Efficacy and (Pharmaco)Kinetics of One Single Dose of Rasburicase in Patients with Chronic Kidney Disease

2008 ◽  
Vol 108 (4) ◽  
pp. c265-c271 ◽  
Author(s):  
E. Sestigiani ◽  
M. Mandreoli ◽  
M. Guardigli ◽  
A. Roda ◽  
E. Ramazzotti ◽  
...  
2020 ◽  
Vol 36 (1) ◽  
pp. 111-120 ◽  
Author(s):  
Sunil Bhandari ◽  
Philip A Kalra ◽  
Mario Berkowitz ◽  
Diogo Belo ◽  
Lars L Thomsen ◽  
...  

Abstract Background The optimal intravenous (IV) iron would allow safe correction of iron deficiency at a single infusion over a short time. The FERWON-NEPHRO trial evaluated the safety and efficacy of iron isomaltoside 1000/ferric derisomaltose (IIM) in patients with non-dialysis-dependent chronic kidney disease and iron deficiency anaemia. Methods In this randomized, open-label and multi-centre trial conducted in the USA, patients were randomized 2:1 to a single dose of 1000 mg IIM or iron sucrose (IS) administered as 200 mg IV injections up to five times within a 2-week period. The co-primary endpoints were serious or severe hypersensitivity reactions and change in haemoglobin (Hb) from baseline to Week 8. Secondary endpoints included incidence of composite cardiovascular adverse events (AEs). Results A total of 1538 patients were enrolled (mean estimated glomerular filtration rate 35.5 mL/min/1.73 m2). The co-primary safety objective was met based on no significant difference in the incidence of serious or severe hypersensitivity reactions in the IIM and IS groups [0.3% versus 0%; risk difference: 0.29% (95% confidence interval: –0.19; 0.77; P > 0.05)]. Incidence of composite cardiovascular AEs was significantly lower in the IIM versus IS group (4.1% versus 6.9%; P = 0.025). Compared with IS, IIM led to a more pronounced increase in Hb during the first 4 weeks (P ≤ 0.021), and change in Hb to Week 8 showed non-inferiority, confirming that the co-primary efficacy objective was met. Conclusions Compared with multiple doses of IS, a single dose of IIM induced a non-inferior 8-week haematological response, comparably low rates of hypersensitivity reactions, and a significantly lower incidence of composite cardiovascular AEs.


Author(s):  
JUNKO YANO ◽  
SAKUYA ITO ◽  
GOH KODAMA ◽  
YOSUKE NAKAYAMA ◽  
YUSUKE KAIDA ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3532-3532
Author(s):  
Anne-Marie Duliege ◽  
Iain Macdougall ◽  
Neill Duncan ◽  
Dawie Wessels ◽  
Julie Iwashita ◽  
...  

Abstract Hematide is a PEGylated synthetic peptidic ESA that binds to and activates the erythropoietin (EPO) receptor. It has demonstrated potent and sustained dose dependent erythropoietic activity in healthy volunteers (HV). Methods: A randomized double-blind placebo controlled dose escalation study is being conducted to assess safety, pharmacokinetics (PK) and pharmacodynamics (PD) of Hematide administered as a single intravenous (IV) injection in EPO-naïve patients with Chronic Kidney Disease (CKD) pre-dialysis. Cohorts of 9 patients each (7 on Hematide and 2 on placebo) are planned with escalating doses. The results of this study are compared with those of a recently completed Phase 1 dose escalating study of Hematide in 4 cohorts of HV in which the pharmacologically active dose (PAD; i.e., the dose resulting in a statistically and clinically significant increase in hemoglobin) of Hematide was defined. Results: 28 day follow up of the 9 patients in Cohort 1 is complete. As of this date, the study remains blinded and we assume that increases in reticulocytes and hemoglobin (Hgb) are due to Hematide. Injections were well tolerated with no adverse events reported as related to study drug injection. In 7 patients whose reticulocytes increased, counts peaked at 7–10 days with a mean change from baseline of 151.4 ± 18.4 x 109/L before returning within baseline at 21 days post-injection. A Hgb increase was also observed in these 7 patients with a mean difference in change from baseline Hgb between these 7 responding and the 2 non-responding subjects of 0.80 g/dL at Day 10 (p = 0.028) and 1.16 g/dL (p = 0.08) at Day 28 post-injection. The PD profile of these 7 responding patients is comparable to that of the 10 HV receiving a PAD of Hematide. For example, the average maximum Hgb increase from baseline was 1.22 g/dL (SD: 0.37) in patients versus 1.36 g/dL (SD: 0.39) in HV. However, the PAD for CKD patients was half the Hematide dose that was required to obtain a similar Hgb effect in HV. Unblinded results from the complete study, including dose responses, follow-up after 28 days, PK data, additional PD data, EPO levels and iron status assessment will be presented and compared to the results in HV. Conclusion: A single IV injection of Hematide administered to patients with CKD was well tolerated and demonstrated potent and sustained erythropoietic activity of the same magnitude as that observed in HV. The PAD in patients with CKD was half of that observed in HV. These preliminary data suggest that patients with CKD may be more sensitive to Hematide and/or might have a different PK profile compared to HV. The sustained increases in Hgb levels for at least one month following a single dose of Hematide are compatible with anticipated monthly dosing of Hematide to maintain Hgb in target range. This is a reduced frequency of administration compared to that of currently available ESAs. These results serve as a basis to plan studies of repeat doses of Hematide in patients with anemia related to cancer or CKD and this plan will be outlined.


2018 ◽  
Vol 34 (1) ◽  
pp. 145-154 ◽  
Author(s):  
Winnie Y. Sohn ◽  
Anthony A. Portale ◽  
Isidro B. Salusky ◽  
Hao Zhang ◽  
Lucy L. Yan ◽  
...  

Abstract Background Calcimimetics, shown to control biochemical parameters of secondary hyperparathyroidism (SHPT), have well-established safety and pharmacokinetic profiles in adult end-stage renal disease subjects treated with dialysis; however, such studies are limited in pediatric subjects. Methods In this study, the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of cinacalcet were evaluated in children with chronic kidney disease (CKD) and SHPT receiving dialysis. Twelve subjects received a single dose of cinacalcet (0.25 mg/kg) orally or by nasogastric or gastric tube. Subjects were randomized to one of two parathyroid hormone (PTH) and serum calcium sampling sequences: [(1) 2, 8, 48 h; or (2) 2, 12, 48 h] and assessed for 72 h after dosing. Results Median plasma cinacalcet t max was 1 h (range 0.5–4.0 h); mean (SD) C max and AUClast were 2.83 (1.98) ng/mL and 11.8 (8.74) h*ng/mL, respectively; mean (SD) half-life (t 1/2) was 3.70 (2.57) h. Dose adjustments, based upon body weight (mg/kg), minimized the effects of age, body weight, body surface area, and body mass index on cinacalcet PK. Reductions in serum PTH levels from baseline were observed at 2 to 8 h post-dose (median 10.8 and 29.6%, respectively), returned towards baseline by 12–72 h and were inversely related to changes in the plasma cinacalcet PK profile. Single-dose cinacalcet was well-tolerated with no unexpected safety findings and a PK/PD, safety profile similar to adults. Conclusions In conclusion, a single 0.25 mg/kg dose of cinacalcet was evaluated to be a safe starting dose in these children aged < 6 years.


2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A216-A217
Author(s):  
Akshan Puar ◽  
Zeb Ijaz Saeed

Abstract Introduction: Denosumab, a monoclonal antibody that inhibits RANK L (receptor activator nuclear factor-kappa beta ligand), is one of the few medications that can be used to treat osteoporosis in patients with chronic kidney disease (CKD). However, its use is associated with a much higher incidence of hypocalcemia in this patient population. What remains unclear is the duration of hypocalcemia after denosumab use. We describe a case of prolonged hypocalcemia of 9 months in a patient with CKD after a single dose of denosumab. Case: A 64-year-old Caucasian man with a history of bilateral lung transplant for interstitial pulmonary fibrosis and CKD Stage IV was referred to the Endocrinology clinic for evaluation of steroid-induced osteoporosis. Bone density scan was consistent with osteoporosis with the lowest T-score of -2.8 at the left femoral neck, which showed a 25.3% decline from a previous one two years prior. His labs upon initial visit: 25 hydroxy Vitamin D: 36.5 ng/mL (30–100), 1, 25 hydroxy vitamin D 32 pg/ml (19.9–79.3), corrected Serum Calcium 8.9 mg/dL (8.5–10.5), Serum Cr 4.38 mg/dL (0.6–1.4), PTH 157 pg/mL (10–65), Serum Alkaline Phosphatase 61 Units/L (25–125), Urine NTX 39 nM BCE/mM creatinine (21–83). After discussing risks and benefits, he was given a dose of subcutaneous denosumab 60 mg. He had been started on Calcium/Vitamin D (600 mg/400 IU BID) prior to receiving his dose. Keeping in mind the increased risk of hypocalcemia given his history of CKD, his corrected serum calcium was checked one week later, and it was 6.5 mg/dL. The patient was asymptomatic. However, given the severity of his hypocalcemia, he was started on calcitriol 0.25 mcg oral BID and calcium carbonate 1200 mg daily. He did show mild improvement in three days to a corrected calcium of 7.0 mg/dL. His calcitriol was briefly increased to 0.5 mcg BID and calcium carbonate was increased to 1800 mg daily. The regimen was weaned to calcitriol 0.25 mcg daily and previous calcium/Vitamin D dosing later that month. Thereafter, his labs were monitored regularly and there were several unsuccessful attempts made to decrease the calcitriol/calcium carbonate. Given persistent hypocalcemia, other bloodwork including a bone specific alkaline phosphatase and celiac screen were checked which were unremarkable. Finally, nine months after his denosumab dose, calcitriol was discontinued safely. Serum calcium levels have remained stable thereafter. Given prolonged hypocalcemia, it was decided not to administer another dose of denosumab. Conclusion: Patients with CKD who receive denosumab are not only at risk for developing severe, but also prolonged hypocalcemia. Therefore, it is imperative to monitor serum calcium levels, not only immediately after receiving a dose, but serially.


2020 ◽  
Vol 21 (22) ◽  
pp. 8868
Author(s):  
Marta Gil-Fernández ◽  
José Alberto Navarro-García ◽  
Almudena Val-Blasco ◽  
Laura González-Lafuente ◽  
José Carlos Martínez ◽  
...  

Risk of cardiovascular disease (CVD) increases considerably as renal function declines in chronic kidney disease (CKD). Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) has emerged as a novel innate immune receptor involved in both CVD and CKD. Following activation, NOD1 undergoes a conformational change that allows the activation of the receptor-interacting serine/threonine protein kinase 2 (RIP2), promoting an inflammatory response. We evaluated whether the genetic deficiency of Nod1 or Rip2 in mice could prevent cardiac Ca2+ mishandling induced by sixth nephrectomy (Nx), a model of CKD. We examined intracellular Ca2+ dynamics in cardiomyocytes from Wild-type (Wt), Nod1−/− and Rip2−/− sham-operated or nephrectomized mice. Compared with Wt cardiomyocytes, Wt-Nx cells showed an impairment in the properties and kinetics of the intracellular Ca2+ transients, a reduction in both cell shortening and sarcoplasmic reticulum Ca2+ load, together with an increase in diastolic Ca2+ leak. Cardiomyocytes from Nod1−/−-Nx and Rip2−/−-Nx mice showed a significant amelioration in Ca2+ mishandling without modifying the kidney impairment induced by Nx. In conclusion, Nod1 and Rip2 deficiency prevents the intracellular Ca2+ mishandling induced by experimental CKD, unveiling new innate immune targets for the development of innovative therapeutic strategies to reduce cardiac complications in patients with CKD.


2018 ◽  
Vol 12 (3) ◽  
pp. 408-413 ◽  
Author(s):  
Taylor V Hunt ◽  
Joshua M DeMott ◽  
Kimberly A Ackerbauer ◽  
William L Whittier ◽  
Gary D Peksa

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4156-4156
Author(s):  
Atif Irfan Khan ◽  
Anam Khan ◽  
Keren Sam ◽  
Fatima Sajid ◽  
Sara Ashraf ◽  
...  

Abstract Background Anemia in chronic kidney disease (CKD) patients develop primarily due to iron and erythropoietin deficiency, factors contributing to this include uremia induced inhibition of erythropoiesis, shortened red cell survival and nutritional deficiency. Erythropoietin stimulating agents (ESA) used in CKD increases iron demand for the production of new red blood cells (RBCs). Oral iron does not provide a reliable route to meet the demand of the iron required and poses the risk of side effects. The intravenous formulation provides the fastest route, although multiple intravenous formulations are available they require multiple dosing and still carry a risk of an adverse event. Ferric derisomaltose/iron isomaltoside (IIM) is an intravenous formulation of iron recently approved by the Food and drug administration (FDA) for patients intolerant to oral iron and non-dialysis dependent CKD patients. FDI is administered in high dose usually in a single administration. In this systematic review, we evaluated the published literature for the efficacy and safety of ferric derisomaltose in both dialysis-dependent and dialysis independent patients. Material/Methods A literature search was done using the following databases: PubMed, Cochrane, Embase, Clinical trials.gov, and Web of Science. The search was completed without using any filter and we used the Mesh Terms for "anemia", "iron deficiency anemia" "chronic kidney disease" and "ferric compounds". Total of 316 which were further screened and we included 2 trials, and 2 prospective studies. Case reports, preclinical trials, meta-analyses, and review articles were excluded. We followed the PRISMA guidelines for literature search and selection of studies. We only included the trial studies that were completed. We excluded the studies that did not report efficacy and safety. Results In total, amongst the four studies, 1558 CKD patients received ferric derisomaltose in a single dose. Patients in all stages of CKD were studied but most patients were between stages I to stage III CKD, stage IV and V(dialysis-dependent) were studied by Kalra et al (2020). A single dose of 1000 mg/dl of IIM, was administered intravenously. The mean pre-treatment haemoglobin level varied from 9.6g/dl to 10.6 g/dl. The mean serum ferritin ranged from 82.4 µg/L to 161.9 µg/L and the mean transferrin saturation ranged in patients from 15.8% to 18.51%. IIM receiving patients were compared with iron sucrose receiving patients by Bhandari et al, and oral iron by Kalra et al (2015). The remaining two studies compared single IIM with multiple IIM dosages. The results are summarised in the table below. Efficacy: The increase in haemoglobin ranged between 0.9 g/dL to 6.58 g/dL from baseline measured at 8 weeks. At the same time, serum ferritin showed an increase in baseline serum ferritin from 104 µg/L to 277 µg/L and transferrin saturation (TSAT) increased in the range of 5.3%-11.4 %. All markers of iron deficiency showed significant improvement. It achieved a greater Hb response irrespective of ESA treatment in both haemodialysis dependent and non-dependent patients. In the comparison group by Bhandari et al, iron sucrose receiving patients also had similar efficacy as IIM. On the other hand, those receiving oral iron in the study by Kalra et al (2015) had much poorer efficacy as compared to IIM Safety: Severe adverse effect was seen in a total of 10 (0.6%) patients who developed hypersensitivity reaction, acute myocardial infarction and injection site-related adverse effects with hypersensitivity being the most common. 15 (0.9%) patients discontinued the treatment. 21 (1.3%) died, and the death was attributed to end-stage renal disease. Conclusion Ferric derisomaltose is an effective parenteral iron therapy to treat iron deficiency when compared to oral iron therapy, however, its efficacy is very similar to other parenteral iron formulations. In terms of safety, being used as a single dose it relatively has lesser adverse events but can seldom lead to discontinuation of the drug. This drug can be recommended for iron therapy especially considering single-dose administration but more studies need to be carried out in comparison to other parenteral formulations to further clarify its efficacy in dialysis-dependent CKD patients. Figure 1 Figure 1. Disclosures Anwer: BMS / Celgene: Honoraria, Research Funding; GlaxoSmithKline: Research Funding; Janssen pharmaceutical: Honoraria, Research Funding; Allogene Therapeutics: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document