Amyotrophic lateral sclerosis By Summer B Gibson MD (Dr. Gibson of University of Utah owns stock in Recursion Pharmaceuticals.) Originally released June 21, 1996; last updated May 9, 2017; expires May 9, 2020

Introduction

This article includes discussion of amyotrophic lateral sclerosis, Charcot disease, Lou Gehrig disease, , Aran-Duchenne disease, Duchenne-Aran disease, familial amyotrophic lateral sclerosis, flaccid , PLS, primary lateral sclerosis, , progressive crural palsy, progressive muscular atrophy, and spastic bulbar palsy. The foregoing terms may include synonyms, similar disorders, variations in usage, and abbreviations.

Overview

Amyotrophic lateral sclerosis is a devastating neurodegenerative disease characterized by progressive muscle weakness without notable sensory loss. There are now 2 U.S. Food and Drug Administration–approved medications that have demonstrated modest effects on slowing disease progression: riluzole, approved in 1995, and edaravone, approved in 2017. In this article, the author reviews clinical manifestations, risk factors, symptomatic management, and clinical trials and provides updates on recent genetic discoveries.

Historical note and terminology

Aran believed this syndrome was a muscular disease and was the first to use the term “progressive muscular atrophy” (Aran 1850). Cruveilhier first noticed the atrophy of the anterior spinal roots and thought progressive muscular atrophy was a myelopathic disorder (Cruveilhier 1853). Charcot and Joffroy proposed the term “amyotrophic lateral sclerosis” when they noticed the involvement of the corticospinal tract (Charcot and Joffroy 1969). Brain used the term “motor neuron disease” to emphasize the connections between progressive muscular atrophy, amyotrophic lateral sclerosis, and progressive bulbar palsy (Brain 1962). The term “motor neuron disease” also highlights the variety of involvement of upper and lower motor neurons. Rowland suggested using the plural form, “motor neuron diseases,” to describe all of the diseases of the anterior horn cells and the motor system, including spinal muscular atrophies (Rowland 1982). Spinal muscular atrophies are clinically and pathologically distinct from amyotrophic lateral sclerosis.

Clinical manifestations

Presentation and course

Amyotrophic lateral sclerosis is characterized by asymmetric progressive weakness without notable sensory symptoms or loss. Common presenting signs correspond to selective involvement of particular motor systems (Table 1). Presenting symptoms can be bulbar, cervical, thoracic, or lumbosacral. The time course can vary from rapid progression, with patients becoming ventilator-dependent in a few months, to slow progression, with patients still living independently more than 10 years after diagnosis.

Table 1. Clinical Manifestations of Amyotrophic Lateral Sclerosis System involved Symptoms and signs Bulbar • dysarthria • dysphagia • sialorrhea (drooling) • atrophy • tongue • spasticity • Babinski signs • jaw jerk • snout reflexes • spread of reflexes • incoordination • weakness • weakness • • fasciculations Cognitive changes • frontotemporal dysfunction or

Bulbar signs and symptoms. Bulbar signs and symptoms include dysarthria, dysphagia, sialorrhea (drooling), tongue atrophy, and tongue . They are caused by the involvement of cranial nerve motor nuclei in the medulla, including VII (facial), IX (glossopharyngeal), and XII (hypoglossal).

Bulbar signs and symptoms develop during the course of the disease, but they may be a presenting feature, especially in middle-aged women. In older women, as in men, limb involvement is more common. Bulbar involvement leads to difficulty in speaking and swallowing and is often closely correlated with reduced vital capacity.

When the medullary lower motor neurons are primarily affected, the condition is called flaccid or paretic bulbar palsy. When upper motor neurons or their descending tracts (corticobulbar) are affected, the condition is called spastic bulbar palsy. A mixed flaccid and spastic bulbar palsy is usually observed.

Upper motor neuron signs and symptoms. Upper motor neuron signs and symptoms include hyperreflexia, spasticity, Babinski signs, jaw jerk, snout reflexes, spread of reflexes, incoordination, and weakness. Upper motor neuron weakness is different from lower motor neuron weakness because the former may not be associated with muscle atrophy. Patients may complain of slowness, inability to control the muscles (incoordination), or stiffness, which is due to spasticity.

Recognition of upper motor neuron involvement in a patient with severe muscle wasting can be challenging. The muscle may be so weak and atrophied that the tendon reflexes are difficult to elicit. However, elicitable tendon reflex in a wasted muscle probably signifies the co-existence of an upper motor neuron lesion. Yet upper motor neuron signs may disappear as the lower motor neuron weakness progresses.

Lower motor neuron signs and symptoms. These include weakness, muscle atrophy, and fasciculations; they may be focal, multifocal, or diffuse. Focal onset of weakness may be misinterpreted as a focal disease process such as cervical or lumbosacral radiculopathy or nerve entrapment. The absence of sensory findings and the presence of hyperactive reflexes and fasciculations argue against entrapment neuropathy or radiculopathy. However, it should be emphasized that some patients may have superimposed entrapment neuropathies or radiculopathies. MRI of the cervical or lumbosacral spine and electrodiagnostic tests are useful diagnostic tools for these patients.

Fasciculations are common and often abundant and widespread, but patients rarely present with fasciculations as an isolated symptom. Rather, they usually seek medical attention because of weakness and may be not aware of fasciculations. It should be noted that fasciculations are not diagnostic as they may result from any lesion of the lower motor neurons.{embed="pagecomponents/media_embed" entry_id="8897"} “Benign fasciculations,” which are intermittent, focal, and aggravated by stress, exercise, or lack of sleep, may occur in normal subjects without associated weakness. When fasciculations occur with weakness or upper motor neuron signs, consider the possibility of amyotrophic lateral sclerosis.

Because many disorders may cause lower motor neuron signs, it is important to perform a detailed physical examination and electrodiagnostic test to search for evidence of other more benign processes, such as radiculopathies, plexopathies, entrapment neuropathies, or myopathy. Cognitive changes. A significant number of patients with amyotrophic lateral sclerosis have cognitive impairment, particularly . Lomen-Hoerth and colleagues examined the relationship between amyotrophic lateral sclerosis and frontotemporal dementia (Lomen-Hoerth et al 2002; Lomen-Hoerth et al 2003). They studied 100 patients with amyotrophic lateral sclerosis using the mini mental status state examination (MMSE) and word generation (Lomen-Hoerth et al 2003). Thirty-one patients had abnormal word generation; 50% of patients with bulbar onset disease had an abnormal score whereas approximately 25% of patients with limb onset disease had an abnormal score. No patient had an abnormal score on the MMSE. Of the 44 patients who agreed to undergo more extensive testing, 18 had results consistent with probable or definite frontotemporal dementia, and 5 patients had possible frontotemporal dementia (Lomen-Hoerth et al 2003). In another study, they performed a detailed neuromuscular examination, nerve conduction studies and EMG in 36 patients with frontotemporal dementia (Lomen- Hoerth et al 2002). According to the El Escorial criteria, 5 patients had definite amyotrophic lateral sclerosis, and 1 patient had neurogenic changes limited to a single limb. Hosler and colleagues have also reported families in whom some members developed amyotrophic lateral sclerosis, others frontotemporal dementia, and still others amyotrophic lateral sclerosis–frontotemporal dementia (Hosler et al 2000).

Important negatives. Sensory symptoms, memory loss, bladder and bowel dysfunction, ocular palsy, and decubiti are rare. Clinically, amyotrophic lateral sclerosis is usually a pure motor neuron syndrome, and motor neurons that innervate extraocular muscles, the bladder, and anal sphincter muscles are typically unaffected.

Although patients generally have no sensory abnormalities in routine neurologic examinations, detailed quantitative measurements show that 18% of patients have abnormally elevated vibratory thresholds, suggestive of subclinical involvement of some sensory neurons (Mulder et al 1983) and up to 20% of patients have abnormal sensory nerve conduction studies in at least 1 nerve (Pugdahl et al 2007).

Memory loss may be evident in about 5% of patients. Moreover, memory loss is a part of the amyotrophic lateral sclerosis--dementia complex in the Western Pacific (Hirano et al 1961). Clinically, approximately 38% of these patients develop parkinsonism and dementia.

Extraocular muscles are typically spared, but the velocity of saccade or smooth pursuit movements may be abnormal due to extrapyramidal or supranuclear dysfunction (Gizzi et al 1992).

Bladder and bowel function in patients is generally thought to be normal because the motor neurons in the Onufrowicz nucleus that control the sphincter muscles are unaffected. Yet 4% to 40% of patients have clinically significant abnormal urodynamics with associated symptoms due to abnormal supranuclear control over sympathetic, parasympathetic, and somatic neurons (Arlandis et al 2017).

Rarely, patients have decubiti, which is possibly due to normal sensory and autonomic functions. In addition, the collagen fibrils in the skin of patients differ from those in healthy people (Ono et al 1986). However, Sasaki and colleagues reported that decubiti are more common in those patients who are kept alive on ventilators for a prolonged period (Sasaki et al 1992).

Subtypes of amyotrophic lateral sclerosis. Several clinical subtypes have been identified and depend on the selectivity of early clinical involvement.

Progressive bulbar palsy. Bulbar palsy (selective dysfunction in lower cranial nerves) is the presenting symptom in about 20% of all patients with amyotrophic lateral sclerosis. It is most commonly seen and has the worst prognosis in menopausal women. Typically, the condition spreads first to the lower cervical-thoracic area and then to the lumbosacral area (Brooks et al 1991). Rarely, the disease is confined to the bulbar region--a true progressive bulbar palsy.

Primary lateral sclerosis. About 2% to 3.7% of all patients with amyotrophic lateral sclerosis present with a pure upper motor neuron syndrome and never go on to develop lower motor neuron signs. The age of onset is most commonly between 50 and 55 years, and the rate of progression may be slow (Pringle et al 1992). Some patients with amyotrophic lateral sclerosis start out with a pure upper motor neuron syndrome and develop lower motor neuron involvement later. These patients are said to have upper motor neuron-onset amyotrophic lateral sclerosis. Gordon and colleagues (Gordon et al 2006) proposed to define primary lateral sclerosis as a disease entity with pure upper motor signs 4 years after symptom onset. Primary lateral sclerosis is a syndrome of slow progression with high level of function.

Progressive muscular atrophy. Progressive muscular atrophy is a syndrome in patients who only develop lower motor neuron signs and never subsequently develop upper motor neuron signs. In a retrospective cohort, about 10% of patients were identified to have progressive muscular atrophy (Kim et al 2009). Older males more commonly develop this syndrome; mean age of onset is 64 years. Patients with progressive muscular atrophy also have improved survival (median survival, 48 months) compared to those with the more prevalent amyotrophic lateral sclerosis (median survival, 36 months).

Flail-arm syndrome. Flail-arm syndrome occurs in up to 10% of patients as relatively symmetric proximal and distal bibrachial wasting with positive Babinski sign (Hu 1998). The age of onset is similar to that in patients with amyotrophic lateral sclerosis, but the male-to-female ratio is 9:1 in the flail-arm group, compared with 1.5:1 in the amyotrophic lateral sclerosis group. There is a trend toward improved survival in the flail-arm group (median survival, 57 months) compared with the amyotrophic lateral sclerosis group (median survival, 39 months).

Mills hemiplegic variant. This is a variant of amyotrophic lateral sclerosis featuring a combination of upper motor neuron and lower motor neuron signs isolated to one side (Gastaut 1994).

The El Escorial World Federation of (WFN) criteria for the diagnosis of amyotrophic lateral sclerosis and Awaji group modifications. In order to enhance clinical and research studies in amyotrophic lateral sclerosis, The Consortium on Clinical Trials in amyotrophic lateral sclerosis, a Subcommittee of the Motor Neuron Diseases Research Group of the WFN, established The El Escorial WFN Criteria for the Diagnosis of Amyotrophic Lateral Sclerosis in 1994 (Brooks 1994). With growing experiences in clinical trials, the same committee revised the El Escorial WFN Criteria at a meeting at Airlie House, Warretnon, Virginia in 1998 (Miller et al 1999a) and provided consensus guidelines for the design and implementation of clinical trials in amyotrophic lateral sclerosis. The revised criteria for the diagnosis of amyotrophic lateral sclerosis require: (A) The presence of: (1) Evidence of lower motor neuron degeneration by clinical, electrophysiological, or neuropathologic examination (2) Evidence of upper motor neuron degeneration by clinical examination (3) Progressive spread of symptoms or signs within a region or to other regions, as determined by history or examination (B) The absence of: (1) Electrophysiological and pathological evidence of other disease processes that might explain the signs of lower or upper motor neuron degeneration (2) Neuroimaging evidence of other disease processes that might explain the observed clinical and electrophysiological signs

The guidelines recommend that a careful history as well as physical and neurologic examination must search for clinical evidence of upper and lower motor neuron signs in 4 regions including and cervical, thoracic, and lumbosacral of the central (Miller et al 1999a). The guidelines also recommend that ancillary tests, such as electrodiagnostic, neurophysiological, neuroimaging, and clinical laboratory studies, should be reasonably applied, as clinically indicated, to exclude other disease processes.

To facilitate clinical trials, the guidelines also provide criteria for the clinical diagnosis of amyotrophic lateral sclerosis at different levels of certainty (Miller 1999). The clinical diagnosis of amyotrophic lateral sclerosis, without pathological confirmation, may be categorized into various levels of certainty by clinical assessment alone depending on the presence of upper and lower motor neuron signs together in the same topographical anatomic region in either the brainstem or the cervical, thoracic, or lumbosacral spinal cord (Table 2).

Table 2. Level of Certainty in the Clinical Diagnosis of Amyotrophic Lateral Sclerosis According to WFN Guidelines Level of certainty Clinical features Clinically definite amyotrophic lateral • Clinical evidence alone by the presence of upper and sclerosis lower motor neuron signs in 3 regions. Clinically probable amyotrophic • Clinical evidence alone by upper and lower motor lateral sclerosis neuron signs in at least 2 regions with some upper motor neuron signs necessarily rostral to (above) the lower motor neuron signs. Clinically probable, laboratory- • Clinical signs of upper and lower motor neuron supported amyotrophic lateral dysfunction in only 1 region or upper motor neuron sclerosis signs alone in 1 region. --And— • Lower motor neuron signs defined by EMG criteria present in at least 2 limbs, with proper application of neuroimaging and clinical laboratory protocols to exclude other causes. Clinically possible amyotrophic • Clinical signs of upper and lower motor neuron lateral sclerosis dysfunction in only 1 region or upper motor neuron signs alone in 2 or more regions. --Or— • Lower motor neuron signs rostral to upper motor neuron signs and the diagnosis of clinically probable, laboratory-supported amyotrophic lateral sclerosis cannot be proven by evidence on clinical grounds in conjunction with electrodiagnostic, neurophysiologic, neuroimaging, or clinical laboratory studies. * Other diagnoses must have been excluded to accept a diagnosis of “clinically possible” amyotrophic lateral sclerosis. Clinically suspected amyotrophic • A pure lower motor neuron syndrome, wherein the lateral sclerosis diagnosis of amyotrophic lateral sclerosis could not be regarded as sufficiently certain to include the patient in a research study. (This category is deleted from the revised El Escorial Criteria for the Diagnosis of Amyotrophic Lateral Sclerosis.)

In 2008, a consensus group met in Awaji, Japan, and evaluated the best use and interpretation of electrophysiological data in the diagnosis of amyotrophic lateral sclerosis and to increase the sensitivity of the diagnostic criteria for clinical trials in amyotrophic lateral sclerosis (de Carvalho et al 2008). They suggested that electrophysiological signs of lower motor neuron dysfunction should be equivalent to clinical signs of lower motor neuron dysfunction in the “definite,” “possible,” and “probable” diagnosis of amyotrophic lateral sclerosis. This would render the “laboratory-supported possible amyotrophic lateral sclerosis” category redundant. Also, the presence of fasciculation potentials together with chronic neurogenic changes in one muscle is evidence of acute denervation and electrophysiologically equivalent to fibrillation potentials and positive sharp waves. These suggestions likely increase the sensitivity of the diagnostic criteria for amyotrophic lateral sclerosis without changing specificity. Many newer clinical trials apply this “Awaji criteria” for the diagnostic criteria of amyotrophic lateral sclerosis and no longer utilize the “laboratory-supported possible amyotrophic lateral sclerosis” category.

It should be emphasized that these criteria are for the diagnosis of amyotrophic lateral sclerosis for the purpose of clinical trials and other areas of amyotrophic lateral sclerosis research and not for everyday clinical practice. These criteria do not address the issue of early diagnosis or staging of the disease. However, they do comprise the best available consensus of opinion concerning diagnosis of amyotrophic lateral sclerosis. These criteria are useful in establishing uniformity of clinical diagnosis in different centers across the world while conducting clinical trials.

Prognosis and complications

The mean duration of disease in amyotrophic lateral sclerosis, defined as the interval between the onset of symptoms and death, is about 3 to 4 years. The 5-year survival rate is about 25%. Roughly 10% of patients survive more than 10 years. Young age at onset and limb onset are good predictors for slower disease progression (Magnus et al 2002). Clinical vignette

A 56-year-old right-handed man noted progressive difficulty with his speech over the past 12 months. At the time of presentation, there were times during the day that his speech was unintelligible. The patient also had begun noticing some problems with dysphagia over the previous 8 months. He needed to "concentrate" while eating to prevent choking. Over the same period of time, he noticed that his right hands tired easily. He also noticed some difficulty walking, and he would trip over his feet. While working around his farm, he was unable to lift barrels of hay, which he had been able to do several months before. He had noticed some intermittent muscle twitching over his thigh. He had some progressively intermittent shortness of breath. He denied any numbness, tingling, or bowel and bladder dysfunction.

Neurologic examination showed that he was extremely dysarthric. His tongue showed some fasciculations and atrophy. The patient had some fasciculations in the right trapezius muscle. He had proximal and distal weakness (4/5) in both upper extremities and distal weakness in the left lower extremity. Reflexes were hyperactive (3/4) diffusely. Nerve conduction velocity/EMG study demonstrated widespread acute and chronic denervation in upper extremities and left lower extremity as well as in the tongue and midthoracic paraspinal muscles.

He was diagnosed with amyotrophic lateral sclerosis. He refused to take riluzole because of the possible liver toxicity. After an intensive search on the Internet, he started taking creatinine, N-acetyl cysteine (NAC), Coenzyme Q10, lipoic acid, glutathione, grape seed extract, vitamin C, vitamin E, and multivitamins.

Biological basis

Etiology and pathogenesis

Genetic causes. Most cases of amyotrophic lateral sclerosis have no family history (aka sporadic amyotrophic lateral sclerosis); however, about10% of cases have a first- or second- degree family history of the disease (aka familial amyotrophic lateral sclerosis) (Siddique et al 1996). Current estimates suggest that about 68% of patients with a family history and about 10% of patients without a family history of amyotrophic lateral sclerosis have an identifiable genetic etiology (Renton et al 2014). Twin studies estimate the heritability of amyotrophic lateral sclerosis to be about 60% (Al-Chalabi et al 2010).

In 1993 the first amyotrophic lateral sclerosis–associated gene was identified (Rosen et al 1993). Since then, more than 30 mostly dominantly inherited amyotrophic lateral sclerosis genes have been reported. SOD1 detoxifies superoxide, creating oxygen and hydrogen peroxide, which can then be cleared by catalase and glutathione peroxidase. Copper is required for SOD1 activity, whereas zinc is thought to stabilize the protein structure. Mutations in SOD1 account for 10% to 20% of cases with a positive family history; to date, more than 155 unique SOD1 mutations have been identified (Robberecht and Philips 2013). The majority of mutations in SOD1 are missense mutations, with a small percentage of deletion and insertion mutations that result in prematurely terminated SOD1 polypeptides. Considerable phenotypic variation in SOD1-mediated amyotrophic lateral sclerosis occurs, including age at onset and severity (Andersen et al 2003). Clinical variation occurs even among patients of the same SOD1 genotype and members of the same family (Andersen et al 1997).

In 2011, the GGGGCC hexanucleotide expansion of C9orf72 was identified as a genetic cause of amyotrophic lateral sclerosis (Renton et al 2011). This mutation is the most common known cause of amyotrophic lateral sclerosis in those with a family history as well as in those without a family history. It accounts for about 7% of all amyotrophic lateral sclerosis cases in a Caucasian population (Majounie et al 2012).

Two genes that play a role in the pathological findings of amyotrophic lateral sclerosis are TAR DNA binding protein (TARDBP) (Kwiatkowski et al 2009) and fused in sarcoma (FUS) (Kwiatkowski et al 2009; Vance et al 2009). Mutations in TARDBP have been found in about 5% of patients with familial amyotrophic lateral sclerosis (Kabashi et al 2008; Sreedharan et al 2008; Yokoseki et al 2008; Del Bo et al 2009). Mutations in the FUS have been found in 3% to 4% of patients with familial amyotrophic lateral sclerosis (Kwiatowski et al 2009; Vance et al 2009). Both FUS and TARDBP are DNA/RNA binding proteins and imply that abnormal DNA/RNA metabolism is a pivotal event (Mackenzie et al 2010). The exact mechanism leading to the accumulation of TDP-43 and FUS accumulation and the subsequent is unknown.

In 2011, abnormalities in the ubiquilin 2 gene were identified as a cause of X-linked dominant juvenile and adult-onset amyotrophic lateral sclerosis (Deng et al 2011). This group of researchers noted that ubiquilin 2 gene mutations caused amyotrophic lateral sclerosis and accumulations of ubiquilin 2 protein without the gene mutations were also associated with the disease. The ubiquilin 2 protein plays a role in cellular maintenance and normally disposes of damaged proteins. This finding bears similarity to other recently discovered gene mutations. Mutations in the genes for the TDP-43 and FUS proteins are known to cause familial amyotrophic lateral sclerosis, but abnormal and possibly disease-contributing accumulation of these 2 proteins also occur in sporadic amyotrophic lateral sclerosis, even in the absence of the mutation.

Juvenile-onset amyotrophic lateral sclerosis genes. There are 3 loci for juvenile onset amyotrophic lateral sclerosis: 1 is autosomal dominant, ALS4 (chromosome 9q34) (Chen et al 2004), and 2 are autosomal recessive, ALS2 (chromosomes 2q33) (Yang et al 2001) and ALS5 (chromosome 15q) (Hentati et al 1998). In general, survival time from diagnosis is longer and disease progression slower in the juvenile-onset cases. The chromosome 2 and chromosome 9 genes have been identified, whereas the chromosome 15 locus remains to be identified. Kanekura and colleagues discovered that the long isoform of alsin/ALS2 specifically binds to mutant—but not to wildtype—SOD1 (Kanekura et al 2004). Expression of the long isoform of alsin/ALS2 protected motor neurons in vitro from mutant SOD1-mediated toxicity. The physical interactions between mutant SOD1 and alsin/ALS2 may link the motor neuron-specific pathways of pathogenesis in these 2 forms of familial amyotrophic lateral sclerosis. The ALS4 locus, mapped to chromosome 9q34, was originally identified in a single large family with autosomal dominant juvenile amyotrophic lateral sclerosis. This family was unusual, because life expectancy was normal, although the clinical criteria were sufficient to diagnose amyotrophic lateral sclerosis. Chen and colleagues identified missense mutations in the senataxin gene in 3 families with autosomal dominant juvenile amyotrophic lateral sclerosis (Chen et al 2004). Each family had a distinct mutation. The exact function of senataxin is not known.

Head trauma. A 2010 study looking at in athletes suffering from chronic traumatic (CTE) made national headlines. This study (McKee et al 2010) evaluated the brain and spinal cord of 12 athletes, all of whom had CTE, and 3 athletes who had CTE and motor neuron disease (MND). The 3 athletes with CTE plus MND had profound muscle weakness, muscle atrophy, spasticity, and diffuse fasciculations. Two of these athletes developed motor symptoms after cognitive and behavioral changes. One developed parkinsonism in addition to dementia, behavioral changes, and MND. Pathologically, these 3 athletes had tau protein and TDP-43 accumulation. When this was compared to sporadic amyotrophic lateral sclerosis controls, the severity and distribution of TDP-43 was distinct from sporadic amyotrophic lateral sclerosis, suggesting that motor neuron disease caused by repetitive head trauma may be pathologically distinct from typical sporadic amyotrophic lateral sclerosis.

Further reports have been conflicting regarding the relationship between head trauma and amyotrophic lateral sclerosis. A case-control study from Italy comparing newly diagnosed amyotrophic lateral sclerosis and healthy controls showed that patients reporting 3 or more traumatic events had higher risk of amyotrophic lateral sclerosis (Pupillo et al 2012). Even if the event occurred 5+ or 10+ years, there was still a strong association between head trauma and risk of amyotrophic lateral sclerosis. In contrast, a report from Rochester, Minnesota, found no increased risk of dementia, Parkinson disease, or amyotrophic lateral sclerosis among 438 football players who played football between 1946 to 1956, when headgear was less protective (Savica et al 2012). Head trauma and its association with amyotrophic lateral sclerosis is still under investigation and is controversial.

Professional athletes. Professional athletes may have an increased risk of developing amyotrophic lateral sclerosis. Many famous professional athletes have developed amyotrophic lateral sclerosis, including Lou Gehrig, the famed New York Yankee who died of the disease at the age of 37. A cohort of 7325 male professional soccer players engaged by a soccer team from the Italian First or Second Division during the period from 1970 to 2001 was studied and evaluated for amyotrophic lateral sclerosis (Chio et al 2005). During the 137,078 person-years follow-up, 5 amyotrophic lateral sclerosis cases were identified (mean age of onset: 43.4 years). Using the standardized morbidity ratio (SMR), there was increased risk for amyotrophic lateral sclerosis onset before 49 years. The findings seem to indicate that playing professional soccer is a strong risk factor for amyotrophic lateral sclerosis. However, the methodology and validity of the conclusion have been questioned (Al-Chalabi and Leigh 2005). A follow-up study indicates that the risk is still present and that it may be soccer-specific as studies have not shown a link between physical activity and the risk for developing amyotrophic lateral sclerosis (Chio et al 2009; Gallo et al 2016). Additionally, the association between head injury, professional athletes, and amyotrophic lateral sclerosis is unclear.

Smoking. Smoking is a probable risk factor for amyotrophic lateral sclerosis. An evidence-based approach evaluating analytic studies of exogenous risk factors for amyotrophic lateral sclerosis published since 1991 found that there was evidence in support of smoking being a probable (“more likely than not”) risk factor for amyotrophic lateral sclerosis (Armon 2003).

Environmental causes. The incidence of amyotrophic lateral sclerosis is 50 to 150 times higher in the Western Pacific than in other areas of the world. There are 3 major foci: (1) the Chamorro people on the islands of Guam, Rota, and Tinian; (2) Japanese villagers on the Kii Peninsula of Honshu Island in Japan; and (3) the people living in West New Guinea (Irian Jaya) of Indonesia (Gajdusek 1982). Western Pacific amyotrophic lateral sclerosis is different from sporadic amyotrophic lateral sclerosis because it is frequently associated with neurofibrillary tangles and parkinsonism-dementia complex. The incidence of the Western Pacific amyotrophic lateral sclerosis has decreased over the last 40 years (Garrutto et al 1985). Genetic factors do not play a role in Western Pacific amyotrophic lateral sclerosis. Environmental factors, including the excitotoxin β-N-methylamino-L-alanine (BMAA) from the cycad seed and mineral imbalances in the soil and water, are being studied as possible causes (Yanagihara et al 1984; Spencer et al 1987; Bradley et al 2013).

A report from northern Italy showed that consumption of drinking water containing one or more mcg/l of inorganic selenium was associated with relative risk for amyotrophic lateral sclerosis of 5.4 (1.1-26, CI 95%) (Vincenti et al 2010). This study was performed in a community reported to have an excess incidence of amyotrophic lateral sclerosis, and further studies are required to determine a causal relationship.

Viral infection. Berger and colleagues detected enterovirus RNA in the spinal cords of patients with amyotrophic lateral sclerosis (Berger et al 2000), but Walker and colleagues could not confirm their report (Walker et al 2001). A small number of patients infected with the human immunodeficiency virus developed a motor neuron disease indistinguishable from amyotrophic lateral sclerosis (Moulignier et al 2001). However, a controlled study revealed no significant elevation of reverse transcriptase in CSF of HIV-negative patients with amyotrophic lateral sclerosis (MacGowan et al 2007), and a controlled pilot study of Indinavir in amyotrophic lateral sclerosis showed significant toxicity along with no effect on progression of disease (Scelsa et al 2005). The potential role of retrovirus has again been raised as Li and colleagues have found that human endogenous retrovirus-K is expressed in cortical and spinal neurons of amyotrophic lateral sclerosis patients, but not in healthy controls (Li et al 2015). More so, they found that animals expressing the envelope protein gene developed progressive symptoms consistent with amyotrophic lateral sclerosis.

Lymphoproliferative diseases. Patients with motor neuron diseases might have higher incidence of lymphoproliferative diseases (Gordon et al 1997). Most of the patients with amyotrophic lateral sclerosis and lymphoproliferative disease had Hodgkin or nonHodgkin , and the rest had myeloma or macroglobulinemia. Among these patients, few had a neurologic response to immunotherapy, and most died of the neurologic disease. Some patients with amyotrophic lateral sclerosis have a monoclonal gammopathy. However, the nature of the association is not known.

Paraneoplastic motor neuron disease. Some patients with cancer and amyotrophic lateral sclerosis tested positive for antineuronal antibody, and the neurologic syndrome disappeared after the removal of the tumor (Khwaja et al 1998; Ferracci et al 1999).

Military service. Studies suggested that military service may be a risk factor for developing amyotrophic lateral sclerosis. Two epidemiologic studies in 2003 (Haley 2003; Horner et al 2003) reported a twofold increase in incidence of amyotrophic lateral sclerosis in Gulf War veterans, particularly in those developing the disease under the age of 45 years. Environmental exposures specific to Gulf War service, such as organophosphate pesticides, chemical nerve agents, or multiple vaccinations with mercury-containing vaccines may have triggered the disease. Weisskopf and colleagues assessed the relationship between United States military service at any time and amyotrophic lateral sclerosis mortality in a large United States cohort enrolled in a nationwide cancer prevention study (Weisskopf et al 2005). More than 500,000 men were followed from 1989 through 1998 and checked for amyotrophic lateral sclerosis mortality using the National Death Index and death certificates. Sixty-three deaths from amyotrophic lateral sclerosis occurred among 126,414 men who did not serve in the military compared to 217 deaths among 281,874 men who did, giving an age and smoking adjusted relative risk of 1.58 (95% CI 1.14 to 2.19). Mortality was higher than expected in virtually all military branches and was independent of the number of years served and the self-reported exposure to a number of lifetime occupational toxins.

In 2008, the United States Department of Veterans' Affairs established amyotrophic lateral sclerosis as a compensable illness. This was based on the 2006 Institute of Medicine (IOM) committee report that concluded that “limited and suggestive” evidence revealed an association between military service and developing amyotrophic lateral sclerosis (Committee on the Review of Scientific Literature on ALS in Veterans 2006).

It is important to note that the National Amyotrophic Lateral Sclerosis (ALS) Registry was initiated by the CDC (Agency for Toxic Substances and Disease Registry) in 2010. It was designed to identify amyotrophic lateral sclerosis cases throughout the United States; patients can self-enroll in the registry. The registry collects critical information about the disease, which will hopefully improve care for people with amyotrophic lateral sclerosis and provide information on what causes the disease, how it can be treated, and even how it can be prevented.

Pathologically, the characteristics of amyotrophic lateral sclerosis include loss of motor neurons in the brain cortex, brainstem, and spinal cord and degeneration of the corticospinal tract. Bunina bodies are characteristic.

We have a limited understanding of the mechanisms of motor neuron injury and cell loss in sporadic amyotrophic lateral sclerosis; however, several hypotheses have been proposed based on the study of the SOD1 transgenic rodent models developed for the familial form of amyotrophic lateral sclerosis (Table 3).

Table 3. Proposed Hypotheses for the Pathogenesis of Amyotrophic Lateral Sclerosis

(1) Glutamate excitotoxicity (2) Free radical-mediated oxidative stress (3) Mitochondrial dysfunction (4) Immune or inflammatory mechanisms (5) Protein aggregation (6) Neurofilament abnormality (7) Peripherin (8) Impaired axonal transport: Dynein and Dynactin (9) Glial cell abnormality (10) Abnormal RNA metabolism

Glutamate excitotoxicity. Glutamate is a prime candidate as a cause of motor neuron excitotoxicity because it is the principal excitatory neurotransmitter in the human motor system, including the corticospinal tract, spinal cord interneurons, and cortical association pathways. Additionally, the concentration of glutamate is about 20,000-fold higher intracellularly than extracellularly. Tightly regulated energy-dependent systems ensure that extracellular glutamate concentrations remain low to prevent cell injury. Therefore, disruption of this steep concentration gradient leads to substantial extracellular accumulation of excitatory amino acids (Rosenberg et al 1992). Furthermore, the proposed mechanism of the only FDA-approved medication to slow the progression, riluzole, is inhibition of glutamatergic transmission.

Free radical-mediated oxidative stress. SOD1 mutations catalyze copper-mediated conversion of hydrogen peroxide to reactive hydrogen radical, promoting oxidative damage (Barber et al 2006). Higher levels of protein carbonyl groups and oxidized nucleic acids in brain homogenates from patients with sporadic amyotrophic lateral sclerosis suggest increased oxidative stress. In addition, fibroblasts from patients with mutant SOD1 appear more sensitive to oxidative stress caused by hydrogen peroxide (Aguirre et al 1998; Barber et al 2006). This has provided the basis for testing antioxidant in amyotrophic lateral sclerosis and developing potential biomarkers of disease.

Mitochondrial dysfunction. Several mechanisms have been proposed that contribute to mutant SOD1-mediated damage to mitochondria. These mechanisms include disruption of energy metabolism, impaired protein import machinery, and impaired calcium buffering (Rothstein 2009). Additionally, there are morphological abnormalities noted in mitochondria of humans with sporadic amyotrophic lateral sclerosis (Manfredi and Beal 2006).

Immune mechanisms. Immune or inflammatory reactions may trigger increased intracellular calcium and motor neuron degeneration (Engelhardt et al 1997; Siklos et al 1998). Evidence for immune mechanisms include a higher incidence of immune disorders in patients (Haverkamp et al 1995), the presence of CD4 and CD8 in the degenerating ventral horn of the spinal cord (Engelhardt et al 1993), the presence of paraproteinemias and (Younger et al 1991), and the presence of IgG within motor neurons (Engelhardt and Appel 1990).

Microglias are immune cells that primarily mediate neuroinflammation within the central nervous system (Kreutzberg 1996). CNS injury activates microglia through the release of cytotoxic and inflammatory substances, such as oxygen radicals, nitrous oxide, glutamate, cytokines, and prostaglandins (Kreutzberg 1996; Hanisch 2002). Activation and proliferation of microglia in regions of motor neuron loss has been observed in amyotrophic lateral sclerosis tissues (Ince et al 1996). Expression of proinflammatory mediators such as TNF-alpha, interleukin-1B, and cyclooxygenase 2 (COX-2) is an early event in mouse models of amyotrophic lateral sclerosis (Alexianu et al 2001; Nguyen et al 2001; Almer et al 2002). Similarly, inhibition of COX-2, a key enzyme in prostaglandin synthesis with celecoxib prolongs survival by 25% in mice with amyotrophic lateral sclerosis (Drachman et al 2002).

Protein aggregation. Intracellular inclusions are a common feature among neurodegenerative diseases. Intracellular, cytoplasmic inclusions have been noted in familial amyotrophic lateral sclerosis mouse models and in both familial and sporadic amyotrophic lateral sclerosis patients (Bruijn and Cudkowicz 2006). Counted among these inclusions are ubiquinated inclusions, Bunina bodies (which are unique to amyotrophic lateral sclerosis), and phosphorylated and nonphosphorylated neurofilament inclusions.

Neurofilament abnormality. Neurofilaments are cytoskeletal elements whose normal production and transport to the termini are critical to the health and integrity of motor neurons. Neurofilament abnormality may play a role in the pathogenesis of amyotrophic lateral sclerosis as neurofilaments accumulate in perikarya and proximal axons (Carpenter 1968). Pathologic features of the disease can be produced in transgenic mice overexpressing various filament protein subunits (Cote et al 1993). In humans, impaired transport of neurofilaments (resulting from a neurofilament gene mutation or an acquired lesion of the neurofilament subunit proteins) may lead to motor neuron degeneration (Wong et al 2000).

Peripherin. Peripherin is an intermediate protein expressed in spinal motor neurons, peripheral sensory neurons, and autonomic nerves. Peripherin is associated with neurofilament proteins in the majority of axonal inclusions in motor neurons of patients with amyotrophic lateral sclerosis (Corbo and Hays 1992). Expression of peripherin 61, a form of peripherin, in primary motor neurons is toxic, even at modest levels (Robertson et al 2003). It is detectable in the lumbar spinal cord of sporadic amyotrophic lateral sclerosis cases but not in nondiseased controls (Robertson et al 2003). Although induced in motor neurons of mutant SOD1 mice, peripherin's role in amyotrophic lateral sclerosis has been called into question because neither elimination of all isoforms by gene deletion nor overexpression of it in an amyotrophic lateral sclerosis model had any effect on survival in SOD1 mutant mouse models (Lariviere et al 2003).

Impaired axonal transport: dynein and dynactin. Motor neurons differ from other neurons by their extreme asymmetry (an axon in the sciatic nerve can be up to 1 meter long) and large volume (up to several thousand times that of a typical cell). Components synthesized in the cell body must be transported into the extended axon via both fast and slow transport system and substances such as trophic factors must be transported back to the cell body via retrograde transport system. Dynein plays an important role in the retrograde transport system. Although cytoplasmic dynein has many cellular roles that include positioning the endoplasmic reticulum and Golgi as well as in assembly of the mitotic spindle, in neurons it is the only known mechanism for retrograde transport. Two dominant point mutations in dynein cause a progressive motor neuron disorder in mice (Hafezparast et al 2003). Similarly, disruption of the dynactin complex, an activator of cytoplasmic dynein, inhibits retrograde axonal transport, provoking a late-onset, progressive motor neuron disease (LaMonte et al 2002). In humans, a dominant point mutation in the p150 subunit of dynactin causes of a lower motor neuron disorder (Puls et al 2003).

Glial cell abnormality. Rao and Weiss proposed a hypothesis that a feed forward cycle involving reciprocal interactions between motor neurons and surrounding astrocytes might play a crucial role in amyotrophic lateral sclerosis pathogenesis (Rao and Weiss 2004). According to this model, damaging reactive oxygen species produced in motor neurons could exit motor neurons and induce oxidative disruption of glutamate transport in surrounding astrocytes. This would exacerbate excitotoxic stress to motor neurons and result in a vicious cycle (Rao and Weiss 2004). This hypothesis suggests that both motor neurons and astrocytes play a crucial role in the pathogenesis of amyotrophic lateral sclerosis. This idea is consistent with the finding that expression of the mutant SOD1 in motor neurons alone did not cause motor neuron disease (Lino et al 2002). Further, studies of mutant SOD1 chimeras indicate the dependence of motor neuron survival on the genotype of nearby nonneuronal cells (Clement et al 2003).

Abnormal RNA metabolism. There have been reports of amyotrophic lateral sclerosis pathogenesis being linked to RNA regulation, specifically associated with the role of microRNA (miRNA) (Williams et al 2009). MicroRNAs are involved with mRNA degradation and associates with both FUS/TLS and TDP-43 (proteins associated with amyotrophic lateral sclerosis) (Strong 2010). Other neurodegenerative diseases, including Alzheimer disease, Parkinson disease, and Huntington disease also implicate miRNA. However, the exact role of the RNA processing defects is still not understood.

Epidemiology"

Amyotrophic lateral sclerosis has a prevalence rate of about 4 per 100,000 and an annual incidence rate of about 1 to 2 per 100,000. The incidence of the disease increases with age, with a peak occurrence between 55 and 75 years of age. It occurs predominantly in men with a male-to-female ratio of about 1.3:1. The average survival period after onset of symptoms is approximately 3 to 5 years. Familial amyotrophic lateral sclerosis comprises about 10% of all cases and most commonly follows an autosomal dominant pattern.

The incidence of amyotrophic lateral sclerosis has been at times 50 to 150 times higher in the Western Pacific than in other areas of the world. There are 3 major foci: (1) the Chamorro people on the islands of Guam, Rota, and Tinian; (2) Japanese villagers on the Kii Peninsula of Honshu Island in Japan; and (3) the people living in West New Guinea (Irian Jaya) of Indonesia (Gajdusek 1982). Western Pacific amyotrophic lateral sclerosis is different from sporadic amyotrophic lateral sclerosis because it is frequently associated with neurofibrillary tangles and parkinsonism- dementia complex. The incidence of the Western Pacific amyotrophic lateral sclerosis has decreased over the last 40 years (Garrutto et al 1985). Genetic factors do not appear to play a role in Western Pacific amyotrophic lateral sclerosis. Environmental factors, including excitotoxin from the cycad seed (β-N-methylamino-L-alanine) and mineral imbalances in the soil and water, are possible causes (Yanagihara et al 1984; Spencer et al 1987).

Prevention

Prevention of amyotrophic lateral sclerosis is unknown.

Differential diagnosis

Table 4. Differential Diagnosis for Amyotrophic Lateral Sclerosis Based on the Symptoms and Signs Bulbar • Myasthenia gravis • Oculopharyngeal dystrophy • Brainstem lesions (tumor, infarction, demyelination) Upper motor neuron • Spondylotic or radiculopathy • Familial spastic paraparesis • HTLV-I associated myelopathy • Subacute combined degeneration of the spinal cord • Adrenoleukodystrophy • Lower motor neuron • Postpolio syndrome • • Benign monomelic • Bulbospinal neuronopathy (Kennedy disease) • Hexosaminidase A deficiency • Multifocal motor neuropathy • Inclusion body myositis

Diseases that have bulbar symptoms and mimic amyotrophic lateral sclerosis.

Myasthenia gravis. Myasthenia gravis mimics amyotrophic lateral sclerosis in that both conditions might have bulbar symptoms and signs, head drop, and respiratory compromise. However, myasthenia gravis patients do not have fasciculation or upper motor neuron signs. Myasthenia gravis patients might have ptosis, ophthalmoplegia, and a positive acetylcholine receptor antibody that are not observed in patients with amyotrophic lateral sclerosis.

Oculopharyngeal muscular dystrophy. Oculopharyngeal muscular dystrophy mimics amyotrophic lateral sclerosis in that they both have bulbar symptoms and signs. However, patients with oculopharyngeal muscular dystrophy have ophthalmoparesis and do not have fasciculation or upper motor neuron findings. Both dominant and recessive oculopharyngeal muscular dystrophies are caused by the expansion of a short GCG triplet repeat coding for polyalanine (Brais et al 1998). Brais and colleagues have proposed the following strict clinical diagnostic criteria for dominant oculopharyngeal muscular dystrophy (Brais et al 1999): (1) a positive family history of oculopharyngeal muscular dystrophy, (2) at least one palpebral fissure at rest smaller than 8 mm (or previous blepharoplasty), and (3) a swallowing time of greater than 7 seconds when asked to drink 80 ml of ice-cold water. DNA testing for oculopharyngeal muscular dystrophy is now available commercially.

Brainstem lesions. Patients who have brainstem lesions such as tumor, , infection or demyelination may present with dysarthria and bilateral pyramidal signs that mimic bulbar symptoms and upper motor neurons of amyotrophic lateral sclerosis. However, an MRI will reveal the pathology in the brainstem, and EMG usually will not show denervation.

Diseases that have upper motor neuron signs and mimic amyotrophic lateral sclerosis.

Spondylotic myelopathy or radiculopathy. Spondylotic myelopathy is caused by the degenerative joint disease at the level of the cervical spine. The major manifestations are causing upper motor neuron signs in the lower extremities, with or without lower motor neuron signs in the upper extremities, caused by cervical root compression. The combination of upper and lower motor neuron signs mimic amyotrophic lateral sclerosis. In some cases, patients present with lower motor neuron findings in the arms and hands without upper motor neuron findings in the lower extremities. An MRI of the cervical spine will confirm the diagnosis of this treatable condition.

Hereditary spastic paraplegia. Most patients with hereditary spastic paraplegia have an autosomal dominant family history, but, occasionally, autosomal recessive inheritance occurs. The disease onset ranges from childhood to late- adult life and progresses slowly as patients develop spastic weakness that begins in the legs. Involvement of the arms is variable (Harding 1981). Distal muscle atrophy is variable and is a late manifestation of the condition. The long history of the condition, absence of bulbar or respiratory involvement, and absence of prominent lower motor neuron findings differentiate hereditary spastic paraplegia from amyotrophic lateral sclerosis;

HTLV-1 associated myelopathy (tropical spastic paraparesis). This chronic myelopathy typically presents during the third or fourth decade of life with slowly progressive spastic paraparesis (Gessain 1992). The condition is acquired through blood transfusion, sexual contact, IV drug abuse, or vertical transfusion. HTLV-1 associated myelopathy is characterized by the presence of upper motor neuron findings including leg weakness, paraplegia with brisk reflexes, and clonus and the absence of lower motor neuron findings including muscle atrophy and fasciculation. The detection of serum and CSF anti-HTLV-1 antibodies establish the diagnosis and differentiate it from amyotrophic lateral sclerosis.

Subacute combined degeneration of the spinal cord. This disorder is one of the manifestations of B12 deficiency. B12 deficiency is associated with megaloblastic anemia, glossitis, dementia, peripheral neuropathy and myelopathy (Beck 1988). Although subacute combined degeneration of the spinal cord shares upper motor neuron signs with amyotrophic lateral sclerosis, the former disease has prominent sensory symptoms. Measurement of serum B12 will establish the diagnosis. In the case of a borderline B12 level, a measurement of serum homocysteine or methylmalonic acid levels is helpful.

Adrenoleukodystrophy. Adrenoleukodystrophy is an X-linked peroxisomal disorder caused by a genetic defect in beta oxidation of saturated very-long-chain fatty acids. The severe form has a disease onset at ages of 4 to 8 years as well as adrenal insufficiency, mental deterioration, and quadriparesis. A relatively mild form, called "adrenomyelodystrophy" occurs in adult men and is characterized by progressive spastic paraparesis, distal muscle weakness, and sensory loss. Because the sensory symptoms may be relatively mild, the upper and lower motor neuron signs of this condition can mimic amyotrophic lateral sclerosis.

Multiple sclerosis. In the classic relapsing and remitting form of multiple sclerosis, the clinical pictures including ocular, sensory, cerebellar, and myelopathic findings have little resemblance to amyotrophic lateral sclerosis. However, in some patients, multiple sclerosis can present as a slowly progressive spastic paraparesis with upper motor neuron weakness. Because these patients may have few sensory disturbances, the diagnosis of amyotrophic lateral sclerosis should be considered. Some multiple sclerosis patients with brainstem involvement may have bulbar findings that mimic amyotrophic lateral sclerosis. The presence of areas of T2 brightening in the MRI of the brain and the spinal cord and the oligoclonal bands in the CSF protein differentiate multiple sclerosis from amyotrophic lateral sclerosis.

Diseases that have lower motor neuron signs and mimic amyotrophic lateral sclerosis. Postpolio syndrome. Postpolio syndrome is a slowly progressive weakness occurring in survivors of the major epidemic of poliomyelitis in the 1950s. The constellation of the neurologic manifestations, including weakness, atrophy, fasciculations, and , is also known as postpoliomyelitis muscular atrophy. Postpoliomyelitis muscular atrophy occurs many years (8 to 71 years) after an acute infection. Weakness occurs insidiously in the previous affected and unaffected muscles with the affected muscles more involved. Postpoliomyelitis muscular atrophy is slowly progressive with strength declining at 1% per year (Dalakas 1986). In EMG studies, postpoliomyelitis muscular atrophy mimics amyotrophic lateral sclerosis in that both have large motor unit potentials of increased duration and reduced interference patterns. However, slowly progressive weakness without upper motor neuron signs in patients with a previous history of poliomyelitis helps to differentiate postpoliomyelitis muscular atrophy from amyotrophic lateral sclerosis.

Spinal muscular atrophy. Current classification of spinal muscular atrophies is based on ages of onset and rates of progression: (1) SMA I (Werdnig-Hoffman disease, acute infantile), (2) SMA II (Intermediate, chronic infantile, arrested), (3) SMA III (Kugelberg-Welander disease, chronic juvenile), and (4) SMA IV (adult onset) (Russman 2002). Neurologists often do not consider SMA I, II, or III in the differential diagnosis of amyotrophic lateral sclerosis, a condition typically associated with the later adult life. However, adult onset SMA IV needs to be considered as a differential diagnosis for amyotrophic lateral sclerosis. The inheritance of SMA IV can be heterogeneous and can be mistaken for familial amyotrophic lateral sclerosis. SMA IV most commonly presents with symmetric, proximal muscle weakness and atrophy and had a slow disease progression. An unusual rapid progression, asymmetry, and upper motor neuron signs are inconsistent with SMA and suggest a diagnosis of amyotrophic lateral sclerosis.

Benign . Benign monomelic amyotrophy is a sporadic disorder most commonly seen in men between 15 and 30 years of age. It presents with focal weakness involving a single limb (Singh 1980). It usually begins with the weakness of the hand intrinsic hand muscles and the C8-T1 myotomes, then spreads over 1 to 2 years to involve forearm flexors and extensors. After the slow progression, the condition usually stabilizes. Respiratory or bulbar muscles are spared. In the early stage, it may be difficult to make a definite diagnosis of benign monomelic amyotrophy. However, EMG testing is helpful because fibrillation potentials and positive waves are sparse in benign monomelic amyotrophy in comparison to classic amyotrophic lateral sclerosis. An MRI of the cervical spine may be useful because it may demonstrate cord atrophy that supports the diagnosis of benign monomelic amyotrophy. Ultimately, the diagnosis of benign monomelic amyotrophy is established when a young man has a stable lower motor neuron weakness restricted to one limb for at least 2 years.

Bulbospinal neuronopathy (Kennedy disease). Bulbospinal neuronopathy is an X-linked disorder that presents with slowly progressive, symmetric, proximal muscle weakness without upper motor neuron findings (Kennedy 1968). More than half of the patients have gynecomastia and infertility associated with muscle atrophy. The genetic basis of this disorder is an unstable CAG trinucleotide repeat in the androgen receptor gene of bulbospinal neuronopathy patients (Spada 1991). Electrodiagnosis demonstrates that both bulbospinal neuronopathy and amyotrophic lateral sclerosis have lower motor neuron findings such as acute denervation (positive waves and fibrillation potentials) and chronic denervation (large motor units, rapid firing, and reduced interference pattern). However, a reduction or absence of sensory nerve action potentials is common in bulbospinal neuronopathy but not in amyotrophic lateral sclerosis. A genetic test for the abnormal CAG repeats confirms the diagnosis of bulbospinal neuronopathy.

Hexosaminidase A deficiency. Hexosaminidase A is a lysosomal enzyme. It degrades GM2 ganglioside, which is highly concentrated in neuronal membrane. The most severe form of hexosaminidase A deficiency results in Tay-Sachs disease. In Tay-Sachs disease, accumulation of GM2 ganglioside in neuronal lysosomes leads to encephalopathy with myoclonic jerk, macular cherry-red spots, and death before 5 years of age. Other mild forms of hexosaminidase A deficiency may begin in infancy or adulthood with syndromes resembling amyotrophic lateral sclerosis and spinomuscular atrophy (Mitsumoto 1985). The spinal muscular atrophy caused by hexosaminidase A deficiency can be differentiated from amyotrophic lateral sclerosis by onset of weakness at a young age, prominent muscle cramp, , dementia, cerebellar atrophy, and sensory nerve involvement. A definite diagnosis is established by markedly reduced hexosaminidase A activity in serum or leukocytes.

Multifocal motor neuropathy with conduction block. Multifocal motor neuropathy with conduction block presents as a slowly progressive, painless, remarkably focal and asymmetric weakness and amyotrophy (Parry 1996). More than half of the patients have elevated titers of antibodies to the GM1 gangliosides. The pure motor involvement of this syndrome mimics the lower motor neuron findings in amyotrophic lateral sclerosis. However, most patients with multifocal motor neuropathy have a long and slowly progressive course. This pattern of progression differs from the rapid progression of amyotrophic lateral sclerosis. It is important to recognize that the disorder often responds to IVIG.

Inclusion body myositis. Inclusion body myositis shares many clinical features with amyotrophic lateral sclerosis such as distal muscle involvement, asymmetric weakness, and difficulty swallowing. However, fasciculation is absent, and there are no upper motor neuron signs. Needle examination demonstrates predominantly myopathic, rather than neurogenic changes in inclusion body myositis. A muscle biopsy to confirm the presence of rimmed vacuoles and intranuclear inclusions establishes the diagnosis of inclusion body myositis (Griggs 1995).

Diagnostic workup

When encountering a patient with the clinical symptoms seemingly indicative of amyotrophic lateral sclerosis, the clinician needs to perform a diagnostic workup to exclude all other possibilities that may cause these clinical symptoms, thus, confirming the diagnosis. Electrodiagnostic tests, neuroimaging of the brain and spinal cord, laboratory studies, and muscle biopsies are useful tools to help the clinician diagnose accurately.

Electrodiagnostic tests. Electrodiagnostic studies are essential for the diagnostic workup in a patient with possible amyotrophic lateral sclerosis. They play important roles in identifying disorders that may resemble amyotrophic lateral sclerosis such as myopathy, neuromuscular transmission disorders, demyelinating polyneuropathy, plexopathy, or radiculopathy. Each of these disorders has typical findings in electrodiagnostic studies (Kimura 2001).

In 1969, Lambert proposed the electrodiagnostic test criteria, which included the following:

(1) Normal sensory nerve conduction studies

(2) Motor nerve conduction velocities, which are normal when recorded from relatively unaffected muscles and not less than 70% of the average normal value when recorded from severely affected muscles

(3) Fibrillation and fasciculation potentials in muscles of the upper and lower extremities or in the muscles of the extremities and the head

(4) Motor unit potentials reduced in number and increased in duration and amplitude (Lambert 1969).

The revised El Escorial criteria for amyotrophic lateral sclerosis require that the needle EMG examination show evidence of active and chronic denervation in at least 2 of 4 regions: (1) brainstem, (2) cervical, (3) thoracic, or (4) lumbosacral. For the brainstem region, needle EMG abnormalities are required in only 1 muscle. For the thoracic region, abnormalities can be in the thoracic paraspinal muscles below the T6 level or in the abdominal muscles. For the cervical and lumbosacral paraspinal regions, abnormalities must be present in more than 2 muscles innervated by 2 different nerve roots and peripheral nerves.

Needle examination of the sternocleidomastoid muscles carries a similar sensitivity as examination of the tongue in patients with bulbar symptoms (Li et al 2002).

Neuroimaging of the brain and spinal cord. Neuroimaging is useful in excluding disorders that have symptoms that mimic amyotrophic lateral sclerosis. For example, bulbar palsy may be seen in patients with brainstem glioma, brainstem stroke, multiple sclerosis, and foramen magnum tumor. In these instances, an MRI of the brainstem will show abnormalities. Neoplastic polyradiculopathy, which has abnormal meningeal enhancement in an MRI, may present with clinical and electrodiagnostic test features that mimic progressive muscular atrophy. Spinal cord tumors, transverse , and spondylotic myelopathy may have upper motor neuron findings that mimic primary lateral sclerosis. MRI of the spinal cord proves useful in differentiating these disorders.

Clinical laboratory testing. Laboratory studies commonly obtained include the following: complete blood count, erythrocyte sedimentation rate, blood chemistry, creatine phosphokinase level, thyroid function test, parathyroid function test, serum rapid plasma reagin test, B12 level, antinuclear antibody test, RA, and serum immunofixation electrophoresis. These studies provide a basic screening for the patient's general health. In addition, special tests may be ordered to exclude amyotrophic lateral sclerosis-related syndromes. These syndromes have unique laboratory- defined characteristics that are linked to the development of the phenotype. These include amyotrophic lateral sclerosis-related syndromes associated with paraproteinemia, anti-GM1 antibodies, lymphoma, endocrinopathies, infections, toxins, ischemia, and cervical spondylosis. The motor neuron disease associated with anti-GM1 antibodies may have pure lower motor neuron or amyotrophic lateral sclerosis features associated with anti-GM1 antibodies. The amyotrophic lateral sclerosis-related syndrome associated with lymphoma presents with lymphadenopathy (check chest x-ray), monoclonal gammopathy (check immunofixation electrophoresis), increased CSF protein (perform lumbar puncture), and abnormal bone marrow biopsy. If a patient is younger than 40 years of age, the possibility of hexosaminidase A deficiency must be considered. Creatine phosphokinase levels are normal to mildly elevated (less than 2000) in amyotrophic lateral sclerosis (Gibson et al 2015). Although many order heavy metal screenings for mercury, lead, and arsenic in blood and urine, there has not been a convincing report of heavy metal- induced motor neuron disease for 25 years (Rowland and Shneider 2001).

Muscle biopsy. Muscle biopsy becomes useful when a confirmation of the neurogenic changes caused by the lower motor neuron degeneration and exclusion of myopathic diseases, such as inclusion body myositis, is necessary.

Management

Because there is no effective treatment for amyotrophic lateral sclerosis, aggressive symptom management and comprehensive palliative or end-of-life care are essential for patients and their families (Mitsumoto and Rabkin 2007). In October 2009, the American Academy of Neurology published a 2-part, evidence-based practice parameter update about care of patients with amyotrophic lateral sclerosis (Miller et al 2009a; Miller et al 2009b), updating the previous evidence-based practice parameter published in 1999 (Miller et al 1999b). Highlights of the update included evidence supporting the use of noninvasive ventilation, percutaneous endoscopic gastrostomy, and riluzole for extending survival or slowing disease progression. The parameter also recommended that amyotrophic lateral sclerosis patients should be managed at a multidisciplinary clinic to optimize health care delivery, prolong survival, and improve quality of life. With regards to symptomatic treatment, botulinum toxin B and low-dose radiation may be considered for refractory sialorrhea. Medication treatments for sialorrhea are usually tried first, and those who do not respond to these medications are then considered refractory. Lastly, dextromethorphan and quinidine should be considered for pseudobulbar affect. This drug manufactured by Avanir (Nuedexta™) is approved for use by the United States Food and Drug Administration based on its clinical trial (Pioro et al 2010). Of note, these practice parameters were published prior to the U.S. Food and Drug Administration approval of edaravone.

Disease-modifying therapy.

Riluzole. Use of riluzole is based on the hypothesis that excitotoxic damage to motor neurons plays a role in the disease as well as observations that riluzole inhibits the presynaptic release of glutamate and neuronal damage in a number of experimental models. The first Cochrane review of 3 randomized controlled trials (Bensimon et al 1994; Bensimon et al 2002; Lacomblez et al 1996) using riluzole to treat patients with amyotrophic lateral sclerosis was published (Miller et al 2002). The review reported a 9% gain in the probability of surviving 1 year (57% in the placebo and 66% in the riluzole group). There was a significant survival advantage with riluzole 100 mg at 6, 9, 12, and 15 months but not at 3 or 18 months. Although muscle strength was not better in the riluzole group, there was a small beneficial effect on both bulbar and limb function. Patients treated with riluzole remained in a more moderately affected health state significantly longer than placebo-treated patients.

The Cochrane review concluded that riluzole 100 mg per day appears to be reasonably safe and probably prolongs survival by about 2 months in patients with amyotrophic lateral sclerosis, although more studies are needed to clarify its effect in older (over 75 years) and more advanced patients. The standard dose for riluzole is 50 mg twice daily 1 hour before or 2 hours after a meal. Side effects of riluzole include fatigue, nausea, vomiting, vertigo, somnolence, and elevated liver function tests. A small number of patients in the riluzole trials developed liver toxicity (defined as LFTs > 5x normal) and severe neutropenia. As a result, serum transaminase levels and complete blood counts should be monitored monthly for the first 3 months of riluzole treatment and then at 3-month intervals. The Quality Standards Subcommittee of the American Academy of Neurology issued a drug advisory (Anonymous 1997) recommending that the drug should be offered to patients.

Edaravone. Endaravone is hypothesized to work as an oxygen free radical scavenger in the central nervous system. In mouse models of amyotrophic lateral sclerosis, it has been demonstrated to prolong survival and to reduce nitration of tyrosine residues in the (Ito et al 2008). In a phase II open-label trial, edaravone appeared safe and effective, reducing 3-nitrotyrosine levels in the cerebrospinal fluid (Abe et al 2014). It has completed 2 randomized controlled phase 3 trials (Sawada 2017). The first phase 3 trial was not significant. The second and final phase 3 trials, which restricted enrollment to amyotrophic lateral sclerosis patients with a relatively short disease duration and preserved vital capacity, demonstrated efficacy in ALSFRS-R scores over 24 weeks.

Endaravone was approved by the U.S. Food and Drug Administration for the treatment of amyotrophic lateral sclerosis in May 2017, although it was previously approved for use in amyotrophic lateral sclerosis in Japan as well as in South Korea. This medication is delivered intravenously the first 14 days of the month, followed by infusions 10 of the first 14 days of the subsequent months. Common reactions include contusions, gait disturbance, and .

Nutritional management: percutaneous endoscopic gastrostomy. Progressive dysphagia and impaired food and fluid intake will develop in patients with amyotrophic lateral sclerosis who have bulbar symptoms. Bulbar muscles are initially affected in approximately 25% of patients. Eventually, almost all patients develop bulbar symptoms. Severe dysphagia results in malnutrition and weight loss, which further worsens muscle weakness and atrophy in patients. Malnutrition increased the relative risk of death 7.7-fold in patients (Desport et al 1999).

The initial AAN Practice Parameter Subcommittee made evidence-based recommendations for nutritional management (Miller et al 1999b):

(1) Percutaneous endoscopic gastrostomy (PEG) is indicated for patients who have symptomatic dysphagia and should be considered soon after symptom onset.

(2) For optimal safety and efficacy, a PEG should be offered and placed when the patient's forced vital capacity is more than 50% of the predicted value. It should be noted that the outcome could be acceptable in some patients even when the forced vital capacity is less than 50%; however, patients with forced vital capacity of less than 50% who undergo PEG may develop more serious pulmonary complications. These points are reiterated in the updated practice parameter in 2009, which adds that a radiologically inserted device (ie, RIG) can be an alternative to PEG, that enteral nutrition helps to stabilize weight, and that PEG is probably effective in prolonging survival. It is important to emphasize that PEG does not eliminate oral feeding.

Respiratory management: noninvasive positive pressure ventilation. Most patients will eventually develop respiratory insufficiency. Early symptoms of respiratory insufficiency include dyspnea on exertion, marked fatigue, supine dyspnea, disturbed sleep, and morning . Nocturnal oximetry is helpful for the detection of nocturnal hypoventilation, and vital capacity should be monitored at 2- to 3-month intervals in patients. Some patients are symptomatic of respiratory insufficiency well before the vital capacity falls to 50% of the predicted value. The practice parameters concluded that nocturnal oximetry and maximal inspiratory pressure (MIP) are more effective than erect forced vital capacity (FVC) in detecting early respiratory insufficiency and that supine FVC is more effective than erect FVC in detecting diaphragm weakness (Miller et al 2009a; Miller et al 2009b).

Noninvasive positive pressure ventilation offers direct clinical benefit. Noninvasive positive pressure ventilation improves respiratory symptoms and quality of life as well as survival (Kleopa et al 1999; Lechtzin et al 2007) and cognitive dysfunction (Newsom-Davis et al 2001). Although tracheostomy and invasive ventilation may increase survival more effectively, the greater financial and care burden must be thoroughly discussed with patients and families (Moss et al 1993). However, long-term mechanical ventilation may be acceptable to some patients, and it enables these patients to lead meaningful lives without regrets about being ventilator-dependent (Moss et al 1996).

A preliminary study suggested that mechanical insufflation-exsufflation can generate clinically effective expiratory flows for effective cough and airway clearance in patients with non-bulbar amyotrophic lateral sclerosis but not in those with bulbar symptoms (Sancho et al 2004).

In 2011, the Food and Drug Administration approved the diaphragm pacing device in patients with amyotrophic lateral sclerosis. The device was approved under the Humanitarian Device Exemption (HDE). To further understand the utility of this device in amyotrophic lateral sclerosis patients, 3 studies were started in the following locations: the U.S., UK, and France. Both the UK and French study have closed early due to safety concerns (McDermott et al 2015).

Communication management. Augmentative and alternative communication devices should be prescribed for patients with severe dysarthria to enable and preserve social closeness and discussion of important issues (Fried-Oken et al 2006). In the last 2 years, there has been growth of development of brain computer interfaces (BCI) for amyotrophic lateral sclerosis. This has major significance for amyotrophic lateral sclerosis because BCI can substitute for loss of communication due to muscle loss and speech loss. BCI allows people with amyotrophic lateral sclerosis to interact though brain signals rather than muscles (Mak et al 2012). BCI measures brain activity and converts it to a computerized output. Theoretically, a “locked in” patient would be able to communicate purely with brain activity and no movement. It is a simple, non-invasive device and is currently in development for amyotrophic lateral sclerosis patients in the Veteran's Administration System.

Exercise. A small study showed that resistance exercise might improve functional level and quality of life in patients with amyotrophic lateral sclerosis (Bello-Haas et al 2007).

Symptomatic treatments. Sialorrhea can be treated with tricyclics such as amitriptyline, doxepin, imipramine, or a small portable suction device. Other options include glycopyrrolate, benztropine, or transdermal scopolamine. Botulinum toxin injected directly into the salivary gland offers another option (Giess et al 2000), especially for those with refractory sialorrhea. Low-dose radiation of the salivary gland tissue can also safely help control secretions in selected patients (Harriman et al 2001).

Emotional liability, such as uncontrollable paroxysms of laughing or crying, can be treated with dextromethorphan/quinidine, tricyclics, fluvoxamine, or a selective serotonin reuptake inhibitor.

Muscle may respond to a benzodiazepine such as diazepam. Fasciculations can be treated with a high-potency, short-duration benzodiazepine, such as lorazepam. Spasticity may be managed with diazepam, baclofen, or dantrolene.

Pain may be a troubling symptom to many patients. , nonsteroidal anti-inflammatory drugs, anticonvulsants, and tricyclic antidepressants, used alone or in combination, are useful in alleviating pain.

Depression affects about 50% of patients, thus, affecting quality of life (Boynton De Sepulveda 2002; Lou et al 2003). Using DSM-IV criteria, Rabkin and colleagues evaluated 80 patients with late-stage amyotrophic lateral sclerosis and found that only 10% had minor depression, 9% had symptoms consistent with major depression, and 81% had no depressive disorders at baseline (Rabkin et al 2005). They reported that 57% of patients never had a depression diagnosis at any visit, implying that 43% of them had a diagnosis of major depression in at least 1 of the visits. Additionally, of 66 patients who filled out the 21-item Becker Depression Inventory at baseline, 62% had scores in the depressive range. Treatment of depression was initiated in 15 patients after the diagnosis of amyotrophic lateral sclerosis was made. In another cross-sectional study of 25 patients with amyotrophic lateral sclerosis at different stages, 44% of the patients scored in the depressive range using CES-D (Center of Epidemiological Study-Depression), and depression was associated with poor quality of life (Lou et al 2003). Because amyotrophic lateral sclerosis is a devastating disease, depressive symptoms are common and associated with poor quality of life in patients with amyotrophic lateral sclerosis. Clinicians need to be sensitive regarding depressive symptoms in these patients and treat them aggressively.

Fatigue is also a common complaint in patients with amyotrophic lateral sclerosis (Heiman-Patterson et al 2001; Lou et al 2003). Amantadine is used widely in multiple sclerosis patients with fatigue, and it should be tried in patients with amyotrophic lateral sclerosis who are complaining of severe fatigue as well.

Table 5. Symptomatic Treatment in Amyotrophic Lateral Sclerosis Symptoms Treatment Fatigue • Modafinil 100 to 300 mg daily • Amantadine, 100 mg twice daily • Methylphenidate, 5 to 10 mg twice daily * Excessive daytime fatigue can be due to nocturnal hypoventilation, and a sleep study is indicated. Cramps • Quinine sulfate, 325 mg twice daily Spasticity • Lioresal, 10 to 20 mg 4 times daily • Dantrolene, 25 mg 3 times daily • Tizanidine, 4 mg 3 times daily • Clonazepam, 0.5 mg 3 times daily • Lorazepam, 0.5 to 1 mg 3 times daily • Diazepam, 2.5 to 5 mg 3 times daily • Phenytoin, 300 mg 4 times daily * These medications may increase weakness and decrease pulmonary function.

Jaw quivering and • Clonazepam, 0.5 mg 3 times daily clenching • Lorazepam, 0.5 to 1 mg 3 times daily • Diazepam, 2.5 to 5 mg 3 times daily Pseudobulbar affect • Dextromethorphan/Quinidine (20 mg/10 mg) 2 times (excessive crying and daily laughter) • Amitriptyline, 10 to 75 mg 4 times daily • Fluoxetine, 10 to 40 mg 4 times daily • Sertraline, 50 to 100 mg at bedtime • Paroxetine, 10 to 40 mg 4 times daily Laryngospasm • Clonazepam, 0.5 mg as needed • Lorazepam, 0.5 to 1mg as needed • Diazepam, 2.5 to 5 mg as needed * Laryngospasm is often triggered by aspiration, cough or gastroesophageal reflexes. Dry mouth and thick • Guaifenesin, 300 mg every 4 hours as needed phlegm • Propranolol 10 to 40 mg, 3 times daily • Nebulized acetylcysteine 10% solution, 2 ml twice daily as needed*consider manually assisted coughing and mechanical insufflation-exsufflation * Dry mouth and thick phlegm is often caused by medications for drooling and depression. Nasal congestion • Pseudoephedrine, 30 to 60 mg, 3 times daily • Diphenhydramine, 25 to 50 mg twice daily • Budesonide nasal spray, 2 sprays twice daily Sleep disturbance • Amitriptyline, 10 to 75 mg every night • Trazodone, 50 mg every night

Pain • Acetaminophen, 500 mg, 3 times daily • NASID • Morphine sulfate, 5 to 10 mg twice daily Depression or anxiety • Amitriptyline, 10 to 75 mg daily • Fluoxetine, 10 to 40 mg daily • Sertraline, 50 to 100 mg at bedtime • Paroxetine, 10 to 40 mg daily Sialorrhea • Glycopyrrolate, 1 to 2 mg 4 times daily • Amitriptyline, 10 to 25 mg, 3 times daily • Diphenhydramine, 25 mg every 8 hours • Oxybutynin, 5 mg 3 times daily • Scopolamine, 0.125 mg, 3 times daily • Trihexyphenidyl, 2 mg twice daily * Botulinum toxin injection and parotid or submandibular glands irradiation may reduce saliva secretion. ** A portable suction device is also useful. Terminal management • Lorazepam, 1 to 3 mg, 3 times daily • Morphine sulfate elixir 20 mg/ml, 0.5 to 2 mg • Morphine SC pump at 1 to 5 mg/hr Agitation or anxiety • Lorazepam, 1 to 3 mg twice daily as needed • Diazepam, 25 to 10 mg twice daily as needed

Human clinical trials. A number of medications with a variety of proposed mechanisms have delayed disease progression in amyotrophic lateral sclerosis animal models, but have failed to show efficacy in human clinical trials (Table 6). Several drugs are still currently in clinical trials or have completed enrollment, but have not released results (www.clinicaltrials.gov). The failure to translate from animals to humans is at least in part due to inherent limitations when using animal models to study human diseases.

Table 6. Medications Tested in Phase III Clinical Trials with No Effect on Human Survival Drugs Proposed Mechanisms References Phase Brain-derived Growth factor (BDNF Study Group III neurotrophic factor 1999) Ceftriaxone Reduction of production and (Cudkowicz et al III increase breakdown of 2014) glutamate Celecoxib Reduction of glutamate (Cudkowicz et al III release and free radicals, 2006) anti-inflammatory Dexpramipexole Reduces apoptosis (Cudkowicz et al III 2013) Minocycline Anti-inflammatory or anti- (Gordon et al 2007) III apoptotic IGF-1 Neurotrophic factor (Sorensen et al 2008) III THC346 Anti-apoptotic (Miller et al 2007) III Gabapentin Glutamate synthesis inhibitor (Miller et al 2001) III Dextromethorphan NMDA receptor inhibitor (Gredal et al 1997) III Branched-chain amino Increased glutamate (Tandan et al 1996) III acids (valine, leucine, dehydrogenase level isoleucine) Verapamil Calcium channel blocker (Miller et al 1996a) III Nimodipine Calcium channel blocker (Miller et al 1996b) III N-acetylcysteine Free radical scavenger (Louwerse et al 1995) III Human ciliary Neurotrophic factor (ALS CNTF III neurotrophic factor Treatment Study (CNTF) Group 1996) Prednisone Immunosuppression (Tan et al 1994) III Cyclophosphamide Immunosuppression (Smith et al 1994) III Total body irradiation Immunosuppression (Drachman et al III 1994) Deprenyl Free radical scavenger (Jossan et al 1994) III Cyclosporine Immunosuppression (Appel et al 1988) III

Alternative medicine. In Germany, about half of the patients with amyotrophic lateral sclerosis use (Wasner et al 2001). The most widely used methods are acupuncture, homeopathy, naturopathy, and esoteric treatments. None of these treatments have been proven to be effective. Use of cannabinoids are also increasingly common, although human clinical trials to support use beyond symptomatic treatment are lacking (Giacoppo and Mazzon 2016). Because the use of alternative medicine is so widespread, physicians need to become more familiar with alternative and educate patients about potential complications.

In the United States, Carter and Bromberg surveyed 53 patients with amyotrophic lateral sclerosis in the University of Utah Motor Neuron Disease Clinic regarding their use of vitamins, herbal supplements and other compounds (Carter and Bromberg 2002). They found that 70% were taking vitamins and 42% were taking herbal supplements. Subjects spent to per month for herbal medicines and vitamins. Because the use of alternative medicine is so widespread in patients with amyotrophic lateral sclerosis, physicians need to become more familiar with alternative medicines and warn the patients of potential complications.

More than 79% of patients with amyotrophic lateral sclerosis take vitamins or nutritional supplements (Miller et al 2009a; Miller et al 2009b). Of the variety of supplements, coenzyme Q10 (COQ10), creatine, and vitamin E have been evaluated for efficacy. High-dose COQ10 was evaluated in a phase II trial and was ineffective in slowing disease progression (Kaufmann et al 2009). Creatine at 10 g/day and 5 g/day failed to alter survival or the rate of functional decline in amyotrophic lateral sclerosis (Groeneveld et al 2003; Shefner et al 2004). Vitamin E 5000 mg/day was ineffective in improving survival or functional outcome, but 1000 mg/day was marginally effective. The current practice parameter concluded that high-dose vitamin E should not be considered as treatment for amyotrophic lateral sclerosis (Miller et al 2009a; Miller et al 2009b) whereas the equivocal evidence for low-dose vitamin permitted no recommendation.

Stem cells. Stem cell therapy is challenging, particularly in amyotrophic lateral sclerosis because of the length of the motor axons and the growing evidence that neurodegeneration in amyotrophic lateral sclerosis may be mediated by neuronal and glial influences. Stem cell studies in rodent models have shown that late-stage fetal cortical neurons could replace apoptotic neurons and fetal motor neurons could migrate and make connections with skeletal muscle (Fricker-Gates et al 2002). There have been 2 phase II stem cell clinical trials in patients with amyotrophic lateral sclerosis. One study delivered autologous mesenchymal stem cells that secrete neurotrophic growth factors intrathecally and intramuscularly (Petrou et al 2016). This study appeared safe, and there was a trend towards clinical benefits. The second study injected neural-derived stem cells (from pluripotent embryonic stem cells) directly into the grey matter of the spinal cord in conjunction with treatment with immunosuppressant medications and appeared generally safe, but no difference in the mean rate of symptom progression was identified in comparison to historical control groups (Glass et al 2016).

Special considerations

Pregnancy

There is no particular risk in pregnancy associated with amyotrophic lateral sclerosis. The respiratory dysfunction and loss of muscle strength may impair labor and delivery, thus, requiring special attention.

Anesthesia

The major risk associated regarding anesthesia is respiratory dysfunction and severity of bulbar involvement.

References cited

Abe K, Itoyama Y, Sobue G, et al. Confirmatory double-blind, parallel-group, placebo-controlled study of efficacy and safety of edaravone (MCI-186) in amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Frontotemporal Degener 2014;15(7-8):610-7. PMID 25286015

Aguirre T, Van Den Bosch L, Goetschalckx K, et al. Increased sensitivity of fibroblasts from amyotrophic lateral sclerosis patients to oxidative stress. Ann Neurol 1998;43:452-7. PMID 9546325

Al-Chalabi A, Fang F, Hanby MF, et al. An estimate of amyotrophic lateral sclerosis heritability using twin data. J Neurol Neurosurg 2010;81(12):1324-6. PMID 20861059

Al-Chalabi A, Leigh PN. Trouble on the pitch: are professional football players at increased risk of developing amyotrophic lateral sclerosis. Brain 2005;128:451-3. PMID 15713848

Alexianu ME, Kozovska M, Appel SH. Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology 2001;57:1282-89. PMID 11591849 Almer G, Teismann P, Stevic Z, et al. Increased levels of the pro-inflammatory prostaglandin PGE2 in CSF from ALS patients. Neurology 2002;58:1277-9. PMID 11971099

ALS CNTF Treatment Study Group. A double-blind placebo-controlled clinical trial of subcutaneous recombinant human ciliary neurotrophic factor (rHCNTF) in amyotrophic lateral sclerosis. Neurology 1996;46(5):1244-9. PMID 8628460

Andersen P, Nilsson P, Keranen M, et al. Phenotypic heterogeneity in motor neuron disease patients with CuZn- superoxide dismutase mutations in Scandinavia. Brain 1997120,1723-37. PMID 9365366

Andersen P, Sims KB, Xin WW, et al. Sixteen novel mutations in the Cu/Zn superoxide dismutase gene in amyotrophic lateral sclerosis: a decade of discoveries, defects and disputes. Amyotroph Lateral Scler Other Motor Neuron Disord 2003;4:62-73. PMID 14506936

Anonymous. Practice advisory on the treatment of amyotrophic lateral sclerosis with riluzole: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 1997;49:657-9. PMID 9305318

Appel SH, Stewart SS, Appel V, et al. A double-blind study of the effectiveness of cyclosporine in amyotrophic lateral sclerosis. Arch Neurol 1988;45:381-6. PMID 3281637

Aran FA. Recherches sur une maladie non “ncore decrite du systeme musculaire (atro- hie musculaire progressive). Arch Gen Med 1850;24:5-35,172-214.

Arlandis S, Vazquez-Costa JF, Martinez-Cuenca E, Sevilla T, Boronat F, Broseta E. Urodynamic findings in amyotrophic lateral sclerosis patients with lower urinary tract symptoms: results from a pilot study. Neurourol Urodyn 2017;38(3):626-31. PMID 26895486

Armon C. An evidence-based medicine approach to the evaluation of the role of exogenous risk factors in sporadic amyotrophic lateral sclerosis. Neuroepidemiology 2003;22(4):217-28. PMID 12792141

Barber S, Mead R, Shaw P. Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim Biophys Acta 2006;1762:1051-67. PMID 16713195

BDNF Study Group. A controlled trial of recombinant methionyl human BDNF in ALS: The BDNF Study Group (Phase III). Neurology 1999;52(7):1427-33. PMID 10227630

Beck WS. Cobalamin and the nervous system. N Eng J Med 1988;318(26):1752-4. PMID 3374546

Bello-Haas VD, Florence JM, Kloos AD. A randomized controlled trial of resistance exercise in individuals with ALS. Neurology 2007;68:2003-7. PMID 17548549

Bensimon G, Lacomblez L, Delumeau JC, Bejuit R, Truffinet P, Meininger V; Riluzole/ALS Study Group II. A study of riluzole in the treatment of advanced stage or elderly patients with amyotrophic lateral sclerosis. J Neurol 2002;249(5):609-15. PMID 12021952

Bensimon G, Lacomblez L, Mekiinger V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group (see comments). N Engl J Med 1994;330:585-91. PMID 8302340

Berger MM, Kopp N, Vital C, et al. Detection and cellular localization of enterovirus RNA sequences in spinal cord of patients with ALS. Neurology 2000;54:20-5. PMID 10636120

Boynton De Sepulveda L. Identification of psychological well-being and depression in an ALS population and factors influencing its presentation. The ALS C.A.R.E. Program Notes, February 2002.

Bradley WG, Borenstein AR, Nelson LM, et al. Is exposure to cyanobacteria an environmental risk factor for amyotrophic lateral sclerosis and other neurodegenerative diseases? Amyotroph Lateral Scler Frontotemporal Degener 2013;14(5-6):325-33. PMID 23286757

Brain WR. Diseases of the nervous system. 6th ed. Oxford: Oxford University Press, 1962.

Brais B, Bouchard J, Xie YG, et al. Short GCG expansions in the PABP2 gene cause oculopharyngeal muscular dystrophy. Nat Genet 1998;18:164-7. PMID 9462747

Brais B, Rouleau GA, Bouchard JP, et al. Oculopharyngeal muscular dystrophy. Semin Neurol 1999;19:59-66. PMID 10711989

Brooks BR. El Escorial World Federation of Neurology criteria for the diagnosis of amyotrophic lateral sclerosis. J Neurol Sci 1994;124:96-107. PMID 7807156

Brooks BR, Sufit RL, DePaul R, et al. Design of clinical therapeutic trials in amyotrophic lateral sclerosis. Adv Neurol 1991;56:521-46. PMID 1853781

Bruijn LI, Cudkowicz M. Therapeutic targets for amyotrophic lateral sclerosis: current treatments and prospects for more effective therapies. Expert Rev Neurother 2006;6(3):417-28. PMID 16533145

Carpenter S. Proximal axonal enlargement in motor neuron disease. Neurology 1968,18:841-51. PMID 4176657

Carter O, Bromberg M. The use of herbal supplements and vitamins by patients with amyotrophic lateral sclerosis. ALS 2002;3(suppl 2):119.

Charcot JM, Joffroy A. Deux cas d'atrophie musculaire progressive avec lesions de la substance grise et des faisceaux antero-lateraux de la moelle epiniere. Arch Physiol Neurol Pathol 1969;2:744.

Chen YZ, Bennet CL, et al. DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4). Am J Hum Genet 2004;74:1128-35. PMID 15106121

Chio A, Benzi G, Dossena M, Mutani R, Mora G. Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players. Brain 2005;128:472-6. PMID 15634730

Chio A, Calvo A, Dossena M, Ghiglione P, Mutani R, Mora G. ALS in Italian professional soccer players: the risk is still present and could be soccer-specific. Amyotroph Lateral Scler 2009;10(4):205-9. PMID 19267274

Clement AM, Nguyen MD, Roberts EA, et al. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 2003;302:113-7. PMID 14526083

Committee on the Review of Scientific Literature on Amyotrophic Lateral Sclerosis in Veterans. Amyotrophic Lateral Sclerosis in Veterans: Review of the Scientific Literature. National Academies Press, 2006. Available at: http://www.nap.edu/openbook.php.recordid=11757

Corbo M, HaysAP. Peripherin and neuro-filament protein coexist in spinal spheroids of motor neuron disease. J Neuropathol Exp Neurol 1992;51:531-7. PMID 1381416

Cote F, Collard JF, Julien JP. Progressing neuronopathy in transgenic mice expressing the human neurofilament heavy gene: a mouse mode1 of amyotrophic lateral sclerosis. Cell 1993,73:35-46. PMID 8462101

Cruveilhier J. Sur la paralysie musculaire, progressive, atrophique. Bull Acad Med (Paris) 1852-1853;18:490-546.

Cudkowicz ME, Shefner JM, Schoenfeld DA, et al. Trial of celecoxib in amyotrophic lateral sclerosis. Ann Neurol 2006;60(1):22-31. PMID 16802291

Cudkowicz ME, Titus S, Kearney M, et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi- stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol 2014;13(11):1083-91. PMID 5297012

Cudkowicz ME, van den Berg LH, Shefner JM, et al. Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial. Lancet Neurol 2013;12(11):1059-67. PMID 24067398

Dalakas M, Elder G, Hallett M, et al. A long-term follow-up study of patients with postpoliomyelitis neuromuscular symptoms. N Eng J Med 1986;314(15):959-63. PMID 3007983 de Carvalho M, Dengler R, Eisen A, et al. Electrodiagnostic criteria for diagnosis of ALS. Clinical Neurophysiol 2008;119(3):497-503. PMID 18164242

Del Bo R, Ghezzi S, Corti S, et al. TARDBP (TDP-43) sequence analysis in patients with familial and sporadic amyotrophic lateral sclerosis: identification of two novel mutations. Eur J Neurol 2009;16(6):727-32. PMID 19236453

Deng HX, Chen W, Hong ST, et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 2011;477(7363):211-5. PMID 21857683

Desport JC, Preux PM, Tmong TC, et al. Nutritional status is a prognostic factor for survival in ALS patients. Neurology 1999;53:1059-63. PMID 10496266

Drachman DB, Chaudhry V, Cornblath D, et al. Trial of immunosuppression in amyotrophic lateral sclerosis using total lymphoid irradiation [see comments]. Ann Neurol 1994;35:142-50. PMID 8109895

Drachman DB, Frank K, Dykes-Hoberg M, et al. Cyclooxygenase 2 inhibition protects motor neurons and prolongs survival in a transgenic mouse model of ALS. Ann Neurol 2002;52:771-8. PMID 12447931

Engelhardt JI, Appel SH. IgG reactivity in the spinal cord and motor cortex in amyotrophic sclerosis. Arch Neurol 1990;47:1210-6. PMID 2122877

Engelhardt JI, Siklos L, Appel SH. Altered calcium homeostasis and ultrastructure in motoneurons of mice caused by passively transferred anti-motoneuronal IgG. J Neuropath Exp Neurol 1997;56:21-39. PMID 8990126

Engelhardt JI, Tajti J, Appel SH. Lymphocytic infiltrates in the spinal cord in amyotrophic lateral sclerosis. Arch Neurol 1993;50:30-6. PMID 8093428

Ferracci F, Fassetta G, Butler MH, Floyd S, Solimena M, De Camilli P. A novel antineuronal antibody in a motor neuron syndrome associated with . Neurology 1999;53:852-5. PMID 10489053

Fricker-Gates RA, Shin JJ, Tai CC, et al. Late stage immature neocortical neurons reconstruct interhemispheric connections and form synaptic contacts with increased efficiency in adult mouse cortex undergoing targeted neurodegeneration. J Neurosci 2002;22:4045-56. PMID 12019324

Fried-Oken M, Fox L, Rau MT, et al. Purposes of AAC device use for persons with ALS as reported by caregivers. Augment Altern Commun 2006;22(3):209-21. PMID 17114164

Gajdusek DC. Foci of motor neuron disease in high incidence in isolated populations of East Asia and the Western Pacific. In: Rowland LP, editor. Human motor neuron diseases. New York: Raven Press, 1982:363-93.

Gallo V, Vanacore N, Bueno-de-Mesquita HB, et al. Physical activity and risk of amyotrophic lateral sclerosis in a prospective cohort study. Eur J Epidemiol 2016;31(3):255-66. PMID 26968841

Garrutto RM, Yanagihara R, Gajdusek DC. Disappearance of high-incidence amyotrophic lateral sclerosis and parkinsonism-dementia on Guam. Neurology 1985;35(2):193-8. PMID 3969206

Gastaut JL, Bartolomei F. Mills' syndrome: ascending (or descending) progressive hemiplegia: a hemiplegic form of primary lateral sclerosis. J Neurol Neurosurg Psychiatry 1994;57:1280-1. PMID 7931406

Gessain A, Gout O. Chronic myelopathy associated with human-T lymphotrophic virus type 1 (HTLV-1). Ann Int Med 1992;117:933-46. PMID 1443956

Giacoppo S, Mazzon E. Can cannabinoids be a potential therapeutic tool in amyotrophic lateral sclerosis? Neural Regen Res 2016;11(12):1896-9. PMID 28197175

Gibson SB, Kasarskis EJ, Hu N, et al. Relationship of creatine kinase to body composition, disease state, and longevity in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2015;16(7-8):473-7. PMID 26312548

Giess R, Naumann M, Werner E, et al. Injections of botulinum toxin A into the salivary glands improve sialorrhea in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2000;69:121-3. PMID 10864618 Gizzi M, DiRocco A, Sivak M, et al. Ocular motor function in motor neuron disease. Neurology 1992;42:1037-46. PMID 1579227

Glass JD, Hertzberg V, Boulis N, et al. Transplantation of spinal cord-derived neural stem cells for ALS: analysis of phase 1 and 2 trials. Neurology 2016;87(4):392-400. PMID 27358335

Gordon PH, Cheng B, Katz IB, Pinto M, Hays AP, Mitsumoto H, Rowland LP. The natural history of primary lateral sclerosis. Neurology 2006;66(5):647-53. PMID 16534101

Gordon PH, Moore, DH, Miller RG, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet Neurol 2007;6(12):1045-53. PMID 17980667

Gordon PH, Rowland LP, Younger DS, et al. Lymphoproliferative disorders and motor neuron disease: an update. Neurology 1997;48:1671-8. PMID 9191785

Gredal O, Werdelin L, Bak S, et al. A clinical trial of dextromethorphan in amyotrophic lateral sclerosis. Acta Neurol Scand 1997;96:8-13. PMID 9262126

Griggs RC, Askanas V, DiMauro S, et al. Inclusion body myositis and myopathies. Ann Neurol 1995;38:705-13. PMID 7486861

Groeneveld JG, Veldink JH, van der Tweel I, et al. A randomized sequential trial of creatine in amyotrophic lateral sclerosis. Ann Neurol 2003;53(4):437-45. PMID 12666111

Hafezparast M, Klocke R, Ruhrberg C, et al. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science 2003;300:808-12. PMID 12730604

Haley RW. Excess incidence of ALS in young Gulf War veterans. Neurology 2003;61:750-6. PMID 14504316

Hanisch UK. Microglia as a source and target of cytokines. Glia 2002;40(2):140-55. PMID 12379902

Harding AE. Hereditary “pure” spastic paraplegia: a clinical and genetic study of the 22 families. J Neurol Neurosurg Psychiatry 1981,44:871-83. PMID 7310405

Harriman M, Morrison M, Hay J, Revonta M, Eisen A, Lentle B. Use of radiotherapy for control of sialorrhea in patients with amyotrophic lateral sclerosis. J Otolaryngol 2001;30(4):242-5. PMID 11771037

Haverkamp LJ, Appel V, Appel SH. Natural history of amyotrophic lateral sclerosis in a database population: validation of a scoring system and a model for survival prediction. Brain 1995;118:707-19. PMID 7600088

Heiman-Patterson T, Rampai N, Branagan TH, Acosta T, Forshew DA, Bromberg MB. The spectrum of patient symptoms in ALS and symptom management. Neurology 2001;56(Supp 3):A199.

Hentati A, Ouahchi K, Pericak-Vance MA, et al. Linkage of a commoner form of recessive amyotrophic lateral sclerosis to chromosome 15q15-q22 markers. Neurogenetics 1998;2:55-60. PMID 9933301

Hirano A, Rurland LT, Krooth RS, et al. Parkinsonism-dementia complex, an endemic disease on the island of Guam. I. Clinical features. Brain 1961;84:642-61. PMID 13907609

Horner RD, Kamins KG, Feussner JR, et al. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology 2003;61:742-9. PMID 14504315

Hosler BA, Siddique T, Sapp PC, et al. Linkage of familial amyotrophic lateral sclerosis with frontotemporal dementia to chromosome 9q21-q22. JAMA 2000;284:1664-9. PMID 11015796

Hu MT, Ellis CM, Al Chalabi A, et al. Flail arm syndrome: a distinctive variant of amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 1998;65:950-1. PMID 9854987

Ince PG, Shaw PJ, Slade JY, Jones C, Hudgson P. Familial amyotrophic lateral sclerosis with a mutation in exon 4 of the Cu/Zn superoxide dismutase gene: pathological and immunocytochemical changes. Acta Neuropathol 1996;92(4):395- 403. PMID 8891072

Ito H, Wate R, Zhang J, et al. Treatment with edaravone, initiated at symptom onset, slows motor decline and decreases SOD1 deposition in ALS mice. Exp Neurol 2008;213(2):448-55. PMID 18718468

Jossan SS, Ekblom J, Gudijonsson O, et al. Double-blind cross over trial with deprenyl in amyotrophic lateral sclerosis. J Neural Transm Suppl 1994;41:237-41. PMID 7931231

Kabashi E, Valdamanis PN, Dion P, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 2008;40(5):572-4. PMID 18372902

Kanekura K, Hashimoto Y, Niikura T, Aiso S, Matsuoka M, Nishimoto I. Alsin, the product of the ALS2 gene, suppresses SOD1 mutant neurotoxicity through RhoGEF domain by interacting with SOD1 mutants. J Biol Chem 2004;279:19247- 56. PMID 14970233

Kaufmann P, Thompson JL, Levy G, et al. Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann Neurol 2009;66:235-44. PMID 19743457

Kennedy WB, Alter M, Sung JG. Report of an X-linked form of spinal muscular atrophy. Neurology 1968;18:671-80.

Khwaja S, Sripathi N, Ahmad BK, Lennon VA. Paraneoplastic motor neuron disease with type 1 Purkinje cell antibodies. Muscle Nerve 1998;21:943-5. PMID 9626256

Kim WK, Liu X, Sandner J, et al. Study of 962 patients indicates progressive muscular atrophy is a form of ALS. Neurology 2009;73(20):1686-92. PMID 19917992

Kimura J. Electrodiagnosis in diseases of nerve and muscle: principles and practice. Oxford University Press, 2001.

Kleopa KA, Sherman M, Neal B, et al. Bipap improves survival and rate of pulmonary function decline in patients with ALS. J Neurol Sci 1999;164:82-8. PMID 10385053

Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci 1996;19:312-8. PMID 8843599

Kwiatkowski TJ, Bosco DA, LeClerc AL, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 2009;323(5918):1205-8. PMID 19251627

Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis/riluzole study group II (see comments). Lancet 1996;347:1425-31. PMID 8676624

Lambert EH. Electromyography in amyotrophic lateral sclerosis. In: Norris FH, Kurland LT, editors. Motor neuron diseases. New York: Grune & Stratton, 1969;135-53.

LaMonte BH, Wallace KE, Holloway BA, et al. Disruption of dynein/dynactin inhibits axonal transport in motor neurons causing late-onset progressive degeneration. Neuron 2002;34:715-27. PMID 12062019

Lariviere RC, Beaulieu JM, Nguyen MD, Julien JP. Peripherin is not a contributing factor to motor neuron disease in a mouse model of amyotrophic lateral sclerosis caused by mutant superoxide dismutase. Neurobiol Dis 2003;13:158-66. PMID 12828939

Lechtzin N, Scott Y, Busse AM, Clawson LL, Kimball R, Wiener CM. Early use of non-invasive ventilation prolongs survival in subjects with ALS. Amyotroph Lateral Scler 2007;8:185-8. PMID 17538782

Li J, Petajan J, Smith G, Bromberg M. Electromyography of sternocleidomastoid muscle in ALS: a prospective study. Muscle Nerve 2002,25:725-8. PMID 11994967

Li W, Lee MH, Henderson L, et al. Human endogenous retrovirus-K contributes to motor neuron disease. Sci Transl Med 2015;7(307):307ra153. PMID 26424568

Lino MM, Schneider C, Caroni P. Accumulation of SOD1 mutants in postnatal motoneurons does not cause motoneuron pathology or motoneuron disease. J Neurosci 2002;22(12):4825-32. PMID 12077179

Lomen-Hoerth C, Anderson T, Miller B. The overlap of amyotrophic lateral sclerosis and frontotemporal dementia. Neurology 2002;59:1077-9. PMID 12370467

Lomen-Hoerth C, Murphy J, Langmore S, et al. Are amyotrophic lateral sclerosis patients cognitively normal. Neurology 2003;60:1094-7. PMID 12682312

Lou JS, Reeves A, Benice T, Sexton G. Fatigue and depression are associated with poor quality of life in ALS. Neurology 2003;60:122-3. PMID 12525733

Louwerse ES, Weverling GJ, Bossuyt PM, et al. Randomized, double-blind controlled trial of acetylcysteine in amyotrophic lateral sclerosis. Arch Neurol 1995;52:559-64. PMID 7763202

MacGowan DJ, Scelsa SN, Imperato TE, Liu KN, Baron P, Polsky B. A controlled study of reverse transcriptase in serum and CSF of HIV-negative patients with ALS. Neurology 2007;68(22):1944-6. PMID 17536052

Mackenzie IR, Rademakers R, Neumann M. TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol 2010;9(10):995-1007. PMID 20864052

Magnus T, Beck M, Giess R, Puls I, Naumann M, Toyka KV. Disease progression in amyotrophic lateral sclerosis: predictors of survival. Muscle Nerve 2002;25:709-14. PMID 11994965

Majounie E, Renton AE, Mok K, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol 2012;11(4):323-30. PMID 22406228

Mak JN, McFarland DJ, Vaughn TM, et al. EEG correlates of P300-based brain-computer interface (BCI) performance in people with amyotrophic lateral sclerosis. J Neural Eng 2012;9(2):026014. PMID 22350501

Manfredi G, Beal M. Mitochondrial dysfunction and energy metabolism in amyotrophic lateral sclerosis. In: Amyotrophic Lateral Sclerosis. Brown RH, Swash M, Passinelli P, editors. London UK: Informa Healthcare, 2006:323-31.

McDermott CJ, Bradburn MJ, Maguire C, et al. Safety and efficacy of diaphragm pacing in patients with respiratory insufficiency due to amyotrophic lateral sclerosis (DiPALS): a multicentre, open-label, randomised controlled trial. Lancet Neurol 2015;14(9):883-92. PMID 26234554

McKee AC, Gavett BE, Stern RA, et al. TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2010;69(9):918-29. PMID 20720505

Miller R, Bradley W, Cudkowicz M, et al. Phase II/III randomized trial of TCH346 in patients with ALS. Neurology 2007;69(8):776-84. PMID 17709710

Miller RG, Jackson CE, Kasarkis EJ, et al. Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: drug, nutritional, and respiratory therapies (an evidence based review). Neurology 2009a;73:1227-33. PMID 19822872

Miller RG, Jackson CE, Kasarkis EJ, et al. Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: multidisciplinary care, symptom management, and cognitive/behavioral impairment (an evidence based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2009b;1218-26. PMID 19822873

Miller R, Michell J, Moore D. Riluzole for amyotrophic lateral sclerosis/motor neuron disease (Cochrane Review). Cochrane Library. Cochrane Database Syst Rev 2002;(2):CD001447. PMID 12076411

Miller RG, Moore D, Gelinas D, et al. Phase III randomized trial of gabapentin in patients with amyotrophic lateral sclerosis. Neurology 2001;56:843-8. PMID 11294919

Miller RG, Munsat TL, Swash M, Brooks BR. Consensus guidelines for the design and implementation of clinical trials in ALS. World Federation of Neurology committee on Research. J Neurol Sci 1999a;169:2-12. PMID 10540001 Miller RG, Rosenberg JA, Gelinas DF, et al. Practice parameter: the care of the patient with amyotrophic lateral sclerosis (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology: ALS Practice Parameters Task Force. Neurology 1999b;52:1311-23. PMID 10227612

Miller RG, Shepherd R, Dao H, et al. Controlled trial of nimodipine in amyotrophic lateral sclerosis. Neuromuscul Disord 1996a;6:101-4. PMID 8664560

Miller RG, Smith SA, Murphy JR. A clinical trial of verapamil in amyotrophic lateral sclerosis. Muscle Nerve 1996b;19:511-5. PMID 8622731

Mitsumoto H, Rabkin JG. for patients with amyotrophic lateral sclerosis: "prepare for the worst and hope for the best." JAMA 2007;298:207-16. PMID 17622602

Mitsumoto H, Sliman RJ, Schafer IA, et al. Motor neuron disease in adult hexosaminidase A deficiency in two families: evidence for multisystem degeneration. Ann Neurol 1985;17:378-85. PMID 3159334

Moss AH, Casey P, Stocking CB, et al. Home ventilation for amyotrophic lateral patients: outcomes, costs, and patient, family, and physician attitudes. Neurology 1993;43:438-43. PMID 8437718

Moss AH, Oppenheimer EA, Casey P, et al. Patients with amyotrophic lateral sclerosis receiving long-term mechanical ventilation: advance care planning and outcomes. Chest 1996;110:249-55. PMID 8681635

Moulignier A, Moulonguet A, Pialoux G, Rozenbaum W. Reversible ALS-like disorder in HIV infection. Neurology 2001;57:995-1001. PMID 11571323

Mulder DW, Bushek W, Spring E, et al. Motor neuron disease (ALS): evaluation of detection thresholds of cutaneous sensation. Neurology 1983;33:1625-7. PMID 6685835

Newsom-Davis IC, Lyall RA, Leigh PN, Moxham J, Goldstein LH. The effect of non-invasive positive pressure ventilation (NIPPV) on cognitive function in amyotrophic lateral sclerosis: a prospective study. J Neurol Neurosurg Psychiatry 2001;71:482-7. PMID 11561031

Nguyen MD, Julien JP, Rivest S. Induction of proinflammatory molecules in mice with amyotrophic lateral sclerosis: no requirement for proapoptotic interleukin-1beta in neurodegeneration. Ann Neurol 2001a;50(5):630-39. PMID 11706969

Ono S, Toyokura Y, Mannen T, et al. Amyotrophic lateral sclerosis: histologic, histochemical and ultrastructural abnormalities of skin. Neurology 1986;36:948-56. PMID 3714056

Parry GJ. AAEM case report #30: multifocal motor neuropathy. Muscle Nerve 1996;19:269-76. PMID 8606689

Petrou P, Gothelf Y, Argov Z, et al. Safety and clinical effects of mesenchymal stem cells secreting neurotrophic factor transplantation in patients with amyotrophic lateral sclerosis: results of phase 1/2 and 2a clinical trials. JAMA Neurol 2016;73(3):337-44. PMID 26751635

Pioro EP, Brooks BR, Cummings J, et al. Dextromethorphan plus ultra-low dose quinidine reduces pseudobulbar affect. Ann Neurol 2010;68(5):693-702. PMID 20839238

Pringle CE, Hudson AJ, Munoz DG, Kiernan JA, Brown WF, Ebers GC. Primary lateral sclerosis: clinical features, neuropathology and diagnostic criteria. Brain 1992;115(Pt 2):495-520. PMID 1606479

Puls I, Jonnakuty C, LaMonte BH, et al. Mutant dynactin in motor neuron disease. Nat Genet 2003;33:455-6. PMID 12627231

Pupillo E, Messina P, Logroscino G, et al. Trauma and amyotrophic lateral sclerosis: a case-control study from a population-based registry. Eur J Neurol 2012;19(12):1509-17. PMID 22537412

Rabkin JG, Albert S, Del Bene ML, et al. Prevalence of depressive disorders and change over time in late-stage ALS. Neurology 2005;65:62-7. PMID 16009886

Pugdahl K, Fuglsang-Frederiksen A, de Carvalho M, et al. Generalised sensory system abnormalities in amyotrophic lateral sclerosis: a European multicentre study. J Neurol Neurosurg Psychiatry 2007;78:746-9. PMID 17575020

Rao SD, Weiss JH. Excitotoxic and oxidative cross-talk between motor neurons and glia in ALS pathogenesis. Trend Neurosci 2004;27:17-23. PMID 14698606

Renton AE, Chio A, Traynor BJ. State of play in amyotrophic lateral sclerosis genetics. Nat Neurosci 2014;17(1):17-23. PMID 24369373

Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011;72(2):257-68. PMID 21944779

Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci 2013;14(4):248-64. PMID 23463272

Robertson J, Doroudchi MM, Nguyen MD, et al. A neurotoxic peripherin splice variant in a mouse model of ALS. J Cell Biol 2003;160:939-49. PMID 12642616

Rosen DR, Siddique T, Patterson D, et al. Mutations Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993;362:59-62. PMID 8332197

Rosenberg PA, Amin S, Leitner M. Glutamate uptake disguises neurotoxic potency of glutamate agonists in in dissociated cell culture. J Neurosci 1992;12:56-61. PMID 1345946

Rothstein JD. Current hypothesis for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol 2009;65(suppl):S3-9. PMID 19191304

Rowland LP. Diverse forms of motor neuron disease. In: Rowland LP, editor. Human motor neuron diseases. New York: Raven Press, 1982:1-13.

Rowland LP, Shneider NA. Amyotrophic lateral sclerosis. New Engl J Med 2001;344:1688-700. PMID 11386269

Russman B. Spinal muscular atrophy. In: Katirji B, Kaminski HJ, Preston DC, Ruff RL, Shapiro BE, editors. Neuromuscular disorders in clinical practice. Boston: Butterworth Heinemann, 2002:445-53.

Sancho J, Servera E, Diaz J, Marin J. Efficacy of mechanical insufflation-exsufflation in medically stable patients with amyotrophic lateral sclerosis. Chest 2004;125:1400-1405. PMID 15078752

Sasaki S, Tsutsumi Y, Tamane K, et al. Sporadic amyotrophic lateral sclerosis with extensive neurological involvement. Acta Neuropathol 1992;84:213-5. PMID 1523976

Savica R, Parisi JE, Wold LE, Josephs KA, Ahlskog JE. High school football and risk of neurodegeneration: a community based study. Mayo Clin PRoc 2012;87(4): 335-40. PMID 22469346

Sawada H. Clinical efficacy of edaravone for the treatment of amyotrophic lateral sclerosis. Expert Opin Pharmacother 2017;18(7):735-8. PMID 28406335

Scelsa SN, MacGowan DJ, Mitsumoto H, et al. A pilot, double-blind, placebo-controlled trial of indinavir in patients with ALS. Neurology 2005;64(7):1298-300. PMID 15824372

Shefner JM, Cudkowicz ME, Shoenfeld D, et al. A clinical trial of creatine in ALS. Neurology 2004;63:1656-61. PMID 15534251

Siddique T, Nijhawan D, Hentati A. Molecular genetic basis of familial ALS. Neurology 1996;47(4 Suppl 2):S27-34. PMID 8858048

Siklos L, Engelhardt JI, Alexianu ME, Gumey ME, Siddique T, Appel SH. Intracellular calcium parallels motoneuron degeneration in SOD-1 mutant mice. J Neuropathol Exp Neurol 1998;57:571-87. PMID 9630237

Singh N, Sachdey K, Susheela AK. Juvenile muscular atrophy localized to arm. Arch Neurol 1980;37:297-9. PMID 7387450 Smith SA, Miller RG, Murphy JR, et al. Treatment of ALS with high dose pulse cyclophosphamide. J Neurol Sci 1994;124(Suppl):84-7. PMID 7807153

Sorensen EJ, Windbank AJ, Mandrekar JN, et al. Subcutaneous IGF-1 is not beneficial in 2 year ALS trial. Neurology 2008;71(22):1770-5. PMID 19029516

Spada AR, Wilson EM, Lubahn DB, et al. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 1991;352:77-9. PMID 2062380

Spencer PS, Ohta M, Palmer VS. Cycad use and motor neuron disease in the Kii Peninsula of Japan. Lancet 1987;2:1462-3. PMID 2892020

Sreedharan J, Blair IP, Tripathi VB, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 2008;319(5870):1668-72. PMID 18309045

Strong MJ. The evidence for altered RNA metabolism in amyotrophic lateral sclerosis (ALS). J Neurol Sci 2010;288:1-12. PMID 19840884

Tan E, Lynn DJ, Amato AA, et al. Immunosuppressive treatment of motor neuron syndromes: attempts to distinguish a treatable disorder. Arch Neurol 1994;51:194-200. PMID 8304845

Tandan R, Bromber MB, Forshew D, et al. A controlled trial of amino acid therapy in amyotrophic lateral sclerosis: I. Clinical, functional, and maximum isometric torque data. Neurology 1996;47:1220-6. PMID 8909433

Vance C, Rogei B, Hortobagyi T, et al. Mutations in FUS, RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 2009;323(5918):1208-11. PMID 19251628

Vincenti M, Bonvicin F, Rothman KJ, Vescovi L, Wang F. The relation between amyotrophic lateral sclerosis and inorganic selenium in drinking water: a population based, case control study. Environ Health 2010;9:77. PMID 21134276

Walker MP, Schlaberg R, Hays AP, Bowser R, Lipkin WI. Absence of echovirus sequences in brain and spinal cord of amyotrophic lateral sclerosis patients. Ann Neurol 2001;49:249-23. PMID 11221802

Wasner M, Klier H, Borasio GD. The use of alternative medicine by patients with amyotrophic lateral sclerosis. J Neurol Sci 2001;191:151-4. PMID 11677007

Weisskopf MG, O'Reilly EJ, McCullough ML, et al. Prospective study of military service and mortality from ALS. Neurology 2005;64(1):32-7. PMID 15642900

Williams AH, Valdez G, Moresi V, et al. MicroRNA-206 delays ALS progression and promotes regeneration of neuromuscular synapse in mice. Science 2009;326;1549-54. PMID 20007902

Wong N, He BP, Strong MJ. Characterization of neuronal intermediate filament protein expression in cervical spinal motor neurons in sporadic amyotrophic lateral sclerosis (ALS). J Neuropathol Exp Neurol 2000;59(11):972-82. PMID 11089575

Yanagihara RT, Garrutto RM, Gajdusek DC, et al. Calcium and vitamin D metabolism in Guamanian Chamorros with amyotrophic lateral sclerosis and parkinsonism-dementia. Ann Neurol 1984;15(1):42-8. PMID 6546847

Yang Y, Hentati A, Deng HX, et al. The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis. Nat Genet 2001;29:160-5. PMID 11586297

Yokoseki A, Shiga A, Tan CF, et al. TDP-43 mutation in familial amyotrophic lateral sclerosis. Ann Neurol 2008;63(4):538-42. PMID 18438952

Younger DS, Rowland LP, Latov N, et al. Lymphoma, motor neuron diseases, and amyotrophic lateral sclerosis. Ann Neurol 1991;29:78-86. PMID 1996882

**References especially recommended by the author or editor for general reading. Former authors

Robert Brown MD (original author), Jau-Shin Lou MD PhD, Jinsy A Andrews MD, and Hiroshi Mitsumoto MD

ICD and OMIM codes

ICD codes

ICD-9: Amyotrophic lateral sclerosis: 335.20

ICD-10: Amyotrophic lateral sclerosis: G12.2

OMIM numbers

Amyotrophic lateral sclerosis 1: #105400

Profile

Age range of presentation

13-18 years 19-44 years 45-64 years 65+ years

Sex preponderance male>female, >2:1 male>female, >1:1

Family history family history may be obtained

Heredity heredity may be a factor

Population groups selectively affected

Chamorro people of the islands of Guam, Rota, and Tinian Japanese villagers of Kii Peninsula, Honshu Island, Japan West New Guineans

Occupation groups selectively affected

Military veterans

Differential diagnosis list amyotrophic lateral sclerosis-parkinsonism-dementia complex benign fasciculations brainstem gliomas in adults brainstem stroke central nervous system complications of radiation cervical spondylosis cervical spondylotic myelopathy Creutzfeldt-Jakob disease familial spastic paraparesis hyperparathyroidism (Myopathies associated with parathyroid disorders) hyperthyroidism (Thyrotoxicosis) inclusion-body myositis inflammatory myopathy Kennedy disease (spinal muscular atrophy) Kugelberg-Welander disease (spinal muscular atrophy) multifocal motor polyneuropathy multiple sclerosis myasthenia gravis paraneoplastic motor neuronopathy post-polio syndrome (Poliomyelitis) radiculopathy spinal muscular atrophy Werdnig-Hoffman disease

Associated disorders

Dementia Depression Parkinson disease

Other topics to consider

Dementia associated with amyotrophic lateral sclerosis Gene therapy of neurodegenerative disorders Riluzole Sleep and neuromuscular and spinal cord disorders Treatment of degenerative disorders with neurotrophic factors

Copyright© 2001-2017 MedLink Corporation. All rights reserved.