You are on page 1of 15

Received: 17 April 2018 | Revised: 9 July 2018 | Accepted: 4 September 2018

DOI: 10.1111/ane.13035

REVIEW ARTICLE

Hereditary transthyretin‐related amyloidosis

Josef Finsterer1 | Stephan Iglseder2 | Julia Wanschitz3 | Raffi Topakian4 |


Wolfgang N. Löscher3 | Wolfgang Grisold5

1
Krankenanstalt Rudolfstiftung, Vienna,
Austria Hereditary transthyretin(TTR)‐related amyloidosis (ATTRm amyloidosis) is an en‐
2
Konventhospital Barmherzige Brüder, Linz, demic/non‐endemic, autosomal‐dominant, early‐ and late‐onset, rare, progressive
Austria disorder, predominantly manifesting as length‐dependent, small fiber dominant, ax‐
3
Department of Neurology, Medical
onal polyneuropathy and frequently associated with cardiac disorders and other mul‐
University Innsbruck, Innsbruck, Austria
4
Department of Neurology, Klinikum Wels‐
tisystem diseases. ATTRm amyloidosis is due to variants in the TTR gene, with the
Grieskirchen, Wels, Austria substitution Val30Met as the most frequent mutation. TTR mutations lead to desta‐
5
Ludwig Boltzmann Institute for bilization and dissociation of TTR tetramers into variant TTR monomers, and forma‐
Experimental und Clinical Traumatology,
Vienna, Austria tion of amyloid fibrils, which are consecutively deposited extracellularly in various
tissues, such as nerves, heart, brain, eyes, intestines, kidneys, or the skin. Neuropathy
Correspondence
Josef Finsterer, Krankenanstalt may not only include large nerve fibers but also small fibers, and not only sensory and
Rudolfstiftung, Vienna, Austria. motor fibers but also autonomic fibers. Types of TTR variants, age at onset, pene‐
Email: fifigs1@yahoo.de
trance, and clinical presentation vary between geographical areas. Suggestive of a
ATTRm amyloidosis are a sensorimotor polyneuropathy, positive family history, auto‐
nomic dysfunction, cardiomyopathy, carpal tunnel syndrome, unexplained weight
loss, and resistance to immunotherapy. If only sensory A‐delta or C fibers are af‐
fected, small fiber neuropathy ensues. Diagnostic tests for small fiber neuropathy
include determination of intraepidermal nerve fiber density, laser‐evoked potentials,
heat‐ and cold‐detection thresholds, and measurement of the electrochemical skin
conductance. Therapy currently relies on liver transplantation and TTR‐stabilizers
(tafamidis, diflunisal).

KEYWORDS
amyloid, neuropathy, small fiber, tafamidis, transplantation, transthyretin

1 | I NTRO D U C TI O N Deposition of wild‐type (wt) TTR (ATTRwt) leads to senile, non‐he‐


reditary amyloidosis,1 with the onset typically >50 years of age. The
Amyloidosis is a multisystem, gain‐of‐toxic function disease due to occurrence of ATTRwt amyloidosis is regarded as an aging‐related
relentless deposition of aggregates of an abnormal protein in var‐ phenomenon, like Aβ deposition in the brain.1
ious tissues.1 More than 30 different abnormal proteins involved ATTRm amyloidosis is a rare, hereditary, autosomal‐domi‐
in the generation of amyloidosis have been reported thus far.1 nant, adult‐onset, multisystem disease (OMIM #105210) due to
Amyloidogenic proteins may have a genetic origin or a non‐genetic deposition of TTR‐amyloid in various organs or tissues. 3 Tissues
origin. One of the >30 proteins degenerating to amyloid is transthyre‐ predominantly affected in ATTRm amyloidosis are the peripheral
tin (TTR). Extracellular deposition of mutated (variant) TTR (ATTRm) nerves, leading to ATTRm‐related polyneuropathy, the meninges
leads to hereditary TTR‐related amyloidosis (ATTRm amyloidosis). 2 (leptomeningeal amyloidosis) and cerebral arteries (amyloid angi‐
opathy), and the heart, leading to TTR‐related familial amyloid car‐
diomyopathy (TTR‐CA). 3 Other organs variably affected are the
Alll authors contributed equally to this study.

92 | © 2018 John Wiley & Sons A/S. wileyonlinelibrary.com/journal/ane Acta Neurol Scand. 2019;139:92–105.
Published by John Wiley & Sons Ltd
FINSTERER et al. | 93

eyes, the gastrointestinal tract, the kidneys, and the skin. ATTRm
3.2 | Clinical presentation
amyloidosis is resistant to immunotherapy. Though ATTRm‐re‐
lated polyneuropathy or TTR‐CA may occur without affection of ATTRm amyloidosis is most frequently dominated by ATTRm‐re‐
another tissue, a variable combination of organ affection (multi‐ lated polyneuropathy including autonomic involvement. In most
organ involvement) is the most frequent phenotype. This review patients, cardiac (TTR‐CA), gastrointestinal, central nervous sys‐
aims at summarizing and discussing previous and recent findings tem, renal, connective tissue, or ocular involvement additionally
concerning the clinical presentation, genetic background, patho‐ develop (Table 1).6 Only rarely may ATTRm amyloidosis present
genesis, diagnosis, treatment, and outcome of ATTRm‐related exclusively as rapidly progressive polyneuropathy without involve‐
polyneuropathy. ment of other organs.7 Generally, the phenotype can be extremely
heterogeneous between geographical areas and even among fam‐
ily members carrying the same mutation. 8 In non‐endemic areas,
2 | M E TH O DS the natural history of ATTRm amyloidosis is more severe than in
endemic areas. 3 Bilateral CTS is reported frequently many years
Data for this review were identified by searches of MEDLINE for before the onset of other symptoms of ATTRm amyloidosis. The
references of relevant articles. Search terms applied were “tran‐ phenotype is not only determined by genetic but also epigenetic
sthyretin,” “gene,” “TTR‐FAP,” or “mutation,” combined with “neu‐ and environmental factors. Generally, three stages of ATTRm
ropathy,” “hereditary,” “autonomic,” “small fiber neuropathy,” and amyloidosis are differentiated: In stage‐I, the patient is ambula‐
“polyneuropathy.” Results of the search were screened for poten‐ tory with one cane; in stage‐II, the patient is ambulatory with two
tially relevant studies by application of inclusion and exclusion canes; and in stage‐III, the patient is confined to a wheelchair or is
criteria for the full texts of the relevant studies. Included were bedridden.9
randomized controlled trials (RCTs), observational studies with
controls, case series, and case reports. Only original articles about
3.2.1 | ATTRm‐related polyneuropathy
humans, and published between 1966 and 2018 were included.
Reviews, editorials, and letters were not considered. Reference ATTRm‐related polyneuropathy is a hallmark of ATTRm amyloidosis
lists of retrieved studies were checked for reports of additional in the early‐onset and late‐onset type and presents as progressive
studies. Websites checked for additional, particularly genetic in‐ (distal to proximal), symmetric, axonal, sensory, autonomic, or motor
formation and for assessing the pathogenicity of TTR mutations neuropathy.4
were the following:
Neuromuscular disease center, Washington University, St. Louis, Motor and sensory fibers
https://neuromuscular.wustl.edu/nother/amyloid.htm#transthyretin. Large non‐myelinated as well as small myelinated fibers are affected.
Genetics Home Reference. Transthyretin amyloidosis. https:// Small fiber neuropathy (SFN) predominates and mainly affects Aδ
ghr.nlm.nih.gov/condition/transthyretin-amyloidosis. and C fibers of sensory nerves.3 TTR‐FAP typically starts by the loss
of temperature and pain sensations in the feet associated with au‐
tonomic dysfunction, whereas proprioception and light touch sen‐
3 | R E S U LT S
sation are initially preserved.10 Burning feet over years due to SFN
is often the initial manifestation. After years, sensory disturbances
3.1 | Classification
spread from the foot soles to the insteps, ankles, lower legs, knees,
ATTRm amyloidosis may be classified as endemic/non‐endemic, and thighs in a length‐dependent manner. Light touch becomes ad‐
as hereditary/non‐hereditary, early‐onset/late‐onset, or as mono‐ ditionally impaired in the distal lower limbs but remains dissociated
system/multisystem (focal/generalized) disease. ATTRm amyloido‐ in the thighs.9 With further progression, motor deficits develop in
sis can be endemic (localized, traceable family history, early onset) the feet and progress in a proximal direction also in a length‐depend‐
or non‐endemic (scattered, frequently negative family history, ent manner.9 In the upper limbs, glove‐type sensory disturbances
4
late onset). Concerning the terminology, there is some confusion may develop.10 Rarely, also the cranial nerves (eg, hypoglossus)
about the optimal designation of the various entities. Some authors may be affected leading to tongue atrophy.11 ATTRm‐related poly‐
use ATTR‐FA synonymously with ATTRm‐related polyneuropathy. neuropathy is the most disabling among the late‐onset hereditary
Other authors use ATTRm synonymously with ATTRm‐related pol‐ neuropathies.3 With recognition of ATTRm‐related polyneuropathy
yneuropathy. In the present review, we use the term ATTRm amy‐ worldwide, classifications according to regional phenotypic variabil‐
loidosis for hereditary TTR‐amyloidosis. If patients with ATTRm ity12 become irrelevant.
amyloidosis manifest with polyneuropathy or cardiomyopathy,
we additionally use the terms ATTRm‐related polyneuropathy Autonomic dysfunction
and TTR‐CA. The term ATTRm is reserved for describing the vari‐ Autonomic functions can be concomitantly impaired from onset of
ant TTR protein, as recommended by the International Society of ATTRm‐related polyneuropathy resulting in cardiocirculatory, geni‐
Amyloidosis. 5 tourinary, or gastrointestinal dysfunction, such as gastroparesis with
94 | FINSTERER et al.

TA B L E 1 Genotype‐phenotype correlation of TTR mutations (modified according to Sekijima 2018)

Mutation PNP CTS SFN CI CNS OI GI Other Reference


4
Ala25Ser y n n n n n n n
4
Val30Leu y n n y n n n n
4
Phe33Val y n n n n n n n
4
Asp38Ala y n n y n n n n
4
Glu42Glya y n n n n n n n
4
Phe44Ser y n n n n n n n
4
Gly47Arg y n n n n n n n
4
Gly47Val y n n n n n n n
4
Thr49Ile y n n n n n n n
4
Thr49Ala y n n n n n n n
4
Ser50Arg y n n y n n n n
4
Glu54Lys y n n n n n n n
4
Leu55Pro y n n n n n n n
4
Glu61Lys y n n n n n n n
4
Val71Ala y n n n n n n n
4
Ser77Tyr y n n n n n n n
4
Ala97Gly y n n n n n n n
4
Ala109Ser y n n n n n n n
4
Val28Ser y n n n n n n n
4
Val28Met y n n n n n n n
4
Ala36Pro y n n n n n n n
4
Ile84Asn y n n n n n n n
4
His88Arg y n n n n n n n
4
Ala120Ser y y n n n n n n
4
Leu58Arg n y n n n n n n
4
Tyr69Ile n y n n n n n n
4
Ile107Val n y n n n n n n
4
Tyr 114His n y n n n n n n
4
Ala120Thr n y n n n n n n
1
Leu58His n y n n n n n n
1
Ile84Ser n y n n n n n n
4
Asp18Glu n n n y n n n n
4
Ala36Asp n n n y n n n n
4
Ala45Asp n n n y n n n n
4
Ser50Ile n n n y n n n n
4
Thr59Arg n n n y n n n n
4,30
Thr60Ala y n n y n n n n
4
Glu89Lys n n n y n n n n
4
Gln92Lys n n n y n n n n
4
Val94Gly n n n y n n n n
4
Val122Ile n n n y n n n n
4
Glu89Gln n n n y n n n n
4
Pro24Ser n n n y n n n n
4
Ala25Thr n n n n y n n n
4
Gly53Glu n n n n y n n n

(Continues)
FINSTERER et al. | 95

TA B L E 1 (Continued)

Mutation PNP CTS SFN CI CNS OI GI Other Reference


4
Tyr114Cys n n n n y n n n
4
Asp18Gly n n n n y n n n
4
Tyr69His n n n n y n n n
92
Ala97Ser y n n y n y n n

CI, cardiac involvement; CNS, central nervous system involvement; CTS, carpal tunnel syndrome; GI, gastrointestinal involvement; OI, ocular involve‐
ment; PNP, polyneuropathy; SFN, small fiber neuropathy.

nausea, followed by vomiting, pos‐tprandial diarrhea, and constipa‐


tion, cardiac conduction defects, orthostatic hypotension, dysuria,
urinary retention with recurrent urinary infections, sweating abnor‐
malities (sudomotor dysfunction), and sexual dysfunctions (eg, erec‐
tile dysfunction).9

3.2.2 | Cardiac involvement in ATTRm amyloidosis


(TTR‐CA)
TTR‐CA develops in about half of the patients with ATTRm amyloi‐
dosis. 8 TTR‐CA is already present at the time of the first positive
biopsy in 51% of the patients.13 TTR‐CA most frequently manifests
clinically as heart failure with normal systolic function, conduction
blocks, ventricular arrhythmias, or sudden cardiac death (SCD).14
Electrocardiographic (ECG) abnormalities typically recorded in
TTR‐CA patients are low voltage ECG, and QS‐pattern in right
precordial leads (suggesting myocardial infarction), but all other
ECG abnormalities may also variably occur, including atrial fibril‐
lation.15 Typically for TTR‐CA is that atrial fibrillation is associated
F I G U R E 1 Long‐axis 4C‐10 min showing diffuse subendocardial
without enlargement of the left atrium.15 SCD may occur in case of
enhancement in hypertrophied ventricles and atria with zebra
involvement of the cardiac conduction system. Echocardiography
stripe appearance [reprint from Kharadi et al Ann Indian Acad Sci
and cardiac MRI typically show myocardial thickening, including 2016, with permission]
the interatrial septum.1 On cardiac MRI, subendocardial late gado‐
linium enhancement (LGE), another characteristic feature can be
found (Figure 1).1 In a study of 263 patients with TTR‐CA, cardiac
MRI revealed asymmetric hypertrophy associated with hyper‐ 3.2.3 | Central nervous system (CNS) ATTRm
trophic cardiomyopathy as the commonest pattern of ventricular amyloidosis
remodeling in ATTR‐CA.16 LGE imaging was typical in all patients CNS manifestations of ATTRm amyloidosis include cerebral amy‐
with TTR‐CA.16 TTR‐CA is most frequently associated with the loid angiopathy and leptomeningeal amyloidosis. Amyloid may be
mutation Val122Ile.1 Since 3% of Afro‐Americans are heterozy‐ also deposited in veins of the subarachnoid space.4 Amyloid depo‐
gous for this mutation, TTR‐CA occurs frequently in this ethnic sition in cerebral amyloid angiopathy can be particularly found in
group. Other TTR mutations associated with TTR‐CA are Ser50Ile, the media and adventitia of medium‐ and small‐sized arteries, ar‐
Leu121Met, Asp18Glu, Ala36Asp, Ala45Asp, Thr59Arg, Thr60Ala, terioles, and veins of the cortex and the leptomeninges.1 Cerebral
Glu89Lys, Gln92Lys, Val94Gly, Glu89Gln, and Pro24Ser (Table 1).1 amyloid angiopathy may lead to ischemic stroke, intracerebral
It is well established that TTR‐CA is associated with an increased hemorrhage, focal subarachnoid bleeding resulting in hydro‐
mortality.13 TTR‐CA is the major cause of death in patients with cephalus, headache, spastic paraparesis, ataxia, seizures, amyloid
ATTRm amyloidosis in non‐endemic areas.17 The extracellular vol‐ spells, psychosis, or progressive dementia similar to Aß‐associated
ume fraction (ECV) correlated with the amyloid burden and was CAA.4,12 Leptomeningeal amyloidosis is infrequent in association
an independent prognostic factor for survival in a cohort of 263 with the mutation Val30Met.1 More frequently, leptomeningeal
16
patients with TTR‐CA. An early marker of cardiac involvement in amyloidosis is associated with the mutations Ala25Thr, Gly53Glu,
ATTRm amyloidosis and prognostic factor of poor outcome is atrial Tyr114Cys, Asp18Gly, or Tyr69His (Table 1).4,12 Leptomeningeal
affection.18 amyloidosis can be even a serious complication after liver
96 | FINSTERER et al.

transplantation (LTX) since the choroid plexus continues to pro‐ at onset depends on the genotype and the gender of the mutation
19
duce ATTRm. carrier.

3.2.4 | Ocular manifestations of ATTRm amyloidosis 3.3.1 | Early‐onset phenotype


Ocular involvement manifests as reduced visual acuity, dry eyes Early‐onset ATTRm amyloidosis starts between late 20 s and the
(sicca syndrome with an abnormal Schirmer test), vitreous opacity, early 40 s and is characterized by progressive, axonal, length‐de‐
pupillary abnormalities (dyscoria (abnormal pupillary shape), scal‐ pendent, sensorimotor polyneuropathy with initially predominant
loped pupils), secondary open‐angle glaucoma, abnormal conjunc‐ loss of small fiber function (nociception, thermal sensation (sensory
tival vessels, keratoconjunctivitis, abnormal tear break‐up time, dissociation)), a positive family history, severe autonomic dysfunction
deposition of amyloid in the lens, or retinal amyloid angiopathy (orthostatic hypotension, impotence, neurogenic bladder, disturbed
(tortuous vessels). 20 The prevalence of all ocular manifestations of bowel contractions), gastrointestinal involvement, and cardiac con‐
ATTRm amyloidosis increases with disease duration. 21 duction defects requiring pacemaker implantation. 26-30 There may
be myocardial thickening and arrhythmias. Vitreous opacities and
bilateral CTS can be also a feature of the early‐onset phenotype. In
3.2.5 | Gastrointestinal involvement
patients carrying the mutations Leu58His, Ile84Ser, and Tyr114His,
Gastrointestinal involvement may manifest as nausea, early satiety, neuropathy starts in the upper extremities as CTS.1 The early‐onset
recurrent vomiting due to gastroparesis, severe, postprandial, wa‐ phenotype is additionally characterized by high penetrance.
tery diarrhea, or severe constipation leading to malnutrition and un‐
intentional weight loss. In more severe cases, ileus may develop. 22
3.3.2 | Late‐onset phenotype
Late‐onset ATTRm amyloidosis starts after age 50 years and mani‐
3.2.6 | Renal involvement
fests initially with progressive, axonal sensorimotor symptoms in
Renal involvement may manifest as nephrotic syndrome or progres‐ the distal lower extremities.4 Both large and small fiber functions
sive renal failure. Renal failure occurs in one‐third of the Portuguese are already impaired initially, and there is rather loss of all sensory
23
patients. Renal involvement is rare in Japanese patients. A compli‐ modalities than sensory dissociation.4 Autonomic involvement is
cation of renal involvement could be renal anemia due to reduced rare but cardiac involvement is frequent. Cardiomyopathy may even
production of erythropoietin. dominate the phenotype.12 Gastrointestinal manifestations are fre‐
quent. Some patients may develop bilateral CTS. There is a marked
male preponderance.4 The late‐onset type is characterized by low
3.2.7 | Involvement of other structures
penetrance and often has a negative family history. Carriers of the
Non‐specific symptoms in ATTRm amyloidosis may be hoarseness, Val30Met in non‐endemic areas and in the endemic focus in Sweden
coldness, Charcot’s joints, or hyperthyroxinemia. Deposition of amy‐ usually present with the late‐onset phenotype.1 Phenotypes at
loid in the transversal carpi ligament may lead to compression neu‐ onset in Japanese patients differ significantly between patients from
ropathy of the median nerve (CTS). endemic and non‐endemic areas.

3.3 | Age and manifestations at onset 3.4 | ATTRwt amyloidosis


Onset of ATTRm amyloidosis is between age 10 and age 90 years Typically, ATTRwt amyloidosis manifests with cardiac abnormali‐
in endemic and non‐endemic areas.8 However, in endemic areas, ties, such as systolic dysfunction, heart failure, atrial fibrillation, or
early onset (age < 50 years; Portugal) or late onset (age > 50 years; ventricular arrhythmias, occasionally requiring implantation of an
4
Sweden), or both (Japan) can be delineated. Mean age at onset implantable cardioverter defibrillator (ICD). Another typical clinical
in Portugal is 33.5 years.4 On the contrary, mean age at onset in manifestation of ATTRwt amyloidosis is CTS, which may occur as the
Sweden is in the 50 s or 60 s.4 Onset in Japan is bimodal, with a peak presenting manifestation of ATTRwt amyloidosis.31 Rarely, ATTRwt
4
between ages 30 and 40 years and a second peak at age > 60 years. amyloidosis may manifest with renal dysfunction. In a Japanese
Early and late onset can be also found in cohorts from the United study of 51 patients with ATTRwt amyloidosis, 76% presented with
States, Switzerland, and Germany.12 In a study of 206 genetically heart failure, 59% with conduction defects, 49% with renal insuffi‐
confirmed patients with ATTRm‐related polyneuropathy from the ciency, 45% with CTS, and 22% with spinal canal stenosis.32
24
United States, (Minnesota) mean age at onset was 63.3 years. In
a Turkish study of 17 patients with ATTRm amyloidosis, mean age
3.5 | Epidemiology and gender
at onset was 40.4 years and the initial manifestation was feet pares‐
thesias in 15 patients. 25 Interestingly, in two patients, ATTRm amy‐ Previously, ATTRm‐related polyneuropathy was regarded to be a
loidosis started with dysarthria and hemiparesis. 25 Generally, age rare disease. Today, it is well acknowledged that there are more
FINSTERER et al. | 97

patients with ATTRm amyloidosis worldwide than previously of TTR mutations are non‐amyloidogenic.4 Recently, it has been
thought.1 ATTRm amyloidosis is currently regarded as the most shown that polymorphisms in various different genes (eg, APCS, AR,
common form of hereditary amyloidosis.1 In Japan, an estimated RBP4) may modify the phenotype since age at onset of ATTRm amy‐
prevalence of 0.87‐1.1 per 1 000 000 has been reported. 33 The loidosis was associated with the presence of polymorphisms in these
global prevalence is 8000‐10000 patients. The estimated inci‐ genes.35
dence is 1:100 000. ATTRwt amyloidosis is also regarded as a fre‐
quent disorder, since autopsy studies of patients >80 years have
3.6.3 | TTR amyloid fibril formation
shown that 10% of them had ATTRwt depositions.1 Of all ATTRm
amyloidosis cases, 72% are male, and of the ATTRwt amyloidosis TTR mutations cause instability of the TTR tetramer. The generation
cases, 85%‐90% are male. Thus, there is a striking male gender of amyloid from normally folded TTR tetramer starts with the dis‐
preponderance.1 sociation of the TTR tetramer into TTR monomers. Then, folded TTR
monomers are transformed into misfolded monomers inducing the
generation of oligomers by aggregation of variant TTR monomers and
3.6 | Etiology and pathogenesis
lastly of amyloid fibrils. TTR mutations destabilize the native quater‐
nary and tertiary structure of TTR, thereby inducing conformational
3.6.1 | Wild‐type TTR
changes leading to dissociation of tetramers and partial unfolding of
TTR is a 55 kDa homo‐tetrameric protein composed of 127‐resi‐ resultant monomers.11 TTR mutations also decrease the thermody‐
due β‐rich subunits.34 TTR is synthesized in the liver (the main namic stability of TTR, thus favoring the shift to misfolded monomer
source of serum TTR), choroid plexus, the retina, and the pancreas. species and kinetic instability. Amyloid formation is regarded as a nu‐
Interestingly, the prevalence of diabetes is not increased in ATTRm cleation‐dependent process, which means that fibril growth requires
amyloidosis. From these organs, it is secreted into blood, cerebro‐ the formation of a high‐energy quaternary oligomeric structure be‐
12
spinal fluid (CSF), and the eyes. The main function of TTR is the fore addition of monomers becomes energetically favorable. These
transport of thyroxin (T4) and of retinol‐binding protein/vitamin‐A destabilizing effects also decrease the secretion of TTR from cells. A
complex (holoRBP).34 Though the TTR tetramer contains two T4 drug known to accelerate TTR amyloidogenesis is heparin.36
34
binding sites, <1% of them are occupied by T4 molecules. This is
because the affinity of T4 to thyroid‐binding globulin is higher, and
3.6.4 | Toxicity of ATTR
the concentration of T4 in the serum is lower compared to that of
TTR. Thus, only 50% of the available TTR is bound to holoRBP. ATTR exhibits its toxic effect via different pathways. In ATTRm amy‐
loidosis, amyloid fibrils cause tissue damage by compression, obstruc‐
tion, or disturbance of local blood circulation.1 Deposition of amyloid
3.6.2 | Mutated TTR
fibrils causes CTS, vitreous opacity, or glaucoma. However, there are
ATTRm amyloidosis is due to mutations in the TTR gene, with in‐ also indications that mature TTR amyloid fibrils are not tissue toxic
complete penetrance.1 More than 130 different mutations in the contrary to low molecular weight monomers. Today, it is assumed that
TTR gene have been thus far detected (Table 1), and considerable oligomers and protofibrils are the actual toxic agent in ATTRm amy‐
genotype‐phenotype relations have been established.12 Most of loidosis and ATTRwt amyloidosis. Additionally, TTR oligomers may
the mutations are point mutations.10 In single cases, compound induce influx of calcium, which itself can be cytotoxic.37 TTR‐induced
heterozygous mutations and microdeletions have been reported. toxicity may be also mediated by receptor for advanced glycation end
Currently, two different nomenclature systems for TTR gene mu‐ products (RAGE).38,39 Activation of RAGE leads to endoplasmatic
tations are applied. According to the old system, which is used for reticulum stress, activation of extracellular signal‐regulated kinases
this review, amino acid changes are located at positions 20 amino (ERK1/2), involved in meiosis, mitosis, and post‐mitotic functions,
acids lower than in the new system. Thus, mutations such as V30M, and caspase‐dependent apoptosis.38,39 Additionally, oxidized forms
T60A, Glu89Lys are now referred as V50M, T80A, and Glu109Lys, of TTR can be cytotoxic to human cardiomyocytes in vitro, suggest‐
respectively. The most common TTR mutation is the substitution ing that age‐related oxidative modifications of TTR may contribute to
Val30Met (c.148G>A).1,12 This mutation is predominantly associated initiation of amyloid formation in late‐onset ATTRwt amyloidosis.40
12
with ATTRm‐related polyneuropathy and responsible for a large
endemic focus in Portugal (Povoa do Varazim, Porto) where ATTRm
3.7 | Diagnosis
amyloidosis due to the Val30Met variant has been first described by
Andrade C in 1951 (“disorder of the small feet”).1 There are strong
3.7.1 | General considerations
indications that the mutation drifted by migration of Portuguese
fisherman to Japan and the United States. In a study of 206 patients The diagnosis ATTRm amyloidosis is established upon the individ‐
with ATTRm amyloidosis from the United States, the most frequent ual and family history, clinical neurologic examination, and instru‐
mutation was the substitution Thr60Ala. 24 The most frequent muta‐ mental investigations, particularly by proof of amyloid deposition
tions causing TTR‐CA are Val122Ile and Ile68Leu.10 A small number on biopsy specimens and identification of a disease‐causing TTR
98 | FINSTERER et al.

FIGURE 2 Criteria for diagnosing


TTR‐FA

mutation.1 ATTRm amyloidosis should be suspected in the pres‐ or neck vein distension. The severity of polyneuropathy can be as‐
ence of a progressive, axonal, length‐dependent polyneuropathy, sessed by application of various polyneuropathy scores. These in‐
initially affecting temperature and pain sensation in association clude the neuropathy impairment score (NIS), the NIS‐lower limb
with one or more of the following manifestations: positive fam‐ (LL) score, the polyneuropathy disability (PND) score, the Norfolk
ily history for neuropathy, autonomic dysfunction, cardiomyopa‐ QOL‐DN total score, the Kumamoto score for assessing sensory dis‐
thy, gastrointestinal disorder, unexplained weight loss, CTS, renal turbances, or the modified Norris score. In case of muscle weakness,
3
impairment, or ocular impairment. These red flags are common quantification by grip strength measurement can be carried out. In a
in early‐onset ATTRm amyloidosis but uncommon in late‐onset retrospective, cross‐sectional study of 283 ATTRm amyloidosis pa‐
cases and thus less appropriate for diagnosing ATTRm amyloido‐ tients, NIS was correlated with functional scores of locomotion, with
sis in non‐endemic areas (Figure 2). 27,41 To establish the diagnosis the PND score, and with the disease stage.43 There was a negative
of ATTRm amyloidosis, two related symptoms and positive biopsy correlation between right‐hand grip strength and the PND score.43
3
findings for ATTRm depositions are required. The most common
combinations are ATTRm‐related polyneuropathy and autonomic
3.7.4 | Blood and CSF tests
dysfunction and ATTRm‐related polyneuropathy and TTR‐CA.42
TTR circulates in the blood as a soluble tetramer. Serum levels of
TTR are reduced in ATTRm amyloidosis. Reference limits for serum
3.7.2 | Individual and family history
TTR are 20‐40 mg/dL. Mutant TTR may be determined in the serum
Patients with ATTRm amyloidosis may report a number of different by application of mass‐spectroscopy after immunoprecipitation with
abnormalities related to polyneuropathy. These include paresthe‐ anti‐TTR antibodies and dissociation of the tetrameric structure into
sias, dysesthesias, allodynia, neuropathic pain, hyperalgesia, exer‐ proamyloidogenic monomers. Immunoprecipitation allows iden‐
cise intolerance, muscle weakness, unexplained weight loss, and tifying 90% of ATTRm variants.12 Serum TTR levels were lower at
muscle wasting. Ocular complaints may include visual field defects, the 1‐year and 2‐year follow‐up after starting TTR‐stabilizers in un‐
ocular pain, visual disturbances, or oversensitivity to light. In case of treated patients with ATTRwt amyloidosis compared to treated pa‐
autonomic involvement, patients may complain about urinary distur‐ tients.44 Small amounts of TTR monomers (0.28‐0.56 µg/mL) can be
bances, erectile dysfunction, fainting and syncopes, or orthostatic detected in the serum of patients with ATTRm amyloidosis.45 Since
intolerance. In case of gastrointestinal involvement, patients may TTR is also produced in the choroid plexus, TTR may be also found in
report diarrhea, or constipation, vomiting, or persisting nausea. If the CSF.9 Frequently, the CSF protein is elevated.
the heart is affected, leg edema, exercise intolerance, or exertional
dyspnea may be reported. In a study of 141 Japanese families with
3.7.5 | Electrophysiology, ultrasound, and MRI of
ATTRm amyloidosis, the family history was positive in 98% of the
large peripheral nerves
early‐onset cohorts and in 48% of the late‐onset cohorts. 27 In non‐
endemic areas, the family history is positive in only half of the cases. Electrophysiological techniques such as nerve conduction studies
and somato‐sensory‐evoked potentials (SSEPs) should be applied to
assess the degree of affection of large sensory and motor fibers in
3.7.3 | Clinical neurologic examination
the disease. Ultrasound of peripheral nerves may show enlargement
The neurologic examination may reveal signs of large and small fiber at entrapment sites and in proximal nerve segments, and segmental
dysfunction, reduced tendon reflexes, weakness, wasting, sensory structural alterations with change or even loss of the physiological
ataxic gait, orthostatic hypotension, leg edema, exertional dyspnea, fascicular structure.46 There may be also increased echogenicity
FINSTERER et al. | 99

within the interfascicular epineurium without substantial enlarge‐


3.7.8 | Biopsy
ment of the nerve.46 Injury of lower limb nerves can be detected
and quantified by high‐resolution magnetic resonance neurography Demonstration of amyloid deposition
(HR‐MRN).47 Application of HR‐MRN to 13 symptomatic patients Demonstration of amyloid on histopathology is a prerequisite for
with ATTRm amyloidosis and asymptomatic mutation carriers re‐ diagnosing ATTRm amyloidosis. This is particularly the case if the
vealed that the number of nerve lesion voxels upon histogram‐based family history is negative for ATTRm amyloidosis. Deposition of
normalization of nerve voxel signal intensities was higher in patients amyloid in affected tissues can be directly demonstrated by Congo
than in asymptomatic carriers or healthy controls.47 red staining of various biopsy materials. Amyloid depositions dis‐
play a characteristic apple‐green birefringence under polarized
light examination. Amyloid may be also detected upon biopsy of the
3.7.6 | Assessment of SFN
subcutaneous fat, sural nerve, the rectal mucosa, labial and other
SFN usually manifests clinically with somato‐sensory manifesta‐ salivary glands (sensitivity up to 92%), the kidneys, the endomyo‐
tions (neuropathic pain, paresthesias) or autonomic manifesta‐ cardium, or tenosynovial tissue obtained during CTS surgery. 51,52
tions.48 SFN often represents a diagnostic challenge. Standard However, organs clinically affected and not mentioned above can
electrophysiological evaluation is usually normal. Quantitative be also biopsied. Biopsy of the sural nerve is less sensitive because
sensory testing (QST) can be carried out to assess small fiber func‐ amyloid deposition is often patchy.12 A common histological finding
tion by determination of thresholds for cold, warm, pressure, or on nerve biopsy is loss of small neurons with/without neighbor‐
pain sensation. The most accurate tool for diagnosing SFN is the ing ATTRm deposition. 53 Additionally, Schwann cell atrophy and
skin biopsy. Of additional help can be pain‐related evoked poten‐ disruption of the blood‐nerve barrier can be found. 53 Negative bi‐
tials (PREP), laser‐evoked potentials (LEP), determination of heat‐ opsy findings do not rule out ATTRm amyloidosis. Immunostaining
and cold‐detection thresholds, the sympathetic skin response of amyloid helps to identify amyloid subtypes, including TTR,
(SSR), and measurement of the electrochemical skin conductance but is not capable to differentiate between ATTRm and ATTRwt.
(ESC) by means of the QSART and the SUDOSCAN. Application Immunohistochemistry and immune‐florescence proteomic analy‐
of the SUDOSCAN allows testing the autonomic innervation of sis are crucial for the correct specification of the amyloid sub‐
the sweat glands.49 In a study of 133 ATTRm amyloidosis patients, type or differentiation between ATTRm and ATTRwt. 2 If biopsy
ESC allowed to distinguish manifesting patients with ANS involve‐ findings are negative but ATTRm amyloidosis is still suspected, it
ment from patients without ANS involvement.47 In this study, the is recommended to perform sequencing of the TTR gene. A diag‐
SUDOSCAN had a sensitivity of 76’% and specificity of 85%.47 nostic challenge is sporadic cases with a negative family history in
Since SFN is often the initial manifestation, it is essential to diag‐ non‐endemic areas. 54-56 Genotypic and phenotypic heterogeneity
nose SFN properly and to start therapy in time. and the non‐specific nature of various symptoms often delay the
diagnosis. 57

3.7.7 | Autonomic testing


Skin biopsy
Autonomic testing is crucial for demonstrating autonomic involve‐ Skin biopsy is usually carried out to diagnose SFN. It is currently the
ment. One of the most frequently applied tests is measuring heart most reliable diagnostic tool to detect SFN.48 The most valuable
rate variability (HRV) in response to deep breathing (six breaths/ parameter in this respect is the intraepidermal nerve fiber density
min).50 In a study of 165 ATTRm amyloidosis patients carrying the (IENFD).48 In a study of 32 patients with ATTRm amyloidosis, IENFD
mutation Val30Met, HRV was reduced in 47%.50 HRV during physi‐ was reduced in symptomatic Val30Met and non‐Val30Met muta‐
ological breathing as assessed by Fourier analysis of the ECG on tion carriers. 58 The IENFD can be also reduced in asymptomatic
the Holter signal allows to detect sympathetic or parasympathetic mutation carriers. 58 IENFD is negatively correlated with disease
over‐ or underactivity. Another test frequently applied is the tilt duration, and positively with the Kumamoto score, heat/pain detec‐
test, which is useful to assess the sympathetic innervation of the tion thresholds, and the sensory nerve action potential (SNAP). 58
myocardium or the precapillary sphincters and to assess orthostatic Sudomotor nerves, stained with protein gene product (PGP9.5)
dysfunction. Autonomic involvement can be also assessed by inves‐ or vasoactive intestinal peptide (VIP), are usually also reduced in
tigations of urinary‐genital functions. Sympathetic innervation of TTR‐SFN. 59
the sweat glands can be assessed by means of the SUDOSCAN and
the NEUROPAD. Autonomic denervation of the myocardium can
3.7.9 | Genetic tests
be best assessed by reduced tracer uptake on the meta‐iodo‐ben‐
zyl‐guanidine (MIBG)‐SPECT, which can also be used to distinguish Documentation of a pathogenic TTR mutation is crucial for di‐
Parkinsonian syndromes. Quantification of sweat glands in skin agnosing ATTRm amyloidosis. A targeted approach is justified if
biopsy allows assessing the sympathetic innervation of the sweat the TTR mutation is already known in another family member. If
glands. Distal vasomotricity can be explored by application of the ATTRm amyloidosis is suspected despite a negative family history,
laser Doppler flowmetry (LDF). sequencing of the entire TTR gene is recommended. The most
100 | FINSTERER et al.

frequent mutations are heterozygous point mutations (Table 1).


3.7.11 | ATTRwt amyloidosis
Occasionally, compound heterozygous point mutations and mi‐
crodeletions have been reported.9 The mutation Val30Met is the To diagnose ATTRwt amyloidosis, it is necessary to prove deposi‐
most common variant in Europe, except in Bulgaria (Glu89Gln) and tion of ATTRwt on a biopsy specimen and to rule out a mutation in
the UK (Thr 60Ala). 54 In sporadic cases, the entire coding regions the TTR gene. ATTRwt can be detected by immunocytochemical
(ie, all four exons) should be sequenced. 42
In a recent Brazilian and proteomic analysis.1 Though ATTRwt amyloidosis is most fre‐
study of 123 patients with ATTRm amyloidosis, 90.6% carried the quently diagnosed upon endomyocardial biopsy, this invasive tech‐
mutation Val30Met and 7 carried non‐Val30Met mutations, such nique is not appealing.1 Though abnormal uptake of 99mrTcPYP
60
as Aps38Tyr, Ile107Val, Val71Ala, and Val121Ile. on myocardial SPECT is highly suggestive of myocardial amyloi‐
dosis and uptake of 99mTcDPD suggestive for ATTRm/ATTRwt
deposition, histopathological confirmation of the suspicion is nec‐
3.7.10 | Other instrumental investigations
essary. If ATTRwt deposition is also found in tenosynovial tissue,
Cardiac involvement confirmation of ATTRwt in other tissues is necessary, since local‐
To assess cardiac involvement, ECG, long‐term ECG, echocardiogra‐ ized deposition of ATTRwt is common in elderly subjects.1
phy, cardiac MRI with contrast medium are useful diagnostic tools.
99 m Technetium‐pyrophosphate (99mTcPYP) SPECT has become
3.7.12 | Differential diagnoses
the most relevant non‐invasive imaging tool for diagnosing cardiac
amyloidosis.61 A possibility to detect ATTRm or ATTRwt is by applica‐ All types of axonal hereditary neuropathies should be consid‐
tion of the 99mTcDPD SPECT, which highly sensitive for TTR‐CA.62 ered as differential diagnoses of ATTRm‐related polyneuropathy.
Autonomic denervation of the myocardium can be best assessed However, the most common misdiagnosis of ATTRm‐related poly‐
by application of the MIBG‐SPECT. MIBG scintigraphy proved to neuropathy is chronic inflammatory demyelinating polyneuropathy
be a strong and independent prognostic marker of poor outcome (CIDP). 66 Thus, if patients with CIDP do not respond to immuno‐
63
in ATTRm amyloidosis. If the MIBG‐SPECT is inconclusive and globulins and if axonal lesions predominate, ATTRm‐related poly‐
the heart the only clinically affected organ, endomyocardial biopsy neuropathy should be suspected. Even autonomic dysfunction may
should be considered. occur in CIDP patients. Other differential diagnoses are lumbar spi‐
nal canal stenosis, idiopathic axonal polyneuropathy, paraneoplas‐
Cerebral involvement tic peripheral neuropathy, toxic neuropathy, vasculitic neuropathy,
To assess CNS involvement, MRI with contrast medium of the brain and motor neuron disease. 67-69 Differential diagnoses of SFN that
or the spinal cord, MRI‐angiography, CT angiography, or biopsy of need to be considered include diabetes, dysimmune syndromes
the meninges may be helpful. TTR may be reduced in the CSF. (Sjögren syndrome, sarcoidosis, monoclonal gammopathy), or ge‐
netic conditions (sodium channel disease, Fabry’s disease, neu‐
Ocular involvement ropathy due to mutations in the prion protein). Hereditary amyloid
Assessment of ocular involvement requires a complete ophthalmo‐ neuropathies other than ATTRm‐related polyneuropathy include
logic workup, including Schirmer test, disk examination, optic coher‐ neuropathy due to mutations in the apolipoprotein A1, gelsolin, or
ence tomography (OCT), and retinal angiography. Recently, it has beta2‐microglobulin genes, respectively.42 Amyloid proteins other
been shown that the corneal nerve fiber length (CNFL) is shortened than ATTRm associated with cardiomyopathy include amyloids
in ATTRm amyloidosis patients on in vivo confocal corneal micros‐ from immunoglobulin light chains (paraprotein, myeloma), serum
copy (IVCM). 64
In a study of 15 ATTRm amyloidosis patients, the amyloid A, atrial natriuretic factor, or from apolipoprotein A1. 2
64
correlation between CNFL and the severity of SFN was negative. For the clinician, it is important to have a high index of suspicion,
There was a positive correlation between CNFL and the SNAP and when red flags are present in a patient with “idiopathic” polyneurop‐
between CNFL and the IENFD.64 athy. Rapid progression, ambulatory loss early on, and autonomic dis‐
turbances may serve as red flags for ATTRm‐related polyneuropathy.
Gastrointestinal involvement However, lack of amyloid in nerve biopsies of patients with ATTRm‐re‐
Gastrointestinal affection requires gastroscopy and colonoscopy, lated polyneuropathy is possible, while other diagnostic pitfalls include
and mucosal biopsy. unspecific electrophysiological and coincident laboratory findings such
as pathologic glucose tolerance or a monoclonal gammopathy.57
General investigations
To screen patients for suspected ATTRm amyloidosis non‐inva‐
3.8 | Treatment
sively, abdominal fat ultrasonography (AFUS) has been recently pro‐
posed.65 In a study of 19 patients with ATTRm amyloidosis, AFUS
3.8.1 | Liver transplantation (LTX)
showed increased mean echogenicity and loss of normal structure of
fat tissue layers.65 Results on AFUS were positively correlated with Until 1990, ATTRm amyloidosis used to be an incurable disease.
the amount of ATTRm deposition in fat tissues.65 In 1990, LTX was introduced as a causative therapy of ATTRm
FINSTERER et al. | 101

amyloidosis. LTX can have a beneficial effect, at least on ATTRm‐ as assessed by the Norfolk Quality of Life scale than untreated pa‐
related polyneuropathy, if carried out early in the disease course tients.80 Tafamidis is currently approved for ATTRm‐related poly‐
1
(<5 years disease duration) and in young patients. LTX results in neuropathy stage‐1.11 TTR‐stabilizers are the first‐line option for
slowing of progression of ATTRm‐related polyneuropathy and of au‐ early‐onset ATTRm‐related polyneuropathy in Europe.4 Another
70
tonomic involvement in some patients. LTX removes the primary TTR‐stabilizer currently under investigation, but not yet approved,
source of ATTRm production, eliminates 95% of ATTRm, slows or is tolcapone.
stops progression of the disease, and improves the disease in single TTR gene silencing drugs are subject to phase‐3 clinical trials.3
4
cases. In a study of 6 patients with ATTRm‐related polyneuropathy, Other compounds currently under investigation include the small
the amount of deposited ATTRm regressed during an observational interfering (si) RNA patisiran, which silences the mRNA, and the anti‐
71
period of 10 years after LTX. In other cases, progression of the dis‐ sense oligonucleotide (AON) inotersen. In these phase‐3 trials, both
ease could be stopped depending on the type of mutation and the inotersen and patisiran have met their primary and secondary end‐
side effects of immunosuppressive drugs. LTX replaces the mutated points for the Neuropathy Impairment Score (NIS) and for quality of
TTR gene by the wt‐gene in the liver, why serum levels of ATTRm life scores. Both are currently moving forward with FDA approval and
1
rapidly decrease after LTX. Accordingly, long‐term follow‐up biop‐ articles about these studies are in press in the New England Journal
sies of patients having undergone LTX showed regression of amyloid of Medicine. These results further emphasize that timely recognition
deposition.72 LTX usually has no benefit on TTR‐CA, leptomenin‐ and accurate diagnosis are crucial as effective treatment is available.
geal involvement, or on ATTRwt amyloidosis.1 New, long‐term re‐ As ATTRm amyloidosis is a progressive disease, it may cause irrevers‐
sults allow selection of best candidates for LTX based on phenotype ible tissue damage if untreated or treated too late.66,75,81
3
and cardiac involvement. LTX is not indicated in late stages, when
there is malabsorption, ileus, or diarrhea, requiring parenteral nutri‐
3.8.3 | Symptomatic measures
tion or if there is TTR‐CA with ventricular arrhythmias. According
to Japanese data, LTX is particularly effective in early‐onset ATTRm In the era prior to LTX and TTR‐stabilizers, treatment of ATTRm
amyloidosis but not in late‐onset ATTRm amyloidosis.73 Interestingly, amyloidosis was restricted to symptomatic measures, such as treat‐
the outcome after LTX is better if living donor liver grafts are im‐ ment of neuropathic pain, implantation of a pacemaker, or parenteral
planted compared to implantation of grafts from deceased donors.74 nutrition. Today, treatment of ATTRm amyloidosis relies on three
The outcome of LTX may be further improved if LTX is combined in pillars: disease‐modifying targeted therapy to prevent further pro‐
tandem with TTR‐stabilizers. Transplantation of livers from patients duction of ATTRm (LTX, TTR‐stabilizers), symptomatic treatment of
with ATTRm amyloidosis to non‐ATTRm patients with liver failure nerve, cardiac, ocular, cerebral, gastrointestinal, and renal involve‐
can subsequently induce amyloidosis in some recipients.5 ment, and genetic counseling and supportive care. Treatment algo‐
LTX for ATTRm amyloidosis has several limitations: (a) Organ dam‐ rithms vary between countries.4 First‐line therapy in Japan is LTX,
age present already prior to LTX is hardly reversed. (b) Outcomes are while European countries adopted pharmacotherapeutic options as
mutation‐specific (patients carrying the Val30Met have a better out‐ first‐line treatment.4 In a recent Italian study, epigallocatechin‐3‐gal‐
come than non‐Val30Met patients). (c) Amyloid deposition continues late failed to improve survival of patients with TTR‐CA.82
after LTX because ATTRwt deposits on existing amyloid. (d) ATTRm
deposition continues because of local production in the choroid
3.8.4 | Gene therapy
plexus or the retinal epithelium. (e) LTX carries the risk of surgery and
long‐term immunosuppression has side effects. (f) Many patients are Recently, the first investigational RNA interference therapeutic
4
not suitable candidates for LTX, and that LTX is not widely available. agent has been shown highly effective in improving multiple dif‐
ferent manifestations of ATTRm amyloidosis.83 In a randomized,
controlled, phase‐3 trial, 225 patients with ATTRm amyloidosis
3.8.2 | TTR‐ stabilizers
with polyneuropathy received 0.3 mg/kg patisiran intravenously
TTR‐tetramer‐stabilizers currently available include tafamidis and every three weeks over 18 months.83 The modified Neuropathy
76
diflunisal. Long‐term randomized controlled trials have shown Impairment Score+7 increased significantly over this period in the
that tetramer stabilizers slowed the progression of ATTRm‐related verum group.83 In a randomized, double‐blind, placebo‐controlled,
polyneuropathy and preserve quality of life in ATTRm‐related poly‐ phase‐3 trial of 112 patients with ATTRm amyloidosis, the antisense
neuropathy patients. Early treatment with tafamidis can delay pro‐ oligonucleotide inotersen (300 mg subcutaneously every week) re‐
gression of ATTRm‐related polyneuropathy over 5.5 years.77 Studies sulted in significant improvement of the clinical neurologic course
on the long‐term effect of tafamidis showed that 40% respond as and the quality of life in the included patient.84
78
reflected on the NIS‐LL score, that biochemical stabilization of
78
TTR can be achieved, that prevention of functional progression
3.9 | Genetic counseling
can be achieved in up to 50% of the patients,67 and that tafamidis
may not prevent the progression of the disease.79 In the THAOS Patients in whom ATTRm amyloidosis was diagnosed and at‐risk
survey, patients under tafamidis had significantly less deterioration family members require genetic counseling. Genetic counseling
102 | FINSTERER et al.

must be carried out by trained professionals.3 Regular monitoring of 4 | CO N C LU S I O N S


asymptomatic carriers is warranted to detect early manifestations of
ATTRm amyloidosis. ATTRm amyloidosis is a multifaceted, fascinating disorder, which still
has a number of surprises to provide to the clinician and the scientist.
Most importantly, ATTRm amyloidosis needs to be diagnosed early
3.10 | Prognosis and outcome
in the course to provide optimal treatment to improve outcome.
ATTRm amyloidosis is the most severe form among the hereditary Current and future studies should thus direct toward the evaluation
neuropathies.46 Without treatment, patients die from progressive and invention of new methods to recognize early manifestations at
organ dysfunction, particularly heart failure, infection, or cachexia onset. This issue not only refers to better characterization of clinical
8
approximately within 10 years of disease onset. Cardiac involve‐ manifestations at onset but also to instrumental investigations that
ment is the most important prognostic factor in patients with gener‐ can reliably confirm the presence of ATTRm or ATTRwt depositions
alized amyloidosis. 2 In a recent retrospective study of 120 patients in clinically affected or unaffected tissues. Currently available, mini‐
with TTR‐CA of whom 29 received tafamidis, application of TTR‐ mally invasive or non‐invasive methods for early diagnosis include
stabilizers was associated with decreased rate of orthotopic heart skin biopsy, IVCM, HR‐MRN, and AFUS. However, future perspec‐
transplantation and death.85 Another major factor influencing the tives should also include the invention of new techniques for early
survival rate of ATTRm amyloidosis patients is the TTR genotype. In diagnosing ATTRm amyloidosis to prolong the survival in this still
patients carrying the Val30Met variant, 5‐year and 10‐year survival progressive and lethal disease.
rates were 82% and 74%, respectively, compared with a 5‐year and
10‐year survival rate of 59% and 44% in non‐Val30Met carriers.86,87
AC K N OW L E D G M E N T S
In early‐onset ATTRm amyloidosis, prognosis in transplanted pa‐
tients is better than in non‐transplanted patients. This effect is no None.
longer present in patients with late‐onset ATTRm amyloidosis. It has
been also reported that the prognosis of females with early‐onset
C O N FL I C T O F I N T E R E S T S
ATTRm amyloidosis carrying the Val30Met mutation is much better
than in males carrying Val30Met with late‐onset ATTRm amyloido‐ There are no conflict of interests.
73
sis. In a study of 206 genetically confirmed patients with ATTRm
amyloidosis, mean survival after diagnosis was 56.8 months. 24 In
AU T H O R C O N T R I B U T I O N
this study, risk factors for death on multivariate analysis were age
at onset, presence of Thr60Ala or Val122Ile mutations, neuropathy, JF designed the study, involved in literature search, and discussed
24
and weight loss. The overall survival rate of patients undergoing and first drafted the manuscript. WL, RT, JW, SI, and WG involved in
LTX is >80% five years after surgery.86 Progression of cardiac amy‐ literature search and made critical review of the manuscript.
loid deposition is more pronounced in non‐Val30Met carriers than
in late‐onset Val30Met liver recipients.88 The mean survival period
ORCID
from the onset of heart failure is 75 months in ATTRwt amyloidosis.
A severe and rapid course with a survival of 6‐7 years after onset has Josef Finsterer http://orcid.org/0000-0003-2839-7305
been reported in patients with late‐onset ATTRm amyloidosis carry‐
ing the mutations Val30Met, Ile107Val, or Thr60Ala.43,89
REFERENCES

1. Sekijima Y. Transthyretin (ATTR) amyloidosis: clinical spectrum,


3.11 | Predictors of survival molecular pathogenesis and disease‐modifying treatments. J Neurol
Neurosurg Psychiatry. 2015;86:1036‐1043.
In patients with ATTRwt amyloidosis, low serum TTR levels were
2. Flodrova P, Flodr P, Pika T et al. Cardiac amyloidosis: from clinical
associated with short survival.44 In a 20‐year retrospective study suspicion to morphological diagnosis. Pathology. 2018; 50:261‐268.
of 1379 patients having undergone LTX, survival was reduced with 3. Adams D, Cauquil C, Labeyrie C. Familial amyloid polyneuropathy.
high body mass index, early‐onset, long disease duration before Curr Opin Neurol. 2017;30:481‐489.
4. Sekijima Y, Ueda M, Koike H, Misawa S, Ishii T, Ando Y. Diagnosis
LTX, and with Val30Met vs. non‐Val30Met mutation.75 In a mono‐
and management of transthyretin familial amyloid polyneuropa‐
centric cohort study of 215 ATTRm amyloidosis patients, predic‐
thy in Japan: red‐flag symptom clusters and treatment algorithm.
tors of death were PND score > II, orthostatic hypotension, NYHA Orphanet J Rare Dis. 2018;13(1):6.
stage > I, QRS duration >120 ms, and thickened interventricular 5. Sipe JD, Benson MD, Buxbaum JN et al. Amyloid fibril proteins
septum.90 Survival was reduced if there was cardiac dysautonomia and amyloidosis: chemical identification and clinical classifica‐
tion International Society of Amyloidosis 2016 Nomenclature
after LTX, or on MIBG‐SPECT and no response to atropine.91 In a
Guidelines. Amyloid. 2016;23:209‐213.
study of 116 patients with ATTRwt amyloidosis, reduced serum TTR 6. Rocha A, Beirão I, Pessegueiro H, Almeida R, Lobato L. End‐stage
values were associated with short survival.44 Low TTR serum levels renal failure due to transthyretin amyloidosis after liver transplanta‐
were an independent predictor of the outcome in these patients.44 tion: outcomes in 19 registry cases. Amyloid. 2017;24(supp1):85‐86.
FINSTERER et al. | 103

7. Schänzer A, Kimmich C, Röcken C, Haverkamp T, Weidner I, Acker 28. Ando Y, Nakamura M, Araki S. Transthyretin‐related familial amyloi‐
T. Krämer HH. A woman with a rare p.Glu74Gly transthyretin muta‐ dotic polyneuropathy. Arch Neurol. 2005;62:1057‐1062.
tion presenting exclusively with a rapidly progressive neuropathy: a 29. Ikeda S, Hanyu N, Hongo M et al. Hereditary generalized amyloido‐
case report. J Med Case Rep. 2014;4(8):403. sis with polyneuropathy. Clinicopathological study of 65 Japanese
8. Ando Y, Coelho T, Berk JL, et al. Guideline of transthyretin‐related patients. Brain. 1987;110:315‐337.
hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31. 30. Nakazato M, Shiomi K, Miyazato M, Matsukura S. Type I familial am‐
9. Martens B, De Pauw M, De Bleecker JL. Single‐centre experience yloidotic polyneuropathy in Japan. Intern Med. 1992;31:1335‐1338.
on transthyretin familial amyloid polyneuropathy: case series and 31. Koike H, Tanaka F, Hashimoto R, et al. Natural history of transthyre‐
literature review. Acta Neurol Belg. 2018; 118:179‐185. tin Val30Met familial amyloid polyneuropathy: analysis of late‐
10. SchmidtHH‐J.Transthyretin‐assoziiertefamiliäreAmyloidpolyneuropathie. onset cases from non‐endemic areas. J Neurol Neurosurg Psychiatry.
Diagnose und Therapie. Thieme Case Rep. 2014;6:1‐20. 2012;83:152‐158.
11. Goyal NA, Mozaffar T. Tongue atrophy and fasciculations in trans‐ 32. Sekijima Y, Yazaki M, Ueda M, Koike H, Yamada M, Ando Y. First na‐
thyretin familial amyloid neuropathy: An ALS mimicker. Neurol tionwide survey on systemic wild‐type ATTR amyloidosis in Japan.
Genet. 2015;1:e18. Amyloid. 2017;28:1‐3.
12. Sekijima Y, Yoshida K, Tokuda T, et al. Familial Transthyretin 33. Kato‐Motozaki Y, Ono K, Shima K, et al. Epidemiology of familial
Amyloidosis. In: Adam MP, Ardinger HH, Pagon RA, et al., eds. amyloid polyneuropathy in Japan: identification of a novel endemic
GeneReviews® [Internet]. Seattle, WA: University of Washington; focus. J Neurol Sci. 2008;270:133‐140.
1993-2018:553-559. 34. Blake CC, Geisow MJ, Swan I. Structure of human plasma prealbu‐
13. Klaassen S, Tromp J, Nienhuis H et al. Frequency of and prognostic min at 2–5 A resolution. A preliminary report on the polypeptide
significance of cardiac involvement at presentation in hereditary chain conformation, quaternary structure and thyroxine binding. J
transthyretin‐derived amyloidosis and the value of N‐terminal Mol Biol. 1974;88:1‐12.
pro‐B‐type natriuretic peptide. Am J Cardiol. 2018;121:107‐112. 35. Santos D, Coelho T, Alves‐Ferreira M, et al. Variants in RBP4 and
14. Delahaye N, Rouzet F, Sarda L et al. Impact of liver transplantation AR genes modulate age at onset in familial amyloid polyneuropathy
on cardiac autonomic denervation in familial amyloid polyneuropa‐ (FAP ATTRV30M). Eur J Hum Genet. 2016;24:756‐760.
thy. Medicine (Baltimore). 2006;85:229‐238. 36. Bourgault S, Solomon JP, Reixach N, et al. Sulfated glycosaminogly‐
15. Henein MY, Suhr OB, Arvidsson S et al. Reduced left atrial myocar‐ cans accelerate transthyretin amyloidogenesis by quaternary struc‐
dial deformation irrespective of cavity size: a potential cause for tural conversion. Biochemistry. 2011;50:1001‐1015.
atrial arrhythmia in hereditary transthyretin amyloidosis. Amyloid. 37. Sorgjerd K, Klingstedt T, Lindgren M, et al. Prefibrillar trans‐
2018;25:1‐8. thyretin oligomers and cold stored native tetrameric transthyre‐
16. Martinez‐Naharro A, Treibel TA, Abdel‐Gadir A et al. Magnetic tin are cytotoxic in cell culture. Biochem Biophys Res Commun.
resonance in transthyretin cardiac amyloidosis. J Am Coll Cardiol. 2008;377:1072‐1078.
2017;70:466‐477. 38. Sousa MM, Du Yan S, Fernandes R, et al. Familial amyloid polyneu‐
17. Ikeda S. Cardiac amyloidosis: heterogenous pathogenic back‐ ropathy: receptor for advanced glycation end products‐dependent
grounds. Intern Med. 2004;43:1107‐1114. triggering of neuronal inflammatory and apoptotic pathways. J
18. Di Bella G, Minutoli F. Atrial impairment in transthyretin cardiac am‐ Neurosci. 2001;21:7576‐7586.
yloidosis: an early marker of cardiac involvement and a prognostic 39. Monteiro FA, Sousa MM, Cardoso I, et al. Activation of ERK1/2
factor. Amyloid. 2018;9:1. MAP kinases in familial amyloidotic polyneuropathy. J Neurochem.
19. Maia LF, Magalhães R, Freitas J et al. CNS involvement in V30M 2006;97:151‐161.
transthyretin amyloidosis: clinical, neuropathological and biochem‐ 40. Zhao L, Buxbaum JN, Reixach N. Age‐related oxidative modifica‐
ical findings. J Neurol Neurosurg Psychiatry. 2015;86:159‐167. tions of transthyretin modulate its amyloidogenicity. Biochemistry.
20. Reynolds MM, Veverka KK, Gertz MA et al. Ocular manifes‐ 2013;52:1913‐1926.
tations of familial transthyretin amyloidosis. Am J Ophthalmol. 41. Samoes R, Taipa R, Valdrez K, et al. Amyloid detection in the
2017;183:156‐162. transverse carpal ligament of patients with hereditary ATTR
21. Beirão JM, Malheiro J, Lemos C, Beirão I, Costa P, Torres P. V30Mamyloidosis and carpal tunnel syndrome. Amyloid.
Ophthalmological manifestations in hereditary transthyretin (ATTR 2017;24:73‐77.
V30M) carriers: a review of 513 cases. Amyloid. 2015;22:117‐122. 42. Conceição I, González‐Duarte A, Obici L et al. "Red‐flag" symptom
22. Wixner J, Suhr OB, Anan I. Management of gastrointestinal com‐ clusters in transthyretin familial amyloid polyneuropathy. J Peripher
plications in hereditary transthyretin amyloidosis: a single‐cen‐ Nerv Syst. 2016;21:5‐9.
ter experience over 40 years. Expert Rev Gastroenterol Hepatol. 43. Mariani LL, Lozeron P, Théaudin M, et al. Genotype‐phenotype cor‐
2018;12:73‐81. relation and course of transthyretin familial amyloid polyneuropa‐
23. Lobato L, Rocha A. Transthyretin amyloidosis and the kidney. Clin J thies in France. Ann Neurol. 2015;78:901‐916.
Am Soc Nephrol. 2012;7:1337‐1346. 44. Hanson J, Arvanitis M, Koch CM et al. Use of serum transthyretin
24. Swiecicki PL, Zhen DB, Mauermann ML et al. Hereditary ATTR amy‐ as a prognostic indicator and predictor of outcome in cardiac amy‐
loidosis: a single‐institution experience with 266 patients. Amyloid. loid disease associated with wild‐type transthyretin. Circ Heart Fail.
2015;22:123‐131. 2018;11(2):e004000.
25. Durmuş‐Tekçe H, Matur Z, Mert Atmaca M, et al. Genotypic and 45. Sekijima Y, Tokuda T, Kametani F, Tanaka K, Maruyama K, Ikeda
phenotypic presentation of transthyretin‐related familial am‐ S. Serum transthyretin monomer in patients with familial amyloid
yloid polyneuropathy (TTRFAP) in Turkey. Neuromuscul Disord. polyneuropathy. Amyloid. 2001;8:257‐262.
2016;26:441‐446. 46. Csillik A, Pozsonyi Z, Soós K, Balogh I, Bodó I, Arányi Z.
26. Koike H, Misu K, Sugiura M, et al. Pathology of early‐ vs late‐ Transthyretin familial amyloid polyneuropathy ‐ three Hungarian
onset TTR Met30 familial amyloid polyneuropathy. Neurology. cases with rare mutations (His88Arg and Phe33Leu). Ideggyogy Sz.
2004;63:129‐138. 2016;69:245‐253.
27. Koike H, Misu K, Ikeda S et al. Type I (transthyretin Met30) familial 47. Castro J, Miranda B, Castro I, et al. The diagnostic accuracy of
amyloid polyneuropathy in Japan: early‐ vs late‐onset form. Arch Sudoscan in transthyretin familial amyloid polyneuropathy. Clin
Neurol. 2002;59:1771‐1776. Neurophysiol. 2016;127:2222‐2227.
104 | FINSTERER et al.

48. Sène D. Small fiber neuropathy: Diagnosis, causes, and treatment. avoiding misdiagnosis of a treatable hereditary neuropathy. J Neurol
Joint Bone Spine 2018t;85:553–559. Neurosurg Psychiatry. 2017;88:457‐458.
49. Lefaucheur JP, Wahab A, Planté‐Bordeneuve V et al. Diagnosis of 68. Lozeron P, Lacroix C, Theaudin M, et al. An amyotrophic lat‐
small fiber neuropathy: A comparative study of five neurophysio‐ eral sclerosis‐like syndrome revealing an amyloid polyneurop‐
logical tests. Neurophysiol Clin 2015;45:445‐455. athy associated with a novel transthyretin mutation. Amyloid.
50. Wiklund U, Kadkhodaee A, Andersson K, Suhr OB, Hörnsten R. 2013;20:188‐192.
Normal scores of deep breathing tests: beware of dysrhythmia in 69. Ishida K, Nishida T, Niimi Y, et al. Elderly onset vitreous opaci‐
transthyretin amyloidosis. Amyloid. 2018;2:1‐8. ties as the initial manifestation in hereditary transthyretin (ATTR
51. Dardiotis E, Koutsou P, Papanicolaou EZ, et al. Epidemiological, Val30Met) carries. Ophthalmic Genet. 2017;38:387‐391.
clinical and genetic study of familial amyloidotic polyneuropathy in 70. Yamamoto S, Wilczek HE, Nowak G, et al. Liver transplantation for
Cyprus. Amyloid. 2009;16:32‐37. familial amyloidotic polyneuropathy (FAP): a single‐center experi‐
52. Jamet MP, Gnemmi V, Hachulla É, et al. Distinctive patterns of ence over 16 years. Am J Transplant. 2007;7:2597‐2604.
transthyretin amyloid in salivary tissue: a clinicopathologic study of 71. Tsuchiya A, Yazaki M, Kametani F, Takei Y, Ikeda S. Marked regres‐
92 patients with amyloid‐containing minor salivary gland biopsies. sion of abdominal fat amyloid in patients with familial amyloid poly‐
Am J Surg Pathol. 2015;39:1035‐1044. neuropathy during long‐term follow‐up after liver transplantation.
53. Koike H, Ikeda S, Takahashi M, et al. Schwann cell and endothe‐ Liver Transpl. 2008;14:563‐570.
lial cell damage in transthyretin familial amyloid polyneuropathy. 72. Yamashita T, Ando Y, Ueda M et al. Effect of liver transplantation
Neurology. 2016;87:2220‐2229. on transthyretin Tyr114Cys‐related cerebral amyloid angiopathy.
54. Parman Y, Adams D, Obici L et al. Sixty years of transthyretin fa‐ Neurology. 2008;70:123‐128.
milial amyloid polyneuropathy (TTR‐FAP) in Europe: where are 73. Okamoto S, Wixner J, Obayashi K et al. Liver transplantation for
we now? A European network approach to defining the epidemi‐ familial amyloidotic polyneuropathy: impact on Swedish patients’
ology and management patterns for TTR‐FAP. Curr Opin Neurol. survival. Liver Transpl. 2009;15:1229‐1235.
2016;29(suppl 1):S3‐S13. 74. Suhr OB, Larsson M, Ericzon BG, Wilczek HE. Survival after trans‐
55. Planté‐Bordeneuve V, Ferreira A, Lalu T et al. Diagnostic pitfalls in plantation in patients with mutations other than Val30Met: ex‐
sporadic transthyretin familial amyloid polyneuropathy (TTR‐FAP). tracts from the FAP World Transplant Registry. Transplantation.
Neurology. 2007;69:693‐698. 2016;100:373‐381.
56. Koike H, Hashimoto R, Tomita M et al. Diagnosis of sporadic trans‐ 75. Ericzon BG, Wilczek HE, Larsson M, et al. Liver transplantation for
thyretin Val30Met familial amyloid polyneuropathy: a practical hereditary transthyretin amyloidosis: after 20 years still the best
analysis. Amyloid. 2011;18:53‐62. therapeutic alternative? Transplantation. 2015;99:1847‐1854.
57. Dohrn MF, Röcken C, De Bleecker JL, et al. Diagnostic hallmarks 76. Hou X, Aguilar MI, Small DH. Transthyretin and familial amyloidotic
and pitfalls in late‐onset progressive transthyretin‐related amyloid polyneuropathy. Recent progress in understanding the molecular
neuropathy. J Neurol. 2013;260:3093‐3108. mechanism of neurodegeneration. FEBS J. 2007;274:1637‐1650.
58. Masuda T, Ueda M, Suenaga G, et al. Early skin denervation in 77. Adams D, Cauquil C, Labeyrie C, et al. TTR kinetic stabilizers and
hereditary and iatrogenic transthyretin amyloid neuropathy. TTR gene silencing: a new era in therapy for familial amyloidotic
Neurology. 2017;88:2192‐2197. polyneuropathies. Expert Opin Pharmacother. 2016;17:791‐802.
59. Chao CC, Huang CM, Chiang HH, et al. Sudomotor innervation in 78. Waddington Cruz M, Amass L, Keohane D, et al. intervention with
transthyretin amyloid neuropathy: pathology and functional cor‐ tafamidis provides long‐term (5.5‐year) delay of neurologic progres‐
relates. Ann Neurol. 2015;78:272‐283. sion in transthyretin hereditary amyloid polyneuropathy. Amyloid.
60. Lavigne‐Moreira C, Marques VD, Magno Gonçalves MV, et al. The 2016;23:178‐183.
genetic heterogeneity of hereditary transthyretin amyloidosis in 79. Planté‐Bordeneuve V, Gorram F, Salhi H et al. Long‐term treat‐
a sample of the Brazilian population. J Peripher Nerv Syst. 2018; ment of transthyretin familial amyloid polyneuropathy with
23:134‐137. tafamidis: a clinical and neurophysiological study. J Neurol.
61. Sperry BW, Gonzalez MH, Brunken R, Cerqueira MD, Hanna M, 2017;264:268‐276.
Jaber WA. Non‐cardiac uptake of technetium‐99m pyrophos‐ 80. Mundayat R, Stewart M, Alvir J et al. Positive effectiveness of
phate in transthyretin cardiac amyloidosis. J Nucl Cardiol. 2018. tafamidis in delaying disease progression in transthyretin famil‐
https://doi.org/10.1007/s12350-017-1166-7 [Epub ahead of ial amyloid polyneuropathy up to 2 years: an analysis from the
print]. transthyretin amyloidosis outcomes survey (THAOS). Neurol Ther.
62. Chen W, Ton VK, Dilsizian V. Clinical phenotyping of transthyretin 2018;7:87‐101.
cardiac amyloidosis with bone‐seeking radiotracers in heart failure 81. Coelho T, Maia LF, Martins da Silva A, et al. Tafamidis for trans‐
with preserved ejection fraction. Curr Cardiol Rep. 2018;20(4):23. thyretin familial amyloid polyneuropathy: a randomized, controlled
63. Treglia G, Glaudemans A, Bertagna F et al. Diagnostic accuracy trial. Neurology. 2012;79:785‐792.
of bone scintigraphy in the assessment of cardiac transthyretin‐ 82. Cappelli F, Martone R, Taborchi G et al. Epigallocatechin‐3‐gallate
related amyloidosis: a bivariate meta‐analysis. Eur J Nucl Med Mol tolerability and impact on survival in a cohort of patients with trans‐
Imaging. 2018; 45):1945‐1955. thyretin‐related cardiac amyloidosis. A single‐center retrospective
64. Rousseau A, Cauquil C, Dupas B, et al. Potential role of in vivo con‐ study. Intern Emerg Med. 2018; 13:873‐880.
focal microscopy for imaging corneal nerves in transthyretin famil‐ 83. Adams D, Gonzalez‐Duarte A, O'Riordan WD et al. Patisiran, an
ial amyloid polyneuropathy. JAMA Ophthalmol. 2016;134:983‐989. RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J
65. Misumi Y, Ueda M, Yamashita T, et al. Novel screening for trans‐ Med. 2018;379:11‐21.
thyretin amyloidosis by using fat ultrasonography. Ann Neurol. 84. Benson MD, Waddington‐Cruz M, Berk JL et al. Inotersen treat‐
2017;81:604‐608. ment for patients with hereditary transthyretin amyloidosis. N Engl
66. Planté‐Bordeneuve V. Update in the diagnosis and manage‐ J Med. 2018;379:22‐31.
ment of transthyretin familial amyloid polyneuropathy. J Neurol. 85. Rosenblum H, Castano A, Alvarez J, Goldsmith J, Helmke S, Maurer
2014;261:1227‐1233. MS. TTR (Transthyretin) stabilizers are associated with improved
67. Cortese A, Vegezzi E, Lozza A, et al. Diagnostic challenges in survival in patients with TTR cardiac amyloidosis. Circ Heart Fail.
hereditary transthyretin amyloidosis with polyneuropathy: 2018;11:e004769.
FINSTERER et al. | 105

86. Wilczek HE, Larsson M, Ericzon BG. Long‐term data from the 91. Algalarrondo V, Antonini T, Théaudin M, et al. Cardiac dysau‐
familial amyloidotic polyneuropathy world transplant registry tonomia predicts long‐term survival in hereditary transthyretin
(FAPWTR). Amyloid. 2011;18(suppl 1):193‐195. amyloidosis after liver transplantation. JACC Cardiovasc Imaging.
87. Herlenius G, Wilczek HE, Larsson M, et al. Familial Amyloidotic 2016;9:1432–1441.
Polyneuropathy World Transplant Registry; Ten years of interna‐ 92. Liu YT, Lee YC, Yang CC, Chen ML, Lin KP. Transthyretin Ala97Ser
tional experience with liver transplantation for familial amyloidotic in Chinese‐Taiwanese patients with familial amyloid polyneu‐
polyneuropathy: results from the familial amyloidotic polyneuropa‐ ropathy: genetic studies and phenotype expression. J Neurol Sci.
thy world transplant registry. Transplantation. 2004;77:64‐71. 2008;267:91–99.
88. 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;255:1526‐1533.
How to cite this article: Finsterer J, Iglseder S, Wanschitz J,
89. Carr AS, Pelayo‐Negro AL, Evans MR, et al. A study of the neurop‐
Topakian R, Löscher WN, Grisold W. Hereditary
athy associated with transthyretin amyloidosis (ATTR) in the UK. J
Neurol Neurosurg Psychiatry. 2016;87:620‐627. transthyretin‐related amyloidosis. Acta Neurol Scand.
90. Algalarrondo V, Antonini T, Théaudin M, et al. Prediction of long‐ 2019;139:92–105. https://doi.org/10.1111/ane.13035
term survival after liver transplantation for familial transthyretin
amyloidosis. J Am Coll Cardiol. 2015;66:2154‐2156.
Copyright of Acta Neurologica Scandinavica is the property of Wiley-Blackwell and its
content may not be copied or emailed to multiple sites or posted to a listserv without the
copyright holder's express written permission. However, users may print, download, or email
articles for individual use.

You might also like