throbber
CMYKP
`
`EDITORIAL
`
`Port J Nephrol Hypert 2012; 26(1): 13-24
`Advance Access publication 12 March 2012
`
`Iron isomaltoside 1000:
`a new high dose option
`for parenteral iron therapy
`
`Philip A. Kalra1, Klaus Bock2, Morten Meldal3
`1 Department of Renal Medicine, Salford Royal Hospital, Salford, United Kingdom.
`2 Department of Chemistry, University of Copenhagen, Copenhagen, Denmark.
`3 Nano Science Center, University of Copenhagen, Copenhagen, Denmark.
`
`Received for publication:
`Accepted:
`
`02/03/2012
`08/03/2012
`
` (cid:132)
`ABSTRACT
`
` (cid:132)
`INTRODUCTION
`
`Iron isomaltoside 1000 (Monofer®) is a new dex-
`tran-free parenteral iron product, currently approved
`in 22 EU countries. Iron isomaltoside 1000 consists
`of iron and a carbohydrate moiety where the iron
`is tightly bound in a matrix structure, which enables
`a controlled and slow release of iron to iron-binding
`proteins, avoiding toxicity. The carbohydrate,
`isomaltoside 1000, is a purely linear chemical struc-
`ture with low immunological activity. Due to the
`structure of iron isomaltoside 1000 and the low
`anaphylactic potential, there is no requirement for
`a test dose, and it can be administered in high
`doses with a maximum dosage of 20 mg/kg within
`30-60 minutes in one visit. Thus, iron isomaltoside
`1000 offers the broadest dosage range compared
`to other parenteral iron products on the market.
`Due to the dose flexibility, the possibility of provid-
`ing full iron correction in one single visit makes iron
`isomaltoside 1000 highly convenient for both the
`health care professional and the patient. Clinical
`studies of iron isomaltoside 1000 show that it is an
`effective and well-tolerated treatment of iron defi-
`ciency anaemia with a favourable safety profile.
`Furthermore, iron isomaltoside 1000 does not seem
`to induce hypophosphataemia.
`
`Key-Words:
`High dose; iron deficiency anaemia; iron isomaltoside;
`iron treatment.
`
`The ability to give high doses of iron is important
`in the management of iron deficiency anaemia (IDA)
`in a number of clinical conditions where demands
`for iron are high, such as chronic kidney disease
`(CKD), chronic blood loss associated with inflamma-
`tory bowel disease (IBD) or other gastrointestinal
`disease, pregnancy, and blood loss during surgery.
`Parenteral iron offers a fast iron correction, and it is
`superior to oral iron in many circumstances, especially
`in the treatment of anaemia associated with chronic
`diseases where the patients may be intolerant of
`oral iron or because the iron absorption may be
`blocked, in cases with large iron deficits as the maxi-
`mum capacity for oral iron absorption is very limited,
`or when patients are treated with erythropoiesis-
`stimulating agents (ESAs).
`
`The currently available parenteral iron preparations
`include high molecular weight iron dextran (Dexfer-
`rum®), low molecular weight iron dextran (Cosmofer®/
`Infed®), iron gluconate (Ferrlecit®), iron sucrose
`(Venofer®), ferumoxytol (Feraheme®), iron carboxy-
`maltose (Ferinject®/Injectafer®), and iron isomaltoside
`1000 (Monofer®). They are generally considered equal-
`ly efficacious, but most of them have limitations in
`dosing, administration (duration and frequency), and
`safety profile. High molecular weight iron dextran has
`been associated with an increased risk of anaphylaxis/
`anaphylactoid reactions and is not available in
`
`
`
`Nefro - 26-1 - MIOLO.indd 13Nefro - 26-1 - MIOLO.indd 13
`
`
`
`14-03-2012 15:49:0814-03-2012 15:49:08
`
` 13
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 1
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`Philip A. Kalra, Klaus Bock, Morten Meldal
`
`CMYKP
`
`Europe1-5. These side effects are significantly reduced
`with low molecular weight iron dextran1-4, but this
`still requires a test dose and has a long infusion time
`of four to six hours for larger doses6. Iron gluconate,
`iron sucrose, and ferumoxytol (only available in US
`and use limited to CKD patients) can only be admin-
`istered in moderate doses since they are limited to
`a maximum total single dose of 125 mg, 200 mg, and
`510 mg, respectively7-9. In addition, treatment with
`iron sucrose requires a test dose in Europe8, and it
`has been found associated with acutely increased
`proteinuria at 100 mg weekly infusions10,11. Iron glu-
`conate has also been found to be associated with a
`mild transient proteinuria in CKD patients11.
