1420706-45-1
Hereditary transthyretin-mediated amyloidosis
Patisiran, marketed under the brand name Onpattro, represents a paradigm shift in the treatment of genetic disorders and marks the clinical translation of a Nobel Prize-winning scientific discovery into a potent therapeutic agent.[1] As the first-ever small interfering RNA (siRNA) therapeutic approved by major global regulatory bodies, Patisiran has fundamentally altered the treatment landscape for hereditary transthyretin-mediated (hATTR) amyloidosis, a rare, progressive, and fatal disease.[4] This report provides a comprehensive analysis of Patisiran, covering its molecular basis, pharmacological profile, clinical development, and its role in the contemporary therapeutic armamentarium.
The mechanism of action of Patisiran is rooted in RNA interference (RNAi), a natural biological process of post-transcriptional gene silencing. The drug is a double-stranded siRNA specifically designed to target the messenger RNA (mRNA) of the transthyretin (TTR) gene. By mediating the catalytic degradation of both mutant and wild-type TTR mRNA within hepatocytes, Patisiran dramatically reduces the hepatic synthesis of the pathogenic TTR protein, thereby addressing the root cause of hATTR amyloidosis.[7] This targeted knockdown of TTR protein production prevents the formation and deposition of amyloid fibrils in tissues, which is the underlying driver of the disease's devastating multisystem pathology.
The clinical efficacy and safety of Patisiran were unequivocally established in the landmark APOLLO Phase 3 clinical trial. This global study demonstrated statistically significant and clinically profound benefits in patients with hATTR amyloidosis with polyneuropathy. Treatment with Patisiran not only halted the relentless progression of neurologic impairment but, remarkably, led to a reversal of neuropathy manifestations from baseline in a majority of patients, as measured by the modified Neuropathy Impairment Score +7 (mNIS+7).[10] Concurrently, patients experienced substantial improvements in quality of life, activities of daily living, and autonomic function, underscoring the drug's holistic, disease-modifying impact.
Administered as an intravenous infusion once every three weeks, Patisiran possesses a well-characterized and manageable safety profile. The most frequently observed adverse events are infusion-related reactions (IRRs) and a reduction in serum vitamin A levels. Both are predictable, mechanism-related effects that are effectively managed through a standardized premedication protocol and routine vitamin A supplementation, respectively.[10]
Patisiran's pioneering journey culminated in historic approvals by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in August 2018, heralding a new era of genetic medicine.[11] While its high cost has positioned it at the center of pharmacoeconomic debates regarding the value of innovation in rare diseases, its clinical impact is undeniable. Patisiran stands as a transformative therapy that offers hope and functional improvement to patients with a previously intractable condition and serves as the definitive proof-of-concept for the entire class of RNAi therapeutics.
A comprehensive understanding of Patisiran requires an appreciation of its precise chemical identity, the pathophysiology of the disease it targets, and the sophisticated formulation science that enables its therapeutic action.
Patisiran is a synthetic, double-stranded small interfering RNA (siRNA) molecule developed and marketed by Alnylam Pharmaceuticals.[5] It is classified as a biotech drug and belongs to the therapeutic classes of transthyretin-directed RNA interaction agents and TTR silencers.[7] Its formal identification and key chemical properties are summarized in Table 1.
Table 1: Patisiran (Onpattro) Key Identifiers and Properties
| Property | Detail | Source(s) |
|---|---|---|
| Generic Name | Patisiran | 7 |
| Brand Name | Onpattro | 4 |
| Drug Type | Biotech; Small Interfering RNA (siRNA) | 7 |
| Manufacturer | Alnylam Pharmaceuticals, Inc. | 5 |
| CAS Number | 1420706-45-1 | 4 |
| DrugBank ID | DB14582 | 4 |
| UNII | 50FKX8CB2Y | 4 |
| Chemical Name | RNA (A-U-G-G-A-A-Um-A-C-U-C-U-U-G-G-U-Um-A-C-dT-dT), complex with RNA (G-Um-A-A-Cm-Cm-A-A-G-A-G-Um-A-Um-Um-Cm-Cm-A-Um-dT-dT) (1:1) | 18 |
| Molecular Formula | C412H520N148O290P40 | 4 |
| Molar Mass | 13424.388 g·mol⁻¹ | 4 |
The drug is also available as Patisiran sodium (CAS: 1386913-72-9, UNII: WO0YM16LKG).[7] The specific chemical structure includes chemically modified nucleotides, such as 2'-O-methoxy-modified sugar residues, to enhance stability and optimize its pharmacological properties.[23]
Patisiran was developed to treat hereditary transthyretin-mediated (hATTR) amyloidosis, a rare, autosomal dominant genetic disorder that is relentlessly progressive and ultimately fatal.[4] The disease is estimated to affect approximately 50,000 individuals worldwide, representing a significant unmet medical need.[4]
The molecular pathology of hATTR amyloidosis originates from mutations in the gene encoding the transthyretin (TTR) protein.[8] TTR is a homotetrameric protein synthesized primarily in the liver that serves as a transporter for thyroxine and retinol binding protein.[7] Over 150 different pathogenic mutations in the
TTR gene have been identified.[23] These mutations destabilize the native tetrameric structure of the TTR protein, promoting its dissociation into individual monomers. These monomers are conformationally unstable and prone to misfolding, leading them to aggregate into insoluble, beta-pleated sheet-rich amyloid fibrils.[8]
These amyloid fibrils deposit extracellularly in a wide range of tissues and organs, causing progressive cellular damage and organ dysfunction.[8] The primary sites of deposition are the peripheral and autonomic nervous systems and the heart. This leads to the characteristic clinical manifestations of the disease: a debilitating sensorimotor and autonomic polyneuropathy, and/or a restrictive cardiomyopathy.[8] The therapeutic rationale for Patisiran is to directly intervene at the source of the pathology by halting the production of the TTR protein, thereby preventing the formation of new amyloid deposits.
A crucial aspect of the disease pathology, which informed the design of Patisiran, is that amyloid deposits are not composed solely of mutant TTR. Wild-type (non-mutated) TTR protein can also become incorporated into the growing amyloid fibrils, contributing significantly to the overall amyloid burden.[23] Therefore, an effective therapeutic strategy must address the production of both forms of the protein. Patisiran was strategically designed to target a sequence in the
TTR mRNA that is common to both wild-type and all known mutant variants, allowing it to comprehensively suppress the production of all amyloidogenic TTR protein, regardless of its genetic origin.[7] This design choice not only enhances its efficacy across a broad spectrum of
TTR mutations but also provides the scientific basis for its investigation in wild-type ATTR amyloidosis (ATTRwt), a related non-hereditary condition.
The therapeutic success of Patisiran is inextricably linked to its advanced formulation. The active siRNA molecule is encapsulated within a lipid nanoparticle (LNP), a sophisticated delivery vehicle that overcomes the fundamental biological barriers to RNAi therapeutics.[5] Naked siRNA administered systemically is rapidly degraded by nucleases in the bloodstream and is inefficiently taken up by target cells due to its size and negative charge.[1] The LNP formulation is therefore not merely a passive carrier but an essential enabling technology that is co-equal to the siRNA itself in achieving the drug's therapeutic effect.
The LNP is a multi-component system precisely engineered for stability and targeted delivery. Its key components include:
This formulation serves two critical functions. First, it protects the siRNA payload from enzymatic degradation in the circulation, ensuring it can reach its target tissue intact.[5] Second, and most importantly, it facilitates targeted delivery to hepatocytes. Upon intravenous administration, the LNP circulates in the bloodstream where it is opsonized by apolipoprotein E (ApoE). The ApoE-coated LNP is then recognized by ApoE receptors, which are highly expressed on the surface of hepatocytes. This interaction triggers receptor-mediated endocytosis, leading to the efficient internalization of the LNP and its siRNA cargo into the liver cells, the primary site of TTR production.[9] This targeted delivery mechanism maximizes the drug's concentration at the site of action while minimizing exposure and potential off-target effects in other tissues.
Patisiran exerts its therapeutic effect by harnessing the cell's own natural machinery for gene regulation through a process known as RNA interference (RNAi). This mechanism allows for the highly specific and potent silencing of the TTR gene at the post-transcriptional level, effectively turning off the production of the disease-causing protein.
Once the LNP has delivered the Patisiran siRNA into the cytoplasm of a hepatocyte, a precise molecular cascade is initiated.[8] This process can be broken down into several key steps:
The guide strand within the activated RISC complex is the key to the drug's specificity. It functions as a template, guiding the RISC to find and bind to messenger RNA (mRNA) molecules that have a complementary nucleotide sequence.[8] The Patisiran guide strand was specifically designed to be perfectly complementary to a sequence within the transthyretin mRNA.
This targeting is highly strategic. The binding site is located in the 3' untranslated region (3'UTR) of the TTR mRNA.[9] This region is not translated into protein but plays a role in mRNA stability and regulation. Pathogenic mutations causing hATTR amyloidosis occur in the protein-coding region of the gene. By targeting a highly conserved sequence in the 3'UTR, a single Patisiran molecule can effectively recognize and silence the mRNA transcripts from both the wild-type allele and virtually all known mutant alleles. This design choice bypasses the complexity of the disease's genetic heterogeneity and makes Patisiran a nearly universal therapy for hATTR amyloidosis.
Once the RISC complex binds to the target TTR mRNA, the Ago2 enzyme within the complex acts as a molecular scissor, cleaving the mRNA strand.[8] This cleavage event renders the mRNA non-functional and flags it for rapid degradation by cellular exonucleases. Without the intact mRNA template, the cell's ribosomes cannot synthesize the TTR protein.[8]
The degradation of TTR mRNA leads to a direct, rapid, and profound reduction in the levels of circulating TTR protein.[7] The RNAi mechanism is catalytic in nature; once a single active RISC complex has cleaved one mRNA target, it is released and can proceed to find and cleave additional mRNA molecules.[9] This molecular amplification explains the potent and durable effect of the drug, allowing for a dosing interval of once every three weeks.
Pharmacodynamic studies in patients with hATTR amyloidosis have quantified this effect. A single intravenous dose of 0.3 mg/kg of Patisiran results in a mean reduction of serum TTR levels by approximately 80% within 10 to 14 days.[9] With chronic dosing every three weeks, this potent knockdown is sustained. After 9 and 18 months of treatment, the mean serum TTR reductions were 83% and 84%, respectively, with a mean maximum reduction of 88% observed over the 1.5-year period.[9] This robust pharmacodynamic response was shown to be consistent regardless of patient age, sex, race, weight, or specific
TTR mutation type, and was not significantly affected by mild to moderate hepatic or renal impairment, highlighting the predictability and reliability of its mechanism of action.[9]
The pharmacokinetic (PK) profile of Patisiran—its absorption, distribution, metabolism, and excretion (ADME)—is largely governed by the properties of its LNP delivery system rather than the siRNA molecule itself. This profile is characterized by targeted delivery to the liver and a metabolic pathway that minimizes the potential for common drug-drug interactions.
Patisiran is administered exclusively via intravenous (IV) infusion, typically over a period of approximately 80 minutes.[25] This route of administration ensures complete bioavailability. The drug exhibits linear and dose-proportional pharmacokinetics within the therapeutic dose range.[7]
With chronic dosing of 0.3 mg/kg every three weeks, steady-state concentrations are achieved by 24 weeks.[7] At steady state, the mean maximum plasma concentration (
Cmax) is 7.15 mcg/mL, the mean trough concentration (Ctrough) is 0.021 mcg/mL, and the mean area under the concentration-time curve (AUC) is 184 mcg·hr/mL.[24]
The distribution of Patisiran is highly influenced by the LNP. Over 95% of the drug in circulation remains encapsulated within the LNP complex.[7] This encapsulation is critical for its biodistribution, as the LNP is designed to be taken up primarily by the liver. The steady-state volume of distribution (
Vd) is low at 0.26 L/kg, which is consistent with the drug being largely confined to the plasma compartment and its target organ, with limited distribution into other tissues.[24] In vitro studies have shown that plasma protein binding of Patisiran is minimal, which is expected for a nanoparticle-based therapeutic.[23] The entire PK profile—from its circulation time to its targeted uptake—is a function of the LNP's interaction with the biological environment, particularly apolipoprotein E and its receptors on hepatocytes.
The metabolic pathway of Patisiran is a key feature that distinguishes it from small-molecule drugs and contributes to its favorable safety profile. As a ribonucleic acid, Patisiran is not metabolized by the cytochrome P450 (CYP) enzyme system, which is the primary pathway for the metabolism of most conventional drugs.[24] Instead, the siRNA component is metabolized by ubiquitous, non-specific nuclease enzymes into smaller, inactive nucleotide fragments of various lengths.[16] This degradation pathway is predictable and has a very low potential for causing or being affected by drug-drug interactions involving the CYP system. This is a significant clinical advantage, particularly for the often-older patient population with hATTR amyloidosis who may be on multiple concomitant medications for comorbidities.
The elimination of Patisiran from the body is primarily driven by this metabolic degradation. The effective half-life (t1/2) of the drug in plasma is approximately 3.2 days, reflecting the clearance of the LNP-siRNA complex.[24] Steady-state clearance is approximately 3.0 L/hr.[24] Excretion of the unchanged drug via the kidneys is negligible, with less than 1% of the administered dose being recovered in the urine.[16] This minimal reliance on renal excretion explains why dosage adjustments are not required in patients with mild or moderate renal impairment.
The pharmacokinetics of Patisiran have been evaluated in several special populations to guide dosing recommendations:
The clinical development of Patisiran culminated in the APOLLO program, a series of Phase 3 trials that provided definitive evidence of its efficacy and safety. The results of these trials not only secured the drug's approval but also established a new benchmark for therapeutic outcomes in hATTR amyloidosis.
The cornerstone of Patisiran's clinical evidence is the APOLLO Phase 3 trial, a global, randomized, double-blind, placebo-controlled study designed to evaluate its efficacy in patients with hATTR amyloidosis with polyneuropathy.[11] The robust design and diverse patient population of this trial lend significant weight to its findings.
The study enrolled 225 patients who were randomized in a 2:1 ratio to receive either Patisiran (n=148) or a placebo (n=77).[12] Patisiran was administered at a dose of 0.3 mg/kg via intravenous infusion once every three weeks for a duration of 18 months.[12] A key strength of the trial was its global reach, enrolling patients from 19 countries with 39 different
TTR genotypes. This broad inclusion criteria ensured that the study population was representative of the diverse clinical and genetic spectrum of the disease seen in real-world practice, thereby enhancing the generalizability of the results.[12]
The APOLLO trial met its primary and all key secondary endpoints with exceptional statistical significance, demonstrating a profound therapeutic effect. The results, summarized in Table 2, represent a fundamental shift in the management of this disease.
Table 2: Summary of Key Efficacy Endpoints from the APOLLO Phase 3 Trial (18 Months)
| Endpoint | Baseline, Mean (SD) | Change from Baseline at 18 Months, LS Mean (SEM) | Treatment Difference (Patisiran - Placebo), LS Mean (95% CI) | p-value |
|---|---|---|---|---|
| Patisiran (N=148) | Placebo (N=77) | Patisiran | Placebo | |
| mNIS+7 (Primary) | 80.9 (41.5) | 74.6 (37.0) | -6.0 (1.7) | +28.0 (2.6) |
| Norfolk QoL-DN (Key Secondary) | 59.6 (28.2) | 55.5 (24.4) | -6.7 (1.8) | +14.4 (2.7) |
Data adapted from.[14] mNIS+7: modified Neuropathy Impairment Score +7 (range 0-304, higher score indicates worse impairment). Norfolk QoL-DN: Norfolk Quality of Life-Diabetic Neuropathy score (range -4 to 136, higher score indicates worse quality of life). LS Mean: Least-Squares Mean. SEM: Standard Error of the Mean. CI: Confidence Interval.
The primary endpoint was the change from baseline in the modified Neuropathy Impairment Score +7 (mNIS+7), a composite measure of neurologic impairment. The results were striking. While the placebo group experienced a substantial and expected worsening of their condition, with an average increase of 28.0 points, the Patisiran group demonstrated an average improvement, with a mean decrease of 6.0 points.[14] The 34.0-point difference between the groups was highly statistically significant (
p<0.001) and clinically transformative.[28]
This outcome represents a paradigm shift. Historically, the goal of treatment for progressive neurodegenerative diseases was to slow the rate of decline. The APOLLO data demonstrated that Patisiran could not only halt progression but actively reverse existing neurologic impairment. This was further evidenced by a key secondary analysis showing that 56% of patients treated with Patisiran experienced an improvement in their mNIS+7 score from their own baseline, a remarkable achievement for a disease previously characterized by inevitable decline.[11] This finding redefined therapeutic expectations, offering patients the potential to regain lost function.
The benefits extended to the key secondary endpoint, the Norfolk Quality of Life-Diabetic Neuropathy (QoL-DN) score. Patisiran-treated patients reported a significant improvement in their quality of life, in stark contrast to the rapid deterioration reported by the placebo group.[10] The strong correlation observed between the degree of TTR protein knockdown and the improvement in clinical measures like mNIS+7 further solidified the evidence, confirming that the observed benefits were a direct consequence of the drug's intended mechanism of action.[12]
Beyond the primary and key secondary endpoints, Patisiran demonstrated consistent and statistically significant benefits across a comprehensive range of measures, reflecting a holistic impact on the patient's condition. These included:
While the primary focus of the APOLLO trial was polyneuropathy, many patients with hATTR amyloidosis also have cardiac involvement. A pre-specified analysis of a cardiac subpopulation within the APOLLO study revealed that Patisiran treatment was associated with favorable effects on exploratory cardiac endpoints, including cardiac biomarkers and echocardiographic parameters, when compared to placebo.[12]
These encouraging findings prompted the APOLLO-B Phase 3 study (NCT03997383), which was specifically designed to evaluate the efficacy of Patisiran in patients with the cardiomyopathy of ATTR amyloidosis (both hereditary and wild-type).[2] The APOLLO-B trial successfully met its primary endpoint, demonstrating a statistically significant and clinically meaningful benefit in functional capacity, as measured by the 6-Minute Walk Test (6-MWT) at 12 months, compared to placebo.[2] The trial also met its first secondary endpoint, showing a significant improvement in health status and quality of life as measured by the Kansas City Cardiomyopathy Questionnaire (KCCQ-OS).[2] Furthermore, exploratory analyses of cardiac biomarkers like NT-proBNP and Troponin I, as well as echocardiographic measures of cardiac structure and function, all favored Patisiran over placebo.[2] These results support the hypothesis that TTR reduction is a viable and effective therapeutic strategy for the cardiac manifestations of the disease.
The clinical utility of a therapeutic agent is determined not only by its efficacy but also by its safety and tolerability. Patisiran has a well-characterized safety profile, with its most notable adverse effects being predictable, mechanism-related, and manageable through established clinical protocols.
Infusion-related reactions (IRRs) are a recognized risk with intravenously administered complex drugs, particularly nanoparticle-based formulations. In the controlled APOLLO study, IRRs were observed in 19% of patients treated with Patisiran, compared to 9% of patients in the placebo group.[10] The majority of these reactions were mild to moderate in severity.[12] Common symptoms include flushing, back pain, nausea, abdominal pain, dyspnea (shortness of breath), and headache.[10]
These reactions are believed to be related to the immune system's response to the LNP components. Due to their predictable nature, a proactive mitigation strategy is employed. A mandatory premedication regimen is a critical component of the administration protocol to reduce the risk and severity of IRRs. This regimen, detailed in Table 3, must be administered at least 60 minutes prior to the start of each infusion.[18]
Table 3: Recommended Premedication Regimen for Patisiran Infusion
| Drug Class | Example Drug(s) | Recommended Dose | Route of Administration |
|---|---|---|---|
| Corticosteroid | Dexamethasone phosphate | 10 mg (or equivalent) | Intravenous (IV) |
| Acetaminophen | Acetaminophen/Paracetamol | 500 mg | Oral |
| H1 Blocker | Diphenhydramine hydrochloride | 50 mg (or equivalent) | Intravenous (IV) |
| H2 Blocker | Ranitidine / Famotidine | 50 mg / 20 mg (or equivalent) | Intravenous (IV) |
Note: Premedications must be administered at least 60 minutes prior to infusion. Oral equivalents may be used if IV preparations are not available or tolerated. Corticosteroid dose may be reduced in tolerating patients. [10]
During the infusion, patients are monitored for signs and symptoms of an IRR. If a reaction occurs, the infusion rate can be slowed or temporarily interrupted. Once symptoms resolve, the infusion may be resumed at a slower rate. In the rare event of a serious or life-threatening IRR, the infusion must be discontinued permanently.[10] This standardized management protocol has proven effective in making the administration of Patisiran safe and tolerable for the vast majority of patients.
The reduction in serum vitamin A levels is a predictable, on-target pharmacological effect of Patisiran.[7] TTR is the primary carrier protein for retinol-binding protein, which in turn transports vitamin A (retinol) in the circulation. By significantly reducing TTR levels, Patisiran indirectly disrupts this transport system, leading to lower measurable levels of vitamin A in the blood.[7]
To manage this effect, all patients receiving Patisiran are advised to take a daily oral supplement of vitamin A at the recommended daily allowance (RDA), which is approximately 2500 IU.[10] It is important to note that serum vitamin A levels during treatment do not accurately reflect the total amount of vitamin A stored in the body. Therefore, patients should not be given doses higher than the RDA in an attempt to normalize their serum levels, as this could lead to vitamin A toxicity.[10] Patients who develop ocular symptoms that could be suggestive of vitamin A deficiency, such as xerophthalmia (dry eyes) or nyctalopia (night blindness), should be referred to an ophthalmologist for evaluation.[13]
Overall, Patisiran demonstrated an encouraging safety and tolerability profile in its pivotal clinical trials. The frequency of serious adverse events and deaths was similar in the Patisiran and placebo arms of the APOLLO study, indicating that the drug did not increase the risk of severe complications.[12]
The most common adverse reactions that occurred more frequently in the Patisiran group compared to placebo were upper respiratory tract infections (29% vs. 21%) and infusion-related reactions (19% vs. 9%).[10] Other adverse events reported in Patisiran-treated patients included peripheral edema (swelling of the limbs), dyspepsia, dyspnea, muscle spasms, arthralgia (joint pain), and bronchitis.[18]
For a therapy intended for chronic, lifelong administration, long-term safety is paramount. Any therapeutic that interacts with fundamental genetic processes raises theoretical concerns about potential long-term risks. Extensive nonclinical toxicology studies were conducted to address these concerns. The results were reassuring, showing no evidence that Patisiran is carcinogenic, mutagenic, or causes impairment of fertility.[16] This suggests that the high specificity of the siRNA for its target minimizes the risk of off-target effects that could lead to deleterious long-term consequences.
The potential for immunogenicity was also assessed. Anti-drug antibodies (ADAs) have been detected in some patients. Notably, these antibodies were found to be specific to one of the lipid components of the LNP (DLin-MC3-DMA), not to the siRNA molecule itself.[7] To date, the presence of these antibodies does not appear to have any impact on the clinical efficacy, safety, pharmacokinetics, or pharmacodynamics of Patisiran. However, the available data are still limited, and this remains an area of ongoing observation.[7]
The journey of Patisiran from laboratory concept to approved medicine was a landmark achievement, not only for the treatment of hATTR amyloidosis but for the entire field of genetic medicine. Its path was characterized by expedited regulatory reviews and a subsequent intense debate on its value and cost.
Recognizing the high unmet medical need in hATTR amyloidosis and the innovative nature of Patisiran's mechanism, regulatory agencies granted it multiple special designations to facilitate its development and review. The U.S. Food and Drug Administration (FDA) granted Patisiran Orphan Drug designation on June 14, 2012, followed by Fast Track, Priority Review, and Breakthrough Therapy designations.[4] These pathways are reserved for drugs that treat serious conditions and demonstrate the potential for substantial improvement over available therapies.
On August 10, 2018, the FDA granted full approval to Onpattro (patisiran) for the treatment of the polyneuropathy of hereditary transthyretin-mediated amyloidosis in adults.[11] This decision was historic, marking the first-ever approval of an RNAi-based therapeutic and the first FDA-approved treatment specifically for this indication.[1]
The regulatory momentum was mirrored in Europe. The European Medicines Agency (EMA) reviewed Patisiran under its accelerated assessment procedure, a pathway granted to medicines deemed to be of major public health interest and therapeutic innovation.[29] Shortly after the FDA decision, on
August 27, 2018, the European Commission granted a centralized marketing authorization for Onpattro for the treatment of hATTR amyloidosis in adult patients with stage 1 or stage 2 polyneuropathy.[14] The rapid, nearly concurrent approvals by the world's two most influential regulatory bodies signaled a strong global consensus on the drug's compelling benefit-risk profile and its transformative impact, based on the strength of the APOLLO trial data.
Following its approvals in the United States and the European Union, Patisiran has gained marketing authorization in numerous other countries. Key approvals include Canada and Japan, where it was approved in June 2019.[3] In Japan, Patisiran also received orphan drug designation, which provides a 10-year period of market exclusivity.[3]
Under a strategic collaboration, Alnylam Pharmaceuticals is responsible for commercializing Patisiran in the U.S., Canada, and Western Europe. Sanofi Genzyme holds the commercialization rights for the rest of the world, including regions in Asia and Latin America.[29]
The clinical breakthrough of Patisiran was accompanied by a significant economic challenge. As a pioneering therapy for a rare disease, it was launched with a very high price tag. The initial list price in the United States was approximately $450,000 per year, with an expected net price of around $345,000 after rebates and discounts.[4] The annual per-patient cost can range from approximately $451,000 to $677,000, depending on the patient's body weight and the corresponding number of vials required for dosing.[5]
This pricing has placed Patisiran at the forefront of the societal debate over the value and affordability of novel treatments for rare genetic diseases. Independent bodies, such as the Institute for Clinical and Economic Review (ICER) in the U.S., have scrutinized the drug's cost-effectiveness. ICER's analysis concluded that, despite its clinical benefits, the price of Patisiran far exceeded commonly accepted thresholds for cost-effectiveness and would need to be substantially lower to align with the value it provides to patients.[39]
This tension between profound clinical value and high economic cost is a defining characteristic of the orphan drug market. Patisiran serves as a key case study in this ongoing healthcare policy conversation, which grapples with how to reward and incentivize groundbreaking R&D for small patient populations while ensuring the sustainability of healthcare systems. In response to these challenges, the manufacturer has established patient support programs, such as Alnylam Assist®, to provide services that help patients navigate insurance coverage and access financial assistance to mitigate out-of-pocket costs.[40]
The approval of Patisiran in 2018 marked the beginning of a therapeutic revolution for hATTR amyloidosis. In the years since, the landscape has evolved rapidly, transforming a condition with virtually no disease-modifying options into one with a complex and growing armamentarium. Understanding Patisiran's role requires a comparative analysis of the available therapeutic strategies.
Modern therapeutic approaches for hATTR amyloidosis can be categorized into two primary mechanisms of action, with a third emerging:
Patisiran is a cornerstone of the TTR gene silencing strategy. The rapid development of multiple drugs across these categories has shifted the clinical challenge from a lack of effective treatments to one of optimal therapy selection, sequencing, and personalization based on patient characteristics and disease phenotype.
Patisiran was the first TTR silencer approved, but it has been joined by several others, each with distinct features related to its molecular class, administration, and dosing frequency.
TTR stabilizers represent a fundamentally different therapeutic approach, aiming to preserve the function of the correctly folded protein rather than eliminating its production.
The availability of these diverse agents has created a nuanced decision-making framework for clinicians. The choice of therapy depends on a multitude of factors, including the predominant clinical phenotype (neuropathy vs. cardiomyopathy), disease severity, patient comorbidities, route of administration preference, and payer coverage. A comparative overview is presented in Table 4.
Table 4: Comparative Overview of Disease-Modifying Therapies for hATTR Amyloidosis
| Drug Name (Brand) | Mechanism of Action | Primary Indication(s) | Route of Administration | Dosing Frequency | Key Clinical Considerations |
|---|---|---|---|---|---|
| Patisiran (Onpattro) | TTR Silencer (siRNA) | Polyneuropathy of hATTR | Intravenous (IV) Infusion | Every 3 weeks | Landmark trial showed reversal of neuropathy; requires premedication and clinic-based infusion. |
| Inotersen (Tegsedi) | TTR Silencer (ASO) | Polyneuropathy of hATTR | Subcutaneous Injection | Once weekly | Self-administered convenience; requires monitoring for thrombocytopenia and glomerulonephritis. |
| Vutrisiran (Amvuttra) | TTR Silencer (siRNA) | Polyneuropathy of hATTR; ATTR Cardiomyopathy | Subcutaneous Injection | Every 3 months | High convenience with infrequent dosing; next-generation siRNA technology. |
| Eplontersen (Wainua) | TTR Silencer (ASO) | Polyneuropathy of hATTR | Subcutaneous Injection | Once monthly | Convenient self-administered monthly dosing. |
| Tafamidis (Vyndamax) | TTR Stabilizer | ATTR Cardiomyopathy; Polyneuropathy of hATTR | Oral Capsule | Once daily | Established efficacy in cardiomyopathy; highest convenience with oral dosing. |
| Acoramidis (Attruby) | TTR Stabilizer | ATTR Cardiomyopathy | Oral Capsule | Twice daily | Next-generation oral stabilizer with strong efficacy in cardiomyopathy. |
Patisiran's potent and proven ability to reverse neuropathy impairment positions it as a foundational therapy for patients with significant polyneuropathy. The development of subcutaneous silencers has introduced options with greater convenience, while oral stabilizers remain a cornerstone, particularly for patients with predominant or exclusive cardiac manifestations. The field continues to evolve, with ongoing studies exploring combination therapies and new therapeutic modalities.
Patisiran (Onpattro) is unequivocally a landmark therapeutic agent. Its approval represents not only a monumental victory for patients with hereditary transthyretin-mediated amyloidosis but also a pivotal moment in the history of medicine. As the first clinically validated RNA interference therapeutic, it has transformed a theoretical biological process into a powerful, life-altering treatment, validating an entirely new class of drugs. The ability of Patisiran to halt and, in many cases, reverse the progression of a devastating neurodegenerative disease has fundamentally altered the natural history of hATTR amyloidosis and redefined the standards of care and therapeutic expectations.
The success of Patisiran extends far beyond its immediate indication. It has served as the definitive clinical proof-of-concept for the entire RNAi platform, demonstrating that it is possible to safely and effectively silence the expression of a specific disease-causing gene in humans with a targeted, durable, and highly potent effect. This achievement has catalyzed a wave of innovation, paving the way for the development of a pipeline of RNAi-based therapies for a wide range of genetic and non-genetic diseases that were previously considered "undruggable."
Looking forward, the journey of TTR-targeted therapies continues to evolve. The positive results from the APOLLO-B study suggest a potential expansion of Patisiran's utility into the treatment of ATTR cardiomyopathy, a major cause of morbidity and mortality in this patient population. Furthermore, the rapid development of second-generation TTR silencers like Vutrisiran, which offer enhanced convenience through less frequent, subcutaneous administration, illustrates the ongoing refinement of this powerful technology. Future research will likely focus on optimizing delivery systems, exploring combination therapies that pair TTR silencers with stabilizers or amyloid removal agents, and expanding the application of RNAi to other organ systems and diseases.
In conclusion, Patisiran is not merely a drug; it is a testament to the power of basic science to yield transformative clinical solutions. It has provided a new lease on life for thousands of patients and has opened the door to a new era of precision genetic medicine, where the root causes of disease can be addressed directly at the molecular level. It stands as a foundational achievement upon which the next generation of genetic therapies will be built.
Published at: August 18, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.