Amyotrophic Lateral Sclerosis

Back

Practice Essentials

Amyotrophic lateral sclerosis (ALS) is the most common degenerative disease of the motor neuron system. Although ALS is incurable and fatal, with median survival of 3 years, treatment can extend the length and meaningful quality of life for patients.

Signs and symptoms

In 75-80% of patients, symptoms begin with limb involvement. Initial complaints in patients with lower limb onset are often as follows:

Initial complaints with upper limb onset include the following:

With bulbar onset (20-25%), initial complaints are as follows:

Emotional and special cognitive difficulties in some ALS patients are as follows:

Features of more-advanced disease are as follows:

Progression of bulbar disease leads to the following:

The image below illustrates the 4 regions of the body.



View Image

The 4 regions or levels of the body. Bulbar (muscles of the face, mouth, and throat); cervical (muscles of the back of the head and the neck, the shou....

See Clinical Presentation for more detail.

Diagnosis

Definitive diagnosis may not be possible with early ALS. Confirmation of the disease may require a period of observation to document its progressive nature and to exclude alternative diagnoses.

The World Federation of Neurology (WFN) has developed a diagnostic algorithm that combines the clinical and, in some cases, electrophysiologic findings.[1] The degree of certainty of diagnosis is increased by the number of body segments that demonstrate upper motor neuron (UMN) and lower motor neuron (LMN) signs. UMN signs are mild weakness, spasticity, and abnormally brisk reflexes; LMN signs are progressive weakness, wasting, and loss of reflexes and muscle tone. WFN categories are as follows:

Hallmark findings in the electrodiagnosis of ALS are normal sensory nerve conduction studies and abnormal motor nerve conduction studies, with reduced motor compound muscle action potentials. The needle exam shows changes characteristic of ongoing denervation and reinnervation of muscles.

In patients with familial ALS, genetic testing may be requested after appropriate counseling. The results of genetic testing may affect not only the patient, but also family members. Tests for the SOD1, TARDBP (coding for TDP-43), FUS, ANG, C9orf72, and FIG4 genes and for the gene causing Kennedy disease are available commercially. Patients with other forms of familial ALS may be referred to centers with a research interest in familial ALS.

See Workup for more detail.

Management

American Academy of Neurology recommendations for management of patients with ALS can be summarized as follows[2, 3] :

Invasive ventilatory support, requiring tracheostomy, may be considered in the following cases:

See Treatment and Medication for more detail.

Background

Amyotrophic lateral sclerosis (ALS) is the most common degenerative disease of the motor neuron system. The disorder is named for its underlying pathophysiology, with “amyotrophy” referring to the atrophy of muscle fibers, which are denervated as their corresponding anterior horn cells degenerate. “Lateral sclerosis” refers to the changes seen in the lateral columns of the spinal cord as upper motor neuron (UMN) axons in these areas degenerate and are replaced by fibrous astrocytes (gliosis).

ALS is a fatal disease, with a median survival period of 3 years from onset of weakness.[4] (See Prognosis.) Aspiration pneumonia and medical complications of immobility contribute to morbidity in patients with the disease.

ALS was first described in 1869 by the French neurologist Jean-Martin Charcot and hence is also known as Charcot disease; however, it gained popular recognition and its best-known eponym in the United States after the baseball player Lou Gehrig announced his diagnosis with the disease in 1939.[5, 6, 7, 8, 9] ALS is also known as motor neuron disease (MND).

The cause of ALS is unknown, although a family history of the disease is obtained in about 5% of patients, and twin studies show a genetic contribution with heritability of about 61%.[10] In some cases, ALS overlaps clinically, pathologically, and biologically with frontotemporal dementia, and it may share common biologic mechanisms with Alzheimer disease, Parkinson disease, and other neurodegenerative diseases.[11, 12, 13, 14] (See Etiology.)

Degenerative effects of ALS

ALS is one of the system degeneration diseases, disorders that cause networks that work together in health to disintegrate together in an organized manner.[15, 16] ALS results from the systematic dismantling of the motor neuron system, with the clinical manifestations in each patient deriving from the site of onset and cell type involved; the relative affinity of the dismantling process for prefrontal, upper and lower motor neurons; and the rate of the disease’s spread within the network.[17]

In its classic form, ALS affects motor neurons at 2 or more levels of the motor neuron network supplying multiple regions of the body. It affects lower motor neurons (LMNs) that reside in the anterior horn of the spinal cord and in the brain stem, corticospinal UMNs that reside in the precentral gyrus, and, frequently, prefrontal motor neurons that are involved in planning or orchestrating the work of the upper and lower motor neurons.[18] (See Pathophysiology.)

Loss of LMNs leads to progressive muscle weakness, wasting (atrophy), and fasciculations, with loss of reflexes and muscle tone. Loss of corticospinal UMNs usually leads to milder weakness associated with stiffness (spasticity), which may be severe, and abnormally brisk reflexes.

Loss of prefrontal neurons may result in special forms of cognitive impairment that include, most commonly, executive dysfunction but that may also include an altered awareness of social implications of one’s circumstances and, consequently, maladaptive social behaviors.[19] In its fully expressed forms, the prefrontal dysfunction meets established criteria for frontotemporal dementia.[20, 21] Loss of ability to integrate motor function (apraxia), a premotor function, is seen at times. It is more noticeable in limbs that are not overly weak.

Types of motor neuron disease

Classic ALS

The term classic ALS is reserved for the form of disease that involves upper and lower motor neurons. The classic form of sporadic ALS usually starts as dysfunction or weakness in one part of the body and spreads gradually within that part and then to the rest of the body.[22] Ventilatory failure results in death 3 years, on average, after the onset of focal weakness. The rate of disease progression varies widely, however, with some patients dying a few months after experiencing their first symptom and others still walking 10 years afterward.

Progressive muscular atrophy and flail limb syndrome

The disease may be restricted to LMNs. When the pattern of LMN involvement is asymmetrical, the disorder is termed progressive muscular atrophy (PMA), and the course is usually indistinguishable from that of classic ALS. Patients with a symmetrical pattern, called flail limb syndrome, have a course that may be far longer.[23]

Primary lateral sclerosis

When only UMNs are involved, the disease is called primary lateral sclerosis (PLS). The course of PLS differs from that of ALS and is usually measured in decades.[24]

Progressive bulbar palsy

Rarely, the disease is restricted to bulbar muscles, in which case it is called progressive bulbar palsy (PBP). In most patients who present with initial involvement of bulbar muscles, the disease evolves to classic ALS.

Familial ALS

Worldwide, a family history of ALS is obtained in about 5% of cases; these patients have familial ALS. Most familial ALS is inherited in an autosomal dominant pattern,[18] often with reduced penetrance, but other patterns, such as X-linked or autosomal recessive inheritance, are seen (see Etiology.)

The fact that in most patients ALS is sporadic does not preclude a genetic contribution to the disease in these cases. ALS as a whole is best thought of as a disease showing complex inheritance.

Complications

Complications of ALS can include the following:

Diagnosis and treatment

The diagnosis of ALS is primarily clinical. Electrodiagnostic testing contributes to the diagnostic accuracy (see Clinical Presentation and Workup). Making a diagnosis is important to patients and families, allowing them to stop the search for alternative causes of a patient's disability and to focus their attention on treatment.

Although ALS is incurable, there are treatments that can extend the length and meaningful quality of life for patients (see Treatment).

Mechanism-specific treatments directed at the processes that cause the disease to evolve after it has expressed itself sufficiently to be diagnosed may, at best, have an ameliorative effect. Treatments that halt the spread of the disease may be more effective than those that try to salvage affected motor neurons. All of these have yet to be realized. Currently, the mainstay of ALS therapy is adaptive treatments directed at the clinical manifestations of the disease.

Pathophysiology

ALS should not be considered a single disease entity, but rather a clinical diagnosis for different pathophysiologic cascades that share the common consequence of causing preferential progressive loss of motor neurons and the orderly dismantling of the motor neuron system.

ALS mechanisms

Previously, research into the mechanisms resulting in sporadic and familial types of ALS had examined several possibilities. For example, excitotoxicity was suggested to occur secondary to overactivation of glutamate receptors.

Oxidative stress linked to free radical formation was also explored as a cause of ALS, owing to the discovery of mutations in the free radical–scavenging enzyme superoxide dismutase 1 (SOD1).[25] Mitochondrial damage was implicated as a possible mechanism as well, as was autoimmunity to calcium ion channels.

The observation of cytoskeletal proteins in cellular inclusions led to consideration of neurofilament defects as another possible cause of ALS. Inclusions in general implicated defects in the proteasome system were considered as a possible unifying mechanism.

More recent research has focused on RNA processing, because several genetic risk factors for ALS are involved in this metabolic pathway, and aggregation of proteins involved in RNA metabolism has been seen in most forms of ALS. Apoptosis has emerged as a possible method of neuronal death, although this is not certain.

Despite such research, no direct mechanism for ALS has been identified. Most investigators and clinicians agree that various factors, possibly a combination of some or all of the above processes, may lead to development of the disease.[26, 27]

If ALS is considered under the umbrella of neuronal system degenerative diseases, then the specificity for the motor system attacked by the disorder can be attributed to a pathologic process that arises within and spreads through the motor neuron system. Similarly, the focal onset (with subsequent spread) can be compared with the pathogenesis of prion disease (focal onset of a misfolded protein and its spread) or malignancy (a single DNA change or summative mutations, with a final one that confers the pathologic activity and its subsequent spread).

Prionlike propagation of misfolding of proteins—in particular, SOD1 and the 43 kDa transactive response DNA binding protein (TDP-43)—has been proposed as a mechanism for the regional spread of ALS symptoms.[25] The accumulation of misfolded proteins has parallels in other neurodegenerative diseases, including Alzheimer, Parkinson, and Huntington disease.

Axonal degeneration

Motor axons die by Wallerian degeneration in ALS, and large motor neurons are affected to a greater extent than smaller ones. This process occurs as a result of the death of the anterior horn cell body, leading to degeneration of the associated motor axon.

As the axon breaks down, surrounding Schwann cells catabolize the axon's myelin sheath and engulf the axon, breaking it into fragments. This forms small ovoid compartments containing axonal debris and surrounding myelin, termed myelin ovoids. Ovoids then are phagocytized by macrophages recruited into the area to clean up debris.

This type of axonal degeneration can be seen in the brain on biopsy as atrophy and pallor of myelinated motor axons in the corticospinal tracts. In cases in which the disease has been active for a long time, atrophy of the primary motor and premotor cortex may be seen as well. On biopsy of the spinal cord, degeneration of the myelinated motor axons with associated atrophy of the anterior motor roots of the spinal cord can be observed.

Wallerian degeneration also occurs peripherally, and collateral branches of surviving axons in the surrounding area can be seen attempting to reinnervate denervated muscle fibers. On muscle biopsy, various stages of atrophy are noted from this pattern of denervation and subsequent reinnervation of muscle fibers.

In typical ALS, certain motor neurons are spared until very late in the disease process. In the brain stem, these include the oculomotor, trochlear, and abducens nerves. In the spinal cord, the posterior columns, spinocerebellar tracts, nucleus of Onuf (which controls bowel and bladder function), and the Clarke column generally are spared, though the Clarke column can be affected in the familial form of the disease.

Pathways to cell death

Pathways that lead to cell death in ALS may be mediated by the following[28] :

Mutations in the copper/zinc superoxide dismutase 1 (SOD1) gene, which encodes an important antioxidant protein, have been seen in up to 20% of familial ALS patients.[29] Studies in transgenic mice carrying the human SOD1 mutation have provided important information on the pathophysiology of ALS.[28] Also, high levels of oxidative damage to proteins have been found in serum, urine, and cerebrospinal fluid samples from patients, as well as in postmortem samples of patients with both sporadic ALS and SOD1 -familial ALS.[30, 31, 32]

Inferences from animal models, including transgenic models of familial disease, to sporadic human disease are tenuous. However, recognition of the role of glutamate excitotoxicity in sporadic disease and in animal models paved the way to the testing and approval of riluzole, the only treatment that has been shown to ameliorate the course of sporadic ALS, extending patients’ lives by 2-3 months.[33, 34]

Derangements of RNA metabolism

The findings below have placed derangements of RNA metabolism at the core of current thinking with regard to the pathophysiology of most types of ALS.

TARDBP gene

In 2006, ubiquitinated inclusions containing pathologic forms of TAR DNA-binding protein-43 (TDP-43) were identified in the cytoplasm of motor neurons of patients with sporadic ALS and in a subset of patients with frontotemporal dementia.[35, 36] TDP-43 is an RNA-processing protein that is normally localized predominantly in the nucleus.

Shortly after their identification in sporadic ALS, TDP-43–positive cytoplasmic inclusions were identified in patients with non-SOD1 familial ALS[37, 38] , and mutations in the gene on chromosome 1 coding for TDP-43 were identified in patients with sporadic and familial ALS.[39, 40, 41, 42, 43, 44]

Mutations in the TARDBP gene, which codes for TDP-43, account for 5% of patients with familial ALS. In addition, TDP-43 inclusions have been found in more than 90% of patients with sporadic ALS, in patients with Guamanian parkinsonism-dementia complex,[45] in the majority of patients with frontotemporal dementia, and in patients with familial British dementia.[46] A review of the continuum of multisystem TDP-43 proteinopathies concluded that the phenotypic expression is linked to the specific cells that are affected by the proteinopathy.[47]

FUS/TLS gene

In 2009, two groups[48, 49] reported that ALS-6, an autosomal dominant form of ALS, results from mutations in the gene for another RNA-processing protein, fused in sarcoma/translated in liposarcoma, or FUS/TLS. (The gene, FUS/TLS, is located on chromosome 16.) Patients these mutations have cytoplasmic inclusions containing FUS/TLS but not TDP-43. Usually, FUS/TLS, like TDP-43, is concentrated in the nucleus. Mutations in FUS/TLS account for 4% of patients with familial ALS.

Additional evidence

Further support for this idea has come from the following[50, 51] :

In 2011, researchers reported that a large hexanucleotide repeat expansion in the noncoding region adjacent to the C9orf72 gene, which is located on the short arm of chromosome 9, accounts for nearly 50% of familial ALS and frontotemporal dementia (FTD) in the Finnish population and more than a third of familial ALS in other groups of European ancestry.[13, 14]  It is the most common mutation seen in patients with sporadic ALS. One effect of this mutation is the formation of nuclear RNA foci containing antisense RNA repeats. In addition, a novel mechanism of poly-dipeptide production, repeat-associated non-ATG translation (RAN)[52] has been shown to occur in carriers of the hexanucleotide expansion.[53] The abnormal polypeptides form cytoplasmic deposits. It is unclear how these deposits cause disease. In particular, since median age of onset of C9orf72-associated ALS is the same as that of sporadic ALS, and the deposits precede clinical disease by years, it is unclear how the nuclear or cytoplasmic deposits cause ALS or FTD.

Disease onset (pathogenesis)

Making the distinction between the pathogenesis and pathophysiology of ALS matters because the mechanisms underlying each of these stages are probably different. This means that interfering with these mechanisms likely requires different approaches. The interventions to prevent disease onset are probably not the same as those required to slow or halt its progression after onset. Prevention of ALS requires modifying or removing factors that are part of disease pathogenesis. A precondition is identifying probable risk factors for ALS.[54]

In contrast to the advances in understanding the pathophysiology of ALS, however, the mechanisms that lead to disease onset (ie, pathogenesis) remain unknown. It is reasonable to assume that the abnormal gene or gene product plays a role in triggering disease onset in familial ALS and may have a role in disease propagation, but having an abnormal gene is neither necessary nor sufficient for developing ALS. Rarely, obligate familial gene carriers do not develop the disease. There is incomplete penetrance, or age-dependent penetrance, of the autosomal dominant genes.

Additional factors must be postulated to intervene between birth and disease onset even in patients who develop familial ALS, because the disease does not appear to start at birth, and within a given family there is great variability in the age of onset. Having normal copies of these genes does not prevent development of sporadic ALS.

Course of the disease

Loss of motor neurons bridges disease pathophysiology and its clinical expression. When advanced, this loss results in the characteristic picture seen in cross-sections of the spinal cord in ALS. At the level of the muscle, loss of discrete LMNs causes loss of innervation of individual motor units.

Early in the disease, surviving nerve fibers establish connections and reinnervate motor units that have lost their connection to axons that have died; as a result, larger motor units are formed. These large motor units manifest in histologic stains as fiber-type grouping. (See the image below.) They also have special characteristics on electromyographic testing. Later in the disease, when the motor neurons that supply the large motor units die, group atrophy ensues.



View Image

Muscle in nerve disease. Image courtesy of Dr. Friedlander, Associate Professor and Chair of Pathology at Kansas City University of Medicine and Biosc....

As long as reinnervation can keep up with denervation, clinical weakness may not be detectable, although loss of dexterity may occur. However, as the motor units grow larger and their numbers decrease, the earliest consequence is that affected muscle may fatigue faster than muscle with normal motor units; consequently, one of the first symptoms of ALS may be fatigability of function in the region of onset (for example, “his speech would become muffled toward the end of his sermons”).

As the number of motor units innervating a muscle decreases further, reinnervation can no longer keep up with denervation, permanent weakness develops and progresses, and the affected muscles gradually atrophy. In general, loss of cortical neurons may also result in weakness, but spasticity manifesting as stiffness is a more prominent and disabling UMN symptom.

Risk factors and triggering of onset

Acquired nucleic acid changes may trigger disease onset in sporadic ALS.[55] The past 10 years have seen an increase in evidence supporting this hypothesis. This hypothesis relies on the observation that smoking is the only established risk factor for sporadic ALS[56, 57] and provides a mechanism by which smoking might cause the disease—namely, by induction of changes in nucleic acids. It is supported by clinical observation that age-specific incidence of ALS increases with age. It follows a similar logic that suggests that the alkylating components in the cycad are responsible for delayed onset of Western Pacific ALS/PDC.[58, 59]

Simultaneous initial involvement of cortical and spinal motor neurons responsible for the same body part in ALS and the independent spread of disease at spinal and cortical levels has been shown.[60] These observations have been replicated,[61] although other patterns of spread are sometimes seen. They establish an irrefutable role for corticospinal neurons in the early spread of ALS and provide an observational foundation for postulating the existence of a focus of onset and one or more "agents of spread."[62]

Further support for this hypothesis comes from a statistical examination of acquired somatic mutation rates in humans. Somatic mutations inevitably occur during the many cell divisions required for the single-cell zygote to develop into a full organism; those mutations result in genetic mosaicism, and a small focus of genetically altered cells could be a trigger for ALS.[63]

The increase, with age, of age-specific incidence of sporadic ALS suggests that as time passes there is greater chance for changes to accumulate in nucleic acids that will lead ultimately to the development of ALS. This observation has been recently subjected to confirmatory quantitative analysis.[64] This analysis showed that ALS incidence increases with age in a logarithmic fashion, and the slope of log incidence vs. log age is 5. This suggests a multi-step process, analogous to carcinogenesis,[61] with 6 steps needed for ALS to be triggered.

An alternative trigger for disease onset might be appearance of a misfolded intracellular protein that induces other proteins to misfold, with cell-to-cell transmission of the misfolded protein within the motor network. The difference from classic prion disease is lack of transmissibility by inoculation to other organisms, and the confinement to a neuronal system, or network, with a common function. Hereditary or acquired nucleic acid changes may increase the likelihood that a gene product would be susceptible to misfolding, thus triggering disease onset.

