Efficacy and safety of tafamidis doses in the Tafamidis in Transthyretin Cardiomyopathy Clinical Trial (ATTR-ACT) and long-term extension study

Thibaud Damy, Pablo Garcia-Pavia, Mazen Hanna, Daniel P Judge, Giampaolo Merlini, Balarama Gundapaneni, Terrell A Patterson, Steven Riley, Jeffrey H Schwartz, Marla B Sultan, Ronald Witteles, Thibaud Damy, Pablo Garcia-Pavia, Mazen Hanna, Daniel P Judge, Giampaolo Merlini, Balarama Gundapaneni, Terrell A Patterson, Steven Riley, Jeffrey H Schwartz, Marla B Sultan, Ronald Witteles

Abstract

Aims: Tafamidis is an effective treatment for transthyretin amyloid cardiomyopathy (ATTR-CM) in the Tafamidis in Transthyretin Cardiomyopathy Clinical Trial (ATTR-ACT). While ATTR-ACT was not designed for a dose-specific assessment, further analysis from ATTR-ACT and its long-term extension study (LTE) can guide determination of the optimal dose.

Methods and results: In ATTR-ACT, patients were randomized (2:1:2) to tafamidis 80 mg, 20 mg, or placebo for 30 months. Patients completing ATTR-ACT could enrol in the LTE (with placebo-treated patients randomized to tafamidis 80 or 20 mg; 2:1) and all patients were subsequently switched to high-dose tafamidis. All-cause mortality was assessed in ATTR-ACT combined with the LTE (median follow-up 51 months). In ATTR-ACT, the combination of all-cause mortality and cardiovascular-related hospitalizations over 30 months was significantly reduced with tafamidis 80 mg (P = 0.0030) and 20 mg (P = 0.0048) vs. placebo. All-cause mortality vs. placebo was reduced with tafamidis 80 mg [Cox hazards model (95% confidence interval): 0.690 (0.487-0.979), P = 0.0378] and 20 mg [0.715 (0.450-1.137), P = 0.1564]. The mean (standard error) change in N-terminal pro-B-type natriuretic peptide from baseline to Month 30 was -1170.51 (587.31) (P = 0.0468) with tafamidis 80 vs. 20 mg. In ATTR-ACT combined with the LTE there was a significantly greater survival benefit with tafamidis 80 vs. 20 mg [0.700 (0.501-0.979), P = 0.0374]. Incidence of adverse events in both tafamidis doses were comparable to placebo.

Conclusion: Tafamidis, both 80 and 20 mg, effectively reduced mortality and cardiovascular-related hospitalizations in patients with ATTR-CM. The longer-term survival data and the lack of dose-related safety concerns support tafamidis 80 mg as the optimal dose.

Clinical trial registration: ClinicalTrials.gov NCT01994889; NCT02791230.

Keywords: Biomarkers; Clinical trial; Mortality; Transthyretin amyloid cardiomyopathy.

© 2020 The Authors. European Journal of Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.

Figures

Figure 1
Figure 1
Change from baseline with tafamidis 80 mg, tafamidis 20 mg, and placebo in (A) 6‐min walk test (6MWT) distance and (B) Kansas City Cardiomyopathy Questionnaire overall summary (KCCQ‐OS) in ATTR‐ACT. P‐values are for the treatment difference vs. placebo for tafamidis 80 mg (shown above the curve) and tafamidis 20 mg (shown below the curve). LS, least squares; SE, standard error.
Figure 2
Figure 2
Forest plot of covariate‐adjusted all‐cause mortality in ATTR‐ACT and ATTR‐ACT combined with the long‐term extension study (LTE). Hazard ratios are for: patients in ATTR‐ACT (30 months follow‐up), tafamidis 80 mg (n = 176) compared with 20 mg (n = 88); patients in ATTR‐ACT combined with the LTE and treatment with tafamidis free acid 61 mg (51 months follow‐up), tafamidis 80 mg/tafamidis free acid 61 mg (n = 230) compared with tafamidis 20 mg/tafamidis free acid 61 mg (n = 116). The unadjusted row was based on the prespecified Cox proportional hazards model with treatment, New York Heart Association baseline classification, and genotype in the model. The single covariate‐adjusted rows were generated by adding age, N‐terminal pro‐B‐type natriuretic peptide (NT‐proBNP) (log transformed), and 6‐min walk test (6MWT) distance separately as covariates to the pre‐specified model. The all‐covariate‐adjusted row was generated by adding all covariates to the pre‐specified model. Patients who discontinued due to heart or combined heart and liver transplantation, or due to implantation of a cardiac mechanical assist device, were counted as death for these analyses. *P‐values for ATTR‐ACT combined with the LTE (51 months of follow‐up) were: unadjusted, P = 0.0374; age‐adjusted, P = 0.0054; NT‐proBNP‐adjusted, P = 0.0099; 6MWT‐adjusted, P = 0.0444; age/NT‐proBNP/6MWT‐adjusted, P = 0.0030. CI, confidence interval.
Figure 3
Figure 3
Least squares (LS) mean (standard error, SE) change in (A) N‐terminal pro‐B‐type natriuretic peptide (NT‐proBNP) and (B) troponin I from baseline to Month 30 in ATTR‐ACT. The differences from placebo at Month 30 in NT‐proBNP and troponin I were significant for tafamidis 80 mg but not tafamidis 20 mg. (C) Cumulative distribution of percent change from baseline in NT‐proBNP at Month 30 with tafamidis 80 mg, tafamidis 20 mg, and placebo in ATTR‐ACT.

References

    1. Ruberg FL, Berk JL. Transthyretin (TTR) cardiac amyloidosis. Circulation 2012;126:1286–1300.
    1. Rapezzi C, Quarta CC, Riva L, Longhi S, Gallelli I, Lorenzini M, Ciliberti P, Biagini E, Salvi F, Branzi A. Transthyretin‐related amyloidoses and the heart: a clinical overview. Nat Rev Cardiol 2010;7:398–408.
    1. Almeida MR, Hesse A, Steinmetz A, Maisch B, Altland K, Linke RP, Gawinowicz MA, Saraiva MJ. Transthyretin Leu 68 in a form of cardiac amyloidosis. Basic Res Cardiol 1991;86:567–571.
    1. Jacobson DR, Pastore RD, Yaghoubian R, Kane I, Gallo G, Buck FS, Buxbaum JN. Variant‐sequence transthyretin (isoleucine 122) in late‐onset cardiac amyloidosis in black Americans. N Engl J Med 1997;336:466–473.
    1. Ranløv I, Alves IL, Ranløv PJ, Husby G, Costa PP, Saraiva MJ. A Danish kindred with familial amyloid cardiomyopathy revisited: identification of a mutant transthyretin‐methionine111 variant in serum from patients and carriers. Am J Med 1992;93:3–8.
    1. Sattianayagam PT, Hahn AF, Whelan CJ, Gibbs SD, Pinney JH, Stangou AJ, Rowczenio D, Pflugfelder PW, Fox Z, Lachmann HJ, Wechalekar AD, Hawkins PN, Gillmore JD. Cardiac phenotype and clinical outcome of familial amyloid polyneuropathy associated with transthyretin alanine 60 variant. Eur Heart J 2012;33:1120–1127.
    1. Damy T, Judge DP, Kristen AV, Berthet K, Li H, Aarts J. Cardiac findings and events observed in an open‐label clinical trial of tafamidis in patients with non‐Val30Met and non‐Val122Ile hereditary transthyretin amyloidosis. J Cardiovasc Transl Res 2015;8:117–127.
    1. Damy T, Maurer MS, Rapezzi C, Plante‐Bordeneuve V, Karayal ON, Mundayat R, Suhr OB, Kristen AV. Clinical, ECG and echocardiographic clues to the diagnosis of TTR‐related cardiomyopathy. Open Heart 2016;3:e000289.
    1. Rapezzi C, Quarta CC, Obici L, Perfetto F, Longhi S, Salvi F, Biagini E, Lorenzini M, Grigioni F, Leone O, Cappelli F, Palladini G, Rimessi P, Ferlini A, Arpesella G, Pinna AD, Merlini G, Perlini S. Disease profile and differential diagnosis of hereditary transthyretin‐related amyloidosis with exclusively cardiac phenotype: an Italian perspective. Eur Heart J 2013;34:520–528.
    1. Coelho T, Merlini G, Bulawa CE, Fleming JA, Judge DP, Kelly JW, Maurer MS, Plante‐Bordeneuve V, Labaudiniere R, Mundayat R, Riley S, Lombardo I, Huertas P. Mechanism of action and clinical application of tafamidis in hereditary transthyretin amyloidosis. Neurol Ther 2016;5:1–25.
    1. Bulawa CE, Connelly S, Devit M, Wang L, Weigel C, Fleming JA, Packman J, Powers ET, Wiseman RL, Foss TR, Wilson IA, Kelly JW, Labaudiniere R. Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc Natl Acad Sci U S A 2012;109:9629–9634.
    1. Maurer MS, Grogan DR, Judge DP, Mundayat R, Packman J, Lombardo I, Quyyumi AA, Aarts J, Falk RH. Tafamidis in transthyretin amyloid cardiomyopathy: effects on transthyretin stabilization and clinical outcomes. Circ Heart Fail 2015;8:519–526.
    1. Saelices L, Johnson LM, Liang WY, Sawaya MR, Cascio D, Ruchala P, Whitelegge J, Jiang L, Riek R, Eisenberg DS. Uncovering the mechanism of aggregation of human transthyretin. J Biol Chem 2015;290:28932–28943.
