Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Meta-Anal. Jun 28, 2021; 9(3): 277-285
Published online Jun 28, 2021. doi: 10.13105/wjma.v9.i3.277
Nusinersen, an exon 7 inclusion drug for spinal muscular atrophy: A minireview
Bijaylaxmi Behera, Department of Neonatology, Chaitanya Hospital, Chandigarh 160044, India
ORCID number: Bijaylaxmi Behera (0000-0001-9061-7677).
Author contributions: Behera B performed the literature review and wrote the manuscript.
Conflict-of-interest statement: The author has no conflicts of interest and has nothing to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Bijaylaxmi Behera, MBBS, MD, Chief Physician, Department of Neonatology, Chaitanya Hospital, Sector 44, Chandigarh 160044, India. jollybubu2008@gmail.com
Received: December 3, 2020
Peer-review started: December 3, 2020
First decision: May 6, 2021
Revised: May 20, 2021
Accepted: June 17, 2021
Article in press: June 17, 2021
Published online: June 28, 2021
Processing time: 221 Days and 2.1 Hours

Abstract

Spinal muscular atrophy is an autosomal recessive neuromuscular disease with incidence of 1 in 5000 to 10000 live births and is produced by homozygous deletion of exons 7 and 8 in the SMN1 gene. The SMN1 and SMN2 genes encode the survival motor neuron protein, a crucial protein for the preservation of motor neurons. Use of the newer drug, Nusinersen, from early infancy has shown improvement in clinical outcomes of spinal muscular atrophy patients.

Key Words: CHERISH, Nusinersen, Spinal muscular atrophy, Survival motor neuron, NURTURE

Core Tip: Spinal muscular atrophy is an autosomal recessive neuromuscular disease, with incidence of 1 in 5000 to 1 in 10000 live births. This review provides an elaborative knowledge regarding the current most effective drug for spinal muscular atrophy, Nusinersen. A brief discussion on other treatment modalities that are under trials is also provided.



INTRODUCTION

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disease, with incidence of 1 in 5000 to 1 in 10000 live births[1,2]. It is caused by homozygous deletion of exons 7 and 8 in the SMN1 gene[1,2]. It is a rare genetic disease that occurs due to degeneration of motor neurons in the spinal cord and brainstem[3]. Clinical manifestations can appear prior to birth to early adulthood and usually manifests with symmetrical, proximal muscle weakness, which is progressive and associated with muscle atrophy. SMA patients have difficulties like inadequate weight gain, restrictive lung disease, scoliosis, joint contractures, and sleep problems but do not have cognitive issues. SMA causes mutation in chromosome 5q11.2-q13.3, affecting the survival motor neuron (SMN) gene, which results in lack of SMN1 exon 7. For the preservation of motor neurons, SMN protein is crucial, which is encoded by SMN1 and SMN2. A homozygous deletion of SMN1 exon 7 is diagnostic confirmation for SMA. A typical patient with SMA can have variable copies of SMN2 but have zero copies of SMN1[3-6]. The severest is type 0 and has a antenatal onset, whereas the commonest type of SMA is type1[4]. SMA type 2 presents around 6 mo to 18 mo, type 3 after 18 mo, and type 4 beyond 5 years[7]. The introduction of Nusinersen, from early infancy, has shown improvement in clinical outcomes of SMA type 1 patients. Open label phase two and phase three trials have demonstrated effective improvement of life expectancy in most of the patients[8].

STRUCTURE AND MECHANISM OF ACTION

Research have shown an intronic splicing silencer N1 (ISS-N1) sequence in intron 7 of SMN2, which has shown involvement in messenger RNA (mRNA) exon 7 skipping. Improvement in SMN2-mRNA exon 7 inclusion has been revealed by inhibiting ISS-N1 by antisense oligonucleotides (ASOs), and thus showing clinical improvement in SMA[9]. Nusinersen is a modified 2’-O-methoxyethyl phosphorothioate ASO. After binding to SMN2, it alters splicing of SMN2 pre-mRNA, which leads to rise in exon 7 incorporation into SMN2 mRNA. This results in an augmented manufacture of standard length SMN protein, which is obligatory for the preservation of motor neurons, but is lacking in SMA patients[10,11].

PHARMADYNAMICS AND PHARMACOKINETICS

Nusinersen is unable to cross the blood-brain barrier, when given through intravenous or subcutaneous routes. It is administered intrathecally as it unambiguously targets the central nervous system[10-12]. 3′ and 5′ exonuclease-mediated hydrolysis is the process of metabolism, and it is eliminated by the kidney[10,11]. The mean plasma and central nervous system terminal elimination half-lives are 63-87 d and 35-177 d[10,11]. The plasma and the cerebrospinal fluid volume of distributions are 29 L and 0.4 L[10,11]. Body weight is the only variable that affects Nusinersen pharmacokinetics. There is no dose-related toxicity established in studies so a fixed dose is recommended[10,11].

USES

In an open label phase 2 study, 20 patients with ages ranging from 3 wk to 7 mo, with infantile-onset SMA symptoms and having SMN1 homozygous gene deletion or mutation, were given several intrathecal doses of Nusinersen (6 mg and 12 mg dose). In the 12 mg dose group, there was improvement in achieving motor milestones (P < 0.0001), enhancements in CHOP-INTEND[13] motor function scores (P = 0.0013) (a dependable score for motor skills in SMA), and amplified compound muscle action potential amplitude of the ulnar nerve (P = 0.0103) and peroneal nerve (P < 0.0001)[8]. None of the patients required prolonged pulmonary support nor were any deaths reported[8]. In NURTURE study, all 25 participants achieved the ability to sit without support, 23/25 (92%) achieved walking with assistance, and 22/25 (88%) achieved walking independently[14]. Another larger “ENDEAR” randomized controlled trial comprising of 122 , ≤ 7 mo genetically proven SMA patients also revealed improvement in motor milestones, assessed by section 2 of the Hammersmith Infant Neurological Examination (HINE-2), after 13 mo of trial[15,16]. In the interim analysis, the group of patients who received Nusinersen had significant enhancement in motor-milestone response [21 of 51 infants (41%) as compared to the control group 0 of 27 (0%), P value of < 0.001]. In the treatment group, 22% achieved full head control, 10% were able to roll over, 8% were able to sit independently, and 1% were able to stand; in the control group, no infants achieved these milestones. The study also concluded that the group of patients treated with Nusinersen had substantial development in motor-milestone response [37 of 73 infants (51%) as compared to control group 0 of 37 (0%)]. In comparison to the control group the patients treated with Nusinersen had a higher likelihood of event-free survival with hazard ratio (HR) for death or the use of permanent assisted ventilation of 0.53 and P = 0.005. The Nusinersen group had a greater likelihood of overall survival (HR for death, 0.37; P = 0.004)[15].

CHERISH study, a phase 3 randomized controlled trial, reported an accelerated progress in the motor milestone achievements at 13 mo, in the treatment group of 126 of 179 screened children of age 2 years to 12 years, having a genetic diagnosis of SMA[17]. In another phase 1b/2a study with SMA children of 2-15 years of age, who received ascending multiple doses (3, 6, 9, 12 mg) of Nusinersen, the Hammersmith-Functional-Motor-Scale–Expanded, Upper-Limb-Module, 6-Minute-Walk-Test (6MWT), compound muscle action potential, and quantitative multipoint incremental motor unit number estimation and safety were used to assess enrolled patients. Eleven children of type II SMA and 17 of type III SMA were enrolled. Mean Hammersmith-Functional-Motor-Scale–Expanded scores, Upper-Limb-Module scores, and 6MWT distances improved by the day 1150 visit[18].

A cohort study of 33 children with SMA1, 8.3 mo to 113.1 mo of age, were treated with Nusinersen. Evaluation was done before starting of treatment (M0), after 2 mo (M2), and 6 mo (M6) of starting treatment. Data of survival, respiratory, nutrition, and motor function assessment with the modified HINE-2 were recorded. Six months after the treatment, median development on the modified HINE-2 score was 1.5 points (P < 0.001), and there was a significant increase in the requirement of respiratory support[19]. No serious or life threatening adverse events (AE) were noted in any trials.

Patients with SMA usually present with respiratory morbidities like hypoventilation, aspirations, repeated lung infections due to stiffened rib cage, micro-collapse of the lungs, which occurs as a result of decreased chest wall movement, and increasing thoracic scoliosis[20]. Polysomnograms have proven early type 1 respiratory failure even in stable SMA patients. Nusinersen therapy might have improvement in functioning of peripheral muscles along with improvement of respiratory muscle and diaphragmatic functions, leading to improved co-ordination of oral secretions, decrease lung aspiration, and decreased airway disease and pneumonia.

DOSE AND ADMINISTRATION

On December 23, 2016, Nusinersen was given approval as a treatment for SMA by the Food and Drug Administration (FDA). Nusinersen 6 mg dose equivalent to be diluted to a concentration 1.2 mg/mL or 12 mg dose equivalent diluted to 2.4 mg/mL with artificial cerebrospinal fluid and administered intrathecally every 14 d for three doses, followed by a fourth dose after 30 d. The FDA recommends to administer maintenance doses once every 4 mo to maintain the tissue concentration. This drug has also been approved by European Medicines Agency[21], Australia[22], New Zealand[23], and Canada[24].

Initial therapy

Individuals who meet all of the following criteria: (1) Documentation of a confirmed diagnosis of SMA by genetic testing; (2) Documentation of ≥ 2 copies of the SMN2 gene by genetic testing; and (3) Onset of SMA-associated symptoms before 20 mo of age.

Continuation therapy

After 6 mo of starting of therapy, continue Nusinersen therapy every 6 mo in patients meeting both criteria (1) and (2): (1) Initial therapy was determined to meet the above criteria; and (2) There is substantial improvement in SMA-associated symptoms during the previous treatment period. Clinical efficacy of Nusinersen has to be evaluated every 6 mo in the treated individual.

AE

As per ENDEAR study, severe AE in Nusinersen in 56% compared to 80% in controls and serious AE were 76% in Nusinersen compared to 95% in control group. Even serious AEs with fatal outcome was 39% in controls compared to only 16% in Nusinersen. Pyrexia, constipation, upper-respiratory infection, pneumonia, or respiratory distress or failure was seen in ≥ 20% in Nusinersen group[15]. Almost similar results were seen in CHERISH study[17].

DILEMAS OF NUSINERSEN
Efficacy of Nusinersen in treating type 0 SMA patients

Till date there are no studies on patients who are symptomatic patients since birth or within 7 d of life being treated with the drug. Neither are there any studies on patients with one copy of SMN2 gene being given Nusinersen therapy. Further studies are essential to demonstrate the effectiveness of this drug in these patients.

Factors affecting the response of the drug

In the CHERISH study[17], the response of Nusinersen treatment varied in SMA cases. Amongst the factors identified were the time interval when the type 1 SMA patients became symptomatic and administration of first dose of drug and the age of type 2 SMA patients. There was an extensive difference in the treatment response amongst the patients, which was not explained by these factors[15].

Type 1 SMA respiratory outcomes

None of the trials reported on swallowing evolution and even the respiratory outcomes were considered as the usage of long-lasting ventilation or not and the number of hours spent ventilated. In order to have an improved evaluation of load for families involved in the treatment of a type 1 SMA child, a better documentation of time spent in the hospital is required.

No reporting of the long-term consequences of patients who received therapy before they became symptomatic

Reports from the NURTURE study[8] have shown that children who were treated before manifesting symptoms, with three copies of SMN2, have a far improved result than when they are treated after they become symptomatic[25]. The ENDEAR study and CHERISH Nusinersen trial[15,17] have not reported on the long-term outcomes, for example the number of patients attaining major milestones of walking, standing, or sitting, whether they will be free of ventilation, and the number of patients that will have severe motor disability. Even their intellectual development assessments were not reported.

Nusinersen in type 3 and type 4 SMA

Insufficient data are available in type 3 SMA patients treated with Nusinersen, and there are no reports on type 4 SMA. A study by Montes et al[25] revealed an unremitting result on the 6MWT in type 3 patients, with a median surge of 98 m in a 1050-d period.

Nusinersen a very costly drug

According to a study the estimated price of care of a type 1 SMA patient who is not on Nusinersen therapy was roughly €100000 per year, and type 2 was nearly €90000 per year[26]. The cost of Nusinersen is nearly €100000 per injection and a full course of six injections are required in the initial year, and three during the subsequent year[27]. This drug has the probability to improve the existence of type 1 SMA patients from 10% to 60%[8,15] and also enhance their functional level; the higher cost is an important factor in deciding the treatment. So, identifying good responders of treatment early by various outcome measures and optimizing the treatment for presymptomatic patients through newborn screening could decrease the cost burden. So the most important challenge is Nusinersen’s remarkably expensive cost[28,29].

OTHER THERAPIES

Other disease modifying therapies like SMN1 gene replacement therapy, AVXS-101 (zolgensma) in trials, when delivered intravenously to SMA type I infants had promising outcomes in in vivo studies[30-32]. Although these studies have shown improved life expectancy, motor milestones, and motor functions, the number of patients were less and additional studies are essential to prove the effectiveness. Adeno-associated virus 9 vector was most efficiently able to cross the blood brain barrier, and was able to infect almost 60% of motor neurons and enhance SMN expression[28,29]. Studies have not clearly shown long term benefits with combined treatment of Zolgensma and Nusinersen[33,34].

Some other SMN2 exon 7 inclusion drug like risdiplam (RG7916), having central and peripheral tissues distributions, have proven to be a beneficial[35,36] and has been investigated by FIREFISH and SUNFISH trials[37,38]. Branaplam (LMI070) is another such drug that interacts with U1 small nuclear ribonucleoprotein and facilitates exon 7 inclusion of SMN2 transcripts and increases SMN protein levels with initial results of ongoing trials showing improvement[39,40]. Small molecular drugs like Celecoxib, a cyclooxygenase 2 inhibitor, have revealed an increase SMN in animal models[41], and an ongoing trial is recruiting patients[42]. Quinazoline (repligen or RG3039), which blocks a decapping scavenger enzyme (DcpS) and demonstrated upsurge in FL-SMN2 transcript by enhancing SMN2 promoter activity in animal studies[43,44], did not show significant results in phase trial[45,46].

Aminoglycoside antibiotics (tobramycin, geneticin, and amikacin) act as SMN protein stabilizers by masking premature stop codon mutations and increasing read-through of exon 8 and thus increasing the SMN protein[47,48] and have only shown in vivo efficacy[49]. In mouse models, BBrm2 (FDA-approved azithromycin) has revealed an upsurge in SMN and enhancement in motor function[49,50]. Similar results were shown in mouse models with Bortezomib, an ubiquitin proteasome inhibitor that inhibits SMN protein degeneration[51].

A proper SMA treatment includes both SMN-dependent and SMN-independent approaches, which will provide the central and peripheral therapies[33,52].

Neuroprotective agents like olesoxime, which has neuroprotective properties in pre-clinical studies, had suggested improvement in the function and survival of neurons[53]. Subsequently, however, they did not show significant results[52,54]. Studies with riluzole and gabapentin, did not show encouraging results[42,55-57].

As muscle weakness is eminent in SMA, SMN-independent therapies lately focus on muscle. Myostatin, a growth factor, inhibits muscle growth and by stalling its signaling pathway, muscle mass can be enhanced[58]. Follistatin[59], adeno-associated virus-mediated soluble inhibitor of activin receptor type IIB (ActRIIB)[60], in SMA mouse models and BIIB 110 (ALG 801) are undergoing a phase 1a trial[61]. In SMA mouse models[62] and phase 2 trials[63], SRK-015, a human monoclonal antibody, has shown improvement in muscle mass.

Initial trials with reldesemtiv, a skeletal muscle troponin activator, has revealed improvement in muscle function[46,64-66]. Trials are ongoing with drugs acting on neuromuscular junction, like pyridostigmine (mestinon)[67,68] and 4-aminopyridine (4-AP or ampyra)[69,70].

Studies have demonstrated that stem cell therapy in SMA provide support to degenerating motor neurons and short of functional cell replacement[71-73].

CONCLUSION

Although many therapies are under trial and some have shown some clinically beneficial results, the level of evidence is very low. Nusinersen, however, has a moderate level of evidence and has been approved by the FDA. Nusinersen can be used in diagnosed type 1, 2, and 3 SMA patients. The earlier Nusinersen is started, the better is the improvement of muscle strength and better will be the improved quality and span of life. Currently this drug is available and approved by FDA, but cost will be a limiting factor.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Medicine, research and experimental

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Kerpel-Fronius S S-Editor: Gao CC L-Editor: Filipodia P-Editor: Wang LL

References
1.  Bogari NM, Bogari FR, Rayes HH, Alqassimi NM, Balto HM, Dannoun A. Molecular Genetic Diagnosis for a family with type 1 spinal muscular atrophy (SMA) via analysis of the survival motor neuron (SMN)gene. J Rare Dis Diagn Ther. 2015;1:21.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Kesari A, Misra UK, Kalita J, Mishra VN, Pradhan S, Patil SJ, Phadke SR, Mittal B. Study of survival of motor neuron (SMN) and neuronal apoptosis inhibitory protein (NAIP) gene deletions in SMA patients. J Neurol. 2005;252:667-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
3.  Mercuri E, Bertini E, Iannaccone ST. Childhood spinal muscular atrophy: controversies and challenges. Lancet Neurol. 2012;11:443-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 249]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
4.  Castro D, Iannaccone ST. Spinal muscular atrophy: therapeutic strategies. Curr Treat Options Neurol. 2014;16:316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
5.  Prior TW, Leach ME, Finanger E. Spinal Muscular Atrophy. 2000 Feb 24 [updated 2020 Dec 3]. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, editors. GeneReviews® [Internet].  Seattle (WA): University of Washington, Seattle; 1993–2021.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Wang CH, Finkel RS, Bertini ES, Schroth M, Simonds A, Wong B, Aloysius A, Morrison L, Main M, Crawford TO, Trela A;  Participants of the International Conference on SMA Standard of Care. Consensus statement for standard of care in spinal muscular atrophy. J Child Neurol. 2007;22:1027-1049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 623]  [Cited by in F6Publishing: 535]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
7.  Butchbach ME. Copy Number Variations in the Survival Motor Neuron Genes: Implications for Spinal Muscular Atrophy and Other Neurodegenerative Diseases. Front Mol Biosci. 2016;3:7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 108]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
8.  Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, Yamashita M, Rigo F, Hung G, Schneider E, Norris DA, Xia S, Bennett CF, Bishop KM. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet. 2016;388:3017-3026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 668]  [Cited by in F6Publishing: 676]  [Article Influence: 84.5]  [Reference Citation Analysis (0)]
9.  Chen TH. New and Developing Therapies in Spinal Muscular Atrophy: From Genotype to Phenotype to Treatment and Where Do We Stand? Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
10.  Cambridge, MA: Biogen Inc.   Spinraza [package insert]. Reference ID: 4625921. 2020. [cited 30 June 2020]. In: Spinraza [Internet]. Available from: https://www.spinraza.com/PI.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Haché M, Swoboda KJ, Sethna N, Farrow-Gillespie A, Khandji A, Xia S, Bishop KM. Intrathecal Injections in Children With Spinal Muscular Atrophy: Nusinersen Clinical Trial Experience. J Child Neurol. 2016;31:899-906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 151]  [Article Influence: 18.9]  [Reference Citation Analysis (0)]
12.  Wood MJA, Talbot K, Bowerman M. Spinal muscular atrophy: antisense oligonucleotide therapy opens the door to an integrated therapeutic landscape. Hum Mol Genet. 2017;26:R151-R159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
13.  Glanzman AM, Mazzone E, Main M, Pelliccioni M, Wood J, Swoboda KJ, Scott C, Pane M, Messina S, Bertini E, Mercuri E, Finkel RS. The Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND): test development and reliability. Neuromuscul Disord. 2010;20:155-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 194]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
14.  De Vivo DC, Bertini E, Swoboda KJ, Hwu WL, Crawford TO, Finkel RS, Kirschner J, Kuntz NL, Parsons JA, Ryan MM, Butterfield RJ, Topaloglu H, Ben-Omran T, Sansone VA, Jong YJ, Shu F, Staropoli JF, Kerr D, Sandrock AW, Stebbins C, Petrillo M, Braley G, Johnson K, Foster R, Gheuens S, Bhan I, Reyna SP, Fradette S, Farwell W;  NURTURE Study Group. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study. Neuromuscul Disord. 2019;29:842-856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 363]  [Article Influence: 72.6]  [Reference Citation Analysis (0)]
15.  Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL, Kirschner J, Chiriboga CA, Saito K, Servais L, Tizzano E, Topaloglu H, Tulinius M, Montes J, Glanzman AM, Bishop K, Zhong ZJ, Gheuens S, Bennett CF, Schneider E, Farwell W, De Vivo DC;  ENDEAR Study Group. Nusinersen vs Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med. 2017;377:1723-1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1151]  [Cited by in F6Publishing: 1307]  [Article Influence: 186.7]  [Reference Citation Analysis (0)]
16.  Kuntz N, Farwell W, Zhong ZJ, Sun P, Gheuens S, Schneider E, Finkel R. Nusinersen in Infants Diagnosed with Spinal Muscular Atrophy (SMA): Study Design and Initial Interim Efficacy and Safety Findings from the Phase 3 International ENDEAR Study (CCI. 002). Neurology. 2017;88:CCI-002.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Mercuri E, Finkel R, Kirschner J, Chiriboga CA, Kuntz N, Darras B, Shieh PB, Saito K, De Vivo DC, Mazzone ES, Montes J. Interim analysis of the phase 3 CHERISH study evaluating nusinersen in patients with later-onset spinal muscular atrophy (SMA): Primary and descriptive secondary endpoints. Eur J Paediatr Neurol. 2017;21: e15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
18.  Darras BT, Chiriboga CA, Iannaccone ST, Swoboda KJ, Montes J, Mignon L, Xia S, Bennett CF, Bishop KM, Shefner JM, Green AM, Sun P, Bhan I, Gheuens S, Schneider E, Farwell W, De Vivo DC;  ISIS-396443-CS2/ISIS-396443-CS12 Study Groups. Nusinersen in later-onset spinal muscular atrophy: Long-term results from the phase 1/2 studies. Neurology. 2019;92:e2492-e2506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 178]  [Article Influence: 35.6]  [Reference Citation Analysis (0)]
19.  Aragon-Gawinska K, Seferian AM, Daron A, Gargaun E, Vuillerot C, Cances C, Ropars J, Chouchane M, Cuppen I, Hughes I, Illingworth M, Marini-Bettolo C, Rambaud J, Taytard J, Annoussamy M, Scoto M, Gidaro T, Servais L. Nusinersen in patients older than 7 mo with spinal muscular atrophy type 1: A cohort study. Neurology. 2018;91:e1312-e1318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
20.  Khouri JM, Payne JR, Arnon SS. More Clinical Mimics of Infant Botulism. J Pediatr. 2018;193:178-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
21.  European Medicines Agency  Spinraza. [cited 30 June 2020]. In: European Medicines Agency [Internet]. Available from: http://www.ema.europa.eu.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Nusinersen for spinal muscular atrophy. Aust Prescr. 2019;42:75-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
23.  Spinraza funding recommended for spinal muscular atrophy in NZ. PharmacoEcon Outcomes News. 2020;844:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
24.  Clinical Review Report: Nusinersen (Spinraza): (Biogen Canada Inc.): Indication: Treatment of patients with 5q SMA [Internet].  Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2018.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Montes J, Young SD, Mazzone E, Pasternak A, Glanzman A, Finkel R, Darras B, Muntoni F, Mercuri E, De Vivo D, Bishop K, Schneider E, Bennett F, Foster R, Farwell W. Ambulatory function and fatigue in nusinersen-treated children with spinal muscular atrophy. Muscle Nerve 2018; 90: P2. 322;.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Klug C, Schreiber-Katz O, Thiele S, Schorling E, Zowe J, Reilich P, Walter MC, Nagels KH. Disease burden of spinal muscular atrophy in Germany. Orphanet J Rare Dis. 2016;11:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 75]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
27.  Prasad V. Nusinersen for Spinal Muscular Atrophy: Are We Paying Too Much for Too Little? JAMA Pediatr. 2018;172:123-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
28.  Thomas K  Costly drug for fatal muscular disease wins FDA approval. December 30, 2016. [cited 30 June 2020]. In: New York Times [Internet]. Available from: https://www.nytimes.com/2016/12/30/business/spinraza-price.html.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Gornall J, Hoey A, Ozieranski P. A pill too hard to swallow: how the NHS is limiting access to high priced drugs. BMJ. 2016;354:i4117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
30.  Shell RD, Kotha K, Al-zaidy S. Gene therapy for spinal muscular atrophy type 1 improves survival and stabilizes pulmonary outcomes in a phase I/IIa safety study. Am J Respir Crit Care Med. 2016;193:A1040.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, Lowes L, Alfano L, Berry K, Church K, Kissel JT, Nagendran S, L'Italien J, Sproule DM, Wells C, Cardenas JA, Heitzer MD, Kaspar A, Corcoran S, Braun L, Likhite S, Miranda C, Meyer K, Foust KD, Burghes AHM, Kaspar BK. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med. 2017;377:1713-1722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1314]  [Cited by in F6Publishing: 1422]  [Article Influence: 203.1]  [Reference Citation Analysis (0)]
32.  Foust KD, Wang X, McGovern VL, Braun L, Bevan AK, Haidet AM, Le TT, Morales PR, Rich MM, Burghes AH, Kaspar BK. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol. 2010;28:271-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 574]  [Cited by in F6Publishing: 567]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
33.  Sumner CJ, Crawford TO. Two breakthrough gene-targeted treatments for spinal muscular atrophy: challenges remain. J Clin Invest. 2018;128:3219-3227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
34.  Lee BH, Collins E, Lewis L, Guntrum D, Eichinger K, Voter K, Abdel-Hamid HZ, Ciafaloni E. Combination therapy with nusinersen and AVXS-101 in SMA type 1. Neurology. 2019;93:640-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
35.  Ratni H, Ebeling M, Baird J, Bendels S, Bylund J, Chen KS, Denk N, Feng Z, Green L, Guerard M, Jablonski P, Jacobsen B, Khwaja O, Kletzl H, Ko CP, Kustermann S, Marquet A, Metzger F, Mueller B, Naryshkin NA, Paushkin SV, Pinard E, Poirier A, Reutlinger M, Weetall M, Zeller A, Zhao X, Mueller L. Discovery of Risdiplam, a Selective Survival of Motor Neuron-2 ( SMN2) Gene Splicing Modifier for the Treatment of Spinal Muscular Atrophy (SMA). J Med Chem. 2018;61:6501-6517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 278]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
36.  Sturm S, Günther A, Jaber B, Jordan P, Al Kotbi N, Parkar N, Cleary Y, Frances N, Bergauer T, Heinig K, Kletzl H, Marquet A, Ratni H, Poirier A, Müller L, Czech C, Khwaja O. A phase 1 healthy male volunteer single escalating dose study of the pharmacokinetics and pharmacodynamics of risdiplam (RG7916, RO7034067), a SMN2 splicing modifier. Br J Clin Pharmacol. 2019;85:181-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 68]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
37.  Baranello1 G, Servais L, Day JW, Deconinck N, Mercuri E, Klein A, Darras B, Masson R, Kletzl H, Cleary Y, El-Khairi M, Seabrook T, Czech C, Gerber M, Somugompely P, Gelblin K, Gorni K, Khwaja O.   FIREFISH part 1: Early clinical results following an increase of SMN protein in infants with type 1 spinal muscular atrophy (SMA) treated with Risdiplam (RG7916). Proceedings of the Communication Presented at MDA Clinical & Scientific Conference; 2019 April 13-17; Orlando, FL, United States.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Roche  Roche’s Risdiplam Meets Primary Endpoint in Pivotal SUNFISH Trial in People with Type 2 or 3 Spinal Muscular Atrophy. [cited 30 June 2020]. In: Roche [Internet]. Available from: https://www.roche.com/media/releases/med-cor-2019-11-11.htm.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Cheung AK, Hurley B, Kerrigan R, Shu L, Chin DN, Shen Y, O'Brien G, Sung MJ, Hou Y, Axford J, Cody E, Sun R, Fazal A, Fridrich C, Sanchez CC, Tomlinson RC, Jain M, Deng L, Hoffmaster K, Song C, Van Hoosear M, Shin Y, Servais R, Towler C, Hild M, Curtis D, Dietrich WF, Hamann LG, Briner K, Chen KS, Kobayashi D, Sivasankaran R, Dales NA. Discovery of Small Molecule Splicing Modulators of Survival Motor Neuron-2 (SMN2) for the Treatment of Spinal Muscular Atrophy (SMA). J Med Chem. 2018;61:11021-11036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
40.  Jevtic S, Carr D, Dobrzycka-Ambrozevicz A.   Branaplam in Type 1 spinal muscular atrophy: Second part of aphase I/II Study. Proceedings of the CureSMA 23rd Annual SMA Researcher Meeting; 2019 June 28-30; Anaheim, CA, United States.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Farooq F, Abadía-Molina F, MacKenzie D, Hadwen J, Shamim F, O'Reilly S, Holcik M, MacKenzie A. Celecoxib increases SMN and survival in a severe spinal muscular atrophy mouse model via p38 pathway activation. Hum Mol Genet. 2013;22:3415-3424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
42.  Wadman RI, van der Pol WL, Bosboom WM, Asselman FL, van den Berg LH, Iannaccone ST, Vrancken AF. Drug treatment for spinal muscular atrophy types II and III. Cochrane Database Syst Rev. 2020;1:CD006282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
43.  Jarecki J, Chen X, Bernardino A, Coovert DD, Whitney M, Burghes A, Stack J, Pollok BA. Diverse small-molecule modulators of SMN expression found by high-throughput compound screening: early leads towards a therapeutic for spinal muscular atrophy. Hum Mol Genet. 2005;14:2003-2018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 126]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
44.  Gogliotti RG, Cardona H, Singh J, Bail S, Emery C, Kuntz N, Jorgensen M, Durens M, Xia B, Barlow C, Heier CR, Plasterer HL, Jacques V, Kiledjian M, Jarecki J, Rusche J, DiDonato CJ. The DcpS inhibitor RG3039 improves survival, function and motor unit pathologies in two SMA mouse models. Hum Mol Genet. 2013;22:4084-4101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
45.  Jędrzejowska M, Kostera-Pruszczyk A. Spinal muscular atrophy - new therapies, new challenges. Neurol Neurochir Pol. 2020;54:8-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
46.  Van Meerbeke J, Gibbs R, Plasterer H, Feng Z, Lin MY, Wee C, Xia B, Jacques V, Rusche J, Ko CP. The Therapeutic Effects of RG3039 in Severe Spinal Muscular Atrophy Mice and NormalHuman Volunteers (S25.003). Neurology. 2012;78:S25.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Heier CR, DiDonato CJ. Translational readthrough by the aminoglycoside geneticin (G418) modulates SMN stability in vitro and improves motor function in SMA mice in vivo. Hum Mol Genet. 2009;18:1310-1322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
48.  Cobb MS, Rose FF, Rindt H, Glascock JJ, Shababi M, Miller MR, Osman EY, Yen PF, Garcia ML, Martin BR, Wetz MJ, Mazzasette C, Feng Z, Ko CP, Lorson CL. Development and characterization of an SMN2-based intermediate mouse model of Spinal Muscular Atrophy. Hum Mol Genet. 2013;22:1843-1855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
49.  Calder AN, Androphy EJ, Hodgetts KJ. Small Molecules in Development for the Treatment of Spinal Muscular Atrophy. J Med Chem. 2016;59:10067-10083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
50.  Greif H, Rosin-Arbesfeld R, Megiddo D.   BBrm2, A Read-through Repurposed Drug, Shows Proof of E_cacy in SMA Treatment. Proceedings of the Cure SMA 19th Annual SMA Researcher Meeting; 2015 June 18-20; Kansas City, MO, United States.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Kwon DY, Motley WW, Fischbeck KH, Burnett BG. Increasing expression and decreasing degradation of SMN ameliorate the spinal muscular atrophy phenotype in mice. Hum Mol Genet. 2011;20:3667-3677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
52.  Wirth B, Karakaya M, Kye MJ, Mendoza-Ferreira N. Twenty-Five Years of Spinal Muscular Atrophy Research: From Phenotype to Genotype to Therapy, and What Comes Next. Annu Rev Genomics Hum Genet. 2020;21:231-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 117]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
53.  Bordet T, Buisson B, Michaud M, Drouot C, Galéa P, Delaage P, Akentieva NP, Evers AS, Covey DF, Ostuni MA, Lacapère JJ, Massaad C, Schumacher M, Steidl EM, Maux D, Delaage M, Henderson CE, Pruss RM. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J Pharmacol Exp Ther. 2007;322:709-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 193]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
54.  Bertini E, Dessaud E, Mercuri E, Muntoni F, Kirschner J, Reid C, Lusakowska A, Comi GP, Cuisset JM, Abitbol JL, Scherrer B, Ducray PS, Buchbjerg J, Vianna E, van der Pol WL, Vuillerot C, Blaettler T, Fontoura P;  Olesoxime SMA Phase 2 Study Investigators. Safety and efficacy of olesoxime in patients with type 2 or non-ambulatory type 3 spinal muscular atrophy: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Neurol. 2017;16:513-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 83]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
55.  Darras BT. Spinal muscular atrophies. Pediatr Clin North Am. 2015;62:743-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 96]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
56.  Haddad H, Cifuentes-Diaz C, Miroglio A, Roblot N, Joshi V, Melki J. Riluzole attenuates spinal muscular atrophy disease progression in a mouse model. Muscle Nerve. 2003;28:432-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 64]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
57.  Merlini L, Solari A, Vita G, Bertini E, Minetti C, Mongini T, Mazzoni E, Angelini C, Morandi L. Role of gabapentin in spinal muscular atrophy: results of a multicenter, randomized Italian study. J Child Neurol. 2003;18:537-541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Pirruccello-Straub M, Jackson J, Wawersik S, Webster MT, Salta L, Long K, McConaughy W, Capili A, Boston C, Carven GJ, Mahanthappa NK, Turner KJ, Donovan A. Blocking extracellular activation of myostatin as a strategy for treating muscle wasting. Sci Rep. 2018;8:2292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 68]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
59.  Feng Z, Ling KK, Zhao X, Zhou C, Karp G, Welch EM, Naryshkin N, Ratni H, Chen KS, Metzger F, Paushkin S, Weetall M, Ko CP. Pharmacologically induced mouse model of adult spinal muscular atrophy to evaluate effectiveness of therapeutics after disease onset. Hum Mol Genet. 2016;25:964-975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 44]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
60.  Liu M, Hammers DW, Barton ER, Sweeney HL. Activin Receptor Type IIB Inhibition Improves Muscle Phenotype and Function in a Mouse Model of Spinal Muscular Atrophy. PLoS One. 2016;11:e0166803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
61.  AliveGen  R&D Pipeline: ALG-801. [cited 20 June 2020]. In: AliveGen [Internet]. Available from: http://www.alivegen.com/r-d-pipeline.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Long KK, O'Shea KM, Khairallah RJ, Howell K, Paushkin S, Chen KS, Cote SM, Webster MT, Stains JP, Treece E, Buckler A, Donovan A. Specific inhibition of myostatin activation is beneficial in mouse models of SMA therapy. Hum Mol Genet. 2019;28:1076-1089.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 71]  [Article Influence: 14.2]  [Reference Citation Analysis (2)]
63.  Chyung Y  Interim Results from a Phase 1 Study of SRK-015, a Fully Human Monoclonal Antibody that Inhibits Myostatin Activation. Proceedings of the CureSMA 23rd Annual SMA Researcher Meeting; 2019 June 28-30; Anaheim, CA, United States.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Hwee DT, Kennedy AR, Hartman JJ, Ryans J, Durham N, Malik FI, Jasper JR. The small-molecule fast skeletal troponin activator, CK-2127107, improves exercise tolerance in a rat model of heart failure. J Pharmacol Exp Ther. 2015;353:159-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
65.  Andrews JA, Miller TM, Vijayakumar V, Stoltz R, James JK, Meng L, Wolff AA, Malik FI. CK-2127107 amplifies skeletal muscle response to nerve activation in humans. Muscle Nerve. 2018;57:729-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
66.  Rudnicki SA, Andrews JA, Duong T, Cockroft BM, Malik FI, Meng L, Wei J, Wolff AA, Genge A, Johnson NE, Tesi-Rocha C, Connolly AM, Darras BT, Felice K, Shieh PB, Mah JK, Statland J, Campbell C, Habib AA, Kuntz NL, Oskoui M, Day JW. Reldesemtiv in Patients with Spinal Muscular Atrophy: a Phase 2 Hypothesis-Generating Study. Neurotherapeutics. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
67.  Wadman RI, Vrancken AF, van den Berg LH, van der Pol WL. Dysfunction of the neuromuscular junction in spinal muscular atrophy types 2 and 3. Neurology. 2012;79:2050-2055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 74]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
68.  Stam M, Wadman RI, Wijngaarde CA, Bartels B, Asselman FL, Otto LAM, Goedee HS, Habets LE, de Groot JF, Schoenmakers MAGC, Cuppen I, van den Berg LH, van der Pol WL. Protocol for a phase II, monocentre, double-blind, placebo-controlled, cross-over trial to assess efficacy of pyridostigmine in patients with spinal muscular atrophy types 2-4 (SPACE trial). BMJ Open. 2018;8:e019932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
69.  Imlach WL, Beck ES, Choi BJ, Lotti F, Pellizzoni L, McCabe BD. SMN is required for sensory-motor circuit function in Drosophila. Cell. 2012;151:427-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 152]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
70.  Pandolfi F, De Vita D, Bortolami M, Coluccia A, Di Santo R, Costi R, Andrisano V, Alabiso F, Bergamini C, Fato R, Bartolini M, Scipione L. New pyridine derivatives as inhibitors of acetylcholinesterase and amyloid aggregation. Eur J Med Chem. 2017;141:197-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
71.  Ebert AD, Yu J, Rose FF Jr, Mattis VB, Lorson CL, Thomson JA, Svendsen CN. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009;457:277-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1098]  [Cited by in F6Publishing: 1026]  [Article Influence: 64.1]  [Reference Citation Analysis (0)]
72.  Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D, Saladino F, Bordoni A, Fortunato F, Del Bo R, Papadimitriou D, Locatelli F, Menozzi G, Strazzer S, Bresolin N, Comi GP. Neural stem cell transplantation can ameliorate the phenotype of a mouse model of spinal muscular atrophy. J Clin Invest. 2008;118:3316-3330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 111]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
73.  Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D, Simone C, Falcone M, Papadimitriou D, Locatelli F, Mezzina N, Gianni F, Bresolin N, Comi GP. Embryonic stem cell-derived neural stem cells improve spinal muscular atrophy phenotype in mice. Brain. 2010;133:465-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 82]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]