43
REVIEW Iron Sucrose: A Wealth of Experience in Treating Iron Deficiency Iain C. Macdougall . Josep Comin-Colet . Christian Breymann . Donat R. Spahn . Ioannis E. Koutroubakis Received: February 18, 2020 / Published online: April 15, 2020 Ó The Author(s) 2020 ABSTRACT Iron deficiency and iron-deficiency anemia are associated with increased morbidity and mor- tality in a wide range of conditions. In many patient populations, this can be treated effec- tively with oral iron supplementation; but in patients who are unable to take or who do not respond to oral iron therapy, intravenous iron administration is recommended. Furthermore, in certain conditions, such as end-stage kidney disease, chronic heart failure, and inflammatory bowel disease, intravenous iron administration has become first-line treatment. One of the first available intravenous iron preparations is iron sucrose (Venofer Ò ), a nanomedicine that has been used clinically since 1949. Treatment with iron sucrose is particularly beneficial owing to its ability to rapidly increase hemoglobin, fer- ritin, and transferrin saturation levels, with an acceptable safety profile. Recently, important new data relating to the use of iron sucrose, including the findings from the landmark PIVOTAL trial in patients with end-stage kidney disease, have been reported. Several years ago, a number of iron sucrose similars became avail- able, although there have been concerns about the clinical appropriateness of substituting the original iron sucrose with an iron sucrose simi- lar because of differences in efficacy and safety. This is a result of the complex and unique physicochemical properties of nanomedicines such as iron sucrose, which make copying the molecule difficult and problematic. In this review, we summarize the evidence accumu- lated during 70 years of clinical experience with iron sucrose in terms of efficacy, safety, and cost-effectiveness. Keywords: Anemia; Iron deficiency; Iron sucrose; Nanomedicine; Venofer Digital Features To view digital features for this article go to https://doi.org/10.6084/m9.figshare.12030552. I. C. Macdougall (&) Department of Renal Medicine, King’s College Hospital, London, UK e-mail: [email protected] J. Comin-Colet Department of Cardiology, Bellvitge University Hospital and IDIBELL, University of Barcelona, Barcelona, Spain C. Breymann Perinatal and Gynecology Center Seefeld Zu ¨rich, Zurich, Switzerland D. R. Spahn Institute of Anaesthesiology, University of Zurich and University Hospital Zu ¨ rich, Zurich, Switzerland I. E. Koutroubakis Department of Gastroenterology, University Hospital Heraklion, Crete, Greece Adv Ther (2020) 37:1960–2002 https://doi.org/10.1007/s12325-020-01323-z

Iron Sucrose: A Wealth of Experience in Treating Iron Deficiency · 2020. 5. 4. · REVIEW Iron Sucrose: A Wealth of Experience in Treating Iron Deficiency Iain C. Macdougall. Josep

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • REVIEW

    Iron Sucrose: A Wealth of Experience in Treating IronDeficiency

    Iain C. Macdougall . Josep Comin-Colet . Christian Breymann .

    Donat R. Spahn . Ioannis E. Koutroubakis

    Received: February 18, 2020 / Published online: April 15, 2020� The Author(s) 2020

    ABSTRACT

    Iron deficiency and iron-deficiency anemia areassociated with increased morbidity and mor-tality in a wide range of conditions. In manypatient populations, this can be treated effec-tively with oral iron supplementation; but inpatients who are unable to take or who do notrespond to oral iron therapy, intravenous ironadministration is recommended. Furthermore,

    in certain conditions, such as end-stage kidneydisease, chronic heart failure, and inflammatorybowel disease, intravenous iron administrationhas become first-line treatment. One of the firstavailable intravenous iron preparations is ironsucrose (Venofer�), a nanomedicine that hasbeen used clinically since 1949. Treatment withiron sucrose is particularly beneficial owing toits ability to rapidly increase hemoglobin, fer-ritin, and transferrin saturation levels, with anacceptable safety profile. Recently, importantnew data relating to the use of iron sucrose,including the findings from the landmarkPIVOTAL trial in patients with end-stage kidneydisease, have been reported. Several years ago, anumber of iron sucrose similars became avail-able, although there have been concerns aboutthe clinical appropriateness of substituting theoriginal iron sucrose with an iron sucrose simi-lar because of differences in efficacy and safety.This is a result of the complex and uniquephysicochemical properties of nanomedicinessuch as iron sucrose, which make copying themolecule difficult and problematic. In thisreview, we summarize the evidence accumu-lated during 70 years of clinical experience withiron sucrose in terms of efficacy, safety, andcost-effectiveness.

    Keywords: Anemia; Iron deficiency; Ironsucrose; Nanomedicine; Venofer

    Digital Features To view digital features for this articlego to https://doi.org/10.6084/m9.figshare.12030552.

    I. C. Macdougall (&)Department of Renal Medicine, King’s CollegeHospital, London, UKe-mail: [email protected]

    J. Comin-ColetDepartment of Cardiology, Bellvitge UniversityHospital and IDIBELL, University of Barcelona,Barcelona, Spain

    C. BreymannPerinatal and Gynecology Center Seefeld Zürich,Zurich, Switzerland

    D. R. SpahnInstitute of Anaesthesiology, University of Zurichand University Hospital Zürich, Zurich, Switzerland

    I. E. KoutroubakisDepartment of Gastroenterology, UniversityHospital Heraklion, Crete, Greece

    Adv Ther (2020) 37:1960–2002

    https://doi.org/10.1007/s12325-020-01323-z

    https://doi.org/10.6084/m9.figshare.12030552http://crossmark.crossref.org/dialog/?doi=10.1007/s12325-020-01323-z&domain=pdfhttps://doi.org/10.1007/s12325-020-01323-z

  • Key Summary Points

    Intravenously administered (IV) iron isimportant for individuals who are unableto tolerate oral iron therapy, who are non-compliant with oral treatment, or inwhom oral preparations are not effective.

    One of the first IV iron preparations to bemanufactured is iron sucrose (Venofer�;Vifor Pharma), which became available forclinical use over 70 years ago in 1949. It isthe most commonly used IV iron therapyworldwide, with clinical experienceencompassing 25 million patient-years.

    Many studies across a broad range oftherapy areas consistently demonstratethat iron sucrose is able to correct irondeficiency, in addition to promotingerythropoiesis and subsequently reducingthe required doses of erythropoiesis-stimulating agents (ESAs). Not only is ironsucrose effective, but it is also welltolerated and rapidly increasesbioavailable iron supplies.

    Since the last review focused on ironsucrose was published in 2014, reassuringclinical data have continued to becomeavailable on iron sucrose from a numberof studies. Most notably, the recentlypublished PIVOTAL trial in patients withend-stage kidney disease compared theefficacy and safety of high- versus low-dose iron sucrose.

    Generic substitution of iron sucrosesimilar for iron sucrose should not beassumed to provide therapeuticequivalence, as the complexphysicochemical properties of the originalagent are extremely difficult to duplicateand studies have not been able toconclusively demonstrate that ironsucrose similars have safety and efficacyequivalent to that of iron sucrose.

    INTRODUCTION

    Iron plays an important role in key cellularmetabolic pathways in addition to its role inheme synthesis and subsequent oxygen trans-port [1]. Iron deficiency and iron-deficiencyanemia typically occur as a consequence of asevere underlying condition, such as inflam-matory bowel disease (IBD), gastrointestinal(GI) bleeding, chronic heart failure (CHF),chronic kidney disease (CKD), or cancer [2–9].Heavy menstrual bleeding and pregnancy mayalso result in iron deficiency [10, 11]. Effectivetreatment of iron deficiency/iron-deficiencyanemia is important to relieve symptoms,including extreme fatigue and tiredness, as wellas to alleviate the adverse impact on quality oflife [11–16]. Furthermore, anemia is a riskamplifier for mortality and morbidity inpatients with heart disease [17] and those withCKD [18–20]. Iron deficiency, even in theabsence of anemia, can be debilitating and canexacerbate any underlying chronic disease,leading to increased morbidity and mortality[21, 22]; for example, iron deficiency can bepredictive of negative disease outcomes and hasbeen associated with a worse prognosis in CHF[23, 24]. Additionally, in patients with irondeficiency undergoing cardiac surgery, mortal-ity is elevated threefold, irrespective of thepresence or absence of anemia [25]. Preopera-tive anemia, which is most commonly causedby iron deficiency, is also associated with anincreased risk of poor outcomes after surgery[26, 27]. Iron deficiency and iron-deficiencyanemia generate considerable financial burdensdue to additional medical costs and loss ofproductivity [28].

    Iron administration is recommended for themanagement of iron deficiency/iron-deficiencyanemia associated with a variety of conditions[2–4, 7–10, 29, 30]. The Network for Advance-ment of Transfusion Alternatives recommendsthat iron deficiency is treated before using ery-thropoiesis-stimulating agents (ESAs) inpatients with anemia [31, 32]. This is alsoadvocated in guidelines for the treatment ofanemia in CKD [33, 34]. Corrective strategies tonormalize iron levels and hemoglobin (Hb) are

    Adv Ther (2020) 37:1960–2002 1961

  • recommended as part of perioperative care tooptimize outcomes in patients undergoing sur-gery [35]. In patients lacking a response to, orunable to tolerate, oral iron therapy, treatmentwith intravenously administered (IV) ironshould be considered [9]. There are also cir-cumstances in which IV iron is recommendedfor use as first-line treatment, including cases ofsevere iron-deficiency anemia [2–4] and whenrapid correction of Hb levels is warranted (e.g.,when less than 6 weeks prior to surgery [35]), orin certain patient groups with iron deficiency/iron-deficiency anemia such as those with CHF[3, 4, 30], hemodialysis-dependent CKD[3, 4, 8], severe IBD, or cancer [7, 29]. Advan-tages of parenteral iron therapy include (1)similar or superior effectiveness, often with amore rapid response [36, 37]; (2) avoidance ofpoor or unreliable intestinal absorption, whichis particularly problematic in patients withincreased hepcidin levels associated withinflammatory conditions [37]; and (3) improvedtolerability, as the GI side effects of oral irontherapy are avoided [38]. Several IV iron carbo-hydrate preparations are available (Table 1).

    One of the first IV iron preparations to bemanufactured is iron sucrose (Venofer�; ViforPharma), which became available for clinicaluse over 70 years ago, in 1949. Indeed, likeaspirin, it is one of the oldest therapeutic agentsstill being used widely today. The active ingre-dient of Venofer is an iron(III) hydroxide–su-crose complex, and the product is nowregistered in 91 countries worldwide. Indeed, itis the most commonly used IV iron formulationworldwide, with clinical experience encom-passing 25 million patient-years [39].

    The aim of this article is to provide a com-prehensive overview of Venofer, focusingmainly on the characteristics, pharmacokinet-ics, efficacy, safety, and limitations associatedwith this iron preparation. While IV Venoferhas a long history, there are a number of reasonswhy a review of its therapeutic use is timely.Firstly, important new data on the use of thisiron sucrose preparation have recently becomeavailable. A large open-label, blinded-endpointstudy in 2141 patients undergoing hemodialysis(PIVOTAL study) has recently demonstrated theclinical benefits of a proactive high-dose iron

    regimen of iron sucrose (Venofer) comparedwith a reactive low-dose iron regimen, includ-ing a lower incidence of death, heart attack,stroke, and hospitalization for heart failure, aswell as reduced ESA dose requirements and useof blood transfusions [40]. Secondly, variousiron sucrose similars have been developed andintroduced as generic alternatives to Venofer.However, several recent studies have suggestedthat the therapeutic equivalence of Venofer andfollow-on products cannot be assumed, giventhat these are complex nanomedicine entitiesthat may have clinically relevant differences intheir physicochemical properties.

    In order to identify peer review publicationsof interest, we searched PubMed using the terms‘‘iron sucrose’’ and filtered results by publicationdate (limited to the last 25 years), article type(clinical trials and reviews), human species, andEnglish language. This review article is based onpreviously conducted studies and does notcontain unpublished data from any studies withhuman participants or animals performed byany of the authors.

    PHARMACOKINETICSAND PHARMACODYNAMICS

    Venofer is rapidly cleared from the serum ofhealthy subjects, with a terminal half-life of5.3 ± 1.6 h and total body clearance of1.23 ± 0.22 L/h (20.5 ± 3.7 mL/min) [41]. Fol-lowing IV administration of iron sucrose intopatients with anemia, iron is rapidly taken upby the liver, spleen, and bone marrow [42]. Themajority (97%) of injected iron is utilized for redblood cell (RBC) synthesis in these patients, andserum ferritin and transferrin saturation (TSAT)increase within 24 h and 1 week, respectively[42]. Furthermore, less than 5% of the overallelimination of Venofer in healthy individualscan be attributed to undesirable renal elimina-tion, due to a relatively large molecular mass(greater than 18 kDa) [21].

    1962 Adv Ther (2020) 37:1960–2002

  • Table1

    Overview

    ofcharacteristicsof

    approved

    IViron

    form

    ulations.Adapted

    from

    Muñ

    ozandMartı́n-Montañez.Reprinted

    byperm

    ission

    ofthepublisher

    (Taylor&

    FrancisLtd,h

    ttps://w

    ww.tand

    fonline.com)[175]

    Iron

    glucon

    ate

    Iron

    sucrose

    LMWID

    Ferric

    carboxym

    altose

    Iron

    isom

    altoside

    1000

    Ferumoxytol

    Brand

    name

    Ferrlecit�

    Venofer�

    Cosmofer�

    Ferinject�,

    Injectafer�

    Monofer�

    FeraHem

    e�

    Manufacturer

    Sanofi-Aventis

    Vifo

    rPh

    arma

    Pharmacosmos

    Vifo

    rPh

    arma,

    American

    Regent

    Pharmacosmos

    AMAG

    Pharmaceuticals

    Carbohydrateshell

    Gluconate

    (monosaccharide)

    Sucrose

    (disaccharide)

    Dextran

    (branched

    polysaccharide)

    Carboxymaltose

    (branched

    polysaccharide)

    Isom

    altoside

    (linear

    oligosaccharide)

    Polyglucosesorbitol

    (branched

    polysaccharide)

    Molecular

    weightmeasuredby

    manufacturer,kD

    a

    289–

    440

    34–6

    0165

    150

    150

    750

    Initialdistribution

    volume,L

    63.4

    3.5

    3.5

    3.4

    3.16

    Terminalhalf-life(dosein

    mgFe)

    [176]

    1.42

    h(125)

    5.3h(100)

    27–3

    0h

    (500–2

    000)

    7.4/9.4h(100/

    1000)

    20.8/22.5h(100/

    200)

    14.7h(316)

    Labile

    iron,%

    ofdose

    3.3

    3.4

    1.9

    0.6

    10.9

    Directiron

    donation

    to

    transferrin,

    %of

    dose

    5–6

    4–5

    1–2

    1–2

    \1

    \1

    Iron

    content,mg/mL

    12.5

    2050

    50100

    30

    Maxim

    alsingledose,m

    g

    (minim

    umadministration

    time)a[176,1

    77]

    125(10–

    60min)

    200 (10–

    30min)

    20mg/kg

    BW

    (4–6

    h)

    1000

    (15min)

    20mg/kg

    BW

    (1h)

    510(15min)

    BW

    body

    weight,IV

    intravenously

    administered,

    LMWID

    lowmolecular

    weightiron

    dextran

    aMostcommon

    maxim

    aldose

    andcorrespond

    ingminim

    aladministrationtime;theexactposology

    may

    vary

    betweenmarkets.Please

    referto

    localprescribing

    inform

    ation

    Adv Ther (2020) 37:1960–2002 1963

    https://www.tandfonline.com

  • Efficacy of Iron Sucrose in VariousConditions

    The efficacy of iron sucrose has been demon-strated in a wide range of conditions that causeor are associated with iron deficiency and iron-deficiency anemia, as summarized below. Acrossthese various conditions, iron sucrose treatmentis associated with improved iron status (ferritin,TSAT); increased Hb levels, with or without ESAtherapy; and improved health-related quality oflife and clinical outcomes. When used in com-bination with ESA therapy, iron sucrose sub-stantially reduces ESA dose requirements.

    Chronic Kidney DiseaseAs summarized in Table 2, studies have consis-tently reported that iron sucrose is an effectivetreatment for iron deficiency/iron-deficiencyanemia in patients with CKD, leading toincreased Hb, serum ferritin, and TSAT inpatients regardless of whether they are under-going hemodialysis or receiving ESAs[40, 43–54].

    In adults undergoing maintenancehemodialysis, differing regimens, includingcontinuous and intermittent administration ofiron sucrose, have been examined[40, 43, 46–48, 50, 55]. While each of thesestudies has demonstrated benefits of ironsucrose in maintenance hemodialysis, the mostconclusive results are reported in the recentPIVOTAL study, a landmark trial that is alreadychanging clinical practice and will almost cer-tainly impact on future clinical guidelines.PIVOTAL was a multicenter, open-label trialwith blinded-endpoint evaluation in 2141hemodialysis patients; it is the largest prospec-tive, controlled clinical trial of IV iron inpatients with CKD [40]. In the PIVOTAL trial,patients were randomized into two groups: onereceiving high-dose iron sucrose proactively(400 mg monthly; with temporary discontinu-ation of treatment if safety cutoffs of ferritin700 lg/L or TSAT of 40% were exceeded) andthe other group receiving low-dose iron sucroseadministered reactively (0–400 mg monthly,adjusted to achieve minimum targets for ferritinand TSAT of 200 lg/L and 20%, respectively)

    [40]. Importantly, within the context of a rig-orous study design, this study showed that therisk of cardiovascular adverse events (AEs),infections, hospitalization, and death was loweror similar with high-dose iron sucrose comparedwith low-dose iron sucrose.

    High-dose iron sucrose was superior to low-dose iron sucrose with respect to the primarycomposite endpoint of non-fatal myocardialinfarction, non-fatal stroke, hospitalization forheart failure, or death (p\0.001 for non-infe-riority; p = 0.04 for superiority) [40]. The effectof high-dose iron sucrose on the primary end-point was consistent across all of the prespeci-fied subgroups (duration of dialysis treatment,diabetes status, and vascular access). In addi-tion, proactive administration of high-dose ironappeared to protect against recurrent cardio-vascular events. An additional benefit of high-dose iron sucrose was the subsequent reductionin ESA dose required by patients in this groupand the reduced likelihood of requiring bloodtransfusions [40].

    Relative to the low-dose strategy, the high-dose approach to iron sucrose administrationwas not associated with any safety signals.There was no difference in vascular accessthrombosis, or in hospitalizations between thedosing groups [40]. Additionally, infection rateswere virtually identical between the high-doseand low-dose iron sucrose groups, suggesting noassociation between iron dose, ferritin/TSAT,and the risk of infection [40, 56]. This is reas-suring because it has been previously suggestedthat IV iron administration may increase therisk of infection [57].

    Studies comparing iron sucrose with oraliron administration consistently show ironsucrose to be more effective in patients withCKD, with significantly better outcomes in Hb,hematocrit, serum ferritin, and TSAT, as well asreduced recombinant human erythropoietin(rhEPO) dose requirements [48, 51].

    Randomized trials have indicated that ironsucrose has comparable efficacy to other IV ironformulations in CKD; for example, ferumoxytoland iron sucrose have similar efficacy inpatients with CKD, including in patientsundergoing hemodialysis [55, 58]. An addi-tional trial suggested superior efficacy of iron

    1964 Adv Ther (2020) 37:1960–2002

  • Table2

    Publishedclinicalstudiesusingiron

    sucrosein

    chronickidn

    eydisease

    Stud

    yInclusioncriteria

    Patients,n

    Patient

    dialysisstatus

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Studiesofiron

    sucrose

    Macdougall

    etal.

    (2019)

    [40]

    ESR

    D

    TSA

    T\

    30%

    SF\

    400ng/m

    L

    High-dose

    IS?

    ESA

    :1093

    Low

    -dose

    IS?

    ESA

    :1048

    Hem

    odialysisB

    12months

    priorto

    screening

    400mg,monthly

    0–400mg,monthly

    2.1years(m

    edian

    follow-up)

    Significantlyfewer

    events(non-fatalMI,non-fatal

    stroke,h

    ospitalizationforheartfailure,ordeath)

    withhigh-doseIS

    versus

    low-doseIS

    (29.3%

    vs.

    32.3%;p\

    0.001[non-in

    feriority];p=0.04

    [superiority])

    Nodifferencesbetweengroups

    inSA

    Es,

    hospitalization,

    infectionrate,o

    rvascular

    access

    thrombosis

    Haddad

    etal.

    (2009)

    [43]

    Intoleranceto

    iron

    dextran

    IS?

    ESA

    :15

    Hem

    odialysis

    100mgq.w.

    Significant

    hematocritincrease

    Nohypersensitivity

    reactionsor

    effectson

    intradialyticbloodpressure

    Mircescu

    etal.

    (2006)

    [44]

    Hb\

    11g/dL

    SF\

    200ng/m

    L

    IS:60

    Nodialysis

    200mg,monthly,

    1year

    Significant,continu

    ous,andprogressiveHb

    increase

    Noworsening

    ofrenalfunction,b

    lood

    pressure

    changes,or

    SAEs

    Tagboto

    etal.

    (2008)

    [45]

    Hb\

    11.5g/dL

    IS:82

    Nodialysis

    49

    200mg,q.w.

    Hbincrease

    in74%

    ofpatients

    Nospecificsafety

    results

    reported

    Denget

    al.

    (2017)

    [46]

    RLS

    TSA

    T\

    20%

    SF\

    200ng/m

    L

    IS:16

    Placebo:

    16

    Hem

    odialysis

    100mg,t.i.w.(1000

    mg

    total)

    2weeks

    afterlastinjection,

    decrease

    inIRLSscore

    wassignificantlygreaterforIS

    versus

    placebo,and

    increasesin

    SF,T

    SAT,and

    Hbweresignificantly

    greaterforIS

    versus

    placebo

    NoAEswereobserved

    Adv Ther (2020) 37:1960–2002 1965

  • Table2

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Patient

    dialysisstatus

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Wan

    and

    Zhang

    (2018)

    [47]

    Hb10.0–1

    3.0g/

    dL

    SF100–

    500ng/

    mL

    iPTH\

    800pg/

    mL

    Continu

    ous

    IS?

    ESA

    :17

    Interm

    ittent

    IS?

