You are on page 1of 7

Current Heart Failure Reports

https://doi.org/10.1007/s11897-020-00457-z

BIOMARKERS OF HEART FAILURE (WH TANG & J GRODIN, SECTION EDITOR)

Disease-Specific Biomarkers in Transthyretin Cardiac Amyloidosis


Nicholas S. Hendren 1 & Lori R. Roth 1 & Justin L. Grodin 1

# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Purpose of Review Transthyretin amyloidosis is an increasingly recognized cause of restrictive cardiomyopathy related to
amyloid fibril deposition in cardiac tissues. As treatment therapies have emerged for transthyretin amyloidosis (ATTR), so has
interest in using biomarkers to identify disease prior to advanced presentation.
Recent Findings Lower levels of transthyretin and retinol binding protein-4 have been demonstrated in patients with pathogenic
mutations of transthyretin either with or without clinical disease. Levels associate with the severity of mutations as well as
response to treatment with transthyretin stabilizers or small interfering RNA molecules which silence transthyretin production.
Transthyretin stability is the rate limiting step of amyloid fibril formation and directly measuring transthyretin kinetic stability has
the potential to identify patients as risk as well as therapeutic response to treatment regardless of pathogenic or wild-type genetics.
In addition, non-antibody protein-based peptide probes have been developed that directedly measure misfolded transthyretin
oligomers due to transthyretin breakdown. Although promising, both TTR kinetic and protein peptide probes remain in early
stages of clinical investigation.
Summary Transthyretin, retinol binding protein-4, transthyretin kinetic stability, and protein-based peptide probes have potential
as biomarkers to facilitate an earlier ATTR diagnosis for patients with pathogenic transthyretin mutations.

Keywords Transthyretin amyloidosis . Retinol binding protein . Biomarkers

Introduction manifestations of ATTR [1]. Cardiac manifestations include


diastolic filling abnormalities, conduction system abnormali-
Cardiac transthyretin amyloidosis (ATTR) is a chronic, progres- ties, arrhythmias, and heart failure [2]. Neurologic involvement
sive, and often fatal disease due to insoluble transthyretin (TTR) from ATTR is a source of substantial morbidity and is charac-
protein deposition in organ systems throughout the body in terized by sensory motor neuropathy, autonomic neuropathy,
older adults (Fig. 1). These misfolded amyloid fibrils principal- and dysautonomia [3]. These symptoms can lead to a substan-
ly occur due to unstable TTR tetramers that can spontaneously tial reduction in quality of life, worsening functional capacity,
occur without a pathogenic mutation (wild-type transthyretin and increased healthcare utilization [4–7].
amyloidosis, ATTRwt) or due to a pathogenic mutation that The diagnosis of ATTR is typically delayed years after
increases TTR structural instability (hereditary transthyretin initiation of symptoms. In a review of 534 patients with
amyloidosis, hATTR). Although a spectrum of symptoms can ATTR, patients presented to the hospital a median of 17
be present, they are often related to cardiac and neurologic [9–27] times with 3 [1–5] in-patient admissions in the
3 years preceding diagnosis [7]. Historically, median sur-
vival at the time of diagnosis has been estimated at approx-
This article is part of the Topical Collection on Biomarkers of Heart Failure
imately 4.8 years for ATTRwt, 2.5 years with V122I
* Justin L. Grodin
hATTR and about 5.8 years for non-V122I hATTR [7].
Justin.Grodin@utsouthwestern.edu Fortunately, recent therapeutic advances have led to the
development of two major therapeutic drug classes with
Nicholas S. Hendren
nicholas.hendren@phhs.org the capacity to slow and halt ATTR disease progression.
TTR stabilizers bind to TTR stabilizing the protein com-
1
Division of Cardiology, Department of Internal Medicine, University plex and reduce protein disassembly that leads to amyloid
of Texas Southwestern Medical Center, Dallas, TX, USA deposition. The second class includes gene silencing
Curr Heart Fail Rep

Fig. 1 Conceptual model of cardiac ATTR progression over time. capacity. *Biomarker Stage defined by the Mayo Clinic for ATTRwt as
Changes in various parameters are shown. The relative scale specific to cardiac troponin T (< 0.05 ng/mL) and amino-terminal pro-B-type
each factor and time course are not proportional. Myocardial amyloid natriuretic peptide (< 3000 pg/mL) with stages I, II, and III defined as
infiltration occurs before clinically manifest changes in ejection having both, one, or neither of the markers below the threshold. ATTR
fraction, cardiac biomarkers, and renal function. Thus, most patients indicates transthyretin amyloidosis; hATTR, hereditary transthyretin
with cardiac ATTR likely have a long latency period before declines in amyloidosis; ATTRwt, wild-type transthyretin amyloidosis; KCCQ,
functional capacity, which can occur rapidly in the context of multiple Kansas City Cardiomyopathy Questionnaire; 6 MWD, 6-min walk
hospitalizations for acute decompensated heart failure and arrhythmias. distance; NYHA, New York Heart Association; QOL, quality of life.
The ideal emerging therapeutic window for novel therapies is Figure and caption used with permission from: Grodin JL, Maurer MS.
hypothesized to be before significant organ dysfunction has occurred The Truth Is Unfolding About Transthyretin Cardiac Amyloidosis.
and before rapid and potentially irreversible declines in functional Circulation. 2019;140 (1):27–30

therapies that silence hepatic production of TTR and dra- including heart failure; however, less is known about bio-
matically reduce amyloid fibril formation [4, 8••, 9]. markers for ATTR. Currently transthyretin, retinol-binding
With the development of new therapies, screening strate- protein-4 (RBP4), measurements of TTR kinetic stability,
gies to detect subclinical ATTR in the presymptomatic patient and non-antibody peptide probes have shown promise as po-
have gained interest. Previously factors such as N-terminal tential biomarkers to identify patients are risk for ATTR. As
pro-B-type natriuretic peptide (NT-proBNP), left ventricular such, we will review possible biomarkers and the evidence for
ejection fraction, uric acid, troponin T, troponin I, and renal their utility.
function have been associated with survival but lack precision
to differentiate ATTR from alternative diseases [10–12].
Consequently, identifying prognostic, cost-effective, and eas- Transthyretin
ily applicable preclinical markers to screen large populations
at risk is crucial for early identification of patients with pre- Commonly known as prealbumin, TTR is a liver-secreted
clinical ATTR that would benefit from treatment (Fig. 1). plasma protein composed of 4 identical 127-amino acid
Biomarkers are well suited to identify patients early in the monomers that are non-covalently bound and circulate as a
disease course. Biomarkers play a major role in the diagnosis, tetramer [13]. TTR functions as a carrier of thyroxine and its
risk stratification, and therapeutic decisions for many diseases binding partner to retinol-binding protein 4 (RBP4) [13].
Curr Heart Fail Rep

ATTR pathogenesis occurs when the TTR tetramer disassem- Transthyretin is a negative acute-phase reactant which re-
bles into monomers which can then form amyloid fibrils sults in lower levels during malnutrition or chronic inflamma-
(Fig. 2). tion [16, 17]. TTR is unusually rich in the amino acid tyrosine
TTR is a promising biomarker for ATTR for several rea- which makes TTR more sensitive to protein deficient diets or
sons. First, TTR has a short half-life allowing for serial mea- catabolic states [17]. Although it was previously used a nutri-
surement, abnormal levels may predate clinical symptoms and tional biomarker, the use of TTR to assess nutritional status is
TTR levels are associated with survival in ATTR [13]. no longer recommended as levels of TTR can be affected by
Second, the principal pathogenesis is an unstable TTR tetra- extreme zinc deficiency, calorie deficiencies or acute and/or
mer leading to amyloid production. It has been observed that chronic inflammation in an unpredictable manner that is inde-
patients with more pathogenic (destabilizing) mutations have pendent of nutritional status [18]. Lastly, higher levels of TTR
lower levels of TTR [13], while patients with TTR stabilizing are observed with glucocorticoid use, and acute liver injury
mutations have higher levels of TTR [14]. Lastly, treatment due to release from damaged hepatic cells [17].
with TTR stabilizers increases serum TTR levels and may A normal TTR concentration is 18–45 mg/dL which is
indicate a positive prognostic response to therapy [13]. dependent on age, sex, and race [12] with a biological half-
Alternately, therapies that silence TTR translation led to dra- life of about 2 days [17]. Multiple commercial labs have avail-
matic reductions of serum TTR levels serving as a different able testing to precisely quantify TTR; however, modest intra-
marker of therapeutic efficacy [4, 9]. and inter-person variability occur can with serial measure-
TTR can be detected in fetal blood within 8 weeks of con- ments. In a small study, serial TTR measurements were ob-
ception and increases to approximately half the adult level in tained weekly for a month in 15 patients with biopsy proven
healthy neonates [12]. At the onset of puberty, TTR levels hATTR and 11 patients with a pathogenic mutation without
increase to adult levels with slightly higher levels in males clinical ATTR [20]. The overall mean TTR value was
compared with females presumably due to higher levels of 20.2 mg/dL with approximately 23% intrasubject variability
testosterone (32.3 ± 4.9 mg/dL vs 28.3 ± 4.3 mg/dL) [15]. and 25% inter subject variability over 1 month [19].
After age 50, TTR levels begin to progressively decline with Several studies have observed significantly lower levels of
age and sex specific differences are eliminated during the sev- TTR with inherited forms of ATTR amyloidosis [12]. For
enth decade of life [15]. As such, although there are sex- many years, it has been observed that serum TTR concentra-
specific differences that attenuate with age, they are modest tions are relatively low in carriers of autosomal dominant
and typically do not overlap with frankly abnormal values. amyloidogenic TTR mutations even before the signs and

Fig. 2 Pathobiology of transthyretin amyloid. The mechanism of with permission from: Ruberg FL, Grogan M, Hanna M, Kelly JW,
transthyretin (TTR) protein dissociation, misfolding, and aggregation as Maurer MS. Transthyretin Amyloid Cardiomyopathy: JACC State-of-
amyloid fibrils is illustrated with resultant end-organ dysfunction. ATTR, the-Art Review. J Am Coll Cardiol. 2019;73 (23):2872–91
transthyretin amyloid; mRNA, messenger RNA. Figure and caption used
Curr Heart Fail Rep

symptoms of familial amyloid polyneuropathy (FAP) become established clinical ATTRwt; however, TTR may not be able
apparent [20]. Total serum TTR concentrations were obtained to discriminate preclinical ATTRwt from healthy subjects at
in 47 Swedish patients with varied states of clinical disease the population level [12]. Additional follow-up was complet-
known to be V30M amyloidogenic allele carriers. TTR levels ed in 23 untreated ATTRwt patients and 12 patients treated
were significantly lower in V30M allele carriers compared with a TTR stabilizer. In the treated group, TTR increased at
with homozygous wild-type individuals (16.8 vs. 22.0 mg/ year 1 and remained significant higher in the treated cohort at
dL, P = 0.005) [20]. Asymptomatic allele carriers were also 2-year follow-up (20 vs. 31 mg/dL, P < 0.001) [12]. This is
found to have an abnormal TTR level (19.3 mg/dL) [20]. This one early observation that increases in TTR levels may also be
observation was similarly reported in V30M carriers in pa- associated with a positive response to treatment.
tients of Portuguese, African, and Japanese ethnicity In general, the more destabilizing the TTR mutation, the
supporting that asymptomatic carriers of pathogenic TTR mu- more penetrant and severe the phenotype [22]. A super-
tations may have abnormal TTR levels [20]. stabilizing mutation (T119M) has been identified that protects
In a second, larger observational study in the US serum carriers from clinical ATTR or FAP by reducing the dissociation
TTR concentrations were also evaluated in a selected cohort rate of TTR by over 33-fold compared with wild-type protein
of patients older than 60 years. Forty-three patients with [22]. Compound heterozygotes carrying both T119M and the
biopsy-proven ATTRwt were compared with 128 Caucasian V30M mutations develop few if any manifestations of ATTR
control patients. Although TTR was slightly numerically [23]. One large observational study of 68,602 patients prospec-
higher in control patients, there was no statistically significant tively identified 321 heterozygotes with the T119M mutation
difference (24.8 vs 25.0 mg/dL; P = 0.72) [21]. There was also (0.47% prevalence) [24]. Compared with non-carriers, hetero-
no statistical difference between in TTR levels between all zygotes for T119M had a mean TTR level of 18.3 mg/dL vs
patients with ATTRwt (N = 45) or controls (N = 128) com- 15.7 mg/dL, (P = 0.007) which represented a 17% difference.
pared with 20 patients with light chain amyloidosis with heart When adjusted for age and sex, a 20% difference remained
failure (237 vs 250 vs 195 mg/dL; P = 0.59, P = 0.24 respec- [24]. Again, a higher TTR level was prognostic as T119M
tively) [21]. TTR levels were then evaluated in 828 Caucasian heterozygotes had a median of 5 more years of life expectancy
and 826 African-American control patients. TTR levels were compared with non-carriers (P = 0.04) [24]. This study supports
each independently higher in men, patients younger than 60, the observation that higher TTR levels and increased kinetic
and Caucasians compared with African-Americans (26.3 vs stability are associated with improved clinical outcomes.
24.8 mg/dL) [21]. Finally, they identified 12 African- Two main mechanisms of treatment have been developed
American patients with a pathogenic V122I mutation. After for ATTR which include TTR stabilizing compounds and
adjusting for age and sex, TTR levels were lower in patients small interfering RNA molecules to silence TTR production.
with a pathogenic mutation (17.1 vs 22.9 mg/dL). This study TTR levels may have prognostic ability for treatment for each
supports a few important observations. First, TTR levels may drug class. Suhr et al. recruited 29 patients with biopsy proven
not differ enough between patients with ATTRwt and healthy ATTRwt or hATTR for a phase II clinical trial of patisiran
controls to serve as a precise biomarker for preclinical (small interfering RNA) with four different dosing regimens
ATTRwt. Second, in a small sample size TTR levels were [25]. Baseline serum TTR concentrations were similar across
similar between patients with AL and control patients despite the four groups; however, more intensive treatment led to
concerns about AL confounding TTR levels. Finally, lower greater reduction of TTR with mean reductions of approxi-
TTR levels in a small selected cohort are supportive that TTR mately 80% and maximal reductions of 90% [25]. In the phase
should be investigated as a possible biomarker for hATTR. III clinical trial of patisiran, patients had a sustained 81%
Importantly, the V122I carriers had significantly lower serum reduction of TTR with clinical improvement of neuropathy
TTR concentrations despite the majority of subjects below the and quality of life [4], indicative that TTR may serve as a
age of clinical risk for ATTR [21]. surrogate marker for clinical response to patisiran.
Hanson et al. retrospectively evaluated 116 patients with Patients treated with TTR stabilizers such as tafamidis or
clinical ATTRwt compared with 30 control patients ≥ 60 years diflunisal have been observed to have higher levels of TTR
old to evaluate TTR levels [12]. Baseline TTR levels were compared with non-treated patients [25]. Tafamidis binds to
prognostic and patients with baseline TTR ≥ 18 mg/dL had a TTR, and stabilizes TTR to reduce dissociation of TTR, which
median survival time of 4.1 years versus 2.8 years in the group is a rate-limiting step in amyloid fibril formation. Tafamidis is
with TTR < 18 mg/dL (P = 0.03, HR = 2.3, 95% CI 1.2–4.3) shown to reduce morbidity and mortality in subjects with
[12]. Higher TTR values in both univariable and multivariable ATTR in a large placebo controlled clinical trial [8••]. In a
analyses were positively associated with longer survival in review of 31 patients with ATTR (25 ATTRwt and 6 hATTR)
patients with ATTRwt [12]. As such, a serum TTR < 18 mg/ treated with tafamidis, serum TTR levels were obtained pre and
dL may have the potential to identify patients at increased risk post medication initiation [26•]. The mean serum TTR levels
for clinical outcomes and associate with prognosis for prestabilize administration were 20.6 ± 5.6 mg/dL and were
Curr Heart Fail Rep

similar between ATTRwt and hATTR patients [26•]. With increased amyloid production [30]. In a small cohort of 47
tafamidis, TTR levels increased in from 18.5 ± 7.2 to 28 ± patients with ATTRwt and 27 patients with hATTR V122I,
7.4 mg/dL in hATTR subjects (increase of 37%; P = 0.046) serum RBP4 concentration was lower compared with non-
and 21.3 ± 4.8 to 29.9 ± 6.0 in ATTRwt (increase of 40%; amyloid controls (31.5 vs 49.4 μg/mL, P < .001), and the dif-
P < 0.0001) [26•]. These increases did not appear to differ be- ference persisted after adjusting for several confounders [31•].
tween men and women. Overall, this study was suggestive that As expected, RBP4 and TTR levels were highly correlated
increased TTR kinetic stability with tafamidis translates to in- (Spearman rho = 0.75, P = 0.001) [31•]. For screening pur-
creased levels of TTR in subjects with clinical ATTR. poses, a RBP4 level < 49.5 μg/mL achieved a high sensitivity,
A second TTR stabilizer, AG10, was tested in a phase II but poor specificity in this exploratory analysis for patients
clinical trial in patients with ATTR (11 hATTR, 38 ATTRwt) with hATTR V122I [31•]. This analysis in combination with
and New York Heart Association Class II-III symptoms [13]. additional observations suggests RBP4 levels are associated
AG10 is a potent TTR stabilizer that stabilizes TTR > 90% by with pathogenic TTR mutations and may be a reasonably
mimicking the T119M variant which forms a stabilizing bond sensitive biomarker to screen for ATTR and that warrants
within the TTR tetramer. In this trial, participants were given a further investigation within the general population.
placebo, 400 mg or 800 mg dose twice daily of AG10 for In a second study, RBP4 levels were obtained in 25 patients
28 days. The baseline median TTR level was 22.0 ± 5.4 mg/ with biopsy proven V122I ATTR and compared with 50 pa-
dL [13]. AG10 was a highly effective TTR stabilizer provid- tients older than 60 years. Serum RBP4 concentrations were
ing > 90% stabilization to the TTR tetramer leading to a 36% found to be significantly lower in patients with V122I ATTR
increase of TTR in the 400 mg group, 50% increase in the compared with patients without amyloidosis (31.5 vs.
800 mg group, and a 7% decline in the placebo group [13]. 49.4 μg/mL, P < 0.001) despite adjustment for age, sex, tro-
There was also a greater increase observed in the hATTR ponin level, nutritional status, and echocardiographic param-
compared with ATTRwt group suggesting that AG10 may eters [32]. As seen previously, RBP4 and TTR levels were
more effective in less stable tetramers. However, the magni- highly correlated (Spearman rho = 0.75, P < 0.001) [32]. In
tude of change in serum TTR was less than the magnitude of this small cohort of patients with V122I ATTR, RBP4 at a
change with tafamidis. threshold of 43 μg/mL maximized the prognostic value with
In sum, TTR is a reproducible, easily quantifiable protein a sensitivity of 62% (95% CI: 47–77%) and a specificity of
associated with clinical outcomes and abnormal in patients 88% (95% CI 76–100%) [35]. For a higher sensitivity a
with both clinical and preclinical ATTR. TTR levels associate threshold of 50 μg/mL maximized sensitivity at 100% (95%
with the degree of TTR instability and increases in TTR levels CI, 100–100%); however, the specificity declined to 38%
are associated with treatment efficacy. (95% CI, 23–51%) [32]. In short, RBP4 merits further inves-
tigation as a biomarker for ATTR in a larger and more patho-
Retinol-Binding Protein logically diverse cohort.
Despite the presence of pathogenic mutations in TTR, the
Retinol is a hydrophobic compound that requires binding to age of onset for clinical disease has considerable variance
retinol binding protein 4 (RBP4) in order to secreted into the within families and can include asymptomatic carriers as old
blood stream and delivered to target organs [27]. Both un- as 95 years old [33]. This suggests that additional factors may
bound retinol and RBP4 are rapidly cleared from the blood- modify disease penetrance. Mutations in RBP4 which either
stream largely via urinary excretion (retinol) or urinary metab- stabilize or destabilize the TTR-RBP4 complex may affect the
olism (RBP4). Retinol-RBP4 complexes then bind to TTR penetrance of either hATTR or ATTRwt. In a review of 318
which stabilizes the tetramer preventing TTR disassembly, patients with a V30M mutation, genetic differences in RBP4
amyloid aggregation, [28], and urinary losses of RBP4 and modified the age of onset for clinical symptoms of amyloid-
retinol [29]. RBP4 is primarily produced in the liver with an osis by as much as 19 years [30]. This is suggestive that RBP4
additional 20% production from adipose tissue [27], and it has may have a pathophysiological link to ATTR which can serve
been observed that RBP4 levels and TTR levels are associat- as protective role against pathological mutations and/or serve
ed. In a phase II clinical trial of patisiran, the level of serum as a marker of more pathological ATTR mutations to influence
TTR reduction was highly correlated with the reduction in the disease penetrance of ATTR. In sum, RBP4 levels are
circulating level of RBP4 (r2 = 0.89, P < 10−15) and vitamin associated with pathogenic mutations and show promise as a
A (r2 = 0.89, P < 10−15) [25]. Thus, lower levels of RBP4 biomarker for ATTR in limited clinical cohorts.
appear to parallel serum TTR levels.
Destabilizing TTR mutations that cause TTR instability Novel Marks of TTR Stability
also translate into fewer circulating RBP4-TTR complexes
and increased urinary excretion of RBP4. However, lower The fundamental problem and rate-limiting step of ATTR re-
RBP4 levels may also destabilize TTR leading towards lates to the disassembly of unstable TTR complexes into
Curr Heart Fail Rep

amyloid fibrils. Two methods currently being evaluated as a rather than expensive genotyping which may discover genetic
biomarker for ATTR include measuring kinetic stability of the mutations of undetermined significance. Additionally, these
TTR homotetramer and using peptide probes to quantify the test results may associate with a response to disease modifying
level of misfolded TTR oligomers. One established method to therapy in future research studies. However, significant uncer-
quantify the stability of TTR tetramers involves measuring the tainly remains about the utility of both tests and it remains to
rate of subunit exchange with the TTR tetramer. The rate of be seen if changes in TTR kinetics or oligomer concentration
exchange is referred to as kinetic stability and may serve as a associate with clinical outcomes.
surrogate biomarker for the prediction of the clinical outcome
and response to therapy [34].
The variability in the age of onset or the aggressiveness of Conclusion
the hereditary TTR amyloidosis can, in part, be attributed to
differences in the thermodynamic and kinetic stability of tet- In sum, ATTR is an increasingly recognized cause of heart
ramers comprising disease-associated and ATTRwt subunits failure. With development of recent pharmacotherapies iden-
[34]. The decreased kinetic stability of TTR tetramers incor- tifying patients earlier in the disease course to modifying dis-
porating pathogenic mutated subunits leads to an increased ease sequalae has become an important area of investigation.
rate of TTR dissociation [34]. Pharmacologic therapies that The identification of a biomarker to identify those at risk of or
stabilize TTR by binding to the native TTR tetramer slow diagnose patients with ATTR in a preclinical stage is a large
the progression of disease by limiting TTR disassembly. unmet need because many centers do not have the facilities or
This is illustrated by tafamidis which stabilizes TTR 4-fold expertise needed to complete cardiac magnetic resonance im-
in patients receiving the drug and reduces the progression of aging or bone scintigraphy scans making them impractical at a
symptoms [34]. national level. Although earlier studies are promising, further
TTR kinetics may also be able to quantify a patient’s re- investigation is needed to clarify if abnormalities in TTR,
sponse to kinetic stabilizer treatment, which is especially valu- RBP4, TTR kinetic stability, and transthyretin oligomers can
able for patients carrying substantially destabilized, rare, or precisely identify patients with preclinical ATTR that merit
currently uncharacterized TTR mutations. With future study, further clinical evaluation.
the quantification of TTR kinetic stability has the potential to
become a surrogate biomarker to predict clinical response to Compliance with Ethical Standards
therapies or screen for asymptomatic patients at risk for
ATTRwt or hATTR [34]. Conflict of Interest Dr. Grodin received research funding from the Texas
A second novel diagnostic tool is the development of pep- Health Resources Clinical Scholarship and receives consulting income
from Pfizer and Eidos Therapeutics. Dr. Nicholas Hendren and Lori Roth
tide probes that selectively bind to pathogenic TTR oligomers declare that they have no conflict of interest.
in the serum of patients with hATTR. These probes work by
binding to specific peptide sequences in misfolded TTR olig- Human and Animal Rights and Informed Consent This article does not
omers which are not available in the more densely bound contain any studies with human or animal subjects performed by any of
native TTR or TTR amyloid fibrils [35]. This allows the the authors.
probes to precisely quantitate the level of TTR oligomers sep-
arate from amyloid fibrils or TTR regardless of the genetic
mutations in TTR or RBP4 [35]. In small highly selected References
cohorts, peptide probes identified higher levels of misfolded
TTR oligomers in patients with clinical FAP, but not in healthy Papers of particular interest, published recently, have been
controls, asymptomatic carriers of mutations associated with highlighted as:
FAP, or patients with cardiac hATTR or ATTRwt [35]. • Of importance
Additional investigation in patients with FAP observed a sig- •• Of major importance
nificant decrease in misfolded oligomers for patients treated
with tafamidis (1.8 fold decline) or liver transplantation (no 1. Rapezzi C, Lorenzini M, Longhi S, Milandri A, Gagliardi C,
Bartolomei I, et al. Cardiac amyloidosis: the great pretender.
detectable pathogenic TTR oligomers) [35]. As such peptide Heart Fail Rev. 2015;20(2):117–24.
probes may be an evolving biomarker for FAP with levels 2. Dungu JN, Anderson LJ, Whelan CJ, Hawkins PN. Cardiac
indicative of a response to therapy, but further investigation transthyretin amyloidosis. Heart. 2012;98(21):1546–54.
will be required. 3. Kapoor M, Rossor AM, Laura M, Reilly MM. Clinical presenta-
To date, both TTR kinetic stability and protein-based pep- tion, diagnosis and treatment of TTR amyloidosis. J Neuromuscul
Dis. 2019;6(2):189–99.
tide probes have had limited study in the clinical and preclin- 4. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M,
ical ATTR population. Both tests could be developed to screen Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary
patients for pathogenic TTR prior to symptom development transthyretin amyloidosis. N Engl J Med. 2018;379(1):11–21.
Curr Heart Fail Rep

5. Coelho T, Maurer MS, Suhr OB. THAOS - The Transthyretin 22. Hammarstrom P, Jiang X, Hurshman AR, Powers ET, Kelly JW.
Amyloidosis Outcomes Survey: initial report on clinical manifesta- Sequence-dependent denaturation energetics: a major determinant
tions in patients with hereditary and wild-type transthyretin amy- in amyloid disease diversity. Proc Natl Acad Sci U S A.
loidosis. Curr Med Res Opin. 2013;29(1):63–76. 2002;99(Suppl 4):16427–32.
6. Cruz MW. Tafamidis for autonomic neuropathy in hereditary 23. Coelho T, Chorao R, Sousa A, Alves A, Torres MF, Saraiva M.
transthyretin (ATTR) amyloidosis: a review. Clin Auton Res. Compound heterozygotes of transthyretin Met30 and transthyretin
2019;29(Suppl 1):19–24. Met119 are protected from the devastating effects of familial amy-
7. Lane T, Fontana M, Martinez-Naharro A, Quarta CC, Whelan CJ, loid polyneuropathy. J Neuromusc Disord. 1996;6(S20).
Petrie A, et al. Natural history, quality of life, and outcome in car- 24. Hornstrup LS, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-
diac transthyretin amyloidosis. Circulation. 2019;140(1):16–26. Hansen A. Genetic stabilization of transthyretin, cerebrovascular
8.•• Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, disease, and life expectancy. Arterioscler Thromb Vasc Biol.
Waddington-Cruz M, et al. Tafamidis treatment for patients with 2013;33(6):1441–7.
transthyretin amyloid cardiomyopathy. N Engl J Med. 25. Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, et al.
2018;379(11):1007–16. This study is a major clinical trial dem- Efficacy and safety of patisiran for familial amyloidotic
onstrating the clinical efficacy of transthyretin stabilizing ther- polyneuropathy: a phase II multi-dose study. Orphanet J Rare Dis.
apies that improve clinical outcomes. 2015;10:109.
9. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck 26.• TS GD, De Los Santos J, Helmke S, Guadalupe S, Maurer M.
PJ, Wang AK, et al. Inotersen treatment for patients with hereditary Tafamidis increases serum TTR (prealbumin) levels in both
transthyretin amyloidosis. N Engl J Med. 2018;379(1):22–31. ATTRh and ATTRwt cardiac amyloidosis. Journal of Cardiac
10. Gillmore JD, Damy T, Fontana M, Hutchinson M, Lachmann HJ, Failure. 2019;25(8):S21. This study associates changes in
Martinez-Naharro A, et al. A new staging system for cardiac transthyretin levels with a response to therapy outlining the
transthyretin amyloidosis. Eur Heart J. 2018;39(30):2799–806. potential for transthyretin to serve as a surrogate for treatment
11. Grogan M, Scott CG, Kyle RA, Zeldenrust SR, Gertz MA, Lin G, efficacy.
et al. Natural history of wild-type transthyretin cardiac amyloidosis 27. Noy N, Li L, Abola MV, Berger NA. Is retinol binding protein 4 a
and risk stratification using a novel staging system. J Am Coll link between adiposity and cancer? Horm Mol Biol Clin Investig.
Cardiol. 2016;68(10):1014–20. 2015;23(2):39–46.
12. Hanson JLS, Arvanitis M, Koch CM, Berk JL, Ruberg FL, 28. Hyung SJ, Deroo S, Robinson CV. Retinol and retinol-binding pro-
Prokaeva T, et al. Use of serum transthyretin as a prognostic indi- tein stabilize transthyretin via formation of retinol transport com-
cator and predictor of outcome in cardiac amyloid disease associ- plex. ACS Chem Biol. 2010;5(12):1137–46.
ated with wild-type transthyretin. Circ Heart Fail. 2018;11(2): 29. Hamilton JA, Benson MD. Transthyretin: a review from a structural
e004000. perspective. Cell Mol Life Sci. 2001;58(10):1491–521.
13. Judge DP, Heitner SB, Falk RH, Maurer MS, Shah SJ, Witteles
30. Santos D, Coelho T, Alves-Ferreira M, Sequeiros J, Mendonca D,
RM, et al. Transthyretin stabilization by AG10 in symptomatic
Alonso I, et al. Variants in RBP4 and AR genes modulate age at
transthyretin amyloid cardiomyopathy. J Am Coll Cardiol.
onset in familial amyloid polyneuropathy (FAP ATTRV30M). Eur J
2019;74(3):285–95.
Hum Genet. 2016;24(5):756–60.
14. Ruberg FL, Maurer MS, Judge DP, Zeldenrust S, Skinner M, Kim
31.• Arvanitis M, Simon S, Chan G, Fine D, Beardsley P, La Valley M,
AY, et al. Prospective evaluation of the morbidity and mortality of
et al. Retinol binding protein 4 (RBP4) concentration identifies
wild-type and V122I mutant transthyretin amyloid cardiomyopa-
V122I transthyretin cardiac amyloidosis. Amyloid.
thy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am
2017;24(sup1):120–1 This study serves as an excellent review of
Heart J. 2012;164(2):222–8 e1.
the role of retinol binding protein 4 in ATTR and the associations of
15. Ingenbleek Y, De Visscher M. Hormonal and nutritional status:
retinol binding protein and clinical outcomes in ATTR.
critical conditions for endemic goiter epidemiology? Metabolism.
32. Arvanitis M, Koch CM, Chan GG, Torres-Arancivia C, LaValley
1979;28(1):9–19.
MP, Jacobson DR, et al. Identification of transthyretin cardiac am-
16. Dickson PW, Howlett GJ, Schreiber G. Metabolism of prealbumin
yloidosis using serum retinol-binding protein 4 and a clinical pre-
in rats and changes induced by acute inflammation. Eur J Biochem.
diction model. JAMA Cardiol. 2017;2(3):305–13.
1982;129(2):289–93.
17. Ingenbleek Y, Young V. Transthyretin (prealbumin) in health and 33. Sousa A, Coelho T, Barros J, Sequeiros J. Genetic epidemiology of
disease: nutritional implications. Annu Rev Nutr. 1994;14:495– familial amyloidotic polyneuropathy (FAP)-type I in Povoa do
533. Varzim and Vila do Conde (north of Portugal). Am J Med Genet.
18. Marcason W. Should albumin and prealbumin be used as indicators 1995;60(6):512–21.
for malnutrition? J Acad Nutr Diet. 2017;117(7):1144. 34. Rappley I, Monteiro C, Novais M, Baranczak A, Solis G, Wiseman
19. Berk J CL, Hankinson E, Falzone R, Figueroa Y, Hutabarat R, RL, et al. Quantification of transthyretin kinetic stability in human
Butler J, Kretschmer M, Sah D, Cehelsky J, Vaishnaw A, Gollob plasma using subunit exchange. Biochemistry. 2014;53(12):1993–
J. Serial measurements of circulating transthyretin (TTR) in sub- 2006.
jects with TTR amyloidosis or carriers of mutant TTR. Meeting of 35. Schonhoft JD, Monteiro C, Plate L, Eisele YS, Kelly JM, Boland D,
the Peripheral-Nerve-Society. 2011;16. et al. Peptide probes detect misfolded transthyretin oligomers in
20. Buxbaum J, Anan I, Suhr O. Serum transthyretin levels in Swedish plasma of hereditary amyloidosis patients. Sci Transl Med.
TTR V30M carriers. Amyloid. 2010;17(2):83–5. 2017;9(407).
21. Buxbaum J, Koziol J, Connors LH. Serum transthyretin levels in
senile systemic amyloidosis: effects of age, gender and ethnicity. Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
Amyloid. 2008;15(4):255–61. tional claims in published maps and institutional affiliations.

You might also like