`
`Iron carboxymaltose does not require a test dose,
`and it can be administered in doses of 20 mg/kg up
`to a maximum of 1000 mg per infusion12. Iron carboxy-
`maltose infusion has been associated with hypophos-
`phataemia of unknown aetiology. The clinical signifi-
`cance of the hypophosphataemia is unknown13.
`
`The newest parenteral iron preparation, iron
`isomaltoside 1000 (Monofer®), was introduced in
`Europe in 2010. The ambition with iron isomaltoside
`1000 was to develop an efficacious parenteral iron
`product with a favourable safety profile without test
`dose requirement and without dose limitations in
`order to optimise dosing flexibility and user conve-
`nience. Iron isomaltoside 1000 fulfils these require-
`ments and can be administered with a maximum
`dosage of 20 mg/kg, no test dose, and within 30-60
`minutes in a single visit14. Due to the dose flexibility
`that iron isomaltoside 1000 offers, the possibility of
`providing full iron correction in a single infusion makes
`it highly convenient for both the health care profes-
`sionals and the patient. The present review describes
`the physiochemical characteristics, pharmacological,
`pharmacokinetic, and immunogenic properties, pre-
`clinical and clinical data, and cost analysis of iron
`isomaltoside 1000.
`
` (cid:132)
` PHYSIOCHEMICAL CHARACTERISTICS
`OF IRON ISOMALTOSIDE 1000
`
`Iron isomaltoside 1000 consists of iron and a car-
`bohydrate moiety. The carbohydrate isomaltoside 1000
`consists predominantly of 3-5 glucose units and origi-
`nates from isomalto-oligosaccharides produced by
`
`hydrolysis of dextran, followed by subsequent fractiona-
`tion and chemical modification to provide a product
`with the desired molecular weight distribution. Further-
`more, isomaltoside 1000 is isolated after chemical
`reduction of the reducing sugar residues to avoid com-
`plications due to redox reactions or degradation of the
`aldehyde group at the anomeric centre. The absence
`of reducing sugar prevents any complex redox reactions
`and thereby degradation of the iron complexes16. Apart
`from differences in molecular weight between dextran
`in iron dextran and isomaltoside 1000, the latter is also
`completely devoid of any branching structures as evi-
`denced by 13C and 1H NMR spectroscopic analysis and
`it does not contain any reducing sugar residues15. Thus,
`although isomaltoside 1000 is manufactured by a chemi-
`cal modification and hydrolysis of dextran, isomaltoside
`1000 is not a dextran. The chemical structure of iso-
`maltoside 1000 is very different from the dextran struc-
`ture, in which the α-(1,3) linked branches of the molecule
`are wound around the main chain α-(1,6) linked polymer
`in a tight helical arrangement15 while isomaltoside 1000
`has a purely linear oligomer structure of α-(1,6) linked
`glucopyranose residues, on average repeating 5.2 times,
`and contains no reducing sugar units.
`
`Electron microscopy16 presented the nano structure
`of iron isomaltoside 1000 as spheroidal while 13C NMR
`and associated molecular modelling have indicated
`that it is composed of a matrix structure in which the
`iron atoms are predominantly bound and dispersed in
`the matrix. From the masses of the components it can
`be calculated that there are approximately 10 iron atoms
`bound per oligosaccharide molecule. It is not yet known
`if these constitute coordinated single iron oxide moie-
`ties or small clusters of coordinated iron oxide. The
`iron isomaltoside 1000 matrix is composed of inter-
`changing strands of linear isomaltoside 1000 with iron
`atoms placed in cavities between and within the iso-
`maltoside molecules15. The matrix structure enables a
`controlled and slow release of iron which attaches to
`iron-binding proteins with little risk of free iron toxicity
`(Fig. 1). This is a unique structure and quite different
`from other iron products which are described as a pure
`iron core surrounded by a carbohydrate shell. The for-
`mation of this molecular matrix structure is possible
`due to the short, linear, and non-ionic isomaltoside
`1000 structure combined with the production technol-
`ogy for complexing iron and isomaltoside 1000.
`
`Iron is tightly bound in the iron isomaltoside 1000
`molecule; assessment of an iron isomaltoside 1000
`
`14 Port J Nephrol Hypert 2012; 26(1): 13-24
`
`
`
`Nefro - 26-1 - MIOLO.indd 14Nefro - 26-1 - MIOLO.indd 14
`
`
`
`14-03-2012 15:49:1014-03-2012 15:49:10
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 2
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`CMYKP
`
`Iron isomaltoside 1000: a new high dose option for parenteral iron therapy
`
`Figure 1
`
`Matrix structure of iron isomaltoside 1000 wich enables a controlled and slow release of iron.
`
`Comparative free iron content in high dose IV iron products
`
`Figure 2
`
`Free iron content in high dose parenteral iron products. The data is obtained without pH adjustment. The detection limit was 0.002 %. [Modified from Jahn et
`al., 2011 (Ref. 16)].
`
`solution equivalent to that administered to patients
`showed very low concentrations of free iron close to
`the detection limit of the assay (Fig. 2). A similar low
`
`concentration of free iron has been found with feru-
`moxytol, while the concentration of free iron in iron
`dextran and iron carboxymaltose solutions is significantly
`
`
`
`Nefro - 26-1 - MIOLO.indd 15Nefro - 26-1 - MIOLO.indd 15
`
`
`
`14-03-2012 15:49:1214-03-2012 15:49:12
`
`Port J Nephrol Hypert 2012; 26(1): 13-24 15
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 3
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`Philip A. Kalra, Klaus Bock, Morten Meldal
`
`CMYKP
`
`higher15. Measurement of labile iron showed that the
`newer iron products (iron carboxymaltose, ferumoxytol,
`and iron isomaltoside 1000) have low labile iron content
`when compared to the older products (iron dextran <<
`iron sucrose and iron gluconate)15.
`
` (cid:132)
` IMMUNOGENIC PROPERTIES
`OF ISOMALTOSIDE 1000
`
`Anaphylactoid/anaphylactic reactions may occur with
`all parenteral iron compounds and were seen relatively
`often with the old high molecular weight iron dextran
`products. The pathogenic mechanisms for these reactions
`is not entirely clear, but the reactions seem to occur both
`through specific and non-specific immune reactions, with
`the carbohydrate carrier playing an important role for
`these reactions16. Thus, an important aim of the develop-
`ment of iron isomaltoside 1000 was to develop a product
`with a low risk of anaphylactoid/anaphylactic reactions.
`In order to achieve this, a carbohydrate carrier with a
`low immunogenic potential was sought. In iron isomal-
`toside 1000, the carbohydrate carrier, isomaltoside 1000,
`is based on a chemical modification of oligomers known
`to prevent dextran-induced anaphylactic reactions.
`
`Since it is well known that homopolymers of glucose
`have very low immunogenic potential17, in product
`design, any residual branching units that were α-linked
`to the 3-position of the main chain were removed,
`and the reducing sugar residue was chemically trans-
`formed quantitatively to non-reducing groups.
`
`Thus, from a theoretical point of view, the immu-
`nogenic potential of iron isomaltoside 1000 is expected
`to be very low, and on this basis, the regulatory authori-
`ties decided that no test dose was required in the first
`clinical studies with iron isomaltoside 1000. These
`studies supported the theoretical rationale for low
`immunogenic activity, and iron isomaltoside 1000 was
`therefore approved for use without a test dose.
`
` (cid:132)
` PHARMACOLOGICAL AND
`PHARMACOKINETIC PROPERTIES
`OF IRON ISOMALTOSIDE 1000
`
`Following IV administration, iron isomaltoside
`1000 is rapidly taken up by the cells in the
`
`reticuloendothelial system (RES), particularly in the
`liver and spleen, from where iron is slowly released.
`The plasma half-life is 5 hours for circulating iron
`and 20 hours for total iron (bound and circulating).
`Circulating iron is removed from the plasma by cells
`of the RES which split the complex into iron and
`isomaltoside 1000. Iron is immediately bound and
`stored, mainly in ferritin. The iron replenishes hae-
`moglobin and depleted iron stores. Negligible quan-
`tities of iron are eliminated in the urine and faeces.
`Due to the size of the nanoparticles (20.5 nm), iron
`isomaltoside 1000 is not eliminated via the kidneys.
`The carbohydrate component, isomaltoside 1000,
`is either metabolised or excreted unchanged via
`the kidney14.