The recent identification of the non-coding C9ORF72 hexanucleotide expansion in many familial and some sporadic ALS cases opens up a possibility that failure of regulation of a normal gene product may underlie ALS initiation and spread.[65, 66, 67, 68, 69, 70] In a broader sense, generation of a faulty regulatory gene product responsible for motor network maintenance, or failure to regulate such a product, may account for the specific disintegration of the motor network. MicroRNAs are particularly attractive candidates for this role.

The affirmation of spread substantiates the concept of a biologic focal onset for ALS. This in turn lends credibility to the concept of a focal trigger for ALS onset that generates the production of 1 or more agents of spread.[55]

Under this hypothesis, disease phenotype in each patient depends on the site of onset and the relative affinity of the specific agent of spread in that patient to motor neurons at the different hierarchical levels of the motor system (prefrontal, corticospinal, spinal/bulbar).[62] The concept of preferential affinity of the agent of spread may apply even to specific motor neurons within a given hierarchy, resulting, for example, in the special phenotypes in which LMNs are predominantly affected (flail arm syndrome, flail leg syndrome).[71]

Most of the biochemical changes found in the upper and lower motor neurons of diseased patients are probably downstream from those that initiate the disease and cause its spread. Some of these changes may represent the processes by which the motor neurons die, but others may reflect the efforts of the motor neurons to survive by compensating for, or “fighting,” the primary pathologic processes driving progression of ALS.

Etiology

Most ALS cases are sporadic, and the specific cause of sporadic ALS is unknown. Many abnormal genes have been identified in familial cases and are considered causal, although the precise mechanism by which they cause ALS is unknown for most.

All of the mutated genes found to give rise to familial ALS have also been found in patients with sporadic ALS. This is to be expected, as the distinction between familial and apparently sporadic disease is based on obtaining a family history, which in turn depends on gene penetrance, family size, age of family members, and the level of knowledge of the person being interviewed.[72]

In addition, first-degree relatives of patients with apparently sporadic disease have an increased risk of ALS. The overall lifetime risk of ALS in these relatives is low, however (approximately 1 in 50).[73]

Familial cases

A family history of ALS is obtained in about 5% of cases and is often consistent with a Mendelian autosomal dominant pattern of inheritance. While most cases of familial ALS are indistinguishable from sporadic disease, others have unique phenotypes.[74]

Juvenile forms of ALS are more commonly familial. Mean age of onset is 10-20 years younger in patients with familial ALS than in patients with apparently sporadic disease, and variability in age of onset between families is greater than variability within families.[18] Age of onset may also be modified by genetic factors independent of the cause of ALS.[75, 76]

Many specific heritable gene mutations have been described in familial ALS (see Table 1, below).[8] A curated, up-to-date list that includes unpublished mutations, genotype-phenotype correlations, and tools for analysis is maintained on the ALSoD website.

Table 1. Familial Forms of ALS[77, 78, 79]



View Table

See Table

About 10-20% of familial ALS cases result from a mutation in the copper/zinc superoxide dismutase 1 (SOD1) gene, also known as ALS1. In general, SOD1 ALS is a LMN form of the disease.[80] The other genes most commonly involved in familial ALS are C9orf72, FUS (ALS6) and TARDBP (ALS10).

SOD1 mutations

More than 140 allelic variations have been seen in SOD1. Some of these variations are characterized by a relatively predictable age of onset or rate of disease progression.

The traditionally used numbering of the amino acids in SOD1, which omits the start codon, is no longer in line with standard practice but will be used here. To translate to the modern numbering, an additional codon should be counted. For example, the D90A mutation frequent in Scandinavia should be p.D91A.

The most common SOD1 mutation in the United States is the A4V mutation, accounting for 50% of SOD1 ALS cases. It causes a rapidly progressive lower motor disease with a mean survival period of 1 year. The North American SOD1 A4V mutation descended from 2 founders (Amerindian and European) 400-500 years ago.[81]

Not all individuals with an SOD1 mutation develop ALS. SOD1 ALS has been shown to be a gain-of-function disease; knockout mice depleted of SOD1 do not develop ALS, and transgenic mice with 1 mutated and 2 normal genes have worse disease than those with 1 normal and 1 mutated gene. Misfolding and precipitation of the abnormal (and normal) SOD1 proteins are thought to be part of the pathophysiology of SOD1 ALS, but why the disease begins when it does and how it causes the LMN ALS phenotype is not clear.

Work in animal models suggests that silencing from birth of the expression of mutant SOD1 by silencing RNA molecules (siRNA) may prevent disease onset in transgenic mouse models.[82] This is an exciting direction for research in humans with SOD1 mutation, but major barriers need to be overcome first, including demonstration that this approach is effective in halting the disease after its clinical onset.[74, 77, 78, 83]

TARDBP and FUS mutations

Gene mutations resulting in abnormalities in proteins that regulate RNA processing have been discovered in patients with autosomal dominant familial ALS. Mutations in the TARDBP gene, which codes for TDP-43, have been found in 5% of patients with familial ALS.[37, 38, 39, 40, 41, 42, 43, 44] Mutations in the FUS gene are found in 3-4% of familial ALS cases.[74]

The mechanism by which these mutations cause ALS is distinct from that which takes place in SOD1 mutations. Although ubiquinated pathologic TDP-43 aggregates have been found in motor neuron cytoplasm of patients with sporadic ALS, they are not specific for this disease and have been found in affected nonmotor cells in patients with Guamanian parkinsonism-dementia complex,[45] British familial dementia,[46] and Alzheimer disease,[84] as well as in most patients with frontotemporal dementia.

Thus, formation of pathologic TDP-43 and its ubiquination may prove to be a mechanism of cell death that is not specific to ALS and is triggered by upstream processes, causing clinical pathology that depends on the cells affected. Conversely, TDP-43 deposition may prove to be a nonspecific defense mechanism involving an unsuccessful attempt to mitigate the action of the true instigators of cell death in a spectrum of neurodegenerative diseases, or it may be an epiphenomenon common to many forms of neurodegenerative diseases. Transgenic animal models with mutated TDP-43 do not develop ALS.

C9orf72 mutation

Studies of families with familial ALS in which some individuals also had frontotemporal dementia found linkage to a region on chromosome 9p21[85, 86, 87] Subsequently, a large genome-wide association study (GWAS) identified single-nucleotide polymorphisms (SNPs) associated with apparently sporadic ALS in the same region. This was confirmed in a further GWAS-wide association study of patients with apparently sporadic ALS and controls from 8 countries,[88] as well as in a Finnish study[89] using family-based samples.

In 2011, two groups reported the discovery of a hexanucleotide (GGGGCC) repeat expansion in the first intron of the chromosome 9 open reading frame 72 (C9orf72) gene­—the function of which is as yet unknown—as the cause of chromosome 9p21–associated ALS and frontotemporal dementia.[65, 66] This expansion of hexanucleotide repeats (from ≤23 in normal individuals to thousands in affected individuals) appears to be the most common genetic abnormality in ALS and in frontotemporal dementia.

The repeat expansion was found in 46.0% of patients with familial ALS, 21.1% with sporadic ALS, and 29.3% with familial frontotemporal dementia, in the Finnish population.[66] In their analysis of an extended North American clinical series, DeJesus-Hernandez et al found the C9orf72 expansion in 23.5% of patients with ALS cases and 11.7% of patients with familial frontotemporal dementia.[65]

More recent reports have found the C9ORF72 repeat expansion in 22-57% of familial ALS patients (depending in part on geographic origin) and in 3.6-7% of sporadic ALS patients.[67, 68, 69, 70] The frequency of the mutation varies widely even within Europe.[90]

Haplotype analysis of 5 European cohorts has shown that the hexanucleotide repeat expansion in C9orf72 had a single founder and arose around 6300 years ago.[90] The haplotype from which the mutation arose is intrinsically unstable, with an increased number of repeats.[90]

The phenotype of C9orf72 -mediated ALS shows distinct pathology, with p62-positive, TDP43-negative inclusions in the cerebellum and hippocampus.[91] Clinically, patients with this mutation have an earlier onset of disease and are more likely to have bulbar-onset disease, cognitive and behavioral impairment, and a family history of frontotemporal dementia than are ALS patients who carry other known mutations.

Other genes in familial ALS

Mutation in the ubiquilin 2 (UBQLN2) gene has been identified as a cause of X-linked dominant familial ALS and ALS with frontotemporal dementia.[92] This finding is of interest because it directly implicates the proteasome pathway in ALS pathogenesis.

Mutation in the profilin 1 (PFN1) gene has been identified in families with familial ALS.[79] The protein encoded by PFN1 plays a critical role in the conversion of monomeric (G)-actin to filamentous (F)-actin. Thus, the identification of this mutation provides further support for the role of the cytoskeleton and axonal transport in ALS pathogenesis.

Sporadic cases

The most widely accepted hypothesis regarding the cause of sporadic ALS posits that interactions between genetic, environmental, and age-dependent risk factors trigger disease onset. (See the image below.) ALS shows complex inheritance, which means that Mendelian, non-Mendelian, and apparently sporadic patterns of inheritance are seen. Smoking is the only environmental risk factor identified to date that may be considered “established.”[56, 57]



View Image

The genetic/environmental/age- and time-dependent interactions hypothesis for amyotrophic lateral sclerosis (ALS). Risk factors operate upstream to a ....

Several lines of evidence support the hypothesis that genetic risk factors may influence disease initiation in apparently sporadic ALS, apart from finding known Mendelian gene mutations in patients with no family history. Twin studies show a genetic contribution to apparently sporadic ALS, with heritability of 0.61.[10] Increased risk for ALS and (in some studies) clustering of non-ALS neurodegenerative disease is found in relatives of patients with apparently sporadic ALS.[73, 93, 94, 95]

Environmental risk factors

Smoking

Cigarette smoking is the only exogenous risk factor that may be considered an established risk factor for ALS[56] (level A conclusion, based on 3 class II studies[96, 97, 98] and 1 class III study[99] ). In addition, a population-based study from the Netherlands demonstrated that current smokers are at increased risk for ALS, with an odds ratio of 1.38, and have shorter survival.[57]

Some aspects of the findings in these studies suggest that smoking may be implicated directly in causing the disease. Overall, studies have shown that active smokers have approximately double the risk of developing ALS compared with never smokers. Former smokers have an intermediate risk.

Identifying smoking as an established risk factor for ALS has the following major implications:

Focusing on processes at the initiation of sporadic ALS and close to its initiation, in order to account for its early spread within the motor system, may provide new avenues to treatments to stop its progression. This approach may augment the current focus on processes that occur relatively late in the course of the disease and cause the death of motor neurons directly.

No other risk factor has attained the level of certainty that smoking has with regard to its association with ALS. Trauma, physical activity, residence in rural areas, and alcohol consumption are probably not risk factors for ALS.[54] Indeed, in the population-based study from the Netherlands noted above, current alcohol consumption was associated with a reduced risk of ALS.[57]

Military service

Putative risk factors include service in the US military during World War II, the Korean War, and Vietnam,[100] as well as deployment to the Persian Gulf in the 1991 Persian Gulf War.[101] However, close scrutiny has cast doubt on the quality of the evidence supporting the role of these factors in triggering ALS.[101, 102, 103, 104, 105] More recently, a 13-year follow-up study found no excess of ALS among Gulf War veterans.[106]

Other putative exogenous risk factors have not risen to the level of probable. These include exposure to pesticides,[107] postmenopausal hormone use,[54, 108] and physical exercise.[109]

Sports

A possible increased risk of ALS in Italian professional soccer players was reported in 2005.[110, 111] Initially, it appeared that the apparent increase in risk may have resulted from underestimation of the expected number of cases of ALS.[112, 113, 114, 115, 116] However, a 5-year extension of the follow-up of this cohort showed an unambiguous excess of cases of ALS; 8 cases (including 3 new ones) were reported, even though the number of expected cases was 1.24.[117]

However, the ALS in the professional soccer players displayed atypical features; ie, 5 of the 8 cases had a bulbar onset, and 5 had been diagnosed between 2000 and 2006 (compared with 3 between 1980 and 1999 and none between 1970 and 1979). The authors concluded that the predilection for soccer players to develop ALS derives from a complex interplay between genetic predisposition for physical endurance and external factors such as drugs or herbicides.[117]

A subsequent report suggested that a form of motor neuron disease might develop in individuals, such as boxers, who have sustained repetitive injuries to the brain and developed chronic traumatic encephalopathy (CTE).[118] However, the claim that this was a novel form of motor neuron disease was challenged; instead, the possibility that these cases represented coincidental co-occurrence of ALS was proposed as more plausible.[119, 120, 121]

More recently, a study in retired US National Football League (NFL) players showed that, while their overall mortality was 50% less than expected, their mortality from neurodegenerative diseases was higher than expected. In particular, mortality from ALS and Alzheimer disease was 4-fold higher than expected.[122] These results are based on 7 individuals who died with Alzheimer disease and 7 individuals who died with ALS, out of a cohort of 3439 individuals.

The interpretation of this number as excessive appears to be a consequence of the method used to calculate the expected rate,[112] which may result in underestimation of that rate. In addition, when an apparent excess of neurodegenerative disease appears in the context of greatly reduced overall morality, the emergence of neurodegenerative deaths due to loss of competing causes of mortality needs to be considered.

These reports raised the question whether trauma to the head may be a risk factor for ALS. An evidence-based review of the literature concluded that for instances of isolated head trauma this was not the case.[123]

A subsequently published national population–based case-control study from Sweden found no association of ALS with severe head injury occurring more than 3 years before ALS diagnosis, nor was ALS associated with subtypes of head injury or repeated injuries occurring more than 3 years before diagnosis.[124] Exclusion of injuries occurring within 3 years of diagnosis is necessary to have some assurance that the injury occurred before biologic onset of the disease, which likely precedes clinical onset by several years.[125]

A population-based study from Rochester, Minnesota, showed no increased risk of neurodegenerative diseases among 438 players who played American football in high school between 1946 and 1956, despite poorer protective equipment and less regard for concussions compared with today, and no rules prohibiting head-first tackling.[126]

The balance of the evidence supports the conclusion that head trauma in general, including repetitive head trauma, is not a risk factor for ALS. The special circumstances of the Italian soccer players are uncertain, as their form of ALS was unusual, with most experiencing bulbar onset.

The special circumstances of the NFL players are also uncertain, as the data are sparse, and it is not clear that the numbers represent a true excess. Risk factors other than the sport itself may be involved, including local environmental risk factors or ingestion of testosterone, anabolic steroids, or other drugs.[117, 127]

One report indicated that head injury does not alter disease progression or neuropathological outcomes in patients with ALS.[128]

Western Pacific ALS

Most of the research on Western Pacific ALS has focused on Guam. Ingestion of food products derived from the false sago palm, Cycas micronesica (recently separated from Cycas circinalis), was proposed by nutritional anthropologist Marjorie Whiting as the process predisposing to the development of this form of ALS.[129] Despite the cycad nut being subjected to an elaborate preparation process to rid it of toxins before being used as a substrate for flour, some toxic factor was presumed to remain.[130]

In addition, the cycad nut is consumed by the flying fox (a type of bat), which used to be part of the Chamorro peoples' diet on Guam. Toxins from the cycad nut may have been concentrated (bioamplified) in the bat and delivered to the human consumer. The consumption of the flying fox was higher in the mid-20th century than it is now.[131] Most flying foxes consumed in Guam currently are imported.

An epidemiologic study in Guam provided evidence consistent with that hypothesis; its conclusions have been challenged,[131] but the challenges themselves have been questioned.[130]

The nature of the putative toxic component of cycad that may be responsible for delayed-onset neurodegenerative disease has also been a matter of intense debate. One hypothesis is that cycad contains excitotoxic amino acids that do not exert an effect until many years after they have been ingested.[132, 133, 134]

An alternative hypothesis is that alkylating components induce changes in nucleic acids[58, 59] that increase the likelihood that subsequent, additional, age-dependent nucleic acid changes trigger disease onset in Guamanian ALS/Parkinson-dementia complex (ALS/PDC).

Epidemiology

Occurrence in the United States

Approximately 5600 people in the United States are diagnosed with ALS each year. The annual incidence is 2-3 per 100,000 population; this is about equal to that of multiple sclerosis and 5 times higher than that of Huntington disease. It is estimated that as many as 18,000 Americans may have ALS at any given time.

The lifetime risk for developing ALS for individuals aged 18 years has been estimated to be 1 in 350 for men and 1 in 420 for women.[112] These estimates are close to those reported from 4 European registries, using different methods.[135, 136, 137]

International occurrence

Age-adjusted European incidence data are similar to those for members of the US population who are of European descent.[135, 138] Most variability between countries has been attributed to different age composition or differences in case finding. More recent data, however, suggest that ethnic variability in disease incidence exists[139, 140] that may not be explained entirely by differences in case finding, with lower incidence in nonwhites or individuals of mixed ethnicity. Although this possibility is not supported by all studies, it merits further examination.[141]

Finland has one of the highest rates of ALS in the world; the disease occurs in the Finnish population nearly twice as frequently as it does in other populations of European ancestry.[89] A study from Finland found 2 clusters of cases based on geographic location at time of death and a cluster based on time of birth that closely matched one of the time-of-death clusters.[142] It was recognized that these results could be consistent with either a genetic or environmental cause. With the discovery of the hexanucleotide repeat expansion in C9orf72, most cases of ALS in Finland have been confirmed to be due to genetic factors.

Race-related demographics

In the United States, ALS affects whites more often than nonwhites; the white-to-nonwhite ratio is 1.6:1.[139] Uncertainty surrounds this finding, however, as it has been considered to be an artifact of reduced case-finding in nonwhites. More convincing evidence for racial differences has come from an epidemiologic study in Cuba.[140]

Small population clusters have been identified that have higher rates of ALS. The Chamorro people of Guam and Marianas Island, residents of the Kii peninsula of Japan’s Honshu Island, and the Auyu and Jakai people of southwest New Guinea have a higher incidence of ALS than is found in populations elsewhere in the world.[8] The Chamorro population in Guam in the mid-20th century had an annual incidence of ALS (often in association with parkinsonism and dementia) as high as 70 cases per 100,000 (see Pathophysiology).[132] The incidence has since decreased to 7 cases per 100,000.

Sex- and age-related demographics

For most of the lifespan, the incidence of ALS is higher in men than in women, with an overall male-to-female ratio of 1.5-2:1.[18] Later in life, the incidence tends to equalize; this occurs at age 40-50 years in some populations and after the age of 65-70 years in others.[143]

Onset of ALS may occur from the teenage years to the late 80s; the incidence rises with increasing age until approximately age 75-80 years. Mean age of onset of sporadic ALS is 65 years; mean age of onset of familial ALS ranges from 46-55 years.

Prognosis

ALS is a fatal disease. Median survival is 3 years from clinical onset of weakness. However, longer survival is not rare. About 15% of patients with ALS live 5 years after diagnosis, and about 5% survive for more than 10 years. Long-term survival is associated with a younger age at onset, being male, and limb (rather than bulbar) symptom onset. Rare reports of spontaneous remission exist.[144] In familial ALS that results from an alanine-to-valine mutation in codon 4 of the SOD1 gene (A4V mutation), average survival is 12 months from disease onset.[81]

Regionally limited forms of motor neuron disease (ie, brachial biplegia, lumbosacral biplegia, and progressive bulbar palsy [PBP] that remains restricted[145] ) progress slower than does classic ALS. Progressive muscular atrophy (PMA), distinct from classic ALS because of lack of upper motor neuron (UMN) findings, progresses at the same rate as classic ALS. UMN-predominant ALS progresses at a slower rate. Survival in cases of primary lateral sclerosis (PLS) is measured in decades. These observations suggest that it is the loss of LMNs that determines the prognosis.

Frontotemporal executive dysfunction may precede or follow the onset of ALS, but most patients with ALS do not have overt dementia, and cognitive impairment is usually subtle.[146] Approximately 15% of patients with ALS meet criteria for frontotemporal dementia (FTD). Patients with ALS associated with FTD have shorter survival than do those with ALS alone.[147, 148]

Measures of disease progression

The most effective predictor of survival is the rate of observed disease progression, estimated via the time between symptom onset and diagnosis or evaluated by employing measures such as the ALS Functional Rating Scale (ALSFRS).[149] Estimates derived as a ratio in which the numerator is a measurement of functional loss—as obtained using the ALSFRS-Revised (ALSFRS-R), the forced vital capacity (FVC) as percent of predicted, strength, or motor unit number estimates (MUNEs)—and the denominator is time elapsed from disease onset to the time of measurement, may provide more individualized prognostic information.[150, 151, 152, 153, 154]

Roche et al have proposed a system of stages, the timing of which is standardized as proportions of elapsed time through the course of ALS.[155] The milestones, and their typical time of occurrence, are as follows:

It may be seen that most patients are diagnosed at a point at which ALS has extended to involve at least 2 regions and that the needs for gastrostomy and noninvasive ventilation (NIV) are usually recognized almost at the same time. These percentages are consistent with the figures of 12 months’ mean time from onset to diagnosis and 3 years’ mean duration of disease from onset to death.

Progressive muscular atrophy

Most patients with PMA have a course indistinguishable from that of patients with classic ALS (except for the absence of UMN findings). However, some patients may have a longer course, particularly those with flail arm or flail leg syndrome.[71]

Kim et al concluded that PMA should be considered a form of ALS.[156] Review of the medical records of 91 patients with PMA and 871 with ALS showed that patients with PMA were more likely to be male, to be older, and to live longer than those with ALS, but risk of death increased with age at onset in both patient groups and UMN signs developed in 22% of patients with PMA within 61 months after diagnosis.[156]

In the study, demographic and other clinical variables did not differ at diagnosis between patients who did or did not develop UMN signs. In PMA, as in ALS, the factors present at diagnosis that predicted shorter survival were greater number of body regions affected, lower FVC, and lower ALSFRS-R score.

Discussing prognosis with the patient

Patients the author has surveyed have indicated ambivalence about being offered individualized information early in the course of the disease (when it may matter most).[157] The author does not offer patients individualized prognostic information at the first visit. When patients do request it, he asks them to consider the possible implications of the answers they may receive and to ask him again at a subsequent visit. It is also helpful to discuss the implications of the rate of disease progression, based on the joint observations of the patient and the physician.

Patient Education

The following education resources are available to patients with ALS:

Informational web sites include the following:

For patient education information, see the Brain and Nervous System Center, as well as Amyotrophic Lateral Sclerosis (ALS, Lou Gehrig’s Disease) and Advance Directives.

History

The diagnosis of amyotrophic lateral sclerosis (ALS) is primarily clinical. When the disease has progressed far in its course and involves many parts of the body, the patient’s appearance and the findings on the neurologic examination may provide sufficient evidence for the diagnosis. When a patient presents with the first symptoms, however, making the diagnosis is not straightforward.[19]

ALS may be suspected whenever an individual develops insidious loss of function or gradual, slowly progressive, painless weakness in 1 or more regions of the body, without changes in the ability to feel, and no other cause is immediately evident.

In lower motor neuron (LMN) involvement, fasciculations may occur early on in the disease, particularly in the tongue and limbs. Patients with upper motor neuron (UMN) involvement generally are hyperreflexic and stiff. Reflexes may be diminished due to LMN involvement. UMN symptoms may include spasms and sudden, uncontrolled straightening movements of the lower limbs.

In 75-80% of patients, symptoms begin with limb involvement, while 20-25% of patients present with bulbar symptoms. For those with limb involvement at presentation, the frequency of upper limb versus lower limb involvement is approximately equal.

Patients with upper limb onset have twice the likelihood for onset in the dominant arm, compared with the nondominant arm. There is equal likelihood for presentation in either lower extremity. Women have a greater frequency of bulbar (speech dysfunction) onset than men. These observations suggest a greater likelihood for network dismantling to start where there is a better-developed, or more complex, cortical network.

Patients who have lower limb onset initially may complain of tripping, stumbling, or awkwardness when running. Foot drop is common, and patients may report a "slapping" gait.

Persons with upper limb onset may experience reduced finger dexterity, cramping, stiffness, and weakness or wasting of intrinsic hand muscles. This may lead to difficulty with actions such as buttoning clothes, picking up small objects, or turning a key. These patients may develop wrist drop.

As ALS progresses, muscle atrophy becomes more apparent, and spasticity may compromise gait and manual dexterity. Immobility, if coupled with spasticity, may lead to the development of painful joint contractures. Muscle cramps are common. In some patients, persistent stiffness or cramping of muscles may stress the related joints and the back. This can usually be ameliorated with medications and physical therapy exercises to relax the muscles and maintain joint range of motion.

Bulbar involvement

A mixture of spastic and flaccid components may characterize speech, resulting in a dysarthria with severe disintegration and slowness of articulation. Hypernasality occurs from palatal weakness, and patients may develop a strained, strangled vocal quality. With time, speech may be lost, and patients may become dependent on other forms of communication, such as writing, communication boards, or speech-generation devices.

Patients with bulbar involvement may develop swallowing difficulties (dysphagia). Swallowing liquids requires the greatest oropharyngeal muscle control; therefore, patients usually report more difficulty with liquids than with solids. Aspiration or choking during a meal may occur.

Drooling affects some patients and results from a combination of excessive salivation, poor swallowing, and poor lip control. The problem can usually be ameliorated effectively with oral medications or a scopolamine patch, but other methods, such as salivary gland irradiation or injection of botulinum toxin, are sometimes required.

Pseudobulbar symptoms

These consist of exaggerated, involuntary emotional responses. The response may be of one type (laughter or crying) or, less commonly, alterations in emotive expressions. Episodes of intense laughter may be followed immediately by tears. The patient's response often does not correspond to an apparent social stimulus or the current psychosocial situation; it may instead be an exaggerated response to a minor trigger. The patient is aware of the lack of control. Symptoms can often be ameliorated by medications.

Preserved functions

Certain motor neurons usually are spared in ALS, which means that some functions are preserved. Most patients retain extraocular movements and bowel and bladder control. With progressive disease, patients may develop problems with urge incontinence and constipation because of weak abdominal musculature, but sphincter control generally is unaffected.

Since the disease primarily involves motor neurons, sensory function typically is preserved, although a minority of patients complains of some numbness and paresthesias. Abnormalities have been reported on sensory nerve conduction studies in a small number of patients with ALS,[159] but these findings often reflect the presence of an unrelated, coexistent condition.

Skin integrity in ALS usually is maintained, primarily due to the combination of preserved sensory function and continued control of bowel and bladder function. Some studies of patients with ALS have found morphologic changes in the skin that are complex and poorly understood but that may contribute to preservation of skin integrity.

Family history

Obtaining a thorough family history is pertinent in the examination of all patients. Patients with a family history of Mendelian ALS may be considered to have definite ALS as soon as any evidence of motor neuron disease arises that cannot be accounted for by an alternative explanation, regardless of the extent of involvement. Some experts, however, require that the abnormal gene be demonstrated in the patient. Genetic testing is typically recommended when a mode of inheritance, most often autosomal dominant, is recognized, but the gene has not yet been identified in the family.

A family history of ALS in a second- or third-degree relative or any family history of frontotemporal dementia should also be regarded as evidence in support of a diagnosis of familial ALS.[160, 161]

Physical Examination

The symptoms that some patients with ALS may experience and the signs that are found on their neurologic examination are summarized below. Not all patients experience all symptoms or have all signs.

Motor neuron manifestations

Findings reflecting upper or lower motor neuron dysfunction include the following:

Findings reflecting UMN dysfunction include the following:

Findings reflecting LMN dysfunction include the following:

The key finding in an involved limb is a combination of upper and lower motor neuron dysfunction, as when a weak, atrophic, fasciculating muscle also has increased tone and hyperreflexia.

Emotional and cognitive symptoms

While the symptoms of motor dysfunction in ALS are best recognized, affecting all patients with the disease, a fair proportion of patients also experience emotional and special cognitive difficulties. Emotional manifestations of ALS include involuntary laughing or crying and/or depression. Cognitive difficulties involve executive function impairment and/or behavioral changes.

Frontotemporal executive dysfunction may precede or follow the onset of ALS, but most patients with ALS do not have overt dementia, and cognitive impairment is usually subtle.[146] Approximately 15% of patients with ALS meet the criteria for frontotemporal dementia (FTD).[147, 148]  An additional 30-40% may have cognitive impairment detectable by special testing. FTD interferes with patients' acceptance of supportive treatment recommendations and makes them harder to manage. On the average, ALS patients who have FTD live 12 months less than comparable patients without FTD.[163]

Bulbar symptoms

Bulbar symptoms manifesting as dysarthria or dysphagia are the most common ALS presentation next to limb involvement, affecting 20-25% of patients. Rarely, patients with ALS may present with respiratory muscle weakness, generalized weakness, or difficulty with head control. Those with respiratory muscle weakness may develop disturbed nocturnal sleep and exhibit excessive daytime sleepiness.

Truncal difficulties

Patients with axial truncal weakness have difficulty maintaining an erect posture when standing; to assist in attaining a standing position, they may support their torso by “walking” their hands up their thighs. Some patients feel more secure when pushing a heavy object on wheels, such as a grocery cart.

Diagnostic Criteria

El Escorial criteria

According to criteria from the El Escorial World Federation of Neurology, the diagnosis of ALS requires the presence of the following:[164]

  1. Signs of lower motor neuron (LMN) degeneration by clinical, electrophysiological or neuropathologic examination,
  2. Signs of upper motor neuron (UMN) degeneration by clinical examination, and
    • Electrophysiological evidence of other disease processes that might explain the signs of LMN and/or UMN degenerations; and
    • Neuroimaging evidence of other disease processes that might explain the observed clinical and electrophysiological signs.
     
     

Awaji criteria

Since the introduction of the revised El Escorial criteria, attempts have been made to permit earlier diagnosis of ALS. Additional approaches have been proposed for the analysis of clinical and electrophysiological data in order to facilitate earlier diagnosis of probable ALS.[165]

The Awaji criteria consider equally the clinical and neurophysiological evidence of LMN involvement. Acute and chronic signs of denervation should also be taken as equivalent, with only one type of change necessary to imply limb involvement. In parallel, it has become evident that most patients with possible ALS who have progressive disease with lower motor neuron findings and in whom other causes of focal disease have been excluded are destined to progress and manifest the full spectrum of the disease, so that they may be considered to have ALS including for the purpose of enrollment into clinical trials. Finally, patients characterized as having clinically suspected ALS, due to absence of upper motor neuron findings, also named PMA (progressive muscular atrophy), have clinical courses that are usually indistinguishable from those of patients with ALS. The exceptions are patients with specific syndromes (e.g., brachial biplegia, lumbosacral biplegia) who tend to have a slower course, and some patients with particularly slow or fast forms of lower motor neuron disease.

In addition, efforts have been made to introduce biological and radiological markers to assist in diagnosing ALS. However, most of the work evaluating these markers has been done in patients in whom the diagnosis was not in question. It is impossible to say if these markers would help make the diagnosis of ALS in individuals who do not meet clinical and electrophysiological criteria for the disease.

In summary, earlier diagnosis of ALS is possible today in individual cases, relative to years past, if alternative causes for early, limited, progressive disease are excluded. The requirements to exclude alternate diagnoses and observe progression are more stringent in very early disease. However, in aggregate reported data of ALS clinic patients and of patients enrolled into clinical trials the average time from clinical disease onset to diagnosis has remained unchanged, and continues to hover around 12 months.  

WFN criteria

The World Federation of Neurology (WFN) has developed an algorithm that combines the clinical and, in some cases, electrophysiologic findings in patients with ALS. This is used to express in each patient the extent of disease involvement at the time of the examination (the revised El Escorial criteria).[1]

The WFN uses adjectives that in usual speech reflect the degree of certainty. However, when these adjectives are applied in the context of diagnosing patients with ALS, they need to be understood as reflective of the degree of clinical involvement rather than the degree of certainty in the diagnosis, particularly if no alternative diagnosis has been found and the disease has progressed beyond a single limb. This distinction can be confusing for patients.

The WFN criteria recognize the following 4 regions, or levels, of the body (see the image below)[19] :

Although the names of these regions appear to describe neurologic segments, the terms actually refer to functional regions of the body. For example, the small muscles of the hands are innervated by thoracic motor neurons but are counted in the cervical region.



View Image

The 4 regions or levels of the body. Bulbar (muscles of the face, mouth, and throat); cervical (muscles of the back of the head and the neck, the shou....

The WFN categories are as follows:

Qualifiers

Originally, it was thought that the degree of certainty of diagnosis was increased by the number of body segments that demonstrated UMN and LMN signs; hence, the choice of qualifying adjectives. The criteria were designed to be usable even if no adjunctive testing was available, in which case the qualifiers did have implications in their literal sense.

However, the qualifiers have effectively lost their meaning, in terms of making the diagnosis, as a result of the following:

The qualifiers continue to reflect the extent of involvement at time of diagnosis or subsequent evaluation. Recruitment to clinical trials previously was limited to patients with definite or probable ALS (including probable, laboratory-supported ALS), but more recent clinical trials have included patients with “possible” ALS. Their progression has been affirmed explicitly.

Patients with progressive bulbar palsy (PBP) may be classified, while the disease is restricted to the bulbar region, as having “suspected ALS” if only UMN or LMN abnormalities are evident, and as “possible ALS” if there is UMN and LMN involvement. When neurophysiologic or clinical spread beyond the bulbar level is evident, the condition would be reclassified as probable; laboratory-supported, probable; or definite ALS.

In day-to-day practice, clinicians will inevitably use the term suspected ALS whenever they believe ALS to be a possibility, regardless of the extent of clinical involvement at the time, and may or may not use the WFN qualifiers when they conclude that the patient has ALS. This leads to clearer terminology in practice; the patient is either suspected of having ALS or confirmed as having the disease, or a different diagnosis is made.

Revisions

A group of experts has proposed that the WFN criteria be further revised, primarily by giving equal weight to clinical and electrophysiologic evidence of denervation.[166] However, even when there is no uncertainty, this system still retains use of the term “probable ALS,” which denotes uncertainty. There are other differences between this system and the previous one, but for the present, abandoning the current WFN would probably be premature.[1]

Classification and prognosis

The term “suspected ALS” has special meaning in the WFN classification system because it is applied to patients with a pure UMN presentation, particularly if they cannot yet be diagnosed with primary lateral sclerosis, and to patients with a pure LMN presentation (particularly early in their presentation) before enough time has elapsed to be sure that their condition will remain restricted to LMNs and might therefore be more precisely described as PMA.

This has a practical implication, because patients with primary lateral sclerosis have a course that is measured in decades (approximately 20 y). Some patients with a predominantly UMN form of ALS may also have a longer course than those with classic ALS.

Since the distinction between a diagnosis of PMA and one of laboratory-supported probable ALS hinges on the identification of 1 UMN sign at some point in the patient’s disease, this distinction may be primarily of significance to researchers. For clinicians and patients, rate of progression is likely to be of greater concern, as it is the factor that determines the patients’ course and outcome.

Approach Considerations

Amyotrophic lateral sclerosis (ALS) may not lend itself to a quick definitive diagnosis early in its presentation. Often, neurologists need many months to exclude all other possible diagnoses in a patient presenting with upper and lower motor neuron signs.

Nerve conduction studies and needle electromyography (EMG) are useful for confirming the diagnosis of ALS and for excluding peripheral conditions that resemble ALS.

Laboratory tests are performed primarily to rule out other disease processes; results generally are normal in ALS.

Biochemical markers in blood are used almost routinely to identify diseases that could mimic ALS. Examination of cerebrospinal fluid usually is not necessary unless the patient has a pure upper motor neuron (UMN) or pure lower motor neuron (LMN) presentation, in which case it can be helpful in excluding inflammatory conditions, neoplastic infiltrations, or infections.

Genetic testing

Genetic testing may be performed to identify genetic defects in some familial types of ALS, as well as other inherited motor neuron diseases. In the future, genetic testing may become more routine, given recent research showing that in some populations the C9orf72 mutation is present in a high proportion of patients with no family history of ALS.

The role of genetic testing in patients with sporadic disease is debated among ALS experts.[167] Patients with sporadic disease who are considering genetic testing should take the time, through genetic counseling, to study the implications for themselves and for their first-degree relatives, Ideally, the first-degree relatives should be involved in the counseling process as the results of genetic testing impact them more than they impact the patient.

Imaging

Imaging studies need to be tailored to the patient’s clinical presentation. Neuroimaging may include computed tomography (CT) scanning or magnetic resonance imaging (MRI) of the brain and spinal cord.

Muscle or nerve biopsy

Muscle biopsy is needed only rarely but may be considered if the presentation of ALS is atypical. The results will confirm the presence of signs of denervation and reinnervation or may lead to an alternative diagnosis.

The presence of small, angular fibers is consistent with neurogenic atrophy (denervation). Fiber-type grouping is consistent with reinnervation.

Needle Electromyography and Nerve Conduction Studies

It is common to examine at least 3 levels—cervical/thoracic/lumbar paraspinal muscles—and bulbar muscles, as follows:

EMG may show fibrillation and fasciculation potentials. The motor units may be polyphasic and have high amplitude and long duration.

Motor unit recruitment

The pattern of recruitment of motor units may be abnormal due to loss of anterior horn cells and a reduction in the number of viable motor axons to activate the muscle(s) involved. This loss results in increased firing frequency of surviving motor units, because fewer anterior horn cells (motor axons) are available to be activated as the amount of effort increases.

Muscle innervation

Recently reinnervated muscles demonstrate variability of morphology on needle EMG examination. This is because of sprouting nerve terminals that are unmyelinated, have slower conduction, and may cause intermittent conduction block. Increased polyphasia of voluntary motor unit action potentials is a result of asynchronous firing of reinnervated, unmyelinated muscle fibers resulting from slowed terminal nerve conduction.

Over time, the voluntary motor unit action potentials increase in size and duration because of collateral sprouting, bringing more muscle fibers into the motor unit. With maturation of the terminal sprouting, voluntary motor unit action potentials increase in size in reinnervated muscle fibers, restoring their size and a greater degree of motor firing synchrony due to myelinization of terminal nerve fibers and more rapid terminal nerve conduction.’

Denervation

Signs of active and chronic denervation are likely to be observed. Fibrillation potentials and positive sharp waves represent active denervation. Chronic denervation is demonstrated by evidence of large motor unit potentials with increased duration and amplitude, as well as polyphasic potentials, reduced recruitment, a reduced interference pattern with firing rates higher than 10 Hz, and unstable motor unit potentials.

Multispike and fasciculation potentials

Complex, repetitive discharges occur in ALS of long duration, as they do in other chronic neurogenic atrophic conditions. These are regularly discharging multispike potentials that are time-locked. Other than an EMG finding associated with a chronic neurogenic atrophic condition, this finding has no other unique significance.

Fasciculation potentials are seen frequently but not invariably in ALS. Their presence is not specific to ALS; they may occur in other conditions, some completely benign.

Conduction studies

Motor and sensory nerve conduction studies are performed primarily to rule out other disorders. In patients with predominantly LMN findings, the presence of conduction block may point to treatable diseases such as multifocal motor neuropathy or motor chronic inflammatory demyelinating polyneuropathy.

Sensory nerve conduction studies are usually normal. Less than 10% of patients with ALS have abnormal sural sensory nerve conduction studies. Patients over age 60 years commonly lose the sural sensory nerve action potential (SNAP), but this is attributable to normal aging.

In late stages of ALS, LMN involvement may be extensive; in such cases, compound muscle action potentials may be reduced. Hallmark findings in the electrodiagnosis of ALS are normal sensory nerve conduction studies and abnormal motor nerve conduction studies, with reduced motor compound muscle action potentials.

Neuromuscular transmission instability of collateral nerve terminal sprouts in ALS patients presents as a decrement with slow repetitive stimulation. This instability is present in less than 50% of patients with ALS. The decrement in ALS is usually less than 10%. In contrast, compound muscle action potential decrements greater than 20% on repetitive stimulation of motor nerves are seen in myasthenia gravis. In ALS, this is usually a late finding, whereas in myasthenia gravis, this is an early finding.

Electrophysiologic features compatible with UMN involvement include an increase of up to 30% in central motor conduction time determined by cortical magnetic stimulation. However, central electrophysiologic studies are currently not part of the routine evaluation of patients with ALS, because whether emergence of central studies abnormalities precedes that of clinical signs of UMN involvement has not been determined.

Low or irregular firing rates of a few voluntary motor unit action potentials on maximal effort may be seen during routine needle examination of UMNs. However, this feature is nonspecific. It may be seen in other settings if patients have difficulty activating specific muscles because of UMN disease, pain inhibition, or poor cooperation.

Motor unit number estimate

The motor unit number estimate (MUNE) is a nerve conduction study technique that can quantify the numbers of motor units innervating an individual muscle.[168] It may be used to help with the diagnostic process in rare cases in which clinical or electrodiagnostic LMN involvement otherwise cannot be shown. For example, MUNE showing numbers below the lower limit of normal in distal upper and lower extremity muscles establishes the diagnosis as ALS.

MUNE may be used to separate patients into faster and slower ALS progression groups.[169] However, other measures of disease progression that are easier to obtain, such as those derived using the ALS Functional Rating Scale-Revised (ALSFRS-R), may serve this purpose. Also, patients the author has surveyed have reflected ambivalence or reluctance at the prospect of receiving prognostic information early in the course of the disease, which is when such information is of greatest relevance.[157]

MUNE is appealing as a quantifiable, physiologic measure of disease progression that is independent of patient effort and is a measure of pure LMN involvement. Nevertheless, the error inherent in the estimation process precludes its use as the primary measure of efficacy of putative, mechanism-specific interventions to slow disease progression.

Characteristics of other neuropathies

Multifocal motor mononeuropathy

Other disease processes may be suggested by their characteristic electrophysiologic presentation. Multifocal motor mononeuropathy may be suggested by the following:

Chronic inflammatory demyelinating polyradiculoneuropathy

This condition may be suggested by the features listed above, together with the following:

Sensorimotor peripheral neuropathy

A generalized axonal sensorimotor peripheral neuropathy may be suggested if motor and sensory fibers appear affected equally without excessive slowing or if sensory fibers are affected more than motor fibers. However, it must be recognized that mild sensory abnormalities may be found occasionally in patients with ALS without sensory symptoms and that the lower limits of normal in persons aged 60 years or older are lower than those for younger individuals.

Inclusion body myositis

Inclusion body myositis may be suggested by a characteristic pattern of distribution of affected muscles and by a mixed pattern of large and small motor unit potentials on needle examination. Other myopathies may also be suggested if small, rapidly firing motor unit potentials are found, rather than those characteristic of a neurogenic process.

Primary lateral sclerosis and monomelic amyotrophy

Primary lateral sclerosis may be suggested if LMN involvement is minimal or absent, particularly if that remains the case 3 years (or more conservatively, 5 years) after clinical onset of disease. Monomelic amyotrophy may be suggested if no evidence of disease is found outside 1 limb several years after its onset.

Laboratory Studies

Laboratory tests sometimes ordered in the evaluation of a patient with possible ALS include anti-ganglioside M1 (anti-GM1) antibodies, as these can be seen in patients with multifocal motor neuropathy with conduction block. Vitamin B12 and folate levels, HIV status, Lyme serology, and creatine phosphokinase (CPK) determinations may also be performed when indicated by clinical circumstances. The CPK level may be elevated in ALS, but this is not a diagnostic finding.

The following tests may also be considered:

If myasthenia gravis is under consideration, antiacetylcholine receptor antibody and anti-muscle specific kinase (MuSK) antibody assays should be ordered.

Urinary 24-hour collections for heavy metals may be requested if there is reason to suspect recent exposure. Hexosaminidase A in urine may be checked when adult Tay-Sachs is suspected strongly.

Lyme disease serology may be considered if clinical data suggest that the patient had untreated Lyme disease. However, the history, rather than laboratory testing, drives the diagnosis in Lyme disease.

Genetic testing

In patients with familial ALS, genetic testing may be requested after appropriate counseling. The results of genetic testing may affect not only the patient, but family members as well.

Tests for the SOD1, TARDBP (coding for TDP-43), FUS, ANG, C9orf72, and FIG4 genes, as well as for the gene causing Kennedy disease, are available commercially. Patients with other forms of familial ALS may be referred to receive further information from centers with a research interest in familial ALS.

CT Scanning and MRI

Brain or spinal MRI may be done to rule out structural lesions and neurologic conditions that sometimes account for early clinical features seen in patients suspected of having ALS (eg, multiple sclerosis, brainstem strokes, tumors, spinal radiculopathy). Results of these studies generally are normal in patients with ALS.

Magnetic resonance spectroscopy may also be used, but it has a high false-negative rate. CT scanning with myelography may be needed in patients in whom an MRI cannot be performed safely (eg, because of the presence of a pacemaker, an implantable defibrillator, or metal fragments).

The value of positron emission tomography (PET) scanning and functional MRI in ALS is being investigated. Imaging studies may not be necessary in patients presenting with advanced disease.

Functional Assessment

ALS typically progresses within the area first affected and then to adjacent, contiguous regions. As it progresses, patients’ function and independence diminish. When respiratory muscles are affected, patients may be supported using noninvasive or invasive measures. The majority of patients die of ventilatory failure, most having chosen not to opt for long-term invasive mechanical ventilation. Less than 5% of patients die of other causes, such as a heart attack, a serious infection, or blood clots that migrate to the lungs.

The pace of disease progression varies from patient to patient, and the symptoms depend on the muscles affected. Regular monitoring of the patient’s course assists in directing treatment.

Standardized assessment of patients with ALS was facilitated by the development of the ALS Functional Rating Scale, a 10-item standardized questionnaire.[170] It was revised to give greater weight to respiratory involvement and became the 12-item ALSFRS-R,[171, 172] which is used extensively.

In the ALSFRS-R, functions mediated by cervical, trunk, lumbosacral, and respiratory muscles are each assessed by 3 items. Each item is scored from 0-4, with 4 reflecting no involvement by the disease and 0 reflecting maximal involvement. The item scores are added to give a total.

Total scores reflect the impact of ALS, as follows:

For an individual patient, loss of only 8-10 points on the ALSFRS-R may have severe implications; eg, if respiratory or bulbar function bears the brunt of the losses. A minor limitation of the scale is that it has a floor effect in terms of measuring disease progression in quadriparetic, ventilator-dependent patients.

Most physicians caring for patients with ALS use a measure of the patient’s breathing ability to follow the course of their disease. In the United Kingdon, a governmental clinical guideline exists for monitoring respiratory weakness, with clear points for referral or intervention.

Of the tests of pulmonary function, vital capacity is used most commonly. Additional measures, such as maximal inspiratory and expiratory pressures, arterial blood gas measurements, and overnight oximetry, may provide earlier evidence of dysfunction. Comparison of vital capacity in the upright and supine positions may also provide an earlier indication of weakening ventilatory muscle strength.

Approach Considerations

Treatment of amyotrophic lateral sclerosis (ALS) may be divided broadly into the following[19, 173, 174] :

Patient education can be enhanced by referral to multidisciplinary clinics staffed by specialists with special interest in ALS, by educational materials prepared for patients and families by national organizations in the United States[175, 176] and other countries or by individual experts,[19] and by patient participation in local support groups.

Inpatient and Outpatient Care

Outpatient care

Most of the care of patients with ALS may be delivered in the outpatient setting. Often, guidance can be provided by a neurologist, physiatrist, or palliative care physician with special interest in the disease.

Multidisciplinary clinics can provide "one-stop shopping", allowing patients to receive all assessments and recommendations in the course of a single visit; most of these clinics provide a combination of on-site and off-site services. In the United States, Muscular Dystrophy Association/ALS centers and ALS Association certified centers have been established at several major medical centers, and in the United Kingdom, the Motor Neurone Disease Association has a network of 18 accredited Motor Neuron Disease Care and Research Centres.

While multidisciplinary clinics are a highly effective method of delivering care to patients with ALS, this approach may not be available in all venues, because of inadequate patient volume or, in some cases, insufficient philanthropic support. Some patients find full-day assessments exhausting; for them, more frequent, but shorter, visits are more acceptable.

In other models of care, the multidisciplinary team works in the community and visits the patient at home or care is provided through a local hospice team. Primary care providers have an important role and in some settings may be able to coordinate all of the care for a patient with ALS.

Inpatient care

Inpatient care may be needed temporarily for patients with ALS who decompensate in the outpatient setting (eg, because of pneumonia). Hospitalization may also be necessary for patients who reach critical ventilatory failure without having appropriate ventilatory support in place or without having made end-of-life decisions (advance directives) declining such ventilatory support and electing for comfort measures instead.

Pharmacologic Treatment

Riluzole

The glutamate pathway antagonist riluzole (Rilutek) is the only medication that has shown efficacy in extending life in ALS. Compared with placebo, riluzole may prolong median tracheostomy-free survival by 2-3 months in patients younger than 75 years with definite or probable ALS who have had the disease for less than 5 years and who have a forced vital capacity (FVC) of greater than 60%.[177]

Studies

This conclusion is based on 2 double-blinded, randomized, placebo-controlled clinical trials.[33, 34] A third clinical trial, in patients who were ineligible to participate in the second pivotal clinical trial, did not show efficacy, possibly due to low power (small number of patients included relative to the magnitude of the possible effect).[34, 178] A fourth clinical trial, conducted in Japan, did not show efficacy; details of the study’s results have not been published in the English literature.[179, 180]

A Cochrane review that pooled data from the first 3 clinical trials reported that the addition of data from the third trial, which included patients who were older and more seriously affected, reduced the overall treatment effect seen in the first 2 trials.[177] Nevertheless, evidence of benefit from riluzole remained statistically significant.

Subsequent reports have claimed greater efficacy for riluzole in clinical practice than in the clinical trial. However, the relevance of these claims has been challenged[181] because the reports pit class IV evidence against class I evidence, which is contrary to the usual evidence-based approach used to judge treatment efficacy.

Prognosis

Patients with ALS who have depression, frontotemporal dementia (FTD), or a milder level of frontal impairment are now recognized as less likely to accept treatment recommendations and have a poorer prognosis. However, although failure to accept riluzole treatment is a risk factor for poor survival, it is not the cause. Furthermore, registries in place since before the introduction of riluzole show no overall extension of survival of patients with ALS.

Adverse effects

The principal clinical side effects some patients with riluzole may experience are stomach upset and asthenia (lack of energy). These problems resolve if the medication is discontinued.

Cases of interstitial lung disease, some of them severe, have been reported in patients treated with riluzole; many of these cases have proved to be hypersensitivity pneumonitis. If respiratory symptoms such as dry cough and/or dyspnea develop, chest radiography should be performed, and if findings suggest interstitial lung disease or hypersensitivity pneumonitis (eg, bilateral diffuse lung opacities), riluzole should be discontinued immediately. In most reported cases, respiratory symptoms resolved after drug discontinuation and symptomatic treatment.

Aminotransferase

Some patients on riluzole develop abnormal liver function test results or neutropenia. Serum levels of aminotransferases, including alanine aminotransferase (ALT), should be measured before and during riluzole therapy, with ALT levels being evaluated every month during the first 3 months of treatment, every 3 months during the remainder of the first year, and periodically thereafter.

Serum ALT levels should be evaluated more frequently in patients who develop elevations. Treatment should be discontinued if ALT levels reach 5 times the upper limit of normal or higher or if clinical jaundice develops.

Edaravone

In May 2017, the pyrazolone free radical scavenger, edaravone (Radicava), was approved to slow the functional decline in patients with ALS. Although the precise mechanism by which edaravone works in ALS is unknown, it may lessen the effects of oxidative stress, which is thought to be a probable factor in ALS onset and progression.

FDA approval of edaravone was based on the pivotal Phase 3 study (MCI186-19), which evaluated 137 people with ALS. Data demonstrated patients who received edaravone for 6 months experienced significantly less decline in physical function (33% reduction or 2.49 ALSFRS-R points; p=0.001).[182]

American Academy of Neurology Guideline

In October 2009, the American Academy of Neurology (AAN) published a 2-part, evidence-based Practice Parameter update about the care of patients with ALS.[2, 3] These publications updated the 1999 evidence-based practice parameter.[173]

NIV, PEG, and riluzole

The chief findings of the practice parameter are that for extending life or slowing disease progression, the evidence is best for use of noninvasive ventilation (NIV), percutaneous endoscopic gastrostomy (PEG), and riluzole. The authors comment that these treatments are often underutilized and recommend that they should be offered to patients, in the case of riluzole, or considered by the physician, in the case of NIV and PEG.

The median extension of life by riluzole in placebo-controlled studies was 2-3 months. The median extension of life by NIV or PEG may be approximately 6 months, provided the treatments are applied early and adhered to. The efficacy of NIV is supported by a randomized, controlled trial.[183]

Multidisciplinary care

The authors also recommend that referral to a multidisciplinary clinic should be considered for managing patients with ALS. Such clinics can optimize healthcare delivery, prolong survival, and enhance quality of life.

Botulinum toxin

In patients with sialorrhea that is refractory to treatment with standard medications, botulinum toxin B injection of the salivary glands should be considered. Alternatively, low-dose radiation therapy to the salivary glands may be considered.

Botulinum toxin for ALS carries a manufacturer's warning regarding possible spread of the toxin to ventilatory muscles (leading to exacerbation of ventilatory failure) or bulbar muscles (exacerbating their weakness). The risk of these adverse effects tilts the balance in terms of safety toward salivary gland irradiation, except in patients with exceptional ventilatory and bulbar reserves.

Dextromethorphan and quinidine

Finally, the guideline recommends that dextromethorphan and quinidine be considered for pseudobulbar affect. A combination of these 2 agents (Nuedexta) is approved by the FDA for this indication.

Clinician and patient summaries, as follow, are available on the AAN website:

Treatment of Symptoms

Limb stiffness

Limb stiffness can be treated with the antispasticity agents baclofen (Lioresal) and tizanidine (Zanaflex). Baclofen can be started at 10 mg/day and titrated up to 10 mg 3 times per day. If adequate efficacy is not attained at a lower dose, baclofen can be titrated to double that dose, if tolerated. Tizanidine can be started at 1 mg 3 times daily and titrated upward, if tolerated, to as high as 8 mg 3 times daily.

The chief risks of both of these agents are that they induce drowsiness in some patients and that relief of spasticity may result in unpredictable loss of tone and falls. Starting at a low dose permits determination of the patient’s tolerance of the medication, before titration to an effective dose.

Intrathecal baclofen may be considered in slowly progressive, upper motor neuron (UMN) ̶ predominant patients with ALS or in patients with primary lateral sclerosis (PLS), who do not respond adequately to oral treatments.

Sialorrhea

Antisialorrhea treatments include the following:

Anticholinergics such as amitriptyline (25-50 mg at bedtime) and trihexyphenidyl (Artane; 0.5-2 mg as needed) may be administered as tolerated. A scopolamine patch may work in patients who have not attained adequate relief from oral anticholinergics. Sympathomimetics such as pseudoephedrine may be tried if tolerated; 30-60 mg may be administered as needed or 120-240 mg/day of the extended-release formulations may be used.

The efficacy of injections of botulinum toxin type B (2500 U) into the salivary glands has been reported in a small, double-blinded, placebo-controlled trial[184] , but there is a concern that the toxin could spread to bulbar or respiratory muscles, and there is a manufacturer’s warning to that effect. Salivary gland irradiation (7.5 Gy) was found to effective in a case series with a standardized outcome measure[185] and does not carry the same risk as botulinum toxin.

Thickened secretions

Mucolytics such as guaifenesin may be used to thin thickened secretions, although removal of secretions may require mechanical suction devices. Adequate hydration and humidification of room air may prove helpful and are recommended.

Depression and anxiety

For treatment of depression, selective serotonin reuptake inhibitors (SSRIs), such as citalopram 10-40 mg/day, work best. If the desired benefit is not achieved with one agent in this class, another may be tried.

For anxiety, lorazepam is commonly used (0.5-1 mg prn). Careful titration is required, as benzodiazepines have the potential to cause respiratory depression.

Pain

Pain may occur in patients with ALS as a secondary consequence of immobility, loss of muscle protection of joints, loss of muscle and fat padding of bony prominences, and overexerted muscles. Nonpharmacologic measures to protect bony prominences (cushions and appropriate bed padding) and support overexerted muscles (neck support, trunk support) should be pursued. Range-of-motion exercises should be employed to prevent frozen shoulders.

If pain develops, its causes need to be assessed and appropriate treatment initiated. Often, nonsteroidal anti-inflammatory drugs (NSAIDs) suffice. If needed, they should be prescribed on a regular schedule.

If stronger analgesics are required, tramadol (Ultram), ketorolac (Toradol), morphine (immediate or extended release), or a fentanyl patch may be considered. (Respiratory depression may occur with opiates.) Careful titration, starting with low doses, is needed. Patients must be assured that they will not be in pain as a result of the disease or its secondary consequences.

A combination of dextromethorphan and quinidine (Nuedexta) has shown efficacy in ameliorating involuntary laughter and crying (the expression of pseudobulbar affect).[186, 187] It has FDA approval for this indication.

Cramps

Cramps are difficult to treat. Quinine sulfate was used in the past; the recommended dose for nocturnal cramps was 200-300 or 324 mg at bedtime. However, quinine does not have FDA approval for this indication, its efficacy compared with placebo has not been well established, and there is a concern regarding its potential adverse cardiac effects. Quinine sulfate is contraindicated in patients with a prolonged QT segment, and has many interactions with other drugs. Its clearance is reduced in patients with renal failure.

Other agents that may be tried for treatment of cramps are benzodiazepines, antispasticity agents, and anticonvulsants (eg, gabapentin, carbamazepine, phenytoin). These agents have not been tested for this indication in controlled studies or approved for it, and their efficacy is uncertain.

Incontinence

Urinary urgency may be treated with tolderodine (Detrol). However, the effectiveness of this agent for incontinence may be limited when the incontinence is due to weakening of the muscles of the pelvic floor.

Sleeping problems

When patients complain of sleeping difficulties, the first step is to determine whether these are due to ventilatory failure; this can be accomplished through overnight polysomnography. In some cases the integrity of sleep can be restored just by introducing noninvasive ventilatory support, which usually consists of bilevel positive airway pressure (often with a backup rate). Adjunctive medications may be considered, but the clinician must bear in mind that some drugs may suppress the respiratory drive (hence the need to initiate noninvasive ventilatory support first).

Loss of appetite

Appetite tends to be reduced as ALS progresses. The following reasons have been imputed:

A full stomach makes it harder to breathe

Breakdown of muscle releases amino acids that produce a false sense of satiety

Metabolism of carbohydrates produces CO2, which requires breathing effort to clear

Reduced activity requires less energy intake

Appetite tends to be lost with reduced intake, creating a vicious circle

Nutritional consultation is advised. Frequent small meals, foods rich in fat and protein, and sometimes a feeding gastrostomy are needed. When initiating supplemental feedings in a patient who has not been nourished adequately, attention should be given to ventilatory support, as the patient may be too weak to blow off the extra CO2 or to breathe against a full stomach.

Ventilatory Support

Noninvasive ventilatory support has been shown to improve patients’ quality of life and to extend life when applied as patients begin to experience the early effects of ventilatory failure, including sleep disruption. Noninvasive ventilatory support is probably more effective than all other treatments for prolonging life in ALS patients.

Overnight polysomnography may identify disruption of the contiguity of sleep, one of the early consequences of ventilatory failure that may precede frank apneas, hypopneas, or nocturnal oxygen desaturation.

Dietary Considerations

The patient’s appetite tends to decline as the disease progresses, and his/her ability to swallow may become impaired. Consultations with a dietician or nutritionist and with a speech therapist may be requested to help the patient compensate for these losses. Dietary supplements may be used to assure adequate caloric intake.

Placement of a feeding gastrostomy may be considered in patients who cannot maintain adequate caloric intake as a result of swallowing difficulties and who have an FVC of greater than 50% of predicted. A gastrostomy can be placed even in patients with an FVC of less than 50% of predicted, but it requires extra care – usually anesthesiology presence. Consultation with a gastroenterologist or surgeon may be requested if PEG placement is being considered.

Activity Restriction

Initially, no activity restriction is necessary. Indeed, early in the course of ALS, encourage patients to continue routine activities. However, patients should not overexert themselves to the point of fatigue or pain.

Patients should maintain a regular exercise regimen if their degree of weakness allows. They need to realize, however, that their muscle reserve will diminish before overt sustained weakness appears, which means that in most cases they should avoid extreme endurance exercises (repetitions). Patients with slowly progressive disease will be able to tolerate exercise and benefit from it more than patients with rapidly progressive disease.

The chief goals of activity are as follows:

As the disease progresses, patients may become unstable and at risk of falls and may need to be counseled to use assistive devices or to not transfer without appropriate support. If they reach a point at which they cannot manage a vehicle safely, including in emergencies, they need to be counseled to stop driving or have modifications made to the car so that they are able to drive safely.

In the United States, some states require mandatory reporting by practitioners to the Department of Motor Vehicle Affairs, and some require disabled patients to pass a driving assessment if they have modifications made to their car (eg, hand controls). In the United Kingdom, any medical condition likely to last more than 3 months requires reporting to the Driver and Vehicle Licensing Agency by the patient.

Alternative Therapies

Many patients with ALS elect to try alternative therapies. If not unsafe or exorbitantly priced, these treatments may be a reasonable approach to giving patients a sense of some control, may confer on them a feeling of calm, and may thus be of benefit, in subjective terms. Since most, if not all, alternative therapies have not been tested against placebo controls, physicians cannot provide blanket recommendations but may be able to be supportive, if circumstances permit, of specific patient choices.

When alternative therapies impose a great burden on patients, in terms of cost or time commitment, they should be avoided. Since 2009, the ALS Untangled group of investigators, who are members of the North American ALS Research Group or the World Federation of Neurology (WFN) ALS Research Group, has evaluated several claims of efficacy of specific alternative therapies (to slow the course of ALS) and shown them to lack adequate foundation.

Deterrence and Prevention

Smoking is the only established environmental risk factor for ALS.[54, 56] Smoking avoidance may result in a decrease in the age-specific incidence of the disease. However, this presumptive benefit may be attenuated if lower mortality from other smoking-related diseases (eg, cancer, cardiovascular diseases, stroke) results in more patients surviving to be at risk for ALS in each age bracket. As the population lives to older ages (in which age-specific incidence of ALS is higher), the crude incidence of ALS may increase.

Individuals with a gene for familial ALS may benefit from genetic counseling if they wish to minimize the risk of transmitting the gene to the next generation.

Informing Patients of the Diagnosis

Doing this properly requires time, motivation, adequate staff, and preparation. The American Academy of Neurology (AAN) 1999 Practice Parameter[173] provided the following suggestions for breaking the news of a diagnosis of ALS to a patient, based on a review of literature pertaining to other diseases:

In this era of readily available information on the Internet, provision of the diagnosis will generate independent activity on the part of patients, families and friends to educate themselves about the disease. Thus, it is helpful if clinicians point patients and the families to those resources that they feel will serve this purpose best, to serve as the starting point (see Patient Education).

The 2009 AAN update to the 1999 practice parameter[2] refers to a 6-step protocol, described by the acronym SPIKES, that was designed for delivering bad news to patients with cancer.[188] The following is summarized from Appendix e-1[2] :

The SPIKES protocol has several advantages and may be further improved by the following 3 measures. First, it is important that relevant family members be present when the patient is informed about the diagnosis, so that the patient is not the one who has to inform them and answer their questions. They can support the patient, and the neurologist can inform and support them directly.

Second, it is very helpful to schedule an early follow-up visit to answer questions that will arise and inform/update family members who were not present when the diagnosis was given. Many patients and families will not remember most of what they are told when they hear the diagnosis for the first time, as they will be in shock. The shock may be less if they have anticipated the bad news, but it cannot be avoided completely. The greater the perceived shock, the more the focus should be on support and discussing the next steps, including the follow-up visit.

Third, it is helpful to have a handout summarizing the information presented in the clinic. The handout should provide links to appropriate Web sites, as many patients will want to take an active role in educating themselves with the use of these resources.

The patient and his/her family may want to tarry a while after the visit. It is helpful to have a nonphysician staff member available to support them and to reinforce the immediate next steps.

Patient Support and Advice

Support groups are available to patients in many communities. Burnout of the primary caregiver needs to be anticipated and avoided by assuring that the primary caregiver is not the only caregiver.

Clinical trials

Patients with ALS may wish to help with the search for treatments for the disease through participation in clinical trials. They should be encouraged to focus on trials that have been listed with the ALS Association, the Muscular Dystrophy Association (MDA), and ClinicalTrials.gov, which is a registry of federally and privately supported clinical trials conducted in the United States and around the world.

Off-label pharmaceuticals

Patients often request prescription of pharmaceuticals for off-label use in the hope of slowing disease progression. Since double-blinded, placebo-controlled studies to date have shown that all such pharmaceuticals either made patients worse or had no benefit, this practice should be avoided.

Government benefits

In the United States, patients are eligible for Social Security and Medicare benefits once they are diagnosed with ALS, without the waiting period required of other patients with chronic diseases. Patients should be advised to apply early.

Since September 2008, ALS has been considered a service-connected condition for US veterans, and they are eligible for care and benefits.[189] Eligible patients should be advised to apply early. Volunteers from local branches of veterans’ organizations may be able to assist in preparing the application and moving it rapidly through the approval process.

End-of-life considerations

End-of-life issues may be discussed and clarified early. However, this may not work well for some patients. The physician should be aware of the individual state or national laws that regulate these issues; encourage, if appropriate for the patient, completion of advance directives; and document the patient’s preferences in the medical records, whether or not formal advance directives have been written.

Resource access

For patients with limited access to home-based resources, social service professionals may be able to assist with placement. Local branches of advocacy organizations (the ALS Association of America and the Muscular Dystrophy Association) may be excellent sources of information on what resources are available locally.

Legal affairs

Patients will have decreasing ability to sign documents or to mobilize themselves for taking care of personal affairs. Some may benefit from legal advice. The earlier they do so, the better, but they should be allowed to recover first from the initial shock of the diagnosis.

Loss of independence

All patients will become at risk for falls as ALS progresses; they should be informed of this and cautioned gently. Most patients tend to sustain several falls before they relinquish the independent activities that led to them.

All patients will become unable to drive as their disease progresses. They need to be alerted when this draws close. Most are gracious about quitting before their driving results in injury. Some states require mandatory reporting by practitioners to the Department of Motor Vehicle Affairs. In the United Kingdom, patients are obliged to inform the Driver and Vehicle Licensing Agency.

Consultations

Consultants are best used within the multidisciplinary model based on patients’ preferences and need to complement the range of services that the patients’ primary care providers and primary neurologists, palliative care physicians, or physiatrists can provide directly. The following consultations may prove helpful:

While many patients and families may benefit from referral to a psychiatrist, psychologist, or counselor, patients may be most likely to use these resources if they have already done so prior to disease onset or if these options are offered within the multidisciplinary model of care.

Long-Term Monitoring

Patients are best cared for in a designated ALS center. Many mechanical aids can help them to overcome disabilities. A patient’s length of survival and quality of life are enhanced by night-time breathing assistance early in the course of the disease and by aggressive application of alternate feeding options to ensure good nutrition once swallowing becomes difficult.[190]

The 2009 AAN practice parameter guideline reviewed available evidence to help maximize the care and quality of life of patients with ALS.[2, 3] The recommendations can be summarized as follows:

Medication Summary

The glutamate pathway antagonist riluzole is the only medication that has shown efficacy in extending life in amyotrophic lateral sclerosis (ALS). The American Academy of Neurology (AAN) guideline recommends that riluzole be offered to patients with ALS.[2]  The pyrazolone free radical scavenger, edaravone (Radicava), was approved to slow the functional decline in patients with ALS.[182]

A variety of other medications may be useful for relief of symptoms associated with ALS. These include the following:

Riluzole (Rilutek)

Clinical Context:  Riluzole (Rilutek)

Riluzole is thought to counteract excitatory amino acid (glutaminergic) pathways, noncompetitively block N-methyl-D-aspartate (NMDA)–mediated responses, and inactivate voltage-dependent sodium channels. Its exact mechanism of action in ALS is unknown. It is a benzothiazole agent that is well absorbed, with average oral bioavailability of 60% and mean elimination half-life of 12 hours; steady state is reached within 5 days with multiple dose administration. Metabolism occurs in the liver (P450-dependent glucuronidation and hydroxylation), with 6 major and a few minor metabolites produced. The recommended dosage for riluzole is 50 mg twice daily. Riluzole is used for extension of time to death or tracheostomy.

Edaravone (Radicava)

Clinical Context:  Edaravone is a pyrazolone free radical scavenger; mechanism by which the drug exerts its therapeutic effects in ALS in unknown. It is theorized to decrease effects of oxidative stress, a likely factor in the onset and progression of ALS. Administration is by IV infusion, requiring it to be given by a healthcare professional and monitoring for infusion-related reactions.

Class Summary

The FDA has approved 2 drugs for ALS as of May 2017.

Baclofen (Lioresal, Gablofen)

Clinical Context:  Baclofen is metabolized in the liver and excreted primarily in urine. This agent is not a controlled substance under the Drug Enforcement Administration (DEA).

Class Summary

These agents relieve spasticity and muscle spasms in patients with symptoms of limb stiffness.

Tizanidine (Zanaflex)

Clinical Context:  Tizanidine is a centrally acting muscle relaxant metabolized in the liver and excreted in urine and feces. It is used in patients with predominantly upper motor neuron (UMN) involvement. It is not a DEA-controlled substance.

Class Summary

Alpha2 adrenergic agonists decrease excitatory input to alpha motor neurons (which are a type of lower motor neuron [LMN]). Common alpha2 adrenergic effects include decrease release of acetylcholine and norepinephrine, contraction of sphincters in the gastrointestinal tract, and inhibition of lipolysis.

Dextromethorphan and quinidine (Nuedexta)

Clinical Context:  Dextromethorphan is a sigma-1 receptor agonist and an uncompetitive NMDA receptor antagonist. Quinidine increases plasma levels of dextromethorphan by competitively inhibiting cytochrome P4502D6, which catalyzes a major biotransformation pathway for dextromethorphan. The mechanism by which dextromethorphan exerts therapeutic effects in patients with PBA is unknown.

This combination is indicated for PBA and symptoms associated with ALS (and multiple sclerosis) that result in involuntary, sudden, and frequent episodes of laughing and/or crying. It is available as a capsule containing dextromethorphan 20 mg and quinidine 10 mg.

Class Summary

The combination of dextromethorphan and quinidine is shown to decrease emotional lability of pseudobulbar affect (PBA).

What is amyotrophic lateral sclerosis (ALS)?What are the initial lower limb symptoms of amyotrophic lateral sclerosis (ALS)?What are the initial upper limb symptoms of amyotrophic lateral sclerosis (ALS)?What are the initial bulbar symptoms of amyotrophic lateral sclerosis (ALS)?What are the emotional and cognitive symptoms of amyotrophic lateral sclerosis (ALS)?What are the symptoms of-advanced amyotrophic lateral sclerosis (ALS)?What are symptoms of progression of bulbar amyotrophic lateral sclerosis (ALS)?Which anatomy is involved by amyotrophic lateral sclerosis (ALS)?What is required for a confirmed diagnosis of amyotrophic lateral sclerosis (ALS)?What is the World Federation of Neurology (WFN) diagnostic algorithm for amyotrophic lateral sclerosis (ALS)?How is amyotrophic lateral sclerosis (ALS) diagnosed?What is the role of genetic testing in the diagnosis of amyotrophic lateral sclerosis (ALS)?What are the American Academy of Neurology recommendations for management of amyotrophic lateral sclerosis (ALS)?When is invasive ventilatory support indicated in the management of amyotrophic lateral sclerosis (ALS)?What is amyotrophic lateral sclerosis (ALS)?What is the prognosis of amyotrophic lateral sclerosis (ALS)?When was amyotrophic lateral sclerosis (ALS) first described?What is the cause of amyotrophic lateral sclerosis (ALS)?What are the degenerative effects of amyotrophic lateral sclerosis (ALS)?How does amyotrophic lateral sclerosis (ALS) affect lower motor neurons (LMNs)?What is classic amyotrophic lateral sclerosis (ALS)?What are progressive muscular atrophy (PMA) and flail limb syndrome in amyotrophic lateral sclerosis (ALS)?What is primary lateral sclerosis in amyotrophic lateral sclerosis (ALS)?What is progressive bulbar palsy in amyotrophic lateral sclerosis (ALS)?What is familial amyotrophic lateral sclerosis (ALS)?What is the role of genetics in the etiology of amyotrophic lateral sclerosis (ALS)?What are the complications of amyotrophic lateral sclerosis (ALS)?How is amyotrophic lateral sclerosis (ALS) diagnosed?What is the goal of treatment for amyotrophic lateral sclerosis (ALS)?What are the treatment options for amyotrophic lateral sclerosis (ALS)?How is amyotrophic lateral sclerosis (ALS) described?Which mechanisms have been suggested are etiologic factors in amyotrophic lateral sclerosis (ALS)?What is the role of oxidative stress in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of cytoskeletal proteins in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of ribonucleic acid (RNA) processing in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the likely pathogenesis of amyotrophic lateral sclerosis (ALS)?What is the role of misfolded proteins in the pathophysiology of amyotrophic lateral sclerosis (ALS)?How does the motor axon degenerate in the pathogenesis of amyotrophic lateral sclerosis (ALS)?What is the role of Wallerian degeneration in the pathogenesis of amyotrophic lateral sclerosis (ALS)?Which motor neurons are initially spared in the pathophysiology of typical amyotrophic lateral sclerosis (ALS)?Which factors may mediate cell death in amyotrophic lateral sclerosis (ALS)?What is the role of the copper/zinc superoxide dismutase 1 (SOD1) gene in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of glutamate excitotoxicity in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of derangements of ribonucleic acid (RNA) metabolism in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of the TAR DNA-binding protein gene (TARDBP) in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the role of the fused in sarcoma/translated in liposarcoma (FUS/TLS) gene in the pathophysiology of amyotrophic lateral sclerosis (ALS)?Where is the support for a fused in sarcoma/translated in liposarcoma (FUS/TLS) gene etiology of amyotrophic lateral sclerosis (ALS)?What is the role of a large hexanucleotide repeat expansion in the pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the distinction between the pathogenesis and pathophysiology of amyotrophic lateral sclerosis (ALS)?What is the disease progression of amyotrophic lateral sclerosis (ALS)?What are characteristics of the early stage of amyotrophic lateral sclerosis (ALS) pathophysiology?Which factors trigger the onset of amyotrophic lateral sclerosis (ALS)?What is the role of aging on the pathogenesis of amyotrophic lateral sclerosis (ALS)?What role does misfolded protein play in the pathogenesis of amyotrophic lateral sclerosis (ALS)?What is the role of non-coding C9ORF72 hexanucleotide expansion in the pathogenesis of amyotrophic lateral sclerosis (ALS)?What is a focal trigger for the onset of amyotrophic lateral sclerosis (ALS)?What is the most common cause of amyotrophic lateral sclerosis (ALS)?What causes familial amyotrophic lateral sclerosis (ALS)?How many SOD1 mutations have been identified in amyotrophic lateral sclerosis (ALS)?Which SOD1 mutation causing amyotrophic lateral sclerosis (ALS) is the most common in the US?What is the role of SOD1 mutations in the etiology of amyotrophic lateral sclerosis (ALS)?What are the roles of the TAR DNA-binding protein (TARDBP) and the fused in sarcoma (FUS) genes in the etiology of amyotrophic lateral sclerosis (ALS)?What is the role of the C9orf72 mutation in the etiology of amyotrophic lateral sclerosis (ALS)?What are less common genetic etiologies of familial amyotrophic lateral sclerosis (ALS)?What causes sporadic amyotrophic lateral sclerosis (ALS)?What evidence support the hypothesis that genetic risk factors are present in sporadic amyotrophic lateral sclerosis (ALS)?What is the role of smoking in the etiology of amyotrophic lateral sclerosis (ALS)?What is the significance of smoking as an established risk factor for amyotrophic lateral sclerosis (ALS)?How does focusing on the processes of initiation of amyotrophic lateral sclerosis (ALS) provide new avenues for treatment?Which factors are unlikely to increase the risk for amyotrophic lateral sclerosis (ALS)?Which suggested risk factors for amyotrophic lateral sclerosis (ALS) are uncertain?What is the risk of amyotrophic lateral sclerosis (ALS) in soccer players?What is the risk of amyotrophic lateral sclerosis (ALS) in US National Football League (NFL) players?What is the association between head injury and amyotrophic lateral sclerosis (ALS)?What are possible non-sport-related risk factors for amyotrophic lateral sclerosis (ALS) in American football players?What causes amyotrophic lateral sclerosis (ALS) in Guam?What is the annual incidence of amyotrophic lateral sclerosis (ALS) in the US?What is the lifetime risk for developing amyotrophic lateral sclerosis (ALS)?What is the global incidence for amyotrophic lateral sclerosis (ALS)?What are the racial predilections for amyotrophic lateral sclerosis (ALS)?How does the incidence of amyotrophic lateral sclerosis (ALS) vary by sex?How does the incidence of amyotrophic lateral sclerosis (ALS) vary by age?What is the prognosis of amyotrophic lateral sclerosis (ALS)?How does the progression of amyotrophic lateral sclerosis (ALS) vary?How is the progression of amyotrophic lateral sclerosis (ALS) measured?What are the milestone stages and corresponding times of occurrence in amyotrophic lateral sclerosis (ALS)?At what disease stage are most patients diagnosed with amyotrophic lateral sclerosis (ALS)?How is progressive muscular atrophy (PMA) distinguished from classic amyotrophic lateral sclerosis (ALS)?When is individualized prognostic information of amyotrophic lateral sclerosis (ALS) discussed with a patient?Which education resources are available to patients with amyotrophic lateral sclerosis (ALS)?When can online patient information about amyotrophic lateral sclerosis (ALS) be found?How is amyotrophic lateral sclerosis (ALS) diagnosed?When is amyotrophic lateral sclerosis (ALS) suspected?What are the motor neuron symptoms of amyotrophic lateral sclerosis (ALS)?What are the initial symptoms of amyotrophic lateral sclerosis (ALS)?What are the clinical differences between upper and lower limb motor neuron symptom onset in amyotrophic lateral sclerosis (ALS)?What are the clinical manifestations of amyotrophic lateral sclerosis (ALS) progression?What are bulbar symptoms of amyotrophic lateral sclerosis (ALS)?What are the pseudobulbar symptoms of amyotrophic lateral sclerosis (ALS)?Which motor functions are preserved in amyotrophic lateral sclerosis (ALS)?What is the focus of family history in suspected amyotrophic lateral sclerosis (ALS)?What are the signs and symptoms of amyotrophic lateral sclerosis (ALS)?Which physical findings are characteristic of upper or lower motor neuron (UMN or LMN) dysfunction in amyotrophic lateral sclerosis (ALS)?Which physical findings reflect upper motor neuron (UMN) dysfunction in amyotrophic lateral sclerosis (ALS)?Which physical findings reflect lower motor neuron (LMN) dysfunction in amyotrophic lateral sclerosis (ALS)?What is the key physical finding characteristic of amyotrophic lateral sclerosis (ALS)?What are the emotional and cognitive symptoms of amyotrophic lateral sclerosis (ALS)?What are the bulbar physical findings characteristic of amyotrophic lateral sclerosis (ALS)?How are axial truncal weakness characterized in amyotrophic lateral sclerosis (ALS)?What is the significance of a suspected amyotrophic lateral sclerosis (ALS) diagnosis?What are the El Escorial World Federation of Neurology diagnostic criteria for amyotrophic lateral sclerosis (ALS)?What are the Awaji diagnostic criteria for amyotrophic lateral sclerosis (ALS)?What are the World Federation of Neurology (WFN) diagnostic criteria for amyotrophic lateral sclerosis (ALS)?How are the regions of the body defined in the World Federation of Neurology (WFN) criteria for amyotrophic lateral sclerosis (ALS)?What are the World Federation of Neurology (WFN) categories for the diagnosis of amyotrophic lateral sclerosis (ALS)?How is the degree of certainty for the diagnosis of amyotrophic lateral sclerosis (ALS) increased?Why have qualifiers lost their meaning in the diagnosis of amyotrophic lateral sclerosis (ALS)?How are qualifiers used in the diagnosis of amyotrophic lateral sclerosis (ALS)?What revisions have been proposed to the WFN diagnostic criteria for amyotrophic lateral sclerosis (ALS)?At what disease stage does the presentation of other neurologic conditions overlap with amyotrophic lateral sclerosis (ALS)?What are the differential diagnoses for amyotrophic lateral sclerosis (ALS) in its early stages?What are the differential diagnoses for more advanced amyotrophic lateral sclerosis (ALS)?What are the differential diagnoses for a rapid onset of amyotrophic lateral sclerosis (ALS)?What conditions may be consider in the differential diagnoses of amyotrophic lateral sclerosis (ALS)?What are the differential diagnoses for Amyotrophic Lateral Sclerosis?What is needed to make a definitive diagnosis of amyotrophic lateral sclerosis (ALS)?How is the diagnosis of amyotrophic lateral sclerosis (ALS) confirmed?What is the role of lab tests in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of biochemical markers in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of genetic testing in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of imaging studies in the diagnosis of amyotrophic lateral sclerosis (ALS)?When is muscle or nerve biopsy indicated in the diagnosis of amyotrophic lateral sclerosis (ALS)?How are needle electromyography and nerve conduction studies performed in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of electromyography (EMG) in the diagnosis of amyotrophic lateral sclerosis (ALS)?Which motor unit recruitment findings indicate amyotrophic lateral sclerosis (ALS)?Which muscle innervation findings indicate amyotrophic lateral sclerosis (ALS)?Which denervation findings indicate amyotrophic lateral sclerosis (ALS)?Which nonspecific EMG findings are common in amyotrophic lateral sclerosis (ALS)?What is the role of motor and sensory nerve conduction studies in the diagnosis of amyotrophic lateral sclerosis (ALS)?What are the electrophysiologic findings characteristic of UMN involvement in amyotrophic lateral sclerosis (ALS)?When are voluntary motor unit action potentials findings suggestive of amyotrophic lateral sclerosis (ALS)?What is the role of the motor unit number estimate (MUNE) in the diagnosis of amyotrophic lateral sclerosis (ALS)?Which EMG findings suggest multifocal motor mononeuropathy in amyotrophic lateral sclerosis (ALS)?Which EMG findings suggest chronic inflammatory demyelinating polyradiculoneuropathy in amyotrophic lateral sclerosis (ALS)?What EMG findings suggests generalized axonal sensorimotor peripheral neuropathy in amyotrophic lateral sclerosis (ALS)?Which EMG findings suggest inclusion body myositis in amyotrophic lateral sclerosis (ALS)?Which EMG findings suggest primary lateral sclerosis or monomelic amyotrophy in amyotrophic lateral sclerosis (ALS)?What is the role of lab studies in the diagnosis of amyotrophic lateral sclerosis (ALS)?Which lab tests may be useful in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of antiacetylcholine receptor antibody and anti-muscle specific kinase (MuSK) antibody assays in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of urinary 24-hour collections in the diagnosis of amyotrophic lateral sclerosis (ALS)?When is Lyme disease serology performed in the evaluation of amyotrophic lateral sclerosis (ALS)?What is the role of genetic testing in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of MRI in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of magnetic resonance spectroscopy in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of CT scanning in the diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of PET scanning and functional magnetic resonance imaging (fMRI) in the diagnosis of amyotrophic lateral sclerosis (ALS)?How does amyotrophic lateral sclerosis (ALS) progress?How does the pace of amyotrophic lateral sclerosis (ALS) progression vary?What is the ALS Functional Rating Scale and how is it used in the assessment of amyotrophic lateral sclerosis (ALS)?How are ALS Functional Rating Scale (ALSFRS) scores interpreted in the assessment of amyotrophic lateral sclerosis (ALS)?How are pulmonary function tests used to monitor amyotrophic lateral sclerosis (ALS) progression?What is the focus of treatment for amyotrophic lateral sclerosis (ALS)?What is the role of patient education in the management of amyotrophic lateral sclerosis (ALS)?What is included in the outpatient care of amyotrophic lateral sclerosis (ALS)?What is included in the inpatient care of amyotrophic lateral sclerosis (ALS)?What is the role of riluzole in the management of amyotrophic lateral sclerosis (ALS)?What is the efficacy of riluzole in the treatment of amyotrophic lateral sclerosis (ALS)?How does treatment with riluzole affect the prognosis of amyotrophic lateral sclerosis (ALS)?What are the adverse effects of riluzole in the treatment of amyotrophic lateral sclerosis (ALS)?What monitoring is needed for patients with amyotrophic lateral sclerosis (ALS) taking riluzole?What is the role of edaravone (Radicava) in the treatment of amyotrophic lateral sclerosis (ALS)?Which organization has published clinical treatment guidelines for amyotrophic lateral sclerosis (ALS)?What are the AAN guidelines for extending life or slowing disease progression in amyotrophic lateral sclerosis (ALS)?What are the benefits of referral to a multidisciplinary clinic for the treatment of amyotrophic lateral sclerosis (ALS)?What are the AAN guidelines for use of botulinum toxin in amyotrophic lateral sclerosis (ALS)?What are the AAN treatment guidelines for pseudobulbar affect in amyotrophic lateral sclerosis (ALS)?Where can AAN guidelines clinician and patient summaries be found for amyotrophic lateral sclerosis (ALS)?What are the treatment options for limb stiffness in amyotrophic lateral sclerosis (ALS)?What are the treatment options for antisialorrhea in amyotrophic lateral sclerosis (ALS)?What is the efficacy of botulinum toxin type B for the treatment of sialorrhea in amyotrophic lateral sclerosis (ALS)?What are the options for managing thickened secretions in amyotrophic lateral sclerosis (ALS)?How is depression treated in amyotrophic lateral sclerosis (ALS)?How is anxiety treated in amyotrophic lateral sclerosis (ALS)?What are the causes of pain in amyotrophic lateral sclerosis (ALS) and how can pain be prevented?How is pain treated in amyotrophic lateral sclerosis (ALS)?What are the treatment options for cramps in amyotrophic lateral sclerosis (ALS)?How is incontinence treated in amyotrophic lateral sclerosis (ALS)?What are the treatment options for sleep difficulties in amyotrophic lateral sclerosis (ALS)?What causes loss of appetite in amyotrophic lateral sclerosis (ALS)?How is loss of appetite managed in amyotrophic lateral sclerosis (ALS)?What are the benefits of noninvasive ventilatory support in the treatment of amyotrophic lateral sclerosis (ALS)?What is the role of overnight polysomnography in the management of amyotrophic lateral sclerosis (ALS)?What is the role of invasive ventilatory support in the treatment of amyotrophic lateral sclerosis (ALS)?What dietary modifications are needed for patients with amyotrophic lateral sclerosis (ALS)?What is the role of exercise in the management of amyotrophic lateral sclerosis (ALS)?What are the chief goals of exercise in amyotrophic lateral sclerosis (ALS)?How do activity restrictions change as amyotrophic lateral sclerosis (ALS) progresses?What is the role of alternative therapies in the treatment of amyotrophic lateral sclerosis (ALS)?How is amyotrophic lateral sclerosis (ALS) prevented?What are the American Academy of Neurology (AAN) recommendations for informing patients of a diagnosis of amyotrophic lateral sclerosis (ALS)?Where should patients and families be directed for information about amyotrophic lateral sclerosis (ALS)?What is the SPIKES protocol for delivering the news of an amyotrophic lateral sclerosis (ALS) diagnosis?What are the advantages to the SPIKES protocol for delivering the news of an amyotrophic lateral sclerosis (ALS) diagnosis?What is the role of a nonphysician staff member immediately after the patient has been given a diagnosis of amyotrophic lateral sclerosis (ALS)?What is the role of support groups in the management of amyotrophic lateral sclerosis (ALS)?What is the role of clinical trials in the management of amyotrophic lateral sclerosis (ALS)?What is the role of off-label pharmaceuticals in the treatment of amyotrophic lateral sclerosis (ALS)?What are the US government benefits available for patients with amyotrophic lateral sclerosis (ALS)?When should end-of-life considerations be addressed in amyotrophic lateral sclerosis (ALS)?Where can patients with amyotrophic lateral sclerosis (ALS) find social services resources?What are the benefits of legal consultation for patients with amyotrophic lateral sclerosis (ALS)?How is the risk of falls reduced in amyotrophic lateral sclerosis (ALS)?When should patients with amyotrophic lateral sclerosis (ALS) cease driving?Which specialist consultations may be helpful in the management of amyotrophic lateral sclerosis (ALS)?What factor may increase the likelihood of consultation with a psychiatrist, psychologist, or counselor for patients with amyotrophic lateral sclerosis (ALS)?What are the advantages of care at a designated ALS center?What are the AAN recommendations to maximize the care and quality of life of patients with amyotrophic lateral sclerosis (ALS)?Which medications are used to slow decline and extend life in patients with amyotrophic lateral sclerosis (ALS)?Which medications are used for symptom relief in amyotrophic lateral sclerosis (ALS)?Which medications in the drug class NMDA Antagonists are used in the treatment of Amyotrophic Lateral Sclerosis?Which medications in the drug class Alpha2 Adrenergic Agonists are used in the treatment of Amyotrophic Lateral Sclerosis?Which medications in the drug class Skeletal Muscle Relaxants are used in the treatment of Amyotrophic Lateral Sclerosis?Which medications in the drug class Neurologics, Other are used in the treatment of Amyotrophic Lateral Sclerosis?

Author

Carmel Armon, MD, MSc, MHS, Chair, Department of Neurology, Assaf Harofeh Medical Center, Tel Aviv University Sackler Faculty of Medicine, Israel

Disclosure: Received research grant from: Neuronix Ltd, Yoqnea'm, Israel<br/>Received income in an amount equal to or greater than $250 from: JNS - Associate Editor. UpToDate - Author Royalties.

Chief Editor

Nicholas Lorenzo, MD, MHA, CPE, Co-Founder and Former Chief Publishing Officer, eMedicine and eMedicine Health, Founding Editor-in-Chief, eMedicine Neurology; Founder and Former Chairman and CEO, Pearlsreview; Founder and CEO/CMO, PHLT Consultants; Chief Medical Officer, MeMD Inc; Chief Strategy Officer, Discourse LLC

Disclosure: Nothing to disclose.

Acknowledgements

Neil A Busis, MD Chief, Division of Neurology, Department of Medicine, Head, Clinical Neurophysiology Laboratory, University of Pittsburgh Medical Center-Shadyside

Neil A Busis, MD is a member of the following medical societies: American Academy of Neurology and American Association of Neuromuscular and Electrodiagnostic Medicine

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

  1. Brooks BR, Miller RG, Swash M, Munsat TL. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000 Dec. 1(5):293-9. [View Abstract]
  2. [Guideline] Miller RG, Jackson CE, Kasarskis EJ et al. Practice Parameter update: The care of the patient with amyotrophic lateral sclerosis: Drug, nutritional, and respiratory therapies (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2009. 73:1218-1226. [View Abstract]
  3. [Guideline] Miller RG, Jackson CE, Kasarskis 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. 2009 Oct 13. 73(15):1227-1233. [View Abstract]
  4. Hardiman O, van den Berg LH, Kiernan MC. Clinical diagnosis and management of amyotrophic lateral sclerosis. Nat Rev Neurol. 2011 Oct 11. 7(11):639-49. [View Abstract]
  5. King SJ, Duke MM, O'Connor BA. Living with amyotrophic lateral sclerosis/motor neurone disease (ALS/MND): decision-making about 'ongoing change and adaptation'. J Clin Nurs. 2009 Mar. 18(5):745-54. [View Abstract]
  6. Eisen A. Amyotrophic lateral sclerosis: A 40-year personal perspective. J Clin Neurosci. 2009 Apr. 16(4):505-12. [View Abstract]
  7. Phukan J, Hardiman O. The management of amyotrophic lateral sclerosis. J Neurol. 2009 Feb. 256(2):176-86. [View Abstract]
  8. Wijesekera LC, Leigh PN. Amyotrophic lateral sclerosis. Orphanet J Rare Dis. 2009 Feb 3. 4:3. [View Abstract]
  9. Brooks BR. Managing amyotrophic lateral sclerosis: slowing disease progression and improving patient quality of life. Ann Neurol. 2009 Jan. 65 Suppl 1:S17-23. [View Abstract]
  10. Al-Chalabi A, Fang F, Hanby MF, Leigh PN, Shaw CE, Ye W, et al. An estimate of amyotrophic lateral sclerosis heritability using twin data. J Neurol Neurosurg Psychiatry. 2010 Dec. 81(12):1324-6. [View Abstract]
  11. Hudson AJ. Clinical Neurology. The motor neuron diseases and related disorders. 1996. Vol. 4: 11-14.
  12. Kanouchi T, Ohkubo T, Yokota T. Can regional spreading of amyotrophic lateral sclerosis motor symptoms be explained by prion-like propagation?. J Neurol Neurosurg Psychiatry. 2012 Jul. 83(7):739-45. [View Abstract]
  13. Renton AE, Majounie E, Waite A, Simón-Sánchez J, Rollinson S, Gibbs JR, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 2011 Oct 20. 72(2):257-68. [View Abstract]
  14. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011 Oct 20. 72(2):245-56. [View Abstract]
  15. Seeley WW, Crawford RK, Zhou J, Miller BL, Greicius MD. Neurodegenerative diseases target large-scale human brain networks. Neuron. 2009 Apr 16. 62(1):42-52. [View Abstract]
  16. Armon C. What is ALS?. Bedlack RS, Mitsumoto H, Eds. Amyotrophic Lateral Sclerosis: A Patient Care Guide for Clinicians. New York: Demos Medical Publishing; 2012. 1-23.
  17. Ravits JM, La Spada AR. ALS motor phenotype heterogeneity, focality, and spread: deconstructing motor neuron degeneration. Neurology. 2009 Sep 8. 73(10):805-11. [View Abstract]
  18. Armon C. Epidemiology of ALS/MND. Shaw P and Strong M, eds. Motor Neuron Disorders. Elsevier Sciences: 2003. 167-206.
  19. Armon C. ALS 1996 and Beyond: New Hopes and Challenges. A manual for patients, families and friends. Fourth Edition. California: Published by the LLU Department of Neurology, Loma Linda; 2007.
  20. Neary D, Snowden JS, Gustafson L, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998 Dec. 51(6):1546-54. [View Abstract]
  21. Clinical and neuropathological criteria for frontotemporal dementia. The Lund and Manchester Groups. J Neurol Neurosurg Psychiatry. 1994 Apr. 57(4):416-8. [View Abstract]
  22. Ravits JM, La Spada AR. ALS motor phenotype heterogeneity, focality, and spread: deconstructing motor neuron degeneration. Neurology. 2009 Sep 8. 73(10):805-11. [View Abstract]
  23. Wijesekera LC, Mathers S, Talman P, Galtrey C, Parkinson MH, Ganesalingam J, et al. Natural history and clinical features of the flail arm and flail leg ALS variants. Neurology. 2009 Mar 24. 72(12):1087-94. [View Abstract]
  24. Almeida V, de Carvalho M, Scotto M, Pinto S, Pinto A, Ohana B, et al. Primary lateral sclerosis: predicting functional outcome. Amyotroph Lateral Scler Frontotemporal Degener. 2013 Mar. 14(2):141-5. [View Abstract]
  25. Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med. 2010 Mar 1. 48(5):629-41. [View Abstract]
  26. Strong MJ. Progress in clinical neurosciences: the evidence for ALS as a multisystems disorder of limited phenotypic expression. Can J Neurol Sci. 2001 Nov. 28(4):283-98. [View Abstract]
  27. Shaw PJ. Molecular and cellular pathways of neurodegeneration in motor neurone disease. J Neurol Neurosurg Psychiatry. 2005 Aug. 76(8):1046-57. [View Abstract]
  28. Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol. 2009 Jan. 65 Suppl 1:S3-9. [View Abstract]
  29. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993 Mar 4. 362(6415):59-62. [View Abstract]
  30. Shaw PJ, Ince PG, Falkous G, Mantle D. Oxidative damage to protein in sporadic motor neuron disease spinal cord. Ann Neurol. 1995 Oct. 38(4):691-5. [View Abstract]
  31. Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology. 2004 May 25. 62(10):1758-65. [View Abstract]
  32. Mitsumoto H, Santella RM, Liu X, Bogdanov M, Zipprich J, Wu HC, et al. Oxidative stress biomarkers in sporadic ALS. Amyotroph Lateral Scler. 2008 Jun. 9(3):177-83. [View Abstract]
  33. Bensimon G, Lacomblez L, Meininger V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. N Engl J Med. 1994 Mar 3. 330(9):585-91. [View Abstract]
  34. 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. Lancet. 1996 May 25. 347(9013):1425-31. [View Abstract]
  35. Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006 Oct 6. 314(5796):130-3. [View Abstract]
  36. Arai T, Hasegawa M, Akiyama H, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006 Dec 22. 351(3):602-11. [View Abstract]
  37. Tan CF, Eguchi H, Tagawa A, et al. TDP-43 immunoreactivity in neuronal inclusions in familial amyotrophic lateral sclerosis with or without SOD1 gene mutation. Acta Neuropathol. 2007 May. 113(5):535-42. [View Abstract]
  38. Mackenzie IR, Bigio EH, Ince PG, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007 May. 61(5):427-34. [View Abstract]
  39. Sreedharan J, Blair IP, Tripathi VB, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008 Mar 21. 319(5870):1668-72. [View Abstract]
  40. Kabashi E, Valdmanis PN, Dion P, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet. 2008 May. 40(5):572-4. [View Abstract]
  41. Van Deerlin VM, Leverenz JB, Bekris LM, et al. TARDBP mutations in amyotrophic lateral sclerosis with TDP-43 neuropathology: a genetic and histopathological analysis. Lancet Neurol. 2008 May. 7(5):409-16. [View Abstract]
  42. Yokoseki A, Shiga A, Tan CF, et al. TDP-43 mutation in familial amyotrophic lateral sclerosis. Ann Neurol. 2008 Apr. 63(4):538-42. [View Abstract]
  43. Rutherford NJ, Zhang YJ, Baker M, et al. Novel mutations in TARDBP (TDP-43) in patients with familial amyotrophic lateral sclerosis. PLoS Genet. 2008 Sep 19. 4(9):e1000193. [View Abstract]
  44. Del Bo R, Ghezzi S, Corti S, et al. TARDBP (TDP-43) sequence analysis in patients with familial and sporadic ALS: identification of two novel mutations. Eur J Neurol. 2009 Jun. 16(6):727-32. [View Abstract]
  45. Hasegawa M, Arai T, Akiyama H, et al. TDP-43 is deposited in the Guam parkinsonism-dementia complex brains. Brain. 2007 May. 130:1386-94. [View Abstract]
  46. Schwab C, Arai T, Hasegawa M, Akiyama H, Yu S, McGeer PL. TDP-43 pathology in familial British dementia. Acta Neuropathol. 2009 Mar 13. [View Abstract]
  47. Geser F, Martinez-Lage M, Robinson J, et al. Clinical and pathological continuum of multisystem TDP-43 proteinopathies. Arch Neurol. 2009 Feb. 66(2):180-9. [View Abstract]
  48. Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009 Feb 27. 323(5918):1205-8. [View Abstract]
  49. Vance C, Rogelj B, Hortobagyi T, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science. 2009 Feb 27. 323(5918):1208-11. [View Abstract]
  50. Simpson CL, Lemmens R, Miskiewicz K, Broom WJ, Hansen VK, van Vught PW, et al. Variants of the elongator protein 3 (ELP3) gene are associated with motor neuron degeneration. Hum Mol Genet. 2009 Feb 1. 18(3):472-81. [View Abstract]
  51. Padhi AK, Kumar H, Vasaikar SV, Jayaram B, Gomes J. Mechanisms of loss of functions of human angiogenin variants implicated in amyotrophic lateral sclerosis. PLoS One. 2012. 7(2):e32479. [View Abstract]
  52. Zu T, Gibbens B, Doty NS, Gomes-Pereira M, Huguet A, Stone MD, et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci U S A. 2011 Jan 4. 108 (1):260-5. [View Abstract]
  53. Zu T, Liu Y, Bañez-Coronel M, Reid T, Pletnikova O, Lewis J, et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc Natl Acad Sci U S A. 2013 Dec 17. 110 (51):E4968-77. [View Abstract]
  54. Armon C. An evidence-based medicine approach to the evaluation of the role of exogenous risk factors in sporadic amyotrophic lateral sclerosis. Neuroepidemiology. 2003 Jul-Aug. 22(4):217-28. [View Abstract]
  55. Armon C. Acquired nucleic acid changes may trigger sporadic amyotrophic lateral sclerosis. Muscle Nerve. 2005 Sep. 32(3):373-7. [View Abstract]
  56. Armon C. Smoking may be considered an established risk factor for sporadic ALS. Neurology. November 17, 2009. 73 (20):1693-1698. [View Abstract]
  57. de Jong SW, Huisman MH, Sutedja NA, van der Kooi AJ, de Visser M, Schelhaas HJ, et al. Smoking, alcohol consumption, and the risk of amyotrophic lateral sclerosis: a population-based study. Am J Epidemiol. 2012 Aug 1. 176(3):233-9. [View Abstract]
  58. Esclaire F, Kisby G, Spencer P, Milne J, Lesort M, Hugon J. The Guam cycad toxin methylazoxymethanol damages neuronal DNA and modulates tau mRNA expression and excitotoxicity. Exp Neurol. 1999 Jan. 155(1):11-21. [View Abstract]
  59. Kisby GE, Standley M, Park T, et al. Proteomic analysis of the genotoxicant methylazoxymethanol (MAM)-induced changes in the developing cerebellum. J Proteome Res. 2006 Oct. 5(10):2656-65. [View Abstract]
  60. Ravits J, Paul P, Jorg C. Focality of upper and lower motor neuron degeneration at the clinical onset of ALS. Neurology. 2007 May 8. 68(19):1571-5. [View Abstract]
  61. Körner S, Kollewe K, Fahlbusch M, et al. Onset and spreading patterns of upper and lower motor neuron symptoms in amyotrophic lateral sclerosis. Muscle Nerve. 2011 May. 43(5):636-42. [View Abstract]
  62. Armon C. From clues to mechanisms: understanding ALS initiation and spread. Neurology. 2008 Sep 16. 71(12):872-3. [View Abstract]
  63. Frank SA. Evolution in health and medicine Sackler colloquium: Somatic evolutionary genomics: mutations during development cause highly variable genetic mosaicism with risk of cancer and neurodegeneration. Proc Natl Acad Sci U S A. 2010 Jan 26. 107 Suppl 1:1725-30. [View Abstract]
  64. Al-Chalabi A, Calvo A, Chio A, Colville S, Ellis CM, et al. Analysis of amyotrophic lateral sclerosis as a multistep process: a population-based modelling study. Lancet Neurol. 2014 Nov. 13 (11):1108-13. [View Abstract]
  65. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011 Oct 20. 72(2):245-56. [View Abstract]
  66. Renton AE, Majounie E, Waite A, Simón-Sánchez J, Rollinson S, Gibbs JR, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 2011 Oct 20. 72(2):257-68. [View Abstract]
  67. Cooper-Knock J, Hewitt C, Highley JR, Brockington A, Milano A, Man S, et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain. 2012 Mar. 135:751-64. [View Abstract]
  68. Chiò A, Borghero G, Restagno G, Mora G, Drepper C, Traynor BJ, et al. Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72. Brain. 2012 Mar. 135:784-93. [View Abstract]
  69. Byrne S, Elamin M, Bede P, Shatunov A, Walsh C, Corr B, et al. Cognitive and clinical characteristics of patients with amyotrophic lateral sclerosis carrying a C9orf72 repeat expansion: a population-based cohort study. Lancet Neurol. 2012 Mar. 11(3):232-40. [View Abstract]
  70. Stewart H, Rutherford NJ, Briemberg H, Krieger C, Cashman N, Fabros M, et al. Clinical and pathological features of amyotrophic lateral sclerosis caused by mutation in the C9ORF72 gene on chromosome 9p. Acta Neuropathol. 2012 Mar. 123(3):409-17. [View Abstract]
  71. Wijesekera LC, Mathers S, Talman P, Galtrey C, Parkinson MH, Ganesalingam J, et al. Natural history and clinical features of the flail arm and flail leg ALS variants. Neurology. 2009 Mar 24. 72(12):1087-94. [View Abstract]
  72. Al-Chalabi A, Lewis CM. Modelling the effects of penetrance and family size on rates of sporadic and familial disease. Hum Hered. 2011. 71(4):281-8. [View Abstract]
  73. Hanby MF, Scott KM, Scotton W, Wijesekera L, Mole T, Ellis CE, et al. The risk to relatives of patients with sporadic amyotrophic lateral sclerosis. Brain. 2011 Dec. 134:3454-7. [View Abstract]
  74. Pasinelli P, Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nat Rev Neurosci. 2006 Sep. 7(9):710-23. [View Abstract]
  75. Fogh I, Rijsdijk F, Andersen PM, Sham PC, Knight J, Neale B, et al. Age at onset in sod1-mediated amyotrophic lateral sclerosis shows familiality. Neurogenetics. 2007 Aug. 8(3):235-6. [View Abstract]
  76. Ahmeti KB, Ajroud-Driss S, Al-Chalabi A, Andersen PM, Armstrong J, et al. The Algen Consortium. Age of onset of amyotrophic lateral sclerosis is modulated by a locus on 1p34.1. Neurobiol Aging. 2013 Jan. 34(1):357.e7-19. [View Abstract]
  77. Andersen PM, Al-Chalabi A. Clinical genetics of amyotrophic lateral sclerosis: what do we really know?. Nat Rev Neurol. 2011 Oct 11. 7(11):603-15. [View Abstract]
  78. Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol. 2011 Nov. 7(11):616-30. [View Abstract]
  79. Wu CH, Fallini C, Ticozzi N, Keagle PJ, Sapp PC, Piotrowska K, et al. Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis. Nature. 2012 Aug 23. 488(7412):499-503. [View Abstract]
  80. Andersen PM. Amyotrophic lateral sclerosis genetics with Mendelian inheritance. Brown RH Jr, Swash M and Pasinelli P, eds. Amyotrophic Lateral Sclerosis. 2nd edition. Informa healthcare; 2006. 187-207.
  81. Saeed M, Yang Y, Deng HX, et al. Age and founder effect of SOD1 A4V mutation causing ALS. Neurology. 2009 May 12. 72(19):1634-9. [View Abstract]
  82. Saito Y, Yokota T, Mitani T, et al. Transgenic small interfering RNA halts amyotrophic lateral sclerosis in a mouse model. J Biol Chem. 2005 Dec 30. 280(52):42826-30. [View Abstract]
  83. Al-Chalabi A, Jones A, Troakes C, King A, Al-Sarraj S, van den Berg LH. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol. 2012 Sep. 124(3):339-52. [View Abstract]
  84. Arai T, Mackenzie IR, Hasegawa M, et al. Phosphorylated TDP-43 in Alzheimer's disease and dementia with Lewy bodies. Acta Neuropathol. 2009 Feb. 117(2):125-36. [View Abstract]
  85. Morita M, Al-Chalabi A, Andersen PM, Hosler B, Sapp P, Englund E, et al. A locus on chromosome 9p confers susceptibility to ALS and frontotemporal dementia. Neurology. 2006 Mar 28. 66(6):839-44. [View Abstract]
  86. Vance C, Al-Chalabi A, Ruddy D, Smith BN, Hu X, Sreedharan J, et al. Familial amyotrophic lateral sclerosis with frontotemporal dementia is linked to a locus on chromosome 9p13.2-21.3. Brain. 2006 Apr. 129:868-76. [View Abstract]
  87. van Es MA, Veldink JH, Saris CG, Blauw HM, van Vught PW, Birve A, et al. Genome-wide association study identifies 19p13.3 (UNC13A) and 9p21.2 as susceptibility loci for sporadic amyotrophic lateral sclerosis. Nat Genet. 2009 Oct. 41(10):1083-7. [View Abstract]
  88. Shatunov A, Mok K, Newhouse S, Weale ME, Smith B, Vance C. Chromosome 9p21 in sporadic amyotrophic lateral sclerosis in the UK and seven other countries: a genome-wide association study. Lancet Neurol. 2010 Oct. 9(10):986-94. [View Abstract]
  89. Laaksovirta H, Peuralinna T, Schymick JC, Scholz SW, Lai SL, Myllykangas L. Chromosome 9p21 in amyotrophic lateral sclerosis in Finland: a genome-wide association study. Lancet Neurol. 2010 Oct. 9(10):978-85. [View Abstract]
  90. Smith BN, Newhouse S, Shatunov A, Vance C, Topp S, Johnson L, et al. The C9ORF72 expansion mutation is a common cause of ALS+/-FTD in Europe and has a single founder. Eur J Hum Genet. 2013 Jan. 21(1):102-8. [View Abstract]
  91. Al-Sarraj S, King A, Troakes C, Smith B, Maekawa S, Bodi I, et al. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011 Dec. 122(6):691-702. [View Abstract]
  92. Deng HX, Chen W, Hong ST, Boycott KM, Gorrie GH, Siddique N, et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature. 2011 Aug 21. 477(7363):211-5. [View Abstract]
  93. Majoor-Krakauer D, Ottman R, Johnson WG, Rowland LP. Familial aggregation of amyotrophic lateral sclerosis, dementia, and Parkinson's disease: evidence of shared genetic susceptibility. Neurology. 1994 Oct. 44(10):1872-7. [View Abstract]
  94. Fallis BA, Hardiman O. Aggregation of neurodegenerative disease in ALS kindreds. Amyotroph Lateral Scler. 2009 Apr. 10(2):95-8. [View Abstract]
  95. Cruz DC, Nelson LM, McGuire V, Longstreth WT Jr. Physical trauma and family history of neurodegenerative diseases in amyotrophic lateral sclerosis: a population-based case-control study. Neuroepidemiology. 1999. 18(2):101-10. [View Abstract]
  96. Nelson LM, McGuire V, Longstreth WT Jr, Matkin C. Population-based case-control study of amyotrophic lateral sclerosis in western Washington State. I. Cigarette smoking and alcohol consumption. Am J Epidemiol. 2000 Jan 15. 151(2):156-63. [View Abstract]
  97. Kamel F, Umbach DM, Munsat TL, Shefner JM, Sandler DP. Association of cigarette smoking with amyotrophic lateral sclerosis. Neuroepidemiology. 1999. 18(4):194-202. [View Abstract]
  98. Gallo V, Bueno-De-Mesquita HB, Vermeulen R, Andersen PM, Kyrozis A, Linseisen J. Smoking and risk for amyotrophic lateral sclerosis: analysis of the EPIC cohort. Ann Neurol. 2009 Apr. 65(4):378-85. [View Abstract]
  99. Sutedja NA, Veldink JH, Fischer K, Kromhout H, Wokke JH, Huisman MH. Lifetime occupation, education, smoking, and risk of ALS. Neurology. 2007 Oct 9. 69(15):1508-14. [View Abstract]
  100. Weisskopf MG, O'Reilly EJ, McCullough ML, et al. Prospective study of military service and mortality from ALS. Neurology. 2005 Jan 11. 64(1):32-7. [View Abstract]
  101. Horner RD, Kamins KG, Feussner JR, et al. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology. 2003 Sep 23. 61(6):742-9. [View Abstract]
  102. Armon C. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology. 2004 Mar 23. 62(6):1027; author reply 1027-9. [View Abstract]
  103. Armon C. Excess incidence of ALS in young Gulf War veterans. Neurology. 2004 Nov 23. 63(10):1986-7; author reply 1986-7. [View Abstract]
  104. Coffman CJ, Horner RD, Grambow SC, Lindquist J. Estimating the occurrence of amyotrophic lateral sclerosis among Gulf War (1990-1991) veterans using capture-recapture methods. Neuroepidemiology. 2005. 24(3):141-50. [View Abstract]
  105. Horner RD, Grambow SC, Coffman CJ, et al. Amyotrophic lateral sclerosis among 1991 Gulf War veterans: evidence for a time-limited outbreak. Neuroepidemiology. 2008. 31(1):28-32. [View Abstract]
  106. Barth SK, Kang HK, Bullman TA, Wallin MT. Neurological mortality among U.S. veterans of the Persian Gulf War: 13-year follow-up. Am J Ind Med. 2009 Sep. 52(9):663-70. [View Abstract]
  107. Sutedja NA, Veldink JH, Fischer K, Kromhout H, Heederik D, Huisman MH, et al. Exposure to chemicals and metals and risk of amyotrophic lateral sclerosis: a systematic review. Amyotroph Lateral Scler. 2009 Oct-Dec. 10(5-6):302-9. [View Abstract]
  108. Popat RA, Van Den Eeden SK, Tanner CM, et al. Effect of reproductive factors and postmenopausal hormone use on the risk of amyotrophic lateral sclerosis. Neuroepidemiology. 2006. 27(3):117-21. [View Abstract]
  109. Beghi E, Logroscino G, Chiò A, Hardiman O, Millul A, Mitchell D, et al. Amyotrophic lateral sclerosis, physical exercise, trauma and sports: results of a population-based pilot case-control study. Amyotroph Lateral Scler. 2010 May 3. 11(3):289-92. [View Abstract]
  110. Chiò A, Benzi G, Dossena M, Mutani R, Mora G. Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players. Brain. 2005 Mar. 128:472-6. [View Abstract]
  111. Belli S, Vanacore N. Proportionate mortality of Italian soccer players: is amyotrophic lateral sclerosis an occupational disease?. Eur J Epidemiol. 2005. 20(3):237-42. [View Abstract]
  112. Armon C. Sports and trauma in amyotrophic lateral sclerosis revisited. J Neurol Sci. 2007 Nov 15. 262(1-2):45-53. [View Abstract]
  113. Chio A, Traynor BJ, Swingler R, eet al. Amyotrophic lateral sclerosis and soccer: a different epidemiological approach strengthen the previous findings. J Neurol Sci. 2008 Jun 15. 269(1-2):187-8; author reply 188-9. [View Abstract]
  114. Belli S, Vanacore N. Sports and amyotrophic lateral sclerosis. J Neurol Sci. 2008 Jun 15. 269(1-2):191; author reply 191-2. [View Abstract]
  115. Armon C. Amyotrophic lateral sclerosis and soccer: a different epidemiological approach strengthen the previous findings. J Neurol Sci. 2008. 269:188-189 (author reply).
  116. Armon C. Response to Belli and Vanacore. J Neurol Sci. 2008. 269:191192 (author reply).
  117. 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 Aug. 10(4):205-9. [View Abstract]
  118. McKee AC, Gavett BE, Stern RA, Nowinski CJ, Cantu RC, Kowall NW, et al. TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 2010 Sep. 69(9):918-29. [View Abstract]
  119. Appel SH, Cwik VA, Day JW. Trauma, TDP-43, and amyotrophic lateral sclerosis. Muscle Nerve. 2010 Dec. 42(6):851-2. [View Abstract]
  120. Bedlack RS, Genge A, Amato AA, Shaibani A, Jackson CE, Kissel JT, et al. Correspondence regarding: TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2010:69;918-29. J Neuropathol Exp Neurol. 2011 Jan. 70(1):96-7; author reply 98-100. [View Abstract]
  121. Armon C, Miller RG. Correspondence regarding: TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2010:69;918-29. J Neuropathol Exp Neurol. 2011 Jan. 70(1):97-8; author reply 98-100. [View Abstract]
  122. Lehman EJ, Hein MJ, Baron SL, Gersic CM. Neurodegenerative causes of death among retired National Football League players. Neurology. 2012 Nov 6. 79(19):1970-4. [View Abstract]
  123. Armon C, Nelson LM. Is head trauma a risk factor for amyotrophic lateral sclerosis? An evidence based review. Amyotroph Lateral Scler. 2012 Jun. 13(4):351-6. [View Abstract]
  124. Peters TL, Fang F, Weibull CE, Sandler DP, Kamel F, Ye W. Severe head injury and amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2013 May. 14(4):267-72. [View Abstract]
  125. Kurland LT, Radhakrishnan K, Smith GE, Armon C, Nemetz PN. Mechanical trauma as a risk factor in classic amyotrophic lateral sclerosis: lack of epidemiologic evidence. J Neurol Sci. 1992 Dec. 113(2):133-43. [View Abstract]
  126. 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. 87:335-340.
  127. Wicks P, Ganesalingham J, Collin C, Prevett M, Leigh NP, Al-Chalabi A. Three soccer playing friends with simultaneous amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2007 Jun. 8(3):177-9. [View Abstract]
  128. Fournier CN, Gearing M, Upadhyayula SR, Klein M, Glass JD. Head injury does not alter disease progression or neuropathologic outcomes in ALS. Neurology. 2015 Apr 28. 84 (17):1788-95. [View Abstract]
  129. Whiting MG. Toxicity of Cycads: A Literature Review. Economic Botany. 1963. 68:270-302.
  130. Borenstein AR, Mortimer JA, Schellenberg GD, Galasko D. The ALS/PDC syndrome of Guam and the cycad hypothesis. Neurology. 2009 Feb 3. 72(5):473, 476; author reply 475-6. [View Abstract]
  131. Steele JC, McGeer PL. The ALS/PDC syndrome of Guam and the cycad hypothesis. Neurology. 2008 May 20. 70(21):1984-90. [View Abstract]
  132. Borenstein AR, Mortimer JA, Schofield E, et al. Cycad exposure and risk of dementia, MCI, and PDC in the Chamorro population of Guam. Neurology. 2007 May 22. 68(21):1764-71. [View Abstract]
  133. Cox PA, Banack SA, Murch SJ. Biomagnification of cyanobacterial neurotoxins and neurodegenerative disease among the Chamorro people of Guam. Proc Natl Acad Sci U S A. 2003 Nov 11. 100(23):13380-3. [View Abstract]
  134. Khabazian I. Isolation of various forms of sterol B-D-glucoside from the seed of Cycas circinalis: neurotoxicity and implications for the ALS-parkinsonian dementia complex. J Neurochem. 2002. 82:516-528.
  135. Chiò A, Mora G, Calvo A, Mazzini L, Bottacchi E, Mutani R. Epidemiology of ALS in Italy: a 10-year prospective population-based study. Neurology. 2009 Feb 24. 72(8):725-31. [View Abstract]
  136. Alonso A, Logroscino G, Jick SS, Hernán MA. Incidence and lifetime risk of motor neuron disease in the United Kingdom: a population-based study. Eur J Neurol. 2009 Jun. 16(6):745-51. [View Abstract]
  137. Johnston CA, Stanton BR, Turner MR, Gray R, Blunt AH, Butt D, et al. Amyotrophic lateral sclerosis in an urban setting: a population based study of inner city London. J Neurol. 2006 Dec. 253(12):1642-3. [View Abstract]
  138. Johnston CA, Stanton BR, Turner MR, Gray R, Blunt AH, Butt D, et al. Amyotrophic lateral sclerosis in an urban setting: a population based study of inner city London. J Neurol. 2006 Dec. 253(12):1642-3. [View Abstract]
  139. Cronin S, Hardiman O, Traynor BJ. Ethnic variation in the incidence of ALS: a systematic review. Neurology. 2007 Mar 27. 68(13):1002-7. [View Abstract]
  140. Zaldivar T, Gutierrez J, Lara G, Carbonara M, Logroscino G, Hardiman O. Reduced frequency of ALS in an ethnically mixed population: a population-based mortality study. Neurology. 2009 May 12. 72(19):1640-5. [View Abstract]
  141. Rojas-Garcia R, Scott KM, Roche JC, Scotton W, Martin N, Janssen A, et al. No evidence for a large difference in ALS frequency in populations of African and European origin: a population based study in inner city London. Amyotroph Lateral Scler. 2012 Jan. 13(1):66-8. [View Abstract]
  142. Sabel CE, Boyle PJ, Löytönen M, et al. Spatial clustering of amyotrophic lateral sclerosis in Finland at place of birth and place of death. Am J Epidemiol. 2003 May 15. 157(10):898-905. [View Abstract]
  143. Manjaly ZR, Scott KM, Abhinav K, Wijesekera L, Ganesalingam J, Goldstein LH, et al. The sex ratio in amyotrophic lateral sclerosis: A population based study. Amyotroph Lateral Scler. 2010 Oct. 11(5):439-42. [View Abstract]
  144. Kimura F, Fujimura C, Ishida S, et al. Progression rate of ALSFRS-R at time of diagnosis predicts survival time in ALS. Neurology. 2006 Jan 24. 66(2):265-7. [View Abstract]
  145. Turner MR, Scaber J, Goodfellow JA, Lord ME, Marsden R, Talbot K. The diagnostic pathway and prognosis in bulbar-onset amyotrophic lateral sclerosis. J Neurol Sci. 2010 Jul 15. 294(1-2):81-5. [View Abstract]
  146. Murphy J, Henry R, Lomen-Hoerth C. Establishing subtypes of the continuum of frontal lobe impairment in amyotrophic lateral sclerosis. Arch Neurol. 2007 Mar. 64(3):330-4. [View Abstract]
  147. Phukan J, Elamin M, Bede P, Jordan N, Gallagher L, Byrne S, et al. The syndrome of cognitive impairment in amyotrophic lateral sclerosis: a population-based study. J Neurol Neurosurg Psychiatry. 2012 Jan. 83(1):102-8. [View Abstract]
  148. Elamin M, Phukan J, Bede P, Jordan N, Byrne S, Pender N, et al. Executive dysfunction is a negative prognostic indicator in patients with ALS without dementia. Neurology. 2011 Apr 5. 76(14):1263-9. [View Abstract]
  149. Chiò A, Logroscino G, Hardiman O, Swingler R, Mitchell D, Beghi E, et al. Prognostic factors in ALS: A critical review. Amyotroph Lateral Scler. 2009 Oct-Dec. 10(5-6):310-23. [View Abstract]
  150. Kimura F, Fujimura C, Ishida S, Nakajima H, Furutama D, Uehara H et al. Progression rate of ALSFRS-R at time of diagnosis predicts survival time in ALS. Neurology. 2006. 66:265-267.
  151. Armon C, Moses D. Linear estimates of rates of disease progression as predictors of survival in patients with ALS entering clinical trials. J Neurol Sci. 1998 Oct. 160 Suppl 1:S37-41. [View Abstract]
  152. Armon C, Graves MC, Moses D, Forté DK, Sepulveda L, Darby SM, et al. Linear estimates of disease progression predict survival in patients with amyotrophic lateral sclerosis. Muscle Nerve. 2000 Jun. 23(6):874-82. [View Abstract]
  153. Armon C, Brandstater ME. Motor unit number estimate-based rates of progression of ALS predict patient survival. Muscle Nerve. 1999 Nov. 22(11):1571-5. [View Abstract]
  154. Czaplinski A, Yen AA, Appel SH. Amyotrophic lateral sclerosis: early predictors of prolonged survival. J Neurol. 2006 Nov. 253(11):1428-36. [View Abstract]
  155. Roche JC, Rojas-Garcia R, Scott KM, Scotton W, Ellis CE, Burman R, et al. A proposed staging system for amyotrophic lateral sclerosis. Brain. 2012 Mar. 135:847-52. [View Abstract]
  156. Kim WK, Liu X, Sandner J, Pasmantier M, Andrews J, Rowland LP, et al. Study of 962 patients indicates progressive muscular atrophy is a form of ALS. Neurology. 2009 Nov 17. 73(20):1686-92. [View Abstract]
  157. Armon C, Schultz JD. Preferences of patients with ALS for accurate prognostic information. Presented at the annual meeting of the International Alliance of Motor Neuron Disease Associations. Philadelphia. December 2004.
  158. Armon C. ALS 1996 and Beyond: New Hopes and Challenges. A Manual for Patients, Families and Friends. Loma Linda University Department of Neurology, Loma Linda, California. Fourth Edition. 2007.
  159. Isaacs JD, Dean AF, Shaw CE, Al-Chalabi A, Mills KR, Leigh PN. Amyotrophic lateral sclerosis with sensory neuropathy: part of a multisystem disorder?. J Neurol Neurosurg Psychiatry. 2007 Jul. 78(7):750-3. [View Abstract]
  160. Byrne S, Elamin M, Bede P, Hardiman O. Absence of consensus in diagnostic criteria for familial neurodegenerative diseases. J Neurol Neurosurg Psychiatry. 2012 Apr. 83(4):365-7. [View Abstract]
  161. Byrne S, Bede P, Elamin M, Kenna K, Lynch C, McLaughlin R, et al. Proposed criteria for familial amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2011 May. 12(3):157-9. [View Abstract]
  162. de Carvalho M, Kiernan MC, Swash M. Fasciculation in amyotrophic lateral sclerosis: origin and pathophysiological relevance. J Neurol Neurosurg Psychiatry. 2017 May 10. [View Abstract]
  163. Woolley SC, Strong MJ. Frontotemporal Dysfunction and Dementia in Amyotrophic Lateral Sclerosis. Neurol Clin. 2015 Nov. 33 (4):787-805. [View Abstract]
  164. Brooks BR. El Escorial World Federation of Neurology criteria for the diagnosis of amyotrophic lateral sclerosis. Subcommittee on Motor Neuron Diseases/Amyotrophic Lateral Sclerosis of the World Federation of Neurology Research Group on Neuromuscular Diseases and the El Escorial "Clinical limits of amyotrophic lateral sclerosis" workshop contributors. J Neurol Sci. 1994 Jul. 124 Suppl:96-107. [View Abstract]
  165. de Carvalho M, Dengler R, Eisen A, England JD, Kaji R, Kimura J, et al. Electrodiagnostic criteria for diagnosis of ALS. Clin Neurophysiol. 2008 Mar. 119 (3):497-503. [View Abstract]
  166. de Carvalho M, Dengler R, Eisen A, et al. Electrodiagnostic criteria for diagnosis of ALS. Clin Neurophysiol. 2008 Mar. 119(3):497-503. [View Abstract]
  167. Chiò A, Battistini S, Calvo A, Caponnetto C, Conforti FL, Corbo M, et al. Genetic counselling in ALS: facts, uncertainties and clinical suggestions. J Neurol Neurosurg Psychiatry. 2014 May. 85 (5):478-85. [View Abstract]
  168. Bromberg MB, Swoboda KJ, Lawson VH. Counting motor units in chronic motor neuropathies. Exp Neurol. 2003 Nov. 184 Suppl 1:S53-7. [View Abstract]
  169. Armon C, Brandstater ME. Motor unit number estimate-based rates of progression of ALS predict patient survival. Muscle Nerve. 1999 Nov. 22(11):1571-5. [View Abstract]
  170. Cedarbaum JM, Stambler N. Performance of the Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS) in multicenter clinical trials. J Neurol Sci. 1997 Oct. 152 Suppl 1:S1-9. [View Abstract]
  171. Cedarbaum JM, Stambler N, Malta E, et al. The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. BDNF ALS Study Group (Phase III). J Neurol Sci. 1999 Oct 31. 169(1-2):13-21. [View Abstract]
  172. Instructions for completing the ALSFRS-R. (ALS Functional Rating Scale). ALS Connection.
  173. [Guideline] 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. 1999 Apr 22. 52(7):1311-23. [View Abstract]
  174. Bedlack RS, Mitsumoto H, Eds. Amyotrophic Lateral Sclerosis: A Patient Care Guide for Clinicians. New York: Demos Medical Publishing; 2012.
  175. Amyotrophic Lateral Sclerosis Association. Available at http://www.alsa.org
  176. Muscular Dystrophy Association. Available at http://als.mdausa.org/
  177. Miller RG, Mitchell JD, Lyon M, Moore DH. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst Rev. 2007 Jan 24. CD001447. [View Abstract]
  178. Bensimon G, Lacomblez L, Delumeau JC, Bejuit R, Truffinet P, Meininger V. A study of riluzole in the treatment of advanced stage or elderly patients with amyotrophic lateral sclerosis. J Neurol. 2002 May. 249(5):609-15. [View Abstract]
  179. Yanagisawa N, Tashiro K, Tohgi H, et al. Efficacy and safety of riluzole in patients with amyotrophic lateral sclerosis: double-blind placebo-controlled study in Japan. Igakuno Ayumi. 1997. 182:851-866. [Article in Japanese].
  180. Yanagisawa N, Shindo M. [Neuroprotective therapy for amyotrophic lateral sclerosis (ALS)]. Rinsho Shinkeigaku. 1996 Dec. 36(12):1329-30. [View Abstract]
  181. Armon C, Guiloff RJ, Bedlack R. Limitations of inferences from observational databases in amyotrophic lateral sclerosis: all that glitters is not gold. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002 Sep. 3(3):109-12. [View Abstract]
  182. Tanaka M, Sakata T, Palumbo J, and Akimoto M. A 24-Week, Phase III, Double-Blind, Parallel-Group Study of Edaravone (MCI-186) for Treatment of Amyotrophic Lateral Sclerosis (ALS) (P3.189). Presented at the American Academy of Neurology Annual Meeting. Vancouver, BC. American Academy of Neurology. Available at http://www.neurology.org/content/86/16_Supplement/P3.189. 2016 Apr 18; Accessed: 2017 May 08.
  183. Bourke SC, Tomlinson M, Williams TL, Bullock RE, Shaw PJ, Gibson GJ. Effects of non-invasive ventilation on survival and quality of life in patients with amyotrophic lateral sclerosis: a randomised controlled trial. Lancet Neurol. 2006. 5:140-147. [View Abstract]
  184. Jackson CE, Gronseth G, Rosenfeld J, et al. Randomized double-blind study of botulinum toxin type B for sialorrhea in ALS patients. Muscle Nerve. 2009 Feb. 39(2):137-43. [View Abstract]
  185. Neppelberg E, Haugen DF, Thorsen L, Tysnes OB. Radiotherapy reduces sialorrhea in amyotrophic lateral sclerosis. Eur J Neurol. 2007 Dec. 14(12):1373-7. [View Abstract]
  186. Brooks BR, Thisted RA, Appel SH, et al. Treatment of pseudobulbar affect in ALS with dextromethorphan/quinidine: a randomized trial. Neurology. 2004 Oct 26. 63(8):1364-70. [View Abstract]
  187. Pioro EP, Brooks BR, Cummings J, Schiffer R, Thisted RA, Wynn D. Dextromethorphan plus ultra low-dose quinidine reduces pseudobulbar affect. Ann Neurol. 2010 Nov. 68(5):693-702. [View Abstract]
  188. Baile WF, Buckman R, Lenzi R, Glober G, Beale EA, Kudelka AP. SPIKES-A six-step protocol for delivering bad news: application to the patient with cancer. Oncologist. 2000. 5(4):302-11. [View Abstract]
  189. VA Secretary Establishes ALS as a Presumptive Compensable Illness. September 23, 2008. VA Secretary Establishes ALS as a Presumptive Compensable Illness. September 23, 2008.
  190. Lo Coco D, Marchese S, Pesco MC, La Bella V, Piccoli F, Lo Coco A. Noninvasive positive-pressure ventilation in ALS: predictors of tolerance and survival. Neurology. 2006 Sep 12. 67(5):761-5. [View Abstract]
  191. Harding A. PET scans distinguish ALS patients from healthy controls. Reuters Health Information. March 11, 2014.
  192. Migliore L, Coppede F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2008 Oct 31. [View Abstract]
  193. Valdmanis PN, Rouleau GA. Genetics of familial amyotrophic lateral sclerosis. Neurology. 2008 Jan 8. 70(2):144-52. [View Abstract]
  194. Van Laere K, Vanhee A, Verschueren J, et al. Value of 18fluorodeoxyglucose-positron-emission tomography in amyotrophic lateral sclerosis: a prospective study. JAMA Neurol. 2014 Mar 10. [View Abstract]

The 4 regions or levels of the body. Bulbar (muscles of the face, mouth, and throat); cervical (muscles of the back of the head and the neck, the shoulders and upper back, and the upper extremities); thoracic (muscles of the chest and abdomen and the middle portion of the spinal muscles); lumbosacral (muscles of the lower back, groin, and lower extremities).

Muscle in nerve disease. Image courtesy of Dr. Friedlander, Associate Professor and Chair of Pathology at Kansas City University of Medicine and Biosciences.

The genetic/environmental/age- and time-dependent interactions hypothesis for amyotrophic lateral sclerosis (ALS). Risk factors operate upstream to a putative biochemical transformation (likely an acquired nucleic acid or protein change), which causes the appearance of altered proteins or nucleic acids or abnormal quantities of normal proteins or nucleid acids. These agents spread within the motor system and cause the downstream disintegration of the motor system and the downstream biochemical, histologic, and clinical consequences of ALS. (Adapted from Armon C. What is ALS? In: Amyotrophic Lateral Sclerosis: A Patient Care Guide for Clinicians. Bedlack RS, Mitsumoto H, Eds. Demos Medical Publishing, New York, 2012:1-23)

The 4 regions or levels of the body. Bulbar (muscles of the face, mouth, and throat); cervical (muscles of the back of the head and the neck, the shoulders and upper back, and the upper extremities); thoracic (muscles of the chest and abdomen and the middle portion of the spinal muscles); lumbosacral (muscles of the lower back, groin, and lower extremities).

The 4 regions or levels of the body. Bulbar (muscles of the face, mouth, and throat); cervical (muscles of the back of the head and the neck, the shoulders and upper back, and the upper extremities); thoracic (muscles of the chest and abdomen and the middle portion of the spinal muscles); lumbosacral (muscles of the lower back, groin, and lower extremities).

The genetic/environmental/age- and time-dependent interactions hypothesis for amyotrophic lateral sclerosis (ALS). Risk factors operate upstream to a putative biochemical transformation (likely an acquired nucleic acid or protein change), which causes the appearance of altered proteins or nucleic acids or abnormal quantities of normal proteins or nucleid acids. These agents spread within the motor system and cause the downstream disintegration of the motor system and the downstream biochemical, histologic, and clinical consequences of ALS. (Adapted from Armon C. What is ALS? In: Amyotrophic Lateral Sclerosis: A Patient Care Guide for Clinicians. Bedlack RS, Mitsumoto H, Eds. Demos Medical Publishing, New York, 2012:1-23)

Muscle in nerve disease. Image courtesy of Dr. Friedlander, Associate Professor and Chair of Pathology at Kansas City University of Medicine and Biosciences.

Gene Locus Protein Inheritance
SOD1



(ALS1)



21q22.11Superoxide dismutase 1 (SOD1)AD*
ALS2 2q33Alsin (ALS2)Juvenile/AR**
ALS3 18q21UnknownAD
ALS4 9q34SETXJuvenile/AD
ALSS 15q15SPG11Juvenile/AR
FUS (ALS6)16p11.2FUSAD
ALS7 20ptel-p13UnknownAD
ALS8 20q13.3VABPAD
ALS9 14q11.2Angiogenin (ANG)AD
TARDBP (ALS10)1p36.2TAR DNA-binding protein (TARDBP)AD
ALS11 6q21FIG4AD
ALS12 10p13OPTNAD; AR
ALS13 12q24ATXN2AD
ALSX Xp11UBQLN2X-linked
C9orf72 (ALS-FTD)9q21-22C9ORF72AD
ALS-FTD 9p13.3SIGMAR1AD; Juvenile/AR
PFL1 17p13.2Profilin 1AD
*AD–autosomal dominant; **AR—autosomal recessive