Skip to main content
Springer logoLink to Springer
. 2025 Sep 2;39(10):949–993. doi: 10.1007/s40263-025-01217-0

Genetic and Mechanistic Insights Inform Amyotrophic Lateral Sclerosis Treatment and Symptomatic Management: Current and Emerging Therapeutics and Clinical Trial Design Considerations

Savannah E Quigley 1, Kellen H Quigg 1, Stephen A Goutman 1,2,
PMCID: PMC12423166  PMID: 40897992

Abstract

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder affecting both upper and lower motor neurons. ALS is classically characterized by painless progressive weakness, causing impaired function of limbs, speech, swallowing, and respiratory function. The disease is fatal within 2–4 years, often the result of respiratory failure. The pathologic hallmark for a majority of ALS cases is aberrant cytoplasmic accumulations of the nuclear protein TAR-DNA binding protein (TDP-43). A total of 10–15% of ALS can be attributed to a single gene mutation, known as genetic or “familial” ALS, while the remainder of cases are termed nongenetic or “sporadic” although heritability has been measured in up to 37% in this population. Complex interactions between genetics, environment, and physiologic susceptibility are thought to contribute to disease. Management is primarily supportive in nature, though there are several approved treatments worldwide. This review details the mechanisms and evidence of approved disease-modifying treatments, relevant measures to track disease burden and progression used in clinical trials, and approaches to pharmacologic management of common symptoms in ALS. As there is not currently a cure for ALS, research into the complex pathophysiologic and genetic alterations contributing to disease is of great interest. This review further discusses the current understanding of genetic etiologies and altered physiology leading to disease, such as neuroinflammation, integrated stress response, aberrant proteostasis and mitochondrial dysfunction, among others. The translation of preclinical discoveries into current investigational therapeutics, novel therapeutic categories such as antisense oligonucleotides and stem cell transplantation, as well as future horizons harnessing the power of artificial intelligence in drug development and clinical trials are discussed.

Key Points

Management of amyotrophic lateral sclerosis is best delivered in a multidisciplinary clinic and utilizes pharmacologic and non-pharmacologic approaches.
An improved understanding of amyotrophic lateral sclerosis pathomechanisms informs new therapeutic targets.
The therapeutic landscape of amyotrophic lateral sclerosis is very active, with numerous clinical trials.

Introduction

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder affecting, most notably, both upper and lower motor neurons in the brain, brainstem, and spinal cord, leading to progressive weakness of limbs and bulbar muscles as well as neuromuscular respiratory failure [1]. Up to 40–50% of patients with ALS demonstrate mild-to-moderate cognitive impairment, primarily involving executive dysfunction and impaired verbal fluency, with 15% of patients meeting criteria for frontotemporal dementia (FTD) [2]. Most patients who forego invasive mechanical ventilation via tracheostomy die within 2–4 years from diagnosis, primarily from respiratory failure and complications of bulbar and diaphragmatic weakness [3]. The observed prevalence of ALS in the USA is 6.6/100,000 with a predominance in male sex, white race, and age > 66 years [4]. Presentation is often insidious and motor and cognitive phenotypes vary greatly between patients, contributing to diagnostic delays of on average 12 months from symptom onset, a timeline that has unfortunately not declined over the last decade [5]. Shortening the time from the onset of ALS symptoms to diagnosis is essential to evaluate and initiate treatments and therapeutics sooner, especially if they exert a meaningful disease-modifying effect. In total, 85–90% of ALS cases are “sporadic,” occurring without known genetic cause or family history. Even in these cases, heritability has been estimated to be up to 36.9% [6]. Alternatively, 10–15% of ALS cases can be attributed to a single gene mutation, and these mutations can be found in patients without a known family history. A total of 5–10% of cases are termed genetic, or “familial,” ALS and have Mendelian inheritance; yet penetrance is incomplete [6], perhaps resulting from environmental exposures. To date, at least 40 genes have been linked to ALS [7], with only a subset felt to be directly causative. Among diverse and complex genetic mechanisms, many mutations lead to protein aggregates with toxic gain of function, though loss of protein function, particularly in the nucleus, is important as well [8].

In > 97% of patients with ALS (and 50% with FTD), a hallmark pathologic finding is accumulation of TAR-DNA binding protein (TDP-43) [9], a nuclear protein normally involved in RNA and cryptic exon splicing [8]. When TDP-43 is mislocalized from the nucleus to the cytoplasm, there is both loss of nuclear function and toxic sequestration of proteins and ribonucleic acids (RNAs) within the aggregates. The superoxide dismutase 1 (SOD1) and fused in sarcoma (FUS) genetic forms of ALS are exceptions as they do not develop TDP-43 aggregates, but instead show cytoplasmic SOD1 and FUS aggregates, respectively [8, 10, 11].

Despite the prominence of aberrant TDP-43 accumulation in most forms of ALS, it is a heterogenous disease with multiple aberrant pathogenic mechanisms implicated in progression, which contributes to challenges in disease modeling and developing a single disease-modifying therapy. The initiating factor of ALS is hypothesized to be a combination of multigenic and environmental factors [7] acting as “multiple hits” toward a threshold at which genetic and epigenetic changes converge into a pathologic cycle of degeneration. Some mechanisms of motor neuron injury include cytoplasmic protein aggregation [12], pathologic RNA processing, dysregulated microglia with associated neuroinflammation, and oxidative stress, among others [8]. Drugs approved to date have focused on glutamate excitotoxicity and oxidative stress, discussed in detail below. Current trials have aimed to target the early state of disease to intervene prior to development of heterogenous mechanisms of neurodegeneration [5, 13]. An improved understanding of genetic forms of disease has more recently facilitated the identification of amenable targets for novel gene therapies in subsets of patients with ALS. Notwithstanding, environmental and epigenetic contributions to disease are still under intense investigation as they may influence disease penetrance and therapeutic responses.

Pharmacologic successes have unfortunately not kept pace with the increasing pathophysiologic insights into ALS, leading to only a few medications approved for use globally, none of which stop or reverse disease progression. Therefore, there is an urgent need for edifying understanding of ALS pathophysiology and genetic underpinnings and developing novel therapeutics. This lack of therapeutic success does result in some patients seeking off-label therapies, supplements, and other “treatments” that lack high-quality levels of evidence but provide hope of altering disease trajectory. These therapies are often not covered by insurance and can be quite costly, leading to inequitable access and variability of formulations and dosing. Patients with ALS may not report these complementary therapies, leading to risks of adverse effects and possible interference with clinical trial eligibility [13].

ALS Genetics

While only a minority of patients with ALS harbor a monogenic cause, the downstream consequences of genetic mutations in patients with ALS have improved mechanistic insights. In turn, the genetic mutations or resulting dysregulated mechanisms provide therapeutic targets. Of the dozens of known mutations, four—SOD1, chromosome 9 open reading frame 72 (C9orf72) hexanucleotide repeat expansion (HRE), transactive response DNA binding protein (TARDBP), and FUS—are most prevalent [14, 15].

SOD1 was the first identified genetic cause of ALS. Discovered in 1993, it is the second most common cause of familial/genetic ALS in those of European ancestry, affecting 12% of patients with familial ALS [7]. Normal SOD1 protein functions as an antioxidant enzyme. In ALS, mutations lead to misfolding or aberrant conformational changes via oxidation leading to toxic gain of function and anomalous protein aggregation [16].The aggregates, in turn, lead to oxidative stress and mitochondrial dysfunction [17]. Therapeutic approaches have focused on reducing SOD1 protein and mRNA levels [18]. Despite the eventual recognition that most forms of ALS have different pathologic hallmarks, the discovery of SOD1 led to the development of SOD1 transgenic mouse models, which continue to be the foundation for studying potential ALS therapies. However, by lacking TDP-43 pathology, they may not accurately predict therapeutic success for all forms of ALS [19]. Please see recent reviews on SOD1 ALS for more details [10].

The most common genetic mutation in both sporadic and familial forms of ALS is the C9orf72 HRE, representing 40% of familial cases [7]. C9orf72 HRE is also the most common genetic form of FTD, thereby linking the two diseases. C9orf72 plays a role in RNA processing and autophagy. In ALS, pathological intronic hexanucleotide GGGGCC expansions in the C9orf72 gene are thought to contribute to neurodegeneration by multiple mechanisms, including toxic cytoplasmic dipeptide repeat protein aggregates, toxic RNA gain-of-function, or loss of function due to haploinsufficiency [20, 21]. The reader is directed to other recent reviews [22] for more detailed information regarding C9orf72 pathophysiology.

Other notable causes of genetic ALS currently undergoing dedicated therapeutic study include FUS and TARDBP [7]. FUS, similar to SOD1, is not a TDP-43 proteinopathy [8]. It is a nuclear RNA binding protein that mediates transport of mRNA to the cytoplasm. Mutated FUS leads to cytoplasmic protein aggregates [12, 23]. Mutations in TANK-binding kinase (TBK1) [24] and NIMA-related kinase 1 (NEK1) [25, 26], though more prevalent than FUS, currently do not have any specific targeted therapeutics in trial. TARDBP is the gene encoding TDP-43 and leads to TDP-43 aggregates, similar to findings in nongenetic ALS, thus making it an intriguing target for intervention.

Clinical Outcomes in ALS

The US Food and Drug Administration (FDA) provides guidance on important outcomes in ALS clinical trials, such as mortality, functional status, respiratory and strength measures, and patient-reported outcomes [27]. Survival and other time-to-event (such as time to death or permanent assisted ventilation) outcomes are standard and discussed elsewhere [28, 29]. Here, we review the most used non-survival outcome measures in ALS clinical trials.

The ALS Functional Rating Scale-Revised (ALSFRS-R) is a 12-point ordinal scale measuring domains of bulbar, upper limb, lower limb, and respiratory function. Scores range from 0 to 48, with lower scores indicating greater functional impairment [30]. The average rate of decline in ALSFRS-R is 1 point per month [31], with more negative slopes correlating with poorer survival [32, 33]. Among experts, a clinically meaningful change in ALSFRS-R in clinical trials would be a reduction of about 20% in the slope of rate of decline [34]. Ordinal scales, such as ALSFRS-R, where total sum scores are compared within participants, sometimes under- or over-value changes (i.e., the difference in scores in patients who go from walking up stairs to using a handrail is considered equivalent to the difference between feeding oneself and relying on others for all feeds). Thus, the Rasch-based method for developing weighted scales was applied to questions related to activities of daily living in patients with ALS to create the 28-question Rasch-built Overall ALS Disability Scale (ROADS) [35, 36]. Nonetheless, the selection of the optimal scale for use in ALS research remains of considerable debate and was the topic of a recent global summit, as the ALSFRS-R exhibits multidimensionality and revision is suggested [3739].

Measures of respiratory function are important markers of disease progression and clinical trial outcomes. Forced vital capacity (FVC), the vital capacity when exhalation is performed with a rapid maximum exhalation, is the most common respiratory measurement in ALS trials and correlates with survival [40, 41]. Worsening rates of decline of predicted vital capacity over time increases the risk of mortality up to 17% [40]. Other measures, such as slow vital capacity (SVC), where the patient exhales slowly into a spirometer, and mean inspiratory pressure, where the patient inhales against an occluded airway, have also been used [42, 43]. SVC is sometimes favored because it is more accurate for patients with concurrent obstructive lung pathology [44]. Unfortunately, most measurements of pulmonary function can be affected by cognition, or more commonly, orofacial weakness, impairing the ability to create a seal with the respiratory measurement interface [45].

Objective measures of strength are clear target outcomes in a disease of progressive weakness. Handheld dynamometry (HHD) has been studied and consists of the evaluator applying confrontational resistance to prespecified muscle groups, measuring outputs against healthy population Z-scores and combining Z-scores into cumulative scores for an extremity. This can be monitored over time more reliably than manual muscle testing [46]. In ALS populations, change in HHD scores was correlated with ALSFRS-R and vital capacity (VC), with coefficient of variation performing better than VC, but not as well as ALSFRS-R [47]. A tool for isometric measurement of limb strength, Accurate Test of Limb Isometric Strength (ATLIS), was developed to eliminate user variability with handheld dynamometers and was found to be more sensitive to change than ALSFRS-R and VC [48]; however, training and accessibility to the ATLIS device has limited widespread use.

The FDA encourages outcomes that combine both survival and function as efforts to identify meaningful interventions persists [27]. The most commonly used combined outcome is the Combined Assessment of Function and Survival (CAFS), which is a combined, nonparametric test assessing change of ALSFRS-R scores at multiple timepoints while accounting for survival, patient drop-out (censorship), or other prespecified “time-to-event” endpoints, similar to a rank sum model [49, 50]. Though useful for accounting for study endpoint events in a rapidly fatal disease (i.e., death versus study completion versus trial withdrawal), combined outcomes suffer from issues with cross-trial validity and weighting the importance of relative contributions to outcome (death versus disease progression versus loss of follow-up) in creating the summed rank model [51]. More recently, other techniques used to combine function and survival include progression-free survival [52] and Bayesian shared parameter analysis [53].

Influenced by the 21st Century Cures Act [54], patient-reported outcome (PRO) measures are playing a more prominent role in clinical trials. Though the number of questionnaires and PROs used across ALS trials is too extensive to discuss here, we highlight a few of the most frequently used scales often included as secondary or exploratory outcomes in recent clinical trials. The Patient-Reported Outcome Measurement Information System (PROMIS-10), which measures a combination of physical, mental, and social determinants of health [55], as well as the NeuroQoL-fatigue, an 8–9 question survey about fatigue [56], correlate with ALSFRS-R scores [57] and are being incorporated more consistently across neurology trials, but are not specific to ALS. The ALS Specific Quality of Life Scale (ALSSQOL) and the ALSSQOL-Short Form (ALSSQOL-SF) were developed to measure quality of life beyond limitations in physical function and instead measure across six domains: negative emotion, interaction with people and the environment, intimacy, religiosity, physical symptoms, and bulbar function. The ALSSQOL is validated in patients with ALS and correlates with ALSFRS-R scores, as well as other measures of quality of life [58, 59]. The 40-item ALS Assessment Questionnaire (ALSAQ-40) has also been used frequently in trials, measuring five domains (eating and drinking, communication, activities of daily living/independence, physical mobility, and emotional reactions) with strong internal reliability, construct validity, and correlations with ALSFRS-R [60]. Lastly, the Center for Neurologic Study Bulbar Function Scale (CNS-BFS) is a 21-question self-administered questionnaire regarding bulbar function, including specific speech, saliva, and swallowing domains that correlates well with clinical findings and other objective measures of speech and swallow [61]. Additional details on qualitative outcomes in ALS can be found elsewhere [62].

ALS clinical trials have also incorporated advanced electrophysiologic studies as exploratory outcomes measures for quantifying lower and upper motor neuron function, although they are limited by generalizability and interrater reliability [63, 64]. For lower motor neuron function, motor unit number estimation (MUNE) and motor unit number index (MUNIX) are strategies that attempt to estimate the number of remaining motor units or lower motor neurons. Both are based on the ratio of compound muscle action potential to an average single motor unit to establish a global index, relating single motor unit function relative to muscle activation, findings of which correlate with disease progression [63, 6567]. MUNIX is generally more reproducible than MUNE, correlates with decreasing ALSFRS-R scores better than standard surface compound muscle action potential (CMAP) measurements, and performs with exceptionally high specificity [68]. Electrical impedance myography (EIM) is a noninvasive measure of muscle via low-intensity current to detect changes in impedance over time [69]. In essence, EIM quantifies the “health” of the muscle tissue and correlates with survival in ALS as well as ALSFRS-R and HHD [70]. Given the difficulty in quantifying upper motor neuron dysfunction on clinical exam alone, some studies have used transcranial magnetic stimulation (TMS) to indirectly measure cortical hyperexcitability as a surrogate for pyramidal cell dysfunction. Though techniques vary, threshold tracking TMS has demonstrated abnormal cortical hyperexcitability as an early feature of sporadic ALS [71]. Other TMS measures, such as short-interval intracortical inhibition (SICI) and resting motor threshold (RMT), have been used in trials with variable success, though issues with reproducibility, reliability, and inconsistency in specific measurement protocols have limited the utility of these in further studies [72, 73]. Detailed review of MUNIX, TMS, and other advanced neurophysiologic studies, such as peripheral threshold tracking nerve conduction studies to measure spinal motor neuron excitability, are reviewed elsewhere [7477].

One of the fundamental principles of therapeutic drug development is confirmation of end-target engagement, measured most readily and ideally via serum biomarkers. Though there is not a single specific serum or plasma biomarker for ALS, higher levels of serum neurofilament light chain (NfL), a neuron-specific marker of axonal degeneration, have been found to differentiate ALS from disease mimics, such as inflammatory demyelinating polyneuropathy or structural polyradiculopathy [78], and correlates with rate of progression in ALS as well as disease severity [79]. NfL is emerging as an important biomarker in ALS clinical trials [80] and has been considered by the FDA in approval decisions as an outcome to measure the effect of an intervention [81]. Further details on NfL in ALS, as well as other promising biomarkers, including markers of oxidative stress and alterations in stress and energy metabolism, are reviewed separately [8285].

Clinical trial design in ALS continues to advance, allowing for better measurement of changes in outcomes via patient stratification, identification of patients most likely to benefit from specific drug mechanisms, and methods for faster drug development and availability to patients. Further details on trial design development in ALS are referenced [53, 8689].

Current ALS Treatment Approaches

The overall treatment approach to the patient with ALS incorporates disease-slowing therapeutics, symptomatic management, and respiratory and nutritional support. Given the absence of disease-modifying therapies that meaningfully slow disease progression, supportive care remains the mainstay of treatment for patients with ALS. Multidisciplinary clinics substantially improve survival and quality of life in patients with ALS and are the “gold standard” of care [90, 91]. This survival benefit likely results from increased use of noninvasive ventilation and feeding tube placement [92], although it should be noted that a recent review did show both negative and positive outcomes with respect to feeding tubes, indicating future research is needed [93]. Though patients often report good quality of life, symptoms such as fatigue, pain, pseudobulbar affect, depression, anxiety, thick secretions, and sialorrhea are reported frequently and contribute to worse quality of life [9496]. Though there is not a standardized approach to multidisciplinary care of patients with ALS, expert consensus statements and society guidelines such as those from the European Academy of Neurology [97], United Kingdom National Institute for Health and Care Excellence (NICE), American Academy of Neurology [98, 99], and Japanese Society of Neurology [100], as well as national guidelines from Canada [100] and Iran [101], provide valuable frameworks for care.

Disease-Modifying Treatments

Only a few medications are approved globally for the treatment of ALS (Fig. 1). A combination of PubMed searches and guideline reviews were used to generate lists of these treatments. A PubMed literature search used keywords, such as “amyotrophic lateral sclerosis” or “ALS,” as well as combinations of words using the controlled vocabulary thesaurus feature, Medical Subject Headings (MeSH). The final PubMed search was completed in February 2025. International society guidelines published prior to this date were reviewed from the American Academy of Neurology (AAN), European Academy of Neurology (EAN), and Japanese Society of Neurology, as well as national guidelines from Canada and Iran. Specific note is made of therapies approved internationally but not currently used in the USA. The primary focus of this review is on current FDA-approved therapies; however, given patient interest in supplements, data on safety and efficacy of supplements were obtained from the Table of Evidence provided by the international research consortium of ALS Untangled [102]. ALS Untangled is a body of international ALS experts who rigorously review alternative therapies and off-label treatments to provide objective and informative information for patients and providers [103]. Filters for “grade A” mechanism, indicating peer-reviewed publications to target relevant mechanisms in humans, and “grade A” trial or case data, indicated by two or more peer-reviewed publications describing benefit in well-designed placebo-controlled human trials, or one or more peer-reviewed publications reporting benefits in validated diagnoses were applied for inclusion in our review. References were thoroughly reviewed for quality and additional primary data. This criteria identified methylcobalamin, acetyl-L-carnitine, and vitamin E, though upon review of the literature, randomized trials of vitamin E in high and “regular” dosing did not meet primary endpoints [104106], and is associated with increased risk of hemorrhage [107] and slight increased risk of prostate cancer [108], and is not recommended by consensus guidelines [98]. It is thus not reviewed in further detail here.

Fig. 1.

Fig. 1

Mechanisms linked to current ALS therapeutics. Sites of action for medications and selected supplements used for the treatment of ALS. Tofersen is limited to patients with SOD1 mutations. ALCAR: acetyl-L-Carnitine; ATP: adenosine triphosphate; EEAT: Excitatory amino acid transporters; HCY: homocysteine; HSP: heat shock protein; MethylCo: methylcobalamin; mRNA: messenger RNA; Mt SOD1: mutant superoxide dismutase 1; ROS: reactive oxidative species; SAM: S-adenosylmethionine. Created in BioRender.

Source: Goutman, S. (2025) https://BioRender.com/h74e601

Riluzole

Riluzole (2-amino-6-(trifluoromethoxy)benzothiazole) was developed to modulate glutaminergic transmission to target excess of the excitatory neurotransmitter, glutamate, at synapses in the central nervous system [109]. The mechanism by which it does this is not fully understood [110], though proposed mechanisms include: (1) antagonism of the persistent sodium (Na+) current, thereby reducing hyperexcitability; (2) blockage of voltage gated calcium cannels to prevent glutamine release [111]; and (3) inhibition of inhibitory gamma-aminobutyric acid (GABA) reuptake [112]. A randomized, double-blind, placebo-controlled, stratified trial of riluzole 100 mg daily, given as 50 mg twice daily, demonstrated a statistically significant survival benefit at 12 and 21 months, with median survival increasing by 83 days [113], with more robust survival effect seen in patients with bulbar onset disease. Primary indications for withdrawal of treatment were asthenia, worsening spasticity, and increases in serum aminotransferase levels and blood pressure. Riluzole (Rilutek) was approved for marketing by the FDA in 1995 [114] and is readily available worldwide [115]. A 2012 Cochrane Review of pooled data from three trials examining tracheostomy-free survival in 974 patients treated with riluzole and 503 placebo-treated patients demonstrated a statistically significant increase in survival from 11.8 months to 14.8 months, but without any benefit on measures of muscle strength. Hazard ratios decreased while using riluzole for the first 6 months of assessment but increased when measured out to 18 months, at which point the survival benefit was no longer statistically significant [116]. Included in the Cochrane Review was a trial evaluating riluzole in patients > 75 years old with disease duration longer than 5 years and vital capacity < 60% of predicted normal, but could not recruit enough to meet their prespecified power requirements and could not comment on survival, though the medication was well tolerated in this population [117].

“Real-world” riluzole data complements clinical trial data, as retrospective studies and ALS registry data also support a riluzole survival benefit ranging from 4 months [118] to 21 months [119, 120]. Some studies suggest that earlier initiation of riluzole (within 1 year of symptom onset) can provide an additional survival benefit of up to 1.9 months [121], especially in patients with an older age at disease onset [122]. Other studies suggest riluzole prolongs time at milder disease stages [123]; however, others indicate greater benefit of riluzole later in disease, except for bulbar-onset patients, when adjusted for age and slope of ALSFRS-R decline at time of diagnosis [124]. Meta-analyses and population-based studies suggest a small benefit in bulbar and limb function, but not muscle strength [116], and contradictory to other data, greater benefit in those with bulbar onset compared with limb onset [122].

Dysphagia is an expected symptom in the progression of ALS and crushing riluzole tablets or ingesting riluzole with high-fat foods results in loss of efficacy [125]. An oral suspension was developed to obviate the need for crushing and mixing with foods, such as yogurts and puddings, and was specifically developed to avoid the side effect of mouth anesthesia that is reported with oral riluzole tablets [126]. The FDA approved the liquid version (marketed as Tiglutik [127]) in 2018 and an oral film (marketed as Exservan [128]) in 2019, though the latter was taken off the market in fall 2024 [129].

Tofersen

Tofersen was developed as an antisense oligonucleotide (ASO) targeting SOD1 mRNA to reduce synthesis of SOD1 proteins in patients with ALS resulting from mutations encoding this gene. SOD1 mutations classically have accounted for about 12% of familial ALS in European countries [7], though recent data suggest this may be an underestimate and that SOD1 mutations are also common in sporadic ALS [130]. The phase 1–2 trial of tofersen [131] demonstrated target efficacy, as measured by reduction of SOD1 in cerebrospinal fluid (CSF), without treatment-related serious adverse events (SAEs) or participant discontinuation in the trial. The phase 3 study was constructed in a three-part sequence, first comparing eight intrathecal doses of 100 mg tofersen versus placebo over 28 weeks, then proceeding into a 52-week open label extension with ongoing data collection planned through 236 weeks [132]. A total of 108 participants were enrolled, with 72 participants assigned to receive tofersen. Endpoints were designed specifically to assess changes among a prespecified “faster progression group,” though others were not excluded from the trial. Final trial results were reported for 60 of the 108 enrolled participants who met criteria for the faster progression group. Faster progression was defined by SOD1 genotype and rate of decline on ALSFRS-R calculated using the deltaFS method [33]. The primary outcome in the placebo-controlled period was change in ALSFRS-R from baseline to week 28 based on a joint rank test, which allows for measurement of treatment effects while accounting for deaths during the trial, though interestingly, there were not many deaths during either the 28- or 52-week period. The primary endpoint for the 52-week open label portion was change in neurofilament light chain (NfL). This portion of the study was not powered to assess clinical outcomes. The placebo-controlled portion of the trial did not meet its primary endpoint, but there was a statistically significant decline in CSF measurement of SOD1 and NfL at week 28 but not at week 52. There was a trend toward reduction in clinical progression at week 40 and beyond in the open-label extension. Serious neurologic adverse events occurred in 6% and 7% of the tofersen trial group and open-label group, respectively, in the form of myelitis, chemical or aseptic meningitis, lumbar radiculopathy, and papilledema, each requiring discontinuation of treatment. The mechanism of myelitis is unclear. Otherwise, headache and procedural pain from the lumbar puncture were the most common adverse events. Tofersen (marketed as Qalsody) was approved under the FDA’s accelerated approval pathway on the basis of the target engagement of reduced NfL in April 2023 [81]. It is also approved in the European Union, Norway, China, Japan, and the UAE, among others [133, 134].

Edaravone

Edaravone (MCI-186) is a free-radical scavenger, originally approved for stroke [135], developed to reduce oxidative stress in and around ischemic tissue in motor neurons. In the ischemic stroke literature, the use of intravenous (IV) edaravone is associated with rash, abnormal liver and abnormal renal function [136], fever, and dizziness; although pooled meta-analyses have not found significant differences as compared with placebo in the acute stroke population [137]. A phase 2 trial of edaravone in patients with ALS demonstrated tolerability of 60 mg, with the most common side effect reported as soft stools and diarrhea [138]. The study found a significant decline in 3-nitrotyrosine, a marker of oxidative stress, but not other inflammatory markers, such as coenzyme Q10, 8-hydroxydeoxyguanosine, or 4-hyroxy-2,3,nonenal [138]. Two subsequent phase 3 trials were completed, which did not meet their endpoints, but post hoc data revealed that a specific subset of the early stage patients may have had a greater response [139]. This led to the MCI186-19 trial, which enrolled patients with early stage disease, defined as symptom onset within 2 years and score of at least 2 out of 4 on each question of the ALSFRS-R, slow progression, defined as < 4 point decline over a 12-week observation period, and forced vital capacity (FVC) > 80%. This study compared 60 mg IV edaravone for 6 cycles (24 weeks) with placebo in 137 patients with ALS and successfully met the primary endpoint of adjusted mean difference of ALSFRS-R [140]. Biomarkers were not part of the analysis in MCI186-19 trial. The study allowed for an open-label extension, and post hoc data of patients who had FVC < 80% during the open-label extension demonstrated an overall reduction in ALSFRS-R decline from baseline to week 48. However, a propensity matched study of 194 patients with motor neuron disease in Germany failed to demonstrate any difference in disease progression as compared with riluzole alone over 1 year [138].

IV formulations are felt to add undue burden on patients with ALS, and thus an oral bioequivalent liquid form of edaravone was developed [141]. In an early phase study of oral edaravone in ALS, the drug demonstrated predictable kinetics, though with variable metabolism of drug metabolites, and was tolerated orally and via gastrostomy tube [142]. An open-label, phase 3 study of oral edaravone over 48 weeks with dosing mimicking the cycle dosing of IV edaravone was completed in 2020, with fatigue, dizziness, headache, nausea, and constipation the most frequently reported treatment-related adverse event [143]. There were no changes in serum lab monitoring or vital signs. The FDA approved oral edaravone in cycle dosing in May 2022 [144]. A phase 3 trial of oral daily dosing of edaravone failed to demonstrate superiority to cycle dosing of oral edaravone (NCT04569084) [145] but full trial results have not yet been published. Similarly, a phase 3 randomized trial of once daily oral edaravone did not demonstrate benefit as measured by change in ALSFRS-R and CAFS, though full results have not been published (NCT05178810) [146].

Sodium Phenylbutyrate-Taurursodiol (AMX0035)

The combination of sodium phenylbutyrate and tauruorsodeoxycholic acid (TUDCA) had brief approval following an encouraging phase 2 study but was removed from the market following negative phase 3 results. Sodium phenylbutyrate is a histone deacetylase inhibitor, acting on histone deacetylases, which inhibit transcription leading to disruption of cellular function and neuronal death. This is thought to be a contributing mechanism of pathogenesis in numerous neurodegenerative disorders, including ALS. Data in Alzheimer’s disease had suggested an anti-inflammatory effect, including reduction of glial activation, using TUDCA [147]. A phase 2 tolerability and dose-finding study of sodium phenylbutyrate alone in ALS demonstrated biomarker engagement and overall tolerability at a dose of 9 g/day [148]. The combination of sodium phenylbutyrate and TUDCA had been shown to reduce neuronal death in experimental models of ALS [149], thus leading to a phase 2 randomized, double-blind, placebo controlled trial in 137 patients of the combination drug, which demonstrated a statistically significant difference of 0.42 points per month decline in ALSFRS-R between treatment and placebo groups (−1.24 and −1.66 points per month, respectively) [149]. The most common side effects requiring discontinuation or dose adjustments were gastrointestinal side effects, primarily diarrhea. In a controversial decision [150]), the FDA approved AMX0035 for treatment of ALS, with the condition that it would be removed if an ongoing phase 3 trial, due to complete in 2024, was negative [151]. Two post hoc analyses of the long-term trial data showed increased overall median survival in the treatment group (25.0 months versus 18.5 months) [152] and reduced risk of death, tracheostomy, permanent assisted ventilation, and hospitalization [153]. Despite optimism from the phase 2 trial, the phase 3 trial did not meet primary or secondary endpoints and thus was voluntarily removed from the market in April 2024 [154]. Full study results have not yet been published as of March 2025.

Methylcobalamin

Methylcobalamin is the active form of cobalamin, or vitamin B12, and reduces oxidative stress [155], glutamate toxicity [156], and apoptosis, and lowers plasma homocysteine levels [156], all of which are demonstrated pathologies in patients with ALS. Given relatively low risk of administration, it has long been used by patients with ALS but has lacked convincing trial evidence to support disease-modifying effects [157]. A phase 2/3 study of ultra-high dose methylcobalamin failed to meet primary survival and functional outcomes [158]. Driven by a subgroup of these results, a phase 3 trial of ultrahigh-dose methylcobalamin was repeated in Japanese patients with symptom onset within 1 year of enrollment, slow decline in ALSFRS-R pretreatment, and limited respiratory involvement (FVC > 60%, no history of NIV or mechanical ventilation, and ambulatory). Results demonstrated a statistically significant difference in decline in ALSFRS-R over 16 weeks [159]. Methylcobalamin was approved in Japan for the treatment of ALS, marketed under the brand name Rozebalamin in 2024 [160]. The long-term open label extension of the Japanese Early-Stage Trial of High Dose Methylcobalamin for Amyotrophic Lateral Sclerosis (JETALS) completed enrollment in March 2025 (NCT03548311), with the medication showing good tolerability at an interim analysis performed in early 2024 [161].

Lenzumestrocel

Lenzumestrocel (marketed as Neuronata-R) is an intrathecal autologous bone-marrow-derived mesenchymal stem cell (MSC) therapy currently approved in South Korea for the treatment of ALS. Early preclinical data showed a prolonged life span and delay in decline in motor performance with preservation of motor neurons in the lumbar spinal cord of SOD1 mouse models [162]. In a phase 1 study, two monthly injections of intrathecal MSC were given to seven patients with ALS. There were no treatment-related serious adverse events, and all adverse events were transient in nature, such as flu-like illnesses, fevers, headache, and back pain. At 6 months, the rate of decline in ALSFRS-R was less than what had been measured in a 3-month lead in period, though this was not statistically significant [163]. The phase 2 randomized, parallel trial similarly followed the protocol of a 3-month lead-in period, followed by two intrathecal injections of MSCs in consecutive months followed by 6 months of clinical outcomes and follow-up. The control group did not undergo sham lumbar punctures or injection, but outcome evaluators were blinded to treatment assignments. A total of 33 patients underwent treatment. Adverse events were nearly identical to the phase 1 study and there were no treatment-related SAEs reported. There was a statistically significant difference in the mean difference in ALSFRS-R scores between groups at both 4 months and 6 months after injection. The mean difference in the slope of decline between lead-in and 6 months after injection was also significantly lower in the treated group. There was no change between groups in FVC or slope of FVC decline [164]. The results from these studies led to conditional approval of the drug in South Korea by the Korean Ministry of Food and Drug Safety (KMFDS) in 2013. Post-marketing surveillance data were published from 170 patients who underwent treatment with lenzumestrocel, with symptom duration at time of injection < 2 years and documented follow-up, though only 157 were included in the final survival analysis. Most patients received two injections (one cycle), but 21 underwent repeated cycle dosing. Treated patients were propensity score matched to the external controls from the PRO-ACT database. Median survival probability was significantly higher in treated patients (single and multiple cycles) compared with controls. In total, 17% of patients experienced adverse drug reactions, again mostly fever, headache, pain, back pain, and nausea, though 5% had persistent pain and allodynia lasting greater than 2 weeks [165]. The FDA and KMFDS approved a phase III, multicenter, randomized, double-blind, sham-procedure-controlled trial, conducted exclusively in South Korea [166] NCT 04745299, KCT0005954, which did not meet primarily efficacy outcomes [167], though long-term follow-up and safety reporting continues through 2026. Detailed results of the primary analysis have not been published.

Disease-Modifying Treatments Pending Regulatory Approval

Three additional agents, masitinib, co-formulated ciprofloxacin and celecoxib (marketed as PrimeC), and debamestrocel-MSC-NTF (marketed as NurOwn), are notable due to requests for regulatory opinions [168170]. Masitinib is an oral tyrosine kinase inhibitor targeting central and peripheral inflammation [171]. Co-formulated ciprofloxacin and celecoxib is a combination of ciprofloxacin and celecoxib, thought to act synergistically to regulate inappropriate microRNA processing and reduce inflammation [172]. NurOwn is an autologous bone-marrow-derived mesenchymal stem cell designed to induce neurotrophic factors [173]. Originally, the FDA did not support the biologics license application submission for NeurOwn [174]; however, it has provided clearance for initiation of a phase 3b trial (NCT06973629) estimated to start in June 2025.

Use of Supplements in Management of ALS

Acetyl-L-Carnitine (ALCAR)

Carnitine is a human amino acid that contributes to mitochondrial fatty acid oxidation. There are multiple proposed mechanisms for its use in ALS, including mitigating oxidative stress by up-regulating nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione, and heme oxygenase 1 (HO-1) [175], promoting mitochondrial function by increasing serum levels of ATP [176], and protecting against glutamate excitotoxicity [177]. It has been demonstrated that baseline fatty acid peroxidation markers are elevated in patients with ALS, whereas antioxidant markers are lower. This finding was reversed after 6 months of treatment with acetyl-L-carnitine, though the same pattern was not seen in nitric oxide measurements [178]. A phase 2 randomized, placebo-controlled trial compared acetyl-L-carnitine with placebo over 48 weeks and did not find a significant difference in primary endpoint of time to non-self-sufficiency [179]. Unfortunately, the study had numerous pitfalls, including protocol deviations during randomization and slow recruitment. Post hoc analysis did show a trend toward prolonged time to non-self-sufficiency but was not statistically significant. A more recent retrospective, case-controlled study found a survival benefit in patients with ALS taking 1.5 g/day of acetyl-L-carnitine, but not for patients taking 3 g/day. Functional outcomes were not significantly different from controls [180]. A phase 2/3 multicenter randomized trial of ALCAR versus placebo is expected to begin enrollment in 2025 (NCT06126315).

Symptomatic Treatment

Symptomatic management of ALS is critical and many of the medications utilized for symptomatic management (Fig. 2) are based on expert guidelines, as randomized clinical trials are lacking [181]. Medications consistently recommended in consensus guidelines—along with their mechanisms of action, important safety monitoring parameters, common adverse effects, and available clinical trial data—are listed in Table 1. In-depth reviews for ALS symptomatic management are published [182, 183].

Fig. 2.

Fig. 2

Therapeutics for the management of ALS symptoms. The mainstay of ALS clinical management is addressing and controlling symptoms. While non-pharmacologic and secondary sources of symptoms should be evaluated at every visit, pharmacologic options are frequently utilized. A range of medications are available to target the diverse symptoms experienced by patients with ALS. Created in BioRender.

Source: Goutman, S. (2025) https://BioRender.com/w04b047

Table 1.

Selected medications used in the treatment of symptoms in ALS

Symptom Medication Mechanism of action Other uses Common side effects Monitoring Special considerations
Sleep/fatigue Modafinil1 Thought to block dopamine transporters, increasing the amount of dopamine in the brain

-Headache

-Nausea

-Gastrointestinal distress

-Regularly monitor heart rate, blood pressure, and weight

-Can exacerbate anxiety and depression

-Increased cardiac monitoring in patients with unstable angina

-Fatigue is a common side effect of riluzole

SNRI* (duloxetine, venlafaxine) Inhibits neuronal serotonin and norepinephrine reuptake

Depression/anxiety

Neuropathic pain

-Nausea

-Headache

-Dose-related drowsiness

-Constipation

-Abnormal dreams

-Blood pressure

-Baseline kidney and liver function

-Can precipitate manic attacks if used as monotherapy in patients with bipolar disorder

-Serotonin syndrome can occur if used with multiple other serotonergic medications

Melatonin* Melatonin agonist at the suprachiasmatic nucleus to reduced evening arousal and promote sleep onset

-Vivid dreams and nightmares

-Dizziness

-Irritability

-Stomach cramps

-Daytime sleepiness may indicate need for dose adjustment or discontinuation -Dietary supplement, thus quantities and properties may vary between formulations
Depression/anxiety Tricyclic antidepressants* (amitriptyline, nortriptyline, imipramine) Prevents reuptake of serotonin and norepinephrine

Neuropathic pain

Pseudobulbar affect

Sialorrhea (amitriptyline)

Insomnia

-Anticholinergic effects (constipation, dry mouth, urinary retention, tachycardia)

-Sedation

-Weight gain

-Cardiac rhythm abnormalities at high doses and those at risk of cardiac arrhythmia

-Heart rate

-Serum sodium in those with poor oral intake

-Side effects can be exacerbated and can increase risk of falls in older adults

-Nortriptyline is thought to have a more modest anticholinergic effect than amitriptyline

Benzodiazepine* Increases GABA-ergic post-synaptic membrane permeability to chloride, enhancing inhibitory GABA effects

Short-acting or end-of-life anxiety

Refractory insomnia

Refractory spasticity

-Sedation

-Dizziness

-Respiratory depression

-Respiratory status

-Heart rate, blood pressure

-Mental status

-Risk of paradoxical reaction in those with preexisting aggression or alcoholism

-Consider implementing benzodiazepines as first-line therapy in patients with advanced disease in whom non-pharmacologic options for therapy are not possible

Gabapentinoids* (gabapentin, pregabalin) Structurally related to GABA, but work on pre-synaptic voltage gated calcium channels to inhibit excitatory neurotransmitter release

Pain

Cramps

Spasticity

-Dizziness

-Drowsiness

-Ataxia

-Fatigue

-Peripheral edema

-Renal function

-Sedation

-Respiratory function

-Initiate at the lowest dose for those with respiratory involvement
Appetite stimulation

Cannabinoids2

(dronabinol [THC])

Acts on CB1 and CB2 receptors in the brain

Pain

Insomnia

Spasticity

-Dizziness

-Drowsiness

-Nausea/vomiting

-Paranoia/changes in thinking

-Euphoria

-Heart rate, blood pressure

-Mood

-May exacerbate depression

-May cause syncope with dose initiation

-Cost may be prohibitive to patients

-Nonregulated forms of THC face legal, safety, and dose-reliability issues

Megestrol acetate* Synthetic progestin with antiestrogenic properties and thought to antagonize metabolic effect of catabolic cytokines

-Dose and duration-dependent hypothalamic-pituitary axis suppression

-Rash

-Impotence/decreased libido

-Flatulence

-Insomnia

-Serum glucose

-Signs of adrenal insufficiency with chronic use

-Blood pressure

-May increase risk of venous thromboembolism

CB1, cannabinoids 1; CB2, cannabinoids 2; GABA, gamma-aminobutyric acid; THC, tetrahydrocannabinol

*Expert/consensus recommendations

1Modafanil was well tolerated in doses up to 300 mg/day and demonstrated improvement in patient-reported measures of fatigue after 8 weeks of use in patients with ALS as compared with placebo [326]

2Moderate evidence supports the use of cannabinoids as an appetite stimulant [327, 328] but it has not been specifically studied in ALS, thus guidelines are variable in recommendations for use in this patient population

Pseudobulbar Affect

Pseudobulbar affect (PBA) represents unintentional or uncontrollable laughing and crying related to impaired corticobulbar function and is a common symptom in ALS. Currently there is one medication, dextromethorphan/quinidine (DMQ) (marketed as Nuedexta), specifically FDA-approved for PBA. Dextromethorphan is a weak non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist resulting in glutamate inhibition and protected from first-pass metabolism by its co-drug, quinidine, which was shown to be well tolerated in patients with ALS [184]. Patient-reported episodes of PBA were reduced in patients taking DMQ compared with quinidine alone with significant improvement in quality-of-life measures over 29 days. The most common side effects were nausea, dizziness, and loose stools. There was a significant difference in QT interval prolongation on electrocardiogram in those receiving DMQ; however, it was not judged to be clinically relevant. Similar findings were reproduced over 12 weeks in a combined study of DMQ for ALS and multiple sclerosis [185]. A small, phase 2 crossover trial of DMQ versus placebo found significantly improved bulbar function as measured by the patient-reported CNS-BFS, as well as the bulbar subdomain of the ALSFRS-R [186]; however, these findings have not been replicated in larger cohorts and DMQ is not currently FDA-approved as a disease-modifying therapy in ALS. Other considerations based on expert guidelines include selective serotonin reuptake inhibitors (SSRI) or tricyclic antidepressants (TCA) [97]. Treatment of PBA should always include an evaluation for underlying mood disorders, such as depression, which may be contributing to emotionality.

Disorders of Saliva (Sialorrhea and Thick Mucus)

Treatment for sialorrhea (excessive saliva) includes anticholinergics, such as amitriptyline, glycopyrrolate, atropine, or scopolamine, with the latter two providing non-tablet administration options [97, 187]. As detailed above, there is patient-reported improvement in overall bulbar function with DMQ [186], and thus, on the basis of EAN guidelines, DMQ is considered for treatment of sialorrhea in those patients with concurrent pseudobulbar affect [97]. A recent Cochrane review identified low-certainty and moderate-certainty evidence for the use of botulinum toxin B injections for patients with “refractory sialorrhea” or “bulbar dysfunction” [187].

The aforementioned medications are helpful for thin, pooled secretions and drooling, but may exacerbate thicker mucus that can arise from poor cough or sinus drainage. Treatment of thick secretions often includes non-pharmacologic therapies, such as insufflation–exsufflation devices or saline nebulizers; however, mucolytics (acetylcysteine, guaifenesin) or beta blockers [188] can be used if these are insufficient.

Carbonated thin liquids reduce penetration and aspiration in patients with neurogenic dysphagia [189], and sour taste has been shown to increase swallowing frequency and prolong cortical response to the initiation of swallow [190]. A variety of foods have been tested to promote intake in neurogenic dysphagia [191], though rigorous trial data are lacking. Anecdotally, grapefruit juice helps relieve symptoms of excessive saliva, whereas papaya and pineapple juice are more helpful for thicker secretions.

Cramps

Up to 95% of patients with ALS experience muscle cramps [192]. A phase 2 trial of mexiletine, a sodium channel blocker, demonstrated dose-dependent reduction in muscle cramps, though 300 mg doses were associated with fewer adverse events. Other medications with effects on sodium channels, such as gabapentinoids and baclofen, are also commonly used, though there is inconsistent evidence on the efficacy of gabapentin for cramps in ALS [181]. The antimalarial medication quinine has long been used for cramp relief, but after increasing reports of side effects, most severely disseminated intravascular coagulopathy [193], the FDA limited and provided guidelines for the amount of quinine readily available in common formulations [194].

Spasticity

Spasticity related to upper motor neuron dysfunction can be treated with classical muscle relaxants, such as baclofen, tizanidine, and cyclobenzaprine. Surveys of patients with ALS report reduction in spasticity with use of cannabis [195]. Focal injections of botulinum toxin are frequently used as well. For intractable spasticity, intrathecal baclofen pumps can be considered [196].

Preclinical Drug Development Disease Models

The early discovery of SOD1 genetic ALS led to the development of SOD1 transgenic mouse models. These mice display motor neuron degeneration and resultant muscle atrophy and weakness similar to human ALS phenotypes. As such, they continue to be the foundation for the study of potential ALS therapeutics [19]. Alternative models for drug development are used less frequently. In silico screens utilize computational biology to predict interactions between drug compounds and target proteins. In vitro drug screens are performed on either cell-free protein aggregates, animal-derived cell lines, or patient-derived motor neurons [7, 197]. Finally, in addition to murine models, in vivo drug screens are performed with transgenic Caenorhabditis elegans, Drosophila melanogaster, and zebrafish (please see reference for review of animal models used in ALS drug discovery [198]).

Investigational Agents in Clinical Trials

A large number of investigational agents are currently in various phases of clinical trials for the treatment of ALS. These investigational agents target a range of mechanisms implicated in ALS. The below is not intended as a comprehensive review of ALS pathophysiology, but instead to highlight mechanisms targeted by drugs currently in phase 1, 2, and 3 studies.

ALS Mechanisms Targeted by Current Investigational Agents

To identify current ALS investigational agents, we accessed the ClinicalTrials.gov database on 1 October 2024. We searched for “amyotrophic lateral sclerosis” but excluded other motor neuron disorders including primary lateral sclerosis, spinal muscular atrophy, and spinal bulbar muscular atrophy. To focus on contemporaneous studies, trials were further filtered for interventional studies with a posted date from 1 January 2020 through 1 October 2024. We restricted the analysis to phase 1, 2, and 3 studies. A total of 210 results were independently reviewed and 150 of these were excluded on the basis of author consensus to scope the review around active trials (Fig. 3). This identified 60 active or recently completed drug trials included in the final review. Additional investigational agents were included from independent sources known to the authors.

Fig. 3.

Fig. 3

Screening criteria for ALS clinical trials included in final review. ClinicalTrials.gov search strategy to identify current investigational therapeutics in phase 1, 2, and 3 trials for ALS; 211 trials were evaluated and ultimately 60 trials were included in the final review

Broad categories of drugs are under investigation as new ALS therapeutics. These include novel small molecules, repurposed drugs, biologics (including antibodies and immune cells), gene therapies, and stem cells. Further, these investigational agents are targeting a range of pathological mechanisms including protein homeostasis and neuroinflammation. Other targeted ALS pathways include neurotrophic factors, oxidative stress, neuromuscular junction and axonal integrity, mitochondrial function and energy expenditure, RNA metabolism, and the gut–brain axis (Fig. 4).

Fig. 4.

Fig. 4

ALS therapeutic targets of drugs in phase 1, 2, and 3 clinical trials. ALS drugs in clinical trials currently target multiple pathophysiologic mechanisms implicated in ALS pathogenesis. These mechanisms include impaired proteostasis and integrated stress response within motor neurons; neuroinflammation mediated by microglia, astrocytes, and blood–brain barrier infiltration of peripheral immune cells, such as regulatory T cells (Tregs); oxidative stress mediated by reactive oxygen species (ROS); impaired neuromuscular junction and axonal transport; mitochondrial dysfunction; alterations in RNA metabolism; excitotoxicity due to glutamate-mediated calcium influx into motor neurons; and changes to the gut microbiome. Additionally, use of stem cells and isolated neurotrophic factors are used to stimulate motor neuron growth and survival. Gene-specific therapies, such as antisense oligonucleotides, are also employed to target various ALS pathophysiologic mechanisms, particularly in genetic ALS. Created in BioRender.

Source: Goutman, S. (2025) j37l770

Gene-Specific Therapies

Genetic forms of ALS offer hope that treatments can be personalized to the specific patients harboring a monogenic form of disease. While this potential shows promise for some genetic forms, such as SOD1 [131, 132] and FUS [199], similar efficacy has unfortunately not been realized to date with others, including C9orf72 HRE [200] and ATXN2 [201]. In cases of genetic ALS, mutations lead to motor neuron pathology via multiple mechanisms, such as oxidative stress with SOD1 and RNA processing with C9orf72. However, even in sporadic ALS, genetic therapies may have amenable targets. An example is the microtubule binding protein stathmin-2 (STMN2), which exhibits reduced expression due to TDP-43 mislocalization [202] and is currently being targeted by an antisense oligonucleotide in clinical trial.

There are multiple approaches under development to target ALS genes. Strategies include antisense oligonucleotides (ASOs), ribonucleic acid (RNA) interference, and viral vectors [203]. ASOs are currently in clinical trials for C9orf72, SOD1, FUS, and coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) ALS [204]. ASOs are single-stranded RNA or DNA sequences of 13–25 nucleotides. They can deliver a normal copy of a mutated (target) gene or knock out a causative gene by targeting its RNA [203]. ASO mechanisms vary and include messenger RNA degradation, splicing modulation, or inhibition of RNA-binding protein-mediated protein translation. Limitations of ASOs include rapid degradation and inability to cross the blood–brain barrier, requiring treatment with repeated intrathecal injections [205]. As a result, the drug is usually administered via repeated lumbar punctures, but other strategies are contemplated [206].

RNA interference molecules, namely small interfering RNAs (siRNA), are also being studied in ALS clinical trials [204]. siRNA are double-stranded, 19–23 nucleotide RNA sequences, which are assembled within the cell to form RNA-induced silencing complex (RISC). The RISC is then used similarly to an ASO to degrade, silence, or alter splicing of messenger RNA to inactivate gene expression [207]. Exogenously delivered double-stranded RNA interference molecules are also rapidly degraded, necessitating high doses. Strategies can be employed to mitigate this. One such strategy is the use of precursor RNAs including short hairpin RNA and microRNAs, which are processed in the cytoplasm to siRNA and are continuously produced within cells. Another strategy for both siRNAs and ASOs is conjugating them to carrier molecules such as peptides or nanoparticles. Finally, viral vectors can be employed to more stably deliver and express these transgenes [203].

Viral vectors, most commonly adeno-associated viral (AAV) vectors, are non-enveloped, single-stranded viruses that have DNA modifications to deliver the desired therapeutic gene. The AAV can either deliver a normal copy of a diseased gene or deliver ASOs or RNA interference molecules, as mentioned previously [205]. One benefit of viral vectors is their ability to target specific cell types allowing for more targeted delivery, known as viral tropism. Specifically, AAV9 has tropism for neurons and is frequently used in ALS drug development [205]. Additional benefits of viral vectors include their persistence in cells as extra-chromosomal plasmids, limiting risk of insertional DNA mutations in the host [203, 208], and their ability to cross the blood–brain barrier, allowing for intravenous delivery [205]. AAV are not without limitations, which include small packaging capacity (5 kilobases), limiting delivery of large genes, and development of antibodies (while AAV is poorly immunogenic, both the capsid and transgene can elicit an immune response). Mechanisms to overcome these limitations are discussed elsewhere [209, 210].

Proteostasis

Protein homeostasis, or proteostasis, is essential for normal cellular function and involves maintaining proteins in appropriate conformation, concentration, and location within the cell [211]. A delicate balance of protein synthesis, trafficking, and degradation requires the interplay of complex cellular factors to achieve proteostasis [212]. A functioning proteome allows a cell to adapt to environmental conditions and cellular stress; without it, a cell becomes dysfunctional and eventually undergoes apoptosis if the stressor is not ameliorated [211].

Prolonged cellular stress conditions within motor neurons dysregulate normal protein homeostasis [212]. Cellular stress leads to both impaired protein production and reduced protein degradation. Affected pathways involved in protein production include messenger RNA dysregulation, protein misfolding, and protein mislocalization. Changes to the unfolded protein response (UPR) and autophagy pathways reduce clearance of misfolded proteins [211]. Overall, downstream effects of an abnormal proteome lead to a vicious cycle of disruption in proteostasis, including endoplasmic reticulum stress, mitochondrial dysfunction, and synapse destruction within motor neurons [213].

The hallmark of defective proteostasis in ALS is cytoplasmic ubiquitinated protein inclusions (such as TDP-43 or FUS) [211, 214]. Some of these proteins, including TDP-43, have prion-like domains that are particularly prone to aggregation [12]. It remains unclear whether these inclusions are the inciting factor or downstream consequence of motor neuron pathology [211]. Motor neurons are perhaps more susceptible to dysregulated proteostasis given their large size [215]. However, protein aggregates are also found in other neurodegenerative disorders, supporting a causal role in neuronal degeneration [12].

Protein mislocalization and cytoplasmic aggregation are prominent features in both genetic and nongenetic ALS, leading to loss of essential cellular functions. Many genes implicated in ALS involve pathways of protein production [211]. As a primary example, TDP-43, a product of the TARDBP gene, is a DNA- and RNA-binding protein that plays an essential role in RNA metabolism including splicing, repression, and messenger RNA nucleocytoplasmic shuttling [12, 216]. Loss of nucleolar function leads to altered levels of more than 600 messenger RNAs [217]. Similarly, FUS is an RNA-binding protein whose loss of function may reduce multiple protein activities [214].

Perhaps more importantly, protein aggregates lead to harmful downstream effects due to toxic gain-of-function. Misfolded or aggregated proteins in the endoplasmic reticulum (ER) lumen trigger the UPR. The UPR is a signaling pathway activated by ER stress that tries to promote protein refolding or clearance, and if unsuccessful, activates apoptotic pathways [218]. Separately, misfolded or aggregated proteins in the cytosol participate in stress granule formation. Stress granules are dynamic, membrane-less clusters of messenger RNAs and RNA-binding proteins (including TDP-43) that assemble in response to cell stress. They redirect translation toward stress-response proteins to restore cellular homeostasis [8]. When stress is resolved, these granules are normally disassembled. However, when mutant FUS, TDP-43, and ATXN2 aggregate within stress granules, they form insoluble aggregates, impacting appropriate transcription [12, 219].

Finally, autophagy, the process of degrading misfolded proteins and damaged organelles, is required for cell survival but becomes aberrant under stress conditions. Normally, autophagosomes deliver cellular constituents (such as TDP-43 aggregates and stress granules) to lysosomes, where they are degraded and recycled [8, 220]. This lysosomal stress response is regulated by mammalian target of rapamycin (mTOR) and transcription factor EB [221]. However, many ALS-implicated genes, including C9orf72, are also involved [8, 220]. For example, C9orf72 is part of a protein complex that controls autophagosome formation and lysosomal fusion, and when impaired, leads to accumulation of TDP-43 aggregates [222]. Similar to autophagy, which is mediated within the cell, neurons can also exocytose protein aggregates, which can be influenced by PIKfyve kinase inhibition [223].

A total of 14 current investigational agents target proteostasis (Table 2) by attempting to reduce mutated proteins prone to aggregation or enhancing cellular mechanisms to clear aggregates [212]. Various mechanisms include targeting protein aggregates (e.g., with ASOs, antibodies, or other methods), enhancing autophagy or exocytosis, or reducing apoptosis in response to ER stress.

Table 2.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting proteostasis

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

Trehalose

NCT05136885

Completed

2/3 Small molecule Activates transcription factor EB and inhibits mTOR Proteostasis: promotes lyososomal biogenesis and induces autophagy. Oxidative stress: free radical scavenger. Neuroinflammation: inhibits proinflammatory cytokines and NF-kB. All 24 weeks Randomized, blinded 161 Failed to meet primary or secondary endpoints. Some signal of slowed ALSFRS-R decline in the TUDCA-sodium phenylbutyrate free group. Ongoing post-hoc analyses. [329] [330]

ION363 (ulefnersen)

NCT04768972

Recruiting

3 Gene therapy ASO versus FUS transcript Proteostasis: reduces FUS aggregates. FUS 60 weeks Randomized, blinded 95 (est) [214]

Lithium carbonate

NCT06008249

Recruiting

3 Repurpose (bipolar disorder) Interacts with multiple neurotransmitters and attenuates calcium influx Proteostasis: increases autophagic vacuoles in mice. Mitochondrial dysfunction: promotes biogenesis of mitochondria. All, with specific UNC13A genotype 24 months Randomized, blinded 171 (est) Prior lithium study in ALS terminated for futility. [331] [332]

3K3A-APC

NCT05039268

Completed

2 Small molecule Recombinant variant of activated protein C with reduced anticoagulant activity Proteostasis: in C9orf72 mouse models, lowers C9orf72 dipeptide repeat protein levels and restores nuclear TDP-43 localization. Rescues the survival of sporadic ALS induced motor neurons. Excitotoxicity: lowers glutamate receptor levels. All 7 days Open label 16 [333]

LAM-002A

NCT05163886

Active, not recruiting

2 Small molecule Inhibits PIKfyve Proteostasis: inhibits transcription factor EB pathway to increase lysosome production and induce autophagy. C9orf72 12 weeks Randomized, blinded 14 Demonstrated CNS penetrance and target engagement. Resulted in the greatest reduction in CSF poly(GP) levels observed to date in C9orf72 clinical trials. [334] [335]

AP-101

NCT05039099

Active, not recruiting

2 Biologic Monoclonal antibody (IgG1) versus SOD1 protein Proteoastasis: targets misfolded SOD1 Sporadic or SOD1 48 weeks Randomized, blinded 63 [336]

Tideglusib

NCT05105958

Not yet recruiting

2 Small molecule Inhibits GSK-3 Proteostasis: Reduces TDP-43 phosphorylation to lower cytoplasmic aggregation and recover nuclear localization. All (excluding SOD1 or FUS mutations) 14 weeks Randomized, blinded 98 (est) [337]

Bosutinib

NCT04744532

Unknown

1/2 Repurpose (chronic myeloid leukemia) Inhibits Src/c-Abl tyrosine kinase Proteostasis: promotes autophagy to reduced protein aggregates and inhibit motor neuron death. Sporadic or SOD1 12 weeks phase 1, 24 weeks phase 2 Open label 25 (est, phase 2 arm) In phase 1 trial, dose-limiting toxicity observed at 400mg dose. ALSFRS-R remained stable or declined in 7 of 9 patients on 100-300mg dosing. [338]

AMT-162

NCT06100276

Recruiting

1/2 Gene therapy AAV vector with microRNA versus SOD1 transcript Proteostasis: microRNA silences SOD1 in lower motor neurons. SOD1 5 years Open label (phase 1) 6–8 (est, phase 2 arm) [339]

Monepantel

NCT04894240

Completed

1 Repurpose (livestock wormicide) Inhibits mTOR Proteostasis: inhibits mTOR, reducing protein accumulation. mTOR inhibition slows progression in preclinical ALS models. All (excluding SOD1 or VCP mutations) 6 months Open label 12 43-58% slowing in decline of ALSFRS-R (secondary endpoint) in high dose cohort in 2-year open label extension. Enrolling in HEALEY trial. [340]

RAG-17

NCT06556394

Recruiting

1 Gene therapy Small interfering RNA bound to accessory oligonucleotide versus SOD1 transcript Proteostasis: reduces mutant SOD1 protein by binding mRNA. SOD1 Unknown Randomized, blinded 32 (est) Well tolerated at all doses. [341]

VRG50635

NCT06215755

Recruiting

1 Small molecule Inhibits PIKfyve Proteostasis: stimulates exocytosis of protein aggregates from motor neurons. All 28 days Randomized, blinded 50 (est) [223]

ALN-SOD

NCT06351592

Recruiting

1 Gene therapy Small interfering RNA versus SOD1 transcript Proteostasis: reduces mutant SOD1 protein by binding mRNA. SOD1 4 weeks Randomized, blinded 42 (est)

Prosetin

NCT05279755

Unknown

1 Small molecule Inhibits MAP4K Proteostasis: reduces apoptotic response to misfolded proteins, which trigger ER stress and the unfolded protein response. All 7 days Randomized, blinded 32 (est) [342]

Abbreviations: AAV, adeno-associated virus; ALSFRS-R, ALS functional rating scale revised; ASO, antisense oligonucleotides; CNS, central nervous system; CSF, cerebral spinal fluid; ER, endoplasmic reticulum; est, estimated; FUS, fused in the sarcoma; GP, glycine-proline; GSK-3, glycogen synthase kinase-3; MAP4K, MAP kinase kinase kinase kinase; mTOR; mechanistic target of rapamycin; NF-kB, nuclear factor kappa-beta; TDP-43, transactive response DNA binding protein; TUDCA, tauroursodeoxycholic acid

Integrated Stress Response

Integrated stress response (ISR) is an integral part of protein homeostasis that is directly targeted by several drugs in clinical trials. Under conditions of cellular stress, including ER stress (via the UPR), cytosolic protein aggregation, starvation, and infection, the ISR is activated to restore cellular homeostasis [224]. It does this by reducing global protein synthesis and enhancing gene expression targeted toward cellular recovery (chaperones, protein folding, etc.) [225, 226].

Specifically, under normal conditions, the eukaryotic initiation factor-2 (eIF2) complex (composed of alpha, beta, and gamma subunits) assists with RNA translation via ribosomal recognition of the AUG start codon [227]. However, during the integrated stress response, this process is halted and instead open reading frame messenger RNAs (mRNAs) are preferentially translated [224]. Of these mRNAs, perhaps most important is ATF4, a transcription factor that binds DNA targets to regulate gene expression, essential for restoring mitochondrial homeostasis [228]. When cellular homeostasis has been restored, the ISR is turned off via protein phosphatase 1 complex (PP1R15A, also known as growth arrest and DNA damage-inducible protein GADD34, and PPP1R15B). This negative feedback loop is important to restore normal protein synthesis [225, 229].

In ALS, the ISR can lead to increased translation of mutant genes. For example, C9orf72 GGGGCC repeat expansions can be translated into toxic dipeptide repeats via pathogenic non-AUG repeat-associated (RAN) translation. RAN translation is enhanced by the preferential translation of open reading frame mRNAs during the ISR [230]. Additionally, as cellular homeostasis cannot be restored in ALS, a prolonged integrated stress response eventually leads to cell death. This is in part mediated by PPR15A/B dephosphorylation of eIF2a, allowing for synthesis of death-inducing proteins [225, 231]. Elevated markers of the integrated stress response and unfolded protein response have been identified in postmortem samples from patients with nongenetic ALS [224, 232].

Investigational agents involving the ISR in current ALS clinical trials target the eIF2 complex, PP1R15A, and pathological RAN translation (Table 3).

Table 3.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting the integrated stress response

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

ABBV-CLS-7262 (fosigotifator)

NCT05740813

Enrollment complete

2/3 Small molecule Activates eIFB2 ISR: inhibits the ISR to restore protein synthesis and dissolve TDP-43 containing stress granules All 24 weeks Randomized, blinded 310 No significant difference in combined analysis of ALSFRS-R and mortality (primary endpoint) or in respiratory function (secondary endpoint). Slower deterioration of handheld dynamometry (secondary endpoint) in high dose group. [343]

DNL 343

NCT05842941

Enrollment complete

2/3 Small molecule Activates eIFB2 ISR: inhibits the ISR to restore protein synthesis and dissolve TDP-43 containing stress granules All 24 weeks Randomized, blinded 240 (est) Failed to meet primary endpoint in decline of ALSFRS-R as well as secondary survival endpoints. Detailed results pending publication. [344]

Metformin

NCT04220021

Active, not recruiting

2 Repurpose (diabetes) Glucose metabolism ISR: reduces repeat-associated non-AUG translation of C9orf72 dipeptide repeats, which occurs more frequently during the ISR. C9orf72 24 weeks Open label 18 [345]

IFB-088 (icerguastat)

NCT05508074

Active, not recruiting

2 Small molecule Inhibits PPP1R15A and NMDA subtype 2B receptors ISR: prolongs ISR to adjust protein production rate and encourage protein folding. Excitotoxicity: inhibits NMDA receptors to reduce glutamate excitotoxicity. All (bulbar-onset only) 26 weeks Randomized, blinded 51 (est) [231, 346]

Abbreviations: ALSFRS-R, ALS functional rating scale revised; eIFB2, eukaryotic initiation factor 2; est, estimated; ISR, integrated stress response; PPP1R15A, protein phosphatase 1, regulatory subunit 15A; TDP-43, transactive response DNA binding protein; NMDA, N-methyl-D-aspartate

Neuroinflammation

Central and systemic inflammation play a role in ALS pathogenesis, with some immune cell targets [8, 233] leading to investigational considerations (Table 4). Astrocytes in the central nervous system (CNS) help to maintain the integrity of the blood–brain barrier and mediate release of antiinflammatory molecules, such as interleukins (IL) and tumor necrosis factor (TNF) [234]. Overproduction of these signals drives necrotic cell death, or necroptosis [235]. Higher levels of these inflammatory cytokines have been found in CSF and serum of patients with ALS and correlate negatively with survival, specifically TNF-alpha, interleukin-10 and interleukin-1 beta [236]. These molecules further activate the nuclear factor-kappa B (NF-kB) pathway, which perpetuates proinflammatory destruction in the CNS, muscle, and neuromuscular junction [237]. Activation of NF-kB has also been linked to both SOD1 gene function [238] and C9orf72 [239]. Of further support, cultured astrocytes from postmortem tissue of patients with ALS are toxic to motor neurons [240].

Table 4.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting neuroinflammation

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

Zilucoplan

NCT04436497

Completed

2/3 Small molecule Inhibits C5 complement Neuroinflammation: inhibits terminal complement cascade (active in SOD1 mouse models) to reduce neuroinflammation. NMJ: reduces NMJ destruction by inhibiting complement activation at motor neuron terminals and Schwann cells. All 24 weeks Randomized, blinded 162 Failed to meet primary endpoint in ALSFRS-R at 24 weeks, therefore terminated for futility. [347349]

Dolutegravir, Abacavir and Lamivudine (Triumeq)

NCT05193994

Active, not recruiting

3 Repurposed (HIV) Combined nucleoside reverse transcriptase inhibitor and integrase strand transfer inhibitor Neuroinflammation: reduces HERV-K, which has increased transcripts in ALS that may drive inflammation. All 24 months Randomized, blinded 390 (est) Previous antiretroviral clinical trials were ineffective (zidovudine monotherapy in 12 patients, indinavir monotherapy in 46 patients). [350]

RAPA-501

NCT04220190

Recruiting

2/3 Biologic T regulatory/Th2 cells Neuroinflammation: ex vivo manufactured cells with epigenetic reprogramming to enrich an anti-inflammatory phenotype. Regulatory T cell expansion prolonged survival in mouse models. All 24 weeks Open label 41 (est) [351]

NP001

NCT02794857

Completed

2b Small molecule Sodium chlorite Neuroinflammation: Downregulates NF-kB expression in macrophages to inhibit production of pro-inflammatory cytokines. All 24 weeks Randomized, blinded 138 Seeking FDA approval as of 10/2024. No difference in ALSFRS-R score (primary endpoint), SVC (secondary endpoint), or percentage of non-progressors overall. Subpopulation of patients 40–65 years old had 36% slower decline in ALSFRS-R and 51% slower decline in SVC. [352, 353]

AT-1501 (tegoprubart)

NCT04322149

Completed

2 Biologic Antibody versus CD40 ligand Neuroinflammation: blocks CD40 ligand on T cells to suppress the adaptive immune system that is overactive in ALS. All 11 weeks Open label 54 Dose-dependent reduction in proinflammatory biomarkers. [354, 355]

ANX005

NCT04569435

Completed

2 Biologic Antibody versus C1q complement Neuroinflammation: inhibits classical complement cascade (elevated in spinal cord and motor cortex in ALS) to reduce inflammation and subsequent neurodegeneration. All 22 weeks Open label 17 Prior study showed Cq1b inhibition had no effect. [356, 357]

ZYIL1 (usnoflast)

NCT05981040

Completed

2 Small molecule Inhibits NLRP3 inflammasome Neuroinflammation: reduces inflammation triggered by pathogen or damage-associated molecular patterns. Validated in inflammatory preclinical animal models. All 12 weeks Randomized, blinded 24 FDA approved US phase IIb study of 210 patients. [358, 359]

TPN-101

NCT04993755

Active, not recruiting

2 Repurposed (HIV) Nucleoside reverse transcriptase inhibitor (derivative of stavudine) Neuroinflammation: Loss of nuclear TDP-43 causes reactivation of virus-derived retrotransposable DNA including LINE-1. Inhibiting LINE-1 reverse transcriptase reduces the antiviral immune response that otherwise drives neurodegeneration. C9orf72 24 weeks Randomized, blinded 42 At 48 weeks, 40% less decline in ALSFRS-R, 50% slower decline in vital capacity, and reduced neurofilament level (secondary endpoints). [360, 361]

WP-0512 (fasudil)

NCT05218668

Active, not recruiting

2 Small molecule Inhibits rho kinase Neuroinflammation: inhibits proinflammatory and apoptotic signaling pathways. Reduces microglial activation. Axonal transport: regulates cytoskeleton including actin and microtubules, involved in vesicle transport among many other functions. All 24 weeks Open label 40 (est) Approved in China and Japan for treating subarachnoid hemorrhage. Prior phase 2 trial on intravenous fasudil showed reduced decline in MUNIX scores versus placebo. [362366]

Dazucorilant

NCT05407324

Active, not recruiting

2 Small molecule Modulates glucocorticoid receptors Neuroinflammation: counteracts high cortisol levels found in ALS and reduces microglial proinflammatory response. All 24 weeks Randomized, blinded 249 No slowing in ALSFRS-R decline (primary endpoint) but did improve survival. Gastrointestinal side effects more common. Ongoing open-label extension. [367, 369]

TCD601 (siplizumab)

NCT06453668

Active, not recruiting

1 Biologic Monoclonal antibody vs CD2 Neuroinflammation: targets CD2 receptor on T cells to reduce mature T cells and increase regulatory T cells. All 52 weeks Open label 48 (est) [368]

Abatacept and Proleukin (IL-2)

NCT06307301

Active, not recruiting

1 Repurposed, biologic (rheumatoid arthritis) CTLA-4 analog and interleukin-2 Neuroinflammation: prevents interaction of antigen presenting cells (CD80/CD86 ligands) with T cells (CD28/CTLA4 receptors) to inhibit T cell activation. All 59 weeks Open label 5 [369]

FB418

NCT05995782

Recruiting

1 Small molecule Inhibits LRRK2 and c-Abl Neuroinflammation: inhibits neuroinflammatory response in microglia and modulates lysosome pathway and autophagy in immune cells. All Unknown Randomized, blinded 64 (est) [370]

CK0803

NCT05695521

Recruiting

1 Biologic Umbilical cord blood derived regulatory T cells Neuroinflammation: regulatory T cells inhibit proinflammatory immune cells and higher cell counts prolong survival in ALS. All 24 weeks Randomized, blinded 66 (est) [371]

Abbreviations: ALSFRS-R, ALS functional rating scale revised; CLTA-4, cytotoxic T-lymphocyte-associated protein 4; est, estimated; HERV-K, human endogenous retrovirus-K; HIV, human immunodeficiency virus; LRRK2, leucine-rich repeat kinase 2; LINE-1, long interspersed nuclear elements 1; MUNIX, motor unit number index; NF-kB, nuclear factor kappa-beta; NLRP3, NOD-like receptor protein 3; NMJ, neuromuscular junction; TDP-43, transactive response DNA binding protein; TH2, T helper cell type 2; SVC, slow vital capacity

Microglia are felt to have a more direct role in pathologic inflammatory changes in ALS. In the M1 state, microglia are thought to be more proinflammatory and toxic, while in the M2 state microglia are anti-inflammatory and more restorative [241]. In SOD1 mouse models, end-stage disease is associated with higher markers of M1 activation (such as NOX2 and other reactive oxidative species) compared with higher levels of M2 activation (such as CD163 and brain-derived neurotrophic factor (BDNF)) at disease onset [242]. M1 macrophages in patients with ALS are found to release more toxic cytokines, such as IL-6 and TNFα, as compared with healthy controls [243]. Positron emission tomography (PET) studies have also demonstrated higher levels of microglial activation in the motor cortices of patients with ALS [244].

There is increasing evidence of peripheral and central immune system cross connectivity contributing to acceleration of a proinflammatory state in ALS [245]. Mechanisms by which the barrier is disrupted include direct changes to the integrity of the blood–brain barrier via impaired tight junctions and adhesion proteins, leading to increased permeability [246, 247] and changes in barrier cell concentration and increased localized secretion of inflammatory proteins [248, 249]. Chronic activation of the peripheral immune system exacerbates disruption of the central and peripheral immune barrier, and more specifically to ALS, levels of specific pro- or anti-inflammatory cytokines and chemokines are inappropriately altered [250]. As a result of the barrier disruption, additional pathologic changes in the peripheral immune system are capable of affecting the central immune response [251]. For example, dendritic cells, which prime T cells as part of the innate immune response, are found in abundance in the ventral horn and corticospinal tracts, specifically, in postmortem spinal cord tissue [252]. Regulatory T cells (Tregs), which act to prevent an excessive immune response or autoimmunity, are found in greater numbers during a slow-progression phase of SOD1 mouse models but increase during rapid disease progression, suggesting that control of Tregs may influence disease progression [253]. Ratios of “classical” monocytes (CD14-/CD16-), which are more responsible for phagocytosis and initiation/priming of the immune system [254], are higher in patients with ALS and pre-symptomatic SOD1 carriers as compared with healthy controls, but function at a decreased efficacy and invade the CNS, though the difference between the roles of monocytes in the CNS versus systemic circulation are still unknown [255]. Evaluation of the complement cascade, the initiation of the innate immune response, is a natural investigation. The findings in mouse models of SOD1 ALS show an increase of complement activators (C1qB, C4, C3, C5, and CD88) and decrease in complement regulators (CD55, CD59a) during disease progression in spinal-cord-tissue supported investigations of complement inhibitors in ALS [256]. Higher levels of circulating serum cortisol are found in patients with nongenetic ALS, though correlation with symptom onset and progression are variable [257, 258]. Glucocorticoid receptor (GR) modulation has been shown to reduce inflammation and glial reactivity in mouse models [259]. Other peripheral immune cells, such as neutrophils [260] and natural killer cells [52, 261263], show associations with ALS and may influence future therapeutic targets [264].

The rapidity of clinical decline and death in ALS has led many to investigate whether there could be an infectious etiology of the disease, primarily leading to research into transposable elements of the genome, meaning parts of genes that can “jump” from one chromosome to another and are present and essential in the healthy human genome [265]. These transposable elements include transposons and retrotransposons, such as endogenous retroviruses, which can essentially “cut and paste” into any portion of the human genome. In humans, the human endogenous retrovirus-K (HERV-K) is the most well studied and has been found to be increased in multiple degenerative disease states [266]. In ALS, expression of HERV-K and its envelope protein are found in higher concentrations in cortical pyramidal cells and spinal anterior horn cells and are specifically not seen in the other spinal cord tracts or nonmotor cortical areas [267].

Neurotrophic Factors and Stem Cells

Neurotrophic factors are proteins that promote a neuroprotective environment to slow neurodegeneration. They are endogenously used in response to injury or disease to enhance neuron survival, axonal growth, synapse formation, and plasticity [268]. The goal of investigational neurotrophic agents is to promote motor neuron growth, repair, and survival regardless of which cellular mechanisms are damaged. Examples of endogenous neurotrophic factors include BDNF, insulin-like growth factor-1, erythropoietin, granulocyte-colony stimulating factor, and hepatocyte growth factor, among others [269]. Many of these factors have been studied in ALS mouse models and found to reduce motor neuron degeneration and improve survival [268]. There are several investigational agents in clinical trials utilizing this approach (Table 5).

Table 5.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials involving neurotrophic factors

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

Nerve Growth Factor

NCT06391645

Not yet recruiting

2/3 Small molecule Nerve growth factor encapsulated with 2-MPC nanocapsules Neurotrophic: enhances neuron regeneration, showing prolonged survival time in SOD1 mouse model. All 12 weeks Open label 60 (est)

VM202 (engensis)

NCT04632225

Completed

2 Biologic Small circular DNA delivering hepatocyte growth factor gene Neurotrophic: hepatocyte growth factor delivered to muscle induces blood vessel and nerve growth, prevents muscle loss. All 4 months Randomized, blinded 18 Safe and well tolerated in phase 2a study. Awaiting further trial analysis to determine next steps in drug development. [372]

MRG-001 (plerixafor and low-dose tacrolimus)

NCT06315608

Not yet recruiting

2 Repurposed (lympoma, multiple myeloma; organ transplant rejection) CXC chemokine receptor type 4 antagonist and calcineurin inhibitor Neurotrophic: mobilizes bone marrow stem cells to sites of injury, stimulating tissue repair. Neuroinflammation: mobilizes immunoregulatory cells to reduce inflammation at sites of injury. All 3 months Open label 10 (est) [373]

Aleeto

NCT06181526

Not yet recruiting

1 Small molecule Protein polymers secreted by stem cells under stress Neurotrophic: enhances neuron repair. All 66 days Randomized, blinded 24 (est)

Abbreviations: est, estimated; MPC, methacryloyloxyethyl phosphorylcholine

There is incredible interest surrounding the role of stem cell therapy for the treatment of ALS [270]. In early trials, stem cells were transplanted into patients with ALS with the goal of generating new motor neurons. However, they were unable to integrate due to a diseased microenvironment and long distance required for axon growth. Thus, while upstream stem cells do have the ability to differentiate into diverse cell types, in ALS, stem cells therapies are now intended to secrete neurotrophic factors, differentiate into astrocytes and microglia, or synapse with diseased motor neurons (Table 6) [271]. Stem cells can be isolated from embryonic stem cells, mesenchymal stem cells (derived from umbilical cord blood), and neural progenitor cells [272]. Challenges to use of stem cells include optimizing the correct source, dose, and delivery method [270].

Table 6.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials involving stem cells

Drug name
NCT
Study status
Phase Drug category Stem cell type Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

Human neural stem cells

NCT06344260

Recruiting

2 Stem cell Human neural stem cells All Single dose Randomized, blinded 30 (est)

Mononuclear cells

NCT04849065

Unknown

2 Stem cell Autologous bone marrow mononuclear cells (including stem cells) All Single dose Randomized, blinded 100 (est) [374]

hUC-MSC-sEV-001

NCT06598202

Recruiting

1/2 Stem cell Exosomes derived from human umbilical cord blood mesenchymal stem cells All 14 days (phase 1), unknown (phase 2) Randomized, blinded 38 (est)

Mesenchymal stem cells

NCT04651855

Unknown

1/2 Stem cell Mesenchymal stem cells isolated from Wharton’s jelly All 60 days Open label 20 (est)

CNS10-NPC-GDNF

NCT05306457

Recruiting

1 Stem cell Human neural progenitor cells differentiated into astrocytes with glial cell line-derived neurotrophic factor All 3 weeks to 3 months Open label 16 (est) [375]

Abbreviations: est, estimated

Oxidative Stress

Oxidative stress plays a key role in neuronal injury and aging by altering the structure of cellular components (lipids, proteins, DNA, and RNA), leading to mitochondrial damage, and ultimately, cell death [8, 273]. It is mediated by reactive oxygen species (ROS) formed by the partial reduction of oxygen, including superoxide, hydrogen peroxide, and hydroxyl radical [273]. Mitochondria are a prominent site of superoxide production [274]. This occurs due to oxygen reduction necessary for ATP generation [275]. Oxidative stress is also produced outside of the mitochondria. In microglia, myeloperoxidase, a heme-containing enzyme involved in microbial response, utilizes hydrogen peroxide to create hypochlorous acid, an extremely potent reactive halogen species. Hypochlorous acid produced in excess causes cytotoxic cellular damage [276, 277].

Under normal conditions, ROS generation is balanced with antioxidant mechanisms, known as “free radical scavengers.” Antioxidant enzymes include both superoxide dismutase (as implicated in SOD1 ALS), which detoxicates superoxide into the less reactive hydrogen peroxide, and peroxisomal enzymes, which then reduce hydrogen peroxide to water [278].

In a healthy cell, ROS are necessary and have roles in cell signaling [277]. However, in ALS, oxidative stress overwhelms typical antioxidant responses. Increased levels of oxidative stress have been identified in patients with ALS [273]. For example, due to high energy demand and impaired glucose metabolism in ALS, neurons and astrocytes switch to fatty acid beta-oxidation as a key energy source. This incurs risk of creating ROS [279]. Increased ROS leads to several downstream effects including TDP-43 mislocalization and aggregation, mitochondrial dysfunction, and excitotoxicity from astrocyte-mediated glutamate release [8]. Additionally, oxidative stress leads to peroxidation of cellular membrane phospholipids (polyunsaturated fatty acids), leading to motor neuron damage via multiple mechanisms [280].

Current investigational drugs target pro-oxidant enzymes, lipid peroxidation, and antioxidant mechanisms (Table 7).

Table 7.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting oxidative stress

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

Verdiperstat

NCT04436510

Completed

2/3 Small molecule Irreversibly inhibits myeloperoxidase enzyme Oxidative stress: inhibits pro-oxidant enzyme in microglia. All 24 weeks Randomized, blinded 167 Failed to meet primary endpoint (ALSFRS-R) or secondary endpoints. [276, 376]

PTC857 (utreloxastat)

NCT05349721

Active, not recruiting

2 Small molecule Inhibits 15-lipooxygenase Oxidative stress: inhibits an enzyme activated by reactive oxygen species that otherwise increases lipid peroxidation and cell death. All 24 weeks Randomized, blinded 307 [377]

RT001

NCT04762589

Unknown

2 Small molecule 11, 11 di-deuterated linoleic ethyl ester Oxidative stress: makes membrane PUFAs resistant to lipid peroxidation. All 24 weeks Randomized, blinded 40 (est) Nonsignificant signals of clinical benefit in ALSFRS-R (primary endpoint). [280, 378]

Tetramethylpyrazine nitrone

NCT04667013

Not yet recruiting

1 Small molecule Free radical scavenger Oxidative stress: improves mitochondrial antioxidant activity. Healthy volunteers 7 days Randomized, blinded 16 (est) Previous phase 2 trial of 155 patients showed slower decline in grip strength but not in ALSFRS-R. [379, 380]

Abbreviations: ALSFRS-R, ALS functional rating scale revised; est, estimated; PUFAs, polyunsaturated fatty acids

Neuromuscular Junction and Axonal Transport

The neuromuscular junction (NMJ) is the crucial connection between the motor neuron axon terminal and the muscle. Terminal Schwann cells (TSCs) are also a critical part of the NMJ, aiding in their development and maturation [281]. NMJ dysfunction and instability is a frequently encountered electrophysiologic finding in the evaluation of ALS [282]. In SOD1 mouse models, endplate denervation was seen prior to clinical weakness and motor neuron loss, suggesting that NMJ compromise occurs early in the pathophysiology of ALS and is often the inciting event prior to lower motor neuron injury (“dying back” hypothesis) [283].

Impaired presynaptic vesicle pool storage and ineffective or malformed presynaptic terminal formation have been demonstrated in multiple animal models of hereditary ALS [284, 285]. Axonal transport is essential to move cellular proteins to and from the NMJ, which requires an eloquent combination of neurofilaments, actin, microtubules, and motor proteins. High levels of vesicles are found in the NMJ of patients with ALS, suggesting dysfunctional axonal transport [286]. A key protein in axonal transport is Stathmin 2 (STMN2), which aids in axonal regrowth after injury and is found to be altered specifically by the toxic accumulations of TDP-43 and is found in lower quantities in spinal cords of mouse models of ALS [202, 287].

Postsynaptic NMJ changes, even preclinically, have also been identified in ALS [288, 289]. Dysregulation of TSCs in ALS is thought to lead to impaired and erroneous NMJ recovery and plasticity [290]. Early in disease, local signaling via miR-206-related pathways may allow for compensatory reinnervation until a certain threshold of functional muscle loss [291]. Ultimately, it seems as though injury to the NMJ is a culmination of mutant protein aggregation at the presynaptic nerve terminal, presynaptic cholinergic dysfunction, impaired axonal transport, reduced synaptic acetylcholinesterase, and diseased muscle itself, among others [292]. Each mechanism of dysfunction at the pre-synaptic, post-synaptic, and peri-synaptic level offers itself as a target for intervention to improve stabilization, junctional signaling, and synapse integrity [213], potentially even before symptomatic weakness in those with genetic susceptibilities. Current therapies under investigation (Table 8) are targeting muscle-specific kinase (MuSK), which promotes post-synaptic clustering, synaptogenesis via F-actin branching and dendritic proliferation [293], and reduction of aberrant sphingolipid regulation to promote NMJ stability [294].

Table 8.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting the neuromuscular junction and axonal transport

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants References

ARGX-119

NCT06441682

Recruiting

2 Biologic Monoclonal antibody NMJ: activates MuSK to promote maturation and stabilization of neuromuscular junctions. All 100 weeks Randomized, blinded 60 (est) [381]

Darifenacin

NCT06249867

Recruiting

2 Repurpose (overactive bladder) M3 muscarinic receptor antagonist NMJ: blocks muscarinic receptors to trigger perisynaptic schwann cells (glial cells) to repair the NMJ. All 24 weeks Randomized, blinded 30 (est)

Ambroxol

NCT05959850

Recruiting

2 Repurpose (cough medication outside US) Inhibits glucocerebrosidase 2 among others NMJ: prevents breakdown of glucosylceramide (sphingolipid) to preserve NMJs in SOD1 mouse models. Neuroinflammation: inhibits microglial-mediated inflammation. Proteostasis: clears misfolded proteins from ER during ER stress and enhances lysosomal function. All 24 weeks Randomized, blinded 50 (est) [294]

SPG302

NCT05882695

Active, not recruiting

1 Small molecule Pegylated benzothiazole derivative NMJ: increases neural synapses that use glutamate, including motor neurons. Regulates actin cytoskeleton and increases dendritic spine density. All 28 days for 3–12 cycles Randomized, blinded 112 (est) [293]

QRL-201

NCT05633459

Recruiting

1 Gene therapy ASO vs stathmin-2 transcript Axonal transport: modulates splicing of stathmin-2, a microtubule binding protein in motor neurons critical for axon stability. Stathmin-2 expression is controlled by nuclear TDP-43 and is therefore reduced in ALS. Sporadic or C9orf72 Unknown Randomized, blinded 64 (est) [202]

Abbreviations: ASO, antisense oligonucleotide; ER, endoplasmic reticulum; est, estimated; MuSK, muscle-specific kinase; NMJ, neuromuscular junction; TDP-43, transactive response DNA binding protein

Mitochondrial Function and Energy Metabolism

Energy metabolism is dysregulated early in the course of ALS as a result of decreased appetite, increased energy expenditure, and hypothalamic dysfunction, among other complex factors [295299]. Hypermetabolism and weight loss are associated with faster disease progression [300, 301]. One strategy to ameliorate this involves introducing a high-calorie diet, which was shown to extend survival in ALS mouse models and patients with fast-progressing ALS [302]. Other strategies focus on optimizing mitochondrial function, as they are the main source of energy production (adenosine triphosphate, ATP) within the cell.

In a healthy cell, mitochondria utilize the electron transport chain for oxidative phosphorylation, the main source of ATP production. Nicotinamide adenine dinucleotide plus hydrogen (NADH) produced during glycolysis (anaerobic glucose metabolism in the cytosol) and the citric acid cycle (in mitochondria) donates electrons to initiate the electron transport chain via NADH dehydrogenase. Increased cellular energy demand leads to calcium influx into the mitochondria, driving increase in the respiratory chain and ATP synthesis [303].

In fasted states, including ALS, skeletal muscles increase utilization of fatty acid oxidation, which in turn reduces glucose metabolism [304, 305]. Additionally, mitochondria are directly compromised in ALS due to multiple mechanisms, including direct effect of protein aggregates or sequestration of mitochondrial microRNAs and proteins. The result of mitochondrial dysfunction includes excess ROS generation, altered energy production, and impaired calcium buffering. Many investigational agents have targeted mitochondrial function, including TUDCA [8, 149].

Current drugs in development target mitochondrial metabolism, including NAD+/NADH ratio, calcium influx, and fatty acid oxidation. There is also an ASO in development for genetic ALS caused by a mitochondrial protein mutation (Table 9).

Table 9.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting mitochondrial function

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

CNM-Au8

NCT04414345

Completed

2/3 Small molecule Gold nanocrystals catalyze oxidation of NADH to NAD+ Mitochondrial function: increases ATP production through glycolysis and oxidative phosphorylation via the mitochondrial electron transport chain to improve energy metabolism. Proteostasis: reduced TDP-43 aggregation in rodent spinal neurons exposed to excitotoxic glutamate levels. All 24 weeks Randomized, blinded 161 After 76-week open label extension, neurofilament levels reduced by 28% and there was 60% decreased risk of death (secondary endpoint) versus controls. [382, 383]

Pridopidine

NCT04615923

Completed

2/3 Small molecule Activates sigma-1 receptor Mitochondrial function: promotes calcium influx from ER to mitochondria under stress conditions to increase ATP and energy production. Excitotoxicity: modulates NMDA receptor function to reduce glutamate-mediated calcium influx. Proteostasis: induces autophagy to clear protein aggregates. All 24 weeks Randomized, blinded 163 Planning phase 3 trial. Failed to meet primary endpoint (ALSFRS-R) or secondary endpoints. Improved speech and articulation rate at 6 months; 41% slowing in ALSFRS-R decline in fast progressors with < 1.5 years of symptoms. [384, 385]

Trimetazidine Dihydrochloride

NCT04788745

Completed

2 Repurpose (angina pectoris outside US) Inhibits long-chain fatty acid oxidation via long-chain mitochondrial ACAA2 Mitochondrial function: inhibits fatty acid uptake to improve glucose utilization and mitochondrial ATP production (energy metabolism). All 12 weeks Open label 21 [304]

nL-CHCHD-001

NCT06392126

Active, not recruiting

1/2 Gene therapy ASO versus CHCHD10 transcript Mitochondrial dysfunction: targets CHCHD10 mutation, which causes toxic-gain-of function in this mitochondrial protein with unknown function. CHCHD10 Unknown Open label 1 [204]

Abbreviations: ACAA2, acetyl-CoA acyltransferase 2; ALSFRS-R, ALS functional rating scale revised; ASO, antisense oligonucleotide; ATP, adenosine triphosphate;  ER, endoplasmic reticulum; NADH, nicotinamide adenine dinucleotide hydride; NMDA, N-methyl-D-aspartate

RNA Metabolism

RNA metabolism involves a complex interplay of RNA splicing, transport, export, and degradation [306]. RNA-binding proteins (RBPs) bind RNA transcripts to regulate their stability, localization, and translation [307]. Messenger RNA (mRNA) is transported from the nucleus to code for proteins and non-coding microRNAs, which can further regulate gene expression [306].

Multiple aspects of mRNA and microRNA metabolism are dysregulated in ALS. TDP-43 and FUS are RBPs and their alteration leads to downstream effects on mRNA stability [8]. Other ALS gene mutations can lead to changes in RNA splicing [308]. Stress granules, which can become insoluble due to ALS protein aggregates, can sequester mRNAs and stall protein translation [306]. Investigational agents involving RNA metabolism are currently targeting microRNA maturation and processing (Table 10).

Table 10.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting RNA metabolism

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Additional drug mechanism(s) Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

PrimeC (ciprofloxacin and celecoxib)

NCT05357950

Active, not recruiting

2b Repurpose (fluoroquinolone and nonsteroidal anti-inflammatory drug) Inhibits bacterial DNA gyrase and inhibits cyclooxygenase-2 RNA metabolism: regulates microRNA processing, important in RNA interference and degradation, and chelates iron. Neuroinflammation: reduces prostaglandin synthesis, which mediates the inflammatory pathway. All 6 months Randomized, blinded 68 Reduced disease progression by 36% and improved complication-free survival by 53% (secondary endpoints). Seeking approval in Canada. Celecoxib was previously not found to be beneficial as a single agent in ALS. [172, 386]

Enoxacin

NCT04840823

Completed

1/2 Repurpose (fluoroquinolone) Inhibits bacterial DNA gyrase RNA metabolism: enhances microRNA maturation, important in RNA interference and RNA degradation. Found to delay neurological symptoms in ALS mice. All 30 days Randomized, blinded 8 No serious adverse events. Global increase in plasma and CSF microRNA levels at all post-treatment timepoints [387, 388]

Abbreviations: CSF, cerebrospinal fluid

Disrupted nucleocytoplasmic RNA transport found with C9orf72 and TDP-43 pathology is another important ALS mechanism. Mislocalization of TDP-43 to the cytoplasm sequesters nuclear pore complexes and transport factors [309]. C9orf72 mRNA sequesters a nucleocytoplasmic transport regulator in the cytoplasm, altering transport multiple RNAs and proteins through the nuclear pore [310]. No drugs in development currently target nuclear export specifically.

Excitotoxicity

Depolarization of upper motor neurons triggers glutamate release into the synapse, activating post-synaptic glutamate receptors (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, AMPA, and N-methyl-D-aspartate, NMDA) [311]. Increased glutamate release leads to motor neuron hyperexcitability, aberrant firing, increased intracellular calcium, and cell death [312]. Primary mechanisms of glutamate excitotoxicity include increased glutamate release, upregulation of AMPA receptors, and reduced astrocyte glutamate reuptake, among others [311]. Other downstream effects include reduced glutathione antioxidant levels leading to oxidative stress, increased intracellular calcium, and ER stress leading to protein aggregation of TDP-43, SOD1, and FUS [8, 311]. Notably, while riluzole mediates glutamate excitotoxicity, not many drugs in development are targeting this pathway. Thus, no drugs with a primary mechanism acting on excitotoxicity are included in a table.

Gut Microbiota

The gut microbiome includes the trillions of bacteria found in the human stomach and small and large intestines. These microbes participate in host metabolism, prevent colonization by pathogens, and facilitate inflammatory and immune functions [313]. Changes to gut microbiome in ALS include increased gut permeability and low levels of butyrate-producing bacteria [314, 315]. Supplementation with butyrate improved survival time in an ALS mouse model [316]. It is also hypothesized that molecules produced by gut microbes could weaken the blood–brain barrier and promote systemic neuroinflammation [313]. Drugs in development target gut permeability (Table 11).

Table 11.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting gut microbiota

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

PLL001

NCT06513546

Not yet recruiting

1/2 Small molecule Poly-L-lysine conjugated with small chain fatty acids (acetate, butyrate, lactate, propionate) Microbiome: replaces gut microbiome and restores gut and blood–brain barrier permeability. All 24 weeks Randomized, blinded 141 (est, phase 2 arm) [389]

MaaT033

NCT05889572

Active, not recruiting

1b Biologic Gut microbiota Microbiome: replaces gut microbiome and restores gut permeability and gut–brain axis. All 56 days Open label 15 (est) Safe and well tolerated. [313, 390]

Abbreviations: est, estimated

Multiple Mechanisms

Some investigational drugs have broad cellular targets, affecting multiple pathways implicated in ALS pathogenesis (Table 12). It is possible that targeting many pathophysiological mechanisms at once could be necessary to make significant progress in slowing disease progression.

Table 12.

Investigational therapeutics in ALS phase 1, 2, and 3 clinical trials targeting multiple mechanisms

Drug name
NCT
Study status
Phase Drug category Drug target Drug mechanism Genetic or sporadic ALS Treatment period Trial design Number of participants Results References

NX210c

NCT06365216

Recruiting

2 Small molecule Glycoprotein derived from subcommissural organ-spondin Multiple: involved in neuron maturation during embryogenesis. Restores blood–brain barrier, reduces glutamate excitotoxicity, and enhances neurotransmission. All 26 days Randomized, blinded 80 (est) Tolerability proven in phase 1b study. [391, 392]

Clenbuterol

NCT04245709

Completed

2 Repurpose (asthma outside US) Beta-2 agonist Multiple: affects multiple downstream signaling pathways in neurons, microglia, and muscle to induce neurotrophic factors, inhibit glutamate excitotoxicity, reduce microglial neuroinflammation, and preserve NMJ. All 24 weeks Open label 25 13 of 25 participants withdrew due to adverse events. Slowing in ALS FRS-R decline (secondary endpoint) but with limited sample size. [393, 394]

Abbreviations: ALSFRS-R, ALS functional rating scale revised; est, estimated; NMJ, neuromuscular junction

Future Directions, Gaps, and Needs

Currently, much of ALS care focuses on symptomatic management using medications described above. Very few drugs are approved to slow disease progression in ALS. Ongoing research is needed to clarify the complex and interacting pathophysiological mechanisms at play, as well as to identify upstream triggers for disease (such as environmental exposures). Of note, many investigational agents have previously been found to be ineffective to slow ALS progression, highlighting the potential need to target multiple pathophysiologic mechanisms simultaneously [205].

Additionally, it is important to optimize strategies to bring new disease-modifying drugs to market. Improvements in drug development, clinical trials, and outcome measures can help bring drugs to market faster. Important strategies to accelerate general drug development include discovering new ALS biomarkers that can be used as clinical trial endpoints; enhancing data sharing between trials; and performing natural history studies in diverse ALS populations, which may be able to serve as controls for future trials [317]. Separately, research to make ALS a more livable disease is also an important future need [318]. Specifically pertaining to clinical trials, the use of platform trials, which use a single control group for multiple interventions, can help streamline evaluation of multiple investigational agents [53]. Finally, artificial intelligence (AI) is starting to be used to optimize multiple stages of drug development.

Artificial Intelligence in Drug Development

AI is being used more frequently in the drug development pipeline in ALS and beyond. Use of AI can be particularly beneficial with rare diseases or targeted therapies, where profitability of a clinical trial is less certain [319]. AI can be implemented in nearly all stages of drug development, including drug screening, biomarker discovery, clinical trials, and data analysis [320].

Preclinically, AI and machine learning can be used to discover new biomarkers and molecular underpinnings in ALS subtypes [321, 322]. In drug development, it can be used in high throughput drug screening. This technique uses computational biology to screen up to millions of potential compounds for predicted effect on target binding, drug metabolism, and central nervous system penetrance. This reduces the need for resource-intensive animal modeling [197].

Within clinical trials, AI can be used to create a synthetic control arm, eliminating the need for a placebo group [323]. AI has been used to stratify patients with ALS by clinical subgroup and to predict disease progression [324, 325]. Regarding recruitment, AI can comb through electronic medical record data to find patients meeting inclusion criteria, summarize eligibility criteria [323], and disseminate information regarding trials to the broader public, improving fair access. In data analysis, AI can determine a heterogeneity effect on outcome measures (e.g., quickly progressing subgroup) or impute missing data [319]. AI can continue to analyze post-market surveillance data to ensure safety and efficacy of approved drugs.

Limitations of AI in ALS drug development currently include lack of high-quality training data for machine learning models and lack of a global regulatory framework [319, 320].

Conclusions

The pharmacologic management of ALS continues to evolve on the basis of approved therapies, symptomatic management, and ongoing clinical trials. Given the incurable nature of ALS, there is incredible hope that new, effective, and clinically meaningful therapeutics are on the horizon. Unfortunately, data showing a disease-modifying effect for several recently approved drugs have not been replicated in larger trials. Nonetheless, ALS therapeutic development is highly active with a large number of ongoing clinical trials. Advancements in ALS outcomes, ALS clinical trials, and drug discovery have the potential to speed drug development for this fatal disease.

Acknowledgements

The authors thank Dr. Emily J. Koubek for expert editorial support and generation of figures.

Declarations

Funding

This work was funded by the National Institutes of Health/National Institute of Neurological Disorders and Stroke (R01NS120926 and R01NS127188 to S.A.G.), the Centers for Disease Control and Prevention/Agency for Toxic Substances and Disease Registry (R01TS000344 and R01TS000327 to S.A.G.), and James and Margaret Hiller and Linda and Eric Novak (S.A.G.).

Conflicts of Interest

S.A.G. is listed as inventor on a patent, issue number US10660895, held by University of Michigan titled “Methods for Treating Amyotrophic Lateral Sclerosis” that targets immune pathways for use in ALS therapeutics. Scientific consulting for Evidera. S.E.Q. and K.H.Q. have no relevant financial or non-financial interests to disclose.

Ethics Approval

Not applicable.

Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Data availability

Not applicable.

Code availability

Not applicable.

Author contributions

S.A.G. conceived the idea for the article. S.E.Q. and K.H.Q. performed the literature search, and all authors drafted, reviewed, and critically revised the final manuscript.

References

  • 1.Feldman EL, et al. Amyotrophic lateral sclerosis. Lancet. 2022;400(10360):1363–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Vignaroli F, et al. The need for biomarkers in the ALS-FTD spectrum: a clinical point of view on the role of proteomics. Proteomes. 2023. 10.3390/proteomes11010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Larson TC, et al. Causes of death among United States decedents with ALS: an eye toward delaying mortality. Ann Clin Transl Neurol. 2023;10(5):757–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mehta P, et al. Prevalence of amyotrophic lateral sclerosis in the United States. Amyotroph Lateral Scler Frontotemporal Degener. 2018;2023:1–7. [DOI] [PubMed] [Google Scholar]
  • 5.Mitsumoto H, Kasarskis EJ, Simmons Z. Hastening the diagnosis of amyotrophic lateral sclerosis. Neurology. 2022;99(2):60–8. [DOI] [PubMed] [Google Scholar]
  • 6.Ryan M, et al. Lifetime risk and heritability of amyotrophic lateral sclerosis. JAMA Neurol. 2019;76(11):1367–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Goutman SA, et al. Emerging insights into the complex genetics and pathophysiology of amyotrophic lateral sclerosis. Lancet Neurol. 2022;21(5):465–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mead RJ, et al. Amyotrophic lateral sclerosis: a neurodegenerative disorder poised for successful therapeutic translation. Nat Rev Drug Discov. 2023;22(3):185–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Umoh ME, et al. A proteomic network approach across the ALS-FTD disease spectrum resolves clinical phenotypes and genetic vulnerability in human brain. EMBO Mol Med. 2018;10(1):48–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Benatar M, Robertson J, Andersen PM. Amyotrophic lateral sclerosis caused by SOD1 variants: from genetic discovery to disease prevention. Lancet Neurol. 2025;24(1):77–86. [DOI] [PubMed] [Google Scholar]
  • 11.Marrone L, et al. Fus pathology in ALS is linked to alterations in multiple ALS-associated proteins and rescued by drugs stimulating autophagy. Acta Neuropathol. 2019;138(1):67–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Blokhuis AM, et al. Protein aggregation in amyotrophic lateral sclerosis. Acta Neuropathol. 2013;125(6):777–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Adams J, Lee M, Peng W. Critical review of complementary and alternative medicine use in amyotrophic lateral sclerosis: prevalence and users’ profile, decision-making, information seeking, and disclosure in the face of a lack of efficacy. Neurodegener Dis. 2018;18(4):225–32. [DOI] [PubMed] [Google Scholar]
  • 14.Goutman SA, et al. ALS identified: two-year findings from a sponsored ALS genetic testing program. Ann Clin Transl Neurol. 2024;11(8):2201–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dharmadasa T, et al. Genetic testing in motor neurone disease. Pract Neurol. 2022;22(2):107–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bosco DA, et al. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat Neurosci. 2010;13(11):1396–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang W, et al. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther. 2023;8(1): 267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Smith RA. Antisense oligonucleotide therapy for neurodegenerative disease. J Clin Invest. 2006;116(8):2290–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ludolph AC, et al. Guidelines for preclinical animal research in ALS/MND: a consensus meeting. Amyotroph Lateral Scler. 2010;11(1–2):38–45. [DOI] [PubMed] [Google Scholar]
  • 20.Babic Leko M, et al. Molecular mechanisms of neurodegeneration related to C9orf72 hexanucleotide repeat expansion. Behav Neurol. 2019;2019:2909168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hao Z, et al. Role of the C9ORF72 gene in the pathogenesis of amyotrophic lateral sclerosis and frontotemporal dementia. Neurosci Bull. 2020;36(9):1057–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ruffo P, Traynor BJ, Conforti FL. Advancements in genetic research and RNA therapy strategies for amyotrophic lateral sclerosis (ALS): current progress and future prospects. J Neurol. 2025;272(3):233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moens TG, et al. Amyotrophic lateral sclerosis caused by FUS mutations: advances with broad implications. Lancet Neurol. 2025;24(2):166–78. [DOI] [PubMed] [Google Scholar]
  • 24.Pozzi L, et al. TBK1 mutations in Italian patients with amyotrophic lateral sclerosis: genetic and functional characterisation. J Neurol Neurosurg Psychiatry. 2017;88(10):869–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pensato V, et al. Exploring NEK1 genetic variability in Italian amyotrophic lateral sclerosis patients. J Neurol. 2025;272(7):469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Riva N, et al. NEK1 variants in a cohort of Italian patients with amyotrophic lateral sclerosis. Front Neurosci. 2022;16: 833051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Amyotrophic lateral sclerosis: developing drugs for treatment guidance for industry. Food and Drug Administration. 2019.
  • 28.van Eijk RPA, et al. Critical design considerations for time-to-event endpoints in amyotrophic lateral sclerosis clinical trials. J Neurol Neurosurg Psychiatry. 2019;90(12):1331–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Marin B, et al. Non-self-sufficiency as a primary outcome measure in ALS trials. Amyotroph Lateral Scler Frontotemporal Degener. 2015;17(1–2):77–84. [DOI] [PubMed] [Google Scholar]
  • 30.Cedarbaum JM, 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;169(1–2):13–21. [DOI] [PubMed] [Google Scholar]
  • 31.Atassi N, et al. The PRO-ACT database: design, initial analyses, and predictive features. Neurology. 2014;83(19):1719–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Labra J, et al. Rate of disease progression: a prognostic biomarker in ALS. J Neurol Neurosurg Psychiatry. 2016;87(6):628–32. [DOI] [PubMed] [Google Scholar]
  • 33.Kimura F, et al. Progression rate of ALSFRS-R at time of diagnosis predicts survival time in ALS. Neurology. 2006;66(2):265–7. [DOI] [PubMed] [Google Scholar]
  • 34.Castrillo-Viguera C, et al. Clinical significance in the change of decline in ALSFRS-R. Amyotroph Lateral Scler. 2010;11(1–2):178–80. [DOI] [PubMed] [Google Scholar]
  • 35.Fournier CN, et al. Development and validation of the Rasch-built overall Amyotrophic Lateral Sclerosis Disability Scale (ROADS). JAMA Neurol. 2020;77(4):480–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fournier CN, James V, Glass JD. Clinically meaningful change: evaluation of the Rasch-built Overall Amyotrophic Lateral Sclerosis Disability Scale (ROADS) and the ALSFRS-R. Amyotroph Lateral Scler Frontotemporal Degener. 2023;24(3–4):311–6. [DOI] [PubMed] [Google Scholar]
  • 37.Genge A, et al. The ALSFRS-R summit: a global call to action on the use of the ALSFRS-R in ALS clinical trials. Amyotroph Lateral Scler Frontotemporal Degener. 2024;25(3–4):382–7. [DOI] [PubMed] [Google Scholar]
  • 38.Franchignoni F, et al. A further rasch study confirms that ALSFRS-R does not conform to fundamental measurement requirements. Amyotroph Lateral Scler Frontotemporal Degener. 2015;16(5–6):331–7. [DOI] [PubMed] [Google Scholar]
  • 39.Franchignoni F, et al. Evidence of multidimensionality in the ALSFRS-R scale: a critical appraisal on its measurement properties using Rasch analysis. J Neurol Neurosurg Psychiatry. 2013;84(12):1340–5. [DOI] [PubMed] [Google Scholar]
  • 40.Traynor BJ, et al. Functional outcome measures as clinical trial endpoints in ALS. Neurology. 2004;63(10):1933–5. [DOI] [PubMed] [Google Scholar]
  • 41.Czaplinski A, Yen AA, Appel SH. Forced vital capacity (FVC) as an indicator of survival and disease progression in an ALS clinic population. J Neurol Neurosurg Psychiatry. 2006;77(3):390–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Allen SC, et al. Performing slow vital capacity in older people with and without cognitive impairment–is it useful? Age Ageing. 2010;39(5):588–91. [DOI] [PubMed] [Google Scholar]
  • 43.Schmidt EP, et al. Pulmonary predictors of survival in amyotrophic lateral sclerosis: use in clinical trial design. Muscle Nerve. 2006;33(1):127–32. [DOI] [PubMed] [Google Scholar]
  • 44.Chhabra SK. Forced vital capacity, slow vital capacity, or inspiratory vital capacity: which is the best measure of vital capacity? J Asthma. 1998;35(4):361–5. [DOI] [PubMed] [Google Scholar]
  • 45.Fiz JA, et al. Spirometry and maximal respiratory pressures in patients with facial paralysis. Chest. 1993;103(1):170–3. [DOI] [PubMed] [Google Scholar]
  • 46.Andres PL, et al. Quantitative assessment of neuromuscular deficit in ALS. Neurol Clin. 1987;5(1):125–41. [PubMed] [Google Scholar]
  • 47.Shefner JM, et al. Quantitative strength testing in ALS clinical trials. Neurology. 2016;87(6):617–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Andres PL, et al. Fixed dynamometry is more sensitive than vital capacity or ALS rating scale. Muscle Nerve. 2017;56(4):710–5. [DOI] [PubMed] [Google Scholar]
  • 49.Berry JD, et al. The Combined Assessment of Function and Survival (CAFS): a new endpoint for ALS clinical trials. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(3):162–8. [DOI] [PubMed] [Google Scholar]
  • 50.Finkelstein DM, Schoenfeld DA. Combining mortality and longitudinal measures in clinical trials. Stat Med. 1999;18(11):1341–54. [DOI] [PubMed] [Google Scholar]
  • 51.Wittkop L, et al. Methodological issues in the use of composite endpoints in clinical trials: examples from the HIV field. Clin Trials. 2010;7(1):19–35. [DOI] [PubMed] [Google Scholar]
  • 52.Murdock BJ, et al. Peripheral immune profiles predict ALS progression in an age- and sex-dependent manner. Neurol Neuroimmunol Neuroinflamm. 2024;11(3): e200241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Quintana M, et al. Design and statistical innovations in a platform trial for amyotrophic lateral sclerosis. Ann Neurol. 2023;94(3):547–60. [DOI] [PubMed] [Google Scholar]
  • 54.21st Century Cures Act. Public Law 114th United States Congress. 2016.
  • 55.Broderick JE, et al. Advances in patient-reported outcomes: the NIH PROMIS((R)) measures. EGEMS (Wash DC). 2013;1(1):1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gershon RC, et al. Neuro-QOL: quality of life item banks for adults with neurological disorders: item development and calibrations based upon clinical and general population testing. Qual Life Res. 2012;21(3):475–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.De Marchi F, et al. Patient reported outcome measures (PROMs) in amyotrophic lateral sclerosis. J Neurol. 2020;267(6):1754–9. [DOI] [PubMed] [Google Scholar]
  • 58.Simmons Z, et al. The ALSSQOL: balancing physical and nonphysical factors in assessing quality of life in ALS. Neurology. 2006;67(9):1659–64. [DOI] [PubMed] [Google Scholar]
  • 59.Felgoise SH, et al. Amyotrophic lateral sclerosis-specific quality of life-short form (ALSSQOL-SF): a brief, reliable, and valid version of the ALSSQOL-R. Muscle Nerve. 2018;58(5):646–54. [DOI] [PubMed] [Google Scholar]
  • 60.Jenkinson C, et al. Development and validation of a short measure of health status for individuals with amyotrophic lateral sclerosis/motor neurone disease: the ALSAQ-40. J Neurol. 1999;246(Suppl 3):III16–21. [DOI] [PubMed] [Google Scholar]
  • 61.Smith RA, et al. Assessment of bulbar function in amyotrophic lateral sclerosis: validation of a self-report scale (Center for Neurologic Study Bulbar Function Scale). Eur J Neurol. 2018;25(7):907-e66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hartmaier SL, et al. Qualitative measures that assess functional disability and quality of life in ALS. Health Qual Life Outcomes. 2022;20(1):12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ahn SW, et al. Reproducibility of the motor unit number index (MUNIX) in normal controls and amyotrophic lateral sclerosis patients. Muscle Nerve. 2010;42(5):808–13. [DOI] [PubMed] [Google Scholar]
  • 64.Higashihara M, et al. Reproducibility of motor unit number index and multiple point stimulation motor unit number estimation in controls. Muscle Nerve. 2018;58(5):660–4. [DOI] [PubMed] [Google Scholar]
  • 65.Nandedkar SD, Barkhaus PE, Stalberg EV. Motor unit number index (MUNIX): principle, method, and findings in healthy subjects and in patients with motor neuron disease. Muscle Nerve. 2010;42(5):798–807. [DOI] [PubMed] [Google Scholar]
  • 66.Nandedkar SD, Barkhaus PE, Stalberg EV. Reproducibility of MUNIX in patients with amyotrophic lateral sclerosis. Muscle Nerve. 2011;44(6):919–22. [DOI] [PubMed] [Google Scholar]
  • 67.Boekestein WA, et al. Motor unit number index (MUNIX) versus motor unit number estimation (MUNE): a direct comparison in a longitudinal study of ALS patients. Clin Neurophysiol. 2012;123(8):1644–9. [DOI] [PubMed] [Google Scholar]
  • 68.Furtula J, et al. MUNIX and incremental stimulation MUNE in ALS patients and control subjects. Clin Neurophysiol. 2013;124(3):610–8. [DOI] [PubMed] [Google Scholar]
  • 69.Rutkove SB, et al. Electrical impedance myography as a biomarker to assess ALS progression. Amyotroph Lateral Scler. 2012;13(5):439–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rutkove SB, et al. Electrical impedance myography correlates with standard measures of ALS severity. Muscle Nerve. 2014;49(3):441–3. [DOI] [PubMed] [Google Scholar]
  • 71.Vucic S, et al. Utility of threshold tracking transcranial magnetic stimulation in ALS. Clin Neurophysiol Pract. 2018;3:164–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wainger BJ, et al. Effect of ezogabine on cortical and spinal motor neuron excitability in amyotrophic lateral sclerosis: a randomized clinical trial. JAMA Neurol. 2021;78(2):186–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Weiss MD, et al. Effects of mexiletine on hyperexcitability in sporadic amyotrophic lateral sclerosis: preliminary findings from a small phase II randomized controlled trial. Muscle Nerve. 2021;63(3):371–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Paganoni S, Cudkowicz M, Berry JD. Outcome measures in amyotrophic lateral sclerosis clinical trials. Clin Investig (Lond). 2014;4(7):605–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ahmed N, Baker MR, Bashford J. The landscape of neurophysiological outcome measures in ALS interventional trials: a systematic review. Clin Neurophysiol. 2022;137:132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.van den Bos MAJ, et al. Pathophysiology and diagnosis of ALS: insights from advances in neurophysiological techniques. Int J Mol Sci. 2019. 10.3390/ijms20112818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kiernan MC, et al. Measurement of axonal excitability: consensus guidelines. Clin Neurophysiol. 2020;131(1):308–23. [DOI] [PubMed] [Google Scholar]
  • 78.Feneberg E, et al. Multicenter evaluation of neurofilaments in early symptom onset amyotrophic lateral sclerosis. Neurology. 2018;90(1):e22–30. [DOI] [PubMed] [Google Scholar]
  • 79.Lu CH, et al. Neurofilament light chain: a prognostic biomarker in amyotrophic lateral sclerosis. Neurology. 2015;84(22):2247–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Benatar M, Wuu J, Turner MR. Neurofilament light chain in drug development for amyotrophic lateral sclerosis: a critical appraisal. Brain. 2023;146(7):2711–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.FDA approves treatment of amyotrophic lateral sclerosis associated with a mutation in the SOD1 gene. Food and Drug Administration. 2023.
  • 82.Gaetani L, et al. Neurofilament light chain as a biomarker in neurological disorders. J Neurol Neurosurg Psychiatry. 2019;90(8):870–81. [DOI] [PubMed] [Google Scholar]
  • 83.Lanznaster D, et al. Metabolomics biomarkers: a strategy toward therapeutics improvement in ALS. Front Neurol. 2018;9: 1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Goutman SA, et al. Recent advances in the diagnosis and prognosis of amyotrophic lateral sclerosis. Lancet Neurol. 2022;21(5):480–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Irwin KE, et al. Fluid biomarkers for amyotrophic lateral sclerosis: a review. Mol Neurodegener. 2024;19(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.van Eijk RP, et al. Comparing methods to combine functional loss and mortality in clinical trials for amyotrophic lateral sclerosis. Clin Epidemiol. 2018;10:333–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Neel DV, et al. Multicenter expanded access program for access to investigational products for amyotrophic lateral sclerosis. Muscle Nerve. 2024;70(2):232–9. [DOI] [PubMed] [Google Scholar]
  • 88.Tzeplaeff L, et al. Unraveling the heterogeneity of ALS-a call to redefine patient stratification for better outcomes in clinical trials. Cells. 2024. 10.3390/cells13050452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Berry JD, et al. Improved stratification of ALS clinical trials using predicted survival. Ann Clin Transl Neurol. 2018;5(4):474–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Van den Berg JP, et al. Multidisciplinary ALS care improves quality of life in patients with ALS. Neurology. 2005;65(8):1264–7. [DOI] [PubMed] [Google Scholar]
  • 91.Rooney J, et al. A multidisciplinary clinic approach improves survival in ALS: a comparative study of ALS in Ireland and Northern Ireland. J Neurol Neurosurg Psychiatry. 2015;86(5):496–501. [DOI] [PubMed] [Google Scholar]
  • 92.Paipa AJ, et al. Survival benefit of multidisciplinary care in amyotrophic lateral sclerosis in Spain: association with noninvasive mechanical ventilation. J Multidiscip Healthc. 2019;12:465–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sulistyo A, et al. Enteral tube feeding for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2023;8(8): CD004030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Conroy E et al. IMPACT-ALS: summary of results from a European survey of people living with ALS. Amyotroph Lateral Scler Frontotemporal Degener, 2023: p. 1-10. [DOI] [PubMed]
  • 95.Simmons Z. Patient-perceived outcomes and quality of life in ALS. Neurotherapeutics. 2015;12(2):394–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bublitz SK et al. Thick mucus in ALS: a mixed-method study on associated factors and its impact on quality of life of patients and caregivers. Brain Sci, 2022;12(2):252. [DOI] [PMC free article] [PubMed]
  • 97.Van Damme P, et al. European Academy of Neurology (EAN) guideline on the management of amyotrophic lateral sclerosis in collaboration with European Reference Network for Neuromuscular Diseases (ERN EURO-NMD). Eur J Neurol. 2024;31(6): e16264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Miller RG, 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;73(15):1227–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Miller RG, 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(15):1218–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Shoesmith C, et al. Canadian best practice recommendations for the management of amyotrophic lateral sclerosis. CMAJ. 2020;192(46):E1453–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Boostani R, et al. Iranian clinical practice guideline for amyotrophic lateral sclerosis. Front Neurol. 2023;14:1154579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bedlack R, Hardiman O. ALSUntangled (ALSU): a scientific approach to off-label treatment options for people with ALS using tweets and twitters. Amyotroph Lateral Scler. 2009;10(3):129–30. [DOI] [PubMed] [Google Scholar]
  • 103.ALS Untangled. [cited 2025]; Available from: https://www.alsuntangled.com/.
  • 104.Kwiecinski H, et al. The effect of selegiline and vitamin E in the treatment of ALS: an open randomized clinical trials. Neurol Neurochir Pol. 2001;35(1 Suppl):101–6. [PubMed] [Google Scholar]
  • 105.Orrell RW, Lane RJ, Ross M. A systematic review of antioxidant treatment for amyotrophic lateral sclerosis/motor neuron disease. Amyotroph Lateral Scler. 2008;9(4):195–211. [DOI] [PubMed] [Google Scholar]
  • 106.Graf M, et al. High dose vitamin E therapy in amyotrophic lateral sclerosis as add-on therapy to riluzole: results of a placebo-controlled double-blind study. J Neural Transm (Vienna). 2005;112(5):649–60. [DOI] [PubMed] [Google Scholar]
  • 107.Stanger MJ, et al. Anticoagulant activity of select dietary supplements. Nutr Rev. 2012;70(2):107–17. [DOI] [PubMed] [Google Scholar]
  • 108.Klein EA, et al. Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2011;306(14):1549–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Mizoule J, et al. 2-Amino-6-trifluoromethoxy benzothiazole, a possible antagonist of excitatory amino acid neurotransmission–I. Anticonvulsant properties. Neuropharmacology. 1985;24(8):767–73. [DOI] [PubMed] [Google Scholar]
  • 110.Cheah BC, et al. Riluzole, neuroprotection and amyotrophic lateral sclerosis. Curr Med Chem. 2010;17(18):1942–199. [DOI] [PubMed] [Google Scholar]
  • 111.Lamanauskas N, Nistri A. Riluzole blocks persistent Na+ and Ca2+ currents and modulates release of glutamate via presynaptic NMDA receptors on neonatal rat hypoglossal motoneurons in vitro. Eur J Neurosci. 2008;27(10):2501–14. [DOI] [PubMed] [Google Scholar]
  • 112.Mantz J, et al. Riluzole, a novel antiglutamate, blocks GABA uptake by striatal synaptosomes. Eur J Pharmacol. 1994;257(1–2):R7-8. [DOI] [PubMed] [Google Scholar]
  • 113.Bensimon G, Lacomblez L, Meininger V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. N Engl J Med. 1994;330(9):585–91. [DOI] [PubMed] [Google Scholar]
  • 114.Miller RG, et al. Clinical trials of riluzole in patients with ALS ALS/Riluzole Study Group-II. Neurology. 1996;47(4 Suppl 2):S86-90 (discussion S90-2). [DOI] [PubMed] [Google Scholar]
  • 115.Hinchcliffe M, Smith A. Riluzole: real-world evidence supports significant extension of median survival times in patients with amyotrophic lateral sclerosis. Degener Neurol Neuromuscul Dis. 2017;7:61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Miller RG, Mitchell JD, Moore DH. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst Rev. 2012;2012(3): CD001447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Bensimon G, et al. A study of riluzole in the treatment of advanced stage or elderly patients with amyotrophic lateral sclerosis. J Neurol. 2002;249(5):609–15. [DOI] [PubMed] [Google Scholar]
  • 118.Traynor BJ, et al. An outcome study of riluzole in amyotrophic lateral sclerosis–a population-based study in Ireland, 1996–2000. J Neurol. 2003;250(4):473–9. [DOI] [PubMed] [Google Scholar]
  • 119.Turner MR, et al. Prognostic modelling of therapeutic interventions in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002;3(1):15–21. [DOI] [PubMed] [Google Scholar]
  • 120.Andrews JA, et al. Real-world evidence of riluzole effectiveness in treating amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2020;21(7–8):509–18. [DOI] [PubMed] [Google Scholar]
  • 121.Thakore NJ, et al. Early initiation of riluzole may improve absolute survival in amyotrophic lateral sclerosis. Muscle Nerve. 2022;66(6):702–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zoccolella S, et al. Riluzole and amyotrophic lateral sclerosis survival: a population-based study in southern Italy. Eur J Neurol. 2007;14(3):262–8. [DOI] [PubMed] [Google Scholar]
  • 123.Riviere M, et al. An analysis of extended survival in patients with amyotrophic lateral sclerosis treated with riluzole. Arch Neurol. 1998;55(4):526–8. [DOI] [PubMed] [Google Scholar]
  • 124.Thakore NJ, et al. Stage-specific riluzole effect in amyotrophic lateral sclerosis: a retrospective study. Amyotroph Lateral Scler Frontotemporal Degener. 2020;21(1–2):140–3. [DOI] [PubMed] [Google Scholar]
  • 125.Casiraghi A et al. Crushing riluzole tablets: evaluation of loss of powder and active principle in a home-simulation experiment. Amyotroph Lateral Scler Frontotemporal Degener, 2023: p. 1-7. [DOI] [PubMed]
  • 126.Dyer AM, Smith A. Riluzole 5 mg/mL oral suspension: for optimized drug delivery in amyotrophic lateral sclerosis. Drug Des Devel Ther. 2017;11:59–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tiglutik: Prescribing Information. Food and Drug Administration. 2018.
  • 128.Exservan: Prescribing Information. Food and Drug Administration. 2019.
  • 129.Mitsubishi Tanabe Pharma America to discontinue Exservan (riluzole). 2024: Mitsubishi Tanabe Pharma America.
  • 130.Brown CA, et al. Estimated prevalence and incidence of amyotrophic lateral sclerosis and SOD1 and C9orf72 genetic variants. Neuroepidemiology. 2021;55(5):342–53. [DOI] [PubMed] [Google Scholar]
  • 131.Miller T, et al. Phase 1–2 Trial of antisense oligonucleotide tofersen for SOD1 ALS. N Engl J Med. 2020;383(2):109–19. [DOI] [PubMed] [Google Scholar]
  • 132.Miller TM, et al. Trial of antisense oligonucleotide tofersen for SOD1 ALS. N Engl J Med. 2022;387(12):1099–110. [DOI] [PubMed] [Google Scholar]
  • 133.Tofersen/Qalsody. 2025: International Alliance of ALS/MND Associations. https://www.als-mnd.org/support-for-pals-cals/approved-drugs/tofersen-qalsody/.
  • 134.Health Technology assessment report: Qalsody (tofersen). 2025: Joint Nordic HTA-Bodies.
  • 135.Edaravone Acute Infarction Study, G., Effect of a novel free radical scavenger, edaravone (MCI-186), on acute brain infarction. Randomized, placebo-controlled, double-blind study at multicenters. Cerebrovasc Dis. 2003;15(3):222–9. [DOI] [PubMed]
  • 136.Tsukamoto Y, et al. Effect of edaravone on the estimated glomerular filtration rate in patients with acute ischemic stroke and chronic kidney disease. J Stroke Cerebrovasc Dis. 2011;20(2):111–6. [DOI] [PubMed] [Google Scholar]
  • 137.Zhao K, et al. Edaravone for acute ischemic stroke: a systematic review and meta-analysis. Clin Ther. 2022;44(12):e29–38. [DOI] [PubMed] [Google Scholar]
  • 138.Witzel S, et al. Safety and effectiveness of long-term intravenous administration of edaravone for treatment of patients with amyotrophic lateral sclerosis. JAMA Neurol. 2022;79(2):121–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kalin A, et al. A safety analysis of edaravone (MCI-186) during the first six cycles (24 weeks) of amyotrophic lateral sclerosis (ALS) therapy from the double-blind period in three randomized, placebo-controlled studies. Amyotroph Lateral Scler Frontotemporal Degener. 2017;18(sup1):71–9. [DOI] [PubMed] [Google Scholar]
  • 140.Writing G and A.L.S.S.G. Edaravone. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2017;16(7): 505–12. [DOI] [PubMed]
  • 141.Shimizu H, et al. Bioequivalence study of oral suspension and intravenous formulation of edaravone in healthy adult subjects. Clin Pharmacol Drug Dev. 2021;10(10):1188–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Shimizu H, et al. Pharmacokinetics of edaravone oral suspension in patients with amyotrophic lateral sclerosis. Clin Ther. 2023;45(12):1251–8. [DOI] [PubMed] [Google Scholar]
  • 143.Genge A, et al. Oral edaravone demonstrated a favorable safety profile in patients with amyotrophic lateral sclerosis after 48 weeks of treatment. Muscle Nerve. 2023;67(2):124–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.FDA approves oral form for the treatment of adults with amyotrophic lateral sclerosis (ALS). Food and Drug Administration. 2022.
  • 145.Mitsubishi Tanabe Pharma America presents final results from the global, phase 3b MT-1186-A02 post-marketing study of radicava (edaravone) in ALS. 2024: Mitsubishi Tanabe Pharma America.
  • 146.Ferrer reports top-line results from Phase III ADORE study in ALS. 2025: Ferrer.
  • 147.Dionisio PA, et al. Amyloid-beta pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol Aging. 2015;36(1):228–40. [DOI] [PubMed] [Google Scholar]
  • 148.Cudkowicz ME, et al. Phase 2 study of sodium phenylbutyrate in ALS. Amyotroph Lateral Scler. 2009;10(2):99–106. [DOI] [PubMed] [Google Scholar]
  • 149.Paganoni S, et al. Trial of sodium phenylbutyrate-taurursodiol for amyotrophic lateral sclerosis. N Engl J Med. 2020;383(10):919–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Mullard A. Amylyx’s ALS therapy secures FDA approval, as regulatory flexibility trumps underwhelming data. Nat Rev Drug Discov. 2022;21(11):786. [DOI] [PubMed] [Google Scholar]
  • 151.Summary review, Relyvrio. Center for Drug Evaluation and Research, Food and Drug Administration. 2022.
  • 152.Paganoni S, et al. Long-term survival of participants in the CENTAUR trial of sodium phenylbutyrate-taurursodiol in amyotrophic lateral sclerosis. Muscle Nerve. 2021;63(1):31–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Paganoni S, et al. Effect of sodium phenylbutyrate/taurursodiol on tracheostomy/ventilation-free survival and hospitalisation in amyotrophic lateral sclerosis: long-term results from the CENTAUR trial. J Neurol Neurosurg Psychiatry. 2022;93(8):871–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Pharmaceuticals A. Amylyx pharmaceuticals announces formal intention to remove relyvrio/albrioza from the market; provides updates on access to therapy, pipeline, corporate restructuring and strategy. 2024.
  • 155.Kikuchi M, et al. Protective effects of methylcobalamin, a vitamin B12 analog, against glutamate-induced neurotoxicity in retinal cell culture. Invest Ophthalmol Vis Sci. 1997;38(5):848–54. [PubMed] [Google Scholar]
  • 156.Hemendinger RA, Armstrong EJ 3rd, Brooks BR. Methyl vitamin B12 but not methylfolate rescues a motor neuron-like cell line from homocysteine-mediated cell death. Toxicol Appl Pharmacol. 2011;251(3):217–25. [DOI] [PubMed] [Google Scholar]
  • 157.Group, A.L. ALSUntangled No. 30: methylcobalamin. Amyotroph Lateral Scler Frontotemporal Degener. 2015;16(7–8):536–9. [DOI] [PubMed]
  • 158.Kaji R, et al. Ultra-high-dose methylcobalamin in amyotrophic lateral sclerosis: a long-term phase II/III randomised controlled study. J Neurol Neurosurg Psychiatry. 2019;90(4):451–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Oki R, et al. Efficacy and safety of ultrahigh-dose methylcobalamin in early-stage amyotrophic lateral sclerosis: a randomized clinical trial. JAMA Neurol. 2022;79(6):575–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Rozebalamin for Injection 25mg (mecobalamin) Approved in Japan for Amyotrophic Lateral Sclerosis. 2024: Eisai Global.
  • 161.Communications P. Abstract Book 35th International Symposium on ALS/MND (Complete printable file). Amyotroph Lateral Scler Frontotemporal Degener. 2024;25(sup1):1–92. [DOI] [PubMed] [Google Scholar]
  • 162.Kim H, et al. Dose-dependent efficacy of ALS-human mesenchymal stem cells transplantation into cisterna magna in SOD1-G93A ALS mice. Neurosci Lett. 2010;468(3):190–4. [DOI] [PubMed] [Google Scholar]
  • 163.Oh KW, et al. Phase I trial of repeated intrathecal autologous bone marrow-derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl Med. 2015;4(6):590–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Oh KW, et al. Repeated intrathecal mesenchymal stem cells for amyotrophic lateral sclerosis. Ann Neurol. 2018;84(3):361–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Nam JY, et al. Long-term survival benefits of intrathecal autologous bone marrow-derived mesenchymal stem cells (Neuronata-R(R): lenzumestrocel) treatment in ALS: propensity-score-matched control, surveillance study. Front Aging Neurosci. 2023;15:1148444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Nam JY, et al. Efficacy and safety of lenzumestrocel (neuronata-R(R) inj) in patients with amyotrophic lateral sclerosis (ALSUMMIT study): study protocol for a multicentre, randomized, double-blind, parallel-group, sham procedure-controlled, phase III trial. Trials. 2022;23(1):415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Han-soo L. Corestem Chemon's stock plunges after failed P3 ALS drug trial. 2024: Korea Biomedical Review.
  • 168.Drugs@FDA: FDA-Approved Drugs. Food and Drug Administration.
  • 169.European Medicines Agency. 2025. https://www.ema.europa.eu/en/medicines.
  • 170.Drug Product Database online query. Government of Canada. [cited 2025]. https://health-products.canada.ca/dpd-bdpp/.
  • 171.Mora JS, et al. Masitinib as an add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: a randomized clinical trial. Amyotroph Lateral Scler Frontotemporal Degener. 2020;21(1–2):5–14. [DOI] [PubMed] [Google Scholar]
  • 172.Salomon-Zimri S, et al. Combination of ciprofloxacin/celecoxib as a novel therapeutic strategy for ALS. Amyotroph Lateral Scler Frontotemporal Degener. 2023;24(3–4):263–71. [DOI] [PubMed] [Google Scholar]
  • 173.Berry JD, et al. NurOwn, phase 2, randomized, clinical trial in patients with ALS: Safety, clinical, and biomarker results. Neurology. 2019;93(24):e2294–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.FDA. Update on Amyotrophic Lateral Sclerosis (ALS) Product Development. 2021. https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/update-amyotrophic-lateral-sclerosis-als-product-development.
  • 175.Calabrese V, et al. Acetylcarnitine induces heme oxygenase in rat astrocytes and protects against oxidative stress: involvement of the transcription factor Nrf2. J Neurosci Res. 2005;79(4):509–21. [DOI] [PubMed] [Google Scholar]
  • 176.Capecchi PL, et al. Carnitines increase plasma levels of adenosine and ATP in humans. Vasc Med. 1997;2(2):77–81. [DOI] [PubMed] [Google Scholar]
  • 177.Nagesh-Babu G, Kumar A, Singh RL. Chronic pretreatment with acetyl-L-carnitine and +/-DL-alpha-lipoic acid protects against acute glutamate-induced neurotoxicity in rat brain by altering mitochondrial function. Neurotox Res. 2011;19(2):319–29. [DOI] [PubMed] [Google Scholar]
  • 178.Grossini E et al. Effects of acetyl-L-carnitine on oxidative stress in amyotrophic lateral sclerosis patients: evaluation on plasma markers and members of the neurovascular unit. Antioxidants (Basel). 2023;12(10). [DOI] [PMC free article] [PubMed]
  • 179.Beghi E, et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for ALS. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(5–6):397–405. [DOI] [PubMed] [Google Scholar]
  • 180.Sassi S, et al. Retrospective observational study on the use of acetyl-L-carnitine in ALS. J Neurol. 2023;270(11):5344–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Ng L, et al. Symptomatic treatments for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2017;1(1): CD011776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Hobson EV, McDermott CJ. Supportive and symptomatic management of amyotrophic lateral sclerosis. Nat Rev Neurol. 2016;12(9):526–38. [DOI] [PubMed] [Google Scholar]
  • 183.Jenkins TM, Hollinger H, McDermott CJ. The evidence for symptomatic treatments in amyotrophic lateral sclerosis. Curr Opin Neurol. 2014;27(5):524–31. [DOI] [PubMed] [Google Scholar]
  • 184.Brooks BR, et al. Treatment of pseudobulbar affect in ALS with dextromethorphan/quinidine: a randomized trial. Neurology. 2004;63(8):1364–70. [DOI] [PubMed] [Google Scholar]
  • 185.Pioro EP, et al. Dextromethorphan plus ultra low-dose quinidine reduces pseudobulbar affect. Ann Neurol. 2010;68(5):693–702. [DOI] [PubMed] [Google Scholar]
  • 186.Smith R, et al. Enhanced bulbar function in amyotrophic lateral sclerosis: the Nuedexta Treatment Trial. Neurotherapeutics. 2017;14(3):762–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.James E, et al. Treatment for sialorrhea (excessive saliva) in people with motor neuron disease/amyotrophic lateral sclerosis. Cochrane Database Syst Rev. 2022;5(5): CD006981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Newall AR, Orser R, Hunt M. The control of oral secretions in bulbar ALS/MND. J Neurol Sci. 1996;139(Suppl):43–4. [DOI] [PubMed] [Google Scholar]
  • 189.Sdravou K, Walshe M, Dagdilelis L. Effects of carbonated liquids on oropharyngeal swallowing measures in people with neurogenic dysphagia. Dysphagia. 2012;27(2):240–50. [DOI] [PubMed] [Google Scholar]
  • 190.Mulheren RW, Kamarunas E, Ludlow CL. Sour taste increases swallowing and prolongs hemodynamic responses in the cortical swallowing network. J Neurophysiol. 2016;116(5):2033–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Bozorgi C, Holleufer C, Wendin K. Saliva secretion and swallowing-the impact of different types of food and drink on subsequent intake. Nutrients. 2020;12(1):256. [DOI] [PMC free article] [PubMed]
  • 192.Caress JB, et al. Natural history of muscle cramps in amyotrophic lateral sclerosis. Muscle Nerve. 2016;53(4):513–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Menakuru SR, et al. Disseminated intravascular coagulation due to drinking tonic water. Cureus. 2021;13(12): e20512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Drug products containing quinine offered over-the-counter (OTC) for the treatment and/or prevention of malaria. Code of Federal Regulations. 1998.
  • 195.Amtmann D, et al. Survey of cannabis use in patients with amyotrophic lateral sclerosis. Am J Hosp Palliat Care. 2004;21(2):95–104. [DOI] [PubMed] [Google Scholar]
  • 196.McClelland S 3rd, et al. Intrathecal baclofen for spasticity-related pain in amyotrophic lateral sclerosis: efficacy and factors associated with pain relief. Muscle Nerve. 2008;37(3):396–8. [DOI] [PubMed] [Google Scholar]
  • 197.McGown A, Stopford MJ. High-throughput drug screens for amyotrophic lateral sclerosis drug discovery. Expert Opin Drug Discov. 2018;13(11):1015–25. [DOI] [PubMed] [Google Scholar]
  • 198.Patten SA, et al. Simple animal models for amyotrophic lateral sclerosis drug discovery. Expert Opin Drug Discov. 2016;11(8):797–804. [DOI] [PubMed] [Google Scholar]
  • 199.Shneider NA, et al. Antisense oligonucleotide jacifusen for FUS-ALS: an investigator-initiated, multicentre, open-label case series. Lancet. 2025;405(10494):2075–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Biogen and Ionis announce topline phase 1 study results of investigational drug in C9orf72 amyotrophic lateral sclerosis. 2022: Biogen.
  • 201.Biogen and Ionis announce topline phase 1/2 study results of investigational drug in amyotrophic lateral sclerosis. 2024: Biogen.
  • 202.Baughn MW, et al. Mechanism of STMN2 cryptic splice-polyadenylation and its correction for TDP-43 proteinopathies. Science. 2023;379(6637):1140–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Cappella M. et al. Gene therapy for ALS-A perspective. Int J Mol Sci, 2019. 20(18):4388. [DOI] [PMC free article] [PubMed]
  • 204.Ito D, Okada K. Rethinking antisense oligonucleotide therapeutics for amyotrophic lateral sclerosis. Ann Clin Transl Neurol. 2024;11(12):3054–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Gittings LM, Sattler R. Recent advances in understanding amyotrophic lateral sclerosis and emerging therapies. Fac Rev. 2020;9:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Seo J, et al. Preferences of patients with amyotrophic lateral sclerosis for intrathecal drug delivery: choosing between an implanted drug-delivery device and therapeutic lumbar puncture. Patient. 2024;17(2):161–77. [DOI] [PubMed] [Google Scholar]
  • 207.Meister G, Tuschl T. Mechanisms of gene silencing by double-stranded RNA. Nature. 2004;431(7006):343–9. [DOI] [PubMed] [Google Scholar]
  • 208.Colella P, Ronzitti G, Mingozzi F. Emerging issues in AAV-mediated in vivo gene therapy. Mol Ther Methods Clin Dev. 2018;8:87–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Naso MF, et al. Adeno-associated virus (AAV) as a vector for gene therapy. BioDrugs. 2017;31(4):317–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Sun L, Li J, Xiao X. Overcoming adeno-associated virus vector size limitation through viral DNA heterodimerization. Nat Med. 2000;6(5):599–602. [DOI] [PubMed] [Google Scholar]
  • 211.Yerbury JJ, Farrawell NE, McAlary L. Proteome homeostasis dysfunction: a unifying principle in ALS pathogenesis. Trends Neurosci. 2020;43(5):274–84. [DOI] [PubMed] [Google Scholar]
  • 212.Perni M, Mannini B. Targeting protein aggregation in ALS. Biomolecules. 2024;14(10):1324. [DOI] [PMC free article] [PubMed]
  • 213.Verma S, et al. Neuromuscular junction dysfunction in amyotrophic lateral sclerosis. Mol Neurobiol. 2022;59(3):1502–27. [DOI] [PubMed] [Google Scholar]
  • 214.Korobeynikov VA, et al. Antisense oligonucleotide silencing of FUS expression as a therapeutic approach in amyotrophic lateral sclerosis. Nat Med. 2022;28(1):104–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Nijssen J, Comley LH, Hedlund E. Motor neuron vulnerability and resistance in amyotrophic lateral sclerosis. Acta Neuropathol. 2017;133(6):863–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Lagier-Tourenne C, Polymenidou M, Cleveland DW. TDP-43 and FUS/TLS: emerging roles in RNA processing and neurodegeneration. Hum Mol Genet. 2010;19(R1):R46-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Polymenidou M, et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat Neurosci. 2011;14(4):459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Roussel BD, et al. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. 2013;12(1):105–18. [DOI] [PubMed] [Google Scholar]
  • 219.Bentmann E, et al. Requirements for stress granule recruitment of fused in sarcoma (FUS) and TAR DNA-binding protein of 43 kDa (TDP-43). J Biol Chem. 2012;287(27):23079–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Valencia M, et al. Neuronal autophagy: characteristic features and roles in neuronal pathophysiology. Biomol Ther (Seoul). 2021;29(6):605–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Lakpa KL, et al. Lysosomal stress response (LSR): physiological importance and pathological relevance. J Neuroimmune Pharmacol. 2021;16(2):219–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Sellier C, et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 2016;35(12):1276–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Hung ST, et al. PIKFYVE inhibition mitigates disease in models of diverse forms of ALS. Cell. 2023;186(4):786–802 (e28). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Marlin E et al. The role and therapeutic potential of the integrated stress response in amyotrophic lateral sclerosis. Int J Mol Sci. 2022;23(14):7823. [DOI] [PMC free article] [PubMed]
  • 225.Pakos-Zebrucka K, et al. The integrated stress response. EMBO Rep. 2016;17(10):1374–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Ron D. Translational control in the endoplasmic reticulum stress response. J Clin Invest. 2002;110(10):1383–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Lomakin IB, Steitz TA. The initiation of mammalian protein synthesis and mRNA scanning mechanism. Nature. 2013;500(7462):307–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Quiros PM, et al. Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals. J Cell Biol. 2017;216(7):2027–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Novoa I, et al. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol. 2001;153(5):1011–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Green KM, et al. RAN translation at C9orf72-associated repeat expansions is selectively enhanced by the integrated stress response. Nat Commun. 2017;8(1):2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Tsaytler P, et al. Selective inhibition of a regulatory subunit of protein phosphatase 1 restores proteostasis. Science. 2011;332(6025):91–4. [DOI] [PubMed] [Google Scholar]
  • 232.Ilieva EV, et al. Oxidative and endoplasmic reticulum stress interplay in sporadic amyotrophic lateral sclerosis. Brain. 2007;130(Pt 12):3111–23. [DOI] [PubMed] [Google Scholar]
  • 233.McCauley ME, Baloh RH. Inflammation in ALS/FTD pathogenesis. Acta Neuropathol. 2019;137(5):715–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Mifflin L et al. A RIPK1-regulated inflammatory microglial state in amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A. 2021;118(13):e2025102118. [DOI] [PMC free article] [PubMed]
  • 235.Yuan J, Amin P, Ofengeim D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 2019;20(1):19–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Olesen MN et al. Inflammatory profiles relate to survival in subtypes of amyotrophic lateral sclerosis. Neurol Neuroimmunol Neuroinflamm. 2020;7(3):e697. [DOI] [PMC free article] [PubMed]
  • 237.Kallstig E, McCabe BD and Schneider BL. The links between ALS and NF-kappaB. Int J Mol Sci. 2021;22(8):3875. [DOI] [PMC free article] [PubMed]
  • 238.Frakes AE, et al. Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81(5):1009–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Fomin V et al. The C9ORF72 gene, implicated in amyotrophic lateral sclerosis and frontotemporal dementia, encodes a protein that functions in control of endothelin and glutamate signaling. Mol Cell Biol. 2018;38(22):e00155-18. [DOI] [PMC free article] [PubMed]
  • 240.Haidet-Phillips AM, et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat Biotechnol. 2011;29(9):824–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Xue J, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Liao B, et al. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp Neurol. 2012;237(1):147–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Du Y, et al. Increased activation ability of monocytes from ALS patients. Exp Neurol. 2020;328: 113259. [DOI] [PubMed] [Google Scholar]
  • 244.Zurcher NR, et al. Increased in vivo glial activation in patients with amyotrophic lateral sclerosis: assessed with [(11)C]-PBR28. Neuroimage Clin. 2015;7:409–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Yu W, et al. Neuroimmune crosstalk between the peripheral and the central immune system in amyotrophic lateral sclerosis. Front Aging Neurosci. 2022;14: 890958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Pan Y, Nicolazzo JA. Altered blood-brain barrier and blood-spinal cord barrier dynamics in amyotrophic lateral sclerosis: Impact on medication efficacy and safety. Br J Pharmacol. 2022;179(11):2577–88. [DOI] [PubMed] [Google Scholar]
  • 247.Waters S, et al. Blood-spinal cord barrier leakage is independent of motor neuron pathology in ALS. Acta Neuropathol Commun. 2021;9(1):144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Winkler EA, et al. Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 2013;125(1):111–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Pieper C, Pieloch P, Galla HJ. Pericytes support neutrophil transmigration via interleukin-8 across a porcine co-culture model of the blood-brain barrier. Brain Res. 2013;1524:1–11. [DOI] [PubMed] [Google Scholar]
  • 250.Sun Q, et al. Inflammatory cytokine levels in patients with sporadic amyotrophic lateral sclerosis. Neurodegener Dis. 2021;21(3–4):87–92. [DOI] [PubMed] [Google Scholar]
  • 251.Appel SH, Beers DR, Zhao W. Amyotrophic lateral sclerosis is a systemic disease: peripheral contributions to inflammation-mediated neurodegeneration. Curr Opin Neurol. 2021;34(5):765–72. [DOI] [PubMed] [Google Scholar]
  • 252.Henkel JS, et al. Presence of dendritic cells, MCP-1, and activated microglia/macrophages in amyotrophic lateral sclerosis spinal cord tissue. Ann Neurol. 2004;55(2):221–35. [DOI] [PubMed] [Google Scholar]
  • 253.Zhao W, et al. Regulatory T lymphocytes from ALS mice suppress microglia and effector T lymphocytes through different cytokine-mediated mechanisms. Neurobiol Dis. 2012;48(3):418–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Kapellos TS, et al. Human monocyte subsets and phenotypes in major chronic inflammatory diseases. Front Immunol. 2019;10:2035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Zondler L, et al. Peripheral monocytes are functionally altered and invade the CNS in ALS patients. Acta Neuropathol. 2016;132(3):391–411. [DOI] [PubMed] [Google Scholar]
  • 256.Lee JD, et al. Dysregulation of the complement cascade in the hSOD1G93A transgenic mouse model of amyotrophic lateral sclerosis. J Neuroinflammation. 2013;10:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Spataro R, et al. Plasma cortisol level in amyotrophic lateral sclerosis. J Neurol Sci. 2015;358(1–2):282–6. [DOI] [PubMed] [Google Scholar]
  • 258.Monachelli GG, et al. Progesterone and cortisol levels in sporadic amyotrophic lateral sclerosis (sALS): correlation with prognostic factors. Horm Mol Biol Clin Investig. 2011;6(1):167–73. [DOI] [PubMed] [Google Scholar]
  • 259.Meyer M, et al. The selective glucocorticoid receptor modulator cort 113176 reduces neurodegeneration and neuroinflammation in wobbler mice spinal cord. Neuroscience. 2018;384:384–96. [DOI] [PubMed] [Google Scholar]
  • 260.Murdock BJ et al. Amyotrophic lateral sclerosis survival associates with neutrophils in a sex-specific manner. Neurol Neuroimmunol Neuroinflamm. 2021;8(2):e953. [DOI] [PMC free article] [PubMed]
  • 261.Murdock BJ et al. In vitro modeling of natural killer cell cytotoxicity to inform personalized ALS therapeutics. Ann Clin Transl Neurol. 2025;0:1–9. [DOI] [PMC free article] [PubMed]
  • 262.Murdock BJ et al. Early immune system changes in amyotrophic lateral sclerosis correlate with later disease progression. Medicine. 2025;6(8):100673. [DOI] [PMC free article] [PubMed]
  • 263.Murdock BJ et al. NK cells associate with ALS in a sex- and age-dependent manner. JCI Insight 2021;6(11):e147129. [DOI] [PMC free article] [PubMed]
  • 264.Figueroa-Romero C, et al. Tofacitinib suppresses natural killer cells in vitro and in vivo: implications for amyotrophic lateral sclerosis. Front Immunol. 2022;13: 773288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Mc CB. The origin and behavior of mutable loci in maize. Proc Natl Acad Sci U S A. 1950;36(6):344–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Li W, et al. Retroviral elements in pathophysiology and as therapeutic targets for amyotrophic lateral sclerosis. Neurotherapeutics. 2022;19(4):1085–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Li W, et al. Human endogenous retrovirus-K contributes to motor neuron disease. Sci Transl Med. 2015;7(307):307–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Gouel F, et al. Past and future of neurotrophic growth factors therapies in ALS: from single neurotrophic growth factor to stem cells and human platelet lysates. Front Neurol. 2019;10:835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Henriques A, Pitzer C, Schneider A. Neurotrophic growth factors for the treatment of amyotrophic lateral sclerosis: where do we stand? Front Neurosci. 2010;4:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Goutman SA, et al. Stem cell treatments for amyotrophic lateral sclerosis: a critical overview of early phase trials. Expert Opin Investig Drugs. 2019;28(6):525–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Goutman SA, et al. Long-term phase 1/2 intraspinal stem cell transplantation outcomes in ALS. Ann Clin Transl Neurol. 2018;5(6):730–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Lunn JS, Sakowski SA, Feldman EL. Concise review: Stem cell therapies for amyotrophic lateral sclerosis: recent advances and prospects for the future. Stem Cells. 2014;32(5):1099–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Cunha-Oliveira T, et al. Oxidative stress in amyotrophic lateral sclerosis: pathophysiology and opportunities for pharmacological intervention. Oxid Med Cell Longev. 2020;2020:5021694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417(1):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Brand MD. The sites and topology of mitochondrial superoxide production. Exp Gerontol. 2010;45(7–8):466–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Ray RS, Katyal A. Myeloperoxidase: bridging the gap in neurodegeneration. Neurosci Biobehav Rev. 2016;68:611–20. [DOI] [PubMed] [Google Scholar]
  • 277.Khan AA, et al. Biochemical and pathological studies on peroxidases -an updated review. Glob J Health Sci. 2014;6(5):87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Park HR and Yang EJ. Oxidative stress as a therapeutic target in amyotrophic lateral sclerosis: opportunities and limitations. Diagnostics (Basel). 2021;11(9):1546. [DOI] [PMC free article] [PubMed]
  • 279.Agrawal I, et al. Deciphering lipid dysregulation in ALS: from mechanisms to translational medicine. Transl Neurodegener. 2022;11(1):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Yerton M, et al. An expanded access protocol of RT001 in amyotrophic lateral sclerosis-Initial experience with a lipid peroxidation inhibitor. Muscle Nerve. 2022;66(4):421–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Robitaille R. Modulation of synaptic efficacy and synaptic depression by glial cells at the frog neuromuscular junction. Neuron. 1998;21(4):847–55. [DOI] [PubMed] [Google Scholar]
  • 282.Jillapalli D, Shefner JM. Single motor unit variability with threshold stimulation in patients with amyotrophic lateral sclerosis and normal subjects. Muscle Nerve. 2004;30(5):578–84. [DOI] [PubMed] [Google Scholar]
  • 283.Fischer LR, et al. Amyotrophic lateral sclerosis is a distal axonopathy: evidence in mice and man. Exp Neurol. 2004;185(2):232–40. [DOI] [PubMed] [Google Scholar]
  • 284.Xia R, et al. Motor neuron apoptosis and neuromuscular junction perturbation are prominent features in a Drosophila model of Fus-mediated ALS. Mol Neurodegener. 2012;7:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Dzieciolowska S, Drapeau P, Armstrong GAB. Augmented quantal release of acetylcholine at the vertebrate neuromuscular junction following tdp-43 depletion. PLoS One. 2017;12(5): e0177005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Campanari ML, et al. Neuromuscular junction impairment in amyotrophic lateral sclerosis: reassessing the role of acetylcholinesterase. Front Mol Neurosci. 2016;9:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Krus KL, et al. Loss of Stathmin-2, a hallmark of TDP-43-associated ALS, causes motor neuropathy. Cell Rep. 2022;39(13): 111001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Yoshihara T, et al. Ultrastructural and histochemical study of the motor end plates of the intrinsic laryngeal muscles in amyotrophic lateral sclerosis. Ultrastruct Pathol. 1998;22(2):121–6. [DOI] [PubMed] [Google Scholar]
  • 289.Clark JA, et al. Axonal degeneration, distal collateral branching and neuromuscular junction architecture alterations occur prior to symptom onset in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. J Chem Neuroanat. 2016;76(Pt A):35–47. [DOI] [PubMed] [Google Scholar]
  • 290.Cappello V and Francolini M. Neuromuscular junction dismantling in amyotrophic lateral sclerosis. Int J Mol Sci. 2017;18(10):2092. [DOI] [PMC free article] [PubMed]
  • 291.Shefner JM, et al. Skeletal muscle in amyotrophic lateral sclerosis. Brain. 2023;146(11):4425–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Moloney EB, de Winter F, Verhaagen J. ALS as a distal axonopathy: molecular mechanisms affecting neuromuscular junction stability in the presymptomatic stages of the disease. Front Neurosci. 2014;8:252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Trujillo-Estrada L, et al. SPG302 reverses synaptic and cognitive deficits without altering amyloid or tau pathology in a transgenic model of Alzheimer’s disease. Neurotherapeutics. 2021;18(4):2468–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Bouscary A, et al. Ambroxol hydrochloride improves motor functions and extends survival in a mouse model of familial amyotrophic lateral sclerosis. Front Pharmacol. 2019;10:883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Guo K et al. Longitudinal metabolomics in amyotrophic lateral sclerosis implicates impaired lipid metabolism. Ann Neurol. 2025;98(1):19–34. [DOI] [PMC free article] [PubMed]
  • 296.Guo K, et al. Gut microbiome correlates with plasma lipids in amyotrophic lateral sclerosis. Brain. 2024;147(2):665–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Goutman SA, et al. Body mass index associates with amyotrophic lateral sclerosis survival and metabolomic profiles. Muscle Nerve. 2023;67(3):208–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Goutman SA, et al. Metabolomics identifies shared lipid pathways in independent amyotrophic lateral sclerosis cohorts. Brain. 2022;145(12):4425–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Goutman SA, et al. Untargeted metabolomics yields insight into ALS disease mechanisms. J Neurol Neurosurg Psychiatry. 2020;91(12):1329–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Burg T, Van Den Bosch L. Abnormal energy metabolism in ALS: a key player? Curr Opin Neurol. 2023;36(4):338–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Ferri A, Coccurello R. What is “Hyper” in the ALS hypermetabolism? Mediators Inflamm. 2017;2017:7821672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Wills AM, et al. Hypercaloric enteral nutrition in patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet. 2014;383(9934):2065–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Osellame LD, Blacker TS, Duchen MR. Cellular and molecular mechanisms of mitochondrial function. Best Pract Res Clin Endocrinol Metab. 2012;26(6):711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Scaricamazza S, et al. Repurposing of trimetazidine for amyotrophic lateral sclerosis: a study in SOD1(G93A) mice. Br J Pharmacol. 2022;179(8):1732–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Houten SM, et al. The biochemistry and physiology of mitochondrial fatty acid beta-oxidation and its genetic disorders. Annu Rev Physiol. 2016;78:23–44. [DOI] [PubMed] [Google Scholar]
  • 306.Strong MJ. The evidence for altered RNA metabolism in amyotrophic lateral sclerosis (ALS). J Neurol Sci. 2010;288(1–2):1–12. [DOI] [PubMed] [Google Scholar]
  • 307.Corley M, Burns MC, Yeo GW. How RNA-binding proteins interact with RNA: molecules and mechanisms. Mol Cell. 2020;78(1):9–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Butti Z, Patten SA. RNA dysregulation in amyotrophic lateral sclerosis. Front Genet. 2018;9:712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Chou CC, et al. TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nat Neurosci. 2018;21(2):228–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Zhang K, et al. Stress granule assembly disrupts nucleocytoplasmic transport. Cell. 2018;173(4):958–71 (e17). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Arnold FJ et al. Revisiting glutamate excitotoxicity in amyotrophic lateral sclerosis and age-related neurodegeneration. Int J Mol Sci. 2024;25(11):5587. [DOI] [PMC free article] [PubMed]
  • 312.Van Den Bosch L, et al. The role of excitotoxicity in the pathogenesis of amyotrophic lateral sclerosis. Biochim Biophys Acta. 2006;1762(11–12):1068–82. [DOI] [PubMed] [Google Scholar]
  • 313.Wright ML, et al. Potential Role of the gut microbiome in ALS: a systematic review. Biol Res Nurs. 2018;20(5):513–21. [DOI] [PubMed] [Google Scholar]
  • 314.Fang X, et al. Evaluation of the microbial diversity in amyotrophic lateral sclerosis using high-throughput sequencing. Front Microbiol. 2016;7:1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Wu S et al. Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model. Physiol Rep. 2015;3(4): e12356. [DOI] [PMC free article] [PubMed]
  • 316.Zhang YG, et al. Target intestinal microbiota to alleviate disease progression in amyotrophic lateral sclerosis. Clin Ther. 2017;39(2):322–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.In Living with ALS, J. Alper, R.A. English, and A.I. Leshner, Editors. 2024: Washington (DC).
  • 318.Feldman EL, et al. Transforming amyotrophic lateral sclerosis into a liveable disease. Lancet Neurol. 2025;24(2):100–1. [DOI] [PubMed] [Google Scholar]
  • 319.Askin S, et al. Artificial intelligence applied to clinical trials: opportunities and challenges. Health Technol (Berl). 2023;13(2):203–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Zhang K, et al. Artificial intelligence in drug development. Nat Med. 2025;31(1):45–59. [DOI] [PubMed] [Google Scholar]
  • 321.Behler A et al. Diffusion tensor imaging in amyotrophic lateral sclerosis: machine learning for biomarker development. Int J Mol Sci. 2023;24(3):1911. [DOI] [PMC free article] [PubMed]
  • 322.Marriott H, et al. Unsupervised machine learning identifies distinct ALS molecular subtypes in post-mortem motor cortex and blood expression data. Acta Neuropathol Commun. 2023;11(1):208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Liu H, et al. A knowledge base of clinical trial eligibility criteria. J Biomed Inform. 2021;117: 103771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Faghri F, et al. Identifying and predicting amyotrophic lateral sclerosis clinical subgroups: a population-based machine-learning study. Lancet Digit Health. 2022;4(5):e359–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Tavazzi E, et al. Artificial intelligence and statistical methods for stratification and prediction of progression in amyotrophic lateral sclerosis: a systematic review. Artif Intell Med. 2023;142: 102588. [DOI] [PubMed] [Google Scholar]
  • 326.Rabkin JG, et al. Modafinil treatment of fatigue in patients with ALS: a placebo-controlled study. Muscle Nerve. 2009;39(3):297–303. [DOI] [PubMed] [Google Scholar]
  • 327.Bilbao A, Spanagel R. Medical cannabinoids: a pharmacology-based systematic review and meta-analysis for all relevant medical indications. BMC Med. 2022;20(1):259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Nelson K, et al. A phase II study of delta-9-tetrahydrocannabinol for appetite stimulation in cancer-associated anorexia. J Palliat Care. 1994;10(1):14–8. [PubMed] [Google Scholar]
  • 329.Sevriev B, et al. Trehalose: neuroprotective effects and mechanisms—An updated review. NeuroSci. 2024;5(4):429–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Mixed results announced for trehalose in ALS study. 2024: Medical Professionals Reference.
  • 331.Fornai F, et al. Autophagy and amyotrophic lateral sclerosis: the multiple roles of lithium. Autophagy. 2008;4(4):527–30. [DOI] [PubMed] [Google Scholar]
  • 332.Pasquali L, et al. Autophagy, lithium, and amyotrophic lateral sclerosis. Muscle Nerve. 2009;40(2):173–94. [DOI] [PubMed] [Google Scholar]
  • 333.Shi Y et al. Identification and therapeutic rescue of autophagosome and glutamate receptor defects in C9ORF72 and sporadic ALS neurons. JCI Insight. 2019;5(15):e127736. [DOI] [PMC free article] [PubMed]
  • 334.Hasegawa J, Inoki K, Weisman LS. PIKFYVE-dependent regulation of MTORC1 and TFEB. Autophagy Reports. 2022;1(1):247–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Babu S, et al. Apilimod dimesylate in C9orf72 amyotrophic lateral sclerosis: a randomized phase 2a clinical trial. Brain. 2024;147(9):2998–3008. [DOI] [PubMed] [Google Scholar]
  • 336.Genge A, et al. A phase 1, multicenter, open label, single-ascending dose study to evaluate safety, tolerability, and pharmacokinetics of AP-101 in familial and sporadic amyotrophic lateral sclerosis (ALS) (4484). Neurology. 2021;96(15_supplement):4484. [Google Scholar]
  • 337.Martínez-González L et al. Tideglusib, a non-ATP competitive inhibitor of GSK-3β as a drug candidate for the treatment of amyotrophic lateral sclerosis. Int J Mol Sci. 2021;22(16):8975. [DOI] [PMC free article] [PubMed]
  • 338.Imamura K, et al. Safety and tolerability of bosutinib in patients with amyotrophic lateral sclerosis (iDReAM study): a multicentre, open-label, dose-escalation phase 1 trial. EClinicalMedicine. 2022;53: 101707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Borel F, et al. Therapeutic rAAVrh10 mediated SOD1 silencing in adult SOD1(G93A) mice and nonhuman primates. Hum Gene Ther. 2016;27(1):19–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Benefits of repurposed monepantel for ALS sustained for up to 2 years. 2024: ALS News Today.
  • 341.Ractigen announces positive clinical data for RAG-17 in ALS-SOD1 treatment from investigator-initiated trial. 2024: Ractigen Therapeutics.
  • 342.Bos PH, et al. Development of MAP4 kinase inhibitors as motor neuron-protecting agents. Cell Chem Biol. 2019;26(12):1703–15 (e37). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Calico provides update on fosigotifator in HEALEY ALS platform trial. 2025: Calico Life Sciences LLC.
  • 344.DNL343 falls short of primary end point in phase 2/3 HEALEY-ALS platform trial. 2025: Neurology Live.
  • 345.Green KM, et al. RAN translation at C9orf72-associated repeat expansions is selectively enhanced by the integrated stress response. Nat Comm. 2017;8(1):2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.InFlectis BioScience awarded $943,000 grant from the ALS association to advance ongoing ALS clinical trial with the support of IRCCS Carlo Besta Neurological Institute. 2024: Yahoo Finance.
  • 347.Lee JD, et al. Pharmacological inhibition of complement C5a–C5a1 receptor signalling ameliorates disease pathology in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Br J Pharmacol. 2017;174(8):689–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.BK B et al. Overexpression of the Cdk5 inhibitory peptide in motor neurons rescue of amyotrophic lateral sclerosis phenotype in a mouse model. Human Mol Gen. 2019;28(19):3175–87. [DOI] [PMC free article] [PubMed]
  • 349.Kjældgaard A-L, et al. Amyotrophic lateral sclerosis: the complement and inflammatory hypothesis. Mol Immunol. 2018;102:14–25. [DOI] [PubMed] [Google Scholar]
  • 350.Gold J, et al. Safety and tolerability of Triumeq in amyotrophic lateral sclerosis: the Lighthouse trial. Amyotroph Lateral Scler Frontotemporal Degener. 2019;20(7–8):595–604. [DOI] [PubMed] [Google Scholar]
  • 351.Sheean RK, et al. Association of regulatory T-cell expansion with progression of amyotrophic lateral sclerosis: a study of humans and a transgenic mouse model. JAMA Neurol. 2018;75(6):681–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Miller RG, et al. NP001 regulation of macrophage activation markers in ALS: a phase I clinical and biomarker study. Amyotroph Lateral Scler Frontotemporal Degener. 2014;15(7–8):601–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Neuvivo seeks FDA approval for its breakthrough ALS treatment NP001. 2024: Neuvivo.
  • 354.Elgueta R, et al. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev. 2009;229(1):152–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Eledon announces positive topline results from phase 2a trial of tegoprubart demonstrating safety, target engagement, and biomarker response in patients living with amyotrophic lateral sclerosis. 2022: Biospace.
  • 356.Carpanini SM, Torvell M, Morgan BP. Therapeutic inhibition of the complement system in diseases of the central nervous system. Front Immunol. 2019;10:362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Lobsiger CS, et al. C1q induction and global complement pathway activation do not contribute to ALS toxicity in mutant SOD1 mice. Proc Natl Acad Sci. 2013;110(46):E4385–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Swanson KV, Deng M, Ting JPY. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Zydus cleared to begin US trial of oral therapy usnoflast for ALS, in Phase 2b trial to involve 210 patients, company says. 2025: ALS News Today.
  • 360.TPN-101. 2024: AlzForum therapeutics databases.
  • 361.TPN-101 Demonstrates disease-modifying effects on C9orf72-Related ALS/FTD. 2024: Neurology Live.
  • 362.Takata M, et al. Fasudil, a rho kinase inhibitor, limits motor neuron loss in experimental models of amyotrophic lateral sclerosis. Br J Pharmacol. 2013;170(2):341–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Coleman ML, et al. Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat Cell Biol. 2001;3(4):339–45. [DOI] [PubMed] [Google Scholar]
  • 364.Lee DH et al. ROCK and PDE-5 Inhibitors for the treatment of dementia: literature review and meta-analysis. Biomedicines 2022;10(6). [DOI] [PMC free article] [PubMed]
  • 365.Guan G et al. Effect of the rho-kinase/ROCK signaling pathway on cytoskeleton components. Genes (Basel). 2023;14(2):272. [DOI] [PMC free article] [PubMed]
  • 366.Fasudil safe, preserves motor neurons in early-stage ALS: Study, in Raya also developed a version of drug for patients with difficulty swallowing. 2024: ALS News Today.
  • 367.Donaldson K, J.C., Hunt HJ. Safety, pharmacokinetics, and pharmacodynamics of the selective glucocorticoid receptor modulator dazucorilant in healthy volunteers. in ENCALS. 2023. Barcelona
  • 368.Corcept’s dazucorilant fails to slow ALS progression in clinical trial, in Phase 2 DAZALS study enrolled 249 adults at multiple sites. 2024: ALS News Today.
  • 369.Podestà MA, et al. Siplizumab selectively depletes effector memory T cells and promotes a relative expansion of alloreactive regulatory T cells in vitro. Am J Transplant. 2020;20(1):88–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Herrero-Beaumont G, Martínez Calatrava MJ, Castañeda S. Abatacept mechanism of action: concordance with its clinical profile. Reumatol Clin. 2012;8(2):78–83. [DOI] [PubMed] [Google Scholar]
  • 371.García-García R et al. Immune signaling kinases in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Int J Mol Sci. 2021;22(24). [DOI] [PMC free article] [PubMed]
  • 372.Evans FL, et al. Protective and regenerative roles of T cells in central nervous system disorders. Front Immunol. 2019;10:2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Engensis gene therapy found safe in small phase 2a trial, in Helixmith's therapy has potential to promote nerve cell and muscle regeneration. 2022: ALS News Today.
  • 374.Ahmadi AR, et al. A phase I, first-in-human study to evaluate the safety and tolerability, pharmacokinetics, and pharmacodynamics of MRG-001 in healthy subjects. Cell Rep Med. 2023;4(9): 101169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Geijo-Barrientos E, et al. Intramuscular injection of bone marrow stem cells in amyotrophic lateral sclerosis patients: a randomized clinical trial. Front Neurosci. 2020;14:195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Baloh RH, et al. Transplantation of human neural progenitor cells secreting GDNF into the spinal cord of patients with ALS: a phase 1/2a trial. Nat Med. 2022;28(9):1813–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Verdiperstat fails to differentiate from placebo in critical HEALY ALS platform trial. 2022: Neurology Live.
  • 378.Gao L, et al. First-in-human studies of pharmacokinetics and safety of utreloxastat (PTC857), a novel 15-lipooxygenase inhibitor for the treatment of amyotrophic lateral sclerosis. Clin Pharmacol Drug Dev. 2023;12(2):141–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.BioJiva reports results of pilot phase 2 clinical trial of RT001 in patients with amyotrophic lateral sclerosis (ALS). 2022: GlobeNewswire.
  • 380.Wen J, et al. Tetramethylpyrazine nitrone improves motor dysfunction and pathological manifestations by activating the PGC-1alpha/Nrf2/HO-1 pathway in ALS mice. Neuropharmacology. 2021;182: 108380. [DOI] [PubMed] [Google Scholar]
  • 381.Liu X, et al. Tetramethylpyrazine nitrone in amyotrophic lateral sclerosis: a randomized clinical trial. JAMA Netw Open. 2025;8(2): e2461055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Annual Report 2023: ARGX-119 development. 2023: Argenx.
  • 383.Vucic S, Kiernan MC. Nanocrystalline gold (CNM-Au8): a novel bioenergetic treatment for ALS. Expert Opin Investig Drugs. 2023;32(9):783–5. [DOI] [PubMed] [Google Scholar]
  • 384.New HEALEY-ALS trial results show CNM-AU8's effect on survival, neurofilament light. 2024: Neurology Live.
  • 385.Herrando-Grabulosa M, et al. Sigma 1 receptor as a therapeutic target for amyotrophic lateral sclerosis. Br J Pharmacol. 2021;178(6):1336–52. [DOI] [PubMed] [Google Scholar]
  • 386.Shefner J, et al. Pridopidine for the treatment of ALS—Results from the phase 2 Healey ALS platform trial (S5.003). Neurology. 2024;102(7_supplement_1):6524. [Google Scholar]
  • 387.NeuroSense Therapeutics plans to file for early commercialization of ALS treatment in Canada. 2024: PR Newswire.
  • 388.Felicetti T, Cecchetti V, Manfroni G. Modulating microRNA processing: enoxacin, the progenitor of a new class of drugs. J Med Chem. 2020;63(21):12275–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Magen I et al. A phase Ib/IIa randomized trial of enoxacin in patients with ALS. medRxiv, 2024: p. 2024.07.31.24311258.
  • 390.PLL Therapeutics obtains authorization to start phase I/II trial in Australia for patients with amyotrophic lateral sclerosis (ALS). 2024: PLL Therapeutics.
  • 391.Treatment for improving gut microbiome found safe in ALS, in Oral Therapy MaaT033 tested in phase 1B trial in patients in France. 2024: ALS News Today.
  • 392.Lemarchant S et al. NX210c peptide promotes glutamatergic receptor-mediated synaptic transmission and signaling in the mouse central nervous system. Int J Mol Sci. 2022;23(16). [DOI] [PMC free article] [PubMed]
  • 393.Axoltis Pharma presents promising results from phase 1B clinical trial of innovative drug candidate for neurodegenerative diseases. 2023: ALA Associates.
  • 394.Bartus RT, et al. beta2-Adrenoceptor agonists as novel, safe and potentially effective therapies for amyotrophic lateral sclerosis (ALS). Neurobiol Dis. 2016;85:11–24. [DOI] [PubMed] [Google Scholar]
  • 395.Li X, et al. Clenbuterol treatment is safe and associated with slowed disease progression in a small open-label trial in patients with amyotrophic lateral sclerosis. J Clin Neuromuscul Dis. 2023;24(4):214–21. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from CNS Drugs are provided here courtesy of Springer

RESOURCES