recommendations based solely on their expert opinions. RESULTS The panel recommends the use of BPs in MM patients suffering from lytic bone disease or severe osteoporosis. Intravenous administration may be preferable; however, oral administration can be considered for patients unable to make hospital visits. Dosing should follow approved indications with adjustments if necessary. In general, BPs are well tolerated, but preventive steps should be taken to avoid renal impairment and osteonecrosis of the jaw (ONJ). The panel agrees that BPs should be given for 2 years, but this may be extended if there is evidence of active myeloma bone disease. Initial therapy of ONJ should include discontinuation of BPs until healing occurs. BPs should be restarted if there is disease progression. CONCLUSIONS BPs are an essential component of MM therapy for minimizing skeletal morbidity. Recent retrospective data indicate that a modified dosing regimen and preventive measures can greatly reduce the incidence of ONJ.


Clinical Immunology and Immunopathology | 1987

Production and clearance of tumor necrosis factor in rats exposed to endotoxin and dexamethasone

Anders Waage

The production and clearance of tumor necrosis factor (TNF) in relation to endotoxinemia was studied by the injection of lipopolysaccharide (LPS) in rats. TNF was released into the circulation as a burst when the serum concentration of LPS was rapidly or gradually increased. The maximum concentration of TNF in serum was attained 60 to 90 min after the injection of LPS. TNF was eliminated from the serum according to a first-order kinetics; the half-life was calculated to be 27 +/- 7 min. No additional release of TNF could be evoked by a persistent high level of LPS. When two LPS injections were given within 3 days, the peak concentration of TNF detected after the second injection was 15% of the concentration detected after the first injection. The results indicate that if TNF is a mediator of septic shock, its role is restricted to the initial phase after the appearance of endotoxin in the circulation. Treatment of the rats with dexamethasone (0.5-2.0 micrograms/g) reduced the LPS-induced peak concentrations of TNF in serum by 70-90%. Maximal suppression of the TNF release was observed when dexamethasone was given 5 hr or more prior to LPS, but was gradually lost at shorter intervals.


Blood | 2012

IMWG consensus on maintenance therapy in multiple myeloma

Heinz Ludwig; Brian G. M. Durie; Philip L. McCarthy; Antonio Palumbo; Jesús F. San Miguel; Bart Barlogie; Gareth J. Morgan; Pieter Sonneveld; Andrew Spencer; Kenneth C. Andersen; Thierry Facon; A. Keith Stewart; Hermann Einsele; Maria-Victoria Mateos; Pierre W. Wijermans; Anders Waage; Meral Beksac; Paul G. Richardson; Cyrille Hulin; Ruben Niesvizky; Henk M. Lokhorst; Ola Landgren; P. Leif Bergsagel; Robert Z. Orlowski; Axel Hinke; Michele Cavo; Michel Attal

Maintaining results of successful induction therapy is an important goal in multiple myeloma. Here, members of the International Myeloma Working Group review the relevant data. Thalidomide maintenance therapy after autologous stem cell transplantation improved the quality of response and increased progression-free survival (PFS) significantly in all 6 studies and overall survival (OS) in 3 of them. In elderly patients, 2 trials showed a significant prolongation of PFS, but no improvement in OS. A meta-analysis revealed a significant risk reduction for PFS/event-free survival and death. The role of thalidomide maintenance after melphalan, prednisone, and thalidomide is not well established. Two trials with lenalidomide maintenance treatment after autologous stem cell transplantation and one study after conventional melphalan, prednisone, and lenalidomide induction therapy showed a significant risk reduction for PFS and an increase in OS in one of the transplant trials. Maintenance therapy with single-agent bortezomib or in combination with thalidomide or prednisone has been studied. One trial revealed a significantly increased OS with a bortezomib-based induction and bortezomib maintenance therapy compared with conventional induction and thalidomide maintenance treatment. Maintenance treatment can be associated with significant side effects, and none of the drugs evaluated is approved for maintenance therapy. Treatment decisions for individual patients must balance potential benefits and risks carefully, as a widely agreed-on standard is not established.


