You are on page 1of 10

Narrative Review

New Treatment Approaches for the Anemia of CKD


Mario Bonomini, MD,1 Lucia Del Vecchio, MD,2 Vittorio Sirolli, MD,1 and
Francesco Locatelli, MD2

Normocytic normochromic anemia is a common complication in chronic kidney disease and is associated
with many adverse clinical consequences. Erythropoiesis-stimulating agents (ESAs) and adjuvant iron therapy
represent the primary treatment for anemia in chronic kidney disease. The introduction of ESAs into clinical
practice was a success story, mediating an increase in hemoglobin concentrations without the risk for recurrent
blood transfusions and improving quality of life substantially. However, recombinant ESAs are still expensive
and require a parenteral route of administration. Moreover, concern has arisen following randomized clinical
trials showing that higher hemoglobin targets and/or high ESA doses may cause significant harm. This,
together with changes in ESA reimbursement policy in some countries, has resulted in a significant reduction in
ESA prescribing and the hemoglobin level targeted during therapy. Several attempts are being made to
develop new drugs with improved characteristics and/or easier manufacturing processes compared with
currently available ESAs, including new treatment approaches that may indirectly improve erythropoiesis. We
give an update on the new investigational strategies for increasing erythropoiesis, examining in depth their
characteristics and possible advantages in the clinical setting and the caveats to be aware of at the present
stage of development.
Am J Kidney Dis. 67(1):133-142. 2016 by the National Kidney Foundation, Inc.

INDEX WORDS: Chronic kidney disease (CKD); renal anemia; erythropoiesis; erythropoiesis stimulating
agent (ESA); erythropoietin (EPO); hypoxia-inducible factor (HIF); HIF stabilizer; prolyl hydroxylase
inhibitor; EPO receptor; gene therapy; activin trap; end-stage renal disease; chronic renal insufficiency;
dialysis; review.

N ormocytic normochromic anemia is one of the


hallmarks of progressive chronic kidney dis-
ease (CKD).1 It is mainly due to an absolute or rela-
with frequent subcutaneous administrations, this
may be cumbersome for long-term treatment in
nondialysis-dependent patients with CKD. In hemo-
tive decrease in erythropoietin (EPO) production by dialysis (HD) patients, the issue is more complex
the failing kidney. However, its pathogenesis is much because intravenous administration increases nurse
more complex. Several other factors (iron and vitamin workload but improves treatment adherence. In addi-
deciency, infection, inammation, occult blood loss, tion, in the past several years, clinical trials4-6 have
oxidative stress, inadequate dialysis, and hyperpara- shown that higher Hb targets and/or application of
thyroidism) often contribute to anemia development high ESA doses may increase cardiovascular risk.
and reduce response to treatment. This, together with changes in ESA reimbursement
The introduction of erythropoiesis-stimulating policy in some countries, has led to reductions in
agents (ESAs) has revolutionized the care of anemic prescribed ESA dose7 and target Hb levels.8,9 Hence,
patients with CKD and almost completely eradicated an increase in blood transfusion requirements and
the severe anemia of end-stage renal disease (ESRD).2 intravenous iron dosing has occurred. Blood trans-
Moreover, ESAs decrease the risk for recurrent blood fusions cannot be considered as an alternative strategy
transfusions and iron overload and may improve to ESAs because they still have some risks, expose
quality of life. The rst-generation ESAs were human patients to large Hb-level uctuations, and by deni-
recombinant EPOs (epoetin alfa and epoetin beta). tion have limited availability. Moreover, they may
Subsequently, 2 second-generation ESAs with a longer
duration of action were developed; darbepoetin alfa
and methoxy-polyethylene glycol-epoetin beta. Bio- From the 1Unit of Nephrology and Dialysis, Department of
similar epoetins, together with epoetin theta, have also Medicine, G. dAnnunzio University, Chieti-Pescara; and 2Depart-
ment of Nephrology and Dialysis, Alessandro Manzoni Hospital,
received marketing authorization in many countries.3 Lecco, Italy.
Today, ESAs and adjuvant iron therapy are the Received April 10, 2015. Accepted in revised form June 29,
main tools for treating the anemia associated with 2015. Originally published online September 12, 2015.
CKD. Available ESAs are very effective drugs, Address correspondence to Mario Bonomini, MD, Nephrology
usually obtaining signicant hemoglobin (Hb) level and Dialysis Unit, SS Annunziata Hospital, Via dei Vestini, 66013
Chieti, Italy. E-mail: m.bonomini@nephro.unich.it
increases. However, recombinant ESAs are still  2016 by the National Kidney Foundation, Inc.
expensive and require cold storage. In addition, they 0272-6386
are administered by the parenteral route. Especially http://dx.doi.org/10.1053/j.ajkd.2015.06.030

Am J Kidney Dis. 2016;67(1):133-142 133


Bonomini et al

enhance the synthesis of alloantigenic antibodies, Box 1. New Strategies Under Development to Stimulate
reducing the likelihood of patients subsequently Erythropoiesis
receiving a kidney transplant. Excessive iron use may Not Directly Targeting the EPO Receptor
cause harm as well.10  HIF stabilizers
In the last 2 decades, recombinant forms of EPO  FG-4592 (Roxadustat)
have been engineered to have structural modications  AKB-6548
 GSK1278863
that increase half-life and accommodate a wider  BAY 85-3934 (Molidustat)
administration schedule. However, health systems  JTZ-951
cannot afford to pay more for a secondary advantage,  DS-1093a
as attested by the limited success of methoxy-  Activin traps

polyethylene glycol-epoetin beta in the European  Sotatercept (ACE-011)


 Luspatercept (ACE-536)
market (though this may have been compounded by a  LY2157299
several-month shortage of the drug). Moreover, the
experience with peginesatide (discussed later) has Targeting the EPO Receptor
 EPO mimetic peptides
highlighted that unexpected adverse events may  Centocor molecules: CNTO 528, CNTO 530, CNTO 531
abruptly halt either the development or commerciali-  AplaGen GmbH: AGEM400(HES)
zation of a given molecule after years of investment.  Peginesatidea
The task is becoming increasingly difcult because  EPO fusion proteins

regulatory authorities rightly require more and more  EPO-EPO dimers


 EPO-CPT
data for safety and hard end points following the  EPO-(CPT)3
concerns surrounding the safety of existing ESAs with  Albumin-EPO
regard to cardiovascular events and cancer worsening.  EPO-hyFc (Genexine GX-E2)
Despite this, the ESA market is huge and still very  Antibody agonists to EPO receptor

appealing from an economic standpoint. Attempts are  Mouse monoclonal IgG


 Ab12 molecule
being made to develop new molecules with improved  Ab12.6 (Abbott Laboratories ABT-007) molecule
characteristics and/or easier manufacturing processes  EPO gene therapy (TARGT EPO)
than those currently available. Their success when  Dimerization of EPO receptor intracellular domain with a CID
entering the ESA market will depend on the balance
Abbreviations: CID, chemical inducer of dimerization; CPT,
among price, advantages, and possible risks. There carboxyl-terminal peptide; EPO, erythropoietin; HES, hydrox-
remains the general sentiment that new treatment yethyl starch; HIF, hypoxia-inducible factor; IgG, immunoglobulin
strategies are needed with a better safety prole than G; TARGT, transducer autologous regenerative gene therapy.
a
existing ESAs. Withdrawn from the market.
In this review, we provide an update on the new
treatment approaches for increasing erythropoiesis HIF2-a is expressed in a cell-restricted manner and is
(Box 1). We report in particular on the new erythro- the key regulator of EPO synthesis and iron meta-
poietic strategies currently in clinical development. bolism.28-30 Under normal oxygen conditions, HIF-a
Agents that have not yet reached this phase are subunits are prolyl hydroxylated by PHD1, PHD2, and
described in Table 1.11-25 PHD3 (2-oxoglutarate-dependent oxygenases that
contain a prolyl-4-hydroxylase domain [PHD]). By
NEW ERYTHROPOIETIC STRATEGIES NOT contrast, under hypoxia, PHD activity decreases and
DIRECTLY TARGETING THE EPO RECEPTOR HIF-a accumulates. HIF-a can then bind to the HIF-b
subunit, resulting in the activation of a large array of
Hypoxia-Inducible Factor Stabilizers target hypoxia-responsive genes.31
Hypoxia-inducible factors (HIFs) coordinate the HIF stabilizers are 2-oxoglutarate competitors that
physiologic response to systemic hypoxia by altering are designed to prevent HIF-a degradation. Thus, HIF
gene expression in certain cell types, leading to an stabilizers primarily function by mimicking the
increase in EPO production in the kidney and liver, hypoxia-driven expression of endogenous EPO in the
improved uptake and use of iron, and changes to the kidney.32 Note that in response to a hypoxic stimulus,
bone marrow microenvironment that encourage damaged kidneys are still able to produce EPO, as
erythroid progenitor maturation and proliferation.26 shown in patients with CKD33 and in experimental
HIFs are heterodimers consisting of an oxygen- models.34 In addition, pharmacologic HIF activation
sensitive a subunit and a constitutively expressed b in adults stimulates the liver to retain its ability to
subunit. There are 3 HIF a subunits in mammals. produce EPO.34,35
HIF1-a is ubiquitous and together with HIF2-a facil- A feature unique to HIF stabilizers is that they are
itates oxygen delivery and cellular adaptation to hyp- orally administered, which may be particularly helpful
oxia by stimulating a number of biological processes.27 in nondialysis-dependent patients with CKD and

134 Am J Kidney Dis. 2016;67(1):133-142


New Treatment Approaches for the Anemia of CKD

Table 1. New Erythropoiesis-Stimulating Agents Not in Clinical Development

Mechanism of
Agents Drugs Action Summary of Preclinical Data Reference

EPO mimetic peptides AGEM400(HES), CNTO 530, EPO receptor Efficient stimulation of hematopoietic 11-14
CNTO 531 activation progenitor cells
EPO fusion proteins EPO-EPO dimers, EPO-CPT, EPO receptor Increased erythropoietic activity and half-life 15-21
EPO-(CPT)3, albumin-EPO activation compared to the native endogenous
hormone in vitro and in mice
Agonistic antibodies to Mouse IgG, Ab12, Ab12.6 EPO receptor Relatively poor EPO receptor activation by 22-24
the EPO receptor (ABT-007) activation mouse monoclonal antibodies; in vitro
potency and in vivo erythropoietic efficacy
in mice by human agonistic antibodies
Ab12 and Ab12.6
Inducers of dimerization Chemical inducer of EPO receptor Exclusive stimulation of erythroid cells upon 25
of EPO receptor dimerization (CID) activation perioral administration to transgenic mice
intracellular domain
Note: See Box 1 for expansions of abbreviations.

those treated with peritoneal dialysis. Moreover, these assigned 1:1:1 to FG-4592 low or high dose or pla-
agents induce physiologic EPO levels, unlike the cebo. At the end of the 8-week treatment period,
abnormally high peak concentrations usually observed participants showed mean maximum Hb level in-
with standard ESA therapy, which may be harmful.36 creases from baseline of 2.6 g/dL in the high-dose
The rst promising molecule of this class was cohort and 1.8 g/dL in the low-dose cohort,
FG-2216 (FibroGen Inc), which was withdrawn compared to 0.7 g/dL in the placebo group. The
following a case of fatal hepatitis. The same com- percentage of patients achieving Hb levels $ 11 g/dL
pany then developed roxadustat (FG-4592). In 117 was higher in those receiving the active drug than
nondialysis-dependent patients with CKD, those who placebo. Interestingly, patients who received roxadu-
were randomly assigned to roxadustat (4 doses esca- stat showed small decreases in blood pressure similar
lating from 0.7 to 2.0 mg/kg administered 2 or 3 times to the placebo group. In still another study, FG-4592
weekly) had a higher Hb response rate than those was found effective in achieving anemia correction in
receiving placebo. Roxadustat also achieved good ESA-naive patients receiving peritoneal dialysis, with
anemia correction (Hb increased on average by 2 g/dL a safety prole similar to the HD population.42
in 3 months) in ESA-naive HD patients who had se- Akebia Therapeutics is developing another prolyl
vere anemia and were iron decient in many cases.37 hydroxylase inhibitor, AKB-6548. After successfully
According to the interim analysis of another phase undergoing phase 2a clinical development in CKD
2 study of HD patients, FG-4592 produced an in- stages 3 and 4, the drug has recently been evaluated in
crease in mean Hb levels of w1 g/dL from baseline in a phase 2b, randomized, double-blind, placebo-
the 2 cohorts receiving the highest doses (1.5 and controlled study.43 This 20-week study enrolled 209
2.0 mg/kg). Conversely, those randomly assigned to nondialysis-dependent patients with CKD stages 3 to
recombinant human EPO experienced a slight 5 who were randomly assigned 2:1 to active treatment
decrease in Hb levels during the 6-week follow-up or placebo. The initial dose was 450 mg once daily,
period.38 In FG-4592treated patients, the improve- with dose adjusted in accordance to the patients Hb
ment in erythropoiesis accompanied a reduction in level response based on the AKB-6548 titration al-
serum hepcidin levels. It is still unknown whether this gorithm. Preliminary results showed that the primary
is a direct or indirect effect related to HIF stabiliza- end point, dened as achieving or maintaining mean
tion. Excess hepcidin is the main cause of functional Hb levels $ 11 g/dL or increasing Hb $ 1.2 g/dL
iron deciency and iron-restricted erythropoiesis in above the pretreatment value, was met in 54.9% who
patients with CKD.39,40 Whatever the underlying received AKB-6548 compared with 10.3% in the
mechanism(s), the possibility that HIF stabilization placebo group. The drug was generally well tolerated.
may decrease hepcidin levels and possibly improve A phase 2 open-label study designed to examine the
iron utilization is promising. In both the above efcacy, safety, and tolerability of AKB-6548 in HD
studies, no liver toxicity was reported. patients is ongoing (ClinicalTrials.gov identier
More recently, data were presented about another NCT02260193).
phase 2 double-blind placebo-controlled trial in GlaxoSmithKline are also developing an HIF
China41 in which 91 nondialysis-dependent patients prolyl hydroxylase inhibitor (GSK1278863). In 2011,
with CKD with Hb levels , 10 g/dL were randomly they concluded a phase 2a, randomized, single-blind,

Am J Kidney Dis. 2016;67(1):133-142 135


Bonomini et al

placebo-controlled, parallel-group study aiming to multiple-ascending-dose, phase 1 study, designed to


evaluate the safety, efcacy, and pharmacokinetics of determine the safety, tolerability, pharmacokinetics, and
GSK1278863 administered in 28-day oral repeat doses pharmacodynamics of daily administration for 15 days
of 10 to 100 mg to 70 anemic nondialysis-dependent to HD patients.49 DS-1093a (Daiichi Sankyo Inc) is
patients with CKD and 37 HD patients.44 Hb levels undergoing evaluation in a phase 1 study of patients with
increased in all dose groups, though the increase in Hb CKD stage 3b or 4, an open noncontrolled parallel-group
level was excessive in a number of participants and led investigation of 3 single doses (6 participants per dose;
to drug treatment being discontinued. Dose-dependent dose allocation being randomly assigned), which is
increases in endogenous EPO were found, the highest currently recruiting participants.50
levels occurring in the 100-mg group. Following A potential downside to prolyl hydroxylase inhibi-
GSK1278863 administration, signicant decreases in tion is that HIF transcription factors are involved in the
hepcidin and ferritin levels were also observed. Prob- regulation of various biological processes. Intermittent
ably due to the excessive increase in Hb levels HIF activation over prolonged periods may have
observed in some patients in the previous trial, an adverse effects, including changes in glucose levels,
open-label parallel-group study has been carried out to fat and cholesterol metabolism, and angiogenesis
evaluate the pharmacokinetics of GSK1278863 (at the promotion.51-54 The primary fear is promotion of tu-
lower dose of 5 mg) and its metabolites (predominant mor growth.55,56 In particular, careful evaluation is
metabolites may contribute to the clinical pharma- being applied to possible treatment-related increases in
cology of the parent drug and may accumulate upon vascular endothelial growth factor (VEGF). This is a
repeat-dose administration in CKD) in healthy in- hypoxia-induced angiogenic protein that exhibits a
dividuals and patients with decreased kidney func- broad range of biological and pathologic effects,
tion.45 The drug was administered once daily for 14 ranging from neoangiogenesis promoting tumor
days (healthy individuals and patients with CKD growth to macular degeneration and worsening of
stages 3 and 4) or 15 days (HD patients). Results proliferative diabetic retinopathy. At present, pre-
showed that the area under the curve (a measure of the liminary data do not suggest that VEGF level increases
drug concentration in plasma) of the parent compound following treatment with HIF stabilizers. Another
is similar in healthy individuals and patients with possible concern is that HIF inhibitors simulate a
CKD; the steady-state metabolite area under the curve chronic hypoxic state, as occurs in people living at
was 1.6- to 2.9-fold higher (depending on the metab- high altitude. The long-term consequence of this is
olite) in patients with CKD, further increasing (2.7- to unknown, but patients with CKD receiving HIF in-
6.2-fold higher) in HD patients on a nondialysis day. hibitors are unlikely to develop symptoms of chronic
Though the parent drug was not cleared by HD, mountain sickness because those symptoms are mainly
exposure to all metabolites was 55% lower on driven by excessive erythrocytosis. Conversely, HIF
GSK1278863 administration on a dialysis day than a inhibitors may cause ongoing enhancement of several
nondialysis day.45 Phase 2b studies with GSK1278863 pathways, including VEGF, that might cause systemic
are active but not yet recruiting.46 vascular dysfunction and predispose to increased sys-
Molidustat (BAY 85-3934; Bayer Pharma) is another temic cardiovascular morbidity.57 The fact that pa-
novel HIF prolyl hydroxylase inhibitor. A recent tients with heart disease may go safely to high altitudes
comprehensive pharmacologic characterization showed for intermittent periods is reassuring.58 An intermittent
the compound to be effective in reversing anemia in hypoxic state may be even an advantage, as suggested
animal models of kidney and inammatory anemia.47 by experimental studies showing improved recovery of
No dose-related changes were found in kidney and postmyocardial infarction function with intermittent
liver samples in the expression of hypoxia-responsive hypobaric hypoxia.59
genes other than EPO. In addition, BAY 85-3934 dis- The outlook for these molecules is still unclear.
played antihypertensive effects in a CKD model.47 They may be an alternative option to treat anemia
Preliminary results of a randomized, single-blind, pla- when used alone or in combination with ESA and/or
cebo-controlled, dose-escalation study of single oral iron. Their main advantage could be that of stimu-
doses (5, 12.5, 25, 37.5, and 50 mg) of BAY 85-3934 in lating erythropoiesis with EPO at physiologic con-
healthy men showed rapid absorption (mean terminal centrations. However, it will need to be proved that
half-life, 4.6-10.4 hours), a dose-dependent increase in they are as safe or even safer than existing ESAs.
endogenous EPO, and an increase in reticulocyte count Accordingly, phase 3 studies will require the enroll-
after 37.5- and 50-mg dose administration.48 Phase 2b ment of thousands of patients to be adequately pow-
studies in patients with anemia of CKD are ongoing. ered to test safety.
At least 2 further HIF stabilizers are currently being HIF stabilizers are much further along in clinical
developed. JTZ-951 (Akros Pharma Inc) has comp- development than most other new therapeutic strate-
leted a randomized, single-blind, placebo-controlled, gies to increase erythropoiesis (Table 2), although data

136 Am J Kidney Dis. 2016;67(1):133-142


New Treatment Approaches for the Anemia of CKD

Table 2. Hypoxia-Inducible Factor Stabilizers Under Clinical Development

Drug Company Stage of Clinical Development

FG-4592 (Roxadustat) Fibrogena Phase 2 studies completed (NDD CKD, HD, PD); phase 3 studies ongoing
(NDD CKD, HD, PD)
AKB-6548 Akebia Therapeutics Phase 2 studies completed (NDD CKD) or ongoing but not recruiting (HD)
GSK1278863 Glaxo Smith Kline Phase 2a studies completed (NDD-CKD, HD); phase 2b studies ongoing, but
not recruiting (NDD CKD)
BAY 85-3934 (Molidustat) Bayer Pharmaceuticals Phase 2b studies ongoing (NDD CKD, HD)
JTZ-951 Akros Pharmaceuticals Phase 1 study completed (HD)
DS-1093a Daiichi Sankyo Phase 1 study ongoing (CKD 3b-4)
Abbreviations: CKD, chronic kidney disease; HD, hemodialysis; NDD, nondialysis-dependent; PD, peritoneal dialysis.
a
Collaboration with Astra Zeneca in the United States and China and with Astellas Pharma for Europe, Japan, the Middle East, and
South Africa.

regarding the use of HIF prolyl hydroxylase inhibitors induce erythroid differentiation.67,68 Two potential
in kidney anemia come only from abstracts. Despite nonexclusive indirect mechanisms have been proposed
the promise of a new class of erythropoietic com- for the effects of sotatercept on human erythropoiesis.69
pounds, careful development is still required due to The rst relates to the binding of ACTRIIA ligands,
the wide range of biological pathways in which HIF resulting in modulation of their function regarding
transcription factors are involved. erythroid development. As a second possible mecha-
nism, by neutralizing TGFb family members, sota-
Activin Traps tercept can modulate the SMAD signaling pathway in
Activins are dimers of inhibin b-type chains and stromal cells, leading to changes in the transcription of
belong to the transforming growth factor b (TGFb) SMAD target genes that encode proteins affecting
superfamily. They trigger growth and differentiation erythroid development.70 For example, the expression
by binding to type I and type II serine-threonine kinase of angiotensin II, which can stimulate erythropoiesis
receptors, which are transmembrane proteins.60 Acti- directly and indirectly by EPO production, is increased,
vin, bone morphogenic proteins, and several other whereas the expression of VEGF, considered an in-
members of the TGFb family contribute to regulation hibitor of erythropoiesis, is suppressed.69 Sotatercept is
of erythropoiesis either by directly affecting erythroid currently being evaluated in patients with CKD 5D
progenitor or precursor cells or by altering the (ClinicalTrials.gov identier NCT01146574) in a
behavior of bone marrow accessory cells.60-62 These phase 2a, randomized, double-blind, placebo-
actions seem to be mediated by a signaling pathway controlled, single-dose (0.1 mg/kg subcutaneously)
involving the SMAD proteins.60 Note that activin A is study, followed by a double-blind, placebo-controlled,
identical to the erythroid differentiation factor, multiple-dose, dose-escalation study (starting doses of
which is able to cause differentiation of immature 0.3, 0.5, or 0.7 mg/kg subcutaneously every 28 days in
erythropoietic progenitors into mature cells.63 a sequential design for up to 8 doses). The rst results of
Sotatercept (ACE-011; Acceleron and Celgene part 2 of this study have recently been presented.71 At
Corp) is a dimeric fusion protein in which the extra- each dose, sotatercept was well tolerated and showed a
cellular domain of the activin receptor type IIA safety prole comparable to placebo, with a mean
(ACTRIIA) is linked to the Fc portion of the human elimination half-life of 21 to 26 days. The mean peak
immunoglobulin G1 (IgG1) antibody. Sotatercept traps Hb response in the rst 28 days was dose related, the
circulating activin and other members of the TGFb highest response (1.0 g/dL) being observed in the
superfamily that signal through ACTRIIA.64 In a group treated with sotatercept at a 0.7-mg/kg dose.
double-blind phase 1 trial of healthy postmenopausal Based on observed changes in Hb levels, drug admin-
women, treatment with a single intravenous injection of istration once every 2 weeks instead of once every 4
sotatercept was accompanied by enhanced bone for- weeks has been suggested.72 This is currently being
mation and decreased bone resorption.65 Surprisingly, investigated in a phase 2 study (ClinicalTrials.gov
this trial showed an unanticipated increase in Hb level, identier; NCT01999582), entailing intravenous and
red blood cell number, and hematocrit.65 Treatment subcutaneous administration of escalating doses of
with sotatercept also increased Hb and hematocrit levels sotatercept every 2 weeks in HD patients.
in a phase 2 trial of patients with multiple myeloma in Sotatercept also has other activities that are of po-
parallel with improvement in bone lesions.66 tential relevance in the CKD setting. The drug can inhibit
The clinical observation that an activin-sequestering hepcidin transcription in hepatocytes73 because the
agent such as sotatercept stimulates erythropoiesis is hepcidin promoter contains bone morphogenic protein
hard to reconcile with the reported ability of activin to responsive SMAD-binding elements.74 Activin B has

Am J Kidney Dis. 2016;67(1):133-142 137


Bonomini et al

been shown to play a critical role in hepcidin induction EPO receptor though their amino acid sequence is
by inammation75 and therefore represents a specic completely different from the hormone.85,86 EMP-1,
target for treatment of anemia due to inammation.72 an oligopeptide containing 20 amino acids joined by
Sotatercept has demonstrated stimulating effects on a disulde bridge between 2 cysteine residues, was
bone formation,64 which are believed to be mediated synthesized in 1998.87 However, its short in vivo half-
through its interaction with activin.76 The administra- life and relatively weak binding to the EPO receptor
tion of ACE-011 and its murine counterpart RAP-011 hampered its clinical use.
cause bone loss and osteoporosis to be reversed in In order to increase effectiveness, peptide di-
various animal models.77,78 In CKD mouse models for mers that use either chemical linkers or polymeric
vascular calcication, RAP-011 inhibits SMAD- polyethylene glycol linkers have been constructed.
dependent signaling, blocks aortic osteoblastic transi- The CNTO molecules (Centocor) were developed
tion, increases vascular smooth muscle function, and by using the MIMETIBODY platform, in which a
decreases CKD-stimulated vascular calcication.79,80 given active peptide is connected with an amino
The effects of sotatercept on bone mineral density acid linker to an immunoglobulin Fc domain. The
and vascular calcication were evaluated by using peptide remains capable of interacting with its
quantitative computed tomography81 in the above receptor, while extended pharmacologic properties
mentioned study in HD patients.71 A small interim data are provided by the Fc fragment. Fc fusion tech-
set showed an apparent dose effect on multiple pa- nology has been introduced to generate long-
rameters of CKDmineral bone disorder, including acting antagonistic drugs, such as those targeting
increasing femoral neck cortical bone mass, decreasing TNFR (tumor necrosis factor receptor) and CTLA-
lumbar spine bone mass, and slowing progression of 4 (cytotoxic T-lymphocyteassociated antigen
vascular calcication.82 Considerable differences in 4).88 CNTO 528, obtained by fusion of 2 EMP-1
baseline characteristics of patients may inuence these molecules with a human IgG1 Fc domain, stimu-
results, and the number of participants was small. Thus, lated erythropoiesis after a single intravenous
though sotatercept may address unmet needs in ESRD administration at ascending doses in phase 1
with favorable effects on CKDmineral bone disorder, studies in healthy volunteers.89,90
these results need to be conrmed in larger populations. Peginesatide (originally named Hematide; Affymax/
Another investigational ligand-trapping fusion pro- Takeda), a small dimeric peptide conjugated to a pe-
tein (Luspatercept; ACE-536; Acceleron/Celgene gylated moiety, was approved by the US Food and
Corp) containing the extracellular domain of human Drug Administration in 2012 for treatment of anemic
activin receptor type IIB (ACTRIIB) modied to reduce adult patients receiving dialysis. It is a long-acting
activin binding shows erythropoietic activity and pro- ESA, with dose requirements irrespective of the
motes the maturation of late-stage erythroid precursors administration route. The drug corrected anemia
in vivo.83 Interestingly, treatment with EPO and ACE- effectively in large studies carried out in both non
536 produces a synergistic erythropoietic response.83 dialysis-dependent patients with CKD91 and those
A phase 1, ascending-dose, randomized, double-blind, with chronic kidney failure treated by dialysis.92
placebo-controlled, clinical trial of ACE-536 in Concerns about cardiovascular safety were raised in
healthy volunteers has recently been completed.84 view of higher incidences of death, unstable angina,
Thirty-two postmenopausal women were randomly and arrhythmia in nondialysis-dependent patients
assigned in sequential cohorts of 8 participants each to with CKD treated with peginesatide.91 However, they
receive up to 2 doses of ACE-536 (0.0625-0.25 mg/kg) were not conrmed by a prespecied combined
or placebo (3:1 randomization) administered subcuta- analysis of the dialysis and nondialysis trials. Given
neously every 2 weeks. ACE-536 was well tolerated and its EPO-unrelated structure, the drug was also used
no serious or severe adverse events occurred. Hb levels for the treatment of EPO-related pure red cell apla-
increased dose dependently, beginning 7 days after sia.93 Just one year after commercialization, Affymax
treatment initiation, and were maintained for several and Takeda instituted a voluntary recall of the drug
weeks following treatment.84 These ndings support following postmarketing reports of serious acute hy-
ongoing phase 2 clinical trials of ACE-536 in patients persensitivity reactions that could be life-threatening
with b-thalassemia and myelodysplastic syndromes. or fatal.94 Approximately 0.02% of patients re-
ceiving the rst dose of the drug intravenously died.
NEW ERYTHROPOIETIC STRATEGIES TARGETING The cause of the hypersensitivity reactions has not
THE EPO RECEPTOR been claried.
EPO Mimetic Peptides Epoetin Fusion Proteins
EPO mimetic peptides (EMPs) are a group of A number of attempts have been made to create
synthetic cyclic peptides capable of stimulating the EPO fusion molecules with enhanced erythropoietic

138 Am J Kidney Dis. 2016;67(1):133-142


New Treatment Approaches for the Anemia of CKD

activity (Box 1). The sole compound in clinical CONCLUSIONS


development is GX-E2 (EPO-hyFc; Genexine), in The development of new strategies to treat anemia is
which EPO is fused with a novel hybrid Fc (hyFc) still an evolving and fascinating area of experimental
consisting of human IgG4 plus the hinge and the and clinical research. At present, the most promising
amino-terminus of the IgD heavy chain isotype 2, class of agents seems to be HIF stabilizers, as evident
which replace the corresponding regions of IgG4 in the number of molecules currently under develop-
without linker amino acids being added.95 Of note, ment. This class of drug stimulates erythropoiesis by
IgD has the highest hinge-fold exibility among physiologic concentrations of endogenous EPO,
human immunoglobulins.96 HyFc is nonimmuno- which may translate into a clinical advantage because
genic, noncytolytic, and exible,95 thus overcoming concerns for ESA safety are higher at the high doses.
shortcomings of both IgG1 Fc (antibody-dependent However, these theoretical advantages will need to be
cellular cytotoxicity or complement-dependent demonstrated clinically in large trials. Conversely, the
cytotoxicity of target cells97) and IgG4 Fc (genera- class needs to be proved safe in light of the potential
tion of monovalent half-molecules by formation of 2 risks for increases in levels of VEGF and related fac-
intrachain disulde bonds98). In vitro and in rats, tors and the possibility of widespread stimulation of
GX-E2 shows higher bioactivity than darbepoe- complex pathways leading to unexpected side effects.
tin.99 The safety, tolerability, pharmacokinetics, The nal judgment of benet/risks of these agents will
and pharmacodynamics of GX-E2 were examined be possible only after the completion of large long-
in a randomized, double-blind, placebo-controlled, term safety studies testing hard end points.
phase 1 study recently completed.100 The drug is EMPs were a promising class. They had the ad-
currently under evaluation in a phase 2 randomized vantages of long-acting ESAs with the potential of
study, darbepoetin being the active comparator, being less expensive than short-acting ESAs. Unfor-
aiming to explore the optimal starting dose and tunately, the experience with peginesatide and the fact
dosing interval (as well as safety) in anemic dialysis that the cause of these adverse events has not yet been
patients.101 discovered seems to be halting development of the
EPO Gene Therapy molecule and seriously endangering further develop-
ment of other agents in that class. Sotatercept, which
Medgenics has developed a novel technology for traps circulating activin, is another interesting drug
the sustained production and delivery of therapeutic given its potential for not only correcting anemia but
proteins (like EPO) using ex vivo gene therapy and also checking osteoporosis. This may be of impor-
the patients own tissue. The technology is based on tance considering that the CKD population is
taking a small tissue explant from the patients skin, becoming older and frailer.
transducing the cells with a viral vector for ongoing
production of EPO, and subcutaneously implanting ACKNOWLEDGEMENTS
this dermal pump back into the patient. Following Support: None.
initial favorable observations in severe combined Financial Disclosure: Dr Del Vecchio has served on the advi-
immunodeciency mice102 and technical improve- sory board of Astellas. Dr Locatelli has served on the advisory
ments (preparation of the second-generation viral boards of Akebia, Amgen, Astellas, Fibrogen, Genzyme, GSK,
Fresenius Medical Care, Janssen Cilag, Pharmacosmos, Keryx,
vector and development of a new implantation pro- and ZS Pharma and has been a speaker at a meeting supported by
tocol), the TARGT (Transducer Autologous Regen- Amgen, Asahi Casei, Roche, and ZS Pharma. The remaining au-
erative Gene Therapy) system (formerly known as thors declare that they have no relevant nancial interests.
BioPump) was produced. An open-label phase 1/2
clinical trial (ClinicalTrials.gov identier NCT REFERENCES
02117427) to examine the safety and efcacy of 1. Kausz AT, Levey AS. The care of patients with chronic
kidney disease. J Gen Intern Med. 2002;17(8):658-662.
EPO delivered by the pump in 3 cohorts (18-25,
2. Winearls CG, Oliver DO, Pippard MJ, Reid C,
35-45, or 55-65 IU/kg/d) of patients with ESRD is Downing MR, Cotes PM. Effect of human erythropoietin derived
ongoing; follow-up is one year. Interim results of the from recombinant DNA on the anaemia of patients maintained by
study, including 3 patients who underwent implan- chronic haemodialysis. Lancet. 1986;2(8517):1175-1178.
tation, were presented in late 2014.103 The platform 3. Jelkmann W. The ESA scenario gets complex: from bio-
delivered sustained physiologically appropriate similar epoetins to activin traps. Nephrol Dial Transplant.
levels of endogenous EPO, in turn maintaining Hb 2015;30(4):553-559.
4. Singh AK, Szczech L, Tang KL, et al. Correction of anemia
levels in the desired range. Further clinical studies
with epoetin alfa in chronic kidney disease. N Engl J Med.
have been planned in patients with ESRD who 2006;355(20):2085-2098.
respond poorly to conventional ESA treatment, as 5. Dreke TB, Locatelli F, Clyne N, et al. Normalization of
well as in anemic transplant recipients and peritoneal hemoglobin level in patients with chronic kidney disease and
dialysis patients. anemia. N Engl J Med. 2006;355(20):2071-2084.

Am J Kidney Dis. 2016;67(1):133-142 139


Bonomini et al

6. Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of dar- 24. Lacy SE, DeVries PJ, Xie N, Fung E, Lesniewski RR,
bepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl Reilly EB. The potency of erythropoietin-mimic antibodies cor-
J Med. 2009;361(21):2019-2032. relates inversely with afnity. J Immunol. 2008;181(2):1282-1287.
7. Pisoni RL, Fuller DS, Bieber BA, et al. The DOPPS Practice 25. Suzuki N, Mukai HY, Yamamoto M. In vivo regulation of
Monitor for US dialysis care: trends through August 2011. Am J erythropoiesis by chemically inducible dimerization of the eryth-
Kidney Dis. 2012;60(1):160-165. ropoietin receptor intracellular domain. PLoS One. 2015;10(3):
8. KDIGO Anemia Work Group. KDIGO clinical practice e0119442.
guideline for anemia in chronic kidney disease. Kidney Int Suppl. 26. Haase VH. Regulation of erythropoiesis by hypoxia-
2012;2:S279-S335. inducible factors. Blood Rev. 2013;27(1):41-53.
9. Locatelli F, Brny P, Covic A, et al; ERA-EDTA ERBP 27. Semenza GL. Hypoxia-inducible factors in physiology and
Advisory Board. Kidney Disease: Improving Global Outcomes medicine. Cell. 2012;148(3):399-408.
guidelines on anaemia management in chronic kidney disease: a 28. Kapitsinou PP, Liu Q, Unger TL, et al. Hepatic HIF-2
European Renal Best Practice position statement. Nephrol Dial regulates erythropoietic responses to hypoxia in renal anemia.
Transplant. 2013;28(6):1346-1359. Blood. 2010;116(16):3039-3048.
10. Hung SC, Tarng DC. ESA and iron therapy in chronic 29. Scortegagna M, Ding K, Zhang Q, et al. HIF-2alpha reg-
kidney disease: a balance between patient safety and hemoglobin ulates murine hematopoietic development in an erythropoietin-
target. Kidney Int. 2014;86(4):676-678. dependent manner. Blood. 2005;105(8):3133-3140.
11. Sathyanarayana P, Houde E, Marshall D, et al. CNTO 530 30. Mastrogiannaki M, Matak P, Keith B, Simon MC,
functions as a potent EPO mimetic via unique sustained effects Vaulont S, Peyssonnaux C. HIF-2alpha, but not HIF-1alpha,
on bone marrow proerythroblast pools. Blood. 2009;113(20): promotes iron absorption in mice. J Clin Invest. 2009;119(5):
4955-4962. 1159-1166.
12. Bugelski PJ, Makropoulos D, Spinka-Doms T, et al. Dif- 31. Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible
ferential effects of long-lived erythropoietin receptor agonists in factors and the response to hypoxic stress. Mol Cell. 2010;40(2):
rats. Pharm Anal Acta. 2011;2(7):133. 294-309.
13. Greindl A, Kessler C, Breuer B, et al. AGEM400(HES), a 32. Rabinowitz MH. Inhibition of hypoxia-inducible factor
novel erythropoietin mimetic peptide conjugated to hydroxyethyl prolyl hydroxylase domain oxygen sensors: tricking the body into
starch with excellent in vitro efcacy. Open Hematol J. 2010;4:1-14. mounting orchestrated survival and repair responses. J Med Chem.
14. Kessler C, Greindl A, Breuer B, et al. Erythropoietin 2013;56(23):9369-9402.
mimetic compound AGEM400(HES) binds to the same receptor as 33. Kato A, Hishida A, Kumagai H, Furuya R, Nakajima T,
erythropoietin but displays a different spectrum of activities. Honda N. Erythropoietin production in patients with chronic renal
Cytokine. 2012;57(2):226-237. failure. Ren Fail. 1994;16(5):645-651.
15. Dalle B, Henri A, Rouyer-Fessard P, et al. Dimeric eryth- 34. Shimizu S, Enoki Y, Sakata S, Kohzuki H, Ohga Y,
ropoietin fusion protein with enhanced erythropoietic activity Matsumura K. Erythropoietin response to acute hypobaric or
in vitro and in vivo. Blood. 2001;97(12):3776-3782. anaemic hypoxia in gentamicin-administered rats. Acta Physiol
16. Sytkowski AJ, Lunn ED, Risinger MA, Davis KL. An Scand. 1994;151(2):225-231.
erythropoietin fusion protein comprised of identical repeating 35. Priyadarshi A, Periyasamy S, Burke TJ, Britton SL,
domains exhibits enhanced biological properties. J Biol Chem. Malhotra D, Shapiro JI. Effects of reduction of renal mass on renal
1999;274(35):24773-24778. oxygen tension and erythropoietin production in the rat. Kidney
17. Fares FA, Ganem S, Hajouj T, Agai E. Development of a long- Int. 2002;61(2):542-546.
acting erythropoietin by fusing the carboxyl-terminal peptide of hu- 36. Unger EF, Thompson AM, Blank MJ, Temple R. Eryth-
man chorionic gonadotropin beta-subunit to the coding sequence of ropoiesis stimulating agentstime for a reevaluation. N Engl J
human erythropoietin. Endocrinology. 2007;48(10):5081-5087. Med. 2010;362(3):189-192.
18. Fares F, Havron A, Fima E. Designing a long acting 37. Besarab A, Chernyavskaya EN, Motylev I, et al. FG-4592, an
erythropoietin by fusing three carboxyl-terminal peptides of hu- oral hypoxia-inducible factor propyl hydroxylase inhibitor, corrects
man chorionic gonadotropin b subunit to the n-terminal coding anemia without iron supplementation in incident dialysis patients
sequence. Int J Cell Biol. 2011;2011:275063. [ASN abstract TH-OR096]. J Am Soc Nephrol. 2012;23:21A.
19. Schriebl K, Trummer E, Lattenmayer C, et al. Biochemical 38. Provenzano R, Goodkin D, Klaus S, et al. Evaluation of
characterization of rhEPO-Fc fusion protein expressed in CHO FG-4592, a novel oral hypoxia-inducible factor prolyl hydroxylase
cells. Protein Expr Purif. 2006;49(2):265-275. inhibitor, to treat anemia in hemodialysis patients [abstract 188].
20. Penno CA, Kawabe Y, Ito A, Kamihita M. Production of Presented at: National Kidney Foundation Conference; April 26-
recombinant human erythropoietin/Fc fusion protein by geneti- 30, 2011; Las Vegas, NV.
cally manipulated chickens. Transgenic Res. 2010;19(2):187-195. 39. Finberg KE. Regulation of systemic iron homeostasis. Curr
21. Joung CH, Shin JY, Koo JK, et al. Production and char- Opin Hematol. 2013;20(3):208-214.
acterization of long-acting recombinant human albumin-EPO 40. Sun CC, Vaja V, Babitt JL, Lin HY. Targeting the
fusion protein expressed in CHO cell. Protein Expr Purif. hepcidin-ferroportin axis to develop new treatment strategies for
2009;68(2):137-145. anemia of chronic disease and anemia of inammation. Am J
22. Schneider H, Chaovapong W, Matthews DJ, et al. Homo- Hematol. 2012;87(4):392-400.
dimerization of erythropoietin receptor by a bivalent monoclonal 41. Qian JQ, Chen N, Chen J, et al. A randomized, double-
antibody triggers cell proliferation and differentiation of erythroid blind, placebo controlled trial of FG-4592 for correction of ane-
precursors. Blood. 1997;89(2):473-482. mia in subjects with chronic kidney disease in China [ASN
23. Elliott S, Lorenzini T, Yanagihara D, Chang D, Elliott G. abstract FR-OR011]. J Am Soc Nephrol. 2013;24:38A.
Activation of the erythropoietin (EPO) receptor by bivalent anti- 42. Besarab A, Tak Mao Chan D, Dua SL, et al. Hypoxia
EPO receptor antibodies. J Biol Chem. 1996;271(40):24691-24697. inducing factor propyl hydroxylase inhibitor FG-4592 corrects

140 Am J Kidney Dis. 2016;67(1):133-142


New Treatment Approaches for the Anemia of CKD

anemia in peritoneal dialysis [ASN abstract SA-OR087]. J Am Soc in heart via Na/Ca21 exchanger in developing rats. Cell Physiol
Nephrol. 2013;24:91A. Biochem. 2014;34(2):313-324.
43. Akebia announces positive top-line results from its phase 60. Maguer-Satta V, Bartholin L, Jeanpierre S, et al. Regulation
2b study of AKB-6548 in non-dialysis patients with anemia related of human erythropoiesis by activin A, BMP2, and BMP4, members
to chronic kidney disease. http://ir.akebia.com/releasedetail.cfm? of the TGFbeta family. Exp Cell Res. 2003;282(2):110-120.
ReleaseID5878191. Accessed March 2, 2015. 61. Yu J, Maderazo L, Shao LE, et al. Specic roles of activin/
44. A phase IIa, randomized, single-blind, placebo-controlled, inhibin in human erythropoiesis in vitro. Ann N Y Acad Sci.
parallel-group study to evaluate the safety, pharmacokinetics, and 1991;628:199-211.
efcacy of 28-day repeat oral doses of GSK 1278863A in anemic pre- 62. Zermati Y, Fichelson S, Valensi F, et al. Transforming
dialysis and hemodialysis-dependent patients. GlaxoSmithKline growth factor inhibits erythropoiesis by blocking proliferation and
website www.gsk-clinicalstudyregister.com/study/112844#rs. accelerating differentiation of erythroid progenitors. Exp Hematol.
Accessed March 2, 2015. 2000;28(8):885-894.
45. A repeat-dose, open-label, parallel-group study to assess 63. Murata M, Eto Y, Shibai H, Sakai M, Muramatsu M.
the pharmacokinetics of GSK1278863 and metabolites in normal Erythroid differentiation factor is encoded by the same mRNA as
subjects and subjects with impaired renal function. GlaxoSmith- that of the inhibin beta A chain. Proc Natl Acad Sci U S A.
Kline website www.gsk-clinicalstudyregister.com/study/115573#rs. 1988;85(8):2434-2438.
Accessed March 2, 2015. 64. Raje N, Vallet S. Sotatercept, a soluble activin receptor
46. Studies ltered by compound GSK 1278863. GlaxoSmith- type 2A IgG-Fc fusion protein for the treatment of anemia and
Kline website www.gsk-clinicalstudyregister.com/compounds/gsk bone loss. Curr Opin Mol Ther. 2010;12(5):586-597.
1278863#ps. Accessed March 2, 2015. 65. Ruckle J, Jacobs M, Kramer W, et al. Single-dose, ran-
47. Flamme I, Oehme F, Ellinghaus P, Jeske M, domized, double-blind, placebo-controlled study of ACE-011
Keldenich J, Thuss U. Mimicking hypoxia to treat anemia: (ActRIIA-IgG1) in postmenopausal women. J Bone Miner Res.
HIF-stabilizer BAY 85-3934 (Molidustat) stimulates erythro- 2009;24(4):744-752.
poietin production without hypertensive effects. PLoS One. 66. Abdulkadyrov K, Salogub G, Khuazheva N, et al. ACE-011, a
2014;9(11):e111838. soluble activin receptor type IIa IgG-FC fusion protein, increases he-
48. Boettcher MF, Lentini S, Kaiser A, Flamme I, Kubitza D, moglobin (Hb) and improves bone lesions in multiple myeloma pa-
Wensing G. First-in-man study with BAY 85-3934a new oral tients receiving myelosuppressive chemotherapy: preliminary analysis.
selective HIF-PH inhibitor for the treatment of renal anemia [ASN Blood. 2009;114(22). (ASH Annual Meeting Abstracts):Abstract 749.
abstract THPO1090]. J Am Soc Nephrol. 2013;24:347A. 67. Yu J, Shao L, Vaughan J, Vale W, Yu AL. Characterization
49. Safety, tolerability, PK and PD study of JTZ-951 in anemic of the potentiation effect of activin on human erythroid colony
subjects with end-stage renal disease. U.S. National Institutes of formation in vitro. Blood. 1989;73(4):952-960.
Health website https://clinicaltrials.gov/ct2/show?term5jtz-951 68. Shao l, Frigon NL Jr, Young AL, et al. Effect of activin A
&rank52. Accessed March 2, 2015. on globin gene expression in puried human erythroid progenitors.
50. Pilot PK/PD study of DS-1093a in patients with chronic Blood. 1992;79(3):773-781.
kidney disease. U.S. National Institutes of Health website https:// 69. Iancu-Rubin C, Mosoyan G, Wang J, Kraus T, Sung V,
clinicaltrials.gov/ct2/show?term5ds-1093a&rank51. Accessed Hoffman R. Stromal cell-mediated inhibition of erythropoiesis can
March 2, 2015. be attenuated by sotatercept (ACE-011), an activin receptor type II
51. Cheng K, Ho K, Stokes R, et al. Hypoxia-inducible factor- ligand trap. Exp Hematol. 2013;41:155-166.
1alpha regulates beta cell function in mouse and human islets. 70. Blank U, Karlsson S. The role of Smad signaling in he-
J Clin Invest. 2010;120(6):2171-2183. matopoiesis and translational hematology. Leukemia. 2011;25(9):
52. Rankin EB, Rha J, Selak MA, et al. Hypoxia-inducible 1379-1388.
factor 2 regulates hepatic lipid metabolism. Mol Cell Biol. 71. El-Shahawy M, Cotton J, Kaupke J, Kopyt N, Choi S,
2009;29(16):4527-4538. Smith WT. Safety and hemoglobin effects of the rst 28-day dose cycle
53. Zehetner J, Danzer C, Collins S, et al. PVHL is a regulator of sotatercept 0.7 mg/kg compared with lower doses and placebo for
of glucose metabolism and insulin secretion in pancreatic beta correction of anemia in hemodialysis subjects: interim analysis [ASN
cells. Genes Dev. 2008;22(22):3135-3146. abstract TH-PO985]. J Am Soc Nephrol. 2014;25:337A.
54. Krishnan J, Suter M, Windak R, et al. Activation of a 72. Besson-Fournier C, Latour C, Kautz L, et al. Induction of
HIF1alpha-PPARgamma axis underlies the integration of glyco- activin B by inammatory stimuli up-regulates expression of the
lytic and lipid anabolic pathways in pathologic cardiac hypertro- iron-regulatory peptide hepcidin through Smad1/5/8 signaling.
phy. Cell Metab. 2009;9(6):512-524. Blood. 2012;120(2):431-439.
55. Semenza GL. HIF-1: upstream and downstream of cancer 73. Finberg KE, Whittlesey RL, Fleming MD, Andrews NC.
metabolism. Curr Opin Genet Dev. 2009;20(1):51-56. Down-regulation of Bmp/Smad signaling by Tmprss6 is required for
56. Ratcliffe PJ. Oxygen sensing and hypoxia signalling maintenance of systemic iron homeostasis. Blood. 2010;115(18):
pathways in animals: the implications of physiology for cancer. 3817-3826.
J Physiol. 2013;591(8):2027-2042. 74. Truksa J, Lee P, Beutler E. Two BMP responsive elements,
57. Rimoldi SF, Rexhaj E, Pratali L, et al. Systemic vascular STAT, and bZIP/HNF4/COUP motifs of the hepcidin promoter are
dysfunction in patients with chronic mountain sickness. Chest. critical for BMP, SMAD1, and HJV responsiveness. Blood.
2012;141(1):139-146. 2009;113(3):688-695.
58. Schmid JP, Nobel D, Brugger N, et al. Short-term high 75. Ganz T. Systemic iron homeostasis. Physiol Rev.
altitude exposure at 3454m is well tolerated in patients with stable 2013;93(4):1721-1741.
heart failure. Eur J Heart Fail. 2015;17(2):182-186. 76. Lotinun S, Pearsall RS, Horne WC, Baron R. Activin re-
59. Ma HJ, Li Q, Ma HJ, et al. Chronic intermittent hypobaric ceptor signaling: a potential therapeutic target for osteoporosis.
hypoxia ameliorates ischemia/reperfusion-induced calcium overload Curr Mol Pharmacol. 2012;5(2):195-204.

Am J Kidney Dis. 2016;67(1):133-142 141


Bonomini et al

77. Lotinun S, Pearsall RS, Davies MV, et al. A soluble activin 90. Perez-Ruixo JJ, Krzyzanski W, Bouman-Thio E, et al.
receptor type IIA fusion protein (ACE-011) increases bone mass Pharmacokinetics and pharmacodynamics of the erythropoietin
via a dual anabolic-antiresorptive effect in Cynomolgus monkeys. Mimetibody construct CNTO 528 in healthy subjects. Clin
Bone. 2010;46(4):1082-1088. Pharmacokinet. 2009;48(9):601-613.
78. Pearsall RS, Canalis E, Cornwall-Brady M, et al. 91. Macdougall IC, Provenzano R, Sharma A, et al. Pegine-
A soluble activin type IIA receptor induces bone formation and satide for anemia in patients with chronic kidney disease not
improves skeletal integrity. Proc Natl Acad Sci U S A. 2008;105(19): receiving dialysis. N Engl J Med. 2013;368(4):320-332.
7082-7087. 92. Fishbane S, Schiller B, Locatelli F, et al. Peginesatide in
79. Fang Y, Agapova OA, Sugatani T, Malluche H, patients with anemia undergoing hemodialysis. N Engl J Med.
Hruska KA. Treatment of the CKD-MBD with a ligand trap for the 2013;368(4):307-319.
activin receptor type 2A [ASN abstract FR-PO816]. J Am Soc 93. Macdougall IC, Rossert J, Casadevall N, et al. A peptide-
Nephrol. 2014;25:557A. based erythropoietin-receptor agonist for pure red-cell aplasia.
80. Hruska KA, Agapova OA, Fang Y, Sugatani T, N Engl J Med. 2009;361(19):1848-1855.
Seifert ME, Sung V. Chronic kidney disease (CKD) stimulates 94. Afmax and Takeda announce a nationwide voluntary recall
activin and endothelial to mesenchymal transition (EnMT), of all lots of OMONTYSR (peginesatide) Injection. http://www.
which causes vascular calcication and is inhibited by an activin takeda.com/news/2013/20130224_5664.html. Accessed February
ligand trap [ASN abstract SA-OR063]. J Am Soc Nephrol. 8, 2015.
2014;25:95A. 95. Im SJ, Yang SI, Yang SH, et al. Natural form of non-
81. Leonard MB. A structural approach to skeletal fragility in cytolytic exible human Fc as a long-acting carrier of agonistic
chronic kidney disease. Semin Nephrol. 2009;29(2):133-143. ligand, erythropoietin. PLoS One. 2011;6:e24574.
82. Malluche H, Hurska K, Singh HN, Smith WT. Sota- 96. Roux KH, Strelets L, Brekke OH, Sandlie I,
tercept: initial signal-seeking quantitative computed tomography Michaelsen TE. Comparisons of the ability of human IgG3 hinge
results for bone mass and vascular calcication in hemodialysis mutants, IgM, IgE, and IgA2, to form small immune complexes:
patients treated with escalating doses: interim analysis of ACE- a role for exibility and geometry. J Immunol. 1998;161(8):
011-REN-001 [ASN abstract TH-PO602]. J Am Soc Nephrol. 4083-4090.
2014;25:245A. 97. Idusogie EE, Wong PY, Presta LG, et al. Engineered an-
83. Suragani RN, Cadena SM, Cawley SM, et al. Transforming tibodies with increased activity to recruit complement. J Immunol.
growth factor-b superfamily ligand trap ACE-536 corrects anemia 2001;166(4):2571-2575.
by promoting late-stage erythropoiesis. Nat Med. 2014;20(4): 98. Schuurman J, Perdok GJ, Gorter AD, Aalberse RG. The
408-414. inter-heavy chain disulde bonds of IgG4 are in equilibrium with
84. Attie KM, Allison MJ, McClure T, et al. A phase 1 study of intra-chain disulde bonds. Mol Immunol. 2001;38(1):1-8.
ACE-536, a regulator of erythroid differentiation, in healthy vol- 99. Yang SH, Yang SI, Chung Y-K. A long-acting erythro-
unteers. Am J Hematol. 2014;89(7):766-770. poietin fused with noncytolytic human Fc for the treatment of
85. Livnah O, Stura EA, Johnson DL. Functional mimicry of a anemia. Arch Pharm Res. 2012;35(5):757-759.
protein hormone by a peptide agonist: the EPO receptor complex 100. Study to evaluate safety, tolerability, and pharmacokinetics/
at 2.8 A. Science. 1996;273(5274):464-471. pharmacodynamics of GX-E2 in healthy subjects (GX-E2-P1).
86. Wrighton NC, Farrell FX, Chang R, et al. Small peptides as U.S. National Institutes of Health website https://clinicaltrials.gov/
potent mimetics of the protein hormone erythropoietin. Science. ct2/show/study/NCT02291991?term5GX-E2&rank5.1 Accessed
1996;273(5274):458-464. March 2, 2015.
87. Johnson DL, Farrell FX, Barbone FP, et al. Identication of 101. Study to evaluate the efcacy and safety of GX-E2 in the
a 13 amino acid peptide mimetic of erythropoietin and description anemic patients diagnosed with chronic kidney disease (CKD).
of amino acids critical for the mimetic activity of EMP1. U.S. National Institutes of Health website https://clinicaltrials.gov/
Biochemistry. 1998;37(11):3699-3710. ct2/show/study/NCT02044653?term5GX-E2&rank52. Accessed
88. Huang C. Receptor-Fc fusion therapeutics, traps, and March 2, 2015.
MIMETIBODY technology. Curr Opin Biotechnol. 2009;20(6): 102. Brill-Almon E, Stern B, Ak D, et al. Ex vivo trans-
692-699. duction of human dermal tissue structures for autologous im-
89. Bouman-Thio E, Franson K, Miller B, et al. A phase I, plantation production and delivery of therapeutic proteins. Mol
single and fractionated, ascending-dose study evaluating the Ther. 2005;12(2):274-282.
safety, pharmacokinetics, pharmacodynamics, and immunoge- 103. Neil G. Prolonged secretion of physiological levels of
nicity of an erythropoietin mimetic antibody fusion protein autologous EPO by TARGT. Presented at: XXII Annual Congress
(CNTO 528) in healthy male subjects. J Clin Pharmacol. of the European Society of Gene and Cell Therapy; The Hague,
2008;48(10):1197-1207. the Netherlands; October 23-26, 2014.

142 Am J Kidney Dis. 2016;67(1):133-142

You might also like