Transthyretin-Related Amyloidosis

Back

Practice Essentials

Transthyretin (TTR) is a protein that functions as a transporter of thyroxine and retinol and is produced chiefly by the liver (> 95%), with additional production within the choroid plexus of the brain and the retinal pigment epithelium. However, it is also associated with the formation of amyloid fibrils, leading to TTR-related amyloidosis (ATTR), in which these fibril proteins are deposited into various organs and tissues, preferentially the nervous system and cardiac tissue, resulting in their inherent dysfunction.

Signs and symptoms

The presenting signs and symptoms in patients with ATTR are fairly nonspecific and often attributed to more common diseases affecting both the heart and the peripheral and autonomic nervous sytems.

Patients with cardiac deposition typically present with the following typical symptoms of chronic heart failure (CHF):

Neuropathic involvement in patients affected by ATTR–familial amyloid polyneuropathy (FAP) is classically a symmetric, ascending length−dependent, sensorimotor, axonal polyneuropathy subtype and may include the following:

Carpal ligament deposits: Weakness and paresthesias of one or both hands (eg, variant TTR L58H, normal-sequence TTR); localized symptomatic carpal ligament deposition sometimes precedes other clinical manifestations by as long as 20 years

Patients with rare TTR variants that cause CNS disease may present with the following features:

See Clinical Presentation for more detail.

Diagnosis

Physical examination findings in patients with ATTR depend on the organ involved, which is affected by the presence and genetic identity of a TTR variant. Symptoms consistent with HFpEF, along with concurrent peripheral/autonomic neuropathy, warrant consideration of ATTR as a diagnosis. A complete family history is of great value.

Biopsy

All types of amyloidosis are diagnosed definitively on the basis of demonstration of Congo red binding material in a biopsy or autopsy specimen. Subcutaneous fat aspiration often provides sufficient tissue for diagnosing amyloid, as well as for further studies (eg, immunostaining). Biopsy of an organ with impaired function (eg, heart, GI tract) can definitively establish a cause-and-effect relationship between organ dysfunction and amyloid deposition. See the image below.



View Image

Congo Red staining of a cardiac biopsy specimen containing amyloid, viewed under polarized light.

Laboratory results for different types of amyloidosis are generally nonspecific, including the following:

Other tests include electrocardiography, nerve conduction studies, and genetic studies (eg, polymerase chain reaction, electrospray ionization mass spectrometry, single-strand conformation polymorphism analysis and/or direct sequencing).

Imaging studies

See Workup for more detail.

Management

Patisiran and inotersen have been approved by the FDA for treatment of polyneuropathy caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adults. Several other medications are under investigation, but liver transplant remains the gold standard.

Diuretics are the mainstay of therapy for amyloid-related CHF, but must be used with caution due to the restrictive physiology involved.

Surgery

Depending on the organ and/or tissue involvement, surgical intervention for patients with ATTR may involve the following:

See Treatment for more detail.

Background

The amyloidoses are a wide range of diseases of secondary protein structure, in which a normally soluble protein forms insoluble extracellular fibril deposits, causing organ dysfunction. All types of amyloid contain a major fibril protein that defines the type of amyloid, plus minor components. Over 20 different fibril proteins have been described in human amyloidosis, each with a different clinical picture (see Amyloidosis, Overview). One such protein that forms human amyloid fibrils is transthyretin (TTR).

TTR acts as a transport protein for thyroxine in plasma. TRR also transports retinol (vitamin A) through its association with the retinol-binding protein. It circulates as a tetramer of four identical subunits of 127 amino acids each. TTR was once called prealbumin because it migrates anodally to albumin on serum protein electrophoresis, but this name was misleading, as TTR is not a precursor of albumin. The TTR monomer contains eight antiparallel beta pleated sheet domains.

TTR can be found in plasma and in cerebrospinal fluid and is synthesized primarily by the liver and the choroid plexus of the brain and, to a lesser degree, by the retina. Its gene is located on the long arm of chromosome 18 and contains 4 exons and 3 introns.[5]

The systemic amyloidoses are designated by a capital A (for amyloid) followed by the abbreviation for the chemical identity of the fibril protein. Thus, TTR amyloidosis is abbreviated ATTR.

Pathophysiology

Normal-sequence transthyretin-related amyloidosis

In contrast to variant ATTR, normal-sequence cardiac ATTR is associated with aging, usually developing in the seventh and eighth decades of life. This disorder is commonly of little or no clinical significance and only noted on autopsies in studies aiming at estimating its prevalance in an otherwise asymptomatic, aging population. In one autopsy study of people >85 years of age, ATTR was present in 25%.[6] The fraction of autopsied patients with clinically significant symptoms is not known.

The stimuli that lead to normal-sequence ATTR are not understood. Normal-sequence TTR forms cardiac amyloidosis predominantly in men above 60 years of age, a disorder termed senile cardiac amyloidosis (SCA). When it was recognized that SCA is often accompanied by microscopic deposits in many other organs, the alternative name senile systemic amyloidosis (SSA) was proposed. Both terms are now used.[5]  The clinical manifestations of severe SCA are similar to those observed in familial ATTR and in cardiac amyloidosis of the immunoglobulin light chain type (AL).

Mutant transthyretin-related amyloidosis

TTR mutations accelerate the process of TTR amyloid formation and are the most important risk factor for the development of clinically significant ATTR. More than 100 amyloidogenic TTR variants cause systemic familial amyloidosis. The age at symptom onset, pattern of organ involvement, and disease course vary, but most mutations are associated with cardiac and/or nerve involvement. The gastrointestinal tract, vitreous, lungs, and carpal ligament are also frequently affected.[5]

In a retrospective cross-sectional study of 284 ATTR and non-ATTR patients, the most common ATTR mutations were as follows[7] :

ATTR is caused by a single-point mutation, of which more than 100 have been described, that promotes destabilazation of the native quarternary structure into beta-pleated sheet predominant, insoluble and inactive form. This conformational change hypothesis has been researched in vitro with a key finding that tetramer dissociation is a required and generally rate-limiting step in amyloid fibril formation.

Energetic studies have suggested that amyloidogenic mutations destabilize the native quaternary and tertiary structures of TTR, thereby inducing conformational changes that lead to dissociation of the tetramers into partially unfolded species, which can subsequently self-assemble into amyloid fibrils. However, the wild-type (wt) TTR form can also result in amyloid deposits found in peripheral nerves and cardiac tissue in patients affected by the disease, usually in older patients. It is expected that the process of amyloid aggregation will be further elucidated in the future to address this and other concerns.[8]

When the peripheral nerves are prominently affected, the disease is termed familial amyloidotic polyneuropathy (FAP). When the heart is involved heavily but the nerves are not, the disease is called familial amyloid cardiomyopathy (FAC).

Epidemiology

United States

The most common amyloidosis-associated TTR variants in the United States are as follows:

Cardiac ATTR amyloidosis has a progressive increase in prevalence in people older than 80 years and is seen in about 15% of autopsies, with one study finding a prevalence of about 25%. In this setting, the deposited TTR is usually of normal sequence (wt-ATTR).

International

A few amyloidosis-associated TTR variants are common in certain populations, although few data indicate population frequencies. The most common TTR variants include the following:

Familial TTR variants

Most variants that cause familial ATTR are rare, but a few are common in certain populations. TTR variants are written, according to convention, by the normal amino acid found at a position in the mature protein, followed by the number of the amino acid from the amino terminal end, and the variant amino acid found, using either the three-letter or single-letter amino acid code. The most widely recognized TTR variants are as follows:

Currently, about 100 TTR variants are known, with varying geographic distributions, degrees of amyloidogenicity, and organ predisposition. Currently known TTR variants are listed in the table below.[5] For organ involvement, the following abbreviations are used: PN = peripheral nerves, AN = autonomic nervous system, H = heart, L = liver, LM = leptomeninges, K = kidney, S = skin, E = eye, GI = gastrointestinal tract, CL = carpal ligament, and CNS = central nervous system.

Known TTR Variants (adapted from Benson[13] and Connors et al[5] )



View Table

See Table

Expression of transthyretin-related amyloidosis

Familial ATTR was traditionally thought of as a group of autosomal dominant diseases, but it is now known that disease expression is more complicated. The most abundant data pertain to TTR V30M; the following observations have been made:

The explanation for the above observations is not well understood. Other genetic and/or environmental variables are thought to be at play. Anticipation, incomplete penetrance, and clinically sporadic cases in kindreds with unaffected allele carriers also have been observed with other TTR variants.[12]

Race

TTR variants occur in all races.

Sex

All TTR variants encoded on chromosome 18 are inherited with equal frequency in males and females. For unknown reasons, disease penetrance is greater and age of onset earlier in males than in females. Individual case reports and several small series suggest that normal-sequence cardiac ATTR is significantly more common in males than in females, although the sex ratio is unknown.[14]

Age

The age of onset varies widely, depending on the presence and identity of the TTR variant.

 

Mortality/Morbidity

Morbidity and mortality from ATTR depends on whether a TTR variant is present and, if so, which variant. Some variants cause clinical disease by age 40 years in all gene carriers and are always fatal within a few years of symptom onset. Other variants typically cause much milder, later onset disease, and some carriers of the variant genes remain asymptomatic until late in life.[15]

Central nervous system (CNS) complications are increasingly noted in liver-transplanted ATTR patients. Atrial fibrillation (AF), is a risk factor for ischemic CNS complications observed after liver transplantation.[16]

Morbidity depends on the organ(s) involved. Neuropathy and cardiomyopathy are most common. The most common immediate cause of death is cardiac failure or fatal arrhythmia.[17]

 

 

Prognosis

TTR-FAP usually proves fatal within 7–12 years from the onset of symptoms, most often due to cardiac dysfunction, infection, or cachexia.[18]  

Within most of the regions in which it is endemic, clinical onset of TTR-FAP often occurs before age 40 years with progressive sensory-motor and autonomic neuropathy, leading to cachexia and eventually death. Length-dependent small-fiber sensory and motor polyneuropathy with life-threatening autonomic dysfunction is a distinguishing feature of TTR-FAP in these areas. In addition, cardiac, renal, and ocular involvement are also common.[19]

In nonendemic areas, and in endemic regions of Sweden, the onset of disease-related symptoms tends to be later in life, from age 50 years onward and with a male predominance for the late-onset TTR-FAP. Neuropathy tends to affect all fibers and may closely resemble chronic inflammatory demyelinating polyneuropathy (CIDP). Typically, sensory and motor neuropathy symptoms of upper and lower extremities occur, associated with mild autonomic symptoms.[19]

Patient Education

N/A

History

The subtype of transthyretin (TTR) protein mutation, its tissue distribution, and the amount of amyloid deposition largely determine the clinical manifestations of TTR-related amyloidosis (ATTR). The key characteristic of ATTR that should raise clinical suspicion for this disorder remains the reliable coexistence of both cardiac and peripheral nervous system (PNS) involvement. This association will require the clinician to adequately interview the patient, who is likely to be presenting with a chief complaint related to one but not both organ systems.

Cardiovascular involvement

Patients with cardiac deposition often present with symptoms suggesting chronic heart failure (ie, dyspnea on exertion, peripheral edema, elevated jugular venous pressure, hepatojuglular reflux) and/or arrhythmias (ie, palpitations, lightheadedness, syncope).[17]

Deposition in the subendothelium of the peripheral vasculature can lead to severe postural hypotension.[17]

Neuropathic involvement

Peripheral nerve problems are the presenting complaints in most cases of ATTR, and can be reliably differentiated from other types of PNS disease by the fact that they are most often symmetric, distal polyneuropathies that typically begin in the lower limbs, progress to the upper limbs, and then affect more proximal aspects of the limbs and the trunk. A family history of a similar polyneuropathy is usually present and hence warrants a rigorous family history discussion as part of the history.

Patients with peripheral nerve deposits note sensorimotor impairment. While the majority present with bilateral, lower-to-upper extremity symptoms, as described above, some TTR variants present as lower-limb neuropathy (eg, TTR V30M), while other variants present as primarily upper-limb neuropathy (eg, TTR I84S, TTR L58H).[1]

Neuropathy in patients with ATTR V30M often presents as lower extremity weakness, pain, and/or impaired sensation. Autonomic dysfunction, often manifested as sexual or urinary dysfunction, is common.[2]

Other manifestations

Patients with gastrointestinal deposits present with alternating diarrhea and constipation. Nausea and vomiting also occur.

Weakness and paresthesias of one or both hands, suggesting carpal ligament involvement, is often the presenting symptom in patients with the variant TTR L58H. It can also be observed in patients with other variants. Carpal tunnel syndrome sometimes precedes other clinical manifestations by as much as 20 years. Patients with normal-sequence TTR also may develop localized symptomatic carpal ligament deposition.

Ophthalmological involvement may present as follows:

A study by Phull et al showed a high prevalence of coexistent monoclonal gammopathy of undetermined significance (MGUS) in patients with ATTR, with a rate higher than the general population.[20]

Physical Examination

As with the history, the physical findings depend on the organ involved, which is affected by the presence and identity of a TTR variant.

Common physical findings in advanced disease include the following:

Cardiac involvement typically results in the following[17] :

Neurologic findings

Typical findings include symmetric sensory impairment and weakness, sometimes accompanied by painless ulcers, similar to the picture in diabetic neuropathy. In the absence of treatment, the peripheral neuropathy is progressive, and motor nerve conduction velocity slowly decreases.[21] Other findings on neurologic examination may include the following:

Central nervous system findings may include the following:

Other systems

On ophthalmologic examination, amyloid deposits may be found in the corpus vitreum. This finding may be the most specific for hereditary transthyretin amyloidosis (as opposed to other systemic amyloidoses).

Cutaneous findings may include purpura, which results from the vascular fragility produced by amyloid deposition in the subendothelium of the small blood vessels.

Approach Considerations

Diagnostic tests for transthyretin (TTR) amyloidosis are listed in the table below (adapted from Ando et al[23] ).

Table. Diagnostic tests for transythyretin (TTR) amyloidosis



View Table

See Table

BSB = 1-Bromo-2,5-bis(3-carboxy-4-hydroxystyryl)benzene; ESI = electrospray ionization; FSB = 1-Fluoro-2,5-bis(3-carboxy-4hydroxystyryl)benzene; FT-ICR = Fourier transform ion cyclotron resonance; LC-MS/MS = liquid chromatography–tandem mass spectrometry; MALDI-TOF = matrix-assisted laser desorption/ionization time-of-flight; PCR = polymerase chain reaction; RFLP = restriction fragment length polymorphism; SELDI-TOF = surface enhanced laser desorption/ionization–TOF; SSCP = single-strand conformation polymorphism 

Laboratory Studies

Nonspecific findings found in different types of amyloidosis include the following:

Protein electrophoresis and serum free light chain measurement can be used to assess for coexisting monoclonal gammopathy of undetermined significance (MGUS).[20]

Biopsy

Amyloidosis (of all types) is diagnosed definitively based on demonstration of Congo red binding material in a biopsy specimen. (See the image below.)



View Image

Congo Red staining of a cardiac biopsy specimen containing amyloid, viewed under polarized light.

For many years, rectal biopsy was the favored procedure when systemic amyloidosis was suspected. Currently, the capillaries in subcutaneous fat are known to be involved often in TTR-related amyloidosis (ATTR) and in some other types of systemic amyloidosis; therefore, subcutaneous fat aspiration often provides sufficient tissue for diagnosing amyloid, as well as for further studies such as immunostaining. On the other hand, biopsy of an organ with impaired function, such as the heart or gastrointestinal tract, has the advantage of definitively establishing a cause-and-effect relationship between organ dysfunction and amyloid deposition.

The sensitivity of detecting ATTR varies by site, as follows[18] :

  1. Labial salivary gland biopsy: 91%
  2. Sural nerve biopsy: 80%
  3. Abdominal fat pad biopsy: 14-83%

ATTR deposition in the peripheral nerves leads to axonal degeneration of the small nerve fibers, causing polyneuropathy. Diagnosis can often be made with sural nerve biopsy, although the deposits may be proximal to the sural nerve and therefore not found in biopsy samples.

Other potential biopsy sites include the following:

Amyloid should not be assumed to be of the TTR type based solely on the Congo red staining and clinical picture. After Congo red staining establishes a diagnosis of amyloidosis, the specific type of amyloidosis must be determined with immunostaining of a biopsy specimen using commercially available antiserum against TTR. Control antisera against other types of amyloid precursors, including immunoglobulin light chains and amyloid A protein, should also be performed to confirm staining specificity. Even patients known to carry a TTR variant should ideally have the diagnosis confirmed with immunostaining to rule out the possibility of a different type of amyloidosis.

Distinguishing between ATTR and AL cardiac amyloidosis on clinical grounds alone is particularly difficult. Without immunologic identification of the deposited protein, an incorrect diagnosis of ATTR in a patient with AL, or the reverse, could lead to ineffective or harmful treatment. Mass spectroscopy can also be used to determine the protein subunit and classify the disease as immunoglobulin light-chain amyloidosis or ATTR.[25]

Cardiac Imaging

Cardiac deposition is, in many patients, the most serious complication of ATTR. For that reason, cardiac involvement usually should be assessed and monitored by imaging studies. Echocardiograms, cardiac magnetic resonance imaging (MRI), and scintigraphy with bone tracers can all help to diagnose infiltrative cardiomyopathy.[18, 26]

Echocardiography enables visualization of increased ventricular wall thickness, increased septal thickness, and an appearance of granular "sparkling." This finding is neither sensitive nor specific enough to be diagnostic but is highly suggestive when present.

Amyloid deposits in the heart occur in the ventricular interstitium, leading to thickening of the ventricular walls and interventricular septum without an increase in the intracardiac volume. Evaluation of diastolic function by Doppler echocardiography reveals impaired ventricular relaxation early in the course of disease, which progresses to short deceleration. The ejection fraction is preserved until late in disease.[25]

Other echocardiographic findings include the following:

Bone scintigraphy using technetium-labelled radiotracers provides very high diagnostic accuracy in the noninvasive assessment of cardiac ATTR.[27] 99mTc-DPD–based single-photon emission computed tomography (SPECT) imaging enables noninvasive diagnosis of cardiac ATTR amyloidosis, particularly in patients rejecting biopsy.

 

Other Tests

Other cardiac testing

The classic finding on electrocardiography is a low-voltage QRS complex in the limb leads, resulting from replacement of normal cardiac tissue by nonconducting amyloid material. In some cases, loss of anterior forces suggests anteroseptal infarction that is not confirmed at autopsy. Various arrhythmias are observed and can be life threatening.[17]

The prevalence of low QRS voltages at the time of diagnosis has been found to be lower than in light chain amyloidosis (AL), despite the finding, in some studies, of greater myocardial infiltration in TTR-related forms.[28]

Holter monitoring and intracardiac electrophysiology study are helpful to detect conduction disorders.[18]

Nerve conduction studies

In patients with amyloid neuropathy, serial nerve conduction studies can be useful for objectively monitoring the course of disease and for assessing response to treatment such as liver transplantation.[29]

Genetic studies

In patients with progressive, length-dependent axonal neuropathy predominantly involving small nerve fibers, genetic testing for TTR gene mutations should be performed during the initial diagnostic workup, to prevent serious consequences from delayed diagnosis.[30] Genetic studies to look for a TTR variant can be helpful in many patients with ATTR, particularly in younger patients not known to belong to a kindred carrying a defined TTR variant. These studies generally are not available through routine clinical laboratories.

One approach is to perform polymerase chain reaction (PCR) testing to look for known, common TTR variants. This approach is most useful if the likely TTR variant can be surmised on the basis of the clinical history and genetic background of the patient. These studies are performed by PCR amplification of regions of the TTR gene followed by digestion with restriction enzymes.

If a TTR variant is suspected but initial screening results for a few common known variants are negative, more comprehensive analysis for a TTR variant can be performed. Either the protein can be isolated from the serum and studied using methods such as electrospray ionization mass spectrometry (ESIMS) or the gene can be studied by PCR and such methods as single-strand conformation polymorphism analysis and/or direct sequencing.

Determination of whether a TTR variant is present is important because the treatment options for variant-sequence ATTR differ from those for normal-sequence ATTR. Information about a TTR variant also can be of use to other family members at risk. 

Ophthalmological assessment

Ophthalmological assessment is warranted to identify any ocular manifestations of TTR-FAP, which may include the following:

Radiolabeled P-component scanning

Radiolabeled P-component scanning is available in a few European centers. Where it is available, radiolabeled P-component scanning is a very useful means of evaluating the total body burden of amyloid and is a sensitive noninvasive means of diagnosing amyloid in most organs. Serial studies are useful for monitoring the response to therapy in many settings.

One drawback of P-component scanning is that it is not useful for diagnosing or monitoring cardiac amyloid, because the concentration of label in the intracardiac blood pool obscures the weaker signal from the labeled molecule bound to myocardial amyloid.

Histologic Findings

Biopsy of an affected organ followed by routine hematoxylin and eosin staining reveals homogeneous interstitial eosinophilic material. Amyloid material stained with Congo red and viewed under polarized light appears bright green. Specific staining with antibodies against TTR proves the diagnosis of ATTR, as opposed to other types of amyloidosis that have similar appearance after hematoxylin and eosin or Congo red staining.

Staging

Once the diagnosis has been made, the neuropathy stage and systemic extension of the disease should be determined to direct the course of management. The three stages of ATTR-FAP severity are graded according to the patient’s walking disability and degree of assistance required, as follows[18, 19] :

Stage 0: Asymptomatic carrier of a known ATTR mutation

Stage I: Sensory polyneuropathy; preserved walking capacity without the need for a walking stick

Stage II: Progressive walking disability; ambulatory, but requires assistance, one to two walking sticks or crutches required

Stage III: Wheelchair bound or bedridden

 

Approach Considerations

Transthyretin-related amyloidosis (ATTR) involves many organs and systems, so an interdisciplinary approach is essential for the management of comorbidities. Patisiran and inotersen are approved by the US Food and Drug Administration (FDA) for treatment of polyneuropathy caused by hereditary transthyretin-related amyloidosis (hATTR) in adults. Several other medications are under investigation, but liver transplantation remains the gold standard for therapy. Ideally, patients should be referred while early in stage I for liver transplantation—or possibly multi-organ transplantation, depending on heart or kidney involvement.  

Medical Care

The US Food and Drug Administration (FDA) has approved patisiran (Onpattro) and inotersen (Tegsedi) for treatment of polyneuropathy caused by hATTR in adults. Tafamidis, diflunisal, revusiran, and tolcapone remain under investigation.

Patisiran

Patisiran utilizes RNA interference, a cellular process in which small interfering RNAs (siRNAs) control gene expression by mediating the cleavage of specific messenger RNAs (mRNAs).[31]  Patisiran comprises siRNAs that are specific for TTR mRNA, formulated in lipid nanoparticles. Administration is via intravenous infusion every 3 weeks.

Approval was based on the APOLLO clinical trial, in which patients taking patisiran (n=148) showed significantly improved scores on the Neuropathy Impairment Score+7 and Norfolk Quality of Life Questionnaire–Diabetic Neuropathy (QOL-DN) at 18 months, compared with those taking placebo (n=77) (P <  0.001).[32]

Inotersen

Inotersen was approved by the FDA in October 2018. Like patisiran, it is indicated for polyneuropathy of hATTR in adults; unlike patisiran, inotersen is given as a once-weekly subcutaneous injection that the patient or caregiver can administer. It is an antisense oligonucleotide that causes degradation of mutant and wild-type transthyretin mRNA by binding TTR mRNA. This action results in reduced TTR protein levels in serum and tissue.

Approval was based on an international, randomized, double-blind, placebo-controlled phase III trial (NEURO-TTR) in which patients with stage 1 or 2 hATTR with polyneuropathy (n=172) were randomly assigned in a 2:1 ratio to receive weekly inotersen or placebo. Scores on the mNIS+7 and the QOL-DN showed improvement in those receiving inotersen (P <  0.001).[33]

Tafamidis

Tafamidis, or 2-(3,5-dichloro-phenyl)-benzoxazole-6-carboxylic acid, selectively binds to TTR with negative cooperativity and kinetically stabilizes wild-type native TTR and mutant TTR. Therefore, tafamidis has the potential to halt the amyloidogenic cascade initiated by TTR tetramer dissociation, monomer misfolding, and aggregation.[34]  Early intervention with tafamidis led to minimal disease progression over 5.5 years in patients with mild ATTR-FAP.[35]

A tafamidis trial in patients with stage I neuropathic ATTR (mobilization without need for support) failed to achieve statistical significance for its primary endpoints of neurological deterioration and quality of life. However, because all measured endpoints indicated that the drug decreased the rate of disease progression, tafamidis was approved by the European Medical Agency in 2011 for patients in stage I of neuropathic ATTR.[36] Since 2011, tafamidis has been approved for use in Japan, Mexico, and Argentina, where it is used as a first-line treatment option for patients with early-stage ATTR–familial amyloid polyneuropathy (FAP).

Diflunisal

Diflunisal is a nonsteroidal anti-inflammatory drug that is FDA approved for treatment of arthritis. At a dosage of 250 mg twice daily, diflunisal successfully complexes to the thyroxine binding site and kinetically stabilizes circulating TTR tetramers, inhibiting release of the TTR monomer required for amyloidogenesis. In a randomized, placebo-controlled trial in patients with stage I-II ATTR-FAP, diflunisal improved quality of life scores and reduced progression of neurological impairment compared with placebo. Its use for this indication remains off-label.[37, 38]

Tolcapone

Tolcapone is FDA approved for treatment of Parkinson disease and has Orphan Drug designation for treatment of ATTR. Tolcapone occupies the T4-binding sites located at the TTR dimer-dimer interface and prevents amyloidoenesis by stabilizing the teramer in vivo in mice and humans.[39]  In addition, an added benefit is it also inhibits TTR cytotoxicity. It has been shown that tolcapone docks better than tafamidis in wt-TTR.

Treatment of cardiac involvement

Diuretic agents must be used with caution. Although diuretics are commonly prescribed for patients with heart failure, their use in amyloidosis is complicated. Due to the restrictive effect of the disease, ventricular compliance is poor and end-diastolic volumes are low. Patients often require a higher filling pressure to distend the stiffened heart, and diuretic therapy reduces preload, which can further reduce stroke volume and systolic blood pressure.[22]

Beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, and angiotensin receptor blockers (ARBs) are poorly tolerated in cardiac amyloidosis and should be avoided. Digoxin binds to amyloid fibrils and can lead to locally high levels; it also must be used with caution.[22]

Given the high incidence of sudden death in patients with TTR cardiac amyloidosis, it is prudent to consider prophylactic placement of an implantable cardioverter defibrillator (ICD).[22]

Surgical Care

Liver transplantation

Prior to 1990, no therapy for TTR-FAP was available. Liver transplantation was first performed for FAP in 1990 and to date more than 2100 liver transplants have been reported to the Familial Amyloid Polyneuropathy World Transplant Registry (FAPWTR).[40] Transplantation replaces the main source of variant TTR with a source of normal-sequence TTR, sometimes leading to gradual fibril reabsorption and disease stabilization, especially of neurologic complications. Liver transplantation seems to halt progression of sensory, motor, and autonomic neuropathy. Ideally, the transplantation should be performed as early in the disease course as possible, before significant neurologic disability has been incurred.[41]

Cardiac, leptomeningeal, gastrointestinal, or ocular involvement often progresses despite transplantation.

Overall 20-year survival after transplantation, all mutations included, was 55.3%. The expected mortality rate decreased on average by approximately 4% per year between 1990 and 2010. Improved survival in TTR Val30Met patients was most pronounced during the first 5-year period, whereas non-TTR Val30Met patient survival improved throughout the 20-year period. The natural history of the disease has a 10-15 year prognosis.[41]

Combination heart and liver or liver and kidney transplantation has been performed in select patients, with variable success, and an 18.1% rate of postoperative cardiac complications has been shown with heart transplantations. Patients undergoing combined transplantation were generally older than those only being treated with liver transplant for TTR amyloidosis and more likely carrying a non-TTRVal30Met mutation.[41]

Carpal tunnel release

Involvement of the carpal ligament is observed not only in ATTR but also, most commonly, in patients undergoing dialysis and in patients with light chain amyloidosis (AL). Treatment is surgical release.

At the time of carpal tunnel release, a biopsy should be performed if a definitive diagnosis has not been established previously so that both Congo red staining and immunostaining can be performed. Why the carpal ligament, or indeed any organ, is a favored location for amyloid deposition is not known.

Vitrectomy

Vitrectomy is useful in patients with vitreous involvement. TTR is known to be produced locally by retinal pigment epithelial and ciliary pigment epithelium cells. The progression of ocular disease after liver transplantation suggests that continued intra-ocular TTR production is relevant in this context. In a review of 513 cases, no differences were found in ocular tests between patients who received liver transplants and nontransplanted patients.[42]

Diet

There is no specific diet for ATTR. A small observational study of 24 men with wt-ATTR cardiomyopathy demonstrated that consumption of green tea extract for 1 year may potentially inhibit amyloid fibril formation in the heart.[43] Patients with associated heart disease can also benefit from a low-sodium diet, and may wish to review American Heart Association recommendations on reducing dietary sodium.

Complications

Central nervous system (CNS) complications are increasingly noted in liver-transplanted ATTR patients. Atrial fibrillation (AF) is a risk factor for ischemic CNS complications observed after liver transplantation.[16]

Consultations

Once the diagnosis of ATTR has been made, a multi-disciplinary approach with the following consultations is advised:

 

Activity

Since polyneuropathy (FAP) is a major constellation of symptoms in ATTR, a loss of function is a trigger for liver transplantation. Early involvement of physical therapy to detect subtle changes in function would be helpful.

Prevention

There are no known primary preventive measures. Once the diagnosis has been made, medical and surgical treatments serve as secondary prevention, and supportive care for complications serve as tertiary prevention.

Long-Term Monitoring

For cardiac follow-up, monitor New York Heart Association (NYHA) class and electrocardiographic (ECG) changes in order to mitigate symptoms and determine the need for ICD placement and possibly accompanying heart transplantation in select cases if liver transplant is indicated. Early detection of cardiac abnormalities is important; the prophylactic implantation of pacemakers was found to prevent 25% of major cardiac events in TTR-FAP patients followed up over an average of 4 years.[44]

For ATTR-FAP, liver transplantation should be considered while the patient is still in stage I FAP.

Nephrologic follow-up involves monitoring for microalbuminuria and possibly nephrotic-range proteinuria, as patients may progress to end-stage renal disease .

Ophthalmological monitoring recommendations, which are the same for liver transplant recipients and non-transplanted patients, set out the following schedule for eye examinations[42] :

 

Medication Summary

Despite recent drug approvals, liver transplantation remains the gold standard for treating transthyretin-related amyloidosis (ATTR). Multi-organ transplantation (heart, liver and kidney) has been successful in slowing the natural course of the disease. In 2018, the US Food and Drug Administration (FDA) approved patisiran and inotersen for treatment of polyneuropathy caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adults.[32, 33] In Europe, tafamidis is approved for stage I FAP. Research continues to identify and refine effective medical treatments for preventing the deposition of amyloid fibrils.

Cautious, closely monitored medical treatment for ATTR cardiomyopathy and consideration of implantable cardioverter-defibrillator (ICD) placement are required, as the restrictive pathophysiology of the heart failure differs from the more common hypertrophic or dilated varieties and standard treatments such as beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), or diuretics could worsen symptoms. Digoxin can increase local amyloid fibril levels in the heart[22]

Patisiran (Onpattro)

Clinical Context:  Patisiran is an siRNA agent that reduces serum transthyretin (TTR) protein and TTR protein deposits in tissues. It is indicated for treatment of polyneuropathy of hereditary transthyretin-mediated amyloidosis (hATTR) in adults.

Class Summary

Anti-transthyretin small interfering ribonucleic acid (siRNA) agents causes degradation of mutant and wild-type TTR mRNA through RNA interference.

Inotersen (Tegsedi)

Clinical Context:  Inotersen is an antisense oligonucleotide that causes degradation of mutant and wild-type transthyretin mRNA by binding TTR mRNA. This action results in reduced TTR protein in serum and tissue. It is indicated for polyneuropathy of hATTR in adults.

Class Summary

In a clinical trial, inotersen, an antisense oligonucleotide, has improved neurological scores compared with placebo.[33]

Complications

Complications of transthyretin-related amyloidosis (ATTR) include the following:

Prognosis

The natural course of TTR-FAP can be classified into the following three stages:

Life expectancy ranges from 7.3 to 11 years from onset.[19] The prognosis depends on the presence and identity of a TTR variant and organ(s) involved. Patients with early-onset of variant-sequence TTR may die within a few years of diagnosis. Older patients with slowly progressive disease can live for decades after the onset of symptoms and may never develop life-threatening disease.[15]

Penetrance of the individual ATTR mutations vary. The penetrance of the same mutation in different geographic areas can also vary, for example, the Portuguese population showing much higher penetrance of the Val30Met mutation during middle age (80% at 50 years) compared with the French population (18% at 50 years).[19]

In contrast to light chain amyloidosis (AL), symptomatic cardiac involvement in ATTR does not necessarily portend a poor prognosis. Median survival in cardiac AL is about 6 months, but is several years in older patients with cardiac ATTR, even in those with a TTR variant.

What is transthyretin-related amyloidosis (ATTR)?What are the cardiac signs and symptoms of transthyretin-related amyloidosis (ATTR)?What are the neuropathic signs and symptoms of transthyretin-related amyloidosis (ATTR)?What are the signs and symptoms of CNS disease in transthyretin-related amyloidosis (ATTR)?How is transthyretin-related amyloidosis (ATTR) diagnosed?What is the role of lab testing in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of imaging studies in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of medications in the treatment of transthyretin-related amyloidosis (ATTR)?Which surgical interventions are used in the treatment of transthyretin-related amyloidosis (ATTR)?What is transthyretin (TTR) and how does it cause amyloidosis?What is the pathophysiology of normal-sequence transthyretin-related amyloidosis (ATTR)?What is the pathophysiology of mutant transthyretin-related amyloidosis (ATTR)?What is the US prevalence of transthyretin-related amyloidosis (ATTR)?What is the global prevalence of transthyretin-related amyloidosis (ATTR)?What are the most common variants of familial transthyretin-related amyloidosis (ATTR)?What is known about familial transthyretin-related amyloidosis (ATTR) variant TTR B30M?What are the racial predilections of transthyretin-related amyloidosis (ATTR)?What are the sexual predilections of transthyretin-related amyloidosis (ATTR)?How does the prevalence of transthyretin-related amyloidosis (ATTR) vary by age?What is the mortality and morbidity associated with transthyretin-related amyloidosis (ATTR)?What is the prognosis of transthyretin-related amyloidosis (ATTR)?Which clinical history findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which cardiac findings suggest transthyretin-related amyloidosis (ATTR)?What is the neuropathic involvement characteristic of transthyretin-related amyloidosis (ATTR)?What are the signs and symptoms of GI involvement in transthyretin-related amyloidosis (ATTR)?Which signs and symptoms suggest carpal ligament involvement in transthyretin-related amyloidosis (ATTR)?What are the signs and symptoms of ophthalmologic involvement in transthyretin-related amyloidosis (ATTR)?Which hematologic condition is associated with transthyretin-related amyloidosis (ATTR)?Which physical findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which cardiac exam findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which neurologic exam findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which ophthalmologic exam findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which cutaneous findings are characteristic of transthyretin-related amyloidosis (ATTR)?Which conditions are included in the differential diagnoses of transthyretin-related amyloidosis (ATTR)?What are the differential diagnoses for Transthyretin-Related Amyloidosis?What are the diagnostic tests for transthyretin-related amyloidosis (ATTR)?Which lab test findings are characteristic of transthyretin-related amyloidosis (ATTR)?What is the role of biopsy in the diagnosis of transthyretin-related amyloidosis (ATTR)?How does the sensitivity of transthyretin-related amyloidosis (ATTR) vary among biopsy sites?What is the role of Congo red staining in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of cardiac imaging in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of bone scintigraphy in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of cardiac testing in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of nerve conduction studies in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of genetic studies in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of ophthalmological assessment in the workup of transthyretin-related amyloidosis (ATTR)?What is the role of radiolabeled P-component scanning in the workup of transthyretin-related amyloidosis (ATTR)?Which histologic findings are characteristic of transthyretin-related amyloidosis (ATTR)?How is transthyretin-related amyloidosis (ATTR) staged?How is transthyretin-related amyloidosis (ATTR) treated?Which medications are FDA-approved for the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of patisiran in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of inotersen in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of tafamidis in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of diflunisal in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of tolcapone in the treatment of transthyretin-related amyloidosis (ATTR)?How are cardiac symptoms of transthyretin-related amyloidosis (ATTR) treated?What is the role of liver transplantation in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of carpal tunnel release surgery in the treatment of transthyretin-related amyloidosis (ATTR)?What is the role of vitrectomy in the treatment of transthyretin-related amyloidosis (ATTR)?Which dietary modifications are used in the treatment of transthyretin-related amyloidosis (ATTR)?What are the possible complications of transthyretin-related amyloidosis (ATTR) after treatment?Which specialist consultations are beneficial to patients with transthyretin-related amyloidosis (ATTR)?Which activity modifications are used in the treatment of transthyretin-related amyloidosis (ATTR)?How is transthyretin-related amyloidosis (ATTR) prevented?What is included in the long-term monitoring of transthyretin-related amyloidosis (ATTR)?What is the schedule for long-term ocular monitoring of patients with transthyretin-related amyloidosis (ATTR)?What is the role of medications in the treatment of transthyretin-related amyloidosis (ATTR)?Which medications in the drug class Antisense Oligonucleotides are used in the treatment of Transthyretin-Related Amyloidosis?Which medications in the drug class siRNA Agents are used in the treatment of Transthyretin-Related Amyloidosis?What are the possible complications of transthyretin-related amyloidosis (ATTR)?What is the disease course of transthyretin-related amyloidosis (ATTR)?What is the prognosis of transthyretin-related amyloidosis (ATTR)?

Author

Jefferson R Roberts, MD, Chief of Rheumatology Service, Tripler Army Medical Center; Assistant Clinical Professor of Medicine, Uniformed Services University of the Health Sciences

Disclosure: Nothing to disclose.

Coauthor(s)

Mary L Lan, MD, MPH, Resident Physician, Department of Medicine, Tripler Army Medical Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Lawrence H Brent, MD, Associate Professor of Medicine, Sidney Kimmel Medical College of Thomas Jefferson University; Chair, Program Director, Department of Medicine, Division of Rheumatology, Albert Einstein Medical Center

Disclosure: Stock ownership for: Johnson & Johnson.

Chief Editor

Emmanuel C Besa, MD, Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Disclosure: Nothing to disclose.

Additional Contributors

,

Disclosure: Nothing to disclose.

Aaron Pumerantz, DO, Fellow Instructor, Department of Medicine, Uniformed Services University of the Health Sciences

Disclosure: Nothing to disclose.

Gregory M Sprowl, MD, Resident Physician, Department of Internal Medicine, Tripler Army Medical Center

Disclosure: Nothing to disclose.

Robert E Wolf, MD, PhD, Professor Emeritus, Department of Medicine, Louisiana State University School of Medicine in Shreveport; Chief, Rheumatology Section, Medical Service, Overton Brooks Veterans Affairs Medical Center

Disclosure: Nothing to disclose.

Acknowledgements

Daniel R Jacobson, MD Professor of Medicine, Boston University School of Medicine; Chief of Oncology, Veterans Affairs Boston Healthcare System

Disclosure: Nothing to disclose.

Mariana J Kaplan, MD Assistant Professor, Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School

Disclosure: Nothing to disclose.

Seetha U Monrad, MD Clinical Instructor, Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School

Seetha U Monrad is a member of the following medical societies: American College of Rheumatology

Disclosure: Nothing to disclose.

Robert John Oglesby, MD Chief of Rheumatology Service, Department of Medicine, Walter Reed Army Medical Center; Associate Professor of Medicine, Uniformed Services University of the Health Sciences

Robert John Oglesby, MD is a member of the following medical societies: American College of Physicians, American College of Rheumatology, and Arthritis Foundation

Disclosure: Nothing to disclose.

References

  1. Ando Y, Nakamura M, Araki S. Transthyretin-related familial amyloidotic polyneuropathy. Arch Neurol. 2005 Jul. 62(7):1057-62. [View Abstract]
  2. Ando Y, Suhr OB. Autonomic dysfunction in familial amyloidotic polyneuropathy (FAP). Amyloid. 1998 Dec. 5(4):288-300. [View Abstract]
  3. Plante-Bordeneuve V, Lalu T, Misrahi M, et al. Genotypic-phenotypic variations in a series of 65 patients with familial amyloid polyneuropathy. Neurology. 1998 Sep. 51(3):708-14. [View Abstract]
  4. Hagiwara K, Ochi H, Suzuki S, et al. Highly selective leptomeningeal amyloidosis with transthyretin variant Ala25Thr. Neurology. April 2009. 72:1358-60. [View Abstract]
  5. Connors LH, Lim A, Prokaeva T, Roskens VA, Costello CE. Tabulation of human transthyretin (TTR) variants, 2003. Amyloid. 2003 Sep. 10(3):160-84. [View Abstract]
  6. Tanskanen M, Peuralinna T, Polvikoski T, Notkola IL, Sulkava R, Hardy J, et al. Senile systemic amyloidosis affects 25% of the very aged and associates with genetic variation in alpha2-macroglobulin and tau: a population-based autopsy study. Ann Med. 2008. 40 (3):232-9. [View Abstract]
  7. Zhen DB, Swiecicki PL, Zeldenrust SR, Dispenzieri A, Mauermann ML, Gertz MA. Frequencies and geographic distributions of genetic mutations in transthyretin- and non-transthyretin-related familial amyloidosis. Clin Genet. 2015 Oct. 88 (4):396-400. [View Abstract]
  8. Bulawa C, Connelly S, DeVit M, et al. Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. PNAS. June 12, 2012. 109:9629-9634. [View Abstract]
  9. Suhr OB, Svendsen IH, Andersson R, Danielsson A, Holmgren G, Ranløv PJ. Hereditary transthyretin amyloidosis from a Scandinavian perspective. J Intern Med. 2003 Sep. 254(3):225-35. [View Abstract]
  10. Jacobson DR, Pastore RD, Yaghoubian R, et al. Variant-sequence transthyretin (isoleucine 122) in late-onset cardiac amyloidosis in black Americans. N Engl J Med. 1997 Feb 13. 336(7):466-73. [View Abstract]
  11. Saraiva MJ, Birken S, Costa PP. Amyloid fibril protein in familial amyloidotic polyneuropathy, Portuguese type. Definition of molecular abnormality in transthyretin (prealbumin). J Clin Invest. 1984 Jul. 74(1):104-19. [View Abstract]
  12. Jacobson DR, Buxbaum JN. Genetic aspects of amyloidosis. Adv Hum Genet. 1991. 20:69-123, 309-11. [View Abstract]
  13. Benson MD. The hereditary amyloidoses. Best Pract Res Clin Rheumatol. December 2003. Volume 17, Issue 6:909–927. [View Abstract]
  14. Rapezzi C, Riva L, Quarta CC, Perugini E, Salvi F, Longhi S. Gender-related risk of myocardial involvement in systemic amyloidosis. Amyloid. 2008 Mar. 15(1):40-8. [View Abstract]
  15. Adams D, Samuel D, Goulon-Goeau C, et al. The course and prognostic factors of familial amyloid polyneuropathy after liver transplantation. Brain. 2000 Jul. 123 (Pt 7):1495-504. [View Abstract]
  16. Wange N, Anan I, Ericzon BG, Pennlert J, Pilebro B, Suhr OB, et al. Atrial Fibrillation and Central Nervous Complications in Liver Transplanted Hereditary Transthyretin Amyloidosis Patients. Transplantation. 2018 Feb. 102(2):e59-e66. [View Abstract]
  17. Rapezzi C, Perugini E, Salvi F, Grigioni F, Riva L, Cooke RM, et al. Phenotypic and genotypic heterogeneity in transthyretin-related cardiac amyloidosis: towards tailoring of therapeutic strategies?. Amyloid. 2006 Sep. 13(3):143-53. [View Abstract]
  18. Adams D, Suhr OB, Hund E, Obici L, et al. First European consensus for diagnosis, management, and treatment of transthyretin familial amyloid polyneuropathy. Curr Opin Neurol. 2016. 29 (suppl1):S000-S000. [View Abstract]
  19. Parman Y, Adams D, Obici L, et al. Sixty years of transthyretin familial amyloid polyneuropathy (TTR-FAP) in Europe: where are we now? A European network approach to defining the epidemiology and management patterns for TTR-FAP. Curr Opin Neurol. 2016. 29 (suppl 1):S000–S000. [View Abstract]
  20. Phull P, Sanchorawala V, Connors LH, Doros G, Ruberg FL, Berk JL, et al. Monoclonal gammopathy of undetermined significance in systemic transthyretin amyloidosis (ATTR). Amyloid. 2018 Mar. 25(1):62-67. [View Abstract]
  21. Simmons Z, Specht, C. The Neuromuscular Manifestations of Amyloidosis. Journal of Clin Neuromuscular Dis. March 2010. 11:145-157. [View Abstract]
  22. Gertz MA, Benson MD, Dyck PJ, et al. Diagnosis, Prognosis, and Therapy of Transthyretin Amyloidosis. J Am Coll Cardiol. 2015. 66:2451-66. [View Abstract]
  23. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet Journal of Rare Diseases. 2013. v.8, 31:[View Abstract]
  24. Klaassen SHC, Tromp J, Nienhuis HLA, van der Meer P, van den Berg MP, Blokzijl H, et al. Frequency of and Prognostic Significance of Cardiac Involvement at Presentation in Hereditary Transthyretin-Derived Amyloidosis and the Value of N-Terminal Pro-B-Type Natriuretic Peptide. Am J Cardiol. 2018 Jan 1. 121(1):107-112. [View Abstract]
  25. Gertz MA, Dispenzieri A, Sher T. Pathophysiology and treatment of cardiac amyloidosis. Nat Rev Cardiol. 2015 Feb. 12 (2):91-102. [View Abstract]
  26. Gillmore JD, Maurer MS, Falk RH, Merlini G, Damy T, Dispenzieri A, et al. Nonbiopsy Diagnosis of Cardiac Transthyretin Amyloidosis. Circulation. June 14, 2016. Volume 133, Issue 24:2404-2412. [View Abstract]
  27. Treglia G, Glaudemans AWJM, Bertagna F, Hazenberg BPC, Erba PA, Giubbini R, et al. Diagnostic accuracy of bone scintigraphy in the assessment of cardiac transthyretin-related amyloidosis: a bivariate meta-analysis. Eur J Nucl Med Mol Imaging. 2018 Oct. 45(11):1945-1955. [View Abstract]
  28. Rapezzi C, Merlini G, Quarta, C, et al. Systemic Cardiac Amyloidosis: Disease Profiles and Clnical Courses of the 3 Main Types. Circulation. Sep 2009. 120:1203-1212. [View Abstract]
  29. Montagna P, Marchello L, Plasmati R, et al. Electromyographic findings in transthyretin (TTR)-related familial amyloid polyneuropathy (FAP). Electroencephalogr Clin Neurophysiol. 1996 Oct. 101(5):423-30. [View Abstract]
  30. Planté-Bordeneuve V, Ferreira A, et al. Diagnostic pitfalls in sporadic transthyretin familial amyloid polyneuropathy. Neurology. Aug 2007. 69:693-698. [View Abstract]
  31. Hawkins, P. N., Ando, Y., Dispenzeri, A., et al. Evolving landscape in the management of transthyretin amyloidosis. Annals of Medicine. 2015 Nov 17. 47(8):625–638. [View Abstract]
  32. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N Engl J Med. 2018 Jul 5. 379 (1):11-21. [View Abstract]
  33. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK1, et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. N Engl J Med. 2018 Jul 5;. 379(1):22-31. [View Abstract]
  34. Cruz MW, Benson MD. A Review of Tafamidis for the Treatment of Transthyretin-Related Amyloidosis. Neurol Ther. 2015. 4:61-79. [View Abstract]
  35. Waddington Cruz M, Amass L, Keohane D, Schwartz J, Li H, Gundapaneni B. Early intervention with tafamidis provides long- term (5.5-year) delay of neurologic progression in transthyretin hereditary amyloid polyneuropathy. Amyloid. 05 Aug 2016. Vol. 23 , Iss. 3:178-183. [View Abstract]
  36. Suhr OB, Larsson M, Ericzon B-G, Wilczek HE. Survival After Transplantation in Patients With Mutations Other Than Val30Met: Extracts From the FAP World Transplant Registry. Transplantation. 2015. 00:1-9. [View Abstract]
  37. Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013 Dec 25. 310(24):2658-67. [View Abstract]
  38. Anderson P. NSAID halts progression in familial amyloid polyneuropathy. December 30, 2013. Medscape Medical News. Available at http://www.medscape.com/viewarticle/818483. Accessed: January 10, 2017.
  39. Sant’Anna, R., Gallego, P., Robinson, L. Z., et al. Repositioning tolcapone as a potent inhibitor of transthyretin amyloidogenesis and associated cellular toxicity. Nature Communications. 2016 Feb 23. 7:[View Abstract]
  40. The Familial Amyloidotic Polyneuropathy World Transplant Registry. Available at http://www.fapwtr.org. February 29, 2016; Accessed: March 16, 2016.
  41. Ericzon B-G, Wilczek HE, Larsson M, et al. Liver transplantation for hereditary transthyretin amyloidosis: after 20 years still the best therapeutic alternative?. Transplantation. 2015. 99:1847-1854. [View Abstract]
  42. Beirao JM, Maleiro J, Lemos C, Beirao I, Costa P, Torres P. Ophthalmological manifestations in hereditary transthyretin (ATTR V30M) carriers: a review of 513 cases. Amyloid. 2015. 22:117-122. [View Abstract]
  43. aus dem Siepen F, Bauer R, Aurich M, Buss SJ, Steen H, Altland K, et al. Green tea extract as a treatment for patients with wild-type transthyretin amyloidosis: an observational study. Drug Design, Development and Therapy. 2015 Dec 4. v9:6319–6325. [View Abstract]
  44. Desai HV, Aronow WS, Peterson SJ, et al. Cardiac Amyloidosis: Approaches to Diagnosis and Management. Cardiology in Review. Jan 2010. 18:1-11. [View Abstract]
  45. Ericzon BG, Larsson M, Herlenius G, Wilczek HE. Report from the Familial Amyloidotic Polyneuropathy World Transplant Registry (FAPWTR) and the Domino Liver Transplant Registry (DLTR). Amyloid. 2003 Aug. 10 Suppl 1:67-76. [View Abstract]
  46. Damas AM, Saraiva MJ. Review: TTR amyloidosis-structural features leading to protein aggregation and their implications on therapeutic strategies. J Struct Biol. 2000 Jun. 130(2-3):290-9. [View Abstract]
  47. Monteiro E, Freire A, Barroso E. Familial amyloid polyneuropathy and liver transplantation. J Hepatol. 2004 Aug. 41(2):188-94. [View Abstract]
  48. Lauro A, Diago Usò T, Masetti M, Di Benedetto F, Cautero N, De Ruvo N, et al. Liver transplantation for familial amyloid polyneuropathy non-VAL30MET variants: are cardiac complications influenced by prophylactic pacing and immunosuppressive weaning?. Transplant Proc. 2005 Jun. 37(5):2214-20. [View Abstract]
  49. Klabunde T, Petrassi HM, Oza VB, et al. Rational design of potent human transthyretin amyloid disease inhibitors. Nat Struct Biol. 2000 Apr. 7(4):312-21. [View Abstract]
  50. Tojo K, Sekijima Y, Kelly JW, et al. Diflunisal stabilizes familial amyloid polyneuropathy-associated transthyretin variant tetramers in serum against dissociation required for amyloidogenesis. Neurosci Res. 2006. 56:441-449. [View Abstract]
  51. Palha JA, Ballinari D, Amboldi N, Cardoso I, Fernandes R, Bellotti V, et al. 4'-Iodo-4'-deoxydoxorubicin disrupts the fibrillar structure of transthyretin amyloid. Am J Pathol. 2000 Jun. 156(6):1919-25. [View Abstract]
  52. Peterson SA, Klabunde T, Lashuel HA, et al. Inhibiting transthyretin conformational changes that lead to amyloid fibril formation. Proc Natl Acad Sci U S A. 1998 Oct 27. 95(22):12956-60. [View Abstract]
  53. Saraiva MJ. Transthyretin amyloidosis: a tale of weak interactions. FEBS Lett. 2001 Jun 8. 498(2-3):201-3. [View Abstract]
  54. Suhr OB, Herlenius G, Friman S. Liver transplantation for hereditary transthyretin amyloidosis. Liver Transpl. 2000 May. 6(3):263-76. [View Abstract]

Congo Red staining of a cardiac biopsy specimen containing amyloid, viewed under polarized light.

Congo Red staining of a cardiac biopsy specimen containing amyloid, viewed under polarized light.

Congo Red staining of a cardiac biopsy specimen containing amyloid, viewed under polarized light.

Variant Geographic Focus (Ethnic Origin) Organs Involved
Gly6SerCaucasianNone
Cys10ArgUnited States (Hungarian)H, PN, AN, E
Leu12ProUnited KingdomCNS, AN, L, LM
Asp18GlyUnited States (Hungarian)CNS, LM
Met13IleGermanyNone
Asp18AsnUnited StatesH
Asp18GluSouth AmericaAN, PN
Asp18GlyHungaryLM
Val20IleUnited States, GermanyH, CL
Ser23AsnUnited States (Portuguese)H, E, PN
Pro24SerUnited StatesPN, H, CL
Ala25SerUnited StatesPN, H, CL
Ala25ThrJapanCNS, PN
Val28MetPortugalAN, PN
Val30MetArgentina, Brazil, China, Finland, France, Germany, Greece, Italy, Japan, Portugal, Sweden, Turkey, United StatesPN, AN, E, LM
Val30AlaUnited States (German)AN, H
Val30LeuJapan, United StatesPN, AN, H, K
Val30GlyUnited StatesE, CNS, LM
Phe33CysUnited StatesCL, E, K, H
Phe33IleIsrael (Polish, Ashkenazi Jewish)PN, E
Phe33LeuUnited States (Polish, Lithuanian)PN, AN, H
Phe33ValUnited Kingdom, Japan, ChinaPN
Arg34ThrItalyPN, H
Lys35AsnFrancePN, H, AN
Ala36ProGreece, Italy, United States (Jewish)PN, E, CNS, CL
Asp38AlaJapanH, PN, AN
Trp41LeuUnited States (Russian)E, PN
Glu42GlyJapan, Russia, United StatesPN, AN, H
Glu42AspFranceH
Phe44SerUnited States, JapanPN, H, AN, E
Ala45ThrItaly, Ireland, United StatesH
Ala45AspUnited States , Ireland, ItalyPN, H
Ala45SerSwedenH
Gly47AlaItaly, Germany, FrancePN, H, AN
Gly47ArgJapanPN, AN
Gly47ValSri LankaH, AN, PN, CL
Gly47GluGermany, Italy, Turkey, United StatesH, K, PN, AN
Thr49AlaFrance, Italy (Sicily)PN, CL, H
Thr49IleJapanPN, H
Thr49ProUnited StatesH
Ser50ArgJapan, France, ItalyPN, H, AN
Ser50IleJapanPN, H, AN
Glu51GlyUnited StatesH
Ser52ProUnited KingdomPN, AN, H, K
Gly53GluBasqueCNS, LM, PN, H
Glu54GlyUnited KingdomPN, E, AN
Glu54LysJapanPN, AN, H
Leu55ProUnited States (Dutch, German), TaiwanPN, E, H, AN
Leu55ArgGermanyPN, LM
Leu55GlnUnited States (Spanish)AN, E, PN
Leu58HisUnited States, GermanyH, CL
His56ArgUnited StatesH
Leu58ArgJapanAN, E, CL, H
Thr59LysItaly, United States (Chinese)H, PN, AN
Thr60AlaIreland, United States (Appalachian), Australia, Germany, United Kingdom, JapanH, PN, GI, CL
Glu61LysJapanPN
Phe64LeuItaly, United StatesPN, H, CL
Phe64SerCanada (Italian), United KingdomCNS, PN, E, LM
Ile68LeuGermany, United StatesH
Tyr69HisUnited States, Scotland, CanadaE, LM
Tyr69IleJapanCL, H, AN
Lys70AsnUnited States, GermanyCL, E, PN
Val71AlaFrance, SpainPN, E , CL
Ile73ValBangladeshPN, AN
Asp74HisGermanyNone
Ser77TyrGermany, France, United KingdomPN, H, K
Ser77PheFrancePN, AN, H
Tyr78PheFrance (Italian)PN, CL, S
Ala81ThrUnited StatesH
Ile84SerUnited States (Swiss), HungaryH, CL, E, LM
Ile84AsnItaly, United StatesE, H, CL
Ile84ThrGermany, United KingdomPN, AN, H
Glu89GlnItaly/SicilyPN, H, CL
Glu89LysUnited StatesPN, H, AN
His90AsnPortugal, GermanyNone
Ala91SerFrancePN, H, CL, AN
Gln92LysJapanH
Ala97GlyJapanH, PN
Ala97SerChina, France, TaiwanPN, H
Gly101SerJapanNone
Arg103SerUnited StatesH
Pro102ArgGermanyNone
Arg104CysUnited StatesNone
Arg104HisJapan, United States (Chinese)None
Ile107MetGermanyH, PN
Ile107ValUnited States(German), JapanPN, H, CL
Ala109ValUnited StatesNone
Ala108AlaPortugalNone
Ala109ThrPortugalNone
Ala109SerJapanPN, AN
Leu111MetDenmarkH, CL
Ser112IleItalyPN, H
Tyr114CysHolland, JapanPN, E, H, LM, AN, CNS
Tyr114HisJapanCL, S
Tyr116SerFrancePN, CL, AN
Thr119MetUnited States, PortugalNone
Ala120SerAfro-CaribbeanPN, H, AN
Val122IleAfrica, United States, PortugalH
Val122AlaUnited States (Alaska), United KingdomPN, H, E
Deletion of 122ValEcuador, United States, SpainPN, CNS, GI, CL, H
Pro125SerItalyNone
MethodMaterialSensitivitySpecificityPurpose
Pathologic    
Congo RedTissueMedium/HighHighDetecting amyloid deposits
BSB, FSB dyesTissueHigh MediumDetecting amyloid deposits
Electron microscopyTissueMediumHighConfirming amyloid fibrils
Immunohistochemistry with anti-TTR antibodiesTissueHighMedium/HighDetecting TTR deposits
Genetic    
PCR-RFLPDNAHighHighDetecting predicted mutations in the TTR gene
Real-time PCR (melting curve analysis)DNAHighHighDetecting predicted mutations in the TTR gene
PCR-SSCPDNAMediumMediumScreening for unknown mutations in the TTR gene
SequencingDNAHighHighDetecting unknown mutations in the TTR gene
Mass Spectrometry (MS)    
MALDI-TOF MS, ESI-MSSerum proteinMedium/HighMediumDetecting variant TTR
FT-ICR MSSerum proteinMedium/HighMedium/HighDetecting variant TTR
SELDI-TOF MSSerum proteinMedium/HighMediumDetecting variant TTR
LC-MS/MSTissueMediumMediumIdentifying precursor proteins of amyloid fibrils, including variant TTR