throbber
Pediatr Nephrol
`DOI 10.1007/s00467-011-1838-5
`
`EDUCATIONAL REVIEW
`
`Hyperammonemia in review: pathophysiology, diagnosis,
`and treatment
`
`Ari Auron & Patrick D. Brophy
`
`Received: 23 September 2010 / Revised: 9 January 2011 / Accepted: 12 January 2011
`# IPNA 2011
`
`Abstract Ammonia is an important source of nitrogen and is
`required for amino acid synthesis. It is also necessary for
`normal acid base balance. When present in high concentra
`tions, ammonia is toxic. Endogenous ammonia intoxication
`can occur when there is impaired capacity of the body to
`excrete nitrogenous waste, as seen with congenital enzymatic
`deficiencies. A variety of environmental causes and medica
`tions may also lead to ammonia toxicity. Hyperammonemia
`refers to a clinical condition associated with elevated
`ammonia levels manifested by a variety of symptoms and
`signs, including significant central nervous system (CNS)
`abnormalities. Appropriate and timely management requires a
`solid understanding of the fundamental pathophysiology,
`differential diagnosis, and treatment approaches available.
`The following review discusses the etiology, pathogenesis,
`differential diagnosis, and treatment of hyperammonemia.
`
`Keywords Hyperammonemia . Dialysis . Urea cycle
`defects . Treatment . Pathophysiology
`
`Introduction
`
`Ammonia is an important source of nitrogen and is required
`for amino acid synthesis. Nitrogenous waste results from
`
`A. Auron
`Blank Memorial Hospital for Children,
`1200 Pleasant St,
`Des Moines, IA 50309, USA
`
`P. D. Brophy (*)
`Department of Pediatrics, Pediatric Nephrology, Dialysis &
`transplantation, University of Iowa Children’s Hospital,
`200 Hawkins Dr,
`Iowa City, IA 52242, USA
`e-mail: patrick-brophy@uiowa.edu
`
`the breakdown and catabolism of dietary and bodily proteins,
`respectively. In healthy individuals, amino acids that are not
`needed for protein synthesis are metabolized in various
`chemical pathways, with the rest of the nitrogen waste being
`converted to urea. Ammonia is important for normal animal
`acid base balance. During exercise, ammonia is produced in
`skeletal muscle from deamination of adenosine monophos
`phate and amino acid catabolism. In the brain, the latter
`processes plus the activity of glutamate dehydrogenase
`mediate ammonia production. After formation of ammonium
`from glutamine, α ketoglutarate, a byproduct, may be
`degraded to produce two molecules of bicarbonate, which
`are then available to buffer acids produced by dietary sources.
`Ammonium is excreted in the urine, resulting in net acid loss.
`The ammonia level generally remains low (<40 mmol/L) due
`to the fact that most ammonia produced in tissue is converted
`to glutamine. Glutamine is also excreted by the kidneys and
`utilized for energy production by gut cells, which convert the
`nitrogen byproduct into alanine, citrulline, and ammonia,
`which are transported to the liver via the bloodstream.
`Ammonia enters the urea cycle in hepatocytes or is ultimately
`converted to glutamine. [1 5]
`Ammonia is toxic when present in high concentrations.
`Endogenous ammonia intoxication can occur when there is
`impaired capacity of the body to excrete nitrogenous waste,
`as seen with congenital enzymatic deficiencies. Patients
`with urea cycle defects (UCD), organic acidemias, fatty
`acid oxidation defects, bypass of
`the major site of
`detoxification (liver) (such as that seen in cirrhosis), Reye
`syndrome, postchemotherapy, or exposure to various toxins
`and drugs can all present with elevations in ammonia.
`Delayed diagnosis or
`treatment of hyperammonemia,
`irrespective of the etiology, leads to neurologic damage
`and potentially a fatal outcome, and thus it becomes a
`medical emergency when present. [6]
`
`Page 1 of 16
`
`Horizon Exhibit 2008
`Par v. Horizon
`IPR2017-01769
`
`

`

`Ammonia toxicity (pathogenesis)
`
`Hyperammonemia refers to a clinical condition character-
`ized by elevated serum ammonia levels and manifests with
`hypotonia, seizures, emesis, and abnormal neurologic
`changes (including stupor). It may also exert a diabetogenic
`effect mediated by ammonia inhibition of
`the insulin
`secretion induced by a glucose load [7]. Hyperammonemia
`can cause irreparable damage to the developing brain, with
`presenting symptoms such as posturing, cognitive impair-
`ment (mental retardation), seizures, and cerebral palsy. The
`total duration of hyperammonemic coma and maximum
`ammonia level (but not the rapidity of ammonia removal),
`is negatively correlated with the patient’s neurological
`outcome. This is concerning when ammonia levels at
`presentation exceed 300 mmol/L. If left untreated,
`the
`outcome can be fatal. [7, 8]
`
`Pathologic brain changes induced by hyperammonemia
`
`Deaths secondary to hyperammonemia reveal upon autop
`sia cadaverum brain edema, brainstem herniation, astrocyt-
`ic swelling, and white-matter damage [8]. Neuropathologic
`findings seen in children with hyperammonemia include
`ventriculomegaly, cerebral cortical atrophy, basal ganglia
`lesions, neuronal loss, gliosis, intracranial bleed, and areas
`of focal cortical necrosis and myelination deficiencies.
`Electroencephalogram (EEG) denotes slow delta-wave
`activity, consistent with a metabolic disturbance [9–19].
`The brains of neonates and infants with hyperammonemia
`demonstrate areas of cortical atrophy or gray/white matter
`hypodensities, which are remnants of lost neuronal fibers.
`There is also an increased number of oligodendroglia and
`microglia in striatum. [17]
`
`Amino acid disturbances in the brain
`
`Glutamine
`
`Glutamine synthesis mediated by the astrocytic enzyme
`glutamine synthetase is the major pathway for ammonia
`detoxification in the brain and cerebrospinal fluid. In
`conditions in which excess ammonia exists, brain osmoti-
`cally active glutamine concentrations increase, leading to
`astrocytic damage and swelling. Consequently, the astro-
`cyte promotes intercellular glutamate release, which
`decreases the glutamate intracellular pool and ultimately
`leads to cell death of the glutamatergic neurons [17, 20–22]
`
`Arginine
`
`Although arginine is an essential amino acid for the fetus
`and the neonate, they still synthesize part of it through
`
`Pediatr Nephrol
`
`intestinal expression of carbamyl phosphate synthetase −1
`(CPS), ornithine transcarbamylase (OTC), argininosucci-
`nate synthetase (AS), and argininosuccinase (AL). In the
`adult, arginine is synthesized through two pathways: in
`the intestine, CPS-1 and OTC synthesize citrulline; and
`AS and AL in the renal proximal
`tubules synthesize
`arginine from citrulline. Thus, patients with UCD (except
`Arginase deficiency I) have low serum arginine levels
`and need this amino acid to be replaced, although
`citrulline is a better replacement choice for patients with
`CPS-1 and OTC [17, 23]
`
`Alterations in neurotransmission systems
`
`Glutamatergic system
`
`An excess of extracellular glutamate accumulates in
`brain when the latter
`is exposed to ammonia. The
`mechanism involves astrocytic swelling, pH and calcium
`(Ca2+)-dependent
`release of glutamate by astrocytes,
`inhibition of glutamate uptake by astrocytes through
`inhibition of
`the glutamine aspartate transporter
`(GLAST), and excess depolarization of glutamatergic
`neurons. Excess extracellular glutamate secondary to
`ammonia exposure promotes toxic cellular hyperexcit-
`ability through activation of N-methyl-D-aspartate
`(NMDA) receptors and leads to alteration in nitric oxide
`(NO) metabolism, disturbances in sodium/potassium
`adenosine triphosphatase (Na+/K+−ATPase),
`(causing
`neuronal sodium/potassium influx), ATP shortage, mito-
`chondrial dysfunctions, free-radical accumulation, and
`oxidative stress, leading ultimately to cell death. Toxic
`stimulation of NMDA receptors can also be caused by
`quinolinic acid, which is an oxidation product of
`tryptophan acting on NMDA receptors and plays a role
`in the neurotoxicity seen in UCD patients (associated with
`death of spiny neurons in the striatum). Tryptophan and
`quinolinic acid concentration increases in brain and
`cerebrospinal
`fluid,
`respectively,
`in the presence of
`hyperammonemia. [17, 24–28]. In experimental animal
`models, as a protective cellular mechanism against excitotox-
`icity, a reduction in NMDA receptors is noted secondary to the
`increased release of glutamate. Nevertheless, in patients with
`UCD, despite the downregulation of NMDA receptors, there
`is still a certain degree of cellular toxicity that leads to cell
`death. Interestingly, prolonged cortical neuron survival has
`been reported to occur in hyperammonemic animals given the
`NMDA receptor antagonists MK-801 and 2-amino-5-phos-
`phonovaleric acid (APV) [29]. The glutamatergic stimuli
`secondary to prolonged brain exposure to ammonia can also
`affect activation of other neurotransmission systems, such as
`gamma-aminobutyric acidd (GABA) or benzodiazepine
`receptors [30]
`
`Page 2 of 16
`
`

`

`Pediatr Nephrol
`
`Cholinergic system
`
`This system plays a key role in cognitive development.
`Cholinergic neurons loss, with decreased choline acetyl-
`transferase (ChAT) activity level, has been observed in
`animal models with hyperammonemia. [17, 31]
`
`Serotonergic system
`
`s e r o t o n i n ) a n d 5 -
`f o r
`Tr y p t o p h a n ( p r e c u r s o r
`hydroxyindoleacetic acid (5-HT) (metabolite of serotonin)
`are elevated in cerebrospinal fluid of children with UCD.
`Experimental hyperammonemic animal models demon-
`strate alterations in serotonergic activity through loss of 5-
`HT2 receptors and an increase in 5-HT1A receptors. These
`physiologic responses may play a role in anorexia and sleep
`disturbances noted in children with UCD [32–34]
`
`Cerebral energy deficit
`
`Hyperammonemic experimental animal models develop
`increased superoxide production and decreased activities
`of antioxidant enzymes in brain cells. Blockage of nitric
`oxide synthase by nitroarginine or NMDA receptor antag-
`onists prevent the latter changes, suggesting therefore that
`ammonia-induced oxidative stress is due to the increased
`nitric oxide formation as a consequence of NMDA receptor
`activation [34]. Indeed, excessive formation of nitric oxide
`seen with ammonia exposure impairs mitochondrial respi-
`ration, depletes ATP reserves, and increases free radicals
`and oxidative stress, which ultimately leads to neuron death
`[17, 26]. Creatine, too, plays a key role in cellular energy
`formation and is decreased in the brain cells of hyper-
`ammonemic animals due to altered creatine transport and
`synthesis. Acetyl-L-carnitine enhances restoration of ATP
`and phosphocreatine levels in ischemia and has been
`reported to enhance the recovery of cerebral energy deficits
`caused by hyperammonemia, thus having a neuroprotective
`effect (through restoration of cytochrome C oxidase activity
`and free-radical scavenging) [35–37]. Cotreatment with
`creatine (while there is exposure to ammonia) protects
`axonal growth, thereby suggesting a possible neuroprotec-
`tive role for this substance. [17, 38]
`
`regulated kinase (ERK)1/2, stress-activated protein kinase/c-
`Jun N-terminal kinase (SAPK/JNK), and p38 in astrocytes.
`Phosphorylation of ERK1/2 and p38 are responsible for
`ammonia-induced astrocyte swelling, and phosphorylation of
`SAPK/JNK and p38 are responsible for ammonia-induced
`inhibition of astrocytic uptake of glutamate. Adjunct treatment
`with CNTF has shown to induce protective effects on
`oligodendrocytes by reversing the demyelination induced by
`hyperammonemia [17, 39, 40]
`
`K+ and water channels
`
`Although the brain edema that develops in hyperammonemic
`patients with UCD has been thought to occur secondary to
`astrocytic swelling, recently, this has been challenged. In
`astrocytes of hyperammonemic animals, it has been noted that
`a reduction of proteins that regulate the K+ and water
`transport at the blood–brain barrier, including connexin 43,
`aquaporin 4 (aquaporins are membrane proteins that mediate
`water movement across the membrane), and the astrocytic
`inward-rectifying K+ channels Kir4.1 and Kir5.1, allows
`ammonia to easily permeate through the aquaporin channels.
`In the setting of hyperammonemia, downregulation of
`aquaporins occurs (reflecting a protective mechanism of
`astrocytes to prevent ammonia entering the cell), but
`simultaneously, the excretion of K+ and water is altered,
`and an increase in brain extracellular K+ and water develops,
`contributing to brain edema formation [41, 42].
`
`Differential diagnosis of hyperammonemia
`
`Diagnostically, the presence of acidosis, ketosis, hypoglyce-
`mia, and low bicarbonate levels indicate that the root cause of
`hyperammonemia is probably due to an organic acidemia,
`systemic carnitine deficiency, Reye syndrome, toxins, drug
`effect, or liver disease. On the other hand, if there is normal
`lactate and no metabolic acidosis associated with the hyper-
`ammonemia, then UCD, dibasic aminoaciduria, or transient
`hyperammonemia of the newborn are more likely. The
`association of elevated liver enzymes with hyperammonemia
`is most consistent after insult from hepatotoxins, Reye
`syndrome, or carnitine deficiency [6] (Table 1).
`
`Cerebral exposure
`
`Urea cycle disorders
`
`Once the brain has been exposed to ammonia, intracerebral
`endogenous protective mechanisms that prevent or limit brain
`damage are triggered. Astrocytes express an injury-associated
`survival protein—ciliary neurotrophic factor (CNTF)—which
`is upregulated by ammonia through p38 mitogen-activated
`protein kinase (MAPK) activation. Ammonia activates a
`variety of kinase pathways including extracellular signal
`
`The urea cycle produces arginine by de novo synthesis. Six
`enzymatic reactions comprise the cycle, which occurs in the
`liver. The first three reactions are located intramitochond-
`rially and the rest are cytosolic. The enzymatic substrates
`are ammonia, bicarbonate, and aspartate. After each turn of
`the cycle, urea is formed from two atoms of nitrogen (see
`Fig. 1) [43]. Key enzymes in the urea cycle include CPS-1,
`
`Page 3 of 16
`
`

`

`Table 1 Differential diagnosis of hyperammonemia
`
`Causes
`
`Disease
`
`Urea cycle disorders
`
`Organic acidemias
`
`Drug related causes
`
`Miscellaneous causes
`
`Argininosuccinic aciduria
`Citrullinemia
`Carbamyl Phosphate Synthetase (CPS)
`deficiency 1
`Ornithine transcarbamylase deficiency
`Argininemia
`Propionic acidemia
`Methylmalonic acidemia
`Isovaleric acidemia
`Maple syrup urine disease
`Dibasic aminoacidurias
`Type 1
`Type 2 (Lysinuric protein intolerance)
`Hyperammonemia hyperornithinemia
`homocitrullinuria
`Transient hyperammonemia of the
`newborn
`Salicylates
`Carbamazepine
`Valproic acid
`Topiramate
`Tranexamic acid
`Chemotherapeutic agents:
`Aspariginase 5 fluorouracil
`Rituximab
`Hypoglycin intoxication
`Pregnancy
`Distal Renal Tubular Acidosis (RTA)
`Carnitine transport defects
`Urinary tract dilatation
`Reye’s syndrome
`
`OTC, AS, AL, and arginase. There are cofactors necessary
`for optimal enzyme activity, the most clinically important
`of which is N-acetyl glutamate (NAG). In UCD, there is an
`increase in glutamine, which transports nitrogen groups to
`the liver for ammonia formation within hepatic mitochon-
`dria, and ammonium combines with bicarbonate in the
`presence of NAG to form carbamyl phosphate. If NAG is
`absent, hyperammonemia develops [1, 6]
`The presence of an enzymatic defect in the urea cycle
`results in arginine becoming an essential amino acid (except in
`arginase deficiency), and waste nitrogen accumulates mainly
`as ammonia and glutamine [6] Measurement of plasma amino
`acids aids in differentiating between the different urea cycle
`enzyme deficiencies. Citrulline is the product of CPS and
`OTC and the substrate for AS and AL, thus its value is of
`critical importance. In CPS and OTC deficiencies, plasma
`citrulline levels are low. These two defects are differentiated
`
`Pediatr Nephrol
`
`that orotic acid is elevated in OTC
`by the fact
`deficiency and absent
`in CPS deficiency.
`In AS
`(citrullinemia) and AL (argininosuccinic aciduria) defi-
`ciencies, plasma citrulline levels are increased (see
`Fig. 2). If the serum citrulline level
`is normal,
`then
`disorders such as argininemia, lysinuric protein intoler-
`ance, and the hyperammonemia-hyperornithinemia-
`homocitrullinuria syndrome should be considered [6]
`The prevalence of UCD is estimated at 1:8,200 in the
`USA. The overall incidence of defects presenting clinically
`is estimated at approximately 1 in 45,000 live births [1, 44].
`Each specific disorder is considered below.
`
`Argininosuccinic aciduria: argininosuccinase deficiency 1
`
`The genetic deficiency responsible for this disorder is
`located on chromosome 7-q11.2. The diagnosis is suspected
`by observing hyperammonemia without acidosis in the
`presence of increased AL levels in plasma and urine.
`Plasma citrulline is also elevated. Measuring AL activity in
`erythrocytes or fibroblasts confirms the diagnosis. Distin-
`guishing clinical
`features include mental
`retardation,
`trichorrhexis nodosa (fragile hair with a nodular appear-
`ance), and an erythematous maculopapular skin rash. The
`latter two are associated with arginine deficiency and
`disappear with arginine supplementation. Liver involve-
`ment is characterized by hepatomegaly and elevated trans-
`aminases, and the associated histopathology displays
`enlarged hepatocytes and fibrosis [6, 45]
`
`Citrullinemia: argininosuccinate synthetase deficiency
`
`The genetic deficiency responsible for this disorder is
`located on chromosome 9q34. Common findings include
`hyperammonemia and low serum urea nitrogen and
`arginine levels, but
`the pathognomonic finding is a
`markedly elevated plasma citrulline level in the absence
`of AS. Affected individuals are able to partially incorporate
`the waste nitrogen into urea-cycle intermediates, which
`makes treatment easier [6, 45]
`
`Carbamyl phosphate synthetase (CPS) deficiency 1
`
`This represents the most severe form of UCD, with early
`development of hyperammonemia in the neonatal period,
`although it can also become apparent in adolescence. The
`genetic deficiency responsible for this disorder is located on
`chromosome 2q35. Biochemical findings in this entity
`include recurrent hyperammonemia, absent citrulline, and
`low levels of arginine, urea nitrogen, and urinary orotic
`acid. Diagnosis is confirmed by demonstrating <10%
`normal CPS activity in hepatic, rectal, or duodenal tissue
`[6, 45, 46].
`
`Page 4 of 16
`
`

`

`Pediatr Nephrol
`
`Fig. 1 Urea cycle pathway. Black arrows indicate primary pathway.
`Yellow arrows show alternative pathways used to eliminate nitrogen in
`patients with urea cycle defects. Enzymes are in blue. CPS
`carbamoylphosphate synthetase, OTC ornithine transcarbamoylase,
`
`AS argininosuccinate synthetase, AL argininosuccinate lyase, ARG
`arginase, NAGS N acetylglutamate synthase. Reproduced with
`permission from Walters and Brophy [43]
`
`Ornithine transcarbamylase deficiency
`
`and elevated urinary orotic acid. Liver biopsy helps confirm
`the diagnosis [1, 45, 47, 48]
`
`This represents the most common urea cycle enzyme
`defect, with an incidence of 1 in 14,000, and is the only
`one inherited as a sex-linked trait. OTC catalyzes the
`reaction of ornithine and carbamoyl phosphate to produce
`the amino acid citrulline. The OTC gene is located on the
`short arm of
`the X chromosome at Xp21, which is
`expressed specifically in the liver and gut. Around 350
`pathological mutations have been reported, and in approx-
`imately 80% of patients, a mutation is found. In girls in
`whom there is partial expression of the X-linked OTC
`deficiency disorder, mild symptoms can be seen. Only 15%
`of female carriers are symptomatic, and most asymptomatic
`carriers have a normal IQ score. Biochemical diagnostic
`findings include hyperammonemia, absent serum citrulline,
`
`Argininemia: arginase deficiency 1
`
`Afflicted patients do not develop symptoms in infancy.
`Children develop toe walking as a characteristic feature,
`which progresses to spastic diplegia. Finding elevated
`serum arginine, urine orotic acid, and ammonia levels
`makes the diagnosis.
`
`Organic acidemias
`
`This group of autosomally recessive inherited disorders
`results from enzyme deficiencies in amino acid degra-
`dation pathways. Most of
`the disorders are due to
`
`Page 5 of 16
`
`

`

`Fig. 2 Etiology of hyperammo
`nemia. Urea cycle defects:
`ornithine transcarbamylase
`(OTC); carbamyl phosphate
`synthetase (CPS);
`argininosuccinate synthetase
`(ASS); transient
`hyperammonemia of the
`newborn (THN). Reproduced
`with permission from Walters
`and Brophy [43]
`
`Pediatr Nephrol
`
`Hyperammonemia
`
`Blood pH and
`HCO3
`
`Acidosis
`
`Urine Organic
`Acids
`
`Organic
`Acidemias
`
`Low
`
`Low or Absent
`
`Increased
`
`Citrulline
`
`Normal or Slightly
`Increased
`
`Argininosuccinic
`Acid
`
`No
`Acidosis
`
`Plasma Amino
`Acids
`
`Significantly
`Increased
`
`Citullinemia
`
`CPS
`
`OTC
`
`Normal
`
`Increased
`
`THN
`
`ASS
`
`defective metabolism of branched-chain amino acids
`(leucine,
`isoleucine, and valine),
`tyrosine, homocys-
`teine, methionine, threonine, lysine, hydroxylysine, and
`tryptophan. This results
`in the accumulation and
`excretion of non-amino (organic) acids in plasma and
`urine. Hallmark findings in organic acid disorders
`include high anion gap metabolic acidosis, ketoacidosis
`(due to the accumulation of lactate and organic acids),
`and pancytopenia. Hyperammonemia is seen and is
`caused by accumulation of coenzyme A (CoA) deriva-
`tives, which inhibit carbamyl phosphate synthetase
`activity [6].
`
`Propionic acidemia
`
`The defect responsible for this disorder is in the enzyme
`propionyl-CoA carboxylase, which converts propionyl CoA
`to D-methylmalonyl CoA. Consequently, plasma ammoni-
`um, propionate, and glycine levels are elevated. The
`diagnosis is confirmed by measuring propionyl-CoA
`carboxylase activity in leukocytes or skin fibroblasts [6].
`
`Methylmalonic acidemia
`
`This disorder results from the inability to convert methyl-
`malonyl CoA to succinyl CoA. Patients develop metabolic
`ketoacidosis, hypoglycemia, hyperglycinemia, pancytope-
`nia, and hyperammonemia. There is also increased urinary
`excretion of methylmalonic acid [6].
`
`Isovaleric acidemia
`
`This entity results from a defect in leucine metabolism
`whereby isovaleryl CoA cannot be dehydrogenated to 3-
`methylcrotonyl CoA. This is characterized by a strong body
`odor resembling “sweaty feet.” Diagnostic features include
`elevated serum ammonia, isovaleric acid, and hypocarniti-
`nemia [6].
`
`Maple syrup urine disease
`
`Maple syrup urine disease is characterized by maple syrup
`odor noted in the urine and elevated serum branched-chain
`amino acid concentrations (leucine, isoleucine, and valine)
`during the first 24 h of life. Hyperammonemia contributes
`to neurologic deterioration. Glutamate levels are decreased,
`with consequent decrease in urea synthesis [15].
`
`Dibasic aminoacidurias
`
`recessive
`Lysinuric protein intolerance This autosomal
`condition is characterized by lysinuria and inadequate urea
`formation with development of hyperammonemia. The
`primary defect resides in renal tubular, intestinal, epithelial,
`and hepatic dibasic amino acid transport mechanisms. This
`results in a drop of available serum levels of ornithine,
`lysine, and arginine, leading eventually to hyperammone-
`mia. There are concomitant increases in urinary excretion
`of lysine, ornithine, arginine, and citrulline [49].
`
`Page 6 of 16
`
`

`

`Pediatr Nephrol
`
`Hyperammonemia hyperornithinemia homocitrullinuria
`
`recessive condition results from low
`This autosomal
`ornithine transport into abnormal mitochondria, resulting
`in decreased flow through OTC. Consequently, plasma
`ornithine levels rise with an associated increase in excretion
`of homocitrulline in the urine and hyperammonemia [6].
`
`Transient hyperammonemia of the newborn
`
`This disorder may be found in infants with a history of
`perinatal asphyxia, and symptoms can develop before the
`infant is 24 h of age. It presents in two forms: asymptom-
`atic and symptomatic. The former is self-limited and is
`thought to occur secondary to a transient deficiency of one
`of the urea cycle enzymes, a renal
`transport defect of
`arginine or ornithine, or a deficient synthesis of ornithine.
`All of these presumably are related to developmental
`immaturity.
`In the second form (unknown etiology),
`patients present with respiratory distress,
`lethargy, and
`coma. In both forms, plasma amino acids show elevated
`glutamine and alanine levels. The neurologic outcome in
`these patients can range from normal intellectual capacity to
`profound mental retardation and seizures [6].
`
`Drug related causes
`
`Salicylate Salicylate overdose can lead to a Reye-like
`clinical presentation,
`including emesis, anorexia, fever,
`hyperventilation, irritability, and hallucinations. In the early
`phase, there is respiratory alkalosis, with later development
`of metabolic acidosis associated with hypoglycemia, hyper-
`ammonemia, elevated serum salicylate levels, and hepato-
`toxicity. The pathogenesis involves mitochondrial
`dysfunction and uncoupling of oxidative phosphorylation
`[6].
`
`Carbamazepine Asterixis and hyperammonemia are un-
`common complications associated with carbamazepine use.
`The mechanism by which hyperammonemia develops is
`unknown, and treatment
`for
`the latter
`is simply to
`discontinue the anticonvulsant [50–52].
`
`Valproic acid Valproic acid use has been associated with
`the induction of hyperammonemic encephalopathy through
`astrocytic edema, renal tubular interference with glutamine
`synthesis, and/or
`increased renal glutaminase activity
`(resulting in enhanced ammoniagenesis). Valproic acid
`depends on carnitine for its metabolism and can deplete
`the latter. Hyperammonemia has been prevented by
`providing supplemental carnitine [53–55]. Once the
`valproic-acid-induced hyperammonemic encephalopathy
`has been identified, discontinuation of the drug results in
`
`recovery after a few days, with normalization of serum
`ammonia levels. In patients with inborn errors of metabo-
`lism who are having seizures, valproate should be avoided,
`as it can worsen the hyperammonemia [56].
`
`Topiramate Topiramate in combination with valproic acid
`has been associated with the development of hyperammo-
`nemia [57] The toxic effects of topiramate may relate to its
`ability to inhibit carbonic anhydrase, which in turn affects
`gamma-aminobutyric-acid-gated chloride ion conductance
`at the GABA A receptor and through its effect on the
`hepatic availability of bicarbonate (HCO3), which is also a
`substrate for carbamoylphosphate synthesis [53, 58]. In
`addition, topiramate inhibits glutamine synthetase activity
`in the brain, leading to toxic levels of ammonia [55].
`
`Tranexamic acid This is an antifibrinolytic agent that has
`been associated with the development of hyperammonemia
`seen as cause or result of convulsions caused by the
`medication [59].
`
`Chemotherapy Hyperammonemic coma has been described
`postinduction chemotherapy for acute myeloid leukemia. It
`has also been described in patients with multiple myeloma
`(as abnormal plasma-cell clones may produce ammonia) in
`those receiving chemotherapy with asparaginase, 5-
`fluorouracil
`(transient hyperammonemia), and in those
`receiving rituximab. The pathogenic mechanism is thought
`to be related to an acquired deficiency of glutamine
`synthetase of unknown cause. [60–65]
`
`Miscellaneous causes of hyperammonemia
`
`Hypoglycin (Jamaican vomiting sickness) This is caused by
`a toxin present in the unripe ackee fruit. The Jamaican
`vomiting sickness, which has a high mortality rate (up to
`50%), is characterized by vomiting, lethargy, hallucinations,
`coma, hypoglycemia, hyperammonemia, and abnormal
`liver enzymes, which occur when this unripe fruit
`is
`ingested [6].
`
`Pregnancy Hyperammonemic presentation occurs mostly
`during puerperium, but it has also been reported during
`pregnancy in patients with OTC and CPS deficiencies.
`During pregnancy,
`the healthy fetus can metabolize
`ammonia, whereas after delivery, this metabolic process is
`absent and ammonia accumulates in the mother. In addition,
`tissue breakdown occurs with postdelivery uterine involu-
`tion, adding more waste nitrogen products to the urea cycle
`[66–68].
`
`Distal renal tubular acidosis (RTA) Most renal production
`of ammonia occurs in the S1 and S2 segments of the
`
`Page 7 of 16
`
`

`

`proximal renal tubule. The renal ammonia production and
`tubular reabsorption are stimulated by chronic metabolic
`acidosis and potassium depletion. Distal renal
`tubular
`acidosis (RTA) has been reported in association with
`hyperammonemia and should be considered in the differ-
`ential diagnosis in infants with distal RTA who are
`hypokalemic [69–71].
`
`Carnitine transport defects Carnitine transport enzyme
`defects manifest with hypoglycemia, lethargy, and poten-
`tially a Reye-like syndrome (hepatomegaly, elevated trans-
`aminases, and ammonia). Carnitine plays a key role as
`cofactor in fatty-acid metabolism and its deficiency can
`lead to abnormal fatty-acid oxidation, leading to develop-
`ment of hyperammonemia and encephalopathy. In primary
`systemic carnitine deficiency, an autosomal
`recessive
`condition, the organic cation transporter gene OCTN2 is
`mutated, resulting in hypocarnitinemia secondary to re-
`duced renal tubular reabsorption of carnitine. Patients with
`this often present with hyperammonemia, encephalopathy,
`hypoglycemia, and myopathies [72].
`
`Urinary tract dilatation Proteus is a bacteria that charac-
`teristically produces an alkaline urine pH due to the
`hydrolysis of urea to ammonia by the bacterial urease.
`Children with massively dilated urinary tracts whose
`urinary tract is infected/colonized by this bacterium may
`develop hyperammonemia [73].
`
`Reye syndrome Reye syndrome is a disease of unknown
`etiology associated with hyperammonemia. Viruses (espe-
`cially influenza B and varicella),
`toxins/drugs (valproic
`acid, salicylate, aflatoxin, pesticides, and bacillus cereus),
`and a genetic component (increased risk of occurrence in
`siblings) have been implicated as causal factors. The key
`physiologic findings noted are a consequence of hepatic
`mitochondrial
`injury (swelling and death) caused by
`inhibition of the mitochondrial respiratory chain. Abnormal
`findings include acute encephalopathy, hyperammonemia,
`lactic acidosis, hypoglycemia, elevated liver enzymes,
`fatty-liver infiltration, and increased intracranial pressure
`[6, 74].
`
`Clinical symptoms of hyperammonemia
`
`Symptomatology varies with patient age and ammonium
`level and may include, among the most common findings:
`hypotonia, vomiting,
`lethargy, seizures, coma, ataxia,
`anorexia, abnormal behavior patterns, dysarthria, weakness,
`liver enlargement, and dementia. Most patients with UCD
`present during the neonatal period with nonspecific
`symptoms (poor feeding, vomiting, somnolence, irritability,
`tachypnea, and lethargy).
`
`Pediatr Nephrol
`
`Hepatic encephalopathy
`
`Acute and chronic hepatic encephalopathy (HE), in which
`hyperammonemia plays a pivotal role, can be seen as a
`complication of acute (i.e., drug toxicity,
`infections) and
`chronic liver disease. Swelling of astrocytes can lead to
`increased intracranial hypertension, cerebral edema, brain-
`stem herniation, and death. Chemical abnormalities include a
`generalized aminoacidemia (except for branched-chain amino
`acid levels, which are normal), hypoglycemia, hypovolemia,
`electrolyte disturbances, and hyperammonemia. Once the
`latter is reduced to normal levels, then clinical manifestations
`of hepatic encephalopathy reverse [22].
`
`Hyperammonemia treatment
`
`Toxin removal, enzyme induction, and anabolism are the
`main goals of emergency treatment. Normal patient growth
`and development should be the main goal of long-term
`treatment. Given the correlation between the duration of
`hyperammonemic coma and prospective neurocognitive
`function, it is imperative to institute treatment for hyper-
`ammonemia as soon as possible to prevent
`further
`neurological damage (even prior to a definitive diagnosis
`being made). The infant/child neurologic status should be
`examined carefully to assess response to treatment and,
`simultaneously,
`to ascertain the degree of neurological
`impairment that has occurred as sequelae from the primary
`disease. Overall, the treatment approach is similar, irre-
`spective of diagnosis. Dialysis should be immediately
`available and started if there is no response to conventional
`treatment (perhaps even at the same time).
`Treatment for acute hyperammonemia should be started
`with the goal of decreasing serum ammonia rapidly by
`reducing the production of nitrogenous waste (achieved by
`discontinuing protein intake, for no more than 48 h) and
`providing a hypercaloric glucose-based solution to enhance
`anabolism [75]. Supportive treatment and correction of
`hydration, nutritional status, mineral (calcium, potassium),
`and electrolyte imbalances should be addressed. In patients
`with UCD, bacterial sepsis can lead to a fatal outcome due
`to catabolism, thus, antibiotic coverage should be consid-
`ered even as prophylaxis, as patients often undergo multiple
`invasive procedures (such as line placement

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket