611
Views
52
CrossRef citations to date
0
Altmetric
Review

Diagnosis and Treatment of Hereditary Transthyretin Amyloidosis (hATTR) Polyneuropathy: Current Perspectives on Improving Patient Care

ORCID Icon, ORCID Icon, , &
Pages 109-123 | Published online: 21 Feb 2020

References

  • Planté-Bordeneuve V, Said G. Familial amyloid polyneuropathy. Lancet Neurol. 2011;10(12):1086–1097. doi:10.1016/S1474-4422(11)70246-0
  • 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
  • Ruberg FL, Berk JL. Transthyretin (TTR) cardiac amyloidosis. Circulation. 2012;126(10):1286–1300. doi:10.1161/CIRCULATIONAHA.111.078915
  • Pinney JH, Whelan CJ, Petrie A, et al. Senile systemic amyloidosis: clinical features at presentation and outcome. J Am Heart Assoc. 2013;2(2):e000098. doi:10.1161/JAHA.113.000098
  • Saraiva MJ, Birken S, Costa PP, Goodman DS. Family studies of the genetic abnormality in transthyretin (prealbumin) in Portuguese patients with familial amyloidotic polyneuropathy. Ann N Y Acad Sci. 1984;435:86–100. doi:10.1111/j.1749-6632.1984.tb13742.x
  • Coelho T, Maia LF, Martins da Silva A, et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology. 2012;79(8):785–792. doi:10.1212/WNL.0b013e3182661eb1
  • Adams D, Cauquil C, Labeyrie C, Beaudonnet G, Algalarrondo V, Théaudin M. TTR kinetic stabilizers and TTR gene silencing: a new era in therapy for familial amyloidotic polyneuropathies. Expert Opin Pharmacother. 2016;17(6):791–802. doi:10.1517/14656566.2016.1145664
  • Adams D, Gonzalez-Duarte A, O’Riordan WD, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11–21. doi:10.1056/NEJMoa1716153
  • Benson MD, Waddington-Cruz M, Berk JL, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22–31. doi:10.1056/NEJMoa1716793
  • Holmgren G, Steen L, Ekstedt J, et al. Biochemical effect of liver transplantation in two Swedish patients with familial amyloidotic polyneuropathy (FAP-met30). Clin Genet. 1991;40(3):242–246. doi:10.1111/j.1399-0004.1991.tb03085.x
  • Andrade C. A peculiar form of peripheral neuropathy; familiar atypical generalized amyloidosis with special involvement of the peripheral nerves. Brain J Neurol. 1952;75(3):408–427. doi:10.1093/brain/75.3.408
  • Dardiotis E, Koutsou P, Papanicolaou EZ, et al. Epidemiological, clinical and genetic study of familial amyloidotic polyneuropathy in Cyprus. Amyloid. 2009;16(1):32–37. doi:10.1080/13506120802676948
  • Reinés JB, Vera TR, Martín MU, et al. Epidemiology of transthyretin-associated familial amyloid polyneuropathy in the Majorcan area: Son Llàtzer Hospital descriptive study. Orphanet J Rare Dis. 2014;9:29. doi:10.1186/1750-1172-9-29
  • Tsuzuki T, Mita S, Maeda S, Araki S, Shimada K. Structure of the human prealbumin gene. J Biol Chem. 1985;260(22):12224–12227.
  • Koike H, Misu K, Ikeda S, et al. Type I (transthyretin Met30) familial amyloid polyneuropathy in Japan: early- vs late-onset form. Arch Neurol. 2002;59(11):1771–1776. doi:10.1001/archneur.59.11.1771
  • Parman Y, Adams D, Obici L, et al. Sixty years of transthyretin familial amyloid polyneuropathy (TTR-FAP) in Europe: where are we now? A European network approach to defining the epidemiology and management patterns for TTR-FAP. Curr Opin Neurol. 2016;29(Suppl 1):S3–S13. doi:10.1097/WCO.0000000000000288
  • Saporta MAC, Zaros C, MW C, et al. Penetrance estimation of TTR familial amyloid polyneuropathy (type I) in Brazilian families. Eur J Neurol. 2009;16(3):337–341. doi:10.1111/j.1468-1331.2008.02429.x
  • Planté-Bordeneuve V, Carayol J, Ferreira A, et al. Genetic study of transthyretin amyloid neuropathies: carrier risks among French and Portuguese families. J Med Genet. 2003;40(11):e120. doi:10.1136/jmg.40.11.e120
  • Hellman U, Alarcon F, Lundgren H-E, Suhr OB, Bonaiti-Pellié C, Planté-Bordeneuve V. Heterogeneity of penetrance in familial amyloid polyneuropathy, ATTR Val30Met, in the Swedish population. Amyloid. 2008;15(3):181–186. doi:10.1080/13506120802193720
  • Soares ML, Coelho T, Sousa A, et al. Haplotypes and DNA sequence variation within and surrounding the transthyretin gene: genotype-phenotype correlations in familial amyloid polyneuropathy (V30M) in Portugal and Sweden. Eur J Hum Genet. 2004;12(3):225–237. doi:10.1038/sj.ejhg.5201095
  • Bonaïti B, Alarcon F, Bonaïti-Pellié C, Planté-Bordeneuve V. Parent-of-origin effect in transthyretin related amyloid polyneuropathy. Amyloid. 2009;16(3):149–150. doi:10.1080/13506120903093944
  • Richardson SJ. Cell and molecular biology of transthyretin and thyroid hormones. Int Rev Cytol. 2007;258:137–193. doi:10.1016/S0074-7696(07)58003-4
  • Ando Y, Coelho T, Berk JL, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31. doi:10.1186/1750-1172-8-31
  • Rapezzi C, Quarta CC, Obici L, 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
  • Maurer MS, Hanna M, Grogan M, et al. Genotype and phenotype of transthyretin cardiac amyloidosis: THAOS (transthyretin amyloid outcome survey). J Am Coll Cardiol. 2016;68(2):161–172. doi:10.1016/j.jacc.2016.03.596
  • Yang N-C-C, Lee M-J, Chao -C-C, et al. Clinical presentations and skin denervation in amyloid neuropathy due to transthyretin Ala97Ser. Neurology. 2010;75(6):532–538. doi:10.1212/WNL.0b013e3181ec7fda
  • González-Duarte A, Soto KC, Martínez-Baños D, et al. Familial amyloidosis with polyneuropathy associated with TTR Ser50Arg mutation. Amyloid. 2012;19(4):171–176. doi:10.3109/13506129.2012.712925
  • Sousa A, Andersson R, Drugge U, Holmgren G, Sandgren O. Familial amyloidotic polyneuropathy in Sweden: geographical distribution, age of onset, and prevalence. Hum Hered. 1993;43(5):288–294. doi:10.1159/000154146
  • Coelho T, Sousa A, Lourenço E, Ramalheira J. A study of 159 Portuguese patients with familial amyloidotic polyneuropathy (FAP) whose parents were both unaffected. J Med Genet. 1994;31(4):293–299. doi:10.1136/jmg.31.4.293
  • Suhr OB, Lundgren E, Westermark P. One mutation, two distinct disease variants: unravelling the impact of transthyretin amyloid fibril composition. J Intern Med. 2017;281(4):337–347. doi:10.1111/joim.12585
  • Koike H, Tanaka F, Hashimoto R, et al. Natural history of transthyretin Val30Met familial amyloid polyneuropathy: analysis of late-onset cases from non-endemic areas. J Neurol Neurosurg Psychiatry. 2012;83(2):152–158. doi:10.1136/jnnp-2011-301299
  • Mariani -L-L, Lozeron P, Théaudin M, 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
  • Ruberg FL, Maurer MS, Judge DP, et al. Prospective evaluation of the morbidity and mortality of wild-type and V122I mutant transthyretin amyloid cardiomyopathy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am Heart J. 2012;164(2):222–228.e1. doi:10.1016/j.ahj.2012.04.015
  • Gillmore JD, Damy T, Fontana M, et al. A new staging system for cardiac transthyretin amyloidosis. Eur Heart J. 2018;39(30):2799–2806. doi:10.1093/eurheartj/ehx589
  • Théaudin M, Lozeron P, Algalarrondo V, et al. Upper limb onset of hereditary transthyretin amyloidosis is common in non-endemic areas. Eur J Neurol. 2019;26(3):497–e36. doi:10.1111/ene.13845
  • Lobato L, Rocha A. Transthyretin amyloidosis and the kidney. Clin J Am Soc Nephrol. 2012;7(8):1337–1346. doi:10.2215/CJN.08720811
  • Beirão JM, Malheiro J, Lemos C, Beirão I, Costa P, Torres P. Ophthalmological manifestations in hereditary transthyretin (ATTR V30M) carriers: a review of 513 cases. Amyloid. 2015;22(2):117–122. doi:10.3109/13506129.2015.1015678
  • Martins AC, Rosa AM, Costa E, Tavares C, Quadrado MJ, Murta JN. Ocular manifestations and therapeutic options in patients with familial amyloid polyneuropathy: a systematic review. Biomed Res Int. 2015;2015:282405. doi:10.1155/2015/282405
  • Hagiwara K, Ochi H, Suzuki S, et al. Highly selective leptomeningeal amyloidosis with transthyretin variant Ala25Thr. Neurology. 2009;72(15):1358–1360. doi:10.1212/WNL.0b013e3181a0fe74
  • JL Z, Greicius MD, Zhu W, AN O, CM A, Plowey ED. Neuropathologic analysis of Tyr69His TTR variant meningovascular amyloidosis with dementia. Acta Neuropathol Commun. 2015;3:43. doi:10.1186/s40478-015-0216-0
  • Mitsuhashi S, Yazaki M, Tokuda T, et al. Biochemical characteristics of variant transthyretins causing hereditary leptomeningeal amyloidosis. Amyloid. 2005;12(4):216–225. doi:10.1080/13506120500352404
  • Adams D, Suhr OB, Hund E, et al. First European consensus for diagnosis, management, and treatment of transthyretin familial amyloid polyneuropathy. Curr Opin Neurol. 2016;29(Suppl 1):S14–S26. doi:10.1097/WCO.0000000000000289
  • Dohrn MF, Röcken C, De Bleecker JL, et al. Diagnostic hallmarks and pitfalls in late-onset progressive transthyretin-related amyloid-neuropathy. J Neurol. 2013;260(12):3093–3108. doi:10.1007/s00415-013-7124-7
  • Luigetti M, Conte A, Del Grande A, et al. TTR-related amyloid neuropathy: clinical, electrophysiological and pathological findings in 15 unrelated patients. Neurol Sci. 2013;34(7):1057–1063. doi:10.1007/s10072-012-1105-y
  • Lozeron P, Lacroix C, Theaudin M, et al. An amyotrophic lateral sclerosis-like syndrome revealing an amyloid polyneuropathy associated with a novel transthyretin mutation. Amyloid. 2013;20(3):188–192. doi:10.3109/13506129.2013.818535
  • Lozeron P, Mariani -L-L, Dodet P, et al. Transthyretin amyloid polyneuropathies mimicking a demyelinating polyneuropathy. Neurology. 2018;91(2):e143–e152. doi:10.1212/WNL.0000000000005777
  • Cortese A, Vegezzi E, Lozza A, et al. Diagnostic challenges in hereditary transthyretin amyloidosis with polyneuropathy: avoiding misdiagnosis of a treatable hereditary neuropathy. J Neurol Neurosurg Psychiatry. 2017;88(5):457–458. doi:10.1136/jnnp-2016-315262
  • Westermark P. Diagnosing amyloidosis. Scand J Rheumatol. 1995;24(6):327–329. doi:10.3109/03009749509095175
  • Do Amaral B, Coelho T, Sousa A, Guimarães A. Usefulness of labial salivary gland biopsy in familial amyloid polyneuropathy Portuguese type. Amyloid. 2009;16(4):232–238. doi:10.3109/13506120903421850
  • Cappellari M, Cavallaro T, Ferrarini M, et al. Variable presentations of TTR-related familial amyloid polyneuropathy in seventeen patients. J Peripher Nerv Syst. 2011;16(2):119–129. doi:10.1111/j.1529-8027.2011.00331.x
  • Koike H, Hashimoto R, Tomita M, et al. Diagnosis of sporadic transthyretin Val30Met familial amyloid polyneuropathy: a practical analysis. Amyloid. 2011;18(2):53–62. doi:10.3109/13506129.2011.565524
  • Koike H, Kawagashira Y, Iijima M, et al. Electrophysiological features of late-onset transthyretin Met30 familial amyloid polyneuropathy unrelated to endemic foci. J Neurol. 2008;255(10):1526–1533. doi:10.1007/s00415-008-0962-z
  • Castro J, Miranda B, Castro I, de Carvalho M, Conceição I. The diagnostic accuracy of Sudoscan in transthyretin familial amyloid polyneuropathy. Clin Neurophysiol. 2016;127(5):2222–2227. doi:10.1016/j.clinph.2016.02.013
  • Lefaucheur J-P, Zouari HG, Gorram F, Nordine T, Damy T, Planté-Bordeneuve V. The value of electrochemical skin conductance measurement using Sudoscan® in the assessment of patients with familial amyloid polyneuropathy. Clin Neurophysiol. 2018;129(8):1565–1569. doi:10.1016/j.clinph.2018.05.005
  • Luigetti M, Bisogni G, Romano A, et al. Sudoscan in the evaluation and follow-up of patients and carriers with TTR mutations: experience from an Italian Centre. Amyloid. 2018;25(4):242–246. doi:10.1080/13506129.2018.1545640
  • Niklasson U, Olofsson BO, Bjerle P. Autonomic neuropathy in familial amyloidotic polyneuropathy. A clinical study based on heart rate variability. Acta Neurol Scand. 1989;79(3):182–187. doi:10.1111/j.1600-0404.1989.tb03736.x
  • Buxbaum JN, Ruberg FL. Transthyretin V122I (pV142I)* cardiac amyloidosis: an age-dependent autosomal dominant cardiomyopathy too common to be overlooked as a cause of significant heart disease in elderly African Americans. Genet Med off J Am Coll Med Genet. 2017;19(7):733–742. doi:10.1038/gim.2016.200
  • Sattianayagam PT, Hahn AF, Whelan CJ, 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
  • Gillmore JD, Maurer MS, Falk RH, et al. Nonbiopsy diagnosis of cardiac transthyretin amyloidosis. Circulation. 2016;133(24):2404–2412. doi:10.1161/CIRCULATIONAHA.116.021612
  • Musumeci MB, Cappelli F, Russo D, et al. Low sensitivity of bone scintigraphy in detecting Phe64Leu mutation-related transthyretin cardiac amyloidosis. JACC Cardiovasc Imaging. 2019. doi:10.1016/j.jcmg.2019.10.015
  • Lehrke S, Steen H, Kristen AV, et al. Serum levels of NT-proBNP as surrogate for cardiac amyloid burden: new evidence from gadolinium-enhanced cardiac magnetic resonance imaging in patients with amyloidosis. Amyloid. 2009;16(4):187–195. doi:10.3109/13506120903421538
  • Coll E, Botey A, Alvarez L, et al. Serum cystatin C as a new marker for noninvasive estimation of glomerular filtration rate and as a marker for early renal impairment. Am J Kidney Dis. 2000;36(1):29–34. doi:10.1053/ajkd.2000.8237
  • 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(5):479–485. doi:10.1111/j.1365-2796.1994.tb01106.x
  • Coutinho PM, Lázaro da Silva A, Lopes J, et al. Forty years of experience with type I amyloid neuropathy. Review of 483 cases. In: editors, Glenner GG, Pinho e Costa P, Falcao de Freitas A. Amyloid and Amyloidosis. Amsterdam: Excerpta Medica; 1980:88–98. Available from: https://www.scienceopen.com/document?vid=3a81c019-a30c-4c5a-9d7a-2db26a111dd2. Accessed September 10, 2019..
  • 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(1):229–239. doi:10.1212/wnl.49.1.229
  • Berk JL, Suhr OB, Obici L, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013;310(24):2658–2667. doi:10.1001/jama.2013.283815
  • Dyck PJB, González-Duarte A, Obici L, 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
  • Suarez GA, Opfer-Gehrking TL, Offord KP, Atkinson EJ, O’Brien PC, Low PA. The autonomic symptom profile: a new instrument to assess autonomic symptoms. Neurology. 1999;52(3):523–528. doi:10.1212/wnl.52.3.523
  • Denier C, Ducot B, Husson H, et al. A brief compound test for assessment of autonomic and sensory-motor dysfunction in familial amyloid polyneuropathy. J Neurol. 2007;254(12):1684–1688. doi:10.1007/s00415-007-0617-5
  • van Nes SI, Vanhoutte EK, van Doorn PA, et al. Rasch-built overall disability scale (R-ODS) for immune-mediated peripheral neuropathies. Neurology. 2011;76(4):337–345. doi:10.1212/WNL.0b013e318208824b
  • Vinik EJ, Hayes RP, Oglesby A, et al. The development and validation of the Norfolk QOL-DN, a new measure of patients’ perception of the effects of diabetes and diabetic neuropathy. Diabetes Technol Ther. 2005;7(3):497–508. doi:10.1089/dia.2005.7.497
  • Adams D, Suhr OB, Dyck PJ, 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
  • Conceição I, Damy T, Romero M, et al. Early diagnosis of ATTR amyloidosis through targeted follow-up of identified carriers of TTR gene mutations. Amyloid. 2019;26(1):3–9. doi:10.1080/13506129.2018.1556156
  • Obici L, Kuks JB, Buades J, et al. Recommendations for presymptomatic genetic testing and management of individuals at risk for hereditary transthyretin amyloidosis. Curr Opin Neurol. 2016;29(Suppl 1):S27–S35. doi:10.1097/WCO.0000000000000290
  • Wilczek HE, Larsson M, Ericzon B-G, FAPWTR. Long-term data from the Familial Amyloidotic Polyneuropathy World Transplant Registry (FAPWTR). Amyloid. 2011;18(Suppl 1):193–195. doi:10.3109/13506129.2011.574354072
  • Ericzon B-G, Wilczek HE, Larsson M, 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
  • Saelices L, Chung K, Lee JH, et al. Amyloid seeding of transthyretin by ex vivo cardiac fibrils and its inhibition. Proc Natl Acad Sci U S A. 2018;115(29):E6741–E6750. doi:10.1073/pnas.1805131115
  • Saelices L, Nguyen BA, Chung K, et al. A pair of peptides inhibits seeding of the hormone transporter transthyretin into amyloid fibrils. J Biol Chem. 2019;294(15):6130–6141. doi:10.1074/jbc.RA118.005257
  • 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
  • Said G, Grippon S, Kirkpatrick P. Tafamidis. Nat Rev Drug Discov. 2012;11(3):185–186. doi:10.1038/nrd3675
  • Lozeron P, Théaudin M, Mincheva Z, 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
  • Cortese A, Vita G, Luigetti M, et al. Monitoring effectiveness and safety of Tafamidis in transthyretin amyloidosis in Italy: a longitudinal multicenter study in a non-endemic area. J Neurol. 2016;263(5):916–924. doi:10.1007/s00415-016-8064-9
  • Planté-Bordeneuve V, Gorram F, Salhi H, 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
  • Maurer MS, Schwartz JH, Gundapaneni B, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379(11):1007–1016. doi:10.1056/NEJMoa1805689
  • Akinc A, Querbes W, De S, et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther J Am Soc Gene Ther. 2010;18(7):1357–1364. doi:10.1038/mt.2010.85
  • Butler JS, Chan A, Costelha S, et al. Preclinical evaluation of RNAi as a treatment for transthyretin-mediated amyloidosis. Amyloid. 2016;23(2):109–118. doi:10.3109/13506129.2016.1160882
  • Coelho T, Adams D, Silva A, et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med. 2013;369(9):819–829. doi:10.1056/NEJMoa1208760
  • Nair JK, Willoughby JLS, Chan A, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958–16961. doi:10.1021/ja505986a
  • Ackermann EJ, Guo S, Benson MD, et al. Suppressing transthyretin production in mice, monkeys and humans using 2nd-Generation antisense oligonucleotides. Amyloid. 2016;23(3):148–157. doi:10.1080/13506129.2016.1191458
  • Ferreira N, Saraiva MJ, Almeida MR. Epigallocatechin-3-gallate as a potential therapeutic drug for TTR-related amyloidosis: “in vivo” evidence from FAP mice models. PLoS One. 2012;7(1):e29933. doi:10.1371/journal.pone.0029933
  • Higaki JN, Chakrabartty A, Galant NJ, et al. Novel conformation-specific monoclonal antibodies against amyloidogenic forms of transthyretin. Amyloid. 2016;23(2):86–97. doi:10.3109/13506129.2016.1148025