Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: British Journal of Haematology, Wiley, Vol. 174, No. 3 ( 2016-08), p. 454-460
    Abstract: Prevention of bleeding and joint damage in severe haemophilia is dependent on adherence to prophylactic replacement therapy. The aim of this study was to assess adherence to prophylaxis, including associations with age, bleeding and clotting factor consumption ( CFC ). In three Dutch haemophilia centres, semi‐structured interviews about adherence to prophylaxis in the previous 2 weeks were conducted with patients or parents of a child with haemophilia. Patients were classified, according to pre‐specified definitions, as adherent, sub‐optimally adherent or non‐adherent based on missing, timing, and dose of infusions. Association of annual bleeding rates, mean CFC , person performing the infusion (parents verus patients) with adherence categories were analysed. Overall, 241 patients with haemophilia using prophylaxis were studied. Parents were more adherent (66%; n  = 48/73) than patients (43%; n  = 72/168). Sub‐optimal adherence occurred in 29% of parents and 37% of patients and was characterized by changes in timing of infusion (mostly from morning to evening), while missing 〈 6% of infusions. Non‐adherence occurred less often: in 5% of parents and 20% of patients. Reduced adherence was associated with lower CFC , but not with joint bleeding. In conclusion, non‐adherence in haemophilia was relatively rare, yet 1/3 of patients struggled to administer prophylaxis at the appropriate time of day.
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 1475751-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Immunology, Springer Science and Business Media LLC, Vol. 42, No. 7 ( 2022-10), p. 1521-1534
    Abstract: Disorders of the long arm of chromosome 11 (11q) are rare and involve various chromosomal regions. Patients with 11q disorders, including Jacobsen syndrome, often present with a susceptibility for bacterial and prolonged viral and fungal infections partially explained by hypogammaglobulinemia. Additional T lymphocyte or granular neutrophil dysfunction may also be present. In order to evaluate infectious burden and immunological function in patients with 11q disorders, we studied a cohort of 14 patients with 11q deletions and duplications. Clinically, 12 patients exhibited prolonged and repetitive respiratory tract infections, frequently requiring (prophylactic) antibiotic treatment ( n  = 7), ear-tube placement ( n  = 9), or use of inhalers ( n  = 5). Complicated varicella infections ( n  = 5), chronic eczema ( n  = 6), warts ( n = 2), and chronic fungal infections ( n  = 4) were reported. Six patients were on immunoglobulin replacement therapy. We observed a high prevalence of low B lymphocyte counts ( n  = 8), decreased T lymphocyte counts ( n  = 5) and abnormal T lymphocyte function ( n  = 12). Granulocyte function was abnormal in 29% without a clinical phenotype. Immunodeficiency was found in patients with terminal and interstitial 11q deletions and in one patient with terminal 11q duplication. Genetically, FLI1 and ETS1 are seen as causative for the immunodeficiency, but these genes were deleted nor duplicated in 4 of our 14 patients. Alternative candidate genes on 11q may have a role in immune dysregulation. In conclusion, we present evidence that inborn errors of immunity are present in patients with 11q disorders leading to clinically relevant infections. Therefore, broad immunological screening and necessary treatment is of importance in this patient group.
