throbber
Drugs (2015) 75:101–127
`DOI 10.1007/s40265-014-0332-3
`
`A D I S D R U G E V A L U A T I O N
`
`Ferric Carboxymaltose: A Review of Its Use in Iron Deficiency
`
`Gillian M. Keating
`
`Published online: 27 November 2014
`Ó Springer International Publishing Switzerland 2014
`


`Abstract Ferric carboxymaltose (Ferinject
`)
`, Injectafer
`is an intravenous iron preparation approved in numerous
`countries for the treatment of iron deficiency. A single high
`dose of ferric carboxymaltose (up to 750 mg of iron in the
`US and 1,000 mg of iron in the EU) can be infused in a
`short time frame (15 min). Consequently, fewer doses of
`ferric carboxymaltose may be needed to replenish iron
`stores compared with some other intravenous iron prepa-
`rations (e.g. iron sucrose). Ferric carboxymaltose improved
`self-reported patient global assessment, New York Heart
`Association functional class and exercise capacity in
`patients with chronic heart failure and iron deficiency in
`two randomized, placebo-controlled trials (FAIR-HF and
`CONFIRM-HF). In other randomized controlled trials,
`ferric carboxymaltose replenished iron stores and corrected
`anaemia in various populations with iron-deficiency anae-
`mia,
`including patients with chronic kidney disease,
`inflammatory bowel disease or heavy uterine bleeding,
`postpartum iron-deficiency anaemia and perioperative
`anaemia. Intravenous ferric carboxymaltose was generally
`well tolerated, with a low risk of hypersensitivity reactions.
`
`The manuscript was reviewed by: E. Bisbe, Department of
`Anaesthesia, Hospital Mar-Esperanc¸a, IMIM Hospital del Mar
`Medical Research Institute, Barcelona, Spain; B. Bokemeyer,
`Gastroenterology Practice, Minden, Germany; M. Lainscak, General
`Hospital Celje, Division of Cardiology, Celje, Slovenia; I.C.
`Macdougall, Renal Unit, King’s College Hospital, London, UK.
`
`G. M. Keating (&)
`Springer, Private Bag 65901, Mairangi Bay 0754, Auckland,
`New Zealand
`e-mail: demail@springer.com
`
`It was generally better tolerated than oral ferrous sulfate,
`mainly reflecting a lower incidence of gastrointestinal
`adverse effects. The most common laboratory abnormality
`seen in ferric carboxymaltose recipients was transient,
`asymptomatic hypophosphataemia. The higher acquisition
`cost of ferric carboxymaltose appeared to be offset by
`lower costs for other items, with the potential for cost
`savings.
`In conclusion,
`ferric carboxymaltose is an
`important option for the treatment of iron deficiency.
`
`carboxymaltose
`Ferric
`summary
`
`in iron deficiency:
`
`a
`
`A single high dose of ferric carboxymaltose can be
`infused in a short time frame (15 min), rapidly
`replenishing iron stores
`
`Compared with some other intravenous preparations,
`fewer doses may be needed to replenish iron stores
`
`Showed improvement in self-reported patient global
`assessment, New York Heart Association functional
`class and exercise capacity in chronic heart failure
`and iron deficiency
`
`Replenished iron stores and corrected anaemia in
`various populations with iron-deficiency anaemia
`
`Generally well tolerated, with a low risk of
`hypersensitivity reactions, and better tolerated than
`oral ferrous sulfate
`
`Associated with transient, asymptomatic
`hypophosphataemia
`
`Potential for cost savings
`
`Pharmacosmos, Exh. 1057, p. 1
`
`

`
`102
`
`1 Introduction
`
`Globally, iron deficiency is the most commonly occurring
`nutritional deficiency and the most common cause of
`anaemia [1, 2]. Iron deficiency is a common feature of
`various chronic diseases [e.g. chronic heart failure (CHF),
`chronic kidney disease] and its aetiology is often multi-
`factorial [3]. Iron deficiency may be associated with poor
`nutrition, increased utilization of iron (e.g. during preg-
`nancy), blood loss [e.g. heavy uterine bleeding, blood loss
`associated with gastrointestinal (GI) disorders, surgical
`blood loss], chronic inflammation or
`impaired iron
`absorption [e.g. in inflammatory bowel disease (IBD)] [3–
`5]. Although particularly prevalent in developing countries,
`anaemia remains a significant problem in developed
`countries [2, 5]. Diagnostic criteria for anaemia vary
`between studies [3], although WHO defines anaemia as a
`haemoglobin level of \130 g/L in men, \120 g/L in non-
`pregnant women and \110 g/L in pregnant women [2].
