Early intervention with tafamidis provides long-term (5.5-year) delay of neurologic progression in transthyretin hereditary amyloid polyneuropathy

Márcia Waddington Cruz, Leslie Amass, Denis Keohane, Jeffrey Schwartz, Huihua Li, Balarama Gundapaneni, Márcia Waddington Cruz, Leslie Amass, Denis Keohane, Jeffrey Schwartz, Huihua Li, Balarama Gundapaneni

Abstract

Transthyretin hereditary amyloid polyneuropathy, also traditionally known as transthyretin familial amyloid polyneuropathy (ATTR-FAP), is a rare, relentless, fatal hereditary disorder. Tafamidis, an oral, non-NSAID, highly specific transthyretin stabilizer, demonstrated safety and efficacy in slowing neuropathy progression in early-stage ATTRV30M-FAP in a 1.5-year, randomized, double-blind, placebo-controlled trial, and 1-year open-label extension study, with a second long-term open-label extension study ongoing. Subgroup analysis of the effectiveness of tafamidis in the pivotal study and its open-label extensions revealed a relatively cohesive cohort of patients with mild neuropathy (i.e. Neuropathy Impairment Score for Lower Limbs [NIS-LL] ≤ 10) at the start of active treatment. Early treatment with tafamidis for up to 5.5 years (≥1 dose of tafamidis meglumine 20 mg once daily during the original trial or after switching from placebo in its extension) resulted in sustained delay in neurologic progression and long-term preservation of nutritional status in this cohort. Mean (95% CI) changes from baseline in NIS-LL and mBMI were 5.3 (1.6, 9.1) points and -7.8 (-44.3, 28.8) kg/m2 × g/L at 5.5 years, respectively. No new safety issues or side effects were identified. These data represent the longest prospective evaluation of tafamidis to date, confirm a favorable safety profile, and underscore the long-term benefits of early intervention with tafamidis.

Trial registration: ClincalTrials.gov Identifier: NCT00409175, NCT00791492, and NCT00925002.

Keywords: ATTRV30M; Amyloidosis; TTR Val30Met; disease modification; transthyretin amyloidosis.

Figures

Figure 1.
Figure 1.
Analysis population and patient disposition. *All randomized patients who received at least one dose of study medication and had at least one follow-up assessment. †One patient who completed the tafamidis arm of the double-blind trial did not enroll in the open-label extension study, as he/she wanted to become pregnant. NIS-LL: Neuropathy Impairment Score for the Lower Limbs.
Figure 2.
Figure 2.
Absolute NIS-LL total scores throughout long-term tafamidis treatment for each of the 71 patients with baseline NIS-LL ≤ 10 at the last assessment before the first dose of tafamidis. NIS-LL: Neuropathy Impairment Score for the Lower Limbs.
Figure 3.
Figure 3.
Mean (95% CI) change from baseline in NIS-LL throughout 5.5 years of tafamidis treatment among 71 patients with baseline NIS-LL ≤ 10 at last assessment before first dose of tafamidis. Analyses were performed using observed cases. Year 5 data were excluded due to limited data (n = 2). Mean change in NIS-LL total score (Panel A). Mean change in three NIS-LL subscale scores, muscle weakness, sensation, and reflexes (Panel B). The subscales range from 0 to 64 (muscle weakness), 0 to 16 (sensation), or 0 to 8 (reflexes), with higher values indicating greater neurologic deficit. Mean (95% CI) baseline NIS-LL sub-scores in the analysis population were 0.2 (0.0–0.4) for muscle weakness, 3.4 (2.8–4.0) for sensation, and 0.5 (0.2–0.8) for reflexes. Subgroup analysis of the mean change in NIS-LL total score in patients with baseline NIS-LL score of 0–5 (n = 50) versus those with a baseline NIS-LL score of >5–10 (n = 21) (Panel C). Mean (95% CI) baseline NIS-LL total scores were 2.5 (2.0–2.9) and 8.1 (7.4–8.8) for the two subgroups. CI: confidence interval; NIS-LL: Neuropathy Impairment Score for the Lower Limbs.
Figure 4.
Figure 4.
Mean (95% CI) change from baseline in mBMI throughout 5.5 years of tafamidis treatment among 71 patients with baseline NIS-LL ≤ 10 at last assessment before first dose of tafamidis. Analyses were performed using observed cases. Year 5 data were excluded due to limited data (n = 2). CI: confidence interval; mBMI: modified body mass index.

References

    1. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, Lewis WD, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31.
    1. Sekijima Y. Transthyretin (ATTR) amyloidosis: clinical spectrum, molecular pathogenesis and disease-modifying treatments. J Neurol Neurosurg Psychiatry. 2015;86:1036–43.
    1. Rowczenio DM, Noor I, Gillmore JD, Lachmann HJ, Whelan C, Hawkins PN, Obici L, et al. Online registry for mutations in hereditary amyloidosis including nomenclature recommendations. Hum Mutat. 2014;35:E2403–12.
    1. Coutinho P, Martins da Silva A, Lopes Lima J, Resende Barbosa A. Forty years of experience with type I amyloid neuropathy: review of 483 cases. In: Glenner GG, Pinho e Costa P, Falcao de Freitas A, editors. Amyloid and amyloidosis. Amsterdam: Excerpta Medica; 1980. pp. 88–98.
    1. Conceição IM, González-Duarte A, Obici L, Harmut HH, Simoneau D, Ong ML, Amass L. “Red-flag” symptom clusters in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst. 2015;21:5–9.
    1. Adams D. Recent advances in the treatment of familial amyloid polyneuropathy. Ther Adv Neurol Disord. 2013;6:129–39.
    1. Planté-Bordeneuve V. Update in the diagnosis and management of transthyretin familial amyloid polyneuropathy. J Neurol. 2014;261:1227–33.
    1. Ericzon BG, Wilczek HE, Larsson M, Wijayatunga P, Stangou A, Pena JR, Furtado E, et al. Liver transplantation for hereditary transthyretin amyloidosis: after 20 years still the best therapeutic alternative? Transplantation. 2015;99:1847–54.
    1. Hanna M. Novel drugs targeting transthyretin amyloidosis. Curr Heart Fail Rep. 2014;11:50–7.
    1. Sekijima Y, Tojo K, Morita H, Koyama J, Ikeda S. Safety and efficacy of long-term diflunisal administration in hereditary transthyretin (ATTR) amyloidosis. Amyloid. 2015;22:79–83.
    1. Scott LJ. Tafamidis: a review of its use in familial amyloid polyneuropathy. Drugs. 2014;74:1371–8.
    1. Waddington Cruz M, Benson M. A review of tafamidis for the treatment of transthyretin-related amyloidosis. Neurol Ther. 2015;4:61–79.
    1. Coelho T, Maia LF, Martins da Silva A, Waddington Cruz M, Planté-Bordeneuve V, Lozeron P, Suhr OB, et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology. 2012;79:785–92.
    1. Coelho T, Maia LF, da Silva AM, Cruz MW, Planté-Bordeneuve V, Suhr OB, Conceicao I, et al. Long-term effects of tafamidis for the treatment of transthyretin familial amyloid polyneuropathy. J Neurol. 2013;260:2802–14.
    1. Coelho T, Merkies I, Vinik A, Vinik EJ, Chan J, Packman J, Grogan DR. Relationship between objective measures of neuropathy and quality of life in stages of severity of transthyretin familial amyloid polyneuropathy. Abstract P-098 of the XII International Symposium on Amyloidosis, April 18–21, 2010, Rome, Italy. Amyloid. 2010;17(Suppl 1):138.
    1. Suhr OB, Conceição IM, Karayal ON, Mandel FS, Huertas PE, Ericzon BG. Post hoc analysis of nutritional status in patients with transthyretin familial amyloid polyneuropathy: impact of tafamidis. Neurol Ther. 2014;3:101–12.
    1. Dyck PJ, Davies JL, Litchy WJ, O’Brien PC. Longitudinal assessment of diabetic polyneuropathy using a composite score in the Rochester Diabetic Neuropathy Study cohort. Neurology. 1997;49:229–39.
    1. Suhr O, Danielsson A, Holmgren G, Steen L. Malnutrition and gastrointestinal dysfunction as prognostic factors for survival in familial amyloidotic polyneuropathy. J Intern Med. 1994;235:479–85.
    1. Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T, Heneghan MA, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013;310:2658–67.

Source: PubMed

3
購読する