Skip to main content

Safety and efficacy of first-in-man intrathecal injection of human astrocytes (AstroRx®) in ALS patients: phase I/IIa clinical trial results

Abstract

Background

Malfunction of astrocytes is implicated as one of the pathological factors of ALS. Thus, intrathecal injection of healthy astrocytes in ALS can potentially compensate for the diseased astrocytes. AstroRx® is an allogeneic cell-based product, composed of healthy and functional human astrocytes derived from embryonic stem cells. AstroRx® was shown to clear excessive glutamate, reduce oxidative stress, secrete various neuroprotective factors, and act as an immunomodulator. Intrathecal injection of AstroRx® to animal models of ALS slowed disease progression and extended survival. Here we report the result of a first-in-human clinical study evaluating intrathecal injection of AstroRx® in ALS patients.

Methods

We conducted a phase I/IIa, open-label, dose-escalating clinical trial to evaluate the safety, tolerability, and therapeutic effects of intrathecal injection of AstroRx® in patients with ALS. Five patients were injected intrathecally with a single dose of 100 × 106 AstroRx® cells and 5 patients with 250 × 106 cells (low and high dose, respectively). Safety and efficacy assessments were recorded for 3 months pre-treatment (run-in period) and 12 months post-treatment (follow-up period).

Results

A single administration of AstroRx® at either low or high doses was safe and well tolerated. No adverse events (AEs) related to AstroRx® itself were reported. Transient AEs related to the Intrathecal (IT) procedure were all mild to moderate. The study demonstrated a clinically meaningful effect that was maintained over the first 3 months after treatment, as measured by the pre-post slope change in ALSFRS-R. In the 100 × 106 AstroRx® arm, the ALSFRS-R rate of deterioration was attenuated from − 0.88/month pre-treatment to − 0.30/month in the first 3 months post-treatment (p = 0.039). In the 250 × 106 AstroRx® arm, the ALSFRS-R slope decreased from − 1.43/month to − 0.78/month (p = 0.0023). The effect was even more profound in a rapid progressor subgroup of 5 patients. No statistically significant change was measured in muscle strength using hand-held dynamometry and slow vital capacity continued to deteriorate during the study.

Conclusions

Overall, these findings suggest that a single IT administration of AstroRx® to ALS patients at a dose of 100 × 106 or 250 × 106 cells is safe. A signal of beneficial clinical effect was observed for the first 3 months following cell injection. These results support further investigation of repeated intrathecal administrations of AstroRx®, e.g., every 3 months.

Trial Registration: NCT03482050.

Background

Amyotrophic lateral sclerosis (ALS) is characterized by the loss of both upper and lower motor neurons (MNs). The symptoms include progressive paralysis of MN target muscles. The disease is incurable, and fatal within 3–5 years of first symptoms, usually due to respiratory failure when the diaphragm is affected [1]. The three FDA-approved drugs for the treatment of ALS, riluzole, edaravone, and the recently approved drug Relivrio™ (a combination of sodium phenylbutyrate/taurursodiol) have a modest effect on survival and disease progression [2,3,4,5,6], thus there is an urgent unmet need for therapies that can further delay the pathogenic process.

The pathological mechanisms of ALS are still not well understood and the proposed mechanisms include inflammation, oxidative stress, glutamate cytotoxicity, and protein aggregation. Although Motor Neurons (MNs) are the main affected cells in the disease, a growing body of evidence suggests the involvement of astrocytes in the pathogenesis of ALS in a non-cell-autonomous pathway [7, 8]. In healthy conditions, astrocytes support neurons in various ways. Astrocytes regulate the concentration of neurotransmitters and ions, supply a variety of metabolites and energy, regulate osmolarity, modulate synaptic activity, secrete neurotrophic and neuroprotective factors, promote neurogenesis [9, 10] and remyelination [11], and play a role in immunomodulation [12]. The contribution of astrocytes to the pathology of ALS is probably a combination of loss of homeostatic functions and/or gain of toxic functions [8, 8,13,14,15]. Recent studies also provide evidence for the beneficial role that astrocytes play in protecting MNs in ALS by reducing TDP-43 aggregates and secretion of neuroprotective factors [16,17,18,19]. Interestingly, correction of a pathogenic germline mutation in astrocytes alone slowed down MN degeneration [20]. Comprehensive preclinical studies demonstrated that transplantation of glial-precursor-cells that were generated from iPSCs, or embryonic-stem-cells (ESC), had the potential to delay disease onset and ameliorate clinical symptoms in rodent models of ALS disease [21,22,23] and shown to be safe [19]. Thus, transplantation of healthy astrocytes into the CNS of ALS patients could potentially compensate for malfunctioning endogenous astrocytes and attenuate the progression of the disease.

Pluripotent human embryonic stem cells are an excellent source for regenerative therapies as they can be produced in high quantities and can differentiate into most cell types of the body, including astrocytes [24], human astrocytes derived from clinical-grade embryonic stem cells demonstrated activities of functional healthy astrocytes, including glutamate uptake, secretion of various neurotrophic factors (e.g. GDNF, BDNF, TIMP-1, TIMP-2, and Midkine), promotion of axon outgrowth, immunomodulation and protection of MNs from oxidative stress [19]. Intrathecal injections of AstroRx® into transgenic hSOD1G93A mice and rats significantly delayed disease onset and improved motor performance, as compared to control animals. A nine-month safety study in immunodeficient mice demonstrated the safety of AstroRx® treatment, as well as the biodistribution and survival of the cells upon intrathecal administration [19].

Here we report on the results of phase I/IIa, open-label, dose-escalating clinical study to evaluate the safety, tolerability, and therapeutic effects of intrathecal injection of AstroRx® cells in patients with amyotrophic lateral sclerosis.

Methods

Standard protocol approvals, registrations, and patient consent.

The study protocols were approved by the Israeli Ministry of Health (IMOH), and the institutional review board of Hadassah Medical Center in Jerusalem, Israel. All patients signed informed consent documents before the screening.

Study objectives

The study aimed to evaluate the safety, tolerability, and therapeutic effects (preliminary efficacy) of a single intrathecal injection of low and high doses of AstroRx®, as a treatment for patients with ALS.

Patients' selection criteria

Eligible participants were aged 18–70 years with a diagnosis of probable or definite ALS by revised El Escorial Criteria, within two years of diagnosis. The ALSFRS-R score was ≥ 30, and slow vital capacity (SVC) was ≥ 70% of the predicted normal value for height, age, and sex. Participants were either not receiving riluzole and/or edaravone or were on a stable dose for ≥ 30 days. Potential patients were excluded for the following reasons: past infection or a positive test for HBV, HCV, or HIV, need for respiratory support, renal failure, impaired hepatic function, Body Mass Index (BMI) of < 18.5 or > 30, significant cardiac disease, diabetes, autoimmune diseases, chronic severe infection, malignant disease or any other disease or condition that may risk the patient or interfere with the ability to interpret the study results. A full list of inclusion and exclusion criteria is shown in Additional file 1.

Study outline

A diagram of the study design is shown in Fig. 1a. The study was conducted under 2 sequential clinical protocols, the interventional protocol Astro-001 and its extension, the non-interventional protocol, Astro-002.

Fig. 1
figure 1

Phase 1/2a study design and study flowchart. A Visit 0 (V0) - screening visit, visit 1 (V1) till visit 4 (V4) presents about 3 months run-in period (pre-treatment), AstroRx injection was performed on V4. V4 till visit V10 is the 6 months follow up time under ASTRO-001 study and additional 6 months follow up was performed under study ASTRO-002 on V10-V12 and by phone call. B Study flow chart of patient allocation, treatment doses of ASTRO-001 and ASTRO-002. V Visit, mo Month, BCV Blood Count Visit, EOS End of Study

Study Astro-001: following enrollment, the patients were monitored monthly during a run-in period of about 3 months to determine the progression rate of their ALS disease. Following the run-in period, patients were administered with 100 × 106 AstroRx® cells (Group A, n = 5 patients) and 250 × 106 cells (Group B, n = 5 patients) by a standard LP procedure. The immunosuppressive drug, Mycophenolate Mofetil (MMF) at 1 gr b.i.d. was given 2 days before the intrathecal cell injection and continued for an additional month (total of 32 days). Patients underwent weekly complete blood count (CBC) during the month of MMF treatment, and twice monthly following MMF cessation, to check for leukopenia.

After intrathecal AstroRx® injection, the patients were monitored monthly during a follow-up of 6 months. Upon completion of study Astro-001 (at a 6-month follow-up), participants were offered enrolment in the extension study Astro-002.

Under the study Astro-002, each patient was followed up monthly for an additional 6 months by either on-site visits or phone calls. The outcome measures were similar to study Astro-001. Safety data were monitored by the study investigators, medical monitor, and the Data and Safety Monitoring board (DSMB), which was independent of the study and sponsor.

The initial study design consisted of two additional arms of repeated doses of 100 × 106 cells and 250 × 106 separated by an interval of 60 days. However, due to the COVID-19 pandemic restrictions and the perception of the potential risks it posed to people with ALS, we adopted the recommendation of the independent DSMB and did not treat these 2 cohorts.

AstroRx® Cell manufacturing

The clinical-grade AstroRx® cell product derived from human embryonic stem cells was manufactured under cGMP conditions in Kadimastem's GMP facility using standard operating procedures. AstroRx® cells were freshly prepared, harvested, and formulated in PlasmaLyte to reach a volume of 5 ml with 100 × 106 or 250 × 106 AstroRx® cells. The formulated drug product was uploaded into a 10 ml syringe and transported from the manufacturing facility to the clinical site in a validated shipping system at a controlled temperature of 2–8 °C and administered to the patient within 24 h from formulation. Validated safety quality control tests were performed before the release of each formulated AstroRx cell product before delivery to the clinical site, including sterility, mycoplasma, endotoxin, and Gram or HBL test performed by external qualified certified GLP laboratory (Hylabs laboratories, Israel). The viability and cell concentration were determined using an automated cell counter Nucleocounter (NC-200™ Chemometec®). The identity of AstroRx cell product was assessed by flow cytometry using the following antibodies: anti-GLAST (Miltenibiotec, 1:100), anti- CD44 (BD Pharmingen, 1:50), and anti-GFAP (Miltenibiotec, 1:50). Antibodies against SSEA-4 and EPCAM (both from Biolegend) were used for the detection of any pluripotent marker impurities. The Flow cytometer FACS Canto II (BD) operated with FACSDIVA software (BD) was used for the analysis. To assess AstroRx® potency in-vitro, AstroRx® cells secretion of Midkine and TIMP-1 was determined by ELISA using Human TIMP-1 Quantikine ELISA Kit (R&D systems) and Human Midkine ELISA Kit (Abcam). The optical density was read using the iMark Microplate reader (Bio-Rad Laboratories). A certificate of analysis was generated and approved by the quality assurance department to ensure that each released product met the release criteria before it was delivered to the clinical site for Intrathecal injection.

Measurement outcomes

The primary objective of this study was to assess the safety and tolerability of AstroRx® in patients with ALS. Safety laboratory assessments were performed, and adverse events (AEs) were recorded at each visit. In addition, CNS imaging by MRI and CT scans was performed around 1 month before treatment, 1 month after treatment (CT), and 6 months after treatment (MRI).

The second objective was to evaluate the efficacy of intrathecal injection of AstroRx® in ALS. For this aim, data on ALSFRS-R, predicted slow vital capacity (SVC), hand-held dynamometry (HHD), and grip strength (using JAMAR plus) were collected at all pre-treatment and post-treatment on-site visits. ALSFRS-R score was also performed during home and phone visits. All tests were performed by trained evaluators who were certified by the Outcomes and Monitoring Center for the Northeast ALS Consortium. In addition, levels of the serum biomarker creatinine, creatine kinase, and neurofilament light chain (Nfl) were assessed in selected visits before and after treatment (Additional file 1).

Serum neurofilament analysis

Serum samples for the analysis of Nfl as a biomarker for ALS progression were collected. In study Astro-001-IL, samples were collected on Visit 2 (day −60), Visit 3 (day −30), Visit 4a (day −1), Visit 4d (day + 1), Visit 5 (day + 30), Visit 7 (day + 90) and Visit EOS (day + 180). In the extension study Astro-002-IL, samples were collected at the Screening Visit (day + 180; EoS Visit in protocol Astro-001-IL), Visit 9 M (day + 270), and EOS Visit (day + 365). The measurement of the concentration of the biomarker in serum was performed using the validated SIOMA (Single Molecule Array) method by Quanterix (US).

Statistical analysis

The primary trial outcome was safety, assessed for the incidence of treatment-emergent AEs (TEAEs) and serious AEs (SAEs), laboratory abnormalities, vital signs, ECGs, physical examinations, and CNS imaging. Medical history and AEs were coded using the Medical Dictionary for Regulatory Activities (MedDRA version 21). The Intent to Treat (ITT) population of the study included all subjects who were eligible to be enrolled to receive any study treatment. The Safety Analysis Set included a subset of the ITT set who had undergone intrathecal injection of AstroRx®.

The efficacy analyses were performed on the Modified ITT (mITT) population, which included a subset of the ITT that had undergone intrathecal injection of AstroRx® and had at least one post-baseline efficacy assessment. The baseline was defined for each subject as the last available, valid, non-missing assessment before the first study treatment administration. Efficacy endpoints were analyzed for the change between pre-treatment (run-in) period and post-treatment period. The nominal α level was 2-sided using α = 0.05. Since the study was an open-label exploratory study, no formal correction for type I error due to multiplicity was performed.

The Slope Analysis compared the slope of the pre-treatment period and the slope of the post-treatment period. The analysis compared the two period's slopes taking into account the treatment groups and each time point within a specific period. The actual parameter value at each time point was analyzed using a Mixed Model for Repeated Measures (MMRM) analysis (SAS®). The model included intercept and the time-point in continuous months (slope) as random effects and the following fixed effects and all their interactions: the period in the study as a class variable and the treatment group as a class. The changes from pre-treatment to post-treatment were explored by the estimated slopes resulting from the triple interaction of time point by period by group. An unstructured covariance structure was assumed and the denominator degrees of freedom were computed using the Kenward-Roger method. The change from baseline was analyzed using MMRM analysis (SAS®). The model included the fixed effects of the treatment group and scheduled visit as a categorical variable and their interaction. The model used the unstructured covariance matrix, the Restricted Maximum-Likelihood (REML) estimation method, and the Kenward-Roger adjustment method for the degrees of freedom.

Results

Twenty-four patients were screened in this clinical study. Six patients failed the screening, mostly because they did not meet the minimum respiratory criterion of predicated SVC ≥ 70% or at least 10/12 in the ALSFRS-R respiratory sub-score. Additional 2 patients were screened but not enrolled, due to study discontinuation following the COVID-19 pandemic outbreak. Five patients enrolled in Group A (a single administration of 100 × 106 AstroRx® cells). Four patients completed the 6-month follow-up under protocol Astro-001-IL, and 3 of them continued to the extension study Astro-002-IL and completed the entire 12-month follow-up after treatment. Eight patients enrolled in Group B (a single administration of 250 × 106 AstroRx® cells) and 5 patients were treated. All 5 patients completed the 6-month follow-up and continued to the extension study, and 3 of them completed the entire 12-month follow-up. Three patients enrolled in Group C (2 administrations of 250 × 106 AstroRx® cells). However, due to the COVID-19 outbreak, it was decided to discontinue the study for Group C, and no patient was treated in this group (Fig. 1b).

Before its injection, each formulated AstroRx® cell product was tested for the number of cells, viability, sterility profile, astrocytic cell identity, impurities, and potency to ensure release criteria defined for clinical batches are met (Table 1).

The baseline characteristics of the patients are presented in Table 2. Nine of the 10 treated patients of Group A and Group B were male, and all patients were white. All patients were stable on riluzole and none was treated with edaravone. The average age of the patients was 63 ± 4.9 in Group A and 61 ± 6.2 in Group B. All 5 patients in Group A had a diagnosis of probable ALS by El Escorial Criteria. In Group B, 3 patients had a diagnosis of probable ALS and 2 patients had a diagnosis of definite ALS. Nine of the 10 patients reported limb-onset of disease and 1 patient (Group B) reported a bulbar onset. The time from diagnosis was 14.5 ± 4.6 and 10.6 ± 2.0 months for Group A and Group B, respectively.

Table 1 Cell information
Table 2 Patients’ Baseline Demographics

Safety

Nine out of 10 (90%) of treated patients completed the 6-month follow-up, and 6 patients (60%) completed the 12-month follow-up. One patient in Group A and 2 patients in Group B died during the study, between 9 to 10 months post-treatment, due to respiratory failure that was associated with the natural progression of ALS. Table 3 summarizes the treatment-emergent adverse events (TEAE) reported in the study. All patients reported a total of 86 treatment-emergent adverse events (TEAE). None of TEAE was deemed to be associated with AstroRx® itself. Sixty-three TEAEs were mild, 19 were moderate, and 4 were severe. Six patients developed a total of 9 serious TEAE after the treatment, 2 patients in Group A and 4 patients in Group B (Additional file 2: Table S1). The most common TEAEs that were reported by at least 20% of the patients from Group A and Group B are shown in Additional file 2: Table S2. The Most frequent TEAE was post lumbar puncture (LP) headache, associated with IT injection procedure of the cells, and reported by 50% of the patients. Additional procedure-related TEAEs included pain in the injection site (30%), arthralgia, back pain, muscle contraction, and pain in the leg, each reported by 10% of the patients (Additional file 2: Table S3). All procedure-related AEs were graded as mild to moderate, and all were resolved. One event of moderate post-LP headache was resolved following a blood patch procedure that required hospitalization and was classified as an SAE. There was no apparent difference in the frequency or the nature of the procedure-related AEs between treatment groups. Three patients reported 4 AEs were related to mycophenolate mofetil, including headache, nausea, anemia, and hyperhidrosis (Additional file 2: Table S4). All the immunosuppression-related AEs were graded as mild to moderate, and all were resolved. No clinically significant changes were observed throughout the study in laboratory assessments, as well as in vital signs, physical examinations, or ECG results. MRI scans of the brain and spinal cord performed 6 months after AstroRx® cell injection showed no tumor formation in the CNS. Results were similar also after 12 months of follow-up, however, the MRI data at 12 months were very limited due to the inability of patients to perform MRI because of their medical condition, and restrictions imposed by the COVID-19 pandemic.

Table 3 Summary of TEAEs

Efficacy

ALSFRS-R

The main outcome efficacy measure in the study was ALSFRS-R. At baseline visit (1 day before treatment) the mean ALSFRS-R score was 35.6 ± 3.7, 34.2 ± 7.0, 34.9 ± 5.3, and 33.4 ± 6.4 for Group A, Group B, combined Group A + B, and Rapid Progressors, respectively. The mean decline in the ALSFRS-R slope for patients in Group A was − 0.88/month during the run-in (3–4 months before treatment). In the first 3 months after AstroRx® cell injection, the mean decline of the ALSFRS-R slope was attenuated to − 0.3/month (p = 0.039), reflecting an attenuation of 66% in ALSFRS-R deterioration (Fig. 2). At 6 and 12 months after treatment, the ALSFRS-R deterioration rate was − 0.76/month and − 0.82/month, respectively-similar to that observed during run-in (Fig. 2). The mean deterioration of ALSFRS-R slope in Group B (− 1.43/month) during the run-in was greater than Group A (− 0.88/month). Similar to Group A, the ALSFRS-R deterioration rate during the first 3 months after treatment decreased to − 0.78/month (p = 0.002), representing an attenuation of 45% in ALSFRS-R decline. As observed in Group A, the attenuation of ALSFRS-R decline over the first 3 months post-treatment was not maintained at 6 and 12 months post-treatment (− 1.59/month and − 1.39/month, respectively) (Fig. 2). Combining the data of both groups demonstrated an attenuation of 53% in ALSFRS-R over the first 3 months post AstroRx® IT injection (p < 0.001), which was not maintained at 6- and 12-month follow-up (Fig. 2). The change in the ALSFRS-R slope was also analyzed in a subpopulation of rapid progressors from both groups (n = 5). Rapid progressors were defined as patients who deteriorated ≥ 1.1 ALSFRS-R points per month during the run-in period [25, 26]. The mean improvement in ALSFRS-R slope between the run-in period and 3-month follow-up in these patients was 58% (− 1.58/month vs. − 0.65/month, p < 0.001). Also in this subpopulation, after 3 months post single dosing the ALSFRS-R slope returned to a similar rate that was recorded before treatment (Fig. 2). An improvement ≥ 25% in the ALSFRS-R slope is considered clinically meaningful [27]. The individual ALSFRS-R slopes (Additional file 2: Figure S1) demonstrated an improvement of at least 25% in ALSFRS-R slope between the run-in and 3-month follow-up in 80% of the patients (4 patients in each group, data not shown).

Fig. 2
figure 2

ALSFRS-R slopes analysis in run-in, and 3-, 6- and 12-month follow up after AstroRx® treatment. The change in slopes between pre-treatment slope ("Run-in") and post-treatment slope over 12 months was analyzed by using a repeated mixed model with fit least squares (LS) means (MMRM analysis). Analysis was performed on Cohort A, Cohort B, Cohort A&B as well as on rapid progressors (defined by ALSFRS-R≤1.1/month during run-in). * = P value=0.039 (Run-in vs. 3-month FU, & = P value=0.002 (Run-in vs. 3-month FU) and # = P value <0.001 (Run-in vs. 3-month FU)

Hand-held dynamometry (HHD)

A comparison of the HHD megascore slope between run-in and 3-month follow-up showed a trend of improvement in both Group A and Group B, which was not statistically significant (Additional file 2: Table S5). In combined Group A + B, the rate of HHD megascore decline in the run-in period was − 0.06 ± 0.028 vs. − 0.02 ± 0.031 during the first 3 months post-treatment (p = 0.24) and in Rapid Progressors − 0.06 ± 0.053 vs. + 0.01 ± 0.060. At 6- and 12-month follow-ups, the HHD megascore returned to the similar decline rate that was recorded in the run-in period.

Slow vital capacity

At the baseline visit, the mean percent of predicted SVC (%SVC) was 77.9 ± 14.2%, 67.8 ± 18.9%, 72.9 ± 16.6%, and 66.6 ± 17.1% for Group A, B, combined A + B, and Rapid Progressors, respectively. A comparison of %SVC rate deterioration between the run-in period and follow-up showed a continuation of %SVC deterioration in both Group A and Group B, and Rapid Progressors (Additional file 2: Table S6). The rate of decline in %SVC rate in combined Group A + B (n = 10) was –1.08 ± 1.04% in run-in vs. − 3.20 ± 1.10% during the first 3 months of follow-up (p = 0.01), and − 3.09% ± 0.59% during the 12- month follow-up (p = 0.01).

Serum neurofilament light chain (Nfl)

Serum samples for the analysis of Nfl as a biomarker for ALS disease progression were collected throughout the study, before and after treatment. Nfl, which was extensively studied in ALS, is proposed as a potential biomarker for ALS diagnosis and prognosis [28, 29]. The proposed cut-off serum Nfl level to differentially distinguish between ALS patients vs. non-neurodegenerative controls is 49 pg/mL [30, 31]. The serum Nfl concentration in six of the patients was greater than 49 pg/ml throughout the study and levels tended to be higher in rapid progressors, as reported by others [32] (data not shown). However, no clear tendency of change in the kinetics of serum Nfl was observed (Additional file 2: Figure S2).

Discussion

Although the pathogenesis of MN death in ALS is not fully elucidated, malfunctioning astrocytes can contribute to the death of MNs and the progression of the disease. A cell therapy approach that includes intrathecal injection of healthy and functional astrocytes may compensate for the diseased endogenous astrocytes and attenuate the disease progression. AstroRx® cell therapy is composed of healthy astrocytes derived from human embryonic stem cells. Intrathecal injection of AstroRx allows the distribution of the cells throughout the neural axis, where it can affect both upper and lower MNs [19]. Moreover, AstroRx® is an allogeneic “off-the-shelf” product that does not require individual production procedures as in autologous cell therapies.

This first-in-human phase I/IIa clinical trial assessed the safety and preliminary efficacy of a single intrathecal injection of AstroRx® in two doses. Ten patients were enrolled in the study, 5 in each treatment dose. No AEs related to the product itself were reported. The most common AEs were related to the intrathecal administration procedure or treatment with MMF. These AEs were mild to moderate, and all resolved either spontaneously or with treatment.

Reported SAEs were related to the expected progression of ALS. Three patients died due to the natural progression of ALS 9 to 10 months post-treatment.

A potential major safety concern in using embryonic stem cells as a source for cell therapy is their potential to form teratomas. Before its intrathecal cell injection, each AstroRx® cell product was tested to meet the acceptance criteria for pluripotent markers. MRI scans of the spinal cord and brain performed 6 months after cell injection did not reveal any tumor or teratoma formation. Overall, these safety data indicate that a single injection of AstroRx® at both tested doses of 100 × 106 and 250 × 106 is safe and well-tolerated.

The patient population enrolled in this study was at a relatively early disease stage (about 18 months from first symptoms) and the ALSFRS-R at baseline was similar between groups. The percentage of male patients in the study was 90%, profoundly greater than the estimated ALS incidence ratio of 1.29:1 between males and females [33], which may be explained by the small number of subjects included in this study. Although the known mechanisms of action of AstroRx® are assumed to influence both genders similarly, in future larger clinical studies, efforts will be made to include patients that better reflects the gender ratio in the ALS population. The disease progression recorded during the run-in period was on average greater in patients of Group B. The progression of ALS was assessed by pre-post analysis of slope analysis, change from baseline, and responder analysis. The analyses were performed also on a subpopulation of rapid progressors (ALSFRS-R ≥ 1.1/month during the run-in period). A clinically meaningful signal of decline in disease progression, as assessed by the ALSFRS-R score, was observed for the first 3 months after treatment, as compared to the pre-treatment period. Although the deterioration in Group B patients was greater than that of Group A, the trend of effect was similar. A similar trend was also observed in the rapid progressor population suggesting that AstroRx® has the potential to be effective in a broader ALS patient population. The additional outcome measures of Muscle strength as measured by HHD showed a trend of slowdown in deterioration for the first 3 months post-treatment as compared to the run-in period but was not statistically significant. In contrast, respiratory function expressed as predicated %SVC continued to deteriorate during the entire follow-up, including the first 3 months post AstroRx® injection. No clear trend of change was observed in serum marker Nfl between the pre- and post-treatment periods.

The interpretation of the efficacy results is limited by the small sample size and the difference in disease progression before treatment between study groups. Yet the trend of attenuation in disease progression for the first 3 months as reflected by ALSFRS-R was observed in 8 out of 10 patients from both groups. Notably, in larger studies supporting FDA market authorization, edaravone and the recently ALS-approved drug and Relivrio™, showed a modest but statistically significant benefit in slowing down ALSFRS-R decline, although they did not demonstrate a significant improvement in other ALS outcome measures [6, 34]. The effect of AstroRx® on ALSFRS-R, as well as the other ALS outcome measures, should be further evaluated in a larger randomized parallel, placebo-controlled clinical trial.

The duration of the effect of AstroRx® may be related to the survival of the cells in the CNS. In a preclinical study in immunodeficient mice, AstroRx® cells were shown to survive in the CNS at the pre-specified endpoint of the study 9 months after the intrathecal injection [19]. Although the CNS is generally considered an immune-privileged site, foreign antigens can still drain from the CNS to the peripheral lymph nodes through the glymphatic system and may trigger an immune reaction [35, 36]. AstroRx® is composed of allogeneic cells which can potentially elicit such an immune attack following their injection into the CSF. Clinical trials involving allogeneic cell transplantation in the CNS implement a single or combined immunosuppression regimen to avoid graft rejection [37,38,39]. In our study, transient mild immunosuppression by oral mycophenolate mofetil for one month following AstroRx® intrathecal injection was applied. Currently, there is no evidence from other clinical trials that immunosuppression changes the course of ALS disease [40,41,42]. Therefore we assume that the therapeutic benefit observed during the first 3 months following AstroRx® is not related to the concomitant immunosuppression over the first month post AstroRx® administration.

The survival of the cells in the CNS of the patient was not investigated in this clinical study. It cannot be excluded that the reduction in the clinical signal of effect after 3 months is a result of the loss of AstroRx® cells. Nevertheless, there is no indication for a systemic immune response following intrathecal AstroRx® cell injection as measured by blood immunoglobulins before and after treatment, or change in ɣ-interferon release by peripheral blood mononuclear cells (PBMC) collected from the treated patient before and after treatment (data not shown). The survival of AstroRx® cells post intrathecal cell injection and the regimen of immunosuppression will further be explored in the next clinical study.

Study limitations

The interpretation of the study results is limited by its exploratory nature and small study sample size. The study was unblinded with no control arm, making it difficult to estimate the net effect of the treatment. In addition, there was an imbalance in the pre-treatment ALSFRS-R deterioration rate between the 2 study cohorts (Cohort A − 0.88/month and B − 1.43/month), which makes it difficult to determine whether there is a difference in the effectiveness of the 2 tested doses. The pre-post analysis of the efficacy outcome measures assumes linearity in the deterioration. However, while ALS clinical trials assume linear deterioration in ALSFRS-R over time [6, 34], there is also evidence suggesting that ALS may progress in a non-linear fashion [43, 44]. This study did not assess transplant engraftment; therefore we have no data about the survival of AstroRx® in patients and the effectiveness of immunosuppression to prevent rejection.

Conclusions

In conclusion, a single IT administration of AstroRx®, an astrocyte cell-based therapy derived from embryonic stem cells, at a dose of 100 × 106 or 250 × 106 cells is considered safe. A signal of beneficial clinical effect was observed over the first 3 months post single treatment. It remains to be investigated whether repeated IT administrations of AstroRx® may prolong its beneficial effect in ALS. To further determine the clinical effect of AstroRx in ALS, additional powered, controlled clinical studies to evaluate repeated administration of AstroRx, e.g. every 3 months, are required.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AE:

Adverse event

ALS:

Amyotrophic lateral sclerosis

ALSFRS-R:

ALS Functional Rating Scale-Revised

ALSAQ-40:

ALS Assessment Questionnaire

APC:

Astrocyte progenitor cells

AQP-4:

Aquaporin-4

BCV:

Blood count visit

BDNF:

Brain-derived neurotrophic factor

b.i.d.:

Twice a day

BMI:

Body mass index

BW:

Body weight

CBC:

Complete blood count

CK:

Creatine kinase

CNS:

Central nervous system

CNTF:

Ciliary neurotrophic factor

Cr:

Creatinine

CSF:

Cerebrospinal fluid

C-SSRS:

Columbia-Suicide Severity Rating Scale

CT:

Computed tomography

CXCR:

C-X-C chemokine receptor

DSMB:

Data safety monitoring board

eCRF:

Electronic case report form

EC:

Ethics committee

ELISA:

Enzyme-linked immunosorbent assay

FU:

Follow-up

GDNF:

Glial derived neurotrophic factor

GFAP:

Glial fibrillary acidic protein

GLAST:

Glutamate aspartate transporter

hESC:

Human embryonic stem cells

HHD:

Hand-held dynamometer

ITT:

Intention to treat

IT:

Intrathecal

LP:

Lumbar puncture

MedDRA:

Medical Dictionary for Regulatory Activities

mITT:

Modified Intent to Treat

MMF:

Mycophenolate mofetil

MMRM:

Mixed-effect model for repeated measures

MN:

Motor neuron

MoH:

Ministry of Health

MRI:

Magnetic resonance imaging

Nfl:

Neurofilament light chain

OCT-4:

Octamer-binding transcription factor-4

pNFH:

Phosphorylated neurofilament heavy chain

PBMC:

Peripheral blood mononuclear cells

SAE:

Serious adverse event

SD:

Standard deviation

SEM:

Standard error of the mean

SSEA:

Stage-specific embryonic antigen

SVC:

Slow vital capacity

References

  1. Rowland LP, Shneider NA. Amyotrophic lateral sclerosis. N Engl J Med. 2001;344(22):1688–700.

    Article  CAS  PubMed  Google Scholar 

  2. Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Lancet. 1996;347(9013):1425–31.

    Article  CAS  PubMed  Google Scholar 

  3. 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.

    Article  CAS  PubMed  Google Scholar 

  4. Abe K. An early history of Japanese amyotrophic lateral sclerosis (ALS) and the current significance. Rinsho Shinkeigaku. 2017;57(4):153–62.

    Article  PubMed  Google Scholar 

  5. Yoshino H. Edaravone for the treatment of amyotrophic lateral sclerosis. Expert Rev Neurother. 2019;19(3):185–93.

    Article  CAS  PubMed  Google Scholar 

  6. Paganoni S, Macklin EA, Hendrix S, Berry JD, Elliott MA, Maiser S, Karam C, Caress JB, Owegi MA, Quick A, Wymer J, Goutman SA, Heitzman D, Heiman-Patterson T, Jackson CE, Quinn C, Rothstein JD, Kasarskis EJ, Katz J, Jenkins L, Ladha S, Miller TM, Scelsa SN, Vu TH, Fournier CN, Glass JD, Johnson KM, Swenson A, Goyal NA, Pattee GL, Andres PL, Babu S, Chase M, Dagostino D, Dickson SP, Ellison N, Hall M, Hendrix K, Kittle G, McGovern M, Ostrow J, Pothier L, Randall R, Shefner JM, Sherman AV, Tustison E, Vigneswaran P, Walker J, Yu H, Chan J, Wittes J, Cohen J, Klee J, Leslie K, Tanzi RE, Gilbert W, Yeramian PD, Schoenfeld D, Cudkowicz ME. Trial of sodium phenylbutyrate-taurursodiol for amyotrophic lateral sclerosis. N Engl J Med. 2020;383(10):919–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Johann S. Astrocytes pathology in ALS: a potential therapeutic target? Curr Pharm Des. 2017;23(33):5022–36.

    CAS  PubMed  Google Scholar 

  8. Izrael M, Slutsky SG, Revel M. Rising stars: astrocytes as a therapeutic target for ALS disease. Front Neurosci. 2020;14:824.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Allen NJ, Eroglu C. Cell biology of astrocyte-synapse interactions. Neuron. 2017;96(3):697–708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Verkhratsky A, Zorec R, Parpura V. Stratification of astrocytes in healthy and diseased brain. Brain Pathol. 2017;27(5):629–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fasciani I, Pluta P, Gonzalez-Nieto D, Martinez-Montero P, Molano J, Paino CL, Millet O, Barrio LC. Directional coupling of oligodendrocyte connexin-47 and astrocyte connexin-43 gap junctions. Glia. 2018;66(11):2340–52.

    Article  PubMed  Google Scholar 

  12. Liddelow SA, Barres BA. Reactive astrocytes: production, function, and therapeutic potential. Immunity. 2017;46(6):957–67.

    Article  CAS  PubMed  Google Scholar 

  13. Yamanaka K, Komine O. The multi-dimensional roles of astrocytes in ALS. Neurosci Res. 2018;126:31–8.

    Article  CAS  PubMed  Google Scholar 

  14. Van Harten ACM, Phatnani H, Przedborski S. Non-cell-autonomous pathogenic mechanisms in amyotrophic lateral sclerosis. Trends Neurosci. 2021;44(8):658–68.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Crabe R, Aimond F, Gosset P, Scamps F, Raoul C. How degeneration of cells surrounding motoneurons contributes to amyotrophic lateral sclerosis. Cells. 2020;9(12):2550.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Smethurst P, Risse E, Tyzack GE, Mitchell JS, Taha DM, Chen YR, Newcombe J, Collinge J, Sidle K, Patani R. Distinct responses of neurons and astrocytes to TDP-43 proteinopathy in amyotrophic lateral sclerosis. Brain. 2020;143(2):430–40.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Reichardt LF. Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci. 2006;361(1473):1545–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Poyhonen S, Er S, Domanskyi A, Airavaara M. Effects of neurotrophic factors in glial cells in the central nervous system: expression and properties in neurodegeneration and injury. Front Physiol. 2019;10:486.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Izrael M, Slutsky SG, Admoni T, Cohen L, Granit A, Hasson A, Itskovitz-Eldor J, Krush Paker L, Kuperstein G, Lavon N, Yehezkel Ionescu S, Solmesky LJ, Zaguri R, Zhuravlev A, Volman E, Chebath J, Revel M. Safety and efficacy of human embryonic stem cell-derived astrocytes following intrathecal transplantation in SOD1(G93A) and NSG animal models. Stem Cell Res Ther. 2018;9(1):152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat Neurosci. 2008;11(3):251–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kondo T, Funayama M, Tsukita K, Hotta A, Yasuda A, Nori S, Kaneko S, Nakamura M, Takahashi R, Okano H, Yamanaka S, Inoue H. Focal transplantation of human iPSC-derived glial-rich neural progenitors improves lifespan of ALS mice. Stem cell reports. 2014;3(2):242–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lepore AC, Dejea C, Carmen J, Rauck B, Kerr DA, Sofroniew MV, Maragakis NJ. Selective ablation of proliferating astrocytes does not affect disease outcome in either acute or chronic models of motor neuron degeneration. Exp Neurol. 2008;211(2):423–32.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Izrael M, Slutsky S, Itskovitz-Eldor J, Revel M. Astrocytes in pathogenesis of neurodegenerative diseases and potential: translation into clinic. InTechOpen: Astrocyte—Physiology and Pathology 2018. ISBN 978-953-51-5760-1, 93–118.

  24. Liu G, David BT, Trawczynski M, Fessler RG. Advances in pluripotent stem cells: history, mechanisms, technologies, and applications. Stem Cell Rev Rep. 2020;16(1):3–32.

    Article  PubMed  Google Scholar 

  25. Mora JS, Genge A, Chio A, Estol CJ, Chaverri D, Hernandez M, Marin S, Mascias J, Rodriguez GE, Povedano M, Paipa A, Dominguez R, Gamez J, Salvado M, Lunetta C, Ballario C, Riva N, Mandrioli J, Moussy A, Kinet JP, Auclair C, Dubreuil P, Arnold V, Mansfield CD, Hermine O, Ab10015 Study, G. 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.

    Article  CAS  PubMed  Google Scholar 

  26. Labra J, Menon P, Byth K, Morrison S, Vucic S. Rate of disease progression: a prognostic biomarker in ALS. J Neurol Neurosurg Psychiatry. 2016;87(6):628–32.

    Article  PubMed  Google Scholar 

  27. Wijesekera LC, Leigh PN. Amyotrophic lateral sclerosis. Orphanet J Rare Dis. 2009;4:3.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Forgrave LM, Ma M, Best JR, DeMarco ML. The diagnostic performance of neurofilament light chain in CSF and blood for Alzheimer’s disease, frontotemporal dementia, and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Alzheimers Dement (Amst). 2019;11:730–43.

    Article  PubMed  Google Scholar 

  29. Zhou YN, Chen YH, Dong SQ, Yang WB, Qian T, Liu XN, Cheng Q, Wang JC, Chen XJ. Role of blood neurofilaments in the prognosis of amyotrophic lateral sclerosis: a meta-analysis. Front Neurol. 2021;12: 712245.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Verde F, Otto M, Silani V. Neurofilament light chain as biomarker for amyotrophic lateral sclerosis and frontotemporal dementia. Front Neurosci. 2021;15: 679199.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Halbgebauer S, Steinacker P, Verde F, Weishaupt J, Oeckl P, von Arnim C, Dorst J, Feneberg E, Mayer B, Rosenbohm A, Silani V, Ludolph AC, Otto M. Comparison of CSF and serum neurofilament light and heavy chain as differential diagnostic biomarkers for ALS. J Neurol Neurosurg Psychiatry. 2022;93(1):68–74.

    Article  PubMed  Google Scholar 

  32. Witzel S, Frauhammer F, Steinacker P, Devos D, Pradat PF, Meininger V, Halbgebauer S, Oeckl P, Schuster J, Anders S, Dorst J, Otto M, Ludolph AC. Neurofilament light and heterogeneity of disease progression in amyotrophic lateral sclerosis: development and validation of a prediction model to improve interventional trials. Transl Neurodegener. 2021;10(1):31.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Choi JH, Chung JY, Yoo DY, Hwang IK, Yoo KY, Lee CH, Yan BC, Ahn JO, Youn HY, Won MH. Cell proliferation and neuroblast differentiation in the rat dentate gyrus after intrathecal treatment with adipose-derived mesenchymal stem cells. Cell Mol Neurobiol. 2011;31(8):1271–80.

    Article  CAS  PubMed  Google Scholar 

  34. Writing G, Edaravone ALSSG. 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.

    Article  Google Scholar 

  35. Louveau A, Harris TH, Kipnis J. Revisiting the mechanisms of CNS immune privilege. Trends Immunol. 2015;36(10):569–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mazzini L, Gelati M, Profico DC, Sgaravizzi G, Projetti Pensi M, Muzi G, Ricciolini C, Rota Nodari L, Carletti S, Giorgi C, Spera C, Domenico F, Bersano E, Petruzzelli F, Cisari C, Maglione A, Sarnelli MF, Stecco A, Querin G, Masiero S, Cantello R, Ferrari D, Zalfa C, Binda E, Visioli A, Trombetta D, Novelli A, Torres B, Bernardini L, Carriero A, Prandi P, Servo S, Cerino A, Cima V, Gaiani A, Nasuelli N, Massara M, Glass J, Soraru G, Boulis NM, Vescovi AL. Human neural stem cell transplantation in ALS: initial results from a phase I trial. J Transl Med. 2015;13:17.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Tadesse T, Gearing M, Senitzer D, Saxe D, Brat DJ, Bray R, Gebel H, Hill C, Boulis N, Riley J, Feldman E, Johe K, Hazel T, Polak M, Bordeau J, Federici T, Glass JD. Analysis of graft survival in a trial of stem cell transplant in ALS. Ann Clin Transl Neurol. 2014;1(11):900–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Salado-Manzano C, Perpina U, Straccia M, Molina-Ruiz FJ, Cozzi E, Rosser AE, Canals JM. Is the immunological response a bottleneck for cell therapy in neurodegenerative diseases? Front Cell Neurosci. 2020;14:250.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Appel SH, Stewart SS, Appel V, Harati Y, Mietlowski W, Weiss W, Belendiuk GW. A double-blind study of the effectiveness of cyclosporine in amyotrophic lateral sclerosis. Arch Neurol. 1988;45(4):381–6.

    Article  CAS  PubMed  Google Scholar 

  41. Fournier CN, Schoenfeld D, Berry JD, Cudkowicz ME, Chan J, Quinn C, Brown RH, Salameh JS, Tansey MG, Beers DR, Appel SH, Glass JD. An open label study of a novel immunosuppression intervention for the treatment of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2018;19(3–4):242–9.

    Article  CAS  PubMed  Google Scholar 

  42. Glass JD, Boulis NM, Johe K, Rutkove SB, Federici T, Polak M, Kelly C, Feldman EL. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: results of a phase I trial in 12 patients. Stem Cells. 2012;30(6):1144–51.

    Article  CAS  PubMed  Google Scholar 

  43. Gordon PH, Cheng B, Salachas F, Pradat PF, Bruneteau G, Corcia P, Lacomblez L, Meininger V. Progression in ALS is not linear but is curvilinear. J Neurol. 2010;257(10):1713–7.

    Article  PubMed  Google Scholar 

  44. Ramamoorthy D, Severson K, Ghosh S, Sachs K, Glass JD, Fournier CN, Pooled Resource Open-Access ALS Clinical Trials Consortium, ALS/MND Natural History Consortium, Todd M, Berry JD, Kenney NG, Fraenkel E. Identifying patterns in amyotrophic lateral sclerosis progression from sparse longitudinal data. Nat Comput Sci 2022; 2: 605–616.

Download references

Acknowledgements

Not applicable.

Funding

Funding from the Office of Chief Scientist, Ministry of Economy, Israel was awarded to Kadimastem Ltd. Award number: 77571.

Author information

Authors and Affiliations

Authors

Contributions

MG, YC, YL, MW: Clinical investigator, contribution to protocol design, conducting the study, and writing the manuscript. SB: Pharmacist of the study. TBH: Consultant for the design of the study and writing manuscript. SGS: Clinical Project Manager, writing of the manuscript, and design and execution of the clinical protocol. MI: Head of R&D, supervision of all activities, writing of the manuscript, and design of the clinical protocol. AH: Design of clinical protocol and establishment of study infrastructures. YS: Global clinical development advisor. GK, EE: QA and QC of the AstroRx®. TS: AstroRx® GMP manufacturing. JC, KM: Quality control support. AR: Medical advisor for the study. MR: AstroRx® invention and development, and design of the clinical protocol.

Corresponding author

Correspondence to Michal Izrael.

Ethics declarations

Ethics approval and consent to participate

The study protocols and informed consent forms were approved by the Israeli Ministry of Health (IMOH), and the institutional review board of Hadassah Medical Center in Jerusalem, Israel. All patients signed informed consent after receiving oral and written information and could withdraw from the study at any time point. The studies were performed in compliance with the World Medical Association Declaration of Helsinki and ICH E6 for Good Clinical Practice.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Supplementary materials and methods.

Additional file 2: Figure S1.

ALSFRS-R slope for each patient in run-in, and 3-, 6- and 12-month follow up after AstroRx® treatment. Figure S2. Serum levels of Nfl for each patient. Table S1. List of serious TEAEs. Table S2. Treatment emergent adverse events (TEAE) reported in at least 20% of patients in both treatment arms. Table S3. AEs related to IT injection of AstroRx cells by lumbar puncture. Table S4. AEs related to immunosuppression by Mycophenolate Mofetil. Table S5. Slope analysis of handheld dynamometer megascore. Table S6. %SVC slopes analysis in run-in, and 3-, 6- and 12-month follow up after AstroRx® treatment

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gotkine, M., Caraco, Y., Lerner, Y. et al. Safety and efficacy of first-in-man intrathecal injection of human astrocytes (AstroRx®) in ALS patients: phase I/IIa clinical trial results. J Transl Med 21, 122 (2023). https://doi.org/10.1186/s12967-023-03903-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-03903-3

Keywords