Back To Search Results

Adrenoleukodystrophy

Editor: Lina Saadeh Updated: 6/22/2024 3:30:09 PM

Introduction

Adrenoleukodystrophy (ALD) is a rare genetic condition characterized by the impaired metabolism of very long-chain fatty acids (VLCFAs), leading to their accumulation in various tissues, particularly the nervous system and adrenal glands. This accumulation arises from mutations in the ABCD1 gene, which encodes a peroxisomal membrane protein involved in VLCFA transport and degradation. Although earlier reports in the 1900s described the clinical presentations suggestive of this disease, the terminology and pathophysiology were not reported until the 1970s.[1] The brain, spinal cord, adrenal glands, and testes are the most commonly affected organs. Clinical manifestations of ALD vary widely but usually include the combination of progressive neurologic dysfunction and adrenal insufficiency.

ALD's inheritance pattern is often X-linked, designated as "X-ALD."[2] A unique neonatal form, classified as a form of Zellweger syndrome, has an autosomal recessive inheritance pattern designated as "N-ALD." An interprofessional team approach is highly recommended, given the condition's multiple organ involvement.

Etiology

Register For Free And Read The Full Article
Get the answers you need instantly with the StatPearls Clinical Decision Support tool. StatPearls spent the last decade developing the largest and most updated Point-of Care resource ever developed. Earn CME/CE by searching and reading articles.
  • Dropdown arrow Search engine and full access to all medical articles
  • Dropdown arrow 10 free questions in your specialty
  • Dropdown arrow Free CME/CE Activities
  • Dropdown arrow Free daily question in your email
  • Dropdown arrow Save favorite articles to your dashboard
  • Dropdown arrow Emails offering discounts

Learn more about a Subscription to StatPearls Point-of-Care

Etiology

X-ALD disease is linked to ABCD1 gene mutation. The ABCD1 gene plays a significant role in the VLCFA transport system in the peroxisomes, where VLCFAs can undergo further metabolism. The ABCD1 gene mutation product interferes with this process, resulting in abnormal accumulation of VLCFA in different body organs and interference with the affected organs' normal physiological function. A mutation in the PTS1 receptor, PXR1, PEX1, PEX10, or PEX13 genes causes the neonatal form.[3] Four main subtypes of ALD have been described based on organs affected and age at presentation: neonatal, childhood cerebral form, adrenomyeloneuropathy, and adrenal insufficiency.

Epidemiology

ALD is the most common genetic disease that affects the peroxisomes. The condition has an approximated prevalence of 1 in 20,000. The disease incidence is higher in patients of Latino or African descent.[4] N-ALD has a prevalence of 1 in 50,000.[5]

Histopathology

On gross examination, affected adrenal glands appear small and atrophied. Histological findings consist of nodular swelling affecting mainly the zona fasciculata and zona reticularis. Cellular vacuoles and clefts may also be visible. The medulla is usually spared and appears normal.[6]

Central nervous system (CNS) pathological findings in ALD consist of symmetrical demyelination of the white matter. These effects commonly occur in the corpus callosum and occipitoparietal region. Severe cases may involve the spinal cord. At the cellular level, swelling and vacuolization are caused by infiltrates of active inflammatory cells (macrophages and astrocytes). These changes usually result in the loss of the myelin sheaths, oligodendrocytes, and neuronal axons. Dystrophic mineralization is ultimately seen on histological examination.[7]

History and Physical

Clinical manifestations vary significantly according to disease severity and the age at presentation. The cases are broadly categorized as N-ALD and X-ALD.

N-ALD can manifest immediately after birth. However, some infants only have mild manifestations, potentially delaying the diagnosis in such cases. Typical N-ALD signs and symptoms include the following:

  • Seizures, hypotonia, and hearing dysfunction
  • Vision loss, cataracts, and optic nerve dysplasia
  • Jaundice and hepatomegaly
  • Failure to thrive and facial dysmorphism (hypertelorism and flat midface) [8]

X-ALD has 3 phenotypes, classified based on the age at presentation and the organs affected. These phenotypes are childhood cerebral ALD, Addison disease, and adrenomyeloneuropathy.

Childhood cerebral ALD typically presents between ages 3 and 10 years. The hallmark feature that characterizes this form is developmental regression. Progressive sensory and severe neurological deficits are generally followed by severe disability, coma, and death. A small percentage of adults may develop symptoms similar to childhood cerebral ALD. 

Adrenal gland dysfunction characterizes the Addison disease phenotype of X-ALD. The manifestations of this subtype result from reduced aldosterone and cortisol production, which include hyponatremia, fatigue, hypotension, dehydration, hypoglycemia, and generalized weakness. Hyperpigmentation of the skin may also occur.

Adrenomyeloneuropathy comprises a milder spectrum of X-ALD. The typical age at presentation is in the 3rd decade of life. Typical manifestations include walking difficulties, unbalanced gait, and bowel or bladder sphincter dysfunction.

Female X-ALD carriers may develop subtler symptoms, such as unbalanced gait, neuropathy, and mild paresis. The adrenal glands and cerebrum are rarely involved.[9]

Evaluation

Typical clinical presentation, characteristics, symptoms, signs, and suggestive family history are considered the starting point for ALD evaluation. Several states started using newborn screening tests to identify newborns with X-ALD. Adding X-ALD to newborn screening identifies newborns with the mutation so they can receive treatment before the symptoms appear.

Laboratory workup may reveal abnormal liver function and reduced response to adrenocorticotropic hormone administration. However, results more suggestive of ALD include increased plasma VLCFA concentration, decreased plasmalogen concentration in red blood cells, and increased pipecolic and phytanic acid concentration in both plasma and fibroblasts.[10]

Brain magnetic resonance imaging (MRI) is essential to the evaluation. The typical MRI findings are white matter demyelination pattern, microgyria, and germinolytic cysts in the caudothalamic groove. Further genetic testing to identify the specific mutation causing the disorder is recommended to confirm the diagnosis.[11]

Treatment / Management

ALD has no effective cure. Supportive care, by optimizing nutrition, occupational therapy, and respiratory support, can help alleviate some of the disorder's severe consequences but typically does not significantly impact survival or long-term outcomes. Corticosteroid and mineralocorticoid replacement therapy is recommended in patients with impaired adrenal gland function. Some studies report that allogeneic hematopoietic cell transplantation (HCT) may have beneficial effects. Favorable outcomes with HCT therapy occur in asymptomatic patients at the time of diagnosis or those with mild symptoms and CNS involvement.[12] 

Recent trials exploring docosahexaenoic acid use to induce peroxisome proliferation are inconclusive.[13][14] Given the multiple organs affected and the various needs of patients with ALD, an interprofessional team approach, involving endocrinologists, neurologists, geneticists, and psychologists at the minimum, is recommended.

Differential Diagnosis

The differential diagnosis of N-ALD is comprehensive and includes other genetic syndromes that present in the neonatal period with neurological signs and symptoms. These conditions include Angelman, Rett, and Prader-Willi syndromes, hypoxic-ischemic encephalopathy, metabolic disorders, and myotonic dystrophy. Meanwhile, X-ALD's differentials include conditions manifesting with demyelination, such as acute disseminated encephalomyelitis and multiple sclerosis.

Pertinent Studies and Ongoing Trials

Two ongoing clinical trials to evaluate potential therapeutic interventions for ALD are actively recruiting. The NCT03727555 clinical trial evaluates the use of lentiviral vector TYF-ABCD1 to correct the defective ABCD1 gene causing ALD symptoms. Another trial, NCT03852498, aims to assess the effect of autologous CD34+ HCT on ALD.

Prognosis

The prognosis for N-ALD and most forms of X-ALD is poor. Treatment is usually limited to symptomatic supportive management. Replacement therapy is effective for patients with Addison disease, while HCT may benefit asymptomatic patients identified through newborn screening or incidental imaging, as well as those with mild symptoms. Severe disabilities and a short life span are the expected outcomes for most patients with N-ALD and X-ALD.

Deterrence and Patient Education

Adrenoleukodystrophy is a peroxisome disease that affects neonates with N-ALD and children and adults with X-ALD, involving multiple organs, commonly the central nervous system and the adrenal glands. Patients and families must be educated about the following:

  • N-ALD is autosomal recessive.
  • X-ALD is X-linked recessive and thus has a male predominance.
  • The clinical presentation of ALD varies, with neurological dysfunction (with symptoms like hypotonia, weakness, developmental regression, and cognitive disabilities) and adrenal insufficiency (with hypotension, fatigue, and hypoglycemia) being the most common clinical manifestations.
  • Diagnosis of ALD requires a detailed history, physical examination, and diagnostic modalities that include VLCFA measurement, brain MRI, and specific genetic testing.
  • No effective treatment exists for most ALD cases, and management is usually supportive. Corticosteroid and mineralocorticoid replacement and HCT may benefit some patients.
  • The prognosis of ALD is unfavorable and carries a high rate of mortality and severe disabilities.
  • Clinical trials are ongoing to evaluate potential gene therapy for ALD.

Genetic testing is imperative for properly counseling female carriers and patients' families. Affected families should be educated to help them understand the risks of transmission, consider prenatal testing or preimplantation genetic diagnosis, and make informed decisions regarding reproductive choices.

Enhancing Healthcare Team Outcomes

ALD is a peroxisomal disorder with variable clinical presentations. The condition has many types based on age, organs involved, and mode of inheritance. An interprofessional team of neurologists, endocrinologists, geneticists, dieticians, and psychologists is recommended to provide comprehensive care to individuals with ALD. The collaborative approach should address both the medical and psychosocial aspects of living with ALD.

References


[1]

Engelen M, Kemp S, de Visser M, van Geel BM, Wanders RJ, Aubourg P, Poll-The BT. X-linked adrenoleukodystrophy (X-ALD): clinical presentation and guidelines for diagnosis, follow-up and management. Orphanet journal of rare diseases. 2012 Aug 13:7():51. doi: 10.1186/1750-1172-7-51. Epub 2012 Aug 13     [PubMed PMID: 22889154]


[2]

Engelen M, Kemp S, Poll-The BT. X-linked adrenoleukodystrophy: pathogenesis and treatment. Current neurology and neuroscience reports. 2014 Oct:14(10):486. doi: 10.1007/s11910-014-0486-0. Epub     [PubMed PMID: 25115486]


[3]

Braverman NE, Raymond GV, Rizzo WB, Moser AB, Wilkinson ME, Stone EM, Steinberg SJ, Wangler MF, Rush ET, Hacia JG, Bose M. Peroxisome biogenesis disorders in the Zellweger spectrum: An overview of current diagnosis, clinical manifestations, and treatment guidelines. Molecular genetics and metabolism. 2016 Mar:117(3):313-21. doi: 10.1016/j.ymgme.2015.12.009. Epub 2015 Dec 23     [PubMed PMID: 26750748]

Level 3 (low-level) evidence

[4]

Bonkowsky JL, Wilkes J, Bardsley T, Urbik VM, Stoddard G. Association of Diagnosis of Leukodystrophy With Race and Ethnicity Among Pediatric and Adolescent Patients. JAMA network open. 2018 Nov 2:1(7):e185031. doi: 10.1001/jamanetworkopen.2018.5031. Epub 2018 Nov 2     [PubMed PMID: 30646379]


[5]

Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Steinberg SJ, Raymond GV, Braverman NE, Moser AB. Zellweger Spectrum Disorder. GeneReviews(®). 1993:():     [PubMed PMID: 20301621]


[6]

Ulrich J, Herschkowitz N, Heitz P, Sigrist T, Baerlocher P. Adrenoleukodystrophy. Preliminary report of a connatal case. Light- and electron microscopical, immunohistochemical and biochemical findings. Acta neuropathologica. 1978 Aug 7:43(1-2):77-83     [PubMed PMID: 209659]

Level 3 (low-level) evidence

[7]

Powers JM, Liu Y, Moser AB, Moser HW. The inflammatory myelinopathy of adreno-leukodystrophy: cells, effector molecules, and pathogenetic implications. Journal of neuropathology and experimental neurology. 1992 Nov:51(6):630-43     [PubMed PMID: 1362438]

Level 2 (mid-level) evidence

[8]

Farrell DF. Neonatal adrenoleukodystrophy: a clinical, pathologic, and biochemical study. Pediatric neurology. 2012 Nov:47(5):330-6. doi: 10.1016/j.pediatrneurol.2012.07.006. Epub     [PubMed PMID: 23044013]

Level 3 (low-level) evidence

[9]

Engelen M, Barbier M, Dijkstra IM, Schür R, de Bie RM, Verhamme C, Dijkgraaf MG, Aubourg PA, Wanders RJ, van Geel BM, de Visser M, Poll-The BT, Kemp S. X-linked adrenoleukodystrophy in women: a cross-sectional cohort study. Brain : a journal of neurology. 2014 Mar:137(Pt 3):693-706. doi: 10.1093/brain/awt361. Epub 2014 Jan 29     [PubMed PMID: 24480483]

Level 2 (mid-level) evidence

[10]

Molzer B, Kainz-Korschinsky M, Sundt-Heller R, Bernheimer H. Phytanic acid and very long chain fatty acids in genetic peroxisomal disorders. Journal of clinical chemistry and clinical biochemistry. Zeitschrift fur klinische Chemie und klinische Biochemie. 1989 May:27(5):309-14     [PubMed PMID: 2474624]


[11]

Boehm CD, Cutting GR, Lachtermacher MB, Moser HW, Chong SS. Accurate DNA-based diagnostic and carrier testing for X-linked adrenoleukodystrophy. Molecular genetics and metabolism. 1999 Feb:66(2):128-36     [PubMed PMID: 10068516]


[12]

Eichler F, Duncan C, Musolino PL, Orchard PJ, De Oliveira S, Thrasher AJ, Armant M, Dansereau C, Lund TC, Miller WP, Raymond GV, Sankar R, Shah AJ, Sevin C, Gaspar HB, Gissen P, Amartino H, Bratkovic D, Smith NJC, Paker AM, Shamir E, O'Meara T, Davidson D, Aubourg P, Williams DA. Hematopoietic Stem-Cell Gene Therapy for Cerebral Adrenoleukodystrophy. The New England journal of medicine. 2017 Oct 26:377(17):1630-1638. doi: 10.1056/NEJMoa1700554. Epub 2017 Oct 4     [PubMed PMID: 28976817]


[13]

Martinez M. Restoring the DHA levels in the brains of Zellweger patients. Journal of molecular neuroscience : MN. 2001 Apr-Jun:16(2-3):309-16; discussion 317-21     [PubMed PMID: 11478386]


[14]

Wei H, Kemp S, McGuinness MC, Moser AB, Smith KD. Pharmacological induction of peroxisomes in peroxisome biogenesis disorders. Annals of neurology. 2000 Mar:47(3):286-96     [PubMed PMID: 10716247]