Вы находитесь на странице: 1из 9

Blood Reviews 24 (2010) 39–47

Contents lists available at ScienceDirect

Blood Reviews
journal homepage: www.elsevier.com/locate/blre

REVIEW

Anemia in renal disease: Diagnosis and management


Christina E. Lankhorst a, , Jay B. Wish b,*
a
Division of Nephrology, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States
b
Case Western Reserve University, Division of Nephrology, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States

a r t i c l e i n f o s u m m a r y

Keywords: Chronic kidney disease (CKD) is a widespread health problem in the world and anemia is a common com-
Anemia plication. Anemia conveys significant risk for cardiovascular disease, faster progression of renal failure
Chronic kidney disease and decreased quality of life. Patients with CKD can have anemia for many reasons, including but not
Erythropoietin invariably their renal insufficiency. These patients require a thorough evaluation to identify and correct
Erythropoiesis-stimulating agents
causes of anemia other than erythropoietin deficiency. The mainstay of treatment of anemia secondary to
Ferritin
Transferrin saturation
CKD has become erythropoiesis-stimulating agents (ESAs). The use of ESAs does carry risks and these
Iron deficiency agents need to be used judiciously. Iron deficiency often co-exists in this population and must be evalu-
Erythropoietin resistance ated and treated. Correction of iron deficiency can improve anemia and reduce ESA requirements. Partial,
but not complete, correction of anemia is associated with improved outcomes in patients with CKD.
Ó 2009 Elsevier Ltd. All rights reserved.

Introduction Pathophysiology of anemia in patients with CKD

Chronic kidney disease (CKD) affects approximately 26 million Anemia is defined by the World Health Organization as a Hb
adults in the United Sates and millions of others are at risk.1 CKD concentration less than 13.0 g/dL in adult males and non-
is associated with significant morbidity and mortality, and these menstruating females and less than 12.0 g/dL in menstruating
patients face many other medical problems related to CKD. One of females.3 Anemia is a common problem in patients with CKD,
the major medical issues facing this population is anemia, which and its incidence increases as glomerular filtration rate declines.
often develops early in the course of CKD and contributes to poor Population studies such as the National Health and Nutrition
quality of life. It has been shown to be strongly predictive of adverse Examination Survey (NHANES) by the National Institutes of Health
effects, including complications and death from cardiovascular and the Prevalence of Anemia in Early Renal Insufficiency (PAERI)
causes.2 Prior to the availability of human recombinant erythro- study suggest that the incidence of anemia is less than 10% in
poietin, patients receiving chronic dialysis treatment frequently CKD stages 1 and 2, 20–40% in CKD stage 3, 50–60% in CKD stage
required blood transfusions, exposing them to iron overload, viral 4 and more than 70% in CKD stage 5.4,5
hepatitis and HIV, and increasing production of antibodies to The cause of anemia in patients with CKD is multifactorial. The
human antigens which can severely limit transplantation options. most well-known cause is inadequate erythropoietin (EPO) pro-
The introduction of recombinant human erythropoietin in the duction, which is often compounded by iron deficiency. As renal
late 1980s drastically changed the treatment of anemia in patients failure progresses, the contribution of EPO deficiency to anemia
with CKD. The benefits of anemia treatment in this population increases. The role of decreased renal EPO synthesis in CKD-
reach far beyond the improvement of fatigue and decreased phys- associated anemia is supported by the severe anemia seen in
ical activity to a broad spectrum of physiologic functions. Thus the anephric patients.6 However, the mechanisms impairing renal
presence of anemia should be sought, diagnosed, and treated early EPO production are not well understood. The production capacity
in patients with CKD. The optimal hemoglobin (Hb) targets are still of EPO remains significant even in end stage renal disease (ESRD)
controversial and studies defining these goals are ongoing. The as these patients have been shown to respond with increased
costs of anemia management in the chronic kidney disease popu- EPO synthesis in the setting of an additional hypoxic stimulus.7
lation are considerable and need to be considered along with the This suggests that the decrease in EPO production in CKD is, in part,
risks and benefits. a physiologic response to achieve a chronically reduced Hb
concentration.
* Corresponding author. Tel.: +1 216 844 3163, mobile: +1 216 849 3950; fax: +1 Typically, EPO is produced in the peritubular capillary endothe-
216 844 3328.
lial cells in the kidney relying on a feed-back mechanism measuring
E-mail addresses: christina.lankhorst@uhhospitals.org (C.E. Lankhorst), jaywish@
earthlink.net (J.B. Wish). total oxygen carrying capacity. Hypoxia inducible factor (HIF),
 
Tel.: +1 216 844 5525, mobile: +1 216 965 9058; fax: +1 216 844 5204. which is produced in the kidney and other tissues, is a substance

0268-960X/$ - see front matter Ó 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.blre.2009.09.001
40 C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47

whose spontaneous degradation is inhibited in the presence of been hypothesized that it is the middle molecules (molecular
decreased oxygen delivery due to anemia or hypoxemia. The weight range of 500–2000 daltons) of uremia that contribute to
continued presence of HIF leads to signal transduction and the bone marrow suppression.12
synthesis of EPO. Therefore the usual response is increased EPO pro-
duction in the setting of anemia. The EPO then binds to receptors on
erythroid progenitor cells in the bone marrow, specifically the Evaluation of anemia in patients with CKD
burst-forming units (BFU-E) and colony-forming units (CFU-E).
With EPO present, these erythroid progenitors differentiate into As noted above, the prevalence of anemia in chronic kidney dis-
reticulocytes and red blood cells (RBCs). The absence of EPO leads ease is as high as 10% in patients with CKD as early as stages 1 and
to pre-programmed apoptosis. This is mediated by the Fas antigen. 2. Since the consequences of untreated anemia can be severe, reg-
The decreased red blood cell production and continued loss of blood ular monitoring of the Hb level is needed for optimal care of this
(by programmed red blood cell death) leads to worsening anemia. population. The 2006 National Kidney Foundation (NKF) Kidney
There are other factors in chronic kidney disease which contrib- Disease Outcomes Quality Initiative (KDOQI) clinical practice
ute to anemia. Acute and chronic inflammatory conditions have a guidelines and clinical practice recommendations for anemia in
significant impact on anemia in the CKD population by pro- CKD advocate annual screening for anemia of all patients with
inflammatory cytokines decreasing EPO production and inducing CKD. Further evaluation should be undertaken if anemia is present
apoptosis in colony-forming unit-erythroid cells (CFU-E). The early (Hb less than 13.5 g/dL in men and 12.0 g/dL in women per KDOQI
induction of apoptosis in CFU-E cells stops the process of develop- guidelines13). Note that the Hb thresholds for evaluation in the
ment into RBC. Inflammatory cytokines have also been found to KDOQI guidelines do not separate menstruating versus non-
induce the production of hepcidin, a recently discovered peptide menstruating women as the WHO guidelines do. This needs to be
generated in the liver, which interferes with RBC production by taken into consideration when evaluating women with early stages
decreasing iron availability for incorporation into erythroblasts. of CKD. These recommendations are also based on the assumption
This also impairs the production of RBC. Fig. 1 illustrates the that these patients had normal blood counts in the past. A
above-mentioned interactions. previous diagnosis of anemia secondary to other causes, such as
Red blood cells also have a decreased life span in patients with thalassemias and sickle cell disease, may alter the decision tree
CKD. While the normal life span of an RBC is about 120 days, it for when and how to evaluate a patient’s anemia. For example, a
has been demonstrated that this is shortened to only 60–90 days patient with sickle cell disease whose baseline Hb is about 8 g/dL
in CKD patients. In patients without CKD, the bone marrow has when not in crisis, prior to any known renal disease, would be
significant capacity to increase red blood cell production and to cor- assessed differently from another patient without previous anemia
rect for the shortened life span, but this response is blunted in pa- who presents with CKD stage 3 and Hb of 10 g/dL.
tients with CKD by the relative EPO deficiency. Uremic toxins have The initial evaluation, when considering anemia related to CKD,
been implicated as contributing to apoptosis as the anemia will of- is geared toward eliminating causes other than EPO deficiency.
ten improve after initiation of dialysis. There have been a number of This is because EPO deficiency is a diagnosis of exclusion. As with
prospective and observational studies that have demonstrated any clinical evaluation, a thorough patient history is paramount.
improved Hb levels and decreased dose of erythropoiesis-stimulat- The history should include any prior diagnosis of anemia and the
ing agents (ESAs) with increased adequacy of dialysis.8–11 It has presence of symptoms related to anemia, such as fatigue, exercise

Fig. 1. Erythropoiesis in chronic kidney disease. Ag, antigen; EPO, erythropoietin; Fe, iron; IFN, interferon; IL, interleukin; RBCs, red blood cells; TNF, tumor necrosis factor.
Courtesy of Iain Macdougall, MD. Reprinted with permission from National Kidney Foundation: Primer on Kidney Diseases, 5th Edition. Philadelphia: Saunders Elsevier 2009;
507.
C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47 41

Fig. 2. Flowchart for the evaluation of the chronic kidney disease patient with anemia. CBC, complete blood count; TIBC, total iron-binding capacity; Fe, iron; TSAT, transferrin
saturation. Adapted from Kidney Disease Outcomes Quality Initiative: Am J Kidney Dis 2006;47(Suppl. 3):S1–145.

intolerance, dyspnea, and changes in mentation. The laboratory erythroid marrow to outstrip the ability of the reticuloendothelial
evaluation should include a complete blood count with red blood system to release iron to circulating transferrin. Supply and demand
cell indices, reticulocyte count, transferrin saturation (TSAT), and are the problems in this state, not a total body iron deficiency.16
serum ferritin. A reticulocyte hemoglobin content (CHr), if The hallmark of functional iron deficiency is that it responds to
available, may also be helpful in the diagnosis of iron deficiency. iron supplements with an increase in hemoglobin and/or decrease
Typically the anemia of EPO deficiency is normocytic (normal in ESA requirements. This is despite the normal or elevated ferritin
mean corpuscular volume, or MCV) and normochromic (normal levels. Patients with normal to elevated serum ferritin levels and
mean corpuscular hemoglobin concentration or MCHC). These low TSAT may not respond to intravenous iron therapy,
can also be assessed when evaluating a peripheral smear. Microcy- presumably due to reticuloendothelial blockade. This suggests that
tosis (or low MCV) is suggestive of iron deficiency, but may also be hepcidin has completely prevented the release of iron from macro-
seen in hemoglobinopathies such as thalassemia. Macrocytosis (or phages to circulating transferrin. Hepcidin is an acute phase
high MCV) could be suggestive of vitamin B12 or folate deficiency. reactant protein produced in the liver. It inhibits intestinal iron
If the MCV is high, or it is normal with a pleomorphic RBC smear or absorption along with iron release from stores. In an inflammatory
high red blood cell distribution width (RDW), folate and vitamin state the lower availability of iron has an antimicrobial effect.17
B12 levels should be checked. Since many microorganisms require iron to reproduce, the
In a steady state, serum ferritin levels correlate with the iron increased level of hepcidin, and the resulting decreased available
bound to tissue ferritin in the reticuloendothelial system. Serum iron, limits reproduction. Zaritsky et al. monitored hepcidin levels
ferritin does not carry or bind to iron. Its function is unknown, in patients with chronic kidney disease and performed a multivar-
but is presumed to limit free iron, which is toxic to cells.14 Serum iate analysis comparing these levels with other indicators of
ferritin is also an acute phase reactant; therefore it increases in the anemia. Their findings suggest that increased hepcidin levels may
setting of acute and chronic inflammation, independent of tissue contribute to abnormal iron regulation and erythropoiesis.18 There
iron stores. It is proposed that the elevated serum ferritin of is no uniform testing or recommendations for testing,19 but this
inflammation decreases available iron and this limits the growth may be important for future evaluation of anemia.20
of pathogens that are iron dependent, such as bacteria and fungi.15 The reticulocyte count is an inexpensive and useful test in the
The TSAT is a measure of circulating iron which is available for evaluation of anemia. Reticulocytes are released into the circula-
delivery to the bone marrow. It is calculated by dividing the serum tion about two days prior to maturation into red blood cells. The
iron concentration by the total iron-binding capacity (TIBC) and reticulocyte count assesses the number and percentage of reticulo-
then multiplying by 100. TIBC measures the maximum amount cytes in circulating in the blood. Normally, about 1–2% of red blood
of iron the blood can carry, which indirectly measures transferrin cells in the circulation are reticulocytes. When the bone marrow is
since it is the most dynamic iron carrier. TIBC is used because it stimulated due to anemia, more reticulocytes are released into the
is less expensive than a direct measurement of transferrin. A TSAT blood, increasing their number and percentage. The reticulocyte
of less than 16% in an anemic patient with CKD is consistent with a count helps distinguish red blood cell underproduction from ane-
functional or an absolute iron deficiency. The normal levels for mia caused by red blood cell loss or destruction. Since the typical
ferritin are 30–300 ng/mL in men and 15–250 ng/mL in women. reticulocyte count is represented as a percentage of total circulat-
Ferritin levels below these ranges are consistent with low total ing red blood cells, a corrected reticulocyte count (or reticulocyte
body iron stores and, along with a low TSAT, are diagnostic of abso- index) needs to be calculated to address the effect of anemia on
lute iron deficiency. Functional iron deficiency is seen when the the count. In anemia the reticulocyte percentage may be elevated,
ferritin levels are within or higher than the above-mentioned but the absolute number of reticulocytes is low. With inadequate
ranges and coincide with a low TSAT. This scenario is often observed or deficient EPO production in patients with CKD, the reticulocyte
with the administration of ESAs, which cause iron demand by the count would be expected to be low (absolute reticulocyte count
42 C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47

less than 40,000–50,000 cells per microliter of whole blood). If a reduced cognitive function and mental acuity, impaired quality
patient is found to have an elevated reticulocyte count, it would of life and the need for blood transfusion.24–27 Treatment of anemia
be inconsistent with anemia related to CKD as the bone marrow associated with CKD has been shown to result in improved exercise
responds appropriately to sufficient amounts of EPO. In a patient capacity, energy, and physical mobility.25 Subjective symptoms
with an elevated reticulocyte count, the physician should pursue also improve; such as quality of life, depression and cognition.26
an evaluation for hemolysis or extravascular blood loss. Evidence supporting regression of LVH, fewer cardiac events or
The reticulocyte Hb content (CHr) is a measure of the amount of decreased mortality from ESA treatment is not compelling and will
Hb in the reticulocytes. The CHr is a reasonably good reflection of be discussed in greater detail below.
how much iron was available to the bone marrow for incorporation
into new red blood cells a few days before, which is a more clinically
relevant time frame than the 60–120 days life span of the entire RBC Erythropoiesis-stimulating agents
population. It has been shown that CHr compares favorably with
serum ferritin and TSAT in predicting a response to intravenous Since the introduction of recombinant human erythropoietin in
iron.21 One study demonstrated that a CHr cutoff of 629 pg tended the late 1980s, ESAs have become the mainstay of treatment of
to miss a number of patients who ultimately responded to intrave- anemia in patients with CKD. Treatment with ESAs corrects the
nous iron. It was concluded that a cutoff of 632 pg showed a much underlying pathophysiology of anemia in CKD while reducing the
greater utility, and a majority of the patients who received intrave- need for transfusions and their associated complications.28 The
nous iron therapy on the basis of a CHr 632 pg had an average of first ESA available was epoetin alfa and it was the only therapeutic
23% reduction in their erythropoietin requirements.22 option for over 10 years. Darbepoetin alfa became available in
In the assessment of anemia related to CHr, it would seem that 2001. While both are approved by the US Food and Drug
the measurement of erythropoietin would confirm the diagnosis of Administration (FDA) for the treatment of anemia in CKD and in
EPO deficiency, but routine testing for EPO is not recommended in cancer patients,29–32 only epoetin alfa is also FDA approved for
CKD patients. This is partly because patients who respond to the the treatment of anemia associated with HIV and prior to major
administration of exogenous ESAs often have normal or even ele- surgeries with the risk for increased blood loss.30,31 Epoetin alfa
vated EPO levels, although this may be an inappropriately low level has a shorter half-life than darbepoetin alfa, although the half-life
for the degree of anemia present. Furthermore, an EPO level is an of both drugs is extended when they are administered subcutane-
expensive test and its result will not necessarily drive therapy. ously. Although the bioavailability of subcutaneous epoetin alfa is
Therefore, EPO deficiency is a diagnosis of exclusion in CKD pa- between 20% and 30%, the prolonged half-life following subcutane-
tients with anemia. If the evaluation outlined above is unrevealing ous injection, compared with IV infusion, potentially allows less
but the level of anemia is out of proportion to the stage of renal frequent injections. The dose of epoetin alfa required to achieve
failure, then other causes must be considered in addition to EPO the same hemoglobin response is about 30% lower with subcutane-
deficiency. Fig. 2 is an overview of anemia assessment in patients ous compared with intravenous administration.33 Darbepoetin alfa
with CKD. does not show any significant difference in Hb dose response be-
tween the subcutaneous and intravenous routes of administration.
The package inserts suggest using epoetin alfa three times a week
Clinical manifestations of anemia and darbepoetin alfa once a week, although darbepoetin has also
been approved by the FDA for use every two weeks.29–31 Both ESAs
Anemia has a profound impact on patients with CKD. The most have been shown to have success with extended dosing intervals. A
common symptoms are fatigue (both with activity and at rest), loss retrospective review of the extended dosing of epoetin alfa with
of libido, dizziness, shortness of breath, and decreased sense of intervals ranging form weekly to biweekly to once every three
well being. These symptoms generally occur when the Hb is less weeks was reported by Germain et al., with the most common dos-
than 10 g/dL and become more severe as Hb levels decrease ing interval every two weeks.34 In this study, hemoglobins were
further. Other more dangerous adverse outcomes include cardio- maintained at greater than 11 g/dL in 82% of patients. The PROMPT
vascular disease with left ventricular hypertrophy (LVH) and con- study was a randomized prospective trial in which patients were
gestive heart failure. These may occur when the patient is administered epoetin alfa at intervals of every one, two, three or
otherwise asymptomatic and contribute to the excess cardiovascu- four weeks.35 Hemoglobin levels were maintained above 11 g/dL
lar morbidity and mortality rate observed among patients with in 90% of patients on every two-week dosing intervals and 75%
CKD. In patients who already have coronary artery disease, in groups with every three-week and every four-week dosing
decreased myocardial oxygen delivery may lead to worsening intervals. Similar studies have been performed with darbepoetin
anginal symptoms. Decreased peripheral oxygen delivery due to alfa. Jadoul et al. examined hemodialysis patients on every two-
anemia also leads to peripheral vasodilation, increased sympa- week dosing and when extended to every four-week dosing, 83%
thetic nervous system activity, increased heart rate and stroke of the patients maintained hemoglobin levels above target.36 This
volume and, ultimately, LVH. It has been demonstrated that LVH was a small study, but a later, larger study confirmed the efficacy
strongly correlates with adverse outcomes (hospitalization and of the extended dosing in CKD patients.37
mortality) in patients with CKD. A 0.5 g/dL decrease in hemoglobin There are ESAs that are currently in use in countries other than
below normal correlates with a 32% increase in LVH risk, whereas a the United States. Epoetin beta is similar to epoetin alfa in its
5 mmHg increase in systolic blood pressure correlates with only an efficacy, safety, pharmacokinetics and pharmacodynamics.
11% increase in LVH risk.23 Continuous erythropoietin receptor activator (CERA) was created
Anemia has also been associated with a decline in renal by integrating a large polymer chain into the erythropoietin mole-
function in some patient groups. Decreased tissue oxygen delivery cule. It has been shown to have an extended half-life of about 130 h
caused by anemia stimulates the renin-angiotensin-aldosterone which is independent of intravenous or subcutaneous administra-
system and contributes to renal vasoconstriction. These factors tion. There are other treatments for the anemia of CKD currently in
can further exacerbate proteinuria, which may lead to worsening clinical development. Hematide is an erythropoietin-mimetic
renal failure. In patients with type 2 diabetes, anemia has been peptide, the amino acid sequence of which is completely unrelated
shown to be an independent risk factor to the progression of renal to native or recombinant erythropoietin but shares the same
disease. Other complications associated with anemia consist of functional and biologic properties.38 Hypoxia inducible factor
C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47 43

(HIF) stabilizers are competitive inhibitors of HIF prolylhydroxy- 15.0–35.0 mL/min (by the Cockcroft–Gault formula) and a blood
lases39,40 and asparagyl-hydroxylase, enzymes involved in the pressure less than 170/95. Patients with advanced cardiovascular
metabolism of HIF and its transcriptional activity. Therefore, HIF disease, non-renal cause of anemia, previous ESA treatment, and
stabilizers cause an increase in endogenous erythropoietin produc- anticipated need for renal replacement therapy within six months
tion and are orally active. were excluded. This was an open-label, randomized two-group
Clinical trials of ESAs in patients with CKD have documented study with parallel group design. Patients with moderate anemia
improved quality of life and functional status as well as relief of (Hb, 11.0–12.5 g/dL) were randomized to receive either early or late
symptoms associated with anemia.41 ESAs also reduce the need treatment with epoetin beta, an ESA primarily used overseas, espe-
for transfusions, thereby decreasing the risks of immunologic sen- cially in Europe. The immediate treatment group had a hemoglobin
sitization, infections and iron overload. Although a survival advan- goal of 13–15 g/dL and the delayed treatment group had a lower
tage among CKD patients treated with ESAs has not been target of 10.5–11.5 g/dL. For the second group, treatment was de-
demonstrated in randomized clinical trials, a two-year historical layed until the hemoglobin dropped below 10.5 g/dL. This trial’s
cohort study showed that those hemodialysis patients treated with primary endpoint was a composite of eight cardiovascular events.
ESAs lived longer than those not treated.42 Observational studies Secondary endpoints included left ventricular mass index, quality
have suggested increased survival among hemodialysis patients of life scores, and the progression of chronic kidney disease. There
with Hb levels greater than 12 g/dL. But randomized control trials was no statistically significant difference in the risk of cardiovascu-
with higher target hemoglobins have not shown a reduction of lar events between the two groups (58 versus 47 in high and low
death or cardiovascular events. The largest of these trials in groups retrospectively, hazard ratio of 0.78, 95% confidence interval
hemodialysis patients demonstrated higher mortality rates in 0.53–1.14, with P value of 0.20), although the trend was toward in-
patients with targeted hemoglobin greater than 14 than those creased events in high-target hemoglobin group. Left ventricular
patients with targeted hemoglobins of 10.43 hypertrophy remained stable in both groups. Quality of life was
There is debate about the effect of ESAs on the progression of the only endpoint that was significantly better in the high-target
renal failure. Roth et al. conducted a 48-week randomized open- treatment arm. All other events were not statistically significant
label trial comparing patients with chronic kidney disease who re- in their differences.
ceived epoetin alfa versus those who did not receive any ESA. CKD The Correction of Hemoglobin and Outcomes in Renal Insuffi-
was defined by serum creatinine of 3–8 mg/dL and serial GFRs ciency (CHOIR) trial was an open-label, prospective, randomized
were measured using 125I-iothalamate clearance. The target trail comparing the effect of correcting hemoglobin to a target of
hematocrit was 35% (equal to hemoglobin of 11.7 g/dL) for those 13.5 g/dL versus a target of 11.3 g/dL.56 This trial enrolled 1432
patients receiving epoetin alfa. The study demonstrated no differ- patients at 130 sites within the US, over 18 years of age, who
ence in the mean change of glomerular filtration rate from initial had hemoglobin less than 11.0 g/dL and CKD defined by an esti-
to the last available calculation.44 The study was not powered to mated glomerular filtration rate of 15–50 ml/min by Modification
assess if different starting GFRs made a difference in the outcome. of Diet in Renal Disease (MDRD) formula. Patients were excluded
A randomized controlled trial by Gouva et al. showed treatment if they had uncontrolled hypertension, active gastrointestinal
with epoetin alfa to a target hemoglobin of greater than 13 g/dL bleeding, iron overload, frequent transfusions in the previous six
in patients with chronic kidney disease slowed the progression of months, active cancer, angina, or previous treatment with epoetin
renal disease. The need for renal replacement therapy was actually alfa. Two randomly assigned groups of patients received epoetin
reduced by 60%.45 More recent data suggest that any benefit on the alfa. Group 1 had a target hemoglobin of 13–13.5 g/dL, which
progression of renal disease from ESA therapy depends on the was later amended to 13.5 g/dL. Group 2 had a target hemoglobin
hemoglobin target. Higher hemoglobin targets have been shown of 10.5–11.0 g/dL, which was later changed to 11.3 g/dL. Epoetin
to increase the rate of initiation of dialysis.46 alfa was initially administered weekly, but was allowed to be de-
Renal anemia causes increased sympathetic nerve activity and creased to every other week if the hemoglobin level was stable.
is linked to cardiovascular complications, such as increased blood The patients in the higher hemoglobin group had a significantly
pressure and left ventricular hypertrophy. Treatment with ESAs higher incidence of the composite end-point (death, myocardial
improves heart failure symptoms and leads to regression of left infarction, hospitalization for congestive heart failure and stroke),
ventricular hypertrophy.24,47,48 This is seen with lower hemoglobin congestive heart failure, death, and hospitalization (cardiovascular
targets (10–11 g/dL). Risks associated with higher hemoglobin tar- and all-cause). There was no difference between the groups in the
gets (greater than 13 g/dL) will be discussed later in this article. rate of stroke, myocardial infarction, or renal replacement therapy
Several studies have suggested that the administration of ESAs or in quality of life.
to anemic CKD patients decreases hospitalizations and thereby re- Treatment with ESAs has been reported to increase the risk of
duces the overall cost of care.49–51 Furthermore, there are data to adverse events in other studies. Patients with congestive heart
suggest that ESA-treated anemic CKD patients are more productive failure or ischemic heart disease treated with ESAs had an in-
workers and consume fewer resources.52,53 creased risk of mortality and of non-fatal MI, vascular access
Despite the numerous physiologic benefits of ESA therapy thrombosis, and other thrombotic events when epoetin alfa was
among patients with CKD, there are risks. Randomized controlled dosed to target hematocrit of 42%.43 Following the publication of
trials have shown increased rates of cardiovascular complications such studies, including CHOIR and CREATE, along with the studies
in hemodialysis patients with high (13–15 g/dL) versus low (10– on cancer patients with higher targeted hemoglobin and worse
11.5 g/dL) hemoglobin targets.43,54,55 The Cardiovascular Risk outcomes, the FDA issued a ‘‘black box” warning. The warning
Reduction of Early Anemia Treatment with Epoetin Beta (CREATE) advises physicians that patients experienced greater risks for death
trial is a prominent randomized clinical trial addressing Hb targets. and serious cardiovascular events when administered ESAs to tar-
In this study 603 patients were enrolled from 94 centers in 22 coun- get higher versus lower Hb levels (12.5 versus 11.3 g/dL; 14 versus
tries and were randomly assigned to two treatment groups to test 10 g/dL). The FDA recommends individualized dosing to achieve
the hypothesis that complete correction of anemia in patients with and maintain Hb levels within the range of 10–12 g/dL. This
stages three to four CKD improves cardiovascular outcomes as FDA recommendation is at odds with the US National Kidney
compared with partial correction of anemia.46 The eligible patients Foundation’s Kidney Disease Outcomes Quality Initiative
were over 18 years of age with mild to moderate chronic anemia re- (NKF/KDOQI) anemia guidelines which recommend a target Hb of
lated to their chronic kidney disease, a glomerular filtration rate of 11–12 g/dL, not to exceed 13 g/dL in anemic CKD patients treated
44 C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47

with ESAs. The bottom line is that the target Hb level should be present in hemodialysis patients due to: bleeding when needles
individualized to the patient’s symptoms, co-morbidities, and are removed from the vascular access, blood infiltration of the vas-
ESA responsiveness within the range specified by the FDA, NKF/ cular access, vascular access procedures, frequent blood testing,
KDOQI or other national evidenced based guideline. and clotting or general blood loss in the extracorporeal circuit.
In its own retrospective analysis of the normal hematocrit trial The iron deficiency observed in patients with CKD not yet on
in hemodialysis patients43 and the CHOIR study of CKD patients56 hemodialysis, and those on peritoneal dialysis, is often due to de-
the FDA noted that the rate of rise of Hb in ESA-treated patients creased oral iron intake from the prescribed dietary protein restric-
was an independent risk factor for adverse outcomes irrespective tion or decreased appetite for red meat. Iron deficiency often
of the ESA dose and target Hb level. As a result, the FDA cautions develops, if it is not present initially, while a patient is receiving
against a Hb rise of >2 g/dL in a four-week period and recommends ESA therapy. This is due to depletion of the existing iron stores with
a 25% ESA dose reduction in such patients. The FDA also recom- the stimulation of new RBC production. Regular monitoring of iron
mends a 25% ESA dose reduction in patients trending toward status is essential during the initiation of ESA therapy with monthly
exceeding the Hb target range ceiling of 12 g/dL. assessment of serum ferritin and TSAT while the Hb level is rising
Although the intention to treat analysis of the CHOIR study indi- and then every three months after the Hb level has stabilized. The
cates a higher risk for the composite outcomes of death and cardio- target levels for serum ferritin and TSAT are higher in ESA-treated
vascular events among patients randomized to the higher Hb target, patients than in the general population because of the development
a secondary analysis of the CHOIR study by Sczcech et al. reveals of functional iron deficiency, in which the demand for iron by the
that when the analysis is adjusted for ESA dose and patients not ESA-stimulated bone marrow exceeds the rate at which normal
achieving their Hb target, the difference in outcomes between the reticuloendothelial iron stores can release it. The NKF/KDOQI ane-
high and low Hb targets becomes non-significant and the use of a mia guidelines recommend a target serum ferritin of greater than
high dose of ESA becomes the major predictor of adverse outcomes 200 ng/mL in hemodialysis patients and greater than 100 ng/mL
in both target groups.57 The analysis cannot distinguish whether in patients with CKD not receiving hemodialysis and those receiv-
this effect is due to toxicity of high doses of ESA per se and/or ing peritoneal dialysis patients when taking ESAs. The target TSAT
whether patient factors resulting in ESA resistance (e.g. inflamma- level is greater than 20%.13 While this is the recommended target,
tion) account for the higher incidence of adverse events. Nonethe- the lowest all-cause and cardiovascular death risks where observed
less, it seems prudent to avoid high doses of ESAs (defined as in patients with TSATs in the 30–50% range.60
>20,000 units/week epoetin alfa in the CHOIR study) in ESA-resis- Iron can be administered orally or intravenously. Oral iron is
tant patients. Occasional excursions of Hb to the 12–13 g/dL range usually sufficient in patients with CKD not yet on dialysis or peri-
in patients receiving modest doses of ESAs are likely not to be harm- toneal patients who do not have the continual blood loss that is
ful if the ESA dose is down-titrated accordingly.58 typical in hemodialysis patients. Yet even in non-hemodialysis
Another reported adverse effect of ESA therapy is hypertension. patients, oral iron may not be effective due to gastrointestinal side
It is caused by multiple mechanisms including: expansion of blood effects, poor compliance or the severity of iron deficiency. Oral iron
volume, reversal of hypoxic vasodilation and increased blood vis- salts such as ferrous sulfate, gluconate and fumarate should be
cosity. A direct vasoconstrictive effect has also been proposed.59 administered one hour before or 2 h after meals to result in the
The hypertension related to treatment is more common in patients greatest absorption. Oral ferrous iron salts must be oxidized to
receiving hemodialysis, associated with a rapid rise in Hb level and the ferric form by stomach acid to allow absorption by the small
usually is seen within the first 90 days of treatment.29,30 Rarely, intestine. This step can be impaired by food, antacids, histamine-
seizures and hypertensive encephalopathy have occurred. Antihy- 2 blockers or proton pump inhibitors. It has been shown that the
pertensive treatment may need to be adjusted, so close monitoring minimal effective dose is 200 mg of elemental iron daily. Each
of the patient’s blood pressure is recommended. 325 mg tablet of ferrous sulfate contains 65 mg of elemental iron;
A rare and serious complication associated with ESAs is pure red therefore three tablets must be administered over the course of
blood cell aplasia (PRCA). PRCA is a result of the production of anti- each day. As a result of inflammation or impaired iron mobilization
erythropoietin antibodies induced by the administration of exoge- from stores, serum ferritin is often elevated in patients with
nous ESAs. This was primarily associated with the subcutaneous chronic kidney disease. This further reduces bioavailability of oral
use of epoetin beta sold outside of the United States. It was thought iron to only 1–2%, so even the most compliant patients may not be
to be secondary to the additive Tween 80 used to stabilize the sub- able to replete their iron stores with oral iron.
stance. Tween 80 was never used in the United States and PRCA In CKD patients with mild to moderate iron deficiency, a three-
remains rare in the US. PRCA should be suspected if a patient has month trial of oral iron is recommended to assess the effectiveness
a sudden weekly drop in hemoglobin of about 1 g/dL or a weekly of that route of administration. For patients with severe iron
transfusion requirement and a low reticulocyte count, despite a deficiency and all iron deficient hemodialysis patients, intravenous
high dose of an ESA for several months. White blood cell and plate- iron supplements are recommended. There are four preparations of
let counts remain unaffected, differentiating this condition from intravenous iron currently available in the US: iron dextran, iron
aplastic anemia. Iron deficiency also should be considered; this will sucrose, iron gluconate and ferumoxytol. The least expensive,
be discussed later in the article. PRCA is definitively diagnosed and the one approved by the FDA to be given in doses as large as
when anti-erythropoietin antibodies are demonstrated in the 1000 mg, is iron dextran, but it has been associated with fatal ana-
blood, or an examination of the bone marrow reveals normal cellu- phylactic reactions.61 As a result, FDA has issued a ‘‘black box”
larity and less than 4% erythroblasts. PRCA is treated by discon- warning recommending that patients undergo a 25 mg test dose
tinuing the ESA and administering immunosuppressive therapy. the first time the drug is given. If a patient does not have an
Patients generally respond after several months of treatment and adverse reaction to this dose, he/she is less likely to have an ana-
usually do not relapse after immunosuppressants are discontinued. phylactic reaction to the therapeutic dose of iron dextran, but fatal
anaphylactic reactions still occur with an uneventful test dose. This
risk notwithstanding, the use of a single iron dextran dose of 500–
Iron therapy 1000 mg is appealing in non-hemodialysis patients who need to
travel long distances in order to receive their treatment. Further-
Many anemic patients with CKD and inadequate EPO production more, fewer IV iron doses also means fewer intravenous line place-
have coexisting iron deficiency. Iron deficiency almost always is ments and therefore the preservation of the veins needed for the
C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47 45

creation of permanent dialysis access for future hemodialysis. Iron evidence has been lacking its efficacy in patients with non-dialysis
sucrose and iron gluconate do not require a test dose because they CKD. There has been some benefit in reducing ESA doses in a sub-
have not been associated with fatal anaphylactic reactions. These group of hemodialysis patients, but it is not recommended for all
two preparations are FDA approved to be given in doses up to patients.74,75 Prior to the advent of ESA therapy, androgens were
250–300 mg, meaning non-hemodialysis patients will need more used with some success in treating anemia in hemodialysis
trips to the clinic or hospital for infusions and more placements patients, but with significant side effects. However evidence is lack-
of intravenous catheters to receive a total repletion dose of ing for any benefit of androgen use in patients receiving adequate
1000 mg. These preparations have some non-fatal adverse reac- ESA therapy and long-term androgen use has the potential for sig-
tions such as nausea, vomiting, and hypotension. Generally, with nificant toxicity.
slower infusions of these medications, there is a decreased risk of Even with sufficient dosing of ESAs and iron therapy, patients
adverse side effects. Ferumoxytol, which was approved by the may still require red blood cell transfusions due to ESA resistance
FDA in June 2009, is an iron dextran nanoparticle which can be or acute blood loss. In patients who are candidates for organ trans-
administered in rapid injection of up to 510 mg over 17 s. In phase plantation, this is the option of last resort because sensitization
3 studies in non-dialysis patients with CKD and iron deficiency from transfusion can decrease the chance of a successful trans-
anemia, it was shown to have no more side effects than oral iron plant. There also still exists the small risk of bloodborne infections
and to be more effective in raising the Hb level than an ESA with such as hepatitis and human immunodeficiency virus. Unlike the
oral iron.62 guidelines for ESA therapy, there is no recommended Hb threshold
Despite the benefits of iron supplementation, concerns have at which a patient should be transfused. This is a clinical decision
been raised about potential toxicity of intravenous iron including based primarily on a patient’s symptoms, but must also take into
cellular and vascular damage resulting from oxidative stress and consideration acuity of Hb decline, concomitant diseases such as
impaired white blood cell function based on in vitro studies. In pa- coronary artery disease, and the patient’s candidacy for future
tients with CKD receiving intravenous iron sucrose, there has been organ transplantation.
evidence of increased urinary excretion of markers of tubular
injury, but no increase in albuminuria. This has not proved to be
Conclusions
clinically significant in other studies. There was no increase in
hospitalizations or mortality seen in observational studies in
Anemia is a common and important complication among
hemodialysis patients receiving less than 400 mg on average of
patients with CKD and health care professionals should be familiar
intravenous iron per month.60,63,64 Intravenous iron was not shown
with the best practices for its screening, evaluation and treatment.
to be a risk factor for bacteremia in hemodialysis patients in a
Untreated anemia places patients at risk for cardiovascular events,
multivariate analysis.65 Serial liver biopsies were done in patients
more rapid progression of chronic kidney disease and significantly
with hemochromatosis and showed no significant organ injury
decreased quality of life. The cause of anemia is multifactorial in
when serum ferritin level was less than 2000 ng/mL.66 The NKF/
patients with CKD, but inadequate production of EPO by the dis-
KDOQI anemia guidelines recommend weighing ESA responsive-
eased kidneys is the common denominator. The anemia generally
ness, hemoglobin concentration, TSAT level, and the patient’s
becomes more severe as a patient’s renal function declines. Once
clinical status when considering giving iron to a patient with a
other treatable causes of anemia are evaluated and addressed, pa-
ferritin greater than 500 ng/mL.
tients who remain anemic will likely need to be treated with an
ESA. According to the NKF/KDOQI guidelines, ESA therapy should
be initiated when the patient’s Hb drops below 10 g/dL, with target
ESA resistance
Hb level for treatment being 11–12 g/dL. It is also recommended
not to treat patients with ESAs to Hb above 13 g/dL as studies have
ESA resistance is defined as a failure to achieve a target hemo-
shown evidence of adverse events in patients with CKD maintained
globin greater than 11.0 g/dL in the setting of an epoetin alfa dose
at normal Hb levels without significant benefit. Both prior to and
of more than 500 units/kg per week or the equivalent of another
during ESA treatment, patients with CKD often require iron supple-
ESA. The causes can include iron deficiency, acute and chronic
mentation. This is frequently given orally in patients with CKD not
inflammatory conditions, severe hyperparathyroidism, aluminum
on hemodialysis and in patients on peritoneal dialysis. Hemodialy-
toxicity, folate deficiency and PRCA. Iron deficiency is the most
sis patients and those unable to respond oral iron require intra-
common cause of ESA resistance, but it is followed closely by
venous iron therapy. Intravenous iron therapy is associated with
inflammation and infection. The source of inflammation or infec-
adverse effects, including potentially fatal anaphylactic reactions
tion may be difficult to elucidate, but it is often associated with
to iron dextran, but it also has significant benefits by increasing
high levels of acute phase reactants, such as ferritin, erythrocyte
Hb levels and decreasing ESA requirements. Despite adequate
sedimentation rate (ESR), and C-reactive protein (CRP). The use
treatment with iron and ESAs, patients may fail to increase Hb
of dialysis catheters for vascular access has been shown to lower
and require transfusion. As more is learned about anemia in
Hb levels and increase ESA requirements. This may reflect an
patients with CKD, treatment methods and goals may continue to
inflammatory state induced by the presence of a catheter and its
evolve, but there is no question that current treatments have
biofilm without evidence of an active infection demonstrated by
significantly improved the quality of life and decreased transfu-
positive cultures.67
sions and their complications in this vulnerable population.
The resistance to ESA therapy in the inflammatory state has led
to exploration of adjuvant treatment with vitamin C. Studies have
not shown significant benefit in patients with CKD not yet on dial- Practice points
ysis treated with vitamin C, although there have been many smaller
studies showing decreased ESA requirements with concomitant use  Anemia is a common complication in patients with chronic
of vitamin C in patients undergoing hemodialysis.68–73 Vitamin C kidney disease (CKD) and is associated with adverse out-
therapy may produce some benefit in patients with CKD or under- comes including poor quality of life, cardiovascular disease
going peritoneal dialysis receiving oral iron, as it may promote and progression of renal failure.
intestinal iron absorption. There have been several studies examin-  Evaluation of CKD patients with anemia is straightforward
ing the use of L-carnitine as an adjuvant therapy for anemia, but the and is directed at ruling out iron deficiency and, if clinically
46 C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47

indicated, other etiologies such as hemoglobinopathies, 20. Ganz T, Olbina G, Girelli D, Nemeth W, Westerman M. Immunoassay for human
serum hepcidin. Blood 2008;112:4292–7.
blood loss and vitamin deficiencies.
21. Mittman N, Sreedhara R, Mushnick R, Chattopadhyay J, Zelmanovic D, Vaseghi
 Most anemic patients with CKD will have erythropoietin defi- M, et al. Reticulocyte hemoglobin content predicts functional iron deficiency in
ciency, which is a diagnosis of exclusion. hemodialysis patients receiving rHuEPO. Am J Kidney Dis 1997;30:912–22.
 Treatment of anemia in patients with CKD includes erythro- 22. Fishbane S, Shapiro W, Dutka P, Valenzuela OF, Faubert OF. A randomized trial
of iron deficiency testing strategies in hemodialysis patients. Kidney Int
poiesis-stimulating agents (ESAs), but attention must be paid 2001;60:2406–11.
to repletion and maintenance of iron stores. 23. Levin A, Thompson C, Ethier J, Carlisle E, Tobe S, Mendelssohn D, et al. Left
 The target hemoglobin level for anemic CKD patients receiv- ventricular mass index increase in early renal disease: impact of decline in
hemoglobin. Am J Kidney Dis 1999;34(1):125–34.
ing ESA therapy is 10–12 g/dL per the US Food and Drug 24. Silverberg DS, Wexler D, Blum M, Tchebiner JZ, Sheps D, Keren G, et al. The
Administration. effect of correction of anaemia in diabetics and non-diabetics with severe
 Maintenance of hemoglobin levels above 13 g/dL with ESA resistant congestive heart failure and chronic renal failure by subcutaneous
erythropoietin and intravenous iron. Nephrol Dial Transplant 2003;18:141–6.
therapy has been associated with and increased frequency 25. Painter P, Moore G, Carlson L, Paul S, Myll J, Phillips W, et al. Effects of exercise
of adverse cardiovascular events in randomized clinical trials. training plus normalization of hematocrit on exercise capacity and health-
 When used according to practice guidelines, ESA therapy has related quality of life. Am J Kidney Dis 2002;39:257–65.
26. Valderrabano F, Jofre R, Lopez-Gomez JM. Quality of life in end-stage renal
been associated with improved quality of life, reduction in disease patients. Am J Kidney Dis 2001;38:443–64.
transfusion requirements and few significant adverse effects. 27. Pisioni RL, Bragg-Gresham JL, Young EW, Akizawa T, Asano Y, Locatelli F, et al.
Anemia management and outcomes from 12 countries in the Dialysis Outcomes
and Practice Patterns Study (DOPPS). Am J Kidney Dis 2004;44:94–111.
Conflicts of interest 28. Popovsky MA, Ransil BJ. Long-term impact of recombinant human erythropoietin
on transfusion support in patients with chronic renal failure. Immunohematology
1996;12(1):1–3.
CL reports no conflicts of interest. JW has served on advisory 29. Aranesp (darbepoetin alfa) for injection [prescribing information]. Thousand Oaks,
boards for Amgen, Centocor Ortho Biotech, Watson, Affymax CA: Amgen Inc. <www.aranesp.com/professionalprescribing_information.jsp>;
2007 [accessed 8.07.09].
and AMAG, in addition to serving on speakers bureaus for Amgen,
30. Procrit (epoetin alfa) for injecton [prescribing information]. Thousand Oaks, CA:
Watson, and AMAG. Amgen Inc. Distributed by Ortho Biotech Products, LP: Raritan, NJ. <www.
procrit.com/ppi.pdf>; 2007 [accessed 8.07.09].
31. Epogen (epoetin alfa) for injection [prescribing information]. Thousand Oaks, CA:
References Amgen Inc, 2007. <www.epogen.com/pdf/epogen_pi.pdf>; 2009 [accessed 8.07.09].
32. Nissenson AR, Swan SK, Lindberg JS, et al. Randomized, controlled trial of
darbepoetin alfa for the treatment of anemia in hemodialysis patients. Am J
1. National Kidney Foundation. Chronic kidney disease. <www.kidney.org/
Kidney Dis 2002;40(1):110–8.
kidneydisease/ckd/index.cfm>; 2009 [accessed 7.07.09].
33. Kaufman JS, Reda DJ, Fye CL, et al. Subcutaneous compared with intravenous
2. Weiner DE, Tighiouart H, Vlagopoulos PT, et al. Effects of anemia and left
epoetin in patients receiving hemodialysis. Department of veterans affairs
ventricular hypertrophy on cardiovascular disease in patients with chronic
cooperative study group on erythropoietin in hemodialysis patients. N Engl J
kidney disease. J Am Soc Nephrol 2005;16:1803–10.
Med 1998;339:578–83.
3. World Health Organization. Database of anemia. Worldwide prevalence of
34. Germain M, Ram CV, Bhaduri S, Tang KL, Klausner M, Curzi M. Extended epoetin
anaemia 1993–2005. <www.who.int/vmnis>; 2009 [accessed 7.07.09].
alfa dosing in chronic kidney disease patients: a retrospective review. Nephrol
4. Hsu CY, McCulloch CE, Curhan GC. Epidemiology of anemia associated with
Dial Transplant 2005;20(10):2146–52.
chronic renal insufficiency among adults in the United States: results from the
35. Provenzano R, Bhaduri S, Singh AKPROMPT Study Group. Extended epoetin alfa
third national health and nutrition examination survey. J Am Soc Nephrol
dosing as maintenance treatment for the anemia of chronic kidney disease: the
2002;13:504–10.
PROMPT study. Clin Nephrol 2005;64(2):113–23.
5. Astor BC, Muntner P, Levin A, Eustace JA, Coresh J. Association of kidney
36. Jadoul M, Vanrenterghem Y, Foret M, Walker R, Gray SJ. Darbepoetin alfa
function with anemia: the third national health and nutrition examination
administered once monthly maintains haemoglobin levels in stable dialysis
survey (1988–1994). Arch Intern Med 2002;162:1401–8.
patients. Nephrol Dial Transplant 2004;19(4):898–903.
6. Fehally J, Floege J, Johnson R. Comprehensive clinical nephrology. Philadelphia:
37. Ling B, Walczyk M, Agarwal A, Carroll W, Liu W, Brenner R. Darbepoetin alfa
Mosby Elsevier; 2003, p. 853–60.
administered once monthly maintains hemoglobin concentrations in patients
7. Eckardt KU. Erythropoietin: oxygen-dependent control of erythropoiesis and its
with chronic kidney disease. Clin Nephrol 2005;63(5):327–34.
failure in renal disease. Nephron 1994;67:7–23.
38. Macdougall IC, Eckardt KU. Novel strategies for stimulating erythropoiesis and
8. Eschbach JW, Adamson JW, Cook JD. Disorders of red cell production in uremia.
potential new treatments for anaemia. Lancet 2006;368:947–53.
Arch Intern Med 1970;126:812–5.
39. Macdougall IC, Padhi D, Jang G. Pharmacology of darbepoetin alfa. Nephrol Dial
9. Koch KM, Patyna D, Shaldon S, Werner E. Anemia of the regular hemodialysis
Transplant 2007;22(Suppl. 4):iv2–9.
patient and its treatment. Nephron 1974;12:405–19.
40. Bunn HF. New agents that stimulate erythropoiesis. Blood 2007;109:868–73.
10. Geerlings W, Morris RW, Brunner FP, et al. Factors influencing anaemia in
41. Kimel M, Leidy NK, Mannix S, Dixon J. Does epoetin alfa improve health-related
dialysis patients. A special survey by the EDTA-ERA registry. Nephrol Dial
quality of life in chronically ill patients with anemia? Summary of trials of
Transplant 1993;8:585–9.
cancer, HIV/AIDS, and chronic kidney disease. Value Health 2008;11(1):57–75.
11. Katzarski KS, Charra B, Luik AJ, et al. Fluid state and blood pressure control in
42. Regidor DL, Kopple JD, Kovesdy CP, et al. Associations between changes in
patients treated with long and short dialysis. Nephrol Dial Transplant
hemoglobin and administered erythropoiesis-stimulating agent and survival in
1999;14:369–75.
hemodialysis patients. J Am Soc Nephrol 2006;17(4):1181–91.
12. Locatelli F, Del Vecchio L. Dialysis adequacy and response to erythropoietic
43. Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with
agents: what is the evidence base? Nephrol Dial Transplant 2003;18(Suppl. 8):
low hematocrit values in patients with cardiac disease who are receiving
viii29–35.
hemodialysis and epoetin. N Engl J Med 1998;339(9):584–90.
13. National Kidney Foundation. KDOQI clinical practice guidelines and clinical
44. Roth D, Smith RD, Schulman G, et al. Effects of recombinant human
practice recommendations for anemia in chronic kidney disease. <www.
erythropoietin on renal function in chronic renal failure predialysis patients.
kidney.org/PROFESSIONALS/kdoqi/guidelines_anemia/guide2.htm#cpr- 11>; 2009
Am J Kidney Dis 1994;24(5):777–84.
[accessed 7.07.09].
45. Gouva C, Nikolopoulos P, Ioannidis JP, Siamopoulos KC. Treating anemia early
14. Orino K, Lehman L, Tsuji Y, Ayaki H, Torti SV, Torti FM. Ferritin and the response
in renal failure patients slows the decline of renal function: a randomized
to oxidative stress. Biochem J 2001;357(1):241–7.
controlled trial. Kidney Int 2004;66(2):753–60.
15. Parkkinen J, von Bonsdorff L, Peltonen S, Gronhagen-Riska C, Rosenlof K.
46. Drueke TB, Locatelli F, Clyne N, et al. CREATE Investigators. Normalization of
Catalytically active iron and bacterial growth in serum of haemodialysis
hemoglobin level in patients with chronic kidney disease and anemia. N Engl J
patients after i.v. iron-saccharate administration. Nephrol Dial Transplant
Med 2006;355(20):2071–84.
2000;15:1827–34.
47. Frank H, Heusser K, Hoffken B, Huber P, Schmieder RE, Schobel HP. Effect of
16. Kovesdy CP. Iron and clinical outcomes in dialysis and non-dialysis dependent
erythropoietin on cardiovascular prognosis parameters in hemodialysis
chronic kidney disease patients. Adv Chronic Kidney Dis 2009;16(2):
patients. Kidney Int 2004;66(2):832–40.
109–16.
48. Ayus JC, Go AS, Valderrabano F, et al. Spanish group for the study of the anemia
17. Paradkar P, De Domenico I, Durchfort N, Zohn I, Kaplan J, McVey Ward D. Iron
and left ventricular hypertrophy in pre-dialysis patients. Effects of
depletion limits intracellular growth in macrophages. Blood 2008;12(3):866–74.
erythropoietin on left ventricular hypertrophy in adults with severe chronic
18. Zaritsky J, Young B, Wang H, Westerman M, Olbina G, Nemeth E, et al. A
renal failure and hemoglobin <10 g/dL. Kidney Int 2005;68(2):788–95.
potential novel biomarker for iron status in chronic kidney disease. Clin J Am
49. Wish J, Schulman K, Law A, Nassar G. Healthcare expenditure and resource
Soc Nephrol 2009;4(6):1051–6.
utilization in patients with anaemia and chronic kidney disease: a retrospective
19. Malyszko J. Hepcidin assays: ironing out some details. Clin J Am Soc Nephrol
claims database analysis. Kidney Blood Press Res 2009;32(2):110–8.
2009;4(6):1015–6.
C.E. Lankhorst, J.B. Wish / Blood Reviews 24 (2010) 39–47 47

50. Maddux FW, Shetty S, del Aguila MA, Nelson MA, Murray BM. Effect of 62. Spinowitz BS, Kausz AT, Baptista J, Noble SD, Sothinathan R, Bernardo MV, et al.
erythropoiesis-stimulating agents on healthcare utilization, costs, and Ferumoxytol for treating iron deficiency anemia in CKD. J Am Soc Nephrol
outcomes in chronic kidney disease. Ann Pharmacother 2007;41(11):1761–9. 2008;19:1599–605.
51. London R, Solis A, Goldberg GA, Wade S, Ryu S. Health care resource utilization 63. Horl WH. Adjunctive therapy in anaemia management. Nephrol Dial Transplant
and the impact of anemia management in patients with chronic kidney disease. 2002;17(Suppl. 5):56–9.
Am J Kidney Dis 2002;40(3):539–48. 64. Brewster UC. Intravenous iron therapy in end-stage renal disease. Semin Dial
52. Papatheofanis F, Bookhart BK, Muser E, Piech CT. An examination of 2006;19(4):285–90.
productivity and resource utilization associated with epoetin alfa treatment 65. Hoen B, Paul-Dauphin A, Kessler M. Intravenous iron administration does not
in employees with predialysis chronic kidney disease. J Occup Environ Med significantly increase the risk of bacteremia in chronic hemodialysis patients.
2008;50(5):584–9. Clin Nephrol 2002;57(6):457–61.
53. Moyneur E, Bookhart BK, Mody SH, Fournier AA, Mallett D, Duh MS. The 66. Admas PC, Deugnier Y, Moirand R, Brissot P. The relationship between iron
economic impact of pre-dialysis epoetin alpha on health care and work loss overload, clinical symptoms, and age in 410 patients with genetic
costs in chronic kidney disease: an employer’s perspective. Dis Manag hemochromatosis. Hepatology 1997;25:162–6.
2008;11(1):49–58. 67. Adamson J. Hyporesponsiveness to erythropoiesis stimulating agents in chronic
54. Furuland H, Linde T, Ahlmen J, Christensson A, Strombom U, Danielson BG. A kidney disease: the many faces of inflammation. Adv Chronic Kidney Dis
randomized controlled trial of haemoglobin normalization with epoetin alfa in 2009;16(2):76–82.
pre-dialysis and dialysis patients. Nephrol Dial Transplant 2003;18(2):353–61. 68. Kamgar M, Zaldivar F, Vaziri ND, Pahl MV. Antioxidant therapy does not
55. Parfrey PS, Foley RN, Wittreich BH, Sullivan DJ, Zagari MJ, Frei D. Double-blind ameliorate oxidative stress and inflammation in patients with end-stage renal
comparison of full and partial anemia correction in incident hemodialysis disease. J Natl Med Assoc 2009;101(4):336–44.
patients without symptomatic heart disease. J Am Soc Nephrol 69. Sirover WD, Siddiqui AA, Benz RL. Beneficial hematologic effects of daily oral
2005;16(7):2180–9. ascorbic acid therapy in ESRD patients with anemia and abnormal iron
56. Singh AK, Szczech L, Tang KL, et al. CHOIR investigators. Correction of anemia homeostasis: a preliminary study. Renal Failure 2008;30(9):884–9.
with epoetin alfa in chronic kidney disease. N Engl J Med 2006;355(20):2085–98. 70. Keven K et al. Randomized, crossover study of the effect of vitamin C on EPO
57. Sczcech LA, Burnhart HXS, Inrig JK, Reddan DN, Sapp S, Califf RM, et al. response in hemodialysis patients. Am J Kidney Dis 2003;41(6):1233–9.
Secondary analysis of the CHOIR trial epoetin-a dose and achieved hemoglobin 71. Attallah N, Osman-Malik Y, Frinak S, Besarab A. Effect of intravenous ascorbic
outcomes. Kidney Int 2008;74:791–8. acid in hemodialysis patients with EPO-hyporesponsive anemia and
58. Besarab A, Frinak S, Yee J. What is so bad about a hemoglobin of 12–13 g/dL for hyperferritinemia. Am J Kidney Dis 2006;47(4):644–54.
chronic kidney disease patients anyway? Adv Chronic Kidney Dis 72. Tarng DC, Huang TP, Wei YH. Erythropoietin and iron: the role of ascorbic acid.
2009;16(2):131–42. Nephrol Dial Transplant 2001;16(Suppl. 5):35–9.
59. Miyashita K, Tojo A, Kimura K, et al. Blood pressure response to erythropoietin 73. Giancaspro V et al. Intravenous ascorbic acid in hemodialysis patients with
injection in hemodialysis and predialysis patients. Hypertens Res 2004;27(2):79–84. functional iron deficiency: a clinical trial. J Nephrol 2000;13(6):444–9.
60. Kalantar-Zadeh K, Regidor DL, McAllister CJ, Michael B, Warnock DG. Time- 74. Kletzmayr J, Mayer G, Legenstein G, et al. Anemia and carnitine supplementation
dependent associations between iron and mortality in hemodialysis patients. J in hemodialyzed patients. Kidney Int 1999;55(Suppl. 69):93–106.
Am Soc Nephrol 2005;16(10):3070–80. 75. Labonia WD. L-Carnitine effects on anemia in hemodialyzed patients treated
61. Fletes R, Lazarus JM, Gage J, Chertow GJ. Suspected iron dextran-related adverse with erythropoietin. Am J Kidney Dis 1995;26:757–64.
drug advents in hemodialysis patients. Am J Kidney Dis 2001;37:743–9.

Вам также может понравиться