    Type of Medium: Online Resource
    ISSN: 0271-9142 , 1573-2592
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2016755-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2021
    In:  Clinical Pharmacokinetics Vol. 60, No. 1 ( 2021-01), p. 1-16
    In: Clinical Pharmacokinetics, Springer Science and Business Media LLC, Vol. 60, No. 1 ( 2021-01), p. 1-16
    Type of Medium: Online Resource
    ISSN: 0312-5963 , 1179-1926
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2043781-X
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: British Journal of Clinical Pharmacology, Wiley, Vol. 88, No. 6 ( 2022-06), p. 2757-2768
    Abstract: Prophylactic treatment of haemophilia A patients with factor VIII (FVIII) concentrate focuses on maintaining a minimal trough FVIII activity level to prevent bleeding. However, due to differences in bleeding tendency, the pharmacokinetic (PK)‐guided dosing approach may be suboptimal. An alternative approach could be the addition of haemostatic pharmacodynamic (PD) parameters, reflecting a patient's unique haemostatic balance. Our aim was to develop a population PK/PD model, based on FVIII activity levels and Nijmegen Haemostasis Assay (NHA) patterns, a global haemostatic assay that measures thrombin/plasmin generation simultaneously. Methods PK/PD measurements were collected from 30 patients treated with standard half‐life FVIII concentrate. The relationship between FVIII activity levels and the thrombin/plasmin generation parameters (thrombin potential, thrombin peak height and plasmin peak height), were described by sigmoidal E max functions. Results The obtained EC 50 value was smallest for the normalized thrombin potential (11.6 IU/dL), followed by normalized thrombin peak height (56.6 IU/dL) and normalized plasmin peak height (593 IU/dL), demonstrating that normalized thrombin potential showed 50% of the maximal effect at lower FVIII activity levels. Substantial inter‐individual variability in the PD parameters, such as EC 50 of thrombin potential (86.9%) was observed, indicating that, despite similar FVIII activity levels, haemostatic capacity varies significantly between patients. Conclusion These data suggest that dosing based on patients' individual PK/PD parameters may be beneficial over dosing solely on individual PK parameters. This model could be used as proof‐of‐principle to examine the application of PK/PD‐guided dosing. However, the relation between the PD parameters and bleeding has to be better defined.
    Type of Medium: Online Resource
    ISSN: 0306-5251 , 1365-2125
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 1498142-7
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: British Journal of Clinical Pharmacology, Wiley, Vol. 87, No. 11 ( 2021-11), p. 4408-4420
    Abstract: Population pharmacokinetic (PK) models are increasingly applied to perform individualized dosing of factor VIII (FVIII) concentrates in haemophilia A patients. To guarantee accurate performance of a population PK model in dose individualization, validation studies are of importance. However, external validation of population PK models requires independent data sets and is, therefore, seldomly performed. Therefore, this study aimed to validate a previously published population PK model for FVIII concentrates administrated perioperatively. Methods A previously published population PK model for FVIII concentrate during surgery was validated using independent data from 87 children with severe haemophilia A with a median (range) age of 2.6 years (0.03–15.2) and body weight of 14 kg (4–57). First, the predictive performance of the previous model was evaluated with MAP Bayesian analysis using NONMEM v7.4. Subsequently, the model parameters were (re)estimated using a combined dataset consisting of the previous modelling data and the data available for the external validation. Results The previous model underpredicted the measured FVIII levels with a median of 0.17 IU mL −1 . Combining the new, independent and original data, a dataset comprising 206 patients with a mean age of 7.8 years (0.03–77.6) and body weight of 30 kg (4–111) was obtained. Population PK modelling provided estimates for CL, V1, V2, and Q: 171 mL h −1  68 kg −1 , 2930 mL 68 kg −1 , 1810 mL 68 kg −1 , and 172 mL h −1  68 kg −1 , respectively. This model adequately described all collected FVIII levels, with a slight median overprediction of 0.02 IU mL −1 . Conclusions This study emphasizes the importance of external validation of population PK models using real‐life data.
    Type of Medium: Online Resource
    ISSN: 0306-5251 , 1365-2125
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 1498142-7
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: British Journal of Clinical Pharmacology, Wiley
    Abstract: Recombinant factor IX Fc fusion protein (rFIX‐Fc) is an extended half‐life factor concentrate administered to haemophilia B patients. So far, a population pharmacokinetic (PK) model has only been published for patients aged ≥12 years. The aim was to externally evaluate the predictive performance of the published rFIX‐Fc population PK model for patients of all ages and develop a model that describes rFIX‐Fc PK using real‐world data. Methods We collected prospective and retrospective data from patients with haemophilia B treated with rFIX‐Fc and included in the OPTI‐CLOT TARGET study (NTR7523) or United Kindom (UK)‐EHL Outcomes Registry (NCT02938156). Predictive performance was assessed by comparing predicted with observed FIX activity levels. A new population PK model was constructed using nonlinear mixed‐effects modelling. Results Real‐world data were obtained from 37 patients (median age: 16 years, range 2–71) of whom 14 were aged 〈 12 years. Observed FIX activity levels were significantly higher than levels predicted using the published model, with a median prediction error of −48.8%. The new model showed a lower median prediction error (3.4%) and better described rFIX‐Fc PK, especially for children aged 〈 12 years. In the new model, an increase in age was correlated with a decrease in clearance ( P   〈  .01). Conclusions The published population PK model significantly underpredicted FIX activity levels. The new model better describes rFIX‐Fc PK, especially for children aged 〈 12 years. This study underlines the necessity to strive for representative population PK models, thereby avoiding extrapolation outside the studied population.