`
`An open-label, cross-over, single-centre study was
`performed in 12 patients (5 men/7 women) with
`inflammatory bowel disease (IBD) to assess phar-
`macokinetics18. The patients were allocated to one
`of two single-dose treatments where iron isomal-
`toside 1000 was administered as a single bolus
`dose of 100 or 200 mg with a four-week interval
`between the two doses. The dose was administered
`at a maximum of 50 mg of iron/minute. Pharma-
`cokinetic (PK) variables were analysed for total iron
`(TI), isomaltoside-bound iron (IBI), and transferrin-
`bound iron (TBI) according to a one-compartment
`model. TI and TBI were measured by the Graphite
`GFAAS system and the Advia chemistry system,
`respectively, and IBI was calculated by subtracting
`TBI from TI, assuming that no free iron was present
`and that quantities of ferritin were negligible, so
`that the only iron forms present in plasma were TI,
`TBI, and IBI. The concentration versus time relation-
`ship for IBI and TI showed first-order kinetics with
`small deviations for dose-linearity, and the PK
`parameters for IBI were close to that of TI (Table I).
`Thus, TI could be used as a marker of iron isoma-
`ltoside 1000 PK in future PK studies. Only approxi-
`mately 1 % of the doses administered were excreted
`in the urine. One of the patients was withdrawn
`after receiving a 100 mg dose because of abdominal
`pain and flushing. No serious adverse events (SAE)
`were reported18.
`
`Presently, there are several PK studies ongoing
`with higher doses of iron isomaltoside 1000 in differ-
`ent patient populations (ClinicalTrial.gov: NCT01213979,
`NCT01280240, NCT01213992, NCT01469078, and
`NCT01213680).
`
`16 Port J Nephrol Hypert 2012; 26(1): 13-24
`
`
`
`Nefro - 26-1 - MIOLO.indd 16Nefro - 26-1 - MIOLO.indd 16
`
`
`
`14-03-2012 15:49:1414-03-2012 15:49:14
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 4
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`CMYKP
`
`Iron isomaltoside 1000: a new high dose option for parenteral iron therapy
`
`Table I
`
`Geometric mean (CV in %) for PK parameters of IBI, TI, and TBI
`Isomaltoside-bound iron
`Treatment
`
`Endpoint
`
`Total iron
`Treatment
`
`Transferrin-bound iron
`Treatment
`
`auc0-end (h*μg/ml)
`auc0-inf (h*μg/ml)
`cmax (μg/ml)
`c0 (μg/ml)
`ke (1/h)
`t1/2 (h)
`vD based on c0 (l)
`
`
`
`100 mg
`809 (24)
`888 (22)
`35.6 (39)
`28.3(32)
`0.033 (12)
`20.8 (12)
`3.5 (32)
`
`200 mg
`1885 (20)
`2141 (23)
`68.6 (26)
`64.5 (29)
`0.031 (24)
`22.5 (24)
`3.1 (30)
`
`100 mg
`894 (21)
`1010 (19)
`37.3 (38)
`28.9 (32)
`0.030 (15)
`23.2 (15)
`3.5 (32)
`
`200 mg
`2017 (19)
`2319 (21)
`71.1 (26)
`66.8 (28)
`0.029 (23)
`23.5 (23)
`3.0 (28)
`
`100 mg
`83 (19)
`163 (67)
`2.1 (30)
`1.7 (37)
`0.011 (85)
`62.2 (85)
`60.6 (36)
`
`200 mg
`129 (15)
`228 (51)
`3.0 (16)
`2.9 (37)
`0.013 (87)
`53.9 (87)
`68.3 (37)
`
` (cid:132)
` EFFICACY AND SAFETY PROFILE
`OF IRON ISOMALTOSIDE 1000
`
`In the recent past, the efficacy and safety of iron
`isomaltoside 1000 in the treatment of IDA has been
`investigated in two phase III clinical studies in
`patients with either chronic kidney disease (CKD) or
`chronic heart failure (CHF)19,20. The primary endpoint
`of these studies was to establish the safety profile
`of iron isomaltoside 1000, whereas efficacy was the
`secondary endpoint. Both were open-label, non-
`comparative, multi-centre studies where the patients
`attended six visits during a study period of eight
`weeks. At the investigator’s discretion, iron isomalto-
`side 1000 was administered either as four repeated
`intravenous (IV) bolus injections with 100-200 mg
`iron per dose administered at baseline and at week
`1, 2, and 4 (the last dose could be administered as
`total remaining dose if the total calculated iron
`requirement exceeded 800 mg) or as a high single
`iron correction dose (total dose infusion (TDI)) at
`baseline. If the TDI requirement exceeded 20 mg
`iron/kg the dose was divided into two and these
`given at an interval of one week. No test dose was
`given. The total calculated iron requirement and
`administered cumulative dose in each patient were
`based on a target Hb of 130 g/L and utilising the
`Ganzoni formula that reflects body weight, the dif-
`ference between actual haemoglobin and target hae-
`moglobin, and the desired level of iron stores (com-
`monly 500 mg)21. The safety assessments consisted
`of type and frequency of adverse events (AEs) and
`SAEs, changes in vital signs (including electrocardio-
`gram (ECG)), and clinical laboratory analyses (bio-
`chemistry: s-sodium, s-potassium, s-creatinine,
`s-albumin, s-urea, s-bilirubin, and alanine aminotrans-
`ferase (ALAT), haematology: leucocytes, complete
`
`blood cell count with differentials, and platelets) 1,
`2, 4, and 8 weeks after baseline. The efficacy assess-
`ments consisted of laboratory monitoring of treat-
`ment effect on haemoglobin (Hb), transferrin satura-
`tion (TSAT), and s-ferritin levels 1, 2, 4, and 8 weeks
`after baseline. In addition, the CHF study included
`s-iron, which was monitored at the same time points,
`and a linear analogue scale assessment (LASA) qual-
`ity of life (QoL) questionnaire measuring QoL 4 and
`8 weeks after baseline. The LASA is a validated QoL
`assessment consisting of 100-mm linear analogue
`scales that measured the patient’s energy level, abil-
`ity to do daily activities, and overall QoL.
`
` (cid:132)
` IRON ISOMALTOSIDE 1000
`ADMINISTERED TO PATIENTS
`WITH CHRONIC KIDNEY DISEASE
`
`The study was conducted at 15 centres in three
`European countries (six in Denmark, seven in Sweden,
`and two in the United Kingdom). A total of 182 CKD
`patients (128 men/54 women) receiving dialysis (n =
`161) or pre-dialysis care (n = 21) were included. The
`vast majority of patients were receiving haemodialysis.
`The patients were generally on ESA treatment (n = 82
`%), and the dosage of ESA was kept constant during
`the study. Patients were either switched from an exist-
`ing parenteral iron maintenance therapy (n = 144) or
`were not currently treated with parenteral iron (n =
`38). The mean ± SD age was 63.3 ± 13.8 years (range:
`21-91 years). Patients not receiving parenteral iron
`treatment when entering the study had a baseline Hb
`of 99.1 ± 9.0 g/L and a s-ferritin of 231 ± 154 μg/L,
`and patients who switched from a parenteral iron
`maintenance regimen had a baseline Hb of 114.9 ±
`
`
`
`Nefro - 26-1 - MIOLO.indd 17Nefro - 26-1 - MIOLO.indd 17
`
`
`
`14-03-2012 15:49:1614-03-2012 15:49:16
`
`Port J Nephrol Hypert 2012; 26(1): 13-24 17
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 5
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`Philip A. Kalra, Klaus Bock, Morten Meldal
`
`CMYKP
`
`10.3 g/L and a s-ferritin of 380 ± 195 μg/L. The mean
`± SD total cumulative dose of iron per patient was
`529 ± 283 mg19. In total, 584 treatments were given
`(523 IV bolus 100 mg, 17 IV bolus 100-200 mg and
`44 TDIs) with single doses as high as 1800 mg22.
`
`Nineteen reported AEs were possibly or probably
`related to the study drug. There was no difference in
`the AE frequencies observed in patients treated with
`bolus doses or TDI. Three subjects (1.6 %) had more
`than one AE related to the study drug: one patient
`had two events of nausea, another patient had diar-
`rhoea, influenza, hyperhidrosis, low s-ferritin, and
`arthralgia, and a third patient had a haemorrhagic
`cyst in the right kidney and pruritus. Two of the AEs
`which were determined by the attending physician to
`be possibly treatment-related, fulfilled the criteria for
`SAEs. The events were sepsis with Staphylococcus
`aureus and unstable angina. Two deaths (one reported
`as due to an unknown cause and the other pneumonia)
`occurred, but these were both considered unrelated
`to the study drug. No acute anaphylactoid/anaphylactic
`or delayed allergic reactions were reported. There
`were no clinically significant changes in vital signs or
`routine safety clinical laboratory tests.
`
`Hb increased from 99.2 ± 9.0 g/L at baseline to
`111.2 g/L ± 14.7 at week 8 in patients not having
`received parenteral iron (p < 0.001) and remained stable
`in patients receiving maintenance iron therapy (114.9
`± 10.3 g/L at baseline, 117.5 ± 11.7 g/L at week 8; p =
`0.05). The mean ± SD maximal increase in Hb in the
`overall mixed CKD population was 7.9 ± 9.9 g/L (p <
`0.001). TSAT and s-ferritin also increased significantly
`from baseline to week 8 (p < 0.001). It was concluded
`that iron isomaltoside 1000 administered to CKD
`patients as repeated bolus injections or TDI without
`a test dose, was safe and well-tolerated and resulted
`in improved markers of iron status and anaemia19.
`
`mean age was 75 years (range: 61-88 years). Baseline
`Hb was 108.2 ± 7.6 g/L and s-ferritin was 180 ± 184
`μg/L. All 20 patients received a high single dose
`infusion with a mean infusion time of 59.8 minutes
`(range: 50-67 minutes) with a mean dose of 868 mg
`(range: 650-1000 mg).
`
`No study drug related AEs were reported, no
`deaths occurred, and no acute anaphylactic/anaphy-
`lactoid or delayed allergic reactions were observed.
`There were no clinically significant changes in routine
`clinical safety laboratory tests or vital signs. New
`clinically significant ECG abnormalities were observed
`on 13 occasions, but these did not indicate any new
`disease or progression of disease and could all be
`explained by the patients’ medical history.
`
`Haemoglobin was increased at every visit com-
`pared to baseline; however, the increase was non-
`significant due to the small patient population. As
`compared with baseline value, s-ferritin was signifi-
`cantly increased at all visits, while a statistical
`increase in s-iron and TSAT were observed one week
`after baseline. All QoL assessments increased sig-
`nificantly four weeks after baseline. The empirical
`mean for “energy level” increased by 49 %, “ability
`to do daily activities” increased by 38 %, and “overall
`QoL” increased by 23 %. Eight weeks after baseline,
`the scores were increased by 34 %, 20 %, and 13
`%, for each of these QoL parameters, respectively,
`but statistical significance was only reached for
`“energy level”. The authors concluded that, despite
`the uncontrolled study design and small sample size,
`iron isomaltoside 1000 was well-tolerated and
`improved QoL in patients with CHF20.
`
` (cid:132)
` HIGH DOSING OF IRON
`ISOMALTOSIDE 1000
`
` (cid:132)
` IRON ISOMALTOSIDE 1000
`ADMINISTERED TO PATIENTS
`WITH CHRONIC HEART FAILURE
`
`The study was conducted at seven centres in two
`European countries (four centres in Denmark and
`three centres in Sweden). A total of 20 CHF patients
`(10 men/10 women) with mild anaemia were included.
`None of the patients received ESA treatment. The
`
`As discussed, iron isomaltoside 1000 can be
`administered in high doses without a test dose due
`to its low immunological activity and low risk of free
`iron related toxicity. Three sub-analyses of safety
`and efficacy parameters of the patients in the CKD
`and CHF studies, who were treated with high dose
`infusion, were performed23-25.
`
`The first analysis included 19 haemodialysis patients
`with CKD and anaemia. All 19 patients received a
`
`18 Port J Nephrol Hypert 2012; 26(1): 13-24
`
`
`
`Nefro - 26-1 - MIOLO.indd 18Nefro - 26-1 - MIOLO.indd 18
`
`
`
`14-03-2012 15:49:1814-03-2012 15:49:18
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 6
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`CMYKP
`
`Iron isomaltoside 1000: a new high dose option for parenteral iron therapy
`
`high single dose infusion, with a mean dosage of
`986 mg (range: 463-1800 mg) over 30-60 minutes.
`A total of 19 AEs were reported in 9 patients (47 %),
`but none of them was considered to be related to
`the study drug by the investigator. No acute anaphy-
`lactoid/anaphylactic or delayed allergic reactions were
`observed, and there were no significant changes in
`safety clinical laboratory tests or vital signs. Efficacy
`markers of IDA improved significantly23.
`
`The second analysis included 21 pre-dialysis
`patients with CKD and anaemia. One SAE that was
`considered to be treatment related was observed.
`The event was angina pectoris in an 80-year-old CKD
`patient with a medical history of angina. It occurred
`10-11 days after the patient had received 1400 mg
`iron isomaltoside 1000. The medical history of the
`patient and the time delay in the occurrence of the
`event made the relationship of the SAE to iron
`isomaltoside 1000 quite unlikely. No acute anaphy-
`lactoid/anaphylactic or delayed allergic reactions were
`observed, and there were no significant changes in
`safety clinical laboratory tests or vital signs. Efficacy
`markers of IDA improved significantly25.
`
`The third analysis consisted of the above 40 CKD
`patients aggregated with the 20 CHF patients. A total
`of 58 out of 60 patients had one single high dose
`infusion, and only two CKD patients required two
`divided doses in order to fulfil their iron needs. The
`mean dosage was 975 mg (range: 462-1800 mg) in
`the CKD patients and 868 mg (range: 650-1000 mg)
`in the CHF patients. One treatment related SAE was
`observed, which was the event of angina pectoris
`discussed above. No acute anaphylactoid/anaphy-
`lactic or delayed allergic reactions were observed,
`and there were no significant changes in safety clini-
`cal laboratory tests or vital signs. Efficacy markers
`of IDA improved significantly24.
`
`In conclusion, iron isomaltoside 1000 administered
`as high doses to CKD and CHF patients was safe, well
`tolerated, and effective in improving markers of IDA.
`
` (cid:132)
` IRON ISOMALTOSIDE 1000
`AND NEPHROTOXICITY
`
`It has been suggested that parenteral iron may
`have a direct toxic effect on renal tubular cells which
`
`could cause renal phosphate wasting26,27. In 2004,
`Zager and colleagues reported a study comparing
`the nephrotoxicity of iron sucrose, iron gluconate,
`iron dextran, and iron isomaltoside 1000 over a broad
`dosage range (0, 30 to 1000 μg iron/mL)28. The iron
`preparations were added to isolated mouse proximal
`tubule segments and cultured proximal tubular
`human kidney cells. Cell injury was assessed by
`lactate dehydrogenase release, adenosine triphos-
`phate reductions, cell cytochrome c efflux, and/or
`electron microscopy. The iron preparations evoked
`in vitro toxicity and up to 30-fold differences in
`severity were observed. The highest toxicity was
`observed in iron sucrose and the lowest in iron
`dextran and iron isomaltoside 1000 (iron sucrose >
`iron gluconate >> iron dextran = iron isomaltoside
`1000). The large differences may be explained by
`the difference in capacities of the irons to gain intra-
`cellular access. According to transmission electron
`microscopy (TEM) studies, the nanostructure of iron
`dextran and iron isomaltoside 1000 are similarly large
`and globular while iron sucrose and iron gluconate,
`in addition to being more soluble formulations, differ
`significantly in structure and display elongated and
`smaller nanostructures15.
`
`Similar data was found with in vivo correlates of
`iron toxicity which included increases in renal malon-
`dialdehyde, renal ferritin, and heme oxygenase-1
`expression in mice. These changes also appeared to
`parallel in vivo glomerular iron uptake (seen with
`iron sucrose and iron gluconate, but not with iron
`dextran and iron isomaltoside 1000)28.
`
` (cid:132)
` IRON ISOMALTOSIDE 1000
`AND S-PHOSPHATE
`
`As some parenteral iron therapies have been found
`to be associated with hypophosphataemia13,26,27,29-32,
`the effect of iron isomaltoside 1000 on s-phosphate
`is being measured in several ongoing clinical studies,
`and interim analyses of s-phosphate data have been
`performed as part of the protocols. At the current
`time, these interim analyses included 25 oncology
`patients and 50 patients with non-dialysis dependent
`chronic kidney disease (NDD-CKD) treated with iron
`isomaltoside 1000 (data on file, Pharmacosmos A/S,
`ClinicalTrial.gov: NCT01145638 (oncology study) and
`NCT01102413 (NDD-CKD study)).
`
`
`
`Nefro - 26-1 - MIOLO.indd 19Nefro - 26-1 - MIOLO.indd 19
`
`
`
`14-03-2012 15:49:1914-03-2012 15:49:19
`
`Port J Nephrol Hypert 2012; 26(1): 13-24 19
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 7
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`Philip A. Kalra, Klaus Bock, Morten Meldal
`
`CMYKP
`
`The phosphate analyses are part of two phase III,
`prospective, open-label, randomised, comparative
`studies. A total of 350 patients with a diagnosis of
`non-myeloid cancer and 350 NDD-CDK patients with
`renal-related anaemia are being randomised 2:1 to
`either IV iron isomaltoside 1000 (group A) or oral
`iron sulphate (group B). The patients in group A are
`equally divided into two sub-groups (A1 and A2).
`Group A1 are treated with IV iron isomaltoside, where
`the full iron replacement dose is administered as
`infusions of maximum 1000 mg iron isomaltoside
`1000 over 15 minutes until full replacement dose is
`achieved. Group A2 are treated with IV bolus injec-
`tions of 500 mg iron isomaltoside 1000 over 2 min-
`utes, administered once per week until full replace-
`ment dose is achieved. Group B are treated with
`200 mg oral iron sulphate daily for 8-12 weeks. For
`the individual patient, the duration of the study is
`8-10 weeks in the NDD-CKD study and 24-26 weeks
`in the oncology study. S-phosphate is measured
`prior to iron administration and at every visit. At
`baseline in the oncology study, s-phosphate was 4.0
`
`± 0.9 mg/dL in group A1 and A2, and 3.6 ± 0.6 mg/
`dL in group B, and in the NDD-CKD study, s-phos-
`phate was 4.5 ± 0.9 mg/dL in group A1 and A2, and
`4.3 ± 0.9 mg/dL in group B.
`
`At interim analysis there was no significant
`change in the s-phosphate levels in any of the IV
`treatment groups in either the oncology or NDD-CKD
`patients. Mean (95 % CI) values of s-phosphate for
`the three treatment arms are shown in Fig. 3 (oncol-
`ogy study) and Fig. 4 (NDD-CKD study). In other
`studies, s-phosphate levels below 2 mg/dL have been
`considered an indicator for hypophosphatemia13,32.
`In the iron isomaltoside 1000 studies, 3 out of 75
`patients experienced a decrease in s-phosphate with
`a value slightly below 2 mg/dL post treatment. The
`first patient was a NDD-CKD patient treated with an
`infusion (1000 mg of iron isomaltoside 1000). The
`patient had a s-phosphate level of 1.8 mg/dL three
`weeks after baseline. The second patient was an oncol-
`ogy patient treated with an infusion (also 1000 mg).
`This patient had a s-phosphate level of 1.9 mg/dL at
`
`Figure 3
`
`Mean (95 % CI) phosphate values in oncology patients during the 24 week study period. Group A1 was treated with iron isomaltoside 1000 infusion, group A2
`was treated with iron isomaltoside 1000 bolus injection, and group B was treated with oral iron sulphate (data on file, Pharmacosmos A/S).
`
`20 Port J Nephrol Hypert 2012; 26(1): 13-24
`
`
`
`Nefro - 26-1 - MIOLO.indd 20Nefro - 26-1 - MIOLO.indd 20
`
`
`
`14-03-2012 15:49:2114-03-2012 15:49:21
`
`Luitpold Pharmaceuticals, Inc., Ex. 2013, p. 8
`Pharmacosmos A/S v. Luitpold Pharmaceuticals, Inc., IPR2015-01495
`
`

`
`CMYKP
`
`Iron isomaltoside 1000: a new high dose option for parenteral iron therapy
`
`Figure 4
`
`Mean (95 % CI) phosphate values in NDD-CKD patients during the 8 week study period. Group A1 was treated with iron isomaltoside 1000 infusion, group A2
`was treated with iron isomaltoside 1000 bolus injection, and group B was treated with oral iron sulphate (data on file, Pharmacosmos A/S).
`
`one and four weeks after baseline. The third patient
`was an oncology patient treated with two bolus injec-
`tions (500 mg + 250 mg of iron isomaltoside 1000).
`The patient already had a low s-phosphate level at
`the screening visit (2.0 mg/dL), and four weeks after
`baseline the s-phosphate level was 1.9 mg/dL. All
`three patients had a s-phosphate which normalised
`at the following visit. No adverse drug reaction was
`reported in the t

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