    1. Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington‐Cruz M, Kristen AV, Grogan M, Witteles R, Damy T, Drachman BM, Shah SJ, Hanna M, Judge DP, Barsdorf AI, Huber P, Patterson TA, Riley S, Schumacher J, Stewart M, Sultan MB, Rapezzi C; ATTR‐ACT Study Investigators. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med 2018;379:1007–1016.
    1. Maurer MS, Elliott P, Merlini G, Shah SJ, Waddington‐Cruz M, Flynn A, Gundapaneni B, Hahn C, Riley S, Schwartz J, Sultan MB, Rapezzi C. Design and rationale of the phase 3 ATTR‐ACT clinical trial (Tafamidis in Transthyretin Cardiomyopathy Clinical Trial). Circ Heart Fail 2017;10:e003815.
    1. Klamerus KJ, Watsky E, Moller R, Wang R, Riley S. The effect of tafamidis on the QTc interval in healthy subjects. Br J Clin Pharmacol 2015;79:918–925.
    1. Lockwood PA, Le VH, O'Gorman MT, Patterson TA, Sultan MB, Tankisheva E, Wang Q, Riley S. The bioequivalence of tafamidis 61‐mg free acid capsules and tafamidis meglumine 4 × 20‐mg capsules in healthy volunteers. Clin Pharmacol Drug Dev 2020;9:849–854.
    1. Finkelstein DM, Schoenfeld DA. Combining mortality and longitudinal measures in clinical trials. Stat Med 1999;18:1341–1354.
    1. ATS Committee on Proficiency Standards for Clinical Pulmonary Function Laboratories . ATS statement: guidelines for the six‐minute walk test. Am J Respir Crit Care Med 2002;166:111–117.
    1. Green CP, Porter CB, Bresnahan DR, Spertus JA. Development and evaluation of the Kansas City Cardiomyopathy Questionnaire: a new health status measure for heart failure. J Am Coll Cardiol 2000;35:1245–1255.
    1. Social Security Administration . Actuarial Life Table 2016. (20 October 2020).
    1. Witteles RM, Bokhari S, Damy T, Elliott PM, Falk RH, Fine NM, Gospodinova M, Obici L, Rapezzi C, Garcia‐Pavia P. Screening for transthyretin amyloid cardiomyopathy in everyday practice. JACC Heart Fail 2019;7:709–716.
    1. Grogan M, Scott CG, Kyle RA, Zeldenrust SR, Gertz MA, Lin G, Klarich KW, Miller WL, Maleszewski JJ, Dispenzieri A. Natural history of wild‐type transthyretin cardiac amyloidosis and risk stratification using a novel staging system. J Am Coll Cardiol 2016;68:1014–1020.
    1. Gillmore JD, Damy T, Fontana M, Hutchinson M, Lachmann HJ, Martinez‐Naharro A, Quarta CC, Rezk T, Whelan CJ, Gonzalez‐Lopez E, Lane T, Gilbertson JA, Rowczenio D, Petrie A, Hawkins PN. A new staging system for cardiac transthyretin amyloidosis. Eur Heart J 2018;39:2799–2806.
    1. Ingle L, Rigby AS, Carroll S, Butterly R, King RF, Cooke CB, Cleland JG, Clark AL. Prognostic value of the 6 min walk test and self‐perceived symptom severity in older patients with chronic heart failure. Eur Heart J 2007;28:560–568.
    1. Cho Y, Baranczak A, Helmke S, Teruya S, Horn EM, Maurer MS, Kelly JW. Personalized medicine approach for optimizing the dose of tafamidis to potentially ameliorate wild‐type transthyretin amyloidosis (cardiomyopathy). Amyloid 2015;22:175–180.
    1. Kristen AV, Maurer MS, Rapezzi C, Mundayat R, Suhr OB, Damy T. Impact of genotype and phenotype on cardiac biomarkers in patients with transthyretin amyloidosis – report from the Transthyretin Amyloidosis Outcome Survey (THAOS). PLoS One 2017;12:e0173086.

Source: PubMed

3
Abonnieren