CKD and Cardiovascular Risks
CKD and Cardiovascular Risks
Cardiovascular Disease in
Chronic Kidney Disease
Peter Stenvinkel, Charles A. Herzog
Diminished estimated glomerular filtration rate (eGFR) and albuminuria       cardiac deaths in the U.S. Renal Data System (USRDS) database are
are powerful graded, independent predictors of cardiovascular (CV)           attributable to arrhythmias.
morbidity and mortality1 and all-cause mortality (Fig. 81.1). Even subtle         Of incident dialysis patients, 75% have LVH and 75% to 85% have
kidney dysfunction, as suggested by albuminuria, increases CV risk           hypertension. Hypertension, anemia, vascular noncompliance, and
because it may reflect microvasculature health, including endothelial        volume overload contribute to LVH. Based on echocardiography, 85%
function. Patients with end-stage renal disease (ESRD) face an extraor-      to 90% of patients have a left ventricular (LV) ejection fraction (EF)
dinary risk for premature death, largely because of CV complications.        of 50% or higher, despite frequent congestive heart failure (CHF), that
However, patients with eGFR below 60 ml/min/1.73 m2 are much more            is, heart failure with preserved EF (HFpEF), not heart failure with reduced
likely to die than to develop ESRD, reflecting the burden of cardiovas-      EF (HFrEF), characterizes most CHF episodes. Therefore, many volume
cular disease (CVD) in this population. The most effective strategy for      overload episodes in dialysis patients may be attributable to diastolic
reducing CV morbidity and mortality would be to target patients with         dysfunction or circulatory congestion. Fig. 81.3 provides a snapshot of
mildly reduced eGFR for prevention and treatment.                            CVD event rates in prevalent dialysis patients.
    Patients with chronic kidney disease (CKD) were often excluded
from randomized controlled trials (RCTs) targeting CVD, possibly             Cardiovascular Disease Is Present Before the Start of
reducing acceptance of evidence-based therapies (validated in nonrenal       Renal Replacement Therapy
patients) and fostering “therapeutic nihilism” in clinicians who treat       In elderly CKD patients at stage 2 or 3, traditional risk factors seem to
CKD patients. Thus novel treatment strategies are urgently needed to         be the major contributors to CV mortality. Atherosclerosis Risk in Com-
reduce the unacceptable high CV event rate.                                  munities (ARIC) data suggest that both traditional and novel risk factors
    Like conventional atheromatous occlusive vascular disease, CKD is        are relevant at CKD stage 4, and novel risk factors are far more prevalent
characterized by generalized vasculopathy, with other characteristics,       in dialysis patients than in the general population (Fig. 81.4).3 The
including left ventricular hypertrophy (LVH), vascular calcification,        Framingham predictive instrument does not accurately predict coronary
and vascular noncompliance. Numerous CVD risk factors are specific           events in CKD. Mild to moderate CKD is associated with increased risk
to CKD and operate in addition to conventional risk factors found in         for venous thromboembolism, supporting the concept of hypercoagu-
the general population.                                                      lability in CKD. Fig. 81.5 shows the burden of CVD in elderly patients
                                                                             with CKD. Because the incidence of CV events is much higher in the
EPIDEMIOLOGY                                                                 first weeks after hemodialysis (HD) initiation, concerns have been raised
                                                                             that the dialysis procedure per se may trigger CV events.4
Prevalence of Cardiovascular Complications in Chronic
Kidney Disease                                                               Racial and International Differences in Cardiovascular
Interpretation of epidemiologic studies of CVD is problematic because        Disease Prevalence
of the difficulty in defining cause of death. Unexpected sudden death        In the United States, survival is better for African American than for
most likely results from arrhythmia, but a subarachnoid hemorrhage,          White dialysis patients. However, overall CV mortality among dialysis
massive embolic stroke, or aortic dissection might be indistinguishable      patients from the United States is significantly greater than is observed
from a primary arrhythmic event without an autopsy. Defining “coronary       in Japan and Europe, even after adjustment for standard risk factors
heart disease” (CHD) is also problematic: in the general population,         and dialysis dose. Higher mortality rates in U.S. dialysis patients may
sudden cardiac death is a primary complication of CHD, but this is           be related to higher prevalence of sicker or diabetic patients, differences
unlikely to be true for dialysis patients. A history of angina cannot        in dialysis practice patterns, cultural habits, differences in diet, or genetic
reliably classify a patient as having CHD because angina (resulting from     variations.
supply-demand mismatch) can occur in patients with LVH and angio-
graphically pristine coronary arteries. This probably relates to the         Reverse Epidemiology
increased myocardial fibrosis, diminished relative capillary density, and    Reverse epidemiology (preferably called confounded epidemiology) refers
increased thickening of the intramyocardial vessel walls in uremia. At       to the paradoxical observation that the association among hypercho-
lower levels of eGFR (especially in dialysis patients), the burden of        lesterolemia, hypertension, obesity, and poor outcomes, including CV
nonatherosclerotic (vs. atherosclerotic) CVD is relatively increased (Fig.   death, in the general population does not exist and may be reversed in
81.2).2 Although occlusive CHD is common in CKD, acute myocardial            CKD. Patients with wasting and inflammation appear to mostly account
infarction (AMI) accounts for only 14% of cardiac deaths; 66% of             for poor survival and confounded epidemiology.
942
                                                  downloaded from www.medicalbr.com
                                                                         CHAPTER 81            Cardiovascular Disease in Chronic Kidney Disease                                 943
       A                                                15                                       14.14
            from any cause (per 100 person-yr)
                                                        14
               Age-standardized rate of death
                                                        13
                                                        12                             11.36
                                                        11
                                                        10
                                                         9
                                                         8                                                                                      Nonatherosclerotic CVD
                                                         7                                                                                      LVH
                                                         6                                                                                      Arrhythmias
                                                                              4.76
                                                         5                                                                                      Sudden cardiac death   Risk
                                                         4                                                                                      Arterial calcification
                                                         3                                                                                      Valve calcification
                                                         2             1.08                                                                     hemorrhagic stroke
                                                         1    0.76
                                                                                                                                                Others
                                                         0
                                                              ≥60     59-45   44-30    29-15      <15
                                                                                                                                        Atherosclerotic CVD event
                                                                Estimated GFR (ml/min/1.73 m2)                                          CAD
        No. of events                                        25,803 11,569    7802     4408      1842
                                                                                                                                        ischemic stroke
                                                                                                                                        PAD
                                                                                                                                       CKD stages
                                                                                                                                                                    Time
       B
            Age-standardized rate of cardiovascular
                                                        30
                                                                                                                                         Risk for fatality after CVD event
                                                        25                             21.80
                                                        20                                                                                                                   Risk
                                                        15
                                                                              11.29
                                                        10
                                                        5             3.65
                                                              2.11
                                                        0                                                  Fig. 81.2 Change in cardiovascular risk during chronic kidney
                                                              ≥60     59-45   44-30    29-15      <15      disease (CKD) progression. Cardiovascular disease (CVD) event (upper
                                                                                                           triangle), contributions of atherosclerotic CVD (tan), nonatherosclerotic
                                                                Estimated GFR (ml/min/1.73 m2)
                                                                                                           CVD (orange), and risk for fatality after CVD event (blue). PAD, Peripheral
        No. of events                                        73,108 34,690 18,580      8809      3824      artery disease. (From reference 2.)
                                                       150
                                                       140
                                                       130                                                 Traditional Risk Factors
                                                       120
                                                       110                                                 Age, Gender, and Smoking
                     (per 100 person-yr)
                                                       100                             86.75               The U.S. National Health and Nutrition Examination Surveys (NHANES)
                                                        90
                                                        80                                                 show the prevalence of CV factors and CVD prevalence in relation to
                                                        70                                                 age and CKD stage. In the United States, the average age at renal replace-
                                                        60                                                 ment therapy (RRT) initiation is 63 years, when CVD is common. An
                                                        50                    45.26
                                                        40                                                 individual-level meta-analysis including more than 2 million participants
                                                        30                                                 showed that low eGFR and high albuminuria were independently asso-
                                                              13.54   17.22
                                                        20                                                 ciated with mortality and ESRD regardless of age. Female gender is
                                                        10
                                                         0                                                 associated with a 4% independent increased risk for mortality in incident
                                                              ≥60     59-45   44-30    29-15      <15      dialysis patients and smoking with a 52% increased risk for death in
                                                                Estimated GFR (ml/min/1.73 m2)             dialysis patients.
300
                                                                                                                         Hemodialysis
                         Rate per 1000 patient-yr
                                                                                                                         Peritoneal dialysis
                                                    200                                                                  Transplant
100
                                                      0
                                                          CHF       CVA/TIA     PAD     Cardiac        AMI        Revasc:     Revasc:      ICD/
                                                                                         arrest                    PCI         surg       CRT-D
                        Fig. 81.3 Event rates of cardiovascular diagnoses and procedures by modality. Point prevalent
                        end-stage renal disease (ESRD) patients on January 1, 2005, age 20 and older, with Medicare as primary
                        payer and survival for 90 days after ESRD diagnosis. AMI, Acute myocardial infarction; CHF, congestive heart
                        failure; CRT-D, cardiac resynchronization therapy defibrillator; CVA, cerebrovascular accident; ICD, implantable
                        cardioverter-defibrillator; PAD, peripheral arterial disease; PCI, percutaneous coronary intervention; TIA, tran-
                        sient ischemic attack. (From U.S. Renal Data System. USRDS 2009 Annual Data Report: Atlas of Chronic
                        Kidney Disease & End-Stage Renal Disease in the United States. Bethesda, MD: National Institutes of Health,
                        National Institute of Diabetes and Digestive and Kidney Diseases; 2009.)
potent independent risk factor for all-cause and CVD-related deaths,                              mean arterial and diastolic pressure may decrease. The relationship
including after coronary revascularization or AMI. Nevertheless the                               between BP and mortality is U shaped; isolated systolic hypertension
rate of incident AMI is even higher for patients with CKD stages                                  and increased pulse pressure probably indicate high long-term risk in
3b to 4 without diabetes than for patients with diabetes and CKD                                  dialysis patients, whereas low mean and diastolic BPs (DBPs) predict
stages 1 or 2.                                                                                    early mortality. CKD patients are frequently “nondippers” and experi-
                                                                                                  ence sleep apnea and sympathetic nervous system activation.
Hypertension
Hypertension is common but variably treated in CKD patients. Of                                   Dyslipidemia
NHANES subjects with CKD stages 3 to 4, 80% had a blood pressure                                  The relationship between hypercholesterolemia, CVD, and mortality
(BP) of 130 or greater/80 or greater mm Hg, and only 20% were aware                               in CKD is weak because some CV abnormalities, such as cardiomyopathy
of it and had BP adequately controlled. In CKD stages 1 and 2, 63%                                and arteriosclerosis, may be less dependent on dyslipidemia than on
were hypertensive and only 11% were adequately controlled. Recent                                 other factors. Low rather than high serum cholesterol level is associated
NHANES data suggest modest improvement in hypertensive people                                     with poor survival in HD patients, likely related to confounding by
with CKD stages 3 or 4.6 Hypertension predicts mortality in CKD patients                          protein-energy wasting and inflammation. After adjustment for C-reactive
before or at dialysis initiation. Isolated systolic hypertension with                             protein (CRP) levels, high cholesterol level predicts risk in noninflamed
increased pulse pressure is by far the most prevalent BP anomaly in                               ESRD patients.
dialysis patients, resulting from arterial medial sclerosis with secondary                            Progressive CKD leads to changes in blood lipids typically associated
stiffening. Stiff vessels cause increased pulse wave velocity, resulting in                       with vascular disease, including decreased apolipoprotein A (apoA)-con-
increased systolic BP (SBP) peak pressure by a prematurely reflected                              taining lipoproteins and increased apoB-containing lipoproteins (Table
pulse wave, progressive LV dysfunction, and finally CHF. At this stage,                           81.1). Serum triglycerides are elevated in most ESRD patients, whereas
Monocyte/macrophage
                                                       ↑ Proinflammatory cytokines
                                                       ↓ Antiinflammatory cytokines
↑ Bone remodeling
↑ Insulin resistance
↑ Muscle catabolism
                      ↑ Acute-phase reactants
                      ↓ Fetuin-A
                                                                                             ↑ Endothelial dysfunction
                                                                                             ↑ Monocyte adhesion
                                                                        ↑                    ↑ Smooth muscle cell proliferation
                                           ↑ Adipocytokine                Appetite
                                                                                             ↑ LDL oxidation
                                             production                 ↑ REE
                                                                                             ↑ Vascular calcification
                     Fig. 81.7 Potential mechanisms by which elevated circulating levels of proinflammatory and
                     antiinflammatory cytokines may promote accelerated atherosclerosis, other uremic complica-
                     tions, and wasting. LDL, Low-density lipoprotein; REE, resting energy expenditure. (From reference 8.)
(ESAs) results in LVH regression, current information suggests no CV                 artery, calcification can occur at young ages. Calciphylaxis (calcific uremic
outcome benefit of normalized hemoglobin (see Chapter 82).                           arteriolopathy) is discussed in Chapter 88.
                                                                                         Vascular calcification is not derived only from passive calcium and
Secondary Hyperparathyroidism and Mineral Metabolism                                 phosphate precipitation. Rather, it involves differentiation of vascular
Disturbances of calcium and phosphate metabolism might accelerate                    smooth muscle cells toward osteoblasts induced by phosphate, calcium,
calcifying atherosclerosis and arteriosclerosis (see also Chapter 85).               and other factors, such as calcitriol and proinflammatory cytokines.
Recent evidence suggests that chronically elevated FGF-23 levels con-                Uremic bone disease and protein-energy wasting may be additional risk
tribute directly to high rates of LVH, atrial fibrillation (AF), and mortality.      factors for vascular calcification.11 One way by which chronic inflam-
In registry data, a strong independent mortality risk is predicted by                mation promotes vascular calcification may involve downregulation of
hyperphosphatemia, an intermediate risk by elevated serum calcium                    fetuin-A, the most potent circulating inhibitor of extraosseous calcifica-
levels, and a weak risk by high or low serum intact parathyroid hormone              tion and a component of calciprotein particles. Apart from fetuin-A,
(PTH) levels. The overall mortality risk prediction attributable to mineral          other inhibitors, such as magnesium, probably counteract unwanted
metabolism disorders is estimated to be about 17% in HD patients.                    calcification. Leptin, matrix GLA protein, FGF-23, pyrophosphates,
                                                                                     bone morphogenic proteins (e.g., BMP-2 and BMP-7), and osteopro-
Cardiovascular Calcification                                                         tegerin may be related to accelerated vascular calcification in ESRD.
CV calcification may affect the arterial media, atherosclerotic plaques,             Deficiency of vitamin K and/or treatment with vitamin K antagonists
myocardium, and heart valves. Medial calcification causes arterial stiff-            (warfarin) may accelerate the vascular calcification process in the
ness and, consequently, increased pulse pressure. The pathophysiologic               uremic milieu.
role of plaque calcification is less clear because soft plaques are assumed
to rupture and cause AMI; atherosclerotic calcification is a potent risk             Advanced Glycation End-Products
marker for CV events, but its utility as a risk marker for clinical man-             AGEs accumulate in CKD patients as a result of nonenzymatic gly-
agement of CKD patients remains controversial. Valvular calcification                cation, oxidative stress, intestinal food components, and diminished
mostly affects the aortic and mitral (annulus) valves in dialysis patients           clearance of AGE precursors. Stable AGE residues of long-lived proteins
and contributes to progressive stenosis and associated morbidity and                 are biomarkers of cumulative metabolic, inflammatory, and oxidative
mortality. In dialysis patients, extensive vascular, especially coronary             stress; carbonyl stress is speculated to contribute to tissue aging and
long-term CKD complications. Whether AGE inhibition may affect CV                                  occurred only in patients with ST-segment elevation myocardial infarc-
disease in CKD is unknown.                                                                         tion (STEMI), specifically related to in-hospital mortality. Rates of
                                                                                                   non-STEMI (NSTEMI) mortality or postdischarge mortality have not
Dialysis Modality                                                                                  improved.12 In-hospital deaths increase with decreasing GFR.13 This
Reports from dialysis registries are inconsistent regarding whether                                poor outcome has been attributed to underrecognition resulting from
HD or PD is associated with better outcomes. Valid mortality compari-                              atypical presentations, underuse of appropriate diagnostic investiga-
sons between HD and PD modalities are not available because this                                   tions, and undertreatment (therapeutic nihilism).14 A U.S. registry of
would require stratification of patients according to underlying ESRD                              dialysis patients hospitalized for AMI found the following14:
cause, age, and level of baseline comorbidity. Cardiac arrhythmias,
such as AF, seem to occur more often on the day of HD compared                                         incorrectly with respect to acute coronary syndrome (ACS).
with PD.
                                                                                                       chest pain.
CLINICAL MANIFESTATIONS AND
                                                                                                       elevation.
NATURAL HISTORY
Fig. 81.8 shows survival of patients with CV diagnoses and procedures,                                 nondialysis patients, were eligible for acute coronary reperfusion.
by RRT modality.
                                                                                                       patients actually received reperfusion.
Chest Pain, Coronary Heart Disease, and Acute
Myocardial Infarction                                                                                 patients. In-hospital cardiac arrest occurred twice as frequently in
AMI in dialysis patients is associated with poor long-term survival. The                              dialysis as in nondialysis patients (11% vs. 5%).
unadjusted 2-year mortality rate is not changing: 71% in 1977 to 1984                                 Similar findings occur across the CKD spectrum; the likelihood of
and 72% in 2008,12 despite dramatic improvements in AMI outcomes                                   increased mortality and lower prevalence of both STEMI and chest
in the general population (Fig. 81.9). Significant improvement has                                 pain are correlated with severity of non–dialysis-dependent CKD in
0.8
0.6
                            0.4       Hemodialysis
                                      Peritoneal dialysis
  Probability of survival
                            0.2
                                      Transplant
                            0.0
                                              AMI                  Coronary revascularization:         Coronary revascularization:                   ICD/CRT-D
                            1.0                                               PCI                               Surgical
0.8
0.6
0.4
0.2
                            0.0
                                  0    3       6       9      12 0        3       6       9       12 0       3        6      9       12 0        3          6    9    12
                                                                                              Months
                                        Fig. 81.8 Survival of patients with cardiovascular diagnoses and procedures, by modality. January
                                        1, 2005, point prevalent end-stage renal disease patients, age 20 and older, with a first cardiovascular diag-
                                        nosis or procedure in 2005-2007. AMI, Acute myocardial infarction; CHF, congestive heart failure; CRT-D,
                                        cardiac resynchronization therapy defibrillator; CVA, cerebrovascular accident; ICD, implantable cardioverter-
                                        defibrillator; PAD, peripheral arterial disease; PCI, percutaneous coronary intervention; TIA, transient ischemic
                                        attack. (Modified from U.S. Renal Data System. USRDS 2009 Annual Data Report: Atlas of Chronic Kidney
                                        Disease & End-Stage Renal Disease in the United States. Bethesda, MD: National Institutes of Health,
                                        National Institute of Diabetes and Digestive and Kidney Diseases; 2009.)
                                                                                                                          systemic embolism
                            Dabigatran 110 mg (n = 1,196)
                                                                                                                               Stroke or
                            Rivaroxaban 15 mg (n = 1,474)
Edoxaban 30 mg (n = 1,302)
                                                                                                                                  Major bleeding
                            Dabigatran 110 mg (n = 1,196)
Rivaroxaban 15 mg (n = 1,474)
Edoxaban 30 mg (n = 1,302)
raised jugular vein pulse, or a third heart sound, or it may be largely          receiving tissue versus mechanical valves and fewer valve-related com-
asymptomatic. Tolerance of large ultrafiltration volumes may indi-               plications with tissue valves. The overall mortality is high, with in-
cate that the dry weight target (see Chapter 96) has not been reached.           hospital mortality about 20% (four times higher than in non-CKD
Reaching an optimal dry weight, however, does not necessarily lead               patients) and 2-year survival of 40%. Transcatheter aortic valve replace-
to immediate BP correction; a lag phase of some weeks can precede                ment may be appropriate in dialysis patients with symptomatic aortic
improvement.                                                                     stenosis who are not good candidates for surgery. In kidney transplant
                                                                                 recipients, in-hospital mortality was 11% for tissue and 15% for mechani-
Pericarditis                                                                     cal valve patients, and 2-year mortality rates were 62% and 60%, respec-
Dialysis-associated pericarditis may be related to intercurrent illnesses        tively. In the entire cohort of kidney transplant patients, the rate of
(including viral infections), fistula recirculation leading to underdialysis,    endocarditis after valve surgery was 5% per year.28
or underlying diseases such as systemic lupus. Fever with pericardial
pain or a rub on heart auscultation, unexplained cardiomegaly on chest           Infective Endocarditis
films, or hemodynamic instability should prompt echocardiography.                Estimated incidence of infective endocarditis in U.S. dialysis patients
An effusion causing overt hemodynamic compromise (i.e., pericardial              is 267 cases per 100,000 patient-years. Vascular access, including tem-
tamponade) or large pericardial effusions judged unlikely to resolve with        porary and semipermanent catheters, is an important source of infection;
conservative measures require echocardiographically guided or com-               heightened risk for bacteremia related to HD therapy is likely an impor-
puted tomography (CT)-guided pericardiocentesis or surgical drainage.            tant aspect of endocarditis risk. Dialysis patients with bacterial endo-
Intensive dialysis is indicated for true uremic pericarditis; the optimal        carditis have poor in-hospital and long-term survival. One-year survival
treatment of dialysis-associated pericarditis is much less clear in patients     for U.S. dialysis patients with endocarditis who subsequently received
without hemodynamic compromise. Citrate-based anticoagulation is                 valve replacement surgery was about 50%.29 A risk model for operative
preferred given the risk for hemorrhage-induced pericardial tamponade.           mortality may be helpful in managing these high-risk patients.
ultrastructure and function, including endothelial dysfunction, interstitial   mortality. As in the general population, CKD patients with LVEF below
fibrosis, decreased perfusion reserve, and diminished ischemia toler-          40% should be evaluated for CHD (exceptions are pediatric or young
ance.31 Low-potassium dialysate (<2 mmol/l) doubles the risk for cardiac       adult patients with nondiabetic CKD and other patients known to be
arrest.32 The rate of cardiac arrest is 50% higher for HD than for PD          at low risk for CHD).
patients 3 months after dialysis initiation but is higher for PD patients          A new classification scheme for CHF staging specifically targeted
at 3 years. The highest rate of sudden cardiac death occurs in the first       for dialysis patients has been proposed by the Acute Dialysis Quality
2 months after HD initiation.33 In cardiac arrests occurring in HD             Initiative (ADQI) XI Workgroup.35 The three key elements are as follows:
centers, the predominant rhythm is ventricular fibrillation (66%), fol-        1. Standardized echocardiographic evidence of structural and/or func-
lowed by pulseless electrical activity (23%), and asystole (10%).                  tional cardiac abnormalities.
    In women with CHD, eGFR below 40 ml/min/1.73 m2 was associ-                2. Dyspnea occurring in the absence of primary lung disease, including
ated with a 2.3-fold increased risk for sudden cardiac death. Despite a            isolated pulmonary hypertension (i.e., not secondary to elevated
graded, incremental risk for arrhythmic death and impaired renal func-             pulmonary capillary wedge pressure).
tion, the overall magnitude of risk in stage 3 CKD patients is small           3. Improvement of congestive symptoms after RRT/ultrafiltration.
compared with that in dialysis patients.                                           The justification for the KDOQI 2005 Guidelines (recommending
    Onsite defibrillation capability in HD centers (preferably with auto-      echocardiograms in incident dialysis patients) and the validity of the
matic external defibrillators) was recommended in a U.S. practice              first element of the proposed ADQI XI heart failure staging scheme
guideline in 2005.27 The role that implantable cardioverter-defibrillators     (standardized echocardiographic evidence of structural and/or functional
(ICDs) may play in reducing mortality in CKD patients is controversial,        heart disease) are both supported by the finding that right ventricular
particularly regarding primary prevention. CKD may attenuate the               dysfunction was associated with a 66% increased risk for death.36
survival advantage of ICDs, but older age and medical comorbidity
should not routinely exclude patients from receiving ICDs. In dialysis         Stress Tests and Screening Renal
patients who survived cardiac arrest, ICD implantation was associated          Transplant Candidates
with a 14% to 42% reduction in long-term mortality.34 The role of              ESRD patients are poorly suited for conventional exercise stress elec-
ICDs in dialysis patients is uncertain: the Wearable Cardioverter Defi-        trocardiography because of limited exercise tolerance and frequent
brillator in Hemodialysis Patients (WED-HED) trial (clinicaltrials.gov,        resting electrocardiographic abnormalities. Accuracies of pharmacologic
NCT02481206) was a prospective RCT, testing wearable defibrillators            stress echocardiographic and nuclear scintigraphic techniques are remark-
for prevention of sudden cardiac death in incident HD patients who             ably variable across the world; they are operator dependent, and the
do not qualify for ICDs under current guidelines. Unfortunately, the           approach of individual sites to cardiac screening should rely on insti-
trial was terminated in 2017 due to low enrollment.                            tutional expertise. Moreover, prediction of the likelihood of future events
                                                                               may differ considerably from prediction of coronary anatomy. In severe
                                                                               CKD, dobutamine stress echocardiography is an independent predictor
DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS                                           of long-term mortality. Sensitivities and specificities for detection of
Key issues in the diagnosis of CVD are underrecognition of symptoms,           CHD in renal transplant candidates have been reported to range from
underuse of appropriate diagnostic investigations, and interpretation          44% to 90%. A meta-analysis concluded that presence of inducible
of those investigations.                                                       myocardial ischemia by any stress-imaging test is independently predic-
                                                                               tive of increased AMI risk and cardiac death, whereas a fixed or resting
Blood Pressure Measurements                                                    defect or abnormality is predictive of cardiac death but not AMI. A
Outcome prediction by ambulatory BP monitoring is not necessarily              subsequent meta-analysis by the Cochrane Collaboration (Fig. 81.11)
better than by office BP measurements. However, ambulatory monitor-            concluded that dobutamine stress echocardiography is probably more
ing is useful to identify high-risk nondippers and inverted dippers,           accurate than myocardial stress nuclear scintigraphy for noninvasive
allowing consequent treatment adjustments.                                     detection of CHD in renal transplant candidates.37
                                                                                   The major problem with “screening” is use of test results for clinical
Electrocardiography and Echocardiography                                       management. The evidence for prophylactic revascularization of asymp-
The National Kidney Foundation’s Kidney Disease Outcomes Quality               tomatic renal transplant candidates (or any other patient group) is
Initiative (KDOQI) clinical practice guidelines for CVD in dialysis            weak.38 The International Study of Comparative Health Effectiveness
patients recommend an electrocardiogram at dialysis initiation and             with Medical and Invasive Approaches–Chronic Kidney Disease Trial
then annually.27 Diabetic HD patients presenting without sinus rhythm          (ISCHEMIA-CKD; clinicaltrials.gov, NCT01985360) is a prospective
(11% of the cohort) were 89% more likely to die and 164% more likely           RCT testing the comparative efficacy of a conservative versus invasive
to sustain a stroke. CKD patients (eGFR <60 ml/min/1.73 m2) with               strategy for treatment of stable CHD in patients with eGFR less than
increased QRS duration had 15% greater risk for incident CHF, 13%              30 ml/min/1.73 m2, including dialysis patients. The best observational
greater risk of incident CHD, and 17% greater risk for mortality per           data supporting the usefulness of preemptive coronary revascularization
10-ms increase; prolongation of the QT interval was independently              showed 3-year cardiac event–free survival of 90% for wait-listed revas-
associated with adverse outcome.                                               cularized patients.39 Optimal medical therapy (which should constitute
    KDOQI guidelines recommend echocardiography in all dialysis                the treatment strategy for all patients) may potentially attenuate the
patients after they achieve “dry weight” targets, preferably 1 to 3 months     putative benefit of prophylactic coronary revascularization. One algo-
after dialysis initiation on an interdialytic day for HD patients and at       rithm for screening and management of CHD in renal transplant can-
3-year intervals thereafter.27 The rationale for this guideline is that        didates is presented in Fig. 81.12.
diminished LV systolic function, an important independent risk factor
for CVD and mortality,27 is not accurately diagnosed by history, physical      Coronary Angiography
examination, or chest radiography. Detection of unsuspected cardio-            Coronary angiography should be considered in stable ESRD patients with
myopathy is also important, given that carvedilol therapy in such patients     evidence for inducible myocardial ischemia, unstable patients with ACS
improved LV systolic function, decreased hospitalization, and reduced          (performed urgently for STEMI), and patients with LVEF below 40%. In
1.0
0.8
                                   Posttest probability
                                                          0.6
                                                          0.4
                                                                                                                     MPS (positive)
                                                                                                                     DSE (positive)
                                                          0.2                                                        MPS (negative)
                                                                                                                     DSE (negative)
                                                          0.0
                                                                0.0       0.2       0.4      0.6        0.8       1.0
Post-test probability
         Test                                             Pretest probability of (%)                  Posttest probability (%)                  Posttest probability (%)
                                                          coronary artery disease                      after positive result                     after negative result*
                    Fig. 81.11 Accuracy of dobutamine stress echocardiography versus myocardial perfusion scin-
                    tigraphy for diagnosing coronary artery disease coronary artery disease in renal transplant
                    candidates. (From reference 67.)
CKD and dialysis patients with residual renal function, fear of contrast                           Imaging of Vascular Calcification
nephropathy may restrain use of coronary angiography (see Chapter                                  Vascular calcification can be visualized by conventional x-ray techniques
70 for preventive measures); one retrospective study of 76 nondialysis                             and by multislice spiral CT or electron-beam CT. Valvular and large-
patients with mean eGFR of 12.5 ml/min/1.73 m2 found no significant                                artery calcification can be visualized by ultrasound techniques, and (if
postangiographic deterioration in renal function. However, patients                                present) predicts worse outcome in dialysis patients, although the value
sustaining acute kidney injury (AKI) after coronary angiography are at                             of coronary artery calcification in dialysis patients as a surrogate for
heightened risk for long-term mortality, ESRD, and hospitalization.40                              CHD severity has been questioned.
However, fear of AKI should not deter clinically mandated coronary
angiography. Echocardiography should be performed before any non-                                  Biomarkers
emergent coronary angiography in CKD patients to diagnose clinically                               Plasma brain natriuretic peptides (BNP and NT-proBNP), cardiac tro-
unsuspected valvular disease or cardiomyopathy, to gauge preprocedure                              ponins (cTnT, cTnI), and high-sensitivity (hs) CRP are prognostic risk
volume status, and to assess LV function (to avoid excessive exposure to                           markers in the evaluation of heart disease in ESRD.42 BNP reflects cardiac
radiocontrast media through unwarranted ventriculography).                                         filling pressures (not limited to the left heart), troponins reflect myocardial
    Noninvasive coronary CT angiography (CCTA) may be problematic                                  cell death (but not necessarily ischemia), and hsCRP reflects inflamma-
in dialysis patients because of medial calcification interfering with                              tion. Elevation of cTnT occurs even in pediatric CKD patients and is
angiographic interpretation. Nevertheless, noninvasive CCTA has diag-                              associated with cardiac dysfunction. Elevated levels of serum troponin
nostic sensitivity to detect obstructive CHD nearly comparable to invasive                         in ESRD patients should not be uncritically attributed to myocardial
coronary angiography and markedly superior to noninvasive nuclear                                  ischemia caused by obstructive coronary artery disease. Elevated levels
single-photon emission computed tomography imaging.41 Generaliz-                                   of cTnT are associated with the presence and severity of HD-induced
ability of these findings must be interpreted in the context of individual                         myocardial stunning.43 The cardiac biomarker–based diagnosis of ACS
institutional expertise. Noninvasive gadolinium-based magnetic reso-                               requires a time-appropriate rise and fall of the biomarker. The most
nance angiographic imaging in patients with severe CKD remains                                     cost-effective combination of biomarkers for risk stratification in dialysis
problematic because of lingering concerns about nephrogenic fibrosing                              patients might be high-sensitivity cardiac troponin and a natriuretic
dermopathy; the usefulness of imaging without contrast media is                                    peptide, but this is speculative. A recent study underscored the robust-
uncertain.                                                                                         ness of IL-6 as a classifier of clinically overt CVD and predictor of
                                                          Two-dimensional
                                                          echocardiography
     Low-risk        High-risk
     patients        patients                                                       Intervention                   Negative               Positive
                                                          No intervention          CABG or PCI
                    Fig. 81.12 Algorithm for management of coronary heart disease in renal transplant candidates.
                    CABG, Coronary artery bypass graft surgery; CAD, coronary artery disease; MI, myocardial infarction; PCI,
                    percutaneous coronary intervention. (Modified from Herzog CA. Acute MI in dialysis patients: How can we
                    improve the outlook? J o Critical Illness 1999;14(11):613-621.)
all-cause mortality in CKD stage 5.44 In CKD, NT-proBNP and BNP are              TREATMENT AND PREVENTION OF
equivalent predictors of decompensated heart failure, but NT-proBNP              CARDIOVASCULAR DISEASE
is a better predictor of survival. Fig. 81.13 graphically displays the rela-
tionship of cTnT and cTnI levels in asymptomatic dialysis patients and           Risk Factor Reduction
long-term survival. On the basis of these data, the U.S. Food and Drug           Lifestyle Factors and Smoking
Administration approved the measurement of cTnT in dialysis patients             Because physical inactivity is associated with albuminuria and CV mor-
for risk stratification (mortality prediction).27                                tality, CKD patients should be advised to stay as physically active as
    High-sensitivity cardiac troponin (hs-cTn) assays, which offer the           possible and to avoid smoking.
advantage of high precision, serve dual, complementary (but distinct)
roles: the diagnosis of AMI, which now includes cardiac biomarkers and           Weight and Diet
risk stratification based on detection of hs-cTn in an asymptomatic,             Lifestyle changes, including balanced diets with regard to saturated fat
nonischemic setting (e.g., perhaps a marker for apoptosis). Hs-cTn–based         and carbohydrates (in diabetic patients), probably reduce CV morbidity
risk stratification should employ reference change values over time,             and should be encouraged. However, in all CKD stages, protein-energy
rather than simple thresholds, particularly when data are inconclusive           wasting must be avoided; especially in dialysis patients, increased body
regarding acceptable normal values in special populations, such as dialysis      mass index has been associated with improved outcomes, possibly
patients, known to have chronically elevated cardiac troponin levels.45          reflecting confounded epidemiology; thus the obesity paradox only
    FGF-23 has also proven to be a strong outcome predictor in CKD               exists in HD patients with inflammation.9
patients, but further studies are needed to demonstrate its role as an
additional clinical biomarker.                                                   Hypertension and Coronary Heart Disease
    An improved CV prediction risk score was recently developed for              BP targets for CKD patients, in particular those with diabetes or pro-
use in HD patients (http://aro-score.askimed.com/).46                            teinuria greater than 1 g/day, are discussed in Chapter 79. A goal of
                                                                                                             Diabetes Mellitus
                 Kaplan-Meier Survival Curves                                                                Optimal glycemic control (see Chapter 32), reaching BP target levels,
                 by Baseline Troponin Cutoffs                                                                and lipid monitoring (with subsequent dyslipidemia treatment) are
                                                                                                             crucial in managing diabetic CKD patients. CHD and other CVD should
                                                                                                             be treated aggressively in this high-risk group. Because the harm associ-
                                           100
                                                                                                             ated with severe hypoglycemia might counterbalance the potential benefit
                 Cumulative survival (%)
catheter use is also important; short daily dialysis with better fluid        Revascularization During Instability in Coronary Artery Disease (FRISC
status was associated with decreasing CRP levels compared with con-           II) trial indicated a superior outcome with an early invasive strategy in
ventional HD. Volume status should be carefully monitored to avoid            ACS compared with conservative management. However, the optimal
inflammation. Altered intestinal microbial flora as a potential risk factor   coronary revascularization method in CKD remains controversial. A post
for systemic uremic inflammation merits further study. It was recently        hoc analysis of CKD patients enrolled in the Arterial Revascularization
demonstrated that medium cut-off dialysis membranes reduce uremic             Therapies Study (ARTS) found similar outcomes for coronary artery
inflammation52; thus larger trials with longer treatment periods are          bypass grafting (CABG) or multivessel percutaneous coronary interven-
encouraged.                                                                   tion (PCI) with non–drug-eluting stents (DESs) for death, myocardial
                                                                              infarction, or stroke. In elderly non–dialysis-dependent CKD patients,
Oxidative Stress                                                              the incidence of ESRD is lower after PCI (5.4% at 3 years vs. 6.8% for
Two placebo-controlled interventional studies showed that vitamin E           CABG), but long-term risk for death (28% 3-year mortality with CABG,
and N-acetylcysteine decreased the number of CV events in HD patients.        33% with PCI) or the combined event of death or ESRD is lower after
Unfortunately, both studies were small and of limited duration, so            CABG. The relative survival advantage of CABG (vs. PCI) occurs only
adequately powered randomized trials are warranted. A recent meta-            more than 6 months after revascularization.63 Dialysis patient survival
analysis showed that use of ultrapure dialysate results in decreased          after CABG is better than after PCI with non-DESs64 and DESs,65 but
markers of inflammation and oxidative stress in HD patients.53 However,       2-year mortality remains high at 44% (vs. 52% for PCI).64 Based on
administration of mixed tocopherols and α-lipoic acid did not influence       three large observational studies,64-67 we recommend the following strat-
biomarkers of inflammation and oxidative stress or the erythropoietic         egy for dialysis patients who do not require acute reperfusion therapy
response in a recent randomized trial in HD patients.54 Because CKD           for STEMI, which would typically be treated with emergent PCI first:
4 patients allocated to the antioxidant synthetic triterpenoid bardoxolone    1. Patients with multivessel CHD (including the left anterior descend-
experienced excessive CVD and especially CHF,55 the beneficial effects            ing coronary artery [LAD]) who are anatomically suitable candidates
of antioxidant treatment strategies in this patient group remains to              for internal mammary artery grafts should undergo CABG surgery;
be proven.                                                                        concomitant ACS additionally favors CABG surgery.
                                                                              2. The CABG survival advantage occurs more than 6 months after
Chronic Kidney Disease–Mineral Bone Disorder                                      revascularization. Patients with limited life expectancy or who are
Recent meta-analyses and the updated KDIGO CKD-MBD guidelines                     concerned about perioperative morbidity (higher with CABG) might
(www.kdigo.org) conclude that calcium-free phosphate binders such                 choose PCI (better outcomes at less than 6 months).
as sevelamer reduce CV events, calcifications, and mortality compared         3. If the left internal mammary graft (to the LAD) is not part of the
with calcium-containing binders (see Chapter 85). Thus, these agents              surgical strategy, CABG likely provides no advantage.
should be preferred for CKD patients with significant life expectancy,            In the general population, advantages of DESs (compared with non-
particularly patients on the transplant waiting list, independent of the      DESs) include lower incidence of in-stent restenosis and improved
presence or absence of calcifications.                                        survival.67 In dialysis patients, reliance on clinical surrogates (e.g., chest
    Because vitamin D treatment is associated with improved survival          pain) leads to underestimation of the true restenosis incidence. The
in dialysis patients, vitamin D insufficiency should be considered.           most complete angiographic follow-up of DESs noted a 22% to 31%
However, results from the Dialysis Outcomes and Practice Patterns             incidence of restenosis with DESs and 24% to 43% with non-DESs.
Study (DOPPS) call into question the survival advantage for HD patients       Clinically silent restenosis is a rationale for surveillance stress imaging
taking vitamin D. In the Paricalcitol Capsules Benefits Renal Failure         (see Fig. 81.12).
Induced Cardiac Morbidity in Subjects With Chronic Kidney Disease
Stage 3/4 (PRIMO) study, 48 weeks of paricalcitol therapy did not alter
LV mass index or improve measures of diastolic dysfunction.56
                                                                              REFERENCES
    The Evaluation of Cinacalcet HCl Therapy to Lower Cardiovascular           1. Go AS, Chertow GM, Fan D, et al. Chronic kidney disease and the risks
Events (EVOLVE) study57 showed that cinacalcet only reduced the risk              of death, cardiovascular events, and hospitalization. N Engl J Med.
for death or major CV events in dialysis patients with moderate-severe            2004;351:1296–1305.
secondary hyperparathyroidism after adjustment for age. In a second            2. Wanner C, Amann K, Shoji T. The heart and vascular system in dialysis.
trial in similar dialysis patients, cinacalcet plus low-dose vitamin D            Lancet. 2016;388:276–284.
sterols tended to attenuate vascular and cardiac valve calcification com-      3. Stenvinkel P, Carrero JJ, Axelsson J, et al. Emerging biomarkers for evaluating
                                                                                  cardiovascular risk in the chronic kidney disease patient: how do new pieces
pared with an exclusively vitamin D sterol–based approach.58
                                                                                  fit into the uremic puzzle? Clin J Am Soc Nephrol. 2008;3:505–521.
    FGF-23, a bone hormone, is cardiotoxic and induces LVH but does            4. Eckardt KU, Gillespie IA, Kronenberg F, et al. High cardiovascular event
not induce vascular calcification.59 In a post-hoc analysis of EVOLVE             rates occur within the first weeks of starting hemodialysis. Kidney Int.
data, treatment of secondary hyperparathyroidism with cinacalcet was              2015;88:1117–1125.
associated with reduced FGF-23 levels and patients with a 30% or 50%           5. U.S. Renal Data System. USRDS 2012 Annual Data Report: Annual Data
drop in FGF-23 levels experienced significantly fewer CV events.60                Report: Atlas of Chronic Kidney Disease & End-State Renal Disease in the
                                                                                  United States. Bethesda, MD: National Institutes of Health, National
Revascularization                                                                 Institute of Diabetes and Digestive and Kidney Diseases; 2012.
CKD is a strong risk factor for death after coronary revascularization         6. Gu Q, Burt VL, Dillon CF, Yoon S. Trends in antihypertensive medication
with percutaneous coronary intervention (PCI) or coronary artery                  use and blood pressure control among United States adults with
                                                                                  hypertension: the National Health And Nutrition Examination Survey,
bypass grafting (CABG).61,62 Coronary revascularization complicated
                                                                                  2001 to 2010. Circulation. 2012;126:2105–2114.
by AKI is associated with excess mortality; operative mortality for non-       7. Kooman JP, Kotanko P, Schols AM, et al. Chronic kidney disease and
ESRD patients who require acute dialysis after cardiac surgery is 44%.            premature ageing. Nat Rev Nephrol. 2014;10:732–742.
    Observational studies suggest lower mortality in CKD (including            8. Vanholder R, Massy Z, Argiles A, et al. Chronic kidney disease as cause of
dialysis) patients undergoing coronary revascularization compared with            cardiovascular morbidity and mortality. Nephrol Dial Transplant. 2005;20:
no revascularization. A post hoc analysis of CKD patients in the Fast             1048–1056.
 9. Stenvinkel P, Gillespie IA, Tunks J, et al. Inflammation Modifies the             29. Leither MD, Shroff GR, Ding S, et al. Long-term survival of dialysis
    Paradoxical Association between Body Mass Index and Mortality in                      patients with bacterial endocarditis undergoing valvular replacement
    Hemodialysis Patients. J Am Soc Nephrol. 2016;27:1479–1486.                           surgery in the United States. Circulation. 2013;128:344–351.
10. Tang WH, Hazen SL. The contributory role of gut microbiota in                     30. Yamada S, Ishii H, Takahashi H, et al. Prognostic value of reduced left
    cardiovascular disease. J Clin Invest. 2014;124:4204–4211.                            ventricular ejection fraction at start of hemodialysis therapy on
11. Chen Z, Qureshi AR, Ripsweden J, et al. Vertebral bone density                        cardiovascular and all-cause mortality in end-stage renal disease patients.
    associates with coronary artery calcification and is an independent                   Clin J Am Soc Nephrol. 2010;5:1793–1798.
    predictor of poor outcome in end-stage renal disease patients. Bone.              31. Foley RN, Gilbertson DT, Murray T, Collins AJ. Long interdialytic interval
    2016;92:50–57.                                                                        and mortality among patients receiving hemodialysis. N Engl J Med.
12. Shroff GR, Li S, Herzog CA. Trends in Mortality Following Acute                       2011;365:1099–1107.
    Myocardial Infarction Among Dialysis Patients in the United States Over           32. Pun PH, Lehrich RW, Honeycutt EF, et al. Modifiable risk factors
    15 Years. J Am Heart Assoc. 2015;4:e002460.                                           associated with sudden cardiac arrest within hemodialysis clinics. Kidney
13. Szummer K, Lundman P, Jacobson SH, et al. Relation between renal                      Int. 2011;79:218–227.
    function, presentation, use of therapies and in-hospital complications in         33. Peer Kidney Care Initiative. Peer Report: Dialysis Care and Outcomes in
    acute coronary syndrome: data from the SWEDEHEART register. J Intern                  The United States. Minneapolis, MN: Chronic Disease Research Group;
    Med. 2010;268:40–49.                                                                  2014:Page9.
14. Herzog CA, Littrell K, Arko C, et al. Clinical characteristics of dialysis        34. Charytan DM, Patrick AR, Liu J, et al. Trends in the use and outcomes of
    patients with acute myocardial infarction in the United States: a                     implantable cardioverter-defibrillators in patients undergoing dialysis in
    collaborative project of the United States Renal Data System and the                  the United States. Am J Kidney Dis. 2011;58:409–417.
    National Registry of Myocardial Infarction. Circulation. 2007;116:                35. Chawla LS, Herzog CA, Costanzo MR, et al. Proposal for a functional
    1465–1472.                                                                            classification system of heart failure in patients with end-stage renal
15. Shroff GR, Frederick PD, Herzog CA. Renal failure and acute myocardial                disease: proceedings of the acute dialysis quality initiative (ADQI) XI
    infarction: clinical characteristics in patients with advanced chronic                workgroup. J Am Coll Cardiol. 2014;63:1246–1252.
    kidney disease, on dialysis, and without chronic kidney disease. A                36. Hickson LJ, Negrotto SM, Onuigbo M, et al. Echocardiography Criteria
    collaborative project of the United States Renal Data System/National                 for Structural Heart Disease in Patients With End-Stage Renal Disease
    Institutes of Health and the National Registry of Myocardial Infarction.              Initiating Hemodialysis. J Am Coll Cardiol. 2016;67:1173–1182.
    Am Heart J. 2012;163:399–406.                                                     37. Wang LW, Fahim MA, Hayen A, et al. Cardiac testing for coronary artery
16. Garimella PS, Hart PD, O’Hare A, et al. Peripheral artery disease and                 disease in potential kidney transplant recipients. Cochrane Database Syst
    CKD: a focus on peripheral artery disease as a critical component of                  Rev. 2011;(12):CD008691.
    CKD care. Am J Kidney Dis. 2012;60:641–654.                                       38. Lentine KL, Costa SP, Weir MR, et al. Cardiac disease evaluation and
17. Herzog CA, Asinger RW, Berger AK, et al. Cardiovascular disease in                    management among kidney and liver transplantation candidates: a
    chronic kidney disease. A clinical update from Kidney Disease: Improving              scientific statement from the American Heart Association and the
    Global Outcomes KDIGO). Kidney Int. 2011;80:572–586.                                  American College of Cardiology Foundation. J Am Coll Cardiol. 2012;
18. Shima H, Mori T, Ooi M, et al. Silent Cerebral Microbleeds and                        60:434–480.
    Longitudinal Risk of Renal and Cardiovascular Events in Patients with             39. Kumar N, Baker CS, Chan K, et al. Cardiac survival after pre-emptive
    CKD. Clin J Am Soc Nephrol. 2016;11:1557–1565.                                        coronary angiography in transplant patients and those awaiting
19. Murray AM, Seliger S, Lakshminarayan K, et al. Incidence of Stroke                    transplantation. Clin J Am Soc Nephrol. 2011;6:1912–1919.
    Before and After Dialysis Initiation in Older Patients. J Am Soc Nephrol.         40. James MT, Ghali WA, Knudtson ML, et al. Associations between acute
    2013;24:1166–1173.                                                                    kidney injury and cardiovascular and renal outcomes after coronary
20. Goldstein BA, Arce CM, Hlatky MA, et al. Trends in the incidence of                   angiography. Circulation. 2011;123:409–416.
    atrial fibrillation in older patients initiating dialysis in the United States.   41. Winther S, Svensson M, Jorgensen HS, et al. Diagnostic Performance of
    Circulation. 2012;126:2293–2301.                                                      Coronary CT Angiography and Myocardial Perfusion Imaging in Kidney
21. Bansal N, Fan D, Hsu CY, et al. Incident Atrial Fibrillation and Risk of              Transplantation Candidates. JACC Cardiovasc Imaging. 2015;8:553–562.
    End-Stage Renal Disease in Adults with Chronic Kidney Disease.                    42. Apple FS, Murakami MM, Pearce LA, Herzog CA. Predictive value of
    Circulation. 2013;127:569–574.                                                        cardiac troponin I and T for subsequent death in end-stage renal disease.
22. Konigsbrugge O, Posch F, Antlanger M, et al. Prevalence of Atrial                     Circulation. 2002;106:2941–2945.
    Fibrillation and Antithrombotic Therapy in Hemodialysis Patients:                 43. Breidthardt T, Burton JO, Odudu A, et al. Troponin T for the detection of
    Cross-Sectional Results of the Vienna InVestigation of AtriaL Fibrillation            dialysis-induced myocardial stunning in hemodialysis patients. Clin J Am
    and Thromboembolism in Patients on HemoDIalysis (VIVALDI). PLoS                       Soc Nephrol. 2012;7:1285–1292.
    ONE. 2017;12(1):e0169400.                                                         44. Sun J, Axelsson J, Machowska A, et al. Biomarkers of Cardiovascular
23. Hart RG, Pearce LA, Asinger RW, Herzog CA. Warfarin in atrial                         Disease and Mortality Risk in Patients with Advanced CKD. Clin J Am
    fibrillation patients with moderate chronic kidney disease. Clin J Am Soc             Soc Nephrol. 2016;11:1163–1172.
    Nephrol. 2011;6:2599–2604.                                                        45. Sandoval Y, Herzog CA, Love SA, et al. Prognostic Value of Serial Changes
24. Hart RG, Eikelboom JW, Ingram AJ, Herzog CA. Anticoagulants in atrial                 in High-Sensitivity Cardiac Troponin I and T over 3 Months Using
    fibrillation patients with chronic kidney disease. Nat Rev Nephrol. 2012;8:           Reference Change Values in Hemodialysis Patients. Clin Chem. 2016;62:
    569–578.                                                                              631–638.
25. Whalley GA, Marwick TH, Doughty RN, et al. Effect of Early Initiation             46. Anker SD, Gillespie IA, Eckardt KU, et al. Development and validation of
    of Dialysis on Cardiac Structure and Function: Results From the Echo                  cardiovascular risk scores for haemodialysis patients. Int J Cardiol.
    Substudy of the IDEAL Trial. Am J Kidney Dis. 2013;61:262–270.                        2016;216:68–77.
26. Park M, Hsu CY, Li Y, et al. Associations between kidney function and             47. ACCORD Study Group: Gerstein HC, Miller ME, Genuth S, et al.
    subclinical cardiac abnormalities in CKD. J Am Soc Nephrol. 2012;23:                  Long-term effects of intensive glucose lowering on cardiovascular
    1725–1734.                                                                            outcomes. N Engl J Med. 2011;364:818–828.
27. KDOQI Workgroup. K/DOQI clinical practice guidelines for                          48. Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL
    cardiovascular disease in dialysis patients. Am J Kidney Dis. 2005;45                 cholesterol with simvastatin plus ezetimibe in patients with chronic
    (4 suppl 3):S1–S153.                                                                  kidney disease (Study of Heart and Renal Protection): a randomised
28. Sharma A, Gilbertson DT, Herzog CA. Survival of kidney transplantation                placebo-controlled trial. Lancet. 2011;377:2181–2192.
    patients in the United States after cardiac valve replacement. Circulation.       49. Chen Z, Qureshi AR, Parini P, et al. Does statins promote vascular
    2010;121:2733–2739.                                                                   calcification in chronic kidney disease? Eur J Clin Invest. 2016;47:137–148.
50. FHN Trial Group, Chertow GM, Levin NW, et al. In-center hemodialysis           60. Moe SM, Chertow GM, Parfrey PS, et al. Cinacalcet, Fibroblast Growth
    six times per week versus three times per week. N Engl J Med. 2010;363:            Factor-23, and Cardiovascular Disease in Hemodialysis: The Evaluation
    2287–2300.                                                                         of Cinacalcet HCl Therapy to Lower Cardiovascular Events (EVOLVE)
51. Jefferies HJ, Virk B, Schiller B, et al. Frequent hemodialysis schedules are       Trial. Circulation. 2015;132:27–39.
    associated with reduced levels of dialysis-induced cardiac injury              61. Best PJM, Lennon R, Ting HH, et al. The impact of renal insufficiency on
    (myocardial stunning). Clin J Am Soc Nephrol. 2011;6:1326–1332.                    clinical outcomes in patients undergoing percutaneous coronary
52. Zickler D, Schindler R, Willy K, et al. Medium Cut-Off (MCO)                       interventions. J Am Coll Cardiol. 2002;39:1113–1119.
    Membranes Reduce Inflammation in Chronic Dialysis Patients-A                   62. Cooper WA, O’Brien SM, Thourani VH, et al. Impact of renal
    Randomized Controlled Clinical Trial. PLoS ONE. 2017;12(1):e0169024.               dysfunction on outcomes of coronary artery bypass surgery: results from
53. Susantitaphong P, Riella C, Jaber BL. Effect of ultrapure dialysate on             the Society of Thoracic Surgeons National Adult Cardiac Database.
    markers of inflammation, oxidative stress, nutrition and anemia                    Circulation. 2006;113:1063–1070.
    parameters: a meta-analysis. Nephrol Dial Transplant. 2013;28:                 63. Charytan DM, Li S, Liu J, Herzog CA. Risks of death and end-stage renal
    438–446.                                                                           disease after surgical compared with percutaneous coronary
54. Himmelfarb J, Ikizler TA, Ellis C, et al. Provision of antioxidant therapy         revascularization in elderly patients with chronic kidney disease.
    in hemodialysis (PATH): a randomized clinical trial. J Am Soc Nephrol.             Circulation. 2012;126(11 suppl 1):S164–S169.
    2014;25:623–633.                                                               64. Shroff GR, Solid CA, Herzog CA. Long-term survival and repeat coronary
55. de Zeeuw D, Akizawa T, Audhya P, et al. Bardoxolone methyl in type 2               revascularization in dialysis patients after surgical and percutaneous
    diabetes and stage 4 chronic kidney disease. N Engl J Med. 2013;369:               coronary revascularization with drug-eluting and bare metal stents in the
    2492–2503.                                                                         United States. Circulation. 2013;127:1861–1869.
56. Thadhani R, Appelbaum E, Pritchett Y, et al. Vitamin D therapy and             65. Chang TI, Shilane D, Kazi DS, et al. Multivessel coronary artery bypass
    cardiac structure and function in patients with chronic kidney disease:            grafting versus percutaneous coronary intervention in ESRD. J Am Soc
    the PRIMO randomized controlled trial. JAMA. 2012;307:674–684.                     Nephrol. 2012;23:2042–2049.
57. EVOLVE Trial Investigators, Chertow GM, Block GA, et al. Effect of             66. Shroff GR, Solid CA, Herzog CA. Impact of acute coronary syndromes
    cinacalcet on cardiovascular disease in patients undergoing dialysis.              on survival of dialysis patients following surgical or percutaneous
    N Engl J Med. 2012;367:2482–2494.                                                  coronary revascularization in the United States. Eur Heart J Acute
58. Raggi P, Chertow GM, Torres PU, et al. The ADVANCE study: a                        Cardiovasc Care. 2016;5:205–213.
    randomized study to evaluate the effects of cinacalcet plus low-dose           67. Shroff GR, Herzog CA. Coronary Revascularization in Patients with
    vitamin D on vascular calcification in patients on hemodialysis. Nephrol           CKD Stage 5D: Pragmatic Considerations. J Am Soc Nephrol. 2016;27:
    Dial Transplant. 2011;26:1327–1339.                                                3521–3529.
59. Scialla JJ, Lau WL, Reilly MP, et al. Fibroblast growth factor 23 is not
    associated with and does not induce arterial calcification. Kidney Int.
    2013;83:1159–1168.
    SELF-ASSESSMENT
    QUESTIONS
1. Which of the following is not an established risk factor for cardio-
   vascular disease in patients with advanced chronic kidney disease?
   A. Inflammation
   B. Smoking
   C. Dyslipidemia
   D. Hyperhomocysteinemia
   E. Diabetes
2. What is not true about cardiovascular complications in end-stage
   renal disease patients?
   A. Left ventricular hypertrophy is present in about 75% of the
      patients.
   B. In dialysis patients the incidence of atrial fibrillation is about
      15% per year.
   C. Low levels of cholesterol predict death.
   D. In dialysis patients, extensive vascular, especially coronary artery,
      calcification can occur even at young ages.
   E. The calcific aortic stenosis progression rate is only slightly higher
      in dialysis patients compared to the general population.
3. What is the most common cause of death in dialysis patients?
   A. Congestive heart failure
   B. Infection
   C. Arrhythmic death
   D. Stroke
   E. Valvular heart disease
4. Which drug has been shown to reduce mortality in dialysis patients
   with dilated cardiomyopathy and systolic heart failure?
   A. Atorvastatin
   B. Digoxin
   C. Carvedilol
   D. Metoprolol
   E. Atenolol
Anemia is an almost universal complication of chronic kidney disease           tive β subunit. The production of HIF-1α and HIF-2α is largely inde-
(CKD). It contributes considerably to reduced quality of life of patients      pendent of oxygen, but their degradation is related to cellular oxygen
with CKD and has been associated with a number of adverse clinical             concentrations. Hydroxylation of specific prolyl and asparagyl residues
outcomes. Before the availability of recombinant human erythropoietin          of HIF-α, for which molecular oxygen is required as a substrate, deter-
(rHuEPO, or epoetin), patients on dialysis frequently required blood           mines proteasomal destruction of HIF and inhibits its transcriptional
transfusions, exposing them to the risks of iron overload, transmission        activity. Apart from EPO, several hundred HIF target genes have been
of viral hepatitis, and human leukocyte antigen (HLA) sensitization,           identified. HIF-2, rather than HIF-1, is the transcription factor primarily
which reduced the chances of successful transplantation. The advent            responsible for the regulation of EPO production.5,6
of rHuEPO in the late 1980s changed this situation completely. The                 The role of renal EPO production in the pathogenesis of renal anemia
ability to correct anemia has had consequences beyond simply an                is supported by the particularly severe anemia in anephric individuals.
improvement in general fatigue and reduced physical capacity, to impact        However, the mechanisms impairing renal EPO production in diseased
on a broad spectrum of physiologic functions. Thus there is a strong           kidneys remain poorly understood. The production capacity for EPO
rationale for managing anemia in CKD patients, and yet the optimal             remains significant, even in end-stage renal disease. Thus patients with
treatment strategies are still incompletely defined. Apart from therapy        anemia and CKD can respond with a significant increase in EPO pro-
with erythropoiesis-stimulating agents (ESAs), iron replacement is             duction to an additional hypoxic stimulus.1 The main problem therefore
essential for anemia management. It is important to note that CKD              appears to be a failure of EPO production to increase in response to
patients on dialysis require target thresholds of iron parameters different    chronically reduced hemoglobin (Hb) concentrations. In line with this
from those for normal individuals to ensure optimal rates of red blood         view, endogenous EPO production can be induced in CKD patients by
cell (RBC) production. The costs of anemia management are consider-            pharmacologic inhibition of HIF degradation (see later discussion).
able, and it has become apparent that full anemia correction may cause             EPO is a glycoprotein hormone consisting of a 165–amino acid
harm; therefore a rational and careful consideration of the risks and          protein backbone and four complex, heavily sialylated carbohydrate
benefits is mandatory.                                                         chains.1 The latter are essential for the biologic activity of EPO in vivo
                                                                               because partially or completely deglycosylated EPO is rapidly cleared
                                                                               from the circulation. This is why rHuEPO has to be manufactured in
PATHOGENESIS                                                                   mammalian cells; bacteria lack the capacity to glycosylate recombinant
Renal anemia is typically an isolated normochromic, normocytic anemia          proteins.
with no leukopenia or thrombocytopenia. Both RBC life span and the                 EPO stimulates RBC production by binding to homodimeric EPO
rate of RBC production are reduced, but the latter is more important.          receptors, which are primarily located on early erythroid progenitor
The normal bone marrow has considerable capacity to increase the rate          cells, the burst-forming units erythroid (BFU-e) and the colony-forming
of erythropoiesis, and compensation normally could be easily made              units erythroid (CFU-e). Binding of EPO to its receptors salvages these
for the reduction in erythrocyte life observed in association with CKD.        progenitor cells and the subsequent earliest erythroblast generation
However, this EPO-induced compensatory increase in erythrocyte pro-            from apoptosis, thereby permitting cell division and maturation into
duction is impaired in CKD. Serum EPO levels remain within the normal          RBCs.7 Inhibition of RBC production by yet unknown uremic inhibi-
range and fail to show the inverse exponential relationship with blood         tors of erythropoiesis may contribute to the pathogenesis of renal
oxygen content characteristic of other types of anemia. EPO is normally        anemia, and dialysis per se can improve renal anemia and the efficacy
produced by interstitial fibroblasts in the renal cortex, in close proximity   of ESAs. Moreover, the interindividual dose requirements for ESAs
to tubular epithelial cells and peritubular capillaries.1,2 In addition,       vary significantly among CKD patients, and the average weekly dose is
hepatocytes and perisinusoidal, or Ito, cells in the liver can produce         much higher than estimated production rates of endogenous EPO in
EPO (Fig. 82.1). Hepatic EPO production dominates during fetal and             healthy individuals. An alternative view to the accumulation of inhibi-
early postnatal life but does not compensate for the loss of renal pro-        tors of erythropoiesis in CKD is that in many patients there is overlap
duction in adult organisms. Subtle changes in blood oxygen content             between renal anemia and the anemia of chronic disease, which is
induced by anemia, reduced environmental oxygen concentrations, and            characterized by inhibition of EPO production and EPO efficacy, as
high altitude stimulate the secretion of EPO through a widespread              well as by reduced iron availability, mediated through inflammatory
system of oxygen-dependent gene expression.2-4 Central to this process         cytokines.8 The hepatic release of hepcidin, the key regulator of iron
is a family of hypoxia-inducible transcription factors (HIFs). The two         metabolism, is upregulated in states of inflammation; it simultaneously
most important members of this family, HIF-1 and HIF-2, are composed           blocks iron absorption from the gut and promotes iron sequestration in
of an oxygen-regulated α subunit (HIF-1α or HIF-2α) and a constitu-            macrophages.9
958
                                                   downloaded from www.medicalbr.com
                                                               CHAPTER 82        Anemia in Chronic Kidney Disease                                                959
                                                                                   Hemoglobin (g/dl)
                                                                                                       15
13
                                                                                                       11
                                  Blood oxygen
                                     content                                                           9                                     95th percentile
                                                                                                                                             Median
                                                                                                       7                                     5th percentile
                                                       Tissue perfusion
0 30 60 90 120 150
                                                                                  B
                                                                                                       17                                  Females
                                                                                   Hemoglobin (g/dl)
                                                                                                       15
                               Hypoxia-inducible
                              factors (HIF-1, HIF-2)
                                                                                                       13
11
                                                                                                       9                                     95th percentile
                                                                                                                                             Median
                            EPO/other HIF target genes                                                 7                                     5th percentile
                              → increases in oxygen
                            supply or hypoxia tolerance
                                                                                                            0     30        60        90        120        150
Fig. 82.1 Feedback control of erythropoiesis. EPO, Erythropoietin.
                                                                                                                 Estimated GFR (ml/min per 1.73 m2)
that thresholds for the diagnosis of anemia and evaluation of the causes     additional causes. Because there is significant variability in the degree
should not be interpreted as being thresholds for treatment of anemia.14     of anemia in relation to the impairment in renal function, no simple
    In addition to the Hb value, the evaluation of anemia in CKD patients    diagnostic criteria can be applied. Causes of anemia other than EPO
should include a complete blood count with RBC indices (mean cor-            deficiency should be considered when (1) the severity of anemia is
puscular Hb concentration [MCHC], mean corpuscular volume [MCV]),            disproportionate to the impairment of renal function, (2) there is evi-
white blood cell count (including differential), and platelet count.         dence of iron deficiency, or (3) there is evidence of leukopenia or
Although renal anemia is typically normochromic and normocytic,              thrombocytopenia. Concomitant conditions such as sickle cell disease
deficiency of vitamin B12 or folate may lead to macrocytosis, whereas        may exacerbate the anemia, as can drug therapy. For example, inhibitors
iron deficiency or inherited disorders of Hb formation (such as thalas-      of the renin-angiotensin system may reduce Hb levels by (1) direct
semia) may produce microcytosis. Macrocytosis with leukopenia or             effects of angiotensin II (Ang II) on erythroid progenitor cells,15 (2)
thrombocytopenia suggests a generalized disorder of hematopoiesis            accumulation of N-acetyl-seryl-lysyl-proline (Ac-SDKP), an endogenous
caused by toxins, nutritional deficit, or myelodysplasia. Hypochromia        inhibitor of erythropoiesis in patients treated with angiotensin-converting
probably reflects iron-deficient erythropoiesis. An absolute reticulocyte    enzyme (ACE) inhibitors,16 and (3) reduction of endogenous EPO
count, which normally ranges between 40,000 and 50,000 cells/μl of           production, possibly because of the hemodynamic effects of Ang II
blood, is a useful marker of erythropoietic activity.                        inhibition. Myelosuppressive effects of immunosuppressants may further
    Iron status tests should be performed to assess the level of iron in     contribute to anemia.17 The measurement of serum EPO concentrations
tissue stores or the adequacy of iron supply for erythropoiesis. Although    is usually not helpful in the diagnosis of renal anemia because there is
serum ferritin is the only available marker of storage iron, several tests   relative rather than absolute deficiency, with a wide range of EPO con-
reflect the adequacy of iron for erythropoiesis, including transferrin       centrations for a given Hb concentration that extends far beyond the
saturation (TSAT), the percentage of hypochromic red blood cells             normal range of EPO levels in healthy, nonanemic individuals. Abnor-
(PHRC), the reticulocyte hemoglobin content (CHr), the MCV, and              malities of other laboratory parameters should be looked for, such as
the MCHC. Storage time of the blood sample may elevate PHRC, and             a very low MCV or MCHC, a high MCV, or an abnormal leukocyte or
MCV and MCHC are below the normal range only after long-standing             platelet count, and further tests should be performed as indicated to
iron deficiency; in clinical practice, TSAT remains the most frequently      explore these potential contributory causes (see earlier). However, when
used parameter.                                                              there are no such pointers to other confounding causes of anemia, and
    It is important to identify anemia in CKD patients because it may        iron deficiency has been excluded, a trial with rHuEPO or its derivatives
signify nutritional deficits, systemic illness, or other conditions that     is warranted, even when the eGFR is only moderately reduced.
warrant attention. Moreover, even at modest degrees, anemia reflects
an independent risk factor for hospitalizations, cardiovascular disease
(CVD), and mortality.12 The diagnosis of renal anemia, that is, anemia
                                                                             CLINICAL MANIFESTATIONS
caused by CKD, requires careful judgment of the degree of anemia in          In the early clinical trials of rHuEPO performed in the late 1980s, the
relation to the degree of renal impairment and exclusion of other or         mean baseline Hb concentration was about 6 to 7 g/dl, and this
progressively increased to about 11 or 12 g/dl after treatment. Patients     studies have indicated increased risks associated with attempts to com-
subjectively felt much better, with reduced fatigue, increased energy        pletely correct anemia. In particular, no survival benefit is evident at a
levels, and enhanced physical capacity, and there were also objective        higher level of anemia correction,20-23 and attempts to normalize Hb
improvements in cardiorespiratory function.18 Thus it is now clear that      concentrations have shown various risks, including increased rates of
many of the symptoms previously attributed to the “uremic syndrome”          thromboembolic events, strokes, and possibly death. Thus there is a
may have been caused by severe anemia associated with CKD (Boxes             possible tradeoff among improved quality of life, reduced transfusion
82.1 and 82.2). Although the avoidance of blood transfusions and             requirements, and risk for harm (see discussion later), and a target Hb
improvement in quality of life are obvious early changes, there are also     level of above 13 g/dl should be avoided.12,14
possible effects on the cardiovascular system (see Box 82.1). The physi-
ologic consequences of long-standing anemia are an increase in cardiac       TREATMENT
output and a reduction in peripheral vascular resistance. Anemia is
associated with the development of left ventricular hypertrophy in CKD       Erythropoiesis-Stimulating Agents
patients and is thought to exacerbate left ventricular dilation. Sustained   Epoetin Therapy
correction of severe anemia in CKD patients tends to reverse most of         Manufacture of rHuEPO is achieved by gene transfer into a suitable
these CV abnormalities, with the notable exception of left ventricular       mammalian cell line such as Chinese hamster ovary (CHO) cells. The
dilation (although anemia correction may prevent further dilation in         early clinical trials of rHuEPO were conducted with both EPO alfa and
some patients19). Other effects of anemia correction reported in clinical    EPO beta, both produced in CHO cells. Like the endogenous hormone,
trials include improvements in quality of life, cognitive function, sleep    rHuEPO consists of a 165–amino acid backbone with one O-linked
patterns, nutrition, sexual function, menstrual regularity, immune           and three N-linked glycosylation chains. Invariably, however, there are
responsiveness, and platelet function. The majority of these trials,         some differences in the glycosylation pattern among different prepara-
however, were not placebo-controlled, so the spectrum and extent of          tions of rHuEPO and the endogenous hormone. “Biosimilar” EPO
possible benefits remain uncertain.                                          preparations have been licensed in Europe after demonstration of their
    Over the years, there has been considerable debate about the optimal     efficacy and safety in an abbreviated trial program.24 Many other “copy”
target range of Hb in CKD patients. A presumed improvement in quality        EPOs are available in several parts of the world, which are not neces-
of life and expectations of positive effects on CV function and renal        sarily produced to the same regulatory standards.
disease progression with increasing Hb concentrations led to sugges-             Before 1998, EPO alfa in Europe was formulated with human serum
tions of a level above 10 to 11 g/dl in all CKD patients,12,13 but several   albumin, but this was replaced with polysorbate 80. EPO beta is for-
                                                                             mulated with polysorbate 20, along with urea, calcium chloride, and
                                                                             five amino acids as excipients. The importance of the formulation of
 BOX 82.1   Cardiovascular Effects Resulting                                 the EPO products was highlighted in 2002 with an upsurge in cases of
 from Anemia Correction                                                      antibody-mediated pure red cell aplasia (PRCA) in association with
                                                                             the subcutaneous use of EPO alfa after its change of formulation. Patients
    Reduction in high cardiac output                                         affected by this complication develop neutralizing antibodies against
    Reduced stroke volume                                                    both rHuEPO and the endogenous hormone, which result in severe
    Reduced heart rate                                                       anemia and transfusion dependence.25 The cause of this serious com-
    Increase in peripheral vascular resistance                               plication in which there is a break in B cell tolerance remains obscure,
    Reduction in anginal episodes                                            although it seems likely that factors such as a breach of the cold storage
    Reduction in myocardial ischemia                                         chain are relevant, and the subcutaneous application route is a prereq-
    Regression of left ventricular hypertrophy                               uisite; circumstantial evidence also suggests that rubber stoppers of
    Stabilization of left ventricular dilation                               prefilled syringes used in one of the albumin-free EPO alfa formulations
    Increase in whole blood viscosity                                        may have released organic compounds that acted as immunologic adju-
                                                                             vants.26 Although this unfortunate combination of adverse factors was
                                                                             specific for one compound, the development of neutralizing anti-EPO
 BOX 82.2             Other Effects of Anemia                                antibodies was subsequently also observed during a clinical trial with
 Correction                                                                  an epoetin alfa biosimilar27 and a low rate of PRCA also occurs with
                                                                             EPO beta and darbepoetin alfa.
  Beneficial
                                                                                 The EPOs are administered either intravenously or subcutaneously.
    Reduced blood transfusions
                                                                             The bioavailability after intraperitoneal administration (in peritoneal
    Increased quality of life
                                                                             dialysis [PD] patients) is too low. The earliest clinical trials of EPO
    Increased exercise capacity
                                                                             used intravenous injections two or three times per week. This was
    Improved cognitive function
                                                                             partly because of the short half-life of EPO (6 to 8 hours after intra-
    Improved sleep patterns
                                                                             venous administration)28 and partly because of the convenience for
    Improved immune function
                                                                             the patient on dialysis. With use of this regimen, 90% of patients show
    Improved muscle function
                                                                             a significant increase in Hb concentration. Good iron management is
    Improved depression
                                                                             pivotal for the success of EPO therapy (see later discussion). Although
    Improved nutrition
                                                                             the bioavailability of subcutaneous EPO is 20% to 30%, the prolonged
    Improved platelet function
                                                                             half-life after subcutaneous administration compared with intravenous
  Adverse                                                                    administration allows less frequent injections. Furthermore, the dose
    Hypertension                                                             required to achieve the same Hb response is about 30% lower with
    Vascular access thrombosis                                               subcutaneous than with intravenous administration.28 There appears
    Increased risk for stroke                                                to be little difference in efficacy among the thigh, arm, or abdomen
                                                                             as injection sites.
                                                                               who are not on iron and ESA therapy and in whom an increase in Hb
Iron Management                                                                concentration is desired and in patients on ESA therapy in whom an
Iron is an essential ingredient for heme synthesis, and adequate amounts       increase in Hb concentration or a decrease in ESA dose is required
of this mineral are required for the manufacture of new RBCs. Thus,            when TSAT is 30% or lower and ferritin level is 500 ng/ml or lower14
under enhanced erythropoietic stimulation, greater amounts of iron             (see Table 82.3). Levels of ferritin above this threshold usually do not
are used, and many CKD patients (particularly those on HD) have                confer any clinical advantage and may exacerbate iron toxicity. The
inadequate amounts of available iron to satisfy the increased demands          optimal TSAT is above 20% to 30% to ensure a readily available supply
of the bone marrow.43 Even before the introduction of ESA therapy,             of iron to the bone marrow. No upper limits of ferritin or TSAT were
many CKD patients were in negative iron balance as a result of poor            specified in the KDIGO anemia guidelines, largely because there is no
dietary intake, poor appetite, and increased iron losses from occult and       robust evidence to determine a threshold beyond which harm or loss
overt blood losses (see Chapter 86). Losses in HD patients are up to 5         of efficacy supervenes. However, until more informative data become
or 6 mg/day, compared with 1 mg in healthy individuals, and this may           available, the treating nephrologist would be well advised to exercise
easily exceed the absorption capacity of the gastrointestinal tract, par-      caution in administering intravenous iron to patients with ferritin levels
ticularly when there is any underlying inflammation. Iron absorption           above 800 ng/ml or TSAT above 30%. Several studies support maintain-
capacity in patients with CKD is considerably lower than in nonuremic          ing the percentage of hypochromic RBCs at less than 6% and the CHr
individuals, particularly in the presence of systemic inflammation, and        at greater than 29 pg/cell. Other measures of iron status, such as serum
this is mediated by hepcidin upregulation.9 For this reason also, tradi-       transferrin receptor levels and erythrocyte zinc protoporphyrin levels,
tional oral iron therapy (e.g., with ferrous sulfate) is ineffective in many   are mainly research tools.
CKD patients, and parenteral iron administration is required, particularly         Oral iron is generally poorly absorbed in uremic individuals, and
in those receiving HD.43 However, recently, a newer preparation of oral        there is a high incidence of gastrointestinal side effects. Intramuscular
iron (ferric citrate) that shows greater absorption of iron from the gut       administration of iron is not recommended in CKD, given the enhanced
has become available in the United States and Japan, and the role that         bleeding tendency, the pain of the injection, and the potential for brown-
ferric citrate may play in iron management in CKD awaits further               ish discoloration of the skin. Thus intravenous administration of iron
elucidation.                                                                   has become the standard of care for many CKD patients, particularly
    An inadequate supply of iron to the bone marrow may be caused              those receiving HD.43 Several intravenous iron preparations are available
by an absolute or a functional iron deficiency.43 Absolute iron deficiency     worldwide, including iron dextran, iron sucrose, iron gluconate, and
occurs when there are low whole-body iron stores, as indicated by a            the newer iron preparations ferric carboxymaltose, ferumoxytol, and
serum ferritin level less than 30 ng/ml. Functional iron deficiency occurs     iron isomaltoside 1000. The last three iron preparations allow higher
when there is ample or even increased storage iron but the iron stores         doses of intravenous iron to be administered more rapidly, without the
fail to release iron rapidly enough to satisfy the demands of the bone         need for a test dose. All of the iron preparations contain elemental iron
marrow. Several markers of iron status are available, but none of them         surrounded by a carbohydrate shell, which allows them to be injected
is ideal (Table 82.3). Serum ferritin is a marker of storage iron but is       intravenously. The lability of iron release from these preparations varies,
spuriously raised in inflammatory conditions and liver disease. TSAT           with iron dextran being the most stable and iron gluconate being the
is a function of the circulating serum iron in relation to the total iron-     least stable. Iron is released from these compounds to plasma transferrin
binding capacity and is often regarded as a better measure of available        and other iron-binding proteins and is then taken up by the reticulo-
iron; however, levels can be highly fluctuant because of significant diurnal   endothelial system.
variation in the measurement of serum iron.43 The percentage of hypo-              In HD patients, it is easy and practical to give low doses of intra-
chromic RBCs and the CHr are RBC and reticulocyte parameters,                  venous iron (e.g., 10 to 20 mg) at every dialysis session or, alternatively,
respectively, that are indirect measures of how much iron is being             100 mg weekly. In PD and nondialysis CKD patients, however, such
incorporated into the newly developing or mature RBC. No one measure           low-dose regimens are impractical and larger doses may be administered.
of iron status is usually adequate to exclude iron deficiency, and the         The more stable the iron preparation, the larger the dose administration
recommended levels for these measures are based on limited scientific          rate that can be used. For example, 1 g of iron dextran may be given
evidence. Functional iron deficiency is usually diagnosed when there           by intravenous infusion, whereas the maximum recommended dose of
is a normal or increased ferritin level and a reduced TSAT (<20%) or           iron gluconate is 125 mg. The usual dose of ferumoxytol is 510 mg,
increased hypochromic RBCs (>10%). The KDIGO guidelines on renal               whereas up to 1 g of ferric carboxymaltose or iron isomaltoside 1000
anemia management suggest a trial of iron in CKD patients with anemia          may be given as a single administration. All intravenous iron prepara-
                                                                               tions carry a risk for immediate hypersensitivity reactions, which may
                                                                               be characterized by hypotension, dizziness, and nausea. These reactions
                                                                               are usually short-lived and caused by too large a dose given in too short
 TABLE 82.3                Recommended Iron Status                             a time. Iron dextran also carries the risk for acute anaphylactic reactions
 Levels in CKD                                                                 because of preformed dextran antibodies, although this was largely a
  Test                                         Recommended Range               problem with the high molecular weight intravenous iron dextran
                                                                               preparations, which have now been withdrawn from the market. Other,
  Serum ferritin                               100-500 μg/l (CKD)
                                                                               longer term concerns about intravenous administration of iron include
                                               200-500 μg/l (HD)
                                                                               the potential for increased susceptibility to infections and oxidative
  Transferrin saturation                       20%-40%                         stress. Much of the scientific evidence for this has been generated from
  Hypochromic red cells                        <10%                            in vitro experiments, the clinical significance of which is unclear.43
  Reticulocyte hemoglobin content (CHr)        >29 pg/cell                         Despite these concerns, the lack of efficacy of traditional oral iron
  Serum transferrin receptor                   Not established                 preparations in HD patients has resulted in the widespread use of intra-
                                                                               venous iron to correct the negative iron balance in this patient popula-
  Erythrocyte zinc protoporphyrin              Not established
                                                                               tion. In general, the greater the use of intravenous iron, the lower is
CKD, Chronic kidney disease; HD, hemodialysis.                                 the dose requirement of ESA therapy; however, the optimum balance
between intravenous iron and ESA therapy remains unknown. This                 17. Winkelmayer WC, Kewalramani R, Rutstein M, et al.
gap in knowledge may be partly addressed by the ongoing PIVOTAL                    Pharmacoepidemiology of anemia in kidney transplant recipients.
RCT, which is comparing a more liberal use of intravenous iron versus              J Am Soc Nephrol. 2004;15:347–352.
a restrictive policy in 2141 HD patients recruited across 50 sites in the      18. Macdougall IC, Lewis NP, Saunders MJ, et al. Long-term
                                                                                   cardiorespiratory effects of amelioration of renal anaemia by
United Kingdom.44
                                                                                   erythropoietin. Lancet. 1990;335:489–493.
    In nondialysis patients, both oral and intravenous iron may be used        19. Parfrey PS, Foley RN, Wittreich BH, et al. Double-blind comparison of
and there are advantages and disadvantages for each route of admin-                full and partial anemia correction in incident hemodialysis patients
istration, which have been compared in two recent RCTs. The FIND-                  without symptomatic heart disease. J Am Soc Nephrol. 2005;16:
CKD study showed that both routes of administration were effective                 2180–2189.
in this patient population, although higher dose intravenous iron resulted     20. Besarab A, Bolton WK, Browne JK, et al. The effects of normal as
in a faster and greater rise in Hb, with no safety concerns.39 The REVOKE          compared with low hematocrit values in patients with cardiac disease
study, however, suggested that the rate of cardiovascular- and infection-          who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339:
related serious adverse events was greater in the intravenous iron treat-          584–590.
ment group compared with the oral iron group.45 The reasons for these          21. Drüeke TB, Locelli F, Clyne N, et al. Normalization of hemoglobin level
                                                                                   in patients with chronic kidney disease and anemia. N Engl J Med. 2006;
discrepant findings remain unknown. Thus the physician has to balance
                                                                                   355:2071–2084.
the low costs and convenience of oral iron treatment with known poor           22. Singh AK, Szczech L, Tang KL, et al. Correction of anemia with
adherence to treatment, gastrointestinal side effects, and reduced efficacy;       epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355:
intravenous iron on the other hand requires special facilities and set-up          2085–2098.
for administration and carries a very small risk for hypersensitivity          23. Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in
reactions and potential additional risks.                                          type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361:
                                                                                   2019–2031.
                                                                               24. Macdougall IC. New anemia therapies: Translating novel strategies from
REFERENCES                                                                         bench to bedside. Am J Kidney Dis. 2012;59:444–451.
                                                                               25. Macdougall IC, Roger SD, de Francisco A, et al. Antibody-mediated
 1. Jelkmann W. Molecular biology of erythropoietin. Intern Med. 2004;43:          pure red cell aplasia in chronic kidney disease patients receiving
    649–659.                                                                       erythropoiesis-stimulating agents: New insights. Kidney Int. 2012;81:
 2. Koury MJ, Haase VH. Anaemia in kidney disease: harnessing hypoxia              727–732.
    responses for therapy. Nat Rev Nephrol. 2015;11:349–410.                   26. Boven K, Stryker S, Knight J, et al. The increased incidence of pure red
 3. Mole DR, Ratcliffe PJ. Cellular oxygen sensing in health and disease.          cell aplasia with an Eprex formulation in uncoated rubber stopper
    Pediatr Nephrol. 2008;23:681–694.                                              syringes. Kidney Int. 2005;67:346–353.
 4. Semenza GL. Oxygen-sensing hypoxia-inducible factors, and disease          27. Haag-Weber M, Eckardt KU, Hörl WH, et al. Safety, immunogenicity and
    pathophysiology. Annu Rev Pathol. 2014;9:47–71.                                efficacy of subcutaneous biosimilar epoetin-α(HX575) in non-dialysis
 5. Warnecke C, Zaborowska Z, Kurreck J, et al. Differentiating the                patients with renal anemia: a multi-center, randomized, double-blind
    functional role of hypoxia-inducible factor (HIF)-1α and HIF-2α                study. Clin Nephrol. 2012;77:8–17.
    (EPAS-1) by the use of RNA interference: Erythropoietin is a HIF-2α        28. Kaufman JS, Reda DJ, Fye CL, et al. Subcutaneous compared with
    target gene in Hep3B and Kelly cells. FASEB J. 2004;18:462–464.                intravenous epoetin in patients receiving hemodialysis. Department
 6. Scortegagna M, Morris MA, Oktay Y, et al. The HIF family member                of Veterans Affairs Cooperative Study Group on Erythropoietin in
    EPAS1/HIF-2alpha is required for normal hematopoiesis in mice. Blood.          Hemodialysis Patients. N Engl J Med. 1998;339:578–583.
    2003;102:634–640.                                                          29. Vanrenterghem Y, Barany P, Mann JF, et al; for the European/Australian
 7. Chen C, Sytkowski AJ. The erythropoietin receptor and its signalling           NESP 970200 Study Group. Randomized trial of darbepoetin alfa for
    cascade. In: Jelkmann WFP, ed. Erythropoietin: Molecular Biology and           treatment of renal anemia at a reduced dose frequency compared with
    Clinical Use. Johnson City, TN: Graham Publishing; 2003:165–194.               rHuEPO in dialysis patients. Kidney Int. 2002;62:2167–2175.
 8. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med.            30. Carrera F, Lok CE, de Francisco A, et al. Maintenance treatment of
    2005;352:11–23.                                                                renal anaemia in haemodialysis patients with methoxy polyethylene
 9. Drakesmith H, Prentice HM. Hepcidin and the iron-infection axis.               glycol-epoetin beta versus darbepoetin alfa administered monthly: A
    Science. 2012;338:768–772.                                                     randomized comparative trial. Nephrol Dial Transplant. 2010;25:
10. Astor BC, Muntner P, Levin A, et al. Association of kidney function with       4009–4017.
    anemia: The Third National Health and Nutrition Examination Survey,        31. Macdougall IC, Rossert J, Casadevall N, et al. A peptide-based
    1988-1994. Arch Intern Med. 2002;162:1401–1408.                                erythropoietin-receptor agonist for pure red-cell aplasia. N Engl J Med.
11. McClellan W, Aronoff SL, Bolton WK, et al. The prevalence of anemia            2009;361:1848–1855.
    in patients with chronic kidney disease. Curr Med Res Opin. 2004;20:       32. Macdougall IC, Provenzano R, Sharma A, et al. Peginesatide for anemia
    501–510.                                                                       in patients with chronic kidney disease not receiving dialysis. N Engl J
12. K/DOQI. Anemia guidelines in CKD patients. Am J Kidney Dis. 2006;              Med. 2013;368:320–332.
    47(suppl 3):S1–S146.                                                       33. Fishbane S, Schiller B, Locatelli F, et al. Peginesatide in patients with
13. Pisoni RL, Bragg-Gresham JL, Young EW, et al. Anemia management and            anemia undergoing hemodialysis. N Engl J Med. 2013;368:307–319.
    outcomes from 12 countries in the Dialysis Outcomes and Practice           34. Maxwell PH, Eckardt KU. HIF prolyl hydroxylase inhibitors for the
    Patterns Study (DOPPS). Am J Kidney Dis. 2004;44:94–111.                       treatment of renal anaemia and beyond. Nat Rev Nephrol. 2016;12:
14. KDIGO Anemia Guideline Work Group. KDIGO clinical practice                     157–168.
    guideline for anemia in chronic kidney disease. Kidney Int Suppl.          35. Bernhardt W, Wiesener MS, Scigalla P, et al. Inhibition of prolyl
    2012;2:279–335.                                                                hydroxylases increases erythropoietin production in ESRD. J Am Soc
15. Cole J, Ertoy D, Lin H, et al. Lack of angiotensin II–facilitated              Nephrol. 2010;21:2151–2156.
    erythropoiesis causes anemia in angiotensin-converting enzyme–deficient    36. Provenzano R, Besarab A, Wright S, et al. Roxadustat (FG-4592) Versus
    mice. J Clin Invest. 2000;106:391–398.                                         Epoetin Alfa for Anemia in Patients Receiving Maintenance
16. Le Meur Y, Lorgeot V, Comte L, et al. Plasma levels and metabolism of          Hemodialysis: A Phase 2, Randomized, 6- to 19-Week, Open-Label,
    AcSDKP in patients with chronic renal failure: Relationship with               Active-Comparator, Dose-Ranging, Safety and Exploratory Efficacy Study.
    erythropoietin requirements. Am J Kidney Dis. 2001;38:10–17.                   Am J Kidney Dis. 2016;67:912–924.
37. Holdstock L, Meadowcroft AM, Maier R, et al. Four-Week Studies of Oral       41. Solomon SD, Uno H, Lewis EF, et al. Erythropoietic response and
    Hypoxia-Inducible Factor-Prolyl Hydroxylase Inhibitor GSK1278863 for             outcomes in kidney disease and type 2 diabetes. N Engl J Med. 2010;363:
    Treatment of Anemia. J Am Soc Nephrol. 2015;27:1234–1244.                        1146–1155.
38. Pergola BE, Spinowitz BS, Hartman CS, et al. Vadadustat, a novel oral        42. Corwin HL, Gettinger A, Fabian TC, et al, EPO Critical Care Trials
    HIF stabilizer, provides effective anemia treatment in nondialysis-              Group. Efficacy and safety of epoetin alfa in critically ill patients. N Engl J
    dependent chronic kidney disease. Kidney Int. 2016;90:1115–1122.                 Med. 2007;357:965–976.
39. Macdougall IC, Bock AH, Carrera F, et al. FIND-CKD: a randomized trial       43. Macdougall IC, et al. Iron management in chronic kidney disease:
    of intravenous ferric carboxymaltose versus oral iron in patients with           conclusions from a “Kidney Disease: Improving Global Outcomes”
    chronic kidney disease and iron deficiency anaemia. Nephrol Dial                 (KDIGO) Controversies Conference. Kidney Int. 2016;89:28–39.
    Transplant. 2014;29:2075–2084.                                               44. https://www.clinicaltrialsregister.eu/ctr-search/trial/2013-002267-25/GB.
40. Canadian Erythropoietin Study Group. Association between recombinant             Accessed November 26, 2016.
    human erythropoietin and quality of life and exercise capacity of patients   45. Agarwal R, Kusek JW, Pappas MK. A randomized trial of intravenous and
    receiving haemodialysis. BMJ. 1990;300:573–578.                                  oral iron in chronic kidney disease. Kidney Int. 2015;88:905–914.
    SELF-ASSESSMENT
    QUESTIONS
1. Which of the following statements is false regarding the character-
   istics of renal anemia?
   A. The red blood cells (RBCs) produced are usually normochromic
       and normocytic.
   B. There is usually no associated leukopenia or thrombocytopenia.
   C. The reticulocyte count is usually around 40,000 to 50,000/μl of
       blood.
   D. Serum erythropoietin levels are usually within the normal range.
   E. The RBC life span is usually normal.
2. Which of the following statements is false?
   A. Dose requirements of epoetin are generally 20% to 30% less
       when the agent is administered subcutaneously compared with
       intravenously.
   B. Erythropoiesis-stimulating agent (ESA) therapy should be used
       with caution in patients with previous or current malignancy.
   C. Patients who are hyporesponsive to ESA therapy have a worse
       prognosis than those who do respond.
   D. The defined upper dose limit of epoetin is 60,000 IU/wk because
       it is known that cardiovascular toxicity occurs above this dose
       level.
   E. Angiotensin-converting enzyme (ACE) inhibitors may confer
       resistance to ESA therapy in some patients.
3. Which of the following statements is false regarding the TREAT
   study?
   A. Patients receiving darbepoetin alfa were randomized to either a
       target hemoglobin (Hb) of 13 or a target Hb of 9 g/dl.
   B. Patients randomized to a target Hb of 13 g/dl had a small increase
       in quality of life compared with the control arm.
   C. There was a significant reduction in the use of RBC transfusions
       in patients randomized to a target Hb of 13 g/dl.
   D. There was a doubling of the rate of stroke in patients random-
       ized to a target Hb of 13 g/dl.
   E. There was a significant increase in cancer-related mortality in
       the subset of patients with a history of malignancy who were
       randomized to a target Hb of 13 g/dl.
4. Which of the following statements is true regarding intravenous
   iron supplementation?
   A. All intravenous iron preparations require a test dose before their
       first administration, as per their product label.
   B. All the newer intravenous iron preparations (ferumoxytol, ferric
       carboxymaltose, and iron isomaltoside 1000) have the advantage
       that up to 1 g may be administered as a single dose.
   C. Intravenous iron preparations vary in their lability and speed
       of iron release from the carbohydrate shell, with iron gluco-
       nate being the most stable and iron dextran the least stable
       compound.
   D. Intravenous iron may improve the anemia of chronic kidney
       disease in up to 30% of patients not receiving ESA therapy who
       have a low ferritin level.
   E. Hypersensitivity reactions are more common with low molecular
       weight iron dextrans than with high molecular weight iron dextran
       compounds.
IMMUNE DYSFUNCTION                                                            often have excess pulmonary fluid, which may impair alveolar bacterial
                                                                              clearance and contribute to the risk for pneumonia. Repeated breaks
Patients with chronic kidney disease (CKD) have a high morbidity and          in the skin barrier by cannulation provide opportunity for bacterial
mortality as a result of infection. This is in part directly caused by        invasion. Finally, dialysis patients frequently receive intravenous iron
alterations in the immune system, although multiple other factors are         preparations, and iron overload is a risk factor for bacterial infection
involved.                                                                     as a result of inhibition of monocyte and macrophage function.
                                                                                                                                                    967
                                                 downloaded from www.medicalbr.com
968             SECTION XVI Chronic Kidney Disease and the Uremic Syndrome
All negative
                                                           Serologic testing
                                                              (anti-HBs)
                                                           4 weeks after inj.
Anti-HBs > 100 IE/L Anti-HBs 10 - 100 IE/L Anti-HBs < 10 IE/L
                                                                                                      Serologic testing
                                                                             Anti-HBs ≥
                     Anti-HBs < 100 IE/L                                                                 (anti-HBs)
                                                                              10 IE/L
                                                                                                      4 weeks after inj.
                                                 Europe                                                                                              USA
                    1500                                                                             400
300
                                                                                   Number of cases
  Number of cases
1000
200
                    500
                                                                                                     100
                      0
                                                          19A
                           19F
12F
22F
                                                                                                             5
                                                                                                                 9V
                                                                                                                      14
                                                                                                                           1
                                                                                                                               18C
                                                                                                                                     23F
                                                                                                                                           4
                                                                                                                                               6B
                                                                                                                                                    19F
                                                                                                                                                          7F
                                                                                                                                                               6A
                                                                                                                                                                    3
                                                                                                                                                                        19A
                                                                                                                                                                              12F
                                                                                                                                                                                    9N
                                                                                                                                                                                         15B
                                                                                                                                                                                                     22F
                                                                                                                                                                                                           15B
                                           7F
                                      14
                                                                                                                                                                                               11A
                                  4
with every stage of CKD. The majority of HD and peritoneal dialysis                                        (IL-10) influence the quantity of its production in response to inflam-
patients periodically or permanently have CRP values above the normal                                      matory stimuli and thus the way a patient can contain and limit
limit in the absence of clinical infection.12                                                              inflammation.16
intermediate monocytes express both CD14 and CD16 (~5% to 7%),                signals. Only T cells with specificity of their T cell receptor toward the
and nonclassic monocytes express high levels of CD16 while showing            particular antigen are activated and proliferate.
limited staining for CD14 (~10% to 12%). Genetic analysis revealed                Major APCs are dendritic cells and their precursors are monocytes.
that CD14++CD16+ intermediate monocytes particularly express markers          Early studies showed that proliferative T cell responses are impaired in
related to antigen presentation, inflammation, and angiogenesis.20            dialysis patients and this impairment is directly correlated with nonre-
    In CKD, the intermediate and nonclassic monocyte subtypes are             sponses to HBV vaccination. Both replacement of the patient’s APCs
significantly expanded. This finding is related to the CV risk21; patients    in in vitro assays with cells from healthy donors and overexpression
with the highest rate of CD14++CD16+ monocytes in blood had the               of costimulatory molecules on the APCs normalize proliferation of T
lowest CV event-free survival.                                                cells, indicating that the major defect leading to reduced T cell activa-
    Mouse models confirm differences between monocyte subpopula-              tion is in the APC.29 However, these rather crude assays did not con-
tions in their ability to invade atherosclerotic plaques.22 In CKD the        sider T cell subpopulations. Helper T cells express the surface marker
expansion of proinflammatory monocyte subsets and their epidemiologic         CD4 and interact with various other cell types. The CD4+ helper T
association with CV events make a causal role of these cells for athero-      cell is particularly needed for activation of B cells for antigen-specific
sclerotic disease likely. Furthermore, monocytes also express components      seroresponses to viral antigens as in HBV vaccination. The CD8+ cyto-
of the angiotensin system. The angiotensin-converting enzyme (ACE)            toxic T cells are important for antiviral defense because they are able
that turns angiotensin I (Ang I) into vasoactive angiotensin II (Ang II)      to lyse infected host cells. In CKD the relation of CD4+/CD8+ T cells
is expressed in atherosclerotic plaques and colocalizes with monocyte-        is reduced.
derived macrophages.23 ACE is also expressed on circulating monocytes,            CD4+ helper cells can be further distinguished into cells that mainly
particularly on those with the CD14++CD16+ phenotype.24 Dialysis              support cellular immune reactions (T helper cells Type 1, Th1) and
patients with high expression of ACE on intermediate monocytes have           others that are more important for immunoglobulin production by
a dramatically enhanced CV mortality risk.24                                  members of the B cell lineage (Th2). These cell types differ in the pattern
    Expression of ACE on monocytes is strongly upregulated by the             of cytokines they produce, with interferon-γ being the major cytokine
uremic milieu.25 A consequence of this might be that monocytes trans-         of Th1 and IL-4 the main cytokine of Th2 cells. CKD leads to a marked
migrate into the subendothelial space of arteries and provide high levels     deviation of T cell differentiation toward the Th1 phenotype.30 Most
of ACE in the atherosclerotic plaque. The local production of Ang II          likely the major cause is elevated production of IL-12 by monocytes
through ACE is thought to contribute to further leukocyte attraction,         and APCs in the context of their inflammatory activation.30
inflammatory activation, and plaque growth. In addition, expression               Another T-lymphocyte subpopulation is regulatory T cells (Tregs)
of ACE on monocytes alters their functional capacities. In vitro assays       that are important for the downregulation of immune responses once
show that a higher expression of ACE leads to stronger endothelial            the aim of an antiinfectious response is reached. They prevent ongoing
adhesion and transmigration of the monocytes. This effect appears to          inflammation and the development of autoimmunity. The typical Treg
be mediated via locally produced Ang II, because adhesion and trans-          cells originate in the thymus and have a distinct pattern of surface
migration could be inhibited by losartan.25                                   molecule expression. In patients with CKD the number of circulating
    Monocytes also express ACE-2, a peptidase that degrades Ang II to         Treg cells is unaltered, but their capacity to downregulate CD4+ helper
Ang1-7, a vasodilatory peptide. Whereas CKD leads to the overexpres-          T cell activity is impaired.31
sion of ACE, the ACE-2 enzyme is downregulated compared with healthy
individuals.26 Experimental overexpression of ACE-2 in a rodent model         B Lymphocytes
of atherosclerosis limited the progression of disease. These findings         Impaired vaccination efficacy in CKD suggests impaired function of
suggest that the uremic milieu alters monocyte function in a strongly         immunoglobulin producing B-lymphocytes and plasma cells; thus a
proatherogenic way.                                                           major dysfunction of this cell type might be expected. However, CKD
    Monocytes are closely related to circulating blood dendritic cells.       patients have normal circulating immunoglobulin levels. Their B cell
Dendritic cells are mainly found in organs and tissues, where they have       lymphopenia is modest and probably not very clinically relevant. It is
strong capabilities in antigen presentation and activation of immune          caused by a higher rate of apoptosis of these cells compared with healthy
reactions. Their immature precursors circulate in blood in low numbers.       individuals.32 Lymphopenia appears to result from reduced numbers
There are different subtypes of circulating dendritic cells, but investiga-   of the majority of B cell subpopulations (naïve B cells, memory B cells,
tors have used different marker sets for their detection. This limits         etc.).33 Taken together, the alterations of B-lymphocytes in CKD appear
comparison among different studies, so that understanding of dendritic        to be less pronounced than alterations of other immune cell types.
cell quantification and pathophysiology remains limited. There is a           Impaired vaccination responses are caused by altered interaction of
relation between elevated numbers of CD14++CD16+ monocytes in the             APCs, helper T lymphocytes, and the cytokine network in CKD rather
blood and the propensity of these cells to differentiate into dendritic       than by abnormalities of B lymphocytes.
cells in cell culture.27 Other studies28 found significantly lower numbers
of dendritic cells in advanced CKD compared with healthy controls.            Granulocytes
The finding may be related to CV disease, because studies in patients         Polymorphonuclear granulocytes (neutrophils) are components of the
with coronary heart disease and normal renal function also reported           antigen-independent innate immune system. Their main activity is to
reduced circulating numbers of dendritic cells.                               kill and phagocytose invading pathogens via numerous enzymes that
                                                                              produce bactericidal substances. Among them are defensins, proteolytic
T Lymphocytes                                                                 enzymes, and enzymes that produce highly active oxygen species such
Impaired vaccination responses against viral antigens such as HBV or          as hypochlorous acid. In CKD patients these nonspecific defense systems
influenza, as well as reduced skin reaction in the Mantoux test, result       are highly activated and the cells spontaneously release more reactive
from impaired T cell activation. T cells are an important component           oxygen species.34 Inflammation, activation of different cell types, anti-
of the antigen-specific adaptive immune defense. Their activation depends     infectious defense, and vascular disease are closely interwoven. Thus
on antigen-presenting cells (APCs) that present foreign antigens with         the elevation of oxygen species H2O2 and malondialdehyde has predictive
major histocompatibility complex and provide important costimulatory          value for CV events and mortality in CKD.35
    Another important function of granulocytes is phagocytosis, which          to fibrin. Subsequent cross-linking of insoluble fibrin results in a stable
is also mildly compromised in CKD.36                                           hemostatic plug.
    It is difficult to establish whether these alterations can be improved
by dialysis. HD inevitably involves marked contact between blood and           Hemorrhagic Diathesis and Uremic Platelet Dysfunction
foreign surfaces. When cellulose-based membranes were still in use,            Patients with CKD have a high risk for bleeding. This hemorrhagic
the activation of the complement system by these membranes led to              diathesis frequently has cutaneous (easy bruising, ecchymoses, or pro-
marked depletion of circulating granulocytes within the first 20 minutes       longed hemorrhage from needle puncture or postoperative sites) and
of a dialysis session.37 Newer synthetic membranes lead to minimal             mucosal (epistaxis; gastrointestinal or gingival bleeding) manifestations.
complement activation, and the leukocyte drop is much less pronounced.         More dramatic—albeit infrequent—manifestations are hemorrhagic
These findings on immune cells relate to CKD. It is important to con-          pericarditis/hemopericardium, hemorrhagic pleural effusion/hemothorax,
sider that some primary diseases, in particular diabetes mellitus, further     and intracranial and retroperitoneal bleeding.38
influence immune cell function.                                                    This hemorrhagic diathesis is not reflected in a prolongation of the
                                                                               prothrombin time or the partial thromboplastin time. Similarly, even
PLATELET DYSFUNCTION AND PLATELET                                              though platelet counts may be moderately decreased because of platelet
                                                                               consumption outperforming platelet production, severe thrombocyto-
INHIBITORS IN CHRONIC KIDNEY DISEASE                                           penia is rarely seen in uremia; the occurrence of very low platelet counts
Normal hemostasis begins with platelet adhesion to vascular endothe-           therefore requires a thorough search for alternative causes.39 Instead,
lium and requires a relatively vasoconstricted vessel wall, integrity of       platelet dysfunction is generally considered as the central contributor
platelet glycoproteins (GPs), and a normal quantity of large molecular         to the high bleeding risk, and a high number of pathophysiologic altera-
weight, multimeric von Willebrand factor (vWF) (Figs. 83.3 and 83.4).          tions have been suggested to contribute, comprising alterations in platelet
Main platelet GPs are GPIb, the platelet receptor for vWF, involved in         function and structure, and extrinsic factors (see Table 83.1). Unfor-
platelet adhesion, and GPIIb/IIIa, the platelet receptor for fibrinogen,       tunately, many studies on platelet functions in CKD date back to early
involved in platelet aggregation.                                              days of clinical nephrology, when clinical care and dialysis treatment
    Under static conditions, GPIb and vWF have no affinity for each            were less sophisticated, and when less advanced laboratory methods
other. However, these molecules develop a specific affinity for each           were available for evaluation of hemostasis. For the various pathophysi-
other at high shear stress, resulting in arterial platelet adhesion. Aggre-    ologic alterations listed in Box 83.1, controversial data have been
gated fibrinogen-platelet mesh acts as a trap for binding and activation       published.
of other plasma clotting factors. The exposure of the preceding clotting           In the search for uremic toxins inducing platelet dysfunction, a
factors to tissue factors, present on damaged endothelial cells, catalyzes     direct pathologic role of urea can be ruled out because no correlation
the conversion of prothrombin to thrombin, which converts fibrinogen           exists between blood urea levels and bleeding time40 and individuals
                              GpIIb/IIIa                                                                           Fibrinogen
                      Platelet
GpIb
                                                                                                                            Fibrin
                                                                                                                            mesh
                                                                                     Platelet
                                                                                                    Coagulation
                                                                                                     cascade    IIa
                                                                       Clotting Cascade
                                                                                             Fibrinogen     Cross-linking
                                                                                      II
                                                  IX    X
                             VII                                VIIIa†
                                                                                                                                 Fibrin
                                                                                                 Va†
                                                                                                                    (Thrombin)
                                                                                                    *        IIa*
                                         Tissue
                                         factor                       VIIa          VIIa IXa Xa
Endothelial cell
                     Fig. 83.4 Clotting cascade. Expansion of the inset in Fig. 83.3 shows the clotting cascade that takes place
                     at the damaged vessel wall. Exposure of subendothelial tissue factor, present on pericytes and fibroblasts,
                     allows eventual activation of prothrombin (factor II) to thrombin. Thrombin converts fibrinogen to fibrin,
                     activates fibrin cross-linking, stimulates further platelet aggregation, and activates anticoagulant protein C.
                     Naturally occurring anticoagulants antithrombin III, protein C, and protein S help maintain control and coun-
                     terbalance on coagulation. *Site of anticoagulant effect for antithrombin III. †Site of anticoagulant effect for
                     protein C–protein S complex. (Courtesy James A. Sloand, MD, FACP, FASN, Baxter Healthcare Corporation,
                     Deerfield, Ill.)
                                                                                    platelets from the axial flow toward the vessel walls. This allows platelets
 BOX 83.1     Proposed Contributors to
                                                                                    to adhere to injured endothelial cells and initiate the formation of a
 Platelet Dysfunction in Uremia                                                     platelet plug. In anemia, platelets are more dispersed, which impairs
 Intrinsic Factors That Contribute to Platelet Dysfunction                          their adherence to the endothelium. Moreover, in CKD, red blood cells
   Dysfunction of glycoprotein IIb/IIIa                                             may affect coagulation by releasing adenosine diphosphate (ADP), by
   Abnormal expression of platelet glycoprotein                                     inactivating PGI2, and by scavenging nitric oxide (NO), which are all
   Altered release of adenosine diphosphate (ADP) and serotonin from platelet       central regulators of platelet function.39
   α-granules
   Faulty arachidonic acid and depressed prostaglandin metabolism, decreased        Treatment of Uremic Platelet Dysfunction
   platelet thromboxane A2 generation                                               Despite the previously discussed pathophysiologic considerations, few
   Abnormal platelet cytoskeletal assembly with reduced incorporation of actin      data establish the extent to which initiation of RRT reduces the risk
   and diminished association of actin binding proteins (α-actin and tropomyosin)   for bleeding in CKD.
   with the cytoskeleton                                                                In the early decades of HD, the interaction of blood with cellulose-
                                                                                    based dialyzer membranes resulted in complement activation and tran-
 Extrinsic Factors That Contribute to Platelet Dysfunction                          sient thrombocytopenia during the dialysis procedure. When using more
   The action of uremic toxins                                                      biocompatible dialyzer membranes, such complement-induced platelet
   Anemia                                                                           reduction no longer has clinical relevance. Nonetheless, HD treatment
   Increased nitric oxide (NO) and cyclic guanosine monophosphate (cGMP)            may still affect bleeding disorders; while potentially removing uremic
   production                                                                       toxins, which affect platelet function, it requires systemic anticoagula-
   Functional von Willebrand factor abnormalities                                   tion and exposes patients to the potential risk for heparin-induced
   Decreased platelet production                                                    thrombocytopenia (HIT). Moreover, HD may disrupt the platelet
   Abnormal interactions between the platelet and the endothelium of the            cytoskeleton, induce repeated platelet stress, decrease the percentage
   vessel wall                                                                      of RNA-rich platelets, and reduce the percentage of available reticu-
                                                                                    lated platelets.42 As RNA-rich and reticulated platelets are more able
Modified from Berns JS, Coutre S. Platelet dysfunction in uremia.
UpToDate, http://www.annemergmed.com/article/S0196-0644(15)
                                                                                    to be activated, the accumulation of less RNA-rich and less reticulated
00034-7/pdf; reference 38.                                                          platelets indicates the presence of less reactive platelets. Although treat-
                                                                                    ment of anemia may improve some parameters of platelet dysfunc-
                                                                                    tion,42 it has not been demonstrated to ameliorate bleeding or risk for
                                                                                    bleeding.
with high serum urea levels but otherwise normal renal function have                    In summary, dialysis and anemia treatment will not completely
no bleeding tendency.41                                                             normalize platelet function. Therefore drug treatment should be
   Among the different extrinsic factors that contribute to platelet                considered for those patients who have active bleeding or are scheduled
dysfunction, the contribution of anemia has gained particular interest.             to undergo an invasive diagnostic or therapeutic procedure with bleed-
Physiologically, erythrocytes occupy the center of a vessel, displacing             ing risk.
HD patients are exposed to potential side effects of heparin, which               likelihood but focused on patients in whom HIT is suspected clinically
comprise an increased bleeding risk and some less frequent adverse                (as suggested by the 4T score; Table 83.2). This is of particular impor-
events, of which the development of HIT is the most serious complica-             tance for HD patients, in whom an incorrect diagnosis may result in
tion. HIT is a clinical syndrome induced by antibodies that bind to               the withdrawal of heparin treatment during HD and in the initiation
heparin and platelet factor IV complexes on the platelet surface and              of less established (and more expensive) anticoagulation strategies. In
thereby cause platelet activation. Clinically, HIT often manifests with           the absence of CKD-specific pathways, the diagnosis of HIT should
arterial or venous thrombosis; less frequent complications are venous             follow recommendations from the general population. Here, the likeli-
limb gangrene, adrenal hemorrhagic necrosis, necrotizing skin lesions             hood of HIT should first be estimated with clinical prediction tools
at heparin injections sites, and acute systemic reactions within a few            and use of antigen assays for confirmation of HIT antibodies should
minutes after exposure to unfractionated heparin or LMWH injections.              focus on patients with high and intermediate clinical likelihood (Fig.
Of note, many patients have heparin-dependent antibodies without                  83.5). Importantly, these antigen assays for detection of HIT antibodies
clinically apparent HIT. Therefore, to avoid over-diagnosing HIT, immu-           have a high sensitivity but poor specificity. Wherever available, functional
nologic tests should not be ordered in patients with a low clinical               assays (serotonin release assays or heparin-induced platelet activation)
                                Moderate                                                  Moderate
            High titre                                                High titre
                                / low titre                                               / low titre
should be used to confirm positive findings from antigen assays in the        anticoagulation with LMWH. When considering indefinite anticoagula-
majority of patients with suspected HIT.                                      tion in patients with advanced CKD, their elevated bleeding risk must
    Once a patient has a high or intermediate clinical probability            be considered.
of HIT, all heparin treatment must be stopped and alternative antico-             For patients with advanced CKD, the use of anticoagulants in atrial
agulation initiated. The most relevant treatment options are listed in        fibrillation is controversial (see also Chapter 81). In the general popula-
Table 83.3. Of note, no VKA should be initiated at this stage because         tion, patients who have one or more risk factors for cerebral stroke or
these agents may reduce activation of anticoagulatory protein C and           systemic embolization (defined as a CHA2DS2-VASc score of ≥1 in men
thus further perpetuate the prothrombotic state.                              and ≥2 in women) should receive oral anticoagulation. Similarly, use
    Additionally, anticoagulants are approved for prevention of throm-        of warfarin (if adjusted to target INR 2.0-3.0) has been shown to reduce
boembolic stroke in patients with atrial fibrillation, for prevention and     stroke risk substantially in patients with CKD 3a/3b.45 In these patients,
treatment of VTE (particularly deep-vein thrombosis and pulmonary             NOACs are at least as efficient as VKA for prevention of thromboembolic
embolism), and for prevention of clotting in patients with mechanical         events, but may cause fewer intracerebral bleeding events. For patients
heart valves. In the latter case, the need for life-long anticoagulation      with GFR <30 ml/min/1.73 m2, few data demonstrate the efficacy (i.e.,
with VKA is indisputable. Similarly, in patients with proximal deep-vein      prevention of thromboembolic events) and safety (i.e., major bleeding
thrombosis and symptomatic pulmonary embolism, anticoagulation is             events) of either VKA or NOAC because most prospective clinical trials
indicated for a minimum of 3 months. Recommendations on extended              excluded patients with advanced CKD. Moreover, VKA accelerates vas-
anticoagulation are mainly derived from the general population, in            cular calcification46 and possibly also CKD progression, in particular
whom indefinite anticoagulation with either VKA or NOAC (Table 83.4)          if overdosed.47 In dialysis patients with atrial fibrillation, retrospective
may be considered in patients with unprovoked proximal deep-vein              cohort studies yielded conflicting findings on whether oral anticoagula-
thrombosis and symptomatic pulmonary embolism, whereas antico-                tion lowers stroke incidence.48
agulation should be stopped after 3 months in patients with provoked              For the time being, patients with atrial fibrillation and at least mod-
VTE with one or more transient risk factors. In patients with active          erate risk for thromboembolism (defined by CHA2DS2-VASc score ≥2
cancer and VTE, data from the general population suggest indefinite           for women, and ≥1 for men) should be offered NOACs if they have
mild to moderate CKD (CKD 1 to 3b), unless they have prohibitive                 (apixaban) has been licensed for use among dialysis patients in the
high risk for bleeding. For CKD 4 and 5 patients with atrial fibrillation,       United States, but not yet in Europe. Compared with other NOACs,
lack of evidence precludes strong recommendations whether to use                 apixaban is characterized by the least accumulation in CKD, but still
NOACs, VKA, or no anticoagulation.                                               requires (like all NOACs) dose adjustment for renal function. Notably,
   Both LMWH and NOACs require dose adjustment in patients with                  pharmacokinetic studies (and subsequent recommendations on NOAC
CKD because they undergo renal excretion. As of 2017, only one NOAC              dosages) used GFR estimated with the Cockcroft-Gault equation. In
clinical practice, eGFR is now mostly calculated using the Modification of         19. Ziegler-Heitbrock L, Ancuta P, Crowe S, et al. Nomenclature of
Diet in Renal Disease (MDRD) or Chronic Kidney Disease–Epidemiology                    monocytes and dendritic cells in blood. Blood. 2010;116:e74–e80.
(CKD-EPI) formula. Substituting the latter for the former may cause                20. Zawada AM, Rogacev KS, Rotter B, et al. SuperSAGE evidence for
large dosage errors, particularly when physicians fail to consider that                CD14++CD16+ monocytes as a third monocyte subset. Blood. 2011;
                                                                                       118:e50–e61.
the more recent equations yield estimates standardized for a body
                                                                                   21. Heine GH, Ulrich C, Seibert E, et al. CD14(++)CD16(+) monocytes but
surface area of 1.73 m2, and nonstandardized measures of renal func-                   not total monocyte numbers predict cardiovascular events in dialysis
tion (as provided by Cockcroft-Gault equation) are suggested for dose                  patients. Kidney Int. 2008;73:622–629.
adjustment.                                                                        22. Tacke F, Alvarez D, Kaplan TJ, et al. Monocyte subsets differentially
                                                                                       employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic
REFERENCES                                                                             plaques. J Clin Invest. 2007;117:185–194.
                                                                                   23. Diet F, Pratt RE, Berry GJ, et al. Increased accumulation of tissue ACE
 1. United States Renal Data System. 2016 USRDS Annual Data Report:                    in human atherosclerotic coronary artery disease. Circulation. 1996;94:
    Epidemiology of Kidney Disease in the United States. Bethesda, MD:                 2756–2767.
    National Institutes of Health, National Institute of Diabetes and Digestive    24. Ulrich C, Heine GH, Seibert E, et al. Circulating monocyte
    and Kidney Diseases; 2016.                                                         subpopulations with high expression of angiotensin-converting enzyme
 2. James MT, Quan H, Tonelli M, et al. CKD and risk of hospitalization and            predict mortality in patients with end-stage renal disease. Nephrol Dial
    death with pneumonia. Am J Kidney Dis. 2009;54:24–32.                              Transplant. 2010;25:2265–2272.
 3. Pisoni RL, Arrington CJ, Albert JM, et al. Facility hemodialysis vascular      25. Trojanowicz B, Ulrich C, Seibert E, et al. Uremic Conditions Drive
    access use and mortality in countries participating in DOPPS: an                   Human Monocytes to Pro-Atherogenic Differentiation via an Angiotensin-
    instrumental variable analysis. Am J Kidney Dis. 2009;53:475–491.                  Dependent Mechanism. PLoS ONE. 2014;9:e102137.
 4. Sester M, Sester U, Clauer P, et al. Tuberculin skin testing underestimates    26. Trojanowicz B, Ulrich C, Kohler F, et al. Monocytic angiotensin-
    a high prevalence of latent tuberculosis infection in hemodialysis                 converting enzyme 2 relates to atherosclerosis in patients with chronic
    patients. Kidney Int. 2004;65:1826–1834.                                           kidney disease. Nephrol Dial Transplant. 2016.
 5. London WT, DiFiglia M, Sutnick AI, Blumberg BS. An epidemic of                 27. Dopheide JF, Zeller GC, Kuhlmann M, et al. Differentiation of
    hepatitis in a chronic hemodialysis unit. N Engl J Med. 1969;281:                  Monocyte Derived Dendritic Cells in End Stage Renal Disease is
    571–578.                                                                           Skewed Towards Accelerated Maturation. Adv Clin Exp Med. 2015;24:
 6. Janssen RS, Mangoo-Karim R, Pergola PE, et al. Immunogenicity and                  257–266.
    safety of an investigational hepatitis B vaccine with a toll-like receptor     28. Agrawal S, Gollapudi P, Elahimehr R, et al. Effects of end-stage renal
    9 agonist adjuvant (HBsAg-1018) compared with a licensed hepatitis B               disease and haemodialysis on dendritic cell subsets and basal and
    vaccine in patients with chronic kidney disease. Vaccine. 2013;31:                 LPS-stimulated cytokine production. Nephrol Dial Transplant. 2010;25:
    5306–5313.                                                                         737–746.
 7. Barraclough KA, Wiggins KJ, Hawley CM, et al. Intradermal Versus               29. Girndt M, Köhler H, Schiedhelm-Weick E, et al. T-cell activation defect
    Intramuscular Hepatitis B Vaccination in Hemodialysis Patients:                    in hemodialysis patients: Evidence for a role of the B7/CD28 pathway.
    A Prospective Open-Label Randomized Controlled Trial in                            Kidney Int. 1993;44:359–365.
    Nonresponders to Primary Vaccination. Am J Kidney Dis. 2009;54:95.             30. Sester U, Sester M, Hauk M, et al. T-cell activation follows Th1 rather
 8. Clinical KDIGO. Practice Guideline for the Evaluation and Management               than Th2 pattern in haemodialysis patients. Nephrol Dial Transplant.
    of Chronic Kidney Disease. Kidney Int Suppl. 2013;3:1.                             2000;15:1217–1223.
 9. Remschmidt C, Wichmann O, Harder T. Influenza vaccination in patients          31. Afzali B, Edozie FC, Fazekasova H, et al. Comparison of regulatory
    with end-stage renal disease: systematic review and assessment of quality          T cells in hemodialysis patients and healthy controls: implications for
    of evidence related to vaccine efficacy, effectiveness, and safety. BMC Med.       cell therapy in transplantation. Clin J Am Soc Nephrol. 2013;8:
    2014;12:244.                                                                       1396–1405.
10. Fuchshuber A, Kuhnemund O, Keuth B, et al. Pneumococcal vaccine in             32. Fernandez-Fresnedo G, Ramos MA, Gonzalez PM, et al. B lymphopenia
    children and young adults with chronic renal disease. Nephrol Dial                 in uremia is related to an accelerated in vitro apoptosis and dysregulation
    Transplant. 1996;11:468–473.                                                       of Bcl-2. Nephrol Dial Transplant. 2000;15:502–510.
11. Bond TC, Spaulding AC, Krisher J, McClellan W. Mortality of dialysis           33. Kim KW, Chung BH, Jeon EJ, et al. B cell-associated immune profiles
    patients according to influenza and pneumococcal vaccination status. Am            in patients with end-stage renal disease (ESRD). Exp Mol Med. 2012;44:
    J Kidney Dis. 2012;60:959–965.                                                     465–472.
12. Miyamoto T, Carrero JJ, Stenvinkel P. Inflammation as a risk factor and        34. Yoon JW, Pahl MV, Vaziri ND. Spontaneous leukocyte activation and
    target for therapy in chronic kidney disease. Curr Opin Nephrol                    oxygen-free radical generation in end-stage renal disease. Kidney Int.
    Hypertens. 2011;20:662–668.                                                        2007;71:167–172.
13. Kudo S, Goto H. Intrarenal handling of recombinant human                       35. Kalantar-Zadeh K, Brennan ML, Hazen SL. Serum myeloperoxidase and
    interleukin-1alpha in rats: mechanism for proximal tubular protein                 mortality in maintenance hemodialysis patients. Am J Kidney Dis. 2006;
    reabsorption. J Interferon Cytokine Res. 1999;19:1161–1168.                        48:59–68.
14. Chen LP, Chiang CK, Chan CP, et al. Does periodontitis reflect                 36. Anding K, Gross P, Rost JM, et al. The influence of uraemia and
    inflammation and malnutrition status in hemodialysis patients? Am J                haemodialysis on neutrophil phagocytosis and antimicrobial killing.
    Kidney Dis. 2006;47:815–822.                                                       Nephrol Dial Transplant. 2003;18:2067–2073.
15. Missailidis C, Hallqvist J, Qureshi AR, et al. Serum Trimethylamine-N-         37. Craddock PR, Fehr J, Dalmasso AP, et al. Hemodialysis leukopenia.
    Oxide Is Strongly Related to Renal Function and Predicts Outcome in                Pulmonary vascular leukostasis resulting from complement activation
    Chronic Kidney Disease. PLoS ONE. 2016;11:e0141738.                                by dialyzer cellophane membranes. J Clin Invest. 1977;59:879–888.
16. Girndt M, Sester U, Sester M, et al. The interleukin-10 promotor               38. Kaw D, Malhotra D. Platelet dysfunction and end-stage renal disease.
    genotype determines clinical immune function in hemodialysis patients.             Semin Dial. 2006;19:317–322.
    Kidney Int. 2001;60:2385–2391.                                                 39. Galbusera M, Remuzzi G, Boccardo P. Treatment of bleeding in dialysis
17. Ziegler-Heitbrock L. Blood Monocytes and Their Subsets: Established                patients. Semin Dial. 2009;22:279–286.
    Features and Open Questions. Front Immunol. 2015;6:423.                        40. Steiner RW, Coggins C, Carvalho AC. Bleeding time in uremia: a useful
18. Auffray C, Fogg D, Garfa M, et al. Monitoring of blood vessels and tissues         test to assess clinical bleeding. Am J Hematol. 1979;7:107–117.
    by a population of monocytes with patrolling behavior. Science. 2007;317:      41. Linthorst GE, Avis HJ, Levi M. Uremic thrombocytopathy is not about
    666–670.                                                                           urea. J Am Soc Nephrol. 2010;21:753–755.
42. Hedges SJ, Dehoney SB, Hooper JS, et al. Evidence-based treatment             46. van Gorp RH, Schurgers LJ. New Insights into the Pros and Cons of the
    recommendations for uremic bleeding. Nat Clin Pract Nephrol. 2007;                Clinical Use of Vitamin K Antagonists (VKAs) Versus Direct Oral
    3:138–153.                                                                        Anticoagulants (DOACs). Nutrients. 2015;7:9538–9557.
43. Ma TK, Chow KM, Kwan BC, et al. Manifestation of tranexamic acid              47. Bohm M, Ezekowitz MD, Connolly SJ, et al. Changes in Renal Function
    toxicity in chronic kidney disease and kidney transplant patients: a report       in Patients With Atrial Fibrillation: An Analysis From the RE-LY Trial.
    of four cases and review of literature. Nephrology (Carlton). 2016.               J Am Coll Cardiol. 2015;65:2481–2493.
44. Palmer SC, Di Micco L, Razavian M, et al. Antiplatelet therapy to prevent     48. Tan J, Liu S, Segal JB, et al. Warfarin use and stroke, bleeding and
    hemodialysis vascular access failure: systematic review and meta-analysis.        mortality risk in patients with end stage renal disease and atrial
    Am J Kidney Dis. 2013;61:112–122.                                                 fibrillation: a systematic review and meta-analysis. BMC Nephrol. 2016;
45. Hart RG, Pearce LA, Asinger RW, Herzog CA. Warfarin in atrial                     17:157.
    fibrillation patients with moderate chronic kidney disease. Clin J Am Soc     49. Dinits-Pensy M, Forrest GN, Cross AS, Hise MK. The use of vaccines in
    Nephrol. 2011;6:2599–2604.                                                        adult patients with renal disease. Am J Kidney Dis. 2005;46:997–1011.
    SELF-ASSESSMENT
    QUESTIONS
1. The following diagnostic finding is reliable (sensitive and specific
   enough to be useful) in patients with chronic kidney disease (CKD)
   even in the presence of their typical immune dysfunction:
   A. Fever as a sign of bacteremia
   B. Radiologic infiltration on chest x-ray film for pneumonia
   C. Mendel-Mantoux skin reaction for tuberculosis
   D. Blood culture for bacteremia
   E. C-reactive protein for infection
2. Viral hepatitis is a typical complication of hemodialysis treatment.
   Which statement describes viral hepatitis in advanced CKD
   correctly?
   A. Hepatitis B virus (HBV) infection leads to chronic infection in
       the majority of affected CKD patients.
   B. Acute HBV infection in CKD patients is typically severe, char-
       acterized by jaundice and fever.
   C. HBV infection can be easily prevented in CKD patients by vac-
       cination as recommended to the general population.
   D. The clinical course of hepatitis C infection is largely different in
       CKD patients and those with normal renal function.
   E. Standard hygienic precautions have largely failed to reduce HBV
       transmission in dialysis centers. Only vaccination succeeded in
       preventing nosocomial transmission.
3. The hemorrhagic diathesis in CKD patients is typically mirrored
   by which of the following?
   A. A prolongation of the prothrombin time
   B. A prolongation of the partial thromboplastin time
   C. Severely thrombocytopenia
   D. Hyperchromic anemia
   E. None of these
4. The spectrum of interventions that may allow reducing hemorrhages
   in advanced CKD does not include which of the following?
   A. Desmopressin
   B. Conjugated estrogens
   C. Tranexamic acid (TXA)
   D. Cryoprecipitates
   E. Clopidogrel
DEFINITION                                                                      PATHOGENESIS
Disturbances of mineral metabolism are common if not ubiquitous in              Several biochemical and hormonal abnormalities associated with CKD
chronic kidney disease (CKD) and lead to serious and debilitating com-          contribute to renal osteodystrophy and can be affected by efforts at
plications unless these abnormalities are addressed and treated. The            prevention and therapy. The major factors may vary as CKD progresses
spectrum of disorders includes abnormal concentrations of serum                 (Fig. 84.3). Similarly, the predominance of one particular pathogenetic
calcium, phosphate, and magnesium and disorders of parathyroid                  mechanism over another may contribute to the heterogeneity of bone
hormone (PTH), fibroblast growth factor-23 (FGF-23), and vitamin D              disorders. We therefore discuss separately the two major entities—high-
metabolism. These abnormalities as well as other factors related to the         and low-turnover osteodystrophy.
uremic state affect the skeleton and result in the complex disorders of
bone known as renal osteodystrophy; it is now recommended that this             OSTEITIS FIBROSA: HYPERPARATHYROIDISM—
term be used exclusively to define the bone disease associated with
CKD. The clinical, biochemical, and imaging abnormalities heretofore
                                                                                HIGH-TURNOVER RENAL BONE DISEASE
identified as correlates of renal osteodystrophy should be defined more         Elevated levels of PTH in blood, hyperplasia of the parathyroid glands,
broadly as a clinical entity or syndrome called chronic kidney disease–         and elevations in FGF-23 are seen once eGFR declines below approxi-
mineral and bone disorder (CKD-MBD).1 The spectrum of skeletal                  mately about 50 ml/min/1.73 m2. Whereas the level of free (i.e., non–
abnormalities seen in renal osteodystrophy includes the following               protein bound) calcium in blood is normally the principal determinant
(Fig. 84.1):                                                                    of PTH secretion, several metabolic disturbances associated with CKD
                                                                       -        also alter the regulation of the secretion of PTH.
    ized by increased osteoclast and osteoblast activity, peritrabecular
    fibrosis, and increased bone turnover.                                      Abnormalities of Calcium Metabolism
                                                                                There are three main body pools of calcium: the bony skeleton (mineral
   formed osteoid most often caused by aluminum deposition; bone                component), the intracellular pool (mostly protein bound), and the
   turnover is decreased.                                                       extracellular pool (see Chapter 10). The calcium in the extracellular
                                                               -                pool is in continuous exchange with that of bone and cells and is
   mally low bone turnover.                                                     altered by diet and excretion. Calcium metabolism depends on the
                                                                                close interaction of two hormonal systems: PTH and vitamin D. Per-
                                                              mixed renal       turbations of both systems occur during the course of CKD, with
   osteodystrophy.                                                              adverse consequences on the skeleton. Total serum calcium tends to
                                                                            -   decrease during the course of CKD as a result of phosphate retention
   dosis, β2                                  β2M amyloidosis]).                and decreased production of 1,25-dihydroxyvitamin D (calcitriol) from
                                                                                the kidney, decreased intestinal calcium absorption, and skeletal resis-
                                                                                tance to the calcemic action of PTH, but the levels of free calcium
EPIDEMIOLOGY                                                                    remain within the normal range in most patients3 as a result of com-
The prevalence of the various types of renal osteodystrophy in patients         pensatory hyperparathyroidism. Because calcium is a major regulator
with end-stage renal disease (ESRD) is illustrated in Fig. 84.2.2 In patients   of PTH secretion, persistent hypocalcemia is a powerful stimulus for
                                                                                development of hyperparathyroidism and also contributes to parathy-
equal frequency. In contrast, in patients on peritoneal dialysis (PD),          roid growth.
a small fraction of cases in either group but is more common in certain         Abnormalities of Phosphate Metabolism
                                                                        -       With progressive CKD, phosphate is retained, at least, transiently, by
ated with CKD start while the estimated glomerular filtration rate (eGFR)       the failing kidney. However, hyperphosphatemia usually does not
is still relatively preserved (~50 ml/min/1.73 m2).                             become evident before CKD stage 4. Until then, compensatory
                                                                                                                                                  979
                                                  downloaded from www.medicalbr.com
980                        SECTION XVI Chronic Kidney Disease and the Uremic Syndrome
                                                                                                   100
                                           Mixed                                                           Calcium <8.4 mg/dl (2.1 mmol/l)
                                                                                                    80     Phosphate >4.6 mg/dl (1.5 mmol/l)
                                                                                    Patients (%)
                                  Hyperparathyroidism                                               60     iPTH >65 pg/ml
                                                                         High
                                                                       turnover
                                                                                                    40
                                   Calcium, calcitriol
     Low                                                                                            20
   turnover                               Aluminum
                                                                                                     0
                                                                                                         >80 79-70 69-60 59-50 49-40 39-30 29-20 <20
Fig. 84.1 The spectrum of renal osteodystrophy. The range of
                                                                                                                        eGFR (ml/min)
skeletal abnormalities in renal bone disease encompasses syndromes
with both high and low bone turnover.
                                                                                                   100
                                                                                                           25(OH)D3 <15 ng/ml
                                                                                                    80     1,25(OH)2D3 <22 pg/ml
                                                                                    Patients (%)
           Prevalence of Renal Osteodystrophy in
                                                                                                    60
           Patients with End-Stage Renal Disease
                                                                                                    40
                      50
                                                                                                    20
                      40                                              HPT                            0
                                                                                                         >80 79-70 69-60 59-50 49-40 39-30 29-20 <20
      % Patients/yr
                      20                                              LTOM                         100
                                                                                                           FGF-23 > 100 RU/ml
                                                                      ABD                                  iPTH >65 pg/ml
                      10                                                                            80
                                                                                                           Phosphate >4.6 mg/dl
                                                                                    Patients (%)
                      0                                                                             60     (1.5 mmol/l)
                       1995 1996 1997 1998 1999 2000 2001
                                                                                                    40
                                     Years
                                                                                                    20
Fig. 84.2 Prevalence of renal osteodystrophy in patients with
end-stage renal disease. ABD, Adynamic bone disease; HPT, high-                                      0
turnover renal osteodystrophy; LTOM, low-turnover osteomalacia; MUO,                                          >70 69-60 59-50 49-40 39-30 29-20 <20
mixed uremic osteodystrophy. (From reference 32.)                                                                    eGFR (ml/min/1.73 m2)
                                                                                  Fig. 84.3 Percentage of patients exhibiting elevated circulating
                                                                                  levels of calcium, phosphate, intact parathyroid hormone (iPTH),
hyperparathyroidism and increases in circulating FGF-23 result in                 25OH-vitamin D3, calcitriol, and fibroblastic growth factor-23
increased phosphaturia, maintaining serum phosphate levels in the                 (FGF-23) in advancing chronic kidney disease (CKD). Particularly
normal range.4                                                                    in early CKD stages there is wide variability at the individual level, and
                                                                         -        some patients, for example, may exhibit increased FGF-23 and normal
parathyroidism is by a decrease in serum free calcium, which in turn              iPTH, whereas others may have elevated iPTH levels with normal FGF-23
stimulates the secretion of PTH (Fig. 84.4). Thus a new steady state is           or elevations of both. (Data from references 54 and 55.)
achieved in which serum phosphate is restored to normal at the expense
of a sustained high level of PTH. This cycle is repeated as renal function        serum calcium or serum calcitriol.5,6 Phosphate may have an effect on
declines until sustained and severe hyperparathyroidism is present.               parathyroid growth independent of serum calcium.7,8 Regardless of the
Second, phosphate retention leads to decreased production of calcitriol           mechanism by which phosphate retention causes hyperparathyroidism,
by the kidney, either directly or by increasing the levels of FGF-23              experimental studies suggest that restriction of dietary phosphate in
(which decreases the activity of 1α-hydroxylase). The decrease in cal-            proportion to the decrease in GFR can prevent development of hyper-
citriol allows increases in PTH gene transcription by direct action and           parathyroidism. Current evidence suggests that FGF-23 also acts directly
also decreases intestinal calcium absorption, leading to hypocalcemia,            on the parathyroid gland and has inhibitory effects on PTH secretion
which in turn stimulates PTH secretion. Third, hyperphosphatemia is               and parathyroid growth.9,10 This suggests that the main effects of FGF-23
associated with resistance to the actions of calcitriol in the parathyroid        on the pathogenesis of hyperparathyroidism are indirect as a result of
glands, which also favors development of hyperparathyroidism and                  the potent effect of FGF-23 to decrease calcitriol production. These
induces resistance to the actions of PTH in bone. Finally, phosphate              various actions may explain, at least in part, the association between
per se appears to affect PTH secretion independently of changes in                higher levels of FGF-23 and adverse clinical outcomes.11
Hyperphosphatemia
                                                       Decreased intestinal
                                                        calcium absorption
Hypocalcemia
↑ PTH secretion
↓ GFR ↑ Pi ↓ 25 (OH)D3
                                                                                                       Abnormal
                                                                                                  parathyroid function                              Hypocalcemia
   A                                                B                                                  C
                    Fig. 84.7 Extraskeletal calcification in chronic renal failure. (A) Arterial calcification (arrows). (B) Pul-
                    monary calcification. (C) Periarticular calcification (arrows).
                                                                                                                                                       -
                                                                             terized by increased osteoid seam width, increase in the trabecular surface
                                                                             covered with osteoid, and decreased bone mineralization as assessed
                                                                             by tetracycline labeling (see Fig. 84.11B). The presence of aluminum
                                                                             can be detected on the mineralization front by specific staining (see
A                                      B                                     C                                  D
                    Fig. 84.11 Bone histology in renal osteodystrophy. (A) Osteitis fibrosa: Characteristic manifestations
                    of severe hyperparathyroidism with increased osteoclast and osteoblast activity and peritrabecular fibrosis
                    (stained blue; arrow). (B) Adynamic bone disease: There is no cellular activity along the bone surface and no
                    osteoid visible. (C) (arrowhead) Mixed renal osteodystrophy: There is evidence of active osteoclasts on one
                    bone surface (arrowhead) and evidence of thickened osteoid (stained red; asterisk) indicating a mineralization
                    defect on the other. (D) Osteomalacia: Marked excess of unmineralized osteoid (stained red) surrounding
                    the mineralized bone (stained blue).
calcium should be measured. The initial approach to therapy for hypo-        Dietary Phosphate Restriction
calcemia in patients with mild to moderate CKD is the administration         In experimental animals with mild CKD, dietary phosphate restriction
of calcium supplements such as calcium carbonate, taken between              can prevent excessive PTH synthesis and secretion, as well as parathyroid
                                                                             cell proliferation, independently of changes in serum calcium and cal-
should be undertaken by measurement of 25-hydroxyvitamin D, and
this should be corrected if it is below 30 ng/ml. The determination of       intake might be considered in patients with CKD stage 2 or 3. The
1,25-dihydroxyvitamin D levels is not helpful in this respect, given its     input of a dietician is essential. Protein restriction and avoidance of
                                                                       -     dairy products (especially processed foods containing high amounts of
                                                                             added phosphate) are the mainstays of the regimen. Phosphate-protein
vitamin D sterols should be considered if hyperparathyroidism or             restriction increases the serum levels of calcitriol in patients with mild
hypocalcemia persists. In patients with ESRD, active vitamin D sterols       to moderate CKD. However, restriction of phosphate by severe dietary
are often required. In dialysis patients, the goal is to achieve levels of   protein restriction below 0.8 g/kg/day may lead to protein-calorie
iPTH that are approximately two to nine times above the upper limit          malnutrition.
of the assay used.23                                                     -
tored. In CKD stages 3 to 5, progressive rises in iPTH above the normal      Phosphate Binders
range should be abrogated by correction of hypocalcemia, vitamin D           Whereas dietary phosphate restriction is usually sufficient in early CKD,
deficiency, and hyperphosphatemia. The latter should be corrected before     the control of phosphate becomes more difficult as renal function dete-
the correction of hypocalcemia.                                              riorates. It then becomes necessary to also use agents that bind ingested
                                                                             phosphate in the intestinal lumen to limit its absorption. Compounds
Control of Phosphate                                                         used for this purpose include calcium-containing antacids, magnesium
Control of phosphate is the cornerstone of effective management of           salts, aluminum hydroxide, and non–calcium-containing, non–
secondary hyperparathyroidism. In mild to moderate CKD, a normal             aluminum-containing phosphate binders (Fig. 84.12). Most phosphate
serum phosphate concentration does not necessarily indicate normal           binders, especially in advanced CKD, have to be given in large numbers
parathyroid status, and except for the late stages of CKD, normophos-        (often accounting for 50% of the daily pill burden) and consequently
phatemia may be maintained at the expense of elevated serum PTH              patient adherence with the medication is a major problem.
and FGF-23. Therefore efforts to control phosphate, including dietary
phosphate restriction and the use of phosphate binders, should not be        in patients with CKD, their long-term use can no longer be recom-
delayed until frank hyperphosphatemia develops.                              mended because of the risk for aluminum toxicity. Ingestion of
                                                                                                          Removed by HD
                          Dietary phosphorus                                                                3 × 800 mg
                                                                          Phosphorus
                                 intake             Absorption                                             2400 mg/wk
                                                                           requiring
                                                                            removal
                              1000 mg/day              60%
                                   or
                                                                          4200 mg/wk                    Phosphorus balance
                              7000 mg/wk
                                                                                                           +1800 mg/wk
Fig. 84.12 Phosphate balance and phosphate binders used in hemodialysis patients.
aluminum-containing antacids together with foods containing citric            lanthanum appears to accumulate in bone, liver, and gastric mucosa.
acid (e.g., fruit juices and foods with sodium, calcium, or potassium         Newly approved iron-containing phosphate binders include ferric citrate
citrate) may significantly increase aluminum absorption and therefore         and sucroferric oxyhydroxide. Ferric citrate allows significant oral iron
should be avoided.                                                            uptake and therapy necessitates monitoring of iron stores, whereas iron
    Calcium carbonate or calcium acetate taken with meals effectively         uptake from sucroferric oxyhydroxide is low.24
binds phosphates and limits their absorption. They are effective phos-
phate binders in 60% to 70% of patients on HD. The doses required             Use of Vitamin D Metabolites
to prevent hyperphosphatemia may vary according to patient compli-            Calcitriol and other 1α-hydroxylated vitamin D sterols, such as
ance with dietary phosphate restriction as well as the CKD stage.             1α-hydroxyvitamin D3 (alfacalcidol), 1α-hydroxyvitamin D2 (doxer-
Hypercalcemia and calcium loading are the major potentially serious           calciferol), and 19-nor-1α,25-dihydroxyvitamin D2 (paricalcitol), are
side effects. Current recommendations are to limit the ingestion of           effective in the control of secondary hyperparathyroidism. Calcitriol
elementary calcium to 1500 mg/day. Consideration of overall calcium           lowers PTH levels and improves bone histologic status. In patients with
balance may be important with the use of calcium-containing phos-             very high levels of PTH and markedly enlarged glands with severe
phate binders.                                                                nodular hyperplasia, the effectiveness of vitamin D metabolites may be
    Magnesium salts are effective phosphate binders for patients who          limited because the levels of vitamin D receptor are low in such tissue.
become hypercalcemic with calcium-containing phosphate binders, but
they should be administered with caution in CKD patients not on               hyperparathyroidism with vitamin D metabolites early in CKD when
dialysis because hypermagnesemia may have serious adverse effects. In         the parathyroid glands are more sensitive to such therapy and thereby
patients on dialysis, magnesium carbonate (elemental magnesium 200
to 500 mg/day) has been used successfully, with prevention of hyper-          D metabolite therapy in treatment of secondary hyperparathyroidism
magnesemia through a reduction in dialysate magnesium concentration.          in patients with mild to moderate CKD has been shown, but the concern
The use of magnesium carbonate also allows reduction of the dose of           with initiation of vitamin D therapy at this stage of CKD is acceleration
calcium carbonate required by about half, but its use is frequently com-      of the progression of renal disease should hypercalcemia occur. Because
plicated by diarrhea.                                                         of the effect of calcitriol to increase intestinal phosphate absorption,
    Nonabsorbable, calcium-free polymers, such as sevelamer hydro-            hyperphosphatemia and elevations in calcium-phosphate product may
chloride in a dose range of 2.4 to 4.8 g/day, provide effective phosphate     predispose patients to the development of metastatic calcification;
                                                                              however, it appears that doses of 1α-hydroxyvitamin D3 or calcitriol
containing phosphate binders in terms of limiting the calcium load,           up to 0.5 mcg/day are not commonly associated with hypercalcemia,
although they are significantly more expensive. Studies have suggested
that the use of sevelamer is associated with decreased progression of         with the use of vitamin D metabolites before dialysis is that oversup-
vascular calcification.24 Sevelamer hydrochloride has largely been replaced   pression of hyperparathyroidism may increase the risk for adynamic
by sevelamer carbonate, which has similar properties. Sevelamer may
be combined with both calcium- and magnesium-containing phosphate             and should not be instituted without documentation of hyperparathy-
binders if necessary. Lanthanum carbonate also is an effective phosphate      roidism, correction of 25-hydroxyvitamin D deficiency, and prior control
binder. No significant toxicity has been observed, although some              of serum phosphate.
Treat acidosis
                         Consider:
                         - Dietary Pi restriction
                         - P binders
                                   CKD 3
                                                       Consider:
                                                       Active vitamin D sterols
                                                       Limiting Ca intake
                                                              CKD 4
                                                                                  Consider:
                                                                                  Parathyroidectomy
                                                                                  Calcimimetic*
                                                                                  Dialysis regimen*
                                                                                  Dialysate calcium*
CKD 5
                    Fig. 84.13 Treatment of renal osteodystrophy at various stages of renal impairment. *Consider
                    in CDK5D (i.e., dialysis dependent) only. Ca, Calcium; CKD, chronic kidney disease; iPTH, intact parathyroid
                    hormone; P, phosphate; Pi, inorganic phosphate. (Modified from reference 56.)
adjusted based on serial measurements of iPTH, with careful attention         combination with the other strategies if iPTH levels do not fall into
                                                                              the target range. Parathyroidectomy should be considered in selected
should be treated with oral sodium bicarbonate because persistent aci-        circumstances. Bone biopsy may be indicated in selected patients, par-
dosis has deleterious effects on the skeleton. The additional sodium
load may require further salt restriction or increases in diuretics.          require chelation therapy with deferoxamine in selected circumstances,
                                                                         -    especially if it is symptomatic, but in most patients the prevention of
thyroidism (iPTH more than two to three times above the upper normal          further aluminum exposure is sufficient to allow a gradual reduction
range of the assay) persists despite these measures, consideration should     in the serum levels of aluminum. During therapy with potent vitamin
be given to the addition of calcitriol (0.25 to 0.5 mcg/day), vitamin D       D metabolites, attention should be given to the dialysate calcium con-
analogues, or vitamin D prohormones to the regimen. This therapy              centrations because high concentrations may aggravate hypercalcemia.
should be monitored carefully to avoid hypercalcemia and acceleration         However, the increasingly frequent use of lower dialysate calcium levels,
of progression of CKD.                                                        such as 1.25 mmol/l, requires careful monitoring of the patient to ensure
    In CKD stages 4 and 5, the preceding therapies may need to be             compliance with calcium-containing phosphate binders and vitamin
intensified and larger amounts of phosphate binders may be required           D metabolites to avoid progressive negative calcium balance. Dialysate
to avoid hyperphosphatemia. The use of aluminum-containing phos-              calcium should remain within the range of 1.25 to 1.75 mmol/l and,
phate binders is particularly undesirable at this stage in view of the        when possible, should be individually prescribed.
increased risk for aluminum accumulation with worsening renal func-
tion. In patients on dialysis, calcitriol therapy can be intensified, with
attention to the serum levels of calcium and phosphate and monitoring
                                                                              LOW-TURNOVER RENAL BONE DISEASE
of iPTH levels. In CKD stage 5, iPTH levels should be maintained                                                                                       -
approximately two to nine times above the upper limit of the assay
used to maintain normal bone turnover.23 Calcitriol may be administered       develops as an adaptive response to counteract the increasing skeletal
orally either daily or intermittently (pulse therapy) or administered
intravenously to patients on HD. During therapy with calcitriol, it is
imperative to ensure serum phosphate remains controlled and eleva-            important in CKD-MBD because of the high percentage of affected
tions of serum calcium do not occur to prevent metastatic calcification.      individuals (>40% in CKD stage 5) and because of its association with
Vitamin D analogues, which are less calcemic and phosphatemic than            CV calcification and mortality.31,32 Furthermore, fracture incidence is
calcitriol and yet retain the ability to suppress the levels of PTH, may      estimated to be twice as high in individuals with low than in those with
be useful. Cinacalcet provides additional effective control of hyperpara-
thyroidism in patients with ESRD and may be used alone or in                  biopsy registries of dialysis patients, which may relate to the increasing
prevalence of its key risk factors—advanced age and diabetes mellitus.              In CKD stages 3 and 4, there are uncertainties about the diagnosis
PD also represents a risk factor, possibly because of an often continuous
exposure to high dialysate calcium as opposed to the cyclic exposure             are required to maintain adequate bone turnover in these stages. It
associated with HD.                                                              seems reasonable to correct vitamin D deficiency, hyperphosphatemia,
                                                                                 and hypocalcemia when PTH levels start to rise, but beyond that, no
Pathogenesis of Adynamic Bone Disease                                            firm recommendations can be given.
Given that the bone develops a relative resistance of the PTH-1 receptor
to its ligand PTH as CKD progresses, PTH levels above the normal                 Bone Biopsy
range are required to maintain adequate bone turnover. Unfortunately,
there are no definite ranges of elevated PTH levels that can reliably            the TMV classification (see earlier discussion),1
differentiate an adaptive response (normal bone turnover) from a mal-            by low turnover, normal (or high secondary) mineralization, and low
adaptive response (increased bone turnover) because PTH resistance               bone (osteoid) volume. The individual indication to perform bone
                                                                       -         biopsy should be considered in symptomatic patients based on incon-
                                                                                 sistencies of biochemical parameters associated with unexplained frac-
cause suppression or cessation of both osteoblast and osteoclast activities,     tures, bone pain, progressive extraosseous calcifications, or hypercalcemia.
resulting in a reduced bone formation rate and low bone mass. Iatro-
genic oversuppression of PTH in CKD mainly results from high-dose                samples and concomitant serum samples to identify biomarker patterns
active vitamin D metabolite treatment, from calcium loading (high
doses of calcium-containing phosphate binders, high dialysate calcium
concentration), or after parathyroidectomy. The effects of calcimimetic
treatment on bone turnover have been prospectively evaluated in HD               from non-high bone formation rate.36 However, there is considerable
patients, and patients in this cohort had significant high-turnover oste-
opathy at baseline (average iPTH >1200 pg/ml). Therapy with cinacalcet
decreased elevated bone formation rate and improved bone histologic              significant aluminum exposure, aluminum bone deposition should be
status.33                                                                        excluded by measurement of serum aluminum and specific staining of
target range of two to nine times the upper reference range of the assay.        a bone biopsy specimen.
Finally, diabetes, uremic toxins, malnutrition, and potentially, C-terminal
PTH fragments may be additional factors favoring a state of low bone             Radiology and Measurements of Bone Density
turnover (Fig. 84.14).
Diagnosis and Differential Diagnosis                                             depending on the primary or secondary mineralization state, but it
Serum Biochemistry                                                               never reflects the actual turnover and is therefore not a helpful diagnostic
Low iPTH levels (<100 to 150 pg/ml) are almost always indicative of
low bone turnover in CKD stage 5D. However, histologically proven                radiographs may raise the suspicion of a low bone turnover state if
                                                                                 accompanying biochemical parameters are compatible with this diagnosis.
300 pg/ml and, in exceptions, of up to 600 pg/ml.34,35 Therefore PTH
                                                                                 calcification in dialysis patients.31
activities of alkaline phosphatase or bone alkaline phosphatase are usually
normal or low; downward trends may indicate the development of                   Treatment of Adynamic Bone Disease
                                                                          -
dent on the choices of cotreatment (phosphate binders, vitamin D                 PTH overexpression and restore adequate PTH levels, without triggering
metabolites) and nutritional status. Particularly in instances of calcium        the progressive development of secondary hyperparathyroidism. Such
and phosphate loading, hypercalcemia and hyperphosphatemia may                   a stepwise treatment approach may include avoidance of calcimimetics,
be pronounced because adynamic bone is unable to buffer calcium and              reduction or withdrawal of active vitamin D metabolites, reduction or
phosphate loads by osseous deposition (Fig. 84.15).                              withdrawal of calcium-containing phosphate binders, and reduction
                                                                              associated with a fall in serum PTH because of the higher calcium load.
                                                                              In an observational study, high-dose calcium-containing phosphate
                                                                              binder intake was associated with both low bone turnover and increased
              Extraskeletal Calcification in                                  aortic calcification.39
                Adynamic Bone Disease
                                                                              studied. They include optimized diabetes control, a change from PD
                         Normal bone turnover                                 to HD to facilitate a more flexible dialysate calcium prescription, the
                                                                              administration of recombinant PTH (e.g., for patients after total para-
           Deposition                            Mobilization
                                                                              thyroidectomy), and calcilytics (agents that antagonize the calcium
                                                                              receptor and thus increase endogenous PTH).40
           Ca2+
                                   Bone
           PO4
                                                                              OSTEOPOROSIS IN CHRONIC KIDNEY DISEASE
                                                                              Whereas abnormal bone is common and fracture risk is increased in
                           Low bone turnover                                  CKD patients, the relative contribution of classic osteoporosis (as defined
                            (adynamic bone)
                                                                              complex is not well defined. Data from studies of antiosteoporosis agents
                                                                              are mostly available for patients in CKD stages 1 to 3, and subjects with
                                                                              features of CKD-MBD were largely excluded. Nevertheless, postmeno-
           Ca2+
                                                                              pausal women and elderly men are highly prevalent in late-stage CKD
                                   Bone
                                                                              populations, and it is thus likely that classic osteoporosis also contributes
           PO4                                                                to their bone disease.
                                                                                                                              Normal or
                                                                 Low bone           Normal              Low bone               increased
                                            Normal                 mass             bone                  mass                bone mass
                                          bone mass                                  mass
             Bone                                                                                                            Decreased
             Composition:                 Normal               Normal                                                        secondary
                                          primary              primary            Abnormal                High              mineralization
                                       mineralization       mineralization         primary             secondary
                                                                                mineralization        mineralization         Increased
                                                                                                                           osteoid volume
Measured BMD: 1.250 g/cm2 0.625 g/cm2 0.625 g/cm2 0.625 g/cm2 0.625 g/cm2
             Densitometry
                                       Normal BMD                                         “Osteoporosis”
             Report:
                    Fig. 84.16 The value of bone density measurements in the assessment of chronic kidney disease
                    (CKD)–related bone disease. Pink boxes indicate mineralized bone; red boxes indicate osteoid. BMD,
                    Bone mineral density. (Courtesy Prof. M. H. Lafage-Proust, St. Etienne, France.)
mineralization and increased osteoid, which is quite atypical for osteo-       hypocalcemia when treated with denosumab; coadministration of vitamin
porosis. Typical pathogenetic factors of osteoporosis, including hypoes-       D analogues may be required to blunt this effect. However, given the
trogenemia, immobilization, and corticosteroid use, are frequent in            growing evidence that antiresorptive therapies may be effective at least
CKD patients, although some postmenopausal women with late-stage               in patients with CKD stages 3 and 4, and the lack of definite evidence
CKD may have relatively normal estrogen levels. However, the sum of
CKD-MBD–related biochemical disturbances probably represents the               to perform a bone biopsy before treatment initiation, if a clinical response
decisive factors as to which bone phenotype predominates. Secondary            is otherwise suspected.
(see Fig. 84.16). However, there is recent evidence that bone mineral          involve three events:
                                                                               1. Pronounced renal retention of β2-microglobulin (11.8 kDa), leading
patients to an extent similar to that in the general population, so this          to plasma levels that can be elevated up to 60-fold in dialysis patients48;
method may become useful in monitoring therapeutic drug effects or                however, even massive overproduction of β2-microglobulin in mice
                                                                                  was not sufficient to induce amyloid deposits and thus further steps
from CKD-MBD–associated bone phenotypes. Peripheral quantitative                  must be important.49
tomography (pQCT) of the radius may be a superior methodology for              2. Modifications of the β2-microglobulin molecule that render it more
assessment of CKD patients in the future but awaits validation in suf-            amyloidogenic, such as limited proteolysis or the formation of dif-
ficiently large patient cohorts.41 The only reliable methodology to diagnose      ferent sugar-protein cross-links.50
osteoporosis and discriminate it from other bone manifestations in             3. Local factors that contribute to and determine the spatial localiza-
CKD patients is bone biopsy. In a large bone biopsy study, including              tion of the amyloidosis.
1429 samples from dialysis patients, osteoporosis was diagnosed in 52%
                                                       42
                                                          These proportions    Epidemiology
may be quite different in patients in earlier CKD stages, but there are        Histologic studies from the 1990s observed amyloid deposits in 100%
no systematic data available on such cohorts.                                  of dialysis patients treated for more than 13 years.51 Most amyloid
                                                                                                                                                  β2M
Treatment of Osteoporosis in Chronic                                           amyloid deposition are age at onset of renal replacement therapy (RRT)
Kidney Disease                                                                 and the duration of (nontransplant) RRT.48-50 β2M amyloid–related
Post hoc analyses of large prospective treatment studies using antios-         symptoms presently are largely confined to patients who have been
teoporotic medications indicate that it is safe and efficacious to treat       dialyzed for more than 15 years.
postmenopausal women in stages CKD 1 to 3 if they have a high risk
                                                                               Clinical Manifestations and Diagnosis
             43-46
CKD-MBD.         In such populations, bisphosphonates, denosumab,                β2M amyloidosis mainly manifests at osteoarticular sites, particularly
raloxifene, and teriparatide appear to be feasible therapeutic options.        synovial membranes; visceral manifestations are rare.48-50 Carpal tunnel
The former three drugs antagonize high bone turnover with an anti-             syndrome occurs and symptoms typically worsen at night and during
resorptive mode of action; teriparatide exerts bone anabolic effects
(PTH analogue). With bisphosphonates, there may be concerns of an              of peripheral joints, resulting from amyloid deposition in periarticular
extended oversuppression of osteoclasts, but only in patients with             bone and the synovial capsule (Fig. 84.17), is characterized by recurrent
                                                                               or persistent arthralgias, stiffness of large and medium-sized joints, and
from the Following Rehabilitation, Economics and Everyday-Dialysis             swelling of capsules and adjacent tendons. Recurrent joint effusions
                                                                               and synovitis, often in the shoulders and knees, may occur. The clinical
reduced fracture risk with denosumab treatment versus placebo, inde-           presentation may vary from frank, acute arthritis to slow, progressive
pendent of the stage of CKD.47 In contrast, for patients in CKD stages         destruction of the affected joints. Destructive spondyloarthropathy (Fig.
3 to 5 with features of CKD-MBD, no data are available on the safety                                     β2M amyloidosis can manifest as asymptomatic
and efficacy of any of these antiosteoporotic medications. In CKD              deposits, radiculopathy, stiffness, “mechanical ache,” and, finally, medul-
                                                                               lary compression with resulting paraplegia or cauda equina syndrome.
osteoclast paralysis. In CKD patients with secondary hyperparathyroid-                                                                                   -
ism, these antiresorptive agents may upregulate PTH secretion. Patients        ing in bent fingers that cannot completely extend or straighten) resulting
with advanced CKD (stages 4 and 5) may develop particularly severe             from amyloid deposits along the flexor tendons of the hands (Fig. 84.19).
Patients undergoing dialysis also can have subcutaneous tumorous                  see also Fig. 84.17). Such bone defects are prone to pathologic fractures.
               β2M amyloid; however, diffuse infiltration of the subcu-                                     β2M amyloid–induced cystic bone radiolucen-
taneous fat or skin has not been observed.                                        cies have been published.52 They include (1) diameter of lesions more
    Case reports of clinically relevant organ manifestations are usually          than 5 mm in wrists and more than 10 mm in shoulders and hips, (2)
in patients treated with HD for more than 15 years and have described             normal joint space adjacent to the bone defect, (3) exclusion of small
heart failure, odynophagia (painful swallowing), intestinal perforation           subchondral cysts in the immediate weight-bearing area of the joint
of both small and large bowel, gastrointestinal bleeding and pseudoob-            and of defects of the “synovial inclusion” type, (4) increase of defect
struction, gastric dilation, paralytic ileus, persistent diarrhea, macroglossia   diameter of more than 30% per year, and (5) presence of defects in at
or functional tongue disturbances (abnormal taste, mobility, articula-            least two joints. Scintigraphy, using either radiolabeled serum amyloid
tion), ureteral stenosis, and renal calculi.                                      P component or β2-microglobulin,53 offers more specific detection of
                                                                                  amyloid deposits but is not widely available. The definitive diagnosis
Diagnosis                                                                              β2M amyloidosis relies on histologic findings. Fat aspiration and
Serum levels of β2M do not distinguish between patients with amy-                                                   β2M amyloidosis, but diagnostic mate-
                                                                        β 2M      rial can be obtained from synovial membranes, synovial fluid, or bone
amyloidosis as thickening of the joint capsules of the hip and knee,              lesions.48
biceps tendons, and rotator cuffs, as well as the presence of echogenic
structures between muscle groups and joint effusions.48-50                        Treatment and Prevention
examination, affected joints may present with single or multiple jux-                                  β2
taarticular, “cystic” (i.e., amyloid-filled) bone radiolucencies (Fig. 84.20;     and physical and surgical measures, such as carpal tunnel
                            A                                                     B
                     Fig. 84.18 Aβ2M amyloidosis–associated spondyloarthropathy. (A) Destruction of an intervertebral
                     disk (arrow) in the neck vertebrae of a long-term hemodialysis patient. (B) Magnetic resonance image of the
                     same patient as in A. Note destruction of the intervertebral space and protrusion of material into the spinal
                     canal (arrow).
A B
         C                                                                  D
                   Fig. 84.20 Peripheral bone cystic radiolucencies in Aβ2M amyloidosis. Radiographic findings in a
                   long-term hemodialysis patient. (A) Multiple cystic lesions (arrows) are present in the hand bones. (B) Large
                   cysts (arrows) in the neck of the femur and adjacent pelvic bones. (C and D) Anterior and lateral views of
                   the head of the tibia with two very large, cystic lesions (arrows) resulting in posterior bulging of the tibial
                   plateau.