Safety, pharmacokinetic, and functional effects of the nogo-a monoclonal antibody in amyotrophic lateral sclerosis: a randomized, first-in-human clinical trial

Vincent Meininger, Pierre-François Pradat, Andrea Corse, Safa Al-Sarraj, Benjamin Rix Brooks, James B Caress, Merit Cudkowicz, Stephen J Kolb, Dale Lange, P Nigel Leigh, Thomas Meyer, Stefano Milleri, Karen E Morrison, Richard W Orrell, Gary Peters, Jeffrey D Rothstein, Jeremy Shefner, Arseniy Lavrov, Nicola Williams, Phil Overend, Jeffrey Price, Stewart Bates, Jonathan Bullman, David Krull, Alienor Berges, Bams Abila, Guy Meno-Tetang, Jens Wurthner, Vincent Meininger, Pierre-François Pradat, Andrea Corse, Safa Al-Sarraj, Benjamin Rix Brooks, James B Caress, Merit Cudkowicz, Stephen J Kolb, Dale Lange, P Nigel Leigh, Thomas Meyer, Stefano Milleri, Karen E Morrison, Richard W Orrell, Gary Peters, Jeffrey D Rothstein, Jeremy Shefner, Arseniy Lavrov, Nicola Williams, Phil Overend, Jeffrey Price, Stewart Bates, Jonathan Bullman, David Krull, Alienor Berges, Bams Abila, Guy Meno-Tetang, Jens Wurthner

Abstract

The neurite outgrowth inhibitor, Nogo-A, has been shown to be overexpressed in skeletal muscle in amyotrophic lateral sclerosis (ALS); it is both a potential biomarker and therapeutic target. We performed a double-blind, two-part, dose-escalation study, in subjects with ALS, assessing safety, pharmacokinetics (PK) and functional effects of ozanezumab, a humanized monoclonal antibody against Nogo-A. In Part 1, 40 subjects were randomized (3∶1) to receive single dose intravenous ozanezumab (0.01, 0.1, 1, 5, or 15 mg/kg) or placebo. In Part 2, 36 subjects were randomized (3∶1) to receive two repeat doses of intravenous ozanezumab (0.5, 2.5, or 15 mg/kg) or placebo, approximately 4 weeks apart. The primary endpoints were safety and tolerability (adverse events [AEs], vital signs, electrocardiogram (ECG), and clinical laboratory tests). Secondary endpoints included PK, immunogenicity, functional endpoints (clinical and electrophysiological), and biomarker parameters. Overall, ozanezumab treatment (0.01-15 mg/kg) was well tolerated. The overall incidence of AEs in the repeat dose 2.5 mg/kg and 15 mg/kg ozanezumab groups was higher than in the repeat dose placebo group and repeat dose 0.5 mg/kg ozanezumab group. The majority were considered not related to study drug by the investigators. Six serious AEs were reported in three subjects receiving ozanezumab; none were considered related to study drug. No study drug-related patterns were identified for ECG, laboratory, or vital signs parameters. One subject (repeat dose 15 mg/kg ozanezumab) showed a weak, positive anti-ozanezumab-antibody result. PK results were generally consistent with monoclonal antibody treatments. No apparent treatment effects were observed for functional endpoints or muscle biomarkers. Immunohistochemical staining showed dose-dependent co-localization of ozanezumab with Nogo-A in skeletal muscle. In conclusion, single and repeat dose ozanezumab treatment was well tolerated and demonstrated co-localization at the site of action. These findings support future studies with ozanezumab in ALS.

Trial registration: ClinicalTrials.gov NCT00875446 GSK-ClinicalStudyRegister.com GSK ID 111330.

Conflict of interest statement

Competing Interests: AL, NW, PO, JP, SB, JB, DK, AB, BA and GM-T are employees of and hold stock in GlaxoSmithKline. JW was a full-time employee of GlaxoSmithKline at the time of study and holds shares in the company. He is now an employee of Novartis. GP was a full-time employee of GlaxoSmithKline at the time of study and holds shares in the company. He is now an employee of Hammersmith Medicines Research. VM has previously received a consultancy fee from GlaxoSmithKline. AC has previously acted as a consultant to GlaxoSmithKline at a single-day advisory board meeting. PNL received travel expenses and consultancy fees for his work on the advisory board of this development. JDR has previously received funding from NIH, MDA, Robert Packard Center for ALS research and has acted as a consultant for Psyadon Pharmaceutical, Biogen Pharma and Cytokinetics. MC is on the GlaxoSmithKline scientific advisory board. P-FP, BRB, JBC, SJK, DL, TM, SM, SA-S and KEM have no conflicts of interest to declare. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Subject disposition flow diagram for…
Figure 1. Subject disposition flow diagram for Parts 1 and 2.
N, total number of subjects in group; n, number of subjects in category; PK, pharmacokinetics; SAE, serious adverse events. *Two doses, received 4 weeks apart.
Figure 2. Mean plasma ozanezumab concentration-time profiles…
Figure 2. Mean plasma ozanezumab concentration-time profiles following single (A) or two (B) IV infusions of ozanezumab.
Figure 3. Co-localization of membrane Nogo-A with…
Figure 3. Co-localization of membrane Nogo-A with ozanezumab in skeletal muscle of individual subjects.
A. Triplicate readings are provided from biopsies in Cohort 7 and 8 (single reading from Cohort 5) dose 2 +D22–26, biopsy taken 22–26 days after the second dose; dose 1 +24H, biopsy taken 24 hours after first dose. B. Nogo-A (red), ozanezumab (green) and co-localization (yellow), in muscle biopsy, 24 hours post-dose.