`Iron metabolism is a complex process and the 25-amino
`acid peptide hepcidin is the key regulator of iron homeo-
`stasis [6]. Hepcidin is upregulated in chronic inflammation
`(as seen in cancer or autoimmune conditions), which
`contributes to the development of anaemia of chronic dis-
`ease [6]. Other factors affecting the regulation of hepcidin
`include iron availability, erythropoiesis and hypoxia [6].
`Iron deficiency may be absolute or functional [7].
`Absolute iron deficiency occurs when iron stores are so low
`that no iron is available for the production of haemoglobin
`[7]. The negative iron balance compromises erythropoiesis
`resulting in iron-deficiency anaemia [4, 8]. In functional
`iron deficiency, body iron stores are normal or increased,
`but there is a failure of these stores to release iron rapidly
`enough to support the demands of the bone marrow [3, 7].
`Iron supplementation is used to correct anaemia and
`replenish iron stores in patients with iron-deficiency

`anaemia [3]. Ferric carboxymaltose (Ferinject
`, Injecta-

`)
`is an intravenous iron preparation approved in
`fer
`numerous countries for the treatment of iron deficiency.
`This article reviews the clinical efficacy, tolerability and
`safety of ferric carboxymaltose in iron deficiency, as well
`as summarizing its pharmacological properties and results
`of pharmacoeconomic analyses. Throughout this article,
`the dose of ferric carboxymaltose and other iron prepara-
`tions is expressed in milligrams of iron.
`
`2 Pharmacodynamic Properties
`
`The pharmacodynamic properties of ferric carboxymaltose
`have been reviewed previously [9]; this section provides a
`brief overview.
`
`G. M. Keating
`
`Ferric carboxymaltose is formulated as a colloidal
`solution of polynuclear iron(III) oxyhydroxide stabilized
`by the carbohydrate polymer carboxymaltose [10, 11]. The
`ferric carboxymaltose solution has isotonic osmolarity and
`a pH of 4.5–7.0 and the complex has a molecular mass of
`&150 kDa [10–12]. Ferric carboxymaltose is designed to
`provide controlled delivery of iron to the macrophages of
`the reticuloendothelial system in the liver, spleen and bone
`marrow, with subsequent delivery to the transport protein
`transferrin, without
`large amounts of ionic iron being
`released into the serum [10, 11].
`The stability of the ferric carboxymaltose complex
`permits the administration of high doses of iron [11]. In
`mice, the 50 % lethal dose (LD50) of intravenous ferric
`carboxymaltose was [1,000 mg iron/kg bodyweight [10],
`compared with LD50 values of 230 mg iron/kg for oral iron
`sulfate, [200 mg iron/kg for intravenous iron sucrose and
`[2,500 mg iron/kg for intravenous iron dextran [13].
`In patients with anaemia receiving radiolabelled ferric
`carboxymaltose, maximum red cell uptake of 59Fe of
`61–99 % was seen after 16–24 days [14]. Intravenous
`ferric carboxymaltose transiently increased total serum iron
`concentrations (see Sect. 3) [15]. Results of clinical trials
`demonstrating the beneficial effects of ferric carboxymal-
`tose on haemoglobin levels, serum ferritin levels and
`transferrin saturation are discussed in Sect. 4.
`Ferric carboxymaltose was associated with transient,
`asymptomatic hypophosphataemia in patients with iron
`deficiency (see Sect. 5). The mechanisms underlying this
`hypophosphataemia are unclear, although one possible
`mechanism is an effect on the phosphate-regulating peptide
`hormone fibroblast growth factor 23 (FGF23). In women
`with heavy uterine bleeding and iron-deficiency anaemia,
`serum levels of intact FGF23 increased from baseline to a
`significantly (p \ 0.05) greater extent with intravenous
`ferric carboxymaltose than with intravenous iron dextran,
`with no significant between-group difference in the change
`from baseline in plasma levels of C-terminal FGF23 [16].
`The transient increase in intact FGF23 was accompanied
`by a transient reduction in serum phosphate levels [16].
`Ferric carboxymaltose lowered elevated platelet counts
`and normalized platelet activation in patients with IBD [17,
`18]. For example, in patients with IBD and secondary
`thrombocytosis in the ThromboVIT trial, mean platelet
`counts were significantly (p \ 0.01) lower at week 6 in
`patients receiving ferric carboxymaltose 500–1,500 mg
`than in placebo recipients, although there was no signifi-
`cant difference in the proportion of ferric carboxymaltose
`versus placebo recipients achieving a reduction in platelet
`count of C25 % [18]. Significant (p \ 0.05) reductions
`from baseline in platelet activation were also seen with
`ferric carboxymaltose [18].
`
`Pharmacosmos, Exh. 1057, p. 2
`
`

`
`Ferric Carboxymaltose: A Review
`
`103
`
`Prolonged exposure to high levels of non-transferrin
`bound iron (as seen in untreated patients with iron overload
`disorders) may trigger oxidative stress in organs such as the
`liver, heart and pancreas [19]. Thus, there has been concern
`that iron compounds that release large amounts of ionic
`iron into the circulation may potentiate oxidative stress [20,
`21]. However, ferric carboxymaltose appears to be asso-
`ciated with a relatively low risk of oxidative stress,
`reflecting the fact that iron is predominantly deposited in
`the reticuloendothelial system [10, 11]. For example, sig-
`nificant (p \ 0.01) increases in oxidative stress and pro-
`inflammatory markers were seen in the liver, heart and
`kidneys of rats administered low-molecular-weight (LMW)
`or high-molecular-weight (HMW) iron dextran or sodium
`ferric gluconate (ferrous gluconate), compared with rats
`administered ferric carboxymaltose or iron sucrose [22]. In
`addition, ferric carboxymaltose had no effect on plasma
`levels of intercellular adhesion molecule (ICAM) or vas-
`cular adhesion molecule or proinflammatory markers in
`patients with nondialysis-dependent chronic kidney disease
`[23].
`However, reactive oxygen species production, ICAM-1
`expression and apoptosis were significantly (p \ 0.005 vs.
`control) increased from baseline when mononuclear cells
`from haemodialysis patients or healthy volunteers were
`cultured in vitro with ferric carboxymaltose, iron sucrose,
`iron dextran or sodium ferric gluconate, with significantly
`(p \ 0.005) greater effects seen in mononuclear cells from
`haemodialysis patients than in mononuclear cells from
`healthy volunteers [20].
`Baseline levels of erythrocyte glutathione peroxidase
`(eGPx)
`(an antioxidant
`enzyme) were
`significantly
`(p = 0.01) higher among patients with nondialysis-depen-
`dent chronic kidney disease and iron-deficiency anaemia
`who responded to a single dose of ferric carboxymaltose
`1,000 mg than in nonresponders [24]. Multivariate analysis
`revealed that eGPx levels independently predicted response
`to ferric carboxymaltose [24].
`
`3 Pharmacokinetic Properties
`
`Total serum iron concentrations increased rapidly following
`the intravenous administration of single ferric carboxy-
`maltose doses of 100, 500, 800 or 1,000 mg of iron to
`patients with mild iron-deficiency anaemia; the 100 mg
`dose was administered as an intravenous bolus and the 500,
`800 and 1,000 mg doses were administered as 15-min
`intravenous infusions [15]. Following single doses of ferric
`carboxymaltose 100, 500, 800 and 1,000 mg, geometric
`mean maximum serum iron concentrations of 37, 156, 319
`and 331 lg/mL, respectively, were reached in a mean time
`of 0.3, 0.3, 1.0 and 1.2 h, respectively [15]. The geometric
`
`mean area under the serum concentration-time curve (AUC)
`from time zero to 24 h was 333 lg h/mL with ferric carb-
`oxymaltose 100 mg, and the AUC from time zero to 72 h
`was 2,345, 5,171 and 6,277 lg h/mL with ferric carboxy-
`maltose 500, 800 and 1,000 mg, respectively [15]. Repeated
`administration of ferric carboxymaltose 500 or 1,000 mg
`did not result in accumulation of iron in serum [25].
`The iron in ferric carboxymaltose had a volume of
`distribution of &3 L [15, 26]. Ferric carboxymaltose did
`not cross the placenta in an in vitro perfusion model [27].
`The iron in ferric carboxymaltose was rapidly cleared
`from plasma [26], with positron emission tomography
`demonstrating that the major portion of a radiolabelled
`injected iron dose was distributed to the bone marrow, with
`uptake also seen in the liver and spleen [14]. Following
`single doses of
`ferric carboxymaltose 100–1,000 mg,
`geometric mean clearance was 2.6–4.3 mL/min [15].
`In vitro experiments showed that the carbohydrate moiety
`underwent hydrolysis to oligoglucose units (e.g. maltotri-
`ose, maltose and glucose) [11].
`Renal elimination of iron following administration of
`ferric carboxymaltose was negligible [26, 28]. Following
`single doses of ferric carboxymaltose 100–1,000 mg, the
`geometric mean terminal
`elimination half-life was
`7.4–12.1 h and the geometric mean residence time was
`11.2–16.6 h [15].
`When intravenous ferric carboxymaltose was adminis-
`tered to women with postpartum iron-deficiency anaemia,
`mean iron concentrations in breast milk increased from
`0.500 mg iron/kg breast milk at baseline to a maximum
`1.447 mg/kg at 24-h postdose [29]. Neither mother nor
`infants had evidence of adverse events associated with the
`administration of ferric carboxymaltose to breast-feeding
`mothers [29].
`
`4 Therapeutic Efficacy in Iron Deficiency
`
`The main focus of this section is the results of randomized
`controlled trials examining the efficacy of ferric carboxy-
`maltose in iron deficiency. Where relevant, results of
`studies in real-world settings are also briefly discussed,
`with a focus on prospective data.
`
`4.1 In Chronic Heart Failure
`
`The efficacy of ferric carboxymaltose in patients with CHF
`and iron deficiency (with or without anaemia) was exam-
`ined in the randomized, double-blind, placebo-controlled,
`multinational FAIR-HF [30] and CONFIRM-HF [31] trials.
`The trials were of 26 [30] or 52 [31] weeks’ duration.
`Inclusion criteria included New York Heart Association
`(NYHA) class II or III CHF [30, 31],
`left ventricular
`
`Pharmacosmos, Exh. 1057, p. 3
`
`

`
`104
`
`G. M. Keating
`
`ejection fraction (LVEF) of B40 % (for NYHA class II
`patients) or B45 % (for NYHA class III patients) [30] or
`B45 % [31], a haemoglobin level of 95–135 g/L [30] or
`\150 g/L [31], iron deficiency (i.e. serum ferritin level of
`\100 ng/mL, or 100–299 ng/mL with a transferrin satu-
`ration of \20 %) [30, 31] and elevated natriuretic peptide
`levels (brain natriuretic peptide level of [100 pg/mL and/
`or N-terminal-pro-brain natriuretic peptide
`level of
`[400 pg/mL) [31].
`In FAIR-HF, patients received intravenous ferric carb-
`oxymaltose 200 mg once weekly until iron repletion was
`achieved (calculated total iron requirement based on the
`Ganzoni formula) and then every 4 weeks during the
`maintenance phase; the maintenance phase started at week
`8 or 12, depending on the iron repletion dose [30]. In
`CONFIRM-HF, patients received ferric carboxymaltose at
`baseline and, if necessary, at week 6 to correct iron defi-
`ciency (cumulative dose of 500–2,000 mg with the dose
`based on bodyweight and haemoglobin levels), followed by
`maintenance doses of 500 mg at weeks 12, 24 and 36 if
`iron deficiency was still present [31].
`Among ferric carboxymaltose and placebo recipients at
`baseline, 17.4 versus 18.7 % in FAIR-HF [30] and 53.3
`versus 60.3 % in CONFIRM-HF [31] were NYHA class
`II, 82.6 versus 81.3 % [30] and 46.7 versus 39.7 % [31]
`were NYHA class III, mean LVEF was 31.9 versus
`33.0 % [30] and 37.1 versus 36.5 % [31], mean haemo-
`globin was 119 versus 119 g/L [30] and 124 versus
`124 g/L [31], mean serum ferritin was 53 versus 60 ng/
`mL [30] and 57 versus 57 ng/mL [31] and mean trans-
`ferrin saturation was 17.7 versus 16.7 % [30] and 20.2
`versus 18.2 % [31].
`The primary endpoints were self-reported patient global
`assessment and NYHA functional class at week 24 [30] and
`the change in 6-min walk test distance from baseline to
`week 24 [31]. Efficacy was assessed in the full analysis set
`[30, 31].
`
`4.1.1 FAIR-HF Trial
`
`Ferric carboxymaltose was beneficial in patients with CHF
`and iron deficiency with or without anaemia [30]. The odds
`ratios (ORs) for improvements in self-reported patient
`global assessment and NYHA functional class both sig-
`nificantly favoured ferric carboxymaltose versus placebo
`recipients at 24 weeks (Table 1). At week 24, 50 % of
`ferric carboxymaltose recipients and 28 % of placebo
`recipients reported much or moderate improvement (based
`on self-reported patient global assessment), and 47 % of
`ferric carboxymaltose recipients and 30 % of placebo
`recipients were NYHA class I or II [30]. ORs for both
`improvement in self-reported patient global assessment and
`NYHA functional class also significantly (p \ 0.001)
`
`favoured ferric carboxymaltose versus placebo recipients at
`weeks 4 and 12 [30].