    Type of Medium: Online Resource
    ISSN: 0306-5251 , 1365-2125
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 1498142-7
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1030-1030
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1030-1030
    Abstract: Introduction Sickle cell disease (SCD) is an autosomally, recessive inherited hemoglobinopathy and multisystem disorder characterized by ongoing hemolytic anemia, episodes of vaso-occlusion and progressive organ failure with ultimately a shortened life expectancy. Despite intensive comprehensive care and improved rates of morbidity and mortality, SCD care is still marked by high utilization of medical resources. Until now, most cost-of care studies have focused on one or two care categories, such as hospitalizations and physician visits [1-4]. Also, few studies have evaluated healthcare expenditures exclusively in children. Estimating cost-of care is important as it can ensure sufficient allocation of resources. In addition, SCD expenditures can be used to raise awareness of disease severity and serve as an incentive for prevention and management of disease complications. Primary aims of this study were to (a) investigate the overall cost of healthcare for pediatric SCD patients and to (b) estimate major cost drivers. Methods All pediatric SCD patients visiting the Erasmus University Medical Center-Sophia Children's Hospital for routine or emergency care from January 1st to December 31st 2017 with a diagnosis of SCD were included. Retrospective data of this cohort were analyzed for 24 months during January 1st 2015 to December 1st 2016. Patients were grouped into four age categories; (A) 0-12 months, (B) 1-5 years, (C) 5-13 years and (D) 13-19 years. For patients born before January 1st 2015, each individual contributed two years of follow-up time. As some children were born during one of the two years during the study time period, the weighted average was calculated based on the time patients were potentially able to make costs. Healthcare utilization of included patients was based upon data from two main sources. The clinical SCD standard treatment guideline was used to determine the expected resource use of routine comprehensive care (planned elective care) and the Erasmus University Medical Center financial claims database was used to estimate real-world resource use associated with acute and inpatient care (additional care). The included items for the SCD guideline and financial claims database per cost category are summarized in Table 1. Results A total of 125 patients were analyzed. The mean age was 8.1 years (SD: 5 years) on December 31st 2015 and 9.9 year (SD: 5 years) on December 31st 2016. Expenditures for the 125 children with SCD averaged €4285.09 (SD: €820.36) per child per year. The majority (49%) of costs was associated with standard treatment (i.e. prophylactic antibiotics); 23% with diagnostics; 19% with inpatient hospital care and 9% with outpatients visits. Annual average costs per patient per age group are depicted in Figure 1. Total expenditures for children with SCD increased per age group, ranging from €2962 (category A), €3726 (category B), and €4087 (category C) to €5890 (category D). This was mostly explained by increases in admission costs. Discussion Although healthcare utilization and costs of pediatric SCD patients have been studied previously [5-7], studies from Europe comprehensive care centers are scarce and have been mostly based on only one aspect of care such as hospitalizations costs [2] . To our knowledge, this is the first study combining standard treatment costs with real world resource use. The total annual coast of healthcare for children with SCD, including inpatient care, outpatient care, diagnostics and treatment averaged €4285.09 per patient per year. This is much lower when compared to costs of healthcare for pediatric patients with SCD reported in other studies. Kauf et al. calculated total costs of healthcare for SCD patients aged 0-9 years to be $10.704 [7] . In addition, inpatient care accounts for a relatively small part of total costs in our study. This finding has been inline with previous research where comprehensive care has been suggested as a means of reducing costs associated with SCD care [8]. A comprehensive, multidisciplinary approach is necessary to address the physical, mental and social needs of each child and their family. Comprehensive care, with effective management in the outpatient setting is able to prevent hospital admission, and is essential for delivery of high quality cost-effective care in SCD. Disclosures Cnossen: Pfizer: Other: Travel Grants, Research Funding; Bayer: Other: Travel Grants, Research Funding; Novo Nordisk: Research Funding; Nordic Pharma: Research Funding; CSL Behring: Other: Travel Grants, Research Funding; Sobi: Research Funding; Baxter: Other: Travel Grants, Research Funding; Shire: Other: Travel Grants, Research Funding; Takeda: Other: Travel Grants, Research Funding; Roche: Other: Travel Grants; NWO: Other: Governmental grants , ZonMW, Innovation fund and Nationale Wetenschapsagenda 2018.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 125, No. 19 ( 2015-05-07), p. 3006-3013
    Abstract: VWFpp discriminates between type 3 VWD patients and severe type 1 VWD patients with very low VWF levels. The pathophysiological mechanisms of all types of VWD can be defined by the combined ratios of VWFpp/VWF:Ag and FVIII:C/VWF:Ag.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 4836-4836
    Abstract: Introduction The most common neurological finding in children and adults with sickle cell disease (SCD) is the presence of silent cerebral infarcts (SCIs). SCIs are, by definition, undetectable by neurologic examination and are recognized on magnetic resonance imaging (MRI) as increased signal intensity in multiple T2-weighted images (1). SCIs are associated with a decline in full-scale IQ (2, 3) and increased risk for future strokes (4). In this paper, we provide an overview of studies that have used magnetic resonance imaging (MRI) to detect SCIs in patients with SCD. We systematically review evidence on SCI prevalence, incidence and associated risk factors. Methods A comprehensive systematic search was performed of Embase, MEDLINE, Cochrane, Web of Science and Google Scholar. Publications through June 1st, 2019 were included. Search terms included synonyms for 'SCIs' and 'SCD' in various combinations. All references listed in identified papers, or noted in footnotes of data tables, were reviewed for additional articles missed by computerized database search. After removal of duplicates, 651 studies were screened on potential eligibility. Studies were included if they concerned patients of all ages with either homozygous or compound heterozygous SCD and assessed for and specifically reported MRI-detected SCIs (Figure 1). We present a weighted prevalence by study size. When multiple articles reported on a single or overlapping population sample, prevalence estimates were obtained from the article with the largest sample size to prevent duplications. Results We included 61 original papers describing results in a total of 3740 individual SCD patients. Most studies were performed in HbSS or HbSβ0thal SCD patients (n=2122). However, five studies separately reported SCI prevalence in other SCD genotypes i.e. HbSC, HbSβ+thal (n=254) and healthy controls (n=52). The pooled prevalence of SCI among all included patients with SCD (n=3740) was 28.4% (95% CI: 4.0;32.9). Data from included studies showed a statistically significant correlation (P 〈 0.001) between mean SCI prevalence % and increasing mean patient sample age (Figure 2). SCIs were more common in patients with HbSS/HbSβ0thal compared to other SCD genotypes (p 〈 0.001) and healthy controls (P 〈 0.001) (Figure 3). Studies focusing on incidence rates were sparse, with only four identified studies providing estimates ranging from 3.1% to 13.6% per year. The majority of included studies analyzed possible risk- or protective factors for SCI in SCD patients, i.e. hematological parameters, α-thalassemia presence and cerebral blood flow velocity (Table 1). Conflicting results are noted for most risk- and protective factors, with equal numbers of similar studies finding significant versus non-significant associations. Conclusions SCIs are common in patients with SCD with a weighted prevalence of 28.4%. Despite advancing neuroimaging technologies and therefore potentially enhanced detection of SCIs, there has been no apparent rise in SCI prevalence over the years. SCIs occur much more frequently in individuals with HbSS or HbSβ0thal than in other SCD genotypes. Analyses of risk- and protective factors showing varying results. The absence of robust findings is probably due to a combination of small sample sizes and weak associations. We believe SCIs are a poorly understood entity in patients with SCD. Despite substantial research efforts into risk- and protective factors, too little attention has been given to underlying pathophysiological mechanisms. Further research should focus on both SCI etiology in SCD as well as on clinical management of patients with evidence of these MRI lesions. Disclosures Cnossen: Pfizer: Other: Travel Grants, Research Funding; Bayer: Other: Travel Grants, Research Funding; Novo Nordisk: Research Funding; Nordic Pharma: Research Funding; CSL Behring: Other: Travel Grants, Research Funding; Sobi: Research Funding; Baxter: Other: Travel Grants, Research Funding; Shire: Other: Travel Grants, Research Funding; Takeda: Other: Travel Grants, Research Funding; Roche: Other: Travel Grants; NWO: Other: Governmental grants , ZonMW, Innovation fund and Nationale Wetenschapsagenda 2018.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 5038-5038
    Abstract: Background: Patients’, parents’ and providers’ preferences with regard to medical interventions may have a major impact on the implementation of innovations, often delaying initiation significantly. Illustratively, as early as 1997, Carlsson et al. suggested that a 30% reduction of consumption of clotting factor concentrate in prophylactic treatment could be attained by dosing based on an individual pharmacokinetic (PK) profile, with a concomitant cost reduction. Therefore, we aim to evaluate do’s and don’ts in hemophilia patients, parents and professionals with regard to individualized dosing according to PK-profile of prophylaxis with clotting factor concentrate. This in order to successfully implement this intervention when imperative. Methods: In this study we included patientswith hemophilia currently or previously on prophylactic treatment with clotting factor concentrate (n=114) and parents of patients aged 12-18 years (n=19) from five Dutch Academic Hemophilia Treatment Centers, and hemophilia professionals attending the World Federation of Haemophia congress 2012 from throughout the world (n=91). Data were analysed using a Discrete Choice Experiment. Patients’, parents’ and professionals’ preferences with regard to the intervention, are measured by specific attributes with varying levels: ‘number of blood samples necessary to construct individual PK-profile’, ‘advised frequency of prophylactic infusions’, ‘frequency of repetitive PK-profiling’, ‘risk of bleeding’, ‘estimated cost reduction of treatment with benefit for society’. Results: For patients and parents (response rate 64%), a higher dosing frequency e.g. daily dosing was an important barrier. They were however willing to infuse more frequently, if bleeding was consequently reduced. ‘Reduction of costs for society’ by implementation of individualized dosing according to PK profile was found relevant and motivating to implement PK-guided dosing. For professionals the most important attributes driving implementation were an acceptable ‘advised frequency of prophylactic infusions’ and reduction of ‘risk of bleeding’. Conclusions: When anticipating implementation of a medical intervention, defining of preferences of those involved is of importance. In case of PK-guided prophylactic dosing in hemophilia conclusions are: realise the impact of daily dosing of clotting factor concentrate, use frequent bleeding as a motivator to initiate PK-guided dosing and actively discuss costs of treatment with those undergoing treatment and the cost reduction that may result from PK-guided dosing. Identification of these preferences will secure successful implementation in the near future. Disclosures Lock: ZonMW: Research Funding; Baxter: Research Funding. Laros-van Gorkum:Sanquin: speakers fee Other; Baxter: Unrestricted educational grant was provided to the Hemophilia Treatment Center of the Radboud university medical center, Unrestricted educational grant was provided to the Hemophilia Treatment Center of the Radboud university medical center Other; CSL Behring: Unrestricted educational grant was provided to the Hemophilia Treatment Center of the Radboud university medical center, Unrestricted educational grant was provided to the Hemophilia Treatment Center of the Radboud university medical center Other. Driessens:Baxter: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; Bayer Schering Pharma: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work, unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; CSL Behring: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work, unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; Eurocept: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work, unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; Novo Nordisk: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; Pfizer: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other; Sanquin: unrestricted grant for meetings and educational courses with hemophilia patients and members of the Netherlands Hemophilia Patient Society, outside the submitted work Other. Fijnvandraat:Baxter: Member of the European Hemophilia Treatment and Standardisation Board sponsored by Baxter Other; CSL Behring: Research Funding; Pfizer: Has given lectures at educational symposiums organised by Pfizer, Has given lectures at educational symposiums organised by Pfizer Other, Research Funding; Bayer Schering Pharma: Has given lectures at educational symposiums organised by Bayer Schering Pharma, Has given lectures at educational symposiums organised by Bayer Schering Pharma Other, Research Funding. Leebeek:CSL Behring: has served on advisory boards of CSL Behring, outside the submitted work Other, Research Funding; Baxter: has served on advisory boards of Baxter, outside the submitted work, has served on advisory boards of Baxter, outside the submitted work Other. Cnossen:Pfizer: Educational funding Other, Research Funding; Bayer Schering Pharma: Educational funding and travel support, Educational funding and travel support Other, Research Funding; Baxter: Educational funding and travel support, Educational funding and travel support Other, Research Funding; Novo Nordisk: Educational funding, Educational funding Other, Research Funding; Novartis: Educational funding and travel support Other, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages