Skip to main content
Therapeutic Advances in Gastroenterology logoLink to Therapeutic Advances in Gastroenterology
. 2017 Apr 26;10(7):553–562. doi: 10.1177/1756283X17705775

The role of sebelipase alfa in the treatment of lysosomal acid lipase deficiency

Angelika L Erwin 1,
PMCID: PMC5484437  PMID: 28804516

Abstract

Lysosomal acid lipase deficiency (LALD) is a lysosomal storage disorder (LSD) characterized either by infantile onset with fulminant clinical course and very poor prognosis or childhood/adult-onset disease with an attenuated phenotype. The disorder is often misdiagnosed or remains undiagnosed in children and adults due to a rather unspecific clinical presentation with dyslipidemia and steatohepatitis. Until recently, no good treatment options were available for LALD. Despite supportive and symptomatic therapies, death occurred before 1 year of age in patients with infantile-onset disease and patients with childhood/adult-onset LALD suffered from significant complications, such as liver cirrhosis, requiring liver transplantation and early-onset cardiovascular disease.

With the recent approval of sebelipase alfa for clinical use in infantile- as well as childhood/adult-onset LALD, a new treatment era for this disorder has begun. Sebelipase alfa is a recombinant human lysosomal acid lipase (LAL), which is administered via the intravenous route. Clinical trials have shown significant improvement of disease parameters such as liver transaminases, hepatomegaly, and dyslipidemia in childhood/adult-onset LALD patients. Treatment of infants with the severe infantile-onset form of the disease has led to improved survival beyond the age of 1 year, and also showed improvement of hepatic and gastrointestinal symptoms, as well as growth. Overall, sebelipase alfa has a favorable safety profile and promises to be a good long-term treatment option for patients with LALD, with significant reduction of disease burden and increased life expectancy.

Keywords: cholesteryl ester storage disease, hyperlipidemia, lysosomal acid lipase deficiency, nonalcoholic fatty liver disease, sebelipase alfa, Wolman disease

Introduction

Lysosomal acid lipase deficiency (LALD) is a rare autosomal recessive lysosomal storage disorder (LSD) caused by absent or deficient activity of the enzyme lysosomal acid lipase (LAL). This enzyme plays an important role in the degradation of triglycerides and cholesteryl esters. Decreased LAL enzyme activity leads to pathological accumulation of lipids in macrophages of various tissues throughout the body.1 Age of onset and phenotypic spectrum are variable and range from an infantile-onset form (Wolman disease) with severe clinical course and death before 1 year of age to childhood/adult-onset disease with milder symptoms, historically also known as cholesteryl ester storage disease.2,3 Both disease entities are part of a continuum and are caused by allelic mutations in the LIPA gene, which encodes the LAL enzyme.4

While the diagnosis in infantile-onset LALD is usually established early on due to the more uniform and severe clinical course, accurate diagnosis is often missed or delayed in individuals with childhood/adult-onset disease given the less specific and milder clinical presentation.5

Historically, treatment options for LALD were limited and based on symptom control. Prognosis for patients with infantile-onset LALD was dismal and individuals with childhood/adult-onset LALD were at significant risk for disease complications such as liver cirrhosis requiring liver transplantation at a young age. Additionally, individuals with childhood/adult-onset LALD frequently present with significant dyslipidemia, predisposing them to accelerated atherosclerosis and increasing their risk for cardiovascular disease.59 With the recent approval of sebelipase alfa (KanumaTM, Alexion Pharmaceuticals, Cheshire, CT, USA), a recombinant human LAL, a new treatment option for patients with LALD has become available. Clinical trials of this enzyme replacement therapy (ERT) have shown promising results, with significant improvement of symptoms and increased life expectancy in affected individuals.

Clinical presentation

Infantile-onset lysosomal acid lipase deficiency (Wolman disease)

Symptoms of the infantile-onset form of LALD occur in the neonatal period, and newborns frequently present with projectile and persistent vomiting in the first week of life.3,10,11 Other common findings are abdominal distention, hepatosplenomegaly, diarrhea, and failure to thrive. Serum transaminases are typically elevated and progressive liver disease with increased bilirubin concentration, coagulopathy, and hypoalbuminemia develops within the first few months of life. Adrenal enlargement with calcification (in 60–70% of cases) visible on abdominal X-ray films is a pathognomonic finding in infantile-onset LALD.3,11 Prognosis is poor, with most infants dying before 1 year of age from malnutrition, liver failure, and adrenal insufficiency. Hematopoietic stem cell transplantation (HSCT) has been attempted, but resulted in early death in most cases.1214

Childhood/adult-onset lysosomal acid lipase deficiency

The childhood/adult-onset form of LALD was first described in 1963 and is likely underdiagnosed due to its often unspecific and variable clinical presentation.2 Age of onset ranges from early childhood to adulthood with the majority of affected individuals presenting between 3 and 12 years of age. Diagnostic investigation in children and adults with LALD is often initiated due to significant dyslipidemia at a young age, complaints of abdominal fullness caused by hepatomegaly, or incidentally detected elevated serum transaminases.5,6

A significant delay between occurrence of first symptoms and establishment of the correct diagnosis is common.5,15 Around 30% of children and adults with LALD have been described to develop gastrointestinal symptoms with nausea, vomiting, abdominal pain, and gallbladder dysfunction.16 However, severe malnutrition and wasting are not usually features of the childhood/adult-onset form of LALD.

The most common disease manifestations of childhood/adult-onset LALD are liver disease and dyslipidemia. Hepatomegaly has been described in 88–99% of cases and splenomegaly has been reported in around 74–79%.5,6 Serum transaminases are frequently elevated, although fluctuation with intermittent normalization of levels has been observed. Hepatic steatosis is common and affected individuals are at a significant risk of developing fibrosis and cirrhosis. Several individuals have been reported who required liver transplantation due to rapid progression to liver failure.5

Dyslipidemia with patterns consistent with type IIb hyperlipoproteinemia or mixed hyperlipidemia is commonly found in individuals with childhood/adult-onset LALD. Patients typically present with elevated low-density lipoprotein cholesterol (LDL-C) and low high-density lipoprotein cholesterol (HDL-C) levels. Total cholesterol and triglycerides can also be increased. While LDL-C levels often respond to treatment with statins or other lipid-lowering medications, some LALD patients may require higher medication doses or combination of different drugs to obtain a satisfactory response.1722 In addition, no beneficial effect of statin therapy on hepatic steatosis or fibrosis has been observed.1,6,23,24 Life expectancy may be decreased due to liver cirrhosis necessitating liver transplantation at a young age. While chronic hyperlipidemia predisposes affected individuals to early-onset atherosclerosis with increased risk for cardiovascular complications, liver disease tends to be the major cause for morbidity and mortality in most of the symptomatic patients with childhood/adult-onset LALD.5,6

Individuals with less severe forms of LALD might have only mild hepatomegaly, intermittent elevation of transaminases, and undetected dyslipidemia. In addition, affected patients with isolated dyslipidemia and absence of hepatomegaly have also been described.8,9 This variable presentation of LALD and significant phenotypic overlap with common disorders such as nonalcoholic fatty liver disease, nonalcoholic steatohepatitis and different forms of dyslipidemia explain why many affected individuals likely remain undiagnosed or are misdiagnosed for many years.

Pathophysiology and prevalence

LALD is an autosomal recessive disorder caused by homozygous or compound heterozygous mutations in the LIPA gene, which encodes the LAL enzyme. LAL plays an important role in the cellular lipid metabolism by degrading cholesteryl esters and triglycerides and thereby providing the cell with free cholesterol and free fatty acids.25 These intracellular lipids interact with sterol regulatory element-binding proteins (SREBPs) which are transcription factors that modulate expression of genes involved in the lipid metabolism.26 A deficiency of the LAL enzyme leads to reduced hydrolysis of cholesteryl esters and triglycerides, which subsequently accumulate in the lysosomes. This lysosomal trapping causes a lack of intracellular free cholesterol, which has several consequences for the lipid metabolism: reduced SREBP-mediated feedback inhibition of the enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase results in increased cholesterol production. Upregulation of LDL-receptor expression occurs, leading to increased cellular uptake of LDL-C. Furthermore, apolipoprotein B, as well as very-low-density lipoprotein cholesterol (VLDL-C) synthesis are markedly increased.27 This metabolic dysregulation ultimately results in elevated total cholesterol, triglycerides, and LDL-C levels.25,2830 Low HDL-C concentration is commonly observed in LALD patients and is thought to be the result of impaired upregulation of the ATP-binding cassette transporter A1 (ABCA1) gene in response to intracellular cholesterol. The membrane lipid transporter ABCA1 promotes phospholipid and cholesterol efflux to apolipoprotein A1, which is the rate-limiting step for HDL-C particle formation. In LALD, cholesteryl esters are trapped in the lysosomes and intracellular free cholesterol is reduced. This results in decreased ABCA1 expression and ultimately leads to insufficient availability of extracellular apolipoprotein A1 needed for HDL-C formation.3133

The accumulation of cholesteryl esters and triglycerides in hepatocytes leads to hepatomegaly and hepatic steatosis. Liver biopsy in patients with childhood/adult-onset LALD typically shows findings consistent with microvesicular steatosis. In addition, foamy macrophages and signs of fibrosis or, in advanced disease, cirrhosis, can be seen on light microscopy.34

LALD is a pan-ethnic disease, although prevalence among the European White population seems to be higher than in other ethnic groups due to a higher carrier frequency (~1 in 200 individuals) of a recurrent splice-junction mutation E8SJM (c. 894G > A) in exon 8.35,36 This particular mutation is associated with the childhood/adult-onset form of LALD and has never been observed in infantile-onset disease, which is likely explained by higher residual enzyme activity of this allele. Overall, more than 100 pathogenic LIPA mutations have been reported so far. The majority of mutations associated with the infantile-onset form of LALD have been shown to produce LAL enzyme with little or no residual enzymatic activity.1,4,6,3740

Diagnosis

The easiest way of establishing the diagnosis of LALD is by measuring the enzymatic activity of LAL in leukocytes. This can be done either on a peripheral blood sample or on a dried blood spot.41 Confirmation of the diagnosis can be pursued by molecular analysis of the LIPA gene for either homozygous or compound heterozygous pathogenic mutations. While a clear genotype–phenotype correlation cannot be established, individuals with infantile-onset LALD tend to show absent or severely diminished residual enzyme activity.4

Clinical management

Like many other LSDs, LALD is a multisystemic disorder and should therefore be managed by an interdisciplinary care team. Historically, very limited treatment options for LALD were available, especially for the infantile-onset form of the disease.

Dietary management and supportive care

In infantile-onset disease, nutritional support is crucial and parenteral nutrition is often necessary due to severe malabsorption. However, due to significant comorbidities, malnutrition often cannot be prevented despite early initiation of dietary modifications such as low-fat formula and supplementation with medium-chain triglycerides.42,43

Lipid-lowering medication

In patients with childhood/adult-onset LALD, correction of high LDL-C and low HDL-C levels is often attempted using HMG-CoA reductase inhibitors (statins). While dyslipidemia often improves, therapeutic response is frequently insufficient, requiring high doses or combinations of statins with other lipid-lowering medication such as ezetimibe or cholestyramine.1722,24,4446 In addition, statin therapy has not uniformly been shown to have a beneficial effect on hepatic steatosis or serum transaminase concentrations, which often remain elevated despite treatment.1,6,23,24 In fact, progression of liver disease under statin treatment has been speculated due to upregulation of LDL receptor expression in hepatocytes and subsequently, increased storage of cholesteryl esters.7

Liver transplantation

Liver transplantation has been reported in several individuals with childhood/adult-onset LALD, most of whom were pediatric patients.4749 Long-term follow-up information for these patients is limited: while several cases have been reported to be doing well between 10 months and 3 years after transplant, one patient had transplant rejection and developed congestive heart failure. Another individual was found to have significant systemic manifestations of LALD with severe lipid accumulation in the renal vascular system and end stage renal disease 7 years after liver transplantation.50,51

Hematopoietic stem cell transplantation

HSCT has been attempted in several individuals with infantile-onset LALD, and while this is a potentially curative approach, outcomes have generally been poor, with significant comorbidities and a mortality rate of >50%.14,52,53 So far, only three children with infantile-LALD have been described in whom HSCT was successfully performed early in life and who were doing reasonably well at 4 and 11 years after transplantation.13,54

Enzyme replacement therapy with sebelipase alfa

General concept

One treatment approach for LSDs is ERT, in which the missing or malfunctioning enzyme is replaced by recombinant enzyme administered usually via the intravenous route. The list of LSDs treatable with ERT is steadily growing as research advances and although the various disorders and therefore enzymatic products are very distinct, they share a common underlying principle. After administration, the recombinant enzyme needs to be taken up by the specific target cells and then directed to the lysosomes, where it can hydrolyze the disease-specific substrate. In order for this process to happen, the recombinant enzyme needs to contain a mannose-6-phosphate moiety that is recognized by mannose-6-phosphate receptors on the surface of the cells targeting the enzyme to the lysosomes.55

Sebelipase alfa

Sebelipase alfa is a recombinant human LAL that is produced in egg white from transgenic Gallus gallus domesticus hens. The attached mannose-6-phosphate residue facilitates its uptake into the lysosomes of hepatocytes and macrophages where it can subsequently hydrolyze cholesteryl esters and triglycerides. Preclinical studies were performed in a LAL-deficient rat model56 and showed 100% survival of treated versus untreated animals, as well as a significant improvement of hepatic lipid storage.57,58

Clinical trials in humans subsequently revealed good tolerability of sebelipase alfa and significant improvement of LALD-related symptoms, as well as improved survival in infantile-onset LALD.5963 The recombinant enzyme was approved for treatment of infantile- as well as childhood/adult-onset LALD by the European Medicines Agency (EMA) in September 2015 and by the US Food and Drug Administration (FDA) in December 2015. Treatment with sebelipase alfa can be initiated as soon as the diagnosis of LALD is established and the recombinant enzyme is administered via the intravenous route. The recommended dosing is 1 mg/kg weekly for infantile-onset LALD and 1 mg/kg every 2 weeks in individuals with childhood/adult-onset LALD.64,65

Sebelipase alfa: clinical trials

To assess safety profile as well as efficacy of treatment with sebelipase alfa, separate clinical trials were conducted or are still ongoing for infantile- and childhood/adult-onset LALD (Table 1).

Table 1.

Outline of the different clinical trials with sebelipase alfa conducted in individuals with infantile- and childhood/adult-onset lysosomal acid lipase deficiency.

Trial name Trial phase Trial description Patients enrolled (n)
Childhood/adult-onset LALD
LAL-CL01 I Open label, single arm 9
LAL-CL04 II Open label, single arm 8
LAL-CL02 (ARISE) III Randomized, double-blind, placebo-controlled period, followed by open-label phase 66
Infantile-onset LALD
LAL-CL03 II/III Open label, single arm 9

LALD, lysosomal acid lipase deficiency.

Childhood/adult-onset lysosomal acid lipase deficiency

The first use of sebelipase alfa in humans was investigated in an open-label phase I clinical trial (LAL-CL01) [ClinicalTrials.gov identifier: NCT01307098]. Nine adult individuals with clinical manifestations of LALD received weekly infusions of sebelipase alfa at different doses (0.35, 1, or 3 mg/kg) over 4 weeks. Outcome measures included safety assessment as well as pharmacodynamic studies. Results revealed a favorable safety profile as well as a statistically significant reduction of liver transaminases in all study subjects.59

Of the nine individuals in LAL-CL01, eight were enrolled in the extension (phase II) clinical trial LAL-CL04 [ClinicalTrials.gov identifier: NCT01488097], which was also an open-label trial. After a washout period of 9–28 weeks, the patients again received weekly sebelipase alfa infusions at the same doses of 0.35, 1, or 3 mg/kg for 4 weeks. After this initial period, they continued to be treated with either 1 or 3 mg/kg recombinant enzyme at biweekly infusion intervals for a total of 52 weeks. During the treatment-free interval between LAL-CL01 and LAL-CL04, patients’ transaminases had reached pretreatment levels. After re-initiation of sebelipase alfa treatment in LAL-CL04, liver enzymes decreased at a similar rate to what had been observed in the phase I trial, and remained stable over the prolonged treatment interval of 52 weeks even after infusion frequency was switched to biweekly. At that time, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) concentrations showed a mean change from baseline of –58% and –40%, respectively. In addition, lipid parameters were assessed and while total cholesterol and LDL-C concentrations had initially increased after 4 weeks of treatment, mean concentrations for both had changed by –39% and –60%, respectively when compared with baseline. HDL-C concentration had changed +29% from baseline. It is speculated that the temporary increase in total cholesterol and LDL-C shortly after ERT initiation was caused by mobilization of accumulated lipids from liver, spleen as well as other tissues. As in the phase I trial, none of the patients experienced clinically concerning adverse events and no antibodies against the infusion drug could be detected.61

The phase III clinical trial (LAL-CL02 or ARISE) [ClinicalTrials.gov identifier: NCT01757184] was a multicenter, randomized, double-blind, placebo-controlled study. A total of 66 patients with childhood/adult-onset LALD were randomized to placebo or sebelipase alfa infusion at 1 mg/kg every other week. The placebo-controlled part of the trial was 20 weeks, followed by an open-label period during which all patients received treatment. The primary endpoint was normalization of ALT concentration and secondary endpoints included further disease-related outcomes, safety, and side-effect profile.

After completion of the placebo-controlled part, 31% of patients treated with sebelipase alfa had reached the primary endpoint as compared with 7% in the placebo group (p = 0.03). Results for AST were similar. Consistent with the improvement in liver function, a significant reduction of hepatic fat content compared with baseline was observed (–32% in sebelipase alfa versus –4.2% in placebo group, p = 0.001). This was determined using multi-echo gradient-echo MRI. For assessment of severity of microvesicular steatosis, liver biopsies at baseline and at 20 weeks were obtained in 16 patients in the sebelipase alfa group and in 10 patients who received placebo. While a reduction in steatosis was more frequently observed in the treatment group than in the placebo group, this finding did not reach statistical significance.

A positive effect of sebelipase alfa treatment on the lipid profile was also reported. While LDL-C improved by –28.4% from baseline in the treatment group, a change of only –6.2% from baseline was seen in the placebo group (p < 0.001). Similar findings were present for HDL-C, which changed by +19.6% from baseline in patients treated with sebelipase alfa compared with –0.3% in the placebo group (p < 0.001).

During the open-label period, after switching from placebo to sebelipase alfa treatment, the pattern of changes in ALT, LDL-C, and HDL-C was very similar to what was observed in the initial sebelipase alfa group. In addition, a further reduction in LDL-C was achieved in the initial treatment group with continued sebelipase alfa infusions.

Overall, sebelipase alfa was well tolerated and only one atypical infusion-related event was reported. Out of the 35 patients in the sebelipase alfa group, five developed antibodies against the infusion drug, but titers were low and nonsustained. They were not found to have an effect on safety or efficacy of sebelipase alfa.60

Infantile-onset lysosomal acid lipase deficiency

A phase II/III clinical trial (LAL-CL03) [ClinicalTrials.gov identifier: NCT01371825] investigating the use of sebelipase alfa in patients with infantile-onset LALD is an open-label trial conducted in Europe, the USA and the Middle East. All patients receive treatment with sebelipase alfa. The primary endpoint is survival at age 12 months and results are compared with data from a historical cohort. Secondary endpoints include safety, growth and liver function, as well as hematological parameters.

By the time of the last report in July 2015, five of the initially nine enrolled patients were still on ERT with sebelipase alfa. Three children had died due to causes unrelated to the infusion drug; the reason for discontinuation of treatment in the fourth child was not reported. All patients started weekly sebelipase alfa infusions at 0.2 or 0.35 mg/kg and the dose was then escalated to 1 or 3 mg/kg per week. The primary endpoint was met in all five patients and their ages ranged between 2 years 5 months and 4 years 7 months at the time the results were published. An improvement was observed of ALT and AST (median percent changes of –45.59% and –39.36% from baseline, respectively) and hepatosplenomegaly as well as gastrointestinal symptoms had improved. In addition, the children’s median weight percentile had increased from 3.59% at baseline to 35.09%. Out of five patients, four had normal scores when evaluating their development using the Denver II screening test.

ERT was reported to be overall well tolerated, with only one patient experiencing a treatment-related serious adverse event consisting of fever, chills, pallor, and tachycardia. Antibodies against the infusion drug were detected in four out of seven patients tested and they were found to be neutralizing in two of them. However, neither the infusion-related reaction nor the development of antibodies led to discontinuation of ERT.62,63

Discussion

LALD is an LSD with variable age of onset and varying degrees of phenotypic severity. It is a rare disorder but owing to the unspecific clinical presentation of the childhood/adult-onset type, LALD is very likely underdiagnosed and its prevalence is expected to be higher than previously reported.35,36 Until the recent EMA and FDA approval of sebelipase alfa, treatment was mainly symptomatic and prognosis for the infantile-onset form of LALD was dismal, with death usually before age 1. Patients with childhood/adult-onset LALD may have decreased life expectancy due to significant liver disease, often requiring liver transplantation at a young age. In addition, accelerated atherosclerosis and an increased risk for early-onset cardiovascular disease are a concern in affected individuals due to unfavorable lipid profiles.5,6,8,9 While so far, no long-term outcome data for treatment with sebelipase alfa is available, clinical trials showed significant reduction of disease burden in both, infants and individuals with childhood/adult-onset disease. This led to significant improvement of survival in the infantile-onset group and is expected to also increase life span and quality of life in patients with childhood/adult-onset LALD. However, longer-term follow up is needed to establish the full extent to which children with infantile-onset LALD benefit from treatment and whether complications such as liver cirrhosis and cardiovascular events can truly be prevented over time in patients with childhood/adult-onset disease.

It should also be noted that individuals with advanced liver disease were not included in the clinical trials of childhood/adult-onset LALD. It is therefore undetermined and remains to be evaluated whether treatment with sebelipase alfa can halt or reverse symptoms of advanced liver disease.

While animal studies did not show a harmful effect on embryogenesis or fetal development, no experience during pregnancy in humans is available so far and no recommendations regarding sebelipase alfa treatment and dosing can therefore be provided at this time.65

In conclusion, sebelipase alfa has been shown as an efficient and safe treatment for infantile- as well as childhood/adult-onset LALD but long-term outcomes remain to be determined. Infantile-onset LALD is usually more easily diagnosable due to the severity of the clinical presentation, and treatment with sebelipase alfa should be initiated as soon as the diagnosis is established by enzymatic screening. Childhood/adult-onset LALD, however, often presents with unspecific clinical features and is therefore frequently undiagnosed or misdiagnosed. With the availability of sebelipase alfa, establishing the correct diagnosis as early as possible is crucial, and broad enzymatic screening for LALD in children and adults with hepatic disease and/or dyslipidemia is recommended. However, given the rarity of this disorder, disease awareness among healthcare providers is low and LALD is often not part of the differential diagnosis. Education, as well as access to enzymatic screening will therefore prove essential to ensure that patients with childhood/adult-onset LALD are diagnosed and started on treatment in a timely fashion.

Footnotes

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of interest statement: Dr Erwin has served as an advisory board member for Alexion Pharmaceuticals.

References

  • 1. Grabowski GA, Charnas L, Du H. Lysosomal acid lipase deficiencies: the Wolman disease/cholesteryl ester storage disease spectrum. In: Valle SD, Beaudet AL, Vogelstein B, et al. (eds) The online metabolic and molecular bases of inherited disease. 8th ed. New York: McGraw-Hill, 2012. [Google Scholar]
  • 2. Fredrickson DS. Newly recognized disorders of cholesterol metabolism. Ann Intern Med 1963; 58: 718. [Google Scholar]
  • 3. Abramov A, Schorr S, Wolman M. Generalized xanthomatosis with calcified adrenals. AMA J Dis Child 1956; 91: 282–286. [DOI] [PubMed] [Google Scholar]
  • 4. Aslanidis C, Ries S, Fehringer P, et al. Genetic and biochemical evidence that CESD and Wolman disease are distinguished by residual lysosomal acid lipase activity. Genomics 1996; 33: 85–93. [DOI] [PubMed] [Google Scholar]
  • 5. Burton BK, Deegan PB, Enns GM, et al. Clinical features of lysosomal acid lipase deficiency. J Pediatr Gastroenterol Nutr 2015; 61: 619–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Bernstein DL, Hulkova H, Bialer MG, et al. Cholesteryl ester storage disease: review of the findings in 135 reported patients with an underdiagnosed disease. J Hepatol 2013; 58: 1230–1243. [DOI] [PubMed] [Google Scholar]
  • 7. Reiner Z, Guardamagna O, Nair D, et al. Lysosomal acid lipase deficiency–an under-recognized cause of dyslipidaemia and liver dysfunction. Atherosclerosis 2014; 235: 21–30. [DOI] [PubMed] [Google Scholar]
  • 8. Elleder M, Chlumska A, Hyanek J, et al. Subclinical course of cholesteryl ester storage disease in an adult with hypercholesterolemia, accelerated atherosclerosis, and liver cancer. J Hepatol 2000; 32: 528–34. [DOI] [PubMed] [Google Scholar]
  • 9. Stitziel NO, Fouchier SW, Sjouke B, et al. Exome sequencing and directed clinical phenotyping diagnose cholesterol ester storage disease presenting as autosomal recessive hypercholesterolemia. Arterioscler Thromb Vasc Biol 2013; 33: 2909–2914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Crocker AC, Vawter GF, Neuhauser EB, et al. Wolman’s disease: three new patients with a recently described lipidosis. Pediatrics 1965; 35: 627–640. [PubMed] [Google Scholar]
  • 11. Marshall WC, Ockenden BG, Fosbrooke AS, et al. Wolman’s disease. A rare lipidosis with adrenal calcification. Arch Dis Child 1969; 44: 331–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Krivit W, Shapiro E, Hoogerbrugge PM, et al. State of the art review. Bone marrow transplantation treatment for storage diseases. Keystone. January 23, 1992. Bone Marrow Transplant 1992; 10(Suppl. 1): 87–96. [PubMed] [Google Scholar]
  • 13. Tolar J, Petryk A, Khan K, et al. Long-term metabolic, endocrine, and neuropsychological outcome of hematopoietic cell transplantation for Wolman disease. Bone Marrow Transplant 2009; 43: 21–27. [DOI] [PubMed] [Google Scholar]
  • 14. Yanir A, Allatif MA, Weintraub M, et al. Unfavorable outcome of hematopoietic stem cell transplantation in two siblings with Wolman disease due to graft failure and hepatic complications. Mol Genet Metab 2013; 109: 224–226. [DOI] [PubMed] [Google Scholar]
  • 15. Zarrilli F, Elce A, Scorza M, et al. An update on laboratory diagnosis of liver inherited diseases. Biomed Res Int 2013; 2013: 697940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Zhang B, Porto AF. Cholesteryl ester storage disease: protean presentations of lysosomal acid lipase deficiency. J Pediatr Gastroenterol Nutr 2013; 56: 682–685. [DOI] [PubMed] [Google Scholar]
  • 17. Fouchier SW, Defesche JC. Lysosomal acid lipase A and the hypercholesterolaemic phenotype. Curr Opin Lipidol 2013; 24: 332–338. [DOI] [PubMed] [Google Scholar]
  • 18. Gasche C, Aslanidis C, Kain R, et al. A novel variant of lysosomal acid lipase in cholesteryl ester storage disease associated with mild phenotype and improvement on lovastatin. J Hepatol 1997; 27: 744–750. [DOI] [PubMed] [Google Scholar]
  • 19. McCoy E, Yokoyama S. Treatment of cholesteryl ester storage disease with combined cholestyramine and lovastatin. Ann N Y Acad Sci 1991; 623: 453–454. [DOI] [PubMed] [Google Scholar]
  • 20. Tadiboyina VT, Liu DM, Miskie BA, et al. Treatment of dyslipidemia with lovastatin and ezetimibe in an adolescent with cholesterol ester storage disease. Lipids Health Dis 2005; 4: 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Tarantino MD, McNamara DJ, Granstrom P, et al. Lovastatin therapy for cholesterol ester storage disease in two sisters. J Pediatr 1991; 118: 131–135. [DOI] [PubMed] [Google Scholar]
  • 22. Yokoyama S, McCoy E. Long-term treatment of a homozygous cholesteryl ester storage disease with combined cholestyramine and lovastatin. J Inherit Metab Dis 1992; 15: 291–292. [DOI] [PubMed] [Google Scholar]
  • 23. Pullinger CR, Stock EO, Movsesyan I, et al. Identification and metabolic profiling of patients with lysosomal acid lipase deficiency. J Clin Lipidol 2015; 9: 716–726. [DOI] [PubMed] [Google Scholar]
  • 24. Leone L, Ippoliti PF, Antonicelli R. Use of simvastatin plus cholestyramine in the treatment of lysosomal acid lipase deficiency. J Pediatr 1991; 119: 1008–1009. [DOI] [PubMed] [Google Scholar]
  • 25. Goldstein JL, Dana SE, Faust JR, et al. Role of lysosomal acid lipase in the metabolism of plasma low density lipoprotein. Observations in cultured fibroblasts from a patient with cholesteryl ester storage disease. J Biol Chem 1975; 250: 8487–8495. [PubMed] [Google Scholar]
  • 26. Jeon TI, Osborne TF. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol Metab 2012; 23: 65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Cummings MH, Watts GF. Increased hepatic secretion of very-low-density lipoprotein apolipoprotein B-100 in cholesteryl ester storage disease. Clin Chem 1995; 41: 111–114. [PubMed] [Google Scholar]
  • 28. Brown MS, Dana SE, Goldstein JL. Receptor-dependent hydrolysis of cholesteryl esters contained in plasma low density lipoprotein. Proc Natl Acad Sci U S A 1975; 72: 2925–2929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Todoroki T, Matsumoto K, Watanabe K, et al. Accumulated lipids, aberrant fatty acid composition and defective cholesterol ester hydrolase activity in cholesterol ester storage disease. Ann Clin Biochem 2000; 37: 187–193. [DOI] [PubMed] [Google Scholar]
  • 30. Kostner GM, Hadorn B, Roscher A, et al. Plasma lipids and lipoproteins of a patient with cholesteryl ester storage disease. J Inherit Metab Dis 1985; 8: 9–12. [DOI] [PubMed] [Google Scholar]
  • 31. Boadu E, Bilbey NJ, Francis GA. Cellular cholesterol substrate pools for adenosine-triphosphate cassette transporter A1-dependent high-density lipoprotein formation. Curr Opin Lipidol 2008; 19: 270–276. [DOI] [PubMed] [Google Scholar]
  • 32. Bowden KL, Bilbey NJ, Bilawchuk LM, et al. Lysosomal acid lipase deficiency impairs regulation of ABCA1 gene and formation of high density lipoproteins in cholesteryl ester storage disease. J Biol Chem 2011; 286: 30624–30635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Oram JF, Heinecke JW. ATP-binding cassette transporter A1: a cell cholesterol exporter that protects against cardiovascular disease. Physiol Rev 2005; 85: 1343–1372. [DOI] [PubMed] [Google Scholar]
  • 34. Hulkova H, Elleder M. Distinctive histopathological features that support a diagnosis of cholesterol ester storage disease in liver biopsy specimens. Histopathology 2012; 60: 1107–1113. [DOI] [PubMed] [Google Scholar]
  • 35. Muntoni S, Wiebusch H, Jansen-Rust M, et al. Prevalence of cholesteryl ester storage disease. Arterioscler Thromb Vasc Biol 2007; 27: 1866–1868. [DOI] [PubMed] [Google Scholar]
  • 36. Scott SA, Liu B, Nazarenko I, et al. Frequency of the cholesteryl ester storage disease common LIPA E8SJM mutation (c.894G>A) in various racial and ethnic groups. Hepatology 2013; 58: 958–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Anderson RA, Byrum RS, Coates PM, et al. Mutations at the lysosomal acid cholesteryl ester hydrolase gene locus in Wolman disease. Proc Natl Acad Sci U S A 1994; 91: 2718–2722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ruiz-Andres C, Selles E, Arias A, et al. Lysosomal acid lipase deficiency in 23 Spanish patients: high frequency of the novel c.966+2T>G mutation in Wolman disease. JIMD Rep. Epub ahead of print 21 February 2017. DOI: 10.1007/8904_2017_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Fasano T, Pisciotta L, Bocchi L, et al. Lysosomal lipase deficiency: molecular characterization of eleven patients with Wolman or cholesteryl ester storage disease. Mol Genet Metab 2012; 105: 450–456. [DOI] [PubMed] [Google Scholar]
  • 40. Sheriff S, Du H, Grabowski GA. Characterization of lysosomal acid lipase by site-directed mutagenesis and heterologous expression. J Biol Chem 1995; 270: 27766–27772. [DOI] [PubMed] [Google Scholar]
  • 41. Dairaku T, Iwamoto T, Nishimura M, et al. A practical fluorometric assay method to measure lysosomal acid lipase activity in dried blood spots for the screening of cholesteryl ester storage disease and Wolman disease. Mol Genet Metab 2014; 111: 193–196. [DOI] [PubMed] [Google Scholar]
  • 42. Hoffman EP, Barr ML, Giovanni MA, et al. Lysosomal acid lipase deficiency. In:Pagon RA, Adam MP, Ardinger HH, et al. (eds) GeneReviews(R). Seattle, WA: University of Washington, Seattle, 1993. [Google Scholar]
  • 43. Alexander MS, Scott KR, Harkless J, et al. Enaminones 11. An examination of some ethyl ester enaminone derivatives as anticonvulsant agents. Bioorg Med Chem 2013; 21: 3272–3279. [DOI] [PubMed] [Google Scholar]
  • 44. Iverson SA, Cairns SR, Ward CP, et al. Asymptomatic cholesteryl ester storage disease in an adult controlled with simvastatin. Ann Clin Biochem 1997; 34: 433–436. [DOI] [PubMed] [Google Scholar]
  • 45. Levy R, Ostlund RE, Jr., Schonfeld G, et al. Cholesteryl ester storage disease: complex molecular effects of chronic lovastatin therapy. J Lipid Res 1992; 33: 1005–1015. [PubMed] [Google Scholar]
  • 46. Rassoul F, Richter V, Lohse P, et al. Long-term administration of the HMG-CoA reductase inhibitor lovastatin in two patients with cholesteryl ester storage disease. Int J Clin Pharmacol Ther 2001; 39: 199–204. [DOI] [PubMed] [Google Scholar]
  • 47. Ferry GD, Whisennand HH, Finegold MJ, et al. Liver transplantation for cholesteryl ester storage disease. J Pediatr Gastroenterol Nutr 1991; 12: 376–378. [DOI] [PubMed] [Google Scholar]
  • 48. Arterburn JN, Lee WM, Wood RP, et al. Orthotopic liver transplantation for cholesteryl ester storage disease. J Clin Gastroenterol 1991; 13: 482–485. [DOI] [PubMed] [Google Scholar]
  • 49. Leone L, Ippoliti PF, Antonicelli R, et al. Treatment and liver transplantation for cholesterol ester storage disease. J Pediatr 1995; 127: 509–510. [DOI] [PubMed] [Google Scholar]
  • 50. Martinez Ibanez V, Iglesias J, Lloret J, et al. [7 years’ experience with hepatic transplantation in children]. Cir Pediatr 1993; 6: 7–10. [PubMed] [Google Scholar]
  • 51. Kale AS, Ferry GD, Hawkins EP. End-stage renal disease in a patient with cholesteryl ester storage disease following successful liver transplantation and cyclosporine immunosuppression. J Pediatr Gastroenterol Nutr 1995; 20: 95–97. [DOI] [PubMed] [Google Scholar]
  • 52. Gramatges MM, Dvorak CC, Regula DP, et al. Pathological evidence of Wolman’s disease following hematopoietic stem cell transplantation despite correction of lysosomal acid lipase activity. Bone Marrow Transplant 2009; 44: 449–450. [DOI] [PubMed] [Google Scholar]
  • 53. Krivit W, Freese D, Chan KW, et al. Wolman’s disease: a review of treatment with bone marrow transplantation and considerations for the future. Bone Marrow Transplant 1992; 10(Suppl. 1): 97–101. [PubMed] [Google Scholar]
  • 54. Stein J, Garty BZ, Dror Y, et al. Successful treatment of Wolman disease by unrelated umbilical cord blood transplantation. Eur J Pediatr 2007; 166: 663–666. [DOI] [PubMed] [Google Scholar]
  • 55. Desnick RJ, Schuchman EH. Enzyme replacement therapy for lysosomal diseases: lessons from 20 years of experience and remaining challenges. Annu Rev Genomics Hum Genet 2012; 13: 307–335. [DOI] [PubMed] [Google Scholar]
  • 56. Yoshida H, Kuriyama M. Genetic lipid storage disease with lysosomal acid lipase deficiency in rats. Lab Anim Sci 1990; 40: 486–489. [PubMed] [Google Scholar]
  • 57. Quinn AG, Leavitt M, Hu W, et al. SBC-102, a recombinant enzyme replacement therapy corrects clinically relevant phenotypic abnormalities in a rat model of lysosomal acid lipase deficiency. Metabol Mol Genet 2011; 102: S37. [Google Scholar]
  • 58. Leavitt M, Burt AD, Hu W, et al. Recombinant lysosomal acid lipase normalizes liver weight, transaminases and histopathological abnormalities in an in vivo model of cholesteryl ester storage disease. J Hepatol 2011; 54(Suppl. 1): S358. [Google Scholar]
  • 59. Balwani M, Breen C, Enns GM, et al. Clinical effect and safety profile of recombinant human lysosomal acid lipase in patients with cholesteryl ester storage disease. Hepatology 2013; 58: 950–957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Burton BK, Balwani M, Feillet F, et al. A phase 3 trial of sebelipase alfa in lysosomal acid lipase deficiency. N Engl J Med 2015; 373: 1010–1020. [DOI] [PubMed] [Google Scholar]
  • 61. Valayannopoulos V, Malinova V, Honzik T, et al. Sebelipase alfa over 52 weeks reduces serum transaminases, liver volume and improves serum lipids in patients with lysosomal acid lipase deficiency. J Hepatol 2014; 61: 1135–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Jones SA, Plantaz D, Vara R, et al. Effect of sebelipase alfa on survival and liver function in infants with rapidly progressive lysosomal acid lipase deficiency. Mol Genet Metab 2015; 114: S59. [Google Scholar]
  • 63. Jones SA, Brassier A, Hughes J, et al. Effect of sebelipase alfa on survival and liver function in infants with rapidly progressive lysosomal acid lipase deficiency: 2-year follow-up data. Mol Genet Metab 2016; 117: S63. [Google Scholar]
  • 64. Shirley M. Sebelipase alfa: first global approval. Drugs 2015; 75: 1935–1940. [DOI] [PubMed] [Google Scholar]
  • 65. European Medicines Agency. Kanuma product information. London, UK: European Medicines Agency, 2015. [Google Scholar]

Articles from Therapeutic Advances in Gastroenterology are provided here courtesy of SAGE Publications

RESOURCES