    ESA

    :17

    Hem

    odialysis

    100mgeverysession

    (1000mgtotal;

    completed

    in1month)

    100mgq.w.(1000

    mg

    total;completed

    in3months)

    SFlevelsincreasedsignificantlyin

    both

    groups

    Hblevelsweresimilarbetweengroups,b

    utinterm

    ittent

    ISsignificantlyreducedHb

    variability

    versus

    continuous

    IS

    NoAEswereobserved

    Mitsopoulos

    etal.

    (2020)

    [54]

    Hb9–

    11.5g/dL

    TSA

    T\

    45%

    SF\

    500ng/m

    L

    IS?

    ESA

    :18

    Stableperitonealdialysis

    200mgloadingdose,

    followed

    by100mg

    monthlyfor

    5months

    HbandSF

    increasedsignificantlyfrom

    baselin

    e

    ESA

    dose

    was

    reducedin

    fivepatientsand

    discontinu

    edin

    one

    Nopatientsexperiencedanyside

    effectsrelatedto

    IS

    Com

    parativestu

    diesofiron

    sucroseversus

    oral

    iron

    form

    ulations

    Liand

    Wang

    (2008)

    [48]

    Hb6.0–

    9.0g/dL

    TSA

    T\

    30%

    SF\

    500ng/m

    L

    IS?

    ESA

    :70

    FeS?

    ESA

    :66

    Hem

    odialysis

    100mgb.i.w

    .thenq.w.

    200mgFeSt.i.d.,

    12weeks

    Significant

    Hbincrease

    versus

    baselin

    ein

    both

    groups

    andwithIS

    versus

    FeS

    NoAEswithIS,3

    3.3%

    GIsymptom

    swithFeS

    Van

    Wyck

    etal.

    (2005)

    [49]

    HbB

    11g/dL

    TSA

    TB

    25%

    SFB

    300ng/m

    L

    IS?

    ESA

    :91

    FeS?

    ESA

    :91

    Nodialysis

    29

    500or

    59

    200mg,14

    days

    65mgt.i.d.,56

    days

    Significantlybetter

    Hbresponse

    (DHbC

    1.0g/

    dL)andincrease

    withIS

    versus

    FeS

    Dysgeusiamostprom

    inentIS-related

    GIcomplaint

    (6.6%)

    Fewer

    GIside

    effectswithIS

    200mg(11.5%

    )and

    IS500mg(3.3%)versus

    FeS(17.6%

    )

    Liand

    Wang

    (2008)

    [51]

    Hb6.0–

    9.0g/dL

    TSA

    T\

    30%

    SF\

    500ng/m

    L

    IS?

    ESA

    :26

    FeS?

    ESA

    :20

    Peritonealdialysis

    200mgq.w.o

    ver

    4weeks

    then

    q.2.w.

    200mgFeSt.i.d.,

    8weeks

    Significant

    Hbincrease

    versus

    baselin

    ein

    both

    groups

    andwithIS

    versus

    FeS

    NoAEswithIS,4

    0%GIsymptom

    swithFeS

    1966 Adv Ther (2020) 37:1960–2002

  • Table2

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Patient

    dialysisstatus

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Agarwal

    etal.

    (2015)

    [52]

    Hb\

    12g/dL

    TSA

    T\

    25%

    SF\

    100ng/m

    L

    IS±

    ESA

    :69

    FeS±

    ESA

    :67

    GFR

    [20

    andB

    60mL/

    min/1.73m

    2(no

    hemodialysis)

    IS200mg,weeks

    0,2,

    4,6,

    8

    FeS325mgt.i.d.,

    8weeks

    24months

    Increasesin

    Hbsimilarbetweengroups

    SFwas

    significantlyhigher

    withIS

    versus

    FeSonly

    from

    baselin

    eto

    6months

    ISwasassociated

    withgreaterrisk

    ofinfections

    and

    cardiovascular

    complications

    versus

    FeS

    GIAEsweremorecommon

    withFeS,whereasgout

    was

    morefrequent

    intheIS

    group

    Com

    parativestu

    diesofiron

    sucroseversus

    otherIV

    iron

    form

    ulations

    Bhand

    ari

    etal.

    (2015)

    [50]

    Hb9.5–

    12.5g/dL

    TSA

    T\

    35%

    SF\

    800ng/m

    L

    IS?

    ESA

    :117

    IIso

    ?ESA

    :234

    Hem

    odialysis

    ISaccordingto

    local

    packageinsert/

    guidelines

    IIso

    500mg,single

    bolus

    IIso

    (splitdose)100mg

    (baseline)

    ?200mg

    (weeks

    2and4)

    6weeks

    (follow-up)

    Nosignificant

    difference

    inHbcontrolbetween

    groups

    Increasesin

    SFfrom

    baselin

    eto

    weeks

    1,2,

    and4

    weresignificantlygreaterforIIso

    versus

    IS

    Frequency,type,and

    severity

    ofAEsweresimilar

    betweengroups

    Macdougall

    etal.

    (2014)

    [58]

    Hb\

    11.0

    and

    C7.0g/L

    TSA

    T\

    30%

    IS:82

    FeruM:80

    Ondialysisandnoton

    dialysis

    200mg9

    5within

    14days(non-dialysis)

    100mgfor10

    consecutivesessions

    (dialysis29

    510mg

    within5days)

    5weeks

    (follow-up)

    Increasesin

    Hbsimilarbetweengroups

    AEprofilessimilarbetweengroups

    Adv Ther (2020) 37:1960–2002 1967

  • Table2

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Patient

    dialysisstatus

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Macdougall

    etal.

    (2019)

    [55]

    Hb\

    11.5g/dL

    TSA

    T\

    30%

    SFB

    800ng/m

    L

    IS:97

    FeruM:196

    Hem

    odialysis

    100mgfor10

    consecutivesessions

    29

    510mgwithin

    5days

    5weeks

    (follow-up)

    Increasesin

    Hbsimilarbetweengroups

    AEprofilessimilarbetweengroups

    Onken

    etal.

    (2014)

    [53]

    Hb\

    11.5g/dL

    TSA

    TB

    30%

    SFB

    100ng/m

    L(orB

    300ng/

    mLifTSA

    Tcriterionmet)

    IS±

    ESA

    :1294

    FCM

    ±ESA

    :1290

    GFR

    \60

    mL/m

    in/

    1.73

    m2 ,non-dialysis-

    depend

    ent

    IS200mgdays

    0,7,

    and14

    ?two

    additionaldoses

    (max.1

    000mg)

    FCM

    15mg/kg

    days

    0and7(m

    ax.

    1500

    mg),d

    ay56

    (end

    oftreatm

    ent)

    Increase

    inHbwas

    statistically

    higher

    withFC

    Mversus

    IS

    Increasesin

    SF,T

    SAT,and

    serum

    iron

    were

    significantlygreaterin

    theFC

    Mgroupversus

    IS

    Protocol-definedhypertensive

    eventswere

    significantlymorecommon

    withFC

    Mversus

    IS,

    whereas

    protocol-definedhypotensiveeventswere

    morecommon

    withIS

    than

    FCM

    Nosignificant

    difference

    intheprim

    arycomposite

    safety

    endpoint

    (all-causedeath,

    non-fatalMI,

    non-fatalstroke,u

    nstableangina

    requiring

    hospitalization,

    congestive

    heartfailure

    requiring

    hospitalizationor

    medicalintervention,cardiac

    arrhythm

    ia,and

    hypertensive

    orhypotensive

    events)betweentreatm

    entgroups

    AEadverseevent,b.i.w

    .twiceweekly,ESA

    erythropoiesis-stimulatingagent,ESR

    Dend-stagerenaldisease,FC

    Mferriccarboxym

    altose,FeruM

    ferumoxytol,FeS

    ferroussuccinate,

    GFR

    glom

    erular

    filtrationrate,GIgastrointestinal,Hbhemoglobin,

    IIso

    iron

    isom

    altoside

    1000,iPTH

    intact

    parathyroidhorm

    one,

    IRLS

    InternationalR

    estlessLegsSynd

    romeStudyGroup

    rating

    scale,IS

    iron

    sucrose,IV

    intravenously

    administered,MImyocardialinfarction,

    q.2.w.every

    2weeks,q.w.

    weekly,RLSrestlesslegs

    synd

    rome,SA

    Eseriousadverseevent,SF

    serum

    ferritin,t.i.d.

    threetimes

    daily,t.i.w.three

    times

    weekly,TSA

    Ttransferrinsaturation

    1968 Adv Ther (2020) 37:1960–2002

  • isomaltose versus iron sucrose in patients withnon-dialysis CKD, although different dosingregimens make a direct comparison of theseproducts problematic [59].

    Limited data are available on the use of IViron in peritoneal dialysis patients. A recentstudy has assessed the safety and efficacy of ironsucrose in this patient population and hasshown that monthly maintenance with ironsucrose may improve iron parameters and ery-thropoietin response [54].

    Chronic Heart FailureIron deficiency and anemia are commonlyobserved in patients with CHF. Anemia waspreviously thought to be a major contributor tothe progression of CHF [60, 61]. In patients withCHF, iron deficiency is associated withincreased mortality [24], increased risk of hos-pitalization [62], and reduced quality of life[14, 63]. A number of studies have evaluated thetreatment effects of iron sucrose in patientswith anemia and patients without anemia, bothalone [64–68] and in combination with rhEPO[60, 69, 70]. A summary of the studies on ironsucrose use in CHF is provided in Table 3.

    Intravenous administration of iron sucrosealone to patients with anemia and CHFimproves the hematological parameters of Hb,ferritin, and TSAT [64–68]. Reductions ininflammatory markers such as natriuretic pep-tides and C-reactive protein (CRP) have alsobeen observed after iron sucrose treatment[64, 66, 70].

    These biochemical changes appear to beassociated with symptom improvement inpatients with CHF, while in one study,echocardiographic evaluation demonstratedsignificant improvements in left ventricularsystolic diameter and left ventricular diastolicdiameter [66]. In line with these findings, animprovement in cardiac remodeling was alsoobserved in another study in patients with CHF[68].

    Inflammatory Bowel Disease and OtherGastrointestinal DisordersA summary of the studies of iron sucrose use inadults with GI disorders is provided in Table 4.

    IBD (including Crohn’s disease and ulcera-tive colitis) is often treated with anti-inflam-matory drugs, including tumor necrosis factor-alpha (TNFa) inhibitors. However, the persis-tence of anemia as a complication despite anti-TNFa treatment highlights the need for irontherapy even when biologics are used [71]. Oraliron therapy, however, is problematic forpatients with IBD as a result of chronic inflam-mation leading to poor absorption and toler-ance. In this respect, and consistent withfindings in patients with CKD, several studies inpatients with iron deficiency and iron-defi-ciency anemia associated with IBD indicate thatiron sucrose is more efficacious than oral irontherapy [72, 73].

    A study in patients with iron-deficiencyanemia and IBD demonstrated significantlybetter outcomes in the iron sucrose arm for twoout of three primary endpoints compared withoral iron sulfate therapy: fewer iron sucrose-treated patients remained anemic and morepatients achieved Hb reference levels [72].

    In a pooled analysis of four trials evaluating atotal of 191 patients with iron-deficiency ane-mia resulting from GI disorders, greater increa-ses in Hb levels were apparent in patientstreated with iron sucrose compared with thosereceiving oral iron therapy [74]. However, theinterpretation of these findings is somewhatproblematic because patients with Hb levelsbelow 10 g/dL were given iron sucrose treat-ment and patients with higher Hb levels (atleast 10 g/dL) were treated with oral ferroussulfate therapy. Iron sucrose treatment inpatients with lower Hb levels (below 10 g/dL)led to 77% of subjects achieving normal Hblevels after 3 months. Oral administration offerrous sulfate in patients with higher baselineHb levels (at least 10 g/dL) resulted in 89%achieving normalized Hb [75]. Although theauthors of this study reported that all patientswere in remission, more details on the overalldisease severity of the two groups would be ofinterest. Patients with IBD and severe anemia(Hb below 10 g/dL) usually have more severeand relapsing disease associated with a higherneed for iron supplementation compared withpatients with mild anemia and inactive disease.Moreover, the recurrence of anemia after iron

    Adv Ther (2020) 37:1960–2002 1969

  • Table3

    Publishedclinicalstudiesusingiron

    sucrosein

    patientswithchronicheartfailure

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Bolger

    etal.

    (2006)

    [65]

    Stablesystolicheartfailure

    and

    HbB

    12g/dL

    IS:16

    200mg(10mL)IV

    bolusover10

    min

    ondays

    1,3,

    and5of

    study

    ISim

    proved

    hematologicalparameters

    (Hb,

    serum

    iron,S

    F,TSA

    T),

    symptom

    s,andexercise

    capacity

    (NYHA

    functionalclass,MLHFQ

    score,6M

    WT).ISC

    was

    welltolerated

    withno

    localor

    system

    icAEs

    Silverberg

    etal.

    (2003)

    [60]

    Octogenarians

    withanem

    iaandsevere,

    resistantCHF

    IS:40

    Com

    bination

    ofSC

    EPO

    (4–5

    KIU

    /week,

    increasing

    to10

    KIU

    /weekto

    atarget

    Hbof

    12.5g/dL

    )andIS

    200mg

    in150mLsalin

    eIV

    infusion

    over

    60min

    every1–

    2weeks

    untilSF

    reached500lg/L

    oriron

    saturation

    reached40%

    orHbreached12.5g/dL

    Mean±

    SDduration

    offollow-upwas

    17.4

    ±10

    months.IS

    incombination

    withEPO

    significantlyincreasedHb,

    serum

    iron,S

    F,and%

    iron

    saturation

    andalso

    improved

    NYHA,L

    VEF,

    and

    VASindex.Significantlyfewer

    hospitalizations

    werereported

    comparedwithacomparableperiod

    priorto

    thestudy

    Silverberg

    etal.

    (2003)

    [69]

    Individualswithandwithout

    diabetes

    withmoderateto

    severe

    resistantCHF

    (and

    CRF);NYHA

    functionalclasses

    I–IV

    andHbrangeof

    9.5–

    11.5g/dL

    IS:179(84

    individuals

    withtype

    2diabetes;95

    without

    diabetes)

    Com

    bination

    ofSC

    EPO

    (4–5

    KIU

    /week,

    increasing

    to10

    KIU

    /weekto

    atarget

    Hbof

    12.5g/dL

    )andIS

    200mg

    in150mLsalin

    eIV

    infusion

    over

    60min

    every1–

    2weeks

    untilSF

    reached500lg/L

    oriron

    saturation

    reached40%

    orHbreached12.5g/dL

    Inindividualswith/without

    diabetes

    with

    severe

    CHFandmild

    tomoderate

    CRF,

    correction

    ofanem

    iaby

    the

    combineduseof

    EPO

    andISC

    was

    associated

    withim

    provem

    entsin

    LVEF

    andcardiacfunctionalstatus

    (NYHA

    andVAS)

    andslo

    wingof

    therate

    ofdeteriorationof

    renalfunction

    Com

    in-

    Colet

    etal.

    (2009)

    [70]

    Stableadvanced

    CHF(N

    YHA

    class

    III–IV)andanem

    ia(m

    en,

    Hb\

    13.0g/dL

    ;wom

    en,

    Hb\

    12.0g/dL

    )andmild

    tomoderate

    renaldysfun

    ction

    IS:27

    EPO

    (4KIU

    /week,

    adjusted

    asnecessary

    toachieveandmaintainatarget

    Hb

    between12.5

    and14.5g/dL

    ).IS

    200mgin

    100–

    200mLsalin

    eIV

    infusion

    over

    60–9

    0min

    q.w.for

    5–6weeks

    untilSF

    reached400lg/L

    orTSA

    Tof

    40%

    orHb[

    14.5g/dL

    .IS

    then

    givenevery4–

    6weeks

    tomaintaintheselevels

    Mean±

    SDfollow-upwas

    15.3

    ±8.6months.Long-term

    combinedtherapywithEPO

    andISC

    increasedHb,

    reducedNT-proBNP,

    andim

    proved

    functionalcapacity

    and

    cardiovascular

    hospitalizationrates

    1970 Adv Ther (2020) 37:1960–2002

  • Table3

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Toblli

    etal.

    (2007,

    2015)

    [64,

    66]

    LVEFB

    35%;NYHA

    functionalclass

    II–IV;anem

    iawithHb\

    12.5g/dL

    formen

    and\

    11.5g/dL

    forwom

    en;

    SF\

    100ng/m

    Land/or

    TSA

    TB

    20%.R

    enalinsufficiency

    (creatinineclearanceB

    90mL/m

    in)

    IS:30

    200mg/200mLISC

    60min

    IVinfusion

    q.w.for

    5weeks

    At6monthspost-treatment,patients

    receivingISC

    infusion

    experienced

    significant

    increasesin

    Hblevel,SF,and

    TSA

    T.H

    eartrateandbody

    massindex

    weresignificantlyreducedin

    theIS

    group,

    whileLVEFwas

    significantly

    increased,

    aswereNYHA

    scores.

    InflammatorymarkersCRPandNT-

    proB

    NPwerealso

    significantlyreduced.

    Echocardiographicevaluation

    show

    edsignificant

    improvem

    entsin

    LVSd

    and

    LVDd,

    suggesting

    apositive

    effect

    oncardiacmuscle.A

    significant

    improvem

    entin

    renalfun

    ctionwasalso

    observed.S

    ideeffectswereminim

    alacrossboth

    groups

    andno

    severe

    drug-

    relatedAEswerereported

    Okonko

    etal.

    (2008)

    [67]

    AgedC

    21yearswithsymptom

    aticCHF

    (NYHAfunctionalclassII–IV;exercise

    limitationpV

    O2B

    18mL/kg/min;

    Hb\

    12.5g/dL

    (group

    withanem

    ia)

    or12.5–1

    4.5g/dL

    (group

    without

    anem

    ia)

    IS:24

    ISadministeredat

    200mgin

    50mL

    salin

    eviaIV

    infusion

    over

    30min

    q.w.

    for16

    weeks.T

    henat

    weeks

    4,8,

    12,

    and16

    thereafter

    ISresultedin

    asignificant

    treatm

    enteffect

    forSF,T

    SAT,and

    Hblevelsin

    the

    patientswithanem

    ia.A

    nim

    provem

    ent

    inexercise

    tolerance(pVO

    2),N

    YHA

    functionalclass,PG

    A,and

    fatiguescore

    wasalso

    observed

    withIS

    inthepatients

    withanem

    ia.A

    Eprofilesweresimilar

    betweengroups:42%

    and64%in

    theIS

    andcontrolgroups,respectively.All

    eventswereun

    related(76%

    )or

    unlikely

    tobe

    related(24%

    )to

    thestudydrug

    Adv Ther (2020) 37:1960–2002 1971

  • Table3

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Beck-da-

    Silva

    etal.

    (2013)

    [178]

    AgedC

    18years;NYHAfunctionalclass

    II–IVandableto

    perform

    ergospirom

    etry;L

    VEF\

    40%;H

    bC

    9andB

    12g/dL

    ;TSA

    T\

    20%;

    SF\

    500lg/L

    IS:10

    FeS:

    7

    Placebo:

    6

    IS200mgIV

    infusion

    over

    30min

    q.w.

    for5weeks

    then

    oralplacebot.i.d.for

    8weeks

    FeS200mgorallyt.i.d.for

    8weeks

    then

    IVplaceboq.w.for

    5weeks

    Oralplacebot.i.d.for

    8weeks,thenIV

    placeboq.w.for

    5weeks

    3-month

    follow-up

    Correctionof

    anem

    ia,d

    efinedas

    Hb[

    12g/dL

    forwom

    enand[

    13g/

    dLformen,w

    asachieved

    bytwo

    patientswithIS,three

    withFeS,

    and

    twowithplacebo.Hbincrease[

    1.5g/

    dLoccurred

    inthreepatientseach

    intheIS

    andFeSgroups.S

    Flevels

    increasedfrom

    baselin

    ewithboth

    ISandFeS(but

    notplacebo),but

    thiswas

    onlystatistically

    significant

    forFeS.

    TSA

    T[

    20%

    was

    achieved

    byfive

    patientsin

    each

    oftheIS

    andFeS

    groups

    pVO

    2increasedby

    3.5mL/kg/min

    with

    ISwhereastherewasno

    change

    inpV

    O2

    withoraliron

    (nostatistically

    significant

    difference

    betweengroups)

    Usm

    anov

    etal.

    (2008)

    [68]

    Hb\

    11g/dL

    ontwooccasionswithin

    1week;

    serum

    creatinine

    \4mg/dL

    andNYHA

    classificationIII/IV

    IS:19NYHAIII

    and13

    NYHA

    IV;22

    healthy

    controls

    IS100mgIV

    q3wfor3weeks,thenq.w.

    for23

    weeks

    (totaldose

    3200

    mgof

    elem

    entaliron

    over

    26weeks)

    Hb,

    serum

    iron,S

    F,andTSA

    Twere

    significantlylower

    inpatientswith

    NYHA

    classificationIII/IV

    versus

    healthycontrolsat

    baselin

    e.The

    mean

    values

    oftheseparameterswere

    significantlyincreasedafter6monthsof

    treatm

    entwithIV

    ISversus

    baselin

    e.Therewereno

    AEsnotedin

    anypatient

    6MWT6-min

    walktest,A

    Eadverseevent,CHFchronicheartfailure,C

    RFchronicrenalfailure,C

    RPC-reactiveprotein,

    EPO

    erythropoietin,F

    eSferroussulfate,

    Hbhemoglobin,

    ISiron

    sucrose,ISCiron

    sucrosecomplex,IVintravenous,LVDdleftventriculardiastolic

    diam

    eter,L

    VEFleftventricularejection

    fraction,L

    VSd

    leftventricularsystolicdiam

    eter,MLHFQ

    Minnesota

    LivingwithHeart

    Failure

    Questionn

    aire,NT-proBNPN-terminalpro-B-typenatriureticpeptide,NYHA

    New

    YorkHeartAssociation,P

    GAPatientGlobalA

    ssessm

    ent,pV

    O2peak

    oxygen

    uptake,q3w

    every3weeks,q.w.w

    eekly,SC

    subcutaneous,SDstandard

    deviation,

    SFserum

    ferritin,t.i.d.

    threetimes

    daily,T

    SATtransferrinsaturation,V

    ASVisualAnalogScale

    1972 Adv Ther (2020) 37:1960–2002

  • Table4

    Publishedclinicalstudiesusingiron

    sucrosein

    gastrointestinaldisorders

    Stud

    yInclusioncriteria

    Patients,

    nIron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Com

    parativestu

    diesofiron

    sucroseversus

    oral

    iron

    form

    ulations

    Lindgrenet

    al.

    (2009)

    [72]

    Hb\

    11.5g/dL

    SF\

    300ng/m

    L

    IS:45

    FeS:

    46

    200mgq.w.o

    rq.2.w.u

    pto

    cumulativedose

    of1000

    mg

    100mgb.i.d.,20

    weeks

    66%

    versus

    47%

    Hbresponse

    toIS

    versus

    FeS(p

    =0.07)

    One

    possiblyrelatedSA

    E(throm

    bocytopenia)

    withIS,A

    EswithFeS

    dominated

    byGIevents

    Lee

    etal.(2017)

    [73]

    TSA

    T\

    16%

    ±SF

    \30

    ng/m

    L(if

    norm

    alCRP)

    or\

    100ng/m

    L(if

    elevated

    CRP)

    IS:36

    FeS:

    36

    39

    300mg(irondeficiency

    only);49

    300mg(with

    anem

    ia)

    300mgb.i.d

    12weeks

    (follow-up)

    HbandTSA

    Tlevelssimilarbetweengroups

    SFsignificantlyhigher

    withIS

    versus

    FeS

    Gut

    microbiotaandmetabolom

    ealtereddifferently

    byFeSandIS

    Gisbertet

    al.

    (2009)

    [75]

    Hb\

    12or

    13g/dL

    a

    TSA

    T\

    12%

    SF\

    30ng/m

    L

    IS:22

    FeS:

    78

    200mgb.i.w

    .ifHb\

    10g/

    dLun

    tilcalculated

    dose

    106mgq.d.

    ifHb[

    10g/

    dL

    77%

    and89%

    response

    rate

    inIS

    andFeSgroup,

    respectively

    NoAEswithIS,5

    .1%

    withoraliron

    therapyintolerance(nausea,

    abdominalpain,and

    constipation),which

    ledto

    discontinu

    ationof

    treatm

    ent

    Com

    parativestu

    diesofiron

    sucroseversus

    otherIV

    iron

    form

    ulations

    Lichtensteinand

    Onken

    (2018)

    [74]

    [Pooledanalysis

    offour

    studies]

    HbB

    12g/dL

    (one

    study);B

    11g/dL

    (three

    studies)

    SFB

    100ng/m

    Lor

    B300ng/m

    Lif

    TSA

    TB

    30%

    (allfour

    studies)

    IS:32

    FCM:

    101

    Oraliron:

    25

    Other

    IViron:61

    15mg/kg

    or750mg

    (variabledosing

    schedules)

    Hb,

    SF,and

    TSA

    Tvalues

    increasedsignificantlyfrom

    baselin

    eforall

    groups,w

    iththeexceptionof

    SFvalues

    intheoraliron

    group

    FCM

    andIS

    resultedin

    greaterincreasesin

    Hbthan

    orallyadministered

    andotherIV

    iron

    products,w

    hich

    may

    bedueto

    lowerbaselin

    eHblevels

    Safety

    profileof

    FCM

    was

    comparablewiththeotheragents

    Incidenceof

    treatm

    ent-relatedSA

    Eswas

    0%withFC

    Mversus

    6.3%

    with

    IS

    Evstatiev

    etal.

    (2011)

    [76]

    Hb7–

    12g/dL

    formen

    and7–

    12g/dL

    forwom

    en

    SF\

    100lg/m

    L

    IS:235

    FCM:

    240

    200mg(upto

    11doses)

    Maxim

    umof

    threeinfusions

    of1000

    or500mg

    MorepatientswithFC

    Mthan

    ISachieved

    Hbresponse(p

    =0.04)or

    Hb

    norm

    alization(p

    =0.015).B

    othtreatm

    entsim

    proved

    qualityof

    life

    scores

    byweek12

    Studydrugswerewelltoleratedanddrug-related

    AEswerein

    linewith

    drug-specific

    clinicalexperience

    AEadverseevent,b.i.d.twicedaily,b.i.w.twiceweekly,CRPC-reactiveprotein,FC

    Mferriccarboxym

    altose,FeS

    ferroussulfate,G

    Igastrointestinal,H

    bhemoglobin,IS

    iron

    sucrose,IV

    intravenous,q.2.w.every

    2weeks,q.d.d

    aily,q.w.w

    eekly,SA

    Eseriousadverseevent,SF

    serum

    ferritin,T

    SATtransferrinsaturation

    aFemaleHb\

    12g/dL

    ,male\

    13g/dL

    Adv Ther (2020) 37:1960–2002 1973

  • treatment may be higher in patients with severeanemia.

    When comparing iron sucrose with ferriccarboxymaltose (FCM), Hb response rates weresimilar to those seen in the studies describedabove and in Table 4; however, outcomes withFCM were significantly better [76]. The pooledanalysis in patients with GI disorders describedabove found that FCM was associated withgreater increases in ferritin and TSAT comparedwith iron sucrose, although increases in Hblevels were similar [74].

    Pediatric studies in children with GI disordersare summarized in the relevant section below.

    Women’s Health, Including Obstetricsand Gynecology

    A summary of the studies investigating ironsucrose use in women’s health (including in thepregnancy and postpartum settings) is providedin Table 5.

    Pregnancy and PostpartumMaintenance of normal iron stores is importantnot only for the health of the prospectivemother,but also for that of her child. A recent summary ofmeta-analyses of the effects of iron status and irontherapy concluded that maternal anemia wasassociated with adverse birth and neonatal out-comes and that iron treatment reduced the risk ofthis [77]. A recent Swedish cohort study ofover 500,000 children born between 1987 and2010 and followedupuntil the end of 2016 foundthat the risks of developing autism, attention-deficit hyperactivity disorder, and intellectualdisabilitywereall increased inchildrenofmotherswho had prenatal anemia during the first30 weeks of pregnancy [78].

    Reflecting results in CKD and IBD, studies inpregnant women with iron deficiency indicatethat iron sucrose is more effective than oral ironadministration and is at least as effective inpregnant women with iron-deficiency anemia.For women with severe iron deficiency duringpregnancy, iron sucrose treatment has beenshown to increase Hb levels more effectivelythan an oral iron polymaltose complex [79],whereas in pregnant women with less severe

    iron-deficiency anemia and in those using ironprophylactically, Hb levels were comparablefollowing either iron sucrose or oral ferroussulfate treatment [80, 81].

    Administering iron sucrose from the thirdtrimester to pre-delivery can significantlyincrease Hb levels and reduce blood transfusionrates in pregnant women with third trimesterHb levels below 9.5 g/dL, compared withpatients who did not receive iron sucrose [83].Treatment with a minimum of three ironsucrose doses at least 2 weeks prior to deliveryappears optimal for increasing Hb levels andtreating antepartum anemia [84].

    Addition of rhEPO to iron sucrose therapycan enhance the effectiveness of treatment ofiron-deficiency anemia in pregnancy. In a studyof pregnant women with iron-deficiency ane-mia resistant to treatment with oral iron ther-apy alone, iron sucrose administered either withor without rhEPO effectively treated anemia.However, increases in reticulocytes and hemat-ocrit were greater, and more patients achievedtarget Hb levels at 4 weeks with the combina-tion of rhEPO and iron sucrose compared withiron sucrose alone. Safety was similar in the twotreatment groups [85]. Selective addition ofrhEPO to iron sucrose for women with severeanemia or an inadequate response to ironsucrose alone has also been shown to be aneffective approach to treatment of iron-defi-ciency anemia in pregnancy [86].

    Furthermore, iron sucrose has demonstratedequal if not superior efficacy compared withoral iron administration in women with iron-deficiency anemia in the postpartum setting.For these women, a study has shown that ironsucrose treatment can achieve higher Hb levelsthan oral ferrous sulfate treatment [88]. Ironsucrose followed by standard oral iron treat-ment after 4 weeks also replenished iron storesmore rapidly and resulted in a greaterimprovement in fatigue score compared withoral iron treatment alone in a randomized studyof women with postpartum iron-deficiencyanemia [90].

    The effectiveness of iron sucrose either withor without rhEPO was assessed in a randomizedstudy in women with severe postpartum ane-mia. Hb returned to near-normal levels within

    1974 Adv Ther (2020) 37:1960–2002

  • Table5

    Publishedclinicalstudiesusingiron

    sucrosein

    obstetrics,gynecology,andwom

    en’shealth

    disorders

    Stud

    yInclusion

    criteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Useofiron

    sucroseduring

    pregnancyandpostpartum

    Al-M

    omen

    etal.(1996)

    [150]

    Gest.

    age\

    32weeks

    Hb\

    9.0g/dL

    SF\

    20ng/m

    L

    IS:52

    FeS:59

    IS:200mgq.d.

    ort.i.w.

    FeS:

    60mgt.i.d.

    Significantlyhigher

    HblevelswithIS

    versus

    FeS

    Self-lim

    itingfeverandtightnessin

    skin

    (one

    each)withIS,3

    0%GI

    eventsand32%

    non-compliancewithFeS,

    6%couldnottolerate

    FeS

    Alet

    al.

    (2005)

    [79]

    Gest.age

    26–3

    4weeks

    Hb8.0–

    10.5g/

    dL

    SF\

    13ng/m

    L

    IS:45

    IPC:45

    IS:200mgun

    tilcalculated

    dose,

    5days

    IPC:100mgt.i.d.,whole

    pregnancy

    Significantlybetter

    Hbincrease

    withIS

    versus

    IPC

    PossiblyrelatedAEsto

    IS:m

    etallic

    taste(n

    =11),hotflush

    (n=12),

    dizziness(n

    =8),n

    ausea(n

    =5),arthralgia(n

    =1),vom

    iting

    (n=1);significantly

    fewerGIsymptom

    swithIS

    versus

    FeS(13.3%

    vs.2

    8.9%

    upperGIevents)

    Bayoumeu

    etal.(2002)

    [80]

    Hb8.0–

    10.0g/

    dL

    SF\

    50ng/m

    L

    IS:24

    FeS:

    23

    IS:69

    200mgun

    tilcalculated

    dose,2

    1days

    FeS:

    80mgt.i.d.,4weeks

    Hbincrease

    inboth

    groups

    without

    significant

    difference

    ‘‘Not-unp

    leasanttaste’’onlyAEwithIS,duringinjection(dysgeusia).

    One

    treatm

    entinterruption

    dueto

    AE(diarrhea)

    withFeS

    Bencaiova

    etal.(2009)

    [81]

    Gest.age

    15–2

    0weeks

    Hb\

    10.5g/dL

    SF\

    100ng/m

    L

    IS:130

    FeS:

    130

    IS:200or

    300mg

    FeS:

    80mgq.d.

    Significant

    Hbincrease

    versus

    baselin

    ein

    both

    groups

    Mild

    anem

    ia(16.2%

    ),infections

    (6.9%),musclepains(2.3%),pruritus

    (2.3%),cough(1.5%),breastdisorders(1.5%)withIS;GIevents

    onlywithFeS(17.7%

    ),14

    SAEsin

    ISgroup(preterm

    contractions

    n=3,

    prem

    atureruptureof

    themem

    branes

    n=3,

    moderate

    anem

    iaafterdeliveryn=2,threatened

    preterm

    deliverybecauseof

    cervicalinsufficiency

    n=2,

    intrauterine

    grow

    threstrictionn=2,

    infectionn=1,

    injury

    n=1),seven

    SAEsin

    FeSgroup(preterm

    contractions

    n=2,

    vaginalbleeding

    n=1,

    prem

    atureruptureof

    themem

    branen=1,

    infectionn=1,

    gestationaldiabetes

    mellitus

    n=1,

    postpartum

    pulmonaryem

    bolism

    n=1)

    Adv Ther (2020) 37:1960–2002 1975

  • Table5

    continued

    Stud

    yInclusion

    criteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Tariq

    etal.

    (2015)

    [82]

    Gest.

    age[

    12weeks

    Hb\

    10.5g/dL

    SF\

    12lg/L

    IS:93

    Iron

    dextran:

    105

    C1antepartum

    300mgIS

    dose

    ISandiron

    dextranwereequally

    effectivein

    treatm

    entof

    IDAduring

    pregnancy

    Nomajor

    side

    effectswereobserved

    ineither

    group

    Ham

    met

    al.

    (2018)

    [83]

    Third

    trim

    ester

    Hb\

    9.5g/dL

    IS:25

    NoIS:364

    C1antepartum

    300mgIS

    dose

    The

    earlierIS

    wasreceived

    beforedelivery,thegreaterthemedianHb

    increase.O

    nlythosereceivingIS[

    2weeks

    before

    deliveryhada

    significant

    increase

    inHblevelfrom

    thirdtrim

    esterto

    delivery.

    Increasing

    administrations

    increm

    entally

    impacted

    Hbdifference

    from

    thirdtrim

    esterto

    delivery,withonlythosereceivingC

    3dosesdemonstrating

    statistically

    significant

    Hbchange

    compared

    withtheno

    ISgroup

    Ham

    met

    al.

    (2019)

    [84]

    Third

    trim

    ester

    Hb\

    9.5g/dL

    IS:65

    NoIS:458

    Exact

    dosesnotstated

    Third

    trim

    esterHbwaslower

    intheIS

    groupcomparedwiththeno

    ISgroup

    Despite

    lower

    starting

    Hbin

    theIS

    group,

    antepartum

    ISreduced

    transfusions

    inpatientswithathirdtrim

    esterHb\

    9.5g/dL

    Breym

    ann

    etal.(2001)

    [85]

    Hb\

    10.0g/dL

    SFB

    15ng/m

    L

    IS:20

    IS?

    ESA

    :20

    200mg,b.i.w

    .Bothregimenswereeffective;increasesin

    hematocritweregreater

    from

    day11

    (p\

    0.01)andthemedianduration

    oftherapywas

    shorter(18vs.2

    5days)withadjuvant

    ESA

    Welltolerated;m

    etallic

    tastewasreported

    in8%

    andwarm

    feelingin

    5%

    Krafftet

    al.

    (2009)

    [86]

    Hb\

    10.0g/dL

    SFB

    15ng/m

    L

    IS:27

    IS?

    ESA

    :57

    200mg,b.i.w

    .IS

    orIS

    ?ESA

    was

    effective:theoverallHblevelaftertherapywas

    11.0g/dL

    (note:32

    patientsrespondedpoorlyinitially

    toIS

    alone,

    thus

    receivingadditionalESA

    )

    Welltolerated;

    mostpatientsreported

    metallic

    taste

    1976 Adv Ther (2020) 37:1960–2002

  • Table5

    continued

    Stud

    yInclusion

    criteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Bencaiova

    etal.(2006)

    [87]

    SFB

    15ng/m

    LIS

    ?ESA

    :19

    100mg,b.i.w

    .Attheendof

    therapy,Hb,

    HCT,and

    serum

    EPO

    werestatistically

    significantlyim

    proved

    cf.atthestart(p

    =0.01,p

    =0.004,

    and

    p=0.006,

    respectively)

    Welltolerated

    andno

    detectablenegative

    effectson

    newborn

    orfetus

    Bhand

    alet

    al.

    (2006)

    [88]

    Hb\

    9.0g/dL

    SF\

    15ng/m

    L

    IS:22

    FeS:

    21

    IS:200mge.o.d.,3

    days

    FeS:

    200mgb.i.d.,6weeks

    Significantlybetter

    Hbincrease

    withIS

    versus

    FeS

    23%

    metallic

    taste,18%

    facialflushingwithIS,3

    3%GIeventswith

    FeS

    NoSA

    Es,no

    hemodynam

    icdisturbances

    during

    orafterinfusion

    Giann

    oulis

    etal.(2009)

    [89]

    Hb\

    8.0g/dL

    SF\

    10ng/m

    L

    IS:78

    FeProtSu:26

    IS:100mgq.d.,3

    days

    FeProtSu:800mgq.d.,2

    8days

    Significantlybetter

    Hbincrease

    withIS

    versus

    FeProtSu

    TwoAEswithIS

    (headache,nausea),11

    AEswithFeProtSu

    (constipationn=5,

    hiccup

    n=4,

    heartburnn=2)

    Westadet

    al.

    (2008)

    [90]

    Hb6.5–

    8.5g/dL

    IS:58

    FeS:

    70

    IS:200mgq.d.,3

    days

    FeS:

    100mgb.i.d.,12

    weeks

    Hbincrease

    inboth

    groups

    FewandtransientAEswithIS

    (phlebitis,p

    ainat

    injectionsite),

    22.9%

    withdrewbecauseof

    drug-related

    AEswithFeS(G

    Ievents

    mostcommon

    reason)

    Perello

    etal.

    (2014)

    [130]

    Hb6–

    8g/dL

    IS?

    FeS:

    36

    Placebo?

    FeS:

    36

    IS:200mgq.d.,2

    days

    FeS:

    525mgb.i.d.,30

    days

    Hbvalues

    werecomparablein

    wom

    enreceivingIS

    orplaceboin

    addition

    tooraliron

    therapyat

    anyof

    thetimepoints.N

    odifferenceswerefoun

    dbetweenclinicalsymptom

    sof

    anem

    ia,

    psychologicalstatus,and

    AEsbetweengroups

    Krafftand

    Breym

    ann

    (2011)

    [91]

    Hb\

    8.5g/dL

    24–4

    8hafter

    delivery

    IS:20

    IS?

    ESA

    :20

    200mgq.d.,4

    days

    Hbvalues

    werecloseto

    norm

    alin

    both

    groups

    within2weeks

    Bothtreatm

    entswerewelltolerated;

    minor

    side

    effectsincluded

    metallic

    tastein

    30%

    ofpatientsandwarm

    flush

    in8%

    ofpatients

    Adv Ther (2020) 37:1960–2002 1977

  • Table5

    continued

    Stud

    yInclusion

    criteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,

    treatm

    entdu

    ration

    Outcomes

    andsafety

    inform

    ation

    Wågström

    etal.(2007)

    [92]

    HbB

    80g/dL

    within72

    hafterdelivery

    IS:15

    IS?

    rhEPO

    1:19

    IS?

    rhEPO

    2:15

    IS:200mgq.d.,d

    ay0,

    day3

    IS?

    rhEPO

    1:IS

    200mgq.d.,day

    0,day3?

    rhEPO

    10,000

    Uon

    day0,

    day3

    IS?

    rhEPO

    2:IS

    200mgq.d.,day

    0,day3?

    rhEPO

    20,000

    Uon

    day0,

    day3

    Hbincreasedsignificantlyin

    allthreegroups

    over

    time(p\

    0.001),

    andtherewereno

    differencesbetweenthetreatm

    entgroups

    onany

    dayof

    evaluation

    (p=0.59).The

    totalmeanincrem

    entin

    Hbin

    allsubjectswas

    18g/dL

    after1weekand28

    g/dL

    after2weeks

    Breym

    ann

    etal.(2000)

    [93]

    Hb\

    10.0g/dL

    IS:20

    IS?

    ESA

    :20

    Oraliron:20

    200mgq.d.,4

    days

    Allthreeregimenswereeffective;day7hematocritincreaseswere

    higher

    withadjuvant

    ESA

    than

    forIS

    ororaliron

    alone:8%

    versus

    5%and4%

    ,respectively(bothcomparisons

    p\

    0.01)

    Welltolerated

    Useofiron

    sucroseforgeneralwom

    en’shealth

    Krayenbuehl

    etal.(2011)

    [94]

    HbC

    12.0g/dL

    SFB

    50ng/m

    L

    IS:43

    Placebo:

    47

    200mgq.d.,4

    days

    Trend

    forbetter

    improvem

    entof

    fatiguescorewithIS

    versus

    placebo

    21%

    drug-related

    AEswithIS

    (nausea,chills,headache,d

    izziness,

    chestpain,d

    ysesthesia,d

    ysgeusia),7%

    AEswithplacebo(nausea,

    headache,d

    izziness,d

    iarrhea)

    Lee

    etal.

    (2019)

    [96]

    Menorrhagia

    Hb\

    10g/dL

    SF\

    30ng/m

    L

    IS:49

    FCM:52

    IS:200mgq.d.

    (B600mg/week)

    FCM:\

    50kg,5

    00mg/

    week;

    C50

    kg,1

    000mg/week

    FCM

    was

    aseffectiveas

    ISin

    achievingHbC

    10g/dL

    within

    2weeks

    afterfirstadministration(78.8%

    vs.7

    2.3%

    ).The

    timeto

    reachHbC

    10g/dL

    was

    significantlyshorterin

    theFC

    Mgroup

    versus

    theIS

    group(7.7days

    vs.1

    0.5days).MeanHblevelswere

    higher

    intheFC

    M-treated

    patientsthan

    intheIS-treated

    patients

    (borderlinesignificance,p=0.013).Q

    oLscores

    didnotdiffer

    betweengroups.N

    odeaths,anaphylacticreactions,or

    transfusion

    requirem

    entswerereported

    ineither

    group.

    AllAEsweremild

    AEadverseevent,b.i.d.twicedaily,b.i.w

    .twiceweekly,e.o.d.everyotherday;

    EPO

    erythropoietin,E

    SAerythropoiesis-stimulatingagent,FC

    Mferrouscarboxy-

    maltose,FeProtSu

    ferrousproteinsuccinylate,FeSferroussulfate,Gest.gestational,GIgastrointestinal,HCT

    hematocrit,Hbhemoglobin,

    IDA

    iron-deficiency

    anem

    ia,IPC

    iron

    polymaltose

    complex,ISiron

    sucrose,q.d.

    daily,Q

    oLqualityof

    life,SA

    Eseriousadverseevent,rhEPO

    recombinant

    human

    erythropoietin,SF

    serum

    ferritin,t.i.d.

    threetimes

    daily,t.i.w.three

    times

    weekly

    1978 Adv Ther (2020) 37:1960–2002

  • 2 weeks in both treatment groups. The findingssuggest that iron sucrose alone is sufficient forthe treatment of postpartum anemia for mostwomen, although additional rhEPO treatmentmay benefit a subgroup of patients with pro-nounced inflammatory responses [91]. A secondstudy in women with postpartum anemia foundthat the addition of rhEPO did not furtherincrease Hb concentrations above thoseachieved with IV iron sucrose treatment alone[92], while another study reported greatesthematocrit increases with rhEPO and ironsucrose compared with either iron sucrose ororal iron treatment alone (both p\0.01) [93].

    General Women’s HealthBesides its use in pregnant women with iron-deficiency anemia, iron treatment can alsobenefit women with low ferritin levels andwithout anemia. In fatigued, premenopausalwomen with low ferritin levels and withoutanemia, iron sucrose treatment (four infusionsof 200 mg iron during the first two study weeks)showed a trend toward a greater improvementin fatigue compared with placebo. The differ-ence between the groups was significant inthose with initial serum ferritin levels of 15 ng/mL or below [94].

    A large number of women are iron deficientand suffer from fatigue. It has therefore beensuggested that parenteral iron administrationcould benefit many women and could becomemore accepted. However, those who will benefitmost, and the doses of iron required for aneffect, need to be further defined [95].

    Two randomized clinical studies havedemonstrated that iron sucrose is effective forthe correction of iron-deficiency anemia due toheavy menstrual bleeding (menorrhagia)[96, 97]; in one study, 72% of women whoreceived treatment with iron sucrose achievedHb levels of at least 10 g/dL within 2 weeks afterthe first administration [96].

    Patient Blood Management

    A summary of the studies assessing iron sucroseuse in patient blood management is provided inTable 6.

    Preoperative Iron TreatmentPreoperative anemia is present in 30–40% ofpatients scheduled for elective surgery and cancomplicate surgery, increasing the likelihood ofblood transfusion or intensive care admission,increasing the risk of infections or throm-boembolic events, and prolonging the length ofhospital stay [35, 98–100]. In addition, and asmentioned previously, in patients undergoingelective cardiac surgery, iron deficiency withand without concomitant anemia is associatedwith a threefold increase in mortality, morepostoperative serious adverse outcomes, moreblood transfusions, and longer intensive careand hospital stay than patients without irondeficiency [25].

    Iron sucrose is an effective pre-surgicaltreatment for iron-deficiency anemia and hasbeen shown to increase Hb, ferritin, and solubletransferrin receptor levels when administeredprior to orthopedic surgery [101]. Increases inHb were greatest 2 weeks after initiating ironsucrose treatment, suggesting that the optimaltime to initiate iron sucrose may be 2–3 weeksprior to elective surgery [101].

    In patients without anemia receivingrhEPO who were due for elective surgery, ironsucrose was associated with significantlyhigher ferritin and reticulocyte counts thanwith oral ferrous sulfate therapy, and pre-vented iatrogenic iron depletion [102]. In aseparate study, there was no differencebetween oral iron therapy and IV iron insupporting preoperative erythropoietin-drivenstimulation of erythropoiesis (transfusionswere not reported) [103].

    A benefit of iron sucrose and an ESA was alsodemonstrated in patients undergoing surgeryfor hip fractures. Combined treatment resultedin increased Hb levels and a lower transfusionrate compared with the use of either ironsucrose or ESA alone [104].

    In surgical patients with iron-deficiencyanemia resulting from heavy menstrual bleed-ing, iron sucrose achieved significantly betterpreoperative Hb and ferritin levels than oraliron protein succinylate treatment [105]. How-ever, the study is limited by the premature ces-sation of the oral iron arm in the study.

    Adv Ther (2020) 37:1960–2002 1979

  • Table6

    Publishedclinicalstudiesusingiron

    sucrosein

    patientbloodmanagem

    ent

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Useofiron

    sucrosein

    thepreoperativesetting

    Theusinger

    etal.

    (2011)

    [101]

    Preoperative

    orthopedic

    surgery

    IS:20

    900mgover

    10days

    HbandSF

    levelsincreasedsignificantly.T

    SAT

    didnot

    change

    significantly.E

    ndogenousEPO

    decreasedfrom

    261to

    190pg/m

    L2weeks

    afterIV

    iron

    treatm

    ent

    (p=0.050,

    notsignificant

    afterBonferronicorrection)

    NoAEsrelatedto

    ISwereobserved

    Rohlin

    get

    al.

    (2000)

    [102]

    Preoperative

    elective

    surgery

    Hb\

    14.0g/dL

    IS?

    ESA

    :6

    FeS?

    ESA

    :6

    IS:200mgb.i.w

    .,3weeks

    FeS:

    160mg/day,3weeks

    Hblevelsincreasedin

    both

    groups;Hblevelincreased

    significantlyover

    3weeks

    withIS

    ?ESA

    butnotwith

    FeS?

    ESA

    Preoperative

    reticulocyte

    coun

    tandSF

    weresignificantly

    higher

    withIS

    ?ESA

    than

    FeS?

    ESA

    Nodose-limitingAEsor

    allergicreactionswereobserved

    TwopatientsreceivingFeSreported

    mild

    GIeffects

    (constipation,

    epigastricpain)

    Olijhoek

    etal.

    (2001)

    [103]

    Preoperative

    elective

    orthopedicsurgery

    HbC

    10.0

    toB

    13.0g/dL

    SFC

    50ng/m

    L

    IS?

    ESA

    :29

    Oraliron

    ?ESA

    :29

    IS?

    placebo:

    25

    Oraliron

    ?placebo:

    27

    IS:200mgday1andday8

    Oraliron:200mg/day,

    2weeks

    IS?

    ESA

    andoraliron

    ?ESA

    significantlyincreasedtotal

    RBC

    production,H

    b,HCT,and

    reticulocytesversus

    respective

    placebogroups;no

    significant

    differencesfor

    IS?

    ESA

    versus

    oraliron

    ?ESA

    IS?

    placebo,

    andoraliron

    ?placebowas

    notassociated

    withincreasesin

    hematologicalvalues

    Incidenceof

    AEssimilarbetweengroups;no

    thrombotic

    and/or

    vascular

    eventswerereported

    1980 Adv Ther (2020) 37:1960–2002

  • Table6

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Braga

    etal.

    (1995)

    [179]

    Preoperative

    stom

    ach

    orcolorectalcancer

    HCT\

    34%

    SF\

    700ng/m

    L

    IS:11

    IS?

    ESA

    :11

    Noiron:11

    (neither

    anem

    icnoriron

    deficient)

    IS:200mgq.d.,1

    2days

    IS?

    ESA

    :200

    mgq.d.,days

    1,4,

    8,12

    IntheIS

    group,no

    patientscoulddonateautologous

    blood

    comparedwithallpatientsin

    theIS

    ?ESA

    group

    36%

    ofpatientsin

    theIS

    groupreceived

    perioperative

    transfusionof

    homologousbloodcomparedwithnone

    intheIS

    ?ESA

    group

    Nomajor

    AEswitheither

    regimen

    Xuet

    al.

    (2019)

    [104]

    Preoperative

    hip

    fracture

    repair

    IS:32

    ESA

    :32

    IS?

    ESA

    :32

    3days

    before

    surgery

    Dosenotstated

    Com

    binedIS

    ?ESA

    was

    moreeffectivethan

    either

    EPO

    orIS

    alone

    HbincreasedsignificantlymoreafterIS

    ?ESA

    than

    after

    either

    EPO

    orIS

    alone

    Nosignificant

    difference

    intheincidenceof

    adversedrug

    reactionsam

    ongthethreegroups

    Kim

    etal.

    (2009)

    [105]

    Menorrhagia

    Hb\

    9.0g/dL

    IS:39

    FeProtSu:37

    200mg,e.o.d.

    t.i.w.

    80mgq.d.,3

    weeks

    Significantlybetter

    Hbincrease

    andresponse

    rate

    withIS

    versus

    FeProtSu

    Twomyalgiaeventsandoneinjectionpain

    eventwithIS;

    oneeventeach

    ofnausea

    anddyspepsiawithFeProtSu;n

    osevere

    AEsreported

    Useofiron

    sucrosein

    theperioperativesetting

    Karkouti

    etal.

    (2006)

    [106]

    Postoperativeanem

    ia

    Hb7–

    9g/dL

    IS:13

    IS?

    ESA

    :12

    Noiron:13

    200mgdays

    1,2,

    3postoperatively

    Withno

    between-groupdifferences,earlypostoperative

    treatm

    entwithIS

    aloneor

    incombination

    withESA

    did

    notappear

    toaccelerateearlyrecovery

    from

    postoperative

    anem

    ia

    Adv Ther (2020) 37:1960–2002 1981

  • Table6

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Madi-Jebara

    etal.

    (2004)

    [107]

    Postoperativecardiac

    surgery

    Hb7–

    10g/dL

    IS:40

    IS?

    ESA

    :40

    Noiron:40

    200mgq.d.

    toreachtotal

    iron

    deficit

    Nosignificant

    difference

    intransfusionrates

    Nosignificant

    difference

    inHbincrease

    PostoperativeIS,w

    ithor

    without

    ESA

    ,isnoteffectiveat

    correcting

    postoperativeanem

    ia

    NoAEswithIS

    Serrano-

    Trenas

    etal.

    (2011)

    [108]

    Perioperativehip

    fracture

    surgery

    IS:99

    Noiron:97

    200mg,39

    e.o.d.

    Nosignificant

    difference

    inRBC

    transfusions

    between

    groups

    3.0%

    AE-related

    treatm

    entsuspension

    withIS

    (one

    skin

    rash,twogeneraldiscom

    fort);overall,14.8%

    infections

    (mainlysuperficialsurgicalwound

    infections

    5.6%

    )

    Garrido-

    Martı́n

    etal.

    (2012)

    [109]

    Perioperativecardiac

    surgery

    Hb\

    8g/dL

    coronary

    patients;\

    7g/dL

    valvesurgerypatients

    IS:54

    Oraliron:53

    Placebo:

    52

    IS:100mg9

    3during

    perioperativeperiod

    Oraliron:105mgq.d.

    pre-

    andpostoperatively,and

    1month

    afterdischarge

    Nobetween-groupdifferencesin

    Hb,HCT,ortransfusions

    NeitherIS

    nororaliron

    therapywereeffectivein

    correcting

    anem

    iaaftercardiacsurgery

    NoAEswithIS

    Shin

    etal.

    (2019)

    [110]

    Perioperative

    orthopedicsurgery

    Meta-analysis

    1869

    patientsfrom

    12clinicaltrials

    Various

    IViron

    PerioperativeIV

    iron

    during

    orthopedicsurgery,especially

    postoperatively,appearsto

    reduce

    theproportion

    ofpatientstransfused

    andun

    itstransfused,w

    ithshorter

    length

    ofhospitalstay

    anddecreasedinfectionrate

    Xuet

    al.

    (2019)

    [111]

    Postoperativecardiac

    valvular

    surgery

    Hb\

    12.0g/dL

    (wom

    en)

    or\

    13.0g/dL

    (men)

    SF30–1

    00ng/m

    Lor

    TSA

    T\

    20%

    IS:75

    Placebo:

    75

    200mge.o.d.,u

    ntiltotal

    iron

    deficiencywas

    achieved

    Hbconcentrationandproportion

    ofpatientswithanem

    iacorrectedor

    achievingHbincrem

    ents[

    2.0g/dL

    were

    significantlygreaterforIS

    than

    placebo14

    days

    postoperatively,butnot7days

    postoperatively

    SFweresignificantlyhigherforIS

    versus

    placebo7daysand

    14days

    postoperatively

    ISwas

    welltolerated:

    noAEsor

    infusion

    reactionswere

    observed

    1982 Adv Ther (2020) 37:1960–2002

  • Table6

    continued

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Muñ

    ozet

    al.

    (2014)

    [112]

    Perioperative

    orthopedicsurgery

    HbC

    10.0g/dL

    IS:1142

    IS?

    EPO

    :351

    FCM:45

    Noiron:1009

    100–

    200mgup

    to39

    (2–5

    days

    preoperatively

    or2–

    3days

    postoperatively)

    Veryshort-term

    perioperativeIV

    iron

    inmajor

    lower

    limb

    orthopedicprocedures

    isassociated

    withreduced

    allogeneicbloodtransfusionratesandlength

    ofhospital

    stay

    Inpatientswithhipfracture,IViron

    isalso

    associated

    with

    areductionin

    postoperativenosocomialinfection

    Nosignificant

    AEswithIV

    iron

    ParejaSierra

    etal.

    (2019)

    [113]

    Perioperativehip

    fracture

    Descriptive

    study

    IS?

    ESA

    :40

    Transfusion:12

    Transfusion

    ?IS

    ?ESA

    :51

    Notreatm

    ent:27

    Hb\

    8.5g/dL

    :transfusion

    Hb8.5–

    10g/dL

    :29

    200mg(48hapart)

    Hb10–1

    1g/dL

    :29

    400mg(48hapart)

    ISdidnotreduce

    theproportion

    oftransfused

    patients

    (56%

    transfused

    vs.44%

    nottransfused

    receivingIS),but

    itdidreduce

    thenu

    mberof

    bloodun

    itsrequired

    Patientswho

    received

    IShadbetterfunctionalrecoverythan

    thosewho

    didnot

    NoAEswithIS

    Montano-

    Pedroso

    etal.

    (2018)

    [114]

    Postoperativebariatric

    abdominoplasty

    Hb\

    11.5g/dL

    SF\

    11ng/m

    L

    IS:28

    Oraliron:28

    IS:200mgim

    mediatelyand

    1daypostoperatively

    Oraliron:100mgb.i.d.

    8days

    Superiorityof

    ISwas

    notshow

    n;theminim

    umclinically

    relevant

    difference

    inHbconcentrations

    wasnotreached

    NoAEsin

    theIS

    group

    Constipation(18%

    ),diarrhea

    (11%

    ),andnausea

    (4%)were

    reported

    withoraliron

    AEadverseevent;b.i.d.twicedaily,b.i.w.twiceweekly,e.o.d.everyotherday,EPO

    erythropoietin,E

    SAerythropoiesis-stimulatingagent,FC

    Mferriccarboxym

    altose,

    FeProtSu

    ferricproteinsuccinylate,FeSferroussulfate,G

    Igastrointestinal,H

    bhemoglobin,

    HCThematocrit,IV

    intravenously

    administered,

    ISiron

    sucrose,q.d.

    everyday,RBCredbloodcell,

    SFserum

    ferritin,t.i.w.three

    times

    weekly,TSA

    Ttransferrinsaturation

    Adv Ther (2020) 37:1960–2002 1983

  • Perioperative Iron TreatmentResults from four randomized controlled trialsdid not find convincing benefits of periopera-tive iron in orthopedic or cardiac surgery[106–109]. In contrast, however, a recent meta-analysis of IV iron therapies, including ironsucrose, showed that perioperative IV irontherapy during orthopedic surgery was associ-ated with several benefits, including reducedtransfusion rates and shorter hospital stays, butwithout impacting the mortality rate [110].Additionally, a separate randomized studyshowed that the use of iron sucrose can signif-icantly increase Hb levels versus placebo controlin patients undergoing cardiac surgery [111].This is important as patients undergoing cardiacsurgery are at risk of postoperative functionaliron deficiency.

    For patients undergoing surgery for fractures,several studies have suggested a benefit of uti-lizing iron sucrose, particularly for hip fractures.A number of improved outcomes have beenassociated with perioperative IV iron (includingiron sucrose) in patients undergoing surgery forhip fracture and arthroplasty, as highlighted ina pooled analysis of observational data, withbenefits including reduced transfusion rates,shorter hospital stays, reduced infection rates(hip fracture only), and lower 30-day mortality(hip fracture only) [112].

    Blood Donors

    Birgegård et al. investigated the frequency andseverity of iron depletion in regular blooddonors and the effects of iron sucrose and oraliron sulfate therapy on iron status [115]. Ironsucrose was found to substitute iron loss inblood donors more efficiently compared withoral iron sulfate treatment, especially inwomen, and also reduced the severity of restlessleg syndrome, which can occur as a complica-tion of iron deficiency.

    Oncology

    A summary of the studies on iron sucrose use inoncology is provided in Table 7.

    Anemia is a common side effect of cancertreatments and ESAs may be used to aid in RBCproduction. In patients with anemia and lym-phoproliferative malignancies who were notreceiving chemotherapy, the addition of ironsucrose to an ESA resulted in a significantlyfaster and greater Hb increase compared withESA therapy alone [116]. Furthermore, usingdata from this study [116], when iron sucrosewas given as an additional therapy, ESA doserequirements were reduced from week 5onward, allowing cost savings over the studyperiod [117].

    In patients with multiple myeloma or lym-phoma who had undergone autologoushematopoietic cell transplantation, supple-mentation of an ESA with iron sucrose increasedthe proportion of patients with Hb responsesfrom 79% to 100%, reduced the medianresponse time, and lowered ESA dose require-ments compared with those receiving no irontreatment [118]. There was no impact of ironsucrose treatment on long-term clinical out-comes or survival [119].

    Utilization of iron sucrose in the absence ofESAs can also benefit some oncology patients byimproving Hb levels and reducing the need forRBC transfusions [120–122].

    Pediatric Populations

    Data on the use of iron sucrose within pediatricpatients are limited. The data that are available,however, suggest that iron sucrose is an effec-tive option (Table 8).

    In children with, or at risk of, iron defi-ciency/iron-deficiency anemia, iron sucroseappears effective in those who cannot tolerateor do not respond to oral iron therapy (recom-mended maximum single dose of 7 mg iron perkg to minimize the risk of AEs) [123]. Whereiron sucrose has been used in children with irondeficiency and iron-deficiency anemia, signifi-cant improvements in Hb levels have beenshown [124, 125].

    Where children also suffer from IBD, the useof oral iron therapy is difficult. In this setting,iron sucrose is effective for the routine man-agement of iron-deficiency anemia. Children

    1984 Adv Ther (2020) 37:1960–2002

  • Table7

    Publishedclinicalstudiesusingiron

    sucrosein

    oncology

    Stud

    yInclusioncriteria

    Patients,n

    Iron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Hedenus

    etal.(2007)

    [116]

    Clin

    ically

    stablelymphoproliferaltive

    malignancy

    Hb9.0–

    11.0g/dL

    IS?

    ESA

    :33

    ESA

    :34

    100mgq.w.(weeks

    0–6);100mgq.2.w.

    (weeks

    8–14)

    16weeks

    (follow-up)

    SignificantlygreaterHbincreasefrom

    week8onwardwithIS

    versus

    noIS

    Proportion

    ofpatientswithHbincrease

    C2g/dL

    wassignificantly

    higher

    withIS

    (93%

    )than

    noIS

    (53%

    ;p=0.001)

    ESA

    dose

    lower

    withIS

    than

    noIS

    from

    week5onward

    AEsweredistributedevenlybetweengroups

    Beguinet

    al.(2013)

    [118]

    AutologousSC

    T

    SF[

    100ng/m

    L

    IS?

    ESA

    :23

    ESA

    :25

    NoESA

    :24

    200mgon

    days28,42,

    and56

    afterSC

    TCom

    pleteHbresponse

    within18

    weeks

    achieved

    by100%

    with

    IS?

    ESA

    ,79%

    withESA

    ,21%

    withno

    ESA

    Erythropoieticresponse

    significantlyhigher

    withIS

    ?ESA

    than

    ESA

    ,resulting

    inlower

    ESA

    use,reduceddrug

    costs,and

    improved

    qualityof

    lifescores;effect

    ontransfusions

    was

    not

    significant

    Safety

    findingsweresimilarbetweengroups

    Jasperset

    al.(2015)

    [119]

    [Long-term

    follow-up

    ofBeguinet

    al.

    (2013)

    [118]]

    AutologousSC

    T

    SF[

    100ng/m

    L

    IS?

    ESA

    :50

    ESA

    :52

    NoESA

    :25

    1.4years(m

    edian

    follow-up)

    Overallsurvival(1-yearand5-year)andprogression-free

    survival(1-

    year

    and5-year)notsignificantlydifferentbetweengroups

    Incidenceof

    infections

    remainedcomparablebetweengroups

    Other

    seriouscomplications

    wereun

    common

    andhardly

    attributableto

    ESA

    orIS

    Dangsuw

    anand

    Manchana(2010)

    [120]

    Hb\

    10.0g/dL

    IS:22

    FeS:

    22

    IS:200mg

    FeS:

    200mgt.i.d.

    Significantlyhigher

    Hbandsignificantlyfewer

    RBC

    transfusions

    withIS

    than

    FeS

    Mostcommon

    AEsweremild

    nausea

    andvomiting;no

    significant

    difference

    inAEsbetweengroups;no

    SAEsor

    hypersensitivity

    reactions

    Kim

    etal.(2007)

    [121]

    Hb\

    12.0g/dL

    IS:30

    Noiron:45

    19

    200mgperCT

    cycle

    Significant

    reductionin

    RBC

    transfusions

    withIS

    versus

    noiron

    Notreatm

    ent-relatedAEswithIS;transfusion-relatedallergic

    reactionswithsimilarfrequencyin

    both

    groups

    AEadverseevent,CTchem

    otherapy,E

    SAerythropoiesis-stimulatingagent,FeSferroussulfate,Hbhemoglobin,

    ISiron

    sucrose,q.2.w.every2weeks,q.w.every

    week,

    RBCredbloodcell,

    SAEseriousadverseevent,SC

    Tstem

    celltransplantation,

    SFserum

    ferritin,t.i.d.

    threetimes

    daily

    Adv Ther (2020) 37:1960–2002 1985

  • Table8

    Publishedclinicalstudiesusingiron

    sucrosein

    pediatricpatients

    Stud

    yInclusioncriteria

    Patients,

    nIron

    dose

    (mg)/dose,

    interval,treatm

    ent

    duration

    Outcomes

    andsafety

    inform

    ation

    Crary

    etal.

    (2011)

    [123]

    AgedB

    18years

    C1dose

    ofIS

    Patientscouldnothave

    CKD

    IS:38

    Mediandose:100mg

    Mediannu

    mberof

    infusions:3

    Patientshadagood

    response

    toIS,w

    ithamedianHbrise

    of1.9–

    3.1g/dL

    depend

    ingon

    theindication

    ISwaswelltolerated

    withonlyoneseriousreaction

    (tem

    poraryandreversible

    hypotension)

    inapatientwho

    hadreceived

    IS500mg,which

    was

    greater

    than

    therecommendeddose

    of300mg

    Akarsuet

    al.

    (2006)

    [124]

    SF\

    12ng/m

    L

    Low

    Hb(adjustedfor

    age)

    IS:62

    Variabledepen