Arzneimittelforschung 2010; 60(6): 362-372
DOI: 10.1055/s-0031-1296301
Ferric Carboxymaltose
Editio Cantor Verlag Aulendorf (Germany)

Pharmacokinetics, safety and tolerability of intravenous ferric carboxymaltose: a dose-escalation study in volunteers with mild iron-deficiency anaemia

Peter Geisser
1   Vifor (International) Inc., St. Gallen, Switzerland
,
José Banké-Bochita
2   Institute of Clinical Pharmacology, Parexel GmbH, Berlin, Germany
› Author Affiliations
Further Information

Publication History

Publication Date:
21 December 2011 (online)

Abstract

Iron-deficiency anaemia (IDA) represents a major burden to public health worldwide. The therapeutic aim for patients with IDA is to return iron stores and haemoglobin (Hb) levels to within the normal range using supplemental iron therapy and erythropoiesis-stimulating agents. Oral and previous intravenous (i. v.) iron formulations have a number of disadvantages, including immunogenic reactions, oxidative stress, low dosages, long administration times and the requirement for a test dose. Ferric carboxymaltose (FCM, Ferinject®) is a novel, next-generation i. v. iron formulation with the potential to overcome these limitations. In this single-centre, randomized, double-blind, placebo-controlled study, the pharmacokinetics (PK), pharmacodynamics (PD), safety and tolerability of single, escalating doses of FCM were investigated.

Four ascending doses were investigated in a total of 24 patients with mild IDA (defined as serum ferritin < 20 µg/l and transferrin saturation [TfS] < 16%): 100 mg iron as FCM given as an i. v. bolus injection, and 500, 800 and 1000 mg iron as FCM given as an i. v. infusion over 15 min. At each dose level six patients received FCM and two received placebo. The decision to escalate to the next dose was based on evaluation of safety and tolerability data from the previous dose. The maximum duration of the study was 5 weeks from screening to final assessment. Assessments were made of PK iron-status parameters up to 168 h post-dose. Safety assessments included incidence of adverse events (AEs), clinical laboratory parameters and vital signs. PK and PD parameters were analysed using descriptive statistics. All analyses were performed on the safety population, which included all patients who received ≥ 1 dose of study medication.

Seventy-seven patients were screened and, of these, 32 male and female patients with pre-study Hb between 9.2 and 11.9 g/dl and serum ferritin < 20 µg/l were included in the study. Two patients had TfS > 16% (19.2% and 17.2%); both patients were considered by the investigator to be eligible for inclusion. Compared with placebo, a rapid, dose-dependent increase in total serum iron was observed across all dose groups. Mean (standard deviation) maximum total serum iron levels ranged between 36.9 (4.4) and 317.9 (42.3) µg/ml in the 100 and 1000 mg groups. Concentration–time curves of total serum iron continuously declined for up to 24 and 72 h post-dose in the 100 and 500–1000 mg groups, respectively. Non-compartmental analysis of PK parameters was truncated at 24 h (100 mg) and 72 h (500–1000 mg doses). A dose-dependent, but not dose-linear, increase in serum ferritin was seen in all treatment groups compared with placebo, with peak levels of a 23–210-fold increase above baseline occurring 48–120 h postdose. Iron-binding capacity was transiently almost fully utilized after doses of 500, 800 and 1000 mg (TfS > 95%). No meaningful changes in serum transferrin or serum transferrin receptor concentrations were observed during this study. The elimination pattern for FCM appeared to be mono-exponential; FCM was cleared from serum with a terminal half-life of approximately 7.4–12.1 h. The percentage of FCM excreted in urine was negligible (0.0005%). FCM was well tolerated; a total of 19 AEs were reported by 8/32 patients (25%), of which three were considered by the investigator to be related to FCM: nausea and vomiting (one patient [100 mg]), and headache (one patient [1000 mg]). The incidence of AEs did not increase with dose. No severe or serious AEs, or deaths occurred. FCM had no significant effect on laboratory safety parameters or vital signs.

This study satisfactorily characterized the PK/PD parameters of single doses of 100, 500, 800 and 1000 mg iron as FCM. The majority of FCM was utilized or eliminated within 24 h of administration of a 100 mg dose and within 72 h of a 500 −1000 mg dose. FCM was generally well tolerated across all doses in patients with mild IDA.

 
  • References

  • 1 Umbreit J. Iron deficiency: a concise review. Am J Hematol. 2005; Mar 78 (3) 225-31
  • 2 Cook JD, Skikne BS, Baynes RD. Iron deficiency: the global perspective. Adv Exp Med Biol. 1994; 356: 219-28
  • 3 World Health Organization. Iron Deficiency Anaemia. Assessment, Prevention, and Control. A guide for programme managers. 2001. Report No.: Document WHO/NHD/01.3.
  • 4 Crichton RR, Danielson BG, Geisser P editors. Iron Therapy –with special emphasis on intravenous administration. 4th ed. Bremen (Germany): UNI-MED; 2008
  • 5 KDOQI Clinical Practice Guideline and Clinical Practice Recommendations for anemia in chronic kidney disease: 2007 update of hemoglobin target Am J Kidney Dis. 2007; Sep 50 (3) 471-530
  • 6 Locatelli F. Appendix B: Assessment of iron stores and functional iron deficiency. Nephrol Dial Transplant. 2004; 19 (Suppl 2) 39-41
  • 7 Cavill I. Iron status as measured by serum ferritin: the marker and its limitations. Am J Kidney Dis. 1999; Oct 34 (4 Suppl 2) S12-S17
  • 8 London: Royal College of Physicians. National Collaborating Centre for Chronic Conditions. Anaemia management in chronic kidney disease: national clinical guideline for management in adults and children. http://www nice org uk/ 2006 [cited Feb 2010]. Available from: http://www.nice.org.uk/nicemedia/pdf/Anaemia_Management_full_guideline.pdf
  • 9 Locatelli F, Aljama P, Barany P, Canaud B, Carrera F, Eckardt KU et al. Revised European best practice guidelines for the management of anaemia in patients with chronic renal failure. Nephrol Dial Transplant. 2004; May 19 (Suppl 2) ii1-47
  • 10 Bothwell THE, Charlton RW, Cook JD, Finch CA. Iron Metabolism in Man. Oxford: Blackwell Scientific Publications; 1979
  • 11 Cavill I, Auerbach M, Bailie GR, Barrett-Lee P, Beguin Y, Kaltwasser P et al. Iron and the anaemia of chronic disease: a review and strategic recommendations. Curr Med Res Opin. 2006; Apr 22 (4) 731-7
  • 12 Locatelli F, Pisoni RL, Akizawa T, Cruz JM, DeOreo PB, Lameire NH et al. Anemia management for hemodialysis patients: Kidney Disease Outcomes Quality Initiative (K/ DOQI) guidelines and Dialysis Outcomes and Practice Patterns Study (DOPPS) findings. Am J Kidney Dis. 2004; Nov 44 (5 Suppl 2) 27-33
  • 13 Vogelzang NJ, Breitbart W, Cella D, Curt GA, Groopman JE, Horning SJ et al. Patient, caregiver, and oncologist perceptions of cancer-related fatigue: results of a tripart assessment survey. The Fatigue Coalition. Semin Hematol. 1997; Jul 34 (3 Suppl 2) 4-12
  • 14 Dresow B, Petersen D, Fischer R, Nielsen P. Non-transferrin-bound iron in plasma following administration of oral iron drugs. Biometals. 2008; Jun 21 (3) 273-6
  • 15 Hutchinson C, Al-Ashgar W, Liu DY, Hider RC, Powell JJ, Geissler CA. Oral ferrous sulphate leads to a marked increase in pro-oxidant nontransferrin-bound iron. Eur J Clin Invest. 2004; Nov 34 (11) 782-4
  • 16 Hamstra RD, Block MH, Schocket AL. Intravenous iron dextran in clinical medicine. JAMA. 1980; May 2 243 (17) 1726-31
  • 17 Kulnigg S, Stoinov S, Simanenkov V, Dudar LV, Karnafel W, Garcia LC et al. A novel intravenous iron formulation for treatment of anemia in inflammatory bowel disease: the ferric carboxymaltose (Ferinject) randomized controlled trial. Am J Gastroenterol. 2008; May 103 (5) 1182-92
  • 18 Fishbane S, Kowalski EA. The comparative safety of intravenous iron dextran, iron saccharate, and sodium ferric gluconate. Semin Dial. 2000; Nov 13 (6) 381-4
  • 19 Fishbane S. Safety in iron management. Am J Kidney Dis. 2003; Jun 41 (5 Suppl) 18-26
  • 20 Fiechter R, Batschwaroff M, Conen D. [Anaphylactic reaction after Fe-injection]. Schweiz Rundsch Med Prax. 2005; Feb 9 94 (6) 209-12
  • 21 Geisser P. The pharmacology and safety profile of ferric carboxymaltose (Ferinject): structure/reactivity relationships of iron preparations. Port J Nephrol Hypert. 2009; 23 (1) 11-6
  • 22 Funk F, Ryle P, Canclini C, Neiser S, Geisser P. The new generation of intravenous iron: chemistry, pharmacology, and toxicology of ferric carboxymaltose. Arzneimittelforschung. 2010; 60 (6a) 345-353
  • 23 Beshara S, Sorensen J, Lubberink M, Tolmachev V, Langstrom B, Antoni G et al. Pharmacokinetics and red cell utilization of 52Fe/59Fe-labelled iron polymaltose in anaemic patients using positron emission tomography. Br J Haematol. 2003; Mar 120 (5) 853-9
  • 24 Geisser P, Rumyantsev V. Pharmacodynamics and safety of ferric carboxymaltose: a mutiple-dose study in patients with iron-deficiency anaemia secondary to a gastrointestinal disorder. Arzneimittelforschung. 2010; 60 (6a) 373-385
  • 25 Ganzoni AM. [Intravenous iron-dextran: therapeutic and experimental possibilities]. Schweiz Med Wochenschr. 1970; Feb 14 (100(7) 301-3
  • 26 Finch CA, Deubelbeiss K, Cook JD, Eschbach JW, Harker LA, Funk DD et al. Ferrokinetics in man. Medicine (Baltimore). 1970; Jan 49 (1) 17-53
  • 27 Beshara S, Lundqvist H, Sundin J, Lubberink M, Tolmachev V, Valind S et al. Pharmacokinetics and red cell utilization of iron(III) hydroxide-sucrose complex in anaemic patients: a study using positron emission tomography. Br J Haematol. 1999; Feb 104 (2) 296-302
  • 28 Danielson BG, Salmonson T, Derendorf H, Geisser P. Pharmacokinetics of iron(III)-hydroxide sucrose complex after a single intravenous dose in healthy volunteers. Arzneimittelforschung. 1996; Jun 46 (6) 615-21