• Family Conference 2023
  • DecNewsletter
  • First Legacy Guardian Peggy Davis


OUR MISSION

In our drive to find a cure for NBIA,
we provide support to families, educate the public and accelerate research with collaborators from around the world.


 

News

NBIAcure prepares to submit for FDA
approval of CoA-Z compound

 

Educational Resources

​Posters, flyers and
other educational handouts

 

MPAN

Mitochondrial-membrane Protein-Associated Neurodegeneration is caused by mutations in the C19orf12 gene. This gene is found on chromosome 19 and is believed to play a role in fatty acid metabolism. Though ultra-rare, it is one of the more common forms of NBIA and has distinctive clinical symptoms that differentiate it from other forms of NBIA. The prevalence of MPAN is roughly estimated at less than one in 1 million.

Onset typically occurs in childhood (ages 3 to 16, considered juvenile onset) to early adulthood (ages 17 to 24, considered adult onset). However, some individuals are reported to have a much later onset, resulting in a slower progression and survival well into adulthood. There are a few instances of later onset with rapid progression of disease. It causes cognitive decline progressing to severe dementia.

MPAN is characterized by spasticity that is more prominent than dystonia. There is weakness in muscles caused by motor axonal neuropathy, optic atrophy, and neuropsychiatric (mental disorder due to disease of the nervous system) changes.

Most affected individuals are still able to walk as they reach adulthood. Psychiatric signs are common, including impulsive or compulsive behavior, depression and frequent mood changes. Unlike most other forms of NBIA, the vast majority of individuals with MPAN develop progressive cognitive decline.

Click on any of the thumbnail images below to view the photos in a larger size;
move cursor to upper right of picture to click on next and view as slideshow

Clinical Diagnosis

The following clinical findings are suggestive of a diagnosis of MPAN:

  • MRI of the brain (T2 –weighted) that shows iron accumulation in the globus pallidus and substantia nigra
  • Impaired gait, or an inability to walk normally
  • Cognitive decline progressing to severe dementia
  • Prominent neuropsychiatric abnormalities, exaggerated emotional responses, depression, anxiety, impulsivity, compulsions, hallucinations, perseveration, inattention and hyperactivity
  • Optic atrophy, a condition in which the optic nerve is damaged, causing vision to be affected
  • Dystonia, a movement disorder in which a person’s muscles, often in the hands and feet, contract uncontrollably
  • Upper motor neuron signs (spasticity, hyperreflexia [overactive reflexes], Babinski sign [when the big toe bends up and back and the other toes fan out]). The lower limbs are usually affected earlier and more significantly than the upper limbs.
  • Lower motor neuron signs, such as muscle weakness and atrophy, hyporeflexia (muscles becoming less responsive to stimuli), and fasciculations (muscle twitches)
  • Parkinsonism, a condition that causes movement abnormalities such as tremor, slow movement, impaired speech or muscle stiffness
  • Dysarthria, a motor speech disorder that causes slow or slurred speech. This typically progresses to anarthria during end-stage disease, in which a person is unable to articulate speech.

Commonly, all age groups experience a decline in cognitive ability that progresses to dementia; prominent neuropsychiatric abnormalities; and movement problems caused by nerve-cell abnormalities. Studies reveal a loss of nerve cells, widespread iron deposits and abnormal axons (a part of nerve cells) called spheroid bodies, in the basal ganglia. Lewy neurites, which are abnormal clusters of protein that develop inside the nerve cells, are present in the globus pallidus, and Lewy bodies and neurites are widespread in other areas of the midbrain and in regions of the globus pallidus called the corpus striatum.

Management

Neuropsychiatric symptoms may require treatment by a psychiatrist.

Medical providers also may seek to manage the disease by performing regular eye exams, as well as neurological tests for dystonia, spasticity and parkinsonism. Children may benefit from a pediatric neurology assessment to establish care and continuity. Adults may benefit from a referral to a movement disorders neurologist or a neuromuscular clinic. This may include evaluation of the affected individual’s ability to walk and speak for possible physical, occupational and/or speech therapy.

For treatment of dystonia/spasticity, consider oral baclofen, trihexyphenidyl, intramuscular botulinum toxin or taking part in a trial of intrathecal baclofen under a physician’s guidance. For parkinsonism, consider pharmacologic treatment under physician guidance. For severe dysphagia (swallowing difficulties), consider nutritional and vitamin supplementation to meet dietary needs or a gastric feeding tube to minimize weight loss and decrease risk of aspiration.

Genetics

MPAN is inherited in an autosomal recessive, or less commonly, in an autosomal dominant manner. The phenotypes, or disease characteristics, are indistinguishable between the two types. Both cause a loss of the function of the C19orf12 protein.

Because most of our genes exist in pairs (one from the mother and one from the father), we normally carry two working copies of each gene. When one copy of a recessive gene has a change (mutation) in it, the person should still have normal health. That person is called a carrier.

Recessive diseases only occur when both parents are carriers for the same condition and then pass their changed genes onto their child, resulting in two disease-causing mutations.

For autosomal recessive inheritance, there is statistically a 25% chance that two carriers would have an affected child. The chances are 50% that the parents will have a child who is an asymptomatic carrier and 25% that the child will not receive the gene mutation. Carrier testing for at-risk relatives and prenatal testing for pregnancies can be done if both disease-causing mutations have been identified in an affected family member.

When a disease is inherited in an autosomal dominant manner, only one copy of the mutated gene coming from either parent is needed to cause the disease. Each child of an MPAN-affected individual has a 50% chance of inheriting the mutation that causes the disorder.

Prenatal Testing

If the disease-causing mutations have been identified in the family, prenatal diagnosis for pregnancies at increased risk can be done. In one test, DNA is extracted from fetal cells obtained by amniocentesis, usually at 15 to 18 weeks’ gestation, and analyzed. Or, sampling is done of the chorionic villus, the tiny finger-like projections on the edge of the placenta, usually at 10 to 12 weeks’ gestation.

Embryo screening, known as preimplantation genetic diagnosis, may be an option for some families in which the disease-causing mutations have been identified.

Note


A main resource for the clinical information provided here is Mitochondrial Membrane Protein-Associated Neurodegeneration - GeneReviews® - NCBI Bookshelf. GeneReviews is primarily for the use of professionals so the terminology and information may be difficult for the general public to understand.


Natural History Studies

The TIRCON International NBIA Registry is housed at Ludwig Maximilian University of Munich, Germany, and was created under an EU grant from 2011 to 2015 called Treat Iron-Related Childhood Onset Neurodegeneration (TIRCON ).

The NBIA Alliance and other sources have provided registry funding since 2015. Clinical centers in 16 countries are participants and provide patient clinical data. There are over 700 entries consisting of NBIA patients and controls as of September, 2021, including MPAN individuals. Clinical centers seeing at least five NBIA patients are eligible to participate. Clinical and natural history data is available to researchers studying NBIA disorders. Contact Anna Baur-Ulatowska at Anna.Baur@med.uni-muenchen.de for more information on this registry.

Therapies Under Investigation

Iron Chelation

Iron chelator drugs are being explored as a potential treatment for MPAN and other related NBIA disorders. One such drug, deferiprone, was used to treat two MPAN patients, including a 13-year-old. A two-year treatment in that person led to reduced iron content in the substantia nigra, while pallidal iron depositions and clinical status remained unchanged [Löbel et al 2014].

A study involving a related NBIA disorder, PKAN, explored whether deferiprone can reduce brain iron and slow disease progression. This study was done over the course of 18 months and was placebo-controlled. The treatment groups were randomized and the study was completed in a double-blind manner in which neither participants nor the researchers knew which patients were in which group. The results indicated that deferiprone did not lead to statistically significant clinical changes in PKAN individuals, although researchers reported a trend toward slower progression on the dystonia scale used for later onset individuals [Klopstock et al 2019].

MPAN Clinical Trials

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for information on clinical studies.

MPAN Research Publications and Articles

Following is a list of recent MPAN research articles.

Other MPAN research articles and studies can be found at Pub Med Central.

2024 - Nazo, the Drosophila homolog of the NBIA-mutated protein–c19orf12, is required for triglyceride homeostasis

2023 - Identification of Autophagy as a Functional Target Suitable for Pharmacological Treatment of Mitochondrial Membrane Protein-Associated Neurodegeneration (MPAN) In Vitro

2020 - The Downregulation of c19orf12 Negatively Affects Neuronal and Musculature Development in Zebrafish Embryos

2020 - Is there heart disease in cases of neurodegeneration associated with mutations in C19orf12?

2020 - Brain iron and metabolic abnormalities in C19orf12 mutation carriers: A 7.0 tesla MRI study in mitochondrial membrane protein-associated neurodegeneration

2019 - Clinical and genetic spectrum of an orphan disease MPAN: a series with new variants and a novel phenotype

2019 - Autosomal dominant mitochondrial membrane protein-associated neurodegeneration (MPAN)

2018 - Mitochondrial membrane protein-associated neurodegeneration: a case report and literature review

2017 - Evolution and novel radiological changes of neurodegeneration associated with mutations in C19orf12

2013 - New NBIA subtype: Genetic, clinical, pathologic, and radiographic features of MPAN

PLAN

PLA2G6-Associated Neurodegeneration, is named for the responsible gene: PLA2G6. This gene is thought to be important in helping cells maintain a healthy membrane (outer layer). It also is involved in lipid (fat) metabolism. It is not yet known how changes in this gene cause the symptoms of PLAN or the excess accumulation of iron in the brain in some affected individuals.

PLAN is made up of three distinct forms with differing characteristics:

  • INAD, or Infantile Neuroaxonal Dystrophy: early onset, rapidly progressive disease
  • Atypical NAD, or atypical neuroaxonal dystrophy: later childhood onset with slower progression and predominant extrapyramidal (nerves that regulate motor control) findings, such as dystonia (involuntary muscle contractions that cause repetitive or twisting movements) and dysarthria (difficulty pronouncing words). It includes a broad range of presentations
  • PLA2G6-related dystonia-parkinsonism: adult-onset dystonia-parkinsonism accompanied by cognitive decline and neuropsychiatric changes (mental disorder due to disease of the nervous system)

Click on any of the thumbnail images below to view the photos in a larger size;
move cursor to upper right of picture to click on next and view as slideshow

Infantile Neuroaxonal Dystrophy (INAD)

Classic INAD starts early in life and progresses rapidly. It usually develops between 6 months and 3 years of age. The first signs are often delays in developing skills, such as walking and talking. Children may be floppy or have low muscle tone early on (hypotonia), but this turns into stiffness (spasticity) as they age, especially in the arms and legs. Eye disease caused by the degeneration of the optic nerve (optic atrophy) is common later on and can cause poor vision and eventual blindness. Seizures and fast rhythms on an EEG may also result.

A loss of cognitive abilities occurs, and many affected children never learn to walk or lose that ability. Many affected children do not survive beyond their first decade, but some survive into their teens and beyond. Supportive care can contribute to a longer life span by reducing the risk of infection and other complications.

Clinical Diagnosis - INAD

An MRI of the brain and an eye exam are keys to diagnosing INAD. In addition to a loss of motor skills, affected individuals also experience cerebellar atrophy, strabismus (crossed eyes) and nystagmus (rapid involuntary eye movements). Abnormal axons (a part of nerve cells), called spheroid bodies, can be seen on biopsies but may not appear until later in the disease as they accumulate with age. Abnormal brain iron accumulation varies among affected individuals and may not be evident on MRI studies.

Management - INAD

Drugs are given to treat spasticity and seizures. Fiber supplements and/or stool softeners are used to treat constipation. A transdermal scopolamine patch may help with mouth secretions. Feeding modifications such as softer foods or a feeding tube may be required to prevent aspiration pneumonia and achieve adequate nutrition.

Atypical NAD

NAD usually starts in early childhood but can occur as late as the end of the second decade. It has a slower progression and a different variety of movement problems than INAD. At first, the individual may have delays in speaking or exhibit features similar to autism. Eventually, difficulty with movement develops, and these individuals usually have dystonia. Behavior changes are common, such as acting impulsively, not being able to pay attention for long periods of time or depression, which may require treatment by a doctor.

Clinical Diagnosis - Atypical NAD

Certain MRI views (T2-weighted images) of the brain show an abnormality in the globus pallidus, a part of the brain that controls movement. The abnormality, called hypointensity, indicates iron accumulation. Consequently, an MRI and eye exam are keys to establishing strong clinical features of NAD.

Predominant features of NAD are onset before age 20, psychomotor regression (i.e. loss of previously acquired skills), language difficulties, autistic-like behavior, cerebellar atrophy, optic atrophy, progressive dystonia and dysarthria. As with INAD, biopsies show evidence of abnormal axons called spheroid bodies. Other common features are psychiatric and behavior abnormalities, spasticity, joint contractures, seizures and nystagmus.

Management - Atypical NAD

Drug therapy is provided for spasticity and seizures. For dystonia associated with atypical NAD, oral or intrathecal baclofen may be tried. Treatment by a psychiatrist is indicated for those with later-onset neuropsychiatric symptoms. Fiber supplements and/or stool softeners are used to treat constipation. A transdermal scopolamine patch may help with mouth secretions. Feeding modifications such as softer foods or a feeding tube may be required to prevent aspiration pneumonia and achieve adequate nutrition.

PLA2G6-related dystonia-parkinsonism

The onset of PLA2G6-related dystonia-parkinsonism varies from childhood to second and third decade of life. These individuals experience dystonia, eye movement abnormalities, slowness, poor balance, rigidity and marked cognitive decline.

Clinical Diagnosis - PLA2G6-related dystonia-parkinsonism

Abnormal brain iron accumulation in the globus pallidus, substantia nigra and/or striatum varies among affected individuals and may not be evident on MRI studies until late in the disease.

The main features are variable onset from childhood to young adulthood; parkinsonism (tremor, bradykinesia [slow movements], rigidity and impaired postural responses); dystonia; cognitive decline; neuropsychiatric changes; and an initial dramatic response to dopaminergic (levodopa) treatment followed by the early development of dyskinesias (diminished voluntary movements and the presence of involuntary movements). Other common features are dysarthria, autonomic involvement and mild cerebral atrophy.

Management - PLA2G6-related dystonia-parkinsonism

Consider treating with dopaminergic agents. Consult a psychiatrist to treat neuropsychiatric symptoms. A physical therapy evaluation may help problems with posture and walking. Occupational therapy can help the person perform activities of daily living. Periodic assessment of vision and hearing may be needed. To prevent secondary complications: Start physical therapy early and orthopedic management to help prevent contractures (tightening of the muscles, tendons, skin and nearby tissues, which cause joints to stiffen) as the disease progresses.

Genetics

PLAN is inherited in an autosomal recessive manner, meaning the affected individual receives two mutated genes, one from each parent. This is how it works:

  • Because most of our genes exist in pairs (one coming from the mother and one coming from the father), we normally carry two working copies of each gene. When one copy of a recessive gene has a change (mutation) in it, the person should still have normal health. That person is called a carrier.
  • Recessive diseases only occur when both parents are carriers for the same condition and then pass their changed genes on to their child. There is a one in four chance that two carriers would have a child with the disorder. There is a two in four chance the parents will have a child who is also a carrier. The chances are one in four that the child will not have the gene mutation.

Carrier testing for at-risk relatives and prenatal testing for pregnancies at risk are suggested if both disease-causing mutations have been identified in an affected family member.

Prenatal Testing

If the disease-causing mutations have been identified in the family, prenatal testing for pregnancies at increased risk can be done. In one test, DNA is extracted from fetal cells obtained by amniocentesis, usually at 15 to 18 weeks’ gestation, and analyzed. Or, sampling is done of the chorionic villus, the tiny finger-like projections on the edge of the placenta, usually at 10 to 12 weeks’ gestation.

Embryo screening, known as preimplantation genetic diagnosis, may be an option for some families in which the disease-causing mutations have been identified.

Note


A main resource for the clinical information provided here is PLA2G6-Associated Neurodegeneration - GeneReviews® - NCBI Bookshelf. GeneReviews is primarily used by genetics professionals so the terminology and information may be difficult to understand for the general public.


Research

The following discussion of research to better understand PLAN is for informational purposes only. We do not endorse specific studies or clinical trials, experimental drugs, procedures, biotech or pharmaceutical companies.

Research has been vital to understanding the role of PLA2G6 and how the loss of function affects nerve health. Understanding the role of this gene is an important step in finding ways to treat the disease. The function of the gene has been investigated in various animal models such as fruit flies and mice, as well as in induced pluripotent stem cell cultures (iPSCs). iPSCs are derived from skin (fibroblasts) or blood cells of healthy or affected individuals and converted to stem cells through overexpression of a group of genes known as Yamanaka factors. These cells can then be converted to any type of cell in the body. The relevant cell type that is used for PLAN research are dopamine neurons. The function of the protein produced by the gene, called iPLA2β, is not fully understood. According to preliminary research, it is thought to be a phospholipase enzyme that controls fatty acid levels in the brain. PLA2G6 mutations appear to cause a loss of catalytic activity and mislocalization of the protein, meaning the protein is not found where it normally is located, in the distal axons and dendrites.

Studies in patient cells show that the loss of PLAG26 function causes an expansion of lysosomes (membrane-covered cell structures that break down cellular waste), aberrant morphology (abnormal shapes) and changes in the mitochondria, and accumulation of glucosylceramide (a basic component of the cell membrane). This is very similar to what is observed in fruit fly models, which suggests that drugs that work in the flies may work in humans. This opens up an avenue for screening drugs that alleviate neurodegeneration.

Researchers have been seeking therapeutic approaches to restore PLA2G6 enzyme function in INAD. Potential therapeutic strategies may include gene therapy, enzyme replacement therapy through modification of the PLA2G6 protein so it can be delivered to the brain, use of pharmacological chaperones to improve the function of mutant PLA2G6 proteins, or stimulation of other enzymes to compensate for the loss of PLA2G6 function such as Acyl CoenzymeA or other phospholipase enzymes. Challenges exist for each of these potential treatments, and all require extensive research before they can be tried in human subjects.

One of the potential therapeutic strategies, pharmacological chaperones, has shown potential in early research. As proteins are produced from DNA, they are folded into configurations that are required for proper function in the cell by small molecules called chaperones. Further research in this area would involve screening many drugs for the potential to act as a chaperone and to stabilize PLA2G6 protein folding.

Another potential therapeutic strategy is stimulation of other enzymes. Scientists have hypothesized that other enzymes called acyl CoA synthetases could compensate when the PLA2G6 enzyme is impaired by mutations. They have observed that a protein that stimulates the activity of acyl CoA synthetases has a beneficial effect on mice with a PLA2G6 mutation.

Gene Therapy

Research into gene therapy for INAD is in the early stages of development. The goal of gene therapy is to deliver healthy copies of the gene, in this case PLA2G6, into a patient’s cells to treat the disease. Genes can be delivered through a viral vector, which is a virus that is scientifically engineered to carry the functional gene into the cell. Before a treatment can be tested in humans, it must first be used in animal models that are genetically engineered to have a PLA2G6 mutation. Scientists have developed a mouse model that is genetically engineered to have a PLA2G6 mutation. These mice develop movement and coordination problems similar to individuals with INAD. Researchers have found that when the mice receive the gene therapy, they have improved outcomes in length of life and mobility.

Researchers are now in a position to publish their findings and seek major grant funding to potentially investigate the therapy in human subjects. The researchers hope this strategy paves the way for future clinical trials in patients with PLAN.

PLAN Natural History Studies and Biobanks

PLANReady Natural History Study

The NBIA Research Group at Oregon Health & Science University has developed a study called PLANready. Its purpose is to help better understand the natural history of PLAN, meaning how symptoms appear and change over time. By studying individuals with PLAN, they also hope to identify disease markers that can be used in future clinical trials. A disease marker is any symptom or measurement that happens reliably in a disease, changes predictably with disease progression and becomes “better” with successful treatment. A disease marker could be an MRI finding, a protein level in the blood, or a rating scale to measure symptoms or function. Natural history studies provide data that serve as the foundation for future drug trials.

To find out more about this natural history study and to contribute to data collection go to: PLANready | NBIA.

TIRCON International NBIA Registry

The TIRCON International NBIA Registry was created under an EU grant called Treat Iron-Related Childhood-Onset Neurodegeneration. Grant funding ran from 2011 to 2015, and the project is housed at Ludwig Maximilian University of Munich, Germany. The NBIA Alliance and other sources have provided registry funding since 2015. Clinical centers from 12 countries currently take part in the registry by entering their patient data. There are over 700 entries consisting of NBIA patients and controls as of July 2020. Clinical centers seeing at least five NBIA patients are eligible to participate. Clinical and natural history data is available to researchers studying NBIA disorders. Contact Anna Baur-Ulatowska at Anna.Baur@med.uni-muenchen.de for more information on this registry.

Retrotope Natural History Study

Retrotope, Inc., is a biopharmaceutical company that began a natural history study in June 2019 with an estimated completion date of December 2021. It is looking at various outcome measures such as INAD mortality and morbidity. The study is not recruiting more participants at this time. For more information, go to A Natural History Study of Infantile Neuroaxonal Dystrophy.

The New York Stem Cell Foundation Research Institute (NYSCF)

The NYSCF’s mission is to accelerate cures for major diseases through stem cell research. The institute is collecting skin cell samples from patients and their families in addition to obtaining medical and family histories. The skin cells are then converted into induced pluripotent stem cells, which are cells that have the ability to differentiate into different types of cells such as neurons. This enables researchers to understand more about the disease and accelerate the rate in which treatments can be discovered. To learn more about this biobank, contact Geoff McGrane at gmcgrane@NYSCF.org

PLAN Clinical Trials

A clinical trial by Retrotope sets out to test a potential drug treatment called RT001 for individuals with INAD. This trial has 19 participants who all received the compound and are being followed over time to observe their response. It began in November 2018 and is expected to be completed by June 2021.

More information can be found at A Study to Assess the Efficacy and Safety of RT001 in Subjects With Infantile Neuroaxonal Dystrophy.

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for information on clinical studies.

PLAN Research Publications and Articles

Following is a list of some recent research articles. Others can be found at Pub Med Central.

2023 - Exploring therapeutic strategies for infantile neuronal axonal dystrophy (INAD/PARK14)

2021 - Compound heterozygous PLA2G6 loss-of-function variants in Swaledale sheep with neuroaxonal dystrophy

2020 - Lack of Association Between PLA2G6 Genetic Variation and Parkinson's Disease: A Systematic Review

2018 - PLA2G6-Associated Neurodegeneration (PLAN): Review of Clinical Phenotypes and Genotypes

2018 - Pantothenate Kinase-Associated Neurodegeneration (PKAN) and PLA2G6-Associated Neurodegeneration (PLAN): Review of Two Major Neurodegeneration with Brain Iron Accumulation (NBIA) Phenotypes

2017 - Disruption of Golgi morphology and altered protein glycosylation in PLA2G6-associated neurodegeneration

PKAN

Pantothenate Kinase-Associated Neurodegeneration is one of the most common forms of NBIA. Approximately 30-35% of the NBIA population has PKAN.

It is caused by mutations in the PANK2 gene on chromosome 20. This gene provides the instruction for making an enzyme called pantothenate kinase. Researchers are investigating how this missing enzyme damages nerve cells in the brain and causes iron to build up.

There are two forms of PKAN: classic and atypical, although some people have symptoms that place them in between the two categories.

Classic PKAN

Individuals with classic disease have a more rapid progression of symptoms. According to published literature, classic PKAN develops before 6 years of age in 90% of patients. The onset age, however, ranges from 6 months to 12 years, with an average onset occurring at 3 years and 4 months.

These children may initially be perceived as clumsy and later develop more noticeable problems with walking. Eventually, falling becomes more common. Because they have trouble protecting themselves during falls, affected children may have repeated injuries to the face and chin.

Many individuals with the classic form of PKAN require a wheelchair by their mid-teens. Most lose the ability to move or walk independently between 10 and 15 years after the beginning of symptoms. Also by this time, they may have enough trouble with chewing and swallowing that a feeding tube becomes necessary.

Dystonia, a movement disorder that causes the muscles to contract and spasm involuntarily, is always present and usually an early manifestation of PKAN. Head and limb dystonia are frequent and may lead to recurrent trauma to the tongue from tongue biting or from the direct impact of falls. Botulinum toxin can be effective in managing oral dystonia and is a first-line treatment to consider. In some extreme cases, removal of all teeth may be necessary. In addition, bone fractures have been reported from the combination of extreme bone stress and osteopenia.

Patients are at risk of premature death. The dystonia can result in swallowing difficulty and poor nutrition. Such secondary effects are more likely to cause premature death than the primary neurodegenerative process. However, life span varies among affected PKAN individuals. With improvements in medical care, more are living into adulthood.

Atypical PKAN

The atypical form of PKAN usually occurs after age 10 and within the first three decades of life. The average age for developing symptoms is 13. The disease progresses more slowly than classic PKAN and generally is less severe.

The symptoms vary among individuals and are more different than those of early-onset disease. The inability to walk typically occurs 15 to 40 years after symptoms develop. Speech is affected early on. Common speech problems are repeating words or phrases (palilalia), rapid speech (tachylalia) and slurring words (dysarthria).

Psychiatric symptoms are more commonly observed in atypical PKAN and can include impulsive behavior, violent outbursts, depression and rapid mood swings. Movement problems are common, although they develop later. Patients often are described as having been clumsy in childhood and adolescence. Similar to Parkinson’s disease, “freezing” while walking may occur, especially when turning a corner or encountering surface variations. Shaking or tremors also have been reported.

Degeneration of the retina may occur, though much less often than with classic PKAN.

Clinical Diagnosis

PKAN is suspected when magnetic resonance imaging (MRI) changes are seen in an individual with typical PKAN symptoms.

All individuals with PKAN have high levels of brain iron, mainly in the globus pallidus. PKAN has a unique characteristic seen on an MRI. Iron accumulation generally makes the brain look dark on certain (T2-weighted) MRI views. In PKAN, this dark area has a very bright spot in the center, called "the eye of the tiger" sign, pictured below. It is rarely seen in other forms of NBIA.

The sign sometimes is absent in early-disease stages. In the Dominican Republic, where over 20 affected individuals have been diagnosed with PKAN and have the same PANK2 mutation, it has been reported that many of these individuals lacked the eye of the tiger, despite their similarities to others in this group.

Some cases with a purported but not actual eye of the tiger sign will be found to instead have Mitochondrial-membrane Protein-Associated Neurodegeneration or MPAN, a different, less-common form of NBIA.

The movement disorder seen in PKAN individuals may include one or more of the following: dystonia, rigidity or choreoanthetosis (twisting and writhing). Other common features include involvement of the corticospinal tract, which is responsible for conducting impulses from the brain to the spinal cord, extensor toe signs that indicate damage to the central nervous system and spasticity, along with retinal degeneration or optic atrophy. Seizures are rare.

When PKAN is suspected, genetic testing is recommended to confirm the diagnosis.

Evaluations Following Initial Diagnosis

To establish the extent of disease in an individual diagnosed with PKAN, the following evaluations are recommended:

  • Neurologic examination for dystonia, rigidity, choreoathetosis and spasticity, including evaluation of walking and speech
  • An eye exam for evidence of retinopathy and optic atrophy
  • Screening to assess if there are delays in development, with referral for more formal testing if delay is indicated
  • Assessment for physical therapy, occupational therapy, and/or speech therapy
  • Medical genetics consultation

Treatment of Dystonia

Dystonia is a movement disorder in which your muscles contract involuntarily, causing repetitive or twisting movements. This is the most debilitating and distressing symptom of the disease. Dystonia and spasticity are usually managed with different drugs such as anticholinergics, benzodiazepines and other anti-spasticity agents. For focal dystonia, which is dystonia in one part of the body, botulinum toxin (Botox) injections can also provide targeted relief of dystonia and spasticity. The first-line drugs that are most commonly effective in PKAN are trihexyphenidyl, clonazepam and baclofen. Second-line drugs for PKAN include clonidine, gabapentin, tetrabenazine and pregabalin. Physical and occupational therapy can be useful to maintain normal joint mobility for as long as possible, particularly for those who are only mildly symptomatic. Some of the most commonly used treatments for dystonia are baclofen, deep brain stimulation (DBS) and Botox.

Baclofen Pump: One of the most consistent forms of relief from dystonia is baclofen. This medication is first taken orally, but a baclofen pump may be an option for some individuals. An evaluation can be done to determine the likelihood a patient would respond positively to a pump, which is surgically implanted under the skin of the abdomen.

DBS: Deep brain stimulation is another option used to treat dystonia in NBIA individuals. It involves the placement of electrodes in the brain, which are attached to wires leading to a battery-operated neurostimulator implanted in the chest. The neurostimulator sends pulses to targeted areas in the brain and takes “off line” the part of the brain that is sending too many signals and causing the muscles to move in painful ways. DBS is the most commonly performed surgical treatment for Parkinson's disease.

Botox: Injection of botulinum toxin into muscles affected by dystonia can provide relief for several months at a time. Botox helps relieve involuntary contractions causing pain, twisting, abnormal posture or changes in a person's voice or speech, by causing temporary weakness in those muscles. Because each affected muscle must be injected, this is most practical when an individual has dystonia significantly affecting a specific body area, such as the hand or jaw. Resistance to Botox is a phenomenon, which may cause the treatment to lose its effectiveness over time. It occurs because the body makes antibodies to combat the toxin.

Please see our Medical Information section for more in-depth information on these therapies.

What to Expect

PKAN is a progressive disorder. Affected individuals may experience episodes of rapid deterioration, often lasting one to two months, interspersed with longer periods of stability. Reasons for this are not clearly understood.

As the disease progresses, episodes of extreme distress may last for days or weeks. It is especially important during these episodes to evaluate for causes of pain so it can be treated. These may include occult GI bleeding, urinary tract infections and bone fractures. Individuals with PKAN are at an especially high risk for fractures without apparent trauma because of osteopenia and stress on long bones from dystonia.

About two out of three individuals with PKAN develop retinal degeneration, a cohort of debilitating conditions characterized by the progressive loss of photoreceptors and neuronal remodeling (the crucial step in sculpting the mature brain). It is more common in classic PKAN. Loss of peripheral vision may contribute to falling and gait problems in the early stages of PKAN. The retinal degeneration follows a typical clinical course, with nyctalopia (night blindness) followed by progressive loss of peripheral visual fields and sometimes eventual blindness. Evaluation by electroretinogram often detects retinal changes that are asymptomatic. Individuals with a normal eye examination at the time of diagnosis generally do not develop retinopathy later. Optic atrophy is only found in 3% of patients and has not been observed in atypical PKAN.

Swallowing evaluation and regular dietary assessments are needed to assure adequate nutrition. Once the individual can no longer maintain an adequate diet orally, a gastrostomy (feeding) tube becomes necessary.

The following should be performed on a regular basis: monitoring of height and weight using appropriate growth curves to screen children for worsening nutritional status; eye assessment; oral assessment for consequences of trauma; assessment of walking and speech abilities.

Two of the main resources for the clinical information are the PKAN - Best Practices which was created in collaboration with many different scientists and clinicians, and PKAN - GeneReviews. GeneReviews is primarily for the use of genetics professionals so the terminology and information may be difficult for the general public to understand.

Metabolism & Energy Requirements in people with PKAN

Connect with Us

Patient & Family Experience with PKAN

Connect with Us

* select to download flyer

Genetics

PKAN is an inherited autosomal recessive disorder. Because most of our genes exist in pairs (one coming from the mother and one coming from the father), we normally carry two working copies of each gene. When one copy of a recessive gene has a change (mutation) in it, the person should still have normal health. That person is called a carrier. Recessive diseases only occur when both parents are carriers for the same condition and pass their changed genes on to their child.

There is a one in four chance that two carriers would have an affected child. The chances are two in four that the couple will have a child who also is a carrier; and they are one in four the child won’t have the gene mutation.

Carrier testing for at-risk relatives and prenatal testing can be obtained if both disease-causing mutations have been identified in an affected family member.

Prenatal Testing

If the disease-causing mutations have been identified in the family, prenatal diagnosis for pregnancies can be done by analyzing DNA extracted from fetal cells in amniocentesis (usually performed at 15 to 18 weeks of gestation) or chorionic villus sampling (usually performed at 10 to 12 weeks of gestation).

Preimplantation genetic diagnosis, used to identify genetic defects in embryos created through in vitro fertilization (IVF),may be an option when the disease-causing mutations have been identified.

Natural History Studies

TIRCON International NBIA Registry

The TIRCON International NBIA Registry was created under an EU grant called Treat Iron-Related Childhood-Onset Neurodegeneration. Grant funding ran from 2011 to 2015, and the project is housed at Ludwig Maximilian University of Munich, Germany. The NBIA Alliance and other sources have provided registry funding since 2015. Clinical centers from 12 countries currently take part in the registry by entering their patient data. There are over 700 entries consisting of NBIA patients and controls as of July 2020. Clinical centers seeing at least five NBIA patients are eligible to participate. Clinical and natural history data is available to researchers studying NBIA disorders. Contact Anna Baur-Ulatowska at Anna.Baur@med.uni-muenchen.de for more information on this registry.

PKANReady

The Oregon Health and Science University has a registry and natural history study that involves patients or guardians entering data remotely by phone, paper or online. It involves the retrospective use of medical records as well as listing PKAN milestones and patient (or parent) reported outcome measures.

The research team plans to use Latent Growth Models (LGM) in which latent growth curves will model disease progression. Understanding the progression of the disease and how it manifests in different cases will increase the knowledge of the disease and ultimately help researchers identify important aspects of the disease to study and treat. PKAN families wishing to participate can find more information and register at PKANready.

PKAN Clinical Trials

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for information on clinical studies.

The PANK2 mutation is thought to result in the reductions of cellular coenzyme A (CoA). CoA is cell autonomous and thus any effective PKAN therapy must penetrate both cellular membranes and the blood-brain barrier (BBB). This presents a challenge in finding a suitable drug that can perform these functions.

Another challenge faced by PKAN researchers has been finding a suitable model on which to perform preliminary research. Using other species as models for the disease causes limitations on the implications of the research. In particular, PKAN researchers have lacked a compelling mammalian model in which an abnormality can be specifically attributed to a defect in PANK2. The state of the models is central to the development of therapeutic treatments.

One clinical trial is currently underway, the CoA-Z Clinical Trial, and another is planned in 2022 by CoA Therapeutics. In addition, other research efforts are in the early stages, such as gene therapy and repurposing drugs already previously approved by the Food and Drug Administration.

CoA-Z Clinical Trial

A recent study showed that a group of proteins called 4’-phosphopantetheine corrected CoA metabolic defects and resolved all secondary abnormalities in mouse and human cell models. With these results, they have demonstrated the effectiveness of representing key features of the human disease in the mouse model and provided evidence in support of 4’-phosphopantetheine as a therapeutic candidate for PKAN.

In the study, oral administration of 4’-phosphopantetheine normalizes brain biomarkers in the living mouse model, which strongly suggests that the molecule is not degraded by intestinal phosphatases, readily crosses membranes and reaches the brain intact. In December 2019, the CoA-Z clinical trial started recruiting participants to test 4’-phosphopantetheine in humans. This research is being done by OHSU through funding by the National Institutes of Health and the Spoonbill and Lepelaar Foundations.

After safety studies were done to prove favorable safety and tolerability profiles with no adverse side effects, the researchers moved on to the Phase 2 portion of the study with the CoA-Z clinical trial.

Phase 2 is being conducted entirely remotely with about 60 participants. The study is testing three different doses of 4’-phosphopantetheine and a placebo. The study is being performed in a double-blind manner, in which neither the participants nor the researchers know which randomly selected participants are receiving a particular treatment or placebo. The treatment for each participant is three years. The trial is in progress. More information about the trial and enrollment information can be found at http://nbiacure.org/coaz-clinical-trial/.

Published papers on this work:

2019 - 4'Phosphopantetheine corrects CoA, iron, and dopamine metabolic defects in mammalian models of PKAN

2019 - CoA‐dependent activation of mitochondrial acyl carrier protein links four neurodegenerative diseases

CoA Therapeutics Clinical Trial

CoA Therapeutics and St. Jude Children’s Research Hospital are developing pantazines for the treatment of PKAN. Pantazines are a series of small drug-like molecules that stimulate CoA synthesis with a promising potential for PKAN therapy. A pantazine compound has been discovered that treats a new mouse model with brain CoA deficiency. Pantazines rescue movement dysfunction and substantially extend the shortened lifespan of the mouse model. Pantazines function by activating other PANK proteins to compensate for mutated PANK2 in PKAN.

CoA Therapeutics is developing pantazines as an oral therapy (e.g. pill or liquid) for the treatment of PKAN with plans to initiate a Phase I study in healthy volunteers in 2021 and a study in PKAN patients in 2022.

Published papers on this work:

2016 - Allosteric Regulation of Mammalian Pantothenate Kinase

2018 - A therapeutic approach to pantothenate kinase associated neurodegeneration

Clinical Trials with Negative Results

Deferiprone Clinical Trial

Iron chelating drugs were thought to be a potential PKAN therapy by removing excess iron from the brain. A long-term deferiprone (iron chelator) study with 88 participants was done from June 2014 to March 2018 through an international trial conducted at clinical centers in the US, Germany, Italy and the United States under a 5.2 million euros (approx 7 million USD) EU grant called Treat Iron-Related Childhood-Onset Neurodegeneration, or TIRCON. This study showed that while the drug successfully reduced the amount of accumulated iron in the brain for PKAN individuals regardless of onset age, the treatment was not effective for PKAN patients in a statistically significant way. The study found a slight indication that deferiprone may slow the progression of the disorder in older patients with later-onset, or atypical PKAN.

Published papers on this work:

2019 - Safety and efficacy of deferiprone for pantothenate kinase-associated neurodegeneration: a randomised, double-blind, controlled trial and an open-label extension study

Retrophin Clinical Trial of Fosmetpantotenate

Seventy-eight PKAN individuals completed a 24-week randomized, double-blind study, meaning that neither the patients nor the doctors knew who was randomly selected to get the drug or placebo. Although the drug was observed to be safe and generally well-tolerated, the study found that it did not meet its primary or secondary endpoints, or outcome measures.

First, the study found no differences between those who received the drug and those who got the placebo. That determination was based on the extent to which individuals improved over the 24-week trial, based on a scale that measures activities of daily living, such as walking, eating and dressing. Those measures were specifically adapted for PKAN individuals using Part II of the comprehensive and widely referenced Unified Parkinson’s Disease Rating Scale.

Second, the study found no measurable change on the same scale’s Part III score, which evaluates motor function, including slowness, stiffness and balance.

No data suggested that a longer course of treatment would change the outcomes, nor were there any differences seen between classic and later-onset PKAN individuals taking part in the trial.

Published papers on this work:

2020 - Fosmetpantotenate Randomized Controlled Trial in Pantothenate Kinase–Associated Neurodegeneration

2019 - The Fosmetpantotenate Replacement Therapy (FORT) randomized, double-blind, Placebo-controlled pivotal trial: Study design and development methodology of a novel primary efficacy outcome in patients with pantothenate kinase-associated neurodegeneration

 

Press Release with Results:

 

2019 - Retrophin Announces Topline Results from Phase 3 FORT Study of Fosmetpantotenate in Patients with PKAN

More information on PKAN and some of the latest research for this disorder can be found at https://nbiascientificsymposium.org for day 1 of the 7th International Symposium on NBIA & Related Disorders held September 30 – October 3, 2020.

PKAN Research Publications and Articles

Following is a list of recent PKAN research articles. Other PKAN research articles and studies can be found at Pub Med Central.

2022 - Bi-Allelic Mutations in Zebrafish pank2 Gene Lead to Testicular Atrophy and Perturbed Behavior without Signs of Neurodegeneration

2021 - Treat Iron-Related Childhood-Onset Neurodegeneration (TIRCON)—An International Network on Care and Research for Patients With Neurodegeneration With Brain Iron Accumulation (NBIA)

2021 - Coenzyme A levels influence protein acetylation, CoAlation and 4′-phosphopantetheinylation: Expanding the impact of a metabolic nexus molecule

2020 - A pantothenate kinase-deficient mouse model reveals a gene expression program associated with brain coenzyme a reduction

2020 - Abnormal Vasculature Development in Zebrafish Embryos with Reduced Expression of Pantothenate Kinase 2 Gene

2020 - Tongue Protrusion Dystonia in Pantothenate Kinase-Associated Neurodegeneration

2020 - Pilot trial on the efficacy and safety of pantethine in children with pantothenate kinase-associated neurodegeneration: a single-arm, open-label study

2020 - Fosmetpantotenate Randomized Controlled Trial in Pantothenate Kinase–Associated Neurodegeneration

2019 - The Fosmetpantotenate Replacement Therapy (FORT) randomized, double-blind, Placebo-controlled pivotal trial: Study design and development methodology of a novel primary efficacy outcome in patients with pantothenate kinase-associated neurodegeneration

2019 - 4'Phosphopantetheine corrects CoA, iron, and dopamine metabolic defects in mammalian models of PKAN

2019 - CoA‐dependent activation of mitochondrial acyl carrier protein links four neurodegenerative diseases

2019 - Safety and efficacy of deferiprone for pantothenate kinase-associated neurodegeneration: a randomised, double-blind, controlled trial and an open-label extension study

2019 - Precision medicine in pantothenate kinase-associated neurodegeneration

2019 - Pantothenate Rescues Iron Accumulation in Pantothenate Kinase-Associated Neurodegeneration Depending on the Type of Mutation

2019 - Deep brain stimulation for pantothenate kinase-associated neurodegeneration: A meta-analysis

2019 - Proposed Therapies for Pantothenate-Kinase-Associated Neurodegeneration

2019 - Inborn errors of coenzyme A metabolism and neurodegeneration

2019 - CoA-dependent activation of mitochondrial acyl carrier protein links four neurodegenerative diseases

What is NBIA?

Neurodegeneration with Brain Iron Accumulation is a group of rare, genetic neurological disorders characterized by abnormal accumulation of iron in the basal ganglia. The basal ganglia is a collection of structures deep within the base of the brain that assist in regulating movements.

The exact relationship between iron accumulation and the symptoms of NBIA is not fully understood. Although we all normally have iron in this area, people with NBIA have extra iron that can be seen on MRI (magnetic resonance imaging). Certain MRI views (T-1 and T2-weighted images) show the iron as dark regions in the brain. High brain iron is most often seen in the part of the basal ganglia called the globus pallidus. It is also often seen in another part called the substantia nigra.

NBIA is progressive and, at this time, there is no cure.

Characteristics of the Disorders

The hallmark clinical manifestations of NBIA relate to the body’s muscle function and feature a progressive movement disorder. There are several descriptive terms for the neuromuscular symptoms associated with all forms of NBIA.

Dystonia describes involuntary muscle cramping that may force certain body parts into unusual, and sometimes painful, movements and positions.

Choreoathetosis is a condition characterized by involuntary, rapid, jerky movements (chorea) occurring in association with relatively slow, sinuous, writhing motions (athetosis).

In addition, there may be stiffness in the arms and legs because of continuous resistance to muscle relaxing (spasticity) and abnormal tightening of the muscles (muscular rigidity). Spasticity and muscle rigidity usually begin in the legs and later develop in the arms.

Parkinsonism is a condition marked by tremor, slowness, rigidity and poor balance. As affected individuals age, they may eventually lose control of voluntary movements. Muscle spasms combined with decreased bone mass can result in bone fractures not caused by trauma or accident.

Dystonia affects the muscles in the mouth and throat, which may cause poor articulation and slurring (dysarthria), and difficulty swallowing (dysphagia). The progression of dystonia in these muscles can result in loss of speech as well as uncontrollable tongue-biting.

Specific forms of dystonia that may occur in association with NBIA include blepharospasm and torticollis. Blepharospasm is a condition in which the muscles of the eyelids do not function properly, resulting in excessive blinking and involuntary closing of the eyelids. Torticollis is a condition in which there are involuntary contractions of neck muscles resulting in abnormal movements and positions of the head and neck.

Most forms of NBIA involve eye disease. The most common problems are degeneration of the retina and optic atrophy. The retina is a thin membrane that lines the back of the eyeball; it helps the eye perceive an image and send it into the brain. In NBIA, early signs of retinal degeneration may be poor night vision or tunnel vision. It can eventually cause significant loss of vision.

Optic atrophy affects the optic nerve, which sends messages between the retina and the brain. The optic nerve is like a cable with thousands of tiny electrical wires, each carrying some visual information to the brain. When the nerve is damaged or breaks down, vision can become blurry, side vision or color vision may be abnormal, the pupil may not work properly, or there may be decreased lightness in one eye compared to the other. Eventually, optic atrophy can cause blindness.

A general loss of brain cells and tissue also are frequently observed, conditions called cerebral atrophy and cerebellar atrophy.

Some forms of NBIA involve delays in development, mainly pertaining to motor skills (movement). Although cognitive decline occurs in some types of the disorder, more often thinking, perception and other mental processes are relatively spared. Intellectual testing may be hampered by the movement disorder; therefore, newer methods of studying intelligence are necessary to determine if there are cognitive features involved.

Onset of NBIA ranges from infancy to adulthood. Progression can be rapid or slow with long periods of stability. Symptoms may vary greatly from case to case, partly because the genetic cause may differ between families. Also, different changes (mutations) within a gene could lead to a more or a less severe presentation.

The factors that influence disease severity and the rate of progression are still unknown. Usually individuals with NBIA develop increasing disabilities during the course of the disease. As the disease progresses, adjustments commonly need to be made to medications and other treatments. It may take several tries before the best combination is found.

Auxiliary devices could become necessary and may include wheelchairs and devices that help with speech.

Individuals with NBIA also all share a finding in the nerve cells that can only be detected by performing electron microscopy on nerve tissue obtained from a biopsy. Nerve cells have long extensions, called axons that transmit messages from one nerve cell to the next. In NBIA, some axons are found to be swollen with collections of cellular debris or "junk" that should not be there. These swellings are called spheroids, spheroid bodies or axonal spheroids. In most forms of NBIA, spheroids are only located in the nerves of the brain and spinal cord. Therefore, they are usually not detected until an autopsy is performed on someone who has passed away.

In infantile neuroaxonal dystrophy, or INAD, however, spheroids are also found in nerves throughout the body and a biopsy can be done on skin, muscle, or other tissue to look for them. In a few cases of MPAN, spheroids have also been found in peripheral nerves.

History

Before 2001, NBIA was called Hallervorden-Spatz disease or syndrome. Researchers changed the name to reflect more closely the characteristics of the disorder and to dissociate from the prior name of two unethical Nazi doctors who identified and studied the disorder.

All forms of NBIA were included under the Hallervorden-Spatz name until 2001, when the first NBIA gene was discovered. That gene causes the second most common form of NBIA Pantothenate Kinase-Associated Neurodegeneration, or PKAN.

Over the years, more genes and disorders have since become a part of the NBIA family. In 2006, the PLA2G6 gene was discovered and another NBIA disorder was identified, now known as PLA2G6-Associated Neurodegeneration, or PLAN. Infantile neuroaxonal dystrophy, or INAD, is under PLAN.

In 2011, the gene C19orf12 was identified as being responsible for Mitochondrial-membrane Protein-Associated Neurodegeneration, or MPAN.

Soon after, in 2012, another disorder was put under the NBIA umbrella Beta-propeller Protein-Associated Neurodegeneration or BPAN. With the use of whole exome sequencing (WES) genetic testing increasing, BPAN is diagnosed more quickly and is now the most common NBIA disorder.

These four subtypes of NBIA are considered the most frequent and are identifiable by their varying symptoms and associated gene changes.

Six other rarer disorders are also under the NBIA umbrella, bringing the current total to ten.

All NBIA disorders have separate symptoms and identifying markers but are alike in that they have iron accumulation in a specific area of the brain and are characterized by a progressive movement disorder.

Affected individuals who have the clinical symptoms of NBIA but no genetic confirmation are considered to have idiopathic NBIA, or NBIA of unknown origin.

Genetics

Of the ten forms of NBIA currently identified, most are recessive. Because most of our genes exist in pairs (one coming from the mother and one coming from the father), we normally carry two working copies of each gene. When one copy of a recessive gene has a change (mutation) in it, the person should still have normal health. That person is called a carrier.

Recessive diseases only occur when both parents are carriers for the same condition and then pass their changed genes on to their child. Statistically, there is a one in four chance that two carriers would have an affected child. There is a two in four chance the parents would have a child who is also a carrier, and there’s a one in four chance they would have a child who did not receive the gene mutation.

Neuroferritinopathy is a dominant condition. A person affected with neuroferritinopathy has one working copy and one copy of the gene that is mutated. This single mutation is enough to cause the disease. There is a 50% chance that an affected individual will pass the gene change on to any of his or her children. Most affected individuals have one parent who is also affected.

Beta-propeller Protein-Associated Neurodegeneration is inherited in an X-linked dominant manner, meaning that a single copy of the mutated gene is enough to cause disease in both males and females. Most affected individuals identified so far have been simplex, or isolated cases; they are the only person in their family to have the disease. The majority are females, indicating the mutations are new, or de novo, and suggesting that mutations may be lethal in most males before birth. Still, parental testing is recommended since recurrence has been reported. If neither parent has the variant, then recurrence is slightly greater than the population risk but still less than one percent . When a parent has the same WDR45 variant and evidence for mosaicism, then the recurrence chance could increase to 50%.

Mitochondrial-membrane Protein-Associated Neurodegeneration (MPAN) is inherited in an autosomal recessive manner and less commonly, in an autosomal dominant manner.

Affected Population

Overall, NBIA affects males and females in equal numbers (BPAN occurs more frequently in females). The frequency of NBIA in the general population is estimated between one to three people per 1 million individuals. Because rare disorders like NBIA often go unrecognized, these disorders may be underdiagnosed or misdiagnosed, making it difficult to determine the accuracy of these estimates.

Therapies

Treatment of NBIA is directed towards the specific symptoms that appear in each individual. Research is focusing on a better understanding of the underlying causes of NBIA, which may eventually reveal a more comprehensive treatment.

Treatment may require the coordinated efforts of a team of specialists. Physicians with whom the family may work include the pediatrician or internist, neurologist, pulmonologist, ophthalmologist, orthopedist, gastroenterologist and clinical geneticist.

A team approach to supportive therapy may include physical therapy, exercise physiology, occupational therapy and speech therapy. In addition, many families may benefit from genetic counseling.

One of the most consistent forms of relief from dystonia is baclofen. This medication is first taken orally. A baclofen pump has been used to administer regular doses automatically into the spinal fluid. The pump may be an option for some NBIA individuals, and an evaluation can be done to determine the likelihood that they would respond positively to a pump.

The anti-cholinergic agent trihexyphenidyl (trade name in some countries is Artane) is a second medication that may be taken alone or in combination with baclofen. The combination of baclofen and artane has been found useful for many people with PKAN.

Levodopa/carbidopa (Sinemet) has been helpful for some patients with idiopathic NBIA, although it has not appeared to be helpful for PKAN patients.

Further muscle-relaxing medication include benzodiazepines such as diazepam (trade name in some countries is Valium) and lorazepam (trade name in some countries is Ativan). Efficacy and tolerability may vary from patient to patient.

Individuals experiencing seizures usually benefit from standard anti-convulsive drugs. In addition, standard approaches to pain management are generally recommended where there is no identifiable treatment for the underlying cause of pain.

Many individuals with NBIA have ongoing constipation due to decreased activity, diet and/or medication side-effects. Over-the-counter fiber supplements and stool softeners can often improve the discomfort.

Drugs that reduce the levels of iron in the body (iron chelation) have been studied for effectiveness in treating PKAN with clinical trials. While the most recent study showed the drug deferiprone to be slightly helpful for some PKAN individuals, it was not found effective for the majority and not approved by regulatory authorities.

Injection of botulinum toxin (Botox) into muscles affected by dystonia can also provide relief for several months at a time. This causes temporary weakness of muscles that have involuntary contractions causing pain, twisting, abnormal posture, or changes in person’s voice or speech. Because each affected muscle must be injected, this is most practical when an individual has dystonia significantly affecting a specific body area, such as the hand or jaw.

Deep Brain Stimulation (DBS) is another treatment used to control dystonia. It is performed by implanting electrodes into the brain with a programmable device (neurostimulator) under the skin of the chest or abdomen. The neurostimulator sends pulses to targeted areas of the brain thus altering the pathological patterns of activity in the basal ganglia that cause the muscles to move in painful ways. DBS has been tried on several NBIA individuals with some good results, although it is unclear whether there is a long-term benefit.

The benefits and limitations of any of the above treatments should be discussed in detail with a physician.

NBIA Disorders

PKAN, or Pantothenic Kinase-Associated Neurodegeneration, is caused by mutations in the PANK2 gene. This is the most common form of NBIA, making up approximately 30% of the NBIA population. This gene provides the instruction for making an enzyme called pantothenate kinase. Current research is investigating how this missing enzyme results in damage to nerve cells in the brain as well as the characteristic iron build-up.

PKAN is generally separated into classic and atypical forms, although some people will have characteristics that place them between these two categories. Individuals with classic disease have a more rapid progression of symptoms. In most cases, atypical disease progresses slowly over several years, and sometimes decades. The symptoms and physical findings vary from case to case.

Children with PKAN typically manifest gait problems around age 3 and later develop progressive dystonia, dysarthria, rigidity, spasticity, hyperreflexia and extensor toe signs. Retinal degeneration is common, particularly in classic PKAN. Individuals with later-onset PKAN are likely to present with speech difficulty. Psychiatric symptoms are more frequent in the later onset form.

PLAN, or PLA2G6-Associated Neurodegeneration, is named for the responsible gene, PLA2G6. The group includes INAD, or Infantile Neuroaxonal Dystrophy, NAD, or atypical neuroaxonal dystrophy, which starts a few years later, and PLA2G6-related dystonia-parkinsonism in which onset varies from childhood to second and third decade of life.

Classic INAD has early onset and rapid progression. Affected individuals usually develop signs and symptoms of the disease between 6 months and age 3. The first signs are often delays in developing skills, like walking and talking. Children may be floppy or have low muscle tone early on (hypotonia), but this later turns into stiffness (spasticity) as they get older, especially in the arms and legs. Eye disease caused by degeneration of the optic nerve (optic atrophy) is common and can cause poor vision and eventual blindness.

NAD usually starts at a later age than INAD, typically during early childhood, although it can be as late as the second decade. It has a slower progression and a different variety of movement problems than INAD. At the outset, children may have speech delay or features similar to autism. Eventually difficulty with movement develops. Unlike classic INAD, these “atypical” individuals usually have dystonia. They are also more likely to have behavior changes, such as being impulsive, not being able to pay attention for long periods of time or becoming depressed, which may require treatment by a doctor.

PLA2G6-related dystonia-parkinsonism onset varies from childhood to second and third decade of life. These individuals experience dystonia, eye movement abnormalities, slowness, poor balance, rigidity and marked cognitive decline.

MPAN, or Mitochondrial-membrane Protein-Associated Neurodegeneration, is caused by the autosomal recessive gene, C19orf12. Onset usually occurs in childhood to early adulthood with dystonia, spasticity, weakness, optic atrophy and neuropsychiatric changes.

BPAN, or Beta-propeller Protein-Associated Neurodegeneration, is caused by mutations in the gene WDR45, located on the X chromosome. Most affected individuals identified so far have been simplex, or isolated cases; they are the only person in their family to have the disease. The majority are females, indicating the mutations are new, or de novo, and suggesting that mutations may be lethal in most males before birth. There have been instances of multiple siblings in a family affected with BPAN. In these cases, the mutation was inherited from a mildly affected parent.

Affected individuals have global developmental delay during childhood with slow motor and cognitive gains. However, during adolescence or adulthood, they experience a relatively sudden onset of progressive dystonia-parkinsonism and dementia.

Aceruloplasminemia has mainly been studied in Japan, where it occurs in about one per 2 million adults. It is unclear how often it occurs in other populations. The gene responsible is CP. It is unusual from other forms of NBIA because iron accumulates not just in the brain, but in other organs, including the liver. The main symptoms are retinal degeneration, diabetes, and neurologic disease related to iron build-up in the basal ganglia. Movement problems include face and neck dystonia, blepharospasm, tremors, and jerky movements.

FAHN, or Fatty Acid Hydroxylase-associated Neurodegeneration, is caused by a mutation in the FA2H gene. Onset occurs in childhood featuring leg dystonia, weakness and falling. Affected individuals also experience optic atrophy, profound cerebellar atrophy and white matter changes in the brain, in addition to high brain iron.

Kufor-Rakeb is named for the village in Jordan where it was first described in 1994. In 2010, a mutation in the ATP13A2 gene was deemed responsible. Two individuals in the United States have been found to have this form of NBIA, and there are a few in South America, the Middle East, Asian countries and one from Italy. It has been suggested that only a portion of cases may have iron accumulation; it may develop late in disease course, or it may only be associated with more severe mutations. Symptoms include juvenile parkinsonism, dementia, abnormal eye movements and involuntary jerking of facial and finger muscles.

Neuroferritinopathy is a genetically dominant form of NBIA. It typically starts during adulthood with dystonia, jerky movements (chorea), and mild changes in thinking (cognitive effects). Within 20 years it usually begins to affect movement in all the limbs and causes difficulty speaking and resembles Huntington’s disease. Although the prevalence is unknown, only about 100 cases have been found and most of these share the same gene change, suggesting they have descended from a common ancestor. It is caused by mutations in the FTL gene, which stands for ferritin light. This refers to one of two protein subunits that make up ferritin, a protein in the body that helps store and detoxify iron. Affected individuals have MRIs that are different from those of other NBIA patients.

Woodhouse-Sakati Syndrome is described in 12 Saudi Arabian families. A founder mutation in DCAF17 accounts for the cases in the Saudi Arabian population. Two individuals in the United States have recently been diagnosed with this disorder. Affected individuals have high brain iron and dystonia in addition to hair loss, diabetes, hearing loss, gonadal dysfunction and mental retardation.

CoPan, or COASY Protein-Associated Neurodegeneration is caused by a mutation in the COASY gene. At this time only a few cases have been identified with this rare form of NBIA. At present, it appears that onset usually occurs in childhood and spasticity and dystonia of the lower limbs are present early on, while dystonia of the mouth and jaw appears later in the disease process. Speech problems are also seen, including stuttering and slurry of words, caused by dysarthria.

Idiopathic NBIA is a type of unknown origin that is suspected to be genetic. It’s likely that there are still several additional, less common genes to be found. For many families, the person diagnosed with NBIA is the first and only affected individual, so it is difficult to know whether there is a specific pattern of inheritance. It is thought that most of these cases are probably recessive because there are some families with more than one affected child and because idiopathic NBIA is more common in families where the parents are related, such as distant cousins. This makes it more likely that they share a common recessive gene. The symptoms in this group are more varied because there are probably several different causes of neurodegeneration in this group. As with other forms of NBIA, there are both early-onset and late-onset types.

Please see more information on each of the forms of NBIA under their separate headings in the drop-down menu at the top of the page.

NBIA Disorders

GeneReviews ®

Connect With Us

Connect With Us

Partners

nbia alliance logo1NBIA Cure logoRare Disease Day PartnerTIRCON

Genetic Alliance logoNORDEURORDISGlobal Genes


Disclaimer    |     Privacy Policy    |     Financials    |     Contact Us

Give While You Shop!

Igive.com

G-CFB5FV5NLL