References

    1. Cleveland DW (1999) From Charcot to SOD1: mechanisms of selective motor neuron death in ALS. Neuron 24: 515–520.
    1. Miller RG, Jackson CE, Kasarskis EJ, England JD, Kalra S, et al. (2009) Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: drug, nutritional, and respiratory therapies (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 73: 1218–1226.
    1. Miller RG, Mitchell JD, Moore DH (2012) Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst Rev (Online) 3: CD001447.
    1. Talbot K (2009) Motor neuron disease: the bare essentials. Pract Neurol 9: 303–309.
    1. Ringholz GM, Appel SH, Bradshaw M, Cooke NA, Mosnik DM, et al. (2005) Prevalence and patterns of cognitive impairment in sporadic ALS. Neurology 65: 586–590.
    1. Turner MR, Hardiman O, Benatar M, Brooks BR, Chio A, et al. (2013) Controversies and priorities in amyotrophic lateral sclerosis. Lancet Neurol 12: 310–322.
    1. Rothstein JD (1996) Therapeutic horizons for amyotrophic lateral sclerosis. Curr Opin Neurobiol 6: 679–687.
    1. Pandya RS, Zhu H, Li W, Bowser R, Friedlander RM, Wang X (2013) Therapeutic neuroprotective agents for amyotrophic lateral sclerosis. Cell Mol Life Sci 70: 4729–4745.
    1. Bellingham MC (2011) A review of the neural mechanisms of action and clinical efficiency of riluzole in treating amyotrophic lateral sclerosis: what have we learned in the last decade? CNS Neurosci Ther 17: 4–31.
    1. Schwab ME (2004) Nogo and axon regeneration. Curr Opin Neurobiol 14: 118–124.
    1. Schwab ME (2010) Functions of Nogo proteins and their receptors in the nervous system. Nat Rev Neurosci 11: 799–811.
    1. Dupuis L, Gonzalez de Aguilar J-L, di Scala F, Rene F, da Tapia M, et al. (2002) Nogo provides a molecular marker for diagnosis of amyotrophic lateral sclerosis. Neurobiol Dis 10: 358–365.
    1. Jokic N, Gonzalez de Aguilar J-L, Pradat P-F, Dupuis L, Echaniz-Laguna A, et al. (2005) Nogo expression in muscle correlates with amyotrophic lateral sclerosis severity. Ann Neurol 57: 553–536.
    1. Pradat P-F, Bruneteau G, Gonzalez de Aguilar J-L, Dupuis L, Jokic N, et al. (2007) Muscle Nogo-a expression is a prognostic marker in lower motor neuron syndromes. Ann Neurol 62: 15–20.
    1. Tågerud S, Libelius R, Magnusson C (2007) Muscle Nogo-A: a marker for amyotrophic lateral sclerosis or for denervation? Ann Neurol 62: 676.
    1. Magnusson C, Libelius R, Tågerud S (2003) Nogo (Reticulon 4) expression in innervated and denervated mouse skeletal muscle. Mol Cell Neurosci 22: 298–307.
    1. Askanas V, Wojcik S, Engel WK (2007) Expression of Nogo-A in human muscle fibers is not specific for amyotrophic lateral sclerosis. Ann Neurol 62: 676–677.
    1. Wojcik S, Engel WK, Askanas V (2006) Increased expression of Noga-A in ALS muscle biopsies is not unique for this disease. Acta Myol 25: 116–118.
    1. Jokic N, Gonzalez de Aguilar JL, Dimou L, Lin S, Fergani A, et al. (2006) The neurite outgrowth inhibitor Nogo-A promotes denervation in an amyotrophic lateral sclerosis model. EMBO Rep 7: 1162–1167.
    1. Fischer LR, Culver DG, Tennant P, Davis AA, Wang M, et al. (2004) Amyotrophic lateral sclerosis is a distal axonopathy: evidence in mice and man. Exp Neurol 185: 232–240.
    1. Weinmann O, Schnell L, Ghosh A, Montani L, Wiessener C, et al. (2006) Intrathecally infused antibodies against Nogo-A penetrate the CNS and downregulate the endogenous neurite growth inhibitor Nogo-A. Mol Cell Neurosci 32: 161–173.
    1. Brooks BR, Miller RG, Swash M, Munsat TL (2000) El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord 1: 293–299.
    1. Cedarbaum JM, Stambler N, Malta E, Fuller C, Hilt D, et al. (1999) The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. J Neurological Sci 169: 13–21.
    1. Sanjak M, Salachas F, Frija-Orvoen E, Theys P, Hutchinson D, et al. (2010) Quality control of vital capacity as a primary outcome measure during phase III therapeutic clinical trial in amyotrophic lateral sclerosis. Amyotroph Lateral Scler 11: 383–388.
    1. Great Lakes ALS Study Group (2003) A comparison of muscle strength testing techniques in amyotrophic lateral sclerosis. Neurology 61: 1503–1507.
    1. Shefner JM, Watson ML, Simionescu L, Caress JB, Burns TM, et al. (2011) Multipoint incremental motor unit number estimation as an outcome measure in ALS. Neurology 77: 235–241.
    1. Cudkowicz M, Bozik ME, Ingersoll EW, Miller R, Mitsumoto H, et al. (2011) The effects of dexpramipexole (KNS-760704) in individuals with amyotrophic lateral sclerosis. Nat Med 17: 1652–166.
    1. Cudkowicz ME, van den Berg LH, Shefner JM, Mitsumoto H, Mora JS, et al. (2013) Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial. Lancet Neurol 12: 1059–1067.

Source: PubMed

3
Abonnieren