Safety, tolerability, pharmacokinetics, and efficacy of AMG 403, a human anti-nerve growth factor monoclonal antibody, in two phase I studies with healthy volunteers and knee osteoarthritis subjects

Jason M Gow, Wayne H Tsuji, Gary J Williams, Daniel Mytych, David Sciberras, Shawn L Searle, Tim Mant, John P Gibbs, Jason M Gow, Wayne H Tsuji, Gary J Williams, Daniel Mytych, David Sciberras, Shawn L Searle, Tim Mant, John P Gibbs

Abstract

Introduction: Nerve growth factor plays a key role in the pathology of osteoarthritis (OA) related chronic pain. The aim of these studies was to evaluate the safety, tolerability, pharmacokinetics, and clinical response of AMG 403, a human anti-nerve growth factor monoclonal antibody, in healthy volunteers and subjects with knee OA.

Methods: Two phase I, randomized, placebo-controlled, double-blind studies were conducted. The single-ascending dose study randomized healthy volunteers (n = 48) 3:1 to receive AMG 403 (1, 3, 10, or 30 mg intravenously; or 10 or 30 mg subcutaneously; n = 8 per group) or placebo. The multiple-ascending dose study randomized knee OA subjects (n = 18) 3:1 to receive AMG 403 (3, 10, or 20 mg subcutaneously once monthly for four doses) or placebo. Safety, tolerability, and pharmacokinetics (PK) were assessed for both studies. Patient's and physician's disease assessments and total WOMAC score were determined in knee OA subjects.

Results: AMG 403 appeared to be well-tolerated after single and multiple doses, except for subject-reported hyperesthesia, pain, and paresthesia (mild to moderate severity). These treatment-emergent neurosensory events showed evidence of reversibility and a possible dose-dependence. Three serious adverse events were reported in AMG 403 treated subjects, but were not considered treatment related. AMG 403 PK was linear with an estimated half-life of 19.6 to 25.8 days. After multiple doses, AMG 403 PK showed modest accumulation (≤2.4-fold increase) in systemic exposure. Knee OA diagnosis, body weight, and anti-drug antibody development did not appear to affect AMG 403 PK. Patient's and physician's disease assessments and total WOMAC score showed improvement in AMG 403 treated knee OA subjects compared with placebo.

Conclusions: AMG 403 was generally safe and well-tolerated in both healthy volunteers and knee OA patients, and exhibited linear pharmacokinetics. Preliminary clinical efficacy was observed in knee OA subjects.

Trial registration: ClinicalTrials.gov NCT02348879 . Registered 23 December 2014. Clintrials.gov NCT02318407 . Registered 2 December 2014.

Figures

Fig. 1
Fig. 1
AMG 403 concentration-time profiles following single and repeat dose administration. AMG 403 was administered intravenously (IV) or subcutaneously (SC) and serum AMG 403 levels were measured pre-dose through end of study. Each point represents the mean (+SD) AMG 403 concentration. a Healthy volunteers received a single IV or SC dose of AMG 403 (1–30 mg, LLOQ = 0.0016 μg/mL). b Patients with knee osteoarthritis received a once-monthly SC dose of AMG 403 (3–20 mg, LLOQ = 0.0051 μg/mL) for up to four dose administrations. Arrows represent dose administration
Fig. 2
Fig. 2
Visual predictive check of the AMG 403 population pharmokinetics (PK) model. A two-compartment model was fit simultaneously to individual AMG 403 PK data from healthy volunteers (HV) and patients with knee osteoarthritis (OA). The model parameter estimates were used to simulate 1000 trials for a visual predictive check. The predicted median PK (solid line) and 80 % prediction interval (shaded area) are shown with the observed individual concentrations (solid dot). Note the different x-axis scales for the single-dose and multiple-dose regimens. IV intravenous, SC subcutaneous
Fig. 3
Fig. 3
Covariate effects on AMG 403 clearance (CL) and (Vc). Possible covariate effects were explored for individual CL and Vc parameters derived from simultaneous population pharmacokinetics modeling of data from healthy volunteers (HV) and patients with knee osteoarthritis (OA). Effect of body weight on CL (a) and Vc (b) is illustrated with a loess regression line. c The effect of anti-drug antibody (ADA) status (negative (n = 34) or positive (n = 14)) or disease status (HV (n = 30) or patients with knee OA (n = 18)) on CL is presented in box plots (median, 25th, and 75th percentiles; whiskers are 1.5 times the inner-quartile range and dots are outliers)
Fig. 4
Fig. 4
Clinical effect of AMG 403 in patients with knee osteoarthritis. Mean change (+ standard error) from baseline values over time for each dose group were plotted for total Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) (a) score and visual analog score (VAS) (b) for patient disease assessment. Arrows represent subcutaneous dose administrations (SC)

