
Tetramethylpyrazine Nitrone in Amyotrophic Lateral Sclerosis
A Randomized Clinical Trial
Xiaolu Liu,MD
Huifang Shang,MD
Qianqian Wei,MD
Xiaoli Yao,MD, PhD
Ling Lian,MD
Jingxia Dang,MD
Rui Jia,MD
Zhiying Wu,MD, PhD
Hongfu Li,MD, PhD
Xi Cheng,MD
Zhangyu Zou,MD, PhD
Sheng Chen,MD, PhD
Min Zhang,MD, PhD
Yang Liu,MD, PhD
Yaling Liu,MD
Qi Liu,MD
Xusheng Huang,MD, PhD
Hongfen Wang,MD
Honglin Feng,MD
Shuyu Wang,MD
Dongsheng Fan,MD, PhD
Accepted for Publication: December 17, 2024.
Published: February 24, 2025. doi:10.1001/jamanetworkopen.2024.61055
Open Access: This is an open access article distributed under the terms of theCC-BY-NC-ND License, which does not permit alteration or commercial use, including those for text and data mining, AI training, and similar technologies. © 2025 Liu X et al.JAMA Network Open.
Corresponding Author: Dongsheng Fan, MD, PhD, Department of Neurology, Peking University Third Hospital, No. 49 North Garden Rd, Beijing 100191, China (dsfan@sina.com).
Author Contributions: Dr Fan had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.
Concept and design: X. Liu, Shang, Yao, Dang, Zou, Zhang, Q. Liu, Fan.
Acquisition, analysis, or interpretation of data: X. Liu, Wei, Lian, Jia, Wu, Li, Niu, Cheng, Zou, Chen, Yang Liu, Yaling Liu, Huang, H. Wang, Feng, S. Wang, Fan.
Drafting of the manuscript: X. Liu, Yao, Dang, Li, Zou, Yang Liu, H. Wang, Fan.
Critical review of the manuscript for important intellectual content: X. Liu, Shang, Wei, Lian, Jia, Wu, Niu, Cheng, Zou, Chen, Zhang, Yaling Liu, Q. Liu, Huang, Feng, S. Wang, Fan.
Statistical analysis: X. Liu, Wei, Yao, Li, Zou, H. Wang, Fan.
Obtained funding: Cheng, Fan.
Administrative, technical, or material support: Shang, Lian, Dang, Jia, Wu, Niu, Cheng, Zhang, Yang Liu, Yaling Liu, Q. Liu, Huang, Feng, S. Wang, Fan.
Supervision: Shang, Zhang, Fan.
Conflict of Interest Disclosures: None reported.
Funding/Support: This trial was sponsored by Guangzhou Magpie Pharmaceuticals Co, Ltd. and partially supported by grant 2022YFA1303003 from the Ministry of Science and Technology of China (Dr Fan).
Role of the Funder/Sponsor: The funder had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Group Information:The nonauthor members of the TBNALS group are listed inSupplement 3.
Data Sharing Statement: SeeSupplement 4.
Additional Contributions: The authors thank all the patients, investigators, and study site personnel involved in this trial. The authors also thank Ms Linda Wang, BS (Guangzhou Magpie Pharmaceuticals Co, Ltd), for proofreading the manuscript for language and factuality. She was compensated for her contribution.
Corresponding author.
Received 2024 Aug 6; Accepted 2024 Dec 17; Collection date 2025 Feb.
This is an open access article distributed under the terms of the CC-BY-NC-ND License, which does not permit alteration or commercial use, including those for text and data mining, AI training, and similar technologies.
Key Points
Question
Is treatment with tetramethylpyrazine nitrone (TBN) safe and effective for improving functional outcomes in patients with amyotrophic lateral sclerosis (ALS)?
Findings
In this randomized clinical trial of 155 patients who were given low-dose tetramethylpyrazine nitrone, high-dose tetramethylpyrazine nitrone, or placebo, the occurrence of adverse events was comparable among all groups. The primary measure of effectiveness, the ALS Functional Rating Scale–Revised score, did not show a significant improvement with tetramethylpyrazine nitrone treatment, while high-dose tetramethylpyrazine nitrone showed a significant slowing of the decline in grip strength, especially in patients younger than 65 years with slowly progressing ALS.
Meaning
These findings demonstrate that tetramethylpyrazine nitrone is safe and may have potential benefits in slowing the decline in grip strength, although its impact on the primary outcome measure was not statistically significant.
This randomized clinical trial evaluates the safety of oral tetramethylpyrazine nitrone and its effect on disease progression among patients in China with amyotrophic lateral sclerosis.
Abstract
Importance
Tetramethylpyrazine nitrone has exhibited promising results in improving motor dysfunction in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS).
Objective
To evaluate the safety and efficacy of orally administered tetramethylpyrazine nitrone in patients with ALS.
Design, Setting, and Participants
This phase 2, multicenter, double-masked, placebo-controlled, randomized clinical trial was conducted from December 24, 2020, through July 14, 2023, in 11 centers in China, with a 180-day follow-up. Patients aged 45 to 70 years, with ALS onset within 2 years, ALS Functional Rating Scale–Revised (ALSFRS-R) scores of at least 2 points on each item, and forced vital capacity (FVC) of at least 80% were included. Patients experienced a 1- to 4-point decrease in ALSFRS-R score during a 3-month screening period.
Interventions
Patients were randomly assigned 1:1:1 to receive low-dose tetramethylpyrazine nitrone (600 mg twice daily), high-dose tetramethylpyrazine nitrone (1200 mg twice daily), or placebo (twice daily) for 180 days.
Main Outcomes and Measures
The primary outcome was change in ALSFRS-R score (range of 0-48, with lower scores indicating worse function) from baseline to 180 days. The secondary outcomes were changes in FVC, grip strength, ALS Assessment Questionnaire-40 (ALSAQ-40) score, and end point events. Safety outcomes included adverse events.
Results
A total of 155 patients (mean [SD] age, 55.0 [6.5] years; 115 men [74.2%]) were randomized (51 [32.9%] to the low-dose tetramethylpyrazine nitrone group, 52 [33.6%] to the high-dose tetramethylpyrazine nitrone group, and 52 [33.6%] to the placebo group). No significant differences were observed in ALSFRS-R score changes between low-dose tetramethylpyrazine nitrone (least squares [LS] mean difference, −0.89 points; 95% CI −3.25 to 1.48 points) and high-dose tetramethylpyrazine nitrone (LS mean difference, −0.20 points; 95% CI −2.48 to 2.07 points) compared with placebo. High-dose tetramethylpyrazine nitrone showed a significantly slower decline in grip strength at day 180 (LS mean difference, 2.46 kg; 95% CI, 0.15-4.76 kg). In a subgroup of patients younger than 65 years with slower disease progression, tetramethylpyrazine nitrone significantly attenuated the decline in grip strength (LS mean difference, 3.63 kg; 95% CI, 0.84-6.41 kg), bulbar scores (LS mean difference, 0.66 points; 95% CI, 0.03-1.29 points), and respiratory scores (LS mean difference, 0.54 points; 95% CI, 0.03-1.06 points). Adverse events were mostly mild or moderate, with no severe treatment-related adverse events or deaths.
Conclusions and Relevance
This randomized clinical trial demonstrates that tetramethylpyrazine nitrone is safe and well-tolerated in patients with ALS. There was no difference in the primary end point across the low-dose, high-dose, and placebo groups, with significant benefits in a subgroup of younger patients with slower disease progression.
Trial Registration
ChiCTR Identifier:ChiCTR2000039689
Introduction
Amyotrophic lateral sclerosis (ALS) is a degenerative neuromuscular disease affecting motor neurons within the cerebral cortex, brainstem, and spinal cord. The onset of ALS usually begins with local morbidity and then spreads throughout the body, with a median survival of 3 to 5 years.1 Ultimately, progressive muscle weakness and respiratory failure lead to death in most patients with ALS.2
Currently, 4 drugs (riluzole, edaravone, AMX0035, and tofersen) are approved by the US Food and Drug Administration (FDA) for ALS treatment, but their clinical benefits are limited. Riluzole, approved in 1995, extends survival by 2 to 3 months.3,4,5 Edaravone and AMX0035 delay disease progression in patients with ALS to a limited extent. Improvements in grip strength and respiratory function, 2 key markers of patient quality of life, with edaravone or AMX0035 were not statistically significant compared with placebo.6,7 AMX0035 was approved by the FDA for ALS treatment after the phase 2 CENTAUR trial, but the phase 3 PHOENIX study led to the discontinuation of AMX0035’s marketing authorization. In clinical trials, tofersen reduced the level of neurofilament light chains in patients with ALS associated with alterations inSOD1 (SOD1 ALS) but did not show improvement in its primary (ALS Functional Rating Scale–Revised [ALSFRS-R] score) or secondary (grip strength and respiratory function) end points.8 Although many new ALS treatments have entered the clinical development stage, the efficacy and safety of these treatments still remain to be demonstrated.
The exact causes of ALS are not fully understood, but research has suggested multiple contributing factors, including oxidative stress, calcium overload, mitochondrial dysfunction, excitotoxicity, protein aggregation, and neuroinflammation.9,10,11 A nitrone derivative of tetramethylpyrazine, from the Chinese herb Chuanxiong, shows multifunctional neuroprotective effects by reducing calcium overload, improving mitochondrial function, activating autophagy, and providing anti-inflammatory benefits.12,13,14 The nitrone group in tetramethylpyrazine nitrone, also found in NXY-059, shows promise in reducing disability from acute ischemic stroke.15 Nonetheless, NXY-059 failed in phase 3 clinical trials due to poor blood-brain barrier permeability, with only 1% brain tissue concentration.16,17 In contrast to NXY-059, tetramethylpyrazine nitrone readily penetrates the blood-brain barrier and reaches therapeutic levels in brain tissues, with concentrations reaching approximately 68% of plasma levels.18 Importantly, tetramethylpyrazine nitrone has been proven to improve motor dysfunction in various neurodegenerative disease animal models, including ALS.19,20,21,22
Aside from readily penetrating the blood-brain barrier, a necessary feature for targeting central nervous system diseases, tetramethylpyrazine nitrone also has a multifunctional mechanism of action. In various cellular and animal models of ALS, tetramethylpyrazine nitrone blocked intracellular calcium overload, reduced reactive oxygen species, upregulated the AMPK/PGC-1α/NRF2/HO-1 (adenosine 5′-monophosphate–activated protein kinase/peroxisome proliferator–activated receptor-γ coactivator 1α/nuclear factor erythoid 2–related factor 2/heme oxygenase-1) pathway to improve mitochondrial function, and inhibited mammalian target of rapamycin to activate autophagy, leading to the degradation of superoxide dismutase 1 (SOD1) and TAR DNA-binding protein 43 (TDP-43) aggregates.21,22,23,24 Based on these findings, the aim of our study is to investigate the safety and efficacy of tetramethylpyrazine nitrone in patients with ALS.
Methods
Study Design and Patients
This phase 2, double-masked, placebo-controlled, randomized clinical trial was conducted from December 24, 2020, through July 14, 2023, at 11 centers in China. Details of the trial rationale, design, and methods have been described previously and are provided in the trial protocol (Supplement 1). The trial was conducted in conformity with the Declaration of Helsinki,25 the Chinese Good Clinical Practice guidelines,26 and local regulations. The institutional review board of each participating center approved the protocol, and all patients provided written informed consent. This study adhered to the Consolidated Standards of Reporting Trials (CONSORT) reporting guideline.
All patients had a diagnosis of definite or probable ALS per revised El Escorial criteria. Eligible patients were aged 45 to 70 years with an ALS duration of 2 years or less, a decrease in ALSFRS-R score of 1 to 4 points within a 3-month screening period, a score of at least 2 points on each ALSFRS-R item, normal respiratory function (4 points), and a forced vital capacity (FVC) of at least 80% at baseline. The ALSFRS-R is a 12-subdomain self-assessment for functionality scored on a scale of 0 to 48, with lower scores indicating worse function. The use of standard of care, such as riluzole, was permitted, but edaravone was contraindicated. Exclusion criteria included familial ALS, cognitive impairment, dysphagia, severe hepatic impairment (alanine aminotransferase or aspartate aminotransferase >3 times the upper limit of normal), severe kidney impairment (creatinine clearance <30 mL/min), cancer, hypersensitivity to the study drug or tetramethylpyrazine, or participation in other trials within 30 days. Coordinating centers and clinical site information are available in eTable 1 inSupplement 2. Detailed inclusion and exclusion criteria are shown in eTable 2 inSupplement 2.
Randomization and Masking
Dynamic randomization by minimization was used to balance sample sizes and prognostic factors across groups. Patients were stratified by ALS diagnosis (definite vs probable), change in ALSFRS-R score within the 3-month screening period (−1 or −2 vs −3 or −4), age (<65 vs ≥65 years), and riluzole use (yes vs no). Eligible patients were randomly assigned (1:1:1) to a low-dose tetramethylpyrazine nitrone group (600 mg orally twice daily), high-dose tetramethylpyrazine nitrone group (1200 mg orally twice daily), or placebo group using an interactive web response system, which only authorized personnel could access for randomization codes; treatment assignments followed these codes. The tetramethylpyrazine nitrone and placebo tablets were packaged using random codes. All patients and study personnel were masked to treatment assignments.
Procedures
Before randomization, potential participants entered a screening period of up to 3 months. Patients with a decrease in ALSFRS-R score of 1 to 4 points within this period were randomly assigned to the low-dose tetramethylpyrazine nitrone, high-dose tetramethylpyrazine nitrone, or placebo group. The tetramethylpyrazine nitrone or placebo tablets were administered on an empty stomach at least 1 hour before or 2 hours after meals. The double-masked treatment period was from baseline to day 180. Study patients had visits on day 30, 90, and 180 after the baseline visit, with a telephone interview done on days 7, 60, 120, and 150. Data on demographics, clinical characteristics, medical history, concomitant medications, adverse events (AEs), serious AEs, vital signs, physical examinations, electrocardiograms, and laboratory tests were collected and updated throughout the trial. All data were uploaded to an electronic data capture system.
Outcomes
The primary efficacy end point was the change in ALSFRS-R score from baseline to day 180. Secondary end points included the total score of the ALSFRS-R evaluated on days 30, 90, and 180, as well as the corresponding changes in grip strength (calculated as the mean of both hands) measured at these time points. Secondary end points included the proportion of end point events, changes in FVC, and variations in ALS Assessment Questionnaire-40 (ALSAQ-40) scores (scale of 0-500, with lower scores indicating better function). The prespecified end point events included death, tracheotomy, or invasive or persistent noninvasive ventilation (daily usage time ≥22 hours and duration ≥10 days). Safety outcomes included AEs, serious AEs, and changes in vital signs and laboratory data before and after treatment.
Statistical Analysis
The sample size was not statistically predetermined, as this was an exploratory study on safety and efficacy, estimating 150 total patients and 50 per group. Analysis of covariance (ANCOVA) was used to analyze the primary end point in the full analysis set. The full analysis set was defined as randomized patients with at least 1 dose of investigational medicine product and 1 postbaseline efficacy assessment. The dependent variable was the change in ALSFRS-R score from baseline to day 180. Patients were included as a random effect in the analysis of the change in ALSFRS-R scores from baseline, which was evaluated on days 30, 90, and 180. Covariates included the baseline ALSFRS-R score, site, sex, ethnicity (Han Chinese, other), ALS diagnostic grade (clinical definite diagnosis vs clinical probable diagnosis), ALSFRS-R decline during screening (−1 or −2 vs −3 or −4), age (<65 vs ≥65 years), and riluzole use (yes vs no). The ethnicity of the participants was determined according to the Chinese citizenship registration system. In addition to Han Chinese patients, patients from Miao, Dong, and Manchu ethnic groups were also enrolled. Ethnicity was collected to show the high homogeneity of the population. Group assignment was the fixed effect. At the outset of this study, a prespecified fixed sequential testing approach was used. The comparison between the high-dose group and the placebo group was tested first, followed by the low-dose vs placebo comparison, contingent upon the success of the high-dose test. Least squares (LS) means and 95% CIs were calculated for each group. The primary efficacy end point, ie, the change in ALSFRS-R score from baseline to day 180, underwent Bonferroni correction to adjust for multiple comparisons. Analyses were also conducted on a per-protocol set using ANCOVA.
The ALSFRS-R subdomain scores followed the total score methodology. Secondary end points, including changes in ALSFRS-R scores on days 30, 90, and 180 from baseline and FVC and grip strength, were analyzed using a mixed model for repeated measures in the full analysis set. The ALSAQ-40 score was analyzed using ANCOVA. End point event proportions and time-to-event end points were analyzed using Fisher exact test and Wilcoxon rank sum test, respectively. Safety was assessed in patients receiving at least 1 dose of investigational medicine product. To address missing data, we used multiple imputation techniques to fill in missing values based on observed data. This approach ensures that the analysis remains robust and minimizes the impact of missing data on the study’s findings. Statistical analyses were performed using SAS, version 9.4 (SAS Institute Inc).P < .05 was set as the threshold for statistical significance (2-sided).
Results
Randomization and Baseline Characteristics
A total of 259 patients with ALS were screened, and 155 (mean [SD] age, 55.0 [6.5] years; 115 men [74.2%] and 40 women [25.5%]; 152 of Han Chinese [98.1%] and 3 of other [1.9%] ethnicity) were randomly assigned to the low-dose tetramethylpyrazine nitrone group (51 patients [32.9%]), high-dose tetramethylpyrazine nitrone group (52 patients [33.6%]), or placebo group (52 patients [33.6%]). Among these 155 patients, 22 discontinued early due to voluntary withdrawal, AEs, or progressive disease. Ultimately, 42 patients (82.4%) in the low-dose tetramethylpyrazine nitrone group, 46 (88.5%) in the high-dose tetramethylpyrazine nitrone group, and 45 (86.5%) in the placebo group completed the 180-day treatment (Figure 1).
Figure 1. Enrollment and Randomization of the Patients.

aPatient required invasive ventilator support.
The demographic and baseline characteristics of the full analysis set (148 patients) were similar across groups except for prior riluzole and/or edaravone use (Table 1). Prior use of riluzole alone was the most common among all groups (39 patients [84.8%] in the low-dose tetramethylpyrazine nitrone group, 33 [63.5%] in the high-dose tetramethylpyrazine nitrone group, and 39 [78.0%] in the placebo group). Only 1 patient (2.0%) with a history of edaravone use alone was present in the placebo group. The number of patients with a history of using both riluzole and edaravone was different across the groups, with 3 (6.5%) in the low-dose tetramethylpyrazine nitrone group, 15 (28.8%) in the high-dose tetramethylpyrazine nitrone group, and 10 (20.0%) in the placebo group (Table 1). Most patients were male (110 [74.3%] vs 38 female [25.7%]) and younger than 65 years (130 [87.8%] vs 18 [12.2%] aged ≥65 years). The mean (SD) ALS duration was 14.4 (5.0) months. During screening, 111 patients (75.0%) had an ALSFRS-R score change of −1 or −2 points, and 37 (25.0%) had a change of −3 or −4 points. The mean (SD) baseline grip strength was 15.8 (10.2) kg, and mean (SD) baseline FVC was 98.4% (13.1%). The median duration of drug exposure for patients was 178.5 days (IQR, 170.0-181.0 days) in the low-dose tetramethylpyrazine nitrone group, 178.8 days (IQR, 173.5-181.5 days) in the high-dose tetramethylpyrazine nitrone group, and 178.0 days (IQR, 172.5-181.5 days) in the placebo group. During the trial, riluzole was administered to 45 patients (88.2%) in the low-dose tetramethylpyrazine nitrone group, 46 (88.5%) in the high-dose tetramethylpyrazine nitrone group, and 48 (92.3%) in the placebo group.
Table 1. Demographics and Clinical Characteristics at Baseline in the Full Analysis Set.
| Characteristic | Participants, mean (SD) | ||
|---|---|---|---|
| Placebo group (n = 50) | Low-dose tetramethylpyrazine nitrone group (n = 46) | High-dose tetramethylpyrazine nitrone group (n = 52) | |
| Sex, No. (%) | |||
| Female | 15 (30.0) | 13 (28.3) | 10 (19.2) |
| Male | 35 (70.0) | 33 (71.7) | 42 (80.8) |
| Age, y | 53.4 (6.7) | 55.6 (6.4) | 55.5 (6.3) |
| Age group, No. (%), y | |||
| <65a | 45 (90.0) | 40 (87.0) | 45 (86.5) |
| ≥65a | 5 (10.0) | 6 (13.0) | 7 (13.5) |
| Ethnicity, No. (%) | |||
| Han Chinese | 50 (100.0) | 45 (97.8) | 50 (96.2) |
| Otherb | 0 | 1 (2.2) | 2 (3.8) |
| Height, cm | 165.9 (7.7) | 165.2 (8.2) | 166.4 (6.5) |
| Body weight, kg | 64.0 (8.7) | 63.4 (11.4) | 65.5 (10.8) |
| BMI, kg/cm2 | 23.2 (2.8) | 23.1 (3.1) | 23.5 (3.0) |
| ALS diagnostic criteriac | |||
| Definitea | 28 (56.0) | 24 (52.2) | 29 (55.8) |
| Probablea | 22 (44.0) | 22 (47.8) | 23 (44.2) |
| Duration of disease, y | 1.4 (0.4) | 1.2 (0.4) | 1.2 (0.4) |
| Duration groups, No. (%), y | |||
| <1 | 15 (30.0) | 20 (43.5) | 21 (40.4) |
| ≥1 | 35 (70.0) | 26 (56.5) | 31 (59.6) |
| ALSAQ-40 scored | 80.9 (53.1) | 76.6 (56.4) | 80.4 (50.7) |
| ALSFRS-R scoree | |||
| Day 90 (before screening period) | 43.5 (2.1) | 43.6 (2.5) | 43.7 (2.1) |
| Day 0 (at baseline) | 41.8 (2.2) | 41.8 (2.5) | 41.8 (2.3) |
| Change during the screening period, No. (%) | |||
| −2 or −1a | 39 (78.0) | 34 (73.9) | 38 (73.1) |
| −4 or −3a | 11 (22.0) | 12 (26.1) | 14 (26.9) |
| Maximum grip strength, kgf | 16.1 (7.8) | 14.5 (10.4) | 16.5 (12.0) |
| FVC, % | 98.3 (12.1) | 99.2 (13.2) | 97.9 (14.0) |
| History of riluzole and/or edaravone use, No. (%) | |||
| Riluzole only | 39 (78.0) | 39 (84.8) | 33 (63.5) |
| Edaravone only | 1 (2.0) | 0 (0.0) | 0 (0.0) |
| Both riluzole and edaravone | 10 (20.0) | 3 (6.5) | 15 (28.8) |
Abbreviations: ALS, amyotrophic lateral sclerosis; ALSAQ-40, Amyotrophic Lateral Sclerosis Assessment Questionnaire-40; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised; BMI, body mass index (calculated as weight in kilograms divided by height in meters squared); FVC, forced vital capacity.
Factor used in dynamic allocation.
Details of ethnicity are not disclosed because of the small number of participants.
According to revised El Escorial criteria.
Scored on a scale of 0 to 500, with lower scores indicating better function.
Scored on a scale of 0 to 48, with lower scores indicating worse function.
Mean of both hands.
Efficacy Outcomes
For the primary end point, the LS mean difference vs placebo for change from baseline in ALSFRS-R scores at 180 days was −0.89 points (95% CI, −3.25 to 1.48 points;P = .40) with low-dose tetramethylpyrazine nitrone and −0.20 (95% CI, −2.48 to 2.07;P = .84) with high-dose tetramethylpyrazine nitrone (Table 2). There were no significant differences in ALSFRS-R score changes between the tetramethylpyrazine nitrone and placebo groups. The ALSFRS-R score trends over time are shown inFigure 2A and further detailed in eTables 3 and 4 inSupplement 2. The ALSFRS-R subdomain scores also did not differ among groups at day 180 (Table 2). The eFigure inSupplement 2 shows the results of a prespecified subgroup analysis of change from baseline to day 180 in ALSFRS-R score. A prespecified subgroup analysis suggested potential treatment interactions favoring tetramethylpyrazine nitrone by day 180 in patients consistently using riluzole.
Table 2. Primary and Secondary Outcomes in the Full Analysis Set.
| Outcome | Placebo group, LS mean change from baseline (95% CI) | Low-dose, LS mean change from baseline (95% CI) | Difference, low dose vs placebo (95% CI) | P value | High-dose, LS mean change from baseline (95% CI) | Difference high dose vs placebo (95% CI) | P value |
|---|---|---|---|---|---|---|---|
| Primary outcome | |||||||
| ALSFRS-R score, pointsa,b | −7.47 (−10.38 to −4.57) | −8.36 (−11.16 to −5.57) | −0.89 (−3.25 to 1.48) | .40 | −7.68 (−10.44 to −4.92) | −0.20 (−2.48 to 2.07) | .84 |
| Bulbar score | −1.26 (−1.76 to −0.76) | −1.32 (−1.83 to −0.82) | −0.06 (−0.69 to 0.56) | .84 | −1.12 (−1.61 to −0.64) | 0.14 (−0.47 to 0.74) | .66 |
| Respiratory score | −1.21 (−1.64 to −0.77) | −1.12 (−1.56 to −0.67) | 0.09 (−0.49 to 0.67) | .76 | −0.96 (−1.38 to −0.53) | 0.25 (−0.31 to 0.82) | .38 |
| Fine motor score | −2.69 (−3.49 to −1.90) | −3.14 (−3.94 to −2.34) | −0.45 (−1.46 to 0.56) | .38 | −3.17 (−3.94 to −2.41) | −0.48 (−1.46 to 0.50) | .33 |
| Gross motor score | −2.46 (−3.11 to −1.81) | −2.56 (−3.20 to −1.92) | −0.10 (−0.86 to 0.66) | .80 | −2.63 (−3.26 to −2.00) | −0.17 (−0.91 to 0.57) | .65 |
| Secondary outcomes | |||||||
| FVC, %a | −13.23 (−18.41 to −8.04) | −14.65 (−19.79 to −9.51) | −1.43 (−7.47 to 4.62) | .64 | −13.07 (−18.11 to −8.02) | 0.16 (−5.76 to 6.08) | .96 |
| Grip strength, kga,c | −7.60 (−9.53 to −5.67) | −6.53 (−8.45 to −4.62) | 1.06 (−1.31 to 3.44) | .38 | −5.14 (−6.99 to −3.30) | 2.46 (0.15 to 4.76) | .04 |
| ALSAQ-40 score, pointsd | 90.85 (48.27 to 133.43) | 93.77 (52.81 to 134.72) | 2.92 (−31.91 to 37.75) | .87 | 85.69 (44.49 to 126.89) | −5.16 (−38.37 to 28.05) | .76 |
| Incidence of end point events, No. (%) | 7 (14.0) | 9 (19.6) | NA | NA | 5 (9.6) | NA | .35e |
| Time-to-event end points, median (IQR), mo | 3.3 (3.0-5.3) | 5.0 (3.0-5.9) | NA | NA | 6.1 (5.1-6.2) | NA | NA |
| Primary and secondary outcomes in the slower progression subgroupf | |||||||
| ALSFRS-R score, points | −5.76 (−7.88 to −3.64) | −5.01 (−6.93 to −3.09) | 0.75 (−1.40 to 2.90) | .49 | −3.93 (−5.94 to −1.92) | 1.83 (−0.29 to 3.95) | .09 |
| Bulbar score | −1.02 (−1.63 to −0.42) | −0.67 (−1.23 to −0.10) | 0.36 (−0.27 to 0.98) | .26 | −0.36 (−0.95 to 0.22) | 0.66 (0.03 to 1.29) | .04 |
| Fine motor score | −2.50 (−3.52 to −1.49) | −2.30 (−3.26 to −1.34) | 0.20 (−0.86 to 1.26) | .71 | −1.80 (−2.80 to −0.80) | 0.70 (−0.35 to 1.75) | .19 |
| Gross motor score | −2.03 (−2.89 to −1.16) | −2.02 (−2.81 to −1.23) | 0.01 (−0.89 to 0.90) | .99 | −1.95 (−2.79 to −1.11) | 0.08 (−0.80 to 0.95) | .86 |
| Respiratory score | −0.71 (−1.21 to −0.22) | −0.17 (−0.64 to 0.29) | 0.54 (0.03 to 1.06) | .04 | −0.11 (−0.59 to 0.38) | 0.61 (0.09 to 1.12) | .02 |
| FVC, % | −14.46 (−21.02 to −7.90) | −10.16 (−16.60 to −3.73) | 4.29 (−2.32 to 10.90) | .20 | −9.06 (−15.58 to −2.54) | 5.39 (−1.26 to 12.05 | .11 |
| Grip strength, kgc | −7.24 (−9.96 to −4.52) | −4.74 (−7.31 to −2.16) | 2.51 (−0.36 to 5.37) | .09 | −3.62 (−6.33 to −0.90) | 3.63 (0.84 to 6.41) | .01 |
Abbreviations: ALSAQ-40, Amyotrophic Lateral Sclerosis Assessment Questionnaire–40; ALSFRS-R, Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised; FVC, forced vital capacity; NA, not applicable.
Missing data imputed using a multiple imputation procedure.
Scored on a scale of 0 to 48, with lower scores indicating worse function.
Mean for the left and right hands.
Scored on a scale of 0 to 500, with lower scores indicating better function.
Fisher exact test.
The subset of individuals below the age of 65, who exhibited a decline in ALSFRS-R score of 1-2 points within a 3-month screening period, was designated as the slower-progression subgroup.
Figure 2. Mean Amyotrophic Lateral Sclerosis Functional Rating Scale–Revised (ALSFRS-R) Scores and Grip Strength in the Full Analysis Set.
Shown are the mean (SD) of observed values over time. A, The ALSFRS-R is scored on a scale of 0 to 48, with lower scores indicating worse function. For patients with missing ALSFRS-R scores, data were imputed by multiple imputation. B, Grip strength values were calculated as the mean of both hands. Missing data were not imputed for grip strength.
From baseline to day 180, the LS mean difference vs placebo in FVC was −1.43% (95% CI, −7.47% to 4.62%;P = .64) with low-dose tetramethylpyrazine nitrone and 0.16% (95% CI, −5.76% to 6.08%;P = .96) with high-dose tetramethylpyrazine nitrone (Table 2). In the full analysis set, grip strength changes from baseline to day 180 were not significantly different between the low-dose tetramethylpyrazine nitrone and placebo groups (difference, 1.06 kg; 95% CI, −1.31 to 3.44 kg;P = .38). However, the high-dose tetramethylpyrazine nitrone group showed a significant attenuation of the decline in grip strength compared with placebo (LS mean difference, 2.46 kg; 95% CI, 0.15-4.76 kg;P = .04) (Figure 2B). Similarly, in the per-protocol set, the high-dose tetramethylpyrazine nitrone group exhibited a significant attenuation of grip strength decline compared with placebo at day 180 (LS mean difference, 2.42 kg; 95% CI, 0.04-4.80 kg;P = .046) (eTable 5 inSupplement 2). Sensitivity analyses confirmed the robustness of the grip strength results. At day 180, the ALSAQ-40 scores increased slightly in the low-dose tetramethylpyrazine nitrone group compared with the placebo group (LS mean difference, 2.92 points; 95% CI, −31.91 to 37.75 points;P = .87) but decreased in the high-dose tetramethylpyrazine nitrone group (LS mean difference, −5.16 points; 95% CI, −38.37 to 28.05 points;P = .76); however, neither of these differences were statistically significant.
End point events occurred in 9 patients (19.6%) in the low-dose tetramethylpyrazine nitrone group, 5 (9.6%) in the high-dose tetramethylpyrazine nitrone group, and 7 (14.0%) in the placebo group, with no significant differences (Fisher exact testP = .35) (Table 2). The median time to end point events was 3.3 months (IQR, 3.0-5.3 months) in the placebo group, 5.0 months (IQR, 3.0-5.9 months) in the low-dose tetramethylpyrazine nitrone group, and 6.1 months (IQR, 5.1-6.2 months) in the high-dose tetramethylpyrazine nitrone group (Table 2). Compared with the placebo group, the time to end point events increased by 51.5% in the low-dose tetramethylpyrazine nitrone group (from 3.3 to 5.0 months) and by 84.8% in the high-dose tetramethylpyrazine nitrone group (from 3.3 to 6.1 months).
Subgroup Analysis
Since the rate of disease progression and patient age may affect drug response, we stratified patients according to their initial progression rate and age in a sensitivity analysis. The subset of individuals younger than 65 years who had a decline in ALSFRS-R score of 1 to 2 points within the 3-month screening period was designated as the slower progression subgroup. This subgroup included 90 of 155 (58.1%) patients, with 28 (18.1%) receiving low-dose tetramethylpyrazine nitrone, 29 (18.7%) receiving high-dose tetramethylpyrazine nitrone, and 33 (21.3%) receiving placebo. The primary and all secondary end points, excluding ALSAQ-40 and proportion of end point events, were evaluated within the slower progression subgroup.
For ALSFRS-R scores at day 180, the LS mean difference vs placebo was 0.75 points (95% CI, −1.40 to 2.90 points;P = .49) with low-dose tetramethylpyrazine nitrone and 1.83 points (95% CI, −0.29 to 3.95 points;P = .09) with high-dose tetramethylpyrazine nitrone (Table 2). Moreover, high-dose tetramethylpyrazine nitrone showed significant positive effects on ALSFRS-R bulbar subdomain scores compared with placebo (LS mean difference, 0.66 points; 95% CI, 0.03-1.29 points;P = .04). For the ALSFRS-R respiratory subdomain scores, the LS mean difference vs placebo was 0.54 points (95% CI, 0.03-1.06 points;P = .04) with low-dose tetramethylpyrazine nitrone and 0.61 points (95% CI, 0.09-1.12 points;P = .02) with high-dose tetramethylpyrazine nitrone. At day 180, grip strength also exhibited a significantly slower decline with high-dose tetramethylpyrazine nitrone (LS mean difference, 3.63 kg; 95% CI, 0.84-6.41;P = .01). Grip strength also exhibited a slower decline with low-dose tetramethylpyrazine nitrone (LS mean difference, 2.51 kg; 95% CI, −0.36 to 5.37 kg;P = .09), although the difference was not significant.
Safety Outcomes
Of the 155 randomized patients, 153 were included in the safety analysis set. Treatment-emergent AEs of any grade occurred in 115 patients (75.2%). As detailed inTable 3, 5 of 50 patients (10.0%) in the low-dose tetramethylpyrazine nitrone group, 3 of 52 (5.8%) in the high-dose tetramethylpyrazine nitrone group, and 3 of 51 (5.9%) in the placebo group experienced treatment-emergent AEs of grade 3 or higher. Treatment-related AEs were noted in 12 of 50 patients (24.0%) in the low-dose group, 18 of 52 (34.6%) in the high-dose group, and 9 of 51 (17.6%) in the placebo group and were mostly mild or moderate in intensity. No grade 3 or higher treatment-related AEs or deaths occurred. Treatment-related AEs leading to drug interruption were rare. Serious AEs were similar in incidence among all study groups, with none related to the investigational medicine product (Table 3).
Table 3. Adverse Events in the Safety Analysis Set.
| Outcome | Placebo group (n = 51) | Low-dose tetramethylpyrazine nitrone group (n = 50) | High-dose tetramethylpyrazine nitrone group (n = 52) | |||
|---|---|---|---|---|---|---|
| Events, No. | Patients, No. (%) | Events, No. | Patients, No. (%) | Events, No. | Patients, No. (%) | |
| All TEAEs | ||||||
| Any | 120 | 40 (78.4) | 81 | 39 (78.0) | 103 | 36 (69.2) |
| Severity (grade ≥3) | 3 | 3 (5.9) | 5 | 5 (10.0) | 4 | 3 (5.8) |
| TRAEs | 16 | 9 (17.6) | 19 | 12 (24.0) | 36 | 18 (34.6) |
| Severity (grade ≥3) | 0 | 0 | 0 | 0 | 0 | 0 |
| Leading to drug interruption | 1 | 1 (2.0) | 4 | 1 (2.0) | 0 | 0 |
| Leading to study discontinuation | 0 | 0 | 1 | 1 (2.0) | 2 | 1 (1.9) |
| Leading to drug discontinuation | 0 | 0 | 1 | 1 (2.0) | 0 | 0 |
| TEAEs reported in ≥5% of patients | ||||||
| COVID-19 | 3 | 3 (5.9) | 5 | 5 (10.0) | 5 | 4 (7.7) |
| Urinary tract infection | 5 | 5 (9.8) | 3 | 3 (6.0) | 2 | 2 (3.8) |
| Upper respiratory tract infection | 6 | 5 (9.8) | 3 | 3 (6.0) | 1 | 1 (1.9) |
| ALT elevation | 4 | 3 (5.9) | 2 | 2 (4.0) | 6 | 5 (9.6) |
| AST elevation | 3 | 2 (3.9) | 0 | 0 | 4 | 4 (7.7) |
| GGT elevation | 0 | 0 | 0 | 0 | 4 | 4 (7.7) |
| TSH elevation | 2 | 1 (2.0) | 3 | 3 (6.0) | 0 | 0 |
| Body weight gain | 0 | 0 | 3 | 3 (6.0) | 0 | 0 |
| Diarrhea | 3 | 3 (5.9) | 2 | 1 (2.0) | 1 | 1 (1.9) |
| Constipation | 0 | 0 | 1 | 1 (2.0) | 6 | 3 (5.8) |
| Dizziness | 3 | 3 (5.9) | 7 | 7 (14.0) | 1 | 1 (1.9) |
| Headache | 6 | 4 (7.8) | 0 | 0 | 1 | 1 (1.9) |
| Influenza-like illness | 3 | 3 (5.9) | 0 | 0 | 5 | 3 (5.8) |
| Fever | 3 | 3 (5.9) | 0 | 0 | 0 | 0 |
| Limb pain | 4 | 3 (5.9) | 0 | 0 | 1 | 1 (1.9) |
| Back pain | 1 | 1 (2.0) | 0 | 0 | 3 | 3 (5.8) |
| Expectoration | 3 | 3 (5.9) | 0 | 0 | 0 | 0 |
| Abnormal liver function | 1 | 1 (2.0) | 4 | 4 (8.0) | 2 | 2 (3.8) |
| Rash | 1 | 1 (2.0) | 3 | 3 (6.0) | 1 | 1 (1.9) |
| Hypertension | 4 | 4 (7.8) | 0 | 0 | 2 | 2 (3.8) |
| SAEs | 1 | 1 (2.0) | 3 | 3 (6.0) | 3 | 3 (5.8) |
| Epidural hematoma | 0 | 0 | 0 | 0 | 1 | 1 (1.9) |
| Femoral neck fracture | 0 | 0 | 1 | 1 (2.0) | 0 | 0 |
| Clavicle fracture | 0 | 0 | 0 | 0 | 1 | 1 (1.9) |
| Infectious pneumonia | 0 | 0 | 1 | 1 (2.0) | 0 | 0 |
| Gastroenteritis | 0 | 0 | 0 | 0 | 1 | 1 (1.9) |
| Syncope | 1 | 1 (2.0) | 0 | 0 | 0 | 0 |
| Nephrolithiasis | 0 | 0 | 1 | 1 (2.0) | 0 | 0 |
Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, γ-glutamyl transferase; SAE, serious adverse event; TEAE, treatment-emergent adverse event; TRAE, treatment-related adverse event; TSH, thyroid-stimulating hormone.
Discussion
The ALSFRS-R scoring system is a frequently used scale consisting of 12 subparameters that assess the functional status and disease progression of patients with ALS.27,28 In this phase 2 randomized clinical trial, tetramethylpyrazine nitrone did not significantly improve the primary end point of ALSFRS-R score at 180 days, and the results were similar across all prespecified sensitivity analyses.
Although each subparameter contributes equally to the total ALSFRS-R score, the importance of the 12 subparameters varies in clinical practice. Bulbar and respiratory functions are highly correlated with the quality of life and survival of patients with ALS. Nearly all patients with ALS eventually die from respiratory failure. The ALSFRS-R bulbar and respiratory subdomain scores exhibited differences of 11.1% and 20.7% between the high-dose tetramethylpyrazine nitrone and placebo groups, respectively. High-dose tetramethylpyrazine nitrone slightly delayed the deterioration of FVC in patients with ALS.
In the slower progression subgroup, high-dose tetramethylpyrazine nitrone significantly decreased the decline in ALSFRS-R bulbar subdomain scores. Additionally, both tetramethylpyrazine nitrone groups showed significant reductions in the decline of ALSFRS-R respiratory subdomain scores, with placebo-adjusted improvements of 76.1% and 85.9%, respectively. These results suggest that tetramethylpyrazine nitrone may delay the deterioration of bulbar and respiratory function, especially in patients experiencing slow progression. Compared with placebo, the time to end point events increased by 51.5% in the low-dose tetramethylpyrazine nitrone group and by 84.8% in the high-dose tetramethylpyrazine nitrone group. Additional clinical trials are needed to confirm the beneficial effects seen during this trial.
Muscle weakness and atrophy are the main characteristics of ALS, leading to functional impairment and respiratory failure.24 Grip strength, a measure of muscle strength, is associated with ALS progression. Currently, FDA-approved drugs for ALS do not meaningfully improve grip strength. Riluzole prolongs survival by a few months but does not enhance symptoms or functionality. Edaravone’s efficacy is inconsistent, and it is not approved in Europe due to limited effectiveness.6,29,30 AMX0035 failed to meet its end points in a phase 3 trial.31 Tofersen, an antisense oligonucleotide, reduced SOD1 levels but did not show significant clinical improvements in ALSFRS-R scores, FVC, or grip strength during the initial 6 months of treatment. However, in the long-term follow-up of the open-label extension study, a potential benefit for patients who began tofersen treatment earlier in the disease course was indicated.8 In this trial, high-dose tetramethylpyrazine nitrone significantly slowed patients’ decline in mean grip strength between both hands compared with placebo in both the full analysis and per-protocol sets. This effectiveness of tetramethylpyrazine nitrone in grip strength was more pronounced in the slower progression subgroup for both sets, highlighting the robustness of this assessment. The positive impact of tetramethylpyrazine nitrone on grip strength is clinically significant, suggesting potential quality-of-life benefits for patients with ALS.
As ALS is a progressive neurodegenerative disease that affects motor neurons, leading to muscle weakness, paralysis, and death, drugs that increase the force of contraction of skeletal muscle may be of benefit. The ability of tetramethylpyrazine nitrone to penetrate the blood-brain barrier and achieve therapeutic concentrations in the brain is unique. Preclinical studies have shown tetramethylpyrazine nitrone’s multifunctional mechanism in protecting motor neurons and enhancing muscle strength by scavenging reactive oxygen species, upregulating the AMPK/PGC-1α/Nrf2/HO-1 pathway, and inhibiting mammalian target of rapamycin to induce autophagy, leading to SOD1 and TDP-43 protein aggregate degradation.23,24 Despite not meeting the primary end point of ALSFRS-R change, tetramethylpyrazine nitrone significantly delayed grip strength deterioration, an important secondary end point and key marker of ALS disease progression and patients’ quality of life.
Adverse events and laboratory changes were similar across all groups. In addition, the safety outcomes are consistent with the previous phase 2 clinical trials of oral tetramethylpyrazine nitrone for diabetic kidney disease, where no significant safety concerns were noted.32 It should be noted that patients with severe hepatic and kidney impairment were excluded from this study.
Limitations
This study has several limitations. In SOD1G93A ALS mice, tetramethylpyrazine nitrone mitigated the loss of spinal motor neurons, the glial response, muscle fiber denervation, and fibrosis.23 Additionally, tetramethylpyrazine nitrone extended survival in TDP-43M337V mice injected bilaterally during the early stages of ALS.24 The objective of this phase 2 study was to maintain functionality and quality of life for patients with early-stage ALS, and long-term survival outcomes were not assessed due to the constraints on treatment and observation periods. Oxidative stress, mitochondrial dysfunction, and impaired autophagy are all potential factors contributing to ALS pathophysiology, from its initial to advanced stages. This study did not evaluate the effectiveness of tetramethylpyrazine nitrone in a broader cohort of patients with advanced-stage ALS. Furthermore, there was a lack of data on neurofilament light chains in cerebrospinal fluid or plasma, which would be necessary to assess the impact of tetramethylpyrazine nitrone on neurodegeneration.
Conclusions
In this multicenter, phase 2, double-masked, placebo-controlled randomized clinical trial, tetramethylpyrazine nitrone was both safe and well-tolerated in patients with ALS. Although there was no significant improvement in the primary end point of change in ALSFRS-R score from baseline to day 180, patients treated with tetramethylpyrazine nitrone achieved a significant delay in the deterioration of grip strength compared with placebo. Subgroup analysis demonstrated that tetramethylpyrazine nitrone was particularly effective in enhancing motor and respiratory function in patients younger than 65 years with slow disease progression.
Trial Protocol
eTable 1. Clinical Sites and Investigator Information
eTable 2. Inclusion and Exclusion Criteria
eTable 3. Change in ALSFRS-R Scores From Baseline at Days 30, 90, and 180 in the Full Analysis Set Population
eTable 4. Sensitivity Analysis: Changes in ALSFRS-R Scores From Baseline at Days 30, 90, and 180 in the Full Analysis Set Population
eTable 5. Sensitivity Analyses for Primary and Secondary End Points in the Per-Protocol Population
eFigure. Forest Plot of ALSRS-R Score Changes With Tetramethylpyrazine Nitrone From Baseline to Day 180 in Subgroups of the Full Analysis Set
Nonauthor Collaborators
Data Sharing Statement
References
- 1.Hardiman O, van den Berg LH, Kiernan MC. Clinical diagnosis and management of amyotrophic lateral sclerosis. Nat Rev Neurol. 2011;7(11):639-649. doi: 10.1038/nrneurol.2011.153 [DOI] [PubMed] [Google Scholar]
- 2.Brown RH, Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med. 2017;377(2):162-172. doi: 10.1056/NEJMra1603471 [DOI] [PubMed] [Google Scholar]
- 3.Bensimon G, Lacomblez L, Meininger V; ALS/Riluzole Study Group . A controlled trial of riluzole in amyotrophic lateral sclerosis. N Engl J Med. 1994;330(9):585-591. doi: 10.1056/NEJM199403033300901 [DOI] [PubMed] [Google Scholar]
- 4.Dharmadasa T, Kiernan MC. Riluzole, disease stage and survival in ALS. Lancet Neurol. 2018;17(5):385-386. doi: 10.1016/S1474-4422(18)30091-7 [DOI] [PubMed] [Google Scholar]
- 5.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: 10.1002/14651858.CD001447.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Writing G, Edaravone ALSSG; Writing Group; Edaravone (MCI-186) ALS 19 Study Group . 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):505-512. doi: 10.1016/S1474-4422(17)30115-1 [DOI] [PubMed] [Google Scholar]
- 7.Paganoni S, Hendrix S, Dickson SP, et al. Long-term survival of participants in the CENTAUR trial of sodium phenylbutyrate-taurursodiol in amyotrophic lateral sclerosis. Muscle Nerve. 2021;63(1):31-39. doi: 10.1002/mus.27091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Miller TM, Cudkowicz ME, Genge A, et al. ; VALOR and OLE Working Group . Trial of antisense oligonucleotide tofersen forSOD1 ALS. N Engl J Med. 2022;387(12):1099-1110. doi: 10.1056/NEJMoa2204705 [DOI] [PubMed] [Google Scholar]
- 9.Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol. 2011;7(11):616-630. doi: 10.1038/nrneurol.2011.152 [DOI] [PubMed] [Google Scholar]
- 10.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: 10.3389/fnins.2014.00252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Robelin L, Gonzalez De Aguilar JL. Blood biomarkers for amyotrophic lateral sclerosis: myth or reality?Biomed Res Int. 2014;2014:525097. doi: 10.1155/2014/525097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lin J, Wang Q, Zhou S, Xu S, Yao K. Tetramethylpyrazine: a review on its mechanisms and functions. Biomed Pharmacother. 2022;150:113005. doi: 10.1016/j.biopha.2022.113005 [DOI] [PubMed] [Google Scholar]
- 13.Sun Y, Jiang J, Zhang Z, et al. Antioxidative and thrombolytic TMP nitrone for treatment of ischemic stroke. Bioorg Med Chem. 2008;16(19):8868-8874. doi: 10.1016/j.bmc.2008.08.075 [DOI] [PubMed] [Google Scholar]
- 14.Meng Z, Chen H, Meng S. The roles of tetramethylpyrazine during neurodegenerative disease. Neurotox Res. 2021;39(5):1665-1677. doi: 10.1007/s12640-021-00398-y [DOI] [PubMed] [Google Scholar]
- 15.Lees KR, Zivin JA, Ashwood T, et al. ; Stroke-Acute Ischemic NXY Treatment (SAINT I) Trial Investigators . NXY-059 for acute ischemic stroke. N Engl J Med. 2006;354(6):588-600. doi: 10.1056/NEJMoa052980 [DOI] [PubMed] [Google Scholar]
- 16.Zivin JA. Clinical trials of neuroprotective therapies. Stroke. 2007;38(2):791-793. doi: 10.1161/01.STR.0000252090.44428.82 [DOI] [PubMed] [Google Scholar]
- 17.Kuroda S, Tsuchidate R, Smith ML, Maples KR, Siesjö BK. Neuroprotective effects of a novel nitrone, NXY-059, after transient focal cerebral ischemia in the rat. J Cereb Blood Flow Metab. 1999;19(7):778-787. doi: 10.1097/00004647-199907000-00008 [DOI] [PubMed] [Google Scholar]
- 18.Zhang Z, Zhang G, Sun Y, et al. Tetramethylpyrazine nitrone, a multifunctional neuroprotective agent for ischemic stroke therapy. Sci Rep. 2016;6:37148. doi: 10.1038/srep37148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhang T, Gu J, Wu L, et al. Neuroprotective and axonal outgrowth-promoting effects of tetramethylpyrazine nitrone in chronic cerebral hypoperfusion rats and primary hippocampal neurons exposed to hypoxia. Neuropharmacology. 2017;118:137-147. doi: 10.1016/j.neuropharm.2017.03.022 [DOI] [PubMed] [Google Scholar]
- 20.Zhang G, Zhang T, Li N, et al. Tetramethylpyrazine nitrone activates the BDNF/Akt/CREB pathway to promote post-ischaemic neuroregeneration and recovery of neurological functions in rats. Br J Pharmacol. 2018;175(3):517-531. doi: 10.1111/bph.14102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhang G, Zhang T, Wu L, et al. Neuroprotective effect and mechanism of action of tetramethylpyrazine nitrone for ischemic stroke therapy. Neuromolecular Med. 2018;20(1):97-111. doi: 10.1007/s12017-018-8478-x [DOI] [PubMed] [Google Scholar]
- 22.Zhang G, Zhang F, Zhang T, et al. Tetramethylpyrazine nitrone improves neurobehavioral functions and confers neuroprotection on rats with traumatic brain injury. Neurochem Res. 2016;41(11):2948-2957. doi: 10.1007/s11064-016-2013-y [DOI] [PubMed] [Google Scholar]
- 23.Wen J, Li S, Zheng C, et al. Tetramethylpyrazine nitrone improves motor dysfunction and pathological manifestations by activating the PGC-1α/Nrf2/HO-1 pathway in ALS mice. Neuropharmacology. 2021;182:108380. doi: 10.1016/j.neuropharm.2020.108380 [DOI] [PubMed] [Google Scholar]
- 24.Huang C, Li J, Zhang G, et al. TBN improves motor function and prolongs survival in a TDP-43M337V mouse model of ALS. Hum Mol Genet. 2021;30(16):1484-1496. doi: 10.1093/hmg/ddab101 [DOI] [PubMed] [Google Scholar]
- 25.World Medical Association . World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi: 10.1001/jama.2013.281053 [DOI] [PubMed] [Google Scholar]
- 26.Announcement of the issuance of the quality management norms for drug clinical trials. National Medical Products Administration and National Health Commission ; 2020. Accessed January 17, 2025.https://www.gov.cn/zhengce/zhengceku/2020-04/28/content_5507145.htm
- 27.The ALS CNTF Treatment Study (ACTS) Phase I-II Study Group . The Amyotrophic Lateral Sclerosis Functional Rating Scale. assessment of activities of daily living in patients with amyotrophic lateral sclerosis. Arch Neurol. 1996;53(2):141-147. doi: 10.1001/archneur.1996.00550020045014 [DOI] [PubMed] [Google Scholar]
- 28.Cedarbaum JM, Stambler N, Malta E, et al. ; BDNF ALS Study Group (Phase III) . The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. J Neurol Sci. 1999;169(1-2):13-21. doi: 10.1016/S0022-510X(99)00210-5 [DOI] [PubMed] [Google Scholar]
- 29.Witzel S, Maier A, Steinbach R, et al. ; German Motor Neuron Disease Network (MND-NET) . Safety and effectiveness of long-term intravenous administration of edaravone for treatment of patients with amyotrophic lateral sclerosis. JAMA Neurol. 2022;79(2):121-130. doi: 10.1001/jamaneurol.2021.4893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lunetta C, Moglia C, Lizio A, et al. ; EDARAVALS Study Group . The Italian multicenter experience with edaravone in amyotrophic lateral sclerosis. J Neurol. 2020;267(11):3258-3267. doi: 10.1007/s00415-020-09993-z [DOI] [PubMed] [Google Scholar]
- 31.Paganoni S, Macklin EA, Hendrix S, et al. Trial of sodium phenylbutyrate-taurursodiol for amyotrophic lateral sclerosis. N Engl J Med. 2020;383(10):919-930. doi: 10.1056/NEJMoa1916945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yang J. Safety and efficacy of tetramethylpyrazine nitrone in treating patients with type 2 diabetes and microalbuminuria—a multicenter, randomized, double-blind, placebo-controlled phase II clinical trial. Diabetes. 2022;71(suppl 1):15-LB. doi: 10.2337/db22-15-LB [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Trial Protocol
eTable 1. Clinical Sites and Investigator Information
eTable 2. Inclusion and Exclusion Criteria
eTable 3. Change in ALSFRS-R Scores From Baseline at Days 30, 90, and 180 in the Full Analysis Set Population
eTable 4. Sensitivity Analysis: Changes in ALSFRS-R Scores From Baseline at Days 30, 90, and 180 in the Full Analysis Set Population
eTable 5. Sensitivity Analyses for Primary and Secondary End Points in the Per-Protocol Population
eFigure. Forest Plot of ALSRS-R Score Changes With Tetramethylpyrazine Nitrone From Baseline to Day 180 in Subgroups of the Full Analysis Set
Nonauthor Collaborators
Data Sharing Statement
