Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

21  Urologic Aspects of Pediatric Nephrology

Michael C. Braun, MD, and Chester J. Koh, MD

INTRODUCTION Glomerular Filtration Rate


Although many parents and patients consider them to be the same Glomerular filtration rate (GFR) refers to the volume of filtrate formed
specialty, pediatric urologists and pediatric nephrologists train through by the movement of plasma across the glomerular filtration barrier
very different pathways and often provide different viewpoints on per unit of time. GFR is influenced by a number of factors, one of
urinary tract abnormalities. This is especially with regard to surgical which is renal blood flow. This relationship between renal blood
and medical management, respectively, of the kidneys and the urinary flow and GFR is evident as changes in both occur during fetal and
tract, where the two specialties are tightly partnered in the treatment postnatal development (Schell et al., 2014). Prenatally, the percentage
of the anatomic and functional effects of urinary tract malformations of cardiac output received by the placenta is significantly greater
and acquired diseases. This chapter is designed to highlight topics than the fetal kidney. Postnatally, renal blood flow increases from
and viewpoints in pediatric nephrology of which pediatric urologists 3% to 4% of total cardiac output to between 12% to 16% in the
should be aware, where the collaborative expertise of pediatric first year of life (Quigley, 2012). The increase in neonatal renal blood
nephrologists and pediatric urologists is usually needed for optimal flow is caused by a combination of increased cardiac output and a
acute and chronic patient care. marked decrease in renal vascular resistance relative to the increase
in cardiac output. Several factors mediate the decrease in renal vascular
resistance. These include a redistribution of intrarenal blood flow
distribution, with an increase in intrarenal blood flow from the
RENAL FUNCTION, FLUID, AND ELECTROLYTE juxtamedullary glomeruli located in deeper regions of the renal cortex
HOMEOSTASIS to glomeruli in the outer renal cortex. Furthermore, the renin-
Renal Development angiotensin system (RAS) plays an important role regarding changes
in renal blood flow. Increasing levels of angiotensin II increase mean
Renal morphogenesis is a complex, temporally and spatially regulated arterial pressure and vasoconstriction of the renal efferent arteriole,
process by which precursor cells develop into a structurally and which in turn increases GFR by increasing glomerular capillary
functionally normal kidney. Abnormal or dysregulated renal develop- pressure. Plasma renin, angiotensin II, and angiotensin-converting
ment results in a wide range of renal abnormalities collectively known enzyme (ACE) levels increase in late gestation and postnatally
as congenital anomalies of the kidney and urinary tract (CAKUT) are higher in the first 2 weeks of life than in adulthood. Additionally,
and that compose the most common cause of end-stage renal prostaglandins and nitric oxide exert vasodilatory effects, lower renal
disease (ESRD) in children. In humans, kidney and urinary tract vascular resistance, and counterbalance the vasoconstrictive effects
development begins at approximately 3 weeks’ gestation with the of the RAS activation (Gleason, 1987).
formation of the initial urinary excretory precursor, the pronephros, Prenatal and neonatal GFR is significantly lower than adult
which undergoes complete involution (Combes et al., 2015; Little GFR levels (Schwartz et al., 2009). Because of placental effects,
et al., 2012; Short and Smyth, 2016). At roughly 4 weeks’ gestation, plasma creatinine concentrations in the first 48 hours after delivery
the mesonephros forms, followed at 5 weeks by the metanephros. reflects maternal rather than neonatal renal function. Under normal
Unlike the pronephros, the mesonephros is both functional and circumstances, by 7 days of life, creatinine levels fully reflect neonatal
integral to the mature urinary tract. In males, residual segments of renal function and are normally less than 0.5 mg/dL in term infants.
the mesonephric duct form the vas deferens, prostate, the seminal Subsequently, GFR doubles in term infants over the first 2 weeks of
vesicles, as well as part of the epididymis; segments of the mesonephric life and increases steadily until reaching adult levels by 2 years of age.
tubules form the testicular efferent ductules. In females, the meso-
nephric ducts persist as the epoophoron and paroophoron. The Tubular Function
metanephros, arising from metanephric mesenchyme, forms the
mature nephron segments. In contrast, the ureteric bud gives rise to Glomerular filtration is only the first step in the homeostatic processes
the renal collecting duct, the renal pelvis, and the ureters. By 34 to required of the kidney. Concurrent with the maturation of GFR,
36 weeks’ gestation, nephrogenesis is complete, and the structural there are morphologic and physiologic changes, particularly with
and functional relationship of each nephron segment is fully devel- respect to the renal tubules, that lead to maturation of the ability
oped. On average, 1 million (range 0.2 to 2.7 million) individual to maintain fluid and electrolyte homeostasis.
nephrons in each kidney arise from embryonic precursor cells. Both term and preterm neonates possess a higher total body
Concurrent with the morphogenesis of the kidney, functional water than older children and adults that is as much as 70% to
development of the fetal kidney also progresses with increasing 90% of total body weight (Gattineni and Baum, 2015). Although
gestational age. Prenatally, the placenta controls fluid and electrolyte neonates have the ability to dilute urine to as low as 50 mOsm/L,
homeostasis, and the primary function of the fetal kidney is the their urinary concentrating ability is limited, thus promoting negative
production of urine to maintain amniotic fluid volume. In the later free water balance. Normal free water loss is as much as 1% to 2%
stages of gestation, urine output, tubular function, and glomerular loss of total body weight per day over the first 5 days of life in term
filtration increase with gestational age. neonates and even higher in preterm neonates. There are a number
After birth, the neonatal kidney undergoes physiologic changes of factors that reduce the ability of the neonatal kidney to generate
to adapt to the extrauterine environment; rapid changes occur over a concentrated urine including decreased responsiveness to antidiuretic
the first several weeks and continue until reaching adult levels at 1 hormone (ADH) and immaturity of the renal tubules. The immaturity
to 2 years of life. of the tubules is both structural and functional. Structurally, the

341
342 PART III  Pediatric Urology

renal tubules increase in length with age, particularly the thick critical factor affecting this process is the postnatal increase in glu-
ascending limb of the loop of Henle. At birth, the activity of the cocorticoid production, which has been shown to promote an increase
sodium-potassium ATPase (Na+,K+-ATPase) exchanger, which drives in the number of NH3 antiporters, which are essential for ammonia
sodium transport, is low but increases in the immediate neonatal generation.
period, with a peak increase at around 2 to 3 weeks of life. Finally, At birth, neonatal serum levels of calcium are higher than
there is an increase in the concentration of urea in the medullary those seen in adults (Schell-Feith et al., 2010). There is a fall in
interstitium, which is required for the generation and maintenance serum calcium levels in the first day of life. The maturation of renal
of the countercurrent multiplier that is essential for the formation tubular calcium regulation is not fully understood. Although there
of concentrated urine. is a rise in serum parathyroid hormone (PTH) levels associated with
the falling calcium levels, neonates are typically in negative calcium
Developmental Changes in Glomerular Filtration Rate and balance marked by relative hypercalciuria. It is likely that neonates
Tubular Function have a blunted response to the effects of PTH either directly or
indirectly, which limits proximal tubule calcium reabsorption through
The maintenance of the balance of sodium is an important function both paracellular and transcellular pathways and calcium reabsorption
of the neonatal kidney. The ability of a neonate to conserve sodium in the distal tubule by the TRPV6 channel. Neonates have a much
in the first week of life is limited with an ensuing initial negative higher renal reabsorption of phosphate compared with adults. Renal
sodium balance during the first week of life, which shifts to a positive tubular reabsorption of phosphate can be as high as 99% immediately
sodium balance by 2 to 3 weeks of age. The natriuresis seen during after birth and remain greater than 90% during the neonatal period
the first week of life is mediated primarily by increased levels of (Kaskal et al., 1988). The reabsorption of phosphorus is mediated
atrial natriuretic peptide. The maturational responses to achieve largely via the tubular transporters, such as the 2Na-Pi IIc antiporter,
sodium homeostasis include elongation of the tubular segments, which are more active in the immature kidney. As circulating levels
which creates a larger surface area for reabsorption and upregulation and tubular responses to PTH increase after birth, the level of
of the number or activity of transporters in renal tubular cell mem- phosphorus reabsorption falls as the kidney matures.
branes. Additionally, endocrine and paracrine mechanisms including
the renin-angiotensin-aldosterone system (RAAS), catecholamines,
cortisol, and thyroid hormone drive sodium homeostasis. Elevated KEY POINTS
levels of renin and aldosterone stimulate sodium reabsorption in • CAKUT is the most common cause of ESRD in children.
the proximal tubule, the thick ascending limb of the loop of Henle, • Prenatal and neonatal GFR is significantly lower than adult
and cortical collecting ducts. Similarly, increased levels of catechol- GFR levels.
amines directly or indirectly through the RAAS pathway stimulate • The maintenance of the balance of sodium is an important
sodium reabsorption. Furthermore, increases in both cortisol and function of the neonatal kidney.
thyroid hormone stimulate tubular sodium transporters. • The newborn kidney has a limited capacity to excrete
The newborn kidney has a limited capacity to excrete potassium potassium and tends to have higher serum potassium
and tends to have higher serum potassium values than older children. values than in older children.
The limited renal capacity of the neonatal kidney to excrete potassium
is thought to be caused by the reduced number and responsiveness
of the Na+,K+-ATPase exchanger and the potassium transporters in HEMATURIA (MICROSCOPIC AND GROSS)
the distal tubule and cortical collecting ducts. Normally, 65% to 75%
of filtered potassium is passively reabsorbed by the proximal tubule Hematuria is one of the most common reasons for referral to pediatric
(Palmer, 2015; Zhou et al., 2004). The remainder of potassium nephrologists and urologists. Hematuria can present as either gross
reabsorption occurs in the thick ascending limb of the loop of Henle hematuria with red or “tea-colored” urine, or microscopic hematuria
via NKCC2, and much smaller amounts are reabsorbed in the distal with yellow urine with a positive dipstick for blood. Microscopic
tubule and cortical collecting ducts via the Na+,K+-ATPase exchanger. hematuria is defined as clear yellow urine with a positive urine
Urinary potassium excretion however is predominantly mediated via dipstick for blood with greater than five red blood cells per high-power
the apical renal outer medullary potassium (ROMK) channel of the field in a freshly spun urine sample. Isolated microscopic hematuria
cortical collecting ducts. Although the levels of circulating aldosterone is reported to occur in approximately 1% of school-age children
are noted to be higher in neonates, the neonatal kidney has a decreased (Dodge et al., 1976; Vehaskari et al., 1979). Less than 25% of these
responsiveness to aldosterone activation. The postnatal increase in children will have hematuria on repeated testing, and most will
potassium secretory capacity is largely related to increased activity of have spontaneous resolution of hematuria within 1 year of pre-
the Na+,K+-ATPase exchanger and an increase in the number of ROMK sentation. The incidence of gross hematuria is difficult to ascertain,
channels in the cortical collecting ducts, which in turn increases tubular however, it is estimated to account for slightly more than 0.1% of
responsiveness to aldosterone. pediatric emergency room visits. Although isolated microscopic
In the neonate, the mechanisms of urinary acidification and hematuria is most commonly a benign condition, the differential
bicarbonate reabsorption are immature, and neonatal serum diagnosis is quite broad, and a definitive diagnosis is determined
bicarbonate levels are lower than that seen in adults. As renal in less than 30% of patients. Gross hematuria typically prompts
acidification and bicarbonate reclamation improve over the first 3 more urgent referral and evaluation, and in the majority of cases,
to 4 weeks of life, there is a steady rise in net acid excretion and an underlying diagnosis can be made.
plasma bicarbonate reabsorption (Quigley et al., 2004). The primary A detailed history should be obtained to determine if the hematuria
site of bicarbonate reabsorption is the proximal tubule, where 80% is painless, intermittent or persistent, and microscopic or gross.
of filtered bicarbonate is reabsorbed. The sodium-hydrogen exchanger Associated symptoms should be sought including a history of trauma,
3 (NHE3) and H+-ATPase, which both secrete H+ into the proximal abdominal pain, fever, dysuria, voiding dysfunction, rash, joint pain,
tubular lumen, drive HCO3 reabsorption in the proximal tubule. weight gain, swelling, exercise, headaches, or visual changes. A detailed
Carbonic anhydrase IV (CA), which catalyzes the conversion of family history should also be obtained with a focus on hematuria,
secreted H+ and filtered bicarbonate (HCO3) into carbon dioxide bleeding disorders, hearing loss, urolithiasis, sickle cell disease, and
(CO2) and water, are less active in the newborn, limiting the ability renal disease. Vital signs including blood pressure should be carefully
to absorb HCO3 and secrete an acid load. With maturation, the measured. A thorough physical examination should be performed,
activity of these mechanisms increases, resulting in a concurrent with particular attention to blood pressure, rashes, joint abnormalities,
increase in serum HCO3 concentration. The final step in urinary edema, abdominal or flank pain, abdominal masses, and perineal
acidification occurs through the secretion of H+ via the apical H+- abnormalities.
ATPase in the cortical collecting ducts. Maturational increases in the The critical step in the evaluation of hematuria is the examination
activity of this transporter is essential to acid-base homeostasis. A of a freshly voided urine sample that includes microscopic assessment.
Chapter 21  Urologic Aspects of Pediatric Nephrology 343

Grossly bloody or red urine is indicative of nonglomerular bleeding,


and brown or “tea-colored” urine is suggestive of a glomerular origin BOX 21.1  Causes of Hematuria in Children
of the hematuria. In all patients with gross hematuria, urine MACROSCOPIC
microscopy is essential to confirm the presence of red blood cells
Transient
(RBCs). The absence of RBCs in grossly bloody urine with a positive
urine dipstick for heme is strongly suggestive of hemoglobinuria Hypercalciuria/nephrolithiasis
or myoglobinuria. The urine sediment should be examined closely Glomerulonephritis (all types)
for the presence of crystals, which are suggestive of urolithiasis, and Cystitis
the presence of white blood cells or bacteria are suggestive of urinary Exercise
tract infection. A semiquantitative assessment of RBC number should Congenital anomalies of the urinary tract
be done, including RBC morphology, as uniformly normal RBC Benign urethrorrhagia
morphology is suggestive of lower urinary tract bleeding, and dys- Wilms tumor or bladder tumor
morphic or “crenallated” RBCs are suggestive of glomerular disease. Bleeding dyscrasia
Urine sediment should be carefully examined for the presence of Renal vein thrombosis
RBC casts, which define glomerular nephritis. In patients with
Papillary necrosis
suspected glomerular disease, a quantitative assessment of proteinuria
should be performed. Based on the history, physical findings, and Nutcracker syndrome
complete assessment of the urine, a logical and structured evaluation
can be performed. MICROSCOPIC
Patients with findings suggestive of glomerular disease, pro- Transient
teinuria, hypertension, edema, or evidence of systemic disease Hypercalciuria/nephrolithiasis
should have a baseline evaluation of their renal function, serum Glomerulonephritis (all types)
electrolytes, complete blood counts, serum albumin, complement Cystitis/pyelonephritis
C3 and C4 levels, and ASO or anti-DNase B titers. Referral to a Exercise
pediatric nephrologist is often warranted. Renal cysts, most commonly Congenital anomalies of the urinary tract
in polycystic kidney disease, can also present with gross hematuria.
Thin basement membrane disease
Patients with fever, abdominal pain, dysuria, or frequency should
have urine cultures done as urinary tract infections are the most Drugs
common cause of gross hematuria in children. A history of trauma, Interstitial nephritis
abdominal or flank pain associated with gross hematuria often Sickle cell disease/trait
accompanied with frank clots should prompt urologic evaluation,
and imaging by computed tomography (CT) is often warranted. In
patients with a family history of urolithiasis, gross hematuria,
abdominal or flank pain, and a history of passing gravel, imaging KEY POINTS
such as renal ultrasonography or CT should be done and also may
include a comprehensive evaluation for renal stones. Other rare • Isolated microscopic hematuria is reported to occur in
causes include urinary obstruction, tumors, coagulopathies, and approximately 1% of school-age children.
vascular malformations. • Most children with microscopic hematuria will have
The causes of hematuria in children are numerous (Box 21.1), spontaneous resolution within 1 year of presentation.
and the assessment of children with microscopic hematuria is based • Gross hematuria and a urinalysis with no RBCs and a
on associated history and physical features combined with urinary positive heme dipstick is strongly suggestive of
findings. In patients with an unremarkable history including family hemoglobinuria or myoglobinuria.
history and who are asymptomatic and have only isolated microscopic
hematuria without proteinuria, the evaluation can be limited to
testing the parents for hematuria and measurement of urinary calcium
excretion. The value of urinary tract imaging in children with ratio in a newborn is less than 0.7 mg/mg, and less than 0.2 mg/
microscopic hematuria is controversial. The yield of routine ultra- mg in older children. If a timed collection is obtained, physiologic
sonography is low, and reported findings are frequently of little urinary protein excretion is typically less than 4 mg/m2/h. Healthy
clinical significance. When microscopic hematuria persists for several adolescents and adults excrete less than 150 mg of protein in a
months, decisions on the need for urinary tract imaging should be 24-hour period. Urinary protein consists of both filtered plasma
based on associated clinical findings. Voiding cystourethrograms proteins and secreted tubular proteins. Although most filtered proteins
and cystoscopy are rarely helpful and are not indicated for the routine are of low molecular weight, albumin accounts for nearly 30% of
evaluation of children with isolated microscopic hematuria. In patients the total protein found in “normal” urine.
with a positive family history, personal history, abnormal physical Proteinuria is categorized as either transient, orthostatic, or fixed
findings, or urinary findings in addition to hematuria, a more (Box 21.2) (Hogg et al., 2000; Vehaskari and Rapola, 1982). Transient
comprehensive evaluation is recommended. Typically this would proteinuria is characterized by disappearance of proteinuria after
include a complete blood count, serum electrolytes, creatinine, a positive test and accounts for more than 75% of patients with
albumin, C3, C4, ASO titer, antinuclear antibody (ANA) testing, isolated proteinuria. Typically, transient proteinuria occurs in
and a quantitative measurement of urinary protein excretion. association with fever or exercise. In an otherwise healthy child,
Additional testing such as hearing screening, hemoglobin electro- transient proteinuria is a benign condition. Orthostatic proteinuria
phoresis, imaging, and genetic testing should be individualized. commonly occurs in adolescents and is defined as proteinuria only
when the patient is in an upright position. Patients are otherwise
healthy and do not have other evidence of renal disease. A diagnosis
PROTEINURIA of orthostatic proteinuria is made by the measurement of urinary
protein in the first-void urine and in a second sample after the patient
Proteinuria is another common finding that prompts referral to a has been upright for several hours (Abitbol et al., 1990). In orthostatic
pediatric subspecialist. It is important to recognize that urine proteinuria, the initial first-void urine sample has normal protein
contains protein under physiologic conditions and that children values (protein-to-creatinine ratio <0.2 mg/mg), and the upright
excrete urine free of protein frequently when evaluated by the urine has increased levels of urine protein. Orthostatic proteinuria is
semiquantitative dipstick method. Urinary protein excretion varies usually transient and benign in nature; however patients should be
by body mass and renal maturity. A normal urine protein-to-creatinine closely evaluated to confirm that they are otherwise asymptomatic and
344 PART III  Pediatric Urology

BOX 21.2  Causes of Proteinuria BOX 21.3  Common Age Ranges for Presentation of
Glomerular Diseases
Transient
Exercise YOUNGER THAN 1 YEAR OF AGE
Fever Congenital infections
Stress/illness Diffuse mesangial sclerosis
Orthostatic Genetic diseases:
Fixed Congenital nephrotic syndrome (CNS) and infantile nephrotic
Drugs (chemotherapy, aminoglycosides, heavy metal syndrome
intoxication) Denys-Drash and Frasier syndromes
Tubular disease (acute tubular necrosis, interstitial nephritis,
cystic kidney diseases, Fanconi syndrome, graft-versus- 1 TO 10 YEARS OF AGE
host disease) Acute poststreptococcal glomerulonephritis (GN)
Reflux nephropathy Minimal change nephrotic syndrome (MCNS)
Glomerulonephritis: acute and chronic, all forms, including Focal segmental glomerulosclerosis (FSGS)
minimal change nephrotic syndrome Henoch-Schönlein purpura (HSP)
Other chronic renal disease (obstructive uropathy, congenital Hemolytic uremic syndrome (HUS)
renal dysplasia, permanent residual dysfunction from
acute disease [e.g., cortical necrosis, hemolytic uremic 7 YEARS OF AGE AND OLDER
syndrome, glomerulonephritis]) Systemic lupus erythematosus (SLE)
Diabetes mellitus Systemic vasculitis
Protein overload syndromes (hemolysis, rhabdomyolysis, Idiopathic crescentic GN
hypergammaglobulinemia) Membranoproliferative GN (MPGN)
Immunoglobulin A nephropathy
Idiopathic and secondary FSGS
Membranous nephropathy (MN)
have normal renal function and blood pressure. It is recommended Alport syndrome
that patients with orthostatic proteinuria be followed annually to Nail-patella syndrome
ensure that there is no progression in their clinical findings. Fixed Pulmonary-renal syndromes (Goodpasture syndrome
proteinuria is persistent and not dependent on postural changes. [antiglomerular basement memebrane disease])
Protein excretion should be quantified either by a timed collection
if possible or by first-void urine protein-to-creatinine ratio. Fixed
proteinuria requires a more thorough evaluation.
The differential diagnosis of proteinuria is quite broad, and the
evaluation of patients with persistent proteinuria should be focused GLOMERULAR DISEASE
on identifying the underlying etiology of the proteinuria. It is essential
to obtain a complete and thorough history and physical examination Glomerular diseases in children have several etiologies, and they
including a comprehensive family history with attention to family commonly present in specific age ranges (Box 21.3).
members with autoimmune disease, renal disease, diabetes, and
ESRD. Associated symptoms should be sought including a history Nephrotic Syndrome
of fever, gross hematuria, excessive urinary foaming, growth failure,
rash, joint pain, weight gain, swelling, headaches, or visual changes. Nephrotic syndrome is a constellation of clinical findings common
Vital signs including blood pressure should be carefully measured. to a number of glomerular diseases. These features include proteinuria
A thorough physical examination should be performed with particular greater than 40 mg/m2/h or a urinary protein-to-creatinine ratio
attention to blood pressure, rashes, joint abnormalities, and edema. greater than 2.0 mg/mg, hypoalbuminemia, and hyperlipidemia.
Laboratory investigation should be tailored to findings on the history The reason for the increased hepatic production of lipoproteins
and physical examination, but often include measurement of serum during the nephrotic state is not entirely understood, but it appears
creatinine, albumin, electrolytes, complete blood count, cholesterol, to be associated with low plasma oncotic pressure and alterations
and triglycerides. If the patient has evidence of systemic disease in lipoprotein metabolism. Although nephrotic syndrome may
or glomerular nephritis, testing should include C3, C4, and ANA present as a feature of glomerular nephritis, it presents more
screening. Further diagnostic testing will be dependent on the results commonly in children in isolation.
of these screening tests. If the screening studies are normal and the Nephrotic syndrome of childhood has been divided into three
patient has persistent proteinuria, renal biopsy may be warranted broad groups: congenital/infantile, primary (inherited or idiopathic),
to determine the underlying diagnosis. and secondary. Approximately 10% of children have an identifiable
secondary cause of nephrotic syndrome. The histologic lesions that are
associated with secondary causes of nephrotic syndrome are frequently
indistinguishable from the idiopathic lesions, but the treatment is
targeted to the primary underlying cause in these cases. Over the past
KEY POINTS two decades, mutations in nearly 30 genes have been identified in
• It is important to recognize that urine contains protein either inherited or sporadic primary nephrotic syndrome.
under physiologic conditions and especially with fever and Primary nephrotic syndrome is the occurrence of the constellation
exercise. of clinical findings that define NS in the absence of an identifiable
• Transient proteinuria accounts for more than 75% of causative agent or disease. Primary NS is classified into four categories
patients with isolated proteinuria. based on histologic findings: minimal change nephrotic syndrome
• If a patient with proteinuria has evidence of systemic (MCNS), mesangial proliferation (MES), focal segmental glomeru-
disease or glomerular nephritis, testing should include C3, losclerosis (FSGS), and membranous nephropathy (MN). Nephrotic
C4, and ANA screening. syndrome is classified by responsiveness to steroid therapy and
histopathologic features if a biopsy is performed.
Chapter 21  Urologic Aspects of Pediatric Nephrology 345

Steroid-sensitive nephrotic syndrome (SSNS) typically presents The peak incidence is in preschool children, with a median age of
with dependent edema with variable swelling of the eyelids, scrotum, 2.5 years. MCNS is usually idiopathic in childhood, although there
and labia. Hypertension is not typical. Microscopic hematuria is is a wide range of secondary causes of MCNS and an increasing
seen in approximately 20% of patients. Macroscopic hematuria is number of genetic mutations linked to MCNS. The cornerstone of
unusual. therapy for MCNS is corticosteroids, which will induce remission
Corticosteroids will induce a remission in the vast majority in nearly 90% of children. Up to 85% of patients achieve long-lasting
of children with nephrotic syndrome. Therefore, for those with a remission. However, a subset of patients will continue to experience
typical clinical presentation, a kidney biopsy is not indicated before relapses into adulthood.
initiating steroid therapy. Atypical features that may require a kidney Focal segmental glomerulosclerosis (FSGS) is seen in the majority
biopsy before treatment include age younger than 1 year at presenta- of children with SRNS. FSGS may be primary or secondary. As opposed
tion, macroscopic hematuria, hypertension, hypocomplementemia, to MCNS, less than 25% of patients with FSGS respond to steroid
and extrarenal symptoms such as a rash or arthritis. therapy. In children, primary FSGS typically presents between 2 and
Edema is mild in most patients and can be managed with dietary 7 years of age, with a higher incidence among African-Americans,
restriction (Teoh et al., 2015). The current standard of care is to treat Hispanics, and Asians. Genetic etiologies of steroid-resistant FSGS
these children with corticosteroids. Most children receive initial have been identified with increasing frequency. The vast majority of
treatment with prednisone (60 mg/m2/day or 2 mg/kg/day). The children with idiopathic FSGS present with nephrotic syndrome that
current definition of steroid responsiveness is a resolution of pro- is indistinguishable from SSNS. Definitive diagnosis requires kidney
teinuria within 8 weeks of starting steroid therapy. Once the urine biopsy. Current guidelines recommend initial treatment of primary
become negative for protein, the prednisone is switched to alternate FSGS with high-dose prednisone for durations of 4 to 16 weeks or
days and tapered over 6 weeks or longer. Almost 50% of children until complete remission. CNIs are often recommended for patients
with SSNS will experience multiple relapses (Teeninga et al., 2012). who are resistant or intolerant to corticosteroids. FSGS can recur
Frequently-relapsing nephrotic syndrome (FRNS) is defined as after kidney transplantation. Treatment with plasmapheresis, B-cell
two or more relapses within 6 months of initial response to steroid depletion, and CNIs has been reported to be effective in some patients.
therapy or four or more relapses within any 12-month period. In However, many will lose their graft and return to dialysis.
this setting, the potential adverse effects of the cumulative steroid Membranous nephropathy (MN) is a rare cause of nephrotic
doses required to achieve sustained remission begin to exceed the syndrome in childhood. The majority of childhood cases of MN are
benefits of maintaining nephrotic syndrome remission. Steroid- secondary to an underlying disorder; autoantibodies to phospholipase
sparing strategies have also been developed to treat FRNS including A2 receptor (aPLA2R) are frequently seen in children and adolescents
alkylating agents such as oral cyclophosphamide, mycophenolate with primary MN (Cossey et al., 2013). Hepatitis serology, serum
mofetil (MMF), B-cell depletion with rituximab, and calcineurin complement levels, and ANA testing should be obtained. A kidney
inhibitors (CNIs). biopsy is required to establish a diagnosis. On biopsy, MN is character-
Steroid-dependent nephrotic syndrome (SDNS) is defined when ized by the presence of subepithelial immune deposits. There are no
a child with NS develops two consecutive relapses during steroid universally accepted treatment guidelines for children with MN. The
therapy or when the ability to achieve and/or remain in remission use of ACE inhibitor or angiotensin receptor blocker (ARB) therapy
from nephrotic syndrome requires continuous administration of and a low-sodium diet are recommended. Corticosteroids alone can
corticosteroids. Steroid-sparing strategies in this population include decrease proteinuria, but rarely leads to long-term remission when
low-dose alternative-day corticosteroids, MMF, B-cell depletion with used in isolation. For this reason, corticosteroids are often used
rituximab, and CNIs. Children with SDNS tend not to respond as combined with an alkylating agent. CNIs are also commonly used.
well to alkylating agents as the FRNS subgroup. Recent data suggest that MMF and rituximab may be efficacious in
Steroid-resistant nephrotic syndrome (SRNS) is seen in 15% MN secondary to PLA2R-AB. MN is typically a slowly progressive
to 20% of children with idiopathic NS and in a small subset of disease, with 5% of children developing ESRD 5 years after diagnosis.
patients who were initially steroid responsive. It is defined as a Persistence of nephrotic syndrome is associated with significant
failure to achieve remission after 6 to 8 weeks of full-dose daily morbidity and even mortality. Because nephrotic syndrome frequently
prednisone. A biopsy is often recommended for steroid-resistant follows a chronic relapsing course, children are at risk for several
patients before initiating alternative therapies as there is a much recognized long-term complications. Spontaneous bacterial peritonitis
higher prevalence of FSGS in this subset of patients (Lovric et al., occurs in 2% to 6% of these children and is typically caused by
2015). A wide range of therapies has been utilized in patients with Streptococcus pneumoniae or gram-negative organisms. Thromboem-
SRNS, with varying degrees of efficacy including intravenous pulse bolism is one of the most serious complications of nephrotic
steroids, combined treatment with high-dose corticosteroids and syndrome. Virtually all nephrotic patients are in a hypercoagulable
alkylating agents, and cyclosporine or tacrolimus (Büscher et al., state, and as many as 20% experience thrombotic events. Mortality,
2016). Patients who achieve remission with CNIs are reported to although rare, is most frequently caused by infectious complications
have a high frequency of relapse when these agents are discontinued. and less commonly by thromboembolic events.
Patients who are refractory to steroids and CNIs rarely respond to
other therapies. Glomerulonephritis
Congenital nephrotic syndrome (CNS) is defined as nephrotic
syndrome presenting within 3 months after birth. CNS can be caused As opposed to isolated nephrotic syndrome, glomerulonephritis
by either genetic defects or perinatal infections. Proteinuria often classically presents with hematuria (gross or microscopic), pro-
begins in utero and can be detectable in the first urine sample after teinuria, hypertension, and acute kidney injury. Inflammation
birth. In cases of CNS, immunosuppressive therapy is not recom- leads to decreased renal function and retention of salt and water
mended as patients have been shown to be nonresponsive to these with ensuing hypertension and edema. Urinary features typically
agents. Conservative management of edema consists of sodium and include “tea-colored” or “cola-colored” urine, with a urine sediment
fluid restriction and intermittent intravenous albumin and loop characterized by dysmorphic erythrocytes and red blood cell casts
diuretics. The management of these patients also includes a hyper- (Kidney Disease Improving Global Outcomes [KDIGO], 2012).
caloric diet, thyroid hormone replacement, monitoring for thrombotic Although glomerular diseases can occur in almost any age group,
episodes, and aggressive management of infectious complications. most have a characteristic age range for disease onset. A number of
Minimal change nephrotic syndrome (MCNS) is the most systemic diseases may have their initial presentation with glomeru-
common cause of nephrotic syndrome in childhood, accounting lonephritis, so a careful examination for signs and symptoms of
for 90% of patients presenting before 10 years of age and 70% to multisystem disease should be sought. It is essential to recognize
80% of all pediatric patients with nephrotic syndrome. MCNS is that many patients with chronic glomerulonephritis may be
characterized by response to corticosteroids and often a chronic asymptomatic in the early stages of disease progression. Therefore
relapsing course. Most patients have an excellent long-term prognosis. it is essential to determine if there is evidence of chronic kidney
346 PART III  Pediatric Urology

disease (CKD) at the time of initial evaluation. Features of CKD progressive glomerulonephritis. Usually other systemic manifestations
include anemia, growth failure, and the hallmarks of chronic kidney of SLE are apparent. The presence of 4 out of 11 American College
disease/mineral bone disease (CKD/MBD): hyperparathyroidism, of Rheumatology classification criteria for SLE confers a 96% sensitivity
hypocalcemia, and hyperphosphatemia. Initial laboratory evaluation and specificity for the diagnosis of SLE. Almost all patients have
of children with glomerulonephritis is focused on defining the severity hypocomplementemia, positive antinuclear antibodies, and anti–
of acute kidney injury, any metabolic derangements associated with double-stranded DNA titers. No combination of serologic markers
decreased renal function, and whether the child has evidence of effectively predicts the histologic type or severity of kidney involve-
hypocomplementemia which, if present, significantly narrows the ment. The treatment of patients with mild renal disease (classes I
differential diagnosis. Hypocomplementemia, defined by a low C3 and II) should be determined by the extrarenal manifestations and
level when combined with normal C4 levels, is strongly supportive typically have a favorable renal prognosis. However, children with
of a diagnosis of acute postinfectious glomerulonephritis or C3 more extensive renal involvement (class III and IV) can have more
glomerulopathy, and a low C3 level and a low C4 level focuses the progressive renal dysfunction if left untreated. Consensus treatment
diagnosis on either lupus nephritis or membranoproliferative glo- recommendations developed for children with SLE include the use
merulonephritis. Normocomplementemia is a feature of the other of corticosteroids combined with immunosuppressive drugs in this
common forms of glomerulonephritis that are seen in the pediatric patient population (Mina et al., 2012). However, only 80% of patients
population. with class IV achieve sustained remission, and up to 50% of patients
Acute poststreptococcal glomerulonephritis is the most common will experience at least one relapse.
form of post-infectious glomerulonephritis (PIGN) in childhood.
However, many other bacterial, viral, and parasitic pathogens are Normocomplementemic Glomerulonephritis
also associated with PIGN. Typically the child has a history of
pharyngitis or pyoderma that precedes the presentation of glomeru- Immunoglobulin A (IgA) nephropathy is the most common primary
lonephritis by several weeks. Acute poststreptococcal glomerulone- cause of glomerulonephritis throughout the world including in
phritis is primarily a disease of school-age children. It is rare before the pediatric population. Patients with this condition commonly
3 years of age and may occur as a sporadic or epidemic disease. present in the second and third decades of life, and it is rare in the
Clinical symptoms begin abruptly, with most patients presenting first decade of life. The true incidence of IgA nephropathy is uncertain,
with edema and gross hematuria. Proteinuria is typically mild; however as there are no reliable serologic markers, and a renal biopsy is the
some patients can present with features of nephrotic syndrome. Mild only way to definitively make the diagnosis. IgA nephropathy is
to moderate hypertension is common, although some patients with defined by the presence of dominant IgA deposits in the glomerulus.
severe disease present with hypertensive urgency or emergency. Acute These deposits are polymeric IgA1 with aberrant glycosylation patterns
kidney injury is typically mild, although in severe cases, rapidly in the hinge region of the antibody (Kiryluk et al., 2011).
progressive glomerulonephritis (RPGN) can occur, necessitating renal Most commonly, these patients present with recurrent episodes
replacement therapy (RRT). Laboratory testing supportive of a of painless macroscopic hematuria. The onset of gross hematuria is
diagnosis of PIGN includes a serum C3 level usually below 50% of often concurrent with or immediately proceeded by an upper respira-
normal levels, and an elevated ASO or DNase B titer. In the vast tory tract infection. The latency period between the onset of infectious
majority of cases, with conservative management, spontaneous symptoms and gross hematuria is considerably shorter in IgA
improvement typically begins within 1 week, with resolution of the nephropathy than that seen in patients with PIGN. The gross
edema occurring in 5 to 10 days, and resolution of the hypertension hematuria episodes typically resolve within a few days, but microscopic
occurring in 2 to 3 weeks. In the majority of patients, the clinical hematuria and variable degrees of proteinuria may persist. Occasion-
presentation and laboratory testing that includes complement levels ally there are complaints of flank or loin pain associated with the
is usually sufficient to make the diagnosis of PIGN, and there is no gross hematuria. Many patients will also present with incidental
indication to perform a kidney biopsy to confirm the diagnosis. C3 findings of microscopic hematuria and mild proteinuria. Less com-
levels typically return to normal within 8 to 12 weeks after presenta- monly, patients will present with isolated nephrotic syndrome. Most
tion. However, if a child has persistently low C3 levels or has an of the acute episodes resolve without specific therapy, and the
atypical clinical course, a kidney biopsy may be indicated. Residual long-term prognosis remains relatively good. There are currently no
proteinuria may be present of up to 6 months after the acute injury, diagnostic markers for IgA nephropathy. A renal biopsy for suspected
and microscopic hematuria may persist for more than 1 year. IgA nephropathy may be considered in patients who have impaired
Two other forms of hypocomplementemic nephritis, although kidney function, hypertension, or significant proteinuria, as these
rare, can present in childhood: Membranoproliferative glomeru- individuals are at increased risk for developing ESRD. IgA nephropathy
lonephritis (MPGN) and C3 glomerulopathy (C3G), which is further is a disease with a highly variable outcome. For patients with self-
subdivided into dense deposit disease and C3-dominant glomeru- limited episodes of recurrent gross hematuria who do not develop
lonephritis (C3GN). These diseases typically present in late childhood significant proteinuria, the long-term outcome is quite good. Risk
and are rare before 5 years of age. The presentation of MPGN/C3G factors for progressive disease include persistent and significant degrees
is varied and includes asymptomatic hematuria or proteinuria, of proteinuria, hypertension, and an elevated serum creatinine. In
nephrotic syndrome, and features that initially mimic PIGN, including the high-risk patient population, kidney function slowly deteriorates.
low serum C3 levels in the remainder (Pickering et al., 2013). The Current data suggest that up to 20% to 30% of this population will
presence of nephrotic syndrome and a depressed serum C4 level are develop ESRD and another 20% have progressive CKD.
early clues that differentiate MPGN from PIGN. Persistent hypo- Henoch-Schönlein purpura (HSP) is a self-limited systemic
complementemia in a patient with suspected PIGN is highly unusual vasculitis syndrome of childhood characterized by symptoms that
and is suggestive of either G3G or MPGN. Although many MPGN may occur asynchronously over a period of several days to weeks.
patients have evidence of activation of the classical complement These features include a purpuric rash, arthralgia, abdominal pain,
cascade with low serum C3 levels and low C4 levels, and patients and glomerulonephritis. Nephritis rarely, if ever, precedes the onset
with C3G patients commonly have signs of alternative pathway of purpura. Histologically, the glomerulonephritis in HSP is indis-
activation with persistently low C3 and normal C4 levels, these tinguishable from idiopathic IgA nephropathy. Whereas IgA
distinctions are not absolute and a kidney biopsy is required to nephropathy is exclusively a renal disorder, HSP is characterized by
establish the diagnosis of MPGN/C3G. its extrarenal manifestations. The pathophysiology is similar in its
The final form of hypocomplementemic nephritis that presents involvement of glomerular deposition of polymeric IgA1 immune
in childhood is systemic lupus erythematosus (SLE) and should complexes (Kiryluk et al., 2011). Most patients with HSP nephritis
be considered a likely diagnosis in any child with evidence of present with asymptomatic hematuria and mild proteinuria, although
systemic disease, glomerulonephritis, and low C3 and C4 levels up to one-fourth of these patients develop gross hematuria. A small
(Hiraki et al., 2012). The clinical presentation of SLE ranges from subset of patients will present with an acute nephritis and/or nephrotic
asymptomatic microscopic hematuria or proteinuria to rapidly syndrome, and presentation with RPGN is rare. HSP remains a clinical
Chapter 21  Urologic Aspects of Pediatric Nephrology 347

diagnosis as there are no specific diagnostic markers for this condition. function at diagnosis. Some patients present with RPGN. ANCA are
Kidney biopsy is not necessary for diagnosis but can be helpful in detected in 85% to 95% of patients with active GPA (PR3-cANCA)
making treatment decisions when patients develop nephrotic syn- and 70% with MPA (MPO-pANCA). ANCA-associated systemic
drome or RPGN. HSP usually spontaneously resolves within 1 to 2 vasculitis is an aggressive disease. Without treatment, mortality rates
weeks, but recurrent episodes occur fairly commonly for the first 4 are 90% within 2 years. Patient and kidney survival have improved
months. Ultimately the vast majority of patients achieve full clinical dramatically with intensive immunosuppressive therapy, however
remission (Dudley et al., 2013). Persistent urinary abnormalities, relapses are common. CKD persists in as many as one-half of patients,
low-grade proteinuria, and microscopic hematuria is commonly seen with nearly one-fourth of patients eventually developing ESRD.
for up to 3 months after presentation and for up to 2 years in 10% Antiglomerular basement membrane (Anti-GBM) disease is an
to 20% of children. A rapidly progressive clinical course with ensuing aggressive form of glomerulonephritis that is rarely seen in childhood.
ESRD can be seen in a small percentage of patients. Patients with It is mediated by antibodies to the glomerular basement membrane.
persistent heavy proteinuria may see progression over a period of Known as Goodpasture syndrome when associated with pulmonary
several years. In a long-term follow-up study of patients with HSP, hemorrhage, it is rare in the first decade of life but has been reported
44% of patients with nephrotic syndrome or acute nephritis at in children as young as 2 years of age. Anti-GBM nephritis may occur
presentation developed hypertension or progressive CKD, compared as a rare complication after extracorporeal shock wave lithotripsy
with normal renal function in 82% of patients who presented with (ESWL), membranous nephropathy, or minimal-change disease.
isolated hematuria. Anti-GBM nephritis may also occur in a small subset of patients
Alport syndrome is a genetic disorder of the collagen type IV with Alport syndrome after kidney transplantation. The Goodpasture
α3, α4, or α5 chains that form the mature glomerular basement target antigen is the carboxy-terminal region (NC1 domain) of the
membrane. The primary presenting clinical finding in all forms of α3 chain of collagen type IV, the structural component of glomerular
Alport syndrome is persistent microscopic hematuria. Intermittent and pulmonary basement membranes. Patients present with acute
gross hematuria is also commonly seen. Sensorineural deafness is nephritis or less commonly with symptoms of CKD. Pulmonary
a common feature that distinguishes Alport syndrome from other hemorrhage presenting as hemoptysis often precedes or accompanies
forms of glomerular disease. Hearing deficits typically occur in late the onset of nephritis; these episodes of hemoptysis may be life-
childhood, are bilateral, and initially involves high-frequency hearing threatening. More than 90% of patients have anti-GBM antibodies
loss that gradually worsens over time. A family history of kidney at the time of clinical presentation. Progression to irreversible kidney
failure or deafness in a child with hematuria is highly suggestive of injury can occur within weeks. The mainstay of therapy is plasma-
Alport syndrome. Ocular anomalies may develop later in childhood, pheresis to remove the circulating antibody combined with
including macular disease and anterior lenticonus. Other rare associa- immunosuppressive therapy to reduce new antibody production.
tions include leiomyomata of the esophagus, trachea, and the external The kidney disease is unlikely to be reversible unless treatment is
genitalia in females. Alport syndrome has X-linked, autosomal started early. Patients who recover from the acute disease generally
dominant, and recessive forms of inheritance (Savige et al., 2016). do well, and disease relapses are extremely rare.
The majority of patients have X-linked mutations in COL4A5, the RPGN is a rare disorder in childhood that is traditionally divided
gene encoding the α5 chain of collagen type IV. Autosomal disease into three groups; anti-GBM nephritis, immune complex nephritis,
is caused by mutations in COL4A3 or COL4A4. Heterozygous muta- and pauci-immune disease associated with ANCA. Less commonly,
tions in COL4A3 or COL4A4 are inherited in a dominant fashion, other forms of proliferative glomerulonephritis may present as severe
and patients with mutations in both alleles present in a recessive crescentic disease. This clinical syndrome is characterized by sudden
manner. Kidney function is usually well preserved during the first onset and rapid decline in kidney function. It typically presents with
decade of life in males with X-linked Alport syndrome. Frank pro- gross hematuria, edema, anemia, and hypertension. Isolated nephrotic
teinuria frequently appears during late childhood or early adolescence syndrome is uncommon. Pulmonary hemorrhage suggests a diagnosis
and is followed by development of hypertension and renal insuf- of anti-GBM nephritis or ANCA vasculitis and may be fatal if treatment
ficiency (Savige et al., 2013). About 50% of males with X-linked is delayed. Serologic studies including a C3, C4, ANA, ANCA, and
disease reach ESRD by 25 years of age. Patients with autosomal anti-GBM antibodies should be obtained; however, a biopsy confirma-
recessive disease follow a course similar to males with X-linked tion is essential and establishes the diagnosis in most patients. The
Alport syndrome. There are no disease-specific treatments for Alport renal biopsy commonly shows extensive crescent formation. Therapy
syndrome, and therapy is focused on reducing the rate of progression is directed to the specific disease entity. Spontaneous resolution of
with RAAS inhibition to reduce proteinuric renal injury and to control RPGN is extremely unlikely. The prognosis is poor unless aggressive
hypertension (Kashtan et al., 2013). therapy is started early in the course of the disease.
Antineutrophilic cytoplasmic antibody (ANCA)-associated Hemolytic uremic syndrome (HUS) is a common cause of severe
glomerulonephritis is a rare disease in childhood. Two target ANCA acute kidney injury in a previously healthy young child. The majority
antigens have been identified: proteinase-3, or cANCA, and myelo- of children with HUS have an antecedent diarrheal illness caused
peroxidase, or pANCA. ANCA-associated glomerulonephritis in by Shiga toxin–producing Escherichia coli (STEC). Approximately
childhood is divided into two small-vessel vasculitic syndromes: 10% of HUS cases are not associated with STEC infections and are
microscopic polyangiitis (MPA) and granulomatosis with polyangiitis referred to as atypical HUS (aHUS). HUS, aHUS, and thrombotic
(GPA) (Cabral et al., 2016). Eosinophilic granulomatosis with thrombocytopenic purpura (TTP) are classified as forms of thrombotic
polyangiitis (Churg-Strauss syndrome) is extremely rare in childhood. microangiopathy (TMA), however the triggers, primary target organs,
Although ANCA-associated glomerulonephritis occurs primarily in response to treatment, and clinical outcomes are distinct. HUS occurs
adults, it can present in adolescence and occasionally during child- in both sporadic and epidemic patterns. More than 70% of children
hood. Nephritis is seen in 70% to 80% of children with either GPA in the United States with STEC-positive HUS have been infected
or MPA. The features of GPA and MPA overlap where both typically with E. coli 0157:H7. The primary reservoir for E. coli 0157:H7 is
present with a flulike prodrome and systemic symptoms such as farm animals, although contamination of fruits, juices, and vegetables
fever, anorexia, malaise, weight loss, myalgias, arthritis and/or is also common. HUS is initiated by damage to the endothelium
arthralgias, and skin lesions (purpura or urticaria). Lung disease with microthrombi formation, leading to the diagnostic triad of
occurs in both syndromes and can include life-threatening pulmonary microangiopathic hemolytic anemia, thrombocytopenia, and acute
hemorrhage. Granulomatous involvement of the upper airway, sinuses, kidney injury. In patients with aHUS, defects in regulation of the
nasal passages, and ears is present in many patients with GPA but alternative complement pathway are commonly found (Loirat et al.,
not in MPA. A small subset of patients present with renal-limited 2015). These include mutations in complement factor H, factor I,
vasculitis. Histologically, the principal finding is pauci-immune focal membrane cofactor protein, C3, complement factor B, and autoan-
necrotizing glomerulonephritis, which is similar in both GPA and tibodies against complement factor H and factor I. In young children,
MPA and is often severe. All patients with nephritis have hematuria, aHUS can occur as a complication of S. pneumoniae infection. Unlike
most have proteinuria, and more than 70% have decreased renal other patients with HUS, these patients have a positive Coombs
348 PART III  Pediatric Urology

anemia. Of note, the use of fresh-frozen plasma is contraindicated TUBULOPATHIES


in patients with pneumococcal-associated HUS.
After ingestion of E. coli–contaminated food, most affected children Renal tubular disorders frequently present during routine laboratory
develop abdominal pain and bloody diarrhea. The colitis is usually investigation of children with failure to thrive. Tubular diseases can
self-limited, but complications may occur, including rectal prolapse, be isolated defects as in the case of distal renal tubular acidosis
toxic megacolon, bowel wall necrosis, and bowel perforation. The (RTA), or generalized defects as seen in Fanconi syndrome, and are
onset of HUS is usually abrupt, occurring within 1 week after the primary or secondary in nature (Batlle and Haque, 2012). Tubu-
onset of the diarrhea. The presenting symptoms are often caused by lopathies such as renal glycosuria can result in solute wasting, and
acute kidney injury. The severity of the acute kidney injury is highly others such as Gordon syndrome result in excess solute reabsorption
variable, but one-half will develop oligoanuria and require dialysis. (O’Shaughnessy, 2015). Evaluation of patients with suspected tubular
Hypertension is variable but may be severe. The mean duration of dysfunction begins with a careful history and physical examination
the acute kidney injury is 2 weeks. In aHUS, the antecedent illness focusing on a family history of renal or metabolic disease, and signs
is more nonspecific, often associated with fever, upper respiratory of failure to thrive, poor feeding, recurrent vomiting, dehydration,
symptoms, and vomiting. Sometimes a prodromal illness is not irritability, tetany, seizures, or icterus. The initial evaluation of patients
apparent. Neurologic involvement is common and can mimic TTP. with suspected tubular disease should include a complete blood
Most patients are very irritable and somnolent, and seizures and count; a comprehensive metabolic panel including electrolytes, renal
coma can occur in 10% of children. Virtually any organ can become function, calcium, magnesium, and phosphorus; a venous or arterial
damaged by the microvascular thrombi. Anemia is characterized by blood gas; and urine for quantitative assessment of urine potassium,
the presence of schistocytes, negative Coombs test, elevated reticu- sodium, calcium, phosphorus, and magnesium excretion. In patients
locytosis and lactose dehydrogenase levels, and a low serum hapto- with suspected RTA, a metered urine pH should be obtained and
globin. The prothrombin and partial thromboplastin time are usually urine sodium, potassium, and chloride concentrations to measure
normal. Serum C3 level and CH50 may be depressed in both the urine anion gap. Additional testing including genetic testing
STEC-positive HUS and aHUS but is more common in those with should be based on the history and physical examination in combina-
aHUS; an abnormal serum C3 level does not rule out the possibility tion with underlying metabolic disturbances. RTA is defined as an
of a genetic complement deficiency. In patients with clinical features inappropriately elevated urine pH in the setting of metabolic acidosis.
suggestive of STEC-positive HUS, confirmation of a Shiga toxin– The hallmark of RTA is hyperchloremic metabolic acidosis (normal
producing E. coli either by culture or the presence of the Shiga toxin anion gap acidosis); the presence of metabolic acidosis must be
is recommended. When aHUS is a consideration, it is important to confirmed by measurement of venous or arterial pH. In the setting
measure ADAMT13 activity and inhibitor testing to rule out TTP of normal anion gap metabolic acidosis, a urine pH greater than
and to evaluate the complement system by protein and genetic testing. 5.5 and an elevated urine anion gap are highly supportive of RTA
For STEC-positive HUS, supportive medical care is by far the most (Carlisle et al., 1991).
important aspect of treatment. Meticulous attention must be paid Proximal RTA is defined by a decrease in the proximal tubule
to fluid and electrolyte management. Severe acute kidney injury bicarbonate reabsorptive capacity, resulting in a low bicarbonate
requiring dialysis still develops in nearly one-half of patients. threshold with ensuing bicarbonate loss. Patients present with
Packed red blood cell transfusions should be reserved for patients failure to thrive and hyperchloremic metabolic acidosis. However,
with clinical indications of severe anemia. Platelet transfusions should patients with proximal RTA can reabsorb filtered bicarbonate below
be avoided unless the patient is actively bleeding or the platelets the lower resorption threshold. Thus patients with proximal RTA
are needed in preparation for an invasive procedure. During the can maintain their serum bicarbonate level, although at a lower
prodromal phase of colitis, antimotility drugs are contraindicated, level than normal (typically 14 to 17 mEq/L). Because the distal
and antibiotics should be avoided as they have been reported to tubule is intact, when the serum bicarbonate level is at or below
increase the probability of developing HUS. For patients with aHUS, the absorptive threshold of the proximal tubule, patients with proximal
Eculizumab, a monoclonal antibody to the complement component RTA can excrete urine with a pH less than 5.5. Isolated proximal
C5, is the therapy of choice. It has been shown to be highly effective RTA is rare and is more commonly seen in conjunction with Fanconi
in patients with complement dysregulation. Acute mortality in syndrome. Isolated proximal RTA can be caused by autosomal recessive
STEC-positive HUS is related to severe complications such as neu- mutations in the sodium bicarbonate cotransporter. Patients with
rologic disease, cardiac failure, and multiorgan involvement. Kidney this disorder usually have ocular defects, short stature, and intellectual
function typically improves spontaneously, and almost all patients impairment. Treatment of proximal RTA is challenging when exog-
are able to discontinue dialysis. However, long-term follow-up studies enous alkali is not absorbed by the proximal tubule. Therefore,
suggest that 5% to 25% of patients will develop CKD. aHUS generally patients commonly require 10 to 15 mEq/kg/day of alkali with only
follows a more severe clinical course and is more likely to relapse modest improvement in serum bicarbonate levels.
in the absence of chronic complement inhibition. Recurrent disease Fanconi syndrome is a generalized disorder of the proximal tubule.
in either native kidneys or after kidney transplant is extremely rare Patients with Fanconi syndrome typically present with failure to
in STEC-positive HUS but is quite common in aHUS. Eculizumab thrive, hyperchloremic metabolic acidosis with hypophosphatemia,
treatment is also recommended for aHUS patients undergoing kidney and hypokalemia (Moe et al., 2009). Typical urinary findings include
transplantation. glucosuria, aminoaciduria, and phosphaturia. Cystinosis is a common
cause of Fanconi syndrome in children. It is an autosomal recessive
disorder resulting in accumulation of free cysteine in intracellular
lysosomes (Nesterova and Gahl, 2013). Presenting clinical features
typically include polydipsia, polyuria, constipation, and failure to
KEY POINTS thrive. Many patients have evidence of rickets caused by renal
• Corticosteroids will induce a remission in the vast majority phosphate wasting. Treatment of Fanconi syndrome is twofold;
of children with nephrotic syndrome. initially, the treatment of the underlying primary disease, if possible,
• Focal segmental glomerulosclerosis is seen in the majority and secondly, the replacement of urinary solute losses and the cor-
of children with steroid-resistant nephrotic syndrome. rection of acidosis.
• Minimal change nephrotic syndrome is the most common Children with distal RTA present with short stature and failure
cause of nephrotic syndrome in childhood. to thrive, and they often have rickets. Classically, they have hyper-
• IgA nephropathy is the most common primary cause of chloremic metabolic acidosis, hypokalemia, hypercalciuria, and
glomerulonephritis throughout the world. nephrocalcinosis. If the hypokalemia is severe, they may present
• Hemolytic uremic syndrome is a common cause of severe with muscle weakness, cramps, and paralysis. The chronic metabolic
acute kidney injury in a previously healthy young child. acidosis results in bone demineralization. Patients with distal RTA are
unable to excrete urine with a pH less than 5.5. Distal RTA may be
Chapter 21  Urologic Aspects of Pediatric Nephrology 349

either a primary or a secondary disorder. Primary distal RTA has been


linked to autosomal dominant or autosomal recessive mutations in the
KEY POINTS
basolateral Cl–/HCO3– exchanger and autosomal recessive mutations • The hallmark of RTA is hyperchloremic metabolic acidosis
in H+-ATPase that are also associated with hearing loss. Secondary (normal anion gap acidosis).
causes of distal RTA include medications and rheumatologic disorders. • Children with distal RTA classically have hyperchloremic
Distal RTA is treated by providing alkali equivalent to the amount of metabolic acidosis, hypokalemia, hypercalciuria, and
H+ ion normally secreted by the distal nephron. Typically 3 to 5 mEq/ nephrocalcinosis.
kg/day of alkali is sufficient in most children to normalize the serum • Hypokalemic metabolic alkalosis is the defining feature of
bicarbonate levels and correct the associated metabolic disturbances. Bartter syndrome.
RTA associated with hyperkalemia is also known as type 4 RTA. It • RTA associated with hyperkalemia is also known as type
is associated with a number of disorders of either mineralocorticoid 4 RTA.
deficiency or mineralocorticoid resistance. Decreased mineralocorti-
coid production is seen most commonly with 21-hydroxylase deficiency
because Addison disease is rare in children. Angiotensin-converting
enzyme inhibitor use can also result in functional hypoaldosteronism.
Hyporeninemic hyperaldosteronism in children can be the result of heterogeneous disorders is varied. Many patients are diagnosed
drugs such as calcineurin inhibitors, nonsteroidal anti-inflammatory prenatally or during the evaluation of hypertension, gross or micro-
drugs (NSAIDs), and tubulointerstitial disease. Other common causes scopic hematuria, abnormal renal function, polyuria, or incidentally
of type 4 RTA in children include obstructive uropathy and interstitial as part of an evaluation of abdominal pain or constipation. Often
renal disease. In these disorders, aldosterone secretion is normal, but a specific diagnosis can be determined by imaging alone or by family
damage to the cortical collecting tubule results in functional resistance history. However, genetic testing to achieve a definitive diagnosis is
to aldosterone. Typically the degree of hyperkalemia is out of proportion preferred. Currently there are no disease-specific treatment options
to the severity of the renal insufficiency. Evaluation and management that can alter disease progression or correct the underlying disorders.
of type 4 RTA should be focused on defining the underlying etiology Therefore, therapy is conservative in nature and focused on manage-
and treating the primary disease. Patients with glucocorticoid and ment of complications that might accelerate CKD progression.
mineralocorticoid deficiency should receive replacement therapy. Most Multicystic dysplastic kidney (MCDK) disease appears on
cases of type 4 RTA caused by obstructive uropathy need urologic ultrasonography as a large reniform mass with noncommunicat-
evaluation and surgery. ing cysts. The MCDK is nonfunctional and is usually unilateral;
Dent disease is an X-linked recessive disorder characterized by although rare, if MCDK is bilateral, neonatal mortality is nearly
low-molecular-weight proteinuria, nephrocalcinosis, recurrent universal. Urologic anomalies are seen in up to 25% of patients with
urolithiasis, and progressive renal insufficiency. Dent disease results MCDK, including renal hypoplasia/dysplasia, vesicoureteral reflux,
from a defect in the renal proximal tubule Cl–/H+ exchanger designated and ureteropelvic junction obstruction. Typically there is gradual
as CLC-5 (Devuyst et al., 2010). Patients with Dent disease have reduction in the size of an MCDK with eventual involution over a
low–molecular weight proteinuria, not glomerular proteinuria (i.e., period of months to years. The contralateral kidney typically shows
albumin). Treatment is directed at preventing renal calculi. Patients compensatory hypertrophy. Because of the association of MCDK
should have adequate fluid intake and eat a low-sodium diet. A with VUR, voiding cystourethrography (VCUG) is recommended,
thiazide diuretic may be helpful to decrease renal calcium excretion. particularly in patients with hydronephrosis and/or hydroureter.
ESRD usually occurs between the third and fifth decade of life. Serial monitoring of renal function, blood pressure, and renal
Bartter syndrome is an autosomal recessive disorder, which usually ultrasonography is generally recommended at an interval of 3 months
presents in the first year of life with failure to thrive. Typically, patients for the first year of life and then every 6 months up to involution,
have a prenatal history of polyhydramnios and prematurity, and or at least up to 5 years. There is emerging data that patients with
polydipsia and polyuria as infants and young children. A history of unilateral MCDK are at risk for development of early-onset hyper-
constipation, salt craving, and muscle cramps are also quite common. tension and possible CKD. As a result, there is growing consensus
Hypokalemic metabolic alkalosis is the defining feature of Bartter that these patients should be monitored annually until well into
syndrome (Seyberth and Schlingmann, 2011). Blood pressure is adulthood.
typically normal, but plasma renin and aldosterone levels are elevated Autosomal recessive polycystic kidney disease (ARPKD) is caused
as a result of volume depletion. Bartter syndrome is commonly by a mutation in the PKHD1 gene and occurs in approximately
caused by mutations in the Na/K/2Cl cotransporter or in Barttin, 1 : 40,000 live births (Bergmann et al., 2004). The kidneys are
which is associated with sensory neural deafness. Treatment of patients bilaterally enlarged as a result of fusiform cysts arising from the
with Bartter syndrome is centered on replacement of urinary solute collecting ducts. The principal extrarenal manifestation of ARPKD
loss and management of polyuria. is congenital hepatic fibrosis; over time, cystic dilation of the biliary
Gitelman syndrome is an autosomal recessive disorder that also tree develops with ensuing portal hypertension and liver failure. In
presents with hypokalemic alkalosis and can be confused with Bartter addition to complications related to CKD, patients with ARPKD are
syndrome (Seyberth and Schlingmann, 2011). Patients with Gitelman at risk for ascending cholangitis, recurrent urinary tract infections,
syndrome typically do not have failure to thrive, and there is no hypertension, nephrolithiasis, and bleeding from esophageal varices.
antenatal history of polyhydramnios. Hypomagnesemia is nearly Neonatal presentation with oligohydramnios/anhydramnios has a
universal in Gitelman syndrome, and urinary calcium excretion is reported mortality rate of 25% to 50%. Over 75% of patients with
low in Gitelman syndrome as opposed to Bartter syndrome, in which ARPKD progress to ESRD by 5 years of age.
urinary calcium excretion is elevated. Many patients with Gitelman Autosomal dominant polycystic kidney disease (ADPKD) is
syndrome have a history of tetany and muscle cramps secondary to caused by mutations in the polycystin-1 (PKD-1) or polycystin-2
hypomagnesemia, which is an uncommon feature of Bartter syndrome. (PKD2) gene, and it has a prevalence of roughly 1 : 1000 (Igarashi
Gitelman syndrome is caused by mutations in the thiazide-sensitive and Somlo, 2002; Torres et al., 2007). ADPKD usually presents in
NaCl cotransporter (NCC) found in the distal convoluted tubule. adulthood but can lead to clinical symptoms in older children and
The keystone of treatment for Gitelman syndrome is magnesium young adults. ARPKD is often identified in childhood either because
and potassium supplementation to replace urinary loss. of a positive family history, as an incidental finding on imaging for
abdominal complaints (pain or constipation), or as part of an
evaluation for hypertension or urinary tract infections. Similar to
CYSTIC RENAL DISEASE ARPKD, there is no disease-specific therapy approved in children.
However, hypertension and urinary tract infections should be treated
Cystic renal diseases are a common cause of both CKD and eventually aggressively to limit disease progression. In patients with a family
ESRD in the pediatric population. Clinical presentation of these history of cerebral aneurysm, routine screening is currently not
350 PART III  Pediatric Urology

recommended in the pediatric population unless there are clinical frequency, hesitancy, and nonspecific abdominal or flank pain.
signs or symptoms that warrant imaging. Although increasingly less common in the United States, stones may
Nephronophthisis in distinction to ARPKD or ADPKD is be identified as part of the evaluation of urinary tract infections. It
characterized by cyst formation at the corticomedullary junction, is not unusual for nephrolithiasis to be noted incidentally as part
but the kidney size is typically normal. Most patients with neph- of a diagnostic evaluation for nonspecific complaints such as recurrent
ronophthisis have a history of polyuria, polydipsia, anemia, and abdominal pain or failure to thrive. Children rarely present with
growth retardation, and eventually reach ESRD by early adolescence. the typical “renal colic” described in adults. In the United States,
Many forms of recessive nephronophthisis are syndromic in nature, the majority of stones are found in the kidney, and presentation
and extrarenal manifestations are common; these include retinitis with bladder stones is more common in those with underlying
pigmentosa in Senior-Løken syndrome; cerebellar vermis aplasia urologic abnormalities.
and colobomas in Joubert syndrome; obesity, diabetes mellitus, and Hypercalciuria is the most common risk factor identified in
polydactyly in Bardet-Biedl syndrome; and skeletal dysplasias in children with urolithiasis, and the vast majority have idiopathic
Juene and Ellis-van Creveld syndromes. In patients with suspected hypercalciuria. Systemic hypercalcemia is less common and is
recessive nephronophthisis, comprehensive genetic analysis is war- associated with hyperparathyroidism, immobilization hypercalcemia,
ranted as this can help to predict both complications and disease thyroid disease, idiopathic hypervitaminosis D, corticosteroid use,
progression, and importantly can help with caregiver education. Williams syndrome, and rarely, mutation of the calcium-sensing
Currently there are more than 100 distinct genes that have been receptor. Children with hypercalciuria are much more commonly
associated with nephronophthisis. normocalcemic, and with the exception of those on loop diuretics,
Other types of cystic kidney disease include medullary cystic most will have idiopathic hypercalciuria. There is, however, an
kidney disease, which is an autosomal dominant disorder associated increasing understanding of the genetic underpinnings of tubular
with hyperuricemia, gout, and ESRD in adults. Mutations in the dysfunction as a cause of hypercalciuria. Examples of this include
gene encoding uromodulin/Tamm-Horsfall protein have been linked mutations in CLCN5 or ORCL1 seen in Dent disease; mutations in
with one of the two types of medullary cystic kidney disease (type SLC12A1, KCNJ1, CLCNKB, BSND, or CASR seen in Bartter syndrome;
2). In addition, glomerulocystic kidney disease is characterized by and mutations in CLDN16 or CLDN19 with familial hypomagnesemia
multiple small cortical cysts that result from cystic dilation of the with hypercalciuria and nephrocalcinosis.
Bowman space; the lack of tubular involvement distinguishes it from Hyperoxaluria is a less common cause of urolithiasis and can be
other forms of cystic disease. And medullary sponge kidney is a rare either primary or secondary. Secondary causes include enteric
congenital cystic disorder in which there is ectasia of cortical ducts hyperoxaluria, excess vitamin C intake, and oxalate-rich diets. Primary
within the inner medulla of the kidney, resulting in a spongelike hyperoxaluria (PH) is rare autosomal recessive disorder of oxalate
appearance on radiograph and is associated with nephrolithiasis metabolism of which there are three subtypes (Nazzal et al., 2016).
and urinary tract infection. PH type 1, which is the most common and most severe, is caused
by aniline/glyoxylate aminotransferase (AGT) deficiency leading to
increased systemic levels and excretion of glycolate and oxalate. PH
KEY POINTS type 2 is less common and is caused by mutations of glyoxylate
reductase, leading to increased excretion of L-glycerate and oxalate.
• Urologic anomalies are seen in up to 25% of patients with The clinical course of PH type 2 is usually less severe than in PH
MCDK, including renal hypoplasia/dysplasia, VUR, and type 1. PH type 3 is the rarest and least severe form and is caused
ureteropelvic junction obstruction. by mutations in HOGA1. Uric acid stones caused by hyperuricosuria
• Autosomal recessive polycystic kidney disease (ARPKD) is can be either a primary disorder seen with inborn errors of purine
caused by a mutation in the PKHD1 gene. metabolism (Lesch-Nyhan syndrome) or, more commonly, a second-
• ADPKD is caused by a mutation in the polycystin-1 ary process associated with hematologic malignancies, gout, high
(PKD-1) or polycystin-2 (PKD2) gene. purine intake, tubular disorders, or diabetes mellitus.
Other rare causes of pediatric urolithiasis include cystinuria and
adenine phosphoribosyltransferase (APRT) deficiency. Cystinuria is
UROLITHIASIS: MEDICAL MANAGEMENT a rare autosomal recessive disorder caused by mutations in either
SCL3A1 or SLC7A9, which result in decreased tubular reabsorption
Accurate data on the incidence and prevalence of urolithiasis in the of cystine and other dibasic amino acids (ornithine, arginine, and
pediatric population in the United States are not readily available. lysine), leading to excessive excretion of cystine and stone formation.
Although not as common as in the adult population, it is estimated that APRT deficiency is a rare autosomal recessive inborn error of adenine
nephrolithiasis accounts for between 1 in 600 and 1 in 800 pediatric metabolism that results in the generation and urinary excretion of
hospital admissions in the United States. Recent data suggest that the large amounts of poorly soluble 2,8-dihydroxyadenine with ensuing
incidence of urolithiasis in the adolescent population has increased stone formation.
significantly over the past 10 years from roughly 20 per 100,000 in The evaluation of nephrolithiasis should begin with a history
the late 1980s to more than 50 per 100,000 in 2014; this number is focused on symptoms associated with stones and predisposing factors
likely to be an underestimate as urolithiasis remains under-diagnosed including recurrent urinary tract infection, familial history of neph-
because many children are asymptomatic at the time of diagnosis rolithiasis, gout, and conditions that increase enteric oxalate absorption:
(Braun et al., 2016; Sas, 2011). Concurrent with the increase in the inflammatory bowel disease, short gut syndrome, or cystic fibrosis
frequency of stone disease has been a shift in the types of stones seen (Edvardsson et al., 2013). A through dietary history should be taken
at presentation. There has been a marked drop in the incidence of including sodium, calcium, fluid intake, and vitamin supplementation.
infectious stones from more than 60% before the 1980s to now less Initial screening studies should focus on identifying a metabolic etiology
than 20%; there are similar reported reductions in the frequency of for the kidney stones and should include serum electrolytes, blood
uric acid and ammonium acid urate stones. The calcium stones are urea nitrogen (BUN), creatinine, calcium, phosphorus, uric acid, and
now the most prevalent. The reasons behind these shifts are not a venous blood gas (for evaluation of possible RTA). In the presence
clear, however in the United States, it has been proposed that changes of hypercalciuria, hypercalcemia, or hypophosphatemia, serum
in dietary practices, changes in types of fluids used for hydration, parathyroid hormone and vitamin D levels should be obtained. Urine
and the obesity epidemic underlie the rise in pediatric stone disease. microscopy can be quite useful, particularly in the diagnosis of cystinuria
Reported rates for stone recurrence in the pediatric population are or ARPT deficiency as the urinary crystals seen in these conditions
variable, ranging from 15% to 20%, but may be as high as 30% in are disease-specific. If the stone is available, it should be sent for
individuals with underlying metabolic disorders. analysis by infrared spectroscopy or radiographic diffraction. A urine
The clinical presentation of nephrolithiasis in children is varied culture should be done to rule out the presence of infection. Timed
and includes microscopic or gross hematuria, urgency, dysuria, urine collections for calcium, oxalate, urate, citrate, and sodium should
Chapter 21  Urologic Aspects of Pediatric Nephrology 351

be performed for any child with stones if possible. For children in especially for distal ureteral stones greater than 10 mm. Open surgery
whom 24-hour urine collections are not feasible, spot ratios of urine is usually reserved for very young children with complex stones,
solute to urine creatinine can be substituted. It is important to remember children with orthopedic issues that limit positioning, or those who
that normative data for urinary solute excretion vary with age. For have failed primary therapy.
example, a normal urine calcium-to-creatinine ratio for a 2-month-old Urolithiasis has a high rate of recurrence, especially if an underlying
child is in the 0.4 to 0.5 mg/mg range, and a ratio of 0.2 mg/mg is metabolic disorder exists. Prevention with appropriate medical therapy
the upper limit of normal for a 5-year-old child. One of the primary and high fluid intake should be encouraged to limit morbidity.
goals of the initial evaluation is to identify patients with underlying Long-term follow up is advisable as some children may develop
metabolic disease for whom specific therapies will be beneficial and CKD and progress to ESRD.
to determine which individuals are at risk for recurrent disease. In
terms of imaging, to limit radiation exposure, ultrasonography is
commonly employed in children for initial screening (Smith-Bindman KEY POINTS
et al., 2014). In patients with a nondiagnostic ultrasound examination, • Urolithiasis remains underdiagnosed because many
nonenhanced helical CT is highly sensitive and specific for the diagnosis children are asymptomatic at the time of diagnosis.
of nephrolithiasis. Standard abdominal radiographs remain useful • Calcium stones are now the most prevalent stones in
to determine stone burden and for longitudinal assessment of response children.
to therapy. Genetic testing is an increasingly important part of the • Cystinuria results in decreased tubular reabsorption of
evaluation of pediatric stone disease, as monogenic disorders may cystine, ornithine, arginine, and lysine.
underlie urolithiasis or nephrocalcinosis in 20% of children.
Medical therapy should be directed toward the underlying meta-
bolic cause, if one has been identified, to prevent recurrence and HYPERTENSION
growth of existing stones (Hernandez et al., 2015). Increasing fluid
intake with a minimum of 2 L/1.73 m2/day should be encouraged Hypertension in the pediatric population is defined as blood pressure
to decrease the chance of crystallization and growth of calculi. on repeat measurements greater than the 95th percentile for age,
With hypercalciuria, salt restriction will reduce urinary calcium gender, and height in comparison to normative population-based
losses by promoting reabsorption of sodium and calcium. Avoiding data. The estimated prevalence of hypertension in children is
excessive protein intake to reduce the acid load is also recommended much lower than the prevalence seen in the adult population;
in recurrent stone formers. A thiazide diuretic can be used in the however, data suggests the increase in the prevalence in hyperten-
normocalcemic hypercalciuric patient to increase distal tubular sion seen in the pediatric population in recent years is closely
reabsorption of calcium. linked to increasing obesity rates in children (Din-Dzietham et al.,
For primary hyperoxaluria, the goal of therapy is to decrease 2007, Falkner, 2010; Muntner, 2004). Weight-related disorders such
oxalate production, increase urine calcium oxalate solubility, and as hypertension are now increasingly common in pediatric patients
decrease crystal deposition in the kidney. Potassium citrate, ortho- and are likely to account for the rise in the prevalence of hypertension
phosphate, or magnesium citrate formulations are used to decrease from 2.7% noted in the early 1990s to 3.7% seen in the 2000s, with
urinary calcium oxalate supersaturation. A trial of pyridoxine should elevated blood pressures (previously known as prehypertension)
be given in PH type 1. About 20% of patients with PH type 1 will observed in approximately 10% of children and adolescents.
respond to pyridoxine treatment with normalization of urine oxalate An accurate measurement of blood pressure is essential to the
concentration, and about 30% will have a partial response. However, diagnosis of hypertension (National High Blood Pressure Education
pyridoxine is not effective in PH types 2 and 3 (Ben-Shalom and Program, 2004). Accurate blood pressure measurement requires a cuff
Frishberg, 2015). Therefore, only successful liver transplantation that is appropriate to the size of the child’s upper arm; the bladder
can offer a potential cure for PH type 1. width should be at least 40% of the mid-arm circumference with a
In individuals with hypocitraturia, a diet rich in fruits and veg- cuff length that covers at least two-thirds of the upper arm. Systolic
etables, and/or supplemental potassium citrate is recommended. blood pressure is defined by the first Korotkoff sound, with the disap-
Sodium citrate should be avoided because the excess salt may increase pearance of the Korotkoff sounds defining diastolic blood pressure. As
hypercalciuria. In cystinuria, alkalization of the urine to a pH greater accurate auscultatory measurement of blood pressure is often difficult
than 7.5 with potassium citrate will decrease cystine supersaturation. in infants and young children, the use of automated (oscillometric)
If stone formation continues despite high fluid intake and alkaliniza- blood pressure measurement devices is now widespread; however
tion of the urine, D-penicillamine or tiopronin can also be used. it should be noted that pediatric normative blood pressure data is
However, these medications are not always well tolerated. In based on auscultatory and not oscillometric standards.
hyperuricosuria, allopurinol, alkalinization of urine, and avoidance A recent clinical practice guidelines for management of hyperten-
of protein excess are recommended. Struvite calculi often require sion in children and adolescents (Flynn et al., 2017) that was
surgical removal and prolonged treatment with antibiotics. published by the American Academy of Pediatrics (AAP) update and
For children who present with acute renal colic, analgesia and refine reference data from the 4th Report of the National Institutes
adequate hydration with appropriate monitoring of fluid and of Health Working Group for the management of blood pressure
electrolyte status are the mainstays of therapy. Radiologic evaluation in children (2004). A major focus of this report was improving the
should be performed, and in the setting of urinary obstruction, recognition of hypertension by simplifying the initial screening blood
infection, acute kidney injury, or incontrollable pain, urologic pressure table. Furthermore, normative data by age, gender, and
intervention may be needed. For small ureteral stones (< 10 mm), height have been revised, including only those children with a normal
management with observation with or without medical expulsive BMI. The blood pressure classification scheme has also been revised
therapy is recommended. Urologic interventions such as ESWL, to align the classification system for adolescents with the absolute
percutaneous nephrolithotripsy, ureteroscopy, and open surgery have blood pressure values used in adult blood pressures guidelines
all been successful therapies in children with urolithiasis. The choice (Redwine et al., 2012). Current AAP guidelines now distinguish
of intervention is typically guided by stone size, composition, and between children 1 to 13 years of age and adolescents older than
location. Complete removal is important in children as residual 13 years of age, with normal blood pressure defined as less than
small fragments are associated with regrowth and adverse outcomes. the 90th percentile for children and less than 120/80 mmHg for
Medical expulsive therapy with an α-blocker has been successful in adolescents (Flynn et al., 2017). The term prehypertension has been
the management of small ureteral stones. ESWL is often employed abandoned and replaced with elevated blood pressures to be consistent
for intermediate-size stones (10 to 20 mm) within the ureter or with the adult guidelines.
collecting system. Percutaneous nephrolithotomy is the recommended Staging of hypertension is useful in determining which children
treatment for large renal pelvic or caliceal stones (>20 mm). Ure- require more immediate attention for diagnostic evaluation and
teroscopy is increasingly becoming a first-line procedure of choice, management. In addition, staging can also predict the presence of
352 PART III  Pediatric Urology

end-organ damage from hypertension. Two other clinically relevant creatinine, and a lipid profile. Although commonly utilized, the current
designations for severity of hypertension are important: (1) hyper- AAP guidelines do not recommend screening renal ultrasonography
tensive emergency, when a patient has end-organ injury such as for patients older than 6 years of age unless there is an abnormal
encephalopathy, pulmonary edema, or cerebral hemorrhage or clinical urinalysis or abnormal renal function. Additional testing such as
symptoms like blurred vision, headache, or nausea, which can be renin and aldosterone levels; catecholamines; cortisol levels; genetic
associated with severely elevated blood pressure; and (2) hypertensive testing for forms of monogenic hypertension; or advanced imaging
urgency, when blood pressure is severely elevated without clear studies including VCUG, dimercaptosuccinic acid (DMSA) renal
evidence of end-organ involvement or overt symptoms. scans, CT angiography, magnetic resonance angiography (MRA), or
It also should be recognized that not every child with elevated angiography are often required based on an individual patient’s
blood pressure readings in the office setting has true hypertension. clinical findings, initial screening studies, and the severity of the
Up to one-fourth of such patients have normal blood pressures hypertension. Echocardiograms are no longer recommended as part
outside of the office setting and are classified as having “white of the initial screening of patients with confirmed hypertension.
coat” hypertension. The recent AAP care guidelines promote the However, baseline echocardiography is now recommended at the
use of 24-hour ambulatory blood pressure monitoring (ABPM) initiation of pharmacologic therapy for all patients regardless of the
in the evaluation of children and adolescents with suspected etiology of their hypertension.
hypertension. ABPM is not only useful in identifying patients with For patients with stage 1 hypertension or those with primary
“white-coat” hypertension, but also it can identify individuals with (essential) hypertension without end-organ damage, therapeutic
so called “masked” hypertension: those with normal blood pres- lifestyle changes are the initial intervention of choice (Lurbe et al.,
sure readings in the office setting, but who are truly hypertensive. 2009). There is clear data that support dietary modification (DASH
Furthermore, ABPM data is extremely useful in accessing effectiveness diet), weight loss, and exercise in reducing blood pressure in children
of antihypertensive regimens. Unfortunately, access to 24-hour ABPM and adolescents. However, like all behavioral interventions, the
is often limited and may require referral to pediatric subspecialists. long-term success of therapeutic lifestyle changes requires engagement
The causes of hypertension are classified as either primary not only of the patient, but of family members as well.
(essential) or secondary when a specific etiology is identified. Pharmacologic therapy should be considered in patients with
Historically, secondary causes of hypertension have been thought to primary (essential) hypertension who do not have improvement in
predominate in the pediatric population (Vehaskari, 2009). However, their blood pressure after a trial of therapeutic lifestyle changes, or
with the rising rates of childhood obesity, the AAP guidelines have in patients with stage 2 hypertension, evidence of end-organ damage,
recognized that primary (essential) hypertension is now the most symptomatic hypertension, or secondary hypertension (Flynn and
common cause of hypertension in obese children older than 6 Daniels, 2006). Unlike adult hypertension guidelines, there are no
years of age with a positive family history of hypertension. data or evidence-based guidelines regarding which drug to select
Secondary causes of hypertension vary with age. In infants younger when initiating pharmacologic therapy. Acceptable first-line agents
than 1 year of age, the majority of cases are caused by either coarctation include diuretics, β-blockers, ACE inhibitors, ARBs, and calcium
of the aorta or renovascular disease. Renovascular disease may also channel blockers. Rather than standardized approaches to pharma-
present later in childhood. In school-age children, the dominant cologic therapy, most practitioners individualize therapy with the
secondary causes of hypertension are renal parenchymal diseases underlying renal, cardiac, or endocrine conditions guiding the choice
including reflux nephropathy. Malignancy-associated hypertension, of the initial agent. This would include the use of RAAS blockage
primarily with Wilms tumor and nephroblastoma, typically presents for those with glomerulonephritis and other forms of CKD because
in infancy and early childhood, and pheochromocytoma is seen of the additional benefit in terms of preventing CKD progression.
more frequently in the adolescent population. Secondary causes of Along similar lines is the use of RAAS inhibition in patients with
hypertension should still be sought in adolescents, however, this type 1 or type 2 diabetes mellitus as these agents may slow or prevent
population, much like adults, is far more likely to have primary the development of diabetic nephropathy. Conversely, the use of
(essential) hypertension. β-blockers in patients with asthma is not recommended. Adverse
Although hypertension is becoming more common in children, effects of each agent and available formulations need to be considered,
the differential diagnosis of childhood hypertension is quite broad. because many standard adult hypertensive medications are not
Features from the history, the physical, the age of the patient, and available in liquid formulations or are in fixed ratios that are not
even the blood pressure values themselves can narrow the diagnostic suitable for the pediatric population. Patients and families should
focus. A secondary cause for hypertension should always be sought be counseled about the potential side effects of the medications
in infants younger than 1 year of age, and secondary causes should before initiation of therapy; this is especially important for adolescents
also be investigated thoroughly in preadolescent children as renal who are engaged in organized sports and may be at risk for dehydra-
disease is the most common secondary cause of hypertension in tion and acute kidney injury while on RAAS or diuretic therapy, and
this age group. The new AAP guidelines recognize that the impact for sexually active adolescents on RAAS therapy regarding the potential
of obesity extends even into the early school-age population as for ACE fetopathy. Once therapy is initiated, blood pressure should
the previous guidelines recommended only a limited investigation be monitored closely, including laboratory monitoring, both for
into secondary causes of hypertension in patients older than 6 years efficacy of the antihypertensive medication and for potential side
of age with obesity, a strong family history of essential hypertension, effects. The goals of therapy for patients without diabetes or CKD
and an unrevealing history and physical examination. Review of the have also been revised in the current AAP guidelines. Based on data
patient’s history should be focused on two areas; firstly, the presence suggesting that end-organ damage is present in patients with blood
of symptoms suggestive of hypertensive injury such as headaches, pressures greater than the 90th percentile but less than the 95th
dizziness, diplopia, or vomiting; and secondly, information that
provides insight into underlying causes of hypertension including
neonatal history, urinary tract infections or unexplained fevers, signs
and symptoms of renal or cardiac disease, chronic illness, medications, KEY POINTS
and family history of hypertension, kidney disease, early-onset • The increase in prevalence of pediatric hypertension in
cardiovascular disease, and diabetes. recent years is closely linked to increasing obesity rates in
The patient’s weight, height, and growth percentiles should be children.
defined. Ideally blood pressure measurements should be obtained • Up to one-fourth of patients have normal blood pressures
in both upper extremities and in at least one lower extremity. The outside of the office setting (“white coat” hypertension).
physical examination should focus on identifying signs of underlying • Therapeutic lifestyle changes are the initial intervention of
disorders that are associated with hypertension. choice for patients with primary hypertension without
Recommended initial diagnostic testing in all patients with end-organ damage.
confirmed hypertension includes a urinalysis, electrolytes, BUN,
Chapter 21  Urologic Aspects of Pediatric Nephrology 353

TABLE 21.1  KDIGO Criteria for Acute Kidney Injury


BOX 21.4  Causes of Acute Kidney Injury
SERUM CREATININE URINE OUTPUT
Decreased true intravascular volume
STAGE CHANGES CHANGES
• Third-space losses: sepsis, trauma, burns, nephrotic
I SCr increase ≥0.3 mg/dL in <0.5 mL/kg/h for syndrome
48 h or 6–12 h • Gastrointestinal losses
1.5–1.9 times baseline • Salt wasting: renal or adrenal
• Diabetes insipidus: central or renal
II SCr increase 2.0–2.9 times <0.5 mL/kg/h for
Decreased apparent intravascular volume: congestive heart
baseline 12 h
failure
III SCr ≥3.0 increase or < 0.5 mL/kg/h Acute tubular necrosis/cortical necrosis
SCr >4.0 mg/dL or for 24 h or • Hypoxic-ischemic insults
If <18 years of age, estimated <0.3 mL/kg/h for • Medications: aminoglycosides, intravenous contrast
creatinine clearance 12 h agents, nonsteroidal anti-inflammatory drugs,
<35 mL/min/1.73 m2 chemotherapeutics
• Exogenous toxins: ethylene glycol, methanol
h, Hour; KDIGO, Kidney Disease Improvng Global Outcomes; SCr, serum • Endogenous toxins: hemoglobin, myoglobin
creatinine.
Tumor lysis syndrome and uric acid nephropathy
Modified from Kellum JA, Lameire N; KDIGO AKI Guideline Work Group.
Interstitial nephritis
Diagnosis, evaluation, and management of acute kidney injury: a KDIGO
summary (Part 1). Crit Care 17(1):204, 2013. • Medication induced: anticonvulsants, antibiotics
• Idiopathic
Glomerulonephritis: all causes
percentile, the current guidelines recommend a target blood pressure Vascular disorders
goal of less than 90%, or less than 130/80 for adolescents. Lower
• Hemolytic uremic syndrome
target goals are recommended for patients with CKD or diabetes
mellitus (<50th percentile). • Renal artery or vein thrombosis
Infection
• Pyelonephritis
• Sepsis
ACUTE KIDNEY INJURY Obstructive lesions
Acute kidney injury (AKI) is currently defined as an abrupt reduc- • Obstructed single kidney
tion in kidney function characterized by an absolute increase in • Bilateral ureteral obstruction
serum creatinine of more than 0.3 mg/dL, a relative increase in • Urethral obstruction
serum creatinine of 50% from baseline, or 6 hours or more of
oliguria (< 0.5 mL/kg/h of urine output). This precise definition
is a result of a large body of work that recognized that AKI is a
continuum renal injury wherein reductions in renal function are (KIM-1), and interleukin-18. Although there is substantial data
linked to adverse outcomes (Table 21.1 and Box 21.4) and shifts the in the adult population regarding the sensitivity and specificity of
focus to earlier detection and treatment of AKI before the development these biomarkers, and although there are emerging pediatric data,
of significant morbidity, as noted in KDIGO staging table (Devarajan, these biomarkers have not yet entered widespread clinical use, and
2013; Mehta et al., 2016; Selewski et al., 2014). Recent data suggest the application of the data from adult studies is challenging in the
that the incidence of AKI is roughly 4 cases per 1000 pediatric hospital pediatric population.
admissions, where as AKI is seen in up to one-third of all children Differentiating between functional and intrinsic AKI is essential.
admitted to intensive care units and one-half of pediatric patients Functional AKI is a sodium-avid state with maximal reabsorption
with shock (Lameire et al., 2017). Patients with AKI have been of solutes and water by the intact proximal tubule; on the other
reported to have a nearly 15-fold increased mortality risk compared hand, the tubular damage typical of intrinsic AKI results in impaired
with patients without AKI (15% vs. 0.6%) (Fitzgerald et al., 2016; reabsorptive capacity of the proximal tubule. Functional AKI is
Kaddourah et al., 2017). The estimated incidence of AKI requiring typically characterized by fractional excretion of sodium (FENa) less
dialysis ranges from 1% to 2% in critically ill children (Slater et al., than 1%, and greater than 3% is characteristic of intrinsic AKI.
2016; Sutherland et al., 2013). AKI can result from ischemic, toxic, However, the use of FENa in patients on diuretic therapy is unreliable.
or inflammatory mechanisms and can be further classified as caused In this setting, the fractional excretion of urea (FEUrea) is very useful,
by renal hypoperfusion (prerenal), structural injury to the kidneys with FEUrea less than 35% being indicative of prerenal AKI.
(intrinsic), or urinary tract obstruction (postrenal). Primary renal AKI results in the disruption of normal electrolyte and fluid
disease is no longer the predominate cause of intrinsic AKI and has homeostasis. Common complications include hyponatremia caused
been supplanted by a heterogeneous mix of pathologic conditions by excessive free water retention, hypernatremia from sodium
including multiple organ failure, renal ischemia, nephrotoxic injury, administration, hyperkalemia caused by reduction in glomerular
congenital heart disease, bone marrow transplantation, and sepsis. filtration and tubular potassium secretion, anion gap metabolic
The diagnosis of AKI is dependent on increases in serum creatinine acidosis secondary to impaired renal hydrogen ion excretion and
compared with baseline levels. As opposed to the adult population, concurrent impairment of bicarbonate reabsorption and regeneration,
normal serum creatinine levels vary in the pediatric population based hyperphosphatemia caused by reduced renal excretion, and hypo-
on age, muscle mass, and body size; therefore there is no absolute calcemia secondary to increased serum phosphate and impaired
creatinine level that defines pediatric AKI. Typically renal function vitamin D metabolism. Fluid overload in patients with AKI is chal-
needs to decrease by 50% before a change in serum creatinine can lenging as adequate fluid resuscitation is essential for tissue perfusion
be measured, and changes in creatinine commonly lag behind the in patients with prerenal AKI. However, there is a clear association
reduction in renal function (GFR) by 24 to 72 hours. This lack of between excessive fluid accumulation and adverse outcomes in the
sensitivity and delayed response has presented challenges to early pediatric population.
identification of patients with AKI. A number of biomarkers to Prerenal AKI requires prompt and vigorous fluid resuscitation
identify early AKI have been developed; these include neutrophil with isotonic fluids. The use of potassium-containing fluids should
gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 be avoided until urine output is established and the severity of AKI
354 PART III  Pediatric Urology

is defined. If oliguria is present, conservative fluid management TABLE 21.2 Classification of Chronic Kidney Disease
should be instituted. Fluid intake should therefore be limited to for Children Older Than 2 Years of Age
match output including insensible fluid loss with replacement of
output. If patients are fluid overloaded, then urine output should STAGE DEFINITION GFR (mL/min/1.73 m2)
not be replaced, and diuretics are likely indicated. If adequate urine
output cannot be achieved with diuretic therapy, RRT may be required. I Normal or ↓ GFR (with ≥90
In addition, aggressive nutritional support is essential for all patients kidney damage)
with AKI (Kyle et al., 2013). Adequate calories should be provided II Mild ↓ GFR (with 60–89
that meet maintenance requirements and to limit or prevent excessive kidney damage)
catabolism. Protein intake should not be restricted. If adequate III Moderate ↓ GFR 30–59
nutrition cannot be achieved because of fluid restriction, early IV Severe ↓ GFR 15–29
institution of RRT should be considered. Nephrotoxic agents should V Kidney failure <15 or dialysis
be avoided in AKI, as they may worsen the injury or delay recovery
of function. Medications should be adjusted based on residual renal
GFR, Glomerular filtration rate.
function. Importantly, patients in the early phase of AKI should be
assumed to have a GFR of less than 15 mL/min/1.73 m2, regardless
of the absolute value of the serum creatinine.
Indications for initiating RRT in AKI typically include fluid overload
that is unresponsive to diuretics, the need to deliver adequate nutri- of age. A normal term infant has a GFR of approximately 40 mL/
tion, or to address hyperkalemia or metabolic disturbances that are min/1.73 m2, and, as part of normal renal developmental processes,
refractory to medical therapy, and symptomatic uremia. RRT options GFR does not reach final adult levels until nearly 2 years of age, thus
include hemodialysis, peritoneal dialysis, and continuous renal GFR increases during the first 2 years of life can make definitive CKD
replacement therapy (CRRT). The choice of dialysis modality depends staging in patients younger than 2 years of age problematic. Secondly,
on the patient’s clinical status, the available clinical expertise, and the requirement for renal abnormalities to persist for more than 3
the availability of appropriate resources. Hemodialysis provides the months to meet the criterion for CKD makes this staging system
most efficient and rapid correction of fluid and electrolyte distur- inapplicable to newborns and infants younger than 3 months of age.
bances; however hemodialysis can be poorly tolerated in patients Causes of CKD in children are very different from those in
with hemodynamic instability. Therefore, CRRT via hemodialysis adults and vary by age group within the pediatric population. In
has become the standard of care as it delivers slow continuous younger children, the most common causes are CAKUT: renal
correction of fluid overload and electrolyte imbalances. Peritoneal hypoplasia/dysplasia and obstructive lesions such as posterior urethral
dialysis is also a CRRT modality and has been preferred in neonates valves and prune belly syndrome. Renal cystic diseases, including
and small infants when central venous access can prove challenging. multicystic kidneys, cystic renal dysplasia, juvenile nephronophthisis,
The advantages of peritoneal dialysis include ease of performance and autosomal recessive polycystic kidney disease, are also common
and avoiding the requirement for specialized equipment, personnel, causes of CKD during childhood. Glomerular diseases such as
or anticoagulation. In critically ill children with respiratory failure, nephrotic syndrome, focal segmental glomerulosclerosis, hemolytic
dwell volumes in the peritoneal cavity may be poorly tolerated, and uremic syndrome, membranoproliferative glomerulonephritis,
previous abdominal surgery with compromise of the peritoneum, membranous nephropathy, and lupus nephritis are uncommon causes
intra-abdominal infection, and ostomy placement can limit the of CKD in early childhood, but by late childhood and early adoles-
application of peritoneal dialysis. cence, they become the predominant causes of CKD. Although the
Data suggest that children who have recovered from an episode prevalence of ESRD, including renal transplantation and chronic
of AKI are at significant risk for CKD, as more than one-third have dialysis, is well-defined, no incidence or prevalence data for pediatric
been reported to have either reduced kidney function or were dialysis- CKD are available from North American registries.
dependent on hospital discharge. Furthermore, nearly two-thirds of The frequency and severity of the complications of CKD increase
the patients with long-term follow-up had evidence of CKD. These as CKD progresses. The classification of CKD is based on five stages
data suggest that long-term follow-up is warranted for children who (Table 21.2), and stage-based care guidelines for the management
survive an AKI episode. of these complications are detailed in consensus recommendation
by the KDOQI and KDIGO initiatives (Inker et al., 2014; KDIGO,
2009; KDOQI, 2009).
KEY POINTS Protein malnutrition is seen in up to 20% of pediatric patients
with any stage of CKD and is more common in advanced CKD and
• Hospitalized pediatric patients with AKI have a nearly in infants and younger children. In younger children, the key factor
15-fold increased mortality risk compared with patients contributing to malnutrition is inadequate intake. Although the
without AKI. underlying cause of malnutrition in children with CKD is often
• Prerenal AKI requires prompt and vigorous fluid multifactorial, it is extremely important that this be recognized and
resuscitation with isotonic fluids. treated promptly and aggressively, as malnutrition has been strongly
• CRRT via hemodialysis has become the standard of care as associated with mortality. Children with CKD often have an impaired
it delivers slow continuous correction of fluid overload appetite, and this can be further complicated by nausea and vomiting.
and electrolyte imbalances. They may also require the restriction of specific dietary components,
especially potassium and phosphate. Specialized formulas and diets
designed for children with CKD are often required. A pediatric dietitian
CHRONIC KIDNEY DISEASE with experience in CKD/ESRD is essential to optimize caloric intake,
which is the overriding treatment goal for malnutrition in children
CKD is defined by the persistence of structural or functional abnor- with CKD.
malities of the kidney for more than 3 months. The primary marker of Growth impairment is a common complication in children with
kidney damage using the Kidney Disease Outcomes Quality Initiative CKD and can have a major psychological impact on a child while
(KDOQI) staging system is a decrease in GFR. This staging system negatively affecting their quality of life (QoL). Children with CKD
was developed to stratify the severity of CKD and to guide clinical are approximately 1.5 standard deviations below normal for height,
management, as the complications of CKD commonly develop at and this height deficit is greatest in the youngest children. With
specific GFR levels irrespective of the etiology of CKD. There are a aggressive treatment of nutritional deficiencies and renal osteodys-
number of caveats with respect to applying this system to pediatrics. trophy, a normal growth pattern is usually reestablished until the
First, the staging of CKD excludes patients younger than 2 years child reaches ESRD (CKD stage V). The causes of growth delay in
Chapter 21  Urologic Aspects of Pediatric Nephrology 355

children with CKD include malnutrition/protein-energy wasting, Contributing risk factors include chronic hypertension, dyslipidemia,
chronic acidosis, severe renal osteodystrophy, sodium depletion, and abnormal mineral metabolism. Hypertension has been reported
and growth hormone resistance. Where possible, treatment should in more than one-half of children with CKD, and, even with anti-
be directed to correction of these metabolic disturbances before the hypertensive medications, less than 50% were well controlled (ESCAPE
introduction of growth hormone (Haffner et al., 2000). Children Trial Group et al., 2009; Mitsnefes, 2012; Taler et al., 2013). Left
with CKD are not growth hormone deficient, but rather have ventricular hypertrophy is common in children with CKD, with
abnormalities along the growth hormone activation pathway including nearly 60% to 80% of children who are initiating dialysis having
reduced production of insulin-like growth factor-1 (IGF-1), increased left ventricular hypertrophy as well. Dyslipidemia is also seen in
circulating levels of IGF-binding proteins that reduce IGF-1 activity, approximately 39% to 65% of children with CKD. The most common
and resistance of bone to growth hormone activity. Despite the abnormality is hypertriglyceridemia, followed by elevated non-HDL
consensus recommendations for children with growth impairment cholesterol and lower HDL cholesterol levels. It is estimated that
and CKD, only about 1 in 4 children are prescribed recombinant one-fourth of children have more than one lipid abnormality. Initial
growth hormone. treatment options usually involve nutrition and dietary modification
CKD/mineral bone disease, a term now employed to replace renal (Sarnak et al., 2015).
osteodystrophy, is a systemic disorder characterized by alterations in Electrolyte abnormalities in children with CKD are very common
mineral metabolism, bone deformities, and calcific cardiovascular and include metabolic acidosis from renal tubular dysfunction
abnormalities in CKD. Insufficiency of 1,25(OH)2 vitamin D in CKD and/or progressive uremia with ensuing retention of phosphate
can be a result of deficient activation of cholecalciferol to the active or sulfate anions. Treatment consists of correction of the acidosis
form of 1,25-dihydrocholecalciferol in the kidney or a result of low with supplemental bicarbonate or citrate solutions and is particularly
cholecalciferol levels as a result of dietary insufficiency. In the later important for growth and bone health. Hyponatremia may also
stages of CKD, retention of phosphate worsens as renal function occur as a result of urinary sodium wasting and is commonly seen
declines. Both the elevation of phosphate levels in plasma and a in the setting of CKD associated with CAKUT. Hyperkalemia is
fall in plasma calcium values stimulate parathyroid hormone secretion; frequently seen as a result of impaired renal potassium excretion
the parathyroid hormone then increases phosphate excretion by the and can be aggravated by concurrent metabolic acidosis; potassium
suppression of tubular phosphate reabsorption, thereby returning control is often focused on dietary restriction and modification.
phosphate values to normal but at the expense of persistently elevated The natural course of CKD is gradual deterioration of kidney
parathyroid hormone values. It has been shown that skeletal min- function. This decline does not occur in a linear fashion as the
eralization defects start in pediatric patients as early as CKD stage later stages of CKD are associated with a more rapid progression.
II, even before defects of bone turnover or any biochemical abnormal- Although there are no clear therapeutic interventions to prevent or
ity, except for an increase in circulating FGF-23 levels. The effects of reverse progression of CKD, several factors have been identified that
metabolic bone disease on the epiphysis are unique to children with may slow progression. In adults, intake of a reduced protein diet
CKD and include growth failure, slipped epiphysis, and rickets. may reduce proteinuria and slow the decline of kidney function.
There are a number of therapeutic approaches to prevent and However pediatric trials have shown that lower protein intake does
treat renal bone disease in patients with CKD. Initially, if plasma not provide any benefit as measured by the rate of decline in creatinine
levels of 25-OH vitamin D are low, then vitamin D3 (cholecalciferol) clearance over a 2-year period. Therefore, it is not recommended to
supplements should be prescribed. Calcitriol (Rocaltrol), active 1,25 restrict protein intake in pediatric patients with CKD unless evaluation
vitamin D, is typically prescribed if the parathyroid hormone levels of an individual’s diet or laboratory values suggests that their protein
exceeds the recommended limits and the calcium level is not elevated. intake is clearly excessive. Strict blood pressure control is the only
In the presence of elevated plasma phosphate values, if dietary intervention that has been shown to decrease progression of CKD
phosphate restriction is ineffective, phosphate binders such as calcium in children. Based on this, KDIGO guidelines recommend that the
carbonate, calcium acetate, or sevelamer should be prescribed. target blood pressure for children should be the 50th percentile for
Calcium-containing compounds are available in liquid formulations age, gender, and height, and particularly if children have proteinuria.
and, therefore, are most easily used in young children. However, Children with obstructive uropathies, particularly those with inad-
excessive total calcium ingestion may lead to hypercalcemia, which equate bladder emptying including those who require intermittent
has been associated with vascular calcification, even in children. catheterization, as well as increasing hydronephrosis, which may
Multiple studies have evaluated QoL and social behavior in children indicate partial lower tract obstruction, should be dealt with promptly
with CKD and have shown lower health-related QoL scores when by a pediatric urologist. Similarly, for those with repeated urinary
compared with healthy controls and other children with chronic tract infections, efforts should be focused on prevention either by
medical conditions; in fact, patients with CKD/ESRD have lower assuring appropriate bladder drainage or by using prophylactic daily
QoL scores than children with cancer. Children with CKD are also antibiotics.
at risk for neurocognitive impairment. Developmental problems
have been reported in as many as 25% of infants with renal disease
and correlates with the severity of renal dysfunction. Cognitive deficits KEY POINTS
are also noted in older children, and the overall IQ of school-age
children with CKD is lower than the normal population. • Causes of CKD in children are very different from those in
Anemia is nearly universal in advanced CKD. The most common adults and vary by age group within the pediatric
cause of anemia with CKD is a deficiency of erythropoietin. population.
However, erythropoietin deficiency in CKD is often confounded by • The most common cause of anemia with CKD is a
other causes of anemia including iron deficiency, blood loss associated deficiency of erythropoietin.
with surgical procedures, and frequent phlebotomy. Erythropoietin- • With CKD, kidney function declines in a nonlinear
stimulating agents are essential for the management of the anemia fashion as the later stages are associated with a more rapid
in all CKD patients and especially in pediatric CKD patients (Kliger progression.
et al., 2013). Either epogen or darbepoetin can be used to successfully
treat anemia in the vast majority of children with CKD. Iron therapy
is also required for nearly all children with CKD, however iron END-STAGE RENAL DISEASE: DIALYSIS AND
therapy is affected by gastrointestinal side effects, and there is RENAL TRANSPLANTATION
decreased absorption with concomitant use of phosphate-binding
agents or proton pump inhibitors. ESRD is defined by the need for either chronic dialysis or kidney
Children with CKD are in the highest risk category for cardio- transplantation to sustain life. There is no absolute creatinine or
vascular disease, and registry data indicates that cardiovascular disease BUN level that mandates the initiation of chronic dialysis. Typically
accounts for nearly one-half of all deaths of children on dialysis. initiation of dialysis is considered in children who have reached
356 PART III  Pediatric Urology

CKD stage V (GFR <15 mL/min/1.73 m2) or when CKD is com- in younger children for whom a central hemodialysis catheter may
plicated by uncontrolled hypertension, volume overload, metabolic be necessary. Hemodialysis catheters are preferentially placed in the
derangements, poor growth, or complications of uremia (Green- internal jugular veins; the subclavian veins should be avoided because
baum and Shaefer, 2012). There are two chronic dialytic modalities of the risk for subclavian stenosis.
available for infants, children, and adolescents: peritoneal dialysis Hemodialysis removes metabolic waste products, solutes, and
and hemodialysis (Chand et al., 2017; Fischbach et al., 2005). excess fluid via diffusion, convection, and ultrafiltration. Diffusion
Selection of hemodialysis or peritoneal dialysis is dependent on a characterizes the solute movement across the semipermeable dialysis
number of factors including the needs of the patient and family, membrane against a concentration gradient. Convection is the
patient size, age, lifestyle choices, and psychosocial risk. movement of solutes with plasma water; it is determined by the
Peritoneal dialysis is the most common initial modality for amount of ultrafiltration and characteristics of the dialysis membrane.
children younger than 9 years of age and less than 20 kg, as the Ultrafiltration is the movement of water across the dialysis membrane
treatment is provided at home and does not require central venous by convective flow caused by a pressure or osmotic gradient. Hemo-
access. For older children, it offers the advantage of greater flexibility dialysis fluid removal is determined by the transmembrane pressure
in the timing of treatments and better social integration. PD can be gradient across the dialysis membrane and the permeability of a
successfully performed in many patients with prior urologic surgeries given dialysis membrane to water, and the dialysate solution contains
including vesicostomies, however the presence of omphalocele, electrolytes in physiologic concentrations to normalize electrolytes
gastroschisis, diaphragmatic hernia, or bladder exstrophy are con- and correct acid-base abnormalities. Typically, pediatric patients
traindications to peritoneal dialysis. receive in-center hemodialysis 3 to 4 times per week for 4 hours at
Peritoneal dialysis is performed via a process of repeated instillation a time to achieve adequate metabolic control and fluid balance.
and drainage of dialysis solution both into and out of the peritoneal Home hemodialysis is also an option for select patients and caregivers;
space via a surgically implanted peritoneal dialysis catheter. The similar to peritoneal dialysis, home hemodialysis is provided daily
dialysate solutions contain electrolyte additives in physiologic at night to provide more consistent metabolic control and fluid
concentrations, with the exception of potassium and phosphorus, balance than achieved with the thrice-weekly in-center hemodialysis
to facilitate correction of electrolyte abnormalities and acid-base treatments (Hothi et al., 2016).
abnormalities. The glucose concentration in the dialysis solution For the vast majority of pediatric patients with ESRD, the
creates an osmotic gradient that mediates the net movement of water ultimate treatment goal is renal transplantation, not long-term
into the peritoneal cavity, which is referred to as ultrafiltration. In dialysis. Pediatric patients younger than 18 years of age account for
peritoneal dialysis, solute clearance occurs via a process of diffusion less than 5% of all kidney transplantations performed in the United
whereby solutes move from a compartment of higher concentration States (Dharnidharka et al., 2014; Lau et al., 2012; United States
to a compartment of lower concentration across the peritoneal Renal Data System [USRDS], 2016). Of the approximately 900 annual
membrane and by convection whereby solutes move via solvent pediatric kidney transplants, about two-thirds are from deceased
drag with the movement of fluid between the blood and dialysate donors and one-third are from living donors (primarily parents).
compartments. Peritoneal dialysis is typically performed overnight For patients with an identified living related donor, a preemptive
for 8 to 12 hours and thus achieves more steady metabolic and transplant before the need for dialysis is often recommended. The
blood pressure control than hemodialysis. Peritoneal dialysis can transplant community has recognized the unique benefits that renal
be prescribed either manually (continuous ambulatory peritoneal transplantation has for pediatric ESRD patients, including improved
dialysis) or using an automated machine (continuous cycling growth, neurocognitive development, and QoL. Therefore, children
peritoneal dialysis). Continuous cycling peritoneal dialysis is pre- on the transplant waiting list for deceased-donor organs before 18
scribed for the majority of pediatric patients in the United States. years of age receive priority compared with the average adult deceased-
The peritoneal dialysis prescription, including volumes, dwell times, donor transplantation candidate. Furthermore, children are also
and dextrose concentration, is tailored to each child’s specific needs. preferentially allotted kidneys from younger donors in an attempt
Infections, such as peritonitis and tunnel infections, are the to optimize the life of the allograft. Overall outcomes for pediatric
most common complications of peritoneal dialysis (Warady kidney transplants are excellent, with 50% of deceased-donor
et al., 2012). Peritonitis typically occurs more frequently in younger transplants functioning 12.5 years post-transplant and 50% of living-
children. Peritonitis typically presents with acute onset of abdominal donor kidneys functioning 20 years post-transplant. Almost all
pain and cloudy dialysis fluid, and fluid with a white blood cell children with ESRD should be considered for kidney transplantation;
count greater than 100/mm3 and at least 50% neutrophils meet absolute contraindications are few but include active malignancy or
the presumptive criteria for peritonitis. Gram-positive organisms are uncontrolled infections. Cognitive impairment or intellectual disability
the most common cause of peritonitis in the pediatric population, should not automatically exclude a patient from consideration of
followed by culture-negative, and then gram-negative organisms. kidney transplantation (Goldberg et al., 2015).
Fungal peritonitis, although rare, can require discontinuation of There are a number of unique considerations relating to renal
peritoneal dialysis, and transition to hemodialysis. Peritonitis can lead transplantation in the pediatric population. In infants, because of
to the development of intra-abdominal adhesions, and ultimately technical issues and the increased risk for graft thrombosis, many
peritoneal membrane failure. Peritoneal dialysis catheter exit site and centers wait until the child is at least 10 kg and 65 cm in length
tunnel infections are also important risk factors for peritonitis and before proceeding with transplantation. As CAKUT is a common
can require catheter removal if persistent or recurrent. Noninfectious cause of ESRD in the pediatric population, urinary tract repair or
complications of peritoneal dialysis include the development of reconstruction of anomalies needs to be addressed before transplanta-
dialysate leak, abdominal hernias, hydrothorax, or hemoperitoneum. tion to optimize outcomes. Compared with adults, immune-mediated
In children older than 10 years of age, hemodialysis is the glomerular disease and focal segmental glomerulosclerosis are
predominant chronic dialysis modality in the United States. As common causes of ESRD in children, thus the risk for disease recur-
opposed to peritoneal dialysis, hemodialysis requires chronic central rence in the allograft must be considered both in terms of pretransplant
venous access. The ability to maintain hemodialysis access in infants evaluation and postoperative care. This is particularly of concern for
and smaller children presents a major challenge for pediatric ESRD primary focal segmental glomerulosclerosis, membranoproliferative
care. Options for hemodialysis access include arteriovenous fistula; glomerulonephritis, C3 glomerulopathy, and atypical hemolytic
arteriovenous graft, which uses a synthetic material to create a conduit uremic syndrome. Adequate immunization before renal transplant
between an artery and a vein; and central venous hemodialysis is also a necessity. Surgical removal of native kidneys is often con-
catheters. Arteriovenous fistulas have better survival rates and lower sidered in the peritransplant period; indications for nephrectomy
rates of infection compared with arteriovenous grafts or hemodialysis include chronic or recurrent pyelonephritis, hypertension refractory
catheters and are recommended for children greater than 20 kg for to medical therapy, and uncontrolled nephrotic syndrome.
whom renal transplantation is not eminent (Ma et al., 2013). However For most children less than 20 kg, the allograft is placed intra-
arteriovenous fistulas are not always technically possible, particularly abdominally and anastomosed to the aorta and the inferior vena
Chapter 21  Urologic Aspects of Pediatric Nephrology 357

cava. For children more than 20 kg, a retroperitoneal approach is used SUMMARY
with the allograft placed in the iliac fossa and vascular anastomoses
to the common or external iliac vessels. An antirefluxing anastomosis Effective partnership between pediatric urologists and pediatric
for the donor ureter into the recipient bladder is common as this nephrologists is needed for the optimal care of pediatric patients
will reduce the risk for vesicoureteral reflux into the allograft. The with kidney and urinary tract abnormalities. Although differences in
use of ureteral stents at the time of transplantation to reduce the opinion regarding best approaches may exist, a careful appreciation of
risk for ureteral obstruction appears to be largely center-specific. the expertise of both specialties is needed, as both have expert teams
In the immediate post-transplant period, primary graft dysfunc- of health care providers that can provide the full spectrum of care for
tion is reported to occur in less than 3% of pediatric kidney these complex issues. As a result of these specialties working closely
allografts, with delayed graft function requiring dialysis being together, the management of these disorders has clearly improved
reported in 5% of pediatric related living-donor allografts and to provide future generations of children with improved health and
15% of pediatric deceased-donor allografts. less burden from their kidney and urinary tract diseases.
Immunosuppression, both in terms of induction at the time of
transplantation and chronic maintenance therapy, are needed for
long-term function of the allograft. Most pediatric transplant centers SUGGESTED READINGS
now use antibody-based agents such as thymoglobulin or basiliximab Batlle D, Haque SK: Genetic causes and mechanisms of distal renal tubular
at the time of transplantation for induction therapy. Thereafter, acidosis, Nephrol Dial Transplant 27(10):3691–3704, 2012.
children are typically maintained on long-term immunosuppression Ben-Shalom E, Frishberg Y: Primary hyperoxalurias: diagnosis and treatment,
with a calcineurin inhibitor (tacrolimus or cyclosporine) and an Pediatr Nephrol 30(10):1781–1791, 2015.
antiproliferative agent such as mycophenolate mofetil. In the past Chand D, Swartz S, Tuchman S, et al: Dialysis in children and adolescents:
the pediatric nephrology perspective, Am J Kidney Dis 69(2):278–286,
decade, there has been a reduction in the use of steroids as part 2017.
of maintenance therapy, with many centers adopting steroid-free Dharnidharka VR, Fiorina P, Harmon WE: Kidney transplantation in children,
maintenance immunosuppression for children thought to be at lower N Engl J Med 371(6):549–558, 2014.
immunologic risk for rejection. Dodge WF, West EF, Smith EH, et al: Proteinuria and hematuria in school-
Infectious complications are a concern in all transplant recipients, children: epidemiology and early natural history, J Pediatr 88(2):327–347,
particularly in younger children who may not yet have acquired native 1976.
immunity to cytomegalovirus (CMV), Epstein-Barr virus, or the BK Edvardsson VO, Goldfarb DS, Lieske JC, et al: Hereditary causes of kidney
virus. Transplant centers commonly use valganciclovir as prophylaxis stones and chronic kidney disease, Pediatr Nephrol 28(10):1923–1942,
to reduce the risk for CMV disease. Trimethoprim-sulfamethoxazole is 2013.
ESCAPE Trial Group, Wühl E, Trivelli A, et al: Strict blood-pressure control
often used for prophylaxis against Pneumocystis carinii pneumonia early and progression of renal failure in children, N Engl J Med 361(17):1639–1650,
in the post-transplant period. However, chronic immunosuppression 2009.
also increases the risk for renal cell carcinoma, thyroid carcinoma, and Falkner B: Hypertension in children and adolescents: epidemiology and
cutaneous malignancies by nearly sevenfold in transplant recipients natural history, Pediatr Nephrol 25(7):1219–1224, 2010.
compared with the general pediatric population. Flynn JT, Kaelber DC, Baker-Smith CM, et al: Clinical practice guideline for
In the pediatric renal transplant population, the risk for acute screening and management of high blood pressure in children and
cellular rejection in the first year post-transplant is less than 10% adolescents, Pediatrics 140(3):e20171904, 2017.
in living-donor transplants and approximately 15% in deceased- Hernandez JD, Ellison JS, Lendvay TS: Current trends, evaluation, and
donor transplants. Generally the rates of acute rejection decrease management of pediatric nephrolithiasis, JAMA Pediatr 169(10):964–970,
2015.
over the life of a renal allograft. Although most rejection episodes Inker LA, Astor BC, Fox CH, et al: KDOQI US commentary on the 2012
are reversible, rejection episodes negatively affect long-term graft KDIGO clinical practice guideline for the evaluation and management of
function. Antibody-mediated rejection is increasingly being recognized CKD, Am J Kidney Dis 63(5):713–735, 2014.
as a cause of allograft dysfunction. The impact on antibody-mediated Kaddourah A, Basu RK, Bagshaw SM, et al: Epidemiology of acute kidney
rejection on long-term allograft function and the treatment strategies injury in critically ill children and young adults, N Engl J Med 376(1):11–20,
for antibody-mediated rejection are areas of ongoing investigation; 2017.
many centers are now using a combination of therapies for antibody- Little MH, McMahon AP: Mammalian kidney development: principles, progress,
mediated rejection including intravenous immunoglobulin, rituximab, and projections, Cold Spring Harb Perspect Biol 4(5):a008300, 2012.
and/or apheresis. Over time, all transplants will lose substantial Savige J, Gregory M, Gross O, et al: Expert guidelines for the management
of Alport syndrome and thin basement membrane nephropathy, J Am Soc
function as a result of a process-labeled chronic allograft nephropathy Nephrol 24(3):364–375, 2013.
for which there is no current therapeutic intervention. With this Torres VE, Harris PC, Pirson Y: Autosomal dominant polycystic kidney disease,
progressive decline in allograft function, nearly all pediatric renal Lancet 369(9569):1287–1301, 2007.
transplant recipients will eventually require either retransplantation
or a return to dialysis.
REFERENCES
KEY POINTS The complete reference list is available online at ExpertConsult.com.

• Peritoneal dialysis is the most common initial modality


for dialysis in children younger than 9 years of age and
less than 20 kg.
• The unique benefits of renal transplantation include
improved growth, neurocognitive development, and QoL.
• The risk for acute cellular rejection in the first year after
pediatric renal transplantation is less than 10% in living-
donor transplants.
• The risk for acute cellular rejection in the first year
post-transplant is approximately 15% in deceased-donor
transplants.
Chapter 21  Urologic Aspects of Pediatric Nephrology 357.e1

Group for Growth Hormone Treatment in Chronic Renal Failure, N Engl


REFERENCES J Med 343(13):923–930, 2000.
Abitbol C, Zilleruelo G, Freundlich M, et al: Quantitation of proteinuria Hernandez JD, Ellison JS, Lendvay TS: Current trends, evaluation, and
with urinary protein/creatinine ratios and random testing with dipsticks management of pediatric nephrolithiasis, JAMA Pediatr 169(10):964–970,
in nephrotic children, J Pediatr 116(2):243–247, 1990. 2015.
Batlle D, Haque SK: Genetic causes and mechanisms of distal renal tubular Hiraki LT, Feldman CH, Liu J, et al: Prevalence, incidence, and demo- graphics
acidosis, Nephrol Dial Transplant 27(10):3691–3704, 2012. of systemic lupus erythematosus and lupus nephritis from 2000 to 2004
Ben-Shalom E, Frishberg Y: Primary hyperoxalurias: diagnosis and treatment, among children in the US Medicaid beneficiary population, Arthritis Rheum
Pediatr Nephrol 30(10):1781–1791, 2015. 64(8):2669–2676, 2012.
Bergmann C, Senderek J, Schneider F, et al: PKHD1 mutations in autosomal Hogg RJ, Portman RJ, Milliner D, et al: Evaluation and management of
recessive polycystic kidney disease (ARPKD), Hum Mutat 23(5):453–463, proteinuria and nephritic syndrome in children: recommendation from
2004. a pediatric nephrology panel established at the National Kidney Foundation
Braun DA, Lawson JA, Gee HY, et al: Prevalence of monogenic causes in Conference on proteinuria, albuminuria, risk assessment, detection, and
pediatric patients with nephrolithiasis or nephrocalcinosis, Clin J Am Soc elimination (Parade), Pediatrics 105(6):1242–1249, 2000.
Nephrol 11(4):664–672, 2016. Hothi DK, Stronach L, Sinnott K: Home hemodialysis in children, Hemodial
Büscher AK, Beck BB, Melk A, et al: Rapid response to cyclosporin A and Int 20(3):349–357, 2016.
favorable renal outcome in nongenetic versus genetic steroid-resistant Igarashi P, Somlo S: Genetics and pathogenesis of polycystic kidney disease,
nephrotic syndrome, Clin J Am Soc Nephrol 11(2):245–253, 2016. J Am Soc Nephrol 13(9):2384–2398, 2002.
Cabral DA, Canter DL, Muscal E, et al: Comparing presenting clinical features Inker LA, Astor BC, Fox CH, et al: KDOQI US commentary on the 2012
of 48 children with microscopic polyangiitis against 183 having granulo- KDIGO clinical practice guideline for the evaluation and management of
matosis with polyangiitis. An ARChiVe cohort study, Arthritis Rheumatol CKD, Am J Kidney Dis 63(5):713–735, 2014.
68(10):2514–2526, 2016. Kaddourah A, Basu RK, Bagshaw SM, et al: Epidemiology of acute kidney
Carlisle EJF, Donnelly SM, Halprin ML: Renal tubular acidosis (RTA): recognize injury in critically ill children and young adults, N Engl J Med 376(1):11–20,
the ammonium defect and pHorget the urine pH, Pediatr Nephrol 2017.
5(2):242–248, 1991. Kashtan CE, Ding J, Gregory M, et al: Clinical practice recommendations for
Chand D, Swartz S, Tuchman S, et al: Dialysis in children and adolescents: the treatment of Alport syndrome, Pediatr Nephrol 28(1):5–11, 2013.
the pediatric nephrology perspective, Am J Kidney Dis 69(2):278–286, Kaskal FJ, Kumar FM, Feld LG, et al: Renal reabsorption of phosphate during
2017. development: tubular events, Pediatr Nephrol 2(1):129–134, 1988.
Combes AN, Davies JA, Little MH: Cell-cell interactions driving kidney Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Work
morphogenesis, Curr Top Dev Biol 112:467–508, 2015. Group: KDIGO clinical practice guideline for the diagnosis, evaluation,
Cossey LN, Walker PD, Larsen CP: Phospholipase A2 receptor staining in prevention, and treatment of chronic kidney disease–mineral and bone
pediatric idiopathic membranous glomerulopathy, Pediatr Nephrol disorder (CKD-MBD), Kidney Int Suppl 113:S1–S130, 2009.
28(12):2307–2311, 2013. Kidney Disease: Improving Global Outcomes (KDIGO): Glomerulonephritis
Devarajan P: Pediatric acute kidney injury: different from acute renal failure Work Group. KDIGO clinical practice guideline for glomerulonephritis,
but how and why?, Curr Pediatr Rep 1(1):34–40, 2013. Kidney Int Suppl 2(2):139–274, 2012.
Devuyst O, Thakker RV: Dent’s disease, Orphanet J Rare Dis 5:28, 2010. Kidney Disease Outcomes Quality Initiative (KDOQI) clinical practice guideline
Dharnidharka VR, Fiorina P, Harmon WE: Kidney transplantation in children, for nutrition in children with CKD: 2008 update. Executive summary, Am
N Engl J Med 371(6):549–558, 2014. J Kidney Dis 53(3 Suppl 2):S11–S104, 2009.
Din-Dzietham R, Liu Y, Bielo MV, et al: High blood pressure trends in children Kiryluk K, Moldoveanu Z, Sanders JT, et al: Aberrant glycosylation of IgA1
and adolescents in national surveys, 1963 to 2002, Circulation 116(13):1488– is inherited in both pediatric IgA nephropathy and Henoch-Schönlein
1496, 2007. purpura nephritis, Kidney Int 80(1):79–87, 2011.
Dodge WF, West EF, Smith EH, et al: Proteinuria and hematuria in school- Kliger AS, Foley RN, Goldfarb DS, et al: KDOQI US commentary on the
children: epidemiology and early natural history, J Pediatr 88(2):327–347, 2012 KDIGO Clinical Practice Guideline for Anemia in CKD, Am J Kidney
1976. Dis 62(5):849–859, 2013.
Dudley J, Smith G, Llewelyn-Edwards A, et al: Randomized, double-blind, Kyle UG, Akcan-Arikan A, Orellana RA, et al: Nutrition support among critically
placebo-controlled trial to determine whether steroids reduce the incidence ill children with AKI, Clin J Am Soc Nephrol 8(4):568–574, 2013.
and severity of nephropathy in Henoch-Schönlein purpura, Arch Dis Child Lameire N, Van Biesen W, Vanholder R: Epidemiology of acute kidney injury
98(10):756–763, 2013. in children worldwide, including developing countries, Pediatr Nephrol
Edvardsson VO, Goldfarb DS, Lieske JC, et al: Hereditary causes of kidney 32(8):1301–1314, 2017.
stones and chronic kidney disease, Pediatr Nephrol 28(10):1923–1942, Lau KK, Giglia L, Chan H, et al: Management of children after renal trans-
2013. plantation: highlights for general pediatricians, Transl Pediatr 1(1):35–46,
ESCAPE Trial Group, Wühl E, Trivelli A, et al: Strict blood-pressure control 2012.
and progression of renal failure in children, N Engl J Med 361(17):1639– Little MH, McMahon AP: Mammalian kidney development: principles, progress,
1650, 2009. and projections, Cold Spring Harb Perspect Biol 4(5):a008300, 2012.
Falkner B: Hypertension in children and adolescents: epidemiology and Loirat C, Fakhouri F, Ariceta G, et al: An international consensus approach
natural history, Pediatr Nephrol 25(7):1219–1224, 2010. to the management of atypical hemolytic uremic syndrome in children,
Fischbach M, Edefonti A, Schröder C, et al: European Pediatric Dialysis Working Pediatr Nephrol 31(1):15–39, 2015.
Group. Hemodialysis in children: general practical guidelines, Pediatr Lovric S, Ashraf S, Tan W, et al: Genetic testing in steroid-resistant nephrotic
Nephrol 20(8):1054–1066, 2005. syndrome: when and how?, Nephrol Dial Transpl 31(11):1802–1813,
Fitzgerald JC, Basu RK, Akcan-Arikan A, et al: Acute kidney injury in pediatric 2015.
severe sepsis: an independent risk factor for death and new disability, Crit Lurbe E, Cifkova R, Cruickshank JK, et al: Management of high blood pressure
Care Med 44(12):2241–2250, 2016. in children and adolescents: recommendations of the European Society
Flynn JT, Daniels SR: Pharmacologic treatment of hypertension in children of Hypertension, J Hypertens 27(9):1719–1742, 2009.
and adolescents, J Pediatr 149(6):746–754, 2006. Ma A, Schroff R, Hothi D, et al: A comparison of arteriovenous fistulas and
Flynn JT, Kaelber DC, Baker-Smith CM, et al: Clinical practice guideline for central venous lines for long term chronic hemodialysis, Pediatr Nephrol
screening and management of high blood pressure in children and 28(2):321–326, 2013.
adolescents, Pediatrics 140(3):e20171904, 2017. Mehta RL, Burdmann EA, Cerdá J, et al: Recognition and management of
Gattineni J, Baum M: Developmental changes in renal tubular transport - an acute kidney injury in the International Society of Nephrology 0by25
overview, Pediatr Nephrol 30(12):2085–2098, 2015. Global Snapshot: a multinational cross-sectional study, Lancet
Gleason CA: Prostaglandins and the developing kidney, Semin Perinatol 387(10032):2017–2025, 2016.
11(1):12–21, 1987. Mina R, von Scheven E, Ardoin SP, et al: Consensus treatment plans for
Goldberg AM, Amaral S, Moudgil A: Developing a framework for evaluating induction therapy of newly diagnosed proliferative lupus nephritis in
kidney transplantation candidacy in children with multiple comorbidities, juvenile systemic lupus erythematosus, Arthritis Care Res (Hoboken)
Pediatr Nephrol 30(1):5–13, 2015. 64(3):375–383, 2012.
Greenbaum LA, Shaefer F: The decision to initiate dialysis in a pediatric Mitsnefes MM: Cardiovascular disease in children with chronic kidney disease,
patient. In Warady BA, Schaefer F, Alexander SR, editors: Pediatric dialysis, J Am Soc Nephrol 23(4):578–585, 2012.
New York, NY, 2012, Springer, pp 85–100. Moe OW, Seldin DW, Baum M: The Fanconi syndrome. In Lifton R, Somlo
Haffner D, Schaefer F, Nissel R, et al: Effect of growth hormone treatment S, Seldin DW, et al, editors: Genetic diseases of the kidney, New York, NY,
on the adult height of children with chronic renal failure. German Study 2009, Elsevier, pp 171–198.
357.e2 PART III  Pediatric Urology

Muntner P: Trends in blood pressure among children and adolescents, JAMA Seyberth HW, Schlingmann KP: Bartter- and Gitelman-like syndromes: salt-
291(17):2107, 2004. losing tubulopathies with loop or DCT defects, Pediatr Nephrol 26(10):1789–
National High Blood Pressure Education Program Working Group on High 1802, 2011.
Blood Pressure in Children and Adolescents: The fourth report on the Short KM, Smyth IM: The contribution of branching morphogenesis to kidney
diagnosis, evaluation, and treatment of high blood pressure in children development and disease, Nat Rev Nephrol 12(12):754–767, 2016.
and adolescents, Pediatrics 114(2 Suppl 4th Report):555–576, 2004. Slater MB, Gruneir A, Rochon PA, et al: Risk factors of acute kidney injury
Nazzal L, Puri S, Goldfarb DS: Enteric hyperoxaluria: an important cause of in critically ill children, Pediatr Crit Care Med 17(9):e391–e398, 2016.
end-stage kidney disease, Nephrol Dial Transplant 31(3):375–382, 2016. Smith-Bindman R, Aubin C, Bailitz J, et al: Ultrasonography versus computed
Nesterova G, Gahl WA: Cystinosis: the evolution of a treatable disease, Pediatr tomography for suspected nephrolithiasis, N Engl J Med 371(12):1100–1110,
Nephrol 28(1):51–59, 2013. 2014.
O’Shaughnessy KM: Gordon syndrome: a continuing story, Pediatr Nephrol Sutherland SM, Ji J, Sheikhi FH, et al: AKI in hospitalized children: epidemiol-
30(11):1903–1908, 2015. ogy and clinical associations in a national cohort, Clin J Am Soc Nephrol
Palmer BF: Regulation of potassium homeostasis, Clin J Am Soc Nephrol 8(10):1661–1669, 2013.
10(6):1050–1060, 2015. Taler SJ, Agarwal R, Bakris GL, et al: KDOQI US commentary on the 2012
Pickering MC, D’Agati VD, Nester CM, et al: C3 glomerulopathy: consensus KDIGO clinical practice guideline for management of blood pressure in
report, Kidney Int 84(6):1079–1089, 2013. CKD, Am J Kidney Dis 62(2):201–213, 2013.
Quigley R, Baum M: Neonatal acid base balance and disturbances, Semin Teeninga N, Kist-van Holthe JE, van Rijswijk N, et al: Extending prednisolone
Perinatol 28(2):97–102, 2004. treatment does not reduce relapses in childhood nephrotic syndrome, J
Quigley R: Developmental changes in renal function, Curr Opin Pediatr Am Soc Nephrol 24(1):149–159, 2012.
24(2):184–190, 2012. Teoh CW, Robinson LA, Noone D: Perspective on edema in childhood
Redwine KM, Acosta AA, Poffenbarger T, et al: Development of hypertension nephrotic syndrome, Am J Physiol Renal Physiol 309(7):F575–F582,
in adolescents with pre-hypertension, J Pediatr 160(1):98–103, 2012. 2015.
Sarnak MJ, Bloom R, Muntner P, et al: KDOQI US commentary on the 2013 Torres VE, Harris PC, Pirson Y: Autosomal dominant polycystic kidney disease,
KDIGO clinical practice guideline for lipid management in CKD, Am J Lancet 369(9569):1287–1301, 2007.
Kidney Dis 65(3):354–366, 2015. United States Renal Data System (USRDS): 2016 USRDS annual data report:
Sas DJ: An update on the changing epidemiology and metabolic risk factors in Epidemiology of kidney disease in the United States. Bethesda, MD: National
pediatric kidney stone disease, Clin J Am Soc Nephrol 6(8):2062–2068, 2011. Institutes of Health, National Institute of Diabetes and Digestive and
Savige J, Gregory M, Gross O, et al: Expert guidelines for the management Kidney Diseases, 2016.
of Alport syndrome and thin basement membrane nephropathy, J Am Soc Vehaskari VM: Heritable forms of hypertension, Pediatr Nephrol 24(10):1929–
Nephrol 24(3):364–375, 2013. 1937, 2009.
Savige J, Colville D, Rheault M, et al: Alport syndrome in women and girls, Vehaskari VM, Rapola J: Isolated proteinuria: analysis of a school age popula-
Clin J Am Soc Nephrol 11(9):1713–1720, 2016. tion, J Pediatr 101(5):661–668, 1982.
Schell C, Wanner N, Huber TB: Glomerular development–shaping the multi- Vehaskari VM, Rapola J, Koskimies O, et al: Microscopic hematuria in school
cellular filtration unit, Semin Cell Dev Biol 36:39–49, 2014. children: epidemiology and clinicopathologic evaluation, J Pediatr 95(5
Schell-Feith EA, Kist-van Holthe JE, van der Heijden AJ: Nephrocalcinosis in Pt 1):676–684, 1979.
preterm neonates, Pediatr Nephrol 25(2):221–230, 2010. Warady BA, Bakkaloglu S, Newland J, et al: ISPD guidelines/recommendations:
Schwartz GJ, Muñoz A, Schneider MF, et al: New equations to estimate GFR consensus guideline for the prevention and treatment of catheter-related
in children with CKD, J Am Soc Nephrol 20(3):629–637, 2009. infections and peritonitis in pediatric patients receiving peritoneal dialysis,
Selewski DT, Cornell TT, Heung M, et al: Validation of the KDIGO acute Perit Dial Int 32(Suppl 2):S32–S86, 2012.
kidney injury criteria in a pediatric critical care population, Intensive Care Zhou H, Satlin LM: Renal potassium handling in healthy and sick newborns,
Med 40(10):1481–1488, 2014. Semin Perinatol 28(2):103–111, 2004.

You might also like