`Ferric carboxymaltose had a consistent treatment effect
`in patients with anaemia (haemoglobin level B120 g/L)
`and in those without anaemia [32]. ORs for improved self-
`reported patient global assessment were 2.48 (95 % CI
`1.49–4.14; p \ 0.001) in patients with anaemia and 2.60
`(95 % CI 1.55–4.35; p \ 0.001) in patients without anae-
`mia, and ORs for improvement by one NYHA functional
`class were 1.90 (95 % CI 1.06–3.40; p = 0.03) in patients
`with anaemia and 3.39 (95 % CI 1.70–6.75; p \ 0.001) in
`patients without anaemia [32]. It should be noted that
`haemoglobin levels
`at week 24 were
`significantly
`(p \ 0.001) higher in ferric carboxymaltose than in pla-
`cebo recipients in the overall population and in the sub-
`group of patients with anaemia at baseline, with no
`significant between-group difference in the subgroup of
`patients without anaemia at baseline [30].
`The mean distance achieved on the 6-min walk test was
`significantly (p \ 0.001) longer with ferric carboxymaltose
`than with placebo at weeks 4 (294 vs. 269 m), 12 (312 vs.
`272 m) and 24 (313 vs. 277 m) (mean baseline values were
`274 vs. 269 m) [30]. At week 24, the increase in 6-min
`walk distance was significantly correlated with a reduction
`in red cell distribution width (r = -0.25; p \ 0.0001),
`according to results of a post hoc analysis [33]. Although
`red cell distribution width initially increased in ferric
`carboxymaltose recipients, it was significantly (p \ 0.05)
`lower with ferric carboxymaltose than with placebo at
`week 24 [33].
`Ferric carboxymaltose improved health-related quality
`of life (HR-QOL) in patients with CHF and iron deficiency
`[34]. The mean changes from baseline in European Quality
`of Life-5 Dimensions (EQ-5D) visual analogue scale
`(VAS) scores significantly (p \ 0.001) favoured ferric
`carboxymaltose versus placebo recipients at weeks 4 (6.0
`vs. 0.8), 12 (7.9 vs. 2.4) and 24 (9.1 vs. 3.4) (mean baseline
`values were 54 in both treatment groups), as did the mean
`changes from baseline in the Kansas City Cardiomyopathy
`Questionnaire (KCCQ) overall summary scores at weeks 4
`(9.4 vs. 3.5), 12 (12.2 vs. 4.6) and 24 (12.8 vs. 6.2) (mean
`baseline values were 52 and 53). Ferric carboxymaltose
`improved HR-QOL in patients with and without anaemia
`[34]. Multivariate analysis showed that intravenous iron
`therapy, lower NYHA class and better 6-min walk test
`results were associated with higher HR-QOL, whereas a
`history of stroke and reduced renal function were associ-
`ated with lower HR-QOL [35].
`Ferric carboxymaltose improved renal function in iron-
`deficient patients with CHF, according to an additional
`analysis of FAIR-HF (available as a poster) [36]. At week
`24, the change from baseline in estimated glomerular fil-
`tration rate significantly favoured ferric carboxymaltose
`
`Pharmacosmos, Exh. 1057, p. 4
`
`

`
`Ferric Carboxymaltose: A Review
`
`105
`
`Table 1 Efficacy of ferric carboxymaltose in patients with chronic heart failure and iron deficiency. Shown are the primary endpoints in the
`randomized, double-blind, multinational FAIR-HF [30] and CONFIRM-HF [31] trials
`
`Study (study name)
`
`Endpoint
`
`Anker et al. [30, 125] (FAIR-HF)
`
`Self-reported patient global assessment at 24 weeksa
`
`Much improved (% of pts)
`
`Moderately improved (% of pts)
`
`A little improved (% of pts)
`
`Unchanged (% of pts)
`
`A little, moderately or much worse (% of pts)
`
`Dead (% of pts)
`
`Odds ratio for improvement (95 % CI)
`NYHA functional class at 24 weeksb
`
`Class I (% of pts)
`
`Class II (% of pts)
`
`Class III (% of pts)
`
`Class IV (% of pts)
`
`Dead (% of pts)
`
`FCM
`
`16
`
`34
`
`26
`
`18
`
`3
`
`2
`
`2.51 (1.75–3.61)**
`
`6
`
`41
`
`50
`
`1
`
`2
`
`PL
`
`10
`
`17
`
`28
`
`35
`
`7
`
`3
`
`1
`
`29
`
`65
`
`3
`
`3
`
`Odds ratio for improvement by one class (95 % CI)
`Ponikowski et al. [31, 126] (CONFIRM-HF) LSM change from baseline to week 24 in 6MWT distancec
`(m) [mean baseline value; m]
`
`2.40 (1.55–3.71)**
`
`?18* [288]
`
`-16 [302]
`
`6MWT 6-min walk test, FCM ferric carboxymaltose, LSM least squares mean, NYHA New York Heart Association, PL placebo, pts patients
`* p \ 0.01, ** p \ 0.001 vs. PL
`a 292 FCM recipients and 149 PL recipients were evaluable for self-reported patient global assessment
`b 294 FCM recipients and 150 PL recipients were evaluable for NYHA functional class
`c 150 FCM recipients and 151 PL recipients were in the full analysis set
`
`(between-group difference
`versus placebo recipients
`4.0 mL/min/1.73 m2; p = 0.017) [36].
`
`4.1.2 CONFIRM-HF Trial
`
`A significant improvement from baseline to week 24 in the
`6-min walk test distance was seen with ferric carboxy-
`maltose versus placebo in patients with CHF and iron
`deficiency in the CONFIRM-HF trial (Table 1), with a
`least squares mean between-group difference of 33 m
`(p = 0.002) [31]. The improvement in 6-min walk test
`distance was maintained at later time points; the difference
`between ferric carboxymaltose and placebo recipients was
`?42 m at week 36 and ?36 m at week 52 (both
`p \ 0.001). No significant interaction was seen in terms of
`the treatment effect in patients with anaemia (haemoglobin
`\120 g/L) or without anaemia (haemoglobin C120 g/L)
`(p-value for interaction 0.15) [31].
`Ferric carboxymaltose recipients were significantly
`(p \ 0.05) more likely than placebo recipients to have
`improvements in self-reported patient global assessment at
`weeks 12, 24, 36 and 52 and in NYHA class at weeks 24,
`36 and 52 [31]. Significant (p \ 0.05) improvements were
`seen with ferric carboxymaltose versus placebo in terms of
`the least squares mean change in fatigue scores at weeks
`
`12, 24, 36 and 52 and for KCCQ scores at weeks 12, 36 and
`52. A significant (p = 0.002) between-group difference
`favouring ferric carboxymaltose was seen for the least
`squares mean change in EQ-5D VAS score at week 36,
`with no significant between-group difference seen at other
`time points [31].
`significantly (p \ 0.001)
`Between-group differences
`favoured ferric carboxymaltose versus placebo recipients
`in terms of ferritin levels at week 24 (265 ng/mL) and
`week 52 (200 ng/mL), transferrin saturation at week 24
`(8.9 %) and week 52 (5.7 %) and haemoglobin levels at
`week 24 (6 g/L) and week 52 (10 g/L) [31].
`
`4.2 In Chronic Kidney Disease
`
`Several randomized, open-label, multicentre trials exam-
`ined the efficacy of intravenous ferric carboxymaltose in
`patients with chronic kidney disease and iron-deficiency
`anaemia [37–41]. Patients had nondialysis-dependent
`chronic kidney disease [37–39, 41] and/or were undergoing
`haemodialysis [40, 41]. Patients with nondialysis-depen-
`dent chronic kidney disease had haemoglobin levels of
`B110 g/L [39] or B115 g/L [37, 41] or at least one hae-
`moglobin value of 90–110 g/L within 4 weeks of ran-
`domization [38], and a ferritin level of B100 ng/mL [37,
`
`Pharmacosmos, Exh. 1057, p. 5
`
`

`
`106
`
`G. M. Keating
`
`38],\200 ng/mL with transferrin saturation of B20 % [38]
`or B300 ng/mL with transferrin saturation of B25 % [39]
`or B30 % [37, 41]. Patients with chronic kidney disease
`who were undergoing haemodialysis had a haemoglobin
`level of B115 g/L [40] or B125 g/L [41] and a ferritin
`level of \200 ng/mL or transferrin saturation of \20 %
`[40] or a ferritin level of \500 ng/mL and transferrin sat-
`uration of B30 % [41]. Patients were followed for 30 days
`[41], 120 days [37], 8 weeks [39, 40] or 56 weeks [38].
`Ferric carboxymaltose was compared with oral ferrous
`sulfate [38, 39], intravenous iron sucrose [37, 40] or stan-
`dard medical care [41]. Stable treatment with erythropoi-
`esis-stimulating agents was permitted in four trials [37, 39–
`41], with no erythropoiesis-stimulating agent therapy per-
`mitted in a fifth trial [38].
`Primary endpoints included the mean change from
`baseline to the highest observed haemoglobin level at any
`time up to day 56 [37], the proportion of patients achieving
`an increase in haemoglobin of C10 g/L [39, 40] and the
`time to initiation of other anaemia management or occur-
`rence of a haemoglobin trigger [38]; safety was the primary
`endpoint in one study, with efficacy endpoints considered
`exploratory secondary endpoints [41]. One trial is only
`available as an abstract [40].
`Ferric carboxymaltose was more effective than oral
`ferrous sulfate in patients with nondialysis-dependent
`chronic kidney disease and iron-deficiency anaemia [38,
`39]. In the FIND-CKD trial, patients whose ferric carb-
`oxymaltose dose was adjusted to maintain a higher ferritin
`level had a significantly (p = 0.026) lower risk of initiating
`other anaemia management or experiencing a haemoglobin
`trigger than ferrous sulfate recipients [hazard ratio (HR)
`0.65; 95 % CI 0.44–0.95], with no significant difference
`seen between patients whose ferric carboxymaltose dose
`was adjusted to maintain a higher versus a lower ferritin
`level (Table 2) [38]. High-ferritin ferric carboxymaltose
`recipients were significantly more likely than low-ferritin
`ferric carboxymaltose or
`ferrous sulfate recipients to
`achieve an increase in haemoglobin level of C10 g/L
`(Table 2). A significantly greater
`least squares mean
`increase from baseline to month 12 in haemoglobin level
`was seen in high-ferritin ferric carboxymaltose recipients
`than in ferrous sulfate recipients, with no significant dif-
`ference seen between low-ferritin ferric carboxymaltose
`recipients and ferrous sulfate recipients (Table 2) [38]. The
`least squares mean change from baseline to month 12 in
`serum ferritin levels was significantly greater with high-
`ferritin ferric carboxymaltose than with ferrous sulfate
`(?451 vs. ?137 ng/mL; p \ 0.001), and was significantly
`smaller with low-ferritin ferric carboxymaltose than with
`ferrous sulfate (?81 vs. ?137 ng/mL; p \ 0.001). The
`least squares mean change from baseline to month 12 in
`transferrin saturation did not significantly differ between
`
`high-ferritin ferric carboxymaltose and ferrous sulfate
`recipients (?15.8 vs. ?13.8 %), although it was signifi-
`cantly smaller with low-ferritin ferric carboxymaltose than
`with ferrous sulfate (?8.5 vs. ?13.8 %; p = 0.001). The
`mean number of ferric carboxymaltose doses needed to
`achieve and maintain ferritin levels in the high- and low-
`ferritin groups was 4.0 and 4.8, respectively [38].
`In a second trial, significantly more ferric carboxymal-
`tose than ferrous sulfate recipients achieved an increase in
`haemoglobin level of C10 g/L and the mean increase from
`baseline to the highest observed haemoglobin level was
`significantly greater with ferric carboxymaltose than with
`ferrous sulfate (Table 2) [39]. The mean changes from
`baseline to study end in serum ferritin levels (?358.8 vs.
`?178.4 ng/mL) and transferrin saturation (?12.1 vs.
`?7.0 %) were significantly (p \ 0.001) greater with ferric
`carboxymaltose than with ferrous sulfate. Only one dose of
`ferric carboxymaltose was administered to 58 % of patients
`[39].
`In the REPAIR-IDA trial, ferric carboxymaltose was
`noninferior to iron sucrose in terms of the mean change
`from baseline to the highest observed haemoglobin level up
`to day 56 in patients with nondialysis-dependent chronic
`kidney disease (Table 2)
`[37]. The 95 % CI
`for
`the
`between-group difference was entirely above zero, sug-
`gesting superiority of ferric carboxymaltose versus iron
`sucrose, although assessment of superiority was not pre-
`specified. Ferric carboxymaltose was also noninferior to
`iron sucrose in terms of the proportion of patients achiev-
`ing an increase in haemoglobin level of C10 g/L up to day
`56; the 95 % CI for the between-group difference was
`entirely above zero (Table 2) [37]. The mean changes from
`baseline to the highest observed value in serum ferritin and
`transferrin saturation were significantly greater with ferric
`carboxymaltose than with iron sucrose (p-values not sta-
`ted). Two ferric carboxymaltose infusions were adminis-
`tered to 96.8 % of patients, and five iron sucrose infusions
`were administered to 91.6 % of patients [37].
`In patients with iron-deficiency anaemia undergoing
`haemodialysis, an increase in haemoglobin level of C10 g/
`L had occurred in 46.4 % of ferric carboxymaltose recip-
`ients and in 37.2 % of iron sucrose recipients at week 4
`(Table 2) [40]. The mean change from baseline in hae-
`moglobin levels is shown in Table 2. At study end, mean
`serum ferritin levels were 465.3 ng/mL in ferric carboxy-
`maltose recipients and 397.7 ng/mL in iron sucrose recip-
`ients (baseline levels of 90.4 and 93.1 ng/mL in the
`corresponding treatment groups) [40].
`In patients with chronic kidney disease (either nondial-
`ysis-dependent or undergoing haemodialysis), no signifi-
`cant difference was seen between patients receiving ferric
`carboxymaltose and those receiving standard medical care
`in terms of
`the change from baseline to day 30 in
`
`Pharmacosmos, Exh. 1057, p. 6
`
`

`
`Ferric Carboxymaltose: A Review
`
`107
`
`Table 2 Efficacy of ferric carboxymaltose in patients with chronic kidney disease and iron-deficiency anaemia. Results of randomized, open-
`label, multicentre trials; patients were followed for 30 days [41], 120 days [37], 8 weeks [39, 40] or 56 weeks [38]
`
`Study
`
`Treatment
`
`No. of
`ptsa
`
`Hb
`
`Meanc
`baseline
`value (g/L)
`
`Meanc change
`from baselined
`(g/L)
`
`Increase of C10 g/Le
`(% of pts)
`
`Pts (%) starting other
`anaemia management
`or having an Hb triggerb
`
`Pts with nondialysis-dependent chronic kidney disease
`IV FCMf
`IV ISCi
`IV high-ferritin
`FCMj
`IV low-ferritin
`FCMj
`Oral FSk
`IV FCMl
`Oral FSk
`Pts with chronic kidney disease undergoing haemodialysis
`46.4g
`Schaefer et al. [40]m
`FCMn
`37.2g
`ISCn
`Pts with chronic kidney disease who were nondialysis dependent or undergoing haemodialysis
`FCMo
`SMCp
`
`Onken et al. [37, 127]
`(REPAIR-IDA)
`
`Macdougall et al. [38, 128]
`(FIND-CKD)
`
`Qunibi et al. [39]
`
`Charytan et al. [41]
`
`1,276
`
`1,285
`
`103.1
`
`103.2
`
`153
`
`101
`
`152
`
`102
`
`308
`
`147
`
`103
`
`97
`
`86
`
`249
`
`249
`
`102
`
`101
`
`100
`
`93
`
`93.4
`
`105.9
`
`104.5
`
`?11.3g,h
`?9.2g
`?14*
`
`?9
`
`?10
`
`?131**
`
`?0.83
`
`?12.7
`
`?9.6
`
`?4.9
`
`?3.3
`
`48.6h
`41.0
`56.9** 
`
`34.2
`
`32.1
`60.4**g
`34.7g
`
`25.7
`
`22.1
`
`23.5*g
`
`32.2g
`
`31.8g
`
`FCM ferric carboxymaltose, FS ferrous sulfate, Hb haemoglobin, ISC iron sucrose, IV intravenous, pts patients, SMC standard medical care
`* p \ 0.05, ** p B 0.001 vs. FS;  p \ 0.001 vs. low-ferritin FCM
`a No. of pts in the modified intent-to-treat [37–39, 41] or per-protocol [40] populations
`b Other anaemia management included erythropoiesis-stimulating agents, blood transfusion or use of an alternative iron therapy, and occurrence
`of an Hb trigger was defined as two consecutive Hb values \100 g/L on or after week 8, without an increase of C5 g/L between the two
`measurements
`c Mean [37, 39–41] or least squares mean values [38]
`d Change from baseline to highest observed Hb at any time up to day 56 [37, 39], or change from baseline to day 30 [41], week 8 [40] or month
`12 [38]
`e Increase of C10 g/L at any time in the study period [37, 39, 41], at week 4 [40] or prior to starting other anaemia management [38]
`f FCM was administered on days 0 and 7 (each dose a maximum of 750 mg)
`g Primary endpoint
`h Noninferiority shown for FCM vs. ICS
`i Five doses of ISC 200 mg were administered between days 0 and 14
`j High-ferritin FCM comprised FCM 1,000 mg followed every 4 weeks by FCM 500 mg if ferritin was 200–400 ng/mL or FCM 1,000 mg if
`ferritin was \200 ng/mL. Low-ferritin FCM comprised FCM 200 mg if ferritin was \100 ng/mL followed every 4 weeks by FCM 200 mg if
`ferritin was \100 ng/mL
`k FS 100 mg twice daily to week 52 [38] or 65 mg three times daily to day 56 [39]
`l First maximum dose of 1,000 mg; second and third maximum doses of 500 mg administered on approximately day 17 and/or day 31 if the
`transferrin saturation remained \30 % and ferritin remained \500 ng/mL
`m Available as an abstract
`n FCM or ISC 200 mg two or three times weekly (administered into the venous line of the dialyser) until the total cumulative dose for each pt
`was reached
`o Nondialysis-dependent pts received a maximum IV dose of FCM 1,000 mg and pts undergoing haemodialysis received FCM 200 mg directly
`into the venous line of the dialyser
`p SMC comprised IV iron (63 % of pts), oral iron (30 %) or no iron treatment (8 %)
`
`haemoglobin levels (

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket