MedPath

Tafamidis Advanced Drug Monograph

Published:Aug 12, 2025

Generic Name

Tafamidis

Brand Names

Vyndamax, Vyndaqel

Drug Type

Small Molecule

Chemical Formula

C14H7Cl2NO3

CAS Number

594839-88-0

Associated Conditions

Cardiomyopathy

Tafamidis (DB11644): A Comprehensive Clinical and Pharmacological Monograph

Executive Summary

Tafamidis represents a landmark achievement in the treatment of transthyretin amyloidosis (ATTR), a progressive, debilitating, and previously untreatable fatal disease. As a first-in-class, orally administered transthyretin (TTR) kinetic stabilizer, tafamidis has fundamentally altered the therapeutic landscape for patients with transthyretin amyloid cardiomyopathy (ATTR-CM). Its mechanism of action directly targets the root cause of the disease—the dissociation of the TTR protein tetramer—by stabilizing its native structure and thereby inhibiting the amyloidogenic cascade.

The clinical value of tafamidis was unequivocally established in the pivotal Phase 3 ATTR-ACT trial, which demonstrated a statistically significant and clinically meaningful reduction in the hierarchical composite of all-cause mortality and cardiovascular-related hospitalizations compared to placebo. Long-term extension data further solidified these findings, revealing a profound survival benefit for patients who received continuous treatment, underscoring the critical importance of early diagnosis and intervention.

Marketed by Pfizer as Vyndaqel® (tafamidis meglumine) and Vyndamax™ (tafamidis), the drug is characterized by a remarkably favorable safety profile, with adverse events in clinical trials being comparable to those observed with placebo. This is particularly significant given the elderly, polymedicated patient population it serves. While its interaction profile with the cytochrome P450 system is minimal, clinicians must be aware of its potential to inhibit drug transporters such as BCRP, which can affect the disposition of co-administered medications like rosuvastatin.

With its proven efficacy, convenient once-daily oral dosing, and established safety, tafamidis has become the entrenched standard of care and a foundational therapy in the management of ATTR-CM. Its development and success have not only provided a life-extending treatment for thousands of patients but have also paved the way for a new era of research into amyloid diseases, with a future that may involve combination therapies aimed at both stabilizing TTR and clearing existing amyloid deposits.

1.0 Introduction to Tafamidis and Transthyretin Amyloidosis

1.1 The Pathophysiology of Transthyretin Amyloidosis (ATTR)

Transthyretin amyloidosis (ATTR) is a systemic, progressive, and life-threatening disease driven by the misfolding of the transthyretin (TTR) protein.[1] TTR is a 127-amino acid protein that is produced primarily in the liver and, to a lesser extent, in the choroid plexus and retinal pigment epithelium. In its normal, functional state, it circulates as a soluble homotetramer—a stable complex of four identical subunits.[2] This structure serves a vital physiological role in the transport of thyroxine (a thyroid hormone) and retinol (vitamin A) via its binding to retinol-binding protein.[2]

The pathogenesis of ATTR is initiated by the dissociation of the stable TTR tetramer into its constituent monomers. This dissociation is the crucial, rate-limiting step in the amyloid cascade.[4] Once dissociated, these monomers are conformationally unstable and prone to misfolding. The misfolded monomers then self-aggregate into a variety of structures, including soluble oligomers, protofilaments, and ultimately, insoluble amyloid fibrils.[5] These amyloid fibrils deposit extracellularly in various tissues and organs, disrupting their normal architecture and function, leading to progressive organ failure and death.[2]

There are two primary subtypes of ATTR, distinguished by the underlying cause of TTR tetramer instability:

  • Hereditary Transthyretin Amyloidosis (ATTRv or hATTR): This form is caused by autosomal dominant pathogenic mutations in the TTR gene, which result in the production of a structurally unstable variant TTR protein that is inherently more prone to dissociation.[4] Over 140 different amyloidogenic mutations have been identified. The clinical presentation and age of onset can vary widely depending on the specific mutation, with symptoms appearing as early as the third decade of life in some individuals.[2] In the United States, the Val122Ile (V122I) mutation is the most prevalent, carried by approximately 3-4% of African Americans, and typically manifests with a cardiac phenotype.[2]
  • Wild-Type Transthyretin Amyloidosis (ATTRwt): This form, formerly known as senile systemic amyloidosis, is not associated with a genetic mutation.[4] Instead, it is believed to result from an age-related process wherein the normal, wild-type TTR protein becomes less stable and begins to dissociate and misfold.[2] ATTRwt primarily affects men over the age of 60 and most commonly manifests as cardiomyopathy.[8]

The clinical manifestations of ATTR are heterogeneous and depend on the primary site of amyloid deposition. This leads to distinct, though often overlapping, clinical phenotypes, most notably ATTR with polyneuropathy (ATTR-PN), characterized by peripheral and autonomic nerve damage, and ATTR with cardiomyopathy (ATTR-CM), characterized by amyloid deposition in the myocardium.[4] In ATTR-CM, the infiltration of amyloid fibrils causes the heart walls to become thick and stiff, leading to restrictive cardiomyopathy, heart failure, and arrhythmias.[8]

Until the advent of targeted therapies, the prognosis for ATTR-CM was exceedingly poor, with treatment limited to supportive care for heart failure symptoms.[9] The median survival from the time of diagnosis was estimated to be just 2 to 3.5 years, highlighting a profound unmet medical need for therapies capable of halting the underlying disease process.[2]

1.2 The Emergence of Tafamidis as a First-in-Class Transthyretin Stabilizer

The development of tafamidis marked a paradigm shift in the management of ATTR, moving the field from purely supportive care to targeted, disease-modifying therapy. Developed by FoldRX, a company later acquired by Pfizer, tafamidis was conceived as a pharmacological chaperone designed to address the root cause of the disease.[1] The therapeutic rationale is elegantly simple: if the dissociation of the TTR tetramer is the first and rate-limiting step of the disease, then preventing this step should halt the entire amyloidogenic cascade.[5]

Tafamidis is classified as a transthyretin stabilizer, a novel class of medication designed specifically for this purpose.[12] Its structure is similar to that of diflunisal, a non-steroidal anti-inflammatory drug (NSAID) that was observed to have TTR-stabilizing properties, but tafamidis was rationally designed for greater potency and selectivity without the associated NSAID-related toxicities.[14] The US Food and Drug Administration (FDA) considers tafamidis to be a first-in-class medication, recognizing its unique mechanism and its role in establishing a new therapeutic category.[4] The successful development of tafamidis was the culmination of decades of meticulous research into the natural history and molecular mechanisms of ATTR, combined with innovative clinical trial design that ultimately led to a breakthrough treatment for this devastating disease.[1]

1.3 Overview of Therapeutic Indications

Tafamidis is approved globally for delaying disease progression in adults with specific forms of transthyretin amyloidosis, although the precise indications vary by regulatory jurisdiction.[4]

  • Transthyretin Amyloid Cardiomyopathy (ATTR-CM): In the United States, Europe, Japan, and other regions, tafamidis is indicated for the treatment of the cardiomyopathy of both wild-type (ATTRwt) and hereditary (ATTRv) transthyretin-mediated amyloidosis in adults. The approved indication is specifically to reduce cardiovascular mortality and cardiovascular-related hospitalization.[8] This indication is supported by the landmark ATTR-ACT trial and is fulfilled by two branded formulations: Vyndaqel® and Vyndamax™.
  • Transthyretin Amyloid Polyneuropathy (ATTR-PN): Prior to its approval for ATTR-CM, tafamidis (as Vyndaqel® 20 mg) was first approved in the European Union (in 2011) and more than 40 other countries for the treatment of ATTR in adult patients with stage 1 symptomatic polyneuropathy to delay peripheral neurologic impairment.[4] While an application was submitted in the US for this indication, it did not receive FDA approval.[25]

2.0 Physicochemical Properties and Formulations

2.1 Chemical Identity and Structure

Tafamidis is a synthetic, non-NSAID, small molecule benzoxazole derivative.[14] Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is

2-(3,5-Dichlorophenyl)-1,3-benzoxazole-6-carboxylic acid.[4] Structurally, it belongs to the chemical class of 1,3-benzoxazoles and is also classified as a monocarboxylic acid and a dichlorobenzene.[17] As noted, it bears a structural resemblance to diflunisal, another compound known to stabilize TTR.[14] A comprehensive list of its key chemical identifiers and properties is provided in Table 1.

Table 1: Key Drug Identifiers and Physicochemical Properties

PropertyValueSource(s)
DrugBank IDDB116444
CAS Number594839-88-04
Molecular FormulaC14​H7​Cl2​NO3​4
Molar Mass308.11 g/mol (free acid)4
Monoisotopic Mass306.980298509 Da14
SMILESC1=CC2=C(C=C1C(=O)O)OC(=N2)C3=CC(=CC(=C3)Cl)Cl17
InChIKeyTXEIIPDJKFWEEC-UHFFFAOYSA-N17
SynonymsPF-06291826, FX-1006, Fx-100514

2.2 Physical and Chemical Properties

Tafamidis exists as a white to pink crystalline powder.[26] The molecule can form two distinct crystalline polymorphs as well as an amorphous form; the manufactured drug product utilizes one of the specific crystalline forms.[4] It is characterized as being slightly soluble in water.[4] Laboratory data indicate it is soluble in organic solvents such as dimethyl sulfoxide (DMSO) at concentrations up to 5 mg/mL and in ethanol at 1 mg/mL.[26] Under appropriate storage conditions, the compound is highly stable, with a shelf life of at least four years.[27]

2.3 Marketed Formulations: Tafamidis vs. Tafamidis Meglumine (Vyndamax™ vs. Vyndaqel®)

Tafamidis is commercially available from Pfizer under two distinct brand names, which correspond to two different salt formulations of the same active moiety. This distinction is critical for correct prescribing and dispensing.[4]

  • Vyndaqel®: This formulation contains tafamidis meglumine, an organoammonium salt created by combining one molar equivalent of tafamidis with meglumine, which is 1-deoxy-1-(methylamino)-D-glucitol.[3] Vyndaqel® is supplied as 20 mg soft gelatin capsules. These capsules are yellow and imprinted with "VYN 20" in red ink.[6] The approved dose for ATTR-CM is 80 mg once daily, which requires the patient to take four 20 mg capsules.[30]
  • Vyndamax™: This formulation contains tafamidis free acid, without the meglumine salt component.[4] Vyndamax™ is supplied as a single 61 mg soft gelatin capsule. This capsule is reddish-brown and imprinted with "VYN 61" in white ink.[22] The approved dose for ATTR-CM is one 61 mg capsule once daily.[30]

The development of the 61 mg Vyndamax™ capsule represents a deliberate, patient-centric evolution in the product's lifecycle. The original 80 mg Vyndaqel® dose, requiring four capsules, presented a significant pill burden, particularly for an elderly patient population that is often managing multiple comorbidities with numerous medications. The single-capsule Vyndamax™ formulation was specifically developed to improve patient convenience and likely enhance long-term adherence to this life-saving therapy.[10]

Clinical pharmacology studies established that the 61 mg dose of tafamidis free acid (Vyndamax™) is therapeutically equivalent to the 80 mg dose of tafamidis meglumine (Vyndaqel®).[3] It is imperative to recognize that the two formulations are

not substitutable on a milligram-per-milligram basis due to the difference in molecular weight between the free acid and the meglumine salt.[5]

Both capsule formulations contain the excipient sorbitol (no more than 44 mg per capsule). The presence of sorbitol can potentially affect the bioavailability of other oral medications administered concomitantly, a consideration for prescribers managing patients on multiple therapies.[5]

3.0 Pharmacological Profile

3.1 Mechanism of Action: Kinetic Stabilization of the Transthyretin Tetramer

Tafamidis is a selective and potent kinetic stabilizer of the transthyretin protein.[1] It functions as a pharmacological chaperone, a small molecule that binds to a target protein to promote its correct folding and structural stability. The mechanism of action of tafamidis is highly specific: it binds with high affinity to one or both of the normally unoccupied thyroxine-binding sites located at the interface between the dimers of the native TTR tetramer.[4] The binding exhibits negative cooperativity, meaning the binding of the first tafamidis molecule to one site reduces the affinity for a second molecule to bind at the other site.[5]

This binding event reinforces the weak interactions that hold the four TTR monomers together, significantly strengthening the quaternary structure of the protein.[4] By stabilizing the native tetrameric conformation, tafamidis slows the rate of tetramer dissociation into individual monomers. As this dissociation is the essential, rate-limiting step in the pathogenesis of ATTR, its inhibition effectively halts the amyloid cascade at its origin.[3] This reduces the available pool of amyloidogenic monomers, thereby preventing their misfolding and aggregation into the toxic amyloid fibrils that cause tissue damage.[14]

3.2 Pharmacodynamics: TTR Stabilization and Downstream Effects

The pharmacodynamic effect of tafamidis is the robust stabilization of the TTR tetramer. A key feature of its clinical utility is its broad efficacy across different forms of the TTR protein. In vitro, ex vivo, and clinical studies have demonstrated that tafamidis effectively stabilizes not only the wild-type TTR protein but also a wide array of amyloidogenic TTR variants. It has been clinically proven to stabilize 14 different variants and has shown ex vivo stabilization for an additional 25 variants, confirming its activity across at least 40 different TTR genotypes.[5] This broad-spectrum stabilizing activity is fundamental to its approval for treating both ATTRwt and the diverse genetic forms of ATTRv.

A predictable pharmacodynamic consequence of its mechanism of action is its effect on thyroid hormone measurements. By occupying the thyroxine-binding sites on TTR, tafamidis competitively displaces thyroxine (T4). This can lead to a measurable decrease in serum concentrations of total T4. However, this laboratory finding is not clinically significant. It is not accompanied by compensatory changes in thyroid-stimulating hormone (TSH) or, more importantly, a change in the level of biologically active free T4.[5] Consequently, this effect is considered a benign laboratory artifact directly related to the drug's mechanism and has not been associated with any clinical evidence of thyroid dysfunction.[3]

3.3 Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The pharmacokinetic profile of tafamidis is well-characterized and notable for features that support a convenient once-daily dosing regimen and a favorable safety profile. A summary of key pharmacokinetic parameters is presented in Table 2.

Table 2: Summary of Pharmacokinetic Parameters

ParameterValue / DescriptionSource(s)
Absorption (Tmax)Maximum plasma concentration is reached in approximately 2 hours under fasted conditions and 4 hours under fed conditions.4
Distribution (Vd)The apparent volume of distribution at steady state is modest at 18.5 L, suggesting distribution primarily within the plasma and well-perfused tissues.14
Protein BindingTafamidis is extensively bound to plasma proteins (>99.9%). Its primary binding partner is its own therapeutic target, the transthyretin protein.4
MetabolismMetabolism is not a major route of elimination. It is not significantly metabolized by the cytochrome P450 (CYP) system. The primary metabolic pathway is glucuronidation via UGT enzymes.3
ExcretionElimination occurs primarily through biliary excretion into the feces. Approximately 59% of an administered dose is recovered in the feces (largely as unchanged drug), and approximately 22% is recovered in the urine (mostly as the glucuronide metabolite).4
Half-life (t½)The terminal elimination half-life is long, approximately 49 hours.3
Clearance (CL)The apparent oral clearance is low, at 0.263 L/h.14

The pharmacokinetic profile of tafamidis reveals several clinically advantageous characteristics. The extremely high degree of plasma protein binding (>99.9%) is a remarkable feature. Because the drug's primary binding partner in plasma is its therapeutic target, TTR, this can be viewed as a "self-targeting" mechanism.[4] This extensive on-target sequestration likely contributes significantly to its favorable safety profile by minimizing the free fraction of the drug available for distribution to off-target tissues. This high level of binding to a stable plasma protein also acts as a reservoir, contributing to the long 49-hour elimination half-life. This long half-life, in turn, ensures that a convenient once-daily dosing regimen is sufficient to maintain steady-state concentrations and near-continuous stabilization of the circulating TTR pool, which is the ultimate therapeutic goal.

Furthermore, the minimal involvement of the cytochrome P450 system in its metabolism is a profound clinical benefit.[4] Tafamidis is primarily metabolized via glucuronidation.[4] This lack of reliance on CYP-mediated pathways significantly reduces the potential for drug-drug interactions with a wide range of medications commonly prescribed to the elderly ATTR-CM population, such as certain statins, beta-blockers, antiplatelet agents, and anticoagulants. This simplifies its clinical use, reducing the need for complex dose adjustments and minimizing the risk of adverse events arising from metabolic interactions. This stands in stark contrast to its significant interactions with drug

transporters, which are a key consideration for clinical management.

4.0 Clinical Efficacy in Transthyretin Amyloid Cardiomyopathy (ATTR-CM)

4.1 The Pivotal ATTR-ACT Trial: Design and Methodology

The definitive evidence for the efficacy of tafamidis in ATTR-CM comes from the Transthyretin Amyloidosis Cardiomyopathy Clinical Trial (ATTR-ACT), a landmark study registered under NCT01994889.[7] ATTR-ACT was a robustly designed Phase 3, multicenter, international, double-blind, placebo-controlled, randomized trial that followed patients for a duration of 30 months.[7]

The trial enrolled 441 patients between the ages of 18 and 90 who had a confirmed diagnosis of ATTR-CM, including both the wild-type (ATTRwt) and hereditary (ATTRv) forms, and a history of heart failure.[9] The patient population was representative of the typical ATTR-CM demographic, with a mean age of 75 years and a vast majority (90%) being male.[40] Patients were randomized in a 2:1:2 ratio to one of three arms: tafamidis 80 mg once daily, tafamidis 20 mg once daily, or a matching placebo.[9]

A crucial aspect of the trial's design was its exclusion criteria. Notably, patients with New York Heart Association (NYHA) Class IV heart failure were excluded from participation.[9] This indicates that the trial primarily investigated the efficacy of tafamidis in patients with mild to moderate (NYHA Class I-III) heart failure, and the results may not be directly generalizable to those with the most advanced, end-stage disease.

4.2 Primary and Secondary Endpoint Analysis: Reduction in Mortality and Morbidity

The primary analysis of the ATTR-ACT trial was based on a pre-specified hierarchical method, which prioritized the most clinically meaningful outcomes. It first compared all-cause mortality between the pooled tafamidis groups (80 mg and 20 mg) and the placebo group. If this was statistically significant, it then proceeded to compare the frequency of cardiovascular-related hospitalizations.[9]

The trial met its primary endpoint with resounding success. Tafamidis demonstrated a statistically significant superiority over placebo in reducing the composite of all-cause mortality and cardiovascular-related hospitalizations (p<0.001).[9] The key efficacy outcomes are summarized in Table 3.

Table 3: Key Efficacy Outcomes from the ATTR-ACT Trial (Tafamidis vs. Placebo)

EndpointTafamidis (n=264)Placebo (n=177)Hazard Ratio / Risk Ratio (95% CI)p-valueSource(s)
All-Cause Mortality29.5% (78 deaths)42.9% (76 deaths)HR = 0.70 (0.51 - 0.96)0.02640
CV-Related Hospitalizations0.48 per year0.70 per yearRRR = 0.68 (0.56 - 0.81)<0.00139
6-Minute Walk Test (6MWT)Lower rate of declineHigher rate of declineN/A<0.00139
KCCQ-OS ScoreLower rate of declineHigher rate of declineN/A<0.00139

The results showed a 30% reduction in the risk of all-cause mortality and a 32% reduction in the rate of cardiovascular-related hospitalizations for patients treated with tafamidis compared to placebo.[41] In addition to these hard clinical endpoints, tafamidis also demonstrated significant benefits in key secondary endpoints related to functional capacity and quality of life. Patients in the tafamidis group experienced a significantly lower rate of decline in their 6-minute walk test (6MWT) distance and a slower deterioration in their health status as measured by the Kansas City Cardiomyopathy Questionnaire–Overall Summary (KCCQ-OS) score.[39]

An interesting finding from the trial relates to the timing of the clinical benefit. The survival curves for all-cause mortality began to diverge between the tafamidis and placebo groups at approximately 18 months, while the curves for cardiovascular-related hospitalizations began to separate earlier, at around 9 months.[42] This apparent lag between the drug's immediate biochemical action (TTR stabilization) and the eventual clinical benefit is mechanistically plausible. Tafamidis works by preventing the

formation of new amyloid fibrils; it is not known to actively remove existing amyloid deposits. Since the disease is characterized by the slow, progressive accumulation of these deposits, halting new deposition would not be expected to yield an immediate survival benefit. The clinical advantage only becomes statistically apparent over many months as the disease progression is arrested in the treated group, while the condition of the placebo group continues to decline. This understanding is critical for managing the expectations of both patients and clinicians regarding the timeline of therapeutic effect.

4.3 Subgroup Analyses: Efficacy Across Genotypes and Disease Severity

The beneficial effects of tafamidis were remarkably consistent across all pre-specified subgroups, reinforcing the robustness of the primary findings.[7] The treatment was effective in patients with both wild-type (ATTRwt) and hereditary (ATTRv) forms of the disease, with a similar magnitude of mortality reduction observed in both groups (approximately 40%).[43]

The benefit was also observed across different levels of baseline disease severity. However, while still present, the effect was somewhat less pronounced in patients with more advanced (NYHA Class III) heart failure compared to those with milder (NYHA Class I/II) disease.[3] This observation suggests that while tafamidis can still provide a benefit in later-stage disease, its greatest impact is likely achieved when treatment is initiated earlier in the disease course, before irreversible cardiac damage has occurred.

4.4 Long-Term Extension (LTE) Study: Sustained Efficacy and the Imperative of Early Intervention

Following the completion of the 30-month ATTR-ACT trial, eligible patients were offered enrollment in an open-label long-term extension (LTE) study.[7] This provided a unique opportunity to assess the long-term durability of the treatment effect and, critically, to compare the outcomes of patients who received continuous tafamidis from the start of ATTR-ACT against those who were initially on placebo and then switched to active treatment upon entering the LTE.

The results from the LTE study were striking and provided a powerful confirmation of the long-term efficacy of tafamidis. After a median follow-up of approximately 58 months, the survival benefit for patients in the continuous tafamidis group was substantial and sustained.[7] The key long-term mortality data are summarized in Table 4.

Table 4: Long-Term Mortality Data from the ATTR-ACT LTE Study

OutcomeContinuous Tafamidis (n=176)Placebo to Tafamidis (n=177)Hazard Ratio (95% CI)p-valueSource(s)
All-Cause Mortality44.9% (79 events)62.7% (111 events)0.59 (0.44 - 0.79)<0.0017
Median Survival67.0 months35.8 monthsN/AN/A7
5-Year Survival (est.)53.2%32.4%N/AN/A7

These data are more than just a confirmation of long-term efficacy; they provide a profound clinical lesson. The dramatic difference in median survival—nearly doubling from 35.8 months in the placebo-to-tafamidis group to 67.0 months in the continuous tafamidis group—is a stark, quantifiable demonstration of the irreversible clinical cost associated with delaying treatment.[7] This finding highlights that while initiating tafamidis even in more advanced disease can still provide a survival benefit, the opportunity to achieve the maximum potential benefit is lost if treatment is not started early. This evidence transforms the clinical conversation, shifting the focus from simply whether to treat ATTR-CM to how rapidly and aggressively it can be diagnosed to enable early therapeutic intervention. This has far-reaching implications for the development of clinical practice guidelines, the implementation of new diagnostic pathways, and the health economic arguments supporting the use of the drug.

5.0 Safety, Tolerability, and Risk Management

5.1 Clinical Trial Safety Data: An Adverse Event Profile Comparable to Placebo

One of the most remarkable findings from the pivotal ATTR-ACT trial was the favorable safety and tolerability profile of tafamidis. Across the 30-month study period, the frequency and types of adverse events (AEs) reported in patients treated with either the 20 mg or 80 mg dose of tafamidis were similar to those reported in the placebo group.[10] Furthermore, the rates of study discontinuation due to adverse events were also comparable between the tafamidis and placebo arms, with approximately 6-7% of patients in each group discontinuing treatment.[38]

This "placebo-like" safety profile has led to statements in patient-facing materials and prescribing information that there were "no known side effects" that occurred during treatment with Vyndaqel or Vyndamax in the ATTR-CM clinical trials.[8] This statement requires careful interpretation. The ATTR-CM patient population is typically elderly and suffers from severe, multi-system heart disease, leading to a very high background rate of medical events such as falls, edema, infections, and progressive cardiac failure.[40] In the controlled setting of the ATTR-ACT trial, the

incremental rate of these AEs attributable to tafamidis was not statistically distinguishable from the high rate of events occurring naturally in the placebo group. This does not mean the drug is entirely free of side effects, but rather that its adverse event profile is largely masked by the significant morbidity of the underlying disease itself.

5.2 Post-Marketing Surveillance and Real-World Safety Observations

While the pivotal trial data were reassuring, information from other clinical trials (particularly in the ATTR-PN population), post-marketing surveillance, and long-term extension studies provides a more complete picture of potential adverse events.

  • Commonly Reported AEs: In some clinical trials, particularly those involving patients with polyneuropathy, AEs reported in more than 10% of participants included urinary tract infections, vaginal infections, upper abdominal pain, and diarrhea.[4]
  • Post-Marketing Reports: Diarrhea has been specifically identified as an adverse event in post-marketing reports for Vyndaqel/Vyndamax.[44]
  • Long-Term Extension Study AEs: The LTE study, which followed patients for over five years, reported a wide range of AEs consistent with the natural progression of ATTR-CM in an aging population. The most common categories of events included cardiac disorders (e.g., cardiac failure, atrial fibrillation), infections and infestations (e.g., pneumonia, cellulitis, urinary tract infection), respiratory disorders (e.g., pleural effusion, dyspnea), falls, and gastrointestinal issues (e.g., constipation, nausea, diarrhea).[7]

5.3 Clinically Significant Drug Interactions

The primary source of clinically significant drug-drug interactions with tafamidis is not through metabolic pathways but through its inhibition of key drug transporters. As discussed previously, tafamidis has a minimal effect on the cytochrome P450 enzyme system.[4] However, it is a clinically relevant inhibitor of several transporters that are crucial for the disposition of other drugs.

Table 5: Summary of Clinically Significant Drug Interactions

Transporter InhibitedSubstrate ExamplesClinical ImplicationSource(s)
BCRP (Breast Cancer Resistance Protein)Methotrexate, Rosuvastatin, Imatinib, SulfasalazineCo-administration may increase the plasma exposure of the BCRP substrate, heightening the risk of substrate-related toxicities. In a clinical study, tafamidis doubled the exposure (AUC) of rosuvastatin. Dose adjustment and/or enhanced monitoring of the substrate drug may be necessary.4
OAT1 / OAT3 (Organic Anion Transporters)NSAIDs, Furosemide, Bumetanide, Methotrexate, Oseltamivir, CidofovirInhibition of these transporters, which are critical for the renal excretion of many drugs, may decrease the clearance and increase the exposure of their substrates. This can lead to potential drug-drug interactions.4

The clinical implication of these transporter-mediated interactions is significant. While tafamidis is metabolically "clean," its use requires vigilance regarding the potential for increased toxicity from other medications the patient is taking. For example, a patient stabilized on a dose of rosuvastatin may experience an increased risk of myopathy if tafamidis is initiated without a corresponding adjustment or close monitoring of the statin. This shifts the focus of clinical monitoring from tafamidis itself to the potential side effects of co-administered drugs whose clearance is now impaired.

5.4 Effects on Laboratory Parameters

Aside from the previously described benign effect on total thyroxine levels, tafamidis has been associated with minor, non-clinically significant liver test abnormalities in some cases.[17] Importantly, it has not been linked to instances of clinically apparent, severe liver injury.[17]

5.5 Contraindications and Precautions

  • Contraindications: The FDA-approved prescribing information for Vyndaqel and Vyndamax lists no contraindications.[22]
  • Precautions and Warnings:
  • Pregnancy: Tafamidis is not recommended for use during pregnancy. Animal studies have demonstrated the potential for fetal harm, including embryofetal mortality, reduced fetal body weight, and fetal malformations at exposures relevant to the human dose. Women of reproductive potential should be counseled on the risks and advised to use effective contraception during treatment.[4]
  • Lactation: It is unknown if tafamidis is excreted in human milk, but it is present in the milk of lactating rats. Due to the potential for serious adverse reactions in a breastfed infant, breastfeeding is not recommended during treatment with tafamidis.[4]
  • Hepatic Impairment: No dose adjustment is needed for mild to moderate hepatic impairment. However, tafamidis has not been studied in patients with severe hepatic impairment, and caution is advised in this population.[3]
  • Renal Impairment: While no dose adjustment is required, there are limited clinical data on the use of tafamidis in patients with severe renal impairment (creatinine clearance ≤ 30 mL/min).[33]
  • Organ Transplantation: Tafamidis should be discontinued in patients who undergo a liver transplant. Since the liver is the primary source of the circulating pathogenic TTR protein, transplantation effectively removes the source of the disease, obviating the need for a TTR stabilizer.[4]

6.0 Dosing, Administration, and Use in Specific Populations

6.1 Recommended Dosing and Administration Instructions

The initiation and ongoing management of tafamidis therapy should be conducted under the supervision of a physician with expertise in the diagnosis and treatment of transthyretin amyloid cardiomyopathy.[33] The recommended dosing regimens for the two available formulations for the treatment of ATTR-CM are distinct and not interchangeable on a milligram-for-milligram basis.

Table 6: Dosing Regimens for Tafamidis Formulations for ATTR-CM

Brand NameActive IngredientDoseFormulationAdministrationSource(s)
Vyndamax™Tafamidis (free acid)61 mgOne 61 mg capsuleOrally once daily22
Vyndaqel®Tafamidis Meglumine80 mgFour 20 mg capsulesOrally once daily22

Patients should be instructed to swallow the capsule(s) whole. The capsules must not be cut, crushed, or chewed, as this can alter the pharmacokinetic properties of the drug.[30] Tafamidis can be taken once daily with or without food.[5]

6.2 Guidance for Missed Doses and Special Considerations

  • Missed Dose: If a patient misses a dose, they should be instructed to take it as soon as they remember. However, if it is close to the time of their next regularly scheduled dose, they should skip the missed dose and resume their normal schedule. Patients must be counseled not to take a double dose to make up for a missed one.[30]
  • Vomiting: In the event of vomiting after administration, if an intact capsule is identified in the vomitus, an additional dose may be given. If no capsule is identified, no additional dose is necessary, and the patient should resume their normal dosing schedule the following day.[5]

6.3 Use in Geriatric, Hepatic, and Renal Impairment

A significant practical advantage of tafamidis is its simple and robust dosing regimen, which does not require adjustment for the majority of the target patient population. This simplicity is a direct consequence of its pharmacokinetic profile, which relies on non-renal and non-CYP-mediated pathways for elimination. This makes the drug easy to use in the complex, comorbid patient population for which it is intended.

  • Geriatric: No dosage adjustment is required for elderly patients (age ≥65 years). The safety and efficacy of tafamidis were established in a clinical trial population that was predominantly elderly, with a mean age of 75 years.[5]
  • Hepatic Impairment: No dosage adjustment is necessary for patients with mild or moderate hepatic impairment. As it has not been studied in patients with severe hepatic impairment, caution is recommended in this subgroup.[3]
  • Renal Impairment: No dosage adjustment is required for patients with any degree of renal impairment. However, clinical data in patients with severe renal impairment (Creatinine Clearance ≤ 30 mL/min) are limited.[5]

6.4 Use in Pregnancy and Lactation: Risk Summary and Recommendations

The use of tafamidis is not recommended in women who are pregnant or breastfeeding due to potential risks identified in animal studies.

  • Pregnancy: Animal reproduction studies have shown that tafamidis can cause fetal harm, including embryofetal toxicity and malformations.[22] Based on these findings, women of reproductive potential should be advised of the potential risks to a fetus and counseled on the importance of pregnancy planning and the use of effective contraception during treatment.[22]
  • Lactation: Tafamidis is known to be present in the milk of lactating rats. While there are no data on its presence in human milk, it is likely to be excreted there. Due to the potential for serious adverse reactions in a breastfed infant, breastfeeding is not recommended during treatment with tafamidis.[5]

7.0 Regulatory History and Market Landscape

7.1 Developmental Timeline and Key Regulatory Milestones (FDA and EMA)

The regulatory journey of tafamidis was lengthy and marked by different outcomes in different jurisdictions, reflecting the evolving understanding of the disease and the drug's efficacy.

  • Early Development and EU Approval: Tafamidis was developed by FoldRx, which was later acquired by Pfizer.[14] It first gained regulatory approval from the European Medicines Agency (EMA) in November 2011. This initial approval was for Vyndaqel® 20 mg for the treatment of transthyretin amyloidosis in adult patients with stage 1 symptomatic polyneuropathy (ATTR-PN).[10] It was not until December 2019 that the EMA's Committee for Medicinal Products for Human Use (CHMP) adopted a positive opinion for the 61 mg dose for the ATTR-CM indication.[10]
  • US Regulatory Path and FDA Approval: The path to approval in the United States was more complex.
  • In 2012, Pfizer received a Complete Response Letter (CRL) from the FDA for its initial application for the ATTR-PN indication, meaning the drug was not approved for that use in the US at that time.[25]
  • Recognizing the high unmet need in ATTR-CM, the FDA granted Tafamidis Fast Track Designation for this indication in 2017, a process designed to facilitate the development and expedite the review of drugs to treat serious conditions.[1]
  • The New Drug Applications (NDAs) for both Vyndaqel® and the new Vyndamax™ formulation for the treatment of ATTR-CM were submitted to the FDA on November 2, 2018.[35]
  • On May 3, 2019, the FDA approved both Vyndaqel® and Vyndamax™ for the treatment of the cardiomyopathy of wild-type or hereditary transthyretin-mediated amyloidosis in adults to reduce cardiovascular mortality and cardiovascular-related hospitalization.[11] This was a landmark decision, as tafamidis became the first-ever FDA-approved treatment for ATTR-CM.[24]
  • Tafamidis was also granted Orphan Drug Designation in the US, EU, and Japan, a status given to drugs that treat rare diseases, which provides incentives for their development.[1]

7.2 Manufacturer and Commercialization Strategy

Tafamidis is manufactured and marketed globally by Pfizer Inc..[15] A cornerstone of Pfizer's commercial strategy has been a significant investment in disease awareness and education campaigns aimed at healthcare professionals and the public. ATTR-CM has historically been severely underdiagnosed, often mistaken for more common forms of heart failure like hypertensive heart disease or hypertrophic cardiomyopathy.[2]

Pfizer's strategy has been to leverage the powerful clinical data, particularly the long-term extension results, to create a sense of urgency around early diagnosis. The clinical evidence demonstrating that delayed treatment leads to significantly worse survival outcomes provides a compelling rationale for proactive screening and diagnosis.[7] This approach has created a symbiotic relationship between the clinical findings and the commercial strategy: by increasing the rate of early diagnosis, Pfizer not only expands the addressable patient market for its drug but also ensures that patients receive the therapy when it can provide the maximum demonstrated clinical benefit. This mutually reinforcing cycle has been instrumental in the drug's commercial success and has helped establish a strong competitive moat.

7.3 Market Position, Competition, and Cost-Effectiveness Considerations

Since its approval, tafamidis (as Vyndaqel/Vyndamax) has become the "entrenched" market leader for the treatment of ATTR-CM.[52] Its commercial performance has been robust, with sales reaching $2.4 billion in 2022 and analysts projecting a peak revenue of nearly $4 billion by 2029.[52]

Despite this dominant position, the therapeutic landscape is evolving, with several competitors emerging:

  • Acoramidis (BridgeBio): This is another oral TTR stabilizer with a similar mechanism of action to tafamidis. While it has shown strong results in its own clinical trials, it is largely perceived as a "me-too" drug. Its twice-daily dosing regimen is considered a disadvantage compared to the convenient once-daily dosing of tafamidis. It is expected to gain significant market share only if it can offer a substantial cost advantage, as the high price and reimbursement hurdles for tafamidis are a known challenge for patients and healthcare systems.[4]
  • RNA Interference (RNAi) "Silencers" (Alnylam): This class of drugs, including patisiran (Onpattro®) and vutrisiran (Amvuttra®), works via a completely different mechanism. Instead of stabilizing the TTR protein, these therapies "silence" the TTR gene in the liver, reducing the production of the protein itself.[13] They represent a distinct therapeutic approach and are being explored as alternatives or as potential components of future combination therapies.

The high cost of tafamidis has been a point of contention and a barrier to access in some healthcare systems.[13] However, its future as a cornerstone therapy seems secure for the near term. Looking ahead, many experts believe that the optimal treatment for ATTR-CM may involve combination therapies. A future paradigm might involve using a stabilizer like tafamidis to prevent the dissociation of existing TTR, combined with a silencer to reduce the overall protein load. This sentiment, captured by some experts who state that "tafamidis alone is not good enough," reflects a forward-looking perspective where the goal shifts from merely slowing progression to more comprehensively controlling the disease.[13]

8.0 Synthesis and Future Perspectives

8.1 Tafamidis's Transformative Impact on the ATTR-CM Treatment Paradigm

Tafamidis is unequivocally a transformative medicine. It has redefined the prognosis for patients with transthyretin amyloid cardiomyopathy, converting what was once a rapidly fatal condition with only supportive care options into a manageable, chronic disease. As the first-in-class, disease-modifying therapy for ATTR-CM, it has fundamentally altered the natural history of the disease by targeting the molecular root of its pathology.

The clinical achievements of tafamidis are clear and well-documented. The ATTR-ACT trial and its long-term extension study have proven that it significantly reduces all-cause mortality and cardiovascular-related hospitalizations. This robust efficacy is paired with a favorable safety profile that is comparable to placebo, making it suitable for long-term, continuous use in a vulnerable, elderly population. Its convenient once-daily oral administration further solidifies its position as a foundational therapy.

8.2 Unmet Needs and the Evolving Therapeutic Landscape

Despite its success, tafamidis has a "ceiling effect." Its mechanism is to prevent the formation of new amyloid deposits by stabilizing the TTR protein; it does not remove the amyloid fibrils that have already accumulated in the heart and other organs. This represents a key unmet need, particularly for patients who are diagnosed at a more advanced stage of the disease. The exclusion of NYHA Class IV patients from the pivotal trial underscores that those with the most severe disease may derive less benefit from stabilization alone.

This limitation has spurred the development of a new wave of therapies with alternative and potentially complementary mechanisms of action:

  • TTR Silencers (RNAi): Drugs like vutrisiran and patisiran work upstream by reducing the liver's production of the TTR protein, thereby lowering the total amount of protein available to form amyloid.[13]
  • Gene Editing: Investigational therapies using in vivo CRISPR-Cas9 technology aim to make a permanent modification to the TTR gene, offering the potential for a one-time treatment that durably reduces TTR production.[13]
  • Amyloid Fibril Removal: A new frontier of research is focused on developing antibody-based therapies designed to bind to and promote the clearance of existing amyloid deposits from tissues, which could potentially reverse organ damage.

The future of ATTR-CM management is likely to move beyond monotherapy and towards combination or sequential treatment strategies. A plausible future paradigm could involve using a stabilizer like tafamidis as a foundational treatment, with a TTR silencer or gene-editing therapy added to suppress the source protein, and perhaps a fibril-clearing agent to address existing organ damage.

8.3 Concluding Remarks on the Clinical Value and Future Role of Tafamidis

For the foreseeable future, tafamidis will remain a cornerstone of therapy for transthyretin amyloid cardiomyopathy. Its proven mortality benefit, established long-term safety record, and convenient oral once-daily dosing regimen make it the standard of care against which new therapies will be judged.

Perhaps the greatest legacy of tafamidis will be its role as a trailblazer. Its success provided the crucial proof-of-concept that targeting the fundamental molecular basis of amyloidosis is a viable and life-saving therapeutic strategy. This has galvanized the field, paving the way for the development of the next generation of more advanced and potentially curative therapies. As the therapeutic armamentarium expands, the central challenge for the medical community will remain the same one that tafamidis brought to the forefront: the critical need for increased awareness and the early, accurate diagnosis of all patients who can benefit from these transformative treatments.

Works cited

  1. Drug Discovery and Development in Rare Diseases: Taking a Closer Look at the Tafamidis Story, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7987260/
  2. About ATTR Cardiac Amyloidosis | VYNDAMAX (tafamidis) | Safety Info, accessed August 12, 2025, https://vyndamax.pfizerpro.com/about-attrcm/disease-impact
  3. Tafamidis and tafamidis meglumine for amyloid cardiomyopathy - PMC, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8377296/
  4. Tafamidis - Wikipedia, accessed August 12, 2025, https://en.wikipedia.org/wiki/Tafamidis
  5. Vyndaqel, INN-tafamidis - EMA, accessed August 12, 2025, https://www.ema.europa.eu/en/documents/product-information/vyndaqel-epar-product-information_en.pdf
  6. This medicinal product is subject to additional monitoring. This will allow quick identification of - Vyndaqel, INN-tafamidis, accessed August 12, 2025, https://ec.europa.eu/health/documents/community-register/2022/20221109157640/anx_157640_en.pdf
  7. Long-Term Survival With Tafamidis in Patients With Transthyretin ..., accessed August 12, 2025, https://www.ahajournals.org/doi/10.1161/CIRCHEARTFAILURE.120.008193
  8. What is ATTR-CM? | VYNDAMAX® (tafamidis) | Safety Info, accessed August 12, 2025, https://www.vyndamax.com/about-attr-cm
  9. Tafamidis Treatment for Patients with Transthyretin Amyloid ..., accessed August 12, 2025, https://openaccess.sgul.ac.uk/113684/1/nejmoa1805689.pdf
  10. Pfizer Receives Positive CHMP Opinion for VYNDAQEL® for Use in Patients with Transthyretin Amyloid Cardiomyopathy, a Rare and Fatal Disease, accessed August 12, 2025, https://www.pfizer.com/news/press-release/press-release-detail/pfizer_receives_positive_chmp_opinion_for_vyndaqel_for_use_in_patients_with_transthyretin_amyloid_cardiomyopathy_a_rare_and_fatal_disease
  11. U.S. FDA Approves VYNDAQEL® and VYNDAMAX™ for Use in Patients with Transthyretin Amyloid Cardiomyopathy, a Rare and Fatal Disease | Pfizer, accessed August 12, 2025, https://www.pfizer.com/news/press-release/press-release-detail/u_s_fda_approves_vyndaqel_and_vyndamax_for_use_in_patients_with_transthyretin_amyloid_cardiomyopathy_a_rare_and_fatal_disease
  12. medlineplus.gov, accessed August 12, 2025, https://medlineplus.gov/druginfo/meds/a622032.html#:~:text=Tafamidis%20is%20used%20to%20treat,of%20medications%20called%20transthyretin%20stabilizers.
  13. Even on Tafamidis, ATTR-CM Patients Face High Mortality Over Time | tctmd.com, accessed August 12, 2025, https://www.tctmd.com/news/even-tafamidis-attr-cm-patients-face-high-mortality-over-time
  14. Tafamidis: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 12, 2025, https://go.drugbank.com/drugs/DB11644
  15. Vyndaqel® (tafamidis meglumine) - Orsini Specialty Pharmacy, accessed August 12, 2025, https://www.orsini.com/vyndaqel-tafamidis-meglumine/
  16. Tafamidis: MedlinePlus Drug Information, accessed August 12, 2025, https://medlineplus.gov/druginfo/meds/a622032.html
  17. Tafamidis | C14H7Cl2NO3 | CID 11001318 - PubChem, accessed August 12, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Tafamidis
  18. www.ncbi.nlm.nih.gov, accessed August 12, 2025, https://www.ncbi.nlm.nih.gov/books/NBK574508/#:~:text=Tafamidis%20is%20an%20oral%20medication,disease%20progression%20and%20mortality%20significantly.
  19. VYNDAMAX® (tafamidis) Prescription Treatment for ATTR-CM | Safety Info, accessed August 12, 2025, https://www.vyndamax.com/
  20. VYNDAMAX® (tafamidis) | Official HCP Site, accessed August 12, 2025, https://vyndamax.pfizerpro.com/
  21. Annotation of FDA Label for tafamidis and TTR - PharmGKB, accessed August 12, 2025, https://www.clinpgx.org/labelAnnotation/PA166184565
  22. VYNDAQEL and VYNDAMAX - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed August 12, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/211996s001,212161s001lbl.pdf
  23. Search Orphan Drug Designations and Approvals - FDA, accessed August 12, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=363312
  24. Vyndaqel (tafamidis meglumine) and Vyndamax (tafamidis) approved by FDA, accessed August 12, 2025, https://arci.org/vyndaqel-tafamidis-meglumine-and-vyndamax-tafamidis-approved-by-fda-for-attr-amyloidosis-cardiomyopathy/
  25. Vyndaqel (tafamidis meglumine) FDA Approval History - Drugs.com, accessed August 12, 2025, https://www.drugs.com/history/vyndaqel.html
  26. Tafamidis ≥98% (, HPLC) - Sigma-Aldrich, accessed August 12, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml4131
  27. Tafamidis (PF-06291826, CAS Number: 594839-88-0) | Cayman Chemical, accessed August 12, 2025, https://www.caymanchem.com/product/13694/tafamidis
  28. VYNDAQEL® (tafamidis meglumine) Capsules and VYNDAMAX ..., accessed August 12, 2025, https://www.pfizer.com/products/product-detail/vyndaqel_tafamidis_meglumine_capsules_and_vyndamax_tafamidis_capsules
  29. Tafamidis Meglumine | C21H24Cl2N2O8 | CID 24970412 - PubChem, accessed August 12, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Tafamidis-Meglumine
  30. Tafamidis (oral route) - Side effects & dosage - Mayo Clinic, accessed August 12, 2025, https://www.mayoclinic.org/drugs-supplements/tafamidis-oral-route/description/drg-20463401
  31. VYNDAQEL® OR VYNDAMAX® (tafamidis meglumine or tafamidis) Dosage and Administration | Pfizer Medical - US, accessed August 12, 2025, https://www.pfizermedical.com/vyndaqel-or-vyndamax/dosage-admin
  32. tafamidis | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 12, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=8378
  33. Product Information Tafamidis and Tafamidis meglumine - Therapeutic Goods Administration (TGA), accessed August 12, 2025, https://www.tga.gov.au/sites/default/files/auspar-tafamidis-200903-pi-01.docx
  34. Dosing and Side Effects of VYNDAMAX® (tafamidis) | Safety Info, accessed August 12, 2025, https://www.vyndamax.com/dosing-and-safety
  35. 211996Orig1s000 212161Orig1s000 | FDA, accessed August 12, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/211996Orig1s000,%20212161Orig1s000MedR.pdf
  36. pmc.ncbi.nlm.nih.gov, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4919130/#:~:text=Evidence%20showed%20that%20tafamidis%20binds,of%20circulating%20functional%20TTR%20requires
  37. Mechanism of Action and Clinical Application of Tafamidis in, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4919130/
  38. highlights of prescribing information - Pfizer, accessed August 12, 2025, https://labeling.pfizer.com/ShowLabeling.aspx?id=11685
  39. Tafamidis Treatment for Patients with Transthyretin Amyloid Cardiomyopathy - PubMed, accessed August 12, 2025, https://pubmed.ncbi.nlm.nih.gov/30145929/
  40. Tafamidis in Transthyretin Cardiomyopathy Clinical Trial - ATTR-ACT, accessed August 12, 2025, https://www.acc.org/Latest-in-Cardiology/Clinical-Trials/2018/08/25/23/02/ATTR-ACT
  41. Tafamidis Phase 3 Transthyretin Amyloid Cardiomyopathy (ATTR-ACT) Study Results Presented as Late-Breaking Data at the ESC Congress 2018 | Pfizer, accessed August 12, 2025, https://www.pfizer.com/news/press-release/press-release-detail/tafamidis_phase_3_transthyretin_amyloid_cardiomyopathy_attr_act_study_results_presented_as_late_breaking_data_at_the_esc_congress_2018
  42. Tafamidis for Patients with Transthyretin Amyloid Cardiomyopathy: Promising Results, accessed August 12, 2025, https://www.jwatch.org/na47394/2018/08/27/tafamidis-patients-with-transthyretin-amyloid
  43. Long-Term Survival With Tafamidis in Patients With Transthyretin Amyloid Cardiomyopathy, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8763250/
  44. INDICATION VYNDAQEL® (tafamidis meglumine) and VYNDAMAX are indicated for the treatment of the cardiomyopathy of wild-type or h, accessed August 12, 2025, https://vyndamax.pfizerpro.com/api/vc/en/content/material/f4e9215b-91f9-4d15-8ac7-c286dace54ba/VYNDAMAXtafamidis_SafetyInfo_PrescribingInfo_PP-VDM-USA-1281.pdf?download=true
  45. What are the possible side effects of VYNDAQEL and VYNDAMAX? - Pfizer Medical Information, accessed August 12, 2025, https://www.pfizermedicalinformation.com/patient/vyndaqel-or-vyndamax/what-are-the-possible-side-effects-of-vyndaqel-and-vyndamax
  46. IMPORTANT SAFETY INFORMATION AND INDICATION | VYNDAMAX® (tafamidis), accessed August 12, 2025, https://www.vyndamax.com/important/information
  47. pmc.ncbi.nlm.nih.gov, accessed August 12, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC11438280/#:~:text=However%2C%20long%2Dterm%20extension%20(,5%2C%207%2C%208%5D.](https://www.google.com/url?q=https://pmc.ncbi.nlm.nih.gov/articles/PMC11438280/%23:~:text%3DHowever%252C%2520long%252Dterm%2520extension%2520(,5%252C%25207%252C%25208%255D.&sa=D&source=editors&ust=1754986643718473&usg=AOvVaw2fCi0WVora4eicHO_I_QAj)
  48. Tafamidis: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 12, 2025, https://www.rxlist.com/tafamidis/generic-drug.htm
  49. tafamidis | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 12, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=8378
  50. 211996Orig1s000 212161Orig1s000 - accessdata.fda.gov, accessed August 12, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/211996Orig1s000,%20212161Orig1s000Approv.pdf
  51. U.S. FDA Approves VYNDAQEL® and VYNDAMAX™ for Use in Patients with Transthyretin Amyloid Cardiomyopathy, a Rare and Fatal Disease | HFSA, accessed August 12, 2025, https://hfsa.org/us-fda-approves-vyndaqelr-and-vyndamaxtm-use-patients-transthyretin-amyloid-cardiomyopathy-rare-and
  52. Pfizer's 'entrenched' tafamidis franchise will be tough to challenge ..., accessed August 12, 2025, https://www.fiercepharma.com/pharma/pfizer-tafamidis-hard-challenge-despite-potential-heart-disease-rivals-alnylam-bridgebio
  53. Tafamidis | C14H7Cl2NO3 - ChemSpider, accessed August 12, 2025, https://www.chemspider.com/Chemical-Structure.9176510.html
  54. Tafamidis | Transthyretin Stabilizer - StressMarq Biosciences Inc., accessed August 12, 2025, https://www.stressmarq.com/products/small-molecules/inhibitor/tafamidis-sih-584/
  55. Tafamidis Free Acid | CAS#594839-88-0 | CAS#951395-08-7 | Transthyretin Protein Stabilizer - MedKoo Biosciences, accessed August 12, 2025, https://www.medkoo.com/products/8088

Published at: August 12, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.