ArticlePDF Available

Perspectives for the therapy of anemia of chronic diseases

Authors:

Abstract and Figures

The incidence of anemia of chronic disease (ACD) is underestimated, increases with age, and affects about 30% of the elderly. ACD treatment is currently based on the pharmacotherapy of diseases that caused anemia, erythropoiesis-stimulating agents, and parenteral administration of iron supplementation in case of iron deficiency. Increasing knowledge on the pathophysiology of ACD has resulted in the burst of research on the development of new drugs that are focused on three main areas. The first group of drugs includes substances that inhibit hepcidin transcription, namely direct and indirect bone morphogenetic protein 6 (BMP6) inhibitors and/or SMAD signaling pathway inhibitors, and drugs that regulate hepcidin transcription through STAT3 signaling pathway. The second group of drugs includes direct hepcidin inhibitors (e.g., aptamers, anticalin proteins, monoclonal antibodies) or substances that inhibit the binding of hepcidin to ferroportin. The third group of drugs improves erythropoiesis mainly by upregulation of erythropoietin and/or inhibition of proinflammatory cytokines. In the latter group, hypoxia-inducing factor stabilizers and IL-6 or TNFα antagonists are particularly important. This article discusses new drug groups and substances that are in different phases of development, including both preclinical and clinical studies, and focuses on the prospects of their use in ACD. © 2020 Polish Society of Hematology and Transfusion Medicine, Insitute of Hematology and Transfusion Medicine.
Content may be subject to copyright.
125
Perspectives for the therapy of anemia
of chronic diseases
Department of Pharmacology and Toxicology,
Collegium Medicum, University of Zielona Góra,
Zielona Góra, Poland
Sylwia Sulimiera Michalak
REVIEW ARTICLE
Article history:
Received: 31.01.2020
Accepted: 02.03.2020
journal homepage: https://content.sciendo.com/ahp
Abstract
The incidence of anemia of chronic disease (ACD) is underestimated, increases with age, and aects about 30% of the elderly. ACD
treatment is currently based on the pharmacotherapy of diseases that caused anemia, erythropoiesis-stimulating agents, and parenteral
administration of iron supplementation in case of iron deciency. Increasing knowledge on the pathophysiology of ACD has resulted
in the burst of research on the development of new drugs that are focused on three main areas. The rst group of drugs includes
substances that inhibit hepcidin transcription, namely direct and indirect bone morphogenetic protein
6 (BMP6) inhibitors and/or SMAD signaling pathway inhibitors, and drugs that regulate hepcidin transcription through STAT3 signaling
pathway. The second group of drugs includes direct hepcidin inhibitors (e.g., aptamers, anticalin proteins, monoclonal antibodies) or
substances that inhibit the binding of hepcidin to ferroportin. The third group of drugs improves erythropoiesis mainly by upregulation
of erythropoietin and/or inhibition of proinammatory cytokines. In the latter group, hypoxia-inducing factor stabilizers and IL-6 or
TNFα antagonists are particularly important. This article discusses new drug groups and substances that are in dierent phases of
development, including both preclinical and clinical studies, and focuses on the prospects of their use in ACD.
© 2020 Polish Society of Hematology and Transfusion Medicine, Insitute of Hematology and Transfusion Medicine. Published by Sciendo.
All rights reserved.
Keywords:
anemia of chronic disease, hepcidin, cytokine, hypoxia-inducible factor, treatment
Acta Haematologica Polonica 51(3) • September 2020 • 125–132 • DOI: 10.2478/ahp-2020-0024
Introduction
The global prevalence of anemia of chronic disease (ACD), also known
as anemia of inammation, is not fully estimated [1]. The incidence of
ACD increases with age, and in the elderly population it accounts for
approximately one-third of all anemia cases [2, 3]. ACD can develop
in the course of infections, malignancies, and autoimmune diseases.
The etiology of this type of anemia is complex and it is associated with
a decrease in red blood cells (RBCs) production and decreased survival.
The activity of proinammatory cytokines, including tumor necrosis
factor α (TNFα) and interferons, results in decreased availability of
iron for erythropoiesis, reduced response to erythropoietin (EPO),
and decreased EPO synthesis, which in turn inhibit proliferation and
dierentiation of erythroid progenitor cells and increase erythrocyte
turnover and their degradation by macrophages. Hepcidin plays
a central role in reducing the bioavailability of iron, and its expression
is regulated by proinammatory cytokines, in particular, interleukin 6
(IL-6) [4, 5]. However, other molecules, such as IL-10, interferon γ (IFN-γ),
IL-1β, and lipopolysaccharide, also serve as hepcidin inducers [6]. Many
proteins have been described to be involved in the regulation of hepcidin
synthesis, and thus they may become a target for ACD therapy. These
include bone morphogenetic protein (BMP), hemojuvelin (HJV), activin
receptor-like kinases 2 and 3 (ALK-2 and ALK-3), and erythroferrone
(ERFE) [5, 7]. Also EPO can directly reduce hepcidin production by
inhibiting the BMP-small mothers against decapentaplegic proteins
(BMP-SMAD) pathway [8] and also via hypoxia-inducible factor (HIF)
signaling [9].
Currently, ACD therapy is limited to the treatment of the underlying
disease that is responsible for inammatory response. Additionally,
erythropoiesis-stimulating agents (ESAs), in particular human
recombinant erythropoietin (EPO), are also administered. Sometimes
iron supplementation is necessary. Better understanding of the
regulation of iron metabolism in the presence of inammation, with
particular focus on the role of hepcidin, proinammatory cytokines, and
HIF in erythropoiesis, has caused a burst of research on the development
of new drugs for the treatment of ACD. These studies, focusing on
three major research areas, are in various stage of development from
experimental stage through preclinical research to clinical studies. The
rst group of studies evaluates the inuence of various substances
on hepcidin production. The second group examines the substances
that inhibit the eects of hepcidin release. This group contains direct
hepcidin inhibitors (hepcidin neutralizers and anti-hepcidin monoclonal
antibodies) or substances that are able to inhibit binding of hepcidin to
ferroportin. Finally, the third group of studies focuses on erythropoiesis
inducers that upregulate EPO, stabilize HIF, and block proinammatory
cytokines.
Hepcidin production inhibitors
Hepcidin production inhibitors downregulate hepcidin gene (hepcidin
antimicrobial peptide gene [HAMP]) in hepatocytes (Fig. 1). Hepcidin
transcription can be inhibited as a result of the inhibition of SMAD and/
or signal transducer and activator of transcription (STAT) signaling
pathways. It can be achieved in a variety of mechanisms, the most
important of which seems to be BMP or BMP receptor interaction and
blocking of IL-6 or IL-6 receptor. Downstream signaling from BMP
receptor is mediated through SMAD1/5/8 pathway while signaling
from IL-6 receptor is mediated through Janus-activated kinases (JAK)/
Corresponding author: Sylwia Sulimiera Michalak, Department of Pharmacology and Toxicology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland,
phone: +48 68 328 73 77, e-mail: s.michalak@cm.uz.zgora.pl
126
Acta Haematologica Polonica
STAT3 signaling pathway. The BMP-SMAD pathway is activated
by BMP-2 and BMP-6 through BMP receptors forming complexes
with ALK-2 and ALK-3, and HJV as a BMP receptor on liver cells
enhances the transmission of this signal [7, 10].
Drugs/substances aecting BMP6 pathway
Currently, a number of substances aecting BMP6 pathways
are studied. In two models, murine and rat, the use of anti-BMP6
antibodies led to an increase in hemoglobin (Hb), a decrease in iron
deposition in tissues, and a reduction in ESA doses needed to treat
ACD [11]. Recently, LY3113593, a human anti-BMP6 monoclonal
antibody, is gaining more interest. LY3113593 blocks binding of BMP6
to its receptor, which leads to an increase in iron concentration, an
increase in transferrin saturation, and a decrease in hepcidin level. In
a study performed in a group of patients with chronic kidney disease
(CKD), LY3113593 increased Hb and caused a decrease in ferritin
level in comparison with placebo [12].
Other drugs aecting BMP-6 receptor currently tested in clinical trials
include: LDN-193189, modied heparin (e.g., roneparstat), TP-0184,
and a group of drugs related to HJV. LDN-193189 is a selective
inhibitor of the BMP kinase type 1 receptor [13]. TP-0184 is an ALK-2
inhibitor that reduces hepcidin mRNA and improves Hb concentration
in preclinical studies in mice [14]. Modied heparin with reduced
anticoagulation ability binds BMP-6 and blocks hepcidin expression,
as demonstrated in in vitro and in vivo animal models [15, 16].
The ability to inhibit hepatic BMP-SMAD signaling, reduce hepcidin
levels, increase ferroportin expression, and increase serum iron
levels has also been demonstrated for protein-soluble HJVFc (HJV.
Fc). HJV.Fc consists of extracellular domain of HJV conjugated with
human IgG Fc fragment [10, 17]. Currently, other potential drugs, e.g.,
h5F9.23 and h5F9-AM8, which interact with HJV, are experimentally
investigated [18]. Administration of a single dose of ABT-207 or
H5F9-AM8 in rats and monkeys resulted in prolonged suppression of
hepcidin production and an increase in serum iron concentration [19].
H5F9-AM8 was tested in animal models of ACD and iron-refractory
iron-deciency anemia (IRIDA), resulting in reduction of hepcidin and
improvement of anemia in all cases [20].
Druggable targets associated with proinammatory
cytokines
Clinically available drugs include a humanized anti-IL-6 receptor
antibody, tocilizumab, and a chimeric (human–murine) antibody
against IL-6, siltuximab. Tocilizumab is used in rheumatoid arthritis.
As an improvement of RBC parameters have been observed during
the treatment, tocilizumab has become a new potential drug for
ACD [21, 22]. Tocilizumab was successfully used in the treatment
of anemia in patients with Castleman’s disease [22] and in patients
with malignant tumors [23]. Reduced serum hepcidin level and
improvement of anemia in patients with Castleman’s disease [24, 25],
myeloma, and other lymphoproliferative cancers [26] have been
reported for siltuximab. An improvement in RBC parameters has also
been demonstrated in the treatment of solid tumors [27]. Another IL-6
receptor antibody, MR16-1, was tested in murine models. MR16-1 has
been shown to improve Hb parameters in cancer-associated anemia
[23]. Other substances aecting hepcidin production by modulation of
signaling pathways in hepatocytes have been tested experimentally.
In murine models, natural and synthetic STAT3 inhibitors have been
shown to inhibit hepcidin expression in macrophages [28]. MaR1
Fig. 1. Hepcidin transcription inhibitors
ACVR2A – activin receptor type 2A; ALK3/2 – activin receptor-like kinase 3/2; BMP-6 – bone morphogenetic protein 6; BMPR2 – bone
morphogenetic protein receptor 2; ERFE – erythroferron; EGFR – epidermal growth factor receptor; HAMP – hepcidin antimicrobial peptide;
HJV – hemojuvelin; HFE – human hemochromatosis protein; IL-6 – interleukin6; IL-6R – IL-6 receptor; JAK – Janus-activated kinases; SMAD
– small mothers against decapentaplegic proteins; STAT3 – signal transducer and activator of transcription 3; TFR2 – transferrin receptor 2;
TMPRSS6 – transmembrane protease serine 6
Acta Haematologica Polonica
127
inhibits inammatory response, including hepcidin expression, by
aecting the IL-6/STAT3 signaling pathway and then reducing the
severity of anemia symptoms [29].
Potential role of ERFE
ERFE is a physiological regulator of hepcidin, which is synthesized
and secreted by erythroblasts in the bone marrow and other tissues in
response to EPO. ERFE inhibits hepcidin transcription by an unknown
mechanism that involves BMP6 pathway and probably some known
membrane receptors [30, 31]. ERFE antagonists and agonists, and
also other substances involved in the BMP6 signaling pathway,
may prove useful in the prevention and treatment of iron disorders.
However, a recently published study suggested a correlation
between increased ERFE levels and death/cardiovascular events in
hemodialysis patients with CKD, which is further enhanced by the
administration of ESA [30]. Therefore, the potential use of ERFE in
anemia requires further research and careful determination of patient
groups who may benet from such a therapy.
Other possibilities of hepcidin production inhibition
It has been shown that hepcidin production can be inhibited
by thiazolidinediones, testosterone, estrogens, vitamin D,
and substances of plant origin (e.g., Angelica sinensis). New
thiazolidinediones are studied in the treatment of anemia. These
drugs may aect hepcidin expression through various pathways,
including SMAD1/5/8 signaling pathway, extracellular signal-
regulated kinases 1/2 (ERK1/2) and TMPRSS6, and also probably
by indirectly aecting ERFE-mediated hepcidin production [32].
In vitro and in vivo studies have shown that vitamin D inhibits the
expression of proinammatory cytokines and directly inhibits
hepcidin transcription by interaction of vitamin D receptor with the
HAMP promoter [33, 34]. In a population of children with chronic
inammatory bowel disease, high levels of cholecalciferol (≥30 ng/mL)
were associated with higher Hb levels, while low levels correlated
with elevated hepcidin and lower Hb concentration [35]. However, not
all studies conrmed the eect of vitamin D supplementation on the
levels of proinammatory cytokines, ferritin, or hepcidin [36]. A meta-
analysis and systematic review of literature did not conrm the eect
of cholecalciferol on IL-6 or C-reactive protein (CRP) levels [37].
On the other hand, Smith and colleagues showed that high doses
of vitamin D signicantly reduced hepcidin levels in healthy adults
1 week after the administration of a single dose, without aecting the
levels of proinammatory cytokines or ferritin. The authors concluded
that this may suggest that the eect of vitamin D on hepcidin is
independent from proinammatory cytokines or ferritin [38]. In many
clinical trials, the inverse correlation between vitamin D levels and
anemia has been reported. It is now believed that vitamin D is an
important factor involved in the pathogenesis of anemia [39, 40].
Testosterone inhibits hepcidin by upregulation of the epidermal
growth factor receptor (EGFR) signaling in the liver [41] and
downregulates hepcidin transcription by its inuence on BMP-SMAD
signaling pathway [42]. The administration of testosterone increases
the concentration of Hb in men with reduced testosterone levels, in
case of anemia of both known and unknown cause as demonstrated
in a study of nearly 800 men, including 126 with anemia [43].
Similarly, 17-β-estradiol could be considered a drug for anemia in the
elderly women because it has also been shown to inhibit hepcidin
transcription. Inhibition of hepcidin transcription results from the
activity of estrogens on HAMP-associated gene promoters [44].
Angelica sinensis polysaccharide (ASP), a polysaccharide
obtained from the root of the Angelica sinensis plant, inhibits
hepcidin expression and may potentially improve RBC parameters
in patients with anemia. The ecacy of ASP in the treatment of
anemia associated with CKD has been demonstrated in rats. ASP
inhibited hepcidin production, increased the amount of iron available
for erythropoiesis by its mobilization from the liver and spleen, and
increased EPO levels [45]. Previous data have shown that regular
supplementation of Angelica sinensis in hemodialyzed patients
improved anemia in patients resistant to recombinant human EPO
therapy [46]. It is believed that ASP inhibits hepcidin expression
by inhibiting the expression and/or phosphorylation of JAK1/2,
SMAD1/5/8, and ERK1/2 while upregulating SMAD7 [47].
Direct hepcidin inhibitors and agents
preventing hepcidin from binding to
ferroportin
Another group of drugs that aect hepcidin are direct hepcidin
inhibitors and drugs that prevent hepcidin from binding to ferroportin
(Fig. 2). Direct neutralization of hepcidin is possible with monoclonal
antibodies (AB 12B9M) as well as anticalin proteins (e.g., PRS-080),
aptamers (e.g., NOX-H94), or guanosine 5'-diphosphate (GDP).
Anticalins are a class of ligand-binding proteins designed based on a
lipocalin scaold. Aptamers are oligonucleotides (short DNA or RNA
fragments) or peptides that bind specically to a specic molecule.
PRS-080 is a hepcidin-binding anticalin protein [48]. In cynomolgus
monkeys, PRS-080 has been shown to reduce hepcidin and increase
iron levels [49]. However, there are currently ongoing studies
assessing the eects of PRS-080 anticalin proteins in patients with
anemia in the course of CKD (NCT03325621) [50]. On the other
hand, the aptamer NOX-H94 (lexaptepid pegol [LP]) is a pegylated
oligoribonucleotide that binds to hepcidin with high anity and blocks
its biological function. The eects of this aptamer were conrmed in
vitro and in vivo [51]. The pharmacokinetics, pharmacodynamics,
and safety study of NOX-H94 carried out in 64 healthy volunteers has
shown decrease in hepcidin level and increase in iron and transferrin
saturation in comparison with placebo [52]. LP has been used to
treat ACD in patients with multiple myeloma and lymphoma. LP
caused a signicant increase in Hb (≥1 g/dL) in 5 out of 12 patients.
Responders experienced increase in Hb concentration in RBCs and
reticulocytes, and a decrease in soluble transferrin receptor (sTFR)
level. These results conrm the hypothesis on the ecacy of hepcidin
inhibition in the treatment of cancer-related anemia, especially in
functional iron deciency. However, the patients with symptomatic
iron deciency, hypochromic anemia, without excessive levels of
ferritin, and high sTFR levels were more likely to respond to LP [53].
The group of monoclonal antibodies that serve as direct inhibitors
of hepcidin includes Ab12B9m and LY2787106. Ab12B9m is fully
human monoclonal IgG2 antibody that binds both human and
monkey hepcidin, which is currently under clinical development
128
Acta Haematologica Polonica
[54]. LY2787106 is also a fully humanized monoclonal antibody. In
phase 1 clinical trials, the use of LY2787106 in tumor-associated
anemia resulted in a dose-dependent increase in iron concentration
and transferrin saturation, with good tolerance prole [55]. GDP also
seems to be a promising agent. It is a direct hepcidin inhibitor with
ecacy conrmed in in vitro studies. In animal experiments in mice
with ACD, administration of GDP in combination with iron sulfate
resulted in an increase in Hb concentration, iron, and ferroportin
(FPN) expression and decrease in ferritin level [56].
Potential drugs that prevent hepcidin from binding to ferroportin
include: LY2928057, fursultiamine, thioxolone, and chloroquine.
These compounds prevent internalization and lysosomal degradation
of ferroportin. As a result, hepcidin cannot inhibit iron transport in
intestinal epithelial cells or macrophages [57, 58]. LY2928057 is
a fully humanized IgG4 monoclonal antibody that binds with human
ferroportin. In patients with anemia in the course of CKD, LY2928057
improved iron parameters but did not increase Hb level. Therefore,
the need for concomitant ESAs treatment was suggested [59].
Agents aecting EPO and proinammatory
cytokines
Another option for treating ACDs is to enhance erythropoiesis by
aecting EPO pathway or interfere with inammatory processes
(Fig. 3). This mechanism is utilized by HIF prolyl hydroxylase
inhibitors (HIF-PHI), i.e., molecules that stabilize HIF subunits. The
use of HIF-PHI, similar to hypoxia, activates genes that regulate EPO
concentration and contribute to the regulation of iron metabolism.
As a result, there is an increase in EPO level and iron availability
for erythropoiesis [9, 60]. EPO stimulates erythropoiesis, and via
activation of EPO receptor it induces ERFE and blocks hepcidin
activity [61].
HIF hydroxylase inhibitors appear to be benecial, especially
for patients with CKD-associated anemia. By increasing EPO
concentration, the need for ESAs that are associated with increased
risk of cardiovascular events is reduced. In addition, HIF-PHI can be
administered orally which is the preferred route for patient treatment
[60]. The following HIF-PHI are currently tested: molidustat [62],
GSK1278863 [63], or FG-4592 [64]. The research is focused mainly
on patients with CKD. Phase 3 trials of the safety, tolerability,
and ecacy of molidustat in patients with CKD (both on and o
dialysis) are currently ongoing [62, 65]. Phase 3 on the ecacy of
GSK1278863 in patients with CKD showed a signicant increase
in the reticulocyte count and other RBC parameters, indicating that
further testing is warranted [63]. Also, phase 2 trials of roxadustat
(FG-4592) has shown its potential for oral therapy in dialyzed anemic
patients with CKD [66].
The trials with HIF hydroxylase inhibitors focus not only in the
treatment of ACDs but also in other studies. HIF stabilization with
HIF-PHI is important in regulating hematopoietic stem cell (HSC)
proliferation and HSC regeneration processes. These properties
can be used to protect the bone marrow during radiation therapy, or
to increase mobilization in transplant procedures using granulocyte
colony-stimulating factor (GCS-F) and plerixafor. Therefore, HIF
Fig. 2. Direct and indirect hepcidin inhibitors
Acta Haematologica Polonica
129
hydroxylase inhibitors may have a potential to improve the eects
of treatment in transplant recipients or oncology patients [67]. HIF
also participates in the regulation of multiple processes including:
apoptosis, angiogenesis, cell proliferation, pH regulation, cellular
energy metabolism, and glucose transport [9]. Therefore, further
research on the long-term safety of HIF hydroxylase inhibitors is
needed, with particular attention to the assessment of the risk of
tumorigenesis. It should be emphasized that in rat studies, roxadustat
did not show carcinogenic properties [68]. Another potential threat is
that HIF stabilization may be associated with an increase in circulating
broblast growth factor 23 (FGF23) level, and high concentrations
of FGF23 have been associated with cardiovascular complications
and bone mineralization disorders. However, the increase in FGF23
caused by HIF-PHI is lower than its increase after administration of
ESAs [69, 70].
The increase in EPO synthesis can also be caused by monoclonal
antibodies as discussed earlier, i.e., anti-IL-6 siltuximab [24] and
anti-IL-6 receptor tocilizumab [21, 24]. TNFα is a potential target
for anemia of inammatory diseases, especially in the course of
arthritis or inammatory bowel disease (IBD) [22]. Anti-TNFα drugs
include etanercept (ETN), adalimumab (ADA), iniximab (IFX), and
golimumab (GO). ETN is a recombinant receptor protein conjugated
to the Fc fragment of a human immunoglobulin G1. ETN prevents
TNFα from binding to the receptor and blocks its eects. Other
human monoclonal antibodies, ADA and GO, as well as chimeric
monoclonal antibody IFX and humanized pegylated Fab fragment of
monoclonal antibody, certolizumab, exert similar eects. In patients
with IBD, reduced hepcidin level and improved anemia after anti-
TNFα treatment that modulated hepcidin concentration via IL-6
pathway were observed [71]. It has also been reported that the use of
anti-TNFα drugs in IBD patients resolves anemia without the need for
iron supplementation [72]. ETN, ADA, and IFX improved Hb level in
patients with rheumatoid and psoriatic arthritis [73]. Increased risk of
carcinogenesis after anti-TNFα therapy has not been conrmed so far;
however, careful surveillance is needed in the treated patients [74]. It
should be highlighted that the use of biologicals in the course of IBD
is associated with the potential risk of hematological complications
such as neutropenia or the development of lymphomas, especially
when biologicals are used in combination with other drugs. Further
studies are needed to estimate the exact risk of these complications
[75, 76].
Summary
A number of studies on new drugs for the treatment of ACD are
underway, focusing on various factors involved in iron metabolism
and erythropoiesis regulation. Drugs and tested substances are
intended to directly inhibit hepcidin or its transcription, or counteract
hepcidin activity. The eect of various substances on erythropoiesis
is also assessed, mainly by stabilizing the HIF and inhibiting
proinammatory cytokines. Some of these drugs are already used
in the treatment of inammatory bowel disease or in rheumatology,
while others are in the experimental phase and will take many years
before their introduction to the routine practice. Intensive research
Fig. 3. Drugs aecting erythropoietin and pro-inammatory cytokines
EPO – erythropoietin; IL-6 – interleukin 6; IL-6R – IL-6 receptor; TNFα – tumor necrosis factor α; HIF – hypoxia-inducible factor
130
Acta Haematologica Polonica
and a vast array of tested compounds give hope for the development
of more eective ACD treatments in the coming future.
Authors’ contributions
SSM – the only author.
Conict of interest
None.
Financial support
None.
Ethics
The work described in this article has been carried out in accordance
with The Code of Ethics of the World Medical Association (Declaration
of Helsinki) for experiments involving humans; EU Directive 2010/63/
EU for animal experiments; Uniform requirements for manuscripts
submitted to biomedical journals.
[1] Kassebaum NJ, Jasrasaria R, Naghavi M, et al. A systematic analysis of
global anemia burden from 1990 to 2010. Blood 2014;123:615–24.
[2] Guralnik JM, Eisenstaedt RS, Ferrucci L, Klein HG, Woodman RC.
Prevalence of anemia in persons 65years and older in the United
States: evidence for a high rate of unexplained anemia. Blood
2004;104:2263–8.
[3] Michalak SS, Rupa-Matysek J, Gil L. Comorbidities, repeated
hospitalizations, and age 80 years as indicators of anemia
development in the older population. Ann Hematol 2018;97:1337–47.
[4] Park CH, Valore EV, Waring AJ, Ganz T. Hepcidin, a urinary antimicrobial
peptide synthesized in the liver. J Biol Chem 2001;276:7806–10.
[5] Yiannikourides A, Latunde-Dada GO. A short review of iron
metabolism and pathophysiology of iron gisorders. Medicines (Basel)
2019;6:85.
[6] Weiss G, Ganz T, Goodnough LT. Anemia of inammation. Blood
2019;133:40–50.
[7] Silvestri L, Nai A, Dulja A, Pagani A. Hepcidin and the BMP-SMAD
pathway: an unexpected liaison. Vitam Horm 2019;110:71–99.
[8] Gammella E, Diaz V, Recalcati S, et al. Erythropoietin’s inhibiting
impact on hepcidin expression occurs indirectly. Am J Physiol Regul
Integr Comp Physiol 2015;308:R330–5.
[9] Peyssonnaux C, Zinkernagel AS, Schuepbach RA, et al. Regulation
of iron homeostasis by the hypoxia-inducible transcription factors
(HIFs). J Clin Invest 2007;117:1926–32.
[10] Theurl I, Schroll A, Nairz M, et al. Pathways for the regulation of
hepcidin expression in anemia of chronic disease and iron deciency
anemia in vivo. Haematologica 2011;96:1761–9.
[11] Petzer V, Tymoszuk P, Wake M, et al. A fully human anti-BMP6 antibody
reduces the need for erythropoietin stimulating agent in two rodent
anemia of chronic disease models. Blood 2018;132(Suppl 1):1045.
[12] Sheetz M, Barrington P, Callies S, et al. Targeting the hepcidin-
ferroportin pathway in anaemia of chronic kidney disease. Br J Clin
Pharmacol 2019;85:935–48.
[13] Yu PB, Deng DY, Lai CS, et al. BMP type I receptor inhibition reduces
heterotopic ossication. Nat Med 2008;14:1363–9.
[14] Peterson P, Kim W, Haws H, et al. The ALK-2 inhibitor, TP-0184,
demonstrates high distribution to the liver contributing to signicant
preclinical ecacy in mouse models of anemia of chronic disease.
Blood 2016;128:263.
[15] Poli M, Anower-E-Khuda F, Asperti M, et al. Hepatic heparan
sulfate is a master regulator of hepcidin expression and iron
homeostasis in human hepatocytes and mice. J Biol Chem
2019;294:13292–303.
[16] Poli M, Asperti M, Naggi A, et al. Glycol-split nonanticoagulant
heparins are inhibitors of hepcidin expression in vitro and in vivo.
Blood 2014;123:1564–73.
[17] Babitt JL, Huang FW, Xia Y, Sidis Y, Andrews NC, Lin HY. Modulation
of bone morphogenetic protein signaling in vivo regulates systemic
iron balance. J Clin Invest 2007;117:1933–9.
[18] Petzer V, Theurl I, Weiss G. Established and emerging concepts to
treat imbalances of iron homeostasis in inammatory diseases.
Pharmaceuticals (Basel) 2018;11:135.
[19] Böser P, Seemann D, Liguori MJ, et al. Anti-repulsive guidance
molecule C (RGMc) antibodies increases serum iron in rats
and cynomolgus monkeys by hepcidin downregulation. AAPS
J 2015;17:930–8.
[20] Kovac S, Böser P, Cui Y, et al. Anti-hemojuvelin antibody corrects
anemia caused by inappropriately high hepcidin levels.
Haematologica 2016;101:e173–6.
[21] Isaacs JD, Harari O, Kobold U, Lee JS, Bernasconi C. Eect of tocilizumab
on haematological markers implicates interleukin-6 signalling in the
anaemia of rheumatoid arthritis. Arthritis Res Ther 2013;15:R204.
[22] Song S-NJ, Iwahashi M, Tomosugi N, et al. Comparative evaluation
of the eects of treatment with tocilizumab and TNF-α inhibitors on
serum hepcidin, anemia response and disease activity in rheumatoid
arthritis patients. Arthritis Res Ther 2013;15:R141.
[23] Noguchi-Sasaki M, Sasaki Y, Shimonaka Y, Mori K, Fujimoto-Ouchi K.
Treatment with anti-IL-6 receptor antibody prevented increase in
serum hepcidin levels and improved anemia in mice inoculated with
IL-6-producing lung carcinoma cells. BMC Cancer 2016;16:270.
[24] Casper C, Chaturvedi S, Munshi N, et al. Analysis of inammatory and
anemia-related biomarkers in a randomized, double-blind, placebo-
controlled study of Siltuximab (anti-IL6 monoclonal antibody) in patients
with multicentric Castleman disease. Clin Cancer Res 2015;21:4294–304.
[25] van Rhee F, Casper C, Voorhees PM, et al. A phase 2, open-label,
multicenter study of the long-term safety of siltuximab (an anti-
interleukin-6 monoclonal antibody) in patients with multicentric
Castleman disease. Oncotarget 2015;6:30408–19.
References
Acta Haematologica Polonica
131
[26] Kurzrock R, Voorhees PM, Casper C, et al. A phase I, open-label study
of siltuximab, an anti-IL-6 monoclonal antibody, in patients with
B-cell non-Hodgkin lymphoma, multiple myeloma, or Castleman
disease. Clin Cancer Res 2013;19:3659–70.
[27] Angevin E, Tabernero J, Elez E, et al. A phase I/II, multiple-dose, dose-
escalation study of siltuximab, an anti-interleukin-6 monoclonal
antibody, in patients with advanced solid tumors. Clin Cancer Res
2014;20:2192–204.
[28] Nordgren TM, Bauer CD, Heires AJ, et al. Maresin-1 reduces airway
inammation associated with acute and repetitive exposures to
organic dust. Transl Res 2015;166:57–69.
[29] Wang H, Shi P, Huang C, Liu Q. Maresin 1 ameliorates iron-decient
anemia in IL-10-/-mice with spontaneous colitis by the inhibition of
hepcidin expression though the IL-6/STAT3 pathway. Am J Transl Res
2016;8:2758–66.
[30] Spoto B, Kakkar R, Lo L, et al. Serum erythroferrone levels associate
with mortality and cardiovascular events in hemodialysis and in CKD
patients: a two cohorts study. J Clin Med 2019;8:523.
[31] Arezes J, Foy N, McHugh K, et al. Erythroferrone inhibits the induction
of hepcidin by BMP6. Blood 2018;132:1473–7.
[32] Chappell M, Rivella S. New potential players in hepcidin regulation.
Haematologica 2019;104:1691–3.
[33] Bacchetta J, Zaritsky JJ, Sea JL, et al. Suppression of iron-regulatory
hepcidin by vitamin D. J Am Soc Nephrol 2014;25:564–72.
[34] Zughaier SM, Alvarez JA, Sloan JH, Konrad RJ, Tangpricha V. The
role of vitamin D in regulating the iron-hepcidin-ferroportin axis in
monocytes. J Clin Transl Endocrinol 2014;1:19–25.
[35] Syed S, Michalski ES, Tangpricha V, et al. Vitamin D status is associated
with hepcidin and hemoglobin concentrations in children with
inammatory bowel disease. Inamm Bowel Dis 2017;23:1650–8.
[36] Braithwaite VS, Crozier SR, D’Angelo S, et al. The eect of vitamin D
supplementation on hepcidin, iron status, and inammation in
pregnant women in the United Kingdom. Nutrients 2019;11:190.
[37] Calton EK, Keane KN, Newsholme P, Zhao Y, Soares MJ. The impact
of cholecalciferol supplementation on the systemic inammatory
prole: a systematic review and meta-analysis of high-quality
randomized controlled trials. Eur J Clin Nutr 2017;71:931–43.
[38] Smith EM, Alvarez JA, Kearns MD, et al. High-dose vitamin D3
reduces circulating hepcidin concentrations: a pilot, randomized,
double-blind, placebo-controlled trial in healthy adults. Clin Nutr
2017;36:980–5.
[39] Nielsen OH, Soendergaard C, Vikner ME, Weiss G. Rational
management of iron-deciency anaemia in inammatory bowel
disease. Nutrients 2018;10:82.
[40] Smith EM, Tangpricha V. Vitamin D and anemia: insights into
an emerging association. Curr Opin Endocrinol Diabetes Obes
2015;22:432–8.
[41] Latour C, Kautz L, Besson-Fournier C, et al. Testosterone perturbs
systemic iron balance through activation of epidermal growth factor
receptor signaling in the liver and repression of hepcidin. Hepatology
2014;59:683–94.
[42] Guo W, Bachman E, Li M, et al. Testosterone administration inhibits
hepcidin transcription and is associated with increased iron
incorporation into red blood cells. Aging Cell 2013;12:280–91.
[43] Roy CN, Snyder PJ, Stephens-Shields AJ, et al. Association of
testosterone levels with anemia in older men: a controlled clinical
trial. JAMA Intern Med 2017;177:480–90.
[44] Yang Q, Jian J, Katz S, Abramson SB, Huang X. 17β-estradiol inhibits
iron hormone hepcidin through an estrogen responsive element
half-site. Endocrinology 2012;153:3170–8.
[45] Liu J-Y, Zhang Y, You R-X, Zeng F, Guo D, Wang K-P. Polysaccharide
isolated from Angelica sinensis inhibits hepcidin expression in rats
with iron deciency anemia. J Med Food 2012;15:923–9.
[46] Bradley RR, Cunni PJ, Pereira BJ, Jaber BL. Hematopoietic eect of
Radix angelicae sinensis in a hemodialysis patient. Am J Kidney Dis
1999;34:349–54.
[47] Zhang Y, Cheng Y, Wang N, Zhang Q, Wang K. The action of JAK, SMAD
and ERK signal pathways on hepcidin suppression by polysaccharides
from Angelica sinensis in rats with iron deciency anemia. Food Funct
2014;5:1381–8.
[48] Renders L, Budde K, Rosenberger C, et al. First-in-human phase I
studies of PRS-080#22, a hepcidin antagonist, in healthy volunteers
and patients with chronic kidney disease undergoing hemodialysis.
PLoS One 2019;14:e0212023.
[49] Hohlbaum AM, Gille H, Trentmann S, et al. Sustained plasma hepcidin
suppression and iron elevation by Anticalin-derived hepcidin
antagonist in cynomolgus monkey. Br J Pharmacol 2018;175:1054–65.
[50] Rothe C, Skerra A. AnticalinÒ proteins as therapeutic agents in human
diseases. BioDrugs 2018;32:233–43.
[51] Schwoebel F, van Eijk LT, Zboralski D, et al. The eects of the anti-
hepcidin Spiegelmer NOX-H94 on inammation-induced anemia in
cynomolgus monkeys. Blood 2013;121:2311–5.
[52] Boyce M, Warrington S, Cortezi B, et al. Safety, pharmacokinetics
and pharmacodynamics of the anti-hepcidin Spiegelmer lexaptepid
pegol in healthy subjects. Br J Pharmacol 2016;173:1580–8.
[53] Georgiev P, Lazaroiu M, Ocroteala L, et al. Abstract 3847: The anti-
hepcidin Spiegelmer Lexaptepid Pegol (NOX-H94) as treatment of
anemia of chronic disease in patients with multiple myeloma, low grade
lymphoma, and CLL: a phase II pilot study. Cancer Res 2014;74:3847.
[54] Xiao JJ, Krzyzanski W, Wang Y-M, et al. Pharmacokinetics of
anti-hepcidin monoclonal antibody Ab 12B9m and hepcidin in
cynomolgus monkeys. AAPS J 2010;12:646–57.
[55] Vadhan-Raj S, Abonour R, Goldman JW, et al. A rst-in-human phase1
study of a hepcidin monoclonal antibody, LY2787106, in cancer-
associated anemia. J Hematol Oncol 2017;10:73.
[56] Angmo S, Tripathi N, Abbat S, et al. Identication of guanosine
5’-diphosphate as potential iron mobilizer: preventing the hepcidin-
ferroportin interaction and modulating the interleukin-6/Stat-3
pathway. Sci Rep 2017;7:40097.
[57] Katsarou A, Pantopoulos K. Hepcidin therapeutics. Pharmaceuticals
(Basel) 2018;11:127.
[58] Ross SL, Biswas K, Rottman J, et al. Identication of antibody and
small molecule antagonists of ferroportin-hepcidin interaction. Front
Pharmacol 2017;8:838.
[59] Barrington P, Sheetz MJ, Callies S, et al. Safety, tolerability,
pharmacokinetics and pharmacodynamics of an anti-ferroportin
antibody in patients with anemia due to chronic renal failure. Blood
2016;128:1280.
132
Acta Haematologica Polonica
[60] Gupta N, Wish JB. Hypoxia-inducible factor prolyl hydroxylase
inhibitors: apotential new treatment for anemia in patients with CKD.
Am J Kidney Dis 2017;69:815–26.
[61] Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identication
of erythroferrone as an erythroid regulator of iron metabolism. Nat
Genet 2014;46:678–84.
[62] Akizawa T, Taguchi M, Matsuda Y, Iekushi K, Yamada T, YamamotoH.
Molidustat for the treatment of renal anaemia in patients with
dialysis-dependent chronic kidney disease: design and rationale of
three phase III studies. BMJ Open 2019;9:e026602.
[63] Ariazi JL, Duy KJ, Adams DF, et al. Discovery and preclinical
characterization of GSK1278863 (daprodustat), a small molecule
hypoxia inducible factor-prolyl hydroxylase inhibitor for anemia.
JPharmacol Exp Ther 2017;363:336–47.
[64] Chen CX, Li YC, Chan SL, Chan KH. Anaemia and type 2 diabetes:
implications from a retrospectively studied primary care case series.
Hong Kong Med J 2013;19:214–21.
[65] Yamamoto H, Taguchi M, Matsuda Y, Iekushi K, Yamada T, Akizawa T.
Molidustat for the treatment of renal anaemia in patients with non-
dialysis-dependent chronic kidney disease: design and rationale of
two phase III studies. BMJ Open 2019;9:e026704.
[66] Chen N, Qian J, Chen J, et al. Phase 2 studies of oral hypoxia-inducible
factor prolyl hydroxylase inhibitor FG-4592 for treatment of anemia
in China. Nephrol Dial Transplant 2017;32:1373–86.
[67] Forristal CE, Levesque J-P. Targeting the hypoxia-sensing pathway in
clinical hematology. Stem Cells Transl Med 2014;3:135–40.
[68] Beck J, Henschel C, Chou J, Lin A, Del Balzo U. Evaluation of the
carcinogenic potential of roxadustat (FG-4592), a small molecule
inhibitor of hypoxia-inducible factor Prolyl hydroxylase in CD-1 mice
and Sprague Dawley rats. Int J Toxicol 2017;36:427–39.
[69] Flamme I, Ellinghaus P, Urrego D, Krüger T. FGF23 expression in rodents
is directly induced via erythropoietin after inhibition of hypoxia
inducible factor proline hydroxylase. PLoS One 2017;12:e0186979.
[70] Wheeler JA, Clinkenbeard EL. Regulation of broblast growth factor
23 by iron, EPO, and HIF. Curr Mol Biol Rep 2019;5:8–17.
[71] Cavallaro F, Duca L, Pisani LF, et al. Anti-TNF-mediated modulation
of prohepcidin improves iron availability in inammatory bowel
disease, in an IL-6-mediated fashion. Can J Gastroenterol Hepatol
2017;2017:6843976.
[72] Lucendo AJ, Roncero Ó, Serrano-Duenas MT, et al. Eects of anti-TNF-
alpha therapy on hemoglobin levels and anemia in patients with
inammatory bowel disease. Dig Liver Dis 2020;52:400–7.
[73] Corrado A, Di Bello V, d’Onofrio F, Maruotti N, Cantatore FP. Anti-
TNF-α eects on anemia in rheumatoid and psoriatic arthritis. Int J
Immunopathol Pharmacol 2017;30:302–7.
[74] Chen Y, Friedman M, Liu G, Deodhar A, Chu C-Q. Do tumor necrosis
factor inhibitors increase cancer risk in patients with chronic immune-
mediated inammatory disorders? Cytokine 2018;101:78–88.
[75] Bessissow T, Renard M, Homan I, Vermeire S, Rutgeerts P,
Van Assche G. Review article: non-malignant haematological
complications of anti-tumour necrosis factor alpha therapy. Aliment
Pharmacol Ther 2012;36:312–23.
[76] Yang C, Huang J, Huang X, et al. Risk of lymphoma in patients with
inammatory bowel disease treated with anti-tumour necrosis factor
alpha agents: a systematic review and meta-analysis. J Crohns Colitis
2018;12:1042–52.
... Interleukina 6 stymuluje syntezę hepcydyny, która jest regulatorem metabolizmu żelaza i jednocześnie mediatorem procesów zapalnych. Także inne cytokiny prozapalne, zwłaszcza IL1, TNFα, interferon γ, hamują proliferację komórek progenitorowych układu czerwonokrwinkowego i prowadzą do rozwoju anemii chorób przewlekłych (Anemia of Chronic Disease -ACD), która jest częsta w populacji geriatrycznej [3,17]. W procesach starzenia znaczenie ma również stres oksydacyjny, ze zwiększeniem obecności wolnych rodników tlenowych, które doprowadzają do uszkodzeń błon komórkowych, białek i DNA. ...
... W każdym przypadku anemii należy znaleźć i leczyć jej przyczyny. Opis leczenia różnych typów anemii nie jest celem tego artykułu i został zaprezentowany w innych opracowaniach [17,39,48]. ...
Article
Full-text available
Anemia i depresja są powszechne u osób w podeszłym wieku i stanowią istotny problem medyczny, społeczny i ekonomiczny świata. Obydwa zaburzenia wiążą się z niską jakością życia, wielochorobowością, zwiększonym ryzykiem śmierci. Równoczesne występowanie anemii i depresji zostało potwierdzone w badaniach, ale mechanizmy i konsekwencje kliniczne tej koegzystencji nie zostały dotąd w pełni wyjaśnione. Istnieje szereg powiązań pomiędzy anemią a depresją takich jak wspólne czynniki etiologiczne, niski status socjoekonomiczny pacjentów, niskie wykształcenie oraz mniejsza aktywność fizyczna. W obydwu zaburzeniach stwierdza się te same patologiczne zmiany zależne od wieku (zwłaszcza obecność stanu zapalnego i stresu oksydacyjnego, zmiany degeneracyjne tkanek i narządów), niedobory żelaza czy witamin (B12, kwasu foliowego), zaburzenia hormonalne (zwłaszcza choroby tarczycy, niedobory hormonów płciowych). Zarówno anemia jak i depresja u osób starszych są w niewystarczającym stopniu rozpoznawane i w rezultacie często nie są leczone. Diagnostyka i leczenie obu zaburzeń u osób podeszłym wieku wykazują pewne różnice w stosunku do postępowania w innych grupach wiekowych. Niniejsza praca zawiera przegląd literatury dotyczących wspólnego występowania anemii i depresji u osób w podeszłym wieku oraz rekomendacje postępowania.
... The management of ACD involves the utilization of various approaches [13,14]. Currently, the treatment of ACD commonly consists of the application of erythropoiesis-stimulating agents (ESAs) and replenishing iron. ...
... Our study has several limitations. It was a retrospective study, meaning that other possible factors influencing iron metabolism could not be taken into account [14]. Also, the impact of disease stage and other possible therapy complications [15][16][17][18][19] were not analysed. ...
Article
Introduction: Nowadays, a significant number of children with acute leukemia can be cured. Iron overload, related to blood transfusions and its long-term complications, remains a problem. Elevated ferritin concentration is often observed in this group. The aim of this study was to evaluate the prognostic value of serum ferritin on long-term outcomes in children treated for acute leukemia. Material: We studied 71 patients treated for acute lymphoblastic (ALL) or myeloblastic (AML) leukemia between 2005 and 2011. Serum ferritin concentration, serum transaminases activity, lactic dehydrogenase and C-reactive protein levels (CRP) were analysed. Serum ferritin >1,000 μg/L was considered to be a marker of iron overload. Results: Thirty-seven patients (52.1%) had iron overload. Ferritin serum concentration correlated with alanine aminotranferase activity (p =0.001) and CRP concentration (p =0.012). A total of 19 (26.76%) patients died during follow-up. Ferritin level was higher in patients with AML vs. ALL. There was a significant difference in long-term outcomes with respect to high ferritin concentrations, both in patients undergoing haematopoietic cell transplantation (HCT) and in the non-HCT group. Conclusions: In both groups, patients with higher ferritin concentrations had worse overall and event-free survivals and a higher relapse incidence. Ferritin concentration >1,000 μg/L was the strongest determinant of long-term treatment outcome. Ferritin serum concentration >1,000 μg/L is an adverse prognostic marker of survival in children with acute leukemia treated with chemotherapy with or without HCT.
Article
Full-text available
Iron is a vital trace element for humans, as it plays a crucial role in oxygen transport, oxidative metabolism, cellular proliferation, and many catalytic reactions. To be beneficial, the amount of iron in the human body needs to be maintained within the ideal range. Iron metabolism is one of the most complex processes involving many organs and tissues, the interaction of which is critical for iron homeostasis. No active mechanism for iron excretion exists. Therefore, the amount of iron absorbed by the intestine is tightly controlled to balance the daily losses. The bone marrow is the prime iron consumer in the body, being the site for erythropoiesis, while the reticuloendothelial system is responsible for iron recycling through erythrocyte phagocytosis. The liver has important synthetic, storing, and regulatory functions in iron homeostasis. Among the numerous proteins involved in iron metabolism, hepcidin is a liver-derived peptide hormone, which is the master regulator of iron metabolism. This hormone acts in many target tissues and regulates systemic iron levels through a negative feedback mechanism. Hepcidin synthesis is controlled by several factors such as iron levels, anaemia, infection, inflammation, and erythropoietic activity. In addition to systemic control, iron balance mechanisms also exist at the cellular level and include the interaction between iron-regulatory proteins and iron-responsive elements. Genetic and acquired diseases of the tissues involved in iron metabolism cause a dysregulation of the iron cycle. Consequently, iron deficiency or excess can result, both of which have detrimental effects on the organism.
Article
Full-text available
Hepcidin is a liver-derived peptide hormone that controls systemic iron homeostasis. Its expression is regulated by the bone morphogenetic protein 6 (BMP6)/SMAD1/5/8 pathway and by the proinflammatory cytokine interleukin 6 (IL-6). Proteoglycans that function as receptors of these signaling proteins in the liver are commonly decorated by heparan sulfate, but the potential role of hepatic heparan sulfate in hepcidin expression and iron homeostasis is unclear. Here, we show that modulation of hepatic heparan sulfate significantly alters hepcidin expression and iron metabolism both in vitro and in vivo. Specifically, enzymatic removal of heparan sulfate from primary human hepatocytes, CRISPR/Cas9 manipulation of heparan sulfate biosynthesis in human hepatoma cells, or pharmacological manipulation of heparan sulfate-protein interactions using sodium chlorate or surfen dramatically reduced baseline and BMP6/SMAD1/5/8-dependent hepcidin expression. Moreover inactivation of the heparan sulfate biosynthetic gene N-deacetylaseN-sulfotransferase 1 (Ndst1) in murine hepatocytes (Ndst1f/fAlbCre+) reduced hepatic hepcidin expression and caused a redistribution of systemic iron, leading to iron accumulation in the liver and serum of mice. Manipulation of heparan sulfate had a similar effect on IL-6-dependent hepcidin expression in vitro and suppressed IL-6-mediated iron redistribution induced by lipopolysaccharide in vivo. These results provide compelling evidence that hepatocyte heparan sulfate plays a key role in regulating hepcidin expression and iron homeostasis in mice and in human hepatocytes.
Article
Full-text available
Introduction New medications for anaemia associated with chronic kidney disease (CKD) are desirable, owing to the limitations of erythropoiesis-stimulating agents (ESAs), the current standard of care. Molidustat is a novel hypoxia-inducible factor prolyl-hydroxylase inhibitor that stimulates erythropoietin production, predominately in the kidney. We report methodological details of three phase III trials, named MolIdustat once dailY improves renal Anaemia By Inducing erythropoietin (MIYABI), designed primarily to investigate the efficacy of molidustat therapy in adults with renal anaemia and dialysis-dependent CKD. Methods and analysis MIYABI Haemodialysis-Correction (HD-C) is a single-arm trial (24-week treatment duration) in approximately 25 patients on haemodialysis, currently untreated with ESAs. MIYABI Peritoneal Dialysis (PD) is a single-arm trial (36 week treatment duration) in approximately 50 patients on peritoneal dialysis, treated or untreated with ESAs. MIYABI Haemodialysis-Maintenance (HD-M) is a randomised, active-controlled, double-blinded, double-dummy trial (52-week treatment duration) comparing molidustat with darbepoetin alfa in approximately 225 patients on haemodialysis, treated with ESAs. Molidustat (starting dose 75 mg/day) will be titrated 4-weekly to maintain haemoglobin in predetermined target ranges. The primary objective is to evaluate the efficacy of molidustat, using the following measures: the rate of rise in haemoglobin (g/L/week) at the first dose change up to week 8 (MIYABI HD-C); responder rate (MIYABI HD-C and MIYABI PD); mean haemoglobin level during weeks 33–36 and non-inferiority to darbepoetin alfa shown by change in mean haemoglobin level from baseline (MIYABI HD-M). The secondary objectives are to assess safety, pharmacokinetics and pharmacodynamics. These trials will provide the first evaluations of molidustat therapy in patients receiving either peritoneal dialysis or currently untreated with ESAs on haemodialysis, and provide further evidence in patients treated with ESAs on haemodialysis. Ethics and dissemination The protocols were approved by ethics committees at all participating sites. The trials will be conducted in accordance with the Declaration of Helsinki and Good Clinical Practice. Results arising from these studies will be published in peer-reviewed journal(s). Trial registration numbers NCT03351166 ; Pre-results, NCT03418168 ; Pre-results, NCT03543657 ; Pre-results
Article
Full-text available
Introduction Anaemia is a common complication of chronic kidney disease (CKD). Owing to the limitations of erythropoiesis-stimulating agents (ESAs), the current standard of care, there is a need to develop new therapies. Hypoxia-inducible factor prolyl-hydroxylase (HIF-PH) inhibitors might be a promising new treatment option. Molidustat is an oral HIF-PH inhibitor that stimulates the endogenous, predominantly renal, production of erythropoietin and was generally well tolerated in phase IIb clinical trials. Here, we report the design and rationale of two studies from the molidustat phase III programme: MolIdustat once dailY improves renal Anaemia By Inducing erythropoietin (MIYABI). Methods and analysis MIYABI Non-Dialysis-Correction (ND-C) and MIYABI Non-Dialysis-Maintenance (ND-M) are randomised, open-label, parallel-group, multicentre studies that aim to demonstrate the efficacy of molidustat treatment compared with darbepoetin alfa in patients with anaemia and non-dialysis-dependent CKD. The secondary objectives are to assess the safety, pharmacokinetics and pharmacodynamics of molidustat treatment. MIYABI ND-C will recruit patients currently untreated with ESAs, whereas patients treated with an ESA will enter MIYABI ND-M. Each study will recruit 150 patients who will be randomised in a 1:1 ratio to receive either molidustat or darbepoetin alfa for 52 weeks, with efficacy evaluated during weeks 30–36. Study drug doses will be titrated regularly using an interactive voice/web response system with the aim of maintaining the patients’ haemoglobin (Hb) levels between ≥110 and <130 g/L. The primary objective will be achieved if, in molidustat-treated patients, the mean Hb level remains within the target range during the evaluation period, and if the change in the mean Hb level at evaluation time points from baseline is non-inferior to darbepoetin alfa. Ethics and dissemination The protocols were approved by ethics committees at all participating sites. These studies will be conducted in accordance with the Declaration of Helsinki and the Good Clinical Practice guidelines. Results arising from these studies will be published in peer-reviewed journal(s). Trial registration numbers NCT03350321 ; Pre-results, NCT03350347 ; Pre-results.
Article
Anemia of chronic disease (ACD) is the most common cause of anemia in hospitalized patients. The underlying pathophysiological mechanisms are manifold, including reduced Erythropoietin (EPO) availability and sensitivity, direct negative effects of inflammatory cytokines on erythropoiesis and functional iron deficiency due to iron restriction mainly in the reticuloendothelial system (RES). The latter can be ascribed to increased hepcidin levels, a small liver derived peptide that has been found to be the key iron regulator. Up to date, most patients suffering from ACD are treated with a combination of both ESA (Erythropoietin Stimulating Agent) and intravenous (i.v.) iron to maintain hemoglobin (Hb) levels. Despite this combination therapy a significant number of patients require increasing doses of i.v. iron and ESA during their medical history - often resulting in continuous, potential toxic iron overload and ESA doses that exceed the acceptable range. As hepcidin is central to the iron-restrictive phenotype in ACD, several therapeutic approaches of hepcidin modulation have been investigated to overcome iron overload and EPO resistance. Some of these therapies are currently investigated in early clinical trials. We here report the effects of a fully human anti-BMP6 antibody on anemia, iron metabolism, erythropoiesis and ESA dosing in two different, well established rodent models of ACD. As BMPs, mainly BMP2 and BMP6, have been reported to be involved in hepcidin control, with knock out mice showing very low hepcidin levels even in an inflammatory setting, a fully human anti-BMP6 antibody has been developed to suppress hepcidin levels. We tested the antibody treatment in two distinct ACD animal models: First, a rat arthritis model (PG-APS in Lewis rats) and second, a Chronic Kidney Disease (CKD) mouse anemia model (Adenine model in C57BL/6 mice). Both models present with long-lasting anemia as seen in humans suffering from ACD. Mice and rats were treated with different doses of the anti-BMP6 antibody with and without ESA. Whole blood count, serum iron parameters (including hepcidin), bone marrow erythropoiesis determined by FACS analysis, cytokine expression and iron metabolism gene expression in spleen, liver and kidney were analyzed. Anti-BMP6 as well as ESA monotherapy resulted in a net increase in Hb level but only anti-BMP6 treatment significantly increased MCV and MCH, which can be ascribed to effective iron mobilization. In contrast, ESA therapy raised Hb levels by increasing red blood cell numbers. Of note, mere i.v. iron supplementation (sodium ferric gluconate), even at high doses, was not able to restore Hb levels to the same extent as the anti-BMP6 monotherapy. Strikingly, combination of both, ESA and anti-BMP6 treatment, had a synergistic effect on Hb levels, especially in the rat PG-APS model. Combination therapy of low ESA doses that only had a modest effect as a monotherapy, led to a dramatic increase in Hb levels, even exceeding those seen in healthy rats. Based on these results, additional experiments were performed to investigate the potential of this combination treatment to reduce ESA doses. Indeed, when anti-BMP6 was combined with a significantly reduced total ESA dose Hb levels were corrected to normal values in disease animals. Anti-BMP6 treatment also led to a significant decrease of iron deposition in the spleen with no iron deposition in parenchymal organs, indicating that the freed-up iron was effectively used for erythropoiesis and not just distributed elsewhere. In summary, anti-BMP6 therapy worked synergistically with ESA treatment in two different models of ACD leading to significantly increased Hb levels, a reduced ESA need as well as reduced iron overload in the RES. Furthermore, these experiments clearly show that treatment of ACD, being a complex multifactorial disease, benefits from using a combination of diversified approaches to overcome anemia and significantly reduce the dose of each therapeutic. Disclosures Wake: Kymab Ltd.: Employment. Bayliss:Kymab Ltd.: Employment. Papworth:Kymab Ltd.: Employment. Carvalho:Kymab Ltd.: Employment. Deantonio:Kymab Ltd.: Employment. Weiss:Kymab Ltd.: Consultancy. Germaschewski:Kymab Ltd.: Employment. Theurl:Kymab Ltd.: Consultancy, Research Funding.
Article
Background: Tumor necrosis factor- (TNF-)is involved in inducing inflammatory anemia. The potential effect of anti–TNF- agents on anemia in inflammatory bowel diseases (IBD) is still unknown. Methods: Analytical data and disease characteristics from 362 IBD patients [271 CD/91UC) treated with anti-TNF- drugs were retrospectively collected. Effects ondisease activity, bloodmarkers andprevalence of anemia were assessed after 6 and 12 months of therapy. Results: 29.3% patients presented anemia at baseline, and significantly reduced to 14.4% and 7.8% after 6 and 12 months of therapy, respectively. Mean ± SD Hb levels increased significantly at month 6, and this increase was sustained at 12 months. Serum markers of iron metabolism increased significantly compared to baseline, as disease activity measured by C-reactive protein (CRP) was reduced. All these effects were observed independently for CD and UC, and were independent of iron supplementation during treatment. Anemia at baseline (OR 4.09; 95%CI 1.98–8.45) and elevated CRP (OR 3.45; 95CI 1.29–9.22) were independently associated with risk of persistent anemia, as well as iron replacement during therapy (OR 4.36; 95%CI 2.07–9.16). Conclusions: Controlling disease activity with anti-TNF- therapy significantly and independently associated with resolution of anemia in IBD, with no relevant role for iron replacement therapy
Article
Decreased hepcidin mobilizes iron, which facilitates erythropoiesis, but excess iron is pathogenic in beta-thalassemia and other iron-loading anaemias. Erythropoietin (EPO) enhances erythroferrone (ERFE) synthesis by erythroblasts, and ERFE suppresses hepatic hepcidin production, through an unknown mechanism. The BMP/SMAD pathway in the liver is critical for control of hepcidin, and we show that EPO suppressed hepcidin and other BMP target genes in vivo in a partially ERFE-dependent manner. Furthermore, recombinant ERFE suppressed the hepatic BMP/SMAD pathway independently of changes in serum and liver iron, and in vitro, ERFE decreased SMAD 1/5/8 phosphorylation and inhibited expression of BMP target genes in hepatoma cells. ERFE specifically abrogated the induction of hepcidin by BMP5, BMP6 and BMP7, but had no or very little effect on hepcidin induction by BMP2, 4, 9 or Activin B. A neutralising anti-ERFE antibody prevented the ability of ERFE to inhibit hepcidin induction by BMP5, 6 and 7. Cell-free Homogeneous Time Resolved Fluorescence assays showed that BMP5, BMP6 and BMP7 competed with anti-ERFE for binding to ERFE. Biacore analysis showed that ERFE binds to BMP6 with a higher affinity compared to its binding to BMP2, BMP4 or Activin B, and does not bind to GDF15. We propose that ERFE suppresses hepcidin by inhibiting hepatic BMP/SMAD signaling via preferentially binding and impairing the function of an evolutionarily closely related BMP sub-group consisting of BMP5, BMP6 and BMP7. These findings indicate that ERFE can act as a natural ligand trap generated by stimulated erythropoiesis in order to regulate availability of iron. Disclosures Arezes: Pfizer: Research Funding. Foy:Pfizer: Employment. McHugh:Pfizer: Research Funding. Sawant:Pfizer: Employment. Benard:Pfizer: Employment. Quinkert:Pfizer: Research Funding. Terraube:Pfizer: Employment. Brinth:Pfizer: Employment. Tam:Pfizer: Employment. LaVallie:Pfizer: Employment. Cunningham:Pfizer: Employment. Lambert:Pfizer: Employment. Draper:Pfizer: Research Funding. Jasuja:Pfizer: Employment. Drakesmith:La Jolla Pharmaceutical Company: Research Funding; Pfizer: Research Funding; Alnylam: Consultancy; Kymab: Membership on an entity's Board of Directors or advisory committees.
Article
Introduction: The iron transporter ferroportin is a membrane protein expressed in enterocytes that absorb dietary iron, macrophages of the spleen and liver that recycle iron from old red blood cells (RBCs), and hepatocytes that store and release iron according to body needs. Hepcidin (HEPC) regulates the absorption, plasma concentrations, and tissue distribution of iron through interactions with ferroportin, leading to degradation of ferroportin. LY2928057 (LY), a humanized immunoglobulin (IgG4) monoclonal antibody, binds to ferroportin and prevents the HEPC-mediated degradation of ferroportin without affecting iron efflux. Objectives: Objectives were to assess safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) properties of LY in patients with chronic kidney disease (CKD) on hemodialysis (HD) after intravenous (IV) multiple doses of LY. The study, compliant with Helsinki declaration, was approved by institutional ethic review board and subjects provided written informed consent prior to enrolment. Methods: CKD patients (N=21) received IV doses (300, 600, or 1000 mg every 2 weeks; total, 3 doses) of LY following discontinuation (when LY treatment was initiated) of erythropoiesis-stimulating agents (ESAs) and IV iron. Seven CKD patients received placebo. Safety assessments included: standard laboratory safety tests, vital signs, ECGs, and anti-drug antibodies. PD data comprised serum iron, transferrin saturation (TSAT), ferritin, HEPC, RBC count and hemoglobin (Hb). PK samples (up to 3 months post last dose) were assayed using a validated enzyme-linked immunosorbent assay method. LY PK data were analyzed using standard non-compartmental PK analysis. Summary statistics were used to describe LY PD data (parameters and ratios relative to baseline). A Bayesian analysis of the absolute change in Hb at week 6 relative to baseline was performed with the following success criterion: >60 % posterior probability of ≥0.8 g/dL difference between any LY dose group and placebo. Results - Safety CKD patients (19 of 28 were male), with mean (SD) age and weight of 53.6 (8) years and 87.35 (18) kg, participated. LY was well tolerated; serious adverse events (syncope, anemia, hypertension, respiratory failure/staphylococcal sepsis/pneumonia) were reported by 3 patients. These were not attributed to study drug. Results - Pharmacokinetics LY maximum concentrations were generally measured at the end of the 30-min infusion and concentrations decreased thereafter in a multiexponential manner. LY area under the concentration time curve (AUC or exposure) increased greater than dose proportionally from 300 to 600 mg and was roughly dose proportional from 600 to 1000 mg. This observation is consistent with binding, target-mediated clearance. LY volume of distribution was small (mean 4L) and LY clearance was low (mean 0.09 L/h) in the 600 to 1000 mg dose range, leading to a mean terminal half-life of 8 days (ranging from 5.5 to 13 days). The dosing of LY during HD did not significantly alter LY exposure. Results - Pharmacodynamics A dose-related increase in iron, with a maximum effect approximately 24 h after dose, was observed. Iron values returned to baseline in approximately 2-weeks post-LY dose. Concurrently, an increase in TSAT was observed (Figure 1). In addition, a decrease in ferritin levels was observed (Figure 2, after third dose, approximately 12% and 20% decrease relative to baseline at the 600 and 1000 mg doses, respectively). HEPC levels increased following LY administration. This is likely explained by the feed-back regulatory response of the body to the iron increase. Figure 3 illustrates that RBC and Hb declined to a lesser extent with LY 600 mg and 1000 mg dose levels compared to 300 mg dose levels and placebo treatment. The Bayesian analysis determined a posterior probability of 58% (less than 60%), for a greater than 0.8 g/dL difference between LY treatment and placebo, in the absolute change in Hb at week 6 relative to baseline Conclusion LY in CKD patients was well tolerated; no safety signals or trends were identified. Expected changes in PD markers (serum iron, HEPC, TSAT, ferritin, RBC, and Hb) were observed after LY administration; however, the effect on Hb did not meet the pre-defined success criterion. It is possible that co-administration of an ESA with LY is required for the increased iron to be optimally used for Hb synthesis in RBC. Disclosures Barrington: Eli Lilly and Company: Employment, Equity Ownership. Sheetz:Eli Lilly and Company: Employment, Equity Ownership. Callies:Eli Lilly and Company: Employment, Equity Ownership. Waters:Eli Lilly and Company: Employment. Berg:Eli Lilly and Company: Employment, Equity Ownership. Pappas:Eli Lilly and Company: Employment. Marbury:Olando Clinical Research Center: Employment, Equity Ownership. Berg:Davita Clinical Research: Employment, Other: Full time employee of Davita Clinical Research, one of the research sites.
Article
In individuals with chronic inflammatory diseases, such as cancer or rheumatoid arthritis, constitutive signaling through ALK2, a member of the bone morphogenetic protein (TGFβ/BMP) receptor family, leads to debilitating anemia, commonly referred to as anemia of chronic disease (ACD). Activation of ALK2, like other members of the BMP receptor family, leads to the phosphorylation and activation of SMAD family transcription factors via signal transduction and subsequent activation of gene expression. Activation of ALK2 in the liver induces the SMAD-driven transcription of the peptide hormone hepcidin which, by promoting the degradation of the iron transporter ferroportin, leads to reduced serum iron levels and subsequent functional anemia. Lowering constitutively elevated hepcidin levels by inhibiting ALK2 kinase activity is a potentially viable therapeutic strategy for ACD. Current therapeutic approaches for ACD rely on transfusions, intravenous iron and the use of erythropoietin-based therapies, none of which address the underlying pathological deficit of functionally low iron levels. TP-0184 is a small-molecule, selective inhibitor of ALK2 kinase activity (IC50 = 5 nM). TP-0184 has demonstrated profound preclinical activity in three mouse efficacy models for ACD. In model 1, TP-0184 reversed hepcidin induction in mice treated with turpentine oil. In model 2, TP-0184 abrogated reductions in hemoglobin and total red blood cell counts induced by intraperitoneal injection with heat-inactivated Brucella abortus. In model 3, TP-0184 reversed elevated hepcidin levels in TC-1 tumor bearing mice. Plasma and liver pharmacokinetics in mice revealed that TP-0184 has a high volume of distribution (Vd = 30.8) and accumulates at high concentrations in the liver (Cmax of 292 mM following a single oral dose of at 20 mg/kg). In rat multi-dose tolerability studies, TP-0184 caused no adverse effects when dosed at 200 mg/kg for 7 days, far exceeding the dose levels required to produce efficacy (25 mg/kg). These data suggest that favorable distribution to the liver may play a significant role in the preclinical efficacy of TP-0184 and provide evidence of a significant therapeutic window. Collectively these studies support the clinical evaluation of TP-0184 as an alternative treatment for ACD. Disclosures Peterson: Tolero Pharmaceuticals: Employment. Kim:Tolero Pharmaceuticals: Employment. Haws:Tolero Pharmaceuticals: Employment. Whatcott:Tolero Pharmaceuticals: Employment. Siddiqui-Jain:Tolero Pharmaceuticals: Employment. Bearss:Tolero Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties. Warner:Tolero Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties.