A phase II, open-label, extension study of long-term patisiran treatment in patients with hereditary transthyretin-mediated (hATTR) amyloidosis

Teresa Coelho, David Adams, Isabel Conceição, Márcia Waddington-Cruz, Hartmut H Schmidt, Juan Buades, Josep Campistol, John L Berk, Michael Polydefkis, Jing Jing Wang, Jihong Chen, Marianne T Sweetser, Jared Gollob, Ole B Suhr, Teresa Coelho, David Adams, Isabel Conceição, Márcia Waddington-Cruz, Hartmut H Schmidt, Juan Buades, Josep Campistol, John L Berk, Michael Polydefkis, Jing Jing Wang, Jihong Chen, Marianne T Sweetser, Jared Gollob, Ole B Suhr

Abstract

Background: Patisiran, an RNA interference therapeutic, has demonstrated robust reduction of wild-type and mutant transthyretin protein and was able to improve polyneuropathy and quality of life following 18 months of treatment in patients with hereditary transthyretin-mediated (hATTR) amyloidosis. In this 24-month Phase II open-label extension study, we evaluated the effects of patisiran treatment (0.3 mg/kg intravenously every 3 weeks) on safety, serum transthyretin levels, and clinical parameters. Efficacy assessments included modified Neuropathy Impairment Score +7 (mNIS+7) and multiple disease-relevant measures. Cardiac assessments were performed in a pre-specified cardiac subgroup.

Results: Twenty-seven patients entered this study, including 12 (44%) with ambulation difficulties due to their neuropathy and 11 (41%) who met criteria for the cardiac subgroup. During treatment, the majority of adverse events were mild/moderate in severity; there were no drug-related adverse events leading to treatment discontinuation. The most common drug-related adverse events were flushing and infusion-related reactions (22% each). Patisiran resulted in rapid, robust (~ 82%), and sustained reduction of mean transthyretin levels over 24 months. A mean 6.95-point decrease (improvement) in mNIS+7 from baseline was observed at 24 months. Patisiran's impact on mNIS+7 was irrespective of concomitant tafamidis or diflunisal use, sex, or age. Clinical assessments of motor function, autonomic symptoms, disease stage, and quality of life remained stable over 24 months. No significant changes were observed for echocardiographic measures or cardiac biomarkers in the cardiac subgroup. Exploratory analyses demonstrated improvements in nerve-fiber density with corresponding reductions in amyloid burden observed in skin biopsies over 24 months.

Conclusions: Long-term treatment with patisiran had an acceptable safety profile and was associated with halting/improvement of polyneuropathy progression in patients with hATTR amyloidosis.

Trial registration: The study was registered at ClinicalTrials.gov (identifier: NCT01961921 ) on October 14, 2013.

Keywords: ATTR amyloidosis; Cardiomyopathy; Patisiran; Polyneuropathy; RNA interference.

Conflict of interest statement

TC reports compensation for training from Pfizer Pharmaceuticals and Alnylam Pharmaceuticals Inc., and compensation for travel from Pfizer Pharmaceuticals, Alnylam Pharmaceuticals Inc., and IONIS Pharmaceuticals. DA reports grants from Alnylam Pharmaceuticals Inc., and personal fees from Pfizer Pharmaceuticals and Alnylam Pharmaceuticals Inc. outside the submitted work. IC acknowledges financial support as a primary investigator for FoldRx Pharmaceuticals/Pfizer Inc., Alnylam Pharmaceuticals Inc., and IONIS Pharmaceuticals, and serves on the Transthyretin Amyloidosis Outcome Survey (THAOS) scientific advisory board. MWC received honorarium from NHI, Prothena, FoldRx, Akcea Therapeutics, Pfizer Pharmaceuticals, Alnylam Pharmaceuticals Inc., PTC, and Genzyme for travel expenses related to presentations at medical meetings, and for acting as a consultant and as a principal investigator in clinical trials. HHS reports grants and consulting fees from Alnylam Pharmaceuticals Inc. outside the submitted work. JBu reports personal fees from Alnylam Pharmaceuticals Inc. during the conduct of the study for serving as a principal investigator. JCo has nothing to disclose. JBe reports serving as a study investigator for IONIS Pharmaceuticals and Pfizer Pharmaceuticals, and consultant for Intellia Therapeutics Inc. and Corino Therapeutics Inc. outside the submitted work. MP reports grants from Alnylam Pharmaceuticals Inc. during the conduct of the study and honorarium following completion of the study. JJW, JCh, MTS, and JG are all Alnylam employees. OBS reports personal fees from Alnylam Pharmaceuticals Inc. during the conduct of the study, personal fees and non-financial support from Pfizer Pharmaceuticals, and personal fees from Prothena Pharmaceuticals and Intellia Therapeutics, Inc. outside the submitted work.

Figures

Fig. 1
Fig. 1
Serum TTR reduction. Percentage change in serum TTR from baseline over time. Pre- and post-dose (0.3 mg/kg Q3W patisiran) values are indicated on the graph with an x. BSL, baseline; ELISA, enzyme-linked immunosorbent assay; Q3W, every 3 weeks; TTR, transthyretin
Fig. 2
Fig. 2
Mean change from baseline (SEM) in mNIS+7 in the all-treated population over 24 months. Error bars represent the SEM. mNIS+7, modified Neuropathy Impairment Score +7; SEM, standard error of the mean
Fig. 3
Fig. 3
Change in sweat gland nerve-fiber density and dermal amyloid burden of the lower limb. a Change in sweat gland nerve-fiber density in distal thigh and distal leg to 24 months. b Distal thigh sweat gland innervation at baseline and at Month 24 in an individual patient, with nerve fibers immunostained for PGP 9·5 (green), blood vessels immunostained for CD31 (red), and cell nuclei labeled with DAPI (blue). c Change from baseline to 24 months in amyloid burden for the lower limb. The statistical significance (performed as a post hoc analysis) of the change from baseline is shown for each time-point where: *p = 0.01–0.05, **p = 0.001–0.01. BSL, baseline; CD31, cluster of differentiation 31 protein (or platelet endothelial cell adhesion molecule [PECAM1]); DAPI, 4′,6-diamidino-2-phenylindole; PGP, protein gene product

References

    1. Hanna M. Novel drugs targeting transthyretin amyloidosis. Curr Heart Fail Rep. 2014;11(1):50–57. doi: 10.1007/s11897-013-0182-4.
    1. Adams D, Koike H, Slama M, Coelho T. Hereditary transthyretin amyloidosis: a model of medical progress for a fatal disease. Nat Rev Neurol. 2019;15(7):387–404. doi: 10.1038/s41582-019-0210-4.
    1. Soprano DR, Herbert J, Soprano KJ, Schon EA, Goodman DS. Demonstration of transthyretin mRNA in the brain and other extrahepatic tissues in the rat. J Biol Chem. 1985;260(21):11793–11798.
    1. Sekijima Y. Recent progress in the understanding and treatment of transthyretin amyloidosis. J Clin Pharm Ther. 2014;39(3):225–233. doi: 10.1111/jcpt.12145.
    1. Sekijima Y, et al. Hereditary transthyretin amyloidosis. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, et al., editors. GeneReviews®. Seattle: University of Washington; 2001.
    1. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625–638. doi: 10.3109/07853890.2015.1068949.
    1. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31. doi: 10.1186/1750-1172-8-31.
    1. Rapezzi C, Quarta CC, Obici L, Perfetto F, Longhi S, Salvi F, et al. Disease profile and differential diagnosis of hereditary transthyretin-related amyloidosis with exclusively cardiac phenotype: an Italian perspective. Eur Heart J. 2013;34(7):520–528. doi: 10.1093/eurheartj/ehs123.
    1. Coelho T, Maurer MS, Suhr OB. THAOS – the Transthyretin amyloidosis outcomes survey: initial report on clinical manifestations in patients with hereditary and wild-type transthyretin amyloidosis. Curr Med Res Opin. 2013;29(1):63–76. doi: 10.1185/03007995.2012.754348.
    1. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11–21. doi: 10.1056/NEJMoa1716153.
    1. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22–31. doi: 10.1056/NEJMoa1716793.
    1. Swiecicki PL, Zhen DB, Mauermann ML, Kyle RA, Zeldenrust SR, Grogan M, et al. Hereditary ATTR amyloidosis: a single-institution experience with 266 patients. Amyloid. 2015;22(2):123–131. doi: 10.3109/13506129.2015.1019610.
    1. Sattianayagam PT, Hahn AF, Whelan CJ, Gibbs SD, Pinney JH, Stangou AJ, et al. Cardiac phenotype and clinical outcome of familial amyloid polyneuropathy associated with transthyretin alanine 60 variant. Eur Heart J. 2012;33(9):1120–1127. doi: 10.1093/eurheartj/ehr383.
    1. Ericzon BG, Wilczek HE, Larsson M, Wijayatunga P, Stangou A, Pena JR, et al. Liver transplantation for hereditary transthyretin amyloidosis: after 20 years still the best therapeutic alternative? Transplantation. 2015;99(9):1847–1854. doi: 10.1097/TP.0000000000000574.
    1. Carvalho A, Rocha A, Lobato L. Liver transplantation in transthyretin amyloidosis: issues and challenges. Liver Transpl. 2015;21(3):282–292. doi: 10.1002/lt.24058.
    1. Waddington Cruz M, Benson MD. A review of tafamidis for the treatment of transthyretin-related amyloidosis. Neurol Ther. 2015;4(2):61–79. doi: 10.1007/s40120-015-0031-3.
    1. Coelho T, Maia LF. Martins da Silva a, Waddington Cruz M, Plante-Bordeneuve V, Lozeron P, et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology. 2012;79(8):785–792. doi: 10.1212/WNL.0b013e3182661eb1.
    1. Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013;310(24):2658–2667. doi: 10.1001/jama.2013.283815.
    1. Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington-Cruz M, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379(11):1007–1016. doi: 10.1056/NEJMoa1805689.
    1. Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med. 2013;369(9):819–829. doi: 10.1056/NEJMoa1208760.
    1. Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, et al. Efficacy and safety of patisiran for familial amyloidotic polyneuropathy: a phase II multi-dose study. Orphanet J Rare Dis. 2015;10:109. doi: 10.1186/s13023-015-0326-6.
    1. Alnylam Pharmaceuticals Inc . US prescribing information: ONPATTRO (patisiran) lipid complex injection, for intravenous use. 2018.
    1. European Medicines Agency . Summary of product characteristics: Onpattro 2 mg/mL concentrate for solution for infusion. 2018.
    1. Solomon SD, Adams D, Kristen A, Grogan M, Gonzalez-Duarte A, Maurer MS, et al. Effects of patisiran, an RNA interference therapeutic, on cardiac parameters in patients with hereditary transthyretin-mediated amyloidosis. Circulation. 2019;139(4):431–443. doi: 10.1161/CIRCULATIONAHA.118.035831.
    1. Adams D, Suhr OB, Dyck PJ, Litchy WJ, Leahy RG, Chen J, et al. Trial design and rationale for APOLLO, a phase 3, placebo-controlled study of patisiran in patients with hereditary ATTR amyloidosis with polyneuropathy. BMC Neurol. 2017;17(1):181. doi: 10.1186/s12883-017-0948-5.
    1. Liz MA, Mar FM, Franquinho F, Sousa MM. Aboard transthyretin: from transport to cleavage. IUBMB Life. 2010;62(6):429–435.
    1. Chi X, Gatti P, Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today. 2017;22(5):823–833. doi: 10.1016/j.drudis.2017.01.013.
    1. Biesalski H, Reifen R, Furst P, Edris M. Retinyl palmitate supplementation by inhalation of an aerosol improves vitamin A status of preschool children in Gondar (Ethiopia). Br J Nutr. 1999;82(3):179–82.
    1. Mariani LL, Lozeron P, Theaudin M, Mincheva Z, Signate A, Ducot B, et al. Genotype-phenotype correlation and course of transthyretin familial amyloid polyneuropathies in France. Ann Neurol. 2015;78(6):901–916. doi: 10.1002/ana.24519.
    1. Ziegler D, Low PA, Litchy WJ, Boulton AJ, Vinik AI, Freeman R, et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care. 2011;34(9):2054–60.
    1. Adams D, Coelho T, Obici L, Merlini G, Mincheva Z, Suanprasert N, et al. Rapid progression of familial amyloidotic polyneuropathy: a multinational natural history study. Neurology. 2015;85(8):675–682. doi: 10.1212/WNL.0000000000001870.
    1. Lozeron P, Theaudin M, Mincheva Z, Ducot B, Lacroix C, Adams D, et al. Effect on disability and safety of tafamidis in late onset of Met30 transthyretin familial amyloid polyneuropathy. Eur J Neurol. 2013;20(12):1539–1545. doi: 10.1111/ene.12225.
    1. Plante-Bordeneuve V, Gorram F, Salhi H, Nordine T, Ayache SS, Le Corvoisier P, et al. Long-term treatment of transthyretin familial amyloid polyneuropathy with tafamidis: a clinical and neurophysiological study. J Neurol. 2017;264(2):268–276. doi: 10.1007/s00415-016-8337-3.
    1. Dyck PJB, González-Duarte A, Obici L, Polydefkis M, Wiesman JF, Antonino I, et al. Development of measures of polyneuropathy impairment in hATTR amyloidosis: from NIS to mNIS+7. J Neurol Sci. 2019;405:116424. doi: 10.1016/j.jns.2019.116424.
    1. Taubel J, Zimmermann T, Karsten V, Martinez C, Chan A, Wang Y, et al. Phase 1 study of ALN-TTRsc02, a subcutaneously administered investigational RNAi therapeutic for the treatment of transthyretin-mediated amyloidosis. XVIth International Symposium on Amyloidosis (ISA) Kumamoto: Poster; 2018. pp. 13–16.

Source: PubMed

3
購読する