References

    1. Johnson VL, Hunter DJ. The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol. 2014;28:5–15. doi: 10.1016/j.berh.2014.01.004.
    1. Corti MC, Rigon C. Epidemiology of osteoarthritis: prevalence, risk factors and functional impact. Aging Clin Exp Res. 2003;15:359–363. doi: 10.1007/BF03327356.
    1. Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States, Part II. Arthritis Rheum. 2008;58:26–35. doi: 10.1002/art.23176.
    1. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum. 2012;64:1697–1707. doi: 10.1002/art.34453.
    1. Dieppe PA, Lohmander LS. Pathogenesis and management of pain in osteoarthritis. Lancet. 2005;365:965–973. doi: 10.1016/S0140-6736(05)71086-2.
    1. Bellamy N. The WOMAC Knee and Hip Osteoarthritis Indices: development, validation, globalization and influence on the development of the AUSCAN Hand Osteoarthritis Indices. Clin Exp Rheumatol. 2005;23:S148–S153.
    1. McConnell S, Kolopack P, Davis AM. The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC): a review of its utility and measurement properties. Arthritis Rheum. 2001;45:453–461. doi: 10.1002/1529-0131(200110)45:5<453::AID-ART365>;2-W.
    1. Torres L, Dunlop DD, Peterfy C, Guermazi A, Prasad P, Hayes KW, et al. The relationship between specific tissue lesions and pain severity in persons with knee osteoarthritis. Osteoarthritis Cartilage. 2006;14:1033–1040. doi: 10.1016/j.joca.2006.03.015.
    1. Szebenyi B, Hollander AP, Dieppe P, Quilty B, Duddy J, Clarke S, et al. Associations between pain, function, and radiographic features in osteoarthritis of the knee. Arthritis Rheum. 2006;54:230–235. doi: 10.1002/art.21534.
    1. Turk DC, Wilson HD, Cahana A. Treatment of chronic non-cancer pain. Lancet. 2011;377:2226–2235. doi: 10.1016/S0140-6736(11)60402-9.
    1. Seidel MF, Herguijuela M, Forkert R, Otten U. Nerve growth factor in rheumatic diseases. Semin Arthritis Rheum. 2010;40:109–126. doi: 10.1016/j.semarthrit.2009.03.002.
    1. Seidel MF, Wise BL, Lane NE. Nerve growth factor: an update on the science and therapy. Osteoarthritis Cartilage. 2013;21:1223–1228. doi: 10.1016/j.joca.2013.06.004.
    1. Eibl JK, Strasser BC, Ross GM. Structural, biological, and pharmacological strategies for the inhibition of nerve growth factor. Neurochem Int. 2012;61:1266–1275. doi: 10.1016/j.neuint.2012.10.008.
    1. Hefti FF, Rosenthal A, Walicke PA, Wyatt S, Vergara G, Shelton DL, et al. Novel class of pain drugs based on antagonism of NGF. Trends Pharmacol Sci. 2006;27:85–91. doi: 10.1016/j.tips.2005.12.001.
    1. Watson JJ, Allen SJ, Dawbarn D. Targeting nerve growth factor in pain: what is the therapeutic potential? BioDrugs. 2008;22:349–359. doi: 10.2165/0063030-200822060-00002.
    1. Wild KD, Bian D, Zhu D, Davis J, Bannon AW, Zhang TJ, et al. Antibodies to nerve growth factor reverse established tactile allodynia in rodent models of neuropathic pain without tolerance. J Pharmacol Exp Ther. 2007;322:282–287. doi: 10.1124/jpet.106.116236.
    1. Katz N, Borenstein DG, Birbara C, Bramson C, Nemeth MA, Smith MD, et al. Efficacy and safety of tanezumab in the treatment of chronic low back pain. Pain. 2011;152:2248–2258. doi: 10.1016/j.pain.2011.05.003.
    1. Lane NE, Schnitzer TJ, Birbara CA, Mokhtarani M, Shelton DL, Smith MD, et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. N Engl J Med. 2010;363:1521–1531. doi: 10.1056/NEJMoa0901510.
    1. Schnitzer TJ, Lane NE, Birbara C, Smith MD, Simpson SL, Brown MT. Long-term open-label study of tanezumab for moderate to severe osteoarthritic knee pain. Osteoarthritis Cartilage. 2011;19:639–646. doi: 10.1016/j.joca.2011.01.009.
    1. Hawker GA, Mian S, Kendzerska T, French M. Measures of adult pain: Visual analog scale for pain (VAS Pain), Numeric Rating Scale for Pain (NRS Pain), McGill Pain Questionnaire (MPQ), Short-Form McGill Pain Questionnaire (SF-MPQ), Chronic Pain Grade Scale (CPGS), Short Form-36 Bodily Pain Scale (SF-36 BPS), and Measure of Intermittent and Constant Osteoarthritis Pain (ICOAP) Arthritis Care Res (Hoboken) 2011;63:S240–S252. doi: 10.1002/acr.20543.
    1. Sanga P, Katz N, Polverejan E, Wang S, Kelly KM, Haeussler J, et al. Efficacy, safety, and tolerability of fulranumab, an anti-nerve growth factor antibody, in the treatment of patients with moderate to severe osteoarthritis pain. Pain. 2013;154:1910–1919. doi: 10.1016/j.pain.2013.05.051.
    1. Schnitzer TJ, Marks JA. A systematic review of the efficacy and general safety of antibodies to NGF in the treatment of OA of the hip or knee. Osteoarthr Cartil. 2015;23:S8–S17. doi: 10.1016/j.joca.2014.10.003.
    1. Dai S, Schantz A, Clements-Egan A, Cannon M, Shankar G. Development of a method that eliminates false-positive results due to nerve growth factor interference in the assessment of fulranumab immunogenicity. AAPS J. 2014;16:464–477. doi: 10.1208/s12248-014-9581-z.
    1. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet. 2010;49:633–659. doi: 10.2165/11535960-000000000-00000.

Source: PubMed

3
Subskrybuj