Lancet Oncology | 2014

Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: a meta-analysis of individual patient data

Antonio Palumbo; Sara Bringhen; Shaji Kumar; Giulia Lupparelli; Saad Z Usmani; Anders Waage; Alessandra Larocca; Bronno van der Holt; Pellegrino Musto; Massimo Offidani; Maria Teresa Petrucci; Andrea Evangelista; Sonja Zweegman; Ajay K. Nooka; Andrew Spencer; Meletios A. Dimopoulos; Roman Hájek; Michele Cavo; Paul G. Richardson; Sagar Lonial; Giovannino Ciccone; Mario Boccadoro; Kenneth C. Anderson; Bart Barlogie; Pieter Sonneveld; Philip L. McCarthy

BACKGROUND Lenalidomide has been linked to second primary malignancies in myeloma. We aimed to pool and analyse available data to compare the incidence of second primary malignancies in patients with and without lenalidomide exposure. METHODS We identified relevant studies through a search of PubMed and abstracts from the American Society of Clinical Oncology, American Society of Hematology, and the International Myeloma Workshop. Randomised, controlled, phase 3 trials that recruited patients with newly diagnosed multiple myeloma between Jan 1, 2000, and Dec 15, 2012, and in which at least one group received lenalidomide were eligible for inclusion. We obtained individual patient data (age, sex, date of diagnosis, allocated treatment and received treatment, duration of treatment and cause of discontinuation, maintenance treatment, date of first relapse, date of second primary malignancy diagnosis, type of second primary malignancy, date of death or last contact, and cause of death) by direct collaboration with the principal investigators of eligible trials. Primary outcomes of interest were cumulative incidence of all second primary malignancies, solid second primary malignancies, and haematological second primary malignancies, and were analysed by a one-step meta-analysis. FINDINGS We found nine eligible trials, of which seven had available data for 3254 patients. 3218 of these patients received treatment (2620 had received lenalidomide and 598 had not), and were included in our analyses. Cumulative incidences of all second primary malignancies at 5 years were 6·9% (95% CI 5·3-8·5) in patients who received lenalidomide and 4·8% (2·0-7·6) in those who did not (hazard ratio [HR] 1·55 [95% CI 1·03-2·34]; p=0·037). Cumulative 5-year incidences of solid second primary malignancies were 3·8% (95% CI 2·7-4·9) in patients who received lenalidomide and 3·4% (1·6-5·2) in those that did not (HR 1·1 [95% CI 0·62-2·00]; p=0·72), and of haematological second primary malignancies were 3·1% (95% CI 1·9-4·3) and 1·4% (0·0-3·6), respectively (HR 3·8 [95% CI 1·15-12·62]; p=0·029). Exposure to lenalidomide plus oral melphalan significantly increased haematological second primary malignancy risk versus melphalan alone (HR 4·86 [95% CI 2·79-8·46]; p<0·0001). Exposure to lenalidomide plus cyclophosphamide (HR 1·26 [95% CI 0·30-5·38]; p=0·75) or lenalidomide plus dexamethasone (HR 0·86 [95% CI 0·33-2·24]; p=0·76) did not increase haematological second primary malignancy risk versus melphalan alone. INTERPRETATION Patients with newly diagnosed myeloma who received lenalidomide had an increased risk of developing haematological second primary malignancies, driven mainly by treatment strategies that included a combination of lenalidomide and oral melphalan. These results suggest that alternatives, such as cyclophosphamide or alkylating-free combinations, should be considered instead of oral melphalan in combination with lenalidomide for myeloma. FUNDING Celgene Corporation.

Collaboration


Dive into the Anders Waage's collaboration.

Top Co-Authors

Avatar

Anders Sundan

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Pieter Sonneveld

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toril Holien

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Peter Gimsing

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Kristine Misund

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Henrik Hjorth-Hansen

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Therese Standal

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Niels Abildgaard

Odense University Hospital

View shared research outputs
Top Co-Authors

Avatar

Sonja